You are on page 1of 955

NEUROGLIA

This page intentionally left blank


NEUROGLIA
THIRD EDITION

Helmut Kettenmann Bruce R. Ransom


MAX DELBRÜCK CENTER FOR MOLECULAR MEDICINE DE PA RTME NT OF N EUR OLO GY

BERLIN, GERMANY U N I V E R S I T Y O F WA S H I N G T O N S C H O O L O F M E D I C I N E

S E AT T L E , WA

1
3
Oxford University Press is a department of the University of Oxford.
It furthers the University’s objective of excellence in research, scholarship, and education by
publishing worldwide.

Oxford New York


Auckland Cape Town Dar es Salaam Hong Kong Karachi
Kuala Lumpur Madrid Melbourne Mexico City Nairobi
New Delhi Shanghai Taipei Toronto

With offices in
Argentina Austria Brazil Chile Czech Republic France Greece
Guatemala Hungary Italy Japan Poland Portugal Singapore
South Korea Switzerland Thailand Turkey Ukraine Vietnam

Oxford is a registered trade mark of Oxford University Press in the UK and certain other countries.

Published in the United States of America by


Oxford University Press
198 Madison Avenue, New York, NY 10016

© Oxford University Press 2013

All rights reserved. No part of this publication may be reproduced, stored in a retrieval system, or transmitted,
in any form or by any means, without the prior permission in writing of Oxford University Press,
or as expressly permitted by law, by license, or under terms agreed with the appropriate
reproduction rights organization. Inquiries concerning reproduction outside the scope of the above
should be sent to the Rights Department, Oxford University Press, at the address above.

You must not circulate this work in any other form


and you must impose this same condition on any acquirer.

Library of Congress Cataloging-in-Publication Data


Kettenmann, Helmut.
Neuroglia / [edited by] Helmut Kettenmann, Bruce R. Ransom. –3rd ed.
p. ; cm.
Includes bibliographical references and index.
ISBN 978–0–19–979459–1 (hardcover : alk. paper)
I. Kettenmann, Helmut. II. Ransom, Bruce R. III. Title.
[DNLM: 1. Neuroglia. 2. Nervous System Diseases—physiopathology. WL 102]
612.8c1045—dc23
2012016025

9 8 7 6 5 4 3 2 1
Printed in the United States of America
on acid-free paper
To our children, Lucas and Georg Kettenmann, and Rebecca, Christopher and Cole Ransom,
who often missed their busy Dads.
This page intentionally left blank
PREFACE TO THE FIR ST EDITION

Nonneuronal cells, termed neuroglia, were recognized as inde- may be enhanced by including glial cells as factories for the
pendent elements of the nervous system nearly a century and a production of trophic substances. Glial transplants to refur-
half ago by Virchow. These cells are present in primitive nervous bish areas of demyelination may also be possible in the near
systems and, undoubtedly driven by positive evolutionary pres- future. Our capacity to measure the brain’s functional mol-
sures, have persisted in high density and acquired greater diver- ecules and determine their cellular topography has revealed
sity in mammals. Knowledge about glial cells has accumulated a baffling array of neurotransmitters, receptors, ion chan-
at a phenomenal rate in the past 30 years, and has become rel- nels, adhesion molecules, and trophic factors associated with
evant to all fields of neurobiology. With so much new informa- glial cells. These findings are stimulating and broadening the
tion at hand, we felt that this was an important time to assemble field of glial research. They provide critical insights into how
the facts about these cells as we presently understand them. neurons and glial cells might communicate with each other,
Historically, glial cells were viewed as a type of central and reveal an astonishing overlap between the features of the
nervous system connective tissue whose main function was to brain’s two principal cell types that would have been heresy
provide support to the true functional cells of the brain, the not long ago.
neurons. This firmly entrenched concept remained virtually The many experts who wrote chapters for this volume
unquestioned for the better part of a century. But glial cells contributed in other valuable ways as well. Before the writ-
are neither connective tissue nor mere supportive cells. In con- ing began, they provided invaluable advice about what topics
trast to early beliefs, glial cells are now recognized as intimate should be covered. In an unselfish manner, they adjusted the
partners with neurons in virtually every function of the brain scope of their individual contributions so that they fit the con-
and as participants in the pathophysiology of the dysfunc- text of the book as a whole. To enhance the quality and utility
tional or diseased brain. These cells have been challenging to of the chapters, each underwent a stage of peer review and this
study, however, because their functions are not associated with was cheerfully provided by other authors. The editorial burden
easily recorded electrical signals, as is the case with neurons. was significantly lightened by the satisfaction of dealing with
While books about the nervous system have grown in size this uniquely talented and energetically committed group of
and complexity, attempting to accommodate the frantic pro- authors. They share our view of the importance of develop-
duction of new neuroscience information, the incorporation of ing a compendium volume about glial cells and continuously
new facts about glia has not kept pace. One simply cannot learn reinforced our enthusiasm for the project as it moved forward.
about glial cells by turning to the typical neuroscience text- We acknowledge their essential partnership in the making of
book (From Neuron to Brain by Nicholls, Martin, and Wallace, Neuroglia and thank them for their efforts.
is a notable exception). This curious fact has also been a moti- One point should be made in concluding. As impressive as
vation for bringing together in the present volume a detailed our gains in glial cell knowledge have been, the best is yet to
summary of what is currently known about these cells. It will, come. Glial researchers have struggled with our own version
we hope, also encourage better integration of the glial and neu- of the Heisenberg uncertainty principle: how to study the role
ronal information bases, which each suffer in the absence of the of glial cells in the multicellular actions of the nervous system
other. The brain cannot be understood as the functional sum of without interfering with the very functions we wish to under-
two isolated cellular compartments; it must, we think, be seen stand. Our initial efforts were a compromise. We retreated
as a single entity containing neurons and glial cells working in somewhat from the immense complexity of the intact nervous
seamless harmony with one another. Somehow, this essential system in favor of simplified preparations that allowed more
message has gone too long undelivered. rigorous study. This reductionistic approach has produced a
Glial research is at a particularly exciting point in its mountain of provocative information, as detailed here, but
evolution. Great advances in our knowledge about nervous few definitive answers. Consequently, we have long lists of
system diseases have opened the door for thinking about glial cell properties, while the list of proven functions is small.
the role of glial cells in the pathogenesis of these conditions But starting with these demonstrated properties, testable
and in their treatment. Therapies that would literally have hypotheses of glial cell function can now be formulated with
been the stuff of science fiction only a decade ago are now in improved precision, taking full advantage of new or refined
advanced stages of testing. Patients with Parkinson’s disease research technologies. A rich yield of vital new insights into
who no longer respond to our best medicines, for example, the functions of neuroglia should follow, and future editions
have received brain tissue transplants, whose effectiveness of this book will survey those benefits.

vii
This page intentionally left blank
PREFACE TO THE SECOND EDITION

Almost 10 years have passed since the first edition of Neuroglia In many ways the second edition of Neuroglia was more
was published. Neuroglia was warmly received and we were challenging to conceive and produce than the first edition.
pleased that it soon became a standard reference work in this With affordability in mind, we were committed to a second
field. The pace of glial research has continued to accelerate, edition that was smaller than the first edition. We also wanted
however, and we could not ignore the obvious need for a fresh a greater degree of uniformity in terms of subject treatment
assessment and summary of this field. than was true for the first edition. We struggled in the planning
The need for a comprehensive and contemporary book stages with how to condense topic areas without sacrificing
on glial cells has never been greater. All of the motivating fac- crucial content. Accordingly, we urged authors to be concise,
tors mentioned in the preface to the first edition continue to and we limited the number of citations and edited each chap-
operate, including the lack of glial coverage in general neuro- ter based on reviews provided by other contributors. But the
science textbooks. In fact, the practical impact of the second universe of relevant knowledge about glial cells is much larger
edition of Neuroglia may be greater than the first because it today and the need to include new topics while adhering to a
is offered at a time when a majority of neuroscientists would strict page budget came at a price. Some “classic” topics covered
acknowledge that neuroglia are elemental to most, if not all, in detail in the first edition such as glial anatomy and ion chan-
brain functions. This was certainly not the case when the first nel expression were significantly condensed, and the first edi-
edition appeared. tion of Neuroglia will remain useful for exactly that reason.
This book is not simply an edited version of the first edi- We predicted in the first edition preface that “As impres-
tion. We started with the proverbial “clean sheet” of paper in sive as our gains in glial cell knowledge have been, the best is
planning the second edition. Based on advice solicited from yet to come.” The second edition of Neuroglia thoroughly doc-
many of our colleagues, the book was entirely reorganized to uments our progress in understanding these cells and vindi-
more logically assemble current information about glial cells. cates this statement, but we readily admit that this prediction
We recruited many new contributors, reflecting the emer- remains true for the future. Not so long ago it was imagined
gence of new topics and new experts in the field. Less than that glia represented a functionally uniform cell population.
a fourth of the original chapters were retained, and most of This concept was discarded as we recognized that glia are a
these were revised so extensively that they are essentially new diverse and complex cell family whose only common feature
contributions themselves. Novel topics appearing in the sec- is that they are not neurons. Situational plasticity and regional
ond edition include: transmitter release by exocytosis from variability in these cells are emerging themes that promise to
glia, glial derived stem cells, glia and synaptic transmission, further enlarge their range of functions. But the path to the
glia and axon guidance, an entire section on mechanisms of future always starts with a current and accessible base of infor-
glial injury, and several new chapters about the roles of glia in mation. We believe the second edition of Neuroglia provides
different diseases. this important starting point.

ix
This page intentionally left blank
PREFACE TO THE THIRD EDITION

Good reasons must exist to do a third edition of a book. We are calling this book ‘Neuroglia, the third edition’, but
After two editions, the project’s novelty has worn off and any the massive revision and redesign from the second edition
delusions about the glamorous life of a book-editor are dead, could have justified a new name altogether. The changes are
replaced by the grinding reality of assembling and editing a too numerous to detail but some general comments will serve
creation with more than a hundred ‘parents’. We looked hard to illustrate the point. The number of individual chapters has
at the past editions of Neuroglia and asked ourselves if a new been expanded from 47 to 71, a 50% increase, and very few of
edition could possibly be worth the effort. ‘Sprucing up’ the the chapters have been spared a new name and altered content
second edition was not a sufficient reason, even if this might range. At the same time, we pushed for more concise chap-
prove commercially successful. We were simply not motivated ters with a consistent level of presentation. More than 50 new
by the notion of a renewable textbook franchise. What finally authors contributed to this edition guaranteeing the incorpo-
pushed us to action were two undeniable facts: the exciting, ration of new perspectives and knowledge. An unanticipated
upward trajectory of knowledge about glial cells, and the con- benefit of so many new contributors was the lift provided by
tinued void of comprehensive treatises on this subject. their genuine and contagious enthusiasm for this project.
Glial biology as a field has entered a period of extraordi- What is now known about glial cells will not conve-
nary growth and evolution. The number of glial research arti- niently fit in a single volume. In planning the third edition
cles published annually continues to increase, and, while there of Neuroglia, difficult decisions had to be made about what
is no precise metric for judging their scientific impact, it is safe to include, and at what level of detail to present information.
to say that this is rising as well. Some recent developments pro- Compromises were made. We chose to include more topic
vide evidence of this growing vitality. It is now established that areas treated generally rather than a narrower range of topics
in addition to astrocytes, microglia and oligodendrocytes, treated at greater depth. The number of illustrations and refer-
a fourth major class of glial cells exists in the brain, termed ences had to be limited. Our contributors accepted these con-
NG2 cells. The roles of glia in disease are becoming clearer, straints with good humor. We believe the resulting volume has
and remarkably there are no apparent exceptions to the rule achieved the desired balance between breadth, accuracy and
that glial cells participate in all brain pathologies. This is a depth to satisfy the broad range of users we had in mind.
particularly fast-paced area of glial research and one likely to Edited volumes vary greatly in terms of style, tone, and
yield unexpected therapeutic targets for diseases with no pres- content value. Our contributors graciously agreed to more
ent cures and few treatments. Glia may even play important stringent standards and oversight than in previous editions in
mechanistic roles in psychiatric diseases. New findings tell us the interest of a high quality end product. All of the chapters
that glial cells modulate complex brain-mediated behaviors were commissioned and written within about nine months, a
like memory, learning and sleep. These few examples illustrate very ambitious timetable and one that ensured that the con-
the wide range and compelling novelty of the field, and help tent would be up-to-date and relevant when the book was
explain why we acceded to the challenge of creating this third published. Preliminary outlines were solicited and posted on
edition of Neuroglia. Collectively, this scientific progress has a website for review by all participants in an effort to identify
led to a state of current knowledge that is transformative. Many and eliminate redundancies. Each of the 71 chapters was peer-
traditional concepts about glial cells will need to be discarded reviewed by two knowledgeable experts from among our con-
or revised in response to this flood of new information. An tributors, and by the editors. Substantial revisions were often
overarching principle that is now unassailable is that nervous required and no chapter was accepted in final form without
system function is best characterized as a partnership between further review. The logistical issues that arose when there were
glia and neurons.* We hope that this book will faithfully trans- 71 ‘balls in the air’, so to speak, surprised us. The Berlin office**
mit the powerful sense among ‘gliologists’ of forward momen- bore the brunt of the onerous record keeping and communica-
tum in understanding glial functions, and in so doing that it tion, and without their efforts, which were far beyond reason-
will attract and inspire the next generation of glial scientists. able, the third edition would not have appeared on time or in
the well organized form that you see before you.
* The long-held practice of referring to glia as ‘support cells’, therefore, should be
Craig Panner at Oxford University Press (OUP) believed
abandoned in favor of the term ‘partner cells’. in the value of a new edition and was a true partner in its
** We especially want to recognize the superb work of Birgit Jarchow, Cheryl creation. He did not shy from contentious issues that we
Hutton and Lisa Leukel. confronted, sought consensus with us on all the main points

xi
regarding production and worked to bring Neuroglia up to cells partner with neurons in all brain functions. This point of
a modern standard. Consequently, the book is destined to view constitutes a paradigm shift. While the first edition of
appear as a fully electronic volume as part of OUP’s electronic Neuroglia largely focused on glial properties, the theme of the
publishing program. This feature will appeal to contemporary third edition, 17 years later, is more on dynamic interactions
users who will value the greatly enhanced access. It also holds and functions. The myriad roles of glia in brain function are in
open the possibility that over time important content might better focus than ever before, but there are still a vast number
be added as appendices, without the need for formal republi- of unsolved issues in this infinitely satisfying puzzle. The suc-
cation. Reader feedback will influence future decisions on this cess of the third edition of Neuroglia will be measured by the
option. further progress it inspires in our understanding of the harmo-
Past prefaces spoke to the importance of glial cells in neu- nious partnership between glia and neurons underlying brain
roscience. Unconsciously, they offered arguments and justifi- function, and the partnership discords that produce diseases.
cations for attention to this subject. We hope you agree that
this is no longer necessary. To do so, in fact, would disrespect Helmut Kettenmann Berlin, Germany
the enlightened perspective of most neuroscientists that glial Bruce R. Ransom Seattle, USA

xii • P R E FAC E 3 R D E D I T I O N
CONTENTS

Contributors xvii Physiological Properties


16. Physiology of Astrocytes: Ion Channels
and Ion Transporters 185
SECTION 1 Christian Steinhäuser, Gerald Seifert,
P R O P E RT I E S O F N EU R O G L I A L C E L L S and Joachim W. Deitmer
17. Release of Gliotransmitters and Transmitter
Evolution of Glial Cells: Insights from Non-Mammalian Glia Receptors in Astrocytes 197
Helmut Kettenmann and Robert Zorec
1. Evolution of Glial Cells 5
Christian Klämbt 18. Storage and Release of Nontransmitter Signaling
Molecules from Macroglia 212
2. Invertebrate Glia 12
Oliver von Bohlen und Halbach and Klaus Unsicker
Marc R. Freeman
19. Physiology of Microglia 223
3. Nonmammalian Vertebrate Glia 24
Mami Noda and Alexei Verkhratsky
Bruce Appel
20. Physiology of Oligodendrocytes 238
Morphology, Ultrastructure, and Identification
Vittorio Gallo and Jean-Marie Mangin
4. Astrocytes and Ependymal Glia 35
21. Physiological Properties of NG2+ Glial Cells 254
Andreas Reichenbach and Hartwig Wolburg
Dwight E. Bergles
5. Radial Glial Cells 50
22. Cytokine, Chemokine, and Growth Factor
Magdalena Götz
Receptors and Signaling 266
6. Structure and Function of Oligodendrocytes 62 Erik W. G. M. Boddeke, Bart J. L. Eggen,
Arthur M. Butt and Knut P. H. Biber
7. Schwann Cells and Myelin 74 23. Lipids, Lipid Mediators, and Other
Rudolf Martini and Ágnes Patzkó Signaling Molecules 281
8. Microglial Cells 86 Hideki Hayashi
Wolfgang J. Streit 24. Gap Junctions and Hemichannels 292
9. Pericytes of the Central Nervous System 98 Bruce R. Ransom and Christian Giaume
Martin Krüger and Ingo Bechmann 25. Purinergic Mechanisms in Glial Cells 306
10. NG2 Cells (Polydendrocytes) 109 Margaret S. Ho and Shumin Duan
Akiko Nishiyama 26. Calcium Signaling in Neuroglia 320
11. Glial Cells in Autonomic and Sensory Ganglia 122 Alexei Verkhratsky and Vladimir Parpura
Menachem Hanani and David C. Spray 27. The Central Role of Astrocytes
Lineage and Development in Neuroenergetics 333
12. Astrocyte Development 137 Pierre J. Magistretti and Luc Pellerin
James E. Goldman Genomic Profiles
13. Lineage and Development: Oligodendrocytes 148 28. The Astrocyte Transcriptome 347
Katsuhiko Ono and Kazuhiro Ikenaka Ditte Lovatt and Maiken Nedergaard
14. The Schwann Cell Lineage: Cellular Transitions 29. Gene Expression Patterns of Oligodendrocyte
During Development and After Injury 159 Progenitor Cells and Oligodendroglia 358
Kristján R. Jessen, and Rhona Mirsky Fraser J. Sim and Steven A. Goldman
15. Microglia Lineage and Development 172
Marco Prinz

xiii
SECTION 2 48. Factors Controlling Microglial Activation 614
F U N C T I O N S O F N EU R O G L I A L C E L L S Uwe-Karsten Hanisch
49. Roles of Activated Microglia 626
Astrocytes Kelly R. Miller, Stefan Prokop, and Frank L. Heppner
30. Neurogenesis and Outer Subventricular Zone Radial 50. Immune Functions of Microglia 638
Glial Cells 379 Trevor Owens
Xiaoqun Wang and Arnold R. Kriegstein
31. Glial Control of Synaptogenesis 388 SECTION 3
Nicola J. Allen ROLE OF GLIAL CELLS IN DISEASE
32. Neuron Migration and Axon Guidance 402
Andreas Faissner
Mechanisms of Glial Injury
33. The Role of Glia in the Formation and Function of the
51. Astrocyte Responses to Central Nervous System Injury
Blood-Brain Barrier 417
and Disease 653
Istvan Krizbai, Imola Wilhelm, Hans-Christian Bauer,
Michael V. Sofroniew
and Hannelore Bauer
52. Metabolic Injury of Oligodendrocytes and Myelin 665
34. Control of the Extracellular Ionic Environment
Peter K. Stys
and Volume 430
Eva Syková 53. Interaction of Microglia with Neurons and Astrocytes
Under Lesioned Neuronal Conditions 677
35. Amino Acid Neurotransmitter Synthesis
Kazuyuki Nakajima and Shinichi Kohsaka
and Removal, 443
Arne Schousboe, Lasse K. Bak, Karsten K. Madsen, 54. Schwann Cells and Injury 687
and Helle S. Waagepetersen Violetta Zujovic and Alexandros A. Lavdas
36. Glycogen and Energy Metabolism 457 Recovery of Neural Function
Angus Brown 55. Nerve Regeneration in the Peripheral
37. Astrocyte Regulation of Neurovascular Control 470 Nervous System, 701
Clare Howarth, Grant R. J. Gordon, Tessa Gordon and Olawale A. R. Sulaiman
and Brian A. MacVicar 56. Nerve Fiber Regeneration in the Central Nervous
38. Astrocytes: Modulation of Synaptic Function and System of Higher Vertebrates 715
Network Activity 481 Anita D. Buchli and Martin E. Schwab
Andrea Volterra 57. Glial Cell Transplantation for Central Nervous
39. Astrocytic Modulation of Mammalian Synapses: System Repair 728
Circuits and Behaviors 494 Anne Baron-Van Evercooren and Rebecca Matsas
Michael M. Halassa and Philip G. Haydon Ischemia
40. Adult Neurogenesis 504 58. Focal Cerebral Ischemia: The Multifaceted Role
Gerd Kempermann of Glial Cells 745
41. Modulation of Neuroendocrine Systems 515 Ulrich Dirnagl, Bruce R. Ransom, and Josef Priller
Stéphane H. R. Oliet Gliomas
Oligodendrocytes/Schwann Cells 59. Malignant Glioma 759
42. Myelin, Impulse Conduction, and the Shannon Donnola Rebecca Bish, and Dolores
Pathophysiology of Demyelination 529 Hambardzumyan
Lakshmi Bangalore and Stephen G. Waxman 60. Glial Tumors in Neurofibromatosis and Tuberous
43. Transcription Factors in Myelinating Cells 543 Sclerosis Complex 772
Michael Wegner Anthony J. Apicelli and David H. Gutmann
44. Factors Controlling Myelin Formation 555 Demyelinating Diseases
Ruth Stassart, Sandra Goebbels, 61. Multiple Sclerosis 785
and Klaus-Armin Nave Monika Bradl and Hans Lassmann
45. Regulation of Myelination by Functional Activity 573 62. Genetic Mutations Affecting Myelin Formation 798
R. Douglas Fields Steven S. Scherer, M. Laura Feltri, and Lawrence Wrabetz
46. Iron and Glia 586 Neurodegenerative Diseases
James R. Connor 63. Amyotrophic Lateral Sclerosis 811
Microglia Rita Sattler and Jeffrey Rothstein
47. Role of Microglia in the Normal Brain 605 64. Alzheimer Disease 825
Frank Kirchhoff Andrew Kraft and Jin-Moo Lee

xiv • CONTENTS
65. Huntington Disease, Parkinson Disease, and Other 68. Microglia and Pain 876
Neurodegenerative Diseases 837 Simon Beggs, Tuan Trang, and Michael W. Salter
Thomas Möller 69. Genetic Disorders Affecting Astrocytes 884
Infectious Diseases Albee Messing and Michael Brenner
66. Glia in Bacterial and Viral Central Nervous 70. Epilepsy 896
System Infections 847 Detlev Boison
Gwenn Garden 71. Psychiatric Disorders 906
Miscellaneous Diseases Josef Priller
67. Neuroglia in Hepatic Encephalopathy 863
Mireille Bélanger, Javier Vaquero, and Roger F. Butterworth Index 917

CONTENTS • xv
This page intentionally left blank
CONTRIBUTOR S

Nicola J. Allen Ingo Bechmann


Molecular Neurobiology Laboratory Institute for Anatomy
The Salk Institute for Biological Studies University of Leipzig
La Jolla, CA Leipzig, Germany

Anthony J. Apicelli Simon Beggs


Department of Radiation Oncology Program in Neurosciences & Mental Health
Washington University School of Medicine Hospital for Sick Children
St. Louis, MO Toronto, Canada

Bruce Appel Mireille Bélanger


Departments of Pediatrics and Cell and Developmental Neuroscience Research Unit
Biology Centre Hospitalier de l’Université de Montréal
University of Colorado School of Medicine Montreal, Canada
Aurora, CO
Dwight E. Bergles
Lasse K. Bak The Solomon H. Snyder Department of Neuroscience
Department of Drug Design The Johns Hopkins University School of Medicine
and Pharmacology Baltimore, MD
University of Copenhagen
Copenhagen, Denmark Knut P. H. Biber
Department of Psychiatry and Psychotherapy
Lakshmi Bangalore University of Freiburg Medical Center
Department of Neurology and Center for Neuroscience Freiburg, Germany
Research
Yale University School of Medicine Rebecca Bish
New Haven, CT Center for Genomics and Systems Biology
New York University
Anne Baron-Van Evercooren New York, NY
Centre de Recherche de l’Institut du Cerveau et de Moelle
Epinière Erik W. G. M. Boddeke
Paris, France Department of Neuroscience
University Medical Center Groningen
Hannelore Bauer Groningen, The Netherlands
Department of Organismic Biology
University of Salzburg Detlev Boison
Salzburg, Austria Legacy Research Institute
Portland, OR
Hans-Christian Bauer
Department of Organismic Biology Monika Bradl
University of Salzburg Department of Neuroimmunology
Salzburg, Austria Medical University of Vienna
Vienna, Austria

xvii
Michael Brenner Andreas Faissner
Department of Neurobiology Department of Cell Morphology and Molecular
University of Alabama Birmingham Neurobiology
Birmingham, AL Ruhr-University of Bochum
Bochum, Germany
Angus Brown
School of Biomedical Sciences M. Laura Feltri
University of Nottingham Hunter James Kelly Research Institute and Department
Nottingham, United Kindom of Biochemistry
SUNY at Buffalo, School of Medicine
Anita D. Buchli Buffalo, NY
Brain Research Institute
University of Zurich and Federal Institute R. Douglas Fields
of Technology Zurich Nervous System Development and Plasticity Section
Zurich, Switzerland National Institutes of Health, NICHD
Bethesda, MD
Arthur M. Butt
Institute of Biology and Biomedical Sciences Marc R. Freeman
University of Portsmouth Department of Neurobiology
Portsmouth, United Kingdom University of Massachusetts Medical School
Worcester, MA
Roger F. Butterworth
Neuroscience Research Unit Vittorio Gallo
Centre Hospitalier de l’Université de Montréal Center for Neuroscience Research
Montreal, Canada Children’s National Medical Center
Washington, DC
James R. Connor
Department of Neurosurgery Gwenn Garden
Pennsylvania State University Department of Neurology
Hershey, PA University of Washington
Seattle, WA
Joachim W. Deitmer
Department of General Zoology Christian Giaume
University of Kaiserslautern Centre Interdisciplinaire de Recherche en Biologie
Kaiserslautern, Germany Collège de France
Paris, France
Ulrich Dirnagl
Center for Stroke Research Sandra Göbbels
Charité-Universitätsmedizin Berlin Department of Neurogenetics
Berlin, Germany Max Planck Institute of Experimental Medicine
Göttingen, Germany
Shannon Donnola
Department of Stem Cell Biology and Regenerative James E. Goldman
Medicine Department of Pathology and Cell Biology
Cleveland Clinic Columbia University College of Physicians and Surgeons
Cleveland, OH New York, NY

Shumin Duan Steven A. Goldman


Institute of Neurobiology Department of Neurology and the Center for Translational
Zhejiang University School of Medicine Neuromedicine
Hangzhou, China University of Rochester Medical Center
Rochester, NY
Bart J. L. Eggen
Department of Neuroscience Grant R. J. Gordon
University Medical Center Groningen Department of Physiology and Pharmacology
Groningen, The Netherlands University of Calgary
Calgary, Canada

xviii • C O N T R I B U TO R S
Tessa Gordon Clare Howarth
Department of Surgery Department of Psychiatry and the Brain
The Hospital for Sick Children Research Centre
Toronto, Canada University of British Columbia
Vancouver, Canada
Magdalena Götz
Institute of Stem Cell Research Kazuhiro Ikenaka
Munich University Division of Neurobiology and Bioinformatics
Munich, Germany National Institute for Physiological Sciences
Okazaki, Japan
David H. Gutmann
Department of Neurology Kristján R. Jessen,MSc
Washington University School of Medicine Department of Cell and Developmental Biology
St. Louis, MO University College London
London, United Kingdom
Michael M. Halassa
Department of Psychiatry Gerd Kempermann
Massachusetts General Hospital Center for Regenerative Therapies (CRTD)
Boston, MA Technical University of Dresden
Dresden, Germany
Dolores Hambardzumyan
Department of Stem Cell Biology and Helmut Kettenmann
Regenerative Medicine Max Delbrück Center for Molecular Medicine (MDC)
Cleveland Clinic Cellular Neurosciences
Cleveland, OH Berlin, Germany

Menachem Hanani Frank Kirchhoff


Laboratory of Experimental Surgery Department of Molecular Physiology
Hadassah-Hebrew University Medical Center University of Saarland
Jerusalem, Israel Homburg, Germany

Uwe-Karsten Hanisch Christian Klämbt


Institute of Neuropathology Institute of Neuro- and Behavioural Biology
University of Göttingen University of Münster
Göttingen, Germany Münster, Germany

Hideki Hayashi Arnold R. Kriegstein


Priority Organization for Innovation and Excellence Department of Neurology
Kumamoto University University of California San Francisco
Kumamoto, Japan San Francisco, CA

Philip G. Haydon Istvan Krizbai


Department of Neuroscience Institute of Biophysics
Tufts University School of Medicine Hungarian Academy of Sciences
Boston, MA Szeged, Hungary

Frank L Heppner Shinichi Kohsaka


Department of Neuropathology Department of Neurochemistry
Charité-Universitätsmedizin Berlin National Institute of Neuroscience
Berlin, Germany Tokyo, Japan

Margaret S. Ho Andrew Kraft


Department of Anatomy and Neurobiology Department of Neurology
Tongji University School of Medicine Washington University School of Medicine
Shanghai, China St. Louis, MO

C O N T R I B U TO R S • xix
Martin Krüger Albee Messing
Institute for Anatomy Waisman Center and Department of Comparative
University of Leipzig Biosciences
Leipzig, Germany University of Wisconsin-Madison
Madison, WI
Hans Lassmann
Center for Brain Research Kelly R Miller
Medical University of Vienna Department of Neuropathology
Vienna, Austria Charité-Universitätsmedizin Berlin
Berlin, Germany
Alexandros A. Lavdas
Laboratory of Cellular and Molecular Neurobiology Rhona Mirsky
Hellenic Pasteur Institute Department of Cell and Developmental Biology
Athens, Greece University College London
London, United Kingdom
Jin-Moo Lee
Department of Neurology Thomas Möller
Washington University School of Medicine Lundbeck Research USA
St. Louis, MO Neuroinflammation Disease Biology Unit
Paramus, NJ
Ditte Lovatt
Center for Translational Neuromedicine Kazuyuki Nakajima
University of Rochester Medical School Department of Neurochemistry
Rochester, NY National Institute of Neuroscience
Tokyo, Japan
Karsten K. Madsen
Department of Drug Design and Pharmacology Klaus-Armin Nave
University of Copenhagen Department of Neurogenetics
Copenhagen, Denmark Max Planck Institute of Experimental Medicine
Göttingen, Germany
Brian A. MacVicar
Department of Psychiatry and the Brain Maiken Nedergaard
Research Centre Center for Translational Neuromedicine
University of British Columbia University of Rochester Medical School
Vancouver, Canada Rochester, NY

Pierre J. Magistretti Akiko Nishiyama


Brain Mind Institute Department of Physiology and Neurobiology
Ecole Polytechnique Fédérale de Lausanne University of Connecticut
Lausanne, Switzerland Storrs, CT

Jean-Marie Mangin Mami Noda


INSERM U952 Laboratory of Pathophysiology
Université Pierre et Marie Curie Kyushu University
Paris, France Fukuoka, Japan

Rudolf Martini Stéphane H. R. Oliet


Department of Neurology INSERM U862 - Neurocentre Magendie
University of Würzburg Université de Bordeaux
Würzburg, Germany Bordeaux, France

Rebecca Matsas Katsuhiko Ono


Laboratory of Cellular and Molecular Neurobiology Department of Biology
Hellenic Pasteur Institute Kyoto Prefectural University of Medicine
Athens, Greece Kyoto, Japan

xx • C O N T R I B U TO R S
Trevor Owens Rita Sattler
Department of Neurobiology Research Department of Neurology
University of Southern Denmark Johns Hopkins University
Odense, Denmark Baltimore, MD

Vladimir Parpura Steven S. Scherer


Department of Neurobiology Department of Neurology
University of Alabama The Perelman School of Medicine at the University
Birmingham, AL of Pennsylvania
Philadelphia, PA
Ágnes Patzkó
Department of Neurology Arne Schousboe
University of Würzburg Department of Drug Design and Pharmacology
Würzburg, Germany University of Copenhagen
Copenhagen, Denmark
Luc Pellerin
Department of Physiology Martin E. Schwab
University of Lausanne Brain Research Institute
Lausanne, Switzerland University of Zurich and Federal Institute
of Technology Zurich
Josef Priller Zürich, Switzerland
Department of Neuropsychiatry and Laboratory of
Molecular Psychiatry Gerald Seifert
Charité-Universitätsmedizin Berlin Institute of Cellular Neurosciences
Berlin, Germany University of Bonn
Bonn, Germany
Marco Prinz
Department of Neuropathology Fraser J. Sim
University of Freiburg Department of Pharmacology and Toxicology
Freiburg, Germany State University of New York at Buffalo School
of Medicine
Stefan Prokop Buffalo, NY
Department of Neuropathology
Charité-Universitätsmedizin Berlin Michael V. Sofroniew
Berlin, Germany Department of Neurobiology
David Geffen School of Medicine
Bruce R. Ransom University of California, Los Angeles
Department of Neurology Los Angeles, CA
University of Washington School of Medicine
Seattle, WA David C. Spray
Dominick P. Purpura Department
Andreas Reichenbach of Neuroscience
Paul Flechsig Institute for Brain Research Albert Einstein College of Medicine
University of Leipzig Bronx, NY
Leipzig, Germany
Ruth Stassart
Jeffrey D. Rothstein Department of Neurogenetics
Department of Neurology Max Planck Institute of Experimental Medicine
Johns Hopkins University Göttingen, Germany
Baltimore, MD
Christian Steinhäuser
Michael W. Salter Institute of Cellular Neurosciences
Department of Physiology University of Bonn
University of Toronto Centre for the Study of Pain Bonn, Germany
Toronto, Canada

C O N T R I B U TO R S • xxi
Wolfgang J. Streit Helle S. Waagepetersen
Department of Neuroscience Department of Drug Design and Pharmacology
University of Florida College of Medicine University of Copenhagen
Gainesville, FL Copenhagen, Denmark

Peter K. Stys Xiaoqun Wang


Department of Clinical Neurosciences Department of Neurology
University of Calgary University of California San Francisco
Calgary, Canada San Francisco, CA

Olawale A. R. Sulaiman Stephen G. Waxman


Ochsner Health Systems Department of Neurology and Center for Neuroscience
New Orleans, LA Research
Yale University School of Medicine
Eva Syková New Haven, CT
Department of Neuroscience
Charles University Michael Wegner
Prague, Czech Republic Institute of Biochemistry
Friedrich-Alexander-University of Erlangen-Nürnberg
Tuan Trang Erlangen, Germany
Department of Physiology
University of Toronto Centre for Imola Wilhelm
the Study of Pain Institute of Biophysics
Toronto, Canada Hungarian Academy of Sciences
Szeged, Hungary
Klaus Unsicker
Department of Molecular Embryology Hartwig Wolburg
University of Freiburg Institute of Pathology and Neuropathology
Freiburg, Germany University of Tübingen
Tübingen, Germany
Javier Vaquero
Neuroscience Research Unit Lawrence Wrabetz
Centre Hospitalier de l’Université de Montréal Department of Neurology and Biochemistry
Montreal, Canada SUNY at Buffalo, School of Medicine
University of Buffalo
Alexei Verkhratsky Buffalo, NY
Faculty of Life Sciences
The University of Manchester Robert Zorec
Manchester, United Kingdom Institute of Pathophysiology
University of Ljubljana
Andrea Volterra Ljubljana, Slovenia
Department of Cell Biology and Morphology
University of Lausanne Violetta Zujovic
Lausanne, Switzerland INSERM, CR1
Centre de Recherche de l’Institut du Cerveau et de la
Oliver von Bohlen und Halbach Moelle Epinière
Institute of Anatomy and Cell Biology Paris, France
University of Greifswald
Greifswald, Germany

xxii • C O N T R I B U TO R S
NEUROGLIA
This page intentionally left blank
SECTION 1
P R O P E RT I E S O F N E U R O G L I A L C E L L S
This page intentionally left blank
EVOLUTION OF GLIAL CELLS: INSIGHTS FROM
NON-MAMMALIAN GLIA
This page intentionally left blank
1.
EVOLUTION OF GLIAL CELLS
Christian Klämbt

Nothing in Biology makes sense, except in the light of evolution. THEODOSIUS DOBZHANSKY

A B B R E VI AT I O N S instruct us about general aspects of glial biology, but will help to


identify the functional constrains sculpting non-neuronal cells in
APC/C anaphase-promoting complex/cyclosome the nervous system. In contrast, if we assume that evolution has
C. elegans Caenorhabditis elegans invented glial cells only once, this provides us with the opportu-
CNS central nervous system nity to deduce the core functions of glia and indeed learn from
GABA gamma-aminobutyric acid studying more simple, genetically amenable organisms.
gcm glial cells missing
GFP green fluorescent protein
GPI glycosylphosphatidylinositol 2 W H AT A R E G L I A L C E L L S ?
GS glutamine synthetase
NCAM neural cell adhesion molecule Although it is fairly straightforward to define a neuronal cell,
PLP proteolipid protein it is still not satisfactorily clear how we can recognize glial
PNS peripheral nervous system cells without any doubt. In fact, particularly in more simple
PSA polysialic acid invertebrates, cells are quite often named glial because they are
not obviously neurons (Bullock and Horridge 1965; Hartline
2011; Radojcic and Pentreath 1979). These classical studies
1 E S S AY: W H Y S H O U L D W E C A R E provide a minimal set of criteria that qualify a cell as a glial cell:
A B O U T T H E E VO LU T I O N A RY (1) Glial cells should have an intimate morphological associa-
ORIGIN OF GLIAL CELLS? tion with neurons or they should separate neuronal elements
from mesodermal layers; and (2) glial cells should originate
Most animals are able to respond to external stimuli by mov- from the embryonic ectoderm and are referred to collectively
ing. To do so, many phyla have developed complex neural as macroglia.
structures allowing more and more sophisticated computing Microglial cells are not covered here. These cells origi-
of information. As the nervous system changed from a simple nate from the mesoderm and enter the vertebrate brain during
netlike structure to condensed ganglia and centralized brains, embryogenesis to become resident immunologically compe-
a new cell type could be recognized in morphological stud- tent cells acting as macrophages during infection and injury
ies: glial cells (Bullock and Horridge 1965; Hartline 2011). (Kettenmann et al. 2011; Saijo and Glass 2011). In Drosophila,
The importance of this new cell type for a functional brain no mesodermal cells are known to migrate into the nervous sys-
is reflected by the increase in the relative number of glial cells tem and macrophage functions are taken over by macroglial cells
during evolution. Although quantifying glial cell numbers in (Awasaki and Ito 2004; Doherty et al. 2009; Watts et al. 2004).
larger mammalian brains is difficult, there are roughly equal There are many examples in which the preceding criteria
numbers of glial cells and neurons in these species (Azevedo allow an easy definition of glial cells, and in some cases even
et al. 2009; Herculano-Houzel 2011; Hilgetag and Barbas obvious similarities between the mammalian and invertebrate
2009). By contrast, only 10% of invertebrate neural cells as in glia can be seen. Good examples are the peripheral glial cells
Drosophila or C. elegans are glial cells (Beckervordersandforth in insects. The wrapping glial cells of the peripheral nerves
et al. 2008; Hilchen et al. 2008; Oikonomou and Shaham in Drosophila are very large cells, stretching their cytoplasm
2011; Pereanu et al. 2005). about 1 mm around axon fascicles (Sieglitz and Klämbt
But what are glial cells, and are they really comparable in all unpublished). Morphologically and functionally these cells
phyla? Are they the many non-neuronal cells of the brain that very much resemble Remak fibers of the mammalian nervous
we consider as glia truly homologous, or have they appeared system (Nave and Trapp 2008; Rodrigues et al. 2011; Stork
independently several times during evolution? If the latter is the et al. 2008). Likewise, cells with an astrocytic morphology
case, the analysis of different animal species will not necessarily are not confined to the mammalian central nervous system

5
(CNS), but can also be identified in Drosophila (Awasaki et al. al. 2010). Once the sensory neuron was defined, it had to spe-
2008; Stork and Freeman personal communication; Volterra cialize more and more and therefore became dependent on the
and Meldolesi 2005) (see chapter 2). Moreover, these cells support of its neighbors. Such simple isolated nerve cells still
express a related set of genes controlling neurotransmitter exist in coelenterates, in which the molecular networks con-
homeostasis (Stacey et al. 2010). trolling neurogenesis in bilateria are all in place (Galliot et al.
In general, these morphological criteria clearly define 2009; Marlow et al. 2009). In Hydrozoa, for example, sensory
glia in most animal phyla with a condensed nervous system. cells and nematocytes are concentrated in the spherical end
However, sometimes it remains difficult to decide whether or bulbs of the tentacles. Interestingly, the four types of sensory
not a cell can be classified as glial cell. Here, the perineurial cells present in these bulbs are separated by distinct support-
glial cells or the “myelin” forming glial cells of invertebrates ing cells, which resemble accessory cells of complex peripheral
may serve as a paradigm. sense organs in bilateria (Holtmann and Thurm 2001a, b).
Myelination has been considered to be a clear glial func- Thus, neurons may have evolved with supporting cells or glial
tion found in vertebrates and invertebrates (Bullock 2004; cells from the very beginning.
Hartline and Colman 2007; Nave 2010a). Interestingly, two
recent studies cast some doubt on this central dogma of glial
biology. In calanoid copepods myelin-like structures appeared 4 PA I RW I S E E VO LU T I O N O F
before glial cells became associated with axons and their for- NEURONS AND GLIAL CELLS
mation was initiated internal to the axolemma. These find-
ings suggest a nonglial origin of myelinated structures; thus, A pairwise evolution of a neuronal cell and a support cell (the
a typical glial structure is made by nonglial cells (Wilson and pigment cell) has been very well documented for the light
Hartline 2011a,b). sensing organs. Photoreceptor neurons are able to receive sig-
A reverse example can be seen in the insect perineurial glia nals and are generally accompanied by pigment cell(s), which
that abuts the blood-brain barrier forming subperineurial glia provide numerous accessory functions (Arendt 2003; Arendt
(Stork et al. 2008). The perineurial glial cells do not form a and Wittbrodt 2001). Charles Darwin speculated that the eye
tight sheath around the nervous system and appear only rel- contains, as minimal equipment, a neuron to detect light and
atively late during development. Significantly, the perineurial a pigment cell for support and shading. Indeed, such simple
glia lack any contact with neuronal cells. Over a long time eyes can be found in planarians and some polychaete larvae
the glial nature of these cells has been highly controversial (Gehring and Ikeo 1999). Over the centuries, however, the
(Radojcic and Pentreath 1979). The perineurial glia has been evolution of eyes has been a matter of quite some debate. The
carefully analyzed in Drosophila. There is now broad consent traditional view, backed by numerous anatomical studies, sug-
to consider these cells as glial, based on the neural lineage of gested that the eye has evolved independently several times.
these cells and the expression of several glial specific markers, However, modern molecular genetic tools have clearly favored
although proliferation of perineurial glial cells during pupal the monophyletic origin of all photoreceptor cells by unravel-
stages does not appear to depend on the regulatory gene glial ing the surprisingly well-conserved network of transcriptional
cells missing (Gcm) (Awasaki et al. 2008; Stork et al. 2008). regulators directing eye development (Arendt 2003; Bullock
Thus, the assignment of glial identity should not be based and Horridge 1965; Erclik et al. 2008, 2009; Gehring, 2002,
solely on histological arguments, but rather requires the inclu- 2005; Gehring and Ikeo 1999; Nilsson 2004).
sion of as much molecular data as possible to faithfully define In all bilateria, pax6-related genes are core components of
a glial cell type. the genetic circuitry orchestrating eye development (Gehring
and Ikeo 1999). In Cnidaria, which are the earliest metazoans
with a well-developed nervous system, Pax6-like transcription
3 E VO LU T I O N O F N E U R O N S factors do not exist. Nevertheless, many Cnidaria, including
hydrozoa and cubozoa form highly elaborated eyes with cor-
Any serious discussion regarding the origin of glial cells has nea, lens, and retina (Martin 2002). Interestingly, in the cubo-
to be linked to the question about the origin of neurons, zoan jellyfish Tripedalia cystophora Pax-B has been implicated
which are found in most multicellular animals. The inven- in eye development (Kozmik 2008; Kozmik et al. 2003),
tion of multicellularity set the stage for a division of labor, whereas in the hydrozoan jellyfish Cladonema radiatum Pax-A
and organs devoted to feeding, reproduction, or movement rather than Pax-B controls eye formation (Suga et al. 2010).
could be formed. In addition, individual ectodermal cells were These data suggest that the origin of eyes occurred before the
equipped with sensory filters to allow the detection of changes duplication of the Pax genes and further supports the hypo-
in the outside world and subsequent response to them. These thesis of the monophyletic origin of all animal eyes.
properties must have evolved in very early metazoa because How can the likely monophyletic origin of animal eyes
Porifera, which are considered to have not formed neuronal help to address the origin of glia? In this respect it is impor-
cells (Bullock and Horridge 1965; Hartline 2011; Radojcic tant to consider that eye formation always implies the pres-
and Pentreath 1979), use well-conserved molecular mecha- ence of a receptor and a pigment cell; indeed, all pax genes do
nisms (proneural genes encoding atonal related proteins and exactly this, they specify receptor cells as well as pigment cells.
neurogenic genes such as Notch and Delta) to generate an Pigment cells serve a number of metabolic functions necessary
ancient sensory cell type (Richards et al. 2008; Srivastava et for normal vision. Vision requires the detection of a photon

6 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
by a receptor that comprises a protein (opsin) coupled to is seen in the septate junctions in insects and the related
11-cis-retinal, which isomerizes to all-trans-retinal on activation septate-like junctions seen in the paranodes of the mammalian
of the receptor. In vertebrates the regeneration of 11-cis-retinal peripheral nervous system (Banerjee and Bhat 2007; Girault
occurs in pigments cells. A similar visual cycle between pho- and Peles 2002; Sherman and Brophy 2005). In both cases
toreceptor cells and pigment cells recently has been identified morphologically similar structures are formed by homologous
in Drosophila (Arshavsky 2010; Fain et al. 2010; Wang et al. sets of interacting transmembrane proteins. In mice, Caspr,
2010). This underscores the intricate interaction of these two Contactin, and Neurofascin155 were found to be incorpo-
cell types, one being a sensory cell, and the other one being clas- rated into the septate-like junctions (Einheber 1997; Peles et
sified as a support cell—in other words—as a glial cell. al. 1997; Poliak et al. 1999). In Drosophila the homologous
The interaction of a photoreceptor cell and a pigment cell proteins NeurexinIV, dContactin, and Neuroglian form the
is not confined to the re-isomerization of all-trans-retinal. base of septate junctions that are essential to establish the
Neurons generally need a lot of energy and much debate is on blood-brain barrier (Banerjee et al. 2006; Baumgartner et
how neurons and in particular how long axons are metaboli- al. 1996; Stork et al. 2008). In addition, genetic analyses in
cally supported by glial cells for the maintenance of axonal Drosophila have unraveled 12 additional membrane-associated
transport and long-term survival (Brown and Ransom 2007; proteins that associate with the septate junctions (Genova and
Nave 2010b; Nave and Trapp 2008). Metabolic coupling is Fehon 2003; Hijazi et al. 2009; Nilton et al. 2010; Oshima
not only needed between ensheathing glia and long axons, but and Fehon 2011; Syed et al. 2011; Tiklová et al. 2010; Wu and
is likely to reflect a much more ancestral function in which Beitel 2004). Although we still do not know how septate junc-
support cells (glial cells) feed the accompanying neurons. In tions are initially assembled and then maintained, it is certain
this way, it is not surprising that pigment cells in the retina that deeper knowledge on the biology of septate junctions in
of bees take up glucose and supply metabolic products (in invertebrates will help to decipher how septate-like junctions
this case alanine) to the receptor cells. Likewise, mammalian are formed in the mammalian nervous system.
astrocytes are able to provide lactate to fuel neuronal activity Adhesion systems are not only working to ensure glial–
(Brown and Ransom 2007; Coles 1989; Pellerin et al. 2007; glial cell contact, but are also needed to ensure the close
Tsacopoulos and Magistretti 1996; Tsacopoulos et al. 1988). association of glial cells with neurons. Neuron–glia inter-
Assuming that at least sensory neurons have evolved as cell action is obvious and evolution has selected astonishingly
pairs—a sensory specialist and a support specialist—it appears well-conserved adhesion molecules to guarantee this specific
likely that glial-like cells should exist in the most simple organisms interaction (Silies and Klämbt, 2011). The neural cell adhesion
with a fairly well developed nervous system, Cnidaria. However, molecule (NCAM) gene is a quite remarkable example and
the prevailing view in the literature is that Cnidaria lack glial demonstrates not only the evolutionary conservation of gene
cells (Bullock and Horridge 1965; Hartline 2011; Radojcic and functions, but also the conservation of specifically spliced iso-
Pentreath 1979). Within the different Cnidaria classes (Antozoa, forms. In mammals and insects, NCAM encodes three distinct
Hydrozoa, Cubozoa, Scyphozoa, and Staurozoa) an amazing isoforms, which share an identical extracellular domain with
spectrum of eyes has developed during evolution. The complex five immunoglobin (Ig) domains and two fibronectin (FN)
lens eyes of cnidarian species are organized together with other domains, but differ in the way they are linked to the cell mem-
sensory organs in the so-called rhopalia (Martin 2002; Nilsson brane. Two NCAM isoforms carry a transmembrane domain
2004). Sensory input then is integrated in a radial symmetrical but differ in their cytoplasmic domains, and one NCAM iso-
central nervous system, which is arranged in a ring form and allows form is linked to the outer cell membrane by a GPI anchor. In
in part complex behavior of jellyfish, such as courtship behavior vertebrates and invertebrates, the transmembrane-bound iso-
in some medusae (Galliot et al. 2009; Garm et al. 2007; Lewis form is expressed by neurons, whereas the GPI-linked protein
and Long 2005; Marlow et al. 2009). The ring nerve of a 1-cm form is made by glial cells (Higgins et al. 2002; Maness and
large Tripedalia cystophora medusa is generated by about 10.000 Schachner 2007; Noble et al. 1985; Silies and Klämbt 2010;
neurons that process and transmit information in several distinct Wright and Copenhaver 2000, 2001).
subsystems (Garm et al. 2007). Within the ring nerve, special The neural cell adhesion molecule and its orthologous
epithelial cells provide compartmentalization by cellular exten- proteins function as homophilic adhesion proteins and
sions that encircle groups of axons, and thus might provide some among others control glial migration in the developing nerv-
ancestral glial function by isolating the axonal bundles (Garm et ous system (Grenningloh et al. 1991; Hoffman and Edelman
al. 2007; Mackie and Meech 1995). Here, more work is needed to 1983). During this process, modulation of NCAM-mediated
address the question whether these cells can be considered as glial adhesiveness is of clear relevance. Interestingly, although the
cells or not, and in the author’s view the chances are high to find expression of specific isoforms has been highly conserved dur-
these typical characteristics. ing evolution, distinct mechanisms evolved to control the
modulation of NCAM-mediated cell stickiness. In the ver-
tebrate CNS, adhesiveness is altered by the addition of poly-
5 M O L E C U L A R C R I T E R I A TO D E F I N E sialic acid (PSA) moieties to the NCAM protein (Rutishauser
G L I A : A D H E S I VE P R O P E RT I E S 2008; Weinhold et al. 2005). Whereas NCAM mutants are
viable and fertile and have only relatively minor defects during
Important insights in the evolution of glial cells originate from nervous system development (Chazal et al. 2000; Cremer et
analyzing their molecular signatures. A prominent example al. 1994), loss of the two enzymes that transfer PSA to NCAM

1. E VO LU T I O N O F G L I A L C E L L S • 7
causes severe defects during brain development, which can be found in the Drosophila CNS (Altenhein et al. 2006; Besson et
rescued by loss of NCAM (Weinhold et al. 2005). Drosophila al. 1999; Soustelle et al. 2002; Stacey et al. 2010). Interestingly,
follows a quite different molecular approach to reach the not only the uptake of neurotransmitters appears evolutionary
same functional result. During fly development, no polysialic conserved, but also the support of the presynaptic terminals with
acid moieties are attached to extracellular proteins. Instead, metabolic intermediates that allow an efficient and fast resynthesis
the NCAM homolog Fasciclin2 is removed from axons in a of these transmitters. This has been well studied for the uptake of
graded manner through APC/C dependent regulatory endo- glutamate by astrocytes. On intake of glutamate, the astrocytic
cytotic processes (Silies and Klämbt 2010). enzyme glutamine synthetase (GS) converts it to glutamine,
These data demonstrate that on the one hand, too much which is then released to the neuron. The Drosophila genome
adhesion is worse than not enough. On the other hand, these encodes two glutamine synthase genes, one of which (GS2) is
molecular studies suggest that during evolution glial cells and expressed in cells associated with the neuropile, which are likely
neurons were equipped with different NCAM proteins before to exert astrocytic functions (Freeman et al. 2003; Stacey et al.
the split of Protostomia and Deuterostomia. The genomes of 2010; Thomas and van Meyel 2007). Indeed, GS is an evolution-
several cnidarian species have been sequenced in the mean- ary well-conserved protein and is found in the nervous system of
time (Hydra, Nematostella, Clytia) and Ig-domain proteins many animal species (Niva et al. 2008; Roots 1981). The genomes
resembling NCAM have been identified (Chapman et al. of Caenorhabditis elegans and Hydra magnipapillata also encode
2010; Houliston et al. 2010; Marlow et al. 2009; Putnam et al. several well-conserved glutamine synthetase genes, but currently
2007). However, we will have to wait for more sophisticated no studies have disclosed the expression of these genes. Glutamate
annotation tools to learn whether several isoforms of NCAM and GABA receptors are known even in Cnidaria, and thus the
are generated in these simple metazoan as well and whether close metabolic coupling of neurons and glial cells exemplified in
the expression of specific isoforms can be linked to individual the tripartite synapse may reflect an ancient evolutionary devel-
neural cell types. opment and glutamine synthetase thus serves as an evolutionary
Neuron–glial interaction cumulates most spectacularly early marker of astrocytic cells (Delgado et al. 2010; Scappaticci
in the formation of myelin sheaths in the vertebrate nervous and Kass-Simon 2008).
system. Here specific adhesion systems have evolved that may
be considered as evolutionary new inventions as invertebrates
lack several of the “myelin” genes. Of the ten human genes 7 M O L E C U L A R C R I T E R I A TO D E F I N E
known to be required for myelin production, only one gene GLIA : TR ANSCRIPTION
encoding a proteolipid protein was found in the Drosophila
genome (Cravchik et al. 2001; Stecca et al. 2000). The fly The evidence collected in the preceding indicates that glial
proteolipid protein (PLP)–like protein M6 is expressed and cells have indeed been invented very early during the evolution
required during oogenesis. A GFP enhancer/gene trap inser- of metazoa. In particular, when we include molecular criteria,
tion into the promoter of the M6 gene suggests that there is glia appears as a very ancient cell type. However, molecular
additional expression in the developing nervous system and biology also provides us with some arguments against this
shows accumulation of the M6-GFP fusion protein along hypothesis. Gliogenesis in the mammalian nervous system is
axon tracts (Zappia et al. 2011). However, it is presently linked to a complex network of transcription factors such as
unknown whether this protein is made by glial cells or neu- olig or sox10 (Britsch 2001; Richardson et al. 2006; Rowitch
rons and whether any phenotypic abnormalities are found in 2004; Rowitch and Kriegstein 2010; Zhou et al. 2001). In
mutant glial cells. contrast, gliogenesis in Drosophila is based on the presence
In conclusion, glial cells use a variety of conserved adhe- of a single transcription factor, Gcm, which is expressed and
sion systems. As a consequence, glial biology became more required in almost all glial cells (Hosoya et al. 1995; Jones
complex during evolution by recruiting additional adhesion et al. 1995; Kammerer and Giangrande 2001; Vincent et al.
systems to perform specific tasks such as myelination. 1996). Glial cells missing activates the transcription of several
downstream factors that mediate glial differentiation (Giesen
et al. 1997; Granderath et al. 2000; Shandala et al. 2003;
6 M O L E C U L A R C R I T E R I A TO Yuasa et al. 2003). Although gcm is prominently involved in
D E F I N E G L I A : S U P P O RT O F specifying glial cell fate, it is not exclusively expressed by glial
SY N A P T I C T R A N S M I S S I O N cells. Besides additional expression domains outside the nerv-
ous system, gcm is also expressed in the neuronal cells, where
A key function of glial cells resides in the support and modulation it exerts some by now unknown functions (Bernardoni et al.
of synaptic function. Astrocytes cooperate with the presynapse 1997; Chotard et al. 2005; Soustelle and Giangrande 2007;
and postsynapse to maintain synaptic fidelity. Indeed, a synapse Soustelle et al. 2004, 2007). Although the gliogenic functions
can be viewed as a tripartite structure that is found in complex of gcm have not been conserved during evolution, gcm appears
animals such as leech, Drosophila, or mammals (Danjo et al. to trigger the activation of a conserved epigenetic pathway by
2011). In the mammalian system it has been shown that astrocytes regulating histone acetylation (Flici et al. 2011; Wegner and
express several neurotransmitter transporters such as glutamate, Riethmacher 2001). Thus, common regulatory mechanisms
gamma-amino butyric acid (GABA), and glycine transporters underlying glial development may affect the general tran-
(Halassa and Haydon 2010). Several of these transporters are also scriptional profile of glial cells and during evolution different

8 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
regulatory transcription factors have learned to restrain the Besson MT, Soustelle L, Birman S. 1999. Identification and structural
activity of these epigenetic modulators. characterization of two genes encoding glutamate transporter homo-
logues differently expressed in the nervous system of Drosophila mela-
nogaster. FEBS Lett 443:97–104.
Britsch S. 2001. The transcription factor Sox10 is a key regulator of
8 S U M M A RY A N D P E R S P E C T I VE S peripheral glial development. Genes Dev 15:66–78.
Brown AM, Ransom BR. 2007. Astrocyte glycogen and brain energy
Clearly, evolution has provided us with a bewildering diver- metabolism. Glia 55:1263–1271.
Bullock TH. 2004. The natural history of neuroglia: an agenda for com-
sity of neuronal and glial cell types. Yet molecularly, all these parative studies. Neuron Glia Biol 1:97–100.
cells are deviations from a simple basic ground plan, suggest- Bullock TH, Horridge GA. 1965. Structure and function in the nervous
ing that quite likely this cell type has appeared just once. Thus, systems of invertebrates. San Francisco: Freeman.
the analysis of simple and highly derived glial cell types in Chapman JA, Kirkness EF, Simakov O, Hampson SE, Mitros T,
many different species will contribute to our understanding Weinmaier T, et al. 2010. The dynamic genome of Hydra. Nature
464:592–596.
of human glial cells. Furthermore, the integration of results Chazal G, Durbec P, Jankovski A, Rougon G, Cremer H. 2000. Consequences
obtained in Cnidaria and genetically amenable organisms of neural cell adhesion molecule deficiency on cell migration in the ros-
such as Drosophila, C. elegans, zebrafish, or mice is needed to tral migratory stream of the mouse. J Neurosci 20:1446–1457.
define what the first glial cells may have looked like and what Chotard C, Leung W, Salecker I. 2005. glial cells missing and Gcm2 cell
their first functions may have been. autonomously regulate both glial and neuronal development in the
visual system of Drosophila. Neuron 48:237–251.
Coles JA. 1989. Functions of glial cells in the retina of the honeybee
drone. Glia 2:1–9.
AC K N OW L E D G M E N T S Cravchik A, Subramanian G, Broder S, Venter JC. 2001. Sequence analy-
sis of the human genome: implications for the understanding of ner-
vous system function and disease. Arch Neurol 58:1772.
The author is thankful to S. Limmer, S. Thomas, and Cremer H, Lange R, Christoph A, Plomann M, Vopper G, Roes J, et al.
S. Sasse for comments and discussions. The work in the lab 1994. Inactivation of the N-CAM gene in mice results in size reduc-
of C. Klämbt is funded through grants of the Deutsche tion of the olfactory bulb and deficits in spatial learning. Nature
Forschungsgemeinschaft and the EC. 367:455–459.
Danjo R, Kawasaki F, Ordway RW. 2011. A tripartite synapse model in
Drosophila. PLoS ONE 6:e17131.
Delgado LM, Couve E, Schmachtenberg O. 2010. GABA and glutamate
REFERENCES immunoreactivity in tentacles of the sea anemone Phymactis papil-
losa (LESSON 1830). J Morphol 271:845–852.
Altenhein B, Becker A, Busold C, Beckmann B, Hoheisel JD, Technau Doherty J, Logan MA, Taşdemir OE, Freeman MR. 2009. Ensheathing
GM. 2006. Expression profi ling of glial genes during Drosophila glia function as phagocytes in the adult Drosophila brain. J Neurosci
embryogenesis. Dev Biol 296:545–560. 29:4768–4781.
Arendt D. 2003. Evolution of eyes and photoreceptor cell types. Int J Einheber S. 1997. The axonal membrane protein Caspr, a homologue of
Dev Biol 47:563–571. neurexin IV, is a component of the septate-like paranodal junctions
Arendt D, Wittbrodt J. 2001. Reconstructing the eyes of Urbilateria. that assemble during myelination. J Cell Biol 139:1495–1506.
Philos Trans R Soc Lond, B, Biol Sci 356:1545–1563. Erclik T, Hartenstein V, Lipshitz HD, McInnes RR. 2008. Conserved
Arshavsky VY. 2010. Vision: the retinoid cycle in Drosophila. Curr Biol role of the Vsx genes supports a monophyletic origin for bilaterian
20:R96–R98. visual systems. Curr Biol 18:1278–1287.
Awasaki T, Ito K. 2004. Engulfing action of glial cells is required for Erclik T, Hartenstein V, McInnes RR, Lipshitz HD. 2009. Eye evolu-
programmed axon pruning during Drosophila metamorphosis. Curr tion at high resolution: the neuron as a unit of homology. Dev Biol
Biol 14:668–677. 332:70–79.
Awasaki T, Lai S-L, Ito K, Lee T. 2008. Organization and postembryonic Fain GL, Hardie R, Laughlin SB. 2010. Phototransduction and the evo-
development of glial cells in the adult central brain of Drosophila. lution of photoreceptors. Curr Biol 20:R114–R124.
J Neurosci 28:13742–13753. Flici H, Erkosar B, Komonyi O, Karatas OF, Laneve P, Giangrande A.
Azevedo FAC, Carvalho LRB, Grinberg LT, Farfel JM, Ferretti REL, 2011. Gcm/Glide-dependent conversion into glia depends on neural
Leite REP, et al. 2009. Equal numbers of neuronal and nonneuronal stem cell age, but not on division, triggering a chromatin signature
cells make the human brain an isometrically scaled-up primate brain. that is conserved in vertebrate glia. Development 138:4167–4178.
J Comp Neurol 513:532–541. Freeman MR, Delrow J, Kim J, Johnson E, Doe CQ. 2003. Unwrapping
Banerjee S, Bhat MA. 2007. Neuron-glial interactions in blood-brain glial biology: Gcm target genes regulating glial development, diversi-
barrier formation. Annu Rev Neurosci 30:235–258. fication, and function. Neuron 38:567–580.
Banerjee S, Pillai AM, Paik R, Li J, Bhat MA. 2006. Axonal ensheath- Galliot B, Quiquand M, Ghila L, de Rosa R, Miljkovic-Licina M,
ment and septate junction formation in the peripheral nervous sys- Chera S. 2009. Origins of neurogenesis, a cnidarian view. Dev Biol
tem of Drosophila. J Neurosci 26:3319–3329. 332:2–24.
Baumgartner S, Littleton JT, Broadie K, Bhat MA, Harbecke R, Garm A, Poussart Y, Parkefelt L, Ekström P, Nilsson D-E. 2007. The
Lengyel JA, et al. 1996. A Drosophila neurexin is required for sep- ring nerve of the box jellyfish Tripedalia cystophora. Cell Tissue Res
tate junction and blood-nerve barrier formation and function. Cell 329:147–157.
87:1059–1068. Gehring WJ. 2002. The genetic control of eye development and its
Beckervordersandforth RM, Rickert C, Altenhein B, Technau GM. implications for the evolution of the various eye-types. Int J Dev Biol
2008. Subtypes of glial cells in the Drosophila embryonic ventral 46:65–73.
nerve cord as related to lineage and gene expression. Mech Dev Gehring WJ. 2005. New perspectives on eye development and the evolu-
125:542–557. tion of eyes and photoreceptors. J Hered 96:171–184.
Bernardoni R, Vivancos V, Giangrande A. 1997. Glide/gcm is expressed Gehring WJ, Ikeo K. 1999. Pax 6: mastering eye morphogenesis and eye
and required in the scavenger cell lineage. Dev Biol 191:118–130. evolution. Trends Genet 15:371–377.

1. E VO LU T I O N O F G L I A L C E L L S • 9
Genova JL, Fehon RG. 2003. Neuroglian, Gliotactin, and the Na+/K+ Maness PF, Schachner M. 2007. Neural recognition molecules of the
ATPase are essential for septate junction function in Drosophila. J immunoglobulin superfamily: signaling transducers of axon guid-
Cell Biol 161:979–989. ance and neuronal migration. Nat Neurosci 10:19–26.
Giesen K, Hummel T, Stollewerk A, Harrison S, Travers A, Klämbt C. Marlow HQ, Srivastava M, Matus DQ, Rokhsar D, Martindale
1997. Glial development in the Drosophila CNS requires concomi- MQ. 2009. Anatomy and development of the nervous system of
tant activation of glial and repression of neuronal differentiation Nematostella vectensis, an anthozoan cnidarian. Dev Neurobiol
genes. Development 124:2307–2316. 69:235–254.
Girault J-A, Peles E. 2002. Development of nodes of Ranvier. Curr Opin Martin VJ. 2002. Photoreceptors of cnidarians. Can J Zool 80:
Neurobiol 12:476–485. 1703–1722.
Granderath S, Bunse I, Klämbt C. 2000. Gcm and pointed synergisti- Nave K-A. 2010a. Myelination and support of axonal integrity by glia.
cally control glial transcription of the Drosophila gene loco. Mech Nature 468:244–252.
Dev 91:197–208. Nave K-A. 2010b. Myelination and the trophic support of long axons.
Grenningloh G, Rehm EJ, Goodman CS. 1991. Genetic analysis of Nat Rev Neurosci 11:275–283.
growth cone guidance in Drosophila: fasciclin II functions as a neu- Nave K-A, Trapp BD. 2008. Axon-glial signaling and the glial support of
ronal recognition molecule. Cell 67:45–57. axon function. Annu Rev Neurosci 31:535–561.
Halassa MM, Haydon PG. 2010. Integrated brain circuits: astrocytic Nilsson D-E. 2004. Eye evolution: a question of genetic promiscuity.
networks modulate neuronal activity and behavior. Annu Rev Physiol Curr Opin Neurobiol 14:407–414.
72:335–355. Nilton A, Oshima K, Zare F, Byri S, Nannmark U, Nyberg KG, et al.
Hartline DK. 2011. The evolutionary origins of glia. Glia 59:1215–1236. 2010. Crooked, coiled and crimpled are three Ly6-like proteins
Hartline DK, Colman DR. 2007. Rapid conduction and the evolution of required for proper localization of septate junction components.
giant axons and myelinated fibers. Curr Biol 17:R29–R35. Development 137:2427–2437.
Herculano-Houzel S. 2011. Not all brains are made the same: new views Niva CC, Lee JM, Myohara M. 2008. Glutamine synthetase gene expres-
on brain scaling in evolution. Brain Behav Evol 78:22–36 sion during the regeneration of the annelid Enchytraeus japonensis.
Higgins MR, Gibson NJ, Eckholdt PA, Nighorn A, Copenhaver PF, Dev Genes Evol 218:39–46.
Nardi J, et al. 2002. Different isoforms of fasciclin II are expressed Noble M, Albrechtsen M, Møller C, Lyles J, Bock E, Goridis C, et al.
by a subset of developing olfactory receptor neurons and by 1985. Glial cells express N-CAM/D2-CAM-like polypeptides in
olfactory-nerve glial cells during formation of glomeruli in the moth vitro. Nature 316:725–728.
Manduca sexta. Dev Biol 244:134–154. Oikonomou G, Shaham S. 2011. The Glia of Caenorhabditis elegans.
Hijazi A, Masson W, Augé B, Waltzer L, Haenlin M, Roch F. 2009. Glia 59:1253–1263.
Boudin is required for septate junction organisation in Drosophila and Oshima K, Fehon RG. 2011. Analysis of protein dynamics within the
codes for a diff usible protein of the Ly6 superfamily. Development septate junction reveals a highly stable core protein complex that does
136:2199–2209. not include the basolateral polarity protein Discs large. J Cell Sci
Hilchen von CM, Beckervordersandforth RM, Rickert C, Technau GM, 124:2861–2871.
Altenhein B. 2008. Identity, origin, and migration of peripheral glial Peles E, Joho K, Plowman GD, Schlessinger J. 1997. Close similar-
cells in the Drosophila embryo. Mech Dev 125:337–352. ity between Drosophila neurexin IV and mammalian Caspr pro-
Hilgetag CC, Barbas H. 2009. Are there ten times more glia than neu- tein suggests a conserved mechanism for cellular interactions. Cell
rons in the brain? Brain Struct Funct 213:365–366. 88:745–746.
Hoffman S, Edelman GM. 1983. Kinetics of homophilic binding by Pellerin L, Bouzier-Sore A-K, Aubert A, Serres S, Merle M, Costalat R,
embryonic and adult forms of the neural cell adhesion molecule. Proc et al. 2007. Activity-dependent regulation of energy metabolism by
Natl Acad Sci U S A 80:5762–5766. astrocytes: an update. Glia 55:1251–1262.
Holtmann M, Thurm U. 2001a. Mono- and oligo-vesicular synapses and Pereanu W, Shy D, Hartenstein V. 2005. Morphogenesis and prolifera-
their connectivity in a Cnidarian sensory epithelium (Coryne tubu- tion of the larval brain glia in Drosophila. Dev Biol 283:191–203.
losa). J Comp Neurol 432:537–549. Poliak S, Gollan L, Martinez R, Custer A, Einheber S, Salzer JL, et al.
Holtmann M, Thurm U. 2001b. Variations of concentric hair cells in a 1999. Caspr2, a new member of the neurexin superfamily, is local-
Cnidarian sensory epithelium (Coryne tubulosa). J Comp Neurol ized at the juxtaparanodes of myelinated axons and associates with
432:550–563. K+ channels. Neuron 24:1037–1047.
Hosoya T, Takizawa K, Nitta K, Hotta Y. 1995. glial cells missing: a binary Putnam NH, Srivastava M, Hellsten U, Dirks B, Chapman J, Salamov A,
switch between neuronal and glial determination in Drosophila. Cell et al. 2007. Sea anemone genome reveals ancestral eumetazoan gene
82:1025–1036. repertoire and genomic organization. Science 317:86–94.
Houliston E, Momose T, Manuel M. 2010. Clytia hemisphaerica: a jel- Radojcic T, Pentreath VW. 1979. Invertebrate glia. Prog Neurobiol
lyfish cousin joins the laboratory. Trends Genet 26:159–167. 12:115–179.
Jones BW, Fetter RD, Tear G, Goodman CS. 1995. Glial cells miss- Richards G, Simionato E, Perron M, Adamska M, Vervoort M, Degnan
ing: a genetic switch that controls glial versus neuronal fate. Cell B. 2008. Sponge genes provide new insight into the evolutionary ori-
82:1013–1023. gin of the neurogenic circuit. Curr Biol 18:1156–1161.
Kammerer M, Giangrande A. 2001. Glide2, a second glial promoting Richardson WD, Kessaris N, Pringle N. 2006. Oligodendrocyte wars.
factor in Drosophila melanogaster. EMBO J 20:4664–4673. Nat Rev Neurosci 7:11–18.
Kettenmann H, Hanisch U-K, Noda M, Verkhratsky A. 2011. Physiology Rodrigues F, Schmidt I, Klämbt C. 2011. Comparing peripheral glial
of microglia. Physiol Rev 91:461–553. cell differentiation in Drosophila and vertebrates. Cell Mol Life Sci
Kozmik Z. 2008. The role of Pax genes in eye evolution. Brain Res Bull 68:55–69.
75:335–339. Roots BI. 1981. Comparative studies on glial markers. J Exp Biol
Kozmik Z, Daube M, Frei E, Norman B, Kos L, Dishaw LJ, et al. 2003. 95:167–180.
Role of Pax genes in eye evolution: a cnidarian PaxB gene uniting Rowitch DH. 2004. Glial specification in the vertebrate neural tube. Nat
Pax2 and Pax6 functions. Dev Cell 5:773–785. Rev Neurosci 5:409–419.
Lewis C, Long TAF. 2005. Courtship and reproduction in Carybdea siv- Rowitch DH, Kriegstein AR. 2010. Developmental genetics of verte-
ickisi (Cnidaria: Cubozoa). Marine Biol 147:477–483. brate glial-cell specification. Nature 468:214–222.
Mackie G, Meech R. 1995. Central circuitry in the jellyfish Aglantha. I: Rutishauser U. 2008. Polysialic acid in the plasticity of the developing
The relay system. J Exp Biol 198:2261–2270. and adult vertebrate nervous system. Nat Rev Neurosci 9:26–35.

10 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Saijo K, Glass CK. 2011. Microglial cell origin and phenotypes in health required for subtype-specific glial gene expression. Development
and disease. Nat Rev Immunol 11:775–787. 134:591–600.
Scappaticci AA, Kass-Simon G. 2008. NMDA and GABA B receptors Tiklová K, Senti K-A, Wang S, Gräslund A, Samakovlis C. 2010.
are involved in controlling nematocyst discharge in hydra. Comp Epithelial septate junction assembly relies on melanotransferrin iron
Biochem Physiol, Part A Mol Integr Physiol 150:415–422. binding and endocytosis in Drosophila. Nat Cell Biol 12:1071–1077.
Shandala T, Takizawa K, Saint R. 2003. The dead ringer/retained Tsacopoulos M, Evêquoz-Mercier V, Perrottet P, Buchner E. 1988.
transcriptional regulatory gene is required for positioning of the Honeybee retinal glial cells transform glucose and supply the
longitudinal glia in the Drosophila embryonic CNS. Development neurons with metabolic substrate. Proc Natl Acad Sci U S A
130:1505–1513. 85:8727–8731.
Sherman DL, Brophy PJ. 2005. Mechanisms of axon ensheathment and Tsacopoulos M, Magistretti PJ. 1996. Metabolic coupling between glia
myelin growth. Nat Rev Neurosci 6:683–690. and neurons. J Neurosci 16:877–885.
Silies M, Klämbt C. 2010. APC/C(Fzr/Cdh1)-dependent regulation Vincent S, Vonesch JL, Giangrande A. 1996. Glide directs glial fate com-
of cell adhesion controls glial migration in the Drosophila PNS. Nat mitment and cell fate switch between neurones and glia. Development
Neurosci 13:1357–1364. 122:131–139.
Silies M, Klämbt C. 2011. Adhesion and signaling between neurons and Volterra A, Meldolesi J. 2005. Astrocytes, from brain glue to com-
glial cells in Drosophila. Curr Opin Neurobiol 21:11–16. munication elements: the revolution continues. Nat Rev Neurosci
Soustelle L, Besson M-T, Rival T, Birman S. 2002. Terminal glial dif- 6:626–640.
ferentiation involves regulated expression of the excitatory amino Wang X, Wang T, Jiao Y, Lintig von J, Montell C. 2010. Requirement for
acid transporters in the Drosophila embryonic CNS. Dev Biol an enzymatic visual cycle in Drosophila. Curr Biol 20:93–102.
248:294–306. Watts RJ, Schuldiner O, Perrino J, Larsen C, Luo L. 2004. Glia engulf
Soustelle L, Giangrande A. 2007. Novel Gcm-dependent lineages in the degenerating axons during developmental axon pruning. Curr Biol
postembryonic nervous system of Drosophila melanogaster. Dev Dyn 14:678–684.
236:2101–2108. Wegner M, Riethmacher D. 2001. Chronicles of a switch hunt: Gcm
Soustelle L, Jacques C, Altenhein B, Technau GM, Volk T, Giangrande genes in development. Trends Genet 17:286–290.
A. 2004. Terminal tendon cell differentiation requires the glide/Gcm Weinhold B, Seidenfaden R, Röckle I, Mühlenhoff M, Schertzinger F,
complex. Development 131:4521–4532. Conzelmann S, et al. 2005. Genetic ablation of polysialic acid causes
Soustelle L, Trousse F, Jacques C, Ceron J, Cochard P, Soula C, et al. severe neurodevelopmental defects rescued by deletion of the neural
2007. Neurogenic role of Gcm transcription factors is conserved in cell adhesion molecule. J Biol Chem 280:42971–42977.
chicken spinal cord. Development 134:625–634. Wilson CH, Hartline DK. 2011a. The novel organization and development of
Srivastava M, Simakov O, Chapman J, Fahey B, Gauthier MEA, Mitros copepod myelin. I. Ontogeny. J Comp Neurol 519:3259–3280.
T, et al. 2010. The Amphimedon queenslandica genome and the evo- Wilson CH, Hartline DK. 2011b. The novel organization and devel-
lution of animal complexity. Nature 466:720–726. opment of copepod myelin. II. Non-glial origin. J Comp Neurol
Stacey SM, Muraro NI, Peco E, Labbé A, Thomas GB, Baines RA, 519:3281–3305.
et al. 2010. Drosophila glial glutamate transporter Eaat1 is regulated Wright JW, Copenhaver PF. 2000. Different isoforms of fasciclin II play
by fringe-mediated notch signaling and is essential for larval locomo- distinct roles in the guidance of neuronal migration during insect
tion. J Neurosci 30:14446–14457. embryogenesis. Dev Biol 225:59–78.
Stecca B, Southwood CM, Gragerov A, Kelley KA, Friedrich VL, Gow Wright JW, Copenhaver PF. 2001. Cell type-specific expression of fasci-
A. 2000. The evolution of lipophilin genes from invertebrates to tet- clin II isoforms reveals neuronal-glial interactions during peripheral
rapods: DM-20 cannot replace proteolipid protein in CNS myelin. nerve growth. Dev Biol 234:24–41.
J Neurosci 20:4002–4010. Wu VM, Beitel GJ. 2004. A junctional problem of apical proportions:
Stork T, Engelen D, Krudewig A, Silies M, Bainton RJ, Klämbt C. 2008. epithelial tube-size control by septate junctions in the Drosophila tra-
Organization and function of the blood-brain barrier in Drosophila. cheal system. Curr Opin Cell Biol 16:493–499.
J Neurosci 28:587–597. Yuasa Y, Okabe M, Yoshikawa S, Tabuchi K, Xiong W-C, Hiromi Y,
Suga H, Tschopp P, Graziussi DF, Stierwald M, Schmid V, Gehring WJ. et al. 2003. Drosophila homeodomain protein REPO controls glial dif-
2010. Flexibly deployed Pax genes in eye development at the early evo- ferentiation by cooperating with ETS and BTB transcription factors.
lution of animals demonstrated by studies on a hydrozoan jellyfish. Development 130:2419–2428.
Proc Natl Acad Sci U S A 107:14263–14268. Zappia MP, Brocco MA, Billi SC, Frasch AC, Ceriani MF. 2011. M6
Syed MH, Krudewig A, Engelen D, Stork T, Klämbt C. 2011. The CD59 membrane protein plays an essential role in Drosophila oogenesis.
family member leaky/coiled is required for the establishment of the PLoS ONE 6:e19715.
blood-brain barrier in Drosophila. J Neurosci 31:7876–7885. Zhou Q, Choi G, Anderson DJ. 2001. The bHLH transcription factor
Thomas GB, van Meyel DJ. 2007. The glycosyltransferase Fringe pro- Olig2 promotes oligodendrocyte differentiation in collaboration
motes Delta-Notch signaling between neurons and glia, and is with Nkx2.2. Neuron 31:791–807.

1. E VO LU T I O N O F G L I A L C E L L S • 11
2.
INVERTEBRATE GLIA
Marc R. Freeman

A B B R E VI AT I O N S and are now poised to contribute in similar ways to our under-


standing of fundamental properties of glial cells (see chapter 1).
BBB blood-brain barrier This chapter reviews the features of key invertebrate preparations
CEP cephalic used to study glial cell biology, provides a short overview of what
CEPsh cephalic sheath has been learned from these organisms, compares their biol-
CNS central nervous system ogy to that of mammals, and explores how invertebrate systems
DCC deleted in colorectal cancer might be used in the future to probe deeply into the biology of
DRG dorsal root ganglia glial cell types such as the astrocyte.
Flp flippase
FRT flip recombinase target site 2 N O N G E N ET I C I N VE RT E B R AT E
GPR G protein–coupled receptor M O D E L SYS T E M S
ISN intersegmental nerve
MARCM mosaic analysis with a repressible cell marker The first evidence that neurons could signal to glia through
MG midline glia neurotransmitters came from studies of the squid (Loligo
MN motor neuron pealei) giant axon preparation, which includes surrounding
NMJ neuromuscular junction Schwann cells. In contrast to mammalian Schwann cells found
NT neurotransmitter in the peripheral nervous system, Schwann cells surrounding
PG perineurial glia the squid giant axon lay in parallel to the giant axon, and highly
PNS peripheral nervous system interdigitated, and many together form a single sheath that
pSJ pleated septate junction envelopes the axon shaft (Brown et al. 1991). Schwann cells were
RNAi RNA interference found to exhibit a rapid membrane hyperpolarization follow-
SBC segment boundary cell ing stimulation of giant axons (Villegas 1972), which resulted
SPG subperineurial glia from axonal release of glutamate (Lieberman et al. 1989) that
acted on Schwann cell metabotropic glutamate receptors
(Evans et al. 1985). Interestingly, following activation by gluta-
1 INTRODUCTION mate, Schwann cells activated a glial-glial signaling event: They
released acetylcholine, which in turn increased Schwann cell
The study of invertebrate preparations has illuminated a number hyperpolarization (Evans et al. 1985; Villegas 1974; Villegas
of fundamental principles of neuronal function, including the et al. 1987). Glial control of neuronal electrophysiology was
electrophysiological and chemical basis of the action potential, also established using the snail Helix pomatia. Ca2+-dependent
mechanisms of synaptic vesicle release, and pathways governing long-term changes in glial membrane K+ conductance after
neuronal cell fate specification and growth. Invertebrate prepa- neuronal activity was shown to alter K+ tone in local circuits
rations offer a number of advantages for studying the nervous and thereby affect local firing of neurons (Gommerat and Gola
system, including ease of culture, rapid growth, high experimen- 1994). In addition, basic aspects of metabolic coupling between
tal accessibility, and the opportunity to uniquely identify and neurons and glia were first detailed in invertebrates. In studies
manipulate individual cells in vivo. Historically such preparations, of the honeybee, drone retina glia were shown to be respon-
especially larger preparations such as the squid, grasshopper, or sible for metabolizing and storing extracellular glucose in the
Aplysia, were used for detailed studies of single neuron physiol- form of glycogen (Tsacopoulos et al. 1988, 1994). Local neu-
ogy, morphological analysis, or the effects of cell transplanta- ronal activity was found to induce rapid glycogen metabolism
tion. In the past three decades as the focus has shifted toward in glia (Evequoz-Mercier and Tsacopoulos 1991) (see chapter 36),
understanding the molecular basis of neuronal development which resulted in glial release of alanine for use as metabolic
and function, smaller invertebrates amenable to rapid molecular fuel by neurons (Tsacopoulos et al. 1994).
genetic analysis such as Caenorhabditis elegans and Drosophila The nervous system of the medicinal leech (Hirudo medici-
have become extremely popular experimental systems. Worms nalis) houses two uniquely identifiable astrocyte-like glial cells
and flies have since made profound contributions to delineating per segmental ganglion that are enormous: their cell bodies
ancient and conserved pathways governing neuronal biology, are approximately 100 μm across (Lohr and Deitmer 1997,

12
1999) and their fine processes that fill the synaptic neuropil form the intersegmental peripheral nerve route. The growth
cover an area of 300 to 350 μm in diameter (Kuffler and Potter cones of axons that pioneer the intersegmental nerve (ISN)
1964). For comparison, a hippocampal mouse astrocyte cell show a strong affinity for the SBC; at the point at which they
body is approximately10 μm and their processes cover a spatial meet the SBC they exit longitudinal axon tracts and pioneer the
domain of approximately100 μm in diameter (Bushong et al. ISN. Ablation of the SBC glial cells in vivo results in a striking
2004), whereas the average size of human cortical astrocyte failure of these growth cones to enter the ISN tract (Bastiani
ranges from 100 to 400 μm in diameter (Oberheim et al. and Goodman 1986).
2009). Leech giant astrocyte–like glia, like mammalian astro- A major shortcoming of the invertebrate prepara-
cytes, are coupled electrically through gap junctions, thereby tions discussed in the preceding is the absence of incisive
allowing for coordination of electrical responses (Coggeshall molecular-genetic approaches that can be used to explore how
1974; Kuffler and Potter 1964). Because of their large size, one specific genes promote glial development, function, or neuron-
can perform direct electrophysiological recordings, micro- glia interactions. However, genome sequence databases are
fluorometry, or microinjections (e.g., pharmacological agents, becoming available for some of these organisms (e.g., Apis is
or RNAi constructs) of astrocyte-like glia, while simultane- complete, Hirudo and Loligo are underway), and the recent
ously stimulating specific neurons or inducing simple behav- revolution in the use of RNA interference (RNAi) has opened
iors such as fictive swimming in largely intact preparations. the door for a limited amount of genetic manipulation.
The leech astrocyte–like glial cell is an electrophysiologically
active cell that responds to neuron signaling with changes in Ca2+
signaling and membrane potential. These glia exhibit a selective 3 C A E N O R H A B D I T I S E L E GA N S G L I A
K+ conductance and can act as sinks for K+ in spatial buffering
(Kuffler and Nicholls 1966). They actively regulate the pH of
3.1 H I S TO L O GY A N D E X P E R I M E N TA L U T I L IT Y
the intracellular and likely extracellular compartments through
O F C A E NO R H A B D I T I S E LEGA N S G L I A
mechanisms including a Na+-HCO3 cotransporter (Deitmer
1991; Deitmer and Schlue 1987, 1989; Szatkowski and Schlue The nervous system of the nematode Caenorhabditis elegans
1994). By controlling extracellular pH these glia could play a consists of only 302 neurons and 50 glial-like cells based on
direct role in regulating neuronal electrophysiology (Deitmer morphological and functional criteria (Shaham 2005, 2006).
and Schneider 1995; Deitmer et al. 1999). Leech astrocyte–like All worm glia are closely associated with peripheral sensory
glia exhibit dramatic Ca2+ fluxes that are regulated potently by structures; none are associated exclusively with the nerve
membrane potential (Nett and Deitmer 1998). Ca2+ increases ring (which is the worm brain). As such, it has been argued
are mediated largely by entry of extracellular Ca2+ (Deitmer et al. that C. elegans glia may represent a good example of the evo-
1999), although glutamate application can elicit Ca2+ release from lutionarily ancient glial subtype, which might have appeared
intracellular stores (Lohr and Deitmer 1997, 1999). Ca2+ spikes first in the peripheral nervous system (Reichenbach and
can occur throughout the soma, but also occur at the local level Pannicke 2008). Glia in the worm can be divided into two
and in a heterogeneous fashion (Lohr and Deitmer 1999), poten- broad classes: 24 are sheath glia, which ensheathe a subset
tially responding to and regulating neural activity. Astrocyte-like of sensory neuron dendrites; and 26 are socket glia, which
glia indeed respond with rapid changes in membrane potential surround sensory neuron dendrite tips to form a pore to
to a number of neurotransmitters, including glutamate, acetyl- allow for direct exposure of dendrites to the environment
choline, serotonin, or GABA (Deitmer and Rose 1996; Deitmer (Fig. 2.1). A subset of four sheath glia, the cephalic sheath
et al. 1998; Lohr and Deitmer 1999; Muller et al. 2000; Schmidt (CEPsh) glia, are bipolar and not only ensheathe sensory
and Deitmer 1999). This responsiveness likely represents normal dendrite tips but also extend flattened membrane processes
glial function in the leech, because stimulation of even a single toward the nerve ring. Each of the 50 C. elegans glia are
cell, the neuromodulatory Leydig neuron, promotes hyperpolar- uniquely identifiable, associate with well-defined subsets of
ization of the giant astrocyte–like glia with the amplitude and rate sensory neurons, and one can therefore study neuron–glia
of hyperpolarization increasing with the frequency of neuronal interactions in single-neuron pairs.
stimulation (Britz et al. 2002; Schmidt and Deitmer 1999). Worms are completely transparent, thereby allowing for
Additional invertebrate preparations have made important imaging and manipulation of all glial cells in the intact organ-
contributions to our understanding of how glial cells coordi- ism. Promoters have been identified that can be used to drive
nate their developmental program with neurons, or how glia expression of any transgene of interest in very specific sub-
guide key steps in neuronal development. Glia in the antennal sets of glia or neurons. Available lines of experiments range
lobe of the moth Manduca sexta have proved quite useful in from simple imaging of glia during development, through
understanding the cellular basis of how neuronal contact initi- opto-genetic activation of specific neurons in mature circuits
ates glial organization of antennal lobe glomeruli (Tolbert and while imaging glial Ca2+ transients in glia with genetically
Oland 1990), or how glia guide the sorting of olfactory recep- encoded Ca2+ indicators. Because the entire cell lineage of
tor neuron axon fibers to the correct location (Rossler et al. C. elegans is well defined one can perform ablations of single
1999). In some cases even single axon–glia interactions have cells or precursors early in development and then explore
been examined in insects. For example, during normal embry- developmental roles for glia in nervous system assembly, or
onic development of the grasshopper, the segment boundary ablate single glial cells at adult stages and examine glial func-
cell (SBC), a glial cell, establishes a position that will ultimately tion in the mature nervous system (see later for examples).

2. I N VE RT E B R AT E G L I A • 13
Sensory 3.2 G L I A L C O N T RO L O F N EU RO NA L
A Sheath
Socket neuron Nerve ring D EV E L O PM E N T I N T H E WO R M

B Single cell ablation studies in C. elegans have revealed that worm


glia modulate diverse aspects of neuronal development from
neurite extension, dendrite morphogenesis, axon pathfinding,
synaptogenesis, to nerve ring assembly. For example, ablation of
CEPsh glia, which are closely associated with cephalic (CEP)
Dendrites/processes Cell sensory neurons from early developmental stages, led to defects
bodies in CEP neuron dendrite extension and wiring of sensory axons
B in the nerve ring (Yoshimura et al. 2008). Ablation of amphid
sheath glia (those sheath glia that do not extend processes to
(Environment) the nerve ring) had little affect on dendrite extension, but led
Cuticle to many sensory neurons elaborating a very simple dendritic
architectures compared with controls. Interestingly, a sub-
set of sensory neurons appeared unaffected by glial ablations,
Socket
indicating that some neurons can extend and elaborate den-
Glia
Sheath drites properly in the absence of glial input (Bacaj et al. 2008).
Finally, because dendrites and glial processes are organized into
mature sensory structures, glial cells control the precise size of
the sensory compartment that houses dendrite tips through
Cilia the opposing actions of the Patched-related molecule DAF-6,
Sensory which restricts the expansion of the sensory compartment, and
neuron
Dendrite LIT-1, which promotes compartment growth (Oikonomou
et al. 2011). Notably, C. elegans sensory neurons survive in the
Figure 2.1 Caenorhabditis elegans Glial Subtypes and Associations with absence of glia, arguing against a role for worm glia in sustain-
Neurons. A. Anterior tip of the adult stage of C. elegans. Caenorhabditis ing neuronal survival.
elegans glia are highly polarized and extend processes from the cell bodies
to the tips of sensory neurons (blue). Socket glia (green) are unipolar and
C. elegans glia also play an important role in synapto-
only ensheathe sensory neuron tips. Most sheath glia are also unipo- genesis in the nerve ring, and this appears to extend to the
lar, but a subset termed CEPsh glia (red) are bipolar and also extend level of individual synapse placement. The AIY interneu-
processes posteriorly that ensheathe the nerve ring. B. At sensory neuron ron forms synaptic contacts with the RIA interneuron in
tips (box region enlarged from A) socket glia form a pore through which the nerve ring, and these are in close proximity to CEPsh
sensory neuron ciliated endings extend into the environment. Sheath
glia enwrap the tips of sensory neurons: Some sensory neuron ciliated
glia. UNC-40 (worm DCC, deleted in colorectal cancer), a
endings extend into the environment, whereas others are fully or partially receptor for UNC-6, is expressed in AIY and important for
embedded within the sheath glial cell. proper positioning of AIY-RIA synapses (Colon-Ramos et
al. 2007). Interestingly, UNC-6 is expressed in CEPsh glia
(Yoshimura et al. 2008), where it acts to cluster neuronal DCC
Caenorhabditis elegans is also remarkable with respect to the and promote AIY-RIA synapse formation (Colon-Ramos
genetic approaches available. Worms offer the opportunity et al. 2007). In unc-6 mutants, AIY-RIA synaptic connectivity is
to perform powerful, rapid, and unbiased forward genetic defective. Strikingly, simply repositioning of CEPsh glial mem-
screens to identify mutations affecting any aspect of glial branes more posteriorly in the nerve ring is sufficient to pro-
cell biology that can be observed in vivo. Unlike more com- mote the formation of ectopic AIY-RIA synapses within these
plex organisms, C. elegans can survive even when the worm domains, arguing that AIY-RIA form synapses in response to
harbors extremely deleterious mutations—as long as the pha- instructive cues from CEPsh glia (Colon-Ramos et al. 2007).
ryngeal pump is working (which is controlled by a very small
number of neurons), the worm is able to feed and survive to
3.3 G L I A L RO L E S I N B E H AVI O R
late stages even when otherwise paralyzed. In addition because
hermaphrodites self-fertilize they can self-cross and generate Ablation of glial cells in the adult allows for direct assessment
additional progeny without the need for mating. As such, one of their roles in sensory-driven behavior and neuronal signal-
can explore the in vivo function of a number of genes that ing. A particular strength of the worm is behavioral analysis—
would cause early lethality in other animals. This is especially the sensory-driven responses driven by uniquely identifiable
important for C. elegans researchers because genetic mosaic neurons are well described and can be used to directly assess
approaches, although available, are not as straightforward as the function of specific neurons. Bacaj et al. laser ablated
in Drosophila or mice. Finally, in recent years collections of adult sheath glia and assayed the function of three uniquely
whole-genome RNAi libraries that contain constructs that identifiable neurons AWC, AWA, and AWB. Strikingly, glial
target nearly every C. elegans gene can be used for RNAi-based ablation dramatically reduced AWC- and AWA-mediated
screens in which RNAi constructs carried by bacteria are fed chemotaxis toward odors, but had no effect on AWR func-
to animals (Kamath et al. 2003). tion (Bacaj et al. 2008). Because AWR functions normally

14 • NEUROGLIA
without glia, behavioral defects in AWC and AWA cannot A Larval brain-neurons B Larval brain-glia
be explained by a simple structural change in sensory organ (dorsal view) (cross section) wrapping
morphology and the roles for glia in neuronal function astrocyte ensheathing
(peripheral
nerve)
appear to be neuron specific. Moreover, neuronal signaling
molecules (e.g., odorant receptors and G proteins) localized
normally in the absence of glia, arguing against a defect in neuropil
motorneuron

maintenance of sensory machinery. Interestingly, Ca2+ imag- neuronal


ing of osmosensory neurons in the same glial-ablated prepa- cell cortex interneuron cell body
perineurial

rations revealed a loss of stimulus-induced increases in Ca2+ (cortex) subperineurial

levels in neurons that are normally seen within seconds in


controls. Thus glia appear essential even at the earliest stages
of sensory neuron signaling (Bacaj et al. 2008). The molecu- C neuromuscular junction D sensory organ
lar basis by which glia modulate neuronal physiology remains wrapping
unclear, but recent work has revealed that glial ACD-1 subperineurial dendrite shaft
(acid-sensing channel degenerin-like) coordinates with the
neuronal DEG/ENaC channel DEG-1 to modulate acid and socket sheath cell
cell
lysine chemotactic behaviors (Wang et al. 2008, 2012).
muscle
sensory neuron glia (axonal)

4 DROSOPHILA GLIA Figure 2.2 Subtypes, Positions, and Morphology of Drosophila Glia.
A. Overview of the histology of the Drosophila larval central nervous
The Drosophila nervous system is far more complex than that system. The neuronal cell cortex (gray) houses all neuronal and most
of C. elegans—the adult fly nervous system houses approx- glial cell bodies. All CNS synaptic contacts between neurons are found
imately150,000 to 200,000 neurons. The precise number of within the neuropil (light gray). B. Position and morphology of glial
subtypes (green) in the Drosophila larval CNS. Glial morphology in the
glia remains unclear, but likely represents approximately10% adult brain is essentially the same. See text for details for B, C, and D.
of the total population of cells in the nervous system. The Midline glia are not shown. C. Glia at the Drosophila larval neuromuscu-
Drosophila brain is quite sophisticated structurally and func- lar junction. Motoneuron (MN) terminals are buried in the muscle, but
tionally, and compared with C. elegans bears much more subperineurial glia (light green) invade the space between the MN and
resemblance to the mammalian nervous system. Distinct brain muscle. D. Sensory organs in Drosophila contain at least three glial-like
cell types: the socket cell, the sheath cell, and an axon-associated glial cell.
regions are subdivided into lobes, with each lobe coordinating A mechanosensory organ is shown as an example.
specific neurophysiological processes, and many brain regions
are interconnected by well-defined nerves. Fly circuits contain
neurons with electrophysiological properties remarkably sim-
ilar to mammalian neurons: They modulate a diverse behav- et al. 2008), and these are thought to be involved in deposi-
ioral repertoire and exhibit both electrophysiological and tion of a carbohydrate-rich neural lamella that surrounds
behavioral plasticity. As discussed in the following, an array of the CNS and peripheral nerves (Leiserson et al. 2000) that
functional classes of glia exist in the fly, each participating in serves as an additional physical and chemical barrier to entry
its own way in nervous system development and function, and of molecules into the CNS (Carlson et al. 2000). Peripheral
many Drosophila glia appear to perform functions remarkably nerves, although covered by the BBB composed of SPGs and
similar to their mammalian counterparts. PGs, also house a population of glia termed wrapping glia that
directly interact with and wrap motor and sensory neuron
axons (Beckervordersandforth et al. 2008; Stork et al. 2008).
4.1 MO R P H O L O G I C A L C O M P L E X I T Y
At the neuromuscular junction, SPGs dynamically invade the
O F D RO SO PH I L A G L I A
space between the motor neuron terminal and postsynaptic
In the embryo, larva, and adult, the Drosophila nervous system muscle cells and interact closely with growing synaptic fields
can be divided into two major histological regions: the neu- (Fig. 2.2C) (Fuentes-Medel et al. 2009).
ronal cell cortex, where neuronal and glia cell bodies reside; In the CNS, cortex glia are found among neuronal cell
and the neuropil, where axons and dendrites project to form bodies, and by the late embryonic stages appear to ensheathe
neural circuits (Fig. 2.2A). Each of these domains contains each neuronal cell body. Impressively, a single cortex glial cell
multiple classes of glial cells (Fig. 2.2B). The entire Drosophila can surround as many as 100 or more neuronal cell bodies
nervous system is surrounded by a blood-brain barrier (BBB) (Awasaki et al. 2008). The neuronal cell cortex is separated
composed of subperineurial glia (SPGs). They exhibit a flat- from the neuropil by ensheathing glia, which compartmental-
tened morphology, cover the entire surface of the CNS, and izes different regions of the CNS synaptic neuropil and wrap
seal the BBB by forming pleated septate junctions (pSJs) nerves (Ito et al. 1995). Within the synaptic neuropil are also
with one another (Auld et al. 1995; Baumgartner et al. 1996; astrocytes (Awasaki et al. 2008; Doherty et al. 2009), which
Schwabe et al. 2005). At late larval and adult stages an addi- have a highly tufted morphology, have an approximate size
tional layer of glia, termed perineurial glia (PGs), forms on the of 20 to 30 μm in diameter, and associate closely with most
surface of SPGs (Awasaki et al. 2008; Pereanu et al. 2005; Stork synapses.

2. I N VE RT E B R AT E G L I A • 15
Adult Drosophila sensory organs also house glia that are such as gfp or GCaMP, or optogenetic molecules including
quite similar to those found in C. elegans (Fig. 2.2D). For ChR2). In essence one can carefully control the spatiotem-
example, mechanosensory organs house socket glial cells, poral expression of any gene of interest in any cell type for
which form the socket through which sensory dendrites which there is a defined promoter. Multiple Gal4 driver lines
extend, and sheath glial cells that enwrap the neuron and prox- are available for nearly all subtypes of Drosophila glia (Stork
imal dendrites (Walker et al. 2000). An additional glial cell is et al. 2012). Because each of the binary expression systems
also born during development that migrates down the axons function independently, one can combine them in the same
and ensheathes it (Gho et al. 1999). animal to label or manipulate multiple cell types simultane-
ously in vivo. For example, astrocytes can be labeled using
the alrm-Gal4 driver (Doherty et al. 2009), whereas one
4.2 T EC H N I C A L A D VA N TAG E S O F
labels neurons with 201Y-QF (Potter et al. 2010) and astro-
S T U DY I N G D RO SO PH I L A G L I A
cyte–neuron interactions can be assayed in live preparations.
Drosophila offers a number of powerful genetic and molec- Multiple whole-genome UAS-driven RNAi collections are
ular approaches with which to study glial cell development now available in Drosophila and it is therefore possible to
and function in vivo. As with C. elegans, the lineages of knockdown almost any gene with high cellular specificity
Drosophila embryonic glia are fully characterized in both the and assay phenotypic consequences.
CNS and PNS with near single-cell resolution. A number Mosaic analysis is a particular strength of Drosophila and
of molecular markers have been characterized that uniquely has improved dramatically over the last decade. Gene function
label all, or even specific subtypes of Drosophila glia (Stork can be analyzed in the fly using mosaic approaches that allow
et al. 2012). Impressively, multiple binary expression systems for the specific labeling of small, or even single homozygous
are now available in the fly: Gal4/UAS (Brand and Perrimon mutant cell clones. Flippase (Flp)-mediated recombination
1993), LexA/LexAOp (Lai and Lee 2006), and QF/QUAS works extremely well in the Drosophila nervous system with
(Potter et al. 2010) (Fig. 2.3A). Binary expression systems flip recombinase target (FRT) sites and can be used to acti-
allow one to use cell type specific promoters (e.g., astrocyte vate, or inactivate, transgene expression in large groups of cells,
gene promoters) to drive Gal4 expression, which in turn acti- or even in single-cell clones (Fig. 2.3B) (Stork et al. 2012).
vates the tissue specific expression of any transgene placed One can generate a “Flp-out” cassette, in which Flp-mediated
under UAS control (e.g., a mis-expression transgene, markers recombination leads to a change in a gene’s placement relative

A Drosophila binary expression sysytems B Flp/FRT-mediated transgene regulation

glial prometer +++ glial


Gal4 prometer stop reporter gene
UAS (OFF)
FRT FRT
gene X
Flp-medicated
glial prometer recombination
+++
LexA LexAOp
glial
gene Y prometer reporter gene
(ON) FRT
glial prometer +++
QF QUAS
gene Z

C Mosaic analysis with a repressible cell marker (MARCM) D astrocyte MARCM


glial nuclei
mut neuropil
mut mut
mut
astro-Gal4, UAS-GFP
Gal80 Gal4/UAS active
Gal80 Gal80
Gal80
astro-Gal4, UAS-GFP
astro-Gal4, UAS-GFP
Gal4/UAS repressed by Gal80 Gal4/UAS repressed by Gal80

Figure 2.3 Genetic Mosaic Approaches in Drosophila. A. The Gal4/UAS, LexA/LexAOp, and QF/QUAS binary expression systems. Each
functions independently (e.g., Gal4 cannot activate LexAOp or QUAS) in Drosophila. B. Flp-mediated excision of FRT-flanked regions can activate
or inactivate transgene expression. C. Mosaic analysis with a repressible cell marker (MARCM) allows for the production of homozygous mutant,
GFP-labeled clones in an otherwise wild-type nervous system. Gal80 normally represses Gal4/UAS activation. In a MARCM clones after cell division
Gal80 is lost in one daughter cell where the trans chromosomal arm is also made homozygous and Gal4/UAS is de-repressed. D. A MARCM clone in
a Drosophila adult brain astrocyte. Courtesy of Ozge Tasdemir.

16 • NEUROGLIA
to a promoter (ON, putting it near the promoter; or OFF, sep- a rescuing transgene only in cells of interest to demonstrate
arating it from the promoter). Alternatively, one can use Flp autonomy of gene function.
to remove the Gal80 repressor in a cell- and/or developmental It should be emphasized that each of the preceding tech-
stage-specific manner. niques can be further combined with the powerful forward
Mosaic analysis with a repressible cell marker (MARCM) genetic approaches available in Drosophila to study virtually
combines flippase Flp/FRT mediated recombination events any glial function in vivo. In addition, as the wiring diagram of
with Gal4/UAS and the Gal4 repressor Gal80 to tightly the Drosophila nervous system becomes clearer, one can more
control marker gene (e.g., GFP) expression only in mutant specifically manipulate glial genes or glial function and deter-
cells (Fig. 2.3C,D) (Lee and Luo 2001). Briefly, FRT sites are mine how perturbation of glial physiology affects the process-
available at the base of all five major chromosomal arms. In ing of information by neural circuits.
MARCM, a mutation of interest is placed distal to an FRT site
and in trans over a chromosome arm contain the same FRT
with a distal Gal80 element. Also included in the background 4.3 D EVE L O PM E N TA L A N D F U N C T I O NA L
is the Gal4 driver of interest (e.g., a glial Gal4) and a marker P RO P E RT I E S O F D RO SO PH I L A G L I A
gene (e.g., membrane-tethered GFP). Before recombination
the Gal4/UAS system is constitutively repressed by Gal80. If a
4.3.1 Axon Sorting and Wrapping
Flp source is supplied at mitosis, this promotes recombination
at FRT sites, and at some frequency clones are generated that The ensheathment of axons by wrapping glia in peripheral
are homozygous mutant for a gene of interest, and have lost the nerves during Drosophila larval stages is remarkably similar
Gal80 repressor. The Gal4/UAS system is therefore derepressed at the cellular level to radial sorting of axons by oligodendro-
in homozygous mutant clones (but not in other genetic types), cytes or Schwann cells in mammals (see chapters 42 and 44).
and cells are labeled with GFP. This has the important advan- Drosophila motor neuron and sensory neuron axons extend
tage of allowing one to examine homozygous mutant clones projections out of, or into the CNS, respectively, by late embry-
(that are marked) in an otherwise wild-type nervous system. onic stages through the peripheral nerves. At early larval stages
Genetic mosaics can also be generated using Gal4/UAS alone. axons are initially clustered into large bundles that are in close
For example, one can knockdown genes specifically in glial contact with wrapping glia that exhibit a relatively simple mor-
cells by driving glial-specific expression of UAS-RNAi con- phology (Fig. 2.4A). Impressively, during larval stages these
structs. Alternatively, in mutant backgrounds one can resupply wrapping glia do not divide but grow 10- to 20-fold in length,

A late embryo/
3rd instar larva
1st instar larva
axons

subperineurial glia

wrapping
glia axon sorting
B
10-20X growth of
nerve length
perineurial
glia (~3 days)

perineurium

B C Dcont NrxIV Nrg


Wrapping
(hemolymph) glia
Moody
Claudin
subperineurial
glia
Cora
perineurium Ga Gγ
Gβ loco pSJ assembly
axon
(solute cortical action assembly
exchange) membrane interdigitation

Figure 2.4 Formation of Nerve Bundles, Axon Sorting, and Blood-Brain Barrier in Drosophila. A. Morphology of peripheral nerves in the larva
immediately after hatching (first instar) and 3 to 4 days later at third instar larval stages. Note the dramatic sorting of axons (red) into small bundles
by wrapping glia by third instar. B. Morphology of the blood-brain barrier in Drosophila (enlarged from A). Entry into the CNS requires passage
through the carbohydrate-rich perineurium, subperineurial glia (SPG), and often wrapping glia. C. Molecular components required for assembly of
the SPG-SPG pleated septate junction (enlarged from B). See text for details.

2. I N VE RT E B R AT E G L I A • 17
and by the third instar larval stage axons are efficiently sorted by pSJs, and in many cases also wrapping glia (or in the CNS
by wrapping glia into smaller clusters of a few or even sin- cortex glia). Based on their close contact with SPGs, it may
gle axon fibers (Leiserson et al. 2000; Stork et al. 2008). The be that wrapping glia and cortex glia act as a direct conduit
molecular mechanisms underlying this sorting event remain for exchange of material into and out of the CNS. Likewise,
poorly defined, but axonal protein Vein seems to signal to the Drosophila glial cells likely also play a key role in the exchange
EGF receptor in glia to coordinate activation of glial genes and of O2/CO2 in the nervous system since tracheal elements (i.e.,
nerve morphogenesis (Sepp and Auld 2003), much like axonal the fly breathing apparatus) are closely associated with multi-
Neuregulin1 signals to ErbB2/B3 in Schwann cells in mam- ple glial cell types in the CNS (Pereanu et al. 2005).
mals to modulate Schwann cell survival and myelination of
axons (Garratt et al. 2000).
4.3.3 Neural Circuit Assembly, Synaptic Growth,
and Trophic Support
4.3.2 Formation of the Blood-Brain Barrier
Glial cells in Drosophila exert powerful control of neural cir-
The blood-brain barrier (BBB) of Drosophila is first estab- cuit assembly during early axon guidance, and later as circuits
lished in the embryo and insulates the CNS and periph- undergo activity-dependent modification. Perhaps the most
eral nerves from the surrounding hemolymph (see chapter famous example is Drosophila midline glia (MG). Early in
33). The fly CNS appears to be immune privileged in that CNS development these cells are arranged along the midline
peripheral macrophages fail to enter the CNS under nor- and express potent guidance cues including Netrin and Slit
mal conditions. The first layer of the BBB surrounding the that govern nearly all longitudinal and commissural axon
brain (from inside to out) are the subperineurial glia (SPGs) path finding decisions at the Drosophila embryonic mid-
that form pleated septate junctions (pSJs) with one another line, in many ways acting similarly to the mammalian floor
(Fig. 2.4B,C). The injection of dye-coupled dextrans into plate ( Jacobs 2000). At late embryonic stages (initially 12)
the hemolymph revealed that the BBB becomes impenetra- MG compete for trophic factors on commissural axons, most
ble to relatively large dextrans (10kDa) by mid-embryonic undergo programmed cell death, and three MG per segment
stages, a time point that coincides with the SPGs taking on a migrate dorsally and ultimately ensheathe commissural axons
sheet-like morphology and forming pSJs with their neighbors (Bergmann et al. 2002). Axonal target layer selection is also
(Schwabe et al. 2005). The infolding of SPG membranes with modulated by specific classes of glia during visual system
adjacent cells and formation of pSJs is mediated by the Moody development. Photoreceptor axons emanating from the lar-
receptor, an SPG-expressed 7TM GPR (Bainton et al. 2005; val eye disk project into the CNS optic lobe where they must
Schwabe et al. 2005) (Fig. 4.4C). In moody mutants the inter- decide which target layer to innervate. Photoreceptors 1 to 6
digitation of pSJs at points at which pSJs will form is signifi- (R1–6) target the laminar compartment, which is defined by
cantly decreased, and pSJs do not form properly (Schwabe et layers of glial cells, while photoreceptors 7 and 8 (R7 and R8)
al. 2005). Dye-dextran injections demonstrate the embryos project through this glial layer into the medulla. Perturbation
lacking Moody have a permeabilized BBB at both embry- of the layer of glia separating the lamina from medulla dra-
onic (Schwabe et al. 2005) and adult stages (Bainton et al. matically affects axon targeting. If glia are not present R1 to 6
2005), but not to the point at which animals are paralyzed fail to innervate the lamina and instead project more deeply
(as is the case in other mutants with more significantly per- into the medulla compartment, indicating that glial cells pro-
meabilized BBBs), indicating that significant sealing of the vide a spatial cue that selectively promotes the termination
BBB has still occurred. Maintenance of the BBB is an active of R1 to 6 growth, while not affecting R7 or R8 (Poeck et al.
and Moody-dependent process throughout the fly life cycle: 2001). The molecular identity of this cue remains undefined.
Activation of a moody-RNAi construct in adults after nor- Neural circuit assembly in mammals often entails an initial
mal BBB formation led to permeabilization, and, strikingly, phase of overproduction of neurons and hyper-innervation of
turning off expression of the moody-RNAi element resulted in the target areas, followed by selective elimination of exuberant
recovery of BBB function (Bainton et al. 2005). connections and superfluous neurons. Similar events occur in
The components of Drosophila pSJs are molecularly Drosophila during both embryonic and pupal stages of nervous
related to components of the mammalian axon–glial sep- system development and glia play an essential role in the removal
tate junctions found at paranodes. Molecular components of neuronal debris. For example, approximately 450 neurons are
include Drosophila contactin (Dcont), NeurexinIV (NrxIV), generated per embryonic hemisegment, but roughly one-third
Neuroglian (Nrg), Coracle (Cora), and Claudin (Fig. 4.4C) of these undergo programmed cell death (Rogulja-Ortmann et
(Banerjee et al. 2006), and mutations in any of these molecules al. 2007). Multiple subtypes of glial cells rapidly engulf these
compromise pSJ function and permeabilization of the BBB. cell corpses to clear them from the CNS (Sonnenfeld and Jacobs
The consequences of pSJ breakdown are thought to include 1995). At larval stages, Drosophila mushroom body gamma neu-
influx of K+ from the hemolymph (which is K+-rich), per- rons initially project both medial and dorsal processes during
turbed neuronal firing, and animal paralysis (Auld et al. 1995). larval stages, but at metamorphosis both the medial and dorsal
It is likely that the BBB is the key site of regulatory exchange axonal projections and dendrites of gamma neurons are pruned
between the hemolymph and the CNS. For any molecule to to rewire the mushroom body with adult-specific axon mor-
gain entry into or to exit the CNS, it must pass through the phologies. Glial cells invade the mushroom body lobes at the
carbohydrate-rich perineurium, a layer of SPGs that are sealed initiation of axon pruning (Fig. 2.5A) and secrete the TGF-β

18 • NEUROGLIA
molecule Myoglianin, which is essential for activation of axonal presynaptic debris in the form of shed membrane fragments or
degeneration (Awasaki et al. 2011). As axons fragment glial cells aborted synaptic boutons. Closer examination of MN termi-
engulf and degrade degenerating axonal debris, genetic block- nals in controls revealed that wild-type growing synapses nor-
ade of glial engulfing activity potently blocks axonal pruning mally shed large amounts of debris in an activity-dependent
(Awasaki and Ito 2004) and genetically labeled axon fragments manner. At the NMJ shed presynaptic debris is normally
can be found within phagocytic glial cells (Watts et al. 2004). phagocytosed and degraded by the combined activity of glia
Precisely how much axonal and dendritic material is pruned and the postsynaptic muscle cell (Fig. 5C). When efficient
during Drosophila metamorphosis remains unclear, but it seems clearance of presynaptic debris by glia was blocked, its accu-
likely that glial cells are the primary cell type responsible for mulation potently blocked synaptic growth, arguing that effi-
clearing most neuronal debris in the CNS. Interestingly, recent cient clearance is essential for the addition of new synaptic
work has shown that glia can also play a protective role during contacts (Fuentes-Medel et al. 2009). Under some conditions
the pruning of neurites. In the larval peripheral nervous system it also appears that glia can also actively promote synapse loss
proximal sensory dendrites are ensheathed by glial membranes and MN retraction from the NMJ. In mutants defective in the
and dendrite degeneration for pruning initiates precisely where spectrin/ankyrin cytoskeleton NMJ terminals initially form,
the glial sheath ends, suggesting a novel neurite protective role but later exhibit significant retraction of MN synaptic bou-
for glial membranes (Fig. 2.5B) (Han et al. 2011). tons. During this retraction event glial cells secrete the TNF-α
There is increasing interest in the role of glial cells in the molecule Eiger, which binds to the TNF-α receptor Wengen
formation and plasticity of synapses. An excellent preparation and activates a caspase-dependent pro-degenerative signal-
in which to study glial–synapse interactions in Drosophila is ing pathway in the MN terminal to promote retraction. Loss
the larval neuromuscular junction (NMJ). Glial cells dynami- of Eiger, Wengen, or other components of this neuron → glia
cally invade the NMJ and associate closely with synaptic signaling pathway significantly suppressed retraction, arguing
contacts between the motoneuron (MN) and postsynaptic that glia instruct MN terminals during this example of pro-
muscle cell (Fuentes-Medel et al. 2009). The morphology and gressive denervation (Keller et al. 2011).
size of the Drosophila NMJ allows for excellent microscopic In mammals, glial neurotrophins provide critical support
examination, which has led to the discovery that this rapidly for neuronal survival. Obvious components of the neurotro-
growing synaptic field is quite wasteful. In mutants defective phin family long eluded discovery in Drosophila, and it was
in engulfment function (e.g., draper, or dCed-6, see the follow- therefore proposed that glia–neuron trophic support might
ing) MN terminals were found to accumulate large amounts of not be essential for construction of the fly nervous system.

A CNS axon/dendrite pruning B PNS neurite protection

dendrites
neurites degenerate growth of adult-
glia engulf debris specific neurites
dendrites
protected

dendrites
glia degenerate
3rd Instar early pupa mid/late pupa
larva

C clearance of shed neuronal debris D NT uptake/recycling

glia shed debris Gln


Gs
shed bouton
Glu Glu

Glu EAAT

GAT GABA
degradation
motor GABA
muscle cell neuron
glial cell

Figure 2.5 Functional Roles for Glial Cells in Neural Circuit Assembly, Growth, and Function. A. Drosophila glial cells actively engulf axons and
dendrites early in pupal stages during mushroom body gamma neuron pruning. B. In the peripheral nervous system dendritic processes that are housed
within glial sheaths are protected from degeneration at metamorphosis. C. Muscle growth requires significant expansion of the synaptic field at the
Drosophila neuromuscular junction (NMJ). During larval stages glia invade the growing NMJ and, working coordinately with muscle cells, engulf shed
synaptic debris (membrane fragments and shed synapses). D. In mature neural circuits Drosophila glia express channels and neurotransmitter metabo-
lizing enzymes to take up and recycle glutamate and GABA from the synaptic cleft.

2. I N VE RT E B R AT E G L I A • 19
However, multiple studies have demonstrated that ablation of neurodegeneration. Intriguingly, the onset of degeneration
Drosophila glia results in a dramatic loss of neurons, and more and motor defects could be suppressed by application of drugs
recently a number of molecules with neurotrophic properties used to treat excitotoxicity in humans (Rival et al. 2004). Glia
have been identified in Drosophila. The Drosophila neurotro- also express enzymes required for the subsequent metabo-
phin–like family of molecules includes Drosophila neurotro- lism of glutamate, including glutamine synthetase (Freeman
phin 1 (DNT1), 2 (DNT2), Spatzle (Spz) (Zhu et al. 2008), et al. 2003), which converts glutamate to glutamine for sub-
and dmMANF (Palgi et al. 2009). Each of these molecules, sequent redelivery to neurons. Drosophila glia also appear to
based on loss of function studies, appears to signal to neurons closely maintain the ionic balance of the nervous system. fray
to promote maintenance of axonal fascicles and neuronal sur- mutants were initially identified as having large bulges in larval
vival. Spz appears to act through its receptor Toll (Zhu et al. peripheral nerves. Fray was later identified to be a Drosophila
2008), but how the additional fly neurotrophin–like mol- PASK molecule, which in subsequent work was shown to inter-
ecules signal remains to be determined. act with the Na-K-Cl cotransporter Ncc69 and regulate water
volume and likely osmotic homeostasis (Leiserson et al. 2011).
4.3.5 Neurotransmitter Uptake and Recycling,
Maintenance of Ionic Balance 4.3.6 Activation of Glia After Neural Injury,
Glial Phagocytic Activity
The neurotransmitters (NTs) glutamate and GABA are widely
used in signaling in the invertebrate nervous system. After Axotomy induces potent responses from Drosophila glia
synaptic release, rapid clearance of these NTs is essential to in the adult brain. Within hours after axon injury, local
allow for subsequent signaling events to occur properly, and glial cells upregulate expression of the engulfment receptor
avoid excitotoxicity or synaptic spillover. Drosophila CNS Draper, extend membranes to regions of the brain housing
glia express NT transporters for both glutamate and GABA: degenerating axons, and phagocytose axonal debris (Fig. 2.6A)
EAAT1 or EAAT2 (Freeman et al. 2003), or GAT (Thimgan (MacDonald et al. 2006). Draper encodes the Drosophila
et al. 2006) (Fig. 2.5D). Loss of EAAT1 function results in ortholog of the cell corpse engulfment receptor CED-1,
defects in motor behavior in adult flies, and age-dependent which is required in the engulfing cell for the recognition and

A
Reactive responses after neural injury
glial activation
“resting” phagocytosis of axonal debris termination of response
glomerulus

axotomy ~1 week

axons

ensheathing glia
B C

Cell corposes

apoptotic
cell corpose
Draper
-6
ed
ark
dC
Sh

Simu
Draper
P-

Src42a

engulfment and
degradation

Figure 2.6 Phagocytic Roles for Drosophila Glial Cells During Development and Activation After Axonal Injury. A. In response to axon (red)
injury glial cell membranes (green) invade regions containing axonal debris, internalize axon fragments through phagocytosis, and ultimately
return to a resting state. For clarity, only a subset of axons are shown. B. Glial cells engulf and degrade developmentally produced CNS neuronal
cell corpses at embryonic and pupal stages through activation of the Draper signaling pathway. C. Draper (green) is expressed on embryonic glial
membranes and surrounds engulfed neuronal cell corpses (red).

20 • NEUROGLIA
phagocytosis of cell corpses (Zhou et al. 2001). Activation members of the Freeman laboratory for helpful discussions.
of the Draper signaling pathway is essential for glial respon- Figure 2.3D was generously provided by Ozge Tasdemir. The
siveness to axotomy (Fig. 2.6B), and is thought to occur by author apologizes to colleagues in the field whose work he was
clustering of the Draper receptor on engulfment targets unable to cite because of space limitations.
through its extracellular domain. Draper clustering has been
proposed to activate a Src-family signaling cascade composed
of Src42a and Shark, which, together with the PTB-domain REFERENCES
protein dCed-6, promote engulfment (Doherty et al. 2009;
Ziegenfuss et al. 2008). Draper is also important for glial Auld VJ, Fetter RD, Broadie K, Goodman CS. 1995. Gliotactin, a novel
engulfment of pruned axons in the mushroom body (Awasaki transmembrane protein on peripheral glia, is required to form the
et al. 2006; Hoopfer et al. 2006), and embryonic neuronal cell blood-nerve barrier in Drosophila. Cell 81:757–767.
Awasaki T, Huang Y, O’Connor MB, Lee T. 2011. Glia instruct devel-
corpses (e.g., Fig. 2.6C). In the context of cell corpse engulf- opmental neuronal remodeling through TGF-beta signaling. Nat
ment in the embryo Draper requires the co-receptor/bridg- Neurosci 14:821–823.
ing molecule Six-microns-under (Simu) for activity (Kurant Awasaki T, Ito K. 2004. Engulfing action of glial cells is required for
et al. 2008). Recent work has shown that DRG satellite cells programmed axon pruning during Drosophila metamorphosis. Curr
also use MEGF10/Jedi signaling (the mammalian orthologs Biol 14:668–677.
Awasaki T, Lai SL, Ito K, Lee T. 2008. Organization and postembryonic
of Draper/CED-1) to engulf cell corpses during mammalian development of glial cells in the adult central brain of Drosophila.
nervous system development (Wu et al. 2009), arguing that J Neurosci 28:13742–13753.
the glial Draper/MEGF-10/Jedi signaling pathway is a con- Awasaki T, Tatsumi R, Takahashi K, Arai K, Nakanishi Y, Ueda R, et al.
served feature mediating glial phagocytic activity. 2006. Essential role of the apoptotic cell engulfment genes draper
and ced-6 in programmed axon pruning during Drosophila meta-
morphosis. Neuron 50:855–867.
Bacaj T, Tevlin M, Lu Y, Shaham S. 2008. Glia are essential for sensory
5 S U M M A RY A N D P E R S P E C T I VE S organ function in C. elegans. Science 322:744–747.
Bainton RJ, Tsai LT, Schwabe T, DeSalvo M, Gaul U, Heberlein U.
Invertebrate model organisms seem well positioned to help 2005. moody encodes two GPRs that regulate cocaine behav-
move the field of glial biology forward in the coming years iors and blood-brain barrier permeability in Drosophila. Cell
123:145–156.
in major ways. As discussed in this chapter there are a num- Banerjee S, Sousa AD, Bhat MA. 2006. Organization and function of
ber of well-conserved features of glial cells when one com- septate junctions: an evolutionary perspective. Cell Biochem Biophys
pares mammals with Drosophila and even C. elegans. We can 46:65–77.
therefore anticipate studies of glia in these organisms, which Bastiani MJ, Goodman CS. 1986. Guidance of neuronal growth cones in
have only recently been undertaken in a serious way by many the grasshopper embryo. III. Recognition of specific glial pathways.
J Neurosci 6:3542–3551.
groups, will yield fundamental insights into glial biology that Baumgartner S, Littleton JT, Broadie K, Bhat MA, Harbecke R,
are broadly applicable to the more complex nervous system Lengyel JA, et al. 1996. A Drosophila neurexin is required for sep-
of mammals. However, there are notable exceptions impor- tate junction and blood-nerve barrier formation and function. Cell
tant to the field: (1) proper myelin sheaths are not found in 87:1059–1068.
Drosophila nor in C. elegans (although atypical myelin-like Beckervordersandforth RM, Rickert C, Altenhein B, Technau GM.
2008. Subtypes of glial cells in the Drosophila embryonic ven-
structures are indeed found in invertebrate species); (2) tral nerve cord as related to lineage and gene expression. Mech Dev
radial glial cells appear to be a vertebrate specific cell type; 125:542–557.
and (3) the mechanisms of glial cell fate specification have Bergmann A, Tugentman M, Shilo BZ, Steller H. 2002. Regulation of
yet to show significant overlap. Therefore, it seems unlikely cell number by MAPK-dependent control of apoptosis: a mechanism
that these topics will be a focus of studies in invertebrates. for trophic survival signaling. Dev Cell 2:159–170.
Brand AH, Perrimon N. 1993. Targeted gene expression as a means of
Nevertheless, molecular-genetic approaches in invertebrates altering cell fates and generating dominant phenotypes. Development
are beginning to yield exciting insights into how glia develop, 118:401–415.
interact with neurons, regulate synaptic signaling, and mod- Britz FC, Lohr C, Schmidt J, Deitmer JW. 2002. Characterization of
ulate even complex behavioral output. In the future it seems a synaptiform transmission between a neuron and a glial cell in the
likely that even larger questions will be incisively addressed in leech central nervous system. Glia 38:215–227.
Brown ER, Bone Q, Ryan KP, Abbott NJ. 1991. Morphology and electri-
invertebrates including: (1) Are there “glial circuits” impor- cal properties of Schwann cells around the giant axon of the squids
tant for information processing? (2) What is the in vivo rele- Loligo forbesi and Loligo vulgaris. Proc Biol Sci 243:255–262.
vance of glial transmission? (3) What are the precise roles of Bushong EA, Martone ME, Ellisman MH. 2004. Maturation of
astrocyte Ca2+ waves? (4) What role do glia play in the pro- astrocyte morphology and the establishment of astrocyte domains
gression of neurodegenerative disease? during postnatal hippocampal development. Int J Dev Neurosci
22:73–86.
Carlson SD, Juang JL, Hilgers SL, Garment MB. 2000. Blood barriers of
the insect. Annu Rev Entomol 45:151–174.
Coggeshall RE. 1974. Gap junctions between identified glial cells in the
AC K N OW L E D G M E N T S
leech. J Neurobiol 5:463–467.
Colon-Ramos DA, Margeta MA, Shen K. 2007. Glia promote local syn-
The author would like to thank A. Nicole Fox for critical aptogenesis through UNC-6 (netrin) signaling in C. elegans. Science
reading of the manuscript and excellent suggestions, and all 318:103–106.

2. I N VE RT E B R AT E G L I A • 21
Deitmer JW. 1991. Electrogenic sodium-dependent bicarbonate secre- Lai SL, Lee T. 2006. Genetic mosaic with dual binary transcriptional
tion by glial cells of the leech central nervous system. J Gen Physiol systems in Drosophila. Nat Neurosci 9:703–709.
98:637–655. Lee T, Luo L. 2001. Mosaic analysis with a repressible cell marker
Deitmer JW, Rose CR. 1996. pH regulation and proton signalling by (MARCM) for Drosophila neural development. Trends Neurosci
glial cells. Prog Neurobiol 48:73–103. 24:251–254.
Deitmer JW, Rose CR, Munsch T, Schmidt J, Nett W, Schneider HP, Leiserson WM, Forbush B, Keshishian H. 2011. Drosophila glia use a
et al. 1999. Leech giant glial cell: functional role in a simple nervous conserved cotransporter mechanism to regulate extracellular volume.
system. Glia 28:175–182. Glia 59:320–332.
Deitmer JW, Schlue WR. 1987. The regulation of intracellular pH by Leiserson WM, Harkins EW, Keshishian H. 2000. Fray, a Drosophila
identified glial cells and neurones in the central nervous system of the serine/threonine kinase homologous to mammalian PASK, is
leech. J Physiol 388:261–283. required for axonal ensheathment. Neuron 28:793–806.
Deitmer JW, Schlue WR. 1989. Functional role of the sodium-bicarbonate Lieberman EM, Abbott NJ, Hassan S. 1989. Evidence that glutamate
cotransport of glial cells in the leech central nervous system. Acta mediates axon-to-Schwann cell signaling in the squid. Glia 2:94–102.
Physiol Scand Suppl 582:31. Lohr C, Deitmer JW. 1997. Intracellular Ca 2+ release mediated by
Deitmer JW, Schneider HP. 1995. Voltage-dependent clamp of intracel- metabotropic glutamate receptor activation in the leech giant glial
lular pH of identified leech glial cells. J Physiol 485(Pt 1):157–166. cell. J Exp Biol 200:2565–2573.
Deitmer JW, Verkhratsky AJ, Lohr C. 1998. Calcium signalling in glial Lohr C, Deitmer JW. 1999. Dendritic calcium transients in the leech
cells. Cell Calcium 24:405–416. giant glial cell in situ. Glia 26:109–118.
Doherty J, Logan MA, Tasdemir OE, Freeman MR. 2009. Ensheathing MacDonald JM, Beach MG, Porpiglia E, Sheehan AE, Watts RJ, Freeman
glia function as phagocytes in the adult Drosophila brain. J Neurosci MR. 2006. The Drosophila cell corpse engulfment receptor Draper
29:4768–4781. mediates glial clearance of severed axons. Neuron 50:869–881.
Evans PD, Reale V, Villegas J. 1985. The role of cyclic nucleotides in mod- Muller M, Henrich A, Klockenhoff J, Dierkes PW, Schlue WR. 2000.
ulation of the membrane potential of the Schwann cell of squid giant Effects of ATP and derivatives on neuropile glial cells of the leech
nerve fibre. J Physiol 363:151–167. central nervous system. Glia 29:191–201.
Evequoz-Mercier V, Tsacopoulos M. 1991. The light-induced increase Nett W, Deitmer JW. 1998. Intracellular Ca 2+ regulation by the leech
of carbohydrate metabolism in glial cells of the honeybee retina giant glial cell. J Physiol 507(Pt 1):147–162.
is not mediated by K+ movement nor by cAMP. J Gen Physiol Oberheim NA, Takano T, Han X, He W, Lin JH, Wang F, et al. 2009.
98:497–515. Uniquely hominid features of adult human astrocytes. J Neurosci
Freeman MR, Delrow J, Kim J, Johnson E, Doe CQ. 2003. Unwrapping 29:3276–3287.
glial biology: Gcm target genes regulating glial development, diversi- Oikonomou G, Perens EA, Lu Y, Watanabe S, Jorgensen EM, Shaham S.
fication, and function. Neuron 38:567–580. 2011. Opposing activities of LIT-1/NLK and DAF-6/patched-related
Fuentes-Medel Y, Logan MA, Ashley J, Ataman B, Budnik V, Freeman direct sensory compartment morphogenesis in C. elegans. PLoS Biol
MR. 2009. Glia and muscle sculpt neuromuscular arbors by engulf- 9:e1001121.
ing destabilized synaptic boutons and shed presynaptic debris. PLoS Palgi M, Lindstrom R, Peranen J, Piepponen TP, Saarma M, Heino TI.
Biol 7:e1000184. 2009. Evidence that DmMANF is an invertebrate neurotrophic fac-
Garratt AN, Britsch S, Birchmeier C. 2000. Neuregulin, a factor with tor supporting dopaminergic neurons. Proc Natl Acad Sci U S A
many functions in the life of a schwann cell. Bioessays 22:987–996. 106:2429–2434.
Gho M, Bellaiche Y, Schweisguth F. 1999. Revisiting the Drosophila Pereanu W, Shy D, Hartenstein V. 2005. Morphogenesis and prolifera-
microchaete lineage: a novel intrinsically asymmetric cell division tion of the larval brain glia in Drosophila. Dev Biol 283:191–203.
generates a glial cell. Development 126:3573–3584. Poeck B, Fischer S, Gunning D, Zipursky SL, Salecker I. 2001. Glial cells
Gommerat I, Gola M. 1994. Satellite glial cell responses to neuronal firing mediate target layer selection of retinal axons in the developing visual
in the nervous system of Helix pomatia. J Membr Biol 138:209–219. system of Drosophila. Neuron 29:99–113.
Han C, Jan LY, Jan YN. 2011. Enhancer-driven membrane markers for Potter CJ, Tasic B, Russler EV, Liang L, Luo L. 2010. The Q system: a
analysis of nonautonomous mechanisms reveal neuron-glia interac- repressible binary system for transgene expression, lineage tracing,
tions in Drosophila. Proc Natl Acad Sci U S A 108:9673–9678. and mosaic analysis. Cell 141:536–548.
Hoopfer ED, McLaughlin T, Watts RJ, Schuldiner O, O’Leary DD, Luo Reichenbach A, Pannicke T. 2008. Neuroscience. A new glance at glia.
L. 2006. Wlds protection distinguishes axon degeneration follow- Science 322:693–694.
ing injury from naturally occurring developmental pruning. Neuron Rival T, Soustelle L, Strambi C, Besson MT, Iche M, Birman S. 2004.
50:883–895. Decreasing glutamate buffering capacity triggers oxidative stress and
Ito K, Urban J, Technau GM. 1995. Distribution, classification and neuropil degeneration in the Drosophila brain. Curr Biol 14:599–605.
development of Drosophila glial cells in the late embryonic and early Rogulja-Ortmann A, Luer K, Seibert J, Rickert C, Technau GM. 2007.
larval ventral nerve cord. Roux’s Arch Dev Biol 204:284–307. Programmed cell death in the embryonic central nervous system of
Jacobs JR. 2000. The midline glia of Drosophila: a molecular genetic Drosophila melanogaster. Development 134:105–116.
model for the developmental functions of glia. Prog Neurobiol Rossler W, Oland LA, Higgins MR, Hildebrand JG, Tolbert LP. 1999.
62:475–508. Development of a glia-rich axon-sorting zone in the olfactory path-
Kamath RS, Fraser AG, Dong Y, Poulin G, Durbin R, Gotta M, et al. way of the moth Manduca sexta. J Neurosci 19:9865–9877.
2003. Systematic functional analysis of the Caenorhabditis elegans Schmidt J, Deitmer JW. 1999. Peptide-mediated glial responses to leydig
genome using RNAi. Nature 421:231–237. neuron activity in the leech central nervous system. Eur J Neurosci
Keller LC, Cheng L, Locke CJ, Muller M, Fetter RD, Davis GW. 2011. 11:3125–3133.
Glial-derived prodegenerative signaling in the Drosophila neuromus- Schwabe T, Bainton RJ, Fetter RD, Heberlein U, Gaul U. 2005. GPR
cular system. Neuron 72:760–775. signaling is required for blood-brain barrier formation in drosophila.
Kuffler SW, Nicholls JG. 1966. The physiology of neuroglial cells. Ergeb Cell 123:133–144.
Physiol 57:1–90. Sepp KJ, Auld VJ. 2003. Reciprocal interactions between neurons and
Kuffler SW, Potter DD. 1964. Glia in the leech central nervous system: glia are required for Drosophila peripheral nervous system develop-
physiological properties and neuron-glia relationship. J Neurophysiol ment. J Neurosci 23:8221–8230.
27:290–320. Shaham S. 2005. Glia-neuron interactions in nervous system function
Kurant E, Axelrod S, Leaman D, Gaul U. 2008. Six-microns-under acts and development. Curr Top Dev Biol 69:39–66.
upstream of Draper in the glial phagocytosis of apoptotic neurons. Shaham S. 2006. Glia-neuron interactions in the nervous system of
Cell 133:498–509. Caenorhabditis elegans. Curr Opin Neurobiol 16:522–528.

22 • NEUROGLIA
Sonnenfeld MJ, Jacobs JR. 1995. Macrophages and glia participate in the Walker RG, Willingham AT, Zuker CS. 2000. A Drosophila mecha-
removal of apoptotic neurons from the Drosophila embryonic nerv- nosensory transduction channel. Science 287:2229–2234.
ous system. J Comp Neurol 359:644–652. Wang Y, Apicella A Jr, Lee SK, Ezcurra M, Slone RD, Goldmit M, et al.
Stork T, Bernardos R, Freeman MR. 2012. Analysis of glial cell develop- 2008. A glial DEG/ENaC channel functions with neuronal channel
ment and function in Drosophila. Cold Spring Harb Protoc 1:1–17. DEG-1 to mediate specific sensory functions in C. elegans. EMBO J
Stork T, Engelen D, Krudewig A, Silies M, Bainton RJ, Klambt C. 2008. 27:2388–2399.
Organization and function of the blood-brain barrier in Drosophila. Wang Y, D’Urso G, Bianchi L. 2012. Knockout of glial channel
J Neurosci 28:587–597. ACD-1 exacerbates sensory deficits in a C. elegans mutant by
Szatkowski MS, Schlue WR. 1994. Chloride-dependent pH regulation in regulating calcium levels of sensory neurons. J Neurophysiol
connective glial cells of the leech nervous system. Brain Res 665:1–4. 107:148–158.
Th imgan MS, Berg JS, Stuart AE. 2006. Comparative sequence analysis Watts RJ, Schuldiner O, Perrino J, Larsen C, Luo L. 2004. Glia engulf
and tissue localization of members of the SLC6 family of transporters degenerating axons during developmental axon pruning. Curr Biol
in adult Drosophila melanogaster. J Exp Biol 209:3383–3404. 14:678–684.
Tolbert LP, Oland LA. 1990. Glial cells form boundaries for developing Wu HH, Bellmunt E, Scheib JL, Venegas V, Burkert C, Reichardt
insect olfactory glomeruli. Exp Neurol 109:19–28. LF, et al. 2009. Glial precursors clear sensory neuron corpses dur-
Tsacopoulos M, Evequoz-Mercier V, Perrottet P, Buchner E. 1988. ing development via Jedi-1, an engulfment receptor. Nat Neurosci
Honeybee retinal glial cells transform glucose and supply the 12:1534–1541.
neurons with metabolic substrate. Proc Natl Acad Sci U S A Yoshimura S, Murray JI, Lu Y, Waterston RH, Shaham S. 2008. mls-2
85:8727–8731. and vab-3 Control glia development, hlh-17/Olig expression and
Tsacopoulos M, Veuthey AL, Saravelos SG, Perrottet P, Tsoupras G. glia-dependent neurite extension in C. elegans. Development
1994. Glial cells transform glucose to alanine, which fuels the neu- 135:2263–2275.
rons in the honeybee retina. J Neurosci 14:1339–1351. Zhou Z, Hartwieg E, Horvitz HR. 2001. CED-1 is a transmembrane
Villegas J. 1972. Axon-Schwann cell interaction in the squid nerve fibre. receptor that mediates cell corpse engulfment in C. elegans. Cell
J Physiol 225:275–296. 104:43–56.
Villegas J. 1974. Effects of acetylcholine and carbamylcholine on the Zhu B, Pennack JA, McQuilton P, Forero MG, Mizuguchi K, Sutcliffe B,
axon and Schwann cell electrical potentials in the squid nerve fibre. et al. 2008. Drosophila neurotrophins reveal a common mechanism
J Physiol 242:647–659. for nervous system formation. PLoS Biol 6:e284.
Villegas J, Evans PD, Reale V. 1987. Modulation of the membrane poten- Ziegenfuss JS, Biswas R, Avery MA, Hong K, Sheehan AE, Yeung YG,
tial of the Schwann cell of the squid giant nerve fiber. J Physiol (Paris) et al. 2008. Draper-dependent glial phagocytic activity is mediated by
82:322–326. Src and Syk family kinase signalling. Nature 453:935–939.

2. I N VE RT E B R AT E G L I A • 23
3.
NONMAMMALIAN VERTEBRATE GLIA
Bruce Appel

A B B R E VI AT I O N S 2 C H O R DAT E P H Y L O G E N Y

3V third ventricle Chordates, which include urochordates, cephalochordates,


aATEN anterior apical trunk epidermal neuron and vertebrates, are characterized by a dorsal neural tube.
BBB blood-brain barrier Different members of the chordate phylum have neural tubes
ca capillaries that vary greatly in size and complexity, resulting from differ-
CC coronet cell ences in the numbers, types, and associations of neural cells.
CE ciliated ependymal cell Chordate evolution is also typified by gains in body size,
CNS central nervous system requiring a peripheral nervous system capable of transmitting
DCEN dorsocaudal epidermal neurons motor and sensory information rapidly over long distances.
ep ependymal glia Glial cells vary greatly in number, morphology, and function
ES endodermal strand among chordates, indicating that glial cell evolution goes hand
fp floor plate in hand with the evolution of increasingly large and complex
GFAP glial fibrillary acid protein nervous systems.
GI group I photoreceptor Most current phylogenies, drawing on molecular evi-
GIII group III photoreceptor dence, place urochordates, represented by ascidians, as a
GP gut primordium sister group to the vertebrates, and cephalochordates, rep-
GS glutamine synthetase resented by amphioxus, as a more distantly related, basal
LC lens cell group in the chordate lineage (Bourlat et al. 2006) (Fig.
M meningeal surface 3.1). However, extant ascidians have highly derived mor-
MC mesenchyme cell phologies and life histories and their nervous systems have
mg midline glia been reduced in size, cell type, and functional complexity
MS muscle cell relative to other chordates. Therefore, among extant mod-
NC notochord els examined to date, amphioxus might provide the best
NT neural tube insight to the starting point of evolutionary processes that
pATEN posterior apical trunk epidermal neuron produced the vertebrate nervous system (Lacalli 2008).
PC pigment cup Among the vertebrate groups, increasing size, metabolic
PNS peripheral nervous system demands of the brain, and vascularization of the central
rp roof plate nervous system (CNS) appear to be accompanied by glial
VCEN ventrocaudal epidermal neurons cell diversification.
VT tectal ventricle

3 E P E N DY M A L G L I A , R A D I A L G L I A ,
AND ASTROCY TES
1 INTRODUCTION
Among the most striking features of nervous system evolu-
Glial cells are remarkably varied in morphology, distribu- tion among chordates are differences in the number and
tion, and function. This variety reflects evolutionary adapta- diversity of ependymal glia, radial glia, and astrocytes. In
tion, and it has contributed to the evolution of increasingly fact, numerous subtypes of these glia have been described
complex nervous systems and behaviors (see chapter 1). and often given different names such as ependymocytes,
Although glial cells are not preserved in fossilized specimens, ependymoglia, tanycytes, and radial astrocytes (Cuoghi
ideas about how the evolution of glia have contributed to the and Mola 2009). Whether these apparent subtypes reflect
evolution of nervous systems can be gathered from investi- functionally distinct types of glia is not clear. Therefore,
gations of glial cell form, gene expression, and intercellular for simplicity, in this chapter cells lining the ventricles are
relationships in diverse species representing the major chor- ependymal glia, bipolar cells that extend long processes from
date groups. the ventricle to the pial surface of the nerve cord are radial

24
ancestral
chordate

cephalochordates (amphioxus), radial glia

urochordates (tunicates), ependymal glia


hagfish, astrocytes

lamprey, radial glia

placoderms (extinct jawed fish)

sharks, radial glia


skates, rays, astrocytes
bony fish, radial glia
myelin
amphibians, radial glia

reptiles, radial glia


birds, astrocytes

mammals, astrocytes

Figure 3.1 Chordate Phylogeny and Glial Cell Evolution. The tree represents a simplified phylogeny highlighting chordates discussed in this chapter
and their glial cell composition. For each group the predominant, but not necessarily exclusive, cell type among ependymal glia, radial glia, and
astrocytes is indicated. Astrocytes appear to have arisen multiple times in chordate evolution. Compact myelin is a common feature of all jawed
vertebrates.

glia and nonependymal, multiprocess cells that express glial cilia into the lumen (Katz 1983; Konno et al. 2010). The
fibrillary acid protein (GFAP) are astrocytes. Many, but not basal surface of these cells form the exterior of the neural
all, ependymal glia and radial glia also express GFAP. In many tube and are surrounded by basal lamina (Fig. 3.2A, B).
organisms, the cell bodies of radial glia line the ventricles. In Therefore, ependymal glia comprise most of the neural tube,
these cases the terms ependymal glia and radial glia are syn- with a few neurons embedded among the ependymal glia.
onymous. Characteristics of ependymal glia and astrocytes No other types of glial cells have been described for ascid-
are described in greater detail in chapter 4 and radial glia are ians. During metamorphosis, the ascidian nervous system is
the subject of chapter 5. dramatically remodeled. Fate mapping experiments showed
Ependymal glia, cells that line the ventricular system of that larval ependymal glia give rise to ependymal glia and
the nerve cord, are evident throughout the chordate group. neurons of the adult nervous system, indicating that ascid-
Thus, ependymal cells might represent a basal glial cell type ian ependymal glia have stem cell characteristics (Horie
from which other glia arose during evolution. The ventricu- et al. 2011).
lar lumens of neural tubes in tadpoles of the ascidian Ciona Anatomical and molecular investigations of the amphioxus
intestinalis are lined by cuboidal ependymal glia that extend larval and adult nervous system have revealed clear homologies

A B C rp
pATEN
DCEN
eg eg
CE NT
PC eg eg
GI
CC GIII LC
MS NC
GP mg fp
MC ES
EP

VCEN

Figure 3.2 Glia of Tunicates and Amphioxus. Transverse section views at the level of the brain (A) and nerve cord (B) of tadpole larvae of the ascidian
Ciona intenstinalis. Cells comprising the neural parenchyma are green. aATEN, anterior apical trunk epidermal neurons; CC, coronet cell; CE,
ciliated ependymal cell; DCEN, dorsocaudal epidermal neurons; ES, endodermal strand; GI, group I photoreceptor; GIII, group III photoreceptor;
GP, gut primordium; LC, lens cell; MC, mesenchyme cell; MS, muscle cell; NC, notochord; NT, neural tube; pATEN, posterior apical trunk
epidermal neuron; PC, pigment cup; VCEN, ventrocaudal epidermal neurons. C. Transverse section through the nerve cord of larval amphioxus. eg,
ependymal glia; fp, floor plate; mg, midline glia; rp, roof plate. (A,B) From Konno, et al. 2010. (C) Modified from Lacalli TC and Kelly SJ. 2002.
with permission.

3. N O N M A M M A L I A N VE RT E B R AT E G L I A • 25
with vertebrate nervous systems (Lacalli 2008). Unlike sessile hagfish CNS (Wicht et al. 1994). However, GFAP+ radial
adult ascidians, adult amphioxus remain motile throughout glia do not occupy the entire CNS but are found only in por-
life. Therefore, compared with ascidians, the amphioxus nerv- tions of the spinal cord. By contrast, numerous, multiprocess
ous system is quite complex (Fig. 3.2C). However, relative to GFAP+ cells that do not contact the ventricle are distributed
most vertebrates, amphioxus larvae and adults are small and throughout the CNS (Fig. 3.3A). These cells have processes
the CNS is not vascularized, which are important differences that are fairly short, irregular, and with few branches, and
relevant to glial cell functions. they make contacts on both blood vessels and neurons.
Amphioxus have a prominent ventricular system Antibody to glutamine synthetase (GS), a commonly used
(Brocklehurst 1979), which in larvae is surrounded by marker of astrocytes, also labels nonependymal cells with
ependymal glia (Wicht and Lacalli 2005). The ependymal numerous fine processes that form meshworks around blood
glia extend processes to the limiting membrane of the nerve vessels and neurons (Wicht et al 1994). Therefore, the glial
cord and therefore have radial glia character (Bone 1960; cell architecture of hagfish represents a departure from
Lacalli and Kelly 2002). Small glial cells, called Schneider’s that of amphioxus, with a transition from a predominantly
glia, line the ventricle near the dorsal roof of the nerve cord ependymal, radial glial organization to a predominantly non-
(Bone 1960). These cells have short processes, and therefore ependymal, astrocyte-like organization. Very little is known
could be considered as astrocyte-like, but the processes do about glia in lamprey. However, antibodies to GFAP and
not appear to terminate within the limiting membrane of keratin-like proteins detect numerous radial fibers extending
the cord. However, several types of neurons lie within close from the ventricles to the limiting membrane of the nerve
proximity to Schneider’s glia, raising the possibility that cord (Merrick et al. 1995; Wasowicz et al. 1994), indicat-
these glial cells provide some sort of support to neurons. A ing that in contrast to hagfish, lamprey has a predominantly
distinct floor plate and roof plate is evident in amphioxus radial glial organization.
larvae, as well as a small number of other non-neuronal cells Elasmobranchs, a subclass of cartilaginous fish repre-
described as midline glia and axial glia, which extend long sented by sharks, skates, and rays, first appear in the fossil
membrane processes along the ventral nerve cord (Lacalli record about 400 million years ago (see Fig. 3.1). Notably,
2000; Lacalli and Kelly 2002). In the periphery, groups of the BBB of elasmobranchs is not endothelial, as in other ver-
very small cells called Müller’s glia are clustered with the dor- tebrates, but composed of glia cells (Bundgaard and Abbott,
sal nerves (Bone 1960). Neither the origin nor function of 2008; Bundgaard and Cserr 1981). GFAP labeling of dogfish
Müller’s glia are known. shark revealed that radial glia are prominently distributed
Agnathans, or jawless fish, represented by lampreys and throughout the CNS with little or no evidence of stellate
hagfish, comprise the most basal group of vertebrates (see astrocyte-like cells (Kálmán and Gould 2001; Wasowicz
Fig. 3.1), having diverged from the lineage that gave rise to et al. 1999) (Fig. 3.3B). Radial glia make en passant contacts
jawed vertebrates about 600 million years ago. Unlike ascid- onto blood vessels and radial glia endfeet make up the menin-
ians and amphioxus, the hagfish CNS is vascularized (Cecon geal glia limitans of the nerve cord (Kálmán and Gould
et al. 2002). Hagfish and lampreys have a blood-brain bar- 2001). By contrast, GFAP+ radial glia are limited to portions
rier with similar characteristics to mammals, and the BBB of the CNS of skates and rays, with numerous astrocyte-like
is formed by endothelial cells (Bundgaard 1982; Bundgaard cells evident in some parts of the nervous system (Fig. 3.3C).
and Cserr 1981). GFAP+ cells are prominent throughout the These cells appear to form a meshwork around blood vessels,

A B C

Figure 3.3 A. Drawing of GFAP+ astrocyte-like cells and processes in hagfish. In the center a multiprocess cells (2) makes contacts on capillaries (ca).
B. Section obtained from optic tectum of dogfish. Glial fibrillary acid protein labeling reveals radial fibers spanning the distance between the tectal
ventricle (VT) and the meningeal surface (M). C. Cross-section through the diencephalon of skate showing distribution of GFAP. In the center, radial
fibers stretch from the third ventricle (3V) to the basal surface. Only nonependymal astrocyte-like cells are evident laterally. (A) From Wicht et al.
1994. (B,C) from Kálmán M and Gould RM. 2001, with permission.

26 • NEUROGLIA
consistent with a glial BBB, and they form the meningeal glia 2006; Marcus and Easter 1995). Additionally, nonependy-
limitans (Kálmán and Gould 2001; Wasowicz et al. 1999). mal star-shaped cells that make contacts on blood vessels and
Therefore, among the elasmobranchs there is an apparent the subpial surface of the nerve cord are apparent in adult
transition from nervous systems having predominantly radial zebrafish (Kawai et al. 2001). Many radial glia in zebrafish
glia (sharks) to nervous systems having mixtures of radial glia also express GS, as well as aquaporin-4, which in mammals
and astrocytes (skates and rays). Kálmán and Gould (2001) functions as the major astrocyte water channel (Grupp et al.
noted that that the difference in radial glia and astrocyte 2010). Unlike mammals, in which aquaporin-4 channels are
composition between sharks and skates is similar to the dif- mainly localized in astrocyte processes around blood vessels
ferences between reptiles, which have predominantly radial and at the subpial surface, in zebrafish aquaporin-4 is distrib-
glia, and mammals and birds, which have both radial glia and uted along the length of radial glial fibers and not concen-
astrocytes. This indicates that radial glia in different verte- trated in glial endfeet. Therefore, zebrafish radial glia share
brate lineages have undergone independent and parallel evo- some characteristics with mammalian astrocytes, indicating
lution toward astrocyte characteristics. that radial glia provide some basic astrocyte functions in
The glia of several species of bony fish have been surveyed, nonmammalian vertebrates.
and although distinctions of possible subclasses of cell type Amphibians are similar to the elasmobranchs, in that
can be made (Cuoghi and Mola 2009), bony fish clearly have species with primarily radial glia or mixtures of radial glia
predominantly radial glia and relatively few nonependymal, and nonependymal astrocyte-like cells can be identified. For
stellate or radial astrocyte-like glia (Fig. 3.4A). The adult example, GFAP antibody labeling reveals widespread radial
barbel has numerous radial glia throughout the CNS, with glia that project endfeet onto blood vessels and the glia limi-
a few GFAP+ cells, described as radial astrocytes, located tans in salamander but does not detect nonependymal glia
just under the pial surface of the nerve cord with their pro- (Naujoks-Manteuffel and Roth 1989). By contrast, in adult
cesses contributing to the glia limitans (Bodega et al. 1993). toad GFAP expression marks both radial glia and nonependy-
Radial glia in barbel seem to be heterogeneous with respect mal radial astrocyte-like cells, both of which extend processes
to intermediate filament expression. Although most radial on to blood vessels and the subpial glia limitans (Bodega et al.
glia express GFAP some do not and a small subset of radial 1990) (see Fig. 3.4).
glia is labeled by antibody to vimentin (Bodega et al. 1993). The glia of several reptile species have been examined,
Similarly, Iberian barb has numerous GFAP+ radial glia, a few mostly by using GFAP immunohistochemistry. In turtle,
vimentin+ radial glia and some GFAP+, nonependymal radial GFAP antibody labels mostly radial glia and only a relatively
astrocytes (Rubio et al. 1992). Heterogeneity of radial glia is small number of radial astrocytes in the spinal cord (Lazzari
also revealed by NADPH-diaphorase histochemistry, which and Franceschini 2006). Geckos and lizards have numerous
in sunfish labels a subset of radial glia described as tanycytes GFAP+ radial glia throughout the CNS, fewer radial astro-
as well as nonependymal, multiprocess cells that contact cytes in the spinal cord, and rare stellate astrocyte-like cells in
blood vessels, neurons, and the pial surface (Ma 1993). Carp the brain (Lazzari and Franceschini 2001, 2005a,b). In addi-
have numerous GFAP+ radial glia, some of which appear tion to radial glia, stellate GFAP+ astrocytes occupy the snake
to extend long processes along blood vessels, and very few brain (Onteniente et al. 1983). In crocodile, both GFAP+
astrocyte-like cells (Kálmán 1998). Both GFAP immunohis- radial glia and widely distributed astrocyte-like cells are evi-
tochemistry and transgenic reporter gene expression reveal dent, with radial glia much more numerous than astrocytes
numerous radial glia in zebrafish (Bernardos and Raymond (Fig. 3.4B) (Kálmán and Pritz 2001).

A B C

Figure 3.4 A. Drawing of GFAP+ structure in the forebrain of carp. Only radial glia are evident. B. Drawing of cross-section through telencephalon
of crocodile, showing GFAP+ features. Radial glia are predominant, with some irregular fibers evident in lateral regions (4b). C. GFAP labeling of
chicken tectum. Radial fibers at the subventricular zone are evident (small arrows) as well as prominent astrocytes (curved arrows). (A) From Kálmán
1998. (B) from Kálmán and Prinz 2001. (C) from Kálmán et al. 1998, with permission.

3. N O N M A M M A L I A N VE RT E B R AT E G L I A • 27
With birds, the relative contributions of radial glia and astrocyte form and function among mammals (Oberheim
astrocytes to the CNS become more like mammals than other et al. 2012).
nonmammalian vertebrates (Fig. 3.4C). Numerous GFAP+
and vimentin+ radial glia are evident in many, but not all
regions of the brain of chicken, including adults, but non- 5 G L I A , A D U LT N E U R O G E N E S I S ,
ependymal astrocytes, which extend processes on to blood A N D I N J U RY R E S P O N S E
vessels, neurons and the glia limitans of the nerve cord, are
much more pronounced in number than other nonmam- One characteristic that distinguishes many nonmammalian
malian vertebrates (Alvarez-Buylla et al. 1987; Kálmán et al. vertebrates from mammals is widespread neurogenesis in the
1993, 1998). adult brain (Kaslin et al. 2008). For example, bony fish, which
have a predominantly radial glia architecture, maintain high
levels of neural cell proliferation and neurogenesis in adult-
4 GLIAL PHYLOGENY AND hood. Cell proliferation is widespread across the brains of
I M P L I C AT I O N S F O R A S T R O C Y T E adult teleost fish, centered in about 16 distinct proliferation
E VO LU T I O N zones (Adolf et al. 2006; Grandel et al. 2006; Zupanc 2001).
In the adult zebrafish brain, approximately 6000 cells are born
Surveys of glia raise the possibility that astrocytes arose within any 30-minute period, representing about 0.06% of
independently from radial glia in several vertebrate lineages total cells, a substantially higher proportion than in mammals
through evolutionary history. In particular, the agnathan lam- (Hinsch and Zupanc 2007). Numerous studies show that
preys and hagfish, which may have arisen from the vertebrate radial glia are the sources of new neurons that can persist for
lineage independently (see Fig. 3.1), have distinct glial archi- long periods (Adolf et al. 2006; Grandel et al. 2006; Hinsch
tectures, with lamprey having primarily radial glia and hag- and Zupanc 2007; Zupanc et al. 2005). In adult fish, both the
fish having relatively few radial glia but many astrocyte-like numbers and volume of individual muscle fibers increase over
cells. Among elasmobranchs, sharks have primarily radial glia, time (Zimmerman and Lowery 1999), whereas in mammals
whereas skates and rays have both radial glia and astrocyte-like postembryonic growth results from an increase in size but not
cells. Bony fish, amphibians, and reptiles have mostly radial in the number of muscle fibers (Rowe and Goldspink 1969).
glia, with relatively small numbers of astrocytes evident in Additionally, adult fish increase the number of sensory recep-
some species. Birds and mammals are similar in having rela- tor cells and organs with age ( Johns and Easter 1977; Nuñez
tively few radial glia and large numbers of nonependymal, et al. 2009). The continuous addition of peripheral motor
stellate astrocytes. However, birds are more closely related and sensory elements could promote continuous addition of
to alligators, which have primarily radial glia, than to mam- central neurons that process the associated motor and sensory
mals, indicating that the astrocyte-predominant patterns of functions (Zupanc 2001).
birds and mammals evolved independently. An important Close examination of cycling characteristics and molec-
caveat to the multiple, independent origin hypothesis is that ular marker expression reveals heterogeneity among neural
investigations of radial glia and astrocyte in nonmammalian precursors in adult zebrafish. Some precursors do not express
vertebrates have been mostly limited to the use of GFAP as a markers common to radial glia, but instead seem to retain
marker, which might fail to uncover primitive astrocyte-like characteristics of embryonic neuroepithelial precursors
cells. Application of a broader set of histochemical stains (Ganz et al. 2010; Ito et al. 2010; Kaslin et al. 2009; Marz et
known to reveal glial cell morphologies might help to clarify al. 2010). Whether adult glial and nonglial neural precursors
the question of astrocyte origins. represent distinct stages in progression of a precursor cell lin-
If astrocytes arose multiple times from radial glia, what eage, neural precursors can alternate between glial and non-
might have driven the transition? One possible hint is that the glial states, and glial and nonglial precursors have different
transition of radial glia to astrocytes parallels the transition of roles in homeostasis and response to disease and injury are
thin- to thick-walled brains, which reflects increasing brain areas of investigation that might provide important insights
complexity (Kálmán 2002). The formation of thick-walled into the biology of mammalian neural stem cells.
brains might place metabolic demands on glial cells that can- Another distinguishing feature of many nonmamma-
not be supported by very long radial glia. In particular, it has lian vertebrates is a robust ability to regenerate neural tis-
been speculated that extremely elongated radial glia cannot sue following injury. In adult zebrafish, a stab wound to the
maintain normal potassium equilibrium and, consequently, brain or spinal cord transection prompts a large increase in
transform into stellate cells (Reichenbach 1989; Reichenbach cell division by radial glia and formation of new neurons
et al. 1987). However, the correlation between astrocyte appear- (Baumgart et al. 2011; März et al. 2011; Reimer et al. 2008;
ance and brain wall thickness or complexity is not a perfect one Zupanc 2001). Transgenic fate mapping experiments show
among vertebrates (Kálmán and Gould 2001). Furthermore, that radial glia are the source of new neurons following
astrocyte-like cells are evident in the very small and relatively injury (Kroehne et al. 2011). Furthermore, although wound-
simple nervous systems of fruit flies (see chapter 2). Perhaps ing induces features of reactive gliosis, glial scarring is not
the repeated transition of radial glia to astrocytes facilitated evident as in mammals (Baumgart et al. 2011; Kroehne et al.
increasingly complex neural functions, for example, by modu- 2011; März et al. 2011), which might also contribute to the
lating synaptic activity, as has been proposed for evolution of regenerative capacity of fish.

28 • NEUROGLIA
6 M Y E L I N AT I N G G L I A cells and oligodendrocytes must have appeared at the same
time, in placoderms (Zalc 2006). An alternative possibility is
Glial cells wrap axons in many invertebrates (see chapter 2), that one population of glia initially myelinated both central
and some invertebrates such as copepods and earthworms and peripheral axons and the second population arose later
have axons ensheathed by myelin-like membrane (Davis et al. to enable evolution of increasingly large animals with increas-
1999; Roots 2008). Although invertebrate myelin enhances ingly complex motor activities. Consistent with this possi-
nerve conduction velocity (Lenz et al. 2000), similar to verte- bility, in the absence of Schwann cells in zebrafish and mice,
brate myelin, invertebrate and vertebrate myelin have distinct oligodendrocyte progenitor cells migrate from the spinal cord
characteristics, indicating that they evolved independently through motor axon exit points to wrap and myelinate periph-
(Roots 2008). eral motor axons (Coulpier et al. 2010; Kucenas et al. 2009).
Among chordates, CNS axons of ascidians and amphi- Similarly, Schwann cell apparently migrate into the nerve cord
oxus lack compact myelin and, in fact, appear not to be and myelinate central axons in multiple sclerosis patients and
ensheathed by glia (Katz 1983; Wicht and Lacalli 2005). demyelinated rodents (Duncan and Hoffman 1997; Itoyama
Axons are similarly unmyelinated in lamprey and hagfish, et al. 1983), although recent fate mapping experiments show
although in hagfish many central and peripheral axons are that Schwann cells arise from CNS progenitors following
ensheathed by a single turn of glial cell membrane and bun- demyelination (Zawadzka et al. 2010). Oligodendrocytes are
dled axons are often separated by glial processes (Bullock et produced by neural precursors that also give rise to motor
al. 1984). The elasmobranchs, bony fish, amphibians, reptiles, neurons and oligodendrocytes myelinate the proximal por-
and birds have compact myelin with characteristics similar tions of motor axons before they exit the nerve cord. The
to that of mammals (Kitagawa et al. 1993; Schweigreiter selective advantage of myelin likely first had bearing on motor
et al. 2006; Waehneldt et al. 1986), indicating that myelin functions, by permitting rapid escape behaviors. Therefore, it
likely evolved with the placoderms, jawed fish that appeared seems plausible that oligodendrocytes evolved first and that
about 400 million years ago. Indeed, inference of peripheral their initial function was to myelinate motor axons along their
nerve length from examination of fossilized skulls suggests entire length. Subsequently, the ability of neural crest-derived
that nerves of placoderms but not of contemporaneous jaw- glia to myelinate axons might have provided additional selec-
less ostracoderm fish must have been myelinated, raising the tive advantage, thereby leading to their replacement of oligo-
possibility that jaws and myelin evolved coordinately (Zalc dendrocytes as the peripheral myelinating cells.
et al. 2008).
All extant vertebrates have two populations of myelinat-
ing glial cells: oligodendrocytes for the CNS and Schwann 7 S U M M A RY A N D P E R S P E C T I VE S
cells for the peripheral nervous system (PNS). This raises
fascinating questions about whether myelinating glia arose Two features of glia in nonmammalian vertebrates stand
once in evolution and then diverged into oligodendrocyte out as being markedly different from mammalian glia and
and Schwann cell populations or two distinct types of glial therefore potentially instructive for our understanding of
cells independently gained the ability to myelinate axons, glial cell evolution and function. First, a predominantly
and if so, when they evolved relative to each other. Schwann radial glial organization is widespread throughout extant
cells arise from the neural crest, a multipotent precursor chordates with evidence of multiple and independent tran-
population that forms at the boundary between neural and sitions to astrocytes, which are smaller and less extensively
non-neural ectoderm (Knecht and Bronner-Fraser 2002). branched than astrocytes in mammals, particularly humans.
The evolution of neural crest appears to have accompanied Investigation of astrocyte properties in nonmammalian spe-
and enabled the transition from filter feeding to active pre- cies might provide insights to basic astrocyte roles in mam-
dation at early stages of vertebrate evolution (Northcutt mals and a better understanding of how further elaboration
and Gans 1983) consistent with the idea that myelination of of astrocyte functions contribute to human brain function.
peripheral nerves first occurred among the placoderms. By Second, many radial glia have stem cell characteristics in
contrast, oligodendrocytes have a different embryonic ori- adult nonmammalian vertebrates and respond to neural
gin, with most produced by neural precursors that occupy injury by elevating cell division and neuron production.
the ventral neural tube (Rowitch and Kriegstein 2010), well Investigation of radial glia precursors in normal and injured
separated from neural crest. Oligodendrocytes and Schwann brains of genetically tractable organisms such as zebrafish
cells are specified from their precursors by distinct molecular might yield information that can be used to promote repair
regulatory mechanisms, and the manner in which they wrap and injury of human nervous systems damaged by disease
axons is very different. Therefore, the ability to form compact or injury.
myelin on axons likely arose twice at a very early stage in ver-
tebrate evolution.
Many axons cross the nerve cord boundary and are myeli- AC K N OW L E D G M E N T S
nated by both oligodendrocytes and Schwann cells. Because
absence of myelin on one portion of an axon causes a conduc- Thanks to Robert M. Gould for discussion and comments on
tion block, it has been proposed that myelinating Schwann the manuscript.

3. N O N M A M M A L I A N VE RT E B R AT E G L I A • 29
REFERENCES K á lmá n M, Gould RM. 2001. GFAP-immunopositive structures in
spiny dogfish, Squalus acanthias, and little skate, Raia erinacea,
Adolf B, et al. 2006. Conserved and acquired features of adult neurogen- brains: differences have evolutionary implications. Anat Embryol
esis in the zebrafish telencephalon. Dev Biol 295:278–293. 204:59–80.
Alvarez-Buylla A, Buskirk DR, Nottebohm F. 1987. Monoclonal Ká lmán M, Pritz MB. 2001. Glial fibrillary acidic protein-immunopositive
antibody reveals radial glia in adult avian brain. J Comp Neurol structures in the brain of a Crocodilian, Caiman crocodilus, and its
264:159–170. bearing on the evolution of astroglia. J Comp Neurol 431:460–480.
Baumgart EV, et al. 2011. Stab wound injury of the zebrafish telen- Ká lmán M, Székely AD, Csillag A. 1993. Distribution of glial fibrillary
cephalon: a model for comparative analysis of reactive gliosis. Glia acidic protein-immunopositive structures in the brain of the domes-
60:343–357. tic chicken (Gallus domesticus). J Comp Neurol 330:221–237.
Bernardos RL, Raymond PA. 2006. GFAP transgenic zebrafish. Gene Ká lmán M, Székely AD, Csillag A. 1998. Distribution of glial fibril-
Expr Patterns 6:1007–1013. lary acidic protein and vimentin-immunopositive elements in the
Bodega G, et al. 1993. Astroglial pattern in the spinal cord of the adult developing chicken brain from hatch to adulthood. Anat Embryol
barbel (Barbus comiza). Anat Embryol 187:385–398. 198:213–235.
Bodega G, Suárez I, Fernández B. 1990. Radial astrocytes and ependymo- Ká lmán M. 2002. GFAP expression withdraws—a trend of glial evolu-
cytes in the spinal cord of the adult toad (Bufo bufo L.). An immuno- tion? Brain Res Bulletin 57:509–511.
histochemical and ultrastructural study. Cell Tiss Res 260:307–314. Kaslin J, et al. 2009. Stem cells in the adult zebrafish cerebellum: ini-
Bone Q. 1960. The central nervous system in amphioxus. J Comp Neurol tiation and maintenance of a novel stem cell niche. J Neurosci
115:27–64. 29:6142–6153.
Bourlat SJ, et al. 2006. Deuterostome phylogeny reveals monophyletic Kaslin J, Ganz J, Brand M. 2008. Proliferation, neurogenesis and regen-
chordates and the new phylum Xenoturbellida. Nature 444:85–88. eration in the non mammalian vertebrate brain. Philosph Trans R
Brocklehurst G. 1979. The significance of the evolution of the cerebrospi- Soc Lond Series B, Biol Sci 363:101–122.
nal fluid system. Ann R Coll Surg England 61:349–356. Katz MJ. 1983. Comparative anatomy of the tunicate tadpole, Ciona
Bullock TH, Moore JK, Fields RD. 1984. Evolution of myelin intestinalis. Biol Bull 164:1–27.
sheaths: both lamprey and hagfi sh lack myelin. Neurosci Letts Kawai H, Arata N, Nakayasu H. 2001. Three-dimensional distribution
48:145–148. of astrocytes in zebrafish spinal cord. Glia 36:406–413.
Bundgaard M. 1982. Brain barrier systems in the lamprey. I. Ultrastructure Kitagawa K, et al. 1993. A proteolipid protein gene family: expression in
and permeability of cerebral blood vessels. Brain Res 240:55–64. sharks and rays and possible evolution from an ancestral gene encod-
Bundgaard M, Cserr H. 1981. A glial blood-brain barrier in elasmo- ing a pore-forming polypeptide. Neuron 11:433–448.
branchs. Brain Res 226:61–73. Knecht AK, Bronner-Fraser M. 2002. Induction of the neural crest: a
Bundgaard M, Cserr HF. 1981. Impermeability of hagfish cerebral capil- multigene process. Nat Rev Genet 3:453–461.
laries to radio-labelled polyethylene glycols and to microperoxidase. Konno A, et al. 2010. Distribution and structural diversity of cilia in tad-
Brain Res 206:71–81. pole larvae of the ascidian Ciona intestinalis. Dev Biol 337:42–62.
Bundgaard M, Abbott NJ. 2008. All vertebrates started out with a glial Kroehne V, et al. 2011. Regeneration of the adult zebrafish brain
blood-brain barrier 4–500 million years ago. Glia 56:699–708. from neurogenic radial glia-type progenitors. Development
Cecon S, Minnich B, Lametschwandtner A. 2002. Vascularization of 138:4831–4841.
the brains of the Atlantic and Pacific hagfishes, Myxine glutinosa and Kucenas S, et al. 2009. A selective glial barrier at motor axon exit points
Eptatretus stouti: a scanning electron microscope study of vascular prevents oligodendrocyte migration from the spinal cord. J Neurosci
corrosion casts. J Morphol 253:51–63. 29:15187–15194.
Coulpier F. et al. 2010. CNS/PNS boundary transgression by central glia Lacalli TC. 2000. Cell morphology in amphioxus nerve cord may reflect
in the absence of Schwann cells or Krox20/Egr2 function. J Neurosci the time course of cell differentiation. Int J Dev Biol 44:903–906.
30:5958–5967. Lacalli TC. 2008. Basic features of the ancestral chordate brain: a proto-
Cuoghi B, Mola L. 2009. Macroglial cells of the teleost central nervous chordate perspective. Brain Res Bull 75:319–323.
system: a survey of the main types. Cell Tissue Res 338:319–332. Lacalli TC, Kelly SJ. 2002. Floor plate, glia and other support cells in the
Davis AD, et al. 1999. Myelin-like sheaths in copepod axons. Nature anterior nerve cord of amphioxus larvae. Acta Zool 83:87–98.
398:571. Lazzari M, Franceschini V. 2001. Glial fibrillary acidic protein and
Duncan ID, Hoffman RL. 1997. Schwann cell invasion of the central vimentin immunoreactivity of astroglial cells in the central nerv-
nervous system of the myelin mutants. J Anat 190:35–49. ous system of adult Podarcis sicula (Squamata, Lacertidae). J Anat
Ganz J, et al. 2010. Heterogeneity and Fgf dependence of adult neural 198:67–75.
progenitors in the zebrafish telencephalon. Glia 58:1345–1363. Lazzari M, Franceschini V. 2005a. Astroglial cells in the central nervous
Grandel H, et al. 2006. Neural stem cells and neurogenesis in the adult system of the adult brown anole lizard, Anolis sagrei, revealed by inter-
zebrafish brain: origin, proliferation dynamics, migration and cell mediate fi lament immunohistochemistry. J Morphol 265:325–334.
fate. Dev Biol 295:263–277. Lazzari M, Franceschini V. 2005b. Intermediate fi lament immunohis-
Grupp L, Wolburg H, Mack AF. 2010. Astroglial structures in the tochemistry of astroglial cells in the leopard gecko, Eublepharis mac-
zebrafish brain. J Comp Neurol 518:4277–4287. ularius. Anat Embryol 210:275–286.
Hinsch K, Zupanc GKH. 2007. Generation and long-term persistence Lazzari M, Franceschini V. 2006. Glial cytoarchitecture in the central
of new neurons in the adult zebrafish brain: a quantitative analysis. nervous system of the soft-shell turtle, Trionyx sinensis, revealed
Neuroscience 146:679–696. by intermediate fi lament immunohistochemistry. Anat Embryol
Horie T, et al. 2011. Ependymal cells of chordate larvae are stem-like cells 211:497–506.
that form the adult nervous system. Nature 469:525–528. Lenz PH, Hartline DK, Davis AD. 2000. The need for speed. I. Fast
Ito Y, et al. 2010. Characterization of neural stem cells and their progeny reactions and myelinated axons in copepods. J Comp Physiol A
in the adult zebrafish optic tectum. Dev Biol 342:26–38. 186:337–345.
Itoyama Y, et al. 1983. Schwann cell remyelination of demyelinated axons Ma PM. 1993. Tanycytes in the sunfish brain: NADPH-diaphorase his-
in spinal cord multiple sclerosis lesions. Ann Neurol 14:339–346. tochemistry and regional distribution. J Comp Neurol 336:77–95.
Johns PR, Easter SS. 1977. Growth of the adult goldfish eye. II. Increase Marcus RC, Easter SS. 1995. Expression of glial fibrillary acidic protein
in retinal cell number. J Comp Neurol 176:331–341. and its relation to tract formation in embryonic zebrafish (Danio
Ká lmán M. 1998. Astroglial architecture of the carp (Cyprinus carpio) rerio). J Comp Neurol 359:365–381.
brain as revealed by immunohistochemical staining against glial Marz M, et al. 2010. Heterogeneity in progenitor cell subtypes in the ven-
fibrillary acidic protein (GFAP). Anat Embryol 198:409–433. tricular zone of the zebrafish adult telencephalon. Glia 58:870–888.

30 • NEUROGLIA
März M, et al. 2011. Regenerative response following stab injury in the Schweigreiter R, et al. 2006. Understanding myelination through study-
adult zebrafish telencephalon. Dev Dynam 240:2221–2231. ing its evolution. Int Rev Neurobiol 73:219–273.
Merrick SE, et al. 1995. Glial cells of the lamprey nervous system contain Waehneldt TV, Matthieu JM, Jeserich G. 1986. Appearance of myelin
keratin-like proteins. J Comp Neurol 355:199–210. proteins during vertebrate evolution. Neurochem Int 9:463–474.
Naujoks-Manteuffel C, Roth G. 1989. Astroglial cells in a salamander Wasowicz M, et al. 1994. Immunoreactivity to glial fibrillary acid pro-
brain (Salamandra salamandra) as compared to mammals: a glial tein (GFAP) in the brain and spinal cord of the lamprey (Lampetra
fibrillary acidic protein immunohistochemistry study. Brain Res fluviatilis). Journal f ür Hirnforschung 35:71–78.
487:397–401. Wasowicz M, Ward R, Repérant J. 1999. An investigation of astro-
Northcutt RG, Gans C. 1983. The genesis of neural crest and epider- glial morphology in torpedo and scyliorhinus. J Neurocytol
mal placodes: a reinterpretation of vertebrate origins. Q Rev Biol 28:639–653.
58:1–28. Wicht H, Derouiche A, Korf HW. 1994. An immunocytochemical
Nuñez VA, et al 2009. Postembryonic development of the posterior lat- investigation of glial morphology in the Pacific hagfish: radial and
eral line in the zebrafish. Evol Dev 11:391–404. astrocyte-like glia have the same phylogenetic age. J Neurocytol
Oberheim NA, Goldman SA, Nedergaard M. 2012. Heterogeneity of 23:565–576.
astrocytic form and function. Meth Mol Biol 814:23–45. Wicht H, Lacalli TC. 2005. The nervous system of amphioxus: struc-
Onteniente B, Kimura H, Maeda T. 1983. Comparative study of the glial ture, development, and evolutionary significance. Can J Zool
fibrillary acidic protein in vertebrates by PAP immunohistochemis- 83:122–150.
try. J Comp Neurol 215:427–436. Zalc B, Goujet D, Colman D. 2008. The origin of the myelination pro-
Reichenbach A. 1989. Glia:neuron index: review and hypothesis to gram in vertebrates. Curr Biol 18:R511–R512.
account for different values in various mammals. Glia 2:71–77. Zalc B. 2006. The acquisition of myelin: a success story. Novartis Found
Reichenbach A, Neumann M, Brückner G. 1987. Cell length to diameter Symp 276:15–21.
relation of rat fetal radial glia—does impaired K+ transport capacity Zawadzka M, et al. 2010. CNS-resident glial progenitor/stem cells pro-
of long thin cells cause their perinatal transformation into multipolar duce Schwann cells as well as oligodendrocytes during repair of CNS
astrocytes? Neurosci Letts 73:95–100. demyelination. Cell Stem Cell 6:578–590.
Reimer MM, et al. 2008. Motor neuron regeneration in adult zebrafish. Zimmerman A, Lowery M. 1999. Hyperplastic development and hyper-
J Neurosci 28:8510–8516. trophic growth of muscle fibers in the white seabass (Atractoscion
Roots BI. 2008. The phylogeny of invertebrates and the evolution of mye- nobilis). J Exp Zool 284:299–308.
lin. Neuron Glia Biol 4:101–109. Zupanc GK. 2001. Adult neurogenesis and neuronal regeneration
Rowe RW, Goldspink G. 1969. Muscle fibre growth in five different mus- in the central nervous system of teleost fish. Brain Behav Evol
cles in both sexes of mice. J Anat 104:519–530. 58:250–275.
Rowitch DH, Kriegstein AR. 2010. Developmental genetics of verte- Zupanc GK, Hinsch K, Gage FH. 2005. Proliferation, migration, neu-
brate glial–cell specification. Nature 468:214–222. ronal differentiation, and long-term survival of new cells in the adult
Rubio M, et al. 1992. Glial fibrillary acidic protein and vimentin immu- zebrafish brain. J Comp Neurol 488:290–319.
nohistochemistry in the posterior rhombencephalon of the Iberian Zupanc GK. 2008. Adult neurogenesis and neuronal regeneration in the
barb (Barbus comiza). Neurosci Letts 134:203–206. brain of teleost fish. J Physiol 102:357–373.

3. N O N M A M M A L I A N VE RT E B R AT E G L I A • 31
This page intentionally left blank
MORPHOLOGY, ULTRASTRUCTURE, AND IDENTIFICATION
This page intentionally left blank
4.
ASTROCY TES AND EPENDYMAL GLIA
Andreas Reichenbach and Hartwig Wolburg

A B B R E VI AT I O N S principle, radial glia cells are involved in brain development,


by acting as stem and progenitor cells generating neurons and
AQP aquaporins (water channels) glia, as well as by guiding migrating neurons and thereby con-
BMI basal membrane infoldings tributing to the organized arrangement of neurons (Götz and
CA1 region region 1 in the cornu ammonis (hippocampus) Huttner 2005; Howard et al. 2008) (see chapter 5). Although
CNS central nervous system most radial glia cells differentiate into neurons and all macro-
CVO circumventricular organs glial classes, some radial glia cells maintain a radial arrange-
GFAP glial fibrillary acid protein ment in the adult brain; these cells are then called tanycytes
GFP green fluorescent protein (see chapter 12) (in brain: Fig. 4.1I) or Müller cells (in retina:
GLAST glutamate aspartate transporter Fig. 4.2A).
GS glutamine synthetase There are two types of astroglia-like cells in the central
NG2+ cells glial/progenitor cells expressing the chondroi- nervous system (CNS) that will not be subject of this chapter.
tin sulfate proteoglycan, NG2 First, the olfactory ensheathing cells partially occupy the brain
Kir4.1 inwardly rectifying potassium channel type 4.1 but are descendants of the peripheral nervous system. Second,
NKCC1 Na+/K+/2Cl– cotransporter the immature astrocytes/glioblasts (Fig. 4.1, IX) will be sub-
OAP orthogonal arrays of particles ject of chapter 12.
RPE retinal pigment epithelium
SVZ subventricular zone

pia mater
1 M O R P H O L O GY O F T H E P E C U L I A R
III
CELL TYPES molecular
layer
As shown in Figure 4.1, the morphology of astroglia and VIII
ependymoglia is very diverse. In particular, the soma of the II
cells may give rise to one or several “primary” or “stem” pro- blood grey
cesses, from which secondary branches may begin. Much of vessel matter
this diversity is related to structural and functional interac- IV
tions of a given cell with its microenvironment, which includes granular
on the one side the neurons and on the other side, blood ves- V
VII layer
sels, the pia mater, and/or the ventricular space (Wolburg
et al. 2009). Macroglial cells may form a “border sheath” against blood white
the ventricular space, the pia, or blood vessels. This is observed la
vessel matter
blood
in ependymocytes (Fig. 4.1, X), choroid plexus cells (Wolburg vessels
lb VI
subepen-
basal membrane
and Paulus 2010) (Fig. 4.1, XI), and retinal pigment epithelial IX labyrinth(s) dymal zone
cells (Fig. 4.4C) but also, although less obvious, in marginal
XI X ependyma
astrocytes (Fig. 4.1, III), perivascular astrocytes (Fig. 4.1, VII),
and pecten glial cells (Fig. 4.4A, B). Within the brain paren- cilium ventricle
microvilli
chyma proper, typical astrocytes are more or less star-shaped
(Fig. 4.1, IV and VIII), but this may be modified by adjacent Figure 4.1 Semi-schematic survey of the main types of astroglial and
neuronal cell bodies (e.g., Fig. 4.1, V) or axons (Fig. 4.1, VI), or ependymoglial cells, and their localization in different layers/specialized
peculiar relationships to the pial surface (Fig. 4.1, II). The term regions of the central nervous system tissue. I, tanycyte (a, pial; b, vascular);
radial glia should be restricted to bipolar ependymoglial cells II, radial astrocyte (Bergmann glial cell); III, marginal astrocyte; IV,
protoplasmic astrocyte; V, velate astrocyte; VI, fibrous astrocyte; VII,
that extend long processes throughout (most of ) the thickness perivascular astrocyte; VIII, interlaminar astrocyte; IX, immature
of the tissue from one surface (the outer pial surface) to the astrocyte/glioblast; X, ependymocyte; XI choroid plexus cell. From
other surface (the inner ventricular surface) of the brain. In Reichenbach and Wolburg 2005, with permission.

35
1.1 R A D I A L G L I A L C E L L S I N A D U LT C E N T R A L in the retinal tissue; their densities per mm2 retinal surface
N E RVO US SYS T E M ( TA N YC Y T E S ) area range between 1,550 and 2,000 (frog, salamander), 5,000
Throughout the literature, the terms radial astrocyte and to 12,000 (most mammals), and greater than 25,000 (in the
radial glia are often used as synonyms. As the primary (fetal) fovea centralis of primates). Each Müller cell ensheathes and
radial glia consists of bipolar ependymoglial cells, we propose supports a columnar group of retinal neurons. The number of
to reserve this term in adult CNS to persistent ependymoglial neurons per Müller cell ranges between 7 (tree shrew), about
cells such as tanycytes and Müller cells, whereas Bergmann 16 (human nonfoveal retina, rabbit, and other herbivorous
glial cells and similar astrocytic cells (e.g., in the hippocam- mammals), about 30 (rodents and carnivores with strongly
pus), which lack an ependymal process, should be termed rod-dominant retinae), up to 80 (frog), and even more than
radial astrocytes (or radially oriented astrocytes). 200 (deep sea teleosts with lifelong generation of new rod
Radial glial cells, often referred to as tanycytes (Fig. 4.1, Ia photoreceptor cells). The size and shape of Müller cells depend
and Ib) are the most common type of macroglia in the CNS of on animal species and, within a given retina, on retinal topog-
lower vertebrates (and even of deuterostomic invertebrates). raphy. The total length of a Müller cell is determined by the
In adult mammals, they are restricted to certain brain regions (local) thickness of the retina, with a few exceptions in reti-
where the tissue is rather thin, such as some circumventricular nae with thick blood vessels in which individual Müller cells
organs (circumventricular organs [CVOs], e.g., the subcom- may end at the scleral surface of such a vessel, rather than the
missural organ, subfornical organ, area postrema), the stalk inner limiting membrane facing the vitreous body. Generally,
of the hypophysis, and the velum medullare, and to the raphe in rod-dominant retinae, a Müller cell extends just one stem
region of the spinal cord. In the CVOs of all vertebrates except process from soma to the inner limiting membrane. This is the
sharks, the capillaries are fenestrated. In these regions, the case in most fish and mammals. In the cone-dominant retinae
tanycytes (as well as the choroid plexus epithelial cells within of most reptiles and birds (as well as of the tree shrew), the
the choroid plexus) constitute the blood-cerebrospinal fluid vitread stem processes of Müller cells are split into several
barrier by expressing extensive tight junctions. Some of these branches. In species with a native polyploidy (e.g., lungfish and
tanycytes are specialized for the secretion of signaling mol- salamander) not only the cell nucleus, but also the entire cell
ecules, and of the material constituting Reissner’s fiber. In the is huge. This may be advantageous for experiments on single
adult subventricular zone (SVZ), short tanycytes are found cells such as intracellular electrophysiological recordings, dye
which extend a cilium into the ventricle, and form endfeet injections, etc. Literature about Müller cells can be found in
at blood vessels; these cells were shown to function as stem Reichenbach and Bringmann (2010).
cells (see chapter 30). More about tanycytes and the choroid
plexus can be found in Wolburg et al. (2009) and Wolburg 1.3 R A D I A L A S T RO C Y T E S
and Paulus (2010).
Radial astrocytes (Fig. 4.1, II) are common in the spinal cord
and brain of lower vertebrates. As they cross white and gray
matter, the properties of their cell processes may change from
1.2 M Ü L L E R C E L L S
“protoplasmic” to “fibrous.” In some lower vertebrates such
Müller cells are the radial glia of the retina. In many verte- as fish, radial astrocytes possess “velate” processes. Typical
brates, including some mammals (i.e., those with avascular examples of radial astrocytes are shown in Figure 4.2A. Some
retinae), they are the only cells representing the macroglia fam- radial astrocytes are also found in the optic nerve of mammals,
ily. They contact virtually every neuronal and non-neuronal where they are intermingled with the more abundant longitu-
element of the retina with specialized branches of their pro- dinally aligned fibrous astrocytes (Fig. 4.2F).
cesses. In the nuclear layers of the retina (particularly, in the In the mammalian hippocampus, radially oriented astro-
outer nuclear layer containing the somata of the photorecep- cytes occur that do not abut the pia with their processes;
tor cells) the Müller cell processes assume the shape of velate rather, they are confined to the stratum granulare of the den-
astrocytes, whereas in the plexiform layers (where the synapses tate gyrus (Feig and Haberly 2011) and the stratum oriens
of the retinal neurons are located) the Müller cell processes of the CA1 region. Examples of these cells are shown in
resemble those of protoplasmic astrocytes (see Fig. 4.1). In the Figure 4.2B, C (see chapter 5). They should be considered as
central retina close to the optic nerve head, in many species a unique cell type, clearly different from the radial astrocytes
the nerve fiber layer becomes very thick; there, the Müller cell of lower vertebrates (and from Bergmann glial cells), which
processes are thin and smooth like those of fibrous astrocytes. all contact the pia mater.
Müller cells occupy a variable volume fraction of the retinal
tissue, from about 3% (most lower vertebrates), 5% to 8% 1.4 B E RG M A N N G L I A
(most mammals, i.e., those with vascularized retinae) up to
about 20% (mammals with avascular retinae, such as the rab- According to our definition, Bergmann glial cells (also termed
bit and guinea pig). The volume of individual Müller cells may Golgi epithelial cells) are radially oriented astrocytes of the cer-
vary from 400 μm3 (mouse) to greater than 2,000 μm3 (rabbit, ebellum in all vertebrates. However, because their cell bod-
retinal periphery); their surface area is in the range of 6,000 to ies reside in the layer of the Purkinje cell somata, and their
12,000 μm2. Müller cells constitute a fairly uniform “lattice” processes (usually, 3–6 per cell) cross the molecular layer,

36 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
A
f
f
nerve
p p

radially oriented
astrocytes:
p mouse optic nerve

v axon

Bergmann glial cells: radially oriented


chick cerebellum astrocytes:
Müller cells: cow retina turtle spinal cord
50μm 25μm
B C
SM

SG

MCE

MCE

MCE

D E 20μm

100μm

fibrous astrocytes:
F cat optic nerve mouse optic nerve

Figure 4.2 A–C. Radially oriented glial cells. (A) Camera-lucida drawings of Golgi-stained or dye-injected cells. Müller cells represent bona fide radial
glial cells (as the “tanycytes of the retina”), whereas cerebellar Bergmann glial cells and radially oriented astrocytes in other central nervous system
regions miss a contact to the ventricular surface and thus are specialized astrocytes. Note that the character of the processes is determined by their local
microenvironment and may change from complex (“protoplasmic,” p) to smooth (“fibrous,” f ) when they pass from gray to white matter. In nuclear
layers with many small neuronal somata they are velate (v). (B, C) Glial fibrillary acid protein immunofluorescence of the dentate gyrus of
the hippocampus of an adult rat. Astroglial cell processes are radially aligned in the stratum granulare (SG), whereas typical “star-shaped” astrocytes
are found in the stratum moleculare (SM). D–F. Non–radially oriented fibrous astrocytes in the mammalian retina (D, E) and optic nerve (F).
(D, E) Glial fibrillary acid protein-labeled fibrous astrocytes in the flat-mounted murine retina, immunofluorescence. Close to the optic nerve (D) the
astrocytic processes are aligned in parallel to the axon bundles that run between rows of Müller cell endfeet (MCE). In the retinal periphery (E), the
cells are more or less star-shaped and form a regular pattern that is only modified by the contacts to retinal blood vessels (asterisks). (F) Camera-lucida
drawings of fibrous astrocytes from cat (Golgi stained cell) and murine optic nerve (dye-injected cell). The cell processes are mainly aligned
longitudinally (i.e., parallel to the axons). The cells extend fingerlike perinodal processes; an artist’s reconstruction of one of them is shown in
(A, bottom right), based on electron microscopical serial sections of the dye-labeled cell. (B,C) Original confocal microphotographs (courtesy of Gert
Brückner, Leipzig). Modified after Reichenbach (1989a) and Reichenbach and Wolburg. 2005, in which the references can also be found.

4 . A S T R O C Y T E S A N D E P E N DY M A L G L I A • 37
but do not reach the ventricular surface, they do not belong A stem fiber
to radial glial cells in the strictest sense. Rather, they resem-
ble protoplasmic astrocytes. Viewed from the pial surface, the
processes of Bergmann glial cells form rows or palisades that
are aligned parallel to the long axis of the folium, therefore
topologically resembling radial glia cells. Their many elabo-
rate side branches (Fig. 4.3) display complex morphological
and functional interactions with the synapses on the dendrites
of the Purkinje cells (see Fig. 4.3); they are characterized by stem fiber
a high surface-to-volume ratio of up to greater than 20 μm–1
(Grosche et al. 1999, 2002). In rodent cerebellum, there are
about 8 Bergmann glial cells per Purkinje cell (i.e., about 8000
mm–1 cerebellar surface area). It has been estimated that each
Bergmann glial cell ensheathes several thousands (2,000–
6,000) Purkinje cell synapses (Reichenbach et al. 2010).
Bergmann glial cells occupy about 15% to 18% of the volume
of the molecular layer of the cerebellum. An average rodent
Bergmann glial cell has a volume of about 3600 μm3. The size
and shape of Bergmann glial cells differ in dependence on ani-
mal species. The total length of the processes is determined by
the thickness of the molecular layer. Generally, in small spe-
cies (e.g., in shrews) the Bergmann glial cell processes are short
and densely covered with lateral appendages, whereas in large
species (e.g., humans) these processes are much longer but stem fiber
show less dense lateral outgrowths. Figure 4.5B gives a survey
of the Bergmann glial cell morphology in several vertebrates.
Fañana’s cells constitute a subtype of short Bergmann glial B D Bergmann
cells, with their somata being located in the molecular layer glia cell
processes
rather than at the level of the Purkinje cell somata. stalk

head
1.5 P ROTO P L A S M I C A S T RO C Y T E S (stem fiber) neuronal
Astrocytes are the “prototypical” macroglial cells of the mam- Purkinje cell spine element
C
malian brain, although they occur also in lower vertebrates. mito-
Protoplasmic astrocytes (Fig. 4.1, IV; Fig. 4.5C, D) are found chondrion
in the gray matter. Their numerous processes are spread more
stalk
or less radially from the soma, usually occupying a spheroid parallel
stem
volume, and extend many fine and complex lamellar side fiber head fibers
branches. In rodent brain astrocytes, these surface extensions
occupy about 50% of the volume but as much as 80% of the Figure 4.3 Three-Dimensional Reconstruction of a Part of a Bergmann
surface area of an average cell (Vcell ~5,500 μm3; Scell ~80,000 Glial Cell Process in the Murine Cerebellum. A. The living cell was
μm2) (Chao et al. 2002), resulting in high surface-to-volume dye-injected in a perfused cerebellar slice. Then, after fixation and
dye-conversion, about 600 consecutive serial ultrathin sections were
ratios of 10 to 20 μm–1. Thus, although the volume fraction of photographed in the electron microscope, and the images of the
astroglia in the cortical tissue amounts only to 10% to 20%, dye-labeled profiles were reconstructed by a computer program. The
the astrocytic processes and side branches contact much of inset shows a substructure labeled in blue; this part was quantitatively
the neuronal surfaces present in a given brain volume com- analyzed (B, C). B,C. A microdomain of the Bergmann glial cell process
partment (Chao et al. 2002). Human astrocytes are larger shown in (A); reconstruction (B) and schematic drawing of such a
glial microdomain and its relationships to the neuronal elements (C).
and even more complex (Oberheim et al. 2006, 2009) (Fig. D. Three-dimensional reconstruction of a group of neighbored cerebellar
4.5C). Independent on species, at least one of the cell pro- synapses (yellow; synaptic clefts: orange) together with the surrounding
cesses is bearing one or several perivascular endfeet such that leaflets provided by the injected Bergmann glial (blue-green). The
the surfaces of the blood vessels in the CNS are virtually com- arrowheads point to neuronal surfaces not covered by glial sheaths from
pletely ensheathed by astroglial endfoot plates (Mathiisen et the labeled cell. Modified after Reichenbach (1989a) and Reichenbach
and Wolburg 2005, in which the references also can be found.
al. 2010). The density of astrocytes in the cerebral cortex is
high (layers III/IV: 4,000–10,000 mm–3 in lissencephalic
cortices of insectivores [Stolzenburg et al. 1989]; 12,000
≥ 30,000 mm–3 in the rat cortex [Distler et al. 1991]). The from about 0.1 in shrew to about 5 in whale, with interme-
cortical glia-to-neuron index (largely determined by proto- diate values of about 2 in humans (for a survey of the rich
plasmic astrocytes) increases with the thickness of the tissue; literature, see Reichenbach and Wolburg 2009).

38 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
1.6 FI B RO US A S T RO C Y T E S plates. Usually they extend several long, smooth processes
Fibrous astrocytes (Fig. 4.1, VI; Fig. 4.2D–F) are found in down into the neuropil (not unlike the interlaminar astro-
white matter tracts, the optic nerve, and the retinal nerve fiber cytes), but their main function is thought to constitute
layer of mammals with vascularized retinae. Their somata are a glial “limiting zone” below the pia mater. Recently, such
often arranged in rows between the axon bundles. Their pro- surface-associated, gap junction–interconnected astrocytes
cesses are comparatively smooth, and frequently oriented in have been described in the posterior piriform cortex of the
parallel to the axons. For the mouse optic nerve it was shown rat, and were discussed in terms of neurovascular regulation,
that every astrocyte possesses several perivascular and/or sub- interstitial homeostasis, and neuromodulation (Feig and
pial endfeet (see Fig. 4.2F). The fibrous astrocytes in the ret- Haberly 2011).
ina may bear endfeet at both the intraretinal blood vessels and In the human and rabbit retina, perivascular astrocytes
the vitreal surface. As a characteristic feature, the processes of have been described (Fig. 4.1, VII) that are virtually devoid of
fibrous astrocytes extend multiple fingerlike outgrowths into neuron-contacting processes but form extensive endfoot con-
the perinodal space of adjacent axons (Figs. 4.2A, 4.4G). A tacts to retinal blood vessels. They seem to occur also elsewhere
density of about 200,000 fibrous astrocytes mm–3 has been in the brain (e.g., the neurohypophysis), but their occurrence
estimated for the anterior commissure of the mouse (Sturrock and possible functions are poorly studied so far. In analogy to
et al. 1977). The processes of fibrous astrocytes are generally the marginal astrocytes, they may constitute a peculiar “glial
longer (mouse: up to 300 μm) than those of protoplasmic coating” around the blood vessels.
astrocytes (mouse: < 50 μm) but their surface-to-volume ratio
is significantly smaller (~5 μm-1). Examples of fibrous astro- 1.10 E P E N DY M O C Y T E S , C H O RO I D
cytes from different locations are shown in Fig. 4.2D–F. P L E XUS C E L L S , A N D R ET I NA L P I G M E N T
E P IT H E L I A L C E L L S
1.7 VE L AT E A S T RO C Y T E S Ependymocytes, choroid plexus cells, and retinal pigment
Velate astrocytes (see Fig. 4.1, V) were described in the granule epithelial cells are specialized glial cells lining the ventricle
layer of the cerebellum, where each of them surrounds several (or the subretinal space, respectively). At their basal pole,
small neuronal granule cells with velate sheaths (for summary most mature ependymocytes (see Fig. 4.1, X) contact rem-
of the literature, see Reichenbach and Wolburg 2009). Similar nants of embryonic blood vessels (so-called “basement mem-
cells occur in the olfactory bulb, where they ensheathe several brane labyrinths”) rather than intact blood vessels. On their
periglomerular neurons and dendritic segments. Obviously, other (i.e., the ventricular) pole, they possess, in addition to
this cell type develops at sites where many small, densely microvilli, kinocilia to support the stream of the cerebrospi-
packed neurons occur. Morphometric data on velate astro- nal fluid. The latter is mainly secreted by the choroid plexus
cytes are not available, but by comparison with the “velate” cells (see Fig. 4.1, XI), characterized by a high density of
parts of Müller cell processes in the outer nuclear layer, it can Na+/K+-ATPase molecules at their microvillous membrane.
be estimated that their surface-to-volume ratio is very high This secretion requires a high permeability of the fenes-
(20–30 μm–1). trated plexus endothelial cells; thus, the blood-cerebrospinal
fluid barrier is formed by the choroid plexus epithelium
(Wolburg and Paulus 2010). Retinal pigment epithelial
1.8 I N T E R L A M I NA R A S T RO C Y T E S
(RPE) cells (Fig. 4.4C) line the subretinal space opposite
Interlaminar astrocytes (see Fig. 4.1, VIII) have been found in to the neuroretina. Their apical surface extends two types
the supragranular layers of the cerebral cortex of higher pri- of microvilli: long (5–7 μm) thin microvilli maximizing the
mates, including humans; they do not occur in lower mam- membrane area available for transepithelial transport, and
mals. They are similar to protoplasmic astrocytes in the upper specially arranged shorter microvilli termed photoreceptor
cortical layers (I–III), but are characterized by a long (up to 1.0 sheaths. The basal surface of the RPE cells contains numerous
mm in humans) process arising from the internal (deep) side invaginations (basal membrane infoldings, BMI, Fig. 4.4C)
of the cell soma usually located in lamina I, and descending to increase the surface area. The size and shape of the RPE
over at least two laminae (down to lamina IV, where it ends in cells depend on their location in retina; within the human
a small bulb). Collectively they form a visible “palisade” that macula, the cells measure about 14 μm in diameter (12 μm
has been found to be severely disrupted in cases of Alzheimer height) but they become wider (up to 60 μm diameter) and
disease and in response to experimental mechanical lesions in flatter in the periphery. Like the choroid plexus cells, the
monkeys. Although their functional relevance is still unclear, RPE cells (1) are in close apposition to many blood vessels,
it is speculated that they may optimize the modular (colum- (2) are specialized for transmembrane transport, and (3)
nar) organization of the cortex (Colombo 2001). form the blood-cerebrospinal (or, in this case, -subretinal)
fluid barrier by their tight junctions (Fig. 4.4E). Like chor-
1.9 M A RG I NA L G L I A A N D P E R I VA S CU L A R
oid plexus epithelial cells the RPE cells express the water
A S T RO C Y T E S
channel protein aquaporin 1 (AQP1), which is present in
the apical membrane opposite to the photoreceptor outer
Close to the pia mater, specialized astrocytes can be found segments. An important difference between pigment and
(see Fig. 4.1, III) that may form several layers of endfoot choroid plexus epithelial cells is the direction of water flux:

4 . A S T R O C Y T E S A N D E P E N DY M A L G L I A • 39
A B Choroid plexus cells release water across the apical mem-
brane into the ventricle. However, if RPE cells would release
P
water into the analogous space, that is, the subretinal space
vit vit bv (representing an obliterated ventricular cleft), detachment
R
of the neuroretina from the pigment epithelium would be
the disastrous consequence. This is avoided by the inward
R vit
RPE
RPE vit c direction of the Na+/K+/2Cl– cotransporter (NKCC1) and
ONH the Na+/bicarbonate cotransporter in the apical membrane,
500 μm 50 μm followed by AQP1-mediated water flux from the subretinal
space toward the choroid vasculature. This even supports
C D
the apposition of the retina to the pigment epithelium
(Strauss 2005). Another specific feature of RPE cells is the
0.5 μm presence of black (melanin) pigment granules aimed at the
absorption of light that passed the photoreceptor cells, and
E
thus at the avoidance of back-scattering light. By contrast, in
some vertebrates adapted to dark environments (e.g., fishes
living in the deep sea or turbid water), the RPE cells may
0.2 μm
contain guanidine crystals to reflect as much light as pos-
F sible. Even in these instances, however, melanin granules
are present; probably, they serve an important role as sinks
for free radicals and excited oxygen species such as singlet
oxygen. A comprehensive overview about ependymocytes,
G choroid plexus cells, and RPE cells can be found in Wolburg
et al. (2009).

1.11 P EC T E N E A L G L I A L C E L L S
The pecten oculi is a peculiar vascular structure of the avian
0.25 μm 100 nm eye, where it bulges from the optic nerve head into the vitre-
ous body (Fig. 4.4A,B). It is comprised of two types of cells,
Figure 4.4 A–C. Specialized ependymoglia of the eye. A. Survey of the endothelial cells and specialized glial cells. The latter (like
fundus of an avian (blue-and-yellow macaw) eye. Close to the optic nerve the RPE cells) originate in the outer leaflet of the optic cup.
head (ONH), two peculiar forms of ependymoglia can be found: in the
pecten (P), the pecten glial cells, and below the retina (R), the retinal They contain pigment granules (like RPE cells) but lose their
pigment epithelial (RPE) cells. B. Higher magnification of an area of the tight junctions during differentiation (unlike RPE cells) such
pecten shown in (A). Many capillaries (c) and larger blood vessels (bv) are that the blood-retina barrier is maintained by the endothe-
embedded in a tissue that contains endothelial cells and pigmented pecten lial cells of the pecten. Immunocytochemical localization of
glial cells (black arrowheads) that contact the vitreous (vit). The pecten glial the enzyme glutamine synthetase (GS) in these cells is weak
cells contain pigment granules but do not form tight junctions (D, freeze
fracture replica). C. The RPE cells (e.g., from rabbit) contain pigment during embryonic development but increasingly strong after
granula (P). Apically (i.e., toward the outer segments of the photoreceptor hatching. Although in astrocytes and Müller cells, GS expres-
cells, ROS) they extend microvilli, which may enclose the shed tips of sion is thought to be involved in transmitter recycling, in the
ROS (asterisk) as a first step of phagocytosis. Basally, the cells face a basal pecteneal glia it may participate in the pH-regulation of the
lamina (Bruch’s membrane, between the arrows) and display an enlarged avian eye (Wolburg et al. 1999). Very probably, the pecten
surface area resulting from basal membrane enfoldings (BME). Because
the capillaries of the underlying choroid possess a fenestrated endothelium glial cells play important roles in the nutrition and detoxifica-
(black arrowheads), the RPE cells from the blood-retina barrier by the tion of the avian retina (Wolburg et al. 1999).
expression of tight junctions (white arrowheads in C and “networks” in E). In addition to the Müller cells in the avian retina and
D–F. Freeze-fracture replicas demonstrating specific astroglial membrane pecteneal glial cells that are developmentally related to the
features of pecten glial cells (D), RPE cells (E), and a subpial astrocytic RPE, there is a further population of retinal glial cells lin-
endfoot membrane of the adult rat optic nerve (F). The tight junctions
form a regular network in the lateral membranes of chicken RPE cells ing the base of the pecten oculi; it has been described in the
(E), but are rudimentary in pecten glial cells (arrowhead in D). At the line chicken retina as the peripapillary glia, specifically expressing
between the white arrowheads in (E) as indicated by the white arrowheads, R-cadherin (Schuck et al. 2000). This retina-specific type of
the fracture level switches from the protoplasmic (pf ) to the external face glia retains characteristics of radial glia by spanning the dis-
(ef ) of the cell membrane. In addition, gap junctions (encircled) also occur tance from the vitreous to the ventricular cleft. Furthermore,
between RPE cells (and, typically, between astrocytes; not shown). F.
Both Müller cell (not shown) and astrocytic endfoot membranes display it represents the border between the vascularized optic
orthogonal arrays of particles (OAPs; encircled). G. Transmission electron nerve head and the adjacent avascular retina, suggesting
micrograph of a corona of fingerlike astrocytic processes around a nodelike that these cells demarcate the outer limit of vascularization
specialization of an unmyelinated axon (asterisk) in the rat retinal nerve and prevent the ingrowth of vessel sprouts into the retina.
fiber layer (see Fig. 4.2A). Modified after Reichenbach (1989a) and However, after injection of R-cadherin antibodies and pre-
Reichenbach and Wolburg 2005, in which the references also can be found.
absorbed complement to lyse the R-cadherin-positive glial

40 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
Table 4.1 MARKER ANTIGENS
CELL TYPE ANTIGEN DEVELOPING ADULT REACTIVE

Astroglia GFAP (+) ++ +++


Vimentin +++ – ++
Nestin ++ – ++
CytokeratinF,A ++
Glutamine synthetase ++
iNOS + +++
RC1/2 (antibodies) ++ –
RAN-2 + ++
BLBP* ++ –
S-100β – ++
3CB2C
R-cadherinC + ++
Müller glia GFAPF – – +++
Vimentin + ++ +
RAN-2 ++
3CB2 ++
nNOS/iNOS + +++
CA ++ ++
S-100β ++
3CB2 ++
B-cadherinC + ++
F1* ++ ++
Glutamine synthetase ++
CRALBP ++
Bergmann glia GFAP ++
Vimentin ++ +
Nestin ++
Glutamine synthetase ++
S-100β ++
Ependymoglia GFAP (+) ++
Vimentin ++
Cytokeratin +++
RAN-2 ++
Retinal pigment Vimentin ++
epithelium cells CRALBP +++
R-cadherinC ++ –
Pecten glia Vimentin + ++
(only avian) B-cadherin + ++
Glutamine synthetase ++
Choroid plexus Cytokeratin ++
epithelium GFAP – ++
Vimentin ++
Neurofilament* ++

List of “marker antigens” suitable to visualize and/or identify the various types of astroglial and ependymoglial cells during ontogenetic development, in the normal
mature CNS, and during reactive changes in cases of pathology
Whereas the list basically reflects the situation in mammals, many of the antigens can also be found in the particular cell types of other vertebrates. In cases in which an
antigen is only found in non-mammalian cells, it is labeled by an exposed letter: F, fishes; A, amphibians; C, chicken. An asterisk indicates that the listed antigen (or anti-
body, respectively) labels not selectively glial cells, but (in other regions of the CNS) also neurons. Antigens expressed in the cytoplasmic membranes such as ion chan-
nels, receptors, or adhesion molecules are excluded from the list because immunocytochemistry for these antigens usually results in “diffuse” labeling of the neuropile, at
the light microscopical level.
BLBP, brain lipid–binding protein; CA, carbonic anhydrase; CRALBP, cellular retinaldehyde-binding protein; GFAP, glial fibrillary acidic protein; NOS, nitric oxide
synthase (n neuronal, i inducible form); RAN-2, rat neural antigen-2.

4 . A S T R O C Y T E S A N D E P E N DY M A L G L I A • 41
cells, blood vessels were not observed to enter the retina bundles of intermediate filaments can be found that may even
(Gerhardt et al. 2000). be used as “markers” of astroglial cells in electron microscopic
sections. The somata of some types of ependymoglial cells
contain conspicuous melanin pigment granules (e.g., RPE
2 I M MU N O C Y TO C H E M I C A L cells, pecteneal glial cells, and choroid epithelial cells). In most
VI S UA L I Z AT I O N A N D of these epithelioid glial cells (including the ependymocytes
I D E N T I F I C AT I O N O F A S T R O G L I A L but not the pecteneal glial cells), the lateral membranes of the
A N D E P E N DY M O G L I A L C E L L S somata are interconnected by zonulae adherentes and tight
junctions, thus forming the blood-brain (or -retina) barrier
Astrocytic and ependymoglial cells can be visualized (and, (Wolburg et al. 2009).
in many instances, even identified) by immunocytochemical In astroglial and ependymoglial cell nuclei, the nucleo-
labeling of certain antigens that are, at least within the CNS, plasm is rather evenly distributed if compared with that in
restricted to these cells. The expression of these molecules by oligodendrocytes and microglial cells. In some ependymoglial
certain cell types may change with differentiation, or dur- cells such as in many tanycytes, the cell nuclei are very irregu-
ing pathological processes (Table 4.1). Furthermore, not all larly shaped and may display deep incisions. The nuclei (and
members of a given cell population (otherwise considered somata) of Müller cells seem to be “indented” by neighbor-
homogeneous) must express the same antigen (at detectable ing neurons. It can be shown by atomic force microscopy that
levels); for instance, although it may be safely stated that Müller cell somata are “softer” (i.e., possess a lower module of
every glial fibrillary acid protein (GFAP)-immunopositive elasticity) than the somata of the neighboring bipolar neurons
cell in brain is an astrocyte, there seem to be many astro- (Lu et al. 2006).
cytes in brain that are not labeled by antibodies directed to
GFAP. It should also kept in mind that some of the antigens
3.2 S T E M P RO C E S S E S
(e.g., the intermediate filament proteins) allow mainly an
immunocytochemical visualization of the larger stem pro- Stem processes are those cellular processes that directly arise
cesses, whereas antibodies directed to cytoplasmic proteins from soma. Typically they contain bundles of intermediate
(e.g., glutamine synthetase and S-100β) may stain fine side filaments. Particularly high densities of intermediate filaments
branches, and may be used at the electron microscopical are found in the basal (i.e., endfoot-bearing) processes of tany-
level to identify even very thin cytoplasmic leaflets of glial cytes and Müller cells, and in processes of fibrous and radial
sheaths. By contrast, antibodies against membrane proteins astrocytes including Bergmann glia. Microtubules are rarely
(e.g., ion channels, ligand receptors, transporter proteins) found in astroglial cell processes. One of the few exceptions
may label (parts of ) the cell surface. A survey of the com- are the apical (i.e., microvilli-bearing) processes of Müller
monly used immunolabeling procedures, and their results cells. The stem processes usually contain numerous mito-
on specific types of astroglial and ependymoglial cells, is chondria. An interesting exception are Müller cell processes
presented in Table 4.1. It should also be mentioned here in species with avascular retinae that contain mitochondria
that ependymoglial and astroglial cells have the capability only at their apical pole (i.e., close to the choroid, which is the
to accumulate exogenously applied fluorescent dyes such as only source of oxygen supply), whereas their stem processes
sulfo-rhodamine, Fura-2, MitoTracker Orange, and others. are devoid of these organelles. It has been argued that because
Thus, such dyes can be used to monitor Ca2+, glutathione, of their dominant glycolytic energy metabolism, Müller cells
pH, and so on, specifically in living glial cells, and simul- (and perhaps other astroglial cells) are free to move and place
taneously, to study their morphology. Microscopically con- their mitochondria toward sites of high pO2 (Germer et al.
trolled intracellular injections of fluorescent dyes can also 1998a,b; Wolburg et al. 1999) rather than to sites of high
be used to visualize individual glial cells. Finally, astroglial energy demand, as observed in the neurons with their domi-
cells can be induced to express the green fluorescent protein nant aerobic metabolism.
(GFP) or similar fluorescent proteins by coupling this gene The stem processes of astrocytes, tanycytes, and Müller
to a glia-specific promoter (e.g., for GFAP, GS, or GLAST) cells do not show the rather regular dichotomised branching
in transgenic mice. In any of these cases, fluorescent dyes are pattern characteristic of neuronal dendrites, but rather are the
desirable because they can be used with the high-resolution origins of specialized endings or side branches described in the
confocal microscopy. More about suitable staining proce- following sections.
dures in the case of Müller cells can be found in Reichenbach
and Bringmann (2010).
3.3 E N D FE ET
Astrocytic endfeet almost completely cover all basal lami-
3 U LT R A S T RU C T U R A L F E AT U R E S nae within the CNS (along the blood vessels, pia mater, and
vitreous body in the eye, here together with the Müller cell
endfeet). They are often coupled to each other by gap junc-
3.1 C E L L S O M A A N D N U C L EUS
tions (and sometimes by zonulae adherentes) (Iadecola and
The soma of astrocytes is usually rather poor in organelles if Nedergaard 2007; Tam and Watts 2010) constituting the
compared with that of neurons. In most cases, characteristic basis of dynamic interactions between astrocytes, neurons,

42 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
and vascular structures (Figley and Stroman 2011; Giaume et depend on the presence of agrin and the dystrophin–dystro-
al. 2010). In the case of tanycytes and Müller cells, the end- glycan complex (Noell et al. 2009, 2011). Agrin is a heparan
feet are densely filled with smooth endoplasmic reticulum. sulfate proteoglycan of the extracellular matrix, originally dis-
At the ventral surface of the rat brain, near the hypothala- covered as being essential for the clustering of acetylcholine
mus, astrocytes form lamellar stacks of astrocyte processes receptors in the postsynaptic membrane of the motor end-
(Holen 2011). Bundles of intermediate filaments extend into plate; it is also present around brain microvessel (Barber and
the endfeet, but fail to occupy the cytoplasm close to the Lieth 1997). Agrin also binds to α-dystroglycan, a member of
basal lamina–contacting endfoot membrane. These filaments the dystrophin–dystroglycan complex that localizes at glial
consist primarily of vimentin when the endfoot is in contact endfeet membranes in a similar way as OAP/AQP4 (Noell
with cerebrospinal fluid or vitreous humor and glial fibrillary et al. 2011).
protein when a blood vessel is contacted (Pixley and DeVellis When, under pathological or experimental condi-
1984); however, this rule may be modified in some cases. tions, apicolateral membranes of RPE cells, Müller cells,
With the exception of Müller cells in species with completely or ependymocytes are confronted with the mesenchymal
avascular retinae (i.e., missing both intraretinal and suprareti- compartment, they lose their original features and develop
nal blood vessels), the endfeet are rich in mitochondria. The an endfoot-like structure. This response suggests that mes-
occurrence of caveolae, coated pits, and vesicles concerned enchymal contact (i.e., collagen/laminin/agrin and/or other
with endo-/exo-/or pinocytosis indicates active material molecules) stimulates the insertion of potassium channels
exchange with the compartment behind the basal lamina of the Kir4.1 type (Ishii et al. 1997). Although astroglial
(i.e., blood plasma, vitreous body, or subarachnoidal fluid). A and ependymoglial cells are thought to contribute to basal
secretory function has been ascribed to the tanycytes of some lamina formation, this seems to require the presence of adja-
circumventricular organs. cent mesenchymal cells; mature Müller cells cannot restore
The most prominent and distinctive ultrastructural the vitreal basal lamina when it is destroyed by enzymatic
property of endfoot membranes of all astroglial cells in digestion (Halfter 1998) or microsurgical peeling (Miller
higher vertebrates is the occurrence of orthogonal arrays of et al. 1986).
intramembranous particles (OAPs) (Fig. 4.4F), which can be
visualized in the transmission electron microscope by means
of the freeze-fracture technique. The OAPs are accumulated 3.4 V E N T R I C U L A R C O N TAC T S
in those parts of the endfoot membrane that directly contact
the perivascular or superficial basal lamina, whereas within Cell processes contacting the ventricular (or subretinal)
the neuropil, there are few if any OAPs. This polarity devel- space occur in ependymoglial cells such as tanycytes and
ops concomitantly with the maturation of the blood-brain Müller cells, but never in astrocytes. They always display an
barrier, and is lost or severely reduced under pathological enlarged surface area, by extending microvilli into the fluid.
conditions such as tumors or inflammatory diseases, as well Furthermore, the apical pole contains abundant mitochon-
as in cultured glial cells (for a recent overview, see Wolburg
et al. 2011). The molecular identity of the particle-forming dria, features that indicate a high metabolic activity that is
protein(s) has been a matter of debate for many years, but now presumably related to an active exchange of substances with
the water channel–forming protein, aquaporin-4 (AQP4) is the luminal fluid.
considered the main constituent of the OAPs (Wolburg et al. Neighboring glial ventricular contact processes (and
2011). However, in zebrafish telencephalon, radial glial cells adjacent neuronal cell processes, if present) are connected by
have been described that express AQP4 without forming various types of apicollateral junctions. These (in particular,
arrays (Grupp et al. 2010), whereas generally, teleost glial cells desmosomes) are general “markers” of virtually all epithelial
can form OAPs (Wolburg et al. 2011). Still, the preconditions cells and occur very early in development. However, their
for OAP formation are not completely known. Noteworthy, nature varies considerably depending on the local microen-
in mammalian glial cells the inwardly rectifying K+ channel, vironment. In regions in which no endothelial blood-brain
Kir4.1, shows a very similar polarized distribution (Nagelhus barrier exists (e.g., in most circumventricular organs and the
et al. 1999) and may be another constituent of the OAPs,
but this remains to be demonstrated. An important role RPE) but not elsewhere, ependymoglial cells form a cerebro-
of OAP/AQP4 for the water homeostasis of the brain and spinal fluid–brain barrier by expressing tight junctions (Fig.
maintenance of the blood-brain barrier has been shown (e.g., 4.4E). Apicollateral gap junctions normally occur between
Amiry-Moghaddam et al. 2004; Benfenati and Ferroni 2010; Müller cells in frogs but not mammals. When, however, rab-
King et al. 2004; Tait et al. 2009; Wolburg et al. 2009; Yool bit Müller cells form homogeneous cultures in vitro, gap and
2007; Yukutake and Yasui 2010). even tight junctions can be observed (Wolburg et al. 1990).
Given that astroglial cells are highly polarized, it is an Likewise, when the basal pole of Müller cells is directly
important question how cellular polarity is organized at the exposed to the vitreous humor (because of basal lamina
molecular level. The mere morphological observation that the defects in retinal wounds) the cells may send microvilli into
occurrence (or at least density) of OAPs crucially depends on the vitreous cavity (Foos and Gloor 1975). Retinal pigment
basal lamina contact suggested a pivotal role of the extracellu- epithelium cells proliferating under areas of retinal detach-
lar matrix for the molecular arrangement of the endfoot mem- ment may lose their basal lamina contact and face the sub-
brane. Indeed, the OAP/AQP4-related polarity of astrocytes retinal fluid round about; in this case, their entire surface is
as well as the array formation from AQP4 tetramers seems to covered by microvilli (Anderson et al. 1983).

4 . A S T R O C Y T E S A N D E P E N DY M A L G L I A • 43
The apical surface of typical ependymocytes is character- intracellular Ca2+ rises in individual microdomains (Grosche et
ized by the presence of 12 to 60 kinocilia, which vary in num- al. 1999). Furthermore, mathematical simulation of the cable
ber according to the species. The cilia are 10 to 20 μm long, properties reveals that even large (e.g., glutamate-induced)
and are of the 9 x 2 + 2 type. These cilia beat rhythmically at depolarizations of the perisynaptic membranes are not con-
a frequency of about 200 per minute, and appear to assist the ducted over the stalks toward neighboring microdomains
rostro-caudad flow of cerebrospinal fluid. All these observa- or the stalk (Grosche et al. 2002). The energetic demands of
tions suggest that the polarity of ependymoglial cells strongly each individual microdomain may be supported by the mito-
depends on the microenvironment with which the different chondria found in the “head” structures (Grosche et al. 1999,
cellular surface domains are contacted. 2002). The glial microdomains overlap; in every given volume
unit of the molecular layer, at least two microdomains, origi-
nating from different Bergmann glial cells, interdigitate. This
3.5 L A M E L L A R N EU RO N- C O N TAC T P RO C E S S E S :
may fit with the observation that Purkinje cells express two
G L I A L M I C RO D O M A I N S
functionally distinct populations of synaptic spines, and indi-
The end branches of neuropilar astroglial cell processes are the vidual spines are capable of independent activation (Denk et
site of glia–neuron interactions. These processes are character- al. 1995), as the glial microdomains may be adjusted to meet
ized by the formation of flat or lamellar sheaths (Chao et al. this functional diversity of Purkinje cell synapses.
2002; Wolff 1968) that enclose neuronal somata, synapses, or
bundles of axonal internodes, or by fingerlike extensions that
contact the nodal membrane of myelinated axons (Fig. 4.2A). 4 I N T E R S P E C I E S VA R I AT I O N,
Such fingerlike astroglial processes also contact nodelike spe- O N TO G E N ET I C / P H YS I O L O G I C A L
cializations of unmyelinated axons in the retinal nerve fiber P L A S T I C I T Y, A N D H I E R A R C H Y
layer (Fig. 4.4G). They may origin not only from astrocytes OF ASTROGLIA
but also from Müller cells (Reichenbach et al. 1988) and from
the NG2+ cells. In contrast to the stem processes, the lamel- Despite of displaying a very complex structure and ultra-
lae are virtually devoid of organelles (with the exception of structure (Figs. 4.1 to 4.4), it is now clear that astroglial cell
actin filaments), but contain ezrin and radixin (Derouiche processes are by no means unchangeable, static structures.
and Frotscher 2001), two actin-binding proteins that link After the general shape of an astroglial cell has been estab-
the cell membrane to the actin cytoskeleton and may medi- lished in ontogenesis (i.e., after at least one endfoot-bearing
ate the formation and stabilization of the very complex, thin stem process has grown), the neuropilar processes, particu-
side branches with their large surface-to-volume ratios of up larly the lamellar perineuronal sheaths, develop in (mutual)
to more than 20 μm–1 (see Fig. 4.3). Whereas generally a large dependence on the developing neuronal cell processes and
part of all neuronal compartments is ensheathed by such lamel- synapses (Reichenbach et al. 2010). Furthermore, it has been
lar processes, the degree (or even the presence) of ensheathing shown that there occurs a lifelong, activity-dependent plastic-
may vary considerably even within the same area of the CNS. ity of glial cell processes (Henneberger et al. 2010; Hirrlinger
There is an obvious tendency to ensheathe the synapses. In rat et al. 2004). It should be kept in mind, however, that most of
neocortex for example, about 56% of all synaptic perimeters the available data were obtained from studies on small labo-
are covered by astroglia, whereas astroglial membranes make ratory rodents, often at very young stages (to facilitate work
up only 22% of all membranes in the neuropil (Chao et al. on slice preparations) and must not necessarily apply to adult
2002 and references therein). However, on synaptic glomeruli humans. Thus, some comparative and developmental aspects
or “complex synapses” (i.e., specialized subcortical structures are discussed here.
in which multiple synaptic junctions are enclosed in a com-
mon glial sheath) the glial coverage is very high (there are even
4.1 G E N ET I C VE R S US A DA P T I VE C O N T RO L
multilamellar sheaths) but the glia does not penetrate the
O F A S T RO G L I A L C E L L S I Z E
interior of the complexes, and thus cannot seal the individual
synaptic clefts. As an extreme, there are astroglia-free neuropil It has been pointed out that the total size of a mammal cor-
compartments, such as in Rolando’s substantia gelatinosa of relates with the size of its brain as well as with the size of
the spinal cord and the cochlear nucleus, in which thin sensory its neurons (Purves 1988). This has been explained by the
axons terminate in “synaptic nests” that lack intrinsic glia. facts that in a big body: (1) more cells exist and need inner-
In Bergmann glial cells, the existence of microdomains has vation (which requires increasing numbers of neurons and
been demonstrated (Grosche et al. 1999, 2002). These occur of axon collaterals), and (2) the nerves have to bridge larger
as “repetitive units” on the stem processes (or as appendages of distances (which requires an increase of not only length but
another microdomain). Each glial microdomain consists of a also thickness of axons, to accelerate impulse propagation,
thin stalk and a cabbagelike, very complex head structure that and enforces an increasing size of the somata to maintain
bears the lamellar perisynaptic sheaths for about five synapses the voluminous axons). In parallel, not only the absolute
(see Fig. 4.3). It has been shown that these microdomains and relative number of glial cells (Reichenbach 1989b)
may interact with “their” (about five) synapses, independent but also their size increases (Fig. 4.5B, C). This appears to
of each other, and also of the stem process. Stimulation of be genetically controlled, at least partially. One example is
the axons ending in the ensheathed synapses causes transient constituted by animals (e.g., lungfish and some amphibians)

44 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
with polyploidy. The increasing DNA content causes not 4.3 VA R I A B I L IT Y A N D H I E R A RC H Y
only an enlargement of the cell nuclei but also of the cells O F G L I A L D O M A I NS
themselves (Fig. 4.5A). This has been used by electrophysi- It has been shown that astroglial cell processes may form
ologists because a large size facilitates recordings from a cell. repetitive microdomains, each interacting (more or less auton-
Another recent argument was provided by Nedergaard and omously) with a small group of synapses (Grosche et al. 1999)
colleagues, who transplanted human astrocytes into mouse (see Fig. 4.3). It has also been speculated that the cell processes
brains and found that the human astrocytes were much larger of each astrocyte occupy a neuropilar territory that constitutes
than the neighboring murine host astrocytes (Nedergaard the domain of its (autonomous) interactions with neuronal
2011) (see chapter 28). With these few exceptions, it is hard (and vascular) compartments. In several studies, the territories
to say how much of the different size of astrocytes in dif- of neighboring astrocytes were found to abut each other with
ferent animals (Fig. 4.5C) results from genetic instruction minimal overlap (Bushong et al. 2002, 2004; Wilhelmsson
or adaptation to the environment. For instance, cerebellar et al. 2006). It has also been demonstrated that in fact, indi-
Bergmann glial cells have to span the entire thickness of the vidual astrocytes may specifically interact with the neuronal
molecular layer. Depending on the number and thickness synapses within their territory (Henneberger et al. 2010).
of neuronal cell processes (see the preceding, and Purves These individual domains are competitively organized during
1988), this layer is much thinner in small animals than in early postnatal development (Bushong et al. 2004) and may
larger ones. Accordingly, the Bergmann glial cell processes even be demarked by molecules such as chondroitin sulfate
are much elongated in large species (Fig. 4.5B). Of note, the (Horii-Hayashi et al. 2010). However, electron microscopi-
brain and its layers are growing during ontogenetic develop- cal studies have observed a substantial overlap of astrocytic
ment, such that young Bergmann glial cells of a large species domains in murine cerebellum (Grosche et al. 1999, 2002) as
may be smaller than adult cells from a small species (Hanke well as in rat cortex (Wolff 1968). This may depend on the spe-
and Reichenbach 1987). The adaptive growth of astroglial cies and/or brain compartment studied, but also on late post-
cell processes is discussed in the following. natal development. We have found that the domains of murine
cortical astrocytes do not overlap in young (2-month-old) ani-
mals, but show considerable overlap (with a factor of about 2)
4 .2 O N TO G E N ET I C D EV E L O PM E N T in 2-year-old animals (Fig. 4.5E).
A N D A D U LT P L A S T I C I T Y O F A S T RO G L I A L Generally, it is clear that neither the microdomains nor the
CELL SIZE AND SHAPE cellular domains are always autonomously interacting with their
The number, length, and complexity of astrocytic side neuronal partners. Astrocytes form gap junction–coupled syn-
branches grow rapidly during the early ontogenesis (Grosche cytial networks (see chapters 24 and 26) such that rather large
et al. 2002; Hanke and Reichenbach 1987; Senitz et al. 1995) numbers of cells may form bigger, multicellular domains that
(Fig. 4.5D). This may be triggered by the overall growth of concertedly interact with the neurons in this territory. Recently,
the neural tissue, ingrowth of blood vessels, and specific neu- we have proposed a concept of variable, hierarchically organized
ron–glia interactions (for an overview of such interactions, domains at many levels, from substructures at individual synapses
see Figley and Stroman 2011; Giaume et al. 2010). Specialized (nanodomains) to entire cortical columns or even areas (macro-
glia–neuron contacts cannot be elaborated until neurons domains) (Reichenbach and Wolburg, 2009). Depending on
have completed their differentiation (Waxman et al. 1983). the degree and distribution of neuronal excitation, the whole
Indeed, the formation of such processes may be triggered by repertoire of glial arrangements from nanodomains and micro-
the onset of neuronal activity, and their growing filopodia domains up to macrodomains or even “superdomains” (cortical
may be attracted (or repelled) by signals from active neurons gyri or fields) may be switched on in series, within the very same
(e.g., K+ ions, neurotransmitter molecules, and/or growth part of the CNS (Fig. 4.6). At the (pathological) end of this
factors). The performance of these mechanisms during onto- scale, phenomena such as spreading depression may spread over
genesis may be modulated by the strength and/or pattern of the entire cortex, thus involving the entire glial population.
neuronal activity that, in turn, is triggered by sensory inputs At the end of this chapter, the authors hope to leave the
and behavioral requirements. For instance, dependent on reader with the view that much of the morphological diversity
whether rats are kept in enriched environments (Sirevaag of astrocytes and ependymoglial cells results from the differ-
and Greenough 1991) or complete darkness (Stewart et al. ent local microenvironments into which a given cell is born (or
1986) the complexity of their glial cell processes may differ migrating). Inevitably, mesenchymal contact induces the for-
significantly. The same (or very similar) mechanisms seem mation of endfeet with OAP-rich membranes, whereas contact
to be maintained in the mature CNS, in which they may to the cerebrospinal fluid induces the outgrowth of microvilli,
modify the structure of glial cells even in the short-term and the formation of stabilizing cell–cell junctions. Where
range (Henneberger et al. 2010, Hirrlinger et al. 2004) so neuronal elements are contacted, the glial cells form delicate
as to meet the changing needs of their neuronal partners. A side branches that end in lamellar sheaths or fingerlike branch-
recent discussion of adult astroglial plasticity and the tech- lets. The number, size, and shape of these glial “end structures”
nical problems to monitor it unequivocally can be found in are precisely adjusted to the morphological and functional
Reichenbach et al. (2010). The role of astrocytic plasticity features of the adjacent neuronal elements. This adjustment
in the modulation of neuroendocrine systems is discussed continues after ontogeny as a lifelong process of plasticity. In
further in chapter 41. addition, rapid, reversible changes may occur that involve both

4 . A S T R O C Y T E S A N D E P E N DY M A L G L I A • 45
A Müller cells: carp frog C 50 μm
salamander protoplasmic
astrocytes:

mouse man
50 μm

D human cortical
teraploid diploid teraploid diploid astrocytes:
neonatal
B

miniature 50 μm
shrew
adult
rat

Bergmann glial cells:

overlap factor of
E territories:

cortex
young mouse
ca.1

100 μm cortex
man monkey old mouse>>1 50 μm

Figure 4.5 The size of (astro-)glial cells and their occupied territories depends on several factors. A. As exemplified by Müller cells of fish and amphibian
retinae, a species-specific tetraploidy (i.e., three sets of chromosomes) causes not only an enlargement of the cell nucleus, but also of the entire cell.
B,C. As exemplified by cerebellar Bergmann glial cells (B) and brain astrocytes (C), bigger animal species not only have bigger brains, but also bigger
glial cells. D. As shown for human cortical astrocytes, there occurs a significant postnatal increase of the number, length, and complexity of cell
processes. E. The overlap factor (OF) of neuropilar territories occupied by the processes of astroglial cells may vary with age. In young animals the
territories may just touch each other (OF ~1), but in aged mice considerable overlap of astrocytic territories (OF ~2) was found (own unpublished
data). (A,B,D) Modified after Reichenbach (1989a) and Reichenbach and Wolburg (2005), in which the references also can be found; (C) from
Oberheim et al. 2006, with permission.

the neurovascular unit. One important task of future glial


morphological alterations of glial cell processes and a modifica-
cell research will be to elucidate the molecular interactions
tion of the gap junction–mediated coupling of the astrocytic
between glial cells and blood vessels with similar emphasis
networks, allowing for fast switches among subcellular, cellular,
as is currently dedicated to the glial contributions to synap-
and multicellular glio–neuronal interactions. Further studies
tic and cognitive processes. For instance, an increase of brain
of astrocytic plasticity at all these levels are supposed to provide
capillary permeability must not necessarily be restricted to
novel insights into the complexity of astrocytic functions.
pathological processes (stroke, inflammation, tumor, neuro-
degenerative diseases), but may give the healthy brain access
to hematogenous factors in the circulation, so as to modulate
5 S U M M A RY A N D P E R S P E C T I VE S neuronal circuits. Another promising future research area will
be the biomechanics of brain cells and tissue, because it has
The multitude of glial cell types in all areas of the CNS reflects been shown that the viscoelastic properties of neurons, glial
the wealth of glial roles, including such crucial functions as, cells, and the extracellular matrix are important for the growth
for instance, guidance in the development of cortical lay- of neural cell processes. Summarizing the current state of
ering, regulation of extracellular homeostasis, support of (functional-) morphological knowledge given in this chapter,
neuronal energy and neurotransmitter metabolism, and reg- the authors want to leave the reader with the view that much
ulatory processes at the blood-brain barrier. This last example of the morphological diversity of astrocytes and ependymo-
underlines that astrocytes do not only interact with neurons glial cells results from the different local microenvironments
and other glial cells, but also with vascular structures, forming into which a given cell is born (or migrates).

46 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
I mesodomain
Anderson DH, Stern WH, Fisher SK, Erickson PA, Borgula GA. 1983.
Retinal detachment in the cat: the pigment epithelial-photoreceptor
II I interface. Invest Ophthalmol Vis Sci 2:906–926.
II
Barber AJ, Lieth E. 1997. Agrin accumulates in the brain microvascu-
ni lar basal lamina during development of the blood-brain barrier. Dev
cellular Dynamics 208:62–74.
domain 1 III III
BefenatiV, Ferroni S. 2010. Water transport between CNS compart-
ments: functional and molecular interactions between aquaporins
and ion channels. Neuroscience 168:926–940.
IV Bushong EA, Martone ME, Ellisman MH. 2004. Maturation of
microdomain
IV astrocyte morphology and the establishment of astrocyte domains
n1 V during postnatal hippocampal development. Int J Dev Neurosci
nii 22:73–86.
VI
networks Bushong EA, Martone ME, Jones YZ, Ellisman MH. 2002. Protoplasmic
V
astrocytes in CA1 stratum radiatum occupy separate anatomical
nano- VI macrodomain domains. J Neurosci 22:183–192.
cellular Chao TI, Rickmann M, Wolff JR. 2002. The synapse-astrocyte bound-
domain
domain 2 ary: anatomical basis for an integrative role of glia in synaptic trans-
superdomain mission. In: Volterra A, Magistretti P, Haydon P (eds.), Tripartite
“neuron” striate cortex occipital cortex synapses: synaptic transmission with glia. Oxford, England: Oxford
University Press, pp. 3–23.
Figure 4.6 Schematic Representation of the Various Types of Coexisting Colombo JA. 2001. A columnar-supporting mode of astroglial archi-
Glial Domains Showing the Transition (Arrows) Between the Striate and tecture in the cerebral cortex of adult primates? Neurobiology
the Occipital Cortex in a Human Brain. At increasing levels of spatial 9:1–16.
organization, the glial cells provide nanodomains up to superdomains Denk W, Sugimori M, Llinas R. 1995. Two types of calcium response
for their interaction with neurons and blood vessels. With the possible limited to single spines in cerebellar Purkinje cells. Proc Natl Acad
exception of the nanodomains, which probably interact with “their” Sci U S A 92:8279–8282.
neuronal partner structures as long as they exist, the domains are not Derouiche A, Frotscher M. 2001. Peripheral astrocyte processes: moni-
only determined by the (ultra-)structural features of the glial cells, but toring by selective immunostaining for the actin-binding ERM pro-
also by the properties of the signal (or stimulus): (I) (a few) individual teins. Glia 36:330–341.
synapses are associated with their ensheathing glial microdomains but Distler C, Dreher Z, Stone J. 1991. Contact spacing among astrocytes
parallel/strong stimulation of related inputs may finally integrate; and in the central nervous system: an hypothesis of their structural role.
(II) (?oligo-)cellular domains involving the whole (or a few) glial cell and Glia 4:484–494.
their neuronal partners (see Fig. 4.4). It depends on the shape of the glial Feig SL, Haberly LB. 2011. Surface-associated astrocytes, not endfeet,
cells involved whether these domains are columnar (“type 1,” for example, form the glia limitans in posterior piriform cortex and have a spa-
Bergmann glial cells; probably radial astrocytes in hippocampal stratum tially distributed, not a domain, organization. J Comp Neurol
oriens, and hypothetically interlaminar astrocytes in primate cortex), or 519:1952–1969.
rather spherical or ellipsoid (“type 2,” star-shaped astrocytes). Appropriate Figley CR, Stroman PW. 2011. The role(s) of astrocytes and astrocyte
(i.e., strong, frequent, or synchronized) stimulation may then activate, via activity in neurometabolism, neurovascular coupling, and the pro-
gap-junctional coupling, networks consisting of more than 50 astrocytes duction of functional and neuroimaging signals. Eur J Neurosci
(types n1 and n2 depend on the shape of the constituting glial cells). 33:577–588.
These networks, however, cannot be considered as static; any member Foos RY, Gloor BP. 1975. Vitroretinal juncture: healing of experimental
of the coupled network at its margin is per se coupled to other cells that wounds. Albr Graefes Arch klin exp Ophthalmol 196:213–230.
are outside the first coupling range but that would be belong to another Gerhardt H, Rascher G, Schuck J, Weigold U, Redies C, Wolburg H.
(overlapping) network if the dye would have been injected into such a 2000. R- and B-cadherin expression defines subpopulations of glial
cell. Thus, if a neuronal stimulus arrives at such a cell later, or if a Ca2+ cells involved in axonal guidance in the optic nerve head of the
wave was triggered by the first “excited” astrocyte, (III) macrodomains chicken. Glia 31:131–143.
will develop; this mechanism may proceed either radially (n1–nii) or Germer A, Biedermann B, Schousboe A, Wolburg H, Mack A,
tangentially (n1–ni). The size of these macrodomains may vary from small Reichenbach A. 1998a. Distribution of mitochondria within Müller
(macrodomain 1, corresponding for example to the orientation columns cells. I. Correlation with retinal vascularization in different mamma-
of Mountcastle 1957) to large (e.g., ocular dominance columns or barrel lian species. J Neurocytol 27:329–345.
fields; macrodomain 2). A further progression of integration will result Germer A, Wolburg H, Mack A, Reichenbach A. 1998b. Distribution of
in the generation of very large functional units. (IV) superdomains, mitochondria within Müller cells. II. Post-natal development of the
corresponding to entire cortical areas or gyri. Eventually, even whole rabbit retinal periphery in vivo and in vitro: dependence on oxygen
hemispheres associated with huge astrocytic populations may transiently supply. J Neurocytol 27:347–359.
be involved, putatively mediating events such as spreading depression, Giaume C, Koulakoff A, Roux L, Holcman D, Rouach N. 2010.
seizures, and/or widespread neuronal degeneration. I–VI, layers of the Astroglial networks: a step further in neuroglial and gliovascular
cortex. From Reichenbach and Wolburg 2009, with permission. interactions. Nature Rev Neurosci 11:87–99.
Götz M, Huttner WB. 2005. The cell biology of neurogenesis. Nature
Rev Mol Cell Biol 6:777–788.
Grosche J, Kettenmann H, Reichenbach A. 2002. Bergmann glial cells
form distinct morphological structures to interact with cerebellar
neurons. J Neurosci Res 68:138–149.
REFERENCES Grosche J, Matyash V, Möller T, Verkhratsky A, Reichenbach A,
Kettenmann H. 1999. Microdomains for neuron-glia interaction:
Amiry-Moghaddam M, Frydenlund DS, Ottersen OP. 2004. Anchoring parallel fiber signaling to Bergmann glial cells. Nature Neuroscience
of aquaporin-4 in brain: molecular mechanisms and implications for 2:139–143.
the physiology and pathophysiology of water transport. Neuroscience Grupp L, Wolburg H, Mack AF. 2010. Astroglial structures in the
129:999–1010. zebrafish brain. J Comp Neurol 518:4277–4287.

4 . A S T R O C Y T E S A N D E P E N DY M A L G L I A • 47
Halfter W. 1998. Disruption of the retinal basal lamina during early Purves D. 1988. Body and brain. A trophical theory of neural connec-
embryonic development leads to retraction of vitreal endfeet, an tions. Cambridge, MA: Harvard University Press.
increased number of ganglion cells, and aberrant axonal growth. Reichenbach A. 1989a. Attempt to classify glial cells by means of their
J Comp Neurol 397:89–104. process specialization using the rabbit retinal Müller cell as an exam-
Hanke S, Reichenbach A. 1987. Quantitative-morphometric aspects of ple of cytotopographic specialization of glial cells. Glia 2:250–259.
Bergmann glial (Golgi epithelial) cell development in rats. A Golgi Reichenbach A. 1989b. Glia:neuron index: review and hypothesis to
study. Anat Embryol 177:183–188. account for different values in various mammals. Glia 2:71–77.
Henneberger C, Papouin T, Oliet SH, Rusakov DA. 2010. Long-term Reichenbach A, Bringmann A. 2010. Müller cells in the healthy and dis-
potentiation depends on release of D-serine from astrocytes. Nature eased retina. New York: Springer.
463:232–236. Reichenbach A, Derouiche A, Kirchhoff F. 2010. Morphology and
Hirrlinger J, Hülsmann S, Kirchhoff F. 2004. Astroglial processes dynamics of perisynaptic glia. Brain Res Rev 63:11–25.
show spontaneous motility at active synaptic terminals in situ. Eur J Reichenbach A, Schippel K, Schümann R, Hagen E. 1988. Ultrastructure
Neurosci 20:2235–2239. of rabbit nerve fibre layer—neuro-glial relationships, myelination,
Holen T. 2011. The ultrastructure of lamellar stack astrocytes. Glia and nerve fiber spectrum. J Hirnforsch 29:481–491.
59:1075–1083. Reichenbach A, Wolburg H. 2005. Astrocytes and ependymal glia. In:
Horii-Hayashi N, Tatsumi K, Matsusue Y, Okuda H, Okuda A, Hayashi Kettenmann H, Ransom BR (eds.), Neuroglia, 2nd ed. New York:
M, et al. 2010. Chondroitin sulfate demarcates astrocytic territories Oxford University Press, pp. 19–35.
in the mammalian cerebral cortex. Neurosci Letters 483:67–72. Reichenbach A, Wolburg H. 2009. Structural association of astrocytes
Howard BM, Mo Z, Filipowic R, Moore AR, Antic SD, Zecevic N. with neurons and vasculature: defining territorial boundaries. In:
2008. Radial glia cells in the developing human brain. Neuroscientist Parpura V, Haydon PG (eds.), Astrocytes in (patho-) physiology of
14:459–473. the nervous system. Boston: Springer, pp. 245–280.
Iadecola C, Nedergaard M. 2007. Glial regulation of the cerebral micro- Schuck J, Gerhardt H, Wolburg H. 2000. The peripapillary glia of the
vasculature. Nature Neurosci 10:1369–1376. optic nerve head in the chicken retina. Anat Rec 259:263–275.
Ishii M, Horio Y, Tada Y, Hibino H, Inanobe A, Ito M, et al. 1997. Senitz D, Reichenbach A, Smith TG Jr. 1995. Surface complexity of
Expression and clustered distribution of an inwardly rectifying potas- human neocortical astrocytic cells: changes with development, aging,
sium channel, K AB-2/Kir4.1, on mammalian retinal Müller cell mem- and dementia. J Brain Res 36:531–537.
brane: Their regulation by insulin and laminin signals. J Neurosci Sirevaag AM, Greenough WT. 1991. Plasticity of GFAP-immunoreactive
17:7725–7735. astrocyte size and number in visual cortex of rats reared in complex
King LS, Kozono D, Agre P. 2004. From structure to disease: the evolving environments. Brain Res 540:273–278.
tale of aquaporins biology. Nature Rev Mol Cell Biol 5:687–698. Stewart MG, Bourne RC, Gabbott PLA. 1986. Decreased levels of an
Lu Y-B, Franze K, Seifert G, Steinhäuser C, Kirchhoff F, Wolburg H, astrocytic marker, glial fibrillary acidic protein, in the visual cortex
et al. 2006. Viscoelastic properties of individual glial cells and neu- of dark-reared rats: measurement by enzyme-linked immunosorbent
rons in the CNS. Proc Natl Acad Sci U S A 193:17759–17764. assay. Neurosci Letts 63:147–152.
Mathiisen TM, Lehre KP, Danbolt NC, Ottersen OP. 2010. The peri- Stolzenburg J-U, Reichenbach A, Neumann M. 1989. Size and density
vascular astroglial sheath provides a complete covering of the brain of glial and neuronal cells within the cerebral neocortex of various
microvessels: an electron microscopic 3D reconstruction. Glia insectivorian species. Glia 2:78–84.
58:1094–1103. Strauss O. 2005. The retinal pigment epithelium in visual function.
Miller B, Miller H, Patterson R, Ryan SJ. 1986. Retinal wound healing. Physiol Rev 85:845–881.
Cellular activity at the vitreoretinal interface. Arch Ophthalmol Sturrock RR, Smart JL, Dobbing J. 1977. Effect of undernutrition dur-
104:281–285. ing the suckling period on the indisium griseum and rostrat part
Mountcastle VB. 1957. Modality and topographic properties of of the mouse anterior commissure. Neuropathol Appl Neurobiol
single neurons of cat’s somatic sensory cortex. J Neurophysiol 3:369–375.
20:408–434. Tait MJ, Saadoun S, Bell BA, Papadopoulos MC. 2008. Water move-
Nagelhus E A, Horio Y, Inanobe A, Fujita A, Haug FM, Nielsen S, et ments in the bran: role of aquaporins. Trends Neurosci 31:37–43.
al. 1999. Immunogold evidence suggests that coupling of K+ siphon- Tam SJ, Watts RJ. 2010. Connecting vascular and nervous system devel-
ing and water transport in rat retinal Müller cells is mediated by a opment: angiogenesis and the blood-brain barrier. Ann Rev Neurosci
coenrichment of Kir4.1 and AQP4 in specific membrane domains. 33:379–408.
Glia 26:47–54. Waxman SG, Black JA, Foster RE. 1983. Ontogenesis of the axolemma
Nedergaard M. 2011. Complexity of human astrocytes. Plenary Lecture and axoglial relationships in myelinated fibers: electrophysiologi-
at the 10th European Meeting on Glial Cells in Health and Disease. cal and freeze-fracture correlates of membrane plasticity. Int Rev
Glia 59:S6. Neurobiol 24:433–484.
Noell S, Fallier-Becker P, Deutsch U, Mack AF, Wolburg H. 2009. Agrin Wilhelmsson U, Bushong EA, Price DL, Smarr BL, Phung V, Terada M,
defines polarized distribution of orthogonal arrays of particles in et al. 2006. Redefining the concept of reactive astrocytes as cells that
astrocytes. Cell Tissue Res 337:185–195. remain within their unique domains upon reaction to injury. Proc
Noell S, Wolburg-Buchholz K, Mack AF, Beedle AM, Satz JS, Campbell Natl Acad Sci U S A 103:17513–17518.
KP, et al. 2011. Evidence for a role of dystroglycan regulating Wolburg H, Liebner S, Reichenbach A, Gerhardt H. 1999. The pecten
the membrane architecture of astroglial endfeet. Eur J Neurosci oculi of the chicken: a model system for vascular differentiation and
33:2179–2186. barrier maturation. Int Rev Cytol 187:111–159.
Oberheim NA, Takano T, Han X, He W, Lin JHC, Wang F, et al. 2009. Wolburg H, Noell S, Mack A, Wolburg-Buchholz K, Fallier-Becker P.
Uniquely hominid features of adult human astrocytes. J Neurosci 2009. Brain endothelial cells and the glio-vascular complex. Cell
29:3276–3287. Tissue Res 335:75–96.
Oberheim NA, Wang X, Goldman S, Nedergaard M. 2006. Astrocytic Wolburg H, Paulus W. 2010. Choroid plexus: biology and pathology.
complexity distinguishes the human brain. Trends Neurosci Acta Neuropathol 119:75–88.
29:547–553. Wolburg H, Reichelt W, Stolzenburg J-U, Richter W, Reichenbach
Pixley SKR, DeVellis J. 1984. Transition between immature radial glia A. 1990. Rabbit retinal Müller cells in cell culture show gap and
and mature astrocytes studied with a monoclonal antibody to vimen- tight junctions which they do not express in situ. Neurosci Letters
tin. Dev Brain Res 15:201–209. 11:58–63.

48 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
Wolburg H, Wolburg-Buchholz K, Fallier-Becker P, Noell S, Mack A. Wolff J. 1968. The role of astroglia in the brain tissue. Acta Neuropathol
2011. Structure and functions of aquaporin-4-based orthogonal (Suppl IV):33–39.
arrays of particles. Int Rev Cell Mol Biol 287:1–41. Yool AJ. 2007. Aquaporins: multiple roles in the central nervous system.
Wolburg H, Wolburg-Buchholz K, Mack AF, Reichenbach A. 2009. Neuroscientist 13:470–485.
Ependymal cells. In: Squire L (ed.), The new encyclopedia of neuro- Yukutake Y, Yasui M. 2010. Regulation of water permeability through
science. Oxford, England: Academic Press, pp. 1133–1140. aquaporin-4. Neuroscience 168:885–891.

4 . A S T R O C Y T E S A N D E P E N DY M A L G L I A • 49
5.
RADIAL GLIAL CELLS
Magdalena Götz

A B B R E VI AT I O N S A B C

Aldh1L1 aldehyde dehydrogenase 1 family, member L1


B-FABP brain-fatty acid binding protein
BLBP brain lipid binding protein
BM basement membrane
BMP bone morphogenic protein
ChSPG heparan sulphate proteoglycans
CNS central nervous system
CSF cerebrospinal fluid
ECM extracellular matrix
FACS fluorescence-activated cell sorting
FGFR fibroblast growth factor receptor
GFAP glial-fibrillary acidic protein
GFP green fluorescent protein Adherens Basal Body
GLAST glutamate transporter astrocyte-specific D junction of cilium
GLT-1 glutamate transporter 1
GS glutamine synthase
IGF insulin-like growth factor
3-PGDH 3-phosphoglycerate dehydrogenase
SCO subcommissural organ
E β-catenin
SHH sonic hedgehog
SEZ subependymal zone F-Actin α-catenin
SVZ subventricular zone Cadherin G-Actin
TN-C Tenascine-C
WNT released proteins equivalent to the wingless and Cilium with
Par
INT1 proteins in Drosophila Prominin
Complex
Apical/Ventricular surface

Figure 5.1 Radial Glial Cell Polarity. A. Schematic drawing depict-


1 D E F I N I T I O N A N D O VE RVI EW ing the epithelial nature of radial glial cells with their main contacts at
the apical and basal side. B. Freeze-fracture depicting radial glia endfeet
Radial glial cells were first identified in the embryonic chick at the pial surface from a mouse embryo. C. Schematic drawing of the
molecular composition of the radial glia endfeet attachment to the
spinal cord (Bentivoglio and Mazzarello 1999) and named basement membrane underlying the meninges. Note the anchoring
according to their elongated radial morphology. As their name of the radial glia endfoot to the basement membrane components by
implies, besides their radial morphology “radial glial cells” are dystroglycan and integrin complexes. D. Electron microscopy depict-
also glial in nature. Radial glial cells are widespread in the ing the junctional complex linking radial glial cells at the apical side.
developing central nervous system (CNS) of all vertebrates E. Schematic drawing of the molecular composition of apical adherens
junction complexes and their dynamic anchoring to the cytoskeleton
examined so far and are the first glial cell type to develop per- by shuttling of α-catenin. F-actin, filamentous actin; G-actin, globular
sisting to different extent into adulthood. They line the ven- actin. (A) Courtesy of Hartwig Wolburg, University of Tübingen, in
tricle and extend their radial processes throughout the entire collaboration with Benedicte Dehouc, University of Lille; (D) courtesy
radial thickness of the neural tube (Fig. 5.1). Radial glial cells of Michaela Wilsch-Bräuninger and Wieland B. Huttner.
are true epithelial cells with an apical side in contact with the
ventricle and a basal endfoot connected to the basement mem- These junctions delineate the apical membrane domain fac-
brane (BM). Radial glial cells are coupled with neighboring ing the ventricular lumen with characteristic sorting of apical
cells by gap junctions as well as adherens or tight junctional membrane components (e.g., the glycoprotein prominin1) to
complexes (Götz and Huttner 2005; Pinto and Götz 2007). this compartment and a distinct basolateral membrane domain

50
Table 5.1a COMPARISON BETWEEN RADIAL GLIAL CELLS AND OTHER CELL TYPES IN THE MAMMALIAN BRAIN
NEURO-
EPITHELIAL RADIAL GLIA RADIAL GLIA MATURE EPENDYMAL REACTIVE ADULT NEURAL
PROTEIN CELLS EARLY LATE ASTROGLIA CELL ASTROGLIA STEM CELL

GFAP – –/+ +/++ –/++ + +++ +++

GLAST(Slc1a3) – ++ ++ +++ ++ +++ ++

GLT1(Slc1a2) – – + +++ ++ ++ ++

Glutamine – – + +++ +++ ++


synthetase

S100-β – – + ++ +++ +++ +

Connexin 43 – + ++ +++ ++ +++ +++


(Gja1)

Aquaporin 4 Nd Nd Nd +++ + +++ ++

KIR 4.1/2.1 – + ++ +++ ++ ++ +++

Aldlhl1 – – + +++ ++ +++ +

Nestin +++ +++ +++ – ++ +++ +++


(RC1/RC2)

Vimentin – + ++ – +++ +++ +++

BLBP ++ +++ +++ – – +++ +++

TN-C – +++ +++ – – +++ ++

Sox2 +++ +++ +++ +++ ++ +++ +++


Phosphacan/ – +++ +++ – – ++ +++
DSD-1

Note that cell heterogeneity is not incorporated in this table.


Based on Doetsch et al. 1997; Pinto and Götz, 2007; Mori et al. 2005; Liu et al. 2006 and references therein.

containing typically integrins in contact with the BM or extra- as GLAST and GLT-1, as well as glutamine synthase (GS),
cellular matrix components (Götz and Huttner 2005). S100β, and metabolic enzymes that are shared in expression
Besides their radial morphology, these cells also possess between astroglia and radial glia (Beckervordersandforth
glial hallmarks. These distinguish radial glial cells from the et al. 2010; Lovatt et al. 2007; Pinto et al. 2008), including
earlier epithelial cell type in the developing neural tube, the 3-phosphoglycerate dehydrogenase (3-PGDH), an essen-
neuroepithelial cells (Table 5.1) (Götz and Huttner 2005). tial enzyme for L-serine biosynthesis (Yamasaki et al. 2001),
The transition between neuroepithelial cells and radial glia is the aldehyde dehydrogenase family member AldhL1 (Cahoy
a gradual process with some hallmarks coming up earlier than et al. 2008) or brain lipid–binding protein (BLBP, also
others (see Table 5.1). The glial features shared between radial B-FABP). Significantly, radial glial cells share particularly
glia and other glial cells comprise ultrastructural, cell biolog- many aspects with activated or reactive astrocytes, which often
ical, and molecular aspects (Götz and Huttner 2005; Pinto reactivate the expression of proteins previously downregu-
and Götz 2007). Radial glial cells contain glycogen granules lated during maturation. These comprise nestin, vimentin, the
and a high density of 9-nm intermediate filaments reminiscent extracellular matrix protein tenascin-C, BLBP, and others
of astrocytes, especially in their basal endfeet (Pinto and Götz (see Table 5.1). Thus, among the “typical” astroglial proteins,
2007). These filaments comprise nestin, vimentin, and in no difference can be detected between activated astrocytes
many species the glia-fibrillary acidic protein (GFAP), a hall- after brain injury and radial glial cells in the developing brain
mark of many, although not all, astrocytes in the adult brain (see Table 5.1). Genomewide expression analysis, however,
(see chapter 4; Table 5.1). Although GFAP is low in rodent yields important differences (Beckervordersandforth et al.,
radial glial cells increasing only at the time of astrocyte genera- 2010) (see chapter 28) also related to profound differences in
tion from these cells, it is already prominent in radial glial cells function as detailed in the following.
at embryonic stages in developing brains of many other verte- Radial glia also share many hallmarks with ependymal
brates (Mori et al. 2005). In addition, radial glial cells contain cells, such as their contact to the ventricle, glycogen storage,
also astrocyte-specific glutamate-aspartate transporters, such and the expression of the described proteins, which are not

R ADIAL GLIAL CELLS • 51


Table 5.1b COMPARISON BETWEEN RADIAL GLIAL CELLS AND OTHER CELL TYPES IN THE MAMMALIAN BRAIN
ADULT
NEUROEPITHELIAL RADIAL RADIAL MATURE EPENDYMAL REACTIVE NEURAL
FUNCTION CELLS GLIA EARLY GLIA LATE ASTROGLIA CELL ASTROGLIA STEM CELL

Glutamate uptake – + ++ +++ ++ +++ +++

K-conductance – – ++ +++ ++ ++ ++
at rest

Glycogen storage – + ++ +++ +++ +++ ++

Gap-junctions/ Nd +++ +++ +++ ++ ++ +++


Hemichannels/
Ca-waves

Blood vessel – + ++ +++ – +++ +++


contact/blood flow
regulation

Cell division +++ +++ ++ – – ++ ++

Multipotency +++ +++ ++ – – + +++

Self-renewal ++ ++ + – – + +++

Based on Doetsch et al. 1997; Pinto and Götz, 2007; Mori et al. 2005; Liu et al. 2006 and references therein.

only contained in astrocytes, but also in ependymal cells and A Pia mater B OB cortex
differ only at the quantitative level among these cell types (see LV
Table 5.1; chapter 4) (Beckervordersandforth et al. 2010).
Radial glial cells thus share hallmarks with both astrocytes RMS
and ependymal cells not only at the morphological, but also C
at the molecular level. These commonly expressed proteins
SEZ
indeed confer key functional properties shared by astrocytes,
ependymal cells, and radial glia, such as glutamate uptake, cilia
K+-buffering and water transport, Ca2+-waves mediated by
gap junctions and hemichannels (see Table 5.1; chapters 16,
24, 34 and 35). Also adult neural stem cells, which likewise
possess radial glial hallmarks with apical contact and a short- blood
ened basal process (Kriegstein and Alvarez-Buylla 2009), vessels
share these functional hallmarks with astrocytes and ependy- LV
mal cells (Fig. 5.2; see Table 5.1).
According to their similarity to ependymal cells, includ- ventricle
ing ventricular contact, radial glial cells are often referred to
as ependymoglia. In the mammalian CNS, radial glial cells are D
largely transient and disappear at early postnatal stages (see
Fig. 5.2). The ventricle is largely lined by cuboid ependymal
cells, lacking a long radial process. However, in some regions E
ependymal cells with an extended radial morphology are pres- OPC
Radial glia/adult neural stem cell
ent (see chapter 4), oftesn referred to as tanycytes (a term used
for glial cells with access to the ventricle and a longer radial Figure 5.2 Radial Glial Cells in Postnatal Mouse
process), such as in the hypothalamus and the subcommis- Forebrain. A. Fluorescence micrograph depicting GFP-labeled radial
sural organ (SCO) (Rodríguez et al. 1998). Thus, tanycytes are glial cells in the dorsal telencephalon of neonatal mouse. Note that radial
glial cells still span the entire width of the brain parenchyma from the
included in the described morphological definition of radial ventricle to the pial surface (pia mater) at this stage. Scale bar: 100 μm.
glial cells with ventricular access, apico-basal polarity, and an B. Schematic drawing of a sagittal section through the adult mouse brain
extended radial process. In most cases these subtypes of radial depicting the areas indicated in panels A and C with a red square. The
glia possess highly specialized (e.g., secretory) functions, as lower panel depicts the subependymal zone (SEZ). In this region adult
in the SCO, releasing a glycoprotein-rich substance forming neural stem cells have hallmarks of radial glial cells. C. Schematic drawing
depicting the cellular composition of the adult mouse SEZ according to
the Reissner’s membrane in the ventricle. Interestingly, radial Fischer et al. 2011. D,E. Examples of radial glial cells (labeled with GFP
glial cells releasing the same protein have also been observed in hGFAP-eGFP mice) from the wall of the lateral ventricle of adult mice
in the lancelet and the ectoneural system of echinodermata (D, lateral wall; E, medial wall). Scale bars: (A) 100 μm; (D,E) 20 μm.

52 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
A B many radial glial cells are present in the adult amphibian,
reptilian, and avian CNS (Cuoghi and Mola 2009).

1.1 A D U LT N EU R A L S T E M C E L L S A R E
RADIAL GLIA
Also in the adult mammalian brain dividing radial glial cells
are present in very few niches in the forebrain where they
act as adult neural stem cells (Fig. 5.2b; see chapters 30, 40)
C D (Kriegstein and Alvarez-Buylla 2009). This is the case at the
wall of the lateral ventricle, where radial glial cells persist in a
widespread manner (Gubert et al. 2009). These cells possess a
small apical process in contact with the ventricle, junctional cou-
pling to neighboring ependymal cells or other stem cells, and a
radial process contacting the basement membrane surrounding
blood vessels (see Fig. 5.2). Similar to radial glial cells during
development, these cells also have a distinct apical membrane
domain with, for example, prominin1 sorted to microvilli or
E F f
cilia. (Note that radial glial cells typically bear a single cilium,
e in contrast with the multiciliated ependymal cells.) These hall-
marks (Prominin1 and glial expression) allow enrichment of
adult neural stem cells (Beckervordersandforth et al. 2010) as
for radial glial cells in the developing brain (Pinto et al. 2008)
by fluorescence-activated cell sorting (FACS). Notably, radial
glial cells in other niches of the ventricular lining in the adult
rodent brain (e.g., in the hypothalamus) can also resume pro-
liferation and seemingly generate young neurons under some
conditions (Kokoeva et al. 2005; Pérez-Martín et al. 2010).

1.2 R A D I A L A S T RO C Y T E S
Figure 5.3 Examples of Radial Glial Cells in the Zebrafish
Telencephalon. A. Micrograph of transgenic GFP+ radial glial cells in In addition, cells with astroglial properties and radial morphol-
48 hours post fertilization zebrafish forebrain with Dsred transgenic ogy are present in some regions of the adult mammalian CNS,
neurons (NBT-DsRed line). B. Micrograph of GFAP-immunostained
section of an adult zebrafish telencephalon hemisphere. Note the radial such as the spinal cord, dentate gyrus, the cerebellum with the
GFAP+ processes traversing the entire thickness of the telencephalon. Bergmann glia, and the retina with the Müller glia (see chapter
C. Micrograph of GFAP-GFP+ radial glial cells with somata lining the ven- 4). All of these cells lack contact to the ventricle and all except
tricle and processes extending through the everted adult zebrafish telenceph- the Müller glia (see chapter 4) also lack epithelial hallmarks
alon line. D. Micrograph of S100β+ radial glia somata lining the ventricle of with an apical and basolateral membrane domain. Therefore,
the everted adult zebrafish telencephalon. E. Micrograph of a single GFP+
radial glial cell from the adult zebrafish telencephalon 6 days after lipofec- they are referred to as radial or radially oriented astrocytes.
tion. F. Micrograph of a single GFP+ radial glial cell from the adult zebrafish This terminology also applies to the so-called outer or basal
telencephalon 5 days after transduction with a viral vector. Note the differ- “radial glia” (Borell and Reillo 2012), a secondary type of glial
ence in branching between radial glial cells in (E) and (F). Scale bars: cells in some regions of the developing brain that lack access
(A) 16 μm; (B,D) 100μm; (C,E,F) 20 μm.(C) From Bernardos et al. 2006. to the ventricle but are connected by a long radial process to
the basement membrane. These cells appear in the develop-
(Viehweg et al. 1998), suggesting their early emergence in the ing cerebral cortex of rodents and become even more frequent
deuterostome lineage. Thus, radial glial cells are not only the during phylogeny, in carnivores and primates (see chapter 30)
first glial cells to appear in ontogeny, but seemingly also appear (Borell and Reillo 2012; Lui et al. 2011).
at early stages in deuterostome phylogeny (Hartline, 2011).
In many if not most nonmammalian and nonavian ver-
tebrates, radial glial cells persist in a widespread manner 2 R A D I A L G L I A I N D E VE L O PM E N T
into adulthood. Glial cells with a radial morphology and an
apical contact to the ventricle line the adult CNS as during During development, radial glial cells perform various key
development with a notably longer radial process extending functions. They mediate architectonic stability and hence
all the way to the pial surface (Fig. 5.3). Many of these radial allow appropriate morphogenetic movements. This includes
glial cells divide in most CNS regions, such as in the zebrafish formation of boundary structures limiting cell migration. The
telencephalon (Chapouton et al. 2007), and can resume cell radial processes of radial glial cells act as guidance structures for
division in regions where they normally do not divide, such migrating neurons and radial glial cells act as lineage-restricted
as the zebrafish spinal cord (Reimer et al. 2008). Likewise, progenitors as well as multilineage stem cells.

R ADIAL GLIAL CELLS • 53


2.1 R A D I A L G L I A A S S T E M A N D P RO G E N ITO R generate neuronal as well as glial progeny (Malatesta et al.
C E L L S —H ET E RO G E N E IT Y O F R A D I A L G L I A 2000; Miyata et al. 2001; Noctor et al. 2001). Radial glial cells
isolated by FACS comprise different sets of progenitors and
Historically, when radial glial cells were first discovered in
stem cells generating only neurons, only glia, or glial cells and
the developing nervous system, they were suggested to act
neurons (Fig. 5.5) (Malatesta et al. 2000). Live imaging then
as progenitor cells, including a contribution to neurogen-
allowed monitoring directly how neurons are generated from
esis (Bentivoglio and Mazzarello 1999; García-Marin et al.
radial glia with apparent differences in the zebrafish embryo
2007). These suggestions were then neglected because of the
in vivo (Alexandre et al. 2010) and slice preparations of the
misconception of considering cells in different phases of the
forebrain from rodent embryos (see chapter 30) (Kriegstein
cell cycle as distinct cell types (interkinetic nuclear migration
and Alvarez-Buylla, 2009; Miyata et al. 2001; Noctor et al.
causes cells in S-phase to be located at abventricular locations,
2001). Genetic fate mapping further substantiates the contri-
whereas M-phase occurs at the ventricular surface) (Fig. 5.4).
bution of rodent radial glia to neurogenesis as well as other
His proposed in 1902 that the neural tube was composed of
glial lineages (Pinto and Götz 2007). The heterogeneity of
two kinds of committed progenitor cells, neuroblast produc-
radial glial cells in lineage and potential relates to differences in
ing germinal cells (dividing at the ventricular surface) and
gene expression (Pinto et al. 2008, 2009) distinguishing radial
nonproliferative glia, the “spongioblasts” (located at abven-
glia generating neurons directly from radial glial cells generat-
tricular positions). Only with the discovery of the cell cycle
ing glial cells and neurons indirectly via an intermediate set of
phases and their occurrence at different positions (Fujita
progenitors (see Fig. 5.5; chapter 30) (Pinto et al. 2008, 2009).
2003) was this concept questioned, and the “matrix cell the-
These distinct modes of radial glia–derived neurogenesis were
ory” was proposed with a homogeneous set of bilineage or
revealed by live imaging (Haubensak et al. 2004; Miyata et al.
multilineage progenitors or stem cells generating first neurons
2001, 2004; Noctor et al. 2001, 2004) as well as FACS allow-
and later glia (Fujita 2003). However, in the 1960s the glial
ing to separate directly from indirectly neurogenic radial
hallmarks of these cells were also discovered, which stabilized
glial cells (Pinto et al. 2008, 2009). Interestingly, direct and
the concept of distinct sets of neural progenitors and radial
glial cells, the latter serving support functions and the former
generating neurons (Bentivoglio and Mazzarello 1999). In
addition, transitional forms between radial glia and astrocytes A
(Pinto and Götz 2007) further supported the link between
radial glial cells and the generation of glia, namely, astrocytes.
Following the initial misconception of His a century longer,
many authors also suggested that the spongioblast equivalent
radial glial cells would not divide.
Now it is known that virtually all radial glial cells in the
developing brain of mammals and other vertebrates (except
for cells in the floor plate and roof plate because of par-
ticularly high levels of Notch signaling) divide and undergo Pure neuronal Mixed Pure glial
interkinetic nuclear migration (Götz and Huttner 2005). progeny progeny progeny
The concept of nonproliferating radial glia was overcome at
the end of the 1990s, when virtually all radial glial cells were B
shown to divide (Götz et al. 1998; Hartfuss et al. 2001) and

apical radial glia subapical radial glia


SVZ
VZ

Direct Indirect
AS

t t neurogenesis neurogenesis
S- G2- M- M- G1- S- G2- M- M- G1-
phase phase Figure 5.5 Schematic Drawing of Radial Glia Lineage. A. Single cell
progeny of radial glial cells isolated from the cerebral cortex of embryonic
Figure 5.4 Radial Glial Cell Division and Regionalization. Schematic day 14 mice (see Malatesta et al. 2000). B. Different modes of neurogene-
drawing of interkinetic nuclear migration of radial glial cells with exam- sis from radial glial cells either by intermediate progenitors in an indirect
ples of GFP electroporated cells next to it. Note that subapically dividing mode (right) or direct neurogenesis (left). Note that these subtypes can
radial glia (right side) are a subtype specific to a few forebrain regions in be separated on the basis of the level of GFAP-driven GFP and their api-
mice, whereas apically dividing radial glial cells are widespread (left side). cal prominin1 (see Pinto et al. 2008).

54 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
indirect generation of cells has also been observed in astroglio- distinct anlagen in the developing hindbrain. These bound-
genesis (see chapter 12) with astrocytes derived from glial pro- aries are of functional significance because their absence in
genitors in the postnatal brains, but also by direct conversion mutant mice results in increased cell mixing between these
of a radial glial cell to an astrocyte (see sections 2 and 3; see regions and ectopic neurogenesis (Chapouton et al. 1999;
chapter 12). Likewise, oligodendrogliogenesis (see chapter 13) Takahashi and Osumi 2011). Moreover, the roof plate and
occurs by intermediate highly proliferative progenitor migrat- floor plate, key signaling centers in the developing neural tube,
ing throughout the brain in a first wave as well as from radial consist of specialized radial glial cells. These radial glial cells
glial cells upregulating oligodendroglial fate determinants hardly proliferate because of particularly high levels of Notch
(Pinto et al. 2008) at late stages (Kessaris et al. 2006) suppos- activity that inhibits proliferation (Baek et al. 2006). Radial
edly in a more direct manner. Taken together, the concept of glial cells in the floor plate contribute to neurogenesis only in
direct and indirect modes of cell type generation may serve to the ventral midbrain, where they generate the dopaminergic
expand cell numbers at specific stages of development. neurons of the substantia nigra (Bonilla et al. 2008; Ono et al.
2007).
2.2 T H E MU LT I P L E RO L E S O F R A D I A L
G L I A I N PAT T E R N I N G T H E D EV E L O P I N G 2.3 S TA B I L I Z I N G T H E D EVE L O P I N G
C E N T R A L N E RVO US SYS T E M B R A I N: T H E E S S E N T I A L F U N C T I O N O F
A P I C O -BA S A L P O L A R IT Y A N D E P IT H E L I A L
Besides their lineage heterogeneity in one brain region, the
HALLMARKS OF RADIAL GLIA
cerebral cortex of mice, radial glial cells also differ profoundly
in different regions of the developing CNS. For example, in In addition to the described region-specific functions, bound-
the spinal cord they appear only at the onset of gliogenesis ary structures formed by radial glia also regulate morphogen-
and hence hardly contribute to neurogenesis (Pinto and Götz esis and often constrict the neural tube in radial dimension
2007). In the ventral telencephalon indirect neurogenesis (see at the sites of boundary formation leading to an apical sul-
Fig. 5.5) is largely increased and radial glial cells seem to con- cus. Thus, the length of the radial glia fiber determines the
tribute only indirectly to neurogenesis, with daughter cells radial extension of the growing neural tube. Radial glial cells
amplifying first in the ventricular zone and then in the sub- further provide tangential restrictions in tissue extension by
ventricular zone, thereby expanding their progeny in number their tight junctional coupling, thereby limiting the tangen-
(Pilz et al. 2012). Part of this amplifying progenitor cascade tial expansion of a given brain region. Therefore, the num-
is a special type of radial glial cells not undergoing interki- ber of radial glial cells at the apical surface defines the size
netic nuclear migration but dividing at subapical positions of a brain region. The total number of radial glial cells lining
still in the ventricular zone (see Fig. 5.4). Significantly, radial the ventricular surface and restricting the expansion of this
glial cells generate—indirectly or directly—very distinct surface is largely achieved by earlier symmetrical divisions of
types of neurons and glia in different brain regions, thereby apical progenitors at the neuroepithelial stage regulating the
contributing essentially to patterning of the brain (Götz and pool of self-renewing apical progenitor cells, including radial
Campbell 2002). For example, throughout the ventral regions glia. In addition, the self-renewing capacity of radial glial cells
of the developing CNS, radial glial cells generate oligoden- is required to maintain this pool (Cappello et al. 2006). Thus,
drocyte progenitor cells that migrate extensively distributing as radial glial numbers regulate the size of the progenitor pool
throughout the CNS, whereas radial glial cells in the dorsal in a brain region they determine the size of this region and
telencephalon contribute to oligodendrogliogenesis only at simultaneously supply the respective brain region with the
later stages (see chapter 13 and the preceding) (Kessaris et appropriate number of guiding structures for radial neuronal
al. 2006; Pinto et al. 2008). In some regions radial glial cells migration.
generate largely GABAergic neurons and in others exclusively
glutamatergic neurons during development (Malatesta et al.
2.4 A P I C A L A N C H O R I N G A N D S I G NA L I N G
2003). Intriguingly, this regionalization is inherited to the
radial glial cells persisting in these regions as adult neural stem Defects in anchoring of radial glia at the apical surface affect-
cells (Brill et al. 2009; Merkle et al. 2007). Likewise, regional ing the junctional complexes and/or their connections to
differences in radial glial cells are at the source of generating the cytoskeleton result in severe defects in morphogenesis.
regional diversity in astrocytes in the developing spinal cord Junctional complexes (see Fig. 5.1C) are composed by dense
(Hochstim et al. 2008) further supporting the importance of clusters of the transmembrane proteins cadherins whose extra-
radial glia diversity for patterning and function in the adult cellular domains tightly bind two neighboring cells together (see
CNS. Fig. 5.1D). The intracellular domains of cadherins interact with
Radial glial cells contribute further to patterning and catenins (β- and α-catenin) connecting the cadherin complexes
regionalization of the developing CNS by forming unique in a dynamic mode to the actin cytoskeleton (see Fig. 5.1D).
signaling centers and boundary regions. Radial glial cells form The actin cytoskeleton stability is regulated by various signal-
fascicles condensing their radial processes into tight bound- ing molecules, including small Rho-GTPases influencing the
ary structures, for example, at the pallial–subpallial bound- tight balance among individual actin monomers, the globular
ary delineating the dorsal and the ventral telencephalon, the form of G-actin, and their assembly into larger filaments, the
mid-hindbrain boundary or the rhombomere boundaries, filamentous F-actin. Defects in either of these components in

R ADIAL GLIAL CELLS • 55


radial glia, such as defects in cadherin clustering, β- or α-catenin brain disorder of cobblestone lissencephaly, in which neurons
and F-actin formation, result in severe aberrations in neuro- assemble in cobblestone-like extrusions atop the brain sur-
architecture of the affected brain region. For example, the lami- face (Bielas and Gleeson 2004). Thus, many disorders with
nated structure of the cerebral cortex with neurons arranged mislocalization of neurons at ectopic sites do not result from
in horizontal layers is abolished into a random or concentric defects within the migrating neurons themselves, but rather
arrangement of neurons when radial glia anchoring is disrupted arise as a secondary consequence of loss of radial glial anchor-
by affecting either of the preceding components (Cappello et al. ing (Halfter et al. 2002). Rupture of the BM caused by defec-
2006, 2012; Lien et al. 2006; Yokota et al. 2009). Notably, these tive anchoring of the radial glia endfeet reveals their key role
defects observed after conditional gene deletion in the mouse in stabilizing the BM. This function is mediated by integrins
model reflect neuronal dysplasias observed in patients (Bielas in the radial glia endfeet anchoring extracellular matrix com-
and Gleeson 2004); for example, formation of a double-cortex ponents (ECM) of the BM (see Fig. 5.1B). Accordingly, loss
with a second cerebral cortex underlying the white matter of major integrin subunits in radial glial cells leads to BM rup-
(Cappello et al. 2012). Besides providing essential anchors for ture and subsequent migration of neurons through these rup-
tissue architecture, adherens junctions also act as signaling cen- tures (Bielas and Gleeson 2004; Halfter et al. 2002; Haubst
ters enriching components of, for example, Notch-, Par-, and et al. 2006). Anchoring of integrins to the actin cytoskeleton
Wnt-mediated signaling pathways (see Fig. 5.1D), and thereby is likewise critical to avoid BM rupture and neuronal ectopia.
regulate proliferation and differentiation of radial glial cells Integrins binding to ECM components also mediate signaling
(Johansson et al. 2010; Zhang et al. 2010). In addition, gap junc- promoting proliferation (Fietz et al. 2010; Loulier et al. 2009).
tional coupling of radial glial cells is important for communica- Accordingly, maintenance of the basal process after radial glial
tion mediated by Ca2+ or other ions and small molecules (Elias cell division plays a key role in the decision to continue pro-
and Kriegstein 2008). Ca2+-signaling in radial glial cells can also liferation as revealed by live imaging in zebrafish and mouse
be elicited by depolarizing via neurotransmitter receptors (e.g., radial glia (Alexandre et al. 2010; Shitamukai et al. 2011).
GABA or glutamate receptors), which positively regulate radial Thus, like the apical anchoring by junctional complexes, also
glia proliferation (Elias and Kriegstein 2008). Taken together, the basal anchoring mediates key signaling pathways, thereby
the function of radial glial cells as stabilizing structures and stem linking progenitor functions and stabilizing functions in an
and progenitor cells is integrated at the points of adhesion and almost inseparable manner. Taken together, the epithelial
coupling. properties of radial glial cells are not only a defining feature,
Apical junctions of radial glial cells are also critical to delin- but also exert essential roles at both apical and basal sides for
eate the contact of the ventricular fluid to the apical membrane all the key functions of radial glial cells.
domain. Indeed, access to the cerebrospinal fluid (CSF) is a
key hallmark of radial glial cells allowing direct access to key
2.6 R A D I A L G L I A A S GU I D E S F O R
signaling molecules in the CSF, such as insulin-like growth
M I G R AT I N G N EU RO NS
factor (IGF), sonic hedgehog (SHH), bone morphogenic
protein (BMP), or WNT ( Johansson et al. 2010; Lehtinen Apico-basal contacts of radial glia are also critical to maintain
and Walsh 2011). In addition, vesicles of distinct size contain- the radial process, which fails to extend in a normal radial
ing apical membrane components are released into the CSF manner when anchoring of radial glial cells is lost. These radial
by radial glial cells as so-called prominosomes (Marzesco et al. processes provide important guiding structures along which
2009) and may constitute a further signaling component via some neurons migrate from their place of birth to their final
CSF. Notably, during embryonic development the ventricular position. Consistent with first observations (Bentivoglio and
system is still closed, with no connection to the subarachnoid Mazzarello 1999), ultrastructural analysis revealed the tight
space and circulation (also differentiated ependymal cells with alignment of neurons to radial glial processes (Chapter 32),
beating cilia are not yet present). Thus localized release of sig- and live imaging revealed their mode of migration in a salta-
naling molecules into the CSF may form gradients depend- tory manner (Cooper 2008). Guidance of neurons along the
ing on the diffusion of the respective protein. Significantly, radial process depends on a multitude of molecules expressed
the special junctions coupling radial glial cells (Mollgoard by radial glial cells, including the extracellular domains of gap
and Saunders, 1975) restrict diffusion of molecules within the junctional proteins, the connexins, forming hemichannels
CSF into the neural tube. Because the apical side of radial glial along the radial process (Elias and Kriegstein 2008). Molecules
cells, however, has access to these components, radial glial cells regulating radial glia–guided migration are specific and often
act as key mediator of signals to and from the CSF. not involved in other modes of migration, such as tangential
migration or somal translocation (Nadarajah and Parnavelas
2002), which are influenced by other factors (Cooper 2008).
2.5 BA S A L A N C H O R I N G A N D S I G NA L I N G
The basal endfoot of radial glial cells (see Fig. 5.1B, C) is like-
wise important for anchoring and neuronal histogenesis as 3 R A D I A L G L I A I N T H E A D U LT
well as mediating key signaling pathways. Disruption of the C E N T R A L N E RVO U S SYS T E M
radial glia basal endfeet attachment to the BM underlying the
meninges results in rupture of the BM and subsequent migra- Although radial glial cells are the first and the predomi-
tion of neurons into these ectopic sites, reflecting the human nant glial cell type in the developing nervous system of all

56 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
vertebrates analyzed so far, in most regions of the mamma- Table 5.2A COMPARISON BETWEEN RADIAL GLIAL
lian brain they are transient and differentiate into diverse cell CELLS IN THE DEVELOPING AND ADULT ZEBRAFISH
types at the end of neurogenesis and neuronal migration (e.g., BRAIN
shortly after birth in the rodent cerebral cortex). Most radial RADIAL GLIA IN RADIAL GLIA
glia disappear by symmetrical neurogenic divisions, that is, ZEBRAFISH IN ADULT
generating two postmitotic neurons at the end of neurogen- PROTEIN LARVAE ZEBRAFISH
esis, by differentiating into ependymal cells or differentiat-
GFAP ++ +++
ing into astrocytes translocating via their radial process from
the ventricular surface (see chapters 12 and 30) (Kriegstein GLAST(Slc1a2) Nd Nd
and Alvarez-Buylla 2009). In few regions, however, such as GLT1(Slc1a3) Nd Nd
the wall of the lateral ventricle, radial glial cells persist inte-
grated into the otherwise differentiating ependyma. The Glutamine synthetase Nd +++
radial glial soma is located above the ependyma, where also S100-β – +++
all the progenitors, the transit-amplifying progenitor and the Connexin 43 Nd Nd
neuroblasts, comprise the “subependymal zone” (SEZ) (see (Gja1)
Fig. 5.2). In the developing brain, where no ependymal cells
Aquaporin 4 Nd +++
are present, radial glial cells line the ventricle and hence other
progenitors that are not located apically (Götz and Huttner KIR 4.1/2.1 Nd +
2005) form the “subventricular zone” (SVZ). However, most Aldlhl1 Nd Nd
of the SEZ radial glial cells do not maintain a radial process
contacting the pial surface, but rather have a shorter radial Nestin ++ +++
process to the BM surrounding neighboring blood vessels Vimentin + +
(see Fig. 5.2B, C) (Kriegstein and Alvarez-Buylla 2009).
BLBP ++ +++
According to the hallmark of radial glial cells, these cells also
possess access to the ventricle with a small apical endfoot, TN-C Nd Nd
which contains apical membrane proteins, such as promi- Sox2 ++ +++
nin1 (Beckervordersandforth et al. 2010). These radial glial
Aromatase B ++ +++
cells are the adult neural stem cells as isolation of cells with
glial expression (high levels of GFAP-driven GFP) (see Fig. FGFR2/3 ++ ++
5.2C) and the apical membrane protein prominin1 by FACS ChSPG4 Nd Nd
allows enrichment of self-renewing and multipotent stem cells
(Beckervordersandforth et al. 2010). Indeed, genetic fate map- According to Zfin database and Alexandre et al. 2010; Chapouton et al. 2007,
2010; Ganz et al. 2010 ; Grupp et al. 2009; März et al. 2010; Rothenaigner et al.
ping using the split-Cre technology also confirms the stem cell 2011; Tong et al. 2009; Topp et al. 2008.
identity of cells coexpressing GFAP and prominin1 in vivo
(Beckervordersandforth et al. 2010). These stem cells gener- Table 5.2B COMPARISON BETWEEN RADIAL GLIAL
ate a series of progenitors that then migrate to the olfactory CELLS IN THE DEVELOPING AND ADULT ZEBRAFISH
bulb generating diverse types of interneurons life long (see BRAIN
chapter 40) (Brill et al. 2009; Kriegstein and Alvarez-Buylla
RADIAL GLIA RADIAL
2009). Notably, interneurons of the olfactory bulb originate AFTER 48 HPF GLIA IN
during development from radial glia of the lateral wall of the IN ZEBRAFISH ADULT
lateral ventricle, and neurogenesis of these neurons contin- FUNCTION EMBRYOS ZEBRAFISH
ues into adulthood in almost all vertebrates analyzed so far
(Doetsch and Scharff 2001; Kishimoto et al. 2011). Glutamate uptake Nd Nd
K-conductance at rest Nd Nd
3.1 WI D E S P R E A D R A D I A L G L I A I N T H E A D U LT Glycogen storage Nd Nd
B R A I N O F M A N Y V E RT E B R AT E S
Gap-junctions/ Nd Nd
Although radial glial cells persisting in the adult brain are the Hemichannels/
exception in the mammalian and avian brains, this is the rule Ca-waves
in most other vertebrates (Cuoghi and Mola 2009; Kalman Blood vessel contact/blood ++ ++
2002). Most vertebrates maintain GFAP+ cells with long flow regulation
radial processes lining the ventricle, which are also coupled Cell division +++ +++
by junctional complexes delineating apical from basolateral
membrane (Grupp et al. 2009). Notably, these cells have a sin- Multipotency +++ +++
gle cilium, such as radial glial cells, and they also express other Self-renewal +++ +++
typical astrocyte/ependyma hallmarks such as S100β, glu- According to Zfin database and Alexandre et al. 2010; Chapouton et al. 2007,
tamine synthase and BLBP (Table 5.2). However, as for radial 2010; Ganz et al. 2010 ; Grupp et al. 2009; März et al. 2010; Rothenaigner et al.
glial cells in the developing brain, there is regional and subtype 2011; Tong et al. 2009; Topp et al. 2008.

R ADIAL GLIAL CELLS • 57


heterogeneity among radial glial cells. For example, in the very large injuries (Kroehne et al. 2011). This distinct glial reac-
adult zebrafish telencephalon (Ganz et al. 2010; Grupp et al. tivity may at least contribute to the scar-less wound healing
2009; März et al. 2010), a subset of radial glial cells proliferates in the adult zebrafish CNS (Ayari et al. 2010; Baumgart et al.
and nonproliferating radial glia can be activated by blocking 2012; März et al. 2011). Only after very large injuries, some
Notch-signaling or injury (Ayari et al. 2010; Baumgart et al. scarlike lesion remains in correlation to migration of radial
2012; Chapouton et al. 2010; Kroehne et al. 2011; Reimer glia–derived astrocytes to the injury site as well as oligoden-
et al. 2008). Thus, similar to embryogenesis, high levels of drocyte progenitor activation (Kroehne et al. 2011). Notably,
Notch signaling block radial glia proliferation. In some CNS injuries damaging the ventricular surface of radial glial cells
regions, such as the spinal cord, radial glial cells do not prolif- evoke a reaction more similar to reactive astrogliosis (März
erate (Reimer et al. 2008), but can also be activated by injury et al. 2011).
(Echeverri and Tanaka 2002; Reimer et al. 2008, 2009). In Most distinct from the situation after brain injury in mam-
both cases, with or without prior proliferation, these cells mals, however, radial glial cells increase neurogenesis after
are able to replace the damaged neurons, as clearly demon- injury not only in regions of normal neurogenesis (Ayari et al.
strated in the telencephalon and spinal cord. In the absence 2010; Baumgart et al. 2012; Kroehne et al. 2011), but also in
of injury, adult neurogenesis occurs in regions with radial glia the spinal cord where no adult neurogenesis occurs normally
proliferation in a much more widespread manner compared (Reimer et al. 2009). A similar neurogenic repair reaction of
with the mammalian or avian brain (Chapouton et al. 2007; a radial glial-like glia cell, the Müller glia, also occurs in the
García-Verdugo et al. 2002). zebrafish retina (see chapter 4) (Raymond et al. 2006; Yurco
Radial glial cells persist in most CNS regions of the adult and Cameron 2005). Thus, the loss of radial glial cells in adult
teleost CNS, as well as in Chondrichtyes, other Actinopterygii, brains of mammals and birds required by increased size and
Amphibia, and reptiles (Cuoghi and Mola 2009; Kalman complexity is accompanied by a rather dramatic side effect
2002). With the exception of the optic nerve, virtually no free leading to differences in reactive gliosis involved in scar forma-
stellate–shaped astrocytes or cuboid ependymal cells are pre- tion and the loss of regenerative response (Robel et al. 2011).
sent in most of the species analyzed in these vertebrate classes.
Therefore, radial glial cells comprise both functions of astro-
cytes and ependymal cells in the adult CNS. Accordingly, they 5 S U M M A RY A N D P E R S P E C T I VE S
also possess Aquaporin channels, even though these are not
sorted toward the polarized BM interface, as is the case for Taken together, radial glial cells are the first and major glial
astrocytes (see chapter 4) (Grupp et al. 2009). Interestingly, cell type in ontogeny and phylogeny. They combine hallmarks
however, in brain regions with increased complexity and of ependymal cells (ventricular access, epithelial polarity) and
thickness, parenchymal astrocytes appear in all these groups, astrocytes (blood vessel contact; water, glutamate-transport
and these stellate cells reduce GFAP expression (Kalman and K+-buffering) with a progenitor and stem cell function.
2002). For example, the squalomorph shark brain has GFAP+ Accordingly, their delineation from astrocytes and ependymal
radial glia, whereas large areas in the skate brain are popu- cells by so-called “markers” (see Table 5.1) has been difficult
lated by stellate astrocytes mostly lacking GFAP expression because these are largely shared with only quantitative differ-
(Kalman 2002). Taken together, astrocytes seemingly evolved ences between these glial cell types (Beckervordersandforth
in parallel in Agnathi, Chondrichtyes, Actinopterygii, and et al. 2010). Although expression of all of these genes clearly
Sarcopterygii-Amniotes in correlation to increased complexity delineates radial glial cells from neuroepithelial cells (see
and thickness of respective brain regions. Increased size appar- Table 5.1A), difficulties delineating radial glia from astrocytes
ently required support by parenchymal glial cells prompting and ependymal cells have led to a rather confused nomencla-
delamination of radial glial cells and astrocyte formation dur- ture, especially when referring to adult brains. Especially radial
ing phylogeny (Cuoghi and Mola 2009; Kalman 2002). glial cells in the adult CNS, for example, in teleosts, are either
referred to as astroglia (Grupp et al. 2009) or ependymoglia
(Cuoghi and Mola 2009), and sometimes as radial glia (Robel
4 R A D I A L G L I A L C E L L R E AC T I O N et al. 2011). This highlights (1) the need for a clear nomen-
TO I N J U RY clature, and (2) a more comprehensive expression analysis
beyond the use of a few marker proteins.
Reactive astrogliosis is a major hallmark of the injury reaction In regard to a clear nomenclature, glial cells with ventricu-
in the mammalian brain. Thus, gliosis differs profoundly after lar access, a radial morphology, and epithelial characteristics
injury in vertebrate classes with persistent radial glial cells should be referred to as radial glial cells. This definition clearly
lacking stellate astrocytes. After an invasive injury in the adult comprises cells of diverse functions ranging from neural stem
zebrafish forebrain, most radial glial cells do not delaminate and progenitor cells to cells with other specialized functions,
from the ventricle and do not migrate toward the injury site such as secretory radial glia often referred to as tanycytes in
(Baumgart et al. 2012), except in very large injury conditions adult brains. The epithelial hallmarks of apico-basal polarity
(Kroehne et al. 2011). Likewise, the reaction of oligodendro- are central to this definition consistent with their functional
cyte progenitors in the adult brain, prominent in the mam- roles in radial glial cells, providing radial tissue stability as well
malian brain (Robel et al. 2011), largely fails to occur in the as influencing their proliferation, fate, and stable guidance and
injured zebrafish forebrain (Baumgart et al. 2012), except in boundary structure. In regard to extending expression analysis

58 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
of distinct glial cells to a genomewide expression level we now Bernardos RL, Raymond PA. 2006. GFAP transgenic zebrafish. Gene
have access to genomewide expression profiles of various types Expr Patterns 6:1007–1013.
Bielas SL, Gleeson JG. 2004. Cytoskeletal associated proteins in the
of glia (see chapters 28, 29) (Beckervordersandforth et al. migration of cortical neurons. J Neurobiol 58:149–159.
2010; Cahoy et al. 2008; Lovatt et al. 2007), including adult Bonilla S, Hall AC, Pinto L, Attardo A, Götz M, Huttner WB, et al.
and embryonic radial glial cells (Beckervordersandforth et al. 2008. Identification of midbrain floor plate radial glia-like cells as
2010; Pinto et al. 2008) revealing shared and unique expres- dopaminergic progenitors. Glia 56:809–820.
sion patterns of these cells. This comparative analysis also Borell V, Reillo I. 2012. Emerging roles of neural stem cells in cerebral
cortex development and evolution. Developmental Neurobiology,
suggests new selective “markers” that still require appropriate 72:955–971.
antibodies to be generated, but in the future will allow more Brill MS, Ninkovic J, Winpenny E, Hodge RD, Ozen I, Yang R, et al.
unequivocal delineation of these cells types, besides morpho- 2009. Adult generation of glutamatergic olfactory bulb interneurons.
logical and functional criteria. Nat Neurosci 12:1524–1533.
An essential functional difference between radial glial cells Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson
KS, et al. 2008. A transcriptome database for astrocytes, neurons,
and their astrocyte and ependymal cell relatives is their role and oligodendrocytes: a new resource for understanding brain devel-
as stem and progenitor cells contributing to neurogenesis and opment and function. J Neurosci 28:264–278.
gliogenesis in a widespread manner in developing and adult Cappello S, Attardo A, Wu X, Iwasato T, Itohara S, Wilsch-Braeuninger
vertebrate CNS, and a few niches in the adult mammalian M, et al. 2006. The Rho-GTPase cdc42 regulates cortical precursor
forebrain. Obviously, beyond gaining new candidates for spe- fate at the apical surface. Nat Neurosci 9:1099–1107.
Cappello S, Böhringer CRJ, Bergami M, Conzelmann K-K, Ghanem
cific “marker” proteins, comparative analysis of the glial cells A, Tomassy GS, et al. 2012. A radial glia specific role of RhoA in
with and without stem cell properties will allow unravelling double-cortex formation. Neuron 73:911–924.
the core network regulating this key functional property, as Chapouton P, Gärtner A, Götz M. 1999. The role of Pax6 in restrict-
well as the genetic regulation of other core functions of dis- ing cell migration between developing cortex and basal ganglia.
tinct glial cells (see chapters 28, 29). Development 126:5569–5579.
Chapouton P, Jagasia R, Bally-Cuif L. 2007. Adult neurogenesis in
non-mammalian vertebrates.Bioessays 29:745–757. Review.
Chapouton P, Skupien P, Hesl B, Coolen M, Moore JC, Madelaine
AC K N OW L E D G E M E N T S R, et al. 2010. Notch activity levels control the balance between
quiescence and recruitment of adult neural stem cells. J Neurosci
Foremost the author would like to thank the DFG for the 30:7961–7974.
Cooper JA. 2008. A mechanism for inside-out lamination in the neocor-
Leibniz Award, which has provided the freedom to pursue tex. Trends Neurosci 31:113–119.
exciting new avenues in research, such as the stem cell function Cuoghi B, Mola L. 2009. Macroglial cells of the teleost central nervous
of glial cells. The author is also particularly grateful to Joana system: a survey of the main types. Cell Tissue Res 338:319–332.
Barbosa, Emily Baumgart, Ruth Beckervordersandforth, Judith Doetsch F, Scharff C. 2001. Challenges for brain repair: insights
Fischer, Wieland Huttner, Jovica Ninkovic, Gregor Pilz, Stefanie from adult neurogenesis in birds and mammals. Brain Behav Evol
58:306–322.
Robel, Franziska Weinandy, Michaela Wilsch-Bräuninger, and Echeverri K, Tanaka EM. 2002. Ectoderm to mesoderm lineage switch-
Hartwig Wolburg for beautiful examples of radial glia and their ing during axolotl tail regeneration. Science 298:1993–1996.
polarity aspects for the figures; and would also like to thank Elias LA, Kriegstein AR. 2008. Gap junctions: multifaceted regulators
Laure Bally-Cuif, Angelique Bordey, and Andreas Reichenbach of embryonic cortical development. Trends Neurosci 31:243–250.
for valuable comments and suggestions. Fietz SA, Kelava I, Vogt J, Wilsch-Bräuninger M, Stenzel D, Fish JL,
et al. 2010. OSVZ progenitors of human and ferret neocortex are
epithelial-like and expand by integrin signaling. Nat Neurosci
13:690–699.
REFERENCES Fujita S. 2003. The discovery of the matrix cell, the identification of the
multipotent neural stem cell and the development of the central nerv-
Alexandre P, Reugels AM, Barker D, Blanc E, Clarke JD. 2010. Neurons ous system. Cell Struct Funct 28:205–228.
derive from the more apical daughter in asymmetric divisions in the Ganz J, Kaslin J, Hochmann S, Freudenreich D, Brand M. 2010.
zebrafish neural tube. Nat Neurosci 13:673–679. Heterogeneity and Fgf dependence of adult neural progenitors in the
Ayari B, El Hachimi KH, Yanicostas C, Landoulsi A, Soussi-Yanicostas zebrafish telencephalon. Glia 58:1345–1363.
N. 2010. Prokineticin 2 expression is associated with neural García-Marín V, García-López P, Freire M. 2007. Cajal’s contributions to
repair of injured adult zebrafi sh telencephalon. J Neurotrauma glia research. Trends Neurosci 30:479–487. 21
27:959–972. García-Verdugo JM, Ferrón S, Flames N, Collado L, Desfilis E, Font E. 2002.
Baek JH, Hatakeyama J, Sakamoto S, Ohtsuka T, Kageyama R. 2006. The proliferative ventricular zone in adult vertebrates: a comparative
Persistent and high levels of Hes1 expression regulate boundary study using reptiles, birds, and mammals. Brain Res Bull 57:765–775.
formation in the developing central nervous system. Development Götz M, Campbell K. 2002. Radial glia: Multipurpose cells for verte-
133:2467–2476. brate brain development. TINS 25:235–238.
Baumgart EV, Barbosa J, Bally-Cuif L, Götz M, Ninkovic J. 2012. Stab Götz M, Huttner WB. 2005. The cell biology of neurogenesis. Nat Rev
wound injury of the zebrafish telencephalon—a model for compara- Mol Cell Biol 6:777–788.
tive analysis of reactive gliosis. Glia Nov 21, Epub ahead of print. Götz M, Stoykova A, Gruss P. 1998. Pax6 controls radial glia differentia-
Beckervordersandforth R, Tripathi P, Ninkovic J, Bayam E, Lepier A, tion in the cerebral cortex. Neuron 21:1031–1044.
Stempfhuber B, et al. 2010. In vivo fate mapping and expression anal- Grupp L, Wolburg H, Mack AF. 2010. Astroglial structures in the
ysis reveals unique molecular hallmarks of prospectively isolated zebrafish brain. J Comp Neurol 518:4277–4287.
adult neural stem cells. Cell Stem Cell 7:744–758. Gubert F, Zaverucha-do-Valle C, Pimentel-Coelho PM, Mendez-Otero
Bentivoglio M, Mazzarello P. 1999. The history of radial glia. Brain Res R, Santiago MF. 2009. Radial glia-like cells persist in the adult rat
Bull 49:305–315. brain. Brain Res 1258:43–52.

R ADIAL GLIAL CELLS • 59


Halfter W, Dong S, Yip YP, Willem M, Mayer U. 2002. A critical Merkle FT, Mirzadeh Z, Alvarez-Buylla A. 2007. Mosaic organization of
function of the pial basement membrane in cortical histogenesis. neural stem cells in the adult brain. Science 317:381–384. 23
J Neurosci 22:6029–6040. Miyata T, Kawaguchi A, Okano H, Ogawa M. 2001. Asymmetric
Hartfuss E, Galli R, Heins N, Götz M. 2001. Characterization of CNS inheritance of radial glial fibers by cortical neurons. Neuron
precursor subtypes and radial glia. Dev Biol 229:15–30. 31:727–741.
Haubst N, Georges-Labouesse E, DeArcangelis A, Mayer U, Götz M. Miyata T, Kawaguchi A, Saito K, Kawano M, Muto T, Ogawa M. 2004.
2006. Basement membrane attachment is dispensable for radial glial Asymmetric production of surface-dividing and non-surface divid-
cell fate and proliferation, but affects positioning of neuronal sub- ing cortical progenitor cells. Development 131:3133–3145.
types. Development 133:3245–3254. Mollgoard K, Saunders NR. 1975. Complex tight junctions of epi-
Hartline DK. 2011. The evolutionary origins of glia. Glia 59:1215-1236. thelial and of endothelial cells in early fetal brain. J. Neurocytol
Haubensak W, Attardo A, Denk W, Huttner WB. 2004. Neurons arise 4:453–468.
in the basal neuroepithelium of the early mammalian telencephalon: Mori T, Buffo A, Götz M. 2005. The novel roles of glial cells revisited:
a major site of neurogenesis. PNAS 101:3196–3201. The contribution of radial glia and astrocytes to neurogenesis. Curr
Hochstim C, Deneen B, Lukaszewicz A, Zhou Q, Anderson DJ. 2008. Top Dev Biol 69:67–99.
Identification of positionally distinct astrocyte subtypes whose iden- Noctor SC, Flint AC, Weissman TA, Dammerman RS, Kriegstein AR.
tities are specified by a homeodomain code. Cell 133:510–522. 2001. Neurons derived from radial glial cells establish radial units in
Johansson PA, Cappello S, Götz M. 2010 Stem cell niches during develop- neocortex. Nature 409:714–720.
ment—lessons from the cerebral cortex. Curr Opin Neurobiol 20:1–8. Noctor SC, Martínez-Cerdeño V, Ivic L, Kriegstein AR. 2004. Cortical
Kalman M. 2002. GFAP expression withdraws—a trend of glial evolu- neurons arise in symmetric and asymmetric division zones and
tion? Brain Res Bull 57:509–511. migrate through specific phases. Nat Neurosci 7:136–144.
Kessaris N, Fogarty M, Iannarelli P, Grist M, Wegner M, Richardson WD. Nadarajah B, Parnavelas JG. 2002. Modes of neuronal migration in the
2006. Competing waves of oligodendrocytes in the forebrain and post- developing cerebral cortex. Nat Rev Neurosci 3:423–432.
natal elimination of an embryonic lineage. Nat Neurosci 9:173–179. Ono Y, Nakatani T, Sakamoto Y, Mizuhara E, Minaki Y, Kumai M,
Kishimoto N, Alfaro-Cervello C, Shimizu K, Asakawa K, Urasaki A, et al. 2007. Differences in neurogenic potential in floor plate cells
Nonaka S, et al. 2011. Migration of neuronal precursors from the tel- along an anteroposterior location: midbrain dopaminergic neu-
encephalic ventricular zone into the olfactory bulb in adult zebrafish. rons originate from mesencephalic floor plate cells. Development
J Comp Neurol 519:3549–3565. 134:3213–3225.
Kokoeva MV, Yin H, Flier JS. 2005. Neurogenesis in the hypo- Pérez-Martín M, Cifuentes M, Grondona JM, López-Avalos MD,
thalamus of adult mice: potential role in energy balance. Science Gómez-Pinedo U, García-Verdugo JM, et al. 2010. IGF-1 stimu-
310:679–683. lates neurogenesis in the hypothalamus of adult rats. Eur J Neurosci
Kriegstein A, Alvarez-Buylla A. 2009. The glial nature of embryonic and 31:1533–1548.
adult neural stem cells. Annu Rev Neurosci 32:149–184. Pilz G, Shitamukai A, Pacary E, Reillo I, Johnson J, Guillemot F. 2012.
Kroehne V, Freudenreich D, Hans S, Kaslin J, Brand M. 2011. Amplification of progenitors in the mammalian telencephalon
Regeneration of the adult zebrafish brain from neurogenic radial includes a novel radial glia cell type. Nature Neurosci (in revision).
glia-type progenitors. Development 138:4831–4841. Pinto L, Drechsel D, Schmid M-T, Ninkovic J, Irmler M, Brill MS,
Lehtinen MK, Walsh CA. 2011. Neurogenesis at the brain-cerebrospinal et al. 2009. AP2γ regulates basal progenitor fate in a region- and
fluid interface. Annu Rev Cell Dev Biol 27:653–679. layer-specific manner in the developing cortex. Nat Neurosci
Lien WH, Klezovitch O, Fernandez TE, Delrow J, Vasioukhin V. 2006. 12:1229–1237.
Alpha E-catenin controls cerebral cortical size by regulating the Pinto L, Mader MT, Irmler M, Gentilini M, Santoni F, Drechsel D, et
hedgehog signaling pathway. Science 311:1609–1612. al. 2008. Prospective isolation of functionally distinct radial glial
Liu X, Bolteus AJ, Balkin DM, Henschel O, Bordey A.2006. subtypes-Lineage and transcriptome analysis. Mol Cell Neurosci
GFAP-expressing cells in the postnatal subventricular zone display 38:15–42.
a unique glial phenotype intermediate between radial glia and astro- Raymond PA, Barthel LK, Bernardos RL, Perkowski JJ. 2006. Molecular
cytes. Glia 54:394–410. characterization of retinal stem cells and their niches in adult
Loulier K, Lathia JD, Marthiens V, Relucio J, Mughal MR, Tang SC, zebrafish. BMC Dev Biol 6:36.
et al. 2009. beta1 integrin maintains integrity of the embryonic neo- Reimer MM, Kuscha V, Wyatt C, Sorensen I, Frank RE, Knuwer M,
cortical stem cell niche. PLoS Biol 7:e1000176. et al. 2009. Sonic hedgehog is a polarized signal for motor neuron
Lovatt D, Sonnewald U, Waagepetersen HS, Schousboe A, He W, Lin regeneration in adult zebrafish. J Neurosci 29:15073–15082.
JH, et al. 2007. The transcriptome and metabolic gene signature Reimer MM, Sorensen I, Kuscha V, Frank RE, Liu C, Becker CG, et al.
of protoplasmic astrocytes in the adult murine cortex. J Neurosci 2008. Motor neuron regeneration in adult zebrafish. J Neurosci
27:12255–12266. 28:8510–8516.
Lui JH, Hansen DV, Kriegstein AR. 2011. Development and evolution Robel S, Berninger B, Götz M. 2011. The stem cell potential from glia—
of the human neocortex. Cell 146:18–36. lessons from reactive gliosis. Nat Rev Neurosci 12:88–104.
Malatesta P, Hartfuss E, Hack MA, Klinkert W, Kirchhoff F, Rodríguez EM, Rodríguez S, Hein S. 1998. The subcommissural organ.
Kettenmann H, Götz M. 2003. Neuronal or glial progeny: regional Microsc Res Tech 41:98–123.
differences in radial glial fate. Neuron 37:751–764. Rothenaigner I, Krecsmarik M, Hayes JA, Bahn B, Lepier A, Fortin
Malatesta P, Hartfuss E, Götz M. 2000. Isolation of radial glial cells G, et al. 2011. Clonal analysis by distinct viral vectors identifies
by fluorescent-activated cell sorting reveals a neuronal lineage. bona fide neural stem cells in the adult zebrafish telencephalon
Development 127:5253–5263. and characterizes their division properties and fate. Development
März M, Chapouton P, Diotel N, Vaillant C, Hesl B, Takamiya M, et al. 38:1459–1469.
2010. Heterogeneity in progenitor cell subtypes in the ventricular Shitamukai A, Konno D, Matsuzaki F. 2011. Oblique radial glial divi-
zone of the zebrafish adult telencephalon. Glia 58:870–888. sions in the developing mouse neocortex induce self-renewing pro-
März M, Schmidt R, Rastegar S, Strä hle U. 2011. Regenerative response genitors outside the germinal zone that resemble primate outer
following stab injury in the adult zebrafish telencephalon. Dev Dyn subventricular zone progenitors. J Neurosci 31:3683–3695.
240:2221–2231. Takahashi M, Osumi N. 2011. Pax6 regulates boundary-cell specifica-
Marzesco AM, Wilsch-Bräuninger M, Dubreuil V, Janich P, Langenfeld tion in the rat hindbrain. Mech Dev 128:289–302.
K, Th iele C, et al. 2009. Release of extracellular membrane vesicles Tong SK, Mouriec K, Kuo MW, Pellegrini E, Gueguen MM, Brion F,
from microvilli of epithelial cells is enhanced by depleting membrane et al. 2009. A Cyp19a1b-gfp (aromatase B) transgenic zebrafish line
cholesterol. FEBS Lett 583:897–902. that expresses gfp in radial glial cells. Genesis 47:67–73.

60 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
Topp S, Stigloher C, Komisarczuk AZ, Adolf B, Becker TS, Bally-Cuif Yokota Y, Kim WY, Chen Y, Wang X, Stanco A, Komuro Y, et al. 2009.
L. 2008. FGF signalling in the zebrafish adult brain: association of The Adenomatous polyposis coli protein is an essential regulator of
Fgf activity with ventricular zones but not cell proliferation. J Comp radial glial polarity and construction of the cerebral cortex. Neuron
Neurol 510:422–439. 61:42–56.
Viehweg J, Naumann WW, Olsson R. 1998. Secretory radial glia in Yurco P, Cameron DA. 2005. Responses of Müller glia to retinal injury
the ectoneural system of the sea star Asturias rubens. Acta Zool in adult zebrafish. Vis Res 45:991–1002.
79:119–131. Zhang J, Woodhead GJ, Swaminathan SK, Noles SR, McQuinn ER,
Yamasaki M, Yamada K, Furuya S, Mitoma J, Hirabayashi Y, Watanabe Pisarek AJ, et al. 2010. Cortical neural precursors inhibit their own
M. 2001. 3-Phosphoglycerate dehydrogenase, a key enzyme for differentiation via N-cadherin maintenance of beta-catenin signaling.
l-serine biosynthesis, is preferentially expressed in the radial glia/ Dev Cell 18:472–479.
astrocyte lineage and olfactory ensheathing glia in the mouse brain.
J Neurosci 21:7691–7704.

R ADIAL GLIAL CELLS • 61


6.
STRUCTURE AND FUNCTION OF OLIGODENDROCY TES
Arthur M. Butt

A B B R E VI AT I O N S integrate historical studies such as these with more recent


findings using immunohistochemistry, modern imaging tech-
CAII carbonic anhydrase II niques, and innovative experimental paradigms, such as tissue
CNS central nervous system culture, transgenic mice, and zebrafish.
CNP 2′,3′-cyclic nucleotide 3′-phosphodiesterase
Cx connexin
eGFP enhanced green fluorescent protein 2 F R O M R I O H O RT E G A TO T H E
EM electron microscopy P R E S E N T DAY
GFAP glial fibrillary acidic protein
GFP green fluorescent protein
2.1 R I O H O RT EG A’S O L I G O D E N D RO C Y T E
Kv voltage-gated potassium channel
P H E N OT Y P E S I TO I V
MAG myelin associated glycoprotein
MBP myelin basic protein Del Rio Hortega classified oligodendrocytes into types I to
MOG myelin oligodendrocyte glycoprotein IV, based on their morphological characteristics of the num-
MRI magnetic resonance imaging ber and orientation of their cellular processes, shape and
MS multiple sclerosis size of their somata, size of the axons with which they were
Nav voltage-gated sodium channel associated, and their distributions within the CNS (Penfield
Nfasc155 neurofascin 155 1932). Oligodendrocyte phenotypic diversity is largely
OPC oligodendrocyte progenitor cell neglected, but has been confirmed by EM, intracellular dye
PDGFR platelet-derived growth factor receptor injection, immunohistochemistry, and more recently expres-
PLP proteolipid protein sion of reporter genes (Berry et al. 1995; Bjartmar et al.
1994; Butt et al. 1994, 1998a; Murtie et al. 2007; Remahl
and Hildebrand 1990a,b; Stensaas and Stensaas 1968a,b;
1 INTRODUCTION Vinet et al. 2010; Weruaga-Prieto et al. 1996a,b). Our studies
indicated that oligodendrocytes can be broadly subdivided
Oligodendrocytes are defined by their singular function— into two distinct phenotypes defined by the caliber of axons
they produce the myelin sheaths that insulate axons in the within the unit, respectively, below and above a diameter of
central nervous system (CNS). Myelin is one of the most 2 to 4 μm, and corresponding to Del Rio Hortega’s types I/II
complex biological structures and is absolutely essential for and III/IV (Butt et al. 1998a). Type I and II units have small
rapid neuronal communication that underpins the mas- rounded somata, 10 to 12 μm in diameter, from which four or
sive integrative computing power of the human brain. more fine primary processes extend and branch to myelinate
Consequently, the loss of myelin has devastating effects on 10 to 30 small axons, less than 2 μm in diameter (Fig. 6.1A,B).
CNS function. In addition to myelin-forming oligodendro- Type I and II oligodendrocytes are similar but can be dis-
cytes, the CNS contains significant populations of “adult tinguished by the alignment of axons they myelinate, which
oligodendrocyte progenitors” (OPCs) and non-myelinating pass in multiple directions and are more widely distributed in
“perineuronal” or “satellite” oligodendrocytes, which can type I units (see Fig. 6.1A), whereas type II units have paral-
generate myelin-forming oligodendrocytes throughout life lel arrays of myelin segments situated within a short distance
and following injurious insults. This chapter deals with the of a centrally located cell body (see Fig. 6.1B). Type I/II oli-
morphology of oligodendrocytes and their myelinating func- godendrocytes are the most generally studied, because they
tion and is indebted to authoritative articles that regretfully are the main population throughout white and gray matter
are slowly disappearing from the literature. Del Rio Hortega with axons of less than or equal to 2 μm. Three-dimensional
is credited with first identifying oligodendrocytes—naming analyses of immunostained and dye-injected type II oligo-
them from the Greek for cell with few processes (Penfield dendrocytes in the optic nerve, cerebellum, spinal cord, and
1932), and the cellular connection between oligodendrocytes cortex has demonstrated a relative uniformity in their over-
and myelin sheaths was demonstrated by electron microscopy all appearance, but this disguises a high degree of heteroge-
(EM) (Hirano 1968; Peters 1964). The emphasis here is to neity in respect to the number of myelin segments per unit

62
(5–50) and the internodal lengths (50–300 μm) (Berry et al. A C E
1995; Butt et al. 1994,1998a; Weruaga-Prieto et al. 1996a,b).
Similarly, studies in mice expressing green fluorescent protein
(GFP) under the control of the proteolipid protein (PLP)
or CNPase (2′,3′-cyclic nucleotide-3′-phosphodiesterase)
gene promoters, in combination with confocal microscopy
and computerized cell tracing systems, have characterized
the morphology of type I oligodendrocytes in the mouse
frontal cortex and hippocampus (Murtie et al. 2007; Vinet
et al. 2010). In mice expressing CNPase-eGFP, three variants
of type I oligodendrocytes were identified in the hippocam- B D
pus, as ramified, stellar, or smooth oligodendrocytes, distin-
guished by decreasing complexity, and that may represent
stages of a maturation process (Vinet et al. 2010). In mice
expressing PLP-eGFP, type I oligodendrocytes in the frontal
cortex were remarkably homogeneous and appeared more
complex with much shorter myelin internodes than the pat-
terns described for type II oligodendrocytes (Butt et al. 1994;
Murtie et al. 2007). Unlike earlier studies using dye injection
and the Rip antibody, studies using the eGFP reporter did
not demonstrate by EM that the reporter completely fills
the entire internodal length, and this may explain the short
internodes observed. However, short internodes are a char-
acteristic of late-forming oligodendrocytes and of remyelina-
tion, and variants of type I oligodendrocytes in the cortex Figure 6.1 Immunolabeling of oligodendrocyte phenotypes with the
Rip antibody in whole mounts of adult rat anterior medullary velum.
and hippocampus may reflect the high degree of remodelling A,B. Multipolar oligodendrocytes supporting numerous myelin sheaths
that continues long into adulthood in these regions. Type for small-caliber fibers, corresponding to Rio Hortega’s type I (A) and
III oligodendrocytes are localized to areas in which axon type II (B). Type I oligodendrocytes have multiple branching processes
diameters are greater than 2 to 4 μm, such as the cerebral and supporting numerous radially oriented myelin sheaths, whereas type II
cerebellar peduncles, the medulla oblongata, and the spinal oligodendrocytes have fewer branching processes that support parallel
myelin sheaths, but otherwise cell bodies of type I and II are difficult to
cord. Type III oligodendrocytes have larger ovoid, irregular, distinguish. C. Type III oligodendrocyte with a large cell body and small
or elongated cell bodies, 12 to 15 μm diameter, which are number of stout processes (arrows) that engage large-caliber fibers; as in
often applied directly to an axon, with one or more thick this case, type III oligodendrocytes often have a cell body directly applied
primary processes that rarely branch and myelinate a small to a large-caliber fiber, which is a phenotypic characteristic of type IV oli-
number of axons, usually less than five, with external sheath godendrocytes. D. A transitional oligodendrocyte with features of type II
and III phenotypes, with a large stout process extending to a large-caliber
diameters ranging from 4 to 15 μm (Fig. 6.1C). There are also fiber, and multiple fine branching processes that myelinate small-caliber
transitional forms between oligodendrocyte phenotypes I/II fibers. E. Type IV oligodendrocyte with cell body directly applied to a
and III units that contain both small and large-caliber axons single large caliber fiber (asterisk), with a long single internode (nodes
(Fig. 6.1D). Finally, type IV oligodendrocytes are restricted indicated by large arrows). Arrowheads in (B) show Rip staining at points
to tracts containing the largest diameter fibers greater than of engagement of oligodendrocyte processes with internodal myelin
sheaths; this cell appears to extend processes to consecutive internodal
10 μm in diameter, and occur near the entrance of nerve myelin segments (arrowheads) either side of a node (arrow). Spiraling of
roots into the CNS. Type IV unit somata do not have pro- the cytoplasmic tongue process is clear in larger fibers (arrows in D and
cesses and they form a single long myelin sheath over a large arrowheads in E). The number of fibers engaged by each unit is inversely
diameter fiber (Fig. 6.1E). The rarity of type IV oligodendro- proportional to fiber diameters, but diameters vary within individual units
cytes in the rodent brain may be related to the modest size of (C). Scale bar = 25 μm in A–D and 50 μm in E. (A–E) From Berry et al.
1995, with permission.
the largest diameter fibers, compared with humans and other
vertebrates where they are more common (Anderson et al.
1999; Hildebrand and Hahn 1978; Hildebrand et al. 1993;
Remahl and Hildebrand 1990a,b; Stensaas and Stensaas
1968a,b). Hence, oligodendrocyte phenotypes differ in the or small numbers of long myelin sheaths for large diameter
number of axons within the unit, the diameters of their axons. Thus, in rodents, type I oligodendrocytes support 30
myelinated fibers, and their internodal lengths (Berry et al. or more small caliber fibers with short internodal lengths
1995; Bjartmar et al. 1994; Butt et al. 1998a; Hildebrand and of 10 to 50 μm, type II oligodendrocytes support 10 to 30
Hahn 1978; Hildebrand et al. 1993; Remahl and Hildebrand small-caliber fibers with internodal lengths between 50 and
1990a,b). These morphological parameters are related, 350 μm, type III oligodendrocytes support 2–5 large-caliber
whereby oligodendrocytes tend either to support a large fibers with internodal lengths of around 400 μm, and type
number of short myelin sheaths for small diameter axons, IV units support a single large-caliber fiber with internodal

S T RU C T U R E A N D F U N C T I O N O F O L I G O D E N D R O C Y T E S • 63
length of up to 1,000 μm. However, there does not appear to 2.3 T H E G E N E R A L I Z E D
be a strict phenotypic segregation, type I to IV units are the O L I G O D E N D RO C Y T E
main variants of what appears to be a morphological contin-
The generalized view is based on EM, immunohistochem-
uum, and transitional forms are unexceptional (Berry et al.
istry, and intracellular dye-filling of the type II oligo-
1995; Butt et al. 1998a). The phenotypic differences between
dendrocyte phenotype. Visualization of the cytoplasmic
oligodendrocytes are not trivial, because fiber diameter and
compartments in oligodendrocytes and myelin using intra-
internodal length determine axonal speed of conduction.
cellular dye-filling showed that the inner and outer cyto-
Hence, axons within type III/IV units conduct the fastest,
plasmic ridges (tongue processes or mesaxons) of the myelin
with conduction velocities of up to 80 to 120 m/second,
sheath “corkscrew” around the axon (Fig. 6.3A) (Berry et al.
whereas those in type I/II units have much slower conduc-
1995; Butt and Ransom 1989). Dye-filling of oligodendro-
tion velocities, generally less than 20 m/second.
cytes demonstrated distinct networks of cytoplasmic inter-
connections (reticulations) or “pockets” within the myelin
2.2 U LT R A S T RU C T U R E O F sheath that included Schmidt-Lanterman clefts and provide
O L I G O D E N D RO C Y T E S cytoplasmic access to the compacted myelin (Fig. 6.3B)
The ultrastructural characteristics of oligodendrocytes were (Berry et al. 1995; Butt and Ransom 1989; Ransom et al.
defined many years ago and have been reviewed in consider- 1991; Velumian et al. 2011). It takes more than 1 hour for
able detail (Peters et al. 1991). Ultrastructurally, oligoden- small (<500 Da) molecules to diffuse along the whole
drocytes can be identified by their generally round, dark myelin sheet (Velumian et al. 2011). If unwrapped, each
nuclei, which are surrounded by dark cytoplasm contain- myelin sheath would appear as an extraordinarily large trap-
ing short granular endoplasmic reticulum, polyribosomes, ezoid sheet that is an extension of the oligodendroglial cell
short mitochondria, and Golgi apparatus (Sandell and membrane (Ransom et al. 1991). The compacted myelin
Peters 2002). The nuclei contain dense patches of chroma- sheet is circumscribed by a continuous cytoplasmic ridge
tin and there is a dense uneven layer of chromatin beneath that is connected to the cell body via the processes and pro-
the nuclear envelope. Hildebrand et al. provided EM vides a conduit for transport of the myelin constituents from
three-dimensional reconstruction of oligodendrocyte phe- the cell body to the myelin. The myelin sheath is wrapped
notypes I/II (Fig. 6.2A,B) and III/IV (Fig. 6.2C,D), and around the axon to form concentric lamellae and the cyto-
identified no outstanding differences in their ultrastructure plasmic ridges stack up to form the paranodal loops at nodes
(Remahl and Hildebrand 1990a). of Ranvier (Fig. 6.3C,D). Oligodendroglial paranodal loops

A C

B D

Figure 6.2 Electron Micrograph Reconstructions of Oligodendrocytes. A,B. Type II oligodendrocyte from feline corpus callosum, 21 days postnatal.
The oligodendrocyte myelinates 11 nearby axons (A), indicated by asterisks in the electron micrograph (B), which corresponds to the plane marked in
(A). C,D. Type IV oligodendrocyte from feline spinal cord of 47-day-old fetus. The oligodendrocyte is attached to a single axon that it myelinates (A),
indicated by the asterisk in the electron micrograph (B). There are no ultrastructural distinguishing features between cell bodies and nuclei of type II and
IV units (indicated by O in B and D). From Remahl and Hildebrand 1990a, with permission.

64 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
form complex axoglial junctions that appear as “transverse 3 F U N C T I O N A L I M P L I C AT I O N S
bands” in EM (Fig. 6.3D), which are important in the separa- OF OLIGODENDROCYTE
tion of ion channels at nodes of Ranvier (Charles et al. 2002; P O LY M O R P H I S M
Hinman et al. 2006; Mierzwa et al. 2010; Zonta et al. 2008)
and involves a neurofascin155 (Nfasc155)-Caspr-Contactin
3.1 MY E L I N VO LUM E , SY N T H E S I S ,
axoglial adhesion complex.
A N D M ETA B O L I S M
There are direct relationships between fiber diameter (D) and
the number of myelin sheaths per oligodendrocyte unit (n),
A and both the longitudinal (internodal lengths, L) and radial
(number of lamellae, N) dimensions of the myelin sheaths
(Butt et al. 1998a). The relationship between axon diam-
eter and the number of myelin lamella is a strict 1:10 ratio,
defined as the g-ratio (Friede 1972). Each myelin sheath has
dimensions of approximately π.D.N.L. and multiplying this
by the number of myelin segments within the oligodendro-
cyte unit (n) provides the total volume of myelin supported
by oligodendrocytes. There is a clear demarcation between
type I/II and type III/IV units, which respectively support
myelin volumes of approximately 500 and 30,000 μm3 (Butt
et al. 1998a). However, this is not an absolute segregation,
because transitional type II/III units contain both small-
and large-caliber axons and support intermediate volumes of
myelin. As noted, type I units in the cerebral cortex have 30
or so very short internodal lengths for small diameter axons
and so support the smallest volume of myelin (Murtie et al.
2007). In contrast, type IV units in which internodal lengths
attain 1,000 μm support the largest volumes of myelin, as
B
great as 1,50,000 μm3 (Butt et al. 1998a). During active myeli-
nation, myelin biosynthesis in type II oligodendrocytes has
1μm been calculated to be 5 to 50 × 103 μm2/cell per day (Baron
C and Hoekstra 2010), and by extrapolation myelin biogenesis
would be more than 100 times greater in type III/IV oligoden-
drocytes. Greater myelin metabolism and turnover has been
1μm noted in large diameter CNS fibers (Hildebrand et al. 1993),
D and oligodendrocyte polymorphism signifies the greater met-
abolic demand of supporting a much larger mass of myelin in
type III/IV oligodendrocytes compared with type I/II units.
Hence, type III/IV units have large cell bodies, thick radial
processes, and prominent Schmidt-Lantermann incisures,
40 μm 0.1μm which facilitate the substantial distribution of myelin gene
products. In contrast, type I/II units have small somata and
Figure 6.3 Oligodendroglial Cytoplasmic Compartments. Confocal long fine processes, reflecting the lower mass of myelin they
microscopy of oligodendrocyte intracellularly dye filled with lysinated support.
rhodamine dextran (A,B) and freeze-fracture EM (C) of oligodendro-
cyte in the whole-mounted adult rat anterior medullary velum, and
transmission EM in mouse spinal cord (D). A. Intracellular dye-filling 3.2 MY E L I N B I O C H E M I S T RY
resolves the oligodendroglial cytoplasmic compartments of this large AND FUNCTION
type III oligodendrocyte unit, which has five axons with external
sheath diameters of 10 to 15 μm and internodal lengths of around The main constituents of myelin are lipids (70% of its dry
400 μm. Myelin sheaths have prominent cytoplasmic reticulations weight) and proteins (30% of the dry weight), many of which
and the dye-filled cytoplasmic tongue processes spiral along the axons
(large arrows) and delineate the paranodal loops (small arrows). B.
are specific to myelin and are used to identify oligodendro-
High-contrast zoom illustrating internodal cytoplasmic compartments cytes by immunohistochemistry, or their genes are used to
forming pockets between compacted myelin, which is occluded to the drive expression of fluorescent reporter proteins (see chap-
dye and appears black. C. Scanning electron micrograph of myelinat- ters 13 and 44). Central nervous system myelin lipids are rich
ing fiber in the postnatal anterior medullary velum of a 15-day-old rat in the glycosphingolipids galactocerebroside (GalC) and the
showing the paranodal loops (between arrows) spiraling toward the node
of Ranvier (asterisk). D. Electron micrograph showing transverse bands
sulfated derivative recognized by the O4 antibody, which
at paranodes (arrowheads). (A–C) From Berry et al. 1995; (D) from are used as specific markers for oligodendrocytes (Raff et al.
Mierzwa et al. (2010), with permission. 1978; Sommer and Schachner 1981). The major CNS myelin

S T RU C T U R E A N D F U N C T I O N O F O L I G O D E N D R O C Y T E S • 65
proteins are proteolipid protein (PLP, 50% of myelin pro- of the large acid shifts in extracellular pH during axonal
tein) and myelin basic protein (MBP, 30% of myelin protein), activity in CNS white matter (Kettenmann et al. 1990;
which are not specific to CNS myelin (Griffiths et al. 1998a; Sykova 1989), and would provide a protective mechanism
Sternberger et al. 1978). The enzyme CNPase (2′,3′-cyclic for intracellular pH regulation in hypoxic stress and during
nucleotide-3′-phosphodiesterase) is expressed in the CNS lactate uptake (Ransom et al. 1992; Rinholm et al. 2011).
specifically by oligodendrocytes and immunohistochemi- Oligodendrocytes also express acid-sensing ion channels
cally is localized to the cell body and processes rather than that may contribute to their vulnerability to CNS ischemia
compacted myelin (Lappe-Siefke et al. 2003). The myelin (Feldman et al. 2008) (see chapters 20 and 52). In addition,
glycoproteins MOG (myelin oligodendrocyte glycoprotein) oligodendrocytes express the highest levels of iron in the
and MAG (myelin-associated glycoprotein) are also used to brain, where it is a basic requirement for oxidative metabo-
identify oligodendrocytes; MAG is expressed in both CNS lism, and differences in iron-mediated oxidative stress are
and PNS myelin (Schachner and Bartsch 2000), whereas most likely involved in susceptible oligodendrocyte popu-
MOG is CNS-specific and a key autoantigen for primary lations (Todorich et al. 2009) (see chapter 46). A number
demyelination in multiple sclerosis (MS) (Clements et al. of studies indicate that type I/II oligodendrocytes may be
2003). Oligodendrocytes also express many other proteins, more susceptible to injury and demyelination than type III/
such as oligodendrocyte-specific protein OSP/claudin-11 IV oligodendrocytes. Late developing type I/II oligoden-
(third most abundant protein in myelin), gap junction con- drocyte units are most susceptible in periventricular white
nexins (Cx32 and Cx47), Nogo, isoform II of carbonic matter injury caused by perinatal ischemic insult (Riddle
anhydrase (CAII), and transferrin, which have multifarious et al. 2006), and in tauopathies and metachromatic leu-
functions (see chapters 13, 43, 44). kodystrophy (Hildebrand et al. 1993). In the taiep mutant
The majority of studies have been on type II oligoden- rat, tauopathy more severely affected type I/II oligodendro-
drocytes, which express all of the aforementioned proteins, cytes than type III/IV oligodendrocytes (Song et al. 2001).
and there have been few studies on biochemical differences These studies indicate that regional predilections to injury
between oligodendrocyte phenotypes I/II and III/IV (Butt are related to differences in susceptibility of oligodendro-
and Berry 2000). Proteolipid protein (PLP) expression may cyte phenotypes.
vary as a function of axon caliber, the smallest myelinated
fibers containing higher levels of PLP relative to MBP than
the largest myelinated fibers (Hartman et al. 1982). There 4 D E VE L O PM E N T O F
is evidence that PLP serves a distinct function in sup- O L I G O D E N D R O C Y T E P H E N OT Y P E S
porting the integrity of type II units, whereby there was
greater disruption in small-diameter axons in the absence
4.1 O L I G O D E N D RO C Y T E P H E N OT Y P I C
of PLP-DM20 (Griffiths et al. 1998b). Similarly, CNP
D I VE RG E N C E
appeared to be more critical for the formation of a normal
inner tongue process in small diameter axons that are myeli- The mechanisms determining axon growth and oligoden-
nated by type II oligodendrocyte units (Edgar et al. 2009). drocyte phenotype divergence are unresolved, but they are
Studies on oligodendrocyte subtypes have indicated differ- entirely interdependent (see chapters 13, 44, and 45). There
ential phenotypic expression of the small isoform of MAG are direct relationships among the age at which axons are
(S-MAG) and CAII, with type I/II units being S-MAG –/ myelinated, their final diameter in the adult, and the develop-
CAII+ and type III/IV being S-MAG+/CAII– (Butt et al. mental divergence of oligodendrocyte phenotypes (Bjartmar
1998b, 1995). S-MAG is essential for normal myelination et al. 1994; Butt et al. 1997; Hildebrand et al. 1993; Remahl
in the PNS (Schachner and Bartsch 2000), suggesting a and Hildebrand 1990a,b). Thus, prospective large-diameter
specific role for this isoform in maintaining the integrity fibers (>4 μm) are myelinated early in development by type
of type III/IV oligodendrocyte units (Butt et al. 1998b); III/IV oligodendrocytes that diverge before birth in rodents,
the large isoform of MAG (L-MAG) is the most important whereas prospective small diameter fibers (<2 μm) are myeli-
functionally in the CNS (Schachner and Bartsch 2000), nated later in development, by type I/II oligodendrocytes
and is expressed by all oligodendrocyte phenotypes (Butt that differentiate perinatally in rodents (Butt et al. 1997).
et al. 1998b). No major abnormalities were observed in the However, oligodendrocytes are not inherently programmed
brains of CAII-deficient mice (Ghandour et al. 1989), and to myelinate a specific size of axon and there are no evident
the functional significance of CAII, specifically in type I/II morphological or physiological differences between early and
units, is unclear. Carbonic anhydrase is a key enzyme in cel- late developing OPCs (Butt et al. 1997; Tripathi et al. 2011).
lular pH regulation, and fluorescence correlation spectros- Oligodendrocyte progenitor cells that normally develop into
copy analysis of CAII microinjected into oligodendrocytes type I/II oligodendrocytes will myelinate large diameter
revealed it exists both as a freely diff using protein through- fibers when transplanted into experimentally demyelinated
out the cell as well as in microdomains associated with Na+/ spinal cord funiculi of the rat (Fanarraga et al. 1998), and in
H+ exchangers in the somata and Na+/HCO3– cotransporter zebrafish, oligodendrocytes that normally myelinate numer-
in the processes (Ro and Carson 2004). Specific expres- ous smaller caliber axons readily myelinate much larger cali-
sion of CAII in type I/II oligodendrocytes may reflect a ber supernumerary axons (Almeida et al. 2011). The existence
greater capacity for intracellular pH regulation in the face of transitional type II/III oligodendrocyte units containing

66 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
both small and large caliber axons clearly points to local con- 1997; Hardy and Friedrich 1996): (1) axonal contact and
trol of myelinated fiber dimensions (Berry et al. 1995; Butt recognition by OPCs; (2) induction phase, in which OPCs
et al. 1998a; Friede 1972). extend initiator processes along receptive axons to form short
ensheathing segments, triggering the differentiation of OPCs
into a premyelinating oligodendrocyte phenotype and initia-
4.2 I N T E R D E P E N D E N T D EV E L O PM E N T O F
tion of axonal ion channel clustering; (3) remodeling phase,
O L I G O D E N D RO C Y T E -AXO N U N I T S
in which oligodendrocyte-axon unit phenotype is established
Oligodendrocyte phenotypic divergence and myelination by the loss of non-myelinating processes within the unit, and
within units occurs in a series of distinct axoglial interde- radial and longitudinal growth of myelinating processes forms
pendent phases (Fig. 6.4) (Butt and Berry 2000; Butt et al. incipient internodal segments, induces axon radial growth
and establishes nodes of Ranvier; and (4) maturation phase,
in which axons and myelin sheaths within units undergo
interdependent growth to establish adult dimensions of axon
diameter, myelin sheath g-ratios, and intermodal lengths.
Differentiation of oligodendrocyte phenotypes, and there-
fore the size of the myelin sheaths and speeds of conduction
of axons within the axoglial units, are driven by complex axo-
glial interactions that are only now being unraveled (Baron
and Hoekstra 2010) (see chapters 44 and 45). The develop-
mental onset of myelination involves a number of axoglial rec-
ognition and adhesion events that regulate the production of
myelin-related gene products and radial axon growth, thereby
governing phenotypic changes in oligodendrocyte-axon units
(see chapter 44). However, axon contact per se is not the
myelin inducting factor, because many OPCs that contact
cGFP PLP/DM20 SMI31 20μm
axons do not differentiate into myelin-forming oligodendro-
cytes, and within individual OPCs and premyelinating oli-
godendrocyte units, many processes that contact axons do
not go on to form myelin (see Fig. 6.4A–C) (Bjartmar et al.
1994; Butt and Ransom 1993; Butt et al. 1997; Hardy and
Friedrich 1996). It is not known why some axons trigger dif-
ferentiation and myelination in certain OPCs, and the rules
of competition between processes within individual units are
unknown. All axons are submicron at the time of myelin ini-
tiation, and the mechanisms governing the divergent growth
of oligodendrocyte phenotypes and axons within the units are
mystifying. Furthermore, developmental myelination along
individual axons is multifocal and asynchronous (Butt et al.
1997; Remahl and Hildebrand 1990b; Skoff 1978), and it is
10μm not known how axoglial recognition signals are spatiotempo-
rally regulated along the length of individual axons.
Figure 6.4 Axon-Oligodendrocyte Relations in Developing Oligodendrocyte
Units In Vivo (A,B,E) and In Vitro (C,D). A,B. Confocal z-stacks of intact 4.3 AXO N- O L I G O D E N D RO C Y T E
anterior medullary vela from postnatal day 10 rat, double immunofluo-
rescence labeled with NG2 (green) for OPC and neurofilament for axons R EC O G N IT I O N S I G NA L S
(red). A. Two OPC lie serially along an axon that they both contact, with
the OPC on the left extending a process that on contact with the axon
The first step in myelination is a recognition event between
bifurcates and this “initiator” process coils along the axon in a bidirectional OPC processes and an axonal segment that is receptive for
manner before ensheathment, whereas the OPC on the right forms multiple myelination. There is a role for β1 integrin in the axoglial
contacts with the same axon via fine processes that extend short lamel- interactions that initiates myelination and the loss of integ-
lipodia on the axon. B. Single OPC forming multiple initiator processes rin signaling leads to a delay in myelination of small-diameter
along numerous axons within its domain. C,D. Cocultures of neurons with
cyto-GFP labeled oligodendrocytes immunostained for PLP, illustrating
axons (Camara et al. 2009). Interactions between axonal
a z-stack (C) and serial single z-section analysis (D), showing the spiral- Caspr and oligodendroglial Nfasc155 are also indicated
ing cytoplasmic tongue process extending around the PLP+ myelin that in early myelination, axonal Caspr distributing as a helical
ensheathes a segment of the axon. E. Transitional oligodendrocyte in vivo coil that winds around the axon to interact with Nfasc155
in the postnatal day 10 rat anterior medullary velum immunolabeled with on the overlying myelinating process (Pedraza et al. 2009).
Rip, illustrating the cytoplasm-filled processes forming ensheathing cuffs
around the axon. (A,E) From Butt, Ibrahim, and Berry 1997; (C,D) from
Three-dimensional imaging of whole oligodendrocyte units
Ioannidou et al., with permission. (B) Unpublished data from Butt, with in the anterior medullary velum and live cell imaging in
permission. brain slices and in cultures have verified that the initiator

S T RU C T U R E A N D F U N C T I O N O F O L I G O D E N D R O C Y T E S • 67
cytoplasmic processes run in a bidirectional coil along the 4.4 O L I G O D E N D RO C Y T E -AXO N
axon (Fig. 6.4A–D) (Butt et al. 1997; Ioannidou et al. 2012; I N T E R AC T I O NS A N D N O D E S O F R A N VI E R
Sobottka et al. 2011). Subsequently, growth of the ensheath- The formation and maintenance of nodal structure are key
ing process around an individual axon does not occur in a uni- functions of oligodendrocytes. During development, solu-
form and synchronous manner, but there are focal expansions ble factors secreted by oligodendrocytes promote the clus-
of the corkscrew process to form short cytoplasm-filled cuffs tering of Nav1.2 α subunits at immature nodes of Ranvier,
around the axon (Fig. 6.4C–E) (Butt et al. 1997; Ioannidou and myelin ensheathment induces the clustering of Nav1.6
et al. 2012; Remahl and Hildebrand 1990b). A current “liquid α subunits, which are characteristic of mature nodes (Boiko
croissant” model of myelination envisages concentric wrap- et al. 2001; Kaplan et al. 2001). Mature nodes of Ranvier
ping of multiple membrane layers around axons (Sobottka et are further characterized by the formation of axoglial inter-
al. 2011). The myelin sheath appears to wrap around the axon cellular “transverse bands,” which anchor oligodendroglial
in a concertina fashion and, as the number of wraps increases cytoplasmic paranodal loops to the axon (Rasband and
and internodal myelin compaction expands to establish an Trimmer 2001; Rosenbluth 2009). The paranodal axoglial
incipient internode, the spiraling cytoplasmic processes are junctions are formed by an adhesion complex between oli-
squeezed to the paranode and stack up at the developing node godendroglial Nfasc155 and the axonal proteins Caspr and
of Ranvier (see Fig. 6.3C) (Berry et al. 1995; Hildebrand and Contactin (Charles et al. 2002; Poliak et al. 2003; Rios
Waxman 1984; Pedraza et al. 2009; Remahl and Hildebrand et al. 2003). Although not essential for the initial cluster-
1990a,b). Within oligodendrocyte units, secondary pro- ing of sodium channels, paranodal axoglial junctions are
cesses that do not form myelin are retracted during remod- essential for the maintenance of the nodal complex, and
eling (Butt et al. 1997; Butt and Ransom 1993; Hardy and mice lacking either Caspr or contactin have everted para-
Friedrich 1996; Remahl and Hildebrand 1990a). Myelinating nodal loops and absent transverse bands, resulting in dis-
processes engage their axons through interactions involving placement of potassium and sodium channels (Poliak et al.
the oligodendroglial ligands integrin α6β1 and/or dystrogly- 2003; Rios et al. 2003). The periodicity of nodes of Ranvier
can and their respective axonal receptors L1 and laminin-2 along axons is directly and positively related to axon cali-
(Baron and Hoekstra 2010; Camara et al. 2009; Laursen et al. ber (Ibrahim et al. 1995). Internodal lengths are relatively
2009). This is followed by growth of the axon and myelin uniform within oligodendrocyte units, generally less than
sheath, which studies in rodents and zebrafish clearly point 50 μm in cortical type I oligodendrocytes, 100 to 200 μm
to being interdependent (Buckley et al. 2010; Colello et al. in type II units, and 400 to 500 μm in type III units (Butt
1994). Myelin growth depends on laminin2-α6β1integrin et al. 1998a; Murtie et al. 2007), and nodal periodicity is
and neuregulin1-ErbB signaling between axons and oligo- established developmentally before myelin compaction
dendrocytes (Chun et al. 2003; Roy et al. 2007). L1 binding (Butt et al. 1997; Butt and Ransom 1993). This raises the
to oligodendrocytes and interactions between oligodendro- question of how the regular spacing of oligodendrocytes
glial integrin and axonal contactin regulate myelin forma- along axons is determined. As axons grow in length, unmy-
tion by controlling Fyn activity (Laursen et al. 2009; White elinated gaps are myelinated by late developing oligoden-
et al. 2008). Neurons stimulate the transport of PLP to the drocytes, and this continues well into adulthood (Butt
plasma membrane (Trajkovic et al. 2006), and axonal elec- et al. 1997; Murtie et al. 2007; Peters and Sethares 2004;
trical activity stimulates localized synthesis of MBP through Rivers et al. 2008), which might explain the extremely short
Fyn kinase-dependent signaling (Wake et al. 2011) (see chap- internodes observed in oligodendrocytes of late myelinated
ter 45). Likewise, myelin proteins are essential for axon radial areas such as the cortex.
growth. For example, when both OSP/claudin-11 and PLP
genes are knocked out, mice exhibit markedly smaller axon
4.5 AG E -R E L AT E D C H A N G E S I N
diameters (Chow et al. 2005). In addition to controlling axon
O L I G O D E N D RO C Y T E S
growth, oligodendrocytes are also essential for axon integ-
rity (Edgar and Nave 2009). Mice developed widespread Electron microscopy studies have provided evidence of
axon degeneration in the absence of PLP-DM20 (Griffiths age-related degenerative changes in myelin, such as split-
et al. 1998b) or Cnp1 (Lappe-Siefke et al. 2003), and mice ting of sheaths, and blebbing or ballooning of sheaths, and
deficient in both CNP and PLP display more severe axonal both short internodes and thin sheaths can be found, as
degeneration (Edgar et al. 2009). A study in sulfatide-null occurs in remyelination (Peters 2002; Peters et al. 2008).
mice indicated an interesting function for this myelin lipid, The conclusion is that with age some internodal lengths
which played a limited role in myelin development, but was of myelin sheaths degenerate and are replaced by newer
essential for myelin maintenance and axonal integrity in aged and shorter internodal lengths by newly generated oligo-
animals; in the absence of sulfatide displayed degenerating dendrocytes, which originate from endogenous OPCs and
myelin sheaths, deteriorating nodal/paranodal structure and OPCs derived from the subventricular zone (Peters and
decreased axonal caliber (Marcus et al. 2006). Thus, oligoden- Sethares 2004; Rivers et al. 2008). Age-related changes
drocytes and axons are entirely interdependent and should be in paranodal structure have also been noted, and affect
considered functional units rather than separate functional axonal protein localization at nodes of Ranvier, including
entities per se. Kv1.2 and Caspr, resulting in changes in axonal conduction

68 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
(Hinman et al. 2006). The advent of magnetic resonance et al. 1999; Levine and Card 1987; Peters 2004; Peters and
imaging (MRI) provides a noninvasive method for examin- Sethares 2004). Electron microscopy and physiological stud-
ing age-related changes in myelin, and these studies suggest ies show NG2-glia form functional synapses with neurons and
that metabolically vulnerable late-differentiating oligoden- axons and respond to glutamate and GABA released at synapses
drocytes may contribute to the cognitive deficits observed (Fig. 6.5B–E) (Bergles et al. 2000; Butt et al. 1999; Haberlandt
in normal aging and dementias through disruption of rapid et al. 2011; Hamilton et al. 2010; Kukley et al. 2007; Velez-Fort
transmission and the loss of synchronization of higher cog- et al. 2010) (see chapter 21). There is no direct evidence of a
nitive functions (Bartzokis et al. 2004). Oligodendrocyte function for NG2-glia at synapses, but it is presumed that syn-
and myelin dysfunction are also implicated as primary aptic signaling plays a role in sustaining the adult population
changes in schizophrenia, resulting from disruption of syn- or regulating their differentiation into oligodendrocytes (De
aptic formation and function (Takasaki et al. 2010) (see Biase et al. 2010; Kukley et al. 2010; Velez-Fort et al. 2010). The
chapter 71). Neuregulin 1 and its receptor erbB4 are genet- density and distribution of NG2-glia in adult gray matter areas
ically linked with susceptibility to schizophrenia and bipo- such as the hippocampus and cerebral cortex highlight their
lar disorder, and the loss of erbB signaling leads to reduced importance in cognitive function by maintaining oligodendro-
myelin thickness and slower conduction velocity in CNS cytes and myelination throughout life (Bartzokis et al. 2004;
axons, as well as alterations in dopaminergic function Peters et al. 2008). There is increased frequency of degenerative
and behavior relevant to neuropsychiatric disorders (Roy changes in myelin in the normal aging primate cortex, and this
et al. 2007). Ultrastructural analysis has confirmed a 20% is partly offset by a 50% increase in the oligodendrocyte num-
decrease in the number of myelinated nerve fibers of cere- ber between young (4–10 years of age) and old (25–35 years
bral white matter fiber tracts associated with frontal lobe of age) monkeys (Peters and Sethares 2004). Additional oli-
areas critical in cognitive processing in the normal aging godendrocytes are generated from NG2-glia and are required
brain (Bowley et al. 2010). Thus, normal aging, cognitive to remyelinate nerve fibers whose sheaths have broken down,
decline, and several psychiatric disorders are associated which leads to cognitive decline (Peters and Sethares 2004;
with white matter defects, suggesting that oligodendrocyte Peters et al. 2008). In human brains, myelination extends to age
abnormalities underlie some aspects of these diseases (see 50 in cortical regions, and MRI studies suggest myelin break-
chapter 71). down accelerates as aging progresses and underlies age-related
cognitive decline (Bartzokis et al. 2004). It is clear the gen-
eration of oligodendrocytes in adult gray and white matter
5 N O N M Y E L I N AT I N G necessitates a major population of OPC and their diminished
OLIGODENDROCYTES capacity for regeneration in the aging brain is likely to be criti-
cal in cognitive decline.
5.1 A D U LT O L I G O D E N D RO C Y T E
P RO G E N I TO R C E L L S 5.2 P E R I N EU RO NA L O L I G O D E N D RO C Y T E S
Adult oligodendrocyte progenitor cells (OPC) are identified Perineuronal oligodendrocytes are apposed directly to neu-
by their expression of the platelet-derived growth factor recep- ronal cell bodies in various gray matter regions (Fig. 6.5F)
tor alpha (PDGFRαα) and the NG2 chondroitin sulfate pro- (Szuchet et al. 2011; Takasaki et al. 2010). Perineuronal oli-
teoglycan (Levine and Card 1987; Levine and Stallcup 1987; godendrocytes are the progeny of A2B5+/O4+ OPC, but
Stallcup 1981; Stallcup and Beasley 1987) (see chapters 10 and have an antigenic phenotype distinct from both OPC and
21 for further details). The sheer number of OPC throughout myelin-forming oligodendrocytes, expressing CNPase and
adult gray and white matter (Fig. 6.5A) has led many to refer A2B5, but not expressing NG2 and having low expression of
to them as a distinct fourth glial cell type, called NG2-glia Sox10, and using PDGFRαβ in contrast with PDGFRαα in
or polydendrocytes (Butt et al. 2005; Nishiyama et al. 2009; OPCs (Fig. 6.5Fi) (Szuchet et al. 2011; Takasaki et al. 2010).
Peters 2004) (see chapter 10). There has been some debate Ultrastructurally, perineuronal oligodendrocytes are oval or
about whether all NG2-glia in the adult brain are OPCs (Butt polygonal cells, with a round nucleus rich in dark hetero-
et al. 2002; Richardson et al. 2011), but fate-mapping studies chromatin, forming a concave impression on the neuron to
in transgenic mice show that NG2-glia have an oligodendro- which they are tightly attached, with parallel alignment of
glial lineage and provide a pool of slowly proliferating cells that two plasma membranes and a narrow intercellular space (Fig.
generate oligodendrocytes throughout life (Kang et al. 2010; 6.5Fii) (Takasaki et al. 2010). Like astrocytes, perineuronal
Rivers et al. 2008). In gray matter, the cell bodies of NG2-glia oligodendrocytes are immunopositive for glutamine syn-
are often applied directly to neuronal somata (Fig. 6.5B), like thase, but they do not express glial fibrillary acidic protein,
perineuronal oligodendrocytes (Fig. 6.5F), but the two cell or the glutamate transporters GLAST and GLT-1 (Takasaki
types are readily distinguished by their differential expression et al. 2010). The functions of perineuronal oligodendrocytes
of NG2 (Takasaki et al. 2010). Ultrastructurally, NG2-glia have are unclear, but they appear to fulfill a neuron metabolic sup-
elongate nuclei and a thin rim of pale organelle-poor cytoplasm port function (Takasaki et al. 2010) and protect against neu-
containing some short rough endoplasmic reticulum, small ronal apoptosis (Taniike et al. 2002). A recent study provided
mitochondria, and few free polyribosomes (Fig. 6.5E) (Butt

S T RU C T U R E A N D F U N C T I O N O F O L I G O D E N D R O C Y T E S • 69
Figure 6.5 NG2-Glia and Perineuronal Oligodendrocytes. A. Immunoperoxidase NG2 labeling illustrating the dense distribution of NG2-glia in gray
matter of the adult rat cerebral cortex. B. Immunolabeling for NG2 (red) and calbindin (blue) in the cerebral cortex of GFAP-GFP mice, illustrated
in z-stack (Bi) and single z-section (Bii), showing astrocytes and NG2-glia forming overlapping networks over the neuronal somata. C. Serial electron
micrographs illustrating the processes of an NG2-glial cell (black, peroxidase reaction) receiving a synapse (arrow) from a bouton (b) that also gives a
synapse to a dendritic spine(s) in rat hippocampus. D. Confocal microscopic reconstructions in whole-mounts of rat anterior medullary vela triple
immunolabeled for NG2 (green), MBP (blue), and ankyrin G (red), an axoskeletal protein that define nodes of Ranvier, illustrating NG2-glial cell
contacting multiple nodes (Di) and three-dimensional isoformed reconstruction of a cell process that terminates at a node of Ranvier and partly
ensheathes the axon (Dii). E. Pre-embedding EM immunocytochemistry shows the surface labeling of the NG2 membrane-embedded chondroitin
sulfate proteoglycan in a cell that sends a process (filled arrows) that wraps around the axolemma at a nearby node of Ranvier (asterisk), defined by the
paranodal loops of the oligodendroglial myelin sheath (open arrows). F. Perineuronal oligodendrocytes in the adult rat cerebral cortex, identified by
immunolabeling (Fi) and EM (Fii). Fi. CNP+ perineuronal oligodendrocyte (green, arrowhead) attached to a MAP2-positive neuron (blue, asterisk).
Fii. Electron micrograph of a perineuronal oligodendrocyte (PO) tightly contacting the soma of a neuron (Neu). (Bi,Bii) From Wigley and Butt
2009. (Ci–Ciii) from Bergles et al. 2000; (Di, Dii) from Butt et al. 2005; (E) from Butt et al. 1999; (Fi, Fii) from Takasaki et al. 2010, with permission.

evidence for a deficit of perineuronal oligodendrocytes in significant populations of cells that are capable of regen-
the prefrontal cortex in schizophrenia, bipolar disorder, and erating myelin-forming oligodendrocytes throughout life,
major depression, suggesting they may play a key role in the but their sheer number reflects the profound importance
pathophysiology of these severe mental disorders (Vostrikov of maintaining oligodendrocytes and makes them worthy
et al. 2007). of greater study. A wide array of new markers and genetic
models will enable us to unravel the mechanisms of axo-
glial interactions that control the interdependent growth
6 S U M M A RY A N D P E R S P E C T I VE S of myelin and axons within units, and how myelination is
essential for axon integrity and axoglial specialization at
Oligodendrocyte phenotypic differences determine the nodes of Ranvier. In addition, modern imaging techniques
conduction properties of axons within oligodendrocyte will greatly facilitate our understanding of the significance
units and susceptibility to damage. Oligodendrocyte phe- of oligodendrocytes and their progenitors in the aging
notype divergence is regulated by axon-derived factors, brain and pathological conditions. These new techniques
and developmentally the growth of axons and myelin will enable us to begin to understand the profound impor-
sheaths are interdependent. In recent years, major advances tance of oligodendrocytes and the devastating effects of
have been made in the study of adult OPCs and perineu- their loss on integrative brain function, as occurs in MS,
ronal oligodendrocytes, but knowledge of their functions cerebral palsy, leukodystrophies, psychological diseases,
remains scanty. It is not clear why the CNS requires two and cognitive decline in the aging brain.

70 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
AC K N OW L E D G M E N T S Butt AM, Ransom BR. 1989. Visualization of oligodendrocytes and
astrocytes in the intact rat optic nerve by intracellular injection of
lucifer yellow and horseradish peroxidase. Glia 2(6):470–475.
The author would like to thank the funding bodies who have Butt AM, Ransom BR. 1993. Morphology of astrocytes and oligoden-
supported this work, and all coworkers past and present, with drocytes during development in the intact rat optic nerve. J Comp
special thanks to Bruce Ransom and Martin Berry. Neurol 338(1):141–158.
Camara J, Wang Z, Nunes-Fonseca C, Friedman HC, Grove M, Sherman
DL, et al. 2009. Integrin-mediated axoglial interactions initiate myeli-
R E F E R E NC E S nation in the central nervous system. J Cell Biol 185(4):699–712.
Charles P, Tait S, Faivre-Sarrailh C, Barbin G, Gunn-Moore F,
Almeida RG, Czopka T, Ffrench-Constant C, Lyons DA. 2011. Denisenko-Nehrbass N, et al. 2002. Neurofascin is a glial receptor
Individual axons regulate the myelinating potential of single oligo- for the paranodin/Caspr-contactin axonal complex at the axoglial
dendrocytes in vivo. Development 138(20):4443–4450. junction. Curr Biol 12(3):217–220.
Anderson ES, Bjartmar C, Westermark G, Hildebrand C. 1999. Chow E, Mottahedeh J, Prins M, Ridder W, Nusinowitz S, Bronstein
Molecular heterogeneity of oligodendrocytes in chicken white mat- JM. 2005. Disrupted compaction of CNS myelin in an OSP/
ter. Glia 27(1):15–21. Claudin-11 and PLP/DM20 double knockout mouse. Mol Cell
Baron W, Hoekstra D. 2010. On the biogenesis of myelin membranes: Neurosci 29(3):405–413.
sorting, trafficking and cell polarity. FEBS Letts 584(9):1760–1770. Chun SJ, Rasband MN, Sidman RL, Habib AA, Vartanian T. 2003.
Bartzokis G, Lu PH, Mintz J. 2004. Quantifying age-related mye- Integrin-linked kinase is required for laminin-2-induced oli-
lin breakdown with MRI: novel therapeutic targets for preventing godendrocyte cell spreading and CNS myelination. J Cell Biol
cognitive decline and Alzheimer’s disease. J Alzheimers Dis 6(6 163(2):397–408.
Suppl):S53–59. Clements CS, Reid HH, Beddoe T, Tynan FE, Perugini MA, Johns TG,
Bergles DE, Roberts JD, Somogyi P, Jahr CE. 2000. Glutamatergic syn- et al. 2003. The crystal structure of myelin oligodendrocyte glycopro-
apses on oligodendrocyte precursor cells in the hippocampus. Nature tein, a key autoantigen in multiple sclerosis. Proc Natl Acad Sci U S
405(6783):187–191. A 100(19):11059–11064.
Berry M, Ibrahim M, Carlile J, Ruge F, Duncan A, Butt AM. 1995. Colello RJ, Pott U, Schwab ME. 1994. The role of oligodendrocytes and
Axon-glial relationships in the anterior medullary velum of the adult myelin on axon maturation in the developing rat retinofugal path-
rat. J Neurocytol 24(12):965–983. way. J Neurosci 14(5 Pt 1):2594–2605.
Bjartmar C, Hildebrand C, Loinder K. 1994. Morphological heteroge- De Biase LM, Nishiyama A, Bergles DE. 2010. Excitability and synap-
neity of rat oligodendrocytes: electron microscopic studies on serial tic communication within the oligodendrocyte lineage. J Neurosci
sections. Glia 11(3):235–244. 30(10):3600–3611.
Boiko T, Rasband MN, Levinson SR, Caldwell JH, Mandel G, Edgar JM, McLaughlin M, Werner HB, McCulloch MC, Barrie
Trimmer JS, et al. 2001. Compact myelin dictates the differential JA, Brown A, et al. 2009. Early ultrastructural defects of axons
targeting of two sodium channel isoforms in the same axon. Neuron and axon-glia junctions in mice lacking expression of Cnp1. Glia
30(1):91–104. 57(16):1815–1824.
Bowley MP, Cabral H, Rosene DL, Peters A. 2010. Age changes in myeli- Edgar JM, Nave KA. 2009. The role of CNS glia in preserving axon func-
nated nerve fibers of the cingulate bundle and corpus callosum in the tion. Curr Opin Neurobiol 19(5):498–504.
rhesus monkey. J Comp Neurol 518(15):3046–3064. Fanarraga ML, Griffiths IR, Zhao M, Duncan ID. 1998. Oligodendrocytes
Buckley CE, Marguerie A, Alderton WK, Franklin RJ. 2010. are not inherently programmed to myelinate a specific size of axon. J
Temporal dynamics of myelination in the zebrafish spinal cord. Glia Comp Neurol 399(1):94–100.
58(7):802–812. Feldman DH, Horiuchi M, Keachie K, McCauley E, Bannerman P, Itoh
Butt AM, Berry M. 2000. Oligodendrocytes and the control of myeli- A, et al. 2008. Characterization of acid-sensing ion channel expres-
nation in vivo: new insights from the rat anterior medullary velum. sion in oligodendrocyte-lineage cells. Glia 56(11):1238–1249.
J Neurosci Res 59(4):477–488. Friede RL. 1972. Control of myelin formation by axon cali-
Butt AM, Colquhoun K, Tutton M, Berry M. 1994. Three-dimensional ber (with a model of the control mechanism). J Comp Neurol
morphology of astrocytes and oligodendrocytes in the intact mouse 144(2):233–252.
optic nerve. J Neurocytol 23(8):469–485. Ghandour MS, Skoff RP, Venta PJ, Tashian RE. 1989. Oligodendrocytes
Butt AM, Duncan A, Hornby MF, Kirvell SL, Hunter A, Levine JM, express a normal phenotype in carbonic anhydrase II-deficient mice.
et al. 1999. Cells expressing the NG2 antigen contact nodes of J Neurosci Res 23(2):180–190.
Ranvier in adult CNS white matter. Glia 26(1):84–91. Griffiths I, Klugmann M, Anderson T, Thomson C, Vouyiouklis D,
Butt AM, Hamilton N, Hubbard P, Pugh M, Ibrahim M. 2005. Nave KA. 1998a. Current concepts of PLP and its role in the nervous
Synantocytes: the fi fth element. J Anat 207(6):695–706. system. Microsc Res Tech 41(5):344–358.
Butt AM, Ibrahim M, Berry M. 1997. The relationship between devel- Griffiths I, Klugmann M, Anderson T, Yool D, Thomson C, Schwab
oping oligodendrocyte units and maturing axons during myelinogen- MH, et al. 1998b. Axonal swellings and degeneration in mice lacking
esis in the anterior medullary velum of neonatal rats. J Neurocytol the major proteolipid of myelin. Science 280(5369):1610–1613.
26(5):327–338. Haberlandt C, Derouiche A, Wyczynski A, Haseleu J, Pohle J, Karram
Butt AM, Ibrahim M, Berry M. 1998a. Axon-myelin sheath relations of K, et al. 2011. Gray matter NG2 cells display multiple Ca2+-signaling
oligodendrocyte unit phenotypes in the adult rat anterior medullary pathways and highly motile processes. PLoS One 6(3):e17575.
velum. J Neurocytol 27(4):259–269. Hamilton N, Vayro S, Wigley R, Butt AM. 2010. Axons and astrocytes
Butt AM, Ibrahim M, Gregson N, Berry M. 1998b. Differential expres- release ATP and glutamate to evoke calcium signals in NG2-glia.
sion of the L- and S-isoforms of myelin associated glycoprotein Glia 58(1):66–79.
(MAG) in oligodendrocyte unit phenotypes in the adult rat anterior Hardy RJ, Friedrich VL Jr. 1996. Progressive remodeling of the oligo-
medullary velum. J Neurocytol 27(4):271–280. dendrocyte process arbor during myelinogenesis. Dev Neurosci
Butt AM, Ibrahim M, Ruge FM, Berry M. 1995. Biochemical subtypes 18(4):243–254.
of oligodendrocyte in the anterior medullary velum of the rat as Hartman BK, Agrawal HC, Agrawal D, Kalmbach S. 1982. Development
revealed by the monoclonal antibody Rip. Glia 14(3):185–197. and maturation of central nervous system myelin: comparison of
Butt AM, Kiff J, Hubbard P, Berry M. 2002. Synantocytes: new immunohistochemical localization of proteolipid protein and basic
functions for novel NG2 expressing glia. J Neurocytol 31(6–7): protein in myelin and oligodendrocytes. Proc Natl Acad Sci U S A
551–565. 79(13):4217–4220.

S T RU C T U R E A N D F U N C T I O N O F O L I G O D E N D R O C Y T E S • 71
Hildebrand C, Hahn R. 1978. Relation between myelin sheath thickness Peters A. 1964. Observations on the connexions between myelin sheaths
and axon size in spinal cord white matter of some vertebrate species. J and glial cells in the optic nerves of young rats. J Anat 98:125–134.
Neurol Sci 38(3):421–434. Peters A. 2002. The effects of normal aging on myelin and nerve fibers: a
Hildebrand C, Remahl S, Persson H, Bjartmar C. 1993. Myelinated review. J Neurocytol 31(8–9):581–593.
nerve fibres in the CNS. Prog Neurobiol 40(3):319–384. Peters A. 2004. A fourth type of neuroglial cell in the adult central nerv-
Hildebrand C, Waxman SG. 1984. Postnatal differentiation of rat ous system. J Neurocytol 33(3):345–357.
optic nerve fibers: electron microscopic observations on the devel- Peters A, Palay SL, Webster H. 1991. The fine structure of the nervous
opment of nodes of Ranvier and axoglial relations. J Comp Neurol system: neurons and their supporting cells, 3rd ed. New York: Oxford
224(1):25–37. University Press.
Hinman JD, Peters A, Cabral H, Rosene DL, Hollander W, Rasband Peters A, Sethares C. 2004. Oligodendrocytes, their progenitors and
MN, et al. 2006. Age-related molecular reorganization at the node of other neuroglial cells in the aging primate cerebral cortex. Cereb
Ranvier. J Comp Neurol 495(4):351–362. Cortex 14(9):995–1007.
Hirano A. 1968. A confirmation of the oligodendroglial origin of myelin Peters A, Verderosa A, Sethares C. 2008. The neuroglial popula-
in the adult rat. J Cell Biol 38(3):637–640. tion in the primary visual cortex of the aging rhesus monkey. Glia
Ibrahim M, Butt AM, Berry M. 1995. Relationship between myelin 56(11):1151–1161.
sheath diameter and internodal length in axons of the anterior med- Poliak S, Salomon D, Elhanany H, Sabanay H, Kiernan B, Pevny L,
ullary velum of the adult rat. J Neurol Sci 133(1–2):119–127. et al. 2003. Juxtaparanodal clustering of Shaker-like K+ channels
Ioannidou K, Anderson KI, Strachan D, Edgar JM, Barnett SC. 2012. in myelinated axons depends on Caspr2 and TAG-1. J Cell Biol
Time-lapse imaging of the dynamics of CNS glial-axonal interactions 162(6):1149–1160.
in vitro and ex vivo. PLoS ONE 7(1):e30775. Raff MC, Mirsky R, Fields KL, Lisak RP, Dorfman SH, Silberberg
Kang SH, Fukaya M, Yang JK, Rothstein JD, Bergles DE. 2010. NG2+ DH, et al. 1978. Galactocerebroside is a specific cell-surface
CNS glial progenitors remain committed to the oligodendrocyte antigenic marker for oligodendrocytes in culture. Nature
lineage in postnatal life and following neurodegeneration. Neuron 274(5673):813–816.
68(4):668–681. Ransom BR, Butt AM, Black JA. 1991. Ultrastructural identification
Kaplan MR, Cho MH, Ullian EM, Isom LL, Levinson SR, Barres of HRP-injected oligodendrocytes in the intact rat optic nerve. Glia
BA. 2001. Differential control of clustering of the sodium channels 4(1):37–45.
Na(v)1.2 and Na(v)1.6 at developing CNS nodes of Ranvier. Neuron Ransom BR, Walz W, Davis PK, Carlini WG. 1992. Anoxia-induced
30(1):105–119. changes in extracellular K+ and pH in mammalian central white
Kettenmann H, Ransom BR, Schlue WR. 1990. Intracellular pH shifts matter. J Cereb Blood Flow Metab 12(4):593–602.
capable of uncoupling cultured oligodendrocytes are seen only in low Rasband MN, Trimmer JS. 2001. Developmental clustering of ion chan-
HCO3- solution. Glia 3(2):110–117. nels at and near the node of Ranvier. Dev Biol 236(1):5–16.
Kukley M, Capetillo-Zarate E, Dietrich D. 2007. Vesicular glutamate Remahl S, Hildebrand C. 1990a. Relation between axons and oligoden-
release from axons in white matter. Nat Neurosci 10(3):311–320. droglial cells during initial myelination. I. The glial unit. J Neurocytol
Kukley M, Nishiyama A, Dietrich D. 2010. The fate of synaptic input 19(3):313–328.
to NG2 glial cells: neurons specifically downregulate transmit- Remahl S, Hildebrand C. 1990b. Relations between axons and oligo-
ter release onto differentiating oligodendroglial cells. J Neurosci dendroglial cells during initial myelination. II. The individual axon.
30(24):8320–8331. J Neurocytol 19(6):883–898.
Lappe-Siefke C, Goebbels S, Gravel M, Nicksch E, Lee J, Braun PE, et al. Richardson WD, Young KM, Tripathi RB, McKenzie I. 2011.
2003. Disruption of Cnp1 uncouples oligodendroglial functions in NG2-glia as multipotent neural stem cells: fact or fantasy? Neuron
axonal support and myelination. Nat Genet 33(3):366–374. 70(4):661–673.
Laursen LS, Chan CW, ffrench-Constant C. 2009. An integrin-contactin Riddle A, Luo NL, Manese M, Beardsley DJ, Green L, Rorvik DA, et al.
complex regulates CNS myelination by differential Fyn phosphoryla- 2006. Spatial heterogeneity in oligodendrocyte lineage maturation
tion. J Neurosci 29(29):9174–9185. and not cerebral blood flow predicts fetal ovine periventricular white
Levine JM, Card JP. 1987. Light and electron microscopic localization of matter injury. J Neurosci 26(11):3045–3055.
a cell surface antigen (NG2) in the rat cerebellum: association with Rinholm JE, Hamilton NB, Kessaris N, Richardson WD, Bergersen LH,
smooth protoplasmic astrocytes. J Neurosci 7(9):2711–2720. Attwell D. 2011. Regulation of oligodendrocyte development and
Levine JM, Stallcup WB. 1987. Plasticity of developing cerebellar cells myelination by glucose and lactate. J Neurosci 31(2):538–548.
in vitro studied with antibodies against the NG2 antigen. J Neurosci Rios JC, Rubin M, St Martin M, Downey RT, Einheber S, Rosenbluth
7(9):2721–2731. J, et al. 2003. Paranodal interactions regulate expression of sodium
Marcus J, Honigbaum S, Shroff S, Honke K, Rosenbluth J, Dupree JL. channel subtypes and provide a diff usion barrier for the node of
2006. Sulfatide is essential for the maintenance of CNS myelin and Ranvier. J Neurosci 23(18):7001–7011.
axon structure. Glia 53(4):372–381. Rivers LE, Young KM, Rizzi M, Jamen F, Psachoulia K, Wade A,
Mierzwa AJ, Arevalo JC, Schiff R, Chao MV, Rosenbluth J. 2010. et al. 2008. PDGFRA/NG2 glia generate myelinating oligodendro-
Role of transverse bands in maintaining paranodal structure cytes and piriform projection neurons in adult mice. Nat Neurosci
and axolemmal domain organization in myelinated nerve fibers: 11(12):1392–1401.
effect on longevity in dysmyelinated mutant mice. J Comp Neurol Ro HA, Carson JH. 2004. pH microdomains in oligodendrocytes. J Biol
518(14):2841–2853. Chem 279(35):37115–37123.
Murtie JC, Macklin WB, Corfas G. 2007. Morphometric analysis of Rosenbluth J. 2009. Multiple functions of the paranodal junction of
oligodendrocytes in the adult mouse frontal cortex. J Neurosci Res myelinated nerve fibers. J Neurosci Res 87(15):3250–3258.
85(10):2080–2086. Roy K, Murtie JC, El-Khodor BF, Edgar N, Sardi SP, Hooks BM, et al.
Nishiyama A, Komitova M, Suzuki R, Zhu X. 2009. Polydendrocytes 2007. Loss of erbB signaling in oligodendrocytes alters myelin and
(NG2 cells): multifunctional cells with lineage plasticity. Nat Rev dopaminergic function, a potential mechanism for neuropsychiatric
Neurosci 10(1):9–22. disorders. Proc Natl Acad Sci U S A 104(19):8131–8136.
Pedraza L, Huang JK, Colman D. 2009. Disposition of axonal caspr with Sandell JH, Peters A. 2002. Effects of age on the glial cells in the rhesus
respect to glial cell membranes: implications for the process of myeli- monkey optic nerve. J Comp Neurol 445(1):13–28.
nation. J Neurosci Res 87(15):3480–3491. Schachner M, Bartsch U. 2000. Multiple functions of the
Penfield W. 1932. Cytology & cellular pathology of the nervous system. myelin-associated glycoprotein MAG (siglec-4a) in formation and
New York: Hoeber. maintenance of myelin. Glia 29(2):154–165.

72 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
Skoff RP. 1978. The pattern of myelination along the developing rat optic Todorich B, Pasquini JM, Garcia CI, Paez PM, Connor JR. 2009.
nerve. Neurosci Lett 7(2–3):191–196. Oligodendrocytes and myelination: the role of iron. Glia 57(5):
Sobottka B, Ziegler U, Kaech A, Becher B, Goebels N. 2011. CNS live 467–478.
imaging reveals a new mechanism of myelination: the liquid croissant Trajkovic K, Dhaunchak AS, Goncalves JT, Wenzel D, Schneider A, Bunt
model. Glia. G, et al. 2006. Neuron to glia signaling triggers myelin membrane
Sommer I, Schachner M. 1981. Monoclonal antibodies (O1 to O4) to exocytosis from endosomal storage sites. J Cell Biol 172(6):937–948.
oligodendrocyte cell surfaces: an immunocytological study in the Tripathi RB, Clarke LE, Burzomato V, Kessaris N, Anderson PN,
central nervous system. Dev Biol 83(2):311–327. Attwell D, et al. 2011. Dorsally and ventrally derived oligodendro-
Song J, Goetz BD, Kirvell SL, Butt AM, Duncan ID. 2001. Selective cytes have similar electrical properties but myelinate preferred tracts.
myelin defects in the anterior medullary velum of the taiep mutant J Neurosci 31(18):6809–6819.
rat. Glia 33(1):1–11. Velez-Fort M, Maldonado PP, Butt AM, Audinat E, Angulo MC. 2010.
Stallcup WB. 1981. The NG2 antigen, a putative lineage marker: immu- Postnatal switch from synaptic to extrasynaptic transmission between
nofluorescent localization in primary cultures of rat brain. Dev Biol interneurons and NG2 cells. J Neurosci 30(20):6921–6929.
83(1):154–165. Velumian AA, Samoilova M, Fehlings MG. 2011. Visualization of cyto-
Stallcup WB, Beasley L. 1987. Bipotential glial precursor cells plasmic diff usion within living myelin sheaths of CNS white mat-
of the optic nerve express the NG2 proteoglycan. J Neurosci ter axons using microinjection of the fluorescent dye Lucifer Yellow.
7(9):2737–2744. Neuroimage 56(1):27–34.
Stensaas LJ, Stensaas SS. 1968a. Astrocytic neuroglial cells, oligoden- Vinet J, Lemieux P, Tamburri A, Tiesinga P, Scafidi J, Gallo V, et al. 2010.
drocytes and microgliacytes in the spinal cord of the toad. I. Light Subclasses of oligodendrocytes populate the mouse hippocampus.
microscopy. Z Zellforsch Mikrosk Anat 84(4):473–489. Eur J Neurosci 31(3):425–438.
Stensaas LJ, Stensaas SS. 1968b. Astrocytic neuroglial cells, oligodendro- Vostrikov VM, Uranova NA, Orlovskaya DD. 2007. Deficit of perineu-
cytes and microgliacytes in the spinal cord of the toad. II. Electron ronal oligodendrocytes in the prefrontal cortex in schizophrenia and
microscopy. Z Zellforsch Mikrosk Anat 86(2):184–213. mood disorders. Schizophr Res 94(1–3):273–280.
Sternberger NH, Itoyama Y, Kies MW, Webster Hd. 1978. Immuno- Wake H, Lee PR, Fields RD. 2011. Control of local protein synthe-
cytochemical method to identify basic protein in myelin-forming sis and initial events in myelination by action potentials. Science
oligodendrocytes of newborn rat C.N.S. J Neurocytol 333(6049):1647–1651.
7(2):251–263. Weruaga-Prieto E, Eggli P, Celio MR. 1996a. Rat brain oligodendro-
Sykova E. 1989. Activity-related extracellular pH changes in spinal cord. cytes do not interact selectively with axons expressing different
Acta Physiol Scand Suppl 582:62. calcium-binding proteins. Glia 16(2):117–128.
Szuchet S, Nielsen JA, Lovas G, Domowicz MS, de Velasco JM, Weruaga-Prieto E, Eggli P, Celio MR. 1996b. Topographic variations
Maric D, et al. 2011. The genetic signature of perineuronal oli- in rat brain oligodendrocyte morphology elucidated by injection of
godendrocytes reveals their unique phenotype. Eur J Neurosci Lucifer Yellow in fi xed tissue slices. J Neurocytol 25(1):19–31.
34(12):1906–1922. White R, Gonsior C, Krmer-Albers E-M, Stahr N, Hatelmaier S, Trotter
Takasaki C, Yamasaki M, Uchigashima M, Konno K, Yanagawa Y, J. 2008. Activation of oligodendroglial Fyn kinase enhances transla-
Watanabe M. 2010. Cytochemical and cytological properties of tion of mRNAs transported in hnRNP A2-dependent RNA gran-
perineuronal oligodendrocytes in the mouse cortex. Eur J Neurosci ules. J Cell Biol 181(4):579–586.
32(8):1326–1336. Wigley R, Butt AM. 2009. Integration of NG2-glia (synantocytes) into
Taniike M, Mohri I, Eguchi N, Beuckmann CT, Suzuki K, Urade Y. the neuroglial network. Neuron Glia Biol 5(1–2):21–28.
2002. Perineuronal oligodendrocytes protect against neuronal Zonta B, Tait S, Melrose S, Anderson H, Harroch S, Higginson J, et al.
apoptosis through the production of lipocalin-type prostaglan- 2008. Glial and neuronal isoforms of Neurofascin have distinct roles
din D synthase in a genetic demyelinating model. J Neurosci in the assembly of nodes of Ranvier in the central nervous system.
22(12):4885–4896. J Cell Biol 181(7):1169–1177.

S T RU C T U R E A N D F U N C T I O N O F O L I G O D E N D R O C Y T E S • 73
7.
SCHWANN CELLS AND MYELIN
Rudolf Martini and Ágnes Patzkó

A B B R E VI AT I O N S addition, progress in live imaging technologies, combined


with immunocytochemical and transgenic labeling approaches
CAM cell adhesion molecule extended our knowledge of the spatio-temporal dynamics
cAMP cyclic adenosine monophosphate of these cells and their related derivatives, such as the termi-
Caspr contactin-associated protein nal Schwann cells at the neuromuscular junction (Brill et al.
CNP-1 2′,3′-cyclic nucleotide 3′-phosphodiesterase 2011). However, even the more “traditional” approaches, such
isoform 1 as transmission electron microscopy, have been essential for
CNS central nervous system understanding basic mechanisms; a representative example
Cx32 connexin 32 is the systematic analysis of the position and putative roles of
DRP2 dystroglycan-dystrophin-related protein 2 Schwann cell precursors during nerve growth (Wanner et al.
ECM extracellular matrix 2006b). Moreover, the roles of the nonmyelinating Schwann
EGR-2 early growth response protein 2 cells, also called Remak Schwann cells, the usually less consid-
FAK focal adhesion kinase ered Schwann cell population, have also been studied in detail
GABA gamma-aminobutyric acid (Griffin and Thompson 2008).
GAP-43 growth associated protein 43 For many years the nodes of Ranvier and particularly the
Gpr126 G protein–coupled receptor 126 role of Schwann cells in directing the relevant ion channels
MAG myelin-associated glycoprotein have been in the focus of interest; depending on the techno-
MBP myelin basic protein logical approaches applied, these studies led to various, some-
MPZ myelin protein zero times even controversial models describing how axons and the
N-Cadherin neural cadherin glia synergistically interact to warrant the function of nerve
N-CAM neural cell adhesion molecule fibers (Feinberg et al. 2010; Thaxton et al. 2011).
Necl nectin-like The present chapter focuses on the major structural
NGF nerve growth factor characteristics of Schwann cells (Fig. 7.1). Although dis-
NF neurofascin cussed more in detail in other chapters, molecular aspects
Nr-CAM neuron glial-related cell adhesion are also considered, especially where they help to under-
molecule stand the cytoarchitecture and function of these fascinat-
Nrg1 neuregulin 1 isoform ing cells.
N-WASP Neural-Wiskott-Aldrich syndrome
protein
Omgp oligodendrocyte myelin glycoprotein 2 E A R LY D E VE L O PM E N T O F
P0 myelin protein zero S C H WA N N C E L L S
PDZ postsynaptic density protein—disk large—
zonula occludens protein
2.1 S C H WA N N C E L L P R EC U R S O R S
PLP proteolipid protein
PMP22 peripheral myelin protein 22 Being derivatives of the neural crest ( Jessen and Mirsky 2005),
PNS peripheral nervous system embryonic Schwann cells are associated with axon bundles
SCP Schwann cell precursor during development (Billings-Gagliardi et al. 1974). These
TAG-1 transient axonal glycoprotein-1 cells are Schwann cell precursors (i.e., prospective Schwann
cells that still lack the ability to survive in the absence of axons)
( Jessen and Mirsky 2005). They show immunoreactivity for
1 INTRODUCTION the low-affinity NGF receptor p75, and the cell adhesion mol-
ecules L1 and N-CAM (Dong et al. 1999; Martini 1994), but
Recent years have brought a dramatic increase in our knowl- the glial marker S100 is not detectable unless the sensitivity of
edge about the molecular components of Schwann cells in conventional immunohistochemical techniques is improved
development and disease (Berti et al. 2011; Jessen and Mirsky ( Jessen and Mirsky 2005). They also express low levels of the
2005; Salzer et al. 2008; Sherman and Brophy 2005). In myelin components P0, PMP22, and PLP ( Jessen and Mirsky

74
A B

A
Figure 7.2 Electron Micrographs of Schwann Cell Precursors
(SCP). A. Cross-section of an embryonic nerve displaying axons and
growth cones (gc) in association with the SCPs. Schwann cell processes
(asterisks), gliopodia (arrows), and adherens junctions (aj; white arrows)
are indicated. ECS: extracellular space devoid of axons. Bar = 1 μm.
B. Longitudinal section of a growth cone (gc) with actin filaments (a) and
neurofilaments (nf ) at its proximal region. Note tight association with
SCPs. Bar = 1 μm. Reprinted from Wanner et al. 2006, with permission
from Wiley.
B C

Figure 7.1 Two principal forms of peripheral nerve fibers: a myelinated


nerve fiber (A) and two Remak fibers associated with nonmyelinating
Schwann cells (B, C). Saphenus nerve of an adult mouse. The their peripheral targets. The view that glial precursor cells
myelinating Schwann cell is surrounded by a basement membrane may at least “shape” the nerve is supported by an elegant
(arrows). Note that the inner Schwann cell loop contains cytoplasm
(arrowheads). The “outer” cytoplasmic domains (asterisks) appear as report on the developing lateral organ of the zebra fish in
“Cajal bands” in longitudinal views (compare with Fig. 7.4). Double which genetic ablation of these cells lead to defasciculation
arrowheads indicate the outer mesaxon on the right hand site. Bar = (Gilmour et al. 2002). Fasciculation may be mediated by
0.5 μm. (B) Note that most, but not all small-caliber axons are separated the polarized secretion of the repulsive extracellular matrix
from each other by slender processes of the nonmyelinating Schwann cell. molecule tenascin-C that has been found previously to sur-
Bar = 1 μm. (C) This micrograph probably displays the border region of
two adjacent nonmyelinating Schwann cells (SC 1, SC 2). Bar = 0.5 μm. round the abaxonal surfaces of the “mantling” (Wanner et al.
2006b) precursor cells (Martini and Schachner 1991; Wehrle
and Chiquet 1990).
Based on electron microscopic data (Martini 1994;
2005). Schwann cell precursors are not only the likely source Wanner et al. 2006b), the most important morphological fea-
for mature Schwann cells but also pivotal cellular components ture of Schwann cell precursors is the absence of a basal lamina
for axon survival ( Jessen and Mirsky 2005; Riethmacher et al. (Fig. 7.2). They intermingle with fasciculating axons proximal
1997). to the growing nerve tip and extend processes in between of
Because of the position of HNK-1-positive cells “ahead” the axon bundles (Fig. 7.3). In addition, these cells are found at
of growing axons, it was suggested that Schwann cell precur- the margin of the prospective nerve or nerve fascicle and face
sors may guide axons to their targets during development the mesenchyme with their abaxonal surface (see Fig. 7.2).
(Noakes and Bennett 1987). Indeed, recent studies in rats
using electron microscopy, confocal three-dimensional (3D)
reconstructions, and immunohistochemistry against p75 2.2 I M M AT U R E S C H WA N N C E L L S
neurotrophin receptor and N-cadherin, identified Schwann When development proceeds, Schwann cell precursors start
cell precursors as components of the growing nerve front, to form a basal lamina, proliferate, and collectively ensheathe
but not stringently ahead of the growth cones (Wanner et al. fasciculating axons forming so called “Schwann cell families”
2006a,b). There, growth cones make preferential contacts as initially described by Webster and colleagues (see Fig. 7.3)
with precursors rather than with extracellular space (Fig. 7.2) (Peters et al. 1991). The respective glial cells reached the stage
(Wanner et al. 2006b). In this position the precursor cells of “immature Schwann cells,” a term that characterizes the
show lamellipodia and filopodia-like, slim processes, called ability of the Schwann cells to survive independently from
gliopodia (Wanner et al. 2006b). These combined observa- axons resulting from an autocrine survival mechanism ( Jessen
tions suggest that the Schwann cell precursors serve as cellular and Mirsky 2005).
chaperons (Wanner et al. 2006b) leading to nerve compac- At the same time when the immature Schwann cells acquire
tion and may facilitate navigation when axons approach their basal lamina, additional connective tissue elements and

S C H WA N N C E L L S A N D M Y E L I N • 75
A B C 3.1 BA S A L L A M I NA F O R M AT I O N
AND FUNCTION
An important prerequisite for radial sorting and the follow-
ing processes is the generation of a basal lamina around the
Schwann cell. Thus, the Schwann cell gains polarity (Ozcelik et
al. 2010). In longitudinal sections, the Schwann cell basal lam-
ina forms a continuous tube that even bridges the nodal gaps
D Laminin 211, 411 (Fig. 7.5). The major Schwann cell basal lamina constituents
1,5 4,6
Integrin αβ1 dystroglycan are the extracellular matrix components laminin-2 (merosin),
3 2 heparan sulfate proteoglycans (e.g., perlecan, agrin), fibronec-
NrgTypell Akt1 ILK, Rac1 p38 MAPK tin (Chernousov and Carey 2000), collagen type IV, and the
recently discovered type XXVIII (Grimal et al. 2010). For the
radial axonal sorting
first time the fundamental role of basal lamina during myeli-
αβ1 integrins myelination
nation was presented by the Bunge laboratory (Eldridge et al.
dystroglycan 1989). An additional hint reflecting the importance of basal
lamina is provided by the dysmyelinating phenotype of dys-
Figure 7.3 Radial Sorting (A–C; 4-Day-Old Mouse) and the Underlying trophic mice, which are deficient in laminin-2 (Matsumura
Molecular Mechanisms (D). A–C. Immature Schwann cells associated et al. 1997).
with a basement membrane are in contact with smaller caliber axons
(asterisks) and with larger axons (double asterisks) in the process of sorting. Laminin-2 is not only crucial in radial sorting and devel-
Note that some axons have already reached the promyelinating stage (PM) opment of myelinating Schwann cells, but also participates in
or are in the process of myelination (M). Bar = 1 μm. D. Morphogenetic nonmyelinating Schwann cell development and Remak bun-
steps comprising the formation of Schwann cell families (1), Schwann dle formation (Yu et al. 2009).
cell process formation (2, requiring αβ1 integrins), and radial sorting to Many studies confirm the importance of cell substrate sig-
promyelinating stages (steps 4–6, requiring dystroglycan) are shown. The
presumed involvement of signaling pathways is also indicated (see also naling mediated by laminin-2 and the corresponding receptors
text). Reprinted from Berti et al. 2011, with permission from the Society of the abaxonal Schwann cell membrane, including integrin
for Neuroscience. heterodimers such as α6/β1 and α6/β4, dystroglycan and
dystroglycan-dystrophin-related protein 2 (DRP2, Fig. 7.3D).
The latter forms a complex with αβ-dystroglycan (Sherman
and Brophy 2005). Both dystroglycan and integrins play an
blood vessels appear to be generated and intermingle between essential and distinct role in radial sorting. Deletion of dystro-
the prospective nerve fibers ( Jessen and Mirsky 2005). In this glycan in Schwann cells revealed an arrest in radial sorting that
context, it is worthwhile to consider recent views on the ori- was most evident in spinal roots (Berti et al. 2011).
gin of endoneurial fibroblasts. Although there are some argu- Schwann cell–specific gene inactivation of β1-integrin
ments favoring the idea that endoneurial fibroblasts might be leads to a severe dysmyelinating phenotype with impaired
derivatives of neural crest cells or even Schwann cell precur- axonal sorting (Feltri et al. 2002). β4-integrin appears to be
sors ( Jessen and Mirsky 2005; Joseph et al. 2004), there are redundant during myelination, because absence of this com-
also strong arguments supporting the more traditional idea, ponent does not interfere with initial myelin formation in
namely the mesodermal origin of fibroblasts (Mäurer et al. vivo or in vitro (Frei et al. 1999). Nevertheless, in adult nerves,
2003). α6/β4 integrin-null myelin is more prone to abnormal fold-
ing with aging. When the laminin receptor dystroglycan is
also ablated, major folding abnormalities occur, associated
3 E S TA B L I S H I N G T H E M AT U R E S TAG E with acute demyelination and macrophage activation. These
data indicate that α6/β4 integrin confers stability to myelin
Immature Schwann cells develop into two directions. Those in peripheral nerves (Nodari et al. 2008). Another molecule
Schwann cells associated with larger caliber axons (>1 μm) that has lately been linked to α6β1 integrin and ErbB recep-
eventually differentiate into the myelinating phenotype (see tor signaling is focal adhesion kinase (FAK). In case of con-
section 3.3). The Schwann cells remaining associated with ditional FAK inactivation, Schwann cells could interdigitate
small caliber axons collectively ensheathe them. This bidirec- among axon bundles, but radial sorting and Schwann cell
tional development is mediated by Neuregulin-1 (Nrg1) type proliferation were impaired leaving large unsorted bundles
III (see chapter 44). (Grove et al. 2007). Growing evidence indicates that collagens
The multistep process by which Schwann cells segregate perform important functions during the development of the
axons is called radial sorting (Fig. 7.3D). Typically, prospec- peripheral nervous system. Collagen XXVIII is almost exclu-
tive myelinated axons of larger caliber expressing higher sively expressed in the peripheral nervous system and is local-
levels of Nrg1 are “moved” to the periphery of axon bun- ized at the basal lamina associated with unmyelinated fibers.
dles and become associated with a prospective myelinating It is worth noting that collagen XXVIII is also present at the
Schwann cell, generated by mitosis of immature Schwann nodes of Ranvier and in terminal Schwann cells in sensory end
cells. organs (Grimal et al. 2010) (i.e., in subdomains of myelinated

76 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
fibers lacking true myelin). All in all, the Schwann cell basal are downregulated, whereas typical myelin components, such
lamina components and their cognate Schwann cell–related as P0, MAG, PMP22, MBP, Cx32, and periaxin are strongly
receptors are crucial elements for maturation. upregulated (Martini 1994). Important regulators of such mol-
ecules are krox20, which promotes and maintains myelination
(Svaren and Meijer 2008) and c-Jun that is upregulated upon
3.2 N O N MY E L I NAT I N G S C H WA N N C E L L S
dedifferentiation ( Jessen and Mirsky 2008). Cross-inhibition
Nonmyelinating Schwann cells typically ensheathe axons of these transcription factors serves to switch Schwann cells
smaller than 1 μm. Nociceptors, postganglionic sympathetic, between the mature and dedifferentiated phenotype (Salzer
and certain preganglionic sympathetic and parasympathetic et al. 2008).
fibers represent this entity (Griffin and Thompson 2008). The pro-myelin stage is followed by spiral formation of
These axons are separated from each other by slender Schwann one of the Schwann cell processes engulfing the axon (see
cell processes (see Fig. 7.1 B, C) being embedded in pocket-like Fig. 7.3C). The apposition of Schwann cell membranes at the
structures. Of note, one single nonmyelinating Schwann inner, axon-related site of the Schwann cell process is called
cell can “home” thin-caliber axons of distinct modalities, as “inner mesaxon,” whereas the contact of Schwann cell mem-
for instance, sensory and sympathetic axons (Griffin and branes at the endoneurial site is termed “outer mesaxon” (see
Thompson 2008). In the region of the Schwann cell nucleus, Fig. 7.1A). These cell contacts are characterized by the forma-
cross-cut axons are found at the periphery of the Schwann cell tion of E-cadherin-positive adherens (desmosome-like) junc-
soma and show a rosette-like organization around the nucleus. tions (Fannon et al. 1995) and tight junctions (Scherer and
In this position, the axons are often only partially covered by Arroyo 2002).
Schwann cell processes and are in direct apposition with the Pioneering work by the Bunge-laboratory identified in
Schwann cell basal lamina. Mature nonmyelinating Schwann Schwann cell–dorsal root ganglion cocultures that the inner
cells share a couple of cell surface molecules with immature lip of the Schwann cell is turning around the axon (Bunge et
Schwann cells that are not found on myelinating Schwann al. 1989). However, in vivo studies revealed the myelinating
cells, such as the cell adhesion molecules L1, N-CAM, and the process to be more complicated than just a spiral formation of
NGF-receptor p75 ( Jessen and Mirsky 2005; Martini 1994). the inner Schwann cell process around the axon (Peters et al.
Although nonmyelinating Schwann cells are morphologi- 1991).
cally and molecularly distinct from myelinating Schwann cells In myelinating Schwann cells, specialized cytoplasmic
(see Fig. 7.1), they can adopt a myelinating phenotype under structures are “Bands of Cajal” which are channel-like domains
experimental conditions (Weinberg and Spencer 1975). “on top” of the outer aspects of the myelin sheathes that
The biological function of nonmyelinating Schwann cells alternate with DRP2/periaxin rich “patches” or plaques of
may be best understood when compared with molecular fea- Schwann cell membrane apposition with basal lamina without
tures of myelinating Schwann cells (Griffin and Thompson underlying cytoplasm (Fig. 7.4) (Court et al. 2009; Sherman
2008). Nonmyelinating Schwann cells lack inhibitory com- and Brophy 2005). Most importantly, both Cajal bands and
ponents, such as MAG and Omgp, which are constituents of the alternating patches are lost in mice deficient in periaxin,
myelin sheaths and nodes of Ranvier, respectively (Griffin and which is a central component of dystroglycan complexes, that
Thompson 2008). Because nonmyelinating Schwann cells are are important receptors for the basement membrane com-
associated with the highly competent and important warning ponent laminin-2 (see the preceding). The disruption of this
system against noxious insults, the nociceptive fibers, it might component results in the dysregulation of internodal length,
be of advantage to be embedded in an environment that allows which might be related to a disruption of microtubule based
rapid sprouting on injury, thus preventing that extended areas transport within the cytoplasmic Schwann cell channels
remain irresponsive to pain after injury (Griffin and Thompson (Sherman and Brophy 2005). Taking into account that choles-
2008). terol is essential for correct myelinogenesis (Saher et al. 2011),
the Cajal bands could have important functions in addition to
those mentioned, because they harbor the cholesterol trans-
3.3 MY E L I NAT I N G S C H WA N N C E L L S
porter ABCA1 (Albrecht et al. 2008).
AND THE INTERNODE
Which other molecules are involved in the process of mye-
As a result of radial sorting and Schwann cell prolifera- lin formation? mTor, a core kinase, that regulates cell growth
tion, large-caliber axons achieve a so called 1:1 ratio with and differentiation in various mammalian cells, proved to be
immature Schwann cells (see Fig. 7.3). Schwann cells of this essential for myelin formation following the promyelinating
stage are called “promyelinating.” In rodents, the genera- stage, but also mediates axon caliber adjustment and determi-
tion of promyelinating Schwann cells occurs around birth. nation of internodal length (Sherman et al. 2012). Radial sort-
However, myelin formation in the PNS is not a highly syn- ing, however, is not mediated by this kinase (Sherman et al.
chronized event. Axons with large calibers receive their 2012). Among the recently identified myelin-related proteins
myelin sheath earlier than smaller axons so that some pro- G protein–coupled receptor Gpr126 emerged as an important
myelin stages can still be found in mice of approximately 3 regulator as well. In gpr126 mutants, Schwann cells failed to
weeks of age. express oct6 and krox20 and were arrested at the promyeli-
Around the promyelinating stage, many cell surface mol- nating stage. Elevation of cAMP in gpr126 mutants, but not
ecules related to immature or nonmyelinating Schwann cells krox20 mutants, could restore myelination (Monk et al. 2011).

S C H WA N N C E L L S A N D M Y E L I N • 77
A them mediates Schwann cell adhesion and the disruption of
the interaction by their soluble extracellular domains, or the
expression of a dominant-negative Necl4 in Schwann cells,
inhibits myelination (Spiegel et al. 2007).
A striking subcellular feature of myelinating glial cells is
the compaction of the apposing membranes of the myelin
B spiral (see Figs. 7.1A and 7.3). In rodent Schwann cells this
starts after completion of several turns of the Schwann cell
process around the axon. Two morphogenetic events occur
nearly simultaneously, namely (1) the narrowing of the spiral-
ing cell surface membranes from approximately 12 to 2 nm,
and (2) the loss of cytoplasm. The collapsed cytoplasmic sites
of the Schwann cell membranes fuse and form the 3.5-nm
“major dense line”; the membrane leaflets facing the extracel-
lular space of the spiral form the “intraperiod line,” which is
double-stranded because of the 2-nm wide gap separating the
extracellular leaflets (Fig. 7.1B) (Peters et al. 1991).
A pivotal molecular mediator for myelin compaction in
the PNS is the cell adhesion molecule P0, also designated
myelin protein zero (MPZ). It mediates homophilic adhesion
Figure 7.4 A. Bands of Cajal and transverse trabeculae (A, Phalloidin leading to the close apposition of the extracellular aspects of
in green) and complementary expression of DRP2 (red). B. Electron the spiraling Schwann cell membrane forming the intraperiod
micrograph with artificially red-colored domains of presumed DRP-2 line (reviewed in Martini and Schachner 1997). This model
deposition. Cytoplasmic compartments between the red-colored areas
represent Cajal bands (compare with Fig. 7.1A). Reprinted from Court
received strong support by the determination of the 3D struc-
et al. 2009. ture of the extracellular domain of P0 by X-ray crystallography
(Shapiro et al. 1996). The intracellular domain of P0 contains
predominantly basic residues that have been suggested to
interact with negatively charged phospholipids of the adjacent
Another important mediator of the initiation of myelination cytoplasmic parts of the Schwann cell membrane leading to the
is Dicer, a regulatory protein that is responsible for the gen- formation of the major dense line (Ding and Brunden 1994;
eration of micro-RNAs and subsequently for gene expression Kirschner and Ganser 1980; Lemke et al. 1988). Final proof
regulation. It is abundantly expressed at the promyelinating that P0 mediates myelin development and compaction was
stage, when some micro-RNAs are substantially upregulated, provided by mice deficient in this molecule that show severe
others downregulated, reflecting a complex regulatory sce- dysmyelination with disturbed myelin compaction (Giese et
nario at this stage (Gokey et al., 2012). Schwann cell–specific al. 1992), with MBP as a partial compensator for major dense
inactivation or reduction of Dicer resulted in impaired krox20 line formation (Martini et al. 1995a).
expression and the reduction of myelin proteins, arrest- During growth of the myelin sheath, the axon plays a piv-
ing of most fibers at the promyelinating stage (Bremer et al. otal role in the initiation of myelination and the regulation
2010; Dugas and Notterpek 2011; Pereira et al. 2010; Verrier of myelin thickness (see chapter 44). Additionally, a constant
et al. 2010). The transcription factor nuclear factor-kappa ratio exists between axon diameter and the outer aspect of the
B—which is also required for myelin formation—is activated myelin sheath (G-ratio) and the axon diameter and internodal
through cAMP-dependent protein kinase A that phosphory- length in the adult nerve. Quantitative studies in various species
lates the p65 subunit of nuclear factor–kappa B (Yoon et al. revealed that a normal internode of a mature peripheral nerve
2008). Similarly, Neural Wiskott-Aldrich syndrome protein is usually 100 times as long as the diameter of the correspond-
(N-WASP) appears to be necessary for the transition from the ing axon (Friede and Beuche 1985). However, developmental
promyelinating to the myelinating stage (Novak et al. 2011). studies in nerves with a robust growth in length during matura-
Many studies identified several molecules that play an tion in the absence of Schwann cell proliferation modified this
important role in Schwann cell–axon interactions during simple view (Friede and Beuche 1985; Schröder et al. 1988).
myelin formation. Both in vitro and in vivo evidence sug-
gests that β-catenin–N-cadherin interaction contributes to
establishing Schwann cell polarity and the timely initiation 4 S P E C I A L M Y E L I N C O M PA RT M E N T S
of axon-induced Schwann cell proliferation (Gess et al. 2008;
Lewallen et al. 2011). Additional molecules that have been
4.1 N O D E O F R A N VI E R , PA R A N O D E S ,
shown to mediate Schwann cell–axon interaction are cell
A N D JUX TA-PA R A N O D E S A N D T H E I R
adhesion molecules (CAMs) of the nectin-like (Necl) fam-
M O L ECU L A R CO M P O N E N TS
ily. Necl4 is the main Necl expressed by myelinating Schwann
cells and is located along the internodes in direct apposition The molecular and cytological architecture of the node of
to Necl1, which is localized on axons. The interaction between Ranvier has attracted the attention of neurobiologists for

78 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
decades. It became clear that this structure, together with its Na+ channels (Salzer et al. 2008; Schafer and Rasband
neighboring compartments, is a highly organized compart- 2006). Prominent molecular players in this context are dis-
ment that warrants the clustering of ion channels and the tinct axon-related cell adhesion molecules, such as NF186
rapid transmission of action potentials and may seal impor- and Nr-CAM (Koticha et al. 2006). Furthermore, axonal
tant functional compartments against pathogens (Rosenbluth cytoskeletal elements, such as ankyrin G, which binds to the
2009). cytoplasmic (axoplasmic) domain of the voltage-gated Na+
channels and its “anchor” protein βIV-spectrin are important
(Dzhashiashvili et al. 2007; Yang et al. 2007). On the site of
4.1.1 The Node of Ranvier Proper
the Schwann cell microvilli Nr-CAM and the gliomedins
The nodal compartment consists not only of the exposed axo- interact with the extracellular domains of axonal Nr-CAM
lemma, but also of multiple slender Schwann cell protrusions and particularly with NF186 and, in this way, appear to direct
abutting the axolemma, called microvilli (Figs. 7.5A,B, 7.6A). and fix a molecular complex consisting of the two CAMs and
They “dive” into a nodal gap substance (Landon and Williams the nodal cytoskeletal and Na+ channel components (Eshed et
1963) composed of tenascin-C, syndecan-3, collagen V, col- al. 2005, 2007; Feinberg et al. 2010). A proof of principle dem-
lagen XXVIII, and versican V1 (Grimal et al. 2010; Martini onstrating the pivotal role of neurofascins has initially been
et al. 1990; Melendez-Vasquez et al. 2005). Possibly, some of provided by investigating corresponding knock-out mice that
these ECM substances may have nerve growth–repulsive char- show defective Na+ channel clustering at nodes (Sherman et al.
acteristics. The CNS myelin component Omgp has been iden- 2005). Further studies implicated not only the gliomedin-Nr-
tified as possible inhibitor of axon “out”-growth at the PNS CAM-NF186-axis during clustering, but—by in vitro myeli-
node (Huang et al. 2005). nation approaches—showed that supplementary paranodal
A pivotal task of Schwann cell components at the node of junction mechanisms can also rescue lack of axolemmal NF186
Ranvier is their contribution to the clustering of voltage-gated (Feinberg et al. 2010). Subsequent studies in vivo are at some
variance to this model, showing that paranodal domains can-
not compensate for the loss of axonal Nf186 (Thaxton et al.
2011), as it has been initially described (Sherman et al. 2005).
A Moreover paranodal domains may even invade the putative
nodal compartment in the absence of Nf186 (Thaxton et al.
2011). One explanation for the different findings by Feinberg
and Thaxton might be the different experimental approach
(in vitro versus in vivo models) (Thaxton et al. 2011). Recently,
a study focusing on the origin of nodal components revealed
two distinct sources of nodal molecular components. NF186
is initially a ubiquitous and freely diffusible axonal compo-
nent that can thus be “collected” from local sources via inter-
action and diffusion trapping by Schwann cell microvilli. Na+
channels and the cytoskeletal elements, however, need to be
transported to the node (Zhang et al. 2012). This model is
summarized in Figure 7.6F–H.
Other Schwann cell–derived molecules involved in correct
B C
ion channel clustering include laminin 2 and its receptor com-
ponent, dystroglycan (Occhi et al. 2005; Saito et al. 2003).
The tight interaction of the myelinating Schwann cell and
nodal Na+ channels is impressively reflected by spatial abnor-
malities of the channels in myelin mutant mice. Both in the
myelin mutants, P0–/– and trembler mice (carrying a spon-
taneous PMP22 missense mutation), clustering appears pre-
mature (Devaux and Scherer 2005; Ulzheimer et al. 2004).
Moreover, in P0–/–, but not trembler mice, the Nav1.8 iso-
form is ectopically expressed in the abnormal nodes, leading
to pathological conduction properties (Moldovan et al. 2011)
Figure 7.5 Nodes of Ranvier, Paranodes, and Juxtaparanodes in the Adult (Fig. 7.6A–C).
Mouse. A. Longitudinal section of a node (arrow), flanked by paranodal
loops (asterisks). Arrowheads mark Schwann cell microvilli abutting the
nodal axolemma. Bar = 1 μm. B. Cross-section of a node of Ranvier.
Note Schwann cell microvilli (arrowheads) abutting the nodal axolemma,
4.1.2 The Paranodes
which is typically undercoated by electron dense material. Bar = 0.5 The nodes of Ranvier proper are flanked by the paranodal loops
μm. C. Cross-section of a juxtaparanode of a larger-caliber fiber with
the typical trefoil shape of the myelin. Outer Schwann cell cytoplasm (see Fig. 7.5A), that are, cytoplasmic pockets of the respective
“fills” the grooves of the folded myelin and shows an accumulation of myelinating glial cells. Similar to the Schmidt-Lanterman inci-
mitochondria. Bar = 2 μm. sures (see section 4.2), they form a helix-shaped cytoplasmic

S C H WA N N C E L L S A N D M Y E L I N • 79
A

F NaCh NF186 NrCAM G Forming node


AnkG Gliomedin MV
B
PNJ
Premyelinating
Schwann cell

C Axon

H Mature node

Figure 7.6 Teased fiber preparations from wild-type (A–D) and P0–/– mice (B, C, E), prepared for immunocytochemistry using antibodies
against voltage-gated Na+ channels (A–C) and K+ channels (D–E). Figure F–H displays the current view of mechanisms leading to deposition
of Na+ channels at the node of Ranvier. A–C. Clusters of Na+ channels at the nodes of adult P0–/– mice display an immature feature (B,C) in
comparison to the nodes of wild-type mice (A). D,E. In wild-type mice (D) K+ channels are confined to the juxtaparanodes, whereas in the P0–/–
mice, they are shifted to paranodes. Kv1.2 is also located along the internodal, inner mesaxon (black arrows), and Schmidt-Lanterman incisures
(white arrows). Bar = 10 μm. F. Before myelination, components of the node are diffusely expressed along the axon. G. During node assembly
NF186 redistributes via diffusion to the node, where it is “trapped” by interactions with the gliomedin/NrCAM complex on Schwann cell
microvilli (MV). Sodium channels are delivered for exocytosis. H. In mature nodes, all node components are being transported by carrier vesicles,
replenishing components that slowly turn over; NF186 is linked to sodium channels via ankyrin G. Note that paranodal junctions are providing a
diffusion barrier. (A–E) Reprinted from Ulzheimer et al. 2004. Altered expression of ion channel isoforms at the node of Ranvier in P0-deficient
myelin mutants. Mol Cell Neurosci 25:83–94, with permission from Elsevier; (F–H) reprinted from Zhang et al. 2012, with permission from
Elsevier.

band that connects the periaxonal with the abaxonal Schwann The typical molecular features of paranodal loops include
cell cytoplasm. The paranodal loops are linked to the associ- the presence of Nf155, the putative ligand of the axonal com-
ated axolemma by specialized septate junctions. Laterally, ponents contactin and Caspr, contributing to the organiza-
Schwann cell paranodes are connected by adherens junctions tion of the septate junctions (Salzer et al. 2008; Schafer and
(Fannon et al. 1995). Depending on the size of the axon, there Rasband 2006). Mice lacking one of these molecular compo-
are in principle two different forms of paranodal regions nents fail to form paranodal junctions (Sherman and Brophy
(Berthold 1996; Phillips et al. 1972). In smaller-caliber fibers, 2005). Abnormal paranodes develop in the absence of protein
the cytoplasmic pockets approach tangentially to the axon, 4.1.B that interacts with Caspr (Cifuentes-Diaz et al. 2011).
and each pocket reaches the axolemma (see Fig. 7.5A). They Recently, the paranodal loops have been suggested to control
generally appear well organized. In larger-caliber fibers, the the nodal molecular arrangement by forming a molecular bar-
pockets approximate the axolemma in a steep and almost per- rier preventing the “escape” of nodal molecules into the wrong
pendicular fashion and not all pockets reach the axolemma. compartment; however, this model is presently under debate
They are usually smaller and often appear much more electron (Feinberg et al. 2010; Thaxton et al. 2011; see the preceding).
dense than the pockets of the smaller-caliber axons. Because By using fluorescent tracers of different sizes Rosenbluth
of these characteristics they often appear less organized than et al. investigated the tightness of the paranodal junction of
paranodes of the smaller fibers. This is of particular impor- myelinated fibers (Mierzwa et al. 2010). Interestingly, 3- and
tance when investigating pathological alterations in nodal and even 70-kDa tracers have been found to be able to pass from
paranodal aspects, because the high variability of paranodes of the nodal space into the paranodal compartment, most prob-
larger-caliber fibers make a thorough quantification of patho- ably “using” the intercellular “pathway” between the heli-
logical “alterations” necessary. cally arranged paranodal terminal loops. By this pathway an

80 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
exchange of water soluble nutrients and metabolites to and destruction. The disorganization of the juxtaparanodal struc-
from the internodal axon is conceivable. Furthermore, this tures in myelin-related mutants might be a pathological con-
might be an ionic connection from the node to the juxtaparan- sequence of a labile, molecularly specialized structure that not
odal potassium channels. Last, antibodies and other detrimen- only comprises the K+ channel compartment and Caspr2 mol-
tal molecules could reach the internodal axon under various ecules, but also the juxtaparanodal network between axonal
pathological conditions (Mierzwa et al. 2010). An obviously and glial membranes.
important intracellular pathway is realized by reflexive gap
junctions formed by Cx32 molecules that may allow rapid
radial connection between the axonal and abaxonal Schwann 4 .2 S C H M I DT-L A N T E R M A N I N C I S U R E S
cell cytoplasm (Balice-Gordon et al. 1998). This radial path-
way is also found in Schmidt-Lanterman incisures. These are cytoplasmic clefts that connect the periaxonal with
the perinuclear (abaxonal) Schwann cell cytoplasm. They are
believed to mediate communication of adaxonal and abaxonal
4.1.3 Juxtaparanodes
Schwann cell cytoplasm, thus fulfilling partially overlapping
Another region of interest is the compartment neighboring functions with paranodal loops. This may be supported by
the paranodes, the juxtaparanodal region. From the morpho- the finding that both structures are expressing gap junctions
logical point of view, this compartment is particularly con- that are composed of Cx32 molecules (Scherer and Arroyo
spicuous in larger-caliber fibers (see Fig. 7.5C). Because of a 2002). In longitudinal sections of osmium tetroxide–fixed
tapering of larger-caliber axons, the myelin sheath appears tissue, they can be identified by light microscopy as slim,
fluted so that in cross-section the myelin sheath and axon are bright lines (“incisures”). In teased fiber preparations labeled
cross-shaped or acquire the form of a trefoil (see Fig. 7.5C). for markers of noncompacted myelin (e.g., MAG, Cx32),
Here, the outer Schwann cell cytoplasm “fills” the grooves of Schmidt-Lanterman incisures appear as funnel-like profiles
the folded myelin and shows an accumulation of mitochon- (Scherer and Arroyo 2002). Moreover, immunohistochemical
dria. The axon–Schwann cell apposition often shows a com- studies indicated that Caspr and the voltage-gated K+ chan-
plicated organization because of a profound interdigitation nels Kv1.1 and 1.2 demarcate the axonal domain underlying
of inner Schwann cell loop and axolemma, called the axon– the inner loop of the Schmidt-Lanterman incisures (Scherer
Schwann cell network. and Arroyo 2002). Electron microscopy reveals that the inci-
Initially the juxtaparanodal region was not distinguished sures consist of slender pockets of cytoplasm that form a heli-
from the paranode, but regarded as a component of this com- cal funnel. Similarly to paranodal loops, Schmidt-Lanterman
partment (Berthold 1996). However, when focusing on the incisures contain E-cadherin–positive adherens junctions
molecular characteristics, paranodal and juxtaparanodal sites which interact with p120 catenin. This interaction is neces-
became clearly distinct. The juxtaparanodes are molecularly sary for their formation (Tricaud et al. 2005).
defined by the presence of TAG-1, on both the glial and axonal Membranes of these Schmidt-Lanterman incisures con-
site and by Caspr2 and voltage-gated Kv1.1 and Kv 1.2 chan- tain the myelin-associated glycoprotein (MAG) but not P0.
nels on the axonal site (Salzer et al. 2008). Caspr2 binds to the Paradoxically, however, the latter molecule appears to be
cytoskeletal adaptor protein 4.1B that has pivotal functions for essential for their formation. When in oligodendrocytes the
the organization of paranodes and juxtaparanodes, including major CNS myelin component PLP is replaced by P0, these
Caspr2-enrichment (Cifuentes-Diaz et al. 2011). Interestingly, cells express ectopic incisures (Yin et al. 2008). Why this oli-
in myelin mutants devoid of P0, K+ channels and Caspr2 are godendrocytic myelin, however, is detrimental for the myeli-
shifted to the paranode (Ulzheimer et al. 2004) (Fig. 7.6D,E), nated axons, is presently unclear. Moreover, absence of MBP
and in other myelin mutants of the PNS K+ channels are disor- appears to foster the formation of Schmidt-Lanterman inci-
ganized (Groh et al. 2010; Kohl et al. 2010). Caspr2 also binds sures (Gould et al. 1995).
with its PDZ-binding sequence to other Caspr2-molecules
that anchor the K+ channels to their characteristic position
(Salzer et al. 2008). 5 S P E C I A L I Z E D ( N O N -R E M A K )
A typical ultrastructural feature is the juxtaparanodal net- S C H WA N N C E L L S D E VO I D O F M Y E L I N
work between axonal and glial membranes, which is most
obvious in larger-caliber fibers. In longitudinal sections, a
5.1 T E R M I NA L S C H WA N N C E L L S O F T H E
striking asymmetry is often detectable in that the axon–
N EU RO MUS C U L A R J U N C T I O N
Schwann cell network is much more conspicuous at the distal
aspect of the nodal region than at the proximal one. In addi- Both in the CNS and PNS, glial cells have been identified to
tion, axonal lysosomes containing acid phosphatase are more perform pivotal functions during synapse formation, prun-
frequent in the distal regions (Gatzinsky 1996). Interestingly, ing, maintenance, and modulation of activity (Eroglu and
this compartment appears particularly vulnerable to molecu- Barres 2010). In the PNS, specialized Schwann cells that do
lar changes in Schwann cells, such as reduced expression of P0 not myelinate fulfill this task at the neuromuscular junction.
(Martini et al. 1995b), absence of Cx32 (Groh et al. 2012), Based on their position they are synonymously called terminal
or lack of β4 integrin and dystroglycan (Nodari et al. 2008). Schwann cells, perisynaptic Schwann cells, and teloglia (Griffin
Of note, these conditions lead to macrophage-related myelin and Thompson 2008). In the adult stage, these cells form an

S C H WA N N C E L L S A N D M Y E L I N • 81
ensheathing structure around terminal axon branches and of acetylcholine receptors at extrasynaptic sites of denervated
synaptic boutons and are covered by a basal lamina that fuses muscle fibers (Griffin and Thompson 2008).
with that of the muscle fiber and that of the motor endplate. Another interesting function of terminal Schwann cells
Interestingly, the presynaptic membranes of the boutons are occurs during early postnatal development when polyneuronal
preserved from being ensheathed, whereas the axon terminals innervation is eliminated. Combining in vivo imaging and
connecting the boutons are mostly entirely enwrapped by the electron microscopy, terminal Schwann cells have been iden-
cells. Although terminal Schwann cells do not form myelin, tified to phagocytose competing motor axon terminals that
they express typical myelin-related molecules, such as galacto- have to be pruned (Bishop et al. 2004). Of note, similar obser-
cerebroside, CNP-1, MAG, and P0 (Georgiou and Charlton vations have been made in P0-null-mutant peripheral nerves
1999). In addition, terminal Schwann cells express cell adhe- in which nerve Schwann cells also phagocytose end bulbs of
sion molecules that are usually confined to nonmyelinating retrogradely dying axons (Ey et al. 2007), possibly reflecting a
Schwann cells, such as L1 and N-CAM (Sanes and Lichtman general program in axon clearance by Schwann cells.
2001) and collagen XXVIII (Grimal et al. 2010). Recently the
question about the size of the territory occupied by a terminal
5.2 S C H WA N N C E L L S AT S E NS O RY T E R M I NA L S
Schwann cell has been investigated during development, in
the adult normal state and axonal degeneration. By labeling Schwann cell–like cells devoid of myelin and associated with
individual terminal Schwann cells and performing time-lapse terminal regions of myelinated axons are also found in the
imaging experiments on them, adult cells—as expected— sensory part of the nervous system. One example is the mul-
appeared to be arranged in a static fashion, whereas young tilamellar cells of the inner core of Pacinian corpuscles that
terminal Schwann cells intermingled dynamically (Brill et al. express the Schwann cell marker S100. Similar to nonmyeli-
2011) (Fig. 7.7). nating Schwann cells, they constitutively express N-CAM, but
Terminal Schwann cells appear to be necessary for the L1-expression is confined to the developmental stages (Nolte
maintenance of presynaptic terminals under normal con- et al. 1989). Thus, these cells might share molecular features
ditions in adults (Reddy et al. 2003). They contribute to of both myelinating (L1-negativity in adulthood) and nonmy-
the maintenance of ionic homeostasis caused by buffer- elinating Schwann cells (maintenance of N-CAM positivity).
ing high concentrations of K+ released as a consequence of Furthermore, neuregulin receptors of the erb-family have been
high-frequency activities (Griffin and Thompson 2008). described on them (Gonzalez-Martinez et al. 2007). Recently,
Interestingly, they regulate the efficacy of synaptic transmis- these cells have been identified to potentially play a role in sen-
sion, as they express purinergic and muscarinic acetylcholine sory transduction and stimulus modification; implicating neu-
receptors that can trigger the release of internal Ca2+ stores ronal glutamate and Schwann cell–derived GABA, possibly
and thus modulate perisynaptic Ca2+ concentration (Griffin involved in adapting permanent pressure (Pack and Pawson
and Thompson 2008). 2010; Pawson et al. 2009). The lamellar cells of Meissner cor-
On nerve injury, the perisynaptic Schwann cells upregu- puscles also express S100 and transiently the low-affinity NGF
late GAP-43 and nestin and extend multiple processes along receptor p75 also, which might be reminiscent of the down-
the muscle fiber that might guide regrowing motor axons regulation of p75 in myelinating Schwann cells (Albuerne et
toward their denervated endplates (Griffin and Thompson al. 2000).
2008; Kang et al. 2007). Additionally, denervated perisyn-
aptic Schwann cells upregulate the active form of the extra-
cellular matrix component agrin and can induce aggregation 6 S U M M A RY A N D P E R S P E C T I VE S

Derived from the neural crest, the Schwann cell precursor cells
not only give rise to the true Schwann cells in the adult stage,
but may also be involved in shaping embryonic nerves. Two
A B C principal phenotypes emerge from the immature Schwann
cells: the myelinating and nonmyelinating Schwann cells. The
nonmyelinating Schwann cells probably provide an environ-
ment for permanent axonal plasticity, supporting functional
recovery after injury. The myelinating Schwann cells have a
pivotal role in the saltatory propagation of action potentials
and maintenance of axonal structure. The node of Ranvier is
the myelin-related compartment responsible for the genera-
tion and propagation of the action potential. As opposed to
the nonmyelinating Schwann cells, the nodes are “sealed” by
Figure 7.7 A–C. In vivo imaging of the same neuromuscular junction in a molecules repulsive to nerve growth to prohibit unwanted
mouse at 6.5 (A) and 10 months (B,C) of age. Note the increased number axonal sprouting. Nodal, paranodal, and juxtaparanodal sites
of terminal Schwann cells at the later time point (2–4) that are demarcating
their territory. Territory reconstruction of individual terminal SCs is based on
are highly specialized, comprising the functionally impor-
photobleaching in the living animal. Axons are labeled green, acetylcholine tant clustering of Na+ and K+ channels. Special cell contacts
receptors red. Bar = 5 μm. Reprinted from Brill et al. 2011. between axon and Schwann cells might attenuate the access of

82 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
noxious components to the internode. Future studies should Cifuentes-Diaz C, Chareyre F, Garcia M, Devaux J, Carnaud M,
consider alterations of nodal structure and function as part of Levasseur G, et al. 2011. Protein 4.1B contributes to the organization
of peripheral myelinated axons. PloS one 6:e25043.
neuromuscular disorders. Court FA, Hewitt JE, Davies K, Patton BL, Uncini A, Wrabetz L, et al.
The internode consists mainly of compacted myelin. In 2009. A laminin-2, dystroglycan, utrophin axis is required for com-
addition to the well-known, noncompacted structures, para- partmentalization and elongation of myelin segments. J Neurosci
nodal loops, and Schmidt-Lanterman incisures, special cyto- 29:3908–3919.
plasmic channels of the myelinated PNS fiber, the Cajal bands, Devaux JJ, Scherer SS. 2005. Altered ion channels in an animal model of
Charcot-Marie-Tooth disease type IA. J Neurosci 25:1470–1480.
have recently been identified as important structures for fiber Ding Y, Brunden KR. 1994. The cytoplasmic domain of myelin glyco-
development and disease. Future studies will certainly con- protein P0 interacts with negatively charged phospholipid bilayers.
tinue to address these issues with regard of demyelinating or J Biol Chem 269:10764–10770.
dysmyelinating disorders. Both the myelinating and nonmy- Dong Z, Sinanan A, Parkinson D, Parmantier E, Mirsky R, Jessen
elinating Schwann cells are surrounded by a basal lamina that KR. 1999. Schwann cell development in embryonic mouse nerves.
J Neurosci Res 56:334–348.
not only forms the demarcation to the mesenchymal environ- Dugas JC, Notterpek L. 2011. MicroRNAs in oligodendrocyte and
ment within the nerve, but also contains components that are Schwann cell differentiation. Dev Neurosci 33:14–20.
important for Schwann cell differentiation. Less conspicuous Dzhashiashvili Y, Zhang Y, Galinska J, Lam I, Grumet M, Salzer JL.
cells, such as terminal Schwann cells, might fulfill important 2007. Nodes of Ranvier and axon initial segments are ankyrin
functions during the formation of neuromuscular junction G-dependent domains that assemble by distinct mechanisms. J Cell
Biol 177:857–870.
cytoarchitecture and neuromuscular transmission. Applying Eldridge CF, Bunge MB, Bunge RP. 1989. Differentiation of axon-related
live-imaging microscopy and other sophisticated techniques, Schwann cells in vitro: II. Control of myelin formation by basal lam-
more knowledge will certainly emerge about this important ina. J Neurosci 9:625–638.
component of the “tripartite synapse” (Eroglu and Barres Eroglu C, Barres BA. 2010. Regulation of synaptic connectivity by glia.
2010; Parpura et al. 2012). Thus, Schwann cells are essential Nature 468:223–231.
Eshed Y, Feinberg K, Carey DJ, Peles E. 2007. Secreted gliomedin is
determinants of the functional integrity of the peripheral a perinodal matrix component of peripheral nerves. J Cell Biol
part of the axon and it is not surprising that disorders pri- 177:551–562.
marily affecting Schwann cells have serious consequences for Eshed Y, Feinberg K, Poliak S, Sabanay H, Sarig-Nadir O, Spiegel I, et al.
axon structure and survival, nodal organization and synaptic 2005. Gliomedin mediates Schwann cell-axon interaction and the
integrity. molecular assembly of the nodes of Ranvier. Neuron 47:215–229.
Ey B, Kobsar I, Blazyca H, Kroner A, Martini R. 2007. Visualization of
degenerating axons in a dysmyelinating mouse mutant with axonal
loss. Mol Cell Neurosci 35:153–160.
REFERENCES Fannon AM, Sherman DL, Ilynia G, Brophy P, Friedrich VL, Colman
DR. 1995. Novel E-Cadherin-mediated adhesion in peripheral nerve:
Albrecht DE, Sherman DL, Brophy PJ, Froehner SC. 2008. The Schwann cell architecture is stabilized by autotypic adherens junc-
ABCA1 cholesterol transporter associates with one of two distinct tions. J Cell Biol 129:189–202.
dystrophin-based scaffolds in Schwann cells. Glia 56:611–618. Feinberg K, Eshed-Eisenbach Y, Frechter S, Amor V, Salomon D,
Albuerne M, De Lavallina J, Esteban I, Naves FJ, Silos-Santiago I, Vega Sabanay H, et al. 2010. A glial signal consisting of gliomedin and
JA. 2000. Development of Meissner-like and Pacinian sensory cor- NrCAM clusters axonal Na+ channels during the formation of nodes
puscles in the mouse demonstrated with specific markers for corpus- of Ranvier. Neuron 65:490–502.
cular constituents. Ant Rec 258:235–242. Feltri ML, Graus Porta D, Previtali SC, Nodari A, Migliavacca B, Cassetti
Balice-Gordon RJ, Bone LJ, Scherer SS. 1998. Functional gap junctions A, et al. 2002. Conditional disruption of beta 1 integrin in Schwann
in the Schwann cell myelin sheath. J Cell Biol 142:1095–1104. cells impedes interactions with axons. J Cell Biol 156:199–209.
Berthold CH. 1996. Development of nodes of Ranvier in feline nerves: Frei R, Dowling J, Carenini S, Fuchs E, Martini R. 1999. Myelin
an ultrastructural presentation. Microsc Res Techn 34:399–421. formation by Schwann cells in the absence of ß 4 integrin. Glia
Berti C, Bartesaghi L, Ghidinelli M, Zambroni D, Figlia G, Chen ZL, 27:269–274.
et al. 2011. Non-redundant function of dystroglycan and beta1 integ- Friede RL, Beuche W. 1985. A new approach toward analyzing peripheral
rins in radial sorting of axons. Development 138:4025–4037. nerve fiber populations. I. Variance in sheath thickness corresponds
Billings-Gagliardi S, Webster Hd, O’Connel MF. 1974. In vivo and elec- to different geometric proportions of the internodes. J Neuropathol
tron microscopic observations on Schwann cells in developing tad- Exp Neurol 44:60–72.
pole nerve fibers. Am J Anat 141:375–392. Gatzinsky KP. 1996. Node-paranode regions as local degradative centres
Bishop DL, Misgeld T, Walsh MK, Gan WB, Lichtman JW. 2004. Axon in alpha-motor axons. J Microsc Res Techn 34:492–506.
branch removal at developing synapses by axosome shedding. Neuron Georgiou J, Charlton MP. 1999. Non-myelin-forming perisynaptic
44:651–661. Schwann cells express protein zero and myelin-associated glycopro-
Bremer J, O’Connor T, Tiberi C, Rehrauer H, Weis J, Aguzzi A. 2010. tein. Glia 27:101–109.
Ablation of Dicer from murine Schwann cells increases their prolifer- Gess B, Halfter H, Kleffner I, Monje P, Athauda G, Wood PM, et al.
ation while blocking myelination. PloS one 5:e12450. 2008. Inhibition of N-cadherin and beta-catenin function reduces
Brill MS, Lichtman JW, Thompson W, Zuo Y, Misgeld T. 2011. Spatial axon-induced Schwann cell proliferation. J Neurosci Res 86:797–812.
constraints dictate glial territories at murine neuromuscular junc- Giese KP, Martini R, Lemke G, Soriano P, Schachner M. 1992. Mouse
tions. J Cell Biol 195:293–305. P0 gene disruption leads to hypomyelination, abnormal expression of
Bunge RP, Bunge MB, Bates M. 1989. Movements of the Schwann cell recognition molecules, and degeneration of myelin and axons. Cell
nucleus implicate progression of the inner (axon-related) Schwann 71:565–576.
cell process during myelination. J Cell Biol 109:273–284. Gilmour DT, Maischein HM, Nusslein-Volhard C. 2002. Migration
Chernousov MA, Carey DJ. 2000. Schwann cell extracellular matrix and function of a glial subtype in the vertebrate peripheral nervous
molecules and their receptors. Histol Histopathol 15:593–601. system. Neuron 34:577–588.

S C H WA N N C E L L S A N D M Y E L I N • 83
Gokey NG, Srinivasan R, Lopez-Anido C, Krueger C, Svaren J. 2012. Martini R, Schachner M. 1991. Complex expression pattern of tenas-
Developmental regulation of microRNA expression in Schwann cin during innervation of the posterior limb buds of the developing
cells. Mol Cell Biol 32:558–568. chicken. J Neurosci Res 28:261–279.
Gonzalez-Martinez T, Germana A, Catania S, Cobo T, Ochoa-Erena Martini R, Schachner M. 1997. Molecular bases of myelin formation as
FJ, de Carlos F, et al. 2007. Postnatal developmental changes in the revealed by investigations on mice deficient in glial cell surface mol-
expression of ErbB receptors in murine Pacinian cospucles. Neurosci ecules. Glia 19:298–310.
Letts 420:90–95. Martini R, Schachner M, Faissner A. 1990. Enhanced expression of the
Gould RM, Byrd AL, Barbarese E. 1995. The number of Schmidt-Lanterman extracellular matrix molecule J1/tenascin in the regenerating adult
incisures is more than doubled in shiverer PNS myelin sheaths. mouse sciatic nerve. J Neurocytol 19:601–616.
J Neurocytol 24:85–98. Martini R, Zielasek J, Toyka KV, Giese KP, Schachner M. 1995b. Protein
Griffin JW, Thompson WJ. 2008. Biology and pathology of nonmyeli- zero (P0)-deficient mice show myelin degeneration in peripheral
nating Schwann cells. Glia 56:1518–1531. nerves characteristic of inherited human neuropathies. Nat Genet
Grimal S, Puech S, Wagener R, Venteo S, Carroll P, Fichard-Carroll A. 11:281–286.
2010. Collagen XXVIII is a distinctive component of the peripheral Matsumura K, Yamada H, Saito F, Sunada Y, Shimizu T. 1997. Peripheral
nervous system nodes of ranvier and surrounds nonmyelinating glial nerve involvement in merosin-deficient congenital muscular dystro-
cells. Glia 58:1977–1987. phy and dy mouse. Neuromuscul Disord 7:7–12.
Groh J, Heinl K, Kohl B, Wessig C, Greeske J, Fischer S, et al. 2010. Mäurer M, Müller M, Kobsar I, Leonhard C, Martini R, Kiefer R. 2003.
Attenuation of MCP-1/CCL2 expression ameliorates neuropathy Origin of pathogenic macrophages and endoneurial fibroblast-like
in a mouse model for Charcot-Marie-Tooth 1X. Hum Mol Genet cells in an animal model of inherited neuropathy. Mol Cell Neurosci
19:3530–3543. 23:351–359.
Groh J, Weis J, Zieger H, Stanley ER, Heuer H, Martini R. 2012. Melendez-Vasquez C, Carey DJ, Zanazzi G, Reizes O, Maurel P, Salzer
Colony-stimulating factor-1 mediates macrophage-related neural JL. 2005. Differential expression of proteoglycans at central and
damage in a model for Charcot-Marie-Tooth disease type 1X. Brain: peripheral nodes of Ranvier. Glia 52:301–308.
J Neurol 135:88–104. Mierzwa A, Shroff S, Rosenbluth J. 2010. Permeability of the paran-
Grove M, Komiyama NH, Nave KA, Grant SG, Sherman DL, Brophy odal junction of myelinated nerve fibers. J Neurosci: J Soc Neurosci
PJ. 2007. FAK is required for axonal sorting by Schwann cells. J Cell 30:15962–15968.
Biol 176:277–282. Moldovan M, Alvarez S, Pinchenko V, Klein D, Nielsen FC, Wood
Huang JK, Phillips GR, Roth AD, Pedraza L, Shan W, Belkaid W, et al. JN, et al. 2011. Na(v)1.8 channelopathy in mutant mice defi-
2005. Glial membranes at the node of Ranvier prevent neurite out- cient for myelin protein zero is detrimental to motor axons. Brain
growth. Science 310:1813–1817. 134:585–601.
Jessen KR, Mirsky R. 2005. The origin and development of glial cells in Monk KR, Oshima K, Jors S, Heller S, Talbot WS. 2011. Gpr126 is
peripheral nerves. Nat Rev Neurosci 6:671–682. essential for peripheral nerve development and myelination in mam-
Jessen KR, Mirsky R. 2008. Negative regulation of myelination: rel- mals. Development 138:2673–2680.
evance for development, injury, and demyelinating disease. Glia Noakes PG, Bennett MR. 1987. Growth of axons into developing mus-
56:1552–1565. cles of the chick forelimb is preceded by cells that stain with Schwann
Joseph NM, Mukouyama YS, Mosher JT, Jaegle M, Crone SA, cell antibodies. J Comp Neurol 259:330–347.
Dormand EL, et al. 2004. Neural crest stem cells undergo multi- Nodari A, Previtali SC, Dati G, Occhi S, Court FA, Colombelli C, et al.
lineage differentiation in developing peripheral nerves to generate 2008. a6ß 4 integrin and dystroglycan cooperate to stabilize the mye-
endoneurial fibroblasts in addition to Schwann cells. Development lin sheath. J Neurosci 28:6714–6719.
131:5599–5612. Nolte C, Schachner M, Martini R. 1989. Immunocytochemical locali-
Kang H, Tian L, Son YJ, Zuo Y, Procaccino D, Love F, et al. 2007. zation of the neural cell adhesion molecules L1, N-CAM, and J1 in
Regulation of the intermediate fi lament protein nestin at rodent neu- Pacinian corpuscles of the mouse during development, in the adult
romuscular junctions by innervation and activity. J Neurosci: J Soc and during regeneration. J Neurocytol 18:795–808.
Neurosci 27:5948–5957. Novak N, Bar V, Sabanay H, Frechter S, Jaegle M, Snapper SB, et al.
Kirschner DA, Ganser AL. 1980. Compact myelin exists in the absence 2011. N-WASP is required for membrane wrapping and myelination
of basic protein in the shiverer mutant mouse. Nature 283:207–210. by Schwann cells. J Cell Biol 192:243–250.
Kohl B, Fischer S, Groh J, Wessig C, Martini R. 2010. MCP-1/CCL2 Occhi S, Zambroni D, Del Carro U, Amadio S, Sirkowski EE, Scherer
modifies axon properties in a PMP22-overexpressing mouse model for SS, et al. 2005. Both laminin and Schwann cell dystroglycan are nec-
Charcot-Marie-Tooth 1A neuropathy. Am J Pathol 176:1390–1399. essary for proper clustering of sodium channels at nodes of Ranvier.
Koticha D, Maurel P, Zanazzi G, Kane-Goldsmith N, Basak S, Babiarz J, J Neurosci 25:9418–9427.
et al. 2006. Neurofascin interactions play a critical role in clustering Ozcelik M, Cotter L, Jacob C, Pereira JA, Relvas JB, Suter U, et al. 2010.
sodium channels, ankyrin G and beta IV spectrin at peripheral nodes Pals1 is a major regulator of the epithelial-like polarization and the
of Ranvier. Dev Biol 293:1–12. extension of the myelin sheath in peripheral nerves. J Neurosci: J Soc
Landon DN, Williams PL. 1963. Ultrastructure of the node of Ranvier. Neurosci 30:4120–4131.
Nature 199:575–579. Pack AK, Pawson LJ. 2010. Neuroglial modulation in peripheral sensory
Lemke G, Lamar E, Patterson J. 1988. Isolation and analysis of the gene systems. Neuroscientist 16:342–348.
encoding peripheral myelin protein zero. Neuron 1:73–83. Parpura V, Heneka MT, Montana V, Oliet SH, Schousboe A, Haydon
Lewallen KA, Shen YA, De la Torre AR, Ng BK, Meijer D, Chan JR. PG, et al. 2012. Glial cells in (patho)physiology. J Neurochem
2011. Assessing the role of the cadherin/catenin complex at the 121(1):4–27.
Schwann cell-axon interface and in the initiation of myelination. Pawson L, Prestia LT, Mahoney GK, Guclu B, Cox PJ, Pack AK. 2009.
J Neurosci: J Soc Neurosci 31:3032–3043. GABAergic/glutamatergic-glial/neuronal interaction contributes to
Martini R. 1994. Expression and functional roles of neural cell surface rapid adaptation in pacinian corpuscles. J Neurosci: J Soc Neurosci
molecules and extracellular matrix components during development 29:2695–2705.
and regeneration of peripheral nerves. J Neurocytol 23:1–28. Pereira JA, Baumann R, Norrmen C, Somandin C, Miehe M, Jacob C,
Martini R, Mohajeri MH, Kasper S, Giese KP, Schachner M. 1995a. et al. 2010. Dicer in Schwann cells is required for myelination and
Mice doubly deficient in the genes for P0 and myelin basic protein axonal integrity. J Neurosci 30:6763–6775.
show that both proteins contribute to the formation of the major Peters A, Palay SL, Webster Hd. 1991. The fine structure of the nervous
dense line in peripheral nerve myelin. J Neurosci 15:4488–4495. system. New York: Oxford University Press.

84 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
Phillips DD, Hibbs RG, Ellison JP, Shapiro H. 1972. An electron Svaren J, Meijer D. 2008. The molecular machinery of myelin gene tran-
microscopic study of central and peripheral nodes of Ranvier. J Anat scription in Schwann cells. Glia 56:1541–1551.
111:229–238. Thaxton C, Pillai AM, Pribisko AL, Dupree JL, Bhat MA. 2011. Nodes
Reddy LV, Koirala S, Sugiura Y, Herrera AA, Ko CP. 2003. Glial cells of Ranvier act as barriers to restrict invasion of flanking paranodal
maintain synaptic structure and function and promote development domains in myelinated axons. Neuron 69:244–257.
of the neuromuscular junction in vivo. Neuron 40:563–580. Tricaud N, Perrin-Tricaud C, Bruses JL, Rutishauser U. 2005. Adherens
Riethmacher D, Sonnenberg-Rietmacher E, Brinkmann V, Yamaai T, junctions in myelinating Schwann cells stabilize Schmidt-Lanterman
Lewin RG, Birchmeier C. 1997. Severe neuropathies in mice with incisures via recruitment of p120 catenin to E-cadherin. J Neurosci
targeted mutations in the ErbB3 receptor. Nature 389:725–730. 25:3259–3269.
Rosenbluth J. 2009. Multiple functions of the paranodal junction of Ulzheimer JC, Peles E, Levinson SR, Martini R. 2004. Altered expres-
myelinated nerve fibers. J Neurosci Res 87:3250–3258. sion of ion channel isoforms at the node of Ranvier in P0-deficient
Saher G, Quintes S, Nave KA. 2011. Cholesterol: a novel regulatory role myelin mutants. Mol Cell Neurosci 25:83–94.
in myelin formation. Neuroscientist 17:79–93. Verrier JD, Semple-Rowland S, Madorsky I, Papin JE, Notterpek L.
Saito F, Moore SA, Barresi R, Henry MD, Messing A, Ross-Barta SE, 2010. Reduction of Dicer impairs Schwann cell differentiation and
et al. 2003. Unique role of dystroglycan in peripheral nerve myeli- myelination. J Neurosci Res 88:2558–2568.
nation, nodal structure, and sodium channel stabilization. Neuron Wanner IB, Guerra NK, Mahoney J, Kumar A, Wood PM, Mirsky R,
38:747–758. et al. 2006a. Role of N-cadherin in Schwann cell precursors of grow-
Salzer JL, Brophy PJ, Peles E. 2008. Molecular domains of myelinated ing nerves. Glia 54:439–459.
axons in the peripheral nervous system. Glia 56:1532–1540. Wanner IB, Mahoney J, Jessen KR, Wood PM, Bates M, Bunge MB.
Sanes JR, Lichtman JW. 2001. Induction, assembly, maturation and main- 2006b. Invariant mantling of growth cones by Schwann cell precur-
tenance of a postsynaptic apparatus. Nat Rev Neurosci 2:791–805. sors characterize growing peripheral nerve fronts. Glia 54:424–438.
Schafer DP, Rasband MN. 2006. Glial regulation of the axonal mem- Wehrle B, Chiquet M. 1990. Tenascin is accumulated along developing
brane at nodes of Ranvier. Curr Opin Neurobiol 16:508–514. peripheral nerves and allows neurite outgrowth in vitro. Development
Scherer SS, Arroyo EJ. 2002. Recent progress on the molecular organiza- 110:401–415.
tion of myelinated axons. J Peripher Nerv Syst 7:1–12. Weinberg HJ, Spencer PS. 1975. Studies on the control of myelinogen-
Schröder JM, Bohl J, v. Bardeleben U. 1988. Changes of the ratio esis. I. Myelination of regenerating axons after entry into a foreign
between myelin thickness and axon diameter in human developing unmyelinated nerve. J Neurocytol 4:395–418.
sural, femoral, ulnar, facial, and trochlear nerves. Acta Neuropathol Yang Y, Ogawa Y, Hedstrom KL, Rasband MN. 2007. betaIV spectrin is
76:471–483. recruited to axon initial segments and nodes of Ranvier by ankyrinG.
Shapiro L, Doyle JP, Hensley P, Colman DR, Hendrickson WA. J Cell Biol 176:509–519.
1996. Crystal structure of the extracellular domain from P0, Yin X, Kidd GJ, Nave KA, Trapp BD. 2008. P0 protein is required for
the major structural protein of peripheral nerve myelin. Neuron and can induce formation of schmidt-lantermann incisures in myelin
17:435–449. internodes. J Neurosci 28:7068–7073.
Sherman DL, Brophy PJ. 2005. Mechanisms of axon ensheathment and Yoon C, Korade Z, Carter BD. 2008. Protein kinase A-induced phos-
myelin growth. Nat Rev Neurosci 6:683–690. phorylation of the p65 subunit of nuclear factor-kappaB promotes
Sherman DL, Krols M, Wu LM, Grove M, Nave KA, Gangloff YG, et al. Schwann cell differentiation into a myelinating phenotype. J Neurosci
2012. Arrest of myelination and reduced axon growth when Schwann 28:3738–3746.
cells lack mTOR. J Neurosci: J Soc Neurosci 32:1817–1825. Yu WM, Yu H, Chen ZL, Strickland S. 2009. Disruption of laminin
Sherman DL, Tait S, Melrose S, Johnson R, Zonta B, Court FA, et al. in the peripheral nervous system impedes nonmyelinating Schwann
2005. Neurofascins are required to establish axonal domains for sal- cell development and impairs nociceptive sensory function. Glia
tatory conduction. Neuron 48:737–742. 57:850–859.
Spiegel I, Adamsky K, Eshed Y, Milo R, Sabanay H, Sarig-Nadir O, et al. Zhang Y, Bekku Y, Dzhashiashvili Y, Armenti S, Meng X, Sasaki Y, et al.
2007. A central role for Necl4 (SynCAM4) in Schwann cell-axon 2012. Assembly and maintenance of nodes of ranvier rely on distinct
interaction and myelination. Nat Neurosci 10:861–869. sources of proteins and targeting mechanisms. Neuron 73:92–107.

S C H WA N N C E L L S A N D M Y E L I N • 85
8.
MICROGLIAL CELLS
Wolfgang J. Streit

A B B R E VI AT I O N S 2 H I S TO R I C A L P E R S P E C T I VE S

AD Alzheimer disease Following early descriptions of neuroglia by Virchow in the


CNS central nervous system mid nineteenth century, other contemporary pathologists
CX3CL1 fractalkine and psychiatrists, including Nissl and Alzheimer, commented
CX3CR1 fractalkine receptor on the possibility that the developing CNS was populated by
Iba1 ionized calcium binding adaptor molecule 1 cells of non-neuroectodermal origin. Speculation abounded as
LPS lipopolysaccharide to the source of these invading cells, but attention was focused
MHC major histocompatibility complex with increasing consistency on the possibility that mesoder-
NSE nonspecific esterase mally derived cells were the invaders. Eventually this led to the
RhIC rhodamine isothiocyanate formulation by Cajal of el tercer elemento, the third element of
TPPase thiamine pyrophosphatase the CNS, referring to a group of cells that was morphologi-
cally distinct from both first and second elements (neurons
and astrocytic neuroglia). Cajal’s third element, defined by
1 INTRODUCTION
him strictly in morphological terms, received further distinc-
tion into oligodendrocytes and microglia by del Rio-Hortega,
Microglial cell numbers are thought to make up 5% to 20% of
the Spanish neuroanatomist who provided the first systematic
the entire central nervous system (CNS) glial cell population,
investigation on microglial cells. Del Rio-Hortega’s detailed
and, assuming that the brain is composed roughly of equal num-
cytological observations, which remain quite relevant even
bers of neurons and glia, one must conclude that 2.5% to 10%
today, gave rise to a longstanding controversy over the ori-
of all brain cells are microglia. This simple calculation is aston-
gin of microglia that dominated microglial research for sev-
ishing only insofar as the very existence of microglial cells was
eral decades into the 1990s. In recent history, the debate over
questioned by some as recently as 20 years ago (Graeber 2010).
microglial ontogeny has been succeeded by another fervent
Research on microglial cells has grown significantly during
discussion, this one focusing on the functional significance of
the past 25 years. This positive trend can be attributed largely to
activated microglial cells, that is, whether activation of micro-
the development of reliable histological methods for identifying
glial cells is a beneficial or harmful process. The significance
the cells in CNS tissue sections using light microscopy. Most of
of this discussion pertains primarily to the issue of bystander
these methods, which are described in this chapter, can also be
damage, a phenomenon that is thought to be important in
carried to the electron microscopic level allowing verification of
both acute and chronic CNS injury and disease. For exam-
microglial identity through direct comparison of the presence
ple, regarding the pathogenesis of Alzheimer disease (AD),
of a specific marker with ultrastructural morphology. Before
many scientists believe that microglial activation is a critical
the advent of microglia-specific markers, electron microscopy
component in the development of AD reflecting a type of
was almost always needed for positive identification using solely
chronic glial inflammation that is triggered by the presence of
morphological criteria. Modern neurobiology now has available
amyloid-beta proteins and causes neurodegenerative changes,
an assortment of reliable light microscopic techniques that read-
such as neurofibrillary tangles through excessive production
ily enable identification of microglial cells by neuroscientists.
of neurotoxic molecules by activated microglia (Akiyama et
Regarding the terminology used in this chapter, it is impor-
al. 2000). However, treatments with antiinflammatory drugs
tant to point out that microglial cells in the adult CNS can
have failed to show clear benefits, and a new view on AD
assume at least four clearly identifiable states: (1) resting (rami-
pathogenesis focusing on the aging-related, structural dete-
fied) microglia, which are distributed ubiquitously throughout
rioration of microglia (cell senescence) has emerged (Streit
the normal and nonpathological CNS; (2) activated (reactive)
2006) (see final section of this chapter).
microglia, which occur in pathological states, but are not always
phagocytic; (3) phagocytic microglia, which appear as rounded
brain macrophages; and (4) dystrophic microglia, which are
senescent cells. For additional descriptions of these microglial 3 O R I G I N A N D L I N E AG E O F M I C R O G L I A
states the reader is referred elsewhere (see chapter 48) (Morioka
et al. 1992; Streit and Kreutzberg 1988; Streit and Xue 2009; Two related issues were at the heart of a longstanding debate
Streit et al. 2004). over the origin of microglia: (1) Are microglia derived from

86
mesoderm or from neuroectoderm? (2) When and how do These process-bearing embryonic microglia are at an inter-
microglia populate the CNS? As discussed elsewhere (Streit mediate stage of differentiation and have not yet matured to
2001), it appears that both issues have been largely resolved the fully ramified morphology that is characteristic of adult
and it is clear that (1) microglia are of myelomonocytic microglia.
lineage and therefore likely derived from hemangioblastic During perinatal stages in rodents at about embry-
mesoderm, and (2) microglia become part of the CNS paren- onic day 20, unique accumulations of so-called ameboid
chyma early during embryonic development at about the microglial cells become apparent (Ling and Wong 1993) as
time neurulation has been completed. Microglial precursor aggregated clusters of rounded cells in specific anatomical
cells are an integral component of the CNS during embry- locations, most prominently in the supraventricular cor-
onic and postnatal development. Cells that are most aptly pus callosum (Hurley et al. 1999; Ling and Wong 1993).
described as “fetal macrophages” (Takahashi et al. 1989) In the postnatal CNS, ameboid microglia within these
populate the developing neuroectoderm as early as embry- clusters undergo mitosis, and these prominent supraven-
onic day 8 in rodents and late during the first trimester in tricular clusters of proliferating cells were recognized by
humans (Alliot et al. 1999; Ginhoux et al. 2010; Monier et al. early microglial researchers who termed them fountains of
2007). These fetal macrophages can be visualized using lec- microglia (Kettenmann et al. 2011). Contemporary neuro-
tin histochemical markers that also label microglia (Sorokin biologists might be inclined to apply the term microglial
et al. 1992) and therefore they are considered the earliest progenitor cells instead of ameboid microglia to emphasize
detectable microglial precursor cells. Significantly, fetal their status as immature precursor cells. Microglial pro-
macrophages can be found in the primitive neuroectoderm genitor cells in the corpus callosum persist through the first
before it becomes vascularized (Chan et al. 2007; Monier two postnatal weeks and during that time the cells migrate
et al. 2007) (Fig. 8.1), which eliminates the possibility that into the overlying cerebral cortex, differentiating into fully
blood-borne monocytes serve as direct microglial precursors. ramified microglia. This perinatal burst of microgliogen-
It is likely that yolk sac–derived fetal macrophages are direct esis occurs to facilitate microglial colonization of the fore-
precursors for microglia, whereas blood monocytes are bone brain, which undergoes its most expansive growth during
marrow–derived (Prinz et al. 2011) (see also chapter 15). As the postnatal period (see Fig. 8.1). Ameboid microglial pro-
the embryonic CNS develops toward the perinatal stage and genitor cells can be distinguished antigenically from rami-
various neural cell types mature and differentiate, fetal mac- fied microglia in that ameboid cells are readily labeled with
rophages also metamorphose from rounded cells to more the ED1 antibody (directed against rat CD68), whereas
differentiated embryonic microglia with short processes. ramified microglia are not (Milligan et al. 1991). At birth,
ED1-positive cells are abundant in the CNS but they disap-
pear completely by the third postnatal week (Milligan et al.
1991), indicating that differentiation of ameboid cells into
ramified microglia is complete at that point in time. ED1 is
A Embryonic Neuroepithelium a macrophage marker that recognizes an intracytoplasmic,
Fetal macrophages
lysosomal antigen whose expression increases during phago-
cytosis (Dijkstra et al. 1985); thus, disappearance of ED1
immunoreactivity during postnatal development shows that
B Perinatal Brain fully differentiated ramified microglia are in a phagocytoti-
Cluster of ameboid cally quiescent state.
microglia in corpus
callosum
During adult life there is little if any replacement of
microglia from exogenous sources, such as the bone marrow,
as shown in parabiotic studies (Ajami et al. 2011). Microglia
have the greatest mitotic potential of all parenchymal cells in
C Adult Brain the CNS and are therefore capable of self-renewal. Microglial
Ramified microglia
mitosis in the normal CNS occurs at a very low rate, indi-
cating low turnover and a long lifespan of cells (Lawson et
al. 1990). Nonetheless, a small fraction of microglial cells
may undergo replacement by bone marrow–derived pre-
cursors via perivascular cells. The latter are mononuclear
phagocytes that reside in the Virchow-Robin (perivascular)
spaces surrounding medium and small-sized cerebral vessels.
Perivascular cells, which are replaced continuously by bone
Figure 8.1 Summary of Microglial Ontogeny During Three marrow–derived progenitors, on occasion may penetrate the
Developmental Stages A. Fetal macrophages are found in the developing perivascular basement membrane, enter the parenchyma and
neuroectoderm as early as embryonic day 8 in rodents. B. In the differentiate into process-bearing microglia. Studies using
perinatal brain, clusters of dividing ameboid microglia are found in the bone marrow chimeras and localization of major histocom-
supraventricular corpus callosum. The cells migrate from the clusters into
the cerebral cortex and differentiate into ramified microglia. C. Ramified patibility antigens support this idea (Hickey and Kimura
microglia have colonized throughout the adult brain. 1988; Streit et al. 1989).

8. M I C R O G L I A L C E L L S • 87
4 M ET H O D S F O R S TA I N I N G M I C R O G L I A 4.3 I M MU N O H I S TO C H E M I C A L D ET EC T I O N
O F M I C RO G L I A
4.1 S I LVE R C A R B O NAT E M ET H O D The microglial plasma membrane is complex and studded
with a large variety of receptor and adhesion molecules in
The first selective stain for microglia was the weak silver car- addition to enzymatic activities. Because of this large reper-
bonate method developed by del Rio-Hortega. Despite its toire of surface antigens, numerous antibodies are now avail-
capriciousness, this method remained the only useful histo- able to facilitate immunohistochemical staining of microglia.
chemical procedure for at least 50 years. As with many other Interestingly, many of these monoclonal antibodies were not
histochemical techniques involving metallic silver impregna- produced with the intention of specifically marking microg-
tions, Hortega’s weak silver carbonate method for microglia lia, but were meant to target differentiation antigens found
has very specific fixation requirements that do not, however, on cells of the immune system, such as macrophages, thymo-
guarantee reproducible results in every preparation. The results cytes, and lymphocytes. Following initial failures of demon-
obtained are quite variable in terms of numbers of microglia strating presence of monocytic and lymphoid antigens on
stained, and vary also with the animal species used. For rea- human microglia (Oehmichen et al. 1979), it was found later
sons unknown, the method seems to work reliably only in that a mouse macrophage–specific antigen could be localized
rabbit brain. Although it can be carried to the electron micro- on resting microglia with a monoclonal antibody designated
scopic level, its usefulness is limited because of poor structural F4/80 (Hume et al. 1983; Perry et al. 1985). These investigators
preservation and the deposition of metallic precipitates that also succeeded in showing the presence of Fc and complement
can obscure much of the cellular detail. receptors on resting mouse microglia using antibodies 2.4G2
and Mac-1, respectively. Analogously, ramified microglia in rat
and mouse brain can be demonstrated reliably using antibod-
4.2 E N ZY M E H I S TO C H E M I C A L M ET H O D S
ies against the CD11b antigen, also known as the CR3 com-
Thiamine pyrophosphatase (TPPase) and nucleoside diphos- plement receptor (Graeber et al. 1988). It is important to note
phatase (NDPase) are the most reliable and specific enzyme that these receptors are also found on macrophages in non-
histochemical methods for staining resting microglial cells neural tissues, and their presence on microglia underscores the
in a variety of species. These methods have been used suc- phagocytic potential of these cells, as well as their close rela-
cessfully to localize microglia at both light and electron tionship to the myelomonocytic cell lineage. Crossreactivity
microscopic levels (Murabe and Sano 1981; Schnitzer 1989). of macrophage-specific antibodies with microglia and blood
TPPase activity, originally described to be localized to the monocytes has been taken as evidence that microglia are
Golgi apparatus in a variety of cell types, including neurons, derived from monocytes. However, a direct lineage relation-
was later found to be associated specifically with the plasma ship between monocytes and microglia is not likely because
membrane of microglial cells and with blood vessels in the both microglia and monocytes are fully differentiated cell
CNS (Murabe and Sano 1981). Nonspecific esterase (NSE) types that may arise from different precursor cells, as dis-
is an enzyme used to identify microglia in mixed brain cul- cussed. A distinction between microglia and so-called “other
tures (Sawada et al. 1990), but is of little use for detecting brain macrophages” has also been made using flow cytometric
resting microglial cells in tissue sections. Actively prolifer- analyses, which have shown that “other CNS macrophages”
ating, reactive microglial cells in the hypoglossal nucleus are phenotypically distinct (CD11b/c + and CD45hi) from
after peripheral nerve transection do not show any stain- parenchymal microglia (CD11b/c + and CD45low) (Ford et al.
ing for this enzyme (Schelper and Adrian 1980). However, 1995).
nonspecific esterase can be found in microglia-derived and In the human brain, microglia can also be localized using
other brain macrophages that are prevalent in stab wounds. antibodies against macrophage surface receptors (Akiyama
Nonspecific esterase enzyme histochemistry thus supports and McGeer 1990). In addition to the expression of Fc and
the view that microglia in vitro are, in fact, microglia that complement receptors, other cell adhesion molecules are
have transformed into brain macrophages as a consequence expressed constitutively on resting microglia in normal brain.
of having been placed into cell culture. Activated and/or Belonging to the integrin superfamily of adhesion molecules,
phagocytic microglia in cell culture or the pathological brain these include typical lymphocytic antigens, such as lympho-
show increased activities for a number of other enzymes that cyte function antigen, CD4 antigen, as well as leukocyte com-
are absent from resting microglial cells in situ. These include mon antigen (Akiyama and McGeer 1990; Perry and Gordon
acid phosphatase, 5′-nucleotidase, and oxidoreductase. 1987). Species differences among mouse, rat, and human
Other studies have shown the presence of nitric oxide syn- in the constitutive expression of these molecules on resting
thase, cyclooxygenase, lysosomal proteinases, plasminogen microglia have been observed, and these are likely caused by
activator, lysozyme, purine nucleoside phosphorylase, and both antibody specificities, as well as variations in tissue pro-
elastase (Banati et al. 1993; Castellano et al. 1990; Nakajima cessing techniques. B-lymphocyte antigens are detectable on
et al. 1992). It is worth noting that some of these enzymatic human microglial cells using monoclonal antibodies LN-1 and
activities are also found in other glial cells types and there- LN-3, the latter recognizing HLA-DR antigens (Dickson and
fore are not always useful as selective histochemical markers Mattiace 1989; Miles and Chou 1988). Although the LN-1
for microglial cells. antibody may label both astrocytes and microglia depending

88 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
on fixation and tissue processing techniques, antibody LN-3 A
has an exclusive specificity for microglia in both normal and
pathologic human brain. Figures 8.2 and 8.3 provide examples
of LN-3 staining of resting and activated microglia in human
brain. Significantly, staining with LN-3 is fixation-sensitive
and optimal preparations require lightly fixed tissues (Streit
and Sparks 1997).
It is apparent then that the microglial surface membrane
bears molecules usually associated with white blood cells,
including antigens of the major histocompatibility complex
(MHC). Although it was once thought that MHC antigens B
were entirely absent from brain, supporting the notion of the
brain as an immunologically privileged organ, it is now well
established that MHC antigens are expressed in normal brain,
and the principal parenchymal cell type expressing MHC anti-
gens is the microglial cell (Craggs and Webster 1985; Hayes et
al. 1987; Streit et al. 1989). The expression of MHC antigens
in normal brain also includes endothelial and perivascular
cells, and there are considerable species differences in the levels
of constitutive MHC antigen expression on these various cell
types. Major histocompatibility antigen expression on micro- C
glia is increased dramatically under pathological conditions,
but also increases with normal aging in rodents, non-human
primates, and humans (Finch et al. 2002; Perry et al. 1993;
Sheffield and Berman 1998; Streit and Sparks 1997). To date a
truly specific marker for microglia, namely, one that does not

Figure 8.3 Resting and Activated Microglia Stained with LN-3 Antibody
in Human Brain. A. Activated microglia stain more strongly than resting
ones and have a bushy appearance in a 41-year-old individual (arrows).
B. Microglial rod cells in the cerebral cortex of a 94-year-old individual.
C. Bushy microglia in the 94-year-old, possibly representing two or more
cells that have fused to form a microglial cluster. Bar = 100 μm.

cross-react with other macrophages, has not been generated.


All antibodies and lectins that react with resting microglial
B cells also label activated microglia and microglia-derived brain
macrophages, as well as peripheral macrophages. Conversely,
not all antibodies that react with brain or other macrophages
label resting microglia, suggesting that the antigenic repertoire
of resting microglia is smaller than that of activated microglia
and brain macrophages, and/or that the level of expression of
certain antigens in resting cells is below the detection limit of
immunohistochemistry (Table 8.1). One antibody that has
proved to be particularly useful because its reactivity is rela-
tively unaffected by prolonged fixation or the species being
studied (notably mouse, rat, and human) is the one raised
against the ionized calcium binding adaptor molecule 1, Iba1
Figure 8.2 Ramified Resting Microglial Cells. A. Ramified resting (Ito et al. 1998). Immunostaining with Iba1 antibody can be
microglial cell in the nonpathological brain of a 64-year-old human used to visualize all morphological states of microglia (see
visualized with monoclonal antibody LN-3. Note extensive branching of Table 8.1).
cytoplasmic processes. Bar = 20 μm. B. Ramified resting microglial cell in
perivascular position. The blood vessel (BV) is seen coursing horizontally.
Immunohistochemical localization of ramified micro-
LN-3 immunohistochemistry in non-pathological brain of a 68-year-old glia has been achieved through the use of phosphotyrosine
individual. Bar = 20 μm. antibodies (Griffith et al. 2000). This procedure detects the

8. M I C R O G L I A L C E L L S • 89
Table 8.1 IN VIVO STATES OF MICROGLIAL BIOLOGY AND ASSOCIATED PHENOTYPIC CHARACTERISTICS

RESTING ACTIVATED PHAGOCYTIC DYSTROPHIC

Proliferation −/+ + + −

Iba1 + + + +

Griffonia simplicifolia + + + ND
B4-isolectin (rat) and Ricinus
communis lectin (mouse)

Vimentin − + + ND

Macrophage markers (CD68) − −/+ −/+ −

CR3 complement receptor + + + +


(CD11b)

MHC class I antigen − + + ND

MHC class II (Ia) antigen −/+ + + +

CD4 antigen −/+ + + ND

CD8 antigen − −/+ + ND

Leukocyte common antigen − + + ND


(CD45)
Antigens listed are relevant for rat, mouse, and human.
Explanation of symbols: − absent; −/+ weak; + strong; ND not determined.

products of an enzymatic reaction carried out by tyrosine for further defining microglial phenotypes and advancing
kinase. There are functional implications for this observa- understanding of their functional roles.
tion, because it is known that tyrosine kinases are commonly
associated with cell surface receptors, which are plentiful on 4.4 L EC T I N H I S TO C H E M I C A L D ET EC T I O N
the microglial membrane. Various other immunohistochemi- O F M I C RO G L I A
cal methods aimed at detecting somewhat unconventional
antigens, such as vaults, ferritin, and lipocortin-1, have also While investigating the distribution of complex carbohydrates
been described (Chugani et al. 1991; Kaneko et al. 1989; in nervous tissue using lectin histochemistry, it was noted that
McKanna 1993). Vaults, which are multiarched ribonucleo- the B4-isolectin derived from Griffonia simplicifolia resulted
protein particles of unknown function, appear to be enriched in the selective visualization of a population of rat glial cells
in microglia during ontogeny, but mostly disappear in adult that were identified as microglia (Streit et al. 1985). These ini-
cells. Ferritin, on the other hand, is a well-known iron-storage tial observations were confirmed soon thereafter in human
protein, and its detection in microglia suggests that the cells tissue, where it was shown that the lectin from Ricinus com-
actively participate in the trafficking and sequestration of munis could be used as a histochemical marker for microglia
iron. Lipocortin-1 is a Ca2+-binding protein that is thought (Mannoji et al. 1986). Both lectins have similar sugar-binding
to function as an antiinflammatory or immunosuppressive characteristics in recognizing anomeric forms of galactose, with
molecule. Griffonia simplicifolia binding to α-D-galactose and Ricinus
In summary, remarkable progress has been made in the communis recognizing ß-d-galactose residues. An additional
development of immunohistochemical procedures used for ß-d-galactose–binding lectin derived from mistletoe has
the detection and identification of microglia. Given the great been shown to preferentially stain human over rat microglia
variety of receptor molecules known to cover the microglial (Suzuki et al. 1988), emphasizing the subtle difference in gly-
cell surface, it is likely that additional antibodies will be devel- cocalyx composition between rodent and human microglial
oped in the future, and undoubtedly these will be important cells, being one of anomeric configuration. The galactose sugar

90 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
residues occur as terminal sugars in the oligosaccharide side 5 M I C R O G L I A A N D R E L AT E D
chains of nervous system glycoproteins that are embedded CELL TYPES
in the microglial plasma membrane, as revealed by electron
microscopy (Streit and Kreutzberg 1987). Interestingly, the
5.1 D E FI N IT I O NS
lectin from Lycopersicum esculentum (tomato), which has an
affinity for poly-N-acetyl lactosamine residues can also be used Microglial cells, because they can change their morphology
for staining rat microglia (Acarin et al. 1994), indicating diver- and appearance in certain pathological and developmental
sity of carbohydrate domains present on the microglial cell states, have been given various descriptive names and attri-
surface. The specific nature and function of lectin-binding gly- butes, such as rod cells, gitter cells, globoid, and ameboid cells,
coproteins on microglial cells has not yet been resolved; how- to name a few. Even though these terms accurately reflect the
ever, it is likely that the carbohydrate domains of many of the cells’ changed appearance, the descriptive terminology has
surface receptors, described in the preceding section, account somewhat obscured the true identity of the cells associated
in large part for lectin binding. Lectin staining is perhaps the with the term microglia. In the normal adult brain, resting
quickest and most resilient method for visualizing microglia microglia can be defined both in terms of morphology and
in tissue sections because the carbohydrate epitopes, unlike phenotype. They are highly branched (ramified) glial cells
most proteins, are largely unaffected by cross-linking through with a small amount of perinuclear cytoplasm and a small,
aldehyde-based fixation and tissue processing techniques. dense, and heterochromatic nucleus (Figs. 8.2–8.5). They can
be distinguished easily from other glial cells by their surface
immunophenotype; that is, they are the only glial cell type
4.5 OT H E R M ET H O D S F O R L A B E L I N G
that constitutively expresses the CR3 complement recep-
M I C RO G L I A
tor (CD11b antigen) and binds lectins with a specificity for
Among all the parenchymal cell types in the mature CNS, galactose residues. Furthermore, at the ultrastructural level
microglia are the cells with the greatest potential for mitosis. microglia are recognizable as true parenchymal constituents
Their ability to divide and proliferate makes them amenable of the CNS because they are located outside of the vascular
to labeling with 3H-thymidine and other markers of dividing basement membrane. At the same time, they may be consid-
cells, such as 5-bromo-2′-deoxyuridine or proliferating cell ered part of the perivascular glia limitans, because microglial
nuclear antigen. Microglial cell division is usually triggered by cytoplasmic processes are found incorporated intermingled
perturbations in CNS homeostasis, such as neuronal injury, with the layer of astrocytic foot processes (Lassmann et al.
but there is also evidence that microglial cell division occurs 1991). The observation that microglia are frequently found
normally in the rodent brain, albeit at a low rate (Korr et al. in the vicinity of blood vessels has resulted in the use of the
1983). Microglia may also be labeled directly or indirectly term perivascular microglia, which is yet another descriptive
using various dyes and tracer substances. Following intrap- term referring to parenchymal microglial cells, as defined,
eritoneal injection of the fluorescent dye, rhodamine isothio- which happen to be located near a cerebral blood vessel (see
cyanate (RhIC), labeled ameboid microglia, were observed Fig. 8.2). Perivascular microglia are not to be confused with
in the corpus callosum. Subsequently, the ameboid cells were so-called perivascular cells, which, unlike microglia, are not
observed to transform into RhIC-labeled ramified microglial
cells (Leong and Ling 1992), confirming earlier observations
using colloidal carbon introduced in the form of India ink.
An indirect method for labeling microglia makes use of their
ability to phagocytose dead or dying neurons. Following injec-
tion of the appropriate tracer substance into axons, the tracer
is retrogradely transported toward the parent neuron cell bod-
ies. If the injected nerve is also axotomized, in some instances
this will cause degeneration of the parent neurons, followed by
removal of dead neurons by local microglia that phagocytose
not only the neuronal debris, but also the tracer substance and
thus become labeled. Such experiments have been successfully
carried out in various systems, including the visual system,
resulting in the labeling of retinal microglia with the carbocya-
nine dye DiI (Thanos 1991), the dorsal motor nucleus of the
vagus where the neural tracer fluorogold was used (Rinaman
et al. 1991), and also the rat facial nucleus where fluorogold
was used in conjunction with toxic ricin to induce motor
neuron degeneration (Streit and Graeber 1993). Interestingly,
direct injection of fluorogold into the brain does not label Figure 8.4 Ultrastructural Appearance of a Perineuronal Microglial
Cell in the Rat Brain. The cell has a heterochromatic nucleus (Nu) and
ramified microglial cells, but if the cells are maintained in cul- prominent cisternae of rough endoplasmic reticulum (arrowheads). Its
ture, where they undergo macrophage transformation, they do plasma membrane, which is accentuated by lectin staining, is directly
take up fluorogold rather avidly (Pennell and Streit 1998). apposed to the neuronal plasma membrane (arrows). Bar = 2 μm.

8. M I C R O G L I A L C E L L S • 91
hippocampal formation, olfactory telencephalon, portions
of the basal ganglia, and substantia nigra. A total number of
3.5 × 106 microglia is estimated to reside in the adult mouse
brain (Lawson et al. 1990), although that estimate is probably
too conservative. Individual microglial cells typically occupy
a distinct territory; that is, neighboring cells do not contact
each other with their cytoplasmic processes. The morphology
and branching patterns of microglial cells show heterogeneity
among different brain regions, which is perhaps most remark-
able when comparing cells in gray and white matter.
Although microglia in gray matter tend to be profusely
ramified with processes extending into all directions, cells in
the white matter often align their cytoplasmic extensions in
parallel, but also at right angles to nerve fiber bundles. Thus,
the cell shape of microglia adapts to the geometry of the brain
region they populate. The microglial immunophenotype is
heterogeneous and appears to be influenced by the chemical
composition of the microenvironment. For example, MHC
class II–positive, as well as CD4-positive microglia are local-
ized preferentially in white matter of normal brain (Hayes
et al. 1987; Perry and Gordon 1987; Streit et al. 1989). Brain
regions lacking a blood-brain barrier, such as the circumven-
Figure 8.5 Electron Micrograph. The electron micrograph shows a tricular organs, do show microglia and microglia-like cells,
microglial cell (M), an oligodendrocyte (O), and a large dendrite (D). such as the Kolmer cells of the choroid plexus, with a different
Both cell types have a heterochromatic nucleus that is larger in the
oligodendrocyte. Bar = 2 μm. immunophenotype, suggesting that the chemical milieu influ-
ences microglial and macrophage phenotypes. This is sup-
ported further by in vivo studies showing profound changes
part of the CNS parenchyma but are separated from it by a in microglial phenotype after brain lesions, such as forebrain
perivascular basement membrane (see chapter 9). Perivascular ischemia and kainic acid injections that compromise the
cells are components of the vascular wall and located in the blood-brain barrier. Similar changes in microglial immuno-
perivascular spaces. They fit the morphological definition phenotype occur also when the cells are maintained in vitro
of a pericyte (Graeber and Streit 1990b; Mato et al. 1986). using serum-containing culture medium.
Perivascular cells are phagocytic and typically express MHC
antigens and macrophage antigens (CD68) constitutively
5.3 M I C RO G L I A I N C E L L CU LT U R E
(Graeber et al. 1989b; Mato et al. 1986; Streit et al. 1989),
which has made it difficult in certain pathological situations Although the maintenance of microglia/brain macrophages
to distinguish between perivascular cells and perivascular in cell culture was used and described in the 1930s, possibly
microglia. However, in the normal brain these two cell types even earlier, the procedure did not gain widespread popular-
are readily distinguished by their morphology and surface ity until the 1980s. The technique described by Giulian and
immunophenotype. Perivascular cells are seen only in asso- Baker (1986) has been widely used with numerous modifica-
ciation with blood vessels, they are not ramified but have an tions. When culturing microglial cells, perhaps more so than
elongate shape, and they can be specifically labeled in the rat with any other neural cell type, it is apparent that microglia in
with ED1 and ED2 antibodies (Graeber et al. 1989a,b). Thus, vitro are quite different from microglia in vivo. The prepara-
there are at least two clearly definable and indigenous sources tion of primary mixed brain cultures from which microglia are
of brain macrophages present in normal brain: microglia isolated causes the generation of large amounts of tissue debris
and perivascular cells. The term brain macrophage is generic that, together with a high serum content of the growth media,
and encompasses all phagocytic cells in the CNS, including promotes rapid transformation of microglial cells into brain
blood-derived monocytes, which may enter the CNS follow- macrophages. Isolated microglia plated onto plastic culture
ing lesions that disrupt the blood-brain barrier. dishes take on a rounded cell shape resembling immature ame-
boid microglial precursor cells, and it was once widely accepted
that cultured microglia are the same as ameboid microglia.
5.2 FAC TO R S A FFEC T I N G D I S T R I BU T I O N,
Because isolated microglia in vitro are essentially brain mac-
MO R P H O L O GY, A N D P H E N OT Y P E
rophages, it is important to distinguish this advanced func-
O F M I C RO G L I A
tional (phagocytic) state from the precursor state that defines
Microglia are distributed ubiquitously throughout the nor- ameboid microglial cells in the developing CNS. Brain mac-
mal CNS with regional differences having been reported in rophages, like cultured microglia, secrete a variety of cytokines
mouse brain (Lawson et al. 1990). According to these authors, and growth factors, whereas ameboid microglial progenitor
the highest microglial densities are encountered in the cells in situ do not (Hurley et al. 1999).

92 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
Another important consideration in the context of using as M1 (cytotoxic), M2 (reparative), and even a third, “deac-
cultured microglial cells to better understand the cells’ functions tivated” form (Colton 2009; Michelucci et al. 2009; Moon
concerns the issue of microglial activation. Typically, microglia et al. 2011), generating data analogous to those obtained from
in vitro are activated by exposing them to potent immunos- studying peripheral macrophages in vitro (Gordon 2003).
timulatory agents, such as lipopolysaccharide (LPS) and/or With reference to the foregoing discussion in this section, to
interferon-gamma, which means that cells already transformed generalize and assume that the M1/M2 in vitro classification
into brain macrophages by the culturing process itself become applies to the microglial population in situ would be a rather
superactivated when exposed to LPS. This in vitro stimulation indiscriminate and indeed misleading. Neurotoxic (M1) and
produces a different kind of activated microglia than what is seen neuroprotective (M2) microglial phenotypes cannot be dis-
in vivo when resting microglia are activated by neuronal injury. tinguished histopathologically in the brain, and it is a matter
Microglia in vivo progress to become brain macrophages only of common sense to state that purified cells in a petri dish do
if debris from degenerating cells needs to be phagocytosed; in not accurately reproduce the cells’ functions in the complex,
the absence of cell death microglia may become activated in vivo normal, or pathologically altered CNS microenvironment.
but do not necessarily become brain macrophages. Accordingly,
cultured microglia/brain macrophages, before LPS stimulation,
are already at an activation state that is equivalent to what is 6 MICROGLIA IN THE NORMAL
perceived as maximal microglial activation in vivo, that is, the A D U LT B R A I N
brain macrophage stage. Therefore, it is important not to equate
microglial activation in vitro with microglial activation in vivo. Microglia are ubiquitous in the CNS, where they are spaced
Techniques for inducing microglial ramification in vitro, includ- evenly in a networklike fashion throughout the brain and
ing the use of organotypic or slice cultures, co-culturing micro- spinal cord. Unlike astrocytes, microglia are not known to
glia with astrocytes or exposing them to astrocyte-conditioned form connections with each other normally, and each cell
medium, or treating them with ramifying agents, such as vitamin occupies its own individual plot of three-dimensional space,
E and thapsigargin (Eder et al. 1999; Heppner et al. 1998; Kloss which is approximately 50,000 μm3 in volume. In the normal
et al. 1997; Mertsch et al. 2001; Sievers et al. 1994; Tanaka and uninjured CNS the cells are referred to as resting microglia
Maeda 1996; Yagi et al. 1999) may allow researchers to study to set them apart from the activated or reactive microglia
ramified microglia more directly. It remains to be seen, however, that appear after brain injury. Resting microglia have a char-
whether ramified microglia in vitro show the same gene expres- acteristic cell shape marked by finely branched, ramified cell
sion patterns as ramified microglia in vivo and can therefore be processes that extend into all directions (see Fig. 8.2) reflect-
considered equivalent. ing the cells’ recognized function as “sensors of pathology”
Microglia activated in vitro with LPS or other immunos- (Kreutzberg 1996). Apparently, microglia are constantly on
timulants can produce potentially neurotoxic molecules, such the lookout for biochemical or bioelectric changes in their
as nitric oxide, glutamate, reactive oxygen and nitrogen spe- microenvironment that may signal ongoing perturbations
cies, and proinflammatory cytokines. These observations have in brain homeostasis and require them to jump into action
been extrapolated to mean that activated microglia in vivo are (Davalos et al. 2005; Nimmerjahn et al. 2005; Petersen and
harmful and could be responsible for exacerbating damage in Dailey 2004). This role as a sentry is very much analogous
the injured or diseased CNS by producing neurotoxic com- to the roles served by cells of the immune system in the rest
pounds that cause neurodegeneration secondarily. However, of the body. The concept of microglia as “the brain’s immune
this inference from cell culture studies is difficult to reconcile system” (Graeber and Streit 1990a), thus reconciles the dis-
with in vivo observations that show that microglial activa- crepancy between the absence of leukocytes in the brain and
tion is the result of neural tissue damage rather than its cause, the brain’s ability to defend itself against infection, injury,
underscoring the basic concept of inflammation, namely, that and disease. With microglia, evolution has found a way to
inflammation is the cellular response to tissue injury. The idea achieve compatibility between the destructive power of the
of microglia as instigators of bystander damage also clashes immune system and the relative vulnerability of the CNS to
with studies showing that cultured microglial cells can produce injury and disease. Functionally speaking, one might there-
neurotrophic factors and other neuroprotective substances, fore view microglia as a hybrid cell type that combines char-
and that increasingly the cells’ primary function is being acteristics of a neuroprotective glial cell with some of the
viewed as one of neuroprotective glia (Nakajima and Kohsaka attributes of macrophages and lymphocytes. In line with
2004; Polazzi and Monti 2010; Streit 2002). Microglia in vitro their role as sensors of pathology, the microglial cell surface is
behave much like other tissue macrophages in a dish, and as covered with an abundance of receptor molecules that range
such are capable of performing the full repertoire of immune from ion channels to immunological recognition molecules
functions in vitro, including phagocytosis, antigen presenta- to neurotransmitter receptors. Low levels of a unique com-
tion, and cellular cytotoxicity. However, this type of immu- position of many cytokines and their receptors in the normal
nological activity does not necessarily reflect what occurs in CNS may contribute to an immunologically quiescent CNS
the normal or injured CNS where inhibitory influences and microenvironment, although precise functional roles of most
cell–cell interactions may dampen immune responses and cytokines/chemokines and their respective receptors in the
inflammation. Investigators have worked on differentiating normal CNS are largely unknown, and may actually be differ-
in vitro functionally distinct microglial phenotypes classified ent there than in the periphery.

8. M I C R O G L I A L C E L L S • 93
For the normal CNS, one chemokine, termed fractalkine is a promising area of investigation, not only in terms of an
(CX3CL1), is of some interest because both it and its recep- enhanced understanding of normal brain plasticity, but also
tor (CX3CR1) are expressed constitutively in relatively for advancing knowledge about neurodegenerative diseases in
high amounts (Harrison et al. 1998; Nishiyori et al. 1998). which the loss of synaptic connections is a major and consis-
Fractalkine, which is present in both membrane-bound and tent correlate of diminished cognitive function.
secreted forms in neurons of the CNS, is bound by the frac-
talkine receptor, which is present on microglial cells. The
distinct separation in cellular localization of CX3CL1 and 7 MICROGLIAL SENESCENCE,
CX3CR1 suggests a role for fractalkine in mediating neuron– DYS T R O P H Y, A N D AG I N G -R E L AT E D
microglia interactions normally, as well as after injury. It is N E U R O D E G E N E R AT I O N
currently thought that high levels of fractalkine in uninjured
CNS neurons function in a constitutively inhibitory fashion The idea that microglia are subject to cell senescence stems
to help maintain microglia in their resting state. Constitutive from histopathological observations in the human brain
inhibition of microglia in the normal CNS is thought to be showing that with aging an increasing proportion of micro-
mediated by other neuronal molecules as well. The CD200 glial cells display abnormal morphological features, such as
molecule, which is present in neurons, has been implicated in shortened, gnarled, beaded, or fragmented cytoplasmic pro-
suppressing microglial activation through interactions with cesses, as well as loss of fine ramifications and formation of
the CD200 receptor thought to be present on the microglial spheroidal swellings, changes that were designated collectively
cell surface (Hoek et al. 2000). as microglial dystrophy (Streit et al. 2004). Although in the
The examples of fractalkine and CD200 as neuronal mol- normally aged human CNS dystrophic microglia appear only
ecules that may regulate microglial cell activity and activation sporadically and seem to be distributed at random through-
provide not only an illustration of how biochemical signaling out various brain regions, the situation changes dramatically
occurs between neurons and microglia, but also underscores in Alzheimer disease and in Down syndrome where severely
the necessity for neuron-microglia signaling to occur con- dystrophic microglial cells are abundant and appear preferen-
stantly within the CNS. Structural observations of perineu- tially in regions marked by neurofibrillary degeneration, that
ronal microglial satellite cells in the normal CNS support is, tau-positive neurofibrillary tangles, pretangles, neuropil
these molecular studies. Perineuronal microglial satellites are threads, and neuritic plaques (Figs. 8.6 and 8.7) (Streit et al.
microglia that are located vis-à-vis to CNS neurons in such a
way that the glial processes are partially wrapped around the
neuronal somata (see Figs. 8.4 and 8.5). There is close physical A
proximity between microglial satellites and neurons, a spatial
arrangement that is ideal for facilitating specific cell–cell inter-
actions involving the targeted exchange of minute quantities
of signaling molecules. Because not all neurons have perineu-
ronal microglial satellites, it is likely that those neurons that
do may have attracted the cells for a reason. The presence of
perineuronal microglial satellites could signify the need for
increased trophic support from microglia because of height-
ened metabolic demands or increased physiological activity.
In addition to providing trophic support, perineuronal micro-
glial satellites could also be involved in the remodeling of
synaptic contacts on these neurons. Microglia have long been
known to engage in synaptic remodeling, a phenomenon first B
reported more than 40 years ago (Blinzinger and Kreutzberg
1968). This potentially important role of microglia in synap-
tic plasticity received very little attention until quite recently
when a number of laboratories started to reexamine and delve
deeper into this phenomenon. It now appears that microglial
regulation of neuronal connectivity is important during
development, as well as in the normal and injured adult brain
(Paolicelli et al. 2011; Tremblay et al. 2010; Wake et al. 2009).
In fact, microglia are well equipped to participate in synap-
tic remodeling because they generate a number of enzymatic
activities, including matrix metalloproteinase, elastase, and
plasminogen activator. They also produce extracellular matrix Figure 8.6 Comparison of Normal (Ramified) and Degenerating
(Dystrophic) Microglia Using Iba1 Immunostaining in Human Cerebral
molecules, such as laminin, thrombospondin, and keratan sul- Cortex. A. A 22-year-old male subject reveals cells with normal morphology.
fate, as well as neurotrophic factors. Continued research into B. A 48-year-old female subject with Down syndrome shows cells displaying
the possible role of microglia in regulating synaptic plasticity dystrophic cytoplasmic fragmentation (cytorrhexis). Scale bar: 50 μm.

94 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
A that immunological responses generally wane in the elderly,
rendering geriatric patients more susceptible to all kinds of
infections, cancers, and other ailments because of compro-
mised immune functions. The hypothesis that is currently
being pursued claims that microglial senescence is a key fac-
tor contributing to aging-related neurodegenerative changes.
Thus, neurodegeneration occurs because progressively neu-
roprotective microglial cells are being lost, and Alzheimer
disease might be the consequence of a rapidly deteriorating
immune system in the brain.

8 S U M M A RY A N D P E R S P E C T I VE S
B
Tremendous advances in our understanding of microglial
biology have been made in the last two decades, which result
in large part from the availability of numerous markers for
microglia that now make it routine procedure for laborato-
ries to visualize the cells in vivo and in situ. Because of the
abundance of cell surface receptors of every imaginable type
on the microglial cell membrane, continued exploration of
molecular phenotypic changes will help to better define the
dynamic functional states of these cells and thus provide
novel insights into their involvement in diverse processes,
including (but certainly not limited to) neuroprotection,
neuroinflammation, repair, synaptic plasticity, and other tis-
sue remodeling. The recognition of dystrophic microglia in
the aging brain has provided a new and different perspec-
tive on the pathogenesis of aging-related neurodegenerative
diseases that is reshaping our views on neuroinflammation
and neuroprotection and may thus influence how future
Figure 8.7 Colocalization of Microglia with Hallmark approaches toward the treatment and/or prevention of these
Neuropathological Features of Alzheimer Disease. A. Neurons expressing
hyperphosphorylated tau as they are undergoing neurofibrillary conditions are constructed.
degeneration (black reaction product) are surrounded by degenerating
(fragmented) microglial cells (brown reaction product). Immunostaining
with antibodies AT8 and iba1 in a 92-year-old male subject with
Alzheimer disease. B. Deposits of amyloid-β protein (red fluorescence) REFERENCES
are surrounded and infiltrated by ramified microglia (green fluorescence).
Immunostaining with antibodies 10D5 and iba1 in a 49-year-old male Acarin L, Vela JM, Gonzalez B, Castellano B. 1994. Demonstration
subject with Down syndrome. Scale bars: 50 μm. of poly-N-acetyl lactosamine residues in ameboid and ramified
microglial cells in rat brain by tomato lectin binding. J Histochem
Cytochem 42(8):1033–1041.
2009; Xue and Streit 2011). This colocalization of neurode- Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FM. 2011.
generative and gliodegenerative changes has cast the patho- Infi ltrating monocytes trigger EAE progression, but do not contrib-
genesis of neurodegeneration in a new and different light, ute to the resident microglia pool. Nat Neurosci 14(9):1142–1149.
suggesting that a decline rather than an upsurge of immuno- Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, et al.
2000. Inflammation and Alzheimer’s disease. Neurobiol Aging
logical activity within the CNS may be responsible for the 21(3):383–421.
development of aging-related neurodegenerative changes Akiyama H, McGeer PL. 1990. Brain microglia constitutively express
(Croisier and Graeber 2006; Fellner et al. 2011; Streit 2010; beta-2 integrins. J Neuroimmunol 30(1):81–93.
Streit and Xue 2009). Clearly, this idea stands in contrast to Alliot F, Godin I, Pessac B. 1999. Microglia derive from progenitors,
the longstanding and popular notion that an overly active, originating from the yolk sac, and which proliferate in the brain.
Brain Res Dev Brain Res 117(2):145–152.
chronic neuroinflammatory response accounts for neuro- Banati RB, Gehrmann J, Schubert P, Kreutzberg GW. 1993. Cytotoxicity
degeneration in Alzheimer’s and other neurodegenerative of microglia. Glia 7(1):111–118.
diseases (Eikelenboom et al. 2002; Lee et al. 2009; McGeer Blinzinger K, Kreutzberg G. 1968. Displacement of synaptic terminals
and McGeer 2001), but aligns with the fact that clinical trials from regenerating motoneurons by microglial celss. Zeitschrift fur
designed to suppress chronic neuroinflammation with anti- Zellforschung(85):145–157.
Castellano B, Gonzalez B, Finsen BR, Zimmer J. 1990. Histochemical
inflammatory drugs have failed to show clear benefits of such demonstration of purine nucleoside phosphorylase (PNPase)
treatments (Martin et al. 2008). This failure of attempted in microglial and astroglial cells of adult rat brain. J Histochem
immunological suppression is perhaps not surprising given Cytochem 38(11):1535–1539.

8. M I C R O G L I A L C E L L S • 95
Chan WY, Kohsaka S, Rezaie P. 2007. The origin and cell lineage of Hayes GM, Woodroofe MN, Cuzner ML. 1987. Microglia are the major
microglia: new concepts. Brain Res Rev 53(2):344–354. cell type expressing MHC class II in human white matter. J Neurol
Chugani DC, Kedersha NL, Rome LH. 1991. Vault immunofluores- Sci 80(1):25–37.
cence in the brain: new insights regarding the origin of microglia. Heppner FL, Roth K, Nitsch R, Hailer NP. 1998. Vitamin E induces
J Neurosci 11(1):256–268. ramification and downregulation of adhesion molecules in cultured
Colton CA. 2009. Heterogeneity of microglial activation in the microglial cells. Glia 22(2):180–188.
innate immune response in the brain. J Neuroimmune Pharmacol Hickey WF, Kimura H. 1988. Perivascular microglial cells of the
4(4):399–418. CNS are bone marrow-derived and present antigen in vivo. Science
Craggs RI, Webster HD. 1985. Ia antigens in the normal rat nervous sys- 239(4837):290–292.
tem and in lesions of experimental allergic encephalomyelitis. Acta Hoek RM, Ruuls SR, Murphy CA, Wright GJ, Goddard R, Zurawski
Neuropathol 68(4):263–272. SM, et al. 2000. Down-regulation of the macrophage lineage through
Croisier E, Graeber MB. 2006. Glial degeneration and reactive gliosis in interaction with OX2 (CD200). Science 290(5497):1768–1771.
alpha-synucleinopathies: the emerging concept of primary gliodegen- Hume DA, Perry VH, Gordon S. 1983. Immunohistochemical local-
eration. Acta Neuropathol (Berl) 112(5):517–530. ization of a macrophage-specific antigen in developing mouse
Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, et al. 2005. retina: phagocytosis of dying neurons and differentiation of microg-
ATP mediates rapid microglial response to local brain injury in vivo. lial cells to form a regular array in the plexiform layers. J Cell Biol
Nat Neurosci 8(6):752–758. 97(1):253–257.
Dickson DW, Mattiace LA. 1989. Astrocytes and microglia in human Hurley SD, Walter SA, Semple-Rowland SL, Streit WJ. 1999. Cytokine
brain share an epitope recognized by a B-lymphocyte-specific mono- transcripts expressed by microglia in vitro are not expressed by ame-
clonal antibody (LN-1). Am J Pathol 135(1):135–147. boid microglia of the developing rat central nervous system. Glia
Dijkstra CD, Dopp EA, Joling P, Kraal G. 1985. The heterogeneity of 25(3):304–309.
mononuclear phagocytes in lymphoid organs: distinct macrophage Ito D, Imai Y, Ohsawa K, Nakajima K, Fukuuchi Y, Kohsaka S. 1998.
subpopulations in the rat recognized by monoclonal antibodies ED1, Microglia-specific localisation of a novel calcium binding protein,
ED2 and ED3. Immunology 54(3):589–599. Iba1. Brain Res Mol Brain Res 57(1):1–9.
Eder C, Schilling T, Heinemann U, Haas D, Hailer N, Nitsch R. 1999. Kaneko Y, Kitamoto T, Tateishi J, Yamaguchi K. 1989. Ferritin immu-
Morphological, immunophenotypical and electrophysiological prop- nohistochemistry as a marker for microglia. Acta Neuropathol
erties of resting microglia in vitro. Eur J Neurosci 11(12):4251–4261. 79(2):129–136.
Eikelenboom P, Bate C, Van Gool WA, Hoozemans JJ, Rozemuller JM, Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. 2011. Physiology
Veerhuis R, et al. 2002. Neuroinflammation in Alzheimer’s disease of microglia. Physiol Rev 91:461–553.
and prion disease. Glia 40(2):232–239. Kloss CU, Kreutzberg GW, Raivich G. 1997. Proliferation of ramified
Fellner L, Jellinger KA, Wenning GK, Stefanova N. 2011. Glial dysfunc- microglia on an astrocyte monolayer: characterization of stimulatory
tion in the pathogenesis of alpha-synucleinopathies: emerging con- and inhibitory cytokines. J Neurosci Res 49(2):248–254.
cepts. Acta Neuropathol 121(6):675–693. Korr H, Schilling WD, Schultze B, Maurer W. 1983. Autoradiographic
Finch C, Morgan T, Rozovsky I, Xie Z, Weindruch R, Prolla T. 2002. studies of glial proliferation in different areas of the brain of the
Microglia and aging in the brain. New York: Springer Verlag. 14-day-old rat. Cell Tissue Kinet 16(4):393–413.
Ford AL, Goodsall AL, Hickey WF, Sedgwick JD. 1995. Normal adult Kreutzberg GW. 1996. Microglia: a sensor for pathological events in the
ramified microglia separated from other central nervous system mac- CNS. Trends Neurosci 19(8):312–318.
rophages by flow cytometric sorting. Phenotypic differences defi ned Lassmann H, Zimprich F, Vass K, Hickey WF. 1991. Microglial cells
and direct ex vivo antigen presentation to myelin basic protein-reactive are a component of the perivascular glia limitans. J Neurosci Res
CD4+ T cells compared. J Immunol 154(9):4309–4321. 28(2):236–243.
Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. 2010. Lawson LJ, Perry VH, Dri P, Gordon S. 1990. Heterogeneity in the dis-
Fate mapping analysis reveals that adult microglia derive from primi- tribution and morphology of microglia in the normal adult mouse
tive macrophages. Science 330(6005):841–845. brain. Neuroscience 39(1):151–170.
Giulian D, Baker TJ. 1986. Characterization of ameboid microglia isolated Lee JK, Tran T, Tansey MG. 2009. Neuroinflammation in Parkinson’s
from developing mammalian brain. J Neurosci 6(8):2163–2178. disease. J Neuroimmune Pharmacol 4(4):419–429.
Gordon S. 2003. Alternative activation of macrophages. Nat Rev Leong SK, Ling EA. 1992. Amoeboid and ramified microglia: their
Immunol 3(1):23–35. interrelationship and response to brain injury. Glia 6(1):39–47.
Graeber MB. 2010. Changing face of microglia. Science 330(6005): Ling EA, Wong WC. 1993. The origin and nature of ramified and
783–788. amoeboid microglia: a historical review and current concepts. Glia
Graeber MB, Streit WJ. 1990a. Microglia: immune network in the CNS. 7(1):9–18.
Brain Pathol 1(1):2–5. Mannoji H, Yeger H, Becker LE. 1986. A specific histochemical marker
Graeber MB, Streit WJ. 1990b. Perivascular microglia defined. Trends (lectin Ricinus communis agglutinin-1) for normal human microglia,
Neurosci 13(9):366. and application to routine histopathology. Acta Neuropathol (Berl)
Graeber MB, Streit WJ, Kreutzberg GW. 1988. Axotomy of the rat facial 71(3–4):341–343.
nerve leads to increased CR3 complement receptor expression by Martin BK, Szekely C, Brandt J, Piantadosi S, Breitner JC, Craft S,
activated microglial cells. J Neurosci Res 21(1):18–24. et al. 2008. Cognitive function over time in the Alzheimer’s Disease
Graeber MB, Streit WJ, Kreutzberg GW. 1989a. Formation of Anti-inflammatory Prevention Trial (ADAPT): results of a ran-
microglia-derived brain macrophages is blocked by adriamycin. Acta domized, controlled trial of naproxen and celecoxib. Arch Neurol
Neuropathol (Berl) 78(4):348–358. 65(7):896–905.
Graeber MB, Streit WJ, Kreutzberg GW. 1989b. Identity of ED2-positive Mato M, Ookawara S, Saito-Taki T. 1986. Serological determinants of
perivascular cells in rat brain. J Neurosci Res 22(1):103–106. fluorescent granular perithelial cells along small cerebral blood vessels
Griffith R, Soria J, Wood JG. 2000. Regulation of microglial tyrosine in rodent. Acta Neuropathol 72(2):117–123.
phosphorylation in response to neuronal injury. Exp Neurol McGeer PL, McGeer EG. 2001. Inflammation, autotoxicity and
161(1):297–305. Alzheimer disease. Neurobiol Aging 22(6):799–809.
Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, McNamara RK, McKanna JA. 1993. Lipocortin 1 immunoreactivity identifies microglia
et al. 1998. Role for neuronally derived fractalkine in mediating in adult rat brain. J Neurosci Res 36(4):491–500.
interactions between neurons and CX3CR1-expressing microglia. Mertsch K, Hanisch UK, Kettenmann H, Schnitzer J. 2001.
Proc Natl Acad Sci U S A 95(18):10896–10901. Characterization of microglial cells and their response to stimulation in
an organotypic retinal culture system. J Comp Neurol 431(2):217–227.

96 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
Michelucci A, Heurtaux T, Grandbarbe L, Morga E, Heuschling P. Sheffield LG, Berman NE. 1998. Microglial expression of MHC class II
2009. Characterization of the microglial phenotype under specific increases in normal aging of nonhuman primates. Neurobiol Aging
pro-inflammatory and anti-inflammatory conditions: Effects of oligo- 19(1):47–55.
meric and fibrillar amyloid-beta. J Neuroimmunol 210(1–2):3–12. Sievers J, Parwaresch R, Wottge HU. 1994. Blood monocytes and spleen
Miles JM, Chou SM. 1988. A new immunoperoxidase marker for microg- macrophages differentiate into microglia-like cells on monolayers of
lia in paraffi n section. J Neuropath Exp Neurol 47:579–587. astrocytes: morphology. Glia 12(4):245–258.
Milligan CE, Cunningham TJ, Levitt P. 1991. Differential immu- Sorokin SP, Hoyt RF Jr, Blunt DG, McNelly NA. 1992. Macrophage
nochemical markers reveal the normal distribution of brain mac- development: II. Early ontogeny of macrophage populations in brain,
rophages and microglia in the developing rat brain. J Comp Neurol liver, and lungs of rat embryos as revealed by a lectin marker. Anat
314(1):125–135. Rec 232(4):527–550.
Monier A, Adle-Biassette H, Delezoide AL, Evrard P, Gressens P, Verney Streit WJ. 2001. Microglia and macrophages in the developing CNS.
C. 2007. Entry and distribution of microglial cells in human embryonic Neurotoxicology 22(5):619–624.
and fetal cerebral cortex. J Neuropathol Exp Neurol 66(5):372–382. Streit WJ. 2002. Microglia as neuroprotective, immunocompetent cells
Moon ML, McNeil LK, Freund GG. 2011. Macrophages make me of the CNS. Glia 40(2):133–139.
sick: How macrophage activation states influence sickness behavior. Streit WJ. 2006. Microglial senescence: does the brain’s immune system
Psychoneuroendocrinology 36(10):1431–1440. have an expiration date? Trends Neurosci 29(9):506–510.
Morioka T, Baba T, Black KL, Streit WJ. 1992. Response of microglial Streit WJ. 2010. Microglial activation and neuroinflammation in
cells to experimental rat glioma. Glia 6(1):75–79. Alzheimer’s disease: a critical examination of recent history. Front
Murabe Y, Sano Y. 1981. Th iaminepyrophosphatase activity in the Aging Neurosci 2:22.
plasma membrane of microglia. Histochemistry 71(1):45–52. Streit WJ, Braak H, Xue QS, Bechmann I. 2009. Dystrophic (senescent)
Nakajima K, Kohsaka S. 2004. Microglia: neuroprotective and neu- rather than activated microglial cells are associated with tau pathol-
rotrophic cells in the central nervous system. Curr Drug Targets ogy and likely precede neurodegeneration in Alzheimer’s disease.
Cardiovasc Haematol Disord 4(1):65–84. Acta Neuropathol 118(4):475–485.
Nakajima K, Shimojo M, Hamanoue M, Ishiura S, Sugita H, Kohsaka S. Streit WJ, Graeber MB. 1993. Heterogeneity of microglial and perivas-
1992. Identification of elastase as a secretory protease from cultured cular cell populations: insights gained from the facial nucleus para-
rat microglia. J Neurochem 58(4):1401–1408. digm. Glia 7(1):68–74.
Nimmerjahn A, Kirchhoff F, Helmchen F. 2005. Resting microglial cells Streit WJ, Graeber MB, Kreutzberg GW. 1989. Expression of Ia antigen
are highly dynamic surveillants of brain parenchyma in vivo. Science on perivascular and microglial cells after sublethal and lethal motor
308(5726):1314–1318. neuron injury. Exp Neurol 105(2):115–126.
Nishiyori A, Minami M, Ohtani Y, Takami S, Yamamoto J, Kawaguchi Streit WJ, Kreutzberg GW. 1987. Lectin binding by resting and reactive
N, et al. 1998. Localization of fractalkine and CX3CR1 mRNAs microglia. J Neurocytol 16(2):249–260.
in rat brain: does fractalkine play a role in signaling from neuron to Streit WJ, Kreutzberg GW. 1988. Response of endogenous glial cells to
microglia? FEBS Lett 429(2):167–172. motor neuron degeneration induced by toxic ricin. J Comp Neurol
Oehmichen M, Wietholter H, Greaves MF. 1979. Immunological anal- 268(2):248–263.
ysis of human microglia: lack of monocytic and lymphoid membrane Streit WJ, Sammons NW, Kuhns AJ, Sparks DL. 2004. Dystrophic
differentiation antigens. J Neuropathol Exp Neurol 38(2):99–103. microglia in the aging human brain. Glia 45(2):208–212.
Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, Streit WJ, Schulte BA, Balentine DJ, Spicer SS. 1985. Histochemical
et al. 2011. Synaptic pruning by microglia is necessary for normal localization of galactose-containing glycoconjugates in sensory neu-
brain development. Science 333(6048):1456–1458. rons and their processes in the central and peripheral nervous system
Pennell NA, Streit WJ. 1998. Tracing of fluoro-gold prelabeled microglia of the rat. J Histochem Cytochem 33(10):1042–1052.
injected into the adult rat brain. Glia 23(1):84–88. Streit WJ, Sparks DL. 1997. Activation of microglia in the brains of
Perry VH, Gordon S. 1987. Modulation of CD4 antigen on macrophages humans with heart disease and hypercholesterolemic rabbits. J Mol
and microglia in rat brain. J Exp Med 166(4):1138–1143. Med 75(2):130–138.
Perry VH, Hume DA, Gordon S. 1985. Immunohistochemical localiza- Streit WJ, Xue QS. 2009. Life and death of microglia. J Neuroimmune
tion of macrophages and microglia in the adult and developing mouse Pharmacol 4(4):371–379.
brain. Neuroscience 15(2):313–326. Suzuki H, Franz H, Yamamoto T, Iwasaki Y, Konno H. 1988.
Perry VH, Matyszak MK, Fearn S. 1993. Altered antigen expression of Identification of the normal microglial population in human and
microglia in the aged rodent CNS. Glia 7(1):60–67. rodent nervous tissue using lectin-histochemistry. Neuropathol Appl
Petersen MA, Dailey ME. 2004. Diverse microglial motility behaviors dur- Neurobiol 14(3):221–227.
ing clearance of dead cells in hippocampal slices. Glia 46(2):195–206. Takahashi K, Yamamura F, Naito M. 1989. Differentiation, matu-
Polazzi E, Monti B. 2010. Microglia and neuroprotection: from in vitro ration, and proliferation of macrophages in the mouse yolk sac: a
studies to therapeutic applications. Prog Neurobiol 92(3):293–315. light-microscopic, enzyme-cytochemical, immunohistochemical,
Prinz M, Priller J, Sisodia SS, Ransohoff RM. 2011.Heterogeneity of and ultrastructural study. J Leukoc Biol 45(2):87–96.
CNS myeloid cells and their roles in neurodegeneration. Nat Neurosci Tanaka J, Maeda N. 1996. Microglial ramification requires nondiff usible
14(10):1227–1235. factors derived from astrocytes. Exp Neurol 137(2):367–375.
Rinaman L, Milligan CE, Levitt P. 1991. Persistence of fluoro-gold fol- Thanos S. 1991. Specific transcellular carbocyanine-labelling of rat reti-
lowing degeneration of labeled motoneurons is due to phagocytosis nal microglia during injury-induced neuronal degeneration. Neurosci
by microglia and macrophages. Neuroscience 44(3):765–776. Lett 127(1):108–112.
Sawada M, Suzumura A, Yamamoto H, Marunouchi T. 1990. Activation Tremblay ME, Lowery RL, Majewska AK. 2010. Microglial interac-
and proliferation of the isolated microglia by colony stimulating tions with synapses are modulated by visual experience. PLoS Biol
factor-1 and possible involvement of protein kinase C. Brain Res 8(11):e1000527.
509(1):119–124. Wake H, Moorhouse AJ, Jinno S, Kohsaka S, Nabekura J. 2009.
Schelper RL, Adrian EK, Jr. 1980. Non-specific esterase activity in Resting microglia directly monitor the functional state of synapses
reactive cells in injured nervous tissue labeled with 3H-thymidine in vivo and determine the fate of ischemic terminals. J Neurosci
or 125iododeoxyuridine injected before injury. J Comp Neurol 29(13):3974–3980.
194(4):829–844. Xue QS, Streit WJ. 2011.Microglial pathology in Down syndrome. Acta
Schnitzer J. 1989. Enzyme-histochemical demonstration of micro- Neuropathol 122(4):455–466.
glial cells in the adult and postnatal rabbit retina. J Comp Neurol Yagi R, Tanaka S, Koike T. 1999. Thapsigargin induces microglial trans-
282(2):249–263. formation from amoeboid to ramified type in vitro. Glia 28(1):49–52.

8. M I C R O G L I A L C E L L S • 97
9.
PERICY TES OF THE CENTRAL NERVOUS SYSTEM
Martin Krueger and Ingo Bechmann

A B B R E VI AT I O N S 2 TO P O G R A P H Y O F P E R I C Y T E S W I T H I N
T H E N E U R O VA S C U L A R U N I T
Ang-1 angiopoietin 1
BBB blood-brain barrier Although pericytes have been repeatedly under investigation
CNS central nervous system during the last decades, still relatively little facts are known about
GFAP glial fibrillary acid protein their roles in health and disease. Furthermore, much of the data
GFP green fluorescent protein concerning pericytes are difficult to interpret and compare with
MMP matrix metalloproteinase other studies. This problem mainly derives from the fact that
NVU neurovascular unit pericytes are often mixed up with other cell types of neighboring
pAPN pericytic aminopeptidase N compartments, such as vascular smooth muscle cells, perivascular
PDGF-β platelet-derived growth factor β cells, or juxtavascular microglia (Table 9.1). Indeed, pericytes are
PDGFR-β platelet-derived growth factor receptor β difficult to address because they are only defined by their unique
SMA alpha smooth muscle actin position in the outermost vascular basement membrane, whereas
TGF-β transforming growth factor β an unambiguous pericyte marker is lacking. Therefore, three dif-
VEGF vascular endothelial growth factor ferent compartments within the NVU must be distinguished
VRS Virchow-Robin-Spaces (Bechmann et al. 2007). The first is constituted by the vascular
wall consisting of endothelial cells, pericytes, and if present, vas-
cular smooth muscle cells. The second compartment represents
1 INTRODUCTION the perivascular space (Virchow-Robin-Space [VRS]), which is
localized between the outermost vascular basement membrane
More than a century ago the French scientist C. M. Rouget and the basement membrane of the glia limitans. The third is the
was the first to describe a population of cells residing in the juxtavascular parenchyma, which is delineated by the glia limi-
vascular wall of capillary vessels. After a long period of neglect tans and its basement membrane (Fig. 9.1).
and misconception, pericytes have turned into focus of scien- The best way to define pericytes within the NVU is given
tific interest again as they prove to be key players in matura- by their name (peri-, meaning around; cyto-, meaning cell),
tion and regulation of the neurovascular unit (NVU). Being which describes their location around endothelial cells in
part of the vascular wall of central nervous system (CNS) microvessels (Rhodin 1968; Zimmermann 1923). With
capillaries, they are perfectly positioned to conduct criti- their long processes, pericytes follow the vessels in longitu-
cal aspects of proper functioning of the blood-brain barrier dinal direction, whereas smaller, radial arms can also encircle
(BBB), and therefore have become of interest in a variety of the capillary wall. Thus, one might have the idea that blood
cerebral disorders. (see chapter 33) vessels are engirdled or cradled by pericytes. In vitro stud-
Pericytes are positioned on the abluminal surface of the ies demonstrated that both endothelial cell and pericytes
endothelial layer, being ensheathed in their “own” basement are capable of contributing to the production of the base-
membrane. Therefore, they are located in an intermediate posi- ment membrane in which they are situated (Cohen 1980;
tion between two compartments. On the one hand pericytes Mandarino 1993), and often their association is so intimate
are in direct contact to endothelial cells as part of the vascular that the intercellular distance is narrowed down to less than
wall, and on the other hand they are juxtaposed to the astro- 20 nm (Sims 1986). In fact, the vascular and glial basement
cytic endfeet that give rise to the glia limitans and its basement membranes of the NVU are structurally and functionally dis-
membrane. Proper functioning and the ability to respond to tinct (Bechmann et al. 2007; Sixt et al. 2001) and provide
critical systemic and neural demands involve a functional clear-cut morphological borders that can be used for pericyte
network not only consisting of endothelial cells, but also detection. Often pericytic processes are found to interdigi-
pericytes, vascular smooth muscle cells, astrocytes, microglia tate with endothelial cells, thereby forming peg-socket con-
and neurons, thus making up the NVU. Therefore, this chap- tacts that consist of N-cadherin, adherens junction protein,
ter addresses pericytes as part of the NVU, highlights recent and connexin-43 hemichannels (Gerhardt et al. 2000; Li
advances in pericyte research, and illustrates current concepts et al. 2011; Winkler et al. 2011). The latter readily form gap
of their function. junctions with endothelial cells, thus allowing exchange of

98
Table 9.1 DISTINCTION BETWEEN PERICYTES AND ADJACENT CELL TYPES
CELL TYPE LOCATION MARKERS ORIGIN

Pericytes Vascular wall, ensheathed in the PDGFR-β, pAPN, (SMA) Neuroectoderm and mesoderm, turnover by
outermost vascular basement membrane blood-derived cells suggested

Smooth muscle Vascular wall, surrounded by own SMA, desmin Mesoderm


cells basement membranes

Perivascular Inside perivascular spaces CX3CR1hi, Iba-1, F4/80, Blood, high turnover by blood derived myeloid
macrophages CD11b, CD45hi, IL-B4, CD163 cells

Juxtavascular Parenchyma proper, behind and closely CX3CR1hi, Iba-1, F4/80, Yolk sac macrophages, self renewal, no exchange
microglia associated to the glia limitans CD11b, CD45lo, IL-B4 with blood-derived cells during life time

Adopted from Graeber and Streit 1990, Prinz et al. 2011.

A Pericytes and dura mater


the compartments arachnoidea
of the NVU subarachnoid
space
pia mater

glia limitans subarachnoid


vessel

neuropil

non capillary vessel capillary


perivascular space

perivascular space
endothelial cells
smooth muscle cells of the tunica media
pericytes
perivascular macrophages
astrocytes
juxtavascular microglia
dura mater
arachnoidea and pia mater
B

neuropil neuropil neuropil


capillary non capillary vessel subarachnoid vessel
vascular wall: endothelial vascular wall:endothelial cells vascular wall: endothelial cells
cells + pericytes + pericytes+smooth muscle + smooth muscle cells+
cells (if present) pericytes
basement membranes

Figure 9.1 Pericytes and the compartments of the NVU. A. This figure represents a topographic overview of the NVU along the different segments
of CNS vessels, where three distinct compartments can be identified. These compartments are the vascular wall, the perivascular (Virchow-Robin)
space, and the adjacent parenchyma proper, all of which are delineated by distinct basement membranes (Bechmann et al. 2001a; Sixt et al. 2001). B.
In the capillary segment, the vascular wall consists of endothelial cells and pericytes only. Here, the perivascular space is mostly occluded by the fused
gliovascular basement membrane, which allows direct contact of astrocytic endfeet to pericytes and endothelial cells. In noncapillary vessels such
direct contact is hampered by presence of perivascular spaces and smooth muscle cells of the tunica media. Subarachnoid vessels commonly exhibit a
continuous layer of smooth muscle cells, at least in the arterial branch of the vascular tree. In contrast, venous vessels may also show a rather irregular
covering by smooth muscle cells, which are then difficult to distinguish from pericytes. In these areas, pericytes may also be regarded as a transitional
form of smooth muscle cells. Adopted from Krüger and Bechmann 2010.

metabolites, second messengers, and ions between both cell 3 M ET H O D S F O R P E R I C Y T E D ET E C T I O N


types (Bobbie et al. 2010). Pericytes and endothelial cells also
contact the glial basement membrane within the microvascu- Pericytes express several antigens (PDGFR-ß, pAPN/CD13,
lature, whereas in noncapillary vessels such direct contact is α-smooth muscle actin, NG-2, RGS5) allowing their identi-
inhibited by Virchow-Robin-Spaces (VRS). fication (Bondjers et al. 2003; Cho et al. 2003; Krause et al.

P E R I C Y T E S O F T H E C E N T R A L N E RVO U S SYS T E M • 99
1992; Lindahl et al. 1997; Nehls and Drenckhahn 1991; depending on the species, tissue, developmental state, and posi-
Ozerdem et al. 2001; Ruiter et al. 1993). However, none of tion within the vascular tree. In chicken embryos, pericytes of
these markers is specific for this population. First, they may angiogenic vessels are regularly positive for α smooth muscle
also be present in adjacent cell types. Second, their expression actin (Gerhardt et al. 2000), whereas mice and rats do not show
can be upregulated in diverse cell types under experimental any immunoreactivity for this marker (Hellström et al. 1999).
conditions. Precise and unambiguous identification is still Electron microscopy, at least in mature vessels, still rep-
only possible by means of morphology at the ultrastructural resents the most specific tool to identify pericytes (Fig. 9.4).
level and in semi-thin sections. These techniques are indeed Unfortunately, this technique cannot be applied to every study
the only tools to demark their unique position in the vascular design. In experimental conditions of altered basement mem-
wall. Although technically feasible, these methods are rarely branes or during pericyte recruitment toward and away from
combined with immunocytochemistry. Unfortunately, light- the vascular wall, detached pericytes are even ultrastructurally
and fluorescence microscopy do not provide the necessary
resolution to accurately determine the location of putative
pericytes within the compartments of the neurovascular unit,
which is by definition a prerequisite for pericyte identification
(Figs. 9.2 and 9.3).
An antigen that already proved its specificity at the ultra-
structural level is the pericytic aminopeptidase N (pAPN/
CD13), which belongs to the family of matrix metalloprotei-
nases (Alliott et al. 1999; Kunz et al. 1995). These enzymes are
shown to be involved in zinc dependent cleavage of extracellu-
lar matrix- and non-matrix components such as growth factors
or neuropeptides (Sato 2004). The specificity of other markers
often has not been tested at the ultrastructural level. Moreover,
the expression pattern of pericytes seems to vary strongly
A

Figure 9.3 Pericytes and their localization in the vascular wall. A. This
picture represents a confocal three-dimensional reconstruction of a
typical capillary pericyte stained for pAPN. The cell body is widely
elongated showing three major processes in longitudinal orientation.
B. These images show confocal single scans of double fluorescence stain-
ing for pAPN (pericytes; red) and laminin (basement membrane; green).
The merged picture demonstrates the close association of the basement
membrane sheaths in relation to the pericyte body and its processes.
Furthermore, it illustrates the difficulty to differentiate between distinct
vascular compartments, even using confocal microscopy. C. This image is
obtained by standard fluorescence microscopy and depicts a staining for
Figure 9.2 Detection of pericytes. These photos show representative pericytes (pAPN; red) in red and bone marrow–derived perivascular cells
stainings for the pericyte marker pAPN, which proved its specificity at in green (a mouse chimera grafted with gfp-expressing bone marrow;
the ultrastructural level. In the brain, pericytes are regularly found in the green). Again, it is often impossible to distinguish between the different
capillary segment, but also at larger vessels. Their morphology can vary compartments of the NVU. In fact, standard fluorescence microscopy
depending on their position in the vascular tree. The respective shape does not offer the necessary resolution to differentiate between perivascu-
ranges from elongated cells, which are longitudinally oriented along the lar cells and pericytes (arrows). The arrowhead points to a bone marrow–
vascular axis to a form also surrounding the vascular circumference in ves- derived cell, which is clearly “behind” pericytes and thus could be located
sels of larger diameters (arrowheads). Scale bars: (A) 50 μm, (B) 25 μm. within the perivascular space or the juxtavascular parenchyma.

100 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
difficult to differentiate from cells within VRS, such as cells Other studies describe the bone marrow to constitute
of the leptomeninges or other perivascular cells. a reservoir for pericyte precursors (Kokovay et al. 2006;
Lamagna and Bergers 2006). Using green fluorescent protein
(GFP)-positive bone marrow mouse chimeras, Kokovay et al.
4 O R I G I N O F C E N T R A L N E RVO U S
investigated whether pericytes are recruited from the periph-
SYS T E M P E R I C Y T E S
ery to stabilize blood vessels in the model of experimental
stroke. Indeed, blood-derived GFP-positive cells were found
Initial vascularization of the CNS occurs via invading, sprout-
in areas of robust angiogenic response after stroke. However,
ing angiogenesis from a vascular plexus being juxtaposed to
these cells were insufficiently determined to be pericytes by
the neural tube. The latter is penetrated by ingressing endothe-
their expression of desmin, vimentin, and angiogenic factors
lial cells that migrate toward the ventricles. The endothelial
(Kokovay et al. 2006). Ultrastructural identification is lack-
cells are pursued by pericytes sensing cues which recruit these
ing in these studies. Moreover, investigating recruitment of
cells toward the nascent vessels (Bautch and James 2009). In
blood-derived cells in models of bone marrow transplanta-
this early angiogenic phase pericytes of the CNS are shown
tion strictly depends on lethal irradiation of host animals.
to have two distinct origins. On the one hand, several stud-
Today, there are several lines of evidence suggesting that irra-
ies using avian chimeras repeatedly demonstrated that trans-
diation itself may condition the brain to attract myeloid lin-
planted neuroectoderm gives rise to pericytes of the forebrain,
eages. On the one hand this may result from the upregulation
and on the other hand transplanted mesoderm gives rise to
of chemoattractant factors, and on the other hand result from
pericytes of the mid brain, brainstem, spinal cord, and periph-
the loss or downregulation of inhibitory signals (Mildner
eral organs (Etchevers et al. 2001; Korn et al. 2002; Kurz
et al. 2007). Therefore, the physiological turnover of CNS
2009). These studies became possible after the discovery of an
pericytes by blood-derived cells remains to be discussed.
antibody selectively labelling quail angioblasts and endothe-
In the past, pericytes were also repeatedly reported to derive
lial cells without cross-reactions to other species. Therefore,
from the monocyte/macrophage lineage. Several studies dem-
it was possible to trace graft derived quail tissue in develop-
onstrated the expression of myeloid markers as ED1 or CD11b
ing quail-chick chimeras. In areas of vessel formation during
on pericytes (Balabanov et al. 1996; Graeber et al. 1989). These
angiogenesis, endothelial cells are suggested to drive migration
findings were supported by in vitro studies demonstrating the
and attachment of pericytes along the nascent capillary tubes
ability of major histocompatibility class II antigen (MHC-II)
(Abramsson et al. 2007; Hellström et al. 1999; Ozerdem and
expression on stimulation with interferon γ (Dore-Duff y
Stallcup 2003, 2004; Stratman et al. 2010; Stenzel et al. 2009).
and Balabanov 1998). Further studies also addressed their
However, whether these events also take place during vascular
phagocytic potential using antibody-coated zymosan and
remodeling in the adult CNS is still under investigation.
fluorochrome-conjugated polystyrene beads (Balabanov et al.
1996). However, the methods used for identification were rather
imprecise in these studies; thus, the findings may likely relate to
perivascular macrophages. Indeed, this population was shown to
be capable of phagocytosis, act as antigen-presenting cells, and
is supplemented by hematogenous precursors (Bechmann et al.
2001a, b; Greter et al. 2005; Hickey and Kimura 1988; Priller
et al. 2001). Furthermore, the question of whether pure peri-
cyte cultures can be established is not yet answered. Therefore,
potential pericyte precursors remain a subject of further inves-
tigation, and their ability to be recruited to the CNS so as to
renew the resident population during injuries or under normal
conditions remains to be determined.

5 PERICY TE SIGNALING

Central nervous system pericytes are involved in a variety of dif-


ferent signaling cascades. These include platelet-derived growth
factor β (PDGF-β), transforming growth factor β (TGF-β),
Figure 9.4 Identification of pericytes by electron microscopy. This photo
Notch, and angiopoietin. Endothelial cells have been shown to
depicts a pericyte (P) adjacent to an endothelial cell (E) of a capillary secrete PDGF-β, whereas its receptor is expressed on vascular
vessel. Here, the basement membrane of the glia limitans is directly mural cells, including pericytes (Bell et al. 2010; Hellström et al.
attached to the outermost vascular basement membrane thereby forming 1999; Lindahl et al. 1997; Winkler et al. 2010). The secreted
a fused gliovascular basement membrane (dotted line), which occludes ligand readily interacts with heparin sulfate proteoglycans
the perivascular space in the capillary segment. The pericyte also shares
a basement membrane (dashed line) to the endothelial layer. Therefore,
within the extracellular matrix, thereby establishing a steep
pericytes are by definition part of the vascular wall (first compartment). concentration gradient by local retention of PDGF-β (Andrae
Clear-cut identification is only possible at this ultrastructural level. et al. 2008). This gradient was suggested to be pivotal for proper
Arrow, tight junction complex; L, vascular lumen; N, neuropil. pericyte recruitment and attachment to the capillary tube
P E R I C Y T E S O F T H E C E N T R A L N E RVO U S SYS T E M • 101
(Abramsson et al. 2003, 2007; Armulik et al. 2010; Lindblom receptors, Notch1-Notch4, and five ligands, delta-like (DLL)
et al. 2003). In contrast, pericyte proliferation was dependent 1, 3, and 4, and jagged ( JAG) 1 and 2. Because all these pro-
on the freely diffusible ligand (Abramsson et al. 2003, 2007). teins, receptors as well as ligands, represent transmembrane
Nevertheless, the detailed mechanism behind the different proteins, proper cell contacts between endothelial cells
PDGF-β responses remains to be demonstrated. It is impor- and pericytes are necessary to induce signal transduction
tant to note that the density of pericytes within the develop- by binding of ligand and receptor on directly opposing cell
ing neural tube correlates with the levels of PDGFR-β, thereby membranes. After binding, the Notch intracellular domain
emphasizing the importance of this signaling cascade (Tallquist (NICD) is released by proteolytic cleavage and translocates to
et al. 2003). Moreover, recent studies also suggested a role for the nucleus, where it binds the transcription factor RBP-Jк.
PDGFR-β signaling in pericyte survival (Bell et al. 2010). Mice The putative role of this pathway for proper vascular stabiliza-
lacking the ability of PDGF-β/PDGFR-β signaling by homozy- tion is underlined by studies using a specific knockout model
gous deletion of the respective genes are shown to develop lethal of RBP-Jк in endothelial cells, which is shown to display peri-
phenotypes by a complete absence of pericytes in CNS vessels natal hemorrhages. Indeed, morphological analysis revealed
(Hellström et al. 1999; Leveen et al. 1994; Soriano 1994). impaired pericyte adhesion and reduced pericyte coverage of
The source of TGF-β variants in the CNS is commonly the vasculature characterizing this phenotype (Li et al. 2011;
known to be rather widespread. Cellular sources of this Winkler et al. 2011). The potential role of Notch signaling
ligand are represented by glial cells, neurons, endothe- in vessel maturation is further substantiated by the finding
lial cells, and pericytes, all of which produce TGF-β pri- that endothelial contact to mural cells upregulates Notch3 in
marily as protein-bound forms (Gaengel et al. 2009). On pericytes and vascular smooth muscle cells (Liu et al. 2009).
activation by thrombospondin or integrins, TGF-β binds Notch3 has also been suggested to convey survival of mural
to TGF-β receptor type 2, resulting in activation of the cells (Walshe et al. 2011). Therefore, it is not surprising that
activin like kinase 5 (ALK 5)/Smad pathway, leading to Notch may also be involved in the regulation of PDGFR-β
differentiation, migration, and/or proliferation (Lebrin signaling ( Jin et al. 2008). Thus, it is likely that PDGFR-β in
et al. 2005). Vascular development strictly depends on the concert with Notch and its ligands functions in vascular sta-
function of these signaling cascades. Therefore, knocking bilization and pericyte differentiation, which has already been
out one of the respective genes (TGF-β, ALK 5, endoglin, postulated for DLL4 (Stewart et al. 2011).
or Smad4 and Smad 5) results in an embryonically lethal
phenotype in mice, which exhibit defective vascular devel-
opment (Gaengel et al. 2009). Thus, the current concept
suggests that vascular development critically depends on an ECM production
intact bidirectional signaling between endothelial cells and +differentiation
pericytes via TGF-β. On the one hand, TGF-β binding to inhibititon of
proliferation
TGF-β receptor type 2 on pericytes induces production of
extracellular matrix as well as contractile elements, whereas pericyte recruitment
proliferation is inhibited (Sieczkiewicz and Herman 2003;
van Geest et al. 2010). On the other hand, the TGF-β signal- TGF βR
TGF β

ing cascade in concert with Notch signaling in endothelial N-cadherin


cells enhances firm pericyte attachment by upregulation of PDGFR- β
Notch ligand
N-cadherin in peg-socket contacts (Gerhardt et al. 2000; Li TGFβR
TGFβ N-cadherin
et al. 2011; Winkler et al. 2011). Moreover, pericyte attach- PDGF- β Ang1 TGF β
ment and vessel stabilization are also shown to be driven Notch1
by sphingosine-1-phosphate signaling (Gaengel et al. 2009;
Tie-2
Paik et al. 2004). To our current understanding, recruited TGF βR2
pericytes attach to the endothelial layer and promote sta- S1P1

bilization of the capillary tube by TGF-β signal transduc- S1P


tion via the ALK 5—Smad2/3 pathway and inhibition of
endothelial maturation
endothelial proliferation (Li et al. 2011) Furthermore, sev- BBB formation
eral lines of evidence suggest this signaling cascade to be
crucial for vascular maturation, basement membrane forma- Figure 9.5 Pericyte Signaling. Endothelial cells (yellow) secrete
tion, and BBB establishment (Dohgu et al. 2005; van Geest PDGF-β, which is retained in the extracellular matrix. Binding to the
receptor PDGFR-β on pericytes (green) leads to receptor dimerization
et al. 2010; Walshe et al. 2009) (Fig. 9.5). and activation of signaling cascades, resulting in recruitment toward
A variety of data additionally suggest Notch to be a key endothelial cells. Firm attachment is further driven by binding of TGF-β
player during vascular development and angiogenesis, which to its receptor TGFβR2. In pericytes TGFβR2-mediated signaling results
also implies crucial roles for pericytes. Still there are very lit- in an inhibition of proliferation and production of ECM components. In
tle available data addressing this issue. At least recent studies endothelial cells, binding of TGF-β to its receptor also leads to inhibi-
tion of proliferation and acts in concert with Notch and S1P signaling
demonstrate the importance of Notch signaling for pericyte in upregulation of N-cadherin, which mediates formation of so called
attachment and survival (Li et al. 2011; Liu et al. 2009; Regan peg-socket contacts. Pericytes can also contribute to BBB formation in
and Majesky 2009; Stewart and Wiley 2011; Walshe et al. 2011; the endothelium by angiopoietin 1 (Ang-1)/Tie-2 and TGF-β/TGFβR2
Winkler et al. 2011). In mammals, there are four described signaling. Adapted from Winkler et al. 2011.

102 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
6 F U N C T I O N S O F C E N T R A L N E RVO U S paracellular leakage as a consequence of decreased expression
SYS T E M P E R I C Y T E S of tight junction-associated proteins (Armulik et al. 2010; Bell
et al. 2010). In pericyte deficient mice, Bell and colleagues
demonstrated the progressive reduction of pivotal tight junc-
6.1 P E R I C Y T E S : R E GU L ATO R S O F T H E tion proteins such as occludin, claudin-5, and zonula occludens
B L O O D -B R A I N BA R R I E R 1 (Bell et al. 2010). In fact, increased paracellular leakage of
hydrophilic tracers correlated with an age-dependent decrease
Since the discovery of pericytes, their putative functions in
of pericyte density on CNS vessels. Increased paracellular leak-
the CNS have been discussed repeatedly. The NVU is unique,
age and subsequent accumulation of vasculotoxic and neuro-
not only by means of its architecture, but also by means of its
toxic blood-derived molecules in the vascular wall or adjacent
tight, barrier-forming function. Therefore, it has been tempt-
perivascular spaces may likely result in reduced microvascular
ing to link Paul Ehrlich’s early finding that the brain vascu-
perfusion and therefore result in secondary neurodegeneration
lature proved tight to certain hydrophilic molecules with
(Bell et al. 2010; Winkler et al. 2011).
another feature of CNS vessels, that is, the high density of
The contribution of pericytes to vascular stability was
pericytes within the CNS. Indeed, the number of pericytes
further substantiated by addressing the angiopoietins Ang-1,
covering the vascular wall was found to be significantly higher
Ang-2, and their receptor Tie-2 in the model of PDGF-β–
compared with peripheral organs (Shepro and Morel 1993).
deficient mice. Pericytes and perivascular cell populations are
Thus, a cardinal role for pericytes in the maintenance of the
responsible for the production of Ang-1 and Ang-2, whereas
BBB has been suggested, but has also been attributed to astro-
their receptor is expressed on endothelial cells (Davis et al.
cytes (Arthur et al. 1987; Janzer and Raff 1987; Stewart et al.
1996; Suri et al. 1996). Here, Ang-1 restored the vascular struc-
1981). However, later studies succeeded to demonstrate the
ture and function in models of PDGF-β deficiency (Uemura
establishment of tight junction complexes in the absence of
et al. 2002). Therefore, Ang-1 is proposed to act as a stabiliz-
astrocytes (Felts and Smith 1996; Jaeger and Blight 1997).
ing factor, whereas Ang-2 is a merely destabilizing cue, as it
Using mice deficient for the astrocytic protein glial fibril-
binds to its receptor without inducing signal transduction. The
lary acidic protein (GFAP) Balabanov et al. demonstrated
antagonistic model of both ligands has been confirmed by sev-
an increased coverage of pericytes in CNS vessels (Balabanov
eral other studies (Maisonpierre et al. 1997; Sato et al. 1995;
and Dore-Duff y 1998). This finding was interpreted as a pos-
Suri et al. 1996). There is also evidence that in ischemic models
sible counter-regulation against vascular leakage. Later stud-
Ang-1 is capable of reducing BBB leakage (Zhang et al. 2002).
ies substantiated this theory in that addition of pericytes to
Vascular stability is also shown to depend on sphingosine-
endothelial cell monolayers led to an increased barrier func-
1-phosphate (S1P) signaling as described, which involves
tion for hydrophilic molecules and augmented transendothe-
regulation and expression of N-cadherin and VE-cadherin
lial resistance (Dente et al. 2001). This implies that different
in endothelial cells (Armulik et al. 2005). Expression of both
populations of cells of the NVU are involved in maintaining
molecules is induced via binding of S1P to its receptor (Paik
barrier function and, in case of functional impairment of one,
et al. 2004). VE-cadherin is found in interendothelial junc-
the other can take over to prevent further damage.
tions, whereas N-cadherin is engaged in forming peg-socket
The importance of pericytes in the formation of the BBB
contacts between endothelial cells and pericytes. Interfering
mostly derives from studies using mice lacking PDGF-β or its
with S1P signaling regularly results in decreased pericyte and
receptor. These mice display the same vascular phenotypes of
smooth muscle cell coverage of blood vessels, again causing
pericyte-depleted vessels with hemorrhages and microvascu-
vascular abnormalities and neonatal lethality (Liu et al. 2000).
lar leakage consecutively leading to neonatal lethality. Recent
Interestingly, S1P signaling is currently focused as a putative
studies using depleted and hypomorphic alleles for PDGFR-β
drug target for the therapy of multiple sclerosis by Fingolimod
have demonstrated an early impact of pericytes on the devel-
(FTY720). This drug is suggested to downregulate lymphocyte
opment of the BBB. Vascular alterations were found at the
egress from lymphoid tissues, which is thought to reduce infil-
known time of the appearance of pericytes, an event markedly
tration of autoreactive lymphocytes to the CNS. However, also
preceding the formation of astrocytes (Daneman et al. 2010).
the potential (beneficial) influence for the modulation of neu-
This study also provided evidence that pericyte coverage of the
roinflammation at the CNS vasculature should be discussed.
endothelial wall regulates the formation of tight junctions and
transendothelial vesicle trafficking.
6.2 P E R I C Y T E S A N D A N G I O G E N E S I S
It is important to note that viable pericyte-deficient mice
with disturbed PDGFR-β signaling do not lack the expression In general, pericytes are commonly seen as a rather static
of BBB associated genes (Armulik et al. 2010; Li et al. 2011). In population, which is shown to be critically involved in vessel
contrast, they lack genes downregulating vascular leakage such as maturation and stabilization. However, pericytes are recruited
angiopoietin-2 and plasmalemma associated protein. Moreover, to newly forming vascular tubes by sensing PDGF-β, which
pericytes suppress the expression of genes increasing endothelial is secreted by endothelial cells. There are also several lines of
permeability and infiltration of blood-borne leukocytes already evidence strongly suggesting a complex and more dynamic
at a time of a rather immature BBB (Daneman et al. 2010). function during angiogenesis because pericytes are described
However, the crucial role of pericytes in regulating the to express matrix metalloproteinases (MMP-2, MMP-3, and
BBB is not limited to embryogenesis, but remains essential MMP-9) (Candelario-Jalil et al. 2009; Virgintino et al. 2007)
throughout adulthood and aging. In the aging brain, loss of and urokinase plasminogen activator receptor (Dore-Duff y
CNS pericytes has been demonstrated to accompany increased et al. 2000). On the other hand, and quite contrary to the
P E R I C Y T E S O F T H E C E N T R A L N E RVO U S SYS T E M • 103
function of these proteins they also contribute to the synthesis cells (Dore-Duff y et al. 2000; Gerhardt and Betsholtz 2003;
of extracellular matrix components (Diaz-Flores et al. 2009; Ozerdem and Stallcup 2003, 2004; Virgintino et al. 2007).
Stratman et al. 2009; van Geest et al. 2010) and tissue inhibi-
tor of metalloproteinase 3 (TIMP-3), which is a potent inhib-
6.3 P E R I C Y T E S C O N T RO L C A P I L L A RY
itor of a number of MMPs (Saharinen et al. 2008).
D I A M ET E R A N D B L O O D FL OW
Ultrastructural studies indeed demonstrated that pericytes
are the first population within the NVU to respond to brain In contrast with vascular smooth muscle cells, the major pro-
hypoxia and traumatic brain injury, which are strong stimuli for cesses of pericytes are oriented in parallel to the longitudinal
angiogenesis (Dore-Duff y et al. 1999, 2000; Gonul et al. 2002). axis of the vessels. Therefore, they should hardly be capable of
As early as 2 hours after brain hypoxia pericytes showed first constricting capillary vessels. However, pericytes were initially
morphological alterations, whereas no other populations of the described as contractile elements by Rouget in 1873. He distin-
NVU were visibly affected. The authors demonstrated a thick- guished this population from migratory and phagocytic leuko-
ening of the luminal basement membrane, whereas the ablu- cytes (Rouget 1874, 1879), and later investigators confirmed his
minal one was evidently thinned out. Moreover, characteristic assumption (Mayer 1902; Vimtrup 1922; Zimmermann 1923).
“peaks” in the sense of pericytic processes were detectable point- Mayer and Zimmermann even suggested pericytes and smooth
ing toward the parenchyma. These findings were interpreted as muscle cells of the tunica media to be a continuous population
initial signs of migration from the vascular wall (Gonul et al. with transitional forms between the two extremes (Mayer 1902;
2002). Comparable observations were reported after traumatic Zimmermann 1923). Applying an early form of live imaging tech-
brain injury. In this study the authors observed elongated peri- nique, Vimtrup observed capillary contractions, which began at
cytes and confirmed the disappearance of the abluminal base- single pericytes and spread onward in both directions from the
ment membrane at leading pericyte processes. Additionally, cell body (Vimtrup 1922). This study was performed in young
migration of pericytes was confirmed by a decreased density of living larvae, which were constantly rinsed with fresh water at the
pericyte on affected vessels (Dore-Duff y et al. 2000). body, whereas the transparent tail was observed on a microscope
Activation and migration of cells can be linked to the expres- slide. To ensure that what he observed really were capillaries, he
sion of urokinase plasminogen activator (uPA) and its receptor restricted his studies to areas in which the afferent arteriole as
(uPAR) on migrating cells (Blasi 1999; Washington et al. 1996). well as the efferent venule were visible. That way, he was able to
Indeed, pericytes are reported to express uPAR on leading pro- identify the capillary bed in between (Vimtrup 1922).
cesses (Dore-Duffy et al. 2000). It is important to note that the Since then, the discussion of whether pericytes contribute to
complex of ligand and receptor represents an active protease and the regulation of blood flow by contracting capillaries has always
its expression on migrating pericytes might be crucial to over- been alive. More recent studies corroborated the view on pericytes
come the barrier of the ensheathing basement membrane to clear as regulators of capillary blood flow by describing their potential
a way for other cell types, such as endothelial cells. Of note, peri- ability to respond to vasoactive substances, such as nitric oxide,
cytes remaining in position exhibited cytoplasmic and nuclear prostacyclin, angiotensin II, and endothelin-1, as they are reported
alterations suggesting degeneration of nonmigratory pericytes to express corresponding receptors (Chakravarthy and Gardiner
(Dore-Duffy et al. 2000). Moreover, leading tips of migratory 1999; Dehouck et al. 1997; Healy and Wilk 1993). Another
pericytes are shown to interfere with synaptic complexes. This was prerequisite for active contraction is the presence of contractile
interpreted as an initial sign of synaptic stripping originally relat- filaments; in fact, both smooth muscle and non–smooth muscle
ing to the concept of an active displacement of synaptic terminals isoforms were found in CNS pericytes (Herman and D’Amore
by microglial cells (Blinzinger and Kreutzberg 1968; Trapp et al. 1985), albeit with species-dependent variations of the expression
2007). Whether this is indeed an active displacement by pericytes of alpha smooth muscle actin (SMA). For example, in embryonic
or a neuronal process that further leads to appearance of pericytic chicken, SMA can be detected in virtually all pericytes, whereas
processes in a second event is yet to be elucidated. in mice and rats the situation is quite different (Gerhardt et al.
Furthermore, hypoxia is reported to stimulate production 2000; Hellström et al. 1999). In mice, the expression of SMA
of vascular endothelial growth factor (VEGF) in pericytes in cerebral pericytes is restricted to the ones positioned on ves-
(Yamagishi et al. 1999). This molecule is reported to increase sels of a diameter larger than 10 μm. Vessels with a diameter of
pericyte density on newly formed vessels in a dose-dependent less than 10 μm are on the other hand negative for SMA (Alliott
manner (Benjamin et al. 1998). With the expression of VEGF, et al. 1999; Nehls and Drenckhahn 1991). These findings raise
pericytes do also have an impact on endothelial cells by enhancing the question of whether or not pericytes are indeed structurally
endothelial survival, proliferation, and formation of angiogenic capable of contracting capillaries because these proteins are lack-
sprouts (Darland et al. 2003; Ozerdem and Stallcup 2004). ing in the respective segment. Another explanation is that com-
In summary, pericytes appear to have a dual role in angio- mon methods simply fail to detect low levels of SMA in pericytes,
genesis. On the one hand, they seem to be capable of inducing which again would raise the question of whether such low expres-
endothelial cell migration and proliferation during early phases sion under the detection level is functionally relevant.
(Darland et al. 2003; Ozerdem and Stallcup 2003, 2004). On However, the differential expression of contractile elements is
the other hand, they promote vascular stability and reduction nevertheless a remarkable finding because it suggests functional
of proliferation in later stages (Hellström et al. 2001; Li et al. differences of CNS pericytes along the vascular tree. A possible
2011). Therefore, it may be important to clarify whether peri- explanation may be given by the morphology of the NVU within
cytes initiate vascular sprouting and guide endothelial tube the respective vascular segment. At the level of capillaries, astro-
formation or if they are recruited into tubes by endothelial cytic endfeet of the glia limitans are separated only by a basement
104 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
membrane. In non–capillary vessels, direct and specific interac- the finding that hyperglycemia- mediated dephosphorylation of
tion is hampered by the presence of PVS (see Fig. 9.1). Thus, the PDGFR-β led to resistance to endothelially secreted PDGF-β,
loss of astrocytic contact may change the expression pattern to which led to pericyte apoptosis in this model (Geraldes et al.
a rather smooth muscle-like pericyte isoform. However, this 2009). Therefore, it is possible that the PDGFR-β pathway may
potential influence needs to be determined. be target for future pharmacological inventions.
Current textbooks comprehensively claim that blood flow Recently, pericytes have also become drug targets in tumor
is regulated by precapillary arterioles. Therefore, it is notewor- therapy because they are vitally involved in angiogenesis. Of
thy that the majority of the noradrenergic innervation of CNS note, antiendothelial tumor therapy alone by using VEGF
blood vessels is reported to terminate in the vicinity of capillaries inhibitors proved insufficient because pericytes often remained
(Cohen et al. 1997). Indeed, two recent studies using live imaging in position (Baluk et al. 2005). They are believed to provide a
techniques confirmed Rouget’s original concept of pericytes as scaffold for rapidly regrowing tumor vessels after removal of
contractile elements in the capillary bed. In 2006, Peppiatt et al. VEGF inhibition. Therefore, a synergistic therapy was suggested
demonstrated pericyte-driven capillary constriction by ATP and affecting both proliferation of endothelial cells and survival of
noradrenalin in rat retinal and cerebellar slices. Also, administra- pericytes. In respective studies the positive effect in tumor ves-
tion of GABA blockers led to a visible reduction of the capillary sel reduction was confirmed in animal models by combining
diameter, which was readily abrogated by addition of glutamate anti-VEGF and anti-PDGF-β/PDGFR-β signaling (Bergers et
(Peppiatt et al. 2006). The constriction of capillary vessels by al. 2003; Erber et al. 2004). However, inhibition of PDGFR-β
pericytes was further demonstrated in vivo (Fernández-Klett et alone does not result in endothelial regression (Abramsson et
al. 2010). Application of intravital two-photon laser scanning al. 2003), but augments the effect of VEGF inhibitors (Bergers
microscopy in mice did not only allow visualization of constric- et al. 2003) or antimitotic drugs such as Taxol (Pietras et al.
tions of the capillary wall in response to thromboxane analogs, but 2002). Recently, also aminopeptidase N (APN) has become of
also allowed determination of red blood cell perfusion changes interest in tumor therapy as combined treatment with antien-
in constricted capillaries. Furthermore, the authors described a dothelial therapies prolonged survival of human neuroblastoma
significantly higher change of the capillary diameter adjacent to bearing mice (Loi et al. 2010). Therefore, it may be possible that
pericyte bodies compared with areas only reached by pericytic future tumor therapies include antipericyte drugs and hopefully
processes (Fernández-Klett et al. 2010). This finding may reflect enhance outcome by deprivation of tumor angiogenesis.
the unique morphology of pericytes, which better enables them Neuronal function critically depends on an intact NVU;
to retract their longitudinal processes than to constrict the vas- consequently, neurovascular dysfunction is often associated with
cular circumference. Thereby, contraction may lead to shortened neurodegeneration (Zlokovic 2008). Therefore, the function of
processes and consequently thickening of the body, which again the vascular wall and its proper architecture and signaling, which
narrows the capillary lumen. However, the authors also dem- also involve pericytes, is essential to maintain a healthy neuronal
onstrated that capillary dilatation is not required for functional microenvironment. Pericyte-deficient mice have recently been
hyperemia (Fernández-Klett et al. 2010). Thus, the functional reported to express a neurodegenerative phenotype as a conse-
impact of pericytes on cerebral blood flow under physiological quence of BBB breakdown, leading to toxic extravasation of plasma
conditions still remains to be determined, but methods are now proteins and chronic hypoperfusion with consequential hypoxia
available to continue on this path. (Bell et al. 2010). Because vascular lesions in pericytes-deficient
mice precede inflammation and neuronal damage, it is possible
that this secondary inflammation also contributes to the observed
7 PERICY TES IN DISEASES OF THE neuronal damage (Quaegebeur et al. 2010).
C E N T R A L N E RVO U S SYS T E M Nevertheless, the exact role of pericytes in specific neu-
rodegenerative disorders such as Alzheimer disease remains
Diabetic retinopathy represents a widespread complication of to be determined. Accumulations of amyloid-β are known
type I and type II diabetes in which early pericyte loss is a com- to occur around capillaries and pericytes (Vinters et al. 1994;
mon pathological hallmark. Here, a nonproliferative phase can Wisniewski et al. 1992) and addition of amyloid-β peptides to
be distinguished from a proliferative phase (Hammes et al. 2011). isolated human brain pericytes has proved to lead to pericyte
The nonproliferative phase is characterized by vessel regression, death in vitro (Wilhelmus et al. 2007). Furthermore, mouse
occluded and pericyte-deprived capillaries, and microaneurysms, models of amyloid-β overexpression display BBB breakdown
whereas the proliferative phase exhibits massive proliferation of and reduced vascular density before neuronal loss or amyloid
abnormal, rupture-predisposed vessels. The latter often give rise deposition occurs (Paul et al. 2007; Zlokovic 2005). Because
to hemorrhages and retinal detachment. Interestingly, mouse several lines of evidence suggest vascular basement membranes
models of PDGF-β deficiency or Ang-2 overexpression have to be drainage pathways for solutes (Carare et al. 2008; Hawkes
demonstrated a phenotype almost mimicking nonproliferative et al. 2011), impaired pericyte function and contractility by
diabetic retinopathy (Hammes et al. 2004; Pfister et al. 2010). vascular and perivascular amyloid beta deposition may cause
However, in a study selectively deleting PDGF-β in endothelial reduced clearance of soluble amyloid peptides, thus perpetuat-
cells the authors also demonstrated a switch toward a prolifera- ing a vicious cycle. This hypothesis may be corroborated by the
tive phenotype as pericyte coverage dropped below 50%, thereby finding that experimentally induced hypercontraction of cere-
suggesting a threshold for pericyte loss either leading to nonpro- bral arterioles resulted in reduced amyloid clearance (Bell et al.
liferative or proliferative vascular pathologies (Enge et al. 2002). 2009). However, the precise role of pericytes and their potential
The critical role of PDGF-β pathway was further confirmed by role as targets in clinical therapies are yet to be established.

P E R I C Y T E S O F T H E C E N T R A L N E RVO U S SYS T E M • 105


8 S U M M A RY A N D P E R S P E C T I VE S Benjamin LE, Hemo I, Keshet E. 1998. A plasticity window for blood
vessel remodeling is defined by pericyte coverage of the preformed
endothelial network and is regulated by PDGF-β and VEGF.
Although several lines of evidence now confirm a role for peri- Development 125:1591–1598.
cytes in regulating capillary diameters, the lack of a specific Bergers G, Song S, Mayer-Morse N, Bergslan E, Hanahan D. 2003.
marker for these cells still causes confusion. Even though under Benefits of targeting both pericytes and endothelial cells in the tumor
normal conditions pericytes can be identified by their topo- vasculature with kinase inhibitors. J Clin Invest 111:1287–1295.
graphic localization within the vascular wall, it is still impos- Blasi F. 1999. UPA, uPA, PAI-1:Key intersection of proteolytic adhesive
and chemotactic highways. Immunol Today 18:415–417.
sible to unequivocally determine their fate once they detach Blinzinger K, Kreutzberg G. 1968. Displacement of synaptic terminals
from the endothelial basement membrane. This is illustrated from regenerating motoneurons by microglial cells. Z Zellforsch
by the recent description of pericytes playing a role in scar for- Mikrosk Anat 85(2):145–157.
mation after spinal cord injury (Göritz et al. 2011). Although Bobbie MW, Roy S, Trudeau K, Munger SJ, Simon AM. 2010. Reduced
cells expressing pericyte markers were clearly identified within connexion 43 expression and its effect on the development of vas-
cular lesions in retinas of diabetic mice. Invest Ophthalmol Vis Sci
the scar, one cannot exclude that this expression is induced on 51:3758–3763.
cells that never had been pericytes based on the topographic Bondjers C, Kaln M, Hellstrom M, Scheidl SJ, Abramsson A, Renner
definition. Unless both, basement membranes and pericytes O, et al. 2003. Transcription profi ling of PDGF-B deficient embryos
can be visualized in vivo over long periods of time, this issue identifies RGS5 as a novel marker for pericytes and vascular smooth
will remain a matter of debate. Eventually it may turn out that muscle cells. Am J Pathol 162:721–729.
Candelario-Jalil E, Yang Y, Rosenberg GA. 2009. Diverse roles of matrix
the term pericytes is misleading, because it only describes a sta- metalloproteinases and tissue inhibitors of metalloproteinases in neu-
tionary phase of a highly versatile and mobile cell. roinflammation and cerebral ischemia. Neuroscience 158(3):983–994.
Carare RO, Bernardes-Silva M, Newman TA, Page AM, Nicoll JA,
Perry VH, et al. 2008. Solutes, but not cells, drain from the brain
REFERENCES parenchyma along basement membranes of capillaries and arteries:
significance for cerebral amyloid angiopathy and neuroimmunology.
Abramsson A, Lindblom P, Betsholtz C. 2003. Endothelial and non endothe- Neuropathol Appl Neurobiol 34(2):131–144.
lial sources of PDGF-B regulate pericyte recruitment and influence vas- Chakravarthy U, Gardiner TA 1999. Endothelium-derived agents in
cular pattern formation in tumors. J Clin Invest 112:1142–1151. pericyte function/dysfunction. Prog Retina Eye Res 18:511–527.
Abramsson A, Kurup S, Busse M, Yamada S, Lindblom P, Schallmeier E, Cho H, Kozaka T, Bondjers C, Betsholtz C, Kehrl J. 2003. Pericyte spe-
et al. 2007. Defective N-sulfation of heparin sulphate proteoglycans cific expression of RGS5: implications for PDGF and EDG receptor
limits PDGF-BB binding and pericyte recruitment in vascular devel- signalling during vascular maturation. FASEB J 17:440–442.
opment. Genes Dev 21(3):316–331. Cohen MP, Frank RN, Khalifa AA. 1980. Collagen production by cul-
Alliott F, Rutin J, Leenen PJM, Pessac B. 1999. Pericytes and perien- tured retinal capillary pericytes. Invest Ophthalmol Vis Sci 19:90–94.
dothelial cells of brain parenchyma vessels co-express aminopepti- Cohen Z, Molinatti G, Hamel E. 1997. Astroglial and vascular interac-
dase n, aminopeptidase a and nestin. J Neurosc Res 58:367–378. tions of noradrenaline terminals in the rat cerebral cortex. J Cereb
Andrae J, Gallini R, Betsholtz C. 2008. Role of platelet-derived growth Blood Flow Metab 17:894–904.
factors in physiology and medicine. Genes Dev 22:1276–1312. Daneman R, Zhou L, Kebede AA, Barres BA. 2010. Pericytes are
Armulik A, Abramsson A, Betsholtz C. 2005. Endothelial/pericyte required for Blood-brain barrier integrity during embryogenesis.
interactions. Circ Res 97(6):512–523. Nature 468:562–566.
Armulik A, Genove G, Mäe M, Nisancioglu MH, Wallgard E, Niaudet Darland DC, Massingham LJ, Smith SR, Piek E, Saint-Geniez M,
C, et al. 2010. Pericytes regulate the blood-brain barrier. Nature 468 D’Amore PA. 2003. Pericyte production of cell-associated VEGF is
(7323):557–561. differentiation-dependent and is associated with endothelial survival.
Arthur FE, Shivers RR, Bowman PD. 1987. Astrocyte-mediated induc- Dev Biol 264(1):275–288.
tion of tight junctions in brain capillary endothelium: an efficient Davis S, Aldrich TH, Jones PF, Acheson A, Compton DL, Jain V, et al.
in vitro model. Brain Res 433:155–159. 1996. Isolation of angiopoietin-1, a ligand for the TIE2 receptor, by
Balabanov R, Dore-Duff y P. 1998. Role of the CNS microvascular peri- secretion-trap expression cloning. Cell 87:1161–1169.
cyte in the blood-brain barrier. J Neurosci Res 53:637–644. Dehouck MP, Vigne P, Torpier G, Breittmayer JP, Cecchelli R, Frelin C.
Balabanov R, Washington R, Wagnerova J, Dore-Duff y P. 1996. CNS micro- 1997. Endothelin-1 as a mediator of endothelial cell-pericyte interactions
vascular pericytes express macrophage-like function, cell surface integrin in bovine brain capillaries. J Cereb Blood Flow Metab 17:464–469.
alpha M, and macrophage marker ED-2. Microvasc Res 52:127–142. Dente CJ, Steffes CP, Speyer C, Tyburski JG 2001. Pericytes augment the
Baluk P, Hashizume H, McDonald DM. 2005. Cellular abnormalities of capillary barrier in in vitro cocultures. J Surg Res 97:85–91.
blood vessels as targets in cancer. Curr Opin Genet Dev 15:102–111. Diaz-Flores L, Gutierrez R, Madrid JF, Valera H, Valladares F, Acosta
Bautch VL and James JM. 2009. Neurovascular development: the begin- E, et al. 2009. Pericytes. Morphofunction, interactions and pathol-
ning of a beautiful friendship. Cell Adh Migr 3:199–204. ogy in a quiescent and activated mesenchymal cell niche. Histol
Bechmann I, Galea I, Perry VH. 2007. What is the blood-brain barrier Histopathol 24:909–969.
(not)? Trends Immunol 28(1):5–11. Dohgu S, Takata F, Yamauchi A, Nakagawa S, Egawa T, Naito M, et al.
Bechmann I, Kwidzinski E, Kovac AD, Simbürger E, Horvath T, Gimsa 2005. Brain pericytes contribute to the induction and up-regulation
U, et al. 2001a. Turnover of rat brain perivascular cells. Exp Neurol of blood-brain barrier functions through transforming growth
168(2):242–249. factor-beta production. Brain Res 1038:208–215.
Bechmann I, Priller J, Kovac A, Böntert M, Wehner T, Klett FF, et al. Dore-Duff y P, Balabanov R. 1998. The role of CNS microvascular peri-
2001b. Immune surveillance of mouse brain perivascular spaces by cyte in leukocyte polarization of cytokine-secreting phenotype.
blood-borne macrophages. Eur J Neurosci 14:1651–1658. J Neurochem 70:72.
Bell RD, Deane R, Chow N, Long X, Sagare A, Singh I, et al. 2009. SRF Dore-Duff y P, Balabanov R, Beaumont T, Hritz MA, Harik SI, LaManna
and myocardin regulate LRP-mediated amyloid-beta clearance in JC. 1999. Endothelial activation following prolonged brain hypoxia.
brain vascular cells. Nat Cell Biol 11(2):143–153. Microvasc Res 57(2):75–85.
Bell RD, Winkler EA, Sagare AP, Singh I, LaRue B, Deane R, et al. 2010. Dore-Duff y P, Owen C, Balabanov R, Murphy S, Beaumont T, Rafols
Pericytes control key neurovascular functions and neuronal pheno- JA. 2000. Pericyte migration from the vascular wall in response to
type in the adult brain and during brain aging. Neuron 68:409–427. traumatic brain injury. Microvasc Res 60:55–69.

106 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
Enge M, Bjarnegård M, Gerhardt H, Gustafsson E, Kalén M, Asker N. Jaeger CB, Blight AR. 1997. Spinal cord compression injury in guinea
2002. Endothelium-specific platelet-derived growth factor-B abla- pigs: structural changes of endothelium and its perivascular cell asso-
tion mimics diabetic retinopathy. EMBO J 21(16):4307–4316. ciations after blood-brain barrier breakdown and repair. Exp Neurol
Erber R, Thurnher A, Katsen AD, Groth G, Kerger H, Hammes HP, et al. 144:381–399.
2004. Combined inhibition of VEGF and PDGF signalling enforces Janzer RC, Raff MC. 1987. Astrocytes induce blood–brain barrier prop-
tumor vessel regression by interfering with pericyte-mediated endo- erties in endothelial cells. Nature 325:253–257.
thelial cell survival mechanisms. FASEB J 18:338–340. Jin S, Hansson EM, Tikka S, Lanner F, Sahlgren C, Farnebo F, et al.
Etchevers HC, Vincent C, Le Douarin NM, Couly GF. 2001. The 2008. Notch signalling regulates platelet-derived growth factor
cephalic neural crest provides pericytes and smooth muscle cells to all receptor-beta expression in vascular smooth muscle cells. Circ Res
vessels of the face and forebrain. Development 128:1059–1068. 102(12):1483–1491.
Felts PA, Smith KJ. 1996. Blood-brain barrier permeability in Kokovay E, Li L, Cunningham LA. 2006. Angiogenic recruitment of
astrocyte-free regions of the central nervous system remyelinated by pericytes from bone marrow after stroke. J Cereb Blood Flow Metabol
Schwann cells. Neuroscience 75:643–655. 26:545–555.
Fernández-Klett F, Offenhauser N, Dirnagl U, Priller J, Lindauer U. Korn J, Christ B, Kurz H. 2002. Neuroectodermal origin of brain peri-
2010. Pericytes in capillaries are contractile in vivo, but arterioles cytes and vascular smooth muscle cells. J Comp Neurol 442:78–88.
mediate functional hyperemia in the mouse brain. Proc Natl Acad Krause D, Kunz J, Dermietzel R. 1992. Cerebral pericytes—a second
Sci U S A 107:22290–22295. line of defence in controlling blood-brain barrier peptide metabo-
Gaengel K, Genove G, Armulik A, Betsholtz C. 2009. Endothelial-mural lism. In: Drewes LR, Betz AL (eds.), Frontiers in cerebral vascular
cell signalling in vascular development and angiogenesis. Arterioscler biology: transport and its regulation. New York: Raven.
Thromb Vasc Biol 29:630–638. Krüger and Bechmann. 2010. CNS pericytes: concepts, misconceptions
van Geest RJ, Klaassen I, Vogels IM, van Noorden CJ, Schlingemann RO. and a way out. Glia 58:1–10.
2010. Differential TGF-beta signalling in retinal vascular cells: a role Kurz H. 2009. Cell lineages and early patterns of embryonic CNS vascu-
in diabetic retinopathy? Invest Ophthalmol Vis Sci 51: 1857–1865. larization. Cell Adh Migr 3:205–210.
Geraldes P, Hiraoka-Yamamoto J, Matsumoto M, Clermont A, Leitges Kunz J, Krause D, Gehrmann J, Dermietzel R. 1995. Changes in the
M, Marette A, et al. 2009. Activation of PKC-delta and SHP-1 by expression pattern of blood–brain barrier-associated pericytic amin-
hyperglycemia causes vascular cell apoptosis and diabetic retinopa- opeptidase N (pAP N) in the course of acute experimental autoim-
thy. Nat Med 15(11):1298–1306. mune encephalomyelitis. J Immunol 59:41–55.
Gerhardt H, Betsholtz C. 2003. Endothelial-pericyte interactions in Lamagna C, Bergers G. 2006. The bone marrow constitutes a reservoir of
angiogenesis. Cell Tissue Res 314(1):15–23. pericyte progenitors. J Leukoc Biol 80:677–681.
Gerhardt H, Wolburg H, Redies C. 2000. N-cadherin mediates Lebrin F, Deckers M, Bertolino P, Ten Dijke P 2005. TGF- beta receptor
pericytic-endothelial interaction during brain angiogenesis in the function in the endothelium. Cardiovasc Res 65:599–608.
chicken. Dev Dyn 218(3):472–479. Leveen P, Pekny M, Gebre-Medhin S, Swolin B, Larsson E, Betsholtz
Gonul E, Duz B, Kahraman S, Kayali H, Kubar A, Timurkaynak E. C. 1994. Mice deficient for PDGF B show renal, cardiovascular, and
2002. Early response to brain hypoxia in cats: an ultrastructural hematological abnormalities. Genes Dev 8:1875–1887.
study. Microvasc Res 64:116–119. Li F, Lan Y, Wang Y, Wang J, Yang G, Meng F, et al. 2011. Endothelial
Göritz C, Dias DO, Tomilin N, Barbacid M, Shupliakov O, Frisen J. 2011. Smad4 maintains cerebrovascular integrity by activating N-cadherin
A pericyte origin of spinal cord scar tissue. Science 333:238–242. through cooperation with Notch. Dev Cell 20(3):291–302.
Graeber MB, Streit WJ. 1990. Perivascular microglia defined. Trends Lindahl P, Johansson BR, Leven P, Betsholtz C. 1997. Pericyte loss
Neurosci 13(9):366. and microaneurysm formation in PDGF-B-deficient mice. Science
Graeber MB, Streit WJ, Kreutzberg GW. 1989. Identity of ED-2 positive 277:242–245.
perivascular cells in rat brain. J Neurosci Res 22:103. Lindblom P, Gerhardt H, Liebner S, Abramsson A, Jeltsch M, Mitchell C,
Greter M, Heppner FL, Lemos MP, Odermatt BM, Goebels N, Laufer T, et al. 2003. Endothelial PDGF-B retention is required for proper invest-
et al. 2005. Dendritic cells permit immune invasion of the CNS in an ment of pericytes in the microvessel wall. Genes Dev 17:1835–1840.
animal model of multiple sclerosis. Nat Med 11(3):328–334. Liu Y, Wada R, Yamashita T, Mi Y, Deng CX, Hobson JP, et al. 2008.
Hammes HP, Feng Y, Pfister F, Brownlee M. 2011. Diabetic retinopathy: Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate,
targeting vasoregression. Diabetes 60(1):9–16. is essential for vascular maturation. J Clin Invest 106:951–961.
Hammes HP, Lin J, Wagner P, Feng Y, Vom Hagen F, Krzizok T, et al. Liu H, Kennard S, Lilly B. 2009. NOTCH3 expression is induced
2004. Angiopoietin-2 causes pericyte dropout in the normal ret- in mural cells through an autoregulatory loop that requires
ina: evidence for involvement in diabetic retinopathy. Diabetes endothelial-expressed JAGGED1. Circ Res 104(4):466–475.
53(4):1104–1110. Loi M, Marchiò S, Becherini P, Di Paolo D, Soster M, Curnis F, et al.
Hawkes CA, Härtig W, Kacza J, Schliebs R, Weller RO, Nicoll JA, 2010. Combined targeting of perivascular and endothelial tumor
et al. 2011. Perivascular drainage of solutes is impaired in the ageing cells enhances anti-tumor efficacy of liposomal chemotherapy in neu-
mouse brain and in the presence of cerebral amyloid angiopathy. Acta roblastoma. J Control Release 145(1):66–73.
Neuropathol 121(4):431–443. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ,
Healy DP, Wilk S. 1993. Localization of immunoreactive glutamyl Radziejewski C, et al. 1997. Angiopoietin-2, a natural antagonist for
aminopeptidase in rat brain. II. Distribution and correlation with Tie2 that disrupts in vivo angiogenesis. Science 277:55–60.
angiotensin II. Brain Res 606:295–303. Mandarino LJ, Sundarraj N, Finlayson J, Hassell HR. 1993. Regulation
Hellström M, Gerhardt H, Kalen M, Li X, Eriksson U, Wolburg H, of fibronectin and laminin synthesis by retinal capillary endothelial
Betsholtz C. 2001. Lack of pericytes leads to endothelial hyperplasia cells and pericytes in vitro. Exp Eye Res 57:609–621.
and abnormal vascular morphogenesis. J Cell Biol 153:543–553. Mayer S. 1902. Die Muscularisierung der capillaren Blutgefäße. Anat
Hellström M, Kalen M, Lindahl P, Abramsson A, Betsholtz C. 1999. Anz 21:442–455.
Role of PDGF-B and PDGFR-beta in recruitment of vascular smooth Mildner A, Schmidt H, Nitsche M, Merkler D, Hanisch UK, Mack M,
muscle cells and pericytes during embryonic blood vessel formation et al. 2007. Microglia in the adult brain arise from Ly-6ChiCCR2+
in the mouse. Development 126:3047–3055. monocytes only under defined host conditions. Nat Neurosci
Herman IM, D’Amore PA. 1985. Microvascular pericytes contain mus- 10:1544–1553.
cle and nonmuscle actins. J Cell Biol 101:43–52. Nehls V, Drenckhahn D. 1991. Heterogeneity of microvascular pericytes
Hickey WF, Kimura H. 1988. Perivascular microglial cells of the for smooth muscle type alpha-actin. J Cell Biol 113:147–154.
CNS are bone marrow-derived and present antigen in vivo. Science Nehls V, Dreckhahn D. 1993. The versatility of microvascular pericytes:
239:290–292. from mesenchyme to smooth muscle? Histochemistry 99:1–12.

P E R I C Y T E S O F T H E C E N T R A L N E RVO U S SYS T E M • 107


Ozerdem U, Grako KA, Dahlin-Huppe K, Monosov E, Stallcup WB. Stewart KS, Zhou Z, Zweidler-McKay P, Kleinerman ES. 2011. Delta-like
2001. NG2 proteoglycan is expressed exclusively by mural cells dur- ligand 4-Notch signalling regulates bone marrow-derived pericyte/
ing vascular morphogenesis. Dev Dyn 222:218–227. vascular smooth muscle cell formation. Blood 117(2):719–726.
Ozerdem U, Stallcup WB. 2003. Early contribution of pericytes to angi- Stratman AN, Malotte KM, Mahan RD, Davis MJ, Davis GE. 2009. Pericyte
ogenic sprouting and tube formation. Angiogenesis 6:241–249. recruitment during vasculogenic tube assembly stimulates endothelial
Ozerdem U, Stallcup WB. 2004. Pathological angiogenesis is reduced by tar- basement membrane formation. Blood 114(24):5091–5101.
geting pericytes via the NG2 proteoglycan. Angiogenesis 7:269–276. Stratman AN, Schwindt AE, Malotte KM, Davis GE. 2010.
Paik JH, Skoura A, Chae SS, Cowan AE, Han DK, Proia RL, et al. 2004. Endothelial-derived PDGF-BB and HB-EGF coordinately regulate
Sphingosine-1-phosphate receptor regulation on N-cadherin medi- pericyte recruitment during vasculogenic tube assembly and stabili-
ates vascular stabilization. Genes Dev 18:2392–2403. zation. Blood 116:4720–4730.
Paul J, Strickland S, Melchor JP. 2007. Fibrin deposition accelerates Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, Davis S, et al.
neurovascular damage and neuroinflammation in mouse models of 1996. Requisite role of angiopoietin-1, a ligand for the TIE2 receptor,
Alzheimer’s disease. J Exp Med 204(8):1999–2008. during embryonic angiogenesis. Cell 87:1171–1180.
Peppiatt CM, Howarth C, Mobbs P, Attwell D. 2006. Bidirectional con- Tallquist MD, French WJ, Soriano P. 2003. Additive effects of PDGF
trol of CNS capillary diameter by pericytes. Nature 443:700–704. receptor beta signalling pathways in vascular smooth muscle cell
Pfister F, Wang Y, Schreiter K, vom Hagen F, Altvater K, Hoff mann S, development. PloS Biol 1:e52.
et al. 2010. Retinal overexpression of angiopoietin-2 mimics diabetic Trapp BD, Wujek JR, Criste GA, Jalabi W, Yin X, Kidd GJ, et al.
retinopathy and enhances vascular damages in hyperglycemia. Acta 2007. Evidence for synaptic stripping by cortical microglia. Glia
Diabetol 47(1):59–64. 55:360–368.
Pietras K, Rubin K, Sjoblom T, Buchdunger E, Sjoquist M, Heldin C-H et al. Uemura A, Ogawa M, Hirashima M, Fujiwara T, Koyama S, Takagi H,
2002. Inhibition of PDGF receptor signaling in tumor stroma enhances et al. 2002. Recombinant angiopoietin-1 restores higher-order archi-
antitumor effect of chemotherapy. Cancer Res 62:5476–5484. tecture of growing blood vessels in mice in the absence of mural cells.
Priller J, Flugel A, Wehner T, Boentert M, Haas CA, Prinz M, et al. J Clin Invest 110:1615–1617.
2001. Targeting gene-modified hematopoietic cells to the central Vimtrup BJ. 1922. Beiträge zur Anatomie der Capillaren. Ü ber contac-
nervous system: use of green fluorescent protein uncovers microglial tile Elemente in der Gefäßwand der Blutcapillaren. Zeitschr Anat
engraftment. Nat Med 7:1356–1361. Entwickl 65:150–182.
Prinz M, Priller J, Sisodia SS, Ransohoff RM. 2011. Heterogeneity of Vinters HV, Secor DL, Read SL, Frazee JG, Tomiyasu U, Stanley TM,
CNS myeloid cells and their roles in neurodegeneration. Nat Neurosci et al. 1994. Ultrastruct Pathol 18(3):333–348.
14(10):1227–1235. Virgintino D, Girolamo F, Errede M, Capobianco C, Robertson D,
Quaegebeur A, Segura I, Carmeliet P. 2010. Pericytes: blood-brain bar- Stallcup WB, et al. 2007. An intimate interplay between precocious,
rier safeguards against neurodegeneration? Neuron 68(3):321–323. migratine pericytes and endothelial cells governs human fetal brain
Regan JN, Majesky MW. 2009. Building a vessel wall with notch signal- angiogenesis. Angiogenesis 10(1):35–45.
ling. Circ Res 104(4):419–421. Walshe TE, Connell P, Cryan L, Ferguson G, Gardiner T, Morrow
Rhodin JAG. 1968. Ultrastructure of mammalian venovenous capillaries, D, et al. 2011. Microvascular retinal endothelial and pericyte cell
venules and small collecting venules. J Ultrastruc Res 25:452–500. apoptosis in vitro: role of hedgehog and Notch signalling. Invest
Rouget C. 1873. Mémoire sur le développement, la structure et les pro- Ophthalmol Vis Sci 52(7):4472–4483.
prietés physiologiques des capillaires sanguins et lymphatiques. Arch Walshe TE, Saint-Geniez M, Maharaj AS, Sekiyama E, Maldonado AE,
Physiol Norm et Path 5:603–663. D’Amore PA. 2009. TGF-beta is required for vascular barrier func-
Rouget C. 1874. Note sur le développement de la tunique contractile des tion, endothelial survival and homeostasis of the adult microvascula-
vaisseaux. Compt rend acad sci Paris 59:559–562. ture. PLoS One 4(4):e5149.
Rouget C. 1879. Sur la contractilité des capillaires sanguins. Compt rend Washington RA, Becher B, Balabanov R, Antel J, Dore-Duff y P. 1996.
acad sci Paris 88:916–918. Expression of the activation marker urokinase plasminogen-activator
Ruiter DJ, Schlingemann RO, Westphal JR, Denijn M, Rietveld FJ, De receptor in cultured human central nervous system microglia.
Waal RM. 1993. Angiogenesis in wound healing and tumor metasta- J Neurosci Res 45:392–399.
sis. Behring Inst Mitt 258–272. Wilhelmus MM, Otte-Höller I, van Triel JJ, Veerhuis R, Maat-Schieman
Saharinen P, Eklund L, Miettinen J, Wirkkala R, Anisimov A, ML, Bu G, et al. 2007. Lipoprotein receptor-related protein-1 medi-
Winderlich M, . 2008. Angiopoietins assemble distinct Tie2 signal- ates amyloid-beta-mediated cell death of cerebrovascular cells. Am J
ling complexes in endothelial cell-cell and cell-matrix contacts. Nat Pathol 171(6):1989–1999.
Cell Biol 10(5):527–537. Winkler EA, Bell RD, Zlokovic BV. 2010. Pericyte specific expression of
Sato TN, Tozawa Y, Deutsch U, Wolburg-Buchholz K, Fujiwara Y, PDGF beta receptor in mouse models with normal and deficient beta
Gendron-Maguire M, et al. 1995. Distinct roles of the receptor tyrosine receptor signalling. Mol Neurodegener 5:32.
kinases Tie-1 and Tie-2 in blood vessel formation. Nature 376:70–74. Winkler EA, Bell RD, Zlokovic BV. 2011. Lack of Smad or Notch leads
Sato Y. 2004. Role of aminopeptidases in angiogenesis. Biol Pharm Bull to a fatal game of brain pericyte hopscotch. Dev Cell 20:279–280.
27(6):772–776. Winkler EA, Bell RD, Zlokovic BV. 2011. Central nervous system peri-
Shepro D, Morel NM. 1993. Pericyte physiology. FASEB J 7:1031–1038. cytes in health and disease. Nat Neurosci 14(11):1398–1405.
Sieczkewicz GJ, Herman IM. 2003. TGF-beta 1 signalling controls retinal Wisniewski HM, Wegiel J, Wang KC, Lach B. 1992. Ultrastructural
pericyte contractile protein expression. Microvasc Res 66:190–196. studies of the cells forming amyloid in the cortical vessel wall in
Sims DE. 1986. The pericyte—A review. Tissue Cell 18:153–174. Alzheimer’s disease. Acta Neuropathol 84(2):117–127.
Sixt M, Engelhardt B, Pausch F, Hallmann R, Wendler O, Sorokin LM. Yamagishi S, Yonekura H, Yamamoto Y. 1999. Vascular endothelial
2001. Endothelial cell laminin isoforms, laminins 8 and 10, play deci- growth factor acts as a pericyte mitogen under hypoxic conditions.
sive roles in T cell recruitment across the blood–brain barrier in exper- Lab Invest 79:501–509.
imental autoimmune encephalomyelitis. J Cell Biol 153:933–946. Zhang ZG, Zhang L, Croll SD, Chopp M. 2002. Angiopoietin-1 reduces
Soriano P. 1994. Abnormal kidney development and hematological disor- cerebral blood vessel leakage and ischemic lesion volume after focal
ders in PDGF beta-receptor mutant mice. Genes Dev 8:1888–1896. cerebral embolic ischemia in mice. Neuroscience 113:683–687.
Stenzel D, Nye E, Nisancioglu M, Adam RH, Yamaguchi Y, Gerhardt Zimmermann KW. 1923. Der feinere Bau der Blutkapillaren. Z Anat
H. 2009. Peripheral mural cell recruitment requires cell-autonomous Entwicklungsgesch 68:29–109.
heparin sulfate. Blood 214:915–924. Zlokovic BV. 2005. Neurovascular mechanisms of Alzheimer’s neurode-
Stewart PA, Wiley MJ. 1981. Developing nervous tissue induces formation generation. Trends Neurosci 28(4):202–208.
of blood-brain barrier characteristics in invading endothelial cells: a study Zlokovic BV. 2008. The blood-brain barrier in health and chronic neuro-
using quail—chick transplantation chimeras. Dev Biol 84:183–192. degenerative disorders. Neuron 57(2):178–201.

108 • M O R P H O L O GY, U LT R A S T RU C T U R E , A N D I D E N T I F I C AT I O N
10.
NG2 CELLS (POLYDENDROCY TES)
Akiko Nishiyama

A B B R E VI AT I O N S and other glial cell populations. They are the primary source
of myelinating and nonmyelinating oligodendrocytes and are
BrdU 5-bromo-2′-deoxyuridine hence often equated with oligodendrocyte progenitor cells
CNP 2′,3-cyclic nucleotide 3′-phosphodiesterase (OPCs), although some questions remain as to whether all
CNS central nervous system polydendrocytes generate oligodendrocytes. Polydendrocytes
Cspg-4 chondroitin sulfate proteoglycan-4 are distributed throughout both gray and white matter and
EAE experimental autoimmune encephalomyelitis are distinct from neural stem/progenitor cells or radial glia.
Ecrg4 esophageal cancer–related gene 4 This chapter summarizes the current knowledge of polyden-
EGF epidermal growth factor drocytes based primarily on morphological observations,
EGFP enhanced green fluorescent protein and highlights some unsolved questions related to their iden-
GFAP fibrillary acidic protein tity and lineage. A more extensive review of their development
GFP green fluorescent protein and electrophysiological properties is provided in chapters 13
GLAST glutamate aspartate transporter and 21.
H2B histone 2B
HDAC histone deacetylase
Hes-5 hairy and enhancer of split-5 2 I D E N T I F I C AT I O N O F
ID2/4 inhibitor of differentiation 2/4 P O LY D E N D R O C Y T E S
MCSP melanoma chondroitin sulfate proteoglycan
MS multiple sclerosis
OPC oligodendrocyte precursor cell 2.1 I M MU N O H I S TO C H E M I C A L
PDGF platelet-derived growth factor I D E N T I FI C AT I O N
Pdgfra alpha receptor for platelet-derived growth
factor 2.1.1 Cell Surface Antigens
PDZ domain postsynaptic density 95/disk large/zonula-
occludens-1 domain Polydendrocytes are identified by their typical multiprocessed
PLP proteolipid protein morphology and the expression of two defining integral
PSA-NCAM polysialylated form of neural cell adhesion plasma membrane antigens, NG2 and Pdgfra (Table 10.1).
molecule Pdgfra belongs to the family of receptor tyrosine kinases with
SVZ subventricular zone a split intracellular tyrosine kinase domain and transduces sig-
YFP yellow fluorescent protein nal when bound by its cognate dimeric ligands, which include
PDGF-AA, PDGF-BB, and PDGF-CC homodimers and
PDGF-AB heterodimers. Pdgfra mediates survival, prolifera-
1 INTRODUCTION tion and migration of polydendrocytes (Andrae et al. 2008;
Vora et al. 2011; see also chapter 22).
Studies conducted over the last three decades have revealed a NG2 is a product of the Cspg-4 (chondroitin sulfate
glial progenitor cell population in the developing and mature proteoglycan-4) gene, also known as the melanoma chon-
mammalian central nervous system (CNS), which are com- droitin sulfate proteoglycan (MCSP) in humans. It exists as
monly known as NG2 cells. The term polydendrocyte is pro- a 300-kDa single membrane-spanning core glycoprotein or as
posed as a formal name for NG2 cells in recognition of their a heterogeneous high molecular weight proteoglycan. NG2
existence as a normal resident cell population in the adult interacts with many extracellular and intracellular proteins
CNS and to match the nomenclature of other macroglial and has been implicated in the regulation of cell migration and
cells. The name reflects their highly branched morphology proliferation (Nishiyama et al. 2009; Stallcup 2002; Sugiarto
and lineal kinship to oligodendrocytes (Nishiyama 2007). et al. 2011; Trotter et al. 2010).
Polydendrocytes are defined as nonvascular cells in the CNS In the oligodendrocyte lineage, Pdgfra and NG2 are highly
parenchyma that express NG2 and the alpha receptor for expressed on proliferating progenitor cells, and the expression
platelet-derived growth factor (Pdgfra). They comprise 2% of these genes is downregulated as the cells differentiate into
to 9% of the cells in the CNS and are distinct from neurons oligodendrocytes (Fig. 10.1). Neither protein is expressed

109
Table 10.1 SPECIFICITY OF COMMONLY USED ANTIGENS EXPRESSED BY POLYDENDROCYTES
ANTIGEN EXPRESSION

NAME DESCRIPTION NSCs PDs ODs ASTROCYTES NEURONS MICROGLIA

NG2 Cell surface – + – – – – On resting microglia


proteoglycan + On some activated
macrophages

Pdgfra Receptor tyrosine – + – – – –


kinase

Olig2 Transcription factor + In some + + Weaker – On most – –

Sox10 Transcription factor – + + – – –

CNPase Cytosolic protein – – On most + on some + – – –

PLP Membrane protein – – On most + on some + – – –

O4 Membrane – + On most (rat) + – – –


glycolipid + on some (mouse)

S100β Cytosolic calcium- – + In some + In some + – –


binding protein

NSC, neural stem cell; OD, oligodendrocyte; PD, polydendrocyte; – not detected; + detected.

exclusively on polydendrocytes, and outside the CNS they are identified as the two most highly enriched in acutely iso-
expressed on cycling lineage committed progenitor cells of the lated glial progenitor cells from human white matter (Sim
mesenchyme and skin. In the CNS, NG2 is also detected on et al. 2006) (see chapter 29). Similarly, Cspg-4 and Pdgfra tran-
vascular mural cells, most commonly on pericytes and smooth scripts were among the five most enriched in polydendrocytes
muscle cells (Nishiyama et al. 2009). Pdgfra is also expressed isolated from P16 mouse brain by immunopanning for Pdgfra,
by vascular cells (Richardson et al. 2011). compared with mature oligodendrocytes (Cahoy et al. 2008).
One caveat to using these cell surface antigens to identify
polydendrocytes is that both NG2 and Pdgfra are sensitive
2.1.2 Transcription Factors
to overfixation with aldehydes (Mori et al. 2009). However,
with careful control of tissue processing conditions, NG2 Oligodendrocyte lineage transcription factors such as Olig2
and Pdgfra have been successfully detected in postmortem and Sox10 are expressed in polydendrocytes and are often
human CNS tissue, where they share many of the character- used in conjunction with the aforementioned cell surface
istics of rodent polydendrocytes (Chang et al. 2000; Jennings antigens for their identification (see Fig. 10.1; Table 10.1).
and Carroll 2010; Reynolds et al. 2002; Wolswijk 1998). In The basic helix-loop-helix transcription factor Olig2 and the
a microarray analysis, the Cspg-4 and Pdgfr transcripts were product of the adjacently located gene Olig1 are implicated

Neuroepithelial NG2 cells Immature


= polydendrocytes Oligodendrocytes
cells oligodendrocytes
germinal zone parenchyma parenchyma

Olig2
Sox10
NG2, Pdgfra
CNP
PLP

Figure 10.1 Relationship Between Polydendrocytes and Oligodendrocyte Lineage Cells. Box indicates polydendrocytes that are process-bearing cells that
express NG2 and Pdgfra. Thick gray lines indicate the duration of expression of the indicated antigens.

110 • NEUROGLIA
in oligodendrocyte specification and differentiation (Peru 2.2.1 NG2 Transgenic Mouse Lines
et al. 2008). Sox10 belongs to the high mobility group tran-
scription factors and is required for terminal differentiation The two transgenic mouse lines in which the expression of
of polydendrocytes into oligodendrocytes (see chapter 43). a fluorescent protein occurs most faithfully in polydendro-
The expression of Sox10 precedes that of Pdgfra, and the onset cytes throughout all stages of development are the NG2-YFP
of Pdgfra expression is dependent on Sox10 (Finzsch et al. knock-in mice (Karram et al. 2008) and the NG2-DsRed
2008). Although Sox10 expression in the CNS is confined to BAC mice (Zhu et al. 2008; Ziskin et al. 2007). In NG2-YFP
the oligodendrocyte lineage, Olig2 is expressed more broadly transgenic mice, despite the loss of one endogenous NG2
in cells fated to become neurons or astrocytes (see Fig. 10.1) allele because of insertion of the YFP expression cassette,
(Ono et al. 2008; Tekki-Kessaris et al. 2001). Within the oli- there is no discernible developmental or behavioral change
godendrocyte lineage, Olig2 is expressed abundantly in the in heterozygous mice. Additional mouse lines that express
nucleus of all polydendrocytes (Ligon et al. 2006). The level GFP (green fluorescence protein) in the majority of poly-
of Olig2 declines after they terminally differentiate into oligo- dendrocytes include Pdgfra-H2B-GFP and Olig2-EGFP
dendrocytes (Kitada and Rowitch 2006), whereas the level of mice (Table 10.2; Fig. 10.2C).
Sox10 is sustained throughout all stages of the oligodendro-
cyte lineage. 2.2.2 Transgenic Mice with
Oligodendrocyte-Specific Promoters
There are transgenic mouse lines that were designed to
2.2 T R A NS G E N I C MO US E L I N E S T H AT
express reporter genes in mature oligodendrocytes but also
E X P R E S S R E P O RT E R P ROT E I N S I N
express them in polydendrocytes. In CNP-EGFP mice that
P O LY D E N D RO C Y T E S
express EGFP (enhanced green fluorescent protein) driven
Immunohistochemical methods of cell identification rely on by a 3.7-kb promoter of the mouse 2′,3′-cyclic nucleotide
the availability of specific antibodies and adequate preserva- 3′-phosphodiesterase (CNP) gene, EGFP is expressed
tion of the antigen through tissue processing. As an alter- robustly in differentiated oligodendrocytes and weakly
native method of imaging and isolating polydendrocytes, in polydendrocytes (Yuan et al. 2002). The regulatory
particularly in live tissue, a number of transgenic mouse sequences of the myelin proteolipid protein (PLP) gene
lines that express various fluorescent reporter genes in NG2 have been used to generate various lines of PLP transgenic
cells have become available (Table 10.2). None of the lines mice (see Table 10.2). Proteolipid protein appears slightly
express the reporter gene exclusively in polydendrocytes, after CNP in oligodendrocyte lineage cells during devel-
but with an accurate knowledge of the pattern of reporter opment (see Fig. 10.1). In mice with less sensitive reporter
expression of each line, these mouse lines can be used to constructs, the reporter is detected primarily in mature oli-
mark polydendrocytes in a certain anatomical region at the godendrocytes, whereas in mice with enhanced sensitivity
desired age. and reporter expression, the reporter is detected in 50% of

Table 10.2 TRANSGENIC MOUSE LINES THAT EXPRESS FLUORESCENT REPORTER IN POLYDENDROCYTES
GENE MODE OF PERCENT OF EXPRESSION IN OTHER
EXPRESSED INSERTION PDs MARKED CNS CELL TYPES REFERENCES

NG2-DsRed DsRed BAC transgenic ~100% Pericytes Zhu et al. 2008


JAX 008241

NG2-YFP YFP Knock-in ~100% Pericytes Karram et al. 2008

Pdgfra-H2BGFP GFP-H2B Knock-in Vascular cells Hamilton et al. 2003


fusion Homozygous lethal JAX 007669

CNP-EGFP EGFP Promoter Mature oligodendrocytes Yuan et al. 2002

PLP-EGFP GFP DsRed Promoter Mature oligodendrocytes Fuss et al. 2000


PLP-DsRed Hirrlinger et al. 2005

PLP-EGFP EGFP Promoter and 50% Mature oligodendrocytes Mallon et al. 2002
(3′UTR) 3c-UTR

Olig2-EGFP EGFP BAC 100% Neural stem cells Gensat Project


Radial glia
Oligodendrocytes

PD, polydendrocyte.

N G 2 C E L L S ( P O LY D E N D R O C Y T E S ) • 111
Figure 10.2 Morphology of Polydendrocytes in the Developing and Mature Rodent Central Nervous System. A,B. Morphology of polydendrocytes
in adult mouse neocortex. (A) and corpus callosum (B) revealed by membrane-anchored EGFP (mGFP) expressed on isolated polydendrocytes in
NG2creER:mTmG double transgenic mice. C. A polydendrocyte in the adult mouse corpus callosum from Olig2-EGFP transgenic mouse (Gensat
Project) immunolabeled for NG2 (red). The majority of the NG2-negative EGFP+ cells are oligodendrocytes. (Inset) A reactive polydendrocyte in
the adult corpus callosum stained for NG2 following a demyelinating lesion. D. Relationship between polydendrocyte processes immunolabeled for
NG2 (green) and processes of a premyelinating oligodendrocyte immunolabeled for DM20/PLP (red) in the hippocampus of a P9 mouse. Arrowheads
indicate close apposition of polydendrocyte and oligodendrocyte processes. E. Relationship between a polydendrocyte immunolabeled for NG2
(red) and mGFP in an isolated astrocyte (green) in the neocortex of a P8 GFAPcreER:mTmG mouse. Only half of the astrocyte is shown. Astrocyte
processes decorate the long slender processes of the polydendrocyte and a nearby blood vessel (bv). Asterisks indicate adjacent cell bodies of the
astrocyte and polydendrocyte. F. Relationship between polydendrocytes (NG2, red) and axon bundles (L1 axonal cell adhesion molecule, green) in a
longitudinal section of a P11 rat spinal cord. Many polydendrocyte processes encircle bundles of L1+ axons (arrows). Asterisk marks the cell body of
a polydendrocyte. G. Relationship between polydendrocytes (EGFP+ NG2+, blue) and radial glia (GLAST+, red) in the ventral forebrain of E18.5
NG2cre:zeg mouse. Polydendrocytes are process-bearing cells that are distinct from GLAST+ radial glia. bv: blood vessel. H. Relationship between
polydendrocytes (EGFP+ Olig2+, arrowheads) and radial glia/immature astrocytes (GLAST+) around the SVZ of a P0 NG2cre:zeg mouse. Olig2
is expressed in some cells with GLAST+ processes that are EGFP-negative (arrows). bv: blood vessels that are EGFP+. LV: lateral ventricle. Scale bar,
50 μm. I.Morphology of a polydendrocyte in the neocortex of a P21 NG2cre:zeg mouse stained for NG2 ( blue) and Aldh1L1 (red). All scale bars
except (H) are 10 μm. (D) Kindly provided by Dr. Dirk Dietrich, University of Bonn. GFAPcreER mice were obtained from Dr. Frank Kirchhoff
(Universitaet des Saarlandes, Germany); (G) modified from Zhu et al. 2008.

112 • NEUROGLIA
polydendrocytes as well as in oligodendrocytes (Mallon
et al. 2002).

2.2.3 Transgenic Mice with


Astrocyte-Specific Promoters
Fluorescent reporter expression has also been detected in
polydendrocytes in transgenic mouse lines that were gen-
erated with the original intent of targeting astrocytes. In
GFAP-EGFP transgenic mice generated with a 2.2-kb human
glial fibrillary acidic protein (GFAP) promoter, some poly-
dendrocytes with the morphological, antigenic, and elec-
trophysiological characteristics of polydendrocytes express
EGFP, although GFAP protein is not detected in those
NG2+ EGFP+ cells (Matthias et al. 2003). Caution must be
exercised when interpreting expression studies based on tran-
scriptional activity, because promoter activation may occur
in the absence of stable protein accumulation. Moreover,
in transgenic mouse lines generated with short promoter
sequences, the level and fidelity of transgene expression varies Figure 10.3 Ultrastructural Profiles of Polydendrocyte
from line to line and is dependent on the site of integration Processes. Pre-embedding immunolabeling for NG2 showing clusters
of multiple thin profiles of NG2-labeled polydendrocyte processes
of the transgene. (arrowheads) in the neuropil outside the SVZ in an adult mouse brain,
suggesting a convoluted nature of the processes. Scale bar, 2 μm.

3 D I S T R I B U T I O N A N D M O R P H O L O GY
O F P O LY D E N D R O C Y T E S the adult CNS varies from 10 to 140/mm2 (reviewed in
Nishiyama 2007). Polydendrocytes constitute 8% to 9% of
total cells in the white matter and 2% to 3% of total cells
3.1 MO R P H O L O GY
in the gray matter (Dawson et al. 2003). The ratio of poly-
In the mature CNS, polydendrocytes characteristically have dendrocytes to oligodendrocytes ranges from 1:1 in the
small, irregularly shaped elongated cell bodies and long slen- rat hippocampus to 1:10 in the cat spinal cord. In a recent
der processes (Peters 2004). The cell body often tapers into study in which simultaneous labeling and quantification of
a primary process at one end of the soma, creating an asym- all glial cells was performed, 99.5% and 100% of all the glial
metry. Although their morphology varies slightly in different cells in the cat and human optic nerve, respectively, could
anatomical regions and different planes of section (Dawson be accounted for as polydendrocytes, oligodendrocytes,
et al. 2003), polydendrocytes in both gray and white matter astrocytes, or microglia ( Jennings and Carroll 2010). Thus,
extend their processes in multiple directions (Fig. 10.2A–C). polydendrocytes do indeed make up the fourth major nor-
Ultrastructurally, polydendrocytes have a pale, mal resident glial population in the mature CNS, and it is
bean-shaped nucleus with an irregular contour and has unlikely that there are additional previously overlooked cell
clumped chromatin beneath the nuclear envelope. The populations.
nucleus is surrounded by relatively light cytoplasm that is
devoid of intermediate filaments (Peters 2004). Their fine
3.3 E A R LY A P P E A R A N C E O F
structure resembles that of previously reported prolifera-
P O LY D E N D RO C Y T E S
tive cells (Carroll et al. 1998; Reyners et al. 1986; Vaughn
1969). Ultrastructurally polydendrocytes most closely In the embryonic forebrain, Pdgfra mRNA+ and Sox10
resemble astrocytes but differ from the latter by their irreg- mRNA+ cells are detected on embryonic day 13.5 (E13.5) in
ularly shaped nuclei and thin mitochondria (Peters 2004). the anterior entopeduncular area (Tekki-Kessaris et al. 2001).
Pre-embedding immunoelectron microscopy performed At this stage NG2 is expressed only in the vasculature. Cells
with anti-NG2 antibody reveals clusters of thin profiles of that express the NG2 and Pdgfra proteins do not appear
labeled processes in the neuropil, indicative of their complex until after E14.5 in the mouse forebrain. They are found in
branching or looping (Fig. 10.3, arrowheads). the parenchyma outside the ventricular zone and do not
express radial glial antigens such as GLAST (see Fig. 10.2G).
Thus, polydendrocytes represent cells that have become
3.2 D I S T R I BU T I O N
committed to a glial lineage and have begun their migration
In the mature CNS, polydendrocytes are uniformly distrib- away from the germinal zones. By E18.5, many of them have
uted throughout the gray and white matter. They appear already acquired several long slender processes. They are to
to be tiled with their processes occupying nonoverlapping be distinguished from Olig2+ cells in the germinal zones
territories. The estimated density of polydendrocytes in that represent a heterogeneous population of progenitor

N G 2 C E L L S ( P O LY D E N D R O C Y T E S ) • 113
cells that give rise to neurons, oligodendrocytes, and astro- function of polydendrocytes in their oligodendrocyte pro-
cytes (Ono et al. 2008) (see Fig. 10.2H). Polydendrocytes genitor capacity.
in the forebrain are generated from three distinct sources,
each of which originates from a distinct domain marked
by its location and transcription factor code (Kessaris et al. 4.2 O L I G O D E N D RO C Y T E S
2006) (see chapter 13). The neocortex is the last forebrain When polydendrocytes terminally differentiate into oligo-
region to be populated by polydendrocytes. By the end of dendrocytes, they lose the expression of NG2 and Pdgfra and
the first postnatal week, polydendrocytes have reached their acquire the expression of oligodendrocyte antigens (see Fig.
peak density, become uniformly distributed, and acquired 10.1). As polydendrocytes differentiate into oligodendrocytes,
a highly branched morphology resembling of those in the the size of the soma and the volume occupied by their processes
mature CNS (see Fig. 10.2I). increase significantly (Kukley et al. 2010). Polydendrocytes
often contact oligodendrocyte processes (see Fig. 10.2D).
However, the functional significance of such contacts remains
4 R E L AT I O N S H I P B ET W E E N unknown.
P O LY D E N D R O C Y T E S A N D OT H E R
CELL TYPES IN THE CENTRAL
N E RVO U S SYS T E M 4.3 A S T RO C Y T E S

Morphological, immunohistochemical, and functional stud- Polydendrocytes are antigenically distinct from protoplasmic
ies have shown that polydendrocytes are distinct from neurons or fibrous astrocytes (Peters 2004) or the velate astrocytes in
and other glial cells. Following is a summary of the distinction the cerebellar cortex (Chan-Palay and Palay 1972). NG2 and
and spatial relationship between polydendrocytes and other Pdgfra are never detected in cells that express GFAP or the
CNS constituents. astrocyte antigen aldehyde dehydrogenase 1 L1 (Aldh1L1),
which was recently discovered by microarray analysis (Cahoy
et al. 2008). Conversely, GFAP or Aldh1L1 proteins are
4.1 N EU RO N S never detected in polydendrocytes (Nishiyama 2007 or 2009;
Polydendrocytes are functionally distinct from neurons, but Richardson et al. 2011; Zhu et al. 2008). However, GFAP tran-
are intimately associated with them. One such mode of inter- scription seems to occur in some polydendrocytes (reviewed
action involves responding to neurotransmitters released in Nishiyama et al. 2009). Although polydendrocytes never
from neuronal axons via ionotropic receptors expressed by express markers of astrocytes, a subpopulation of polyden-
polydendrocytes in both gray and white matter (see chapter drocytes in the embryonic forebrain generates protoplasmic
21). Ultrastructurally, polydendrocyte processes are found astrocytes in a region-specific manner (see section 5.2.2).
apposed to neuronal presynaptic terminals in gray matter and Polydendrocytes can be morphologically distinguished
surrounding unmyelinated axons in white matter. Synaptic from protoplasmic and fibrous astrocytes by their long, slen-
input onto polydendrocytes disappears as they differenti- der processes that are less densely branched in their proximal
ate into oligodendrocytes (De Biase et al. 2010; Kukley et al. regions than those of astrocytes, thus lacking the characteristic
2010). “bushy, fuzz ball appearance” of astrocytes (see Fig. 10.2E) (see
In addition, polydendrocytes in gray matter exist in a sat- chapters 4 and 12). The morphology of polydendrocytes may
ellite position adjacent to neuronal soma (Karram et al. 2008; be more suited for point-to-point communication with other
Mangin et al. 2008), and electron-dense patches have been cellular entities rather than the global homeostatic function of
observed on the adjacent plasma membranes (Peters 2004). astrocytes, such as the clearance of extracellular potassium ions
In myelinated fiber tracts, polydendrocyte processes contact or neurotransmitters that require them to cover a large volume
the nodes of Ranvier (Butt et al. 1999). Although the precise of the neuropil. Although astrocytes extend their endfeet onto
function of such polydendrocyte–axonal contacts remains blood vessels, polydendrocyte processes terminate on the vas-
unknown, they could allow axonally derived signals to reg- cular wall in a small punctum or do not end on the vasculature
ulate polydendrocyte proliferation and differentiation, and at all (Zerlin and Goldman 1997). Polydendrocyte processes
reciprocally polydendrocytes could provide neurotrophic sup- are extensively intertwined with those of astrocytes (see Fig.
port for neurons. 10.2E; Hamilton et al. 2010), suggestive of some functional
It has been debated as to whether polydendrocytes that interaction. Future studies could be directed toward elucidat-
interact with neurons are a distinct postmitotic popula- ing the nature of such interactions.
tion from those that can proliferate and differentiate into
oligodendrocytes. However, the observations that polyden-
4.4 M I C RO G L I A
drocytes can divide while retaining their synaptic inputs
(Kukley et al. 2008) and that polydendrocytes lose their Polydendrocytes are distinct from resting ramified microglia.
synaptic input as they differentiate into oligodendrocytes In the normal developing and mature CNS, NG2 and Pdgfra
(De Biase et al. 2010; Kukley et al. 2010) do not support this are not expressed on resting ramified microglia that express
view. Instead, these observations suggest that such neuron– CD11b and F4/80. Although both cell types have branched
polydendrocyte interaction could be a normal physiological processes, the processes of microglia are barbed with regularly

114 • NEUROGLIA
occurring short side branches, and those of polydendrocytes rat optic nerves could be reprogrammed to become neurons
are longer, smoother, and branch more distally. In response to after a long period in culture (Kondo and Raff 2000). This
various types of insult to the CNS, both polydendrocytes and led to an intensive search by many labs for evidence that poly-
microglia undergo rapid morphological changes characterized dendrocytes are multipotent cells that can generate neurons
by shortening and thickening of their processes. Often the cell as well as oligodendrocyte lineage cells. However, subsequent
bodies of activated polydendrocytes and microglia are seen genetic fate mapping studies described in the following sec-
adjacent to each other, and their processes are intimately inter- tion revealed that polydendrocytes do not generate neurons
twined (Bu et al. 2001; Dawson et al. 2003; Nishiyama et al. under normal physiological conditions, and that their fate in
1997), suggestive of functional interactions between the two the postnatal CNS is restricted to the oligodendrocyte lin-
cell types (Wu et al. 2010). Under certain pathological condi- eage, although some polydendrocytes in some embryonic
tions involving extensive neuronal damage, NG2 is detected brain regions generate astrocytes as well as oligodendrocyte
on a subpopulation of round macrophages that invade the lineage cells.
lesion (Bu et al. 2001; Kucharova et al. 2011).
5.2 G E N ET I C FAT E M A P P I N G O F
4.5 N EU R A L S T E M C E L L S P O LY D E N D RO C Y T E S
There have been conflicting reports about the relationship Mouse genetic tools were developed over the past decade
between polydendrocytes and multipotential neural stem to directly follow the fate of polydendrocytes. This involves
cells and other progenitor cells in the germinal zones, par- Cre-loxP–mediated site-specific recombination to perma-
ticularly those in the subventricular zone (SVZ) around the nently turn on the expression of a reporter gene specifi-
lateral ventricle where neurogenesis persists in adulthood. cally in polydendrocytes and examining the phenotype of
Polydendrocytes are very sparsely scattered in the SVZ, reporter-expressing (reporter+) cells (Fig. 10.4) (Nishiyama
and they are distinct from GFAP+ neural stem cells, transit 2007). Temporal control of reporter activation can be added
amplifying cells, or migrating neuroblasts (Chojnacki et al. to this system by replacing constitutively active Cre with
2011; Komitova et al. 2009; Platel et al. 2009; Richardson tamoxifen-inducible Cre (CreER).
et al. 2011). This is consistent with the lack of evidence that
polydendrocytes give rise to neurons in the olfactory bulb.
5.2.1 Polydendrocytes in the Postnatal Central
The parenchyma outside the germinal zones is more densely
Nervous System Are Committed to the
populated by regularly spaced polydendrocytes, giving the
Oligodendrocyte Lineage
appearance that polydendrocytes are lined up at the outer
border of the SVZ. In the SVZ along the dorsal wall of the In mice that were double transgenic for NG2-cre, which
lateral ventricles, polydendrocytes appear to be more abun- expresses constitutively active Cre in NG2-expressing cells,
dant, and they may provide a source of new oligodendrocyte and a Cre reporter, the majority of oligodendrocytes in both
lineage cells that populate the corpus callosum (Etxeberria gray and white matter expressed the reporter (Zhu et al.
et al. 2010). 2008). This provided the first direct evidence that polyden-
drocytes generate oligodendrocytes. Several ensuing studies
used transgenic mice that express CreER regulated by the gene
5 T H E FAT E O F N G 2 C E L L S encoding Pdgfra (Kang et al. 2010; Rivers et al. 2008), NG2
(Zhu et al. 2011), or Olig2 (Dimou et al. 2008) and exam-
ined the fate of polydendrocytes at different postnatal time
5.1 H I S TO R I C A L P E R S P E C T I V E
Our understanding of the fate of polydendrocytes has evolved The fate of polydendrocytes in the forebrain
over the past three decades after many twists and turns. prenatal postnatal
A2B5+ cells isolated from rodent optic nerves generate oli-
godendrocytes when grown in serum-free medium and stel- DG oligodendrocytes
late GFAP+ type 2 astrocytes when grown in the presence of WM
serum (Raff et al. 1983; Stallcup and Beasley 1987). This led polydendrocytes
to the hypothesis that polydendrocytes represent bipotential
glial progenitor cells. However, numerous attempts made in VG astrocytes
the 1990s failed to unequivocally demonstrate that polyden-
drocytes can give rise to astrocytes as well as oligodendrocyte Figure 10.4 The Fate of Polydendrocytes in Different Forebrain
in vivo. Thus, the concept of bipotential glial progenitor cells Regions. Right. Postnatal brain. Left. Embryonic brain. Polydendrocytes
was gradually dismissed as a cell culture artifact, and the cells in all the regions of the postnatal forebrain generate only polydendro-
began to be called oligodendrocyte precursor cells (OPCs) by cytes or oligodendrocytes. Polydendrocytes in the ventral gray matter
the end of the 1990s. (VG) of the embryonic brain consist of those that generate astrocytes
(dark gray) and those that generate oligodendrocyte lineage cells
Just as the debate on their oligodendrocyte-astrocyte (oligodendrocytes and polydendrocytes). Polydendrocytes in the embry-
bipotentiality was winding down, another controversy was onic white matter (WM) and dorsal gray matter (DG) generate only
sparked by the discovery that polydendrocytes from perinatal polydendrocytes and oligodendrocytes.

N G 2 C E L L S ( P O LY D E N D R O C Y T E S ) • 115
points. Collectively, the common findings from these studies A significant number of oligodendrocytes continue to be
are that the vast majority of polydendrocytes in the postnatal generated in the adult CNS. It has been estimated that 17% to
CNS generate only oligodendrocyte lineage cells, and their 30% of oligodendrocytes are generated de novo from polyden-
progeny are either polydendrocytes or oligodendrocytes (Fig. drocytes within a period of 2 to 3 months in young adult mice
10.5). There are small variances among these reports regarding (Rivers et al. 2008; Zhu et al. 2011). Whether they replace
whether other cell lineages are derived from polydendrocytes existing or degenerating oligodendrocytes or contribute to a
in the postnatal brain. Rivers et al. (2008) observed a small gradual rise in the total number of oligodendrocytes (Peters
number of neurons in the piriform cortex, whereas the other and Sethares 2004) remains to be determined. It is also unclear
studies did not find any neuronal progeny of polydendro- whether all polydendrocytes are endowed with the ability to
cytes (Dimou et al. 2008; Kang et al. 2010; Zhu et al. 2011). differentiate into oligodendrocytes, or there is a subpopulation
Dimou et al. (2008) observed a small number of astrocytes that permanently remains as polydendrocytes throughout life
that expressed the Cre reporter, whereas astrocyte differentia- and is incapable of generating oligodendrocytes.
tion was not detected from postnatal polydendrocytes in the
other studies (Kang et al. 2010; Rivers et al. 2008; Zhu et al.
5.2.2 A Subpopulation of Polydendrocytes in the
2011). The reason for this discrepancy is not clear at present,
Embryonic Brain Generates Astrocytes
but the majority of evidence indicates that polydendrocytes in
the postnatal forebrain are restricted to the oligodendrocyte Fate mapping studies using constitutively active NG2-cre mice
lineage under normal physiological conditions. revealed that more than 40% of protoplasmic astrocytes in the
The rate of oligodendrocyte differentiation from poly- ventral forebrain but not those in the white matter or dorsal
dendrocytes is higher in white than gray matter and declines forebrain are generated from polydendrocytes (Zhu et al.
with age (Dimou et al. 2008; Kang et al. 2010; Rivers et al. 2008). Using NG2-creER mice it was found that astrocyte dif-
2008; Zhu et al. 2011). Single polydendrocytes at all postnatal ferentiation from polydendrocytes occurs only in the embry-
stages generate two daughter oligodendrocytes, two daughter onic brain and does not occur after birth (see Fig. 10.5) (Zhu
polydendrocytes, or one of each, but those in the mature brain et al. 2011). However, contrary to the initial oligodendrocyte–
generate two daughter polydendrocytes more frequently than astrocyte bipotential glial progenitor theory based on optic
oligodendrocytes (Zhu et al. 2011). Further studies are needed nerve cultures, polydendrocytes in the white matter do not
to determine the precise mechanism of symmetric and asym- generate astrocytes at all ages. Single polydendrocytes in the
metric division of polydendrocytes and their self-renewal abil- embryonic ventral gray matter generate either all astrocytes or
ity (see section 7.2) (Sugiarto et al. 2011). all oligodendrocyte lineage cells, and clusters of polydendro-
cyte progeny that contain both astrocytes and oligodendro-
cyte lineage cells have not been found. Thus, the divergence
Inducible fate mapping of polydendrocytes of the astrocyte and oligodendrocyte fates of polydendrocytes
must occur early, and the embryonic ventral forebrain con-
tains an intermingled population of polydendrocytes that are
fated to become astrocytes and those committed to become
NG2creER:YFP oligodendrocytes (see Fig. 10.5). Olig2 is initially expressed in
double tg mouse polydendrocytes, but is downregulated as they develop into
NG2-creER mouse
astrocytes. The mechanism why only polydendrocytes in ven-
NG2 creER
tral and not dorsal forebrain generate astrocytes is not known.
40HT
NG2+(immunolabeled)
NG2– YFP+(progency of NG2+ cells)
Polydendrocytes originating from different domains of the
NG2+/YFP+ embryonic germinal zone may not be equivalent in their astro-
CA p YFP gliogenic potential.

e.g. ROSA-YFP mouse 5.2.3 Polydendrocytes Do Not Contribute


CA p Stop-pA YFP to Neurogenesis
NG2+/YFP+ NG2–/YFP+
loxP
polydendrocyte oligodendrocyte Initially different findings were reported regarding the neu-
rogenic fate of polydendrocytes. Rivers et al. (2008) found
Figure 10.5 Fate Mapping of Polydendrocytes Using Inducible Cre-loxP that a small subpopulation of Pdgfra+ cells in the piriform
Recombination. Left. A double transgenic mouse line generated by cross- cortex generated cells that morphologically resembled pro-
ing a Cre driver (e.g., NG2-creER) and a Cre reporter (e.g., ROSA-YFP).
The Cre driver line expresses inducible Cre (CreER) under the control of a jection neurons, and a similar observation was made using
polydendrocyte-specific promoter. The Cre reporter contains a constitu- PLP-creER mice (Guo et al. 2010), although the PLP pro-
tively active promoter (CAp) followed by a loxP-flanked transcriptional moter can be activated in some neurons (Miller et al. 2009).
stop sequence, and the cDNA encoding YFP. When Cre is activated By contrast, other studies did not to find any neuronal prog-
by 4-hydroxytamoxifen (4OHT), the sequence between the two target eny of polydendrocytes (Kang et al. 2010; Zhu et al. 2008,
loxP sites is excised and YFP expression is permanently turned on. Right.
Examples of Cre activation in polydendrocyte (NG2+ and YFP+) and its 2011). Because additional studies using Pdgfra-creER have
progeny that no longer expresses NG2 (oligodendrocyte). Modified from not revealed a significant number of neuronal progeny of
Nishiyama 2007. polydendrocytes (Tripathi et al. 2010; Zawadzka et al. 2010),

116 • NEUROGLIA
the current consensus is that polydendrocytes do not contrib- astrocyte precursor cells and polydendrocytes that contribute
ute to the generation of new neurons under normal physiolog- to the glial scar.
ical conditions (Richardson et al. 2011).

7 P O LY D E N D R O C Y T E S P R O VI D E
6 P R O L I F E R AT I O N A N D R E S P O N S E O F AN ENDOGENOUS SOURCE OF
P O LY D E N D R O C Y T E S TO I N J U RY R E M Y E L I N AT I N G C E L L S

Polydendrocytes continue to cycle throughout life and 7.1 P O LY D E N D RO C Y T E S P RO L I FE R AT E A N D


comprise 70% to 90% of the proliferating population in the D I F F E R E N T I AT E I N TO R E MY E L I NAT I N G
adult CNS (Horner et al. 2000; Polito and Reynolds 2005). C E L L S A F T E R D E MY E L I NAT I O N
This is consistent with the earlier reports on the existence
of a radiosensitive proliferative cell population in the adult Remyelination is often incomplete in multiple sclerosis (MS)
CNS (Carroll et al. 1998; Reyners et al. 1986). Throughout and other types of CNS insults, such as spinal cord injury,
all stages of postnatal development, 50% or more polyden- that are accompanied by extensive demyelination. Thus strate-
drocytes remain in cell cycle. The cell cycle time increases gies to improve myelin repair should include identifying the
from 2 to 3 days in the early postnatal brain to greater than endogenous source of remyelinating cells and enhancing their
70 days in the adult brain (Kukley et al. 2008; Psachoulia et ability to produce more oligodendrocytes and myelin. Before
al. 2009), and there appears to be heterogeneity in the cell the recognition of polydendrocytes as a normal resident glial
cycle time among polydendrocytes in the same anatomical cell population, unidentified “small glial progenitor cells”
region (Polito and Reynolds 2005). In addition to various were seen to proliferate around experimentally demyelinated
factors that are known to regulate polydendrocyte prolifera- lesions in the corpus callosum and optic nerve and generate
tion (Franklin and ffrench-Constant 2008), voluntary exer- remyelinating cells (Carroll et al. 1998; Gensert and Goldman
cise precipitates their cell cycle exit in the neocortex (Simon 1997). Subsequently, proliferating cells were identified as
et al. 2011). Polydendrocytes also have been implicated as the NG2-expressing polydendrocytes in an acutely demyelinated
cell of origin of glial tumors (Liu et al. 2011; Persson et al. lesion in the rat spinal cord (Keirstead et al. 1998).
2010). Oligodendrocytes that are likely to have been gener-
Polydendrocytes undergo increased proliferation in ated from proliferated polydendrocytes appear in acute and
response to a variety of insults to the CNS. They proliferate chronic experimentally demyelinated lesions (Mason et al.
robustly in response to changes in the density of myelin or 2004; Watanabe et al. 2002). Genetic fate mapping studies
oligodendrocytes, and their numbers can increase severalfold using Pdgfra-creER have provided more direct evidence that
(Nishiyama 2007). By contrast, the total number of mature polydendrocytes in the spinal cord generate new oligodendro-
oligodendrocytes appears to be under tighter regulation, and cytes that remyelinate acute chemically demyelinated lesions
increased polydendrocyte numbers are often not accompanied (Zawadzka et al. 2010) or demyelinated lesions in experimen-
by a corresponding increase in oligodendrocyte numbers. Thus, tal autoimmune encephalomyelitis (EAE), which is often used
polydendrocytes could be serving as a buffer for the homeo- as a rodent model of inflammatory demyelinating diseases
static regulation of oligodendrocyte density. Polydendrocytes such as MS (Tripathi et al. 2010). In addition to generating
also dramatically alter their morphology in response to a wide oligodendrocytes, polydendrocytes in astrocyte-depleted
variety of insults, which is often seen as shortening and thick- areas of acute chemically demyelinated spinal cord also gen-
ening of their processes and a strong upregulation of NG2 erate remyelinating Schwann cells (Zawadzka et al. 2010).
expression (see Fig. 10.2C, inset), although the morphology However, neither Schwann cell remyelination nor Schwann
varies with the nature of the insult. cell differentiation from polydendrocytes has been observed
There has been a considerable debate as to whether in the forebrain.
polydendrocytes contribute to reactive astrocytes. Earlier
studies that used pulse-chase labeling with BrdU (5-bromo-
7.2 S E L F-R E N EWA L O F P O LY D E N D RO C Y T E S
2′-deoxyuridine) led to claims that reactive astrocytes are
A F T E R R E MY E L I NAT I O N
generated from proliferated polydendrocytes. However,
more recent genetic fate mapping studies did not reveal a For polydendrocytes to provide a continued source of remy-
significant number of reactive astrocytes that were derived elinating cells, they must be able to replenish themselves.
from polydendrocytes (Dimou et al. 2008; Komitova et al. Studies to determine whether polydendrocytes become
2011; Tripathi et al. 2010; Zawadzka et al. 2010). By contrast, depleted after successful remyelination have yielded con-
Tatsumi et al. (2008) observed Cre reporter+ astrocytes fol- flicting results. Some suggest that polydendrocytes become
lowing cryoinjury in the brain. Olig2-fate mapping is com- depleted (Keirstead et al. 1998; Mason et al. 2004), whereas
plicated by the fact that Olig2 is expressed by some reactive others indicate efficient repopulation from neighboring cells
astrocytes (Chen et al. 2008). Further studies are needed to (Chari and Blakemore 2002) or efficient remyelination of
determine whether the extent of astrocyte differentiation repeated demyelinating lesions created after a 10-week inter-
from polydendrocytes differs in response to different types val (Penderis et al. 2003). The conflicting observations may
of injury and the precise relationship between NG2-negative reflect the slow kinetics of polydendrocyte proliferation and

N G 2 C E L L S ( P O LY D E N D R O C Y T E S ) • 117
differentiation after myelin repair has been achieved in adult by proteasome-mediated degradation of cyclin D1 and D3
CNS. Repopulation of differentiation-competent polyden- (Kujuro et al. 2010).
drocytes may not occur efficiently if there is a chronic demand Cell extrinsic mechanisms also play a major role in
for remyelinating cells. the decline of remyelination efficiency with age. The level
of growth factors necessary for polydendrocyte recruit-
ment is not significantly altered with age (Franklin and
7.3 P O LY D E N D RO C Y T E S A N D N EU R A L
ffrench-Constant 2008). However, a recent parabiosis
S T E M C E L L S I N MY E L I N R E PA I R
experiment indicates that macrophages from young animals
In the normal adult brain, immature cells in the SVZ do can stimulate polydendrocyte proliferation and differentia-
not significantly contribute to oligodendrocyte production tion and significantly promote remyelination in old animals
in the corpus callosum (Marshall et al. 2003). However, (Ruckh et al. 2012).
when the corpus callosum is demyelinated, increased
migration of proliferated cells from the adult SVZ into the 7.4.2 Gray Matter Versus White Matter
lesioned corpus callosum has been observed (Menn et al.
2006). The polysialylated form of neural cell adhesion mol- Genetic fate mapping has shown that polydendrocytes in the
ecule (PSA-NCAM) is expressed on such migrating cells adult white matter differentiate into oligodendrocytes at a
(Nait-Oumesmar et al. 2008). Proliferating GFAP+ type B greater rate than those in the gray matter (Dimou et al. 2008;
cells have been suggested as a source of remyelinating cells Kang et al. 2010; Rivers et al. 2008; Zhu et al. 2011). Multiple
in the corpus callosum (Menn et al. 2006). Enhancing epi- sclerosis affects not only white matter but also gray matter,
dermal growth factor (EGF) signal stimulates their recruit- and recently extensive cortical remyelination was observed
ment, although reports differ as to whether EGF enhances in patients with chronic MS (Albert et al. 2007) (see chap-
their differentiation into myelinating oligodendrocytes ter 61). Further studies are needed to determine if there are
(Aguirre et al. 2007; Gonzalez-Perez et al. 2009; Ivkovic differences in the ability of polydendrocytes in white and gray
et al. 2008). Further quantitative studies are needed to deter- matter to generate myelinating oligodendrocytes under nor-
mine the relative contribution of local polydendrocytes and mal and demyelinated conditions.
SVZ-derived cells toward remyelination. Identification of
the most efficient endogenous source of remyelinating cells
will facilitate the design of strategies to promote myelin 8 S U M M A RY A N D P E R S P E C T I VE S
repair.
It has now become widely accepted that polydendrocytes repre-
sent a fourth resident glial cell population in the normal CNS.
7.4 FAC TO R S T H AT I N F LU E N C E
Polydendrocytes in the postnatal CNS generate only oligo-
R E MY E L I NAT I O N E FFI C I E N C Y FRO M
dendrocyte lineage cells and are often equated with oligoden-
P O LY D E N D RO C Y T E P RO G E N Y
drocyte precursor cells, although it is not known whether all
A number of studies have identified cell intrinsic and extrin- polydendrocytes differentiate into oligodendrocytes. During
sic mechanisms that promote oligodendrocyte differentiation embryonic development, a subpopulation of polydendrocytes
and are discussed in many excellent review articles (Chong in the ventral forebrain, which are distinct from radial glia,
and Chan 2010; Franklin and ffrench-Constant 2008; Peru downregulates oligodendrocyte lineage antigens and differ-
et al. 2008) (see chapters 13 and 57). A sampling of some new entiates into protoplasmic astrocytes. Polydendrocytes in the
developments is given in the following. postnatal CNS that receive synaptic inputs are not a separate
terminally differentiated cell population, as previously hypoth-
esized, but are capable of proliferating and differentiating into
7.4.1 Old and Young Brain
oligodendrocytes.
The efficiency of remyelination declines with age. In old ani- Why is there a need for the adult brain to maintain a popula-
mals, remyelination occurs, but the rate of polydendrocyte tion that can only generate oligodendrocytes? The mammalian
accumulation in the lesion and their differentiation into oli- CNS has become critically dependent on proper myelination
godendrocytes is decreased (Franklin and ffrench-Constant of its axons and may have developed a cytoarchitecture that
2008). Both cell-intrinsic and -extrinsic mechanisms have ensures the maintenance of the correct ratio of oligodendro-
been implicated in the age-dependent decline of remyelination cytes to axons. The rapid proliferation of polydendrocytes that
efficiency. Intrinsically, an age-dependent decline in histone occurs in response to deviation from the normal number of oli-
deacetylase (HDAC) activity causes de-repression of the tran- godendrocytes or the amount of myelin may be a part of the
scription of differentiation inhibitors such as the Sox2, Hes5, homeostatic mechanism. It is paradoxical that demyelinated
and ID2/4, and compromises the ability of polydendrocytes lesions in MS and spinal cord injury are often incompletely
to differentiate into remyelinating oligodendrocytes (Shen et repaired. Polydendrocytes, together with astrocytes, may have
al. 2008; Ye et al. 2009). Cell senescence may also contribute evolved in the CNS to carry out the role of nonmyelinating
to reduced remyelination efficiency. Polydendrocytes in the Schwann cells that coexist with myelinating Schwann cells in
aged mouse brain express the inducer of senescence esopha- the peripheral nervous system. It is possible that polydendro-
geal cancer–related gene 4 (Ecrg4), which causes G1 arrest cytes retain some degree of lineage plasticity and can alter their

118 • NEUROGLIA
fate in response to manipulation of key regulatory switches. De Biase, LM, Nishiyama A, Bergles DE. 2010. Excitability and synap-
Future studies may be directed toward understanding the fun- tic communication within the oligodendrocyte lineage. J Neurosci
30:3600–3611.
damental mechanisms that regulate their fate and differentia- Dimou L, Simon C, Kirchhoff F, Takebayashi H, Gotz M. 2008. Progeny
tion and their role in the neural network. of Olig2-expressing progenitors in the gray and white matter of the
adult mouse cerebral cortex. J Neurosci 28:10434–10442.
Etxeberria A, Mangin JM, Aguirre A, Gallo V. 2010. Adult-born SVZ
AC K N OW L E D G M E N T S progenitors receive transient synapses during remyelination in corpus
callosum. Nat Neurosci 13:287–289.
The author thanks Dr. Dirk Dietrich (University of Bonn) Finzsch M, Stolt CC, Lommes P, Wegner M. 2008. Sox9 and Sox10
influence survival and migration of oligodendrocyte precursors
for providing the image shown in Fig. 10.2D, Dr. Frank in the spinal cord by regulating PDGF receptor alpha expression.
Kirchhoff (Universitaet des Saarlandes) for his generous gift Development 135:637–646.
of GFAPcreER mice, and David Serwanski for the electron Franklin RJ, ffrench-Constant C. 2008. Remyelination in the CNS:
micrograph in Fig. 10.3. The author also thanks graduate stu- from biology to therapy. Nat Rev Neurosci 9:839–855.
dents Bobby Hill, Jelena Medved, Kiran Patel, Alex Reiss, Fuss B, Mallon B, Phan T, Ohlemeyer C, Kirchhoff F, Nishiyama A,
et al. 2000. Purification and analysis of in vivo-differentiated oli-
and Hao Zuo for their helpful discussions and tissue prepara- godendrocytes expressing the green fluorescent protein. Dev Biol
tions that were used to generate the figures. The author regrets 218:259–274.
that space limitation did not permit the inclusion of many Gensert JM, Goldman JE. 1997. Endogenous progenitors remyelinate
important references that are not cited here. This work was demyelinated axons in the adult CNS. Neuron 19:197–203.
supported by funds from the NIH and the National Multiple Gonzalez-Perez O, Romero-Rodriguez R, Soriano-Navarro M,
Garcia-Verdugo JM, Alvarez-Buylla A. 2009. Epidermal growth
Sclerosis Society. factor induces the progeny of subventricular zone type B cells to
migrate and differentiate into oligodendrocytes. Stem Cells 27:
2032–2043.
REFERENCES Guo F, Maeda Y, Ma J, Xu J, Horiuchi M, Miers L, et al. 2010. Pyramidal
neurons are generated from oligodendroglial progenitor cells in adult
Aguirre A, Dupree JL, Mangin, JM, Gallo V. 2007. A functional role piriform cortex. J Neurosci 30:12036–12049.
for EGFR signaling in myelination and remyelination. Nat Neurosci Hamilton N, Vayro S, Wigley R, Butt AM. 2010. Axons and astrocytes
10:990–1002. release ATP and glutamate to evoke calcium signals in NG2-glia.
Albert M, Antel J, Bruck W, Stadelmann C. 2007. Extensive corti- Glia 58:66–79.
cal remyelination in patients with chronic multiple sclerosis. Brain Hamilton TG, Klinghoffer RA, Corrin PD, Soriano P. 2003. Evolutionary
Pathol 17:129–138. divergence of platelet-derived growth factor alpha receptor signaling
Andrae J, Gallini R, Betsholtz C. 2008. Role of platelet-derived growth mechanisms. Mol Cell Biol 23:4013–4025.
factors in physiology and medicine. Genes Dev 22:1276–1312. Hirrlinger PG, Scheller A, Braun C, Quintela-Schneider M, Fuss B,
Bu J, Akhtar N, Nishiyama A. 2001. Transient expression of the NG2 Hirrlinger J, et al. 2005. Expression of reef coral fluorescent proteins
proteoglycan by a subpopulation of activated macrophages in an exci- in the central nervous system of transgenic mice. Mol Cell Neurosci
totoxic hippocampal lesion. Glia 34:296–310. 30:291–303.
Butt AM, Duncan A, Hornby MF, Kirvell SL, Hunter A, Levine JM, Horner PJ, Power AE, Kempermann G, Kuhn HG, Palmer TD,
Berry M. 1999. Cells expressing the NG2 antigen contact nodes of Winkler J, et al. 2000. Proliferation and differentiation of progen-
Ranvier in adult CNS white matter. Glia 26:84–91. itor cells throughout the intact adult rat spinal cord. J Neurosci
Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson 20:2218–2228.
KS, et al. 2008. A transcriptome database for astrocytes, neurons, Ivkovic S, Canoll P, Goldman JE. 2008. Constitutive EGFR signaling in
and oligodendrocytes: a new resource for understanding brain devel- oligodendrocyte progenitors leads to diff use hyperplasia in postnatal
opment and function. J Neurosci 28:264–278. white matter. J Neurosci 28:914–922.
Carroll WM, Jennings AR, Ironside LJ. 1998. Identification of the Jennings A, Carroll W. 2010. Quantification of oligodendrocyte progen-
adult resting progenitor cell by autoradiographic tracking of oligo- itor cells in human and cat optic nerve: implications for endogenous
dendrocyte precursors in experimental CNS demyelination. Brain repair in multiple sclerosis. Glia 58:1425–1436.
121:293–302. Kang SH, Fukaya M, Yang JK, Rothstein JD, Bergles DE. 2010. NG2+
Chang A, Nishiyama A, Peterson J, Prineas J, Trapp BD. 2000. CNS glial progenitors remain committed to the oligodendrocyte
NG2-positive oligodendrocyte progenitor cells in adult human brain lineage in postnatal life and following neurodegeneration. Neuron
and multiple sclerosis lesions. J Neurosci 20:6404–6412. 68:668–681.
Chan-Palay V, Palay SL. 1972. The form of velate astrocytes in the cere- Karram K, Goebbels S, Schwab M, Jennissen K, Seifert G, Steinhauser
bellar cortex of monkey and rat: high voltage electron microscopy of C, et al. 2008. NG2-expressing cells in the nervous system revealed by
rapid Golgi preparations. Z Anat Entwickl-Gesch 138:1–19. the NG2-EYFP-knockin mouse. Genesis 46:743–757.
Chari DM, Blakemore WF. 2002. Efficient recolonisation of Keirstead HS, Levine JM, Blakemore WF. 1998. Response of the oligo-
progenitor-depleted areas of the CNS by adult oligodendrocyte pro- dendrocyte progenitor cell population (defined by NG2 labelling) to
genitor cells. Glia 37:307–313. demyelination of the adult spinal cord. Glia 22:161–170.
Chen Y, Miles DK, Hoang T, Shi J, Hurlock E, Kernie SG, et al. 2008. Kessaris N, Fogarty M, Iannarelli P, Grist M, Wegner M, Richardson
The basic helix-loop-helix transcription factor olig2 is critical for WD. 2006. Competing waves of oligodendrocytes in the forebrain
reactive astrocyte proliferation after cortical injury. J Neurosci and postnatal elimination of an embryonic lineage. Nat Neurosci
28:10983–10989. 9:173–179.
Chojnacki A, Mak G, Weiss S. 2011. PDGFR alpha expression distin- Kitada M, Rowitch DH. 2006. Transcription factor co-expression pat-
guishes GFAP-expressing neural stem cells from PDGF-responsive terns indicate heterogeneity of oligodendroglial subpopulations in
neural precursors in the adult periventricular area. J Neurosci adult spinal cord. Glia 54:35–46.
31:9503–9512. Komitova M, Zhu X, Serwanski DR, Nishiyama A. 2009. NG2 cells are
Chong SY, Chan JR. 2010. Tapping into the glial reservoir: cells commit- distinct from neurogenic cells in the postnatal mouse subventricular
ted to remaining uncommitted. J Cell Biol 188:305–312. zone. J Comp Neurol 512:702–716.

N G 2 C E L L S ( P O LY D E N D R O C Y T E S ) • 119
Komitova M, Serwanski DR, Nishiyama A. 2011. NG2 cells are not a Penderis J, Shields SA, Franklin RJ. 2003. Impaired remyelination and
major source of reactive astrocytes in neocortical stab wound. Glia depletion of oligodendrocyte progenitors does not occur following
59:800–809. repeated episodes of focal demyelination in the rat central nervous
Kondo T, Raff M. 2000. Oligodendrocyte precursor cells repro- system. Brain 126:1382–1391.
grammed to become multipotential CNS stem cells. Science 289: Persson AI, Petritsch C, Swartling FJ, Itsara M, Sim FJ, Auvergne R,
1754–1757. et al. 2010. Non-stem cell origin for oligodendroglioma. Cancer Cell
Kucharova K, Chang Y, Boor A, Yong VW, Stallcup WB. 2011. Reduced 18:669–682.
inflammation accompanies diminished myelin damage and repair in Peru RL, Mandrycky N, Nait-Oumesmar B, Lu QR. 2008. Paving the
the NG2 null mouse spinal cord. J Neuroinflammation 8:158. axonal highway: from stem cells to myelin repair. Stem Cell Rev
Kujuro Y, Suzuki N, Kondo T. 2010. Esophageal cancer-related gene 4 is 4:304–318.
a secreted inducer of cell senescence expressed by aged CNS precursor Peters A. 2004. A fourth type of neuroglial cell in the adult central ner-
cells. Proc Natl Acad Sci U S A 107:8259–8264. vous system. J Neurocytol 33:345–357.
Kukley M, Kiladze M, Tognatta R, Hans M, Swandulla D, Schramm J, et al. Peters A, Sethares C. 2004. Oligodendrocytes, their progenitors and
2008. Glial cells are born with synapses. FASEB J 22:2957–2969. other neuroglial cells in the aging primate cerebral cortex. Cereb
Kukley M, Nishiyama A, Dietrich D. 2010. The fate of synaptic input Cortex 14:995–1007.
to NG2 glial cells: neurons specifically downregulate transmit- Platel JC, Gordon V, Heintz T, Bordey A. 2009. GFAP-GFP neural
ter release onto differentiating oligodendroglial cells. J Neurosci progenitors are antigenically homogeneous and anchored in their
30:8320–8331. enclosed mosaic niche. Glia 57:66–78.
Ligon KL, Kesari S, Kitada M, Sun T, Arnett HA, Alberta JA, et al. Polito A, Reynolds R. 2005. NG2-expressing cells as oligodendrocyte
2006. Development of NG2 neural progenitor cells requires Olig progenitors in the normal and demyelinated adult central nervous
gene function. Proc Natl Acad Sci U S A 103:7853–7858. system. J Anat 207:707–716.
Liu C, Sage JC, Miller MR, Verhaak RG, Hippenmeyer S, Vogel H, et al. Psachoulia K, Jamen F, Young KM, Richardson WD. 2009. Cell cycle
2011 Mosaic analysis with double markers reveals tumor cell of origin dynamics of NG2 cells in the postnatal and ageing brain. Neuron
in glioma. Cell 146:209–221. Glia Biol 5:57–67.
Mallon BS, Shick HE, Kidd GJ, Macklin WB. 2002. Proteolipid pro- Raff MC, Miller RH, Noble M. 1983. A glial progenitor cell that devel-
moter activity distinguishes two populations of NG2-positive cells ops in vitro into an astrocyte or an oligodendrocyte depending on
throughout neonatal cortical development. J Neurosci 22:876–885. culture medium. Nature 303:390–396.
Mangin JM, Kunze A, Chittajallu R, Gallo V. 2008. Satellite NG2 pro- Reyners H, Gianfelici de Reyners E, Regniers L, Maisin JR. 1986. A glial
genitor cells share common glutamatergic inputs with associated progenitor cell in the cerebral cortex of the adult rat. J Neurocytol
interneurons in the mouse dentate gyrus. J Neurosci 28:7610–7623. 15:53–61.
Marshall CA, Suzuki SO, Goldman JE. 2003. Gliogenic and neurogenic Reynolds R, Dawson M, Papadopoulos D, Polito A, Di Bello IC,
progenitors of the subventricular zone: who are they, where did they Pham-Dinh D, et al. 2002. The response of NG2-expressing oligo-
come from, and where are they going? Glia 43:52–61. dendrocyte progenitors to demyelination in MOG-EAE and MS.
Mason JL, Toews A, Hostettler JD, Morell P, Suzuki K, Goldman JE, J Neurocytol 31:523–536.
et al. 2004. Oligodendrocytes and progenitors become progres- Richardson WD, Young KM, Tripathi RB, McKenzie I. 2011.
sively depleted within chronically demyelinated lesions. Am J Pathol NG2-glia as multipotent neural stem cells: fact or fantasy? Neuron
164:1673–1682. 70:661–673.
Matthias K, Kirchhoff F, Seifert G, Huttmann K, Matyash M, Rivers LE, Young KM, Rizzi M, Jamen F, Psachoulia K, Wade A, et al. 2008.
Kettenmann H, et al. 2003. Segregated expression of AMPA-type PDGFRA/NG2 glia generate myelinating oligodendrocytes and piri-
glutamate receptors and glutamate transporters defines distinct form projection neurons in adult mice. Nat Neurosci 11:1392–1401.
astrocyte populations in the mouse hippocampus. J Neurosci Ruckh JM, Zhao JW, Shadrach JL, van Wijngaarden P, Rao TN, Wagers
23:1750–1758. AJ, et al. 2012. Rejuvenation of regeneration in the aging central ner-
Menn B, Garcia-Verdugo JM, Yaschine C, Gonzalez-Perez O, Rowitch vous system. Cell Stem Cell 10:96–103.
D Alvarez-Buylla A. 2006. Origin of oligodendrocytes in the subven- Shen S, Sandoval J, Swiss VA, Li J, Dupree J, Franklin RJ, et al. 2008.
tricular zone of the adult brain. J Neurosci 26:7907–7918. Age-dependent epigenetic control of differentiation inhibitors is
Miller MJ, Kangas CD, Macklin WB. 2009. Neuronal expression of the critical for remyelination efficiency. Nat Neurosci 11:1024–1034.
proteolipid protein gene in the medulla of the mouse. J Neurosci Res Sim FJ, Lang JK, Waldau B, Roy NS, Schwartz TE, Pilcher WH, et al.
87:2842–2853. 2006. Complementary patterns of gene expression by human oligo-
Mori T, Wakabayashi T, Takamori Y, Kitaya K, Yamada H. 2009. dendrocyte progenitors and their environment predict determinants of
Phenotype analysis and quantification of proliferating cells in the progenitor maintenance and differentiation. Ann Neurol 59:763–779.
cortical gray matter of the adult rat. Acta Histochem Cytochem Simon C, Gotz M, Dimou L. 2011. Progenitors in the adult cerebral cor-
42:1–8. tex: cell cycle properties and regulation by physiological stimuli and
Nait-Oumesmar B, Picard-Riera N, Kerninon C, Baron-Van Evercooren injury. Glia 59:869–881.
A. 2008. The role of SVZ-derived neural precursors in demyelinat- Stallcup WB. 2002. The NG2 proteoglycan: past insights and future
ing diseases: from animal models to multiple sclerosis. J Neurol Sci prospects. J Neurocytol 31:423–435.
265:26–31. Stallcup WB, Beasley L. 1987. Bipotential glial precursor cells of
Nishiyama A. 2007. Polydendrocytes: NG2 cells with many roles in the optic nerve express the NG2 proteoglycan. J Neurosci 7:
development and repair of the CNS. The Neuroscientist 13:62–76. 2737–2744.
Nishiyama A, Komitova M, Suzuki R, Zhu X. 2009. Polydendrocytes Sugiarto S, Persson AI, Munoz EG, Waldhuber M, Lamagna C, Andor
(NG2 cells): multifunctional cells with lineage plasticity. Nat Rev N, et al. 2011. Asymmetry-defective oligodendrocyte progenitors are
Neurosci 10:9–22. glioma precursors. Cancer Cell 20:328–340.
Nishiyama A, Yu M, Drazba JA, Tuohy VK. 1997. Normal and reactive Tatsumi K, Takebayashi H, Manabe T, Tanaka KF, Makinodan M,
NG2+ glial cells are distinct from resting and activated microglia. Yamauchi T, et al. 2008. Genetic fate mapping of Olig2 progenitors
J Neurosci Res 48:299–312. in the injured adult cerebral cortex reveals preferential differentiation
Ono K, Takebayashi H, Ikeda K, Furusho M, Nishizawa T, Watanabe into astrocytes. J Neurosci Res 86:3494–3502.
K, et al. 2008. Regional- and temporal-dependent changes in Tekki-Kessaris N, Woodruff R, Hall AC, Gaffield W, Kimura S,
the differentiation of Olig2 progenitors in the forebrain, and the Stiles CD, et al. 2001. Hedgehog-dependent oligodendrocyte
impact on astrocyte development in the dorsal pallium. Dev Biol lineage specification in the telencephalon. Development 128:
320:456–468. 2545–2554.

120 • NEUROGLIA
Tripathi RB, Rivers LE, Young KM, Jamen F, Richardson WD. 2010. by disrupting the beta-catenin-TCF interaction. Nat Neurosci
NG2 glia generate new oligodendrocytes but few astrocytes in a 12:829–838.
murine experimental autoimmune encephalomyelitis model of demy- Yuan X, Chittajallu R, Belachew S, Anderson S, McBain CJ, Gallo V.
elinating disease. J Neurosci 30:16383–16390. 2002. Expression of the green fluorescent protein in the oligodendro-
Trotter J, Karram K, Nishiyama A. 2010. NG2 cells: Properties, progeny cyte lineage: a transgenic mouse for developmental and physiological
and origin. Brain Res Rev 63:72–82. studies. J Neurosci Res 70:529–545.
Vaughn JE. 1969. An electron microscopic analysis of gliogenesis in rat Zawadzka M, Rivers LE, Fancy SP, Zhao C, Tripathi R, Jamen F, et al.
optic nerves. Z Zellforsch Mikrosk Anat 94:293–324. 2010. CNS-resident glial progenitor/stem cells produce Schwann
Vora P, Pillai PP, Zhu W, Mustapha J, Namaka MP, Frost EE. 2011. cells as well as oligodendrocytes during repair of CNS demyelination.
Differential effects of growth factors on oligodendrocyte progenitor Cell Stem Cell 6:578–590.
migration. Eur J Cell Biol 90:649–656. Zerlin M and Goldman JE. 1997. Interactions between glial progenitors
Watanabe M, Toyama Y, Nishiyama A. 2002. Differentiation of prolif- and blood vessels during early postnatal corticogenesis: blood vessel
erated NG2-positive glial progenitor cells in a remyelinating lesion. contact represents an early stage of astrocyte differentiation. J Comp
J Neurosci Res 69:826–836. Neurol 387:537–546.
Wolswijk G. 1998. Chronic stage multiple sclerosis lesions contain a Zhu X, Bergles DE, Nishiyama A. 2008. NG2 cells generate both oligo-
relatively quiescent population of oligodendrocyte precursor cells. dendrocytes and gray matter astrocytes. Development 135:145–157.
J Neurosci 18:601–609. Zhu X, Hill RA, Dietrich D, Komitova M, Suzuki R, Nishiyama A.
Wu J, Yoo S, Wilcock D, Lytle JM, Leung PY, Colton CA, et al. 2010. 2011. Age-dependent fate and lineage restriction of single NG2 cells.
Interaction of NG2(+) glial progenitors and microglia/macrophages Development 138:745–753.
from the injured spinal cord. Glia 58:410–422. Ziskin JL, Nishiyama A, Rubio M, Fukaya M, Bergles DE. 2007. Vesicular
Ye F, Chen Y, Hoang T, Montgomery RL, Zhao XH, Bu H, et al. 2009. release of glutamate from unmyelinated axons in white matter. Nat
HDAC1 and HDAC2 regulate oligodendrocyte differentiation Neurosci 10:321–330.

N G 2 C E L L S ( P O LY D E N D R O C Y T E S ) • 121
11.
GLIAL CELLS IN AUTONOMIC AND SENSORY GANGLIA
Menachem Hanani and David C. Spray

A B B R E VI AT I O N S 1 I N T R O D U C T I O N : T H E O R G A N I Z AT I O N
OF PERIPHERAL GANGLIA
ANS autonomic nervous system
AQP4 aquaporin 4 The peripheral nervous system (PNS) consists of nerves ema-
ATP adenosine triphosphate nating from brain and spinal cord, as well as sensory and auto-
BMP bone morphogenetic proteins nomic ganglia (Fig. 11.1). Sensory ganglia contain neuronal cell
cAMP cyclic adenosine monophosphate bodies of afferent nerves responsible for mechanical, thermal,
CD Crohn disease painful, and proprioceptive input (mechano-, thermo-, noci-,
CNS central nervous system and proprioceptors). These neurons have T-shaped axons,
with one branch forming nerve endings in the periphery that
Cx connexin
can be either anatomically specialized or nonspecialized, and
DRG dorsal root ganglion the other branch synapsing onto interneurons within the dor-
dsRNA double stranded RNA sal horn of the spinal cord. The main types of sensory ganglia
EGCs enteric glial cells are the dorsal root ganglia (DRG), which innervate most of
ENS enteric nervous system the body and internal organs; trigeminal ganglia (TG), which
ErbB3 avian erythroblastic leukemia viral innervate the face, head and teeth; and nodose ganglia, which
oncogene homolog3 receive sensory inputs from internal organs such as heart, res-
ET endothelin piratory tract, and stomach through the vagal nerves.
GABA gamma-aminobutyric acid The three main types of glia in the PNS are the satellite
GAT GABA transporter glial cells (SGCs) in sensory, parasympathetic, and sympa-
GDNF glia cell line-derived neurotrophic thetic ganglia that share certain properties with astrocytes,
factor the enteric glia, which seem to be even more similar to CNS
GFAP glial fibrillary acidic protein astroglia, and the myelinating and nonmyelinating Schwann
GGF glial growth factor (now termed cells (see chapters 7 and 14). Schwann cells are dealt with in
neuregulin1, NRG1) detail in other chapters in this volume. The PNS also contains
macro-phages, which are similar to microglia, and are not
GLAST glial glutamate aspartate transporter
discussed here. For a description of more specialized types of
IL interleukin peripheral glia, see Hanani (2010).
NO nitric oxide Satellite glial cells share many properties with astrocytes,
NRG1 neuregulin1 including expression of glutamine synthetase and a variety of
NTPDase ecto-nucleotidase receptors (including purinergic receptors) and transporters
P2 purinergic receptor type 2 characteristic of astrocytes; like astrocytes they are mutually
P2Y4 purinergic receptor type P2Y4 coupled by gap junctions, although the coupling is less exten-
P2X7 purinergic receptor type P2X7 sive in SGCs (Hanani et al. 1999; Hanani et al. 2002). A unique
PAR protease activated receptor feature of SGCs that distinguishes them from astrocytes is that
PNS peripheral nervous system they surround the cell bodies of the sensory, sympathetic, or
S100 calcium binding protein S100 parasympathetic neurons, having an anatomical relationship to
SGC satellite glial cells neurons that is similar to that of Schwann cells. Table 11.1 sum-
marizes some of the main characteristics of glia in the PNS.
SK3 calcium activated small conductance
potassium channel KCNN3
Sox10 transcription factor Sox 10 2 I D E N T I F Y I N G S AT E L L I T E
TTX tetrodotoxin GLIAL CELLS
TG trigeminal ganglion
TNF tumor necrosis factor Identifying SGCs may pose difficulty because these cells form
UTP uridine triphosphate a thin sheath around the neurons, which at places may be
too thin to be viewed under the light microscope (Fig. 11.2).

122
A Dorsal root ganglion When these cells are selectively labeled, their thin profile
and close proximity to the neurons may lead to the mistaken
Periphery conclusion that the label is present in or near the neuronal
plasma membrane. A close look will usually reveal that the
SGC envelope is not smooth as expected from the neuronal
Sympathetic ganglion membrane, but has several thickenings, especially where the
B C nucleus is located (which may be missed when viewing sec-
SGC SGC
tions). In general, confirmatory immunohistochemical label-
ing is recommended for identifying these cells. Satellite glial
N N
cells and Schwann cells may share several proteins (e.g., S100
and laminin); therefore, these markers are not definitive in
cases in which ambiguity may exist (e.g., in tissue culture,
D
Submucosal plexus
where structural relations are disrupted). More selective SGC
Longitudinal SM
markers are glutamine synthetase and several other enzymes
Myenteric plexus involved in cell metabolism (Miller et al. 2002; Weick et al.
Circular SM
Submucosa
2003) (Fig. 11.3A), the glutamate transporter GLAST (Ohara
et al. 2008), the K+ channel SK3 (Ohara et al. 2009), or the
P2 receptor P2X7R (Belzer et al. 2010) (Fig. 11.3B). SGCs in
Figure 11.1 The Organization of Peripheral Ganglia. A. Schematic sympathetic and parasympathetic ganglia can be identified by
diagram of spinal cord, nerve roots, paravertebral sympathetic ganglion
and a dorsal root ganglion. B. The arrangement of satellite glial cells
staining for S100 (Cocchia and Michetti 1981; Hanani et al.
(SGCs) around a neuron in a sensory ganglion. C. A similar arrangement 1999); however, this marker can also be found in neurons and
is found in sympathetic ganglia, except that the neurons receive synapses. Schwann cells (Gonzalez-Martinez et al. 2003). Satellite glial
D. Schematic diagram of a cross section in the intestine showing the cells in urinary bladder ganglia (parasympathetic) are positive
arrangement of the myenteric plexus between the two layers of smooth for glutamine synthetase (Hanani et al. 1999), but no similar
muscle (SM), and of the submucosal plexus. (D) Modified from Heanue
and Pachnis 2007.
information is available for sympathetic ganglia.

Table 11.1 COMPARISON AMONG THE MAIN TYPES OF PERIPHERAL GLIAL CELLS
NONMYELINATING
CELL TYPE/PROPERTY AUTONOMIC SGCS SENSORY SGCS SCHWANN CELLS ENTERIC GLIA

Neurotransmitter transporters + + + +
Vimentin, S100 + + + +
GFAP + ± + +
Glutamine synthetase ?1,2 + + +3
Schwann cell myelin protein − − + −
Coupling by gap junctions + + + +++
P2 receptors P2Y1,2,6 P2X7, P2Y1,2,4,6,12,13 P2X7, P2Y1,2 P2X7, P2Y2,4
Ecto-ATPase + + + +
Calcium waves ? + ? +
Cytokine expression LIF IL-1β, TNFα IL-1β, IL-10, TNFα IL-1β, IL-6, TNFα
Cell processes − − − +++
+
Inward rectifying K channels + + + +
Contacts with blood vessels − − − +
Relation with neuronal somata Form a complete cover Form a complete cover Do not contact somata Form a partial cover

Engagement in phagocytosis + + + ?4

Abbreviations: Vim, Vimentin; BMP, bone morphogenetic proteins; LIF, leukemia inhibitory factor
Notes:
1. The question mark indicates a lack of information in the literature.
2. Glutamine synthetase was found in parasympathetic SGCs (Hanani et al. 1999; Sha et al. 2001), but there is no similar information in sympathetic SGCs.
3. Several authors identified glutamine synthetase in enteric glia, but Rühl (2005) reported a failure to reproduce this result.
4. The only report on phagocytosis by enteric glia is an abstract (Hollenbach E. et al. Gastroenterology 2000;118:A184).
Modified from Hanani (2010). See this reference for further details.

G L I A L C E L L S I N AU TO N O M I C A N D S E N S O RY G A N G L I A • 123
Little is known regarding GFAP expression in SGCs in
parasympathetic ganglia. Hanani et al. (1999) reported an
absence of this marker in the intrinsic ganglia of the guinea pig
bladder, whereas SGCs in ganglia of cat pancreas are GFAP
positive (Sha et al. 2001).

3 G L I A I N S E N S O RY G A N G L I A

In sensory ganglia, individual neurons are generally covered by


a single layer of SGCs (for detailed reviews, see Hanani 2005;
Ohara et al. 2009; Pannese 1981). The space between SGCs
and neuron is about 20 nm, similar to the extracellular space
in the central nervous system (CNS). Despite the absence
of synapses, the SGCs synthesize a number of neuroactive
compounds and express a variety of neurotransmitter recep-
tors (see Table 11.1), implying that they can modify and be
modified by neurons and other SGCs.
Figure 11.2 A Low-Power Electron Micrograph Showing the Gap junctions are likely to have important implications
Neuron-Satellite Glial Cell Unit in Sensory Ganglia Satellite glial for the function of glial cells because they allow the passage of
cell(s) (red). The small empty spaces in the SGC sheath are neuronal ions (electrical currents) and also metabolites of up to molec-
protrusions. Ct, connective tissue space; N, neuronal nuclei; v, blood
vessels. Calibration bar, 4 μm.
ular weight of 1,000 (see chapter 24). Extensive coupling of
glial cells by gap junctions enables astrocytes to control the
Using GFAP to identify SGCs is problematic. Under rest- extracellular concentration of potassium ions (K+ buffering).
ing conditions SGCs in sensory ganglia express very low levels The SGCs covering an individual neuron are well coupled,
of GFAP, but after a variety of insults such as axotomy and local which has been extensively evaluated using Lucifer yellow
inflammation, GFAP expression is greatly increased. In fact, injection to measure gap junction-mediated intercellular
the increase in the number of neurons that are surrounded by communication (Fig. 11.4). This coupled network of SGCs,
GFAP-positive SGCs has been used to quantify the extent of glial together with the neuron that they surround, seems likely
activation (Hanani 2005). Satellite glial cells are quite sensitive to a
variety of insults, and even treatment with a drug such as lidocaine
causes the level of GFAP to increase in them (Puljak et al. 2009). A B
The expression of GFAP is detectable in sympathetic N6

ganglia SGCs under resting conditions, and it is strongly N5 N4


N2
upregulated after injury (for review, Hanani 2010). An inter- N
N3
esting common feature of sensory and sympathetic ganglia
N1
is that following injury, GFAP is elevated not only in SGCs
surrounding the injured neurons, but also increases around
noninjured neurons, indicating a spread of activation signals
within the ganglion (Hanani 2010; Stephenson and Byers C D Control
Inflammation
1995).
Coupling incidence (%)

50 20
Coupling incidence (%)

+CBX
+MFA
40 +PA
A B
30
10
20
10
0 0

Figure 11.4 Changes in Dye Coupling Between Satellite Glial Cells of


Mouse S1 DRG 10 to 12 Days After Induction of Colonic Inflammation
with Dinitrobenzoate Sulfonate Inflammation A. A Lucifer yellow
(LY)−injected SGC is coupled to other SGCs only around the same
neuron. B. Dye coupling between SGCs around different neurons,
Figure 11.3 Immunohistochemical Identification of Satellite Glial Cells observed in dinitrobenzoate sulfonate (DNBS)-treated mouse. The
in Sensory Ganglia A. Satellite glial cells are labeled for glutamine asterisks indicate the LY-injected SGCs. Scale bars, 20 μm. C,D. The
synthetase (red). The green dots are gap junctions that were labeled for histograms show the effect of gap junction blockers: carbenoxolone
connexin 43. B. Labeling of SGCs for the purinergic receptor P2X7 (CBX, 50 μM), meclofenamic acid (MFA, 100 μM), and palmitoleic acid
(red). Satellite glial cell nuclei are labeled with DAPI (blue). The neuronal (PA, 30 μM), on the augmented coupling among SGCs after inflamma-
nuclei were also labeled, but are very faint. Note that in both (A) and (B), tion. C. Incidence of coupling between SGCs around the same neuron.
the neurons are not labeled and are seen as empty spaces. D. Incidence of coupling between SGCs around different neurons.

124 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
to form a functional unit. Bidirectional intercellular signal- ganglion, thereby achieving analgesia in a model of neuro-
ing between sensory neurons and SGCs is apparent in Ca2+ pathic facial pain. However, these authors also reported that
wave studies in dissociated cell culture, in which the Ca2+ wave treatment with Cx43 dsRNA could lead to tactile allodynia
was partially inhibited by purinergic receptor antagonists and in control animals (Ohara et al. 2009), raising concerns about
completely blocked when cocultures were treated with a com- whether there is actually a cause-effect relationship between
bination of suramin (broad spectrum P2 receptor antagonist) coupling or gap junction expression and pain sensitivity.
and heptanol (gap junction blocker) (Suadicani et al. 2010). Pharmacological approaches using the gap junction inhib-
The issue of which gap junction proteins (connexins) itor carbenoxolone have been reported to be quite effective in
form the coupling pathway in sensory ganglia has only been producing analgesia in several pain models in which the tar-
addressed relatively recently. Connexin43 (Cx43) was iden- get was presumed to be sensory ganglia (Dublin and Hanani
tified in the perineuronal SGCs of rat trigeminal ganglion 2007; Hanstein et al. 2010; Huang et al. 2010). Finally, it has
(Ohara et al. 2009) and mouse DRG (Procacci et al. 2008) been reported that the antiepileptic drug tonabersat relieved
(see Fig. 11.3). Both Cx43 and Cx26 were also reported to hypersensitivity in a rodent orofacial pain model and blocked
be present in SGCs of rat trigeminal ganglion (Damodaram both the pain-related enhancement of dye spread between
et al. 2009; Garrett and Durham 2008), whereas trigeminal neurons and SGCs and the upregulation of Cx26 in trigem-
ganglion neurons were reported to express Cx36 and Cx40 inal ganglion (Garrett and Durham 2008). These authors
(Garrett and Durham 2008). The expression of gap junction reported that the increased dye coupling between neurons and
proteins and the strength of gap junction−mediated coupling SGCs was associated with upregulation of the neuronal gap
display extraordinary plasticity in the sensory SGCs, one junction proteins Cx40 and Cx36; notably, Cx43 upregula-
example being the increase in abundance and strength that tion was not affected.
occur with age in DRG of rabbits (Martinelli et al. 2004), and The increased coupling following axon injury or inflamma-
mice (Huang et al. 2006). In contrast, the expression of Cx43 tion is likely to increase communication between SGCs, and
was found to decline in old mice (Procacci et al. 2008), sug- our studies on dissociated ganglia from an orofacial pain model
gesting that the increased amount of gap junctions observed showed enhanced SGC-SGC and SGC-neuronal signaling
in aged animals results from the augmentation of a gap junc- (Suadicani et al. 2010). An important underlying concept when
tion protein other than Cx43. Thus, the plasticity in gap junc- discussing SGCs is that these cells are rarely subject to direct
tions likely also extends to changes in the type of connexins injury. Rather, it is the neurons that are injured by damage to the
expressed. Even more profound changes occur following central or peripheral part of the axon. Thus, any change in SGCs
axotomy, with SGCs extending processes that form bridges must be a secondary change driven by alterations to the neuron,
connecting previously separate perineuronal sheaths, and implying signaling mechanisms between neurons and SGCs.
number of gap junctions between SGCs increasing markedly There are a large number of molecules that could be involved in
(Hanani et al. 2002). The incidence of dye coupling among such signaling, and many substances are released from injured
SGCs also increases, with the increase in the number of gap (and noninjured) neurons, such as nitric oxide, tumor necro-
junctions correlating with this functional increase (Hanani sis factor-α (TNF-α), and adenosine triphosphate (ATP)
et al. 2002). Thus, SGCs that are ordinarily coupled only (Bradman et al. 2010; Hanani 2005; Takeda et al. 2009).
to SGCs surrounding the same neuron become extensively Adenosine triphosphate appears to be involved in pain-
coupled to SGCs enveloping other neurons following axo- related processing through the activation of metabotropic (P2Y
tomy (see Fig. 11.4). Similar results were obtained in studies family) or ionotropic (P2X) purinergic receptors. Satellite glial
of trigeminal ganglion following infraorbital nerve section cells express a variety of purinergic receptors (Gu et al. 2010;
(Cherkas et al. 2004). Other conditions under which gap Kushnir et al. 2011; Villa et al. 2010; Weick et al. 2003), and
junction-mediated coupling among SGCs increases include there is evidence that these receptors are involved in neuron-
compression injury of the DRG (Zhang et al. 2009), intesti- SGC communication, and in particular in nociception-related
nal obstruction or inflammation (Huang and Hanani 2005; processes. The P2Y4 receptor has been shown to be exclusively
Huang et al. 2010), paw inflammation (Dublin and Hanani expressed by SGCs in sensory ganglia (Vit et al. 2006). Adenosine
2007) and neuritis of the sciatic nerve (Ledda et al. 2009). triphosphate released following nerve injury may activate P2Y4
receptors on SGCs, resulting in an increase in intracellular Ca2+,
which in turn can trigger activation of K+ channels. The result-
3.1 T H E RO L E O F S AT E L L IT E G L I A L
ing change in extracellular K+ may increase nociceptive neuron
C E L L S I N C H RO N I C PA I N
excitability through altered membrane potential or activation of
There is considerable evidence that glia contribute to chronic the inflammasome (Silverman et al. 2009).
pain (see chapter 68). As mentioned, there are increases in gap
junctions and dye coupling among sensory SGCs in diverse
types of pain models and it was found that inhibition of cou- 4 O R G A N I Z AT I O N O F G L I A L C E L L S
pling can decrease the hypersensitivity as measured behavior- I N SY M PAT H ET I C A N D
ally. Several studies report that the hypersensitivity is reversed PA R A SY M PAT H ET I C G A N G L I A
by treatment of the animals with gap junction inhibitors. For
example, Ohara et al. (2008) knocked down Cx43 expression The peripheral autonomic nervous system (ANS) is made up
in rat trigeminal ganglion by injecting Cx43 dsRNA into the of three functionally and structurally discrete components,

G L I A L C E L L S I N AU TO N O M I C A N D S E N S O RY G A N G L I A • 125
the sympathetic and parasympathetic ganglia, and the enteric somata, and only a minority are axosomatic. Nevertheless,
nervous system (ENS) (for detailed review see Hanani 2010). ultrastructural studies have shown that the synapses are close
to the soma, and that they are enclosed by SGC processes that
wrap around dendrites emerging from the neuronal cell body
4.1 G L I A I N SY M PAT H ET I C G A N G L I A
(Elfvin 1971; Matthews 1983). Dye injection experiments
Preganglionic neuronal somata of the sympathetic nervous sys- reveal that SGCs send tubelike structures having lengths of at
tem are located in the interomediolateral columns of the T1 to least 30 to 40 μm, which enwrap neuronal processes in mouse
L2 spinal cord and synapse onto paravertebral neurons of the superior cervical ganglion (Fig. 11.6E). Thus, it can be con-
sympathetic chain, adrenal chromaffin cells, and prevertebral cluded that SGCs cover most synapses in sympathetic ganglia,
ganglia consisting primarily of principal neurons that supply and are likely to influence synaptic transmission in these gan-
the ENS, and abdominal and pelvic viscera. Preganglionic glia, just as astrocytes are partners in most aspects of synaptic
parasympathetic somata are located in the brainstem and sacral function in the CNS (Perea et al. 2009).
spinal cord and synapse upon parasympathetic ganglia of the Nevertheless, the organization of glial cells in sympathetic
head (ciliary, submandibular, otic, and pterygopalatine gan- ganglia is quite different than in the CNS. Whereas astrocytes
glia) and on cell bodies in tissues that are targets of vagus and contact numerous neurons, oligodendrocytes, and the vascu-
pelvic nerves. The postganglionic autonomic ganglia contain lature (see Dermietzel and Spray 2012), these connections are
efferent nerves directly innervating smooth muscle or glands, absent in the sympathetic ganglia, where each neuron is sur-
whereas the ENS is totally embedded in the gastrointestinal rounded by several SGCs that are in close contact with each other,
wall. Sympathetic and parasympathetic ganglia are largely con- and are separated from SGCs surrounding other neurons.
trolled by the CNS, whereas the ENS is more autonomous. Elfvin and Forsman (1978) identified gap junctions in
Figure 11.5 shows the basic organization of a neuron in SGCs in paravertebral and prevertebral ganglia of rabbits and
sympathetic ganglia and its attending SGCs, which is simi- guinea pigs. Using intracellular dye injections (see Fig. 11.6),
lar to that of sensory ganglia (Matthews, 1983; Pannese 1981, we showed that SGCs in mouse superior cervical ganglion
2010). Each neuron is surrounded by its own glial cover, and
together they form a distinct unit, largely isolated from other
similar units in the ganglion. A B C
Unlike sensory neurons, sympathetic neurons receive syn-
apses, and SGCs cover axon terminals that make synaptic con-
tacts on or near the neuronal somata (Elfvin 1971; Matthews
1983). Usually most of the synapses in sympathetic ganglia
(at least in the paravertebral ones) are between preganglionic
axons and dendrites or small protrusions from the neuronal D E F

N2
N1
N3
A B Syn

n Figure 11.6 Characterization of the Satellite Glial Cell Envelope in the


Mouse Superior Cervical Ganglion with Intracellular Injection of the
Fluorescent Dye Lucifer Yellow All images were obtained on live tissue
nuc Syn during the injection experiments; the injected cell is marked with an
asterisk. A. A single SGC makes a complete envelope around the neuron.
Here, and in the rest of the images, neurons were not stained. B. The
dye has passed from the injected SGC into another SGC around the
n same neuron. C. An example of an SGC that makes a nearly complete
Ax sheath around a neuron, but a small neuronal region appears not to
be covered by the injected SGC (arrow). This may result from a great
attenuation of the glial envelope in this region, or an incomplete invest-
ment of the neuron by this cell. It is quite possible that another SGC
Figure 11.5 The Organization of the Neuron−Satellite Glial Cell Unit (which was not coupled to the injected cell) covers this region. D. An
in Sympathetic Ganglia A. A low-power electron micrograph showing a example of an SGC making a partial envelope around a neuron. In this
sympathetic neuron (n) surrounded by an envelope consisting of satellite case it is very likely that another SGC covers the left part of the neuron,
glial cells. The borders of the SGCs are traced by a thick black line. Note and again is not coupled to the injected cell. E. The injected SGC fully
that the contour of the SGC is very thin at certain points. The nucleus surrounds the neuron and also wraps around two neuronal processes.
of a SGC is indicated with an arrow. Nuc, nucleus of the neuron. Mouse Note the tubelike glial cover of these processes (arrows). F. An example
superior cervical ganglion. Calibration bar, 2 μm. B. Schematic diagram of interenvelope dye coupling. An SGC around neuron N1 was injected
of a sympathetic neuron (n) surrounded by SGCs (arrows). Note that with dye, which spread to another SGC around this neuron, and also to
the synapses (Syn) are covered by SGC processes and that these processes SGCs around two adjacent neurons (N2 and N3). This type of coupling
extend beyond the neuronal soma and ensheathe an axon (Ax) and is rare (in about 3% of the cells) under normal conditions, but its inci-
a dendrite (d). (A) Courtesy of M. Egle De Stefano and Paola Paggi, dence increases eightfold following peripheral injury. Calibration bars,
La Sapienza University, Rome; (B) from Hanani 2010. 20 μm. From Hanani 2010.

126 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
are coupled around a given neuron. Coupling between SGCs lamina and allowing the possibility that the neural protrusions
surrounding different neurons is normally very rare, but might play a functional role in sensing the ganglionic environ-
increases after peripheral inflammation (Hanani et al. 2010) ment. The neuronal plasma membrane and SGC membranes
(see Fig. 11.6). The connexin type responsible for gap junction- display complex interdigitations, which greatly increase
mediated intercellular communication between sympathetic the surface area of both cells, and can promote intercellular
SGCs remains to be determined, as does the degree to which interactions.
coupling strength can be modulated by agents affecting cou- There is only scarce information on gap junctions in
pling in sympathetic neurons (Kessler et al. 1984). SGCs of parasympathetic ganglia. There is electron-micro-
A major change in sympathetic ganglia following axo- scopic evidence for gap junctions in SGCs in the chicken
tomy is the detachment of the presynaptic terminals from the ciliary ganglion (Forsman et al. 1989). Hanani et al. (1999)
postsynaptic membrane (“synaptic stripping,” see Kreutzberg studied dye coupling among SGCs in the intramural gan-
1995). Using extracellular electrical recordings, Matthews and glia of the guinea pig urinary bladder and found only a small
Nelson (1975) found that after axotomy, synaptic transmission degree of dye coupling among SGC forming the envelope of
in the ganglion was greatly depressed. Similarly, intracellular individual neurons.
recordings from guinea pig superior cervical ganglion after
axotomy demonstrated a 70% reduction in synaptic poten-
tial amplitude (Purves 1975). Matthews and Nelson (1975) 5 ENTERIC GLIAL CELLS
also noted that sympathetic neurons lost most or all of their
synaptic inputs after axotomy and became physically separated Since the 1970s there has been intense interest in the ENS,
from them. Significantly, presynaptic terminals appeared as investigators recognized that it is the main element in the
normal. These findings indicate that the reduction in synap- control of all functions of the digestive system (Goyal and
tic transmission was not caused by changes in the presynap- Hirano 1996). Although most of the research in this field
tic compartment. Matthews and Nelson (1975) noted that was devoted to enteric neurons, there has been a fair amount
glial processes become interposed between the retracting ter- of work on enteric glial cells (EGCs), and these cells are the
minals and the postsynaptic cells and therefore apparently most characterized among the glial cells in peripheral ganglia.
played a role in synaptic stripping. Detached presynaptic The mere name enteric glia indicates that these cells are unique
profiles were often wrapped by one or more narrow lamella to this system, in accord with the special properties of the ENS
of SGC cytoplasm, which enveloped the specialized presyn- within the ANS. It should be noted that most of the current
aptic region. Thus it appears that after injury SGCs formed knowledge on the ENS is derived from work on the guinea
new extensions. The presence of glial processes between pre- pig small intestine; therefore, generalizations to other species,
synaptic and postsynaptic elements after injury was confirmed especially humans, should be made with caution.
by more recent ultrastructural studies (DeStefano et al. 2007). The ENS contains more than 100 million neurons and 4
Overall, this phenomenon of interposed glial process between to 10 times as many glia. (Neunlist et al. 2008). These gan-
presynaptic and postsynaptic neuronal elements is similar to glia are organized as submucosal (Meissner’s) plexus in the
that studied extensively in supraoptic hypothalamus (Oliet submucosa, which control mucosal activity, and myenteric
et al. 2008). (Auerbach) plexus between circular and longitudinal layers
that control these muscles. Markers for these glial cells include
4.2 G L I A I N PA R A SY M PAT H ET I C G A N G L I A Sox10, GFAP, and S100b. It is believed that EGCs are major
regulators of barrier and neuronal functions in the gut. Enteric
Much less research has been conducted on SGCs in parasym- glial cells decrease intestinal mucosal barrier permeability via
pathetic ganglia than in sympathetic ganglia. This is caused in release of S-nitrosoglutathione and regulation of expression
part by most parasympathetic ganglia being relatively inacces- of the tight junction proteins ZO1 and occludin (Savidge
sible. Also, in contrast with sympathetic ganglia, which are et al. 2007).
compact and well-defined structures that are located at a large The depth of understanding of molecular and develop-
distance from their targets, parasympathetic ones are more mental neurobiology of the gut is extensive, with Sox10 and
diffuse, and therefore more difficult to isolate. Notch signaling pathways being critical to gliogenesis in the
The general organization of parasympathetic ganglia is very ENS (see chapters 14 and 43) (Ngan et al. 2011; Taylor et al.
similar to that seen in sympathetic ganglia. The glial cells wrap 2007). Sequences of transcription factor activation have been
around the neurons, and thus are classified as SGCs. The intrin- proposed to underlie differentiation of both neurons and the
sic ganglia of the urinary bladder are typical for this system EGCs (Chalazonitis and Kessler 2011; Chalazonitis et al.
(Gabella 1990), and SGCs in them make a thin sheath around 2011). In this scheme of development of the bowel ganglia,
individual neurons, which is attenuated in some regions, but bone morphogenetic proteins (BMP2 and BMP4) play criti-
is still continuous. In the guinea pig tracheal ganglia, neurons cal roles in differentiation of both neurons at early embryonic
and their processes are almost always covered by a sheath con- times and later glia. The early effect is BPM enhancement
sisting of several SGCs, which can be extremely thin (about of GDNF, thereby favoring neurogenesis, and suppress-
0.2 μm) (Baluk et al. 1985). This sheath is interrupted in ing NRG1 (formerly GGF2) and the development of glia.
only very small areas, in which a thin neuronal process pro- Later, the BMPs increase expression of ErbB3 and enhance
trudes between two SGCs, making contact with the basal NRG1-driven enteric gliogenesis with decreased neurogenic

G L I A L C E L L S I N AU TO N O M I C A N D S E N S O RY G A N G L I A • 127
A1 A3 5.1 S T RU C T U R E O F E N T E R I C G L I A L C E L L S
Enteric glial cells are similar in many respects to astrocytes
of the CNS (Gabella 1981; Jessen and Mirsky 1980). Unlike
A4 SGCs, EGCs do not envelop neurons, but each EGC sends
A2 processes that make contact with several neurons, as seen in
astrocytes. Hanani and Reichenbach (1994) studied the mor-
A5 phology of glial cells in the myenteric plexus of the guinea
pig small intestine using intracellular injection of horseradish
50 μm peroxidase, and found a great complexity of structure, resem-
bling that found in astrocytes (Fig. 11.7). The complexity was
B C D quantified by a fractal dimension of around 1.5, which is simi-
lar to that of astrocytes (Reichenbach et al. 1992). Two main
morphological EGC types were found: stellate cells that send
processes in a symmetrical manner and are found within the
ganglia, and cells sending processes in an elongated pattern
that are found within the fiber bundles connecting the ganglia
(see Fig. 11.7). These types roughly correspond to protoplas-
mic and fibrillary astrocytes, respectively.
E F
Satellite glial cells can make contacts with blood vessels
and/or with the surface of the ganglia via thickenings of the
glial processes that resemble astrocytic endfeet (Gabella 1981;
Hanani and Reichenbach 1999). Some EGC processes form
“windows,” which wrap around nerve bundles (Gershon and
Rothman 1991; Hanani and Reichenbach 1994). Enteric glial
cells share other features with astrocytes such as the pres-
ence of glutamine synthetase, Ran-2 (ceruloplasmin) ( Jessen
30 μm 20 μm and Mirsky 1983), apolipoprotein E (Bolyes et al. 1985). See
Gershon and Rothman (1991) and Rühl (2005) for reviews.
Figure 11.7 Some Morphological, Histochemical, and Functional Enteric glial cell processes (apparently from the submu-
Characteristics of Enteric Glial Cells A1−A5. Morphology of EGCs in
the myenteric plexus of the guinea pig small intestine that were injected cosal plexus) extend into the mucosa to the villous tips, and
intracellularly with horseradish peroxidase (HRP). A1. A stellate EGC, thus might affect mucosal functions such as secretion and
located within the ganglion; several processes of the cell make endfeet influence mucosal permeability (Bush et al. 1998; Cornet
with a blood vessel, and other processes contact the borders of the gan- et al. 2001; Neunlist et al. 2008). These projections might
glion. A2. Another example of a stellate EGC, showing its relationship sense the luminal contents and send appropriate signals to
to a neuron (asterisk). A3. An example of SGC located in the fiber tract
connecting the ganglia. Note the elongated shape of the processes, and the ENS. It has been claimed that a population of glial cells is
the thickenings that contact the border of the fiber tract. A4. Another located in the intestinal mucosa (Bernstein and Vidrich 1994).
example of an EGC in a fiber tract. A5. An EGC whose right part, which This would require that these cells are not associated with
is located within the ganglion is symmetrical (stellate shape), whereas its neuronal somata of the ENS, which would be an exception
left side is in a fiber tract. B. Myenteric ganglion in the guinea pig small for EGCs, because there is no evidence for neurons within the
intestine, where a single EGC (asterisk) was injected with the fluorescent
dye Lucifer yellow. The dye diffused (most likely through gap junctions) mucosa proper. This potentially important observation has
to numerous EGCs in the ganglion. The neurons are not labeled with the not been confirmed.
dye, and are seen as dark circles. C. Injection of an EGC in a submu-
cosal ganglion shows dye spread even to EGCs in neighboring ganglia
(arrowheads). D. An EGC in the submucosal plexus injected with HRP.
Some processes contact the border of the ganglion. E. Double immuno-
5.1.1 Gap Junctions
histochemical labeling of mouse colonic myenteric plexus for aquaporin Gap junctions in EGCs were described by electron micros-
4 (AQP4) and the glial marker GFAP. Some neurons (arrowheads)
and nerve fibers (arrows) are labeled for AQP4. Enteric glial cells are
copy (Gabella 1981). Dye injection studies demonstrated that
GFAP-positive, and do not label for AQP4. Similar results were obtained EGCs are extensively coupled to each other in both myenteric
in the small intestine of mice and rats. F. Immunostaining for AQP4 and submucosal plexuses (Hanani et al. 1989; Maudlej and
and GFAP in the mouse colonic submucosal plexus. (E,F) From Thi Hanani 1992) (see Fig. 11.7). As can be seen in Figure 11.7B,
et al. 2008. the dye coupling in myenteric ganglia can extend over a dis-
tance of several hundred μm, and may cover the whole gan-
glion. In the case of submucosal plexus, coupling even extends
response to GDNF. In the context of interplay between tran- between ganglia (see Fig. 11.7C). The spatial extent of dye cou-
scription factor determination of fates of neurons and glia, pling seen in these studies probably exceeds that observed in
it should be noted that recent reports have indicated that the CNS, which may be caused by numerous gap junctions,
EGCs can differentiate into neurons under specific conditions but can also be explained by the essentially two-dimensional
(Gershon 2011). structure of the enteric ganglia, in which no dilution of the

128 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
dye occurs in three dimensions, as in the CNS. These observa- (e.g., inflammation), these receptors may participate in the
tions suggest that EGCs engage in spatial buffering of K+ and response of EGCs. Also, EGCs contain protease-activated
possibly in clearing other potentially harmful substances. receptors (PAR), which are thought to participate in tissue
responses to injury and inflammation Activation of these
receptors by trypsin and thrombin induced a rise in [Ca2+]in in
5.2 P H YS I O L O GY A N D P H A R M AC O L O GY
cultured myenteric EGCs (Garrido et al. 2002). Enteric glial
OF ENTERIC GLIAL CELLS
cells display additional receptors, among which are receptors
A well-established function of glial cells is to regulate their for glutamate (Nasser et al. 2007), interleukin beta 1 and 6
environment. There is evidence that EGCs contain the GABA (Rühl, 2005), lysophosphatidic acid (Segura et al. 2004a),
transporter GAT2 (Fletcher et al. 2002), whereas astrocytes and sphingosine-1-phosphate (Segura et al. 2004b).
contain GAT1,3. There is evidence for glutamatergic trans- A study on intact segments of guinea pig ileum showed
mission in the ENS (Kirchgessner 2001), and for glutamate that ATP indeed is a messenger between neurons and EGCs
receptors in EGCs (Nasser et al. 2007), but there is so far no (Gulbransen and Sharkey 2009). Neuronal stimulation-
evidence for glutamate transporters in EGCs, which are abun- evoked Ca2+ increase in EGCs, which were blocked by P2
dant in astrocytes. receptor blockers, and EGCs responded to exogenous ATP.
The small size of EGC is one reason the physiology of These authors concluded that EGCs have P2Y4 receptors.
these cells has received little attention. Patch clamp record- In summary, EGCs are endowed with a wide spectrum of
ing is clearly the method of choice for studying the physiol- membrane receptors that enable them to interact with their
ogy of these cells. Broussard et al. (1993) developed a tissue environment. The presence of receptors that are activated with
culture preparation containing a nearly pure population of ligands associated with inflammation (ATP, PAR) supports the
EGC from the myenteric plexus of guinea pig ileum. The reports on the potential protective role of EGCs against inflam-
cells retained expression for the glial markers S100 and glu- mation (Bush 2002). Therefore, EGCs might be an important
tamine synthetase, and for GD3 ganglioside. Broussard et target for medical therapy for gastrointestinal disorders, and
al. (1993) made patch clamp recordings from these cells and inflammatory bowel diseases in particular (see section 5.3).
found that the major ionic conductance is for outward recti-
fying K+ channels. Surprisingly, 8% of the cells had tetrodo-
5.3 E N T E R I C G L I A A N D I N T E S T I NA L D I S E A S E
toxin (TTX)-sensitive Na+ channels. Hanani et al. (2000)
made patch recordings from glia in isolated myenteric gan- Crohn disease (CD) is a debilitating and unexplained inflam-
glia, a preparation that is closer to the intact situation than matory bowel disease. Enteric glial cells from CD patients
cultures. They also identified outward rectifying K+ channels. display abnormal features; they are strongly positive for major
However, when the gap junctions were blocked, the K+ chan- histocompatibility class II antigens (Geboes et al. 1992). Thus,
nel blocker Ba2+ (1 mM) had a strong inhibitory effect on the EGCs may serve as antigen presenting and/or target for T
currents (particularly the inward ones) indicating that inward cells. The functional significance of these observations is not
rectifying channels are also present in EGCs. These channels clear, because so little is known about EGCs in humans.
are believed to be important for K+ buffering in central glia Animal studies suggest that EGCs may be essential for
(Verkhratsky and Steinhäuser 2000), and these results provide the integrity of the digestive tract. In two separate studies,
indirect evidence for such a function of EGCs. These experi- transgenic mice were used to deplete or reduce the numbers
ments show that various ionic conductances may normally of EGCs (Bush et al. 1998; Cornet et al. 2001). This resulted
be masked by dominant “passive” currents, which are largely in intestinal inflammation leading to the animals’ death. The
caused by gap junctions among EGCs (Hanani et al. 1989). clinical and pathological findings indicate an inflammatory
An elegant series of experiments on cultured EGCs bowel disease resembling CD. Interestingly, in CD patients
from the myenteric plexus of the newborn guinea pig ileum both involved and uninvolved intestinal segments showed a
employed Ca2+ imaging to characterize responses to a num- diminished EGC network, suggesting that EGCs control the
ber of putative neurotransmitters. EGCs were found to integrity and permeability of submucosal blood vessels and
respond to ATP and UTP by an elevation of intracellular intestinal mucosa (Cornet et al. 2001). Thus, when EGCs are
Ca2+ concentration ([Ca2+]in), and apparently possess what not present, vascular and mucosal barriers break down, lead-
are now termed P2Y2 and/or P2Y4 receptors (Kimball et al. ing to inflammation. Glial depletion was proposed to lead
1996). These results correlate with the presence of the ecto- to mucosal inflammation (Bush 2002). Bush (2002) empha-
nucleotidase (NTPDase2) in EGCs (Braun et al. 2004), an sized the structural similarities between EGCs and astrocytes,
enzyme that breaks down ATP. Endothelin increased [Ca2+]in which may indicate common functions. Reactive astrocytes
by IP3-dependent release from intracellular stores, appar- release a number of cytokines, growth factors, and other active
ently via ETB receptors (Zhang et al. 1997). Responses to compounds (Watkins and Maier 2002). These substances
both ATP and endothelin were followed by capacitative Ca2+ can affect the neurons directly, by acting on receptors, or
influx (Sarosi et al. 1998; Zhang et al. 1998). In addition to indirectly, for example by altering blood flow. These responses
P2Y receptors, which are metabotropic, ionotropic P2X7 can be beneficial to the survival of neurons, but in some cases
receptors were identified in EGCs by immunohistochem- they may be detrimental.
istry (Vanderwinden et al. 2003). These authors suggested Further evidence on the role of EGCs in inflammatory
that, as ATP levels increase under pathological conditions bowel disease (Rühl et al. 2001), demonstrated that EGCs

G L I A L C E L L S I N AU TO N O M I C A N D S E N S O RY G A N G L I A • 129
from rat myenteric plexus produce the proinflammatory 6 B L O O D − N E RVO U S SYS T E M B A R R I E R S
cytokine IL-6, and this process is regulated by the proinflam- I N T H E P E R I P H E R A L N E RVO U S SYS T E M
matory cytokines IL1-β and IL-6 but not TNF-α. This again
supports the notion that EGCs play an important role in intes- The question of whether a barrier exists between the blood and
tinal inflammatory reactions. The authors argued that myen- neural cells in the periphery has been controversial. On the
teric glia are strategically located to amplify and perpetuate one hand, peripheral nerves are surrounded by a highly imper-
the inflammatory response in the ENS because of their close meable perineurium, creating the so-called blood-nerve bar-
anatomical association with enteric neurons and their location rier, and both enteric and sensory ganglia are also encased in
at the edge of the ganglia. This provides them with the ability an impermeable sheath. However, in contrast to most regions
to intervene between the extraganglionic and intraganglionic of the CNS, the vasculature within the ganglia is fenestrated
compartments. Thus they can receive signals and produce the and leaky, so that the capillaries provide relatively free access
proinflammatory cytokine IL-6. to neurons and glia. This arrangement of a connective tissue
Because the submucosa is more closely associated with the sheath surrounding sensory and sympathetic ganglia providing
mucosa, glial cells in the submucosal plexus are even more an impermeable covering, yet penetration by proteins from the
likely than myenteric plexus glia to be involved in the path- blood creates the interesting paradox in which removal, rather
ological processes discussed in the preceding. However, the than delivery, may be impeded in the ganglia. The ENS is avas-
studies mentioned in the previous sections were mostly done
cular, leading to the proposal that there may be a blood-brain
on glial cells of the myenteric plexus.
barrier in that tissue (Gershon 1981). However, subsequent
Chagas disease, caused by infection with the intracellu-
studies indicated that EGCs and neurons are in contact with
lar protozoan parasite Trypanosoma cruzi, is a major public
circulating factors (Allen and Kiernan 1994). There have also
health problem in Latin America (Tanowitz et al. 2009). It
been reports that penetration within the ganglia of injected
is characterized by a brief acute phase, followed by a chronic
probes may be different in sensory and sympathetic ganglia.
phase that can last several decades. Although the most severe
Thus, a tight junction barrier was proposed by Ten Tusscher
pathology associated with the disease is its cardiac manifesta-
et al. (1989) to be present in SGCs in autonomic but not in
tion, the gastrointestinal tract is involved in 8% to 10% of
sensory ganglia. As pointed out by Kiernan (1996), more recent
infected patients. The main pathology of the gut is the devel-
studies using a variety of probes have indicated that in the gan-
opment of a megaesophagus and/or megacolon with severe
glia of the PNS, neurons and glia are exposed to blood-borne
defects of motility (de Souza et al. 2010). Although modu-
agents. The absence of a blood neural barrier in PNS offers the
lated tone of the gut wall has been widely attributed to loss
of enteric neurons, it has recently been shown that dilated opportunity for exposure of cells to therapeutic agents.
intestines are characterized by reduced EGCs (da Silveira Astrocytes in the CNS send endfoot processes to the vessel
et al. 2009), whereas GFAP-positive EGCs are increased in wall, both signaling for local autoregulation of blood flow and
nondilated portions (Nascimento et al. 2010). Thus, it now establishing a tight barrier through induction of various genes
appears that chagasic megacolon may also represent a gliopa- (see Dermietzel and Spray 2012). A marker of this specialized
thy, in which loss of EGC reduces secretion of neurotrophic astrocyte domain is the water channel protein aquaporin 4
factors. Similarly to CD, it has been suggested that EGCs (AQP4) (Nicchia et al. 2004). Aquaporin 4 is located in astro-
could also act as antigen-presenting cells in Chagas disease cytic endfeet in membranes that contact capillaries and pia,
(da Silveira et al. 2011). Trypanosoma cruzi infection of car- in which water can be exchanged between inside and outside
diac muscle and other cell types leads to decreased gap junc- the brain. Therefore, it was suggested that AQP4 may play a
tion-mediated intercellular signaling (Adesse et al. 2011), and role in the regulation of water homeostasis in the central ner-
whether this is also the case in EGCs will be most interesting vous system (Nicchia et al. 2004). Our immunohistochemi-
to determine. cal studies reveal that AQP4 expression is confined to a small
There is evidence that in another inflammatory bowel number of myenteric neurons and a larger fraction of submu-
disease in humans—ulcerative colitis—EGCs are activated cosal neurons, but was absent in EGCs (Thi et al. 2008) (see
and release increased amounts of nitric oxide (NO), which Fig. 11.7E,F). This is a further example of a difference between
can contribute to mucosal damage (Cirillo et al. 2011). Celiac EGCs and astrocytes.
disease is a common inflammatory disease in which the mucosa
in the small intestine is disrupted (Schuppan et al. 2009). It 7 S U M M A RY A N D P E R S P E C T I VE S
has been found that NO production (via S100B upregula-
tion) by EGCs participates in the inflammatory process in this Peripheral ganglia are much more than relay stations between
disease as well (Esposito et al. 2007). Bassotti and Villanacci the CNS and sensory terminals or target organs such as the
(2011) have reviewed the literature and argue that constipa- heart, smooth muscle, and glands. It is now recognized that
tion should be considered a neurogliopathy because functional these ganglia are the site of processing of neural information,
impairment is associated with reduced EGC number. which takes place mainly via chemical messengers. Other
In summary, the idea that SGCs are involved in gastroin- key players in these ganglia are specialized glial cells—SGCs
testinal disease is quite recent, but the available information in sensory, sympathetic, and parasympathetic ganglia, and
indicates key roles for these cells in a variety of inflammatory EGCs in the ENS. Research on most aspects of this topic is
and other gastrointestinal disorders. only at the very beginning, and we still lack information on

130 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
the basic physiology and pharmacology of these cells and how Chalazonitis A, Kessler JA. 2011. Pleiotropic effects of the bone morpho-
their function is altered under pathological conditions. The genetic proteins on development of the enteric nervous system. Dev
Neurobiol 166(1):35−41.
central question concerning glial cells in peripheral ganglia is Chalazonitis A, Kessler JA. 2012. Pleiotropic effects of the bone mor-
the nature of neuron−glia interactions, and in the ENS, also phogenetic proteins on development of the enteric nervous system.
between EGCs and the intestinal mucosa. The chemical mes- Dev Neurobiol 72:843–56.
sengers mediating these interactions include ATP, but addi- Cherkas PS, Huang TY, Pannicke T, Tal M, Reichenbach A, Hanani M.
tional molecules are very likely to be present. Also, much needs 2004. The effects of axotomy on neurons and satellite glial cells in
mouse trigeminal ganglion. Pain 110:290−298.
to be learned about the role of gap junctions among the glia, Cirillo C, Sarnelli G, Turco F, Mango A, Grosso M, Aprea G, et al. 2011.
and possibly between them and the neurons. In the CNS, glial Proinflammatory stimuli activates human-derived enteroglial cells
cells communicate with blood vessels, but in the periphery vir- and induces autocrine nitric oxide production. Neurogastr Motil
tually nothing is known about this topic. It is hoped that this 23:e372−382.
chapter will stimulate additional interest in these cells, and Cocchia D, Michetti F. 1981. S-100 antigen in satellite cells of the adre-
nal medulla and the superior cervical ganglion of the rat. An immu-
that both ongoing and new research in the area will provide nocytochemical study. Cell Tiss Res 215:103−112.
answers to these questions. Cornet A, Savidge TC, Cabarrocas J, Deng WL, Colombel JF, Lassmann
H, et al. 2001. Enterocolitis induced by autoimmune targeting of
enteric glial cells: a possible mechanism in Crohn’s disease? Proc Natl
AC K N OW L E D G M E N T S Acad Sci U S A 98:13306−13311.
Damodaram S, Thalakoti S, Freeman SE, Garrett FG, Durham PL. 2009.
Work done in the authors’ laboratories was supported by the Tonabersat inhibits trigeminal ganglion neuronal-satellite glial cell
signaling. Headache 49:5−20.
European Community’s 7th Framework Programme through da Silveira AB, de Oliveira EC, Neto SG, Luquetti AO, Fujiwara RT,
the Marie Curie Initial Training Network Edu-GLIA, the Oliveira RC, et al. 2011. Enteroglial cells act as antigen-presenting
Israel Cancer Association, the Israel Science Foundation (grant cells in chagasic megacolon. Hum Pathol 42:522−532.
no. 212/08), by the US−Israel Binational Science Foundation da Silveira AB, Freitas MA, de Oliveira EC, Neto SG, Luquetti AO,
(grant no. 2007311) and by the National Institutes of Health Furness JB, et al. 2009. Glial fibrillary acidic protein and S-100 colo-
calization in the enteroglial cells in dilated and nondilated portions
(NS041282). of colon from chagasic patients. Hum Pathol 40:244−251.
Dermietzel R, Spray DC. 2012. Blood-brain barrier and the neural
vascular unit. In: Scemes E, Spray DC (eds.), Astrocytes wiring the
REFERENCES brain. Boca Raton, FL: CRC Press, pp. 179−203.
de Souza AP, Sieberg R, Li H, Cahill HR, Zhao D, Araújo-Jorge TC,
Adesse D, Goldenberg RC, Fortes FS, Jasmin, Iacobas DA, Iacobas S, et al. 2010. The role of selenium in intestinal motility and morphol-
et al. 2011. Gap junctions and chagas disease. Adv Parasitol 76:63−81. ogy in a murine model of Trypanosoma cruzi infection. Parasitol Res
Allen DT, Kiernan JA. 1994. Permeation of proteins from the blood into 106:1293−1298.
peripheral nerves and ganglia. Neuroscience 59:755−764. DeStefano ME, Leone L, Moriconi C, DelSignore C, DelSignore A,
Baluk P, Fujiwara T, Matsuda S. 1985. The fine structure of the ganglia of Petrucci TC, et al. 2007. Involvement of the plasminogen enzymatic
the guinea-pig trachea. Cell Tiss Res 239:51−60. cascade in the reaction to axotomy of rat sympathetic neurons. Mol
Bassotti G, Villanacci V. 2011. Can “functional” constipation be consid- Cell Neurosci 36:174−178.
ered as a form of enteric neuro-gliopathy? Glia Biol 59:345−350. Dublin P, Hanani M. 2007. Satellite glial cells in sensory ganglia: their
Belzer V, Shraer N, Hanani M. 2010. Phenotypic changes in satellite glial possible contribution to inflammatory pain. Brain Behav Immun
cells in cultured trigeminal ganglia. Neuron Glia Biol 6:237−243. 21:592−598.
Bernstein CN, Vidrich A. 1994. Isolation, identification, and culture of Elfvin LG. 1971. Ultrastructural studies on the synaptology of the infe-
normal mouse colonic glia. Glia 12:108–116. rior mesenteric ganglion of the cat. I. Observations on the cell surface
Boyles JK, Pitas RE, Wilson E, Mahley RW, Taylor JM. 1985. Apolipo- of the postganglionic perikarya. J Ultrastruct Res 37:411−425.
protein E associated with astrocytic glia of the central nervous system Elfvin LG, Forsman C. 1978. The ultrastructure of junctions between
and with nonmyelinating glia of the peripheral nervous system. J Clin satellite cells in mammalian sympathetic ganglia as revealed by
Invest 76:1501−1513. freeze-etching. J Ultrastruct Res 63:261−274.
Bradman MJ, Arora DK, Morris R, Th ippeswamy T. 2010. How do Esposito G, Cirillo C, Sarnelli G, De Filippis D, D’Armiento FP, Rocco
the satellite glia cells of the dorsal root ganglia respond to stressed A, et al. 2007. Enteric glial-derived S100B protein stimulates nitric
neurons?–nitric oxide saga from embryonic development to axonal oxide production in celiac disease. Gastroenterology 133:918−925.
injury in adulthood. Neuron Glia Biol 6:11–17. Fletcher EL, Clark MJ, Furness JB. 2002. Neuronal and glial localiza-
Braun N, Sévigny J, Robson SC, Hammer K, Hanani M, Zimmermann tion of GABA transporter immunoreactivity in the myenteric plexus.
H. 2004. Association of the ecto-ATPase NTPDase2 with glial cells Cell Tiss Res 308:339−346.
of the peripheral nervous system. Glia 45:124−132. Forsman CA, Ohrling M, Elfvin LG. 1989. Membrane specializations in
Broussard DL, Bannerman PG, Tang CM, Hardy M, Pleasure D. 1993. the satellite cells of the chicken ciliary ganglion. J Submicroscp Cytol
Electrophysiologic and molecular properties of cultured enteric glia. Pathol 21:593−596.
J Neurosci Res 34:24−31. Gabella G. 1981. Ultrastructure of the nerve plexuses of the mammalian
Bush TG. 2002. Enteric glial cells. An upstream target for induc- intestine: the enteric glial cells. Neuroscience 6:425−436.
tion of necrotizing enterocolitis and Crohn’s disease? Bioessays Gabella G. 1990. Intramural neurons in the urinary bladder of the guinea
24:130−140. pig. Cell Tiss Res 261:231−237.
Bush TG, Savidge TC, Freeman TC, Cox HJ, Campbell EA, Mucke L, Garrett FG, Durham PL. 2008. Differential expression of connexins in
et al. 1998. Fulminant jejuno-ileitis following ablation of enteric glia trigeminal ganglion neurons and satellite glial cells in response to
in adult transgenic mice. Cell 93:189−201. chronic or acute joint inflammation. Neuron Glia Biol 4:295−306.
Chalazonitis A, D’Autréaux F. Pham TD, Kessler JA, Gershon MD. Garrido R, Segura B, Zhang W, Mulholland M. 2002. Presence of func-
2011. Bone morphogenetic proteins regulate enteric gliogenesis by tionally active protease-activated receptors 1 and 2 in myenteric glia.
modulating ErbB3 signaling. Dev Biol 350:64−79. J Neurochem 83:556−564.

G L I A L C E L L S I N AU TO N O M I C A N D S E N S O RY G A N G L I A • 131
Geboes K, Rutgeerts P, Ectors N, Mebis J, Penninckx F, Vantrappen Kiernan JA. 1996. Vascular permeability in the peripheral autonomic
G, et al. 1992. Major histocompatibility class II expression on the and somatic nervous systems: controversial aspects and comparisons
small intestinal nervous system in Crohn’s disease. Gastroenterology with the blood brain barrier. Microscop Res Techn 35:122−136.
103:439−447. Kimball BC, Mulholland MW. 1996. Enteric glia exhibit P2U recep-
Gershon MD. 1981. The enteric nervous system. Annu Rev Neurosci tors that increase cytosolic calcium by a phospholipase C-dependent
4:227−172. mechanism. J Neurochem 66:604−612.
Gershon MD. 2011. Behind an enteric neuron there may lie a glial cell. Kirchgessner AL. 2001. Glutamate in the enteric nervous system. Curr
J Clin Invest 121:3386−3389. Opin Pathol 1:591−596.
Gershon MD, Rothman TP. 1991. Enteric glia. Glia 4:195−204. Kreutzberg GW. 1995. Reaction of neuronal cell bodies to axonal dam-
Gonzalez-Martinez T, Perez-Piñera P, Díaz-Esnal B, Vega,J. 2003. S-100 age. In: Waxman, SG, Kocsis JD, Stys PK (eds.), The axon. New York:
proteins in the human peripheral nervous system. Microscop Res Oxford University Press, pp. 355−374.
Techn 60:633−638. Kushnir R, Cherkas PS, Hanani M. 2011. Peripheral inflammation
Goyal RK, Hirano I. 1996. The enteric nervous system. NEJM upregulates P2X receptor expression in satellite glial cells of mouse
25:334:1106−1115. trigeminal ganglia: a calcium imaging study. Neuropharmacology
Gu Y, Chen Y, Zhang X, Li GW, Wang C, Huang LY. 2010. Neuronal 61:739−746.
soma-satellite glial cell interactions in sensory ganglia and the partici- Ledda M, Blum E, De Palo S, Hanani M. 2009. Augmentation in gap
pation of purinergic receptors. Neuron Glia Biol 6:53−62. junction-mediated cell coupling in dorsal root ganglia following sci-
Gulbransen BD, Sharkey KA. 2009. Purinergic neuron-to-glia signaling atic nerve neuritis in the mouse. Neuroscience 164:1538−1545.
in the enteric nervous system. Gastroenterology 136:1349−1358. Martinelli C, Sartori P, Ledda M, Pannese E. 2004. Gap junctions
Hanani M. 2005. Satellite glial cells in sensory ganglia: from form to between perineuronal satellite cells increase in number with age in
function. Brain Res Rev 48:457−476. rabbit spinal ganglia. J Submicroscp Cytol Pathol 36:17−22.
Hanani M. 2010. Satellite glial cells in sympathetic and parasympathetic Matthews MR. 1983. The ultrastructure of junctions in sympathetic gan-
ganglia: in search of function. Brain Res Rev 64:304−327. glia of mammals. In: Elfvin LG (ed.), Autonomic ganglia. Chichester,
Hanani M, Caspi A, Belzer V. 2010. Peripheral inflammation augments UK: Wiley, pp. 27−66.
gap junction-mediated coupling among satellite glial cells in mouse Matthews MR, Nelson VH. 1975. Detachment of structurally intact
sympathetic ganglia. Neuron Glia Biol 6:85−89. nerve endings from chromatolytic neurones of rat superior cervical
Hanani M, Francke M, Härtig W, Grosche J, Reichenbach A, Pannicke ganglion during the depression of synaptic transmission induced by
T. 2000. Patch-clamp study of neurons and glial cells in iso- post-ganglionic axotomy. J Physiol 245:91−135.
lated myenteric ganglia. Am J Physiol Gastroint Liver Physiol Maudlej N, Hanani M. 1992. Modulation of dye coupling among glial
278:G644−G651. cells in the myenteric and submucosal plexuses of the guinea pig.
Hanani M, Huang TY, Cherkas PS, Ledda M, Pannese E. 2002. Glial cell Brain Res 578: 94−98.
plasticity in sensory ganglia induced by nerve damage. Neuroscience Miller KE, Richards BA, Kriebel RM. 2002. Glutamine-, glutamine
114:279−283. synthetase-, glutamate dehydrogenase- and pyruvate carboxylase-
Hanani M, Maudlej N, Härtig W. 1999. Morphology and intercel- immunoreactivities in the rat dorsal root ganglion and peripheral
lular communication in glial cells of intramural ganglia from the nerve. Brain Res 945:202−211.
guinea-pig urinary bladder. J Auton Nerv Syst 76:62−67. Nascimento RD, de Souza Lisboa A, Fujiwara RT, de Freitas MA, Adad
Hanani M, Reichenbach A. 1994. Morphology of horseradish peroxi- SJ, Oliveira RC, et al. 2010. Characterization of enteroglial cells and
dase (HRP)-injected glial cells in the myenteric plexus of the guinea- denervation process in chagasic patients with and without megae-
pig. Cell Tissue Res 278:153–160. sophagus. Hum Pathol 41: 528−534.
Hanani M, Reichenbach A. 1999. Morphology of horseradish peroxidase Nasser Y, Keenan CM, Ma AC, McCafferty DM, Sharkey KA. 2007.
(HRP)-injected glial cells in the myenteric plexus of the guinea-pig. Expression of a functional metabotropic glutamate receptor 5 on
Cell Tiss Res 278:153−160. enteric glia is altered in states of inflammation. Glia 55:859−872.
Hanani M, Zamir O, Baluk P. 1989. . Glial cells in the guinea pig myen- Neunlist M, Aubert P, Bonnaud S, Van Landeghem L, Coron E, Wedel
teric plexus are dye coupled. Brain Res 497:245–249. T, et al. 2007. Enteric glia inhibit intestinal epithelial cell prolifera-
Hanstein R, Zhao JB, Basak R, Smith DN, Zuckerman YY, Hanani M, tion partly through a TGF-beta1-dependent pathway. Am J Physiol
et al. 2010. Focal inflammation causes carbenoxolone-sensitive tactile Gastroint Liver Physiol 292:G231−G241.
hypersensitivity in mice. J Open Pain 3:123−133. Neunlist M, Van Landeghem L, Bourreille A, Savidge T. 2008.
Heanue TA, Pachnis, V. 2007. Enteric nervous system development and Neuro-glial crosstalk in inflammatory bowel disease. J Intern Med.
Hirschsprung’s disease: advances in genetic and stem cell studies. Nat 263:577–583.
Rev Neurosci 8:466−479. Ngan ES et al. 2011. Hedgehog/Notch-induced premature gliogenesis
Huang TY, Belzer V, Hanani MG. 2010. Gap junctions in dorsal root represents a new disease mechanism for Hirschsprung disease in mice
ganglia: possible contribution to visceral pain. Eur J Pain 14:49. and humans. J Clin Invest. 21:346.
e1−11. Nicchia GP, Nico B, Camassa LM, Mola MG, Loh N, Dermietzel R, et al.
Huang TY, Hanani M. 2005. Morphological and electrophysiological The role of aquaporin-4 in the blood-brain barrier development and
changes in mouse dorsal root ganglia after partial colonic obstruc- integrity: studies in animal and cell culture models. Neuroscience
tion. Am J Physiol Gastrointest Liver Physiol 289:G670–G678. 129:935−945.
Huang TY, Hanani M, Ledda M, De Palo S, Pannese E. 2006. Aging is Ohara PT, Vit JP, Bhargava A, Jasmin L. 2008. Evidence for a role of
associated with an increase in dye coupling and in gap junction num- connexin 43 in trigeminal pain using RNA interference in vivo.
ber in satellite glial cells of murine dorsal root ganglia. Neuroscience J Neurophysiol 100:3064−3073.
137:1185−1192. Ohara PT, Vit JP, Bhargava A, Romero M, Sundberg C, Charles AC, et al.
Jessen KR, Mirsky R. 1980. Glial cells in the enteric nervous system con- 2009. Gliopathic pain: when satellite glial cells go bad. Neuroscientist
tain glial fibrillary acidic protein. Nature 286:736−737. 15:450−463.
Jessen KR, Mirsky R. 1983. Astrocyte-like glia in the peripheral nervous Oliet SH, Panatier A, Piet R, Mothet JP, Poulain DA, Theodosis DT.
system: an immunohistochemical study of enteric glia. J Neurosci 2008. Neuron-glia interactions in the rat supraoptic nucleus. Progress
3:2206−2218. Brain Res 170:109−117.
Kessler JA, Spray DC. Saez JC, Bennett MV. 1984. Determination of Pannese E. 1981. The satellite cells of the sensory ganglia. Adv Anat
synaptic phenotype: insulin and cAMP independently initiate devel- Embryol Cell Biol 65:1−111.
opment of electrotonic coupling between cultured sympathetic neu- Pannese E. 2010. The structure of the perineuronal sheath of satellite
rons. Proc Natl Acad Sci U S A 81:6235−6239. glial cells (SGCs) in sensory ganglia. Neuron Glia Biol 6:3−10.

132 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Perea G, Navarrete M, Araque A. 2009. Tripartite synapses: astrocytes Suadicani SO, Cherkas PS, Zuckerman J, Smith DN, Spray DC, Hanani
process and control synaptic information. Trends Neurosci 32: M. 2010. Bidirectional calcium signaling between satellite glial cells
421−431. and neurons in cultured mouse trigeminal ganglia. Neuron Glia Biol
Procacci P, Magnaghi V, Pannese E. 2008. Perineuronal satellite cells 6:43−51.
in mouse spinal ganglia express the gap junction protein con- Takeda M, Takahashi M, Matsumoto S. 2009. Contribution of the
nexin43 throughout life with decline in old age. Brain Res Bull activation of satellite glia in sensory ganglia to pathological pain.
75:562−569. Neurosci Biobehav Rev 33:784−792.
Puljak L, Kojundzic SL, Hogan QH, Sapunar D. 2009. Lidocaine injec- Tanowitz HB, Machado FS, Jelicks LA, Shirani J, de Carvalho AC,
tion into the rat dorsal root ganglion causes neuroinflammation. Spray DC, et al. 2009. Perspectives on Trypanosoma cruzi-induced
Anesthes Analges 108:1021−1026. heart disease (Chagas disease). Progr Cardiovasc Dis 51:524−539.
Purves D. 1975. Functional and structural changes in mammalian sym- Taylor MK, Yeager K, Morrison SJ. 2007. Physiological Notch signaling
pathetic neurones following interruption of their axons. J Physiol promotes gliogenesis in the developing peripheral and central ner-
252: 429−463. vous systems. Development 134: 2435−2447
Reichenbach A, Siegel A, Senitz D, Smith TG Jr. 1992. A compara- Ten Tusscher MPM, Klooster J, Vrensen GFJM. 1989. Developmental
tive fractal analysis of various mammalian astroglial cell types. patterns of gene expression of secreted proteins in brain and choroid
Neuroimage 1:69−77. plexus. Brain Res 490:95−102.
Rühl A. 2005. Glial cells in the gut. Neurogastroenterol Motil Th i MM, Spray DC, Hanani M. 2008. Aquaporin-4 water channels in
17:777−790. enteric neurons. J Neurosci Res 86:448−456.
Rühl A, Franzke S, Collins SM, Stremmel W. 2001. Interleukin-6 expres- Vanderwinden JM, Timmermans JP, Schiffmann SN. 2003. Glial cells,
sion and regulation in rat enteric glia cells. Am J Physiol—Gastroint but not interstitial cells, express P2X7, an ionotropic purinergic recep-
Liver Physiol 280:G1163−G1171. tor, in rat gastrointestinal musculature. Cell Tiss Res 312, 149−154.
Sarosi GA, Barnhart DC, Turner DJ, Mulholland MW. 1998. Verkhratsky A, Steinhäuser C. 2000. Ion channels in glial cells. Brain
Capacitative Ca2+ entry in enteric glia induced by thapsigargin and Res Rev 32:380−412.
extracellular ATP. Am J Physiol 275:G550-G555. Villa G, Fumagalli M, Verderio C, Abbracchio MP, Ceruti S. 2010.
Savidge TC, Newman P, Pothoulakis C, Ruhl A, Neunlist M, Bourreille Expression and contribution of satellite glial cells purinoceptors to pain
A, et al. 2007. Enteric glia regulate intestinal barrier function and transmission in sensory ganglia: an update. Neuron Glia Biol 6:31−42.
inflammation via release of S-nitrosoglutathione. Gastroenterology Vit JP, Jasmin L, Bhargava A, Ohara PT. 2006. Satellite glial cells in the
132:1344−1358. trigeminal ganglion as a determinant of orofacial neuropathic pain.
Schuppan D, Junker Y, Barisani D. 2009. Celiac disease: from pathogen- Neuron Glia Biol 2:247–257.
esis to novel therapies. Gastroenterology 137:1912−1933. Watkins LR, Maier SF. 2002. Beyond neurons: evidence that immune
Segura BJ, Zhang W, Cowles RA, Xiao L, Lin TR, Logsdon C, et al. and glial cells contribute to pathological pain states. Physiol Rev
2004. Lysophosphatidic acid stimulates calcium transients in enteric 82:981−1011.
glia. Neuroscience 123:687−693. Weick M, Cherkas PS, Härtig W, Pannicke T, Uckermann O, Bringmann
Segura BJ, Zhang W, Xiao L, Turner D, Cowles RA, Logsdon C, et al. A, et al. 2003. P2 receptors in satellite glial cells in trigeminal ganglia
2004. Sphingosine-1-phosphate mediates calcium signaling in guinea of mice. Neuroscience 120:969−977.
pig enteroglial cells. J Surg Res 2116:42−54. Zhang H, Mei X, Zhang P, Ma C, White FA, Donnelly DF, et al. 2009.
Sha L, Miller SM, Szurszewski JH. 2001. Electrophysiological effects Altered functional properties of satellite glial cells in compressed spi-
of GABA on cat pancreatic neurons. Am J Physiol Gastroint Liver nal ganglia. Glia 57:588−1599.
Physiol 280:G324-G331. Zhang W, Sarosi GA Jr, Barnhart DC, Mulholland, MW. 1998.
Silverman WR, de Rivero Vaccari JP, Locovei S, Qiu F, Carlsson SK, Endothelin-stimulated capacitative calcium entry in enteric glial
et al. 2009. The pannexin 1 channel activates the inflammasome in cells: synergistic effects of protein kinase C activity and nitric oxide.
neurons and astrocytes. J Biol Chem 284: 18143−18151. J Neurochem 71:205−212.
Stephenson JL, Byers MR. 1995. GFAP immunoreactivity in trigemi- Zhang W, Sarosi G Jr, Barnhart D, Yule DI, Mulholland MW. 1997.
nal ganglion satellite cells after tooth injury in rats. Exp Neurol Endothelin-activated calcium signaling in enteric glia derived from
131:11–22. neonatal guinea pig. Am J Physiol 272:G1175−G1185.

G L I A L C E L L S I N AU TO N O M I C A N D S E N S O RY G A N G L I A • 133
This page intentionally left blank
SECTION 2
L I N E AG E A N D D E VE L O PM E N T
This page intentionally left blank
12.
ASTROCY TE DEVELOPMENT
James E. Goldman

A B B R E VI AT I O N S different regions varies (e.g., the white matter of the spinal


cord is at its periphery, whereas the white matter of the
bHLH basic helix-loop-helix transcription factor hemispheres is more centrally located). There seem to be
BLBP brain lipid binding protein several general rules, however: (1) Astrocytes, along with
BMP bone morphogenetic protein neurons and oligodendrocytes, originate from ventricu-
CNTF ciliary neuronotropic factor lar zone (VZ) cells in the embryonic CNS. (2) Astrocytes
CT1 cardiotrophin 1 have at least two different origins—one directly from radial
DNMT DNA (cytosine-5)-methyltransferase glial cells and the other indirectly, through a proliferative
EGF epidermal growth factor and migratory population that populates the subventricular
FGF fibroblast growth factor zone (SVZ). (3). A series of soluble and cell surface mole-
IL-6 interleukin 6 cules regulate astrocyte genesis. (4) These molecules induce
LIF leukemia inhibitory factor different combinations of transcription factors that specify
NF1 nuclear factor 1 astrocyte fate and progressively restrict the fate of immature
NGF nerve growth factor cells.
OSM oncostatin M
PACAP pituitary adenylate cyclase activating
polypeptide 2.1 A S T RO C Y T E D EV E L O PM E N T
SVZ subventricular zone IN THE FOREBRAIN
TGF-ß transforming growth factor-ß
2.1.1 Some Astrocytes Arise Directly from
VZ ventricular zone
Radial Glial Cells
The VZ is a pseudostratified epithelium composed of the
1 INTRODUCTION earliest neuroectodermal cells. At about the time when cor-
tical neurons begin to develop (around E11 in mouse), elon-
The developing central nervous system (CNS) gener- gated, radially oriented cells appear in the VZ that share
ates a wealth of astrocytes of different forms and func- many characteristics in common with astrocytes, such as
tions. Therefore, any model of astrocyte development must the GLAST type of glutamate transporter, glycogen gran-
account both for the origins of astrocytes from the imma- ules, brain lipid–binding protein (BLBP), and GFAP (see
ture neuro-epithelium and the wide variety of astrocytes that chapter 5). Radial glia, the historical term for these cells,
populate the adult CNS (see chapter 4). This chapter sum- span the width of the neural tube from ventricular to pial
marizes astrocyte development in several CNS regions and surface, and are found in all regions of the developing CNS.
discusses the molecular regulation of the commitment to an In the cerebral cortex, radial glia generate neurons, primar-
astrocyte fate. ily projection neurons (Malatesta et al. 2000, 2003; Noctor
et al. 2001) (see chapter 5). At a later time, after neuronal
migration has ceased, some of the radial glial cells trans-
2 PAT T E R N S O F A S T R O C Y T E form into astrocytes (Fig. 12.1). We do not know, however,
D E VE L O PM E N T I N D I F F E R E N T R E G I O N S what proportion of astrocytes are generated in this way or if
O F T H E C E N T R A L N E RVO U S SYS T E M : I S those astrocytes have different properties from those gener-
A S T R O C Y T E D E VE L O PM E N T I D E N T I C A L ated from SVZ cells (see section 2.1.2). The generation of
E VE RY W H E R E I N T H E B R A I N ? astrocytes from radial glia likely occurs in all areas of the
CNS, although a direct visualization of this transformation
It is unlikely that the details of astrocyte development are has been made in forebrain (Gaiano et al. 2000; Ventura
identical in every region of the CNS. Development needs and Goldman 2007; Voigt 1989). Fate tracing of radial
to be placed in an appropriate anatomical context, because glia into astrocytes shows that they can generate both gray
different types of astrocytes need to be generated at differ- matter and white matter astrocytes (Ventura and Goldman
ent times in different regions and because the anatomy of 2007).

137
Glial Precursor Migration and Differentiation in Forebrain

AS
OL

OL

OL(NG2)

AS

AS
RG

Radial Glia Tranform into Astrocytes


Postanatal Glial Precursor Migration Paths

Figure 12.1 Near the end of gestation and into postnatal life, glial precursors migrate from the forebrain SVZ into white matter and cortex to become
astrocytes and oligodendrocytes (myelinating oligodendrocytes, NG2+ cells, and immature oligodendrocytes). NG2+ cells reside in white matter
(not shown) as well as cortex. Some of the radial glia transform directly into astrocytes. AS, astrocyte; OL, oligodendrocyte; OL(NG2) NG2+ glia;
RG, radial glia.

2.1.2 Some Astrocytes Do Not Arise Directly from


they acquire the bushy morphology of mature, protoplasmic
Radial Glia, but from Intermediate, Highly
astrocytes. Astrocyte precursors do not appear to develop syn-
Migratory Precursor Cells
chronously, because during early postnatal development, astro-
Other astrocytes arise from immature cells that emigrate cyte morphologies are heterogeneous and sometimes overlap
from the SVZ in the perinatal period and migrate extensively (Fig. 12.4) (see Bushong et al. 2004 for a study of astrocyte
through forebrain to colonize both gray and white matter (see maturation in the hippocampus). It takes about 1 month to
Fig. 12.1) (Levison and Goldman 1993; Marshall et al. 2003). reach mature shapes and mature cellular domain boundaries
Although these cells initially must originate from radial glial (Bushong et al. 2004) (see chapter 4).
cells, how are they generated and from where do they colonize These migratory SVZ cells arise from both dorsal and ven-
the SVZ? tral areas of the developing forebrain. Ventrally derived SVZ
The forebrain SVZ at this time is populated by a large cells migrate dorsally along the ventricle to form the large SVZ
number of highly migratory and proliferating cells, whose at the dorsolateral angle of the lateral ventricle in perinatal life
fates include neurons and oligodendrocytes and astrocytes (Marshall and Goldman 2002). This dorsolateral SVZ popu-
(Levison and Goldman 1993; Luskin and McDermott 1994; lation gives rise to astrocytes and oligodendrocytes of the cor-
Luskin et al. 1993). Although SVZ cells do not appear to tex, white matter, and striatum, as well as to interneurons of
express well-known astrocyte markers within the SVZ itself, the olfactory bulb (Levison and Goldman 1993; Luskin and
they belong to the olig2+, gliogenic population, which gener- McDermott 1994; Luskin et al. 1993). The ventral origins of
ates astrocytes and oligodendrocytes ( Jang and Goldman 2011; some interneurons, oligodendrocytes, and some astrocytes
Marshall et al. 2005). Those gliogenic SVZ cells that develop raise the question of whether glia and interneurons might
along an astrocyte lineage begin to express early markers such arise from common progenitors. This is difficult to answer
as Zebrin II (aldolase C), vimentin, nestin, and GFAP after now, although cells isolated from the embryonic lateral gang-
they exit from the SVZ (Staugaitis et al. 2001; Zerlin et al. lionic eminence and placed in culture will generate clones
1995). The astrocyte precursors migrate extensively and con- composed of neurons and oligodendrocytes (He et al. 2001),
tinue to divide as they migrate (Fig. 12.2), and then acquire but interestingly, not astrocytes, at least under those particular
further markers depending in part on whether they settle in conditions. In the neonatal period, forebrain SVZ cells placed
white matter or gray matter (see section 2.1.3). in culture give rise to clones containing neurons and glia, but
One of the earliest interactions of astrocyte precursors is under these conditions the glia are both astrocytes and oligo-
with blood vessels (Zerlin and Goldman 1997), which sug- dendrocytes (Levison and Goldman 1997). Thus, at least some
gests they already express molecules that recognize endothelial of the immature cells from these regions have the potential to
cells or basal laminae (Fig. 12.3). Astrocytes then develop far differentiate into both neurons and glia—whether they actu-
more complex shapes with multiple branched processes until ally do in vivo is not known.

138 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
ZII
GLAST, GLT-I
Vimentin (low)
Nestin (low)
ASTROCYTE DEVELOPMENT FROM FOREBRAIN SVZ CELLS GFAP(low)
S100β
YbGST
OLIGODENDROCYTES Aquaporin4
lon Channels
Aldh1

“Protoplasmic”
TO GRAY MATTER
SVZ

PSA-NCAM
ZII
GLAST
OLIG1,2 “Fibrous”
ZII
TO WHITE MATTER
GLAST, GLT-I (low)
Other Transporters
Vimentin (low)
Nestin (low)
GFAP(high)
S100β
YbGST
Aquaporin4
lon Channels
CD44
Aldh1

Figure 12.2 Astrocyte precursors migrate from the neonatal SVZ into white matter and gray matter. Early steps include contact with blood vessels (see
Fig. 12.3). Astrocytes that settle in white matter become “fibrous” astrocytes, and those in gray matter “protoplasmic” astrocytes, to use the classical
terminology. The latter interact with both blood vessels and neurons, including synapses. Some of the proteins found in the cells are given in red. SVZ,
subventricular zone

Astrocytes contact blood vessels at an early stage


of astrocyte development

A B

BV

Figure 12.3 Astrocyte precursors contact blood vessels early in their Figure 12.4 Astrocyte precursors in the cortex do not differentiate
development. Images are from retroviral labeled cells that migrated from synchronously. Image taken 7 days after X-gal–expressing retrovirus was
neonatal forebrain SVZ into the cortex (see Zerlin and Goldman 1997). injected into the neonatal forebrain SVZ. 2 astrocytes have become more
mature and protoplasmic (*); precursor contacting a blood vessel (BV)
(arrow); two precursors next to each other, each with a limited arborization
Glia lineages in vivo have been studied using (arrowhead). Bar: 100 μ.
replication-deficient retroviruses, an approach that can deter-
mine lineage relationships by defining the set of cells that
arise from the viral infection of a single, dividing progeni- early postnatal period, dividing progenitors give rise largely to
tor. In some cases, both neurons and glia arise from the same Muller glia and photoreceptor cells. Individual progenitors in
progenitor. For example, single progenitors in the embryonic the embryonic neocortex and the striatum can generate both
retina give rise to neurons and Muller glia (Turner and Cepko astrocytes and neurons (Halliday and Cepko 1992; Walsh
1987). The developmental potential of retinal cells becomes and Cepko 1993), possibly through radial glial intermedi-
restricted over time, however, and in the late embryonic and ates. Most studies of gliogenesis find that retroviral-labeled

A S T R O C Y T E D E VE L O PM E N T • 139
cells accumulate in groups, or clusters, which are in most 2.2 A S T RO C Y T E D EVE L O PM E N T I N T H E
cases homogeneous—either astrocytic or oligodendrocytic. C E R E B E L LU M
However, a small proportion of clusters, about 15%, are The cerebellum contains a variety of astrocyte forms, includ-
mixed, either containing astrocytes and oligodendrocytes, or ing the fibrous astrocytes of white matter, velate astrocytes of
more rarely, neurons and glia (Grove et al. 1993; Levison and the internal granule cell layer, and Bergmann glia, which send
Goldman 1993; Luskin and McDermott 1994; Luskin et al. radially directed processes from the cell bodies in the Purkinje
1988, 1993; Parnevales 1999; Price and Thurlow 1988). Most cell layer to end at the pial surface (Palay and Chan-Palay
if not all of these clusters are clonal (Zerlin et al. 2004). The 1974).
presence of mixed astrocyte–oligodendrocyte clusters tells Early morphological and 3H-thymidine studies suggested
us that a small proportion of the gliogenic SVZ cells may
that some Bergmann glia are generated from other Bergmann
be specified to a glial lineage, but not necessarily to either
glia (because they incorporate thymidine) and/or some pro-
an astrocyte or an oligodendrocyte lineage as they emigrate
genitor, of an unknown type (Basco et al. 1977; Choi and
from the SVZ.
Lapham 1980). It is likely that some Bergmann glia as well as
Analysis of gliogenesis is complex because of spatial dis-
other astrocytes arise from embryonic radial glia of the cer-
persion of members of a clone. Note that immature astrocytes
ebellum. Some astrocytes, including Bergmann glia, share a
and oligodendrocytes continue to divide as they migrate, but
common lineage with Purkinje cells, as determined by retro-
the two progeny of a dividing cell may not necessarily con-
viral tracing in chick (Lin and Cepko 1999).
tinue to migrate together, and therefore would not necessarily
During the perinatal period, astrocyte progenitors, along
end up in the same cluster. Indeed, members of the same clone
with oligodendrocyte and interneuron progenitors, arise
can disperse widely (Zerlin et al. 2004). A direct visualization
in the base of the cerebellum, just dorsal to the fourth ven-
of migrating glia shows that cells cease migration before they
tricle, and migrate through the white matter in a largely
divide, and recommence migration after they divide, the two
radial direction (Miyake et al. 1995; Zhang and Goldman
progeny moving off in different directions. Thus related astro-
1996) (Fig. 12.5). At least some of these progenitors in the
cytes can become spatially separated.
white matter begin to differentiate into astrocytes as they
migrate, because they express astrocyte characteristics, such
2.1.3 Astrocyte Development in the Optic Nerve as GLAST (Milosevic and Goldman 2002). Astrocyte pre-
cursors in the cerebellum express CD44, a surface recep-
In the optic nerve astrocytes and oligodendrocytes are gener- tor for hyaluronan and osteopontin (Cai et al. 2011), in this
ated around the end of gestation and for the first week or two respect similar to astrocyte precursors in the spinal cord (Liu
of life in rodents. Optic nerve astrocytes appear to be intrinsic
to the nerve, and likely arise from radial glia, which are derived
in turn from the initial optic nerve neuroepithelium. The con-
version of the precursors into astrocytes goes through a stage
during which they express the vimentin type of intermediate
filament and the gangliosides recognized by the monoclonal
antibody, A2B5, but do not express either GFAP or S-100beta,
markers of more mature astrocytes in rodents (Mi and Barres SC
1999). In vivo the vimentin+ cells will eventually also express
GFAP. In culture, the astrocyte progenitors can be induced
bc oI
to express GFAP by ciliary neuronotropic factor (CNTF) or
leukemia inhibitory factor (LIF). as gc
Oligodendrocyte precursors, which originate in the SVZ
at the base of the third ventricle and migrate into and along bg
the nerve (reviewed in Miller 2002), display a glial develop- as
mental plasticity. If removed from the neonatal optic nerve
and cultured in the presence of serum or IL-6/LIF/CNTF
family members they develop into a stellate astrocyte type,
the so-called “type 2 astrocytes” (Raff et al. 1983). The “bipo- oI
tential” nature of these oligodendrocyte precursors reveals
that the environment of the developing optic nerve either Migration and Differentiation of
Neuronal and Glial Precursors
promotes their differentiation into oligodendrocytes and/or from Cerebellar White Matter to
Pro
inhibits their differentiation into astrocytes. This pattern of Cortex
immature glia differentiating largely into oligodendrocytes in
white matter also seems to be followed elsewhere in the fore- Figure 12.5 During late gestation and into postnatal life, glial and
brain, because the large majority of SVZ cells that settle in neuronal precursors (Pro) generated at the base of the cerebellum migrate
through white matter into cortex and differentiate into oligodendrocytes
subcortical white matter in the neonatal period differentiate (ol), several astrocyte forms (including Bergmann glia (bg) and velate
into oligodendrocytes, not astrocytes (Levison and Goldman astrocytes (as), and interneurons. bc, basket cells; gc, granule cells; sc,
1993). stellate cells. From Zhang L, Goldman JE 1996.

140 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
et al. 2004) and chick cord (Alfei et al. 1999). Interestingly, spinal cord, consistent with the idea that these genes inhibit
hyaluronan in the early postnatal cerebellum is concentrated astrocyte development (Lu et al. 2000; Zhou et al. 2000).
in white matter and arranged in fiberlike structures (Baier However, developing astrocytes in other regions of the CNS
et al. 2007), possibly providing migratory guides for astrocyte do express olig2 (Marshall et al. 2005).
precursors. How astrocytes arise in cord is not fully understood. By
analogy to the radial glial–astrocyte transformation in the
forebrain (see the preceding), it seems likely that at least some
2.3 A S T RO C Y T E D EV E L O PM E N T
of the cord astrocytes arise from VZ cells via radial glia.
I N T H E S P I NA L C O R D
The preceding considerations argue for a common lineage
The ventricular zone of the embryonic mammalian spinal cord of oligodendrocytes with some neurons and a separate lineage
is divided into a number of discrete domains along the dor- for astrocytes in the cord. Nevertheless, Rao et al. (1998) have
soventral axis, defined by molecular markers, including tran- isolated a progenitor from embryonic spinal cord that appears
scription factors and growth factor receptors. These markers restricted to glial lineages, able to generate both astrocytes and
have allowed genetic studies to trace the fates of immature oligodendrocytes in culture, but not neurons. These cells bind
cells originating in each of these domains. Thus, the spinal the monoclonal antibody A2B5 and express the hyaluronan
cord has been the major CNS area in which to approach the receptor, CD44 (Liu et al. 2004). They can be isolated from both
question of whether an astrocyte fate is specified by the initial dorsal and ventral regions of the cord. These observations do not
position of a precursor within the ventricular zone. Pringle appear to coincide with observations in vivo, as noted. There are
et al. (2003) inferred a widespread generation of astrocytes at least two ways to harmonize these findings. First, one can argue
from both dorsal and ventral cord, observing a widespread that development is highly regulated in spatial and temporal pat-
expression of fibroblast growth factor receptor type 3 (Fgfr3) terns and thus immature cells are prevented from assuming all of
in the embryonic neuroepithelium. Fgfr3+ cells, which do not their potential fates. Removing progenitors from their normal
represent oligodendrocyte precursors and apparently not neu- environment relieves fate restrictions to some degree and allows
ronal precursors either, likely correspond to very early astrocyte progression through lineages not otherwise taken. Second, one
precursors. However, other observations suggest a positional could argue that there are indeed three lineages for oligodendro-
determination of astrocyte fate. Thus, Muroyama et al. (2005) cytes (which might in fact give rise to different oligodendrocyte
show a generation of astrocytes from the ventral p2 domain, populations), one in common with motor neurons, one from the
which also generates spinal interneurons. Astrocyte develop- p3 domain, and the other in common with astrocytes.
ment in this domain depends on the expression of Scl (Stem
cell leukemia, a bHLH transcription factor). Further evidence
for positional specificity in astrocyte development comes from 3 A S T R O C Y T E D E VE L O PM E N T I N T H E
the observation that subpopulations of astrocytes in the white A D U LT C E N T R A L N E RVO U S SYS T E M
matter of the ventral cord can be distinguished by the combina-
tions of Reelin and Slit expression (Hochstim et al. 2008). The Astrocytes are generated in the adult CNS, although at a low
expression patterns are identical to the patterns of these genes rate. Early studies used 3H-thymidine to estimate glial turn-
in the embryonic neuroepithelium, indicating a topographi- over in the adult rodent brain, concluding that astrocytes
cal representation of neuroepithelium onto its final astrocyte and oligodendrocytes continue to be generated. However, it
products. Reelin expression is dependent on the expression of appears as if the numbers of oligodendrocytes increase slowly,
Pax6, and the deletion of Pax6 greatly reduces the numbers of while the numbers of astrocytes remain approximately con-
Reelin+ astrocytes, but does not alter the numbers of astro- stant (Hommes and Leblond 1967; Kaplan and Hinds 1980;
cytes. Thus, Pax6 specifies one characteristic of a set of white Korr et al. 1973; Paterson 1983). A similar conclusion was
matter astrocytes, but does not disrupt astrocyte development drawn from retroviral labeling, which showed an increase in
per se. These findings indeed link initial positional specifica- the size of oligodendrocyte clonal clusters but not astrocyte
tion to astrocyte subtypes. It is not known, however, whether clusters over time (Levison et al. 1999).
these astrocyte subtypes differ in any other ways. New astrocytes may arise either from the proliferation
Other evidence for a separation of astrocyte from oligo- of mature astrocytes or the differentiation of progenitors.
dendrocyte lineages includes the observation that oligoden- The bulk of evidence strongly favors the latter possibility.
drocytes in the ventral cord require sonic hedgehog (Shh) for Although astrocytes divide in pathological states (Norton
their development (Orentas et al. 1999), whereas astrocytes 1999), there is little evidence that mature astrocytes divide in
do not (Pringle et al. 2003). However, in olig1/olig2 double the unperturbed brain. A variety of immature glial cells, how-
knockout (–/–) mice, in which oligodendrocytes do not ever, populate the adult CNS. Thymidine-labeling studies (see
develop, fate mapping studies show astrocytes arising from the preceding) all described proliferating cells that did not
cells that had originated in the domain in which olig2 would have characteristics of mature astrocytes or oligodendrocytes,
have been normally expressed (Zhou and Anderson 2002). based on nuclear morphology and ultrastructural characteris-
This observation raises the possibility that the Olig factors tics. It was difficult at the time to place all of these cells into
normally repress astrocyte differentiation in a population that specific lineages, however, and the more recent use of antigenic
develops into oligodendrocytes and neurons. Neither olig1 markers has begun to sort out the nature of these progenitors.
nor olig2 appears to be expressed in astrocytes in the normal The thymidine studies estimated that cycling cells constituted

A S T R O C Y T E D E VE L O PM E N T • 141
a small but significant proportion of the total cell number. For to date in gene expression profiling from astrocytes isolated
example, proliferating cells in the adult rodent white matter from the developing and adult mouse forebrain (Cahoy et al.
make up as much as 2% of the total (Paterson 1983). 2008). In general, genes promoting or allowing cell prolifera-
Populations isolated directly from adult CNS (Gensert and tion are decreased and genes referable to astrocytes’ functions
Goldman 2001; Nunes et al. 2003) and enriched for immature in neurotransmitter uptake and processing, lipid synthesis and
cells are heterogeneous, expressing various combinations of mark- other metabolic pathways, and secretion are more strongly
ers, including A2B5, O4, and vimentin, although not mature expressed. Much of the changes in gene expression occur before
glial markers. In culture or after transplantation into brain, these p17 in mouse forebrain, a time at which astrocytes are consid-
cells can generate glia and neurons, although the majority appear ered to have reached a mature state. The Cahoy et al. (2008)
to belong to the oligodendrocyte lineage. Whether astrocytes are paper makes the additional point that some genes expressed by
generated from O4+ cells seems less likely, although Armstrong astrocytes in culture do not match those expressed in acutely
et al. (1992) isolated the rare cell from adult human white mat- isolated cells and vice versa.
ter that became both O4+ and GFAP+ in culture. Similarly, the
A2B5+ cells isolated from the adult CNS differentiate into oli-
godendrocytes in culture, although they can develop into astro- 5 ASTROCY TES ARE
cytes under the appropriate culture conditions (Wolswijk and M O R P H O L O G I C A L LY A N D
Noble 1989, 1992). Thus, most immature cells isolated from the F U N C T I O N A L LY H ET E R O G E N E O U S .
adult CNS appear to be oligodendrocyte precursors, not astro- H OW I S T H I S H ET E R O G E N E I T Y
cyte precursors, although changes in culture environment reveal G E N E R AT E D ?
developmental plasticity. Whether this developmental plasticity
occurs in vivo under pathological circumstances is not clear. Astrocytes comprise a highly heterogeneous population, rang-
ing from radial forms such as Bergmann glia to the bushy pro-
toplasmic astrocytes of gray matter to the less complex fibrous
3.1 G E N E R AT I O N O F A S T RO C Y T E S
astrocytes of white matter (see chapter 4). The molecular het-
FRO M N G2+ G L I A
erogeneity of astrocytes includes electrophysiological charac-
Glia expressing a chondroitin sulfate proteoglycan recognized teristics, neurotransmitter transporters and receptors, levels of
by the NG2 antibody populate the developing and adult CNS, enzymes that catabolize neurotransmitters, gap junction cou-
displaying a lacy shape with many delicate processes (see chapters pling, degree and frequency of calcium transients, GFAP levels,
10, 13, and 21). Although the NG2 marker is found on oligo- and levels of receptors that interact with extracellular matrix
dendrocytes during their early development (Nishiyama et al. (see, for example, chapters 16, 17, 24, and 28; and as reviewed in
1996), the NG2+ glia in the adult brain do not myelinate and do Kimelberg 2010; Zhang and Barres 2010). Transcriptional pro-
not display mature oligodendrocyte or astrocyte characteristics. filing that compared astrocyte cultures with astrocytes derived
In terms of lineage, most investigators believe that the NG2+ from neurospheres to total transcripts from different regions of
population is related to oligodendrocytes, rather than astrocytes. the CNS (Bachoo et al. 2004) showed transcripts that many
Fate mapping of NG2+ cells in the postnatal brain reveals that astrocytes expressed in common as well as transcripts expressed
they generate either more NG2+ cells or more mature oligoden- in a heterogeneous way. More recent transcriptional analyses
drocyte precursors (Zhu et al. 2011). However, NG2+ cells in using astrocyte cultures established from different CNS regions
the embryonic brain produce oligodendrocytes and astrocytes (Yeh et al. 2009) or astrocytes acutely isolated from the adult
that populate the ventral forebrain gray matter (Zhu et al. 2011). mouse neocortex by fluorescence-activated cell sorting (Lovatt
This and other studies in the developing and lesioned CNS indi- et al. 2007), or astrocyte ribosome–associated mRNAs from
cate that NG2+ cells largely belong to the oligodendrocyte lin- the ALDHL1-BAC-TRAP mice (Doyle et al. 2008) have also
eage (Komitova et al. 2011; Zhu et al. 2008). It is possible that demonstrated gene expression common to astrocytes, patterns
the population of NG2+ cells is a heterogeneous one, especially distinctly different from neurons and oligodendrocytes, as well
during the embryonic development of the CNS. as region-specific expression patterns (see chapter 28). The
To make matters more complex, there is a population of differences in astrocyte phenotypes are critical to understand
NG2+/vimentin+ cells in the adult rat spinal cord (Horner astrocyte function, but from a developmental point of view
et al. 2000). These appear to be relatively simple cells that the studies do not tell us whether the astrocyte progenitors are
assume a radial orientation and do not look at all like the lacy programmed because of their initial location to acquire spe-
cells in the brain. Thus, cells expressing NG2 may differ in dif- cific characteristics or whether the local environment regulates
ferent regions of the adult CNS. some of the characteristics, and if so, which ones.
How is this heterogeneity established and when is it estab-
lished during the development of astrocytes? There are two
4 TEMPOR AL CHANGES IN ASTROCY TE general ways of thinking about the development of astrocyte het-
GENE TR ANSCRIPTION DURING erogeneity. In one model, the specification of astrocyte fate and
D E VE L O PM E N T the specific characteristics of a given astrocyte are determined
early, during the patterning of the neuroepithelium. Positional
Astrocyte precursors change their gene expression patterns information, determined by dorsoventral gradients and interac-
as they differentiate. The sequence has best been described tions among transcription factors regulate an astrocytes fate and

142 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
type. In another model, the neuroepithelium generates astrocyte 1991; Kaaijk et al. 1997), but how adult levels are regulated and
precursors and the specific type(s) of astrocytes are determined how they are related to precursor levels is not known.
later, either during their migration or even where they finally
reside. It is likely that both mechanisms exist.
As noted, spinal cord astrocytes are heterogeneous in some 6 A S T R O C Y T E D E VE L O PM E N T I S
ways that reflect positional information and combinatorial R E GU L AT E D BY S E VE R A L C L A S S E S O F
codes of transcription factors in the embryonic neuroepithe- M O L E C U L E S A N D I N T R AC E L LU L A R
lium. Thus, in cord, some degree of astrocyte subtype specifi- PAT H WAYS
cation is determined early. The heterogeneity in Reelin and Slit
expression in cord astrocytes may or may not be accompanied For astrocytes, attention has focused on the IL-6/LIF fam-
by heterogeneity in other astrocyte phenotypes, which could ily of cytokines and the LIF receptor/gp130 pair, the TGF-β
be regulated in their initial position in the neuroepithelium or growth factor family, particularly bone morphogenetic pro-
during precursor migration or at their final destinations. teins (BMPs) and BMP receptors, fibroblast growth factor,
Other observations in other parts of the CNS suggest that the and the Notch and Notch ligand pairs.
fate decision to differentiate along an astrocyte lineage is made
well before the decision to develop into a specific astrocyte sub-
6.1 IL-6 Family Members
type. Thus, there may be a basic astrocyte type, on which is then
layered a series of transcriptional changes to provide astrocytes The IL-6 family of proteins includes ciliary neuronotropic fac-
functionally appropriate for the area in which they reside. In fact, tor (CNTF), leukemia inhibitory factor (LIF), cardiotrophin 1
the final decision of form and function may not be made until (CT1), and oncostatin M (OSM). All of these stimulate astro-
an astrocyte precursor has reached its final destination. Several cytic differentiation, usually defined as the induction of GFAP,
observations suggest this is the case during some astrocyte devel- in cells cultured from the embryonic CNS or from the neo-
opment in the forebrain and possibly cerebellum. Astrocyte natal optic nerve (Bonni et al. 1997; Gard et al. 1995; Hughes
colonization of the forebrain from migratory precursors in the et al. 1988; Johe et al. 1996; Ochiai et al. 2001; Yanagisawa et al.
SVZ appears to be a more random situation than one sees in the 1999). These ligands signal through the LIF receptor/gp130
patterned neuroepithelium of the spinal cord (see the preced- complex (Nakashima et al. 1999a), to activate the JAK-STAT
ing). In neonatal rodent forebrain a clonal analysis of SVZ cells intracellular signaling pathway (Bonni et al. 1997; Kahn et al.
revealed radial migration of glial precursors into white matter 1997). This pathway is linked to GFAP regulation, because
and cortex, with some degree of tangential migration in cortex phosphorylated JAK then phosphorylates STAT3, a transcrip-
(Zerlin et al. 2004) and the generation of astrocytes in white tion factor, which then binds to CBP/p300 complex to bind
matter, cortex, at the pial surface and in the striatum. Thus, a to and activate the S3BE sequence in the GFAP promoter
single precursor can generate different types of astrocytes that (Yanagisawa et al. 2001) (Fig. 12.6). IL-6 family members act
reside in different areas. Fate tracing of dorsal radial glial cells synergistically with the extracellular matrix of mesenchymal
in the neonatal mouse forebrain showed that these generated cells (Lillien and Raff 1990) in inducing GFAP expression. As
astrocytes in both cortex and subcortical white matter (Ventura noted, astrocytes have extensive interactions with basal lami-
and Goldman 2007). A dramatic display of astrocyte plastic- nae of blood vessels and the pial surface of the brain. In fact,
ity comes from experiments in which SVZ cells from neonatal cerebral endothelial cells do express LIF (Mi et al. 2001). Such
rats were transplanted into the neonatal cerebellar white mat- an interaction might help induce astrocyte differentiation, a
ter, where they generated (morphologically) fibrous astrocytes mechanism that could appropriately match the numbers of
in white matter, velate astrocytes in the internal granule layer, astrocytes with vessels (Zerlin and Goldman 1997).
and Bergmann glia (Milosevic et al. 2008). Thus, forebrain glial However, bear in mind that astrocyte differentiation must
precursors can give rise to cerebellar-specific astrocyte forms. involve the induction of many genes other than GFAP. Indeed,
The transplanted astrocytes were not analyzed for other charac- some of the GFAP-negative progenitors that migrate through
teristics, however, so it is not known whether the transplanted the forebrain and cerebellum express astrocyte markers such as
cells were able to generate cerebellar astrocytes with full fidelity. the glutamate transporter, GLAST (Milosevic and Goldman
Finally, astrocyte precursors derived from the neonatal rat SVZ 2002) or zebrin II (Staugaitis et al. 2001) before contacting
(A2B5+, expanded in vitro with FGF) transplanted into neona- basal laminae. Thus, early stages of astrocyte development may
tal rat forebrain SVZ and white matter generate astrocytes with begin before mesenchymal interactions.
a variety of morphologies (Lin and Goldman 2009). The final
phenotype of a given astrocyte thus may be regulated both by its
6.2 Transforming Growth Factor-β Family Members
initial position and by the local environment in which it comes
to reside. All of this speaks to the high degree of developmental The TGF-β family of proteins, particularly BMP2 and BMP7,
plasticity of astrocyte precursors. promote astrocyte development from cells of the embryonic
Given that (some) astrocyte precursors in spinal cord and cer- telencephalon (Gross et al. 1996). These ligands bind to BMP
ebellum express CD44, it is interesting that astrocytes in the adult receptors to activate Smad transcription factors (see Fig. 12.6).
CNS vary considerably in their CD44 levels. In general white The BMPs can act synergistically with IL-6 family members
matter and subpial astrocytes express far higher amounts of CD44 (Nakashima et al. 1999c), inducing a complex that has both
than do gray matter astrocytes (Akiyama et al. 1993; Girgrah et al. Smad and STAT3 (Nakashima et al. 1999b). In addition to

A S T R O C Y T E D E VE L O PM E N T • 143
Induction of Astrocyte Genes by IL-6, TGF-β and Notch Pathways to be early committed neurons (Namihira et al. 2009). This find-
IL-6/LIF/CNTF BMP Notch Ligand
ing provides a rationale for the neocortical developmental pattern
in which projection neurons are generated first from radial glia,
and then followed by astrocytes.
An example of Notch pathway interactions with proglial
transcription factors is given by the transcription factor, nuclear
pSmad Notch Receptor
factor 1 (NF1), which binds to the GFAP promoter (Cebolla
P-JAK
and Vallejo 2006) and appears in the cortex at about the stage of
astrocyte development. In spinal cord, NF1A and B appear ear-
P-STAT
lier, at E10 in the VZ and thereafter serve to promote gliogenesis
Hes activation and repress neurogenesis (Deneen et al. 2006). Nuclear factor
1A function requires Notch activation and the expression of the
downstream Hes genes (Deneen et al. 2006), thus coordinating
CBP/p300
NF1 Transcription NF1A and Notch promotion of astrocyte genesis.
Me Me
Me Me Demethylation
Astrocyte gene promoter Astrocyte Gene Promoter 6.4 F I B RO B L A S T G ROW T H FAC TO R S I G NA L I N G

Figure 12.6 Members of the IL-6, TGF-E (BMP) families induce Members of the fibroblast growth factor (FGF) family have
astrocyte genes through Jak/Stat and Smad activation pathways. Notch important effects on astrocyte development. They appear
activation regulates NF1 binding. Promoter methylation (Me) prevents to have multiple effects, in some contexts regulating the dif-
transcription factor binding. NF1 binds to a different site than does the ferentiation and proliferation of astrocyte precursors, Thus,
CBP/p300 complex. FGF1 can upregulate astrocyte functions such as ATP release
and coupling via gap junctions (Garré et al. 2010) and FGF2
inducing astrocyte differentiation, BMP2 may be responsible upregulates glutamate transport in cultured forebrain astro-
for inhibiting neuronal differentiation—that is, controlling cytes (Figiel et al. 2003). Figiel et al. (2003) also found
a switch from one fate to the other, or promoting astrocyte increases in glutamate transport with other growth factors,
development at the expense of neurogenesis. In favor of this including EGF, PACAP, and TGF-α, suggesting final com-
idea is that BMP2 upregulates Id1, Id3, and Hes-5 in embry- mon pathways for growth factor regulation of transporters.
onic brain cells in culture (Nakashima et al. 2001). The inhibi- Fibroblast growth factor 1 changes astrocyte morphology to
tor of differentiation genes (“Id” genes) plays important roles a stellate shape and increases the release of NGF in cultured
in repressing specific differentiation pathways, in this case, astrocytes (Cassina et al. 2005). Fibroblast growth factor 2 has
neurogenic pathways. These two Ids and Hes-5, which lie in a strong proliferative effect on astrocyte precursors cultured
the Notch signaling pathway, inhibit the expression of the from neonatal rat forebrain, although these precursors turn
neurogenic bHLH genes, mash1 and neurogenin. off the proliferative effect even in the continued presence of
An interesting question is whether IL-6 and TGF-β FGF2 (Lin and Goldman 2009). Other studies suggest that
ligands promote the transcription of identical genes in devel- FGF2 can promote an astrocyte differentiation from imma-
oping astrocytes. This in fact may not be the case, because ture cortical progenitor cells and astrocyte proliferation, the
treatment of glial restricted precursors with CNTF or BMPs latter through MAP kinase signaling (Kang and Song 2010).
results in astrocytes with different protein profiles and differ- Clearly, progenitors are subjected to a large variety of sig-
ent abilities to promote axonal growth and neuronal survival nals and the interactions among these signals results in fate
after spinal cord injury (Davies et al. 2011). Bone morphoge- decisions. For example, when neurogenin1 is overexpressed
netic proteins induce GFAP, N-CAM, and CD44 expression in embryonic neural cells, it not only promoted neurogenesis,
in mouse embryo cells to a higher degree than does TGF-β1 but also inhibited the cells from differentiating into astrocytes,
(D’Alessandro et al. 1994). even when stimulated by LIF (Sun et al. 2001). If neurogenin 1
competes with STATs for binding to the CBP/p300 complex,
then one can think of a model of fate determination in which
6.3 N OTC H A N D N OTC H L I G A N D S
the relative levels of transcription factors are the critical vari-
Members of the Notch family of transmembrane receptors and ables (Sun et al. 2001). Consistent with this idea is the finding
their ligands, jagged and delta, play an important role in the devel- that the mouse double knockout of neurogenin 2 and Mash 1
opment of radial glia and astrocytes. Notch 1, when constitutively did not contain large numbers of astrocytes at the expense of
expressed in an activated form in VZ cells of the E9.5 mouse CNS, neurons (Nieto et al. 2001).
strongly promotes an astrocyte fate (Gaiano et al. 2000), instead
of the normal neuronal and astrocyte fates. Similarly, expressing
6.5 C YC L I C A D E N O S I N E MO N O P H O S P H AT E
Notch 1 in cells of the embryonic retina strongly promotes dif-
R EGU L AT I O N O F A S T RO C Y T E D EVE L O PM E N T
ferentiation of Muller glial cells, apparently at the expense of rod
photoreceptors (Furukawa et al. 2000). Thus, as in invertebrates, Another astrogenic pathway may function through cAMP,
Notch activation appears to promote a glial fate, while inhibiting because the induction of cAMP in embryonic forebrain cells
a neuronal fate. Indeed, the source of the Notch ligands appears induces an astrocytic fate (McManus et al. 1999). Cyclic

144 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
adenosine monophosphate is well known to increase the tran- REFERENCES
scription of GFAP in astrocytes (Masood et al. 1993; Shafit-
Zagardo et al. 1988), possibly via the CREB sequence in the Akiyama H, Tooyama I, Kawamata T, Ikeda K, McGeer PL. 1993.
GFAP promoter (Besnard et al. 1991; Masood et al. 1993). Morphological diversities of CD44 positive astrocytes in the cere-
bral cortex of normal subjects and patients with Alzheimer’s disease.
Pituitary adenylate cyclase-activating polypeptide (PACAP) Brain Res 632:249–259.
promotes GFAP transcription through PAC1 receptor, which Alfei L, Aita M, Caronti B, De Vita R, Margotta V, Medolago Albani
is coupled to adenylate cyclase (Vallejo and Vallejo 2002). L, et al. 1999. Hyaluronate receptor CD44 is expressed by astrocytes
in the adult chicken and in astrocyte cell precursors in early develop-
ment of the chick spinal cord. Eur J Histochem 43:29–38.
7 E P I G E N ET I C R E GU L AT I O N O F Armstrong, RC, Dorn HH, Kufta CV, Friedman E, Dubois-Dalcq ME.
A S T R O C Y T E D E VE L O PM E N T 1992. Pre-oligodendrocytes from adult human CNS. J Neurosci
12:1538–1547.
Bachoo RM, Kim RS, Ligon KL, Maher EA, Brennan C, Billings N, et
The demethylation of astrocyte genes appears to be an impor- al. 2004. Molecular diversity of astrocytes with implications for neu-
tant prerequisite to the onset of astrocyte differentiation. Thus, a rological disorders. Proc Natl Acad Sci U S A 101:8384–8389.
number of astrocyte genes are methylated early in development, Baier C, Baader SL, Jankowski J, Gieselmann V, Schilling K, Rauch U,
including GFAP, Aldolase C, and Kir4.1 (Hatada et al. 2008, et al. 2007. Hyaluronan is organized into fiber-like structures along
who compared methylation status of neural precursors from migratory pathways in the developing mouse cerebellum. Matrix Biol
26:348–358.
E11.5 with those at E14.5 and with P1 astrocytes). Methylation Basco E, Hajos F, Fulop Z. 1977. Proliferation of Bergmann-glia in the
of astrocyte genes in the early embryonic CNS is regulated by developing rat cerebellum. Anat Embryol (Berl) 151:219–222.
the methyl transferase, DNMT1, and serves to repress astrocyte- Besnard F, Brenner M, Nakatani Y, Chao R, Purohit HJ, Freese E. 1991.
specific gene transcription (Fan et al. 2005). Demethylation, Multiple interacting sites regulate astrocyte-specific transcription
which occurs at the beginning of astrocyte development, allows of the human gene for glial fibrillary acidic protein. J Biol Chem
266:18877–18883.
LIF and TGF-β ligands to upregulate astrocyte genes by allow- Bonni A, Sun Y, Nadal-Vicens M, Bhatt A, Frank DA, Rozovsky I, et al.
ing transcription factors downstream of these signaling pathways 1997. Regulation of gliogenesis in the central nervous system by the
to bind to astrocyte gene promoters (see Fig. 12.6) (Fan et al. JAK-STAT signaling pathway. Science 278:477–483.
2005; Hatada et al. 2008; Namihira et al. 2009; Takizawa et al. Bushong EA, Martone ME, Ellisman MH. 2004. Maturation of
2001). As another example of epigenetic control, FGF2 changes astrocyte morphology and the establishment of astrocyte domains
during postnatal hippocampal development. Int J Dev Neurosci
histone methylation at the STAT binding site on the GFAP pro- 22:73–86.
moter, allowing STAT to bind and activate GFAP transcription Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson
by CNTF (Song and Ghosh 2004). KS, et al. 2008. A transcriptome database for astrocytes, neurons,
and oligodendrocytes: a new resource for understanding brain devel-
opment and function. J Neurosci 28:264–278.
8 S U M M A RY A N D P E R S P E C T I VE S Cai N, Kurachi M, Shibasaki K, Okano-Uchida T, Ishizaki Y. 2011.
CD44-positive cells are candidates for astrocyte precursor cells in
Among a number of outstanding questions concerning astro- developing mouse cerebellum. Cerebellum 129(5):1126–1136.
Cassina P, Pehar M, Vargas MR, Castellanos R, Barbeito AG, Estévez
cyte development are the following: AG, et al. 2005. Astrocyte activation by fibroblast growth factor-1
and motor neuron apoptosis: implications for amyotrophic lateral
1. Some genes are transcribed early in astrocyte development sclerosis. J Neurochem 93:38–46.
and some later. What accounts for the temporal sequence Cebolla B, Vallejo M. 2006. Nuclear factor-I regulates glial fibrillary
of transcriptional changes during astrocyte development? acidic protein gene expression in astrocytes differentiated from cor-
tical precursor cells. J Neurochem 97:1057–1070.
Do astrocytes develop first as cells expressing a basic stable Choi B, Lapham L. 1980. Evolution of Bergmann glia in developing
of astrocyte characteristics and then regulate other genes human fetal cerebellum: a Golgi, electron microscopic and immuno-
to provide for a match to their local environments? fluorescent study. Brain Res 190:369–383.
D’Alessandro JS, Yetz-Aldape J, Wang EA. 1994. Bone morphogenetic
2. Astrocytes interact intimately with blood vessels and proteins induce differentiation in astrocyte lineage cells. Growth
neurons. How is astrocyte development matched to vascu- Factors 11:53–69.
lar development? How is astrocyte development linked to Davies SJ, Shih CH, Noble M, Mayer-Proschel M, Davies JE, Proschel C.
neuronal development, particularly the development 2011. Transplantation of specific human astrocytes promotes func-
tional recovery after spinal cord injury. PLosOne 6:e17328.
of dendrites and synapses?
Deneen B, Ho R, Lukaszewicz A, Hochstim CJ, Gronostajski RM,
3. How is the molecular and morphological heterogeneity Anderson DJ. 2006. The transcription factor NFIA controls the onset
of gliogenesis in the developing spinal cord. Neuron 52:953–968.
of astrocytes generated? In what ways are astrocytes
Doyle JP, Dougherty JD, Heiman M, Schmidt EF, Stevens TR, Ma G,
functionally heterogeneous? Bupp S, et al. 2008. Application of a translational profi ling approach
for the comparative analysis of CNS cell types. Cell 135:749–762.
4. How can we characterize the population of immature cells
Fan G, Martinowich K, Chin MH, He F, Fouse SD, Hutnick L,
in the adult CNS that have the potential to develop into et al. 2005. DNA methylation controls the timing of astroglio-
astrocytes? Why is their development normally inhib- genesis through regulation of JAK-STAT signaling. Development
ited? Do adult astrocyte progenitors play any role in the 132:3345–3356.
response of the CNS to neurological diseases and how? Figiel M, Maucher T, Rozyczka J, Bayatti N, Engele J. 2003. Regulation
of glial glutamate transporter expression by growth factors. Exp
5. Could adult astrocyte precursors be sources of astrocytomas? Neurol 183:124–135.

A S T R O C Y T E D E VE L O PM E N T • 145
Furukawa T, Mukherjee S, Bao Z-Z, Morrow EM, Cepko CL. 2000. rax, Korr H, Schultze B, Maurer W. 1973. Autoradiographic investigations
Hes1, and notch1 promote the formation of Muller glia by postnatal of glial proliferation in the brain of adult mice. J Comp Neurol
retinal progenitor cells. Neuron 26:383–394. 150:169–176.
Gaiano N, Nye JS, Fishell G. 2000. Radial glial identity is promoted by Levison SW, Goldman JE. 1993. Both oligodendrocytes and astrocytes
Notch1 signaling in the murine forebrain. Neuron 26:395–404. develop from progenitors in the subventricular zone of postnatal rat
Gard AL, Williams WC 2nd, Burrell MR. 1995. Oligodendroblasts forebrain. Neuron 10:201–212.
distinguished from O-2A glial progenitors by surface phenotype Levison SW, Goldman JE. 1997. Multipotential progenitors co-exist
(O4+GalC-) and response to cytokines using signal transducer LIFR with lineage restricted precursors in the mammalian postnatal
beta. Dev Biol 167:596–608. subventricular zone. J Neurosci Res 48:83–94.
Garré JM, Retamal MA, Cassina P, Barbeito L, Bukauskas FF, Sáez JC, Levison SW, Young GM, Goldman JE. 1999. Cycling cells in the adult rat
et al. 2010. FGF-1 induces ATP release from spinal astrocytes in cul- neocortex produce oligodendrocytes. J Neurosci Res 57:435–446.
ture and opens pannexin and connexin hemichannels. Proc Natl Lillien LE, Raff MC. 1990. Differentiation signals in the CNS: Type-2
Acad Sci USA 107:22659–22664. astrocyte development in vitro as a model system. Neuron 5:111–119.
Gensert JM, Goldman JE. 2001. Heterogeneity of cycling glial progeni- Lin G, Goldman JE. 2009. An FGF-responsive astrocyte precursor iso-
tors in the adult mammalian neocortex and white matter. J Neurobiol lated from the neonatal forebrain. Glia 57:592–603.
48:75–86. Lin JC, Cepko CL. 1999. Biphasic dispersion of clones containing
Girgrah N, Letarte M, Becker LE, Cruz TF, Theriault E, Moscarello MA. Purkinje cells and glia in the developing chick cerebellum. Dev Biol
1991. Localization of the CD44 glycoprotein to fibrous astrocytes in 211:177–197.
normal white matter and to reactive astrocytes in active lesions in Liu Y, Han SS, Wu Y, Tuohy TM, Xue H, Cai J, et al. 2004. CD44 expres-
multiple sclerosis. J Neuropathol Exp Neurol 50:779–792. sion identifies astrocyte-restricted precursor cells. Dev Biol 276:31–46.
Gross, RE, Mehler MF, Mabie PC, Zang Z, Santschi L, Kessler JA. 1996. Lovatt D, Sonnewald U, Waagepetersen HS, Schousboe A, He W, Lin JH,
Bone morphogenetic proteins promote astroglial lineage commitment by et al. 2007. The transcriptome and metabolic gene signature of protoplas-
mammalian subventricular zone progenitor cells. Neuron. 17:595–606. mic astrocytes in the adult murine cortex. J Neurosci 27:12255–12266.
Grove EA, Williams BP, Da-Qing L, Hajihosseini M, Friedrich A, Price Lu QR, Yuk D, Alberta JA, Zhu AZ Pawlitzky I, Chan J, et al. 2000. Sonic
J. 1993. Multiple restricted lineages in the embryonic rat cerebral cor- hedgehog-related oligodendrocyte lineage genes encoding bHLH pro-
tex. Development 117:553–561. teins in the mammalian central nervous system. Neuron 25:317–329.
Halliday AL, Cepko CL. 1992. Generation and migration of cells in the Luskin MB, McDermott K. 1994. Divergent lineages for oligodendro-
developing striatum. Neuron 9:15–26. cytes and astrocytes originating in the neonatal forebrain subven-
Hatada I, Namihira M, Morita S, Kimura M, Horii T, Nakashima K. tricular zone. Glia 11:211–226.
2008. Astrocyte-specific genes are generally demethylated in neural Luskin MB, Parnavelas JG, Barfield JA. 1993. Neurons, astrocytes, and
precursor cells prior to astrocytic differentiation. PLoS One 3:e3189. oligodendrocytes of the rat cerebral cortex originate from separate
He W, Ingraham C, Rising L, Goderie S, Temple S. 2001. Multipotent progenitor cells: An ultrastructural analysis of clonally related cells.
stem cells from the mouse basal forebrain contribute GABAergic J. Neurosci 13:1730–1750.
neurons and oligodendrocytes to the cerebral cortex during embry- Luskin MB, Pearlman AL, Sanes JR. 1988. Cell lineage in the cerebral
ogenesis. J Neurosci 21:8854–8862. cortex of the mouse studied in vivo and in vitro with a recombinant
Hochstim C, Deneen B, Lukaszewicz A, Zhou Q, Anderson DJ. 2008. retrovirus. Neuron 1:635–647.
Identification of positionally distinct astrocyte subtypes whose iden- Malatesta P, Hack MA, Hartfuss E, Kettenmann H, Klinkert W,
tities are specified by a homeodomain code. Cell 133:510–522. Kirchoff F, et al. 2003. Neuronal or glial progeny: regional differences
Hommes OR, Leblond CP. 1967. Mitotic division of neuroglia in the in radial glial fate. Neuron 37:751–764.
normal adult rat. J Comp Neurol 129:269–278. Malatesta P, Hartfuss E, Gotz, M. 2000. Isolation of radial glial cells
Horner PJ, Power AE, Kempermann G, Kuhn HG, Palmer TD, Winkler by fluorescent-activated cell sorting reveals a neuronal lineage.
J, et al. 2000. Proliferation and differentiation of progenitor cells Development 127:5253–5263.
throughout the intact rat spinal cord. J Neurosci 20:2218–2228. Marshall CA, Novitch BG, Goldman JE. 2005. Olig2 directs astrocyte
Hughes SM, Lillien LE, Raff MC, Rohrer H, Sendtner M. 1988. Ciliary and oligodendrocyte formation in postnatal subventricular zone
neurotrophic factor induces type-2 astrocyte differentiation in cul- cells. J Neurosci 25:7289–7298.
ture. Nature 335:70–73. Marshall CAG, Goldman JE. 2002. Subpallial Dlx2-expressing cells
Jang ES, Goldman JE. 2011. Pax6 expression is sufficient to induce a neu- give rise to astrocytes and oligodendrocytes in the cerebral cortex and
rogenic fate in glial progenitors of the neonatal subventricular zone. white matter. J Neurosci 22:9821–9830.
PLoS One 6:e20894. Marshall CAG, Suzuki SO, Goldman JE. 2003. Gliogenic and neuro-
Johe KK, Hazel TG, Muller T, Dugich-Djordjevic MM, McKay RD. genic progenitors of the subventricular zone: who are they, where did
1996. Single factors direct the differentiation of stem cells from fetal they come from, and where are they going? Glia 43:52–61.
and adult central nervous system. Genes Dev 10:3129–3140. Masood K, Besnard F, Su Y, Brenner M 1993. Analysis of a seg-
Kaaijk P, Pals ST, Morsink F, Bosch DA, Troost D. 1997. Differential ment of the human glial fibrillary acidic protein gene that directs
expression of CD44 splice variants in the normal human central ner- astrocyte-specific transcription. J Neurochem 61:160–166.
vous system. J Neuroimmunol 73:70–76. McManus MF, Chen L-C, Vallejo I, Vallejo M. 1999. Astroglial dif-
Kahn MA, Huang CJ, Caruso A, Barresi V, Nazarian R, Con dorelli DF, ferentiation of cortical precursor cells triggered by activation of the
de Vellis J. 1997. Ciliary neurotrophic factor activates JAK/Stat signal cAMP-dependent signaling pathway. J Neurosci 19:9004–9015.
transduction cascade and induces transcriptional expression of glial Mi H, Barres BA. 1999. Purification and characterization of astro-
fibrillary acidic protein in glial cells. J Neurochem 68:1413–1423. cyte precursor cells in the developing rat optic nerve. J Neurosci
Kaplan MS, Hinds JW. 1980. Gliogenesis of astrocytes and oligodendrocytes 19:1049–1061.
in the neocortical grey and white matter of the adult rat: electron micro- Mi HM, Haeberle H, Barres BA. 2001. Induction of astrocyte differen-
scopic analysis of light radioautographs. J Comp Neurol 193:711–727. tiation by endothelial cells. J Neurosci 21:1538–1547.
Kang K, Song MR. 2010. Diverse FGF receptor signaling controls astrocyte spec- Miller RH. 2002. Regulation of oligodendrocyte development in the
ification and proliferation. Biochem Biophys Res Comm 395:324–329. vertebrate CNS. Prog Neurobiol 67:451–467.
Kimelberg HK. 2010. Functions of mature mammalian astrocytes: a cur- Milosevic A, Goldman JE. 2002. Progenitors in the postnatal cerebel-
rent view. Neuroscientist 16:79–106. lar white matter are antigenically heterogeneous. J Comp Neurol
Komitova M, Serwanski DR, Lu QR, Nishiyama A. 2011. NG2 cells are 452:192–203.
not a major source of reactive astrocytes after neocortical stab wound Milosevic A, Noctor SC, Martinez-Cerdeno V, Kriegstein AR, Goldman
injury. Glia 59:800–809. JE. 2008. Progenitors from the postnatal forebrain subventricular zone

146 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
differentiate into cerebellar-like interneurons and cerebellar-specific Song MR, Ghosh A. 2004. FGF2-induced chromatin remodeling regu-
astrocytes upon transplantation. Mol Cell Neurosci 39:324–334. lates CNTF-mediated gene expression and astrocyte differentiation.
Miyake T, Fujiwara T, Fukunaga T, Takemura K, Kitamura T. 1995. Nat Neurosci 7:229–235.
Glial cell lineage in vivo in the mouse cerebellum. Develop Growth Staugaitis SM, Zerlin M, Levine JM, Hawkes R, Goldman JE. 2001.
Differ 37:273–285. Aldolase C/Zebrin II expression in neonatal rat forebrain reveals cel-
Muroyama Y, Fujiwara Y, Orkin SH, Rowitch DH. 2005. Specification lular heterogeneity within the subventricular zone and early astrocyte
of astrocytes by bHLH protein SCL in a restricted region of the neu- differentiation. J Neurosci 21:6195–6205.
ral tube. Nature 438:360–363. Sun Y, Nadal-Vicens M, Misono S, Lin MZ, Zubiaga A, Hua X, et al.
Nakashima K, Takizawa T, Ochiai W, Yanagisawa M, Hisatsune T, 2001. Neurogenin promotes neurogenesis and inhibits glial differen-
Hakafuku M, et al. 2001. BMP2-mediated alteration in the develop- tiation by independent mechanisms. Cell 104:365–376.
mental pathway of fetal mouse brain cells from neurogenesis to astro- Takizawa T, Nakashima K, Namihira M, Ochiai W, Uemura A, Yanagisawa
cytogenesis. Proc Natl Acad Sci U S A 98:5868–5873. M, et al. 2001. DNA methylation is a critical cell-intrinsic determinant
Nakashima K, Wiese S, Yanagisawa M, Arakawa H, Kimura N, of astrocyte differentiation in the fetal brain. Dev Cell 1:749–758.
Hisatsune T, et al. 1999a. Developmental requirement of gp130 sig- Turner D, Cepko C. 1987. Cell lineage in the rat retina: a common progenitor
naling in neuronal survival and astrocyte differentiation. J Neurosci for neurons and glia persists late in development. Nature 328:131–136.
19:5429–5434. Vallejo I, Vallejo M. 2002 Pituitary adenylate cyclase-activating polypep-
Nakashima K, Yanagisawa M, Arakawa H, Kimura N, Hisatsune T, tide induces astrocyte differentiation of precursor cells from develop-
Kawabata M, et al. 1999b. Synergistic signaling in fetal brain by ing cerebral cortex. Mol Cell Neurosci 21:671–683.
STAT3-Smad1 complex bridged by p300. Science 284:479–482. Ventura RE, Goldman JE. 2007. Dorsal radial glia generate olfac-
Nakashima K, Yanagisawa M, Arakawa H, Taga T. 1999c. Astrocyte dif- tory bulb interneurons in the postnatal murine brain. J Neurosci
ferentiation mediated by LIF in cooperation with BMP2. FEBS Lett 27:4297–4302.
457:43–46. Voigt T. 1989. Development of glial cells in the cerebral wall of ferrets:
Namihira M, Kohyama J, Semi K, Sanosaka T, Deneen B, Taga T, et al. direct tracing of their transformation from radial glia into astrocytes.
2009. Committed neuronal precursors confer astrocytic potential on J Comp Neurol 289:74–88.
residual neural precursor cells. Dev Cell 16:245–255. Walsh C, Cepko CL. 1993. Clonal dispersion in proliferating layers of
Nieto M, Schuurmans C, Britz O, Guillemot F. 2001. Neural bHLH developing cerebral cortex. Nature 362:632–635.
genes control the neuronal versus glial fate decision in cortical pro- Wolswijk G, Noble M. 1989. Identification of an adult-specific glial pro-
genitors. Neuron 29:401–413. genitor cell. Development 105:387–400.
Nishiyama A, Lin X-H, Giese N, Heldin C-H, Stallcup WB. 1996. Wolswijk G, Noble M. 1992. Cooperation between PDGF and FGF con-
Co-localization of NG2 proteoglycan and PDGF a-receptor on verts slowly dividing O-2A adult progenitor cells to rapidly dividing
O2A progenitor cells in the developing rat brain. J Neurosci Res cells with characteristics of O-2A perinatal progenitor cells. J Cell
43:299–314. Biol 118:889–900.
Noctor SC, Flint AC, Weissman TA, Dammerman RS, Kriegstein AR. Yanagisawa M, Nakashima K, Taga T. 1999. STAT3-mediated astro-
2001. Neurons derived from radial glial cells establish radial units in cyte differentiation from mouse fetal neuroepithelial cells by mouse
neocortex. Nature 409:714–720. oncostatin M. Neurosci Lett 269:169–172.
Norton WT. 1999. Cell reactions following acute brain injury: a review. Yanagisawa M, Nakashima K, Takizawa T, Ochiai W, Arakawa H,
Neurochem Res 24:213–218. Taga T. 2001. Signaling crosstalk underlying synergistic induction
Nunes MC, Roy NS, Keyoung HM, Goodman RR, McKhann G 2nd, of astrocyte differentiation by BMPs and IL-6 family of cytokines.
Jiang L, et al. 2003. Identification and isolation of multipotential FEBS Lett 489:139–143.
neural progenitor cells from the subcortical white matter of the adult Yeh TH, Lee da Y, Gianino SM, Gutmann DH. 2009. Microarray analyses
human brain. Nat Med 29:439–447. reveal regional astrocyte heterogeneity with implications for neurofibro-
Ochiai W, Yanagisawa M, Takizawa T, Hakashima K, Taga T. 2001. matosis type 1 (NF1)-regulated glial proliferation. Glia 57:1239–1249.
Astrocyte differentiation of fetal neuroepithelial cells involving Zerlin M, Goldman JE. 1997. Interactions between glial progenitors
cardiotrophin-1-induced activation of STAT3. Cytokine 14:264–271. and blood vessels during early postnatal corticogenesis: blood vessel
Orentas DM, Hayes JE, Dyer KL, Miller RH. 1999. Sonic hedgehog sig- contact represents an early stage of astrocyte differentiation. J Comp
naling is required during the appearance of spinal cord oligodendro- Neurol 387:537–546.
cyte precursors. Development 126:2419–2429. Zerlin M, Levison SW, Goldman JE. 1995. Early patterns of migration,
Palay S, Chan-Palay V 1974. Cerebellar cortex, cytology and organiza- morphogenesis, and intermediate filament expression of subventricu-
tion. New York: Springer-Verlag. lar zone cells in the postnatal rat forebrain. J Neurosci 15:7238–7249.
Parnevales JG. 1999. Glial cell lineages in the rat cerebral cortex. Zerlin M, Milosevic A, Goldman JE. 2004. Glial progenitors of the neo-
Exp Neurol 156:418–429. natal subventricular zone differentiate asynchronously, leading to
Paterson JA. 1983. Dividing and newly produced cells in the corpus spatial dispersion of glial clones and to the persistence of immature
callosum of adult mouse cerebrum as detected by light microscopic glia in the adult mammalian CNS. Dev Biol 270:200–213.
radioautography. Anat Anz Jena 153:149–168. Zhang L, Goldman JE 1996. Developmental fates and migratory path-
Price J, Thurlow L. 1988. Cell lineage in the rat cerebral cortex: a study ways of dividing progenitors in the postnatal rat cerebellum. J Comp
using retroviral-mediated gene transfer. Development 104:473–482. Neurol 370:536–550.
Pringle NP, Yu W-P, Howell M, Colvin J, Ornitz DM, Richardson WD. Zhang Y, Barres BA. 2010. Astrocyte heterogeneity: an underappreciated
2003. Fgfr3 expression by astrocytes and their precursors: evidence topic in neurobiology. Curr Opin Neurobiol 20:588–594.
that astrocytes and oligodendrocytes originate in distinct neuroepi- Zhou Q and Anderson DJ. 2002. The bHLH transcription factors Olig2
thelial domains. Development 130:93–102. and Olig1 couple neuronal and glial subtype specification. Cell
Raff MC, Miller RH, Noble M. 1983. A glial progenitor cell that devel- 109:61–73.
ops in vitro into an astrocyte or an oligodendrocyte depending on Zhou Q, Wang S, Anderson DJ. 2000. Identification of a novel family of
culture medium. Nature 303:390–396. oligodendrocyte lineage-specific basic helix-loop-helix transcription
Rao MS, Noble M, Mayer-Proschel M. 1998. A tripotential glial precur- factors. Neuron 25:331–343.
sor cells is present in the developing spinal cord. Proc Natl Acad Sci Zhu X, Bergles DE, Nishiyama A. 2008. NG2 cells generate both oligo-
U S A 95:3996–4001. dendrocytes and gray matter astrocytes. Development 135: 145–157.
Shafit-Zagardo B, Iwaki AK, Goldman JE. 1988. Astrocytes regulate Zhu X, Hill RA, Dietrich D, Komitova M, Suzuki R, Nishiyama A.
GFAP mRNA levels by cAMP and protein kinase C dependent 2011. Age-dependent fate and lineage restriction of single NG2 cells.
mechanisms. Glia 1:346–354. Development 138:745–753.

A S T R O C Y T E D E VE L O PM E N T • 147
13.
LINEAGE AND DEVELOPMENT: OLIGODENDROCY TES
Katsuhiko Ono and Kazuhiro Ikenaka

A B B R E VI AT I O N S mature into OLs. In this chapter, lineage-tracing experiments


of OLs are discussed for this purpose. However, to understand
BMP bone morphogenetic protein the references cited, it is important for the reader to know the
CTGF connective tissue growth factor meaning of markers that are used to label OL lineage cells, and
CNS central nervous system these are described at the beginning of the chapter. The same
CDK2 cyclin-dependent kinase 2 marker could label cells of different developmental stages when
ET-1 endothelin-1 different species are analyzed or even when different regions are
GalC galactocerebroside analyzed. In the second part of this chapter, the mechanisms
GFAP glial fibrillary acidic protein underlying OL development are dissected. Many intrinsic and
GPR G protein–coupled receptor extrinsic factors are known to play a role in OL development:
GRP glia restricted progenitor Some regulate OL specification from neural stem cells, some
hEGFR human epidermal growth factor receptor affect migration, some regulate exit from cell cycle, and oth-
HMG high mobility group ers regulate maturation from OPCs to OLs. Regulation of OL
IGF-1 insulin-like growth factor I development at each of these steps is discussed.
LRR leucin-rich repeat
MAG myelin-associated glycoprotein
MBP myelin basic protein 2 D E VE L O PM E N TA L S TAG E S O F
MRF myelin gene regulatory factor O L I G O D E N D R O C Y T E L I N E AG E C E L L S
NGF nerve growth factor C A N B E VI S UA L I Z E D BY U S I N G S TAG E
O-2A oligodendrocyte-type2 astrocyte SPECIFIC MARKER S
OL oligodendrocyte
OPC oligodendrocyte progenitor cell 2.1 G LYC O L I P I D
PDGF platelet-derived growth factor
Oligodendrocytes generate myelin membranes that are rich in
PDGFRD Platelet-derived growth factor receptor D
lipids. In fact, lipids account for 70% to 85% of the dry weight
PLP proteolipid protein
of compact myelin. Probably because of this, OL lineage cells
pMN motoneuron progenitors
express cell type and cell stage–specific glycolipids even before
PSA-NCAM Poly-Sialated Neural Cell Adhesion Molecule
myelin formation. A2B5 antigen, O4 antigen, sulfatide, and
SRF serum response factor
galactocerebroside (GalC) are often used as stage-specific
VZ ventricular zone
markers for OL lineage cell in vitro and in vivo, including in
YY1 Yin Yang 1
the human fetal tissues (Hajihosseini et al. 1995; Wilson et al.
2003; Zhang et al. 2000). The sequence of the onset of lineage
marker expression is summarized in Figure 13.1.
1 INTRODUCTION
2.1.1 A2B5 Antigen
Oligodendrocytes (OLs) are generated from neural stem cells
present in the ventricular zone (VZ) surrounding the ventricle. A monoclonal antibody, A2B5 (Eisenbarth et al. 1979), was
Although neurons and astrocytes are generated from most, if originally reported as a neuron-specific antibody. Later however,
not all, regions of the central nervous system (CNS), OLs are Raff et al. (1983) reported that in the newborn rat optic nerve
generated from only several restricted areas. Thus, after specifi- cell culture, devoid of neurons, a special type of glial progenitor
cation to OL lineage cells from multipotent neural stem cells, cell was labeled with A2B5 antibody. This special cell differen-
OL progenitor cells (OPCs) need to migrate a long distance tiates in vitro into either an astrocyte (GFAP+/A2B5+; type
in a nonradial fashion before they reach their final destination, 2 astrocyte) or OL depending on culture condition, and was
which is mostly the white matter. There, OPCs cease prolifer- called an oligodendrocyte-type 2 astrocyte (O-2A) progeni-
ation and mature into myelinating OLs. Therefore, to under- tor cell. O-2A progenitor cells at the initial stage of culture are
stand the development of OLs, it is important to know where bipolar in shape and highly motile (Small et al. 1987). When
and when OPCs are generated, and where they migrate to and purified O-2A progenitor cells were transplanted into newborn

148
neuroepithelial OPC late progenitor pre-myelinating myelinating
procursor pro-oligodendrocyte oligodendrocyte oligodendrocyte

morphology migratory
proliferative
process-bearing
bipolar
(rodent/human)

MBP
A2B5 04/OPA 01/GalC
in vitro*

PLP
NG2
PDGFRα

01/GalC
O4(/OPA?)
Chick

Sox 10 MBP
Olig2 PLP
PDGFRα
NKx2.2
Rodent

Olig2/Olig1 NKx2.2 PLP


O4 01/GalC MBP
PDGFRα (O4/OPA)**
Sox 10 NG2
spinal cord

O4(45dpc)
Human

O1/GalC(45dpc) MBP(52dpc)***
PDGFRα(49dpc)

Figure 13.1 Scheme of Sequential Expression of Lineage Markers for Oligodendrocyte Progenitor Cells Details are explained in the main text. *Sequence
of lineage marker expression on human OPC was examined in neurosphere-derived OPCs. **When O4 was applied to open-book cultures of neural
tube or hindbrain, presumed migratory OPCs were labeled with O4. ***First myelin was found in the human spinal cord in a 10-week-old fetus under
TEM; therefore, the first MBP+ cells in the human spinal cord were probably premyelinating oligodendrocytes. From Zhang et al. 2000; Wada
et al. 1982.

rat brain, nearly all cells became OLs (Espinosa de los Monteros proliferative stages (Ono et al. 2001). Therefore, it is probable
et al. 1993), and thus the in vivo equivalent of O-2A progenitor that O4 antibody identifies a more immature, migrating, and
cells are believed to be OPCs with some competence to astrocyte mitotically active stage of OL lineage cells in chick embryo
differentiation. A2B5 antibody also recognizes glia restricted than in the rodent. O4 antibody recognizes sulfatide, semino-
progenitor (GRP) cells that differentiate not only into OLs lipid, sulfated and nonsulfated cholesterol, and POA. In spite
and type 2 astrocytes, but also into type 1 (GFAP+/A2B5–) of much improvement in the technology to analyze lipid anti-
astrocytes (Rao et al. 1998). Although GFAP+/A2B5+ astro- gens, the nature of POA has not been identified yet (Bansal
cytes were reported to be in the developing optic nerve (Miller et al. 1992).
et al. 1985), the presence of two distinct astrocyte lineages was
not confirmed in vivo (Skoff 1990). A2B5 has been reported to
react with a variety of antigens, including multiple ganglioside
2.1.3 O1 Antigen and GalC
and sulfatide (Kundu et al. 1983), and thus the specificity of the A monoclonal antibody O1 was generated in the same
antibody to the OPC or OL lineage cells is not high. series of experiments as the O4 antibody. O1+ cells in vitro
are multiple process bearing and no longer proliferative or
migratory. Therefore, the O1+ cell stage is a relatively mature
2.1.2 O4 Antigens
stage in OL lineage cells, although early O1+ cells do not
A monoclonal antibody O4 was generated by immuniz- show heavy myelination. O1 antibody recognizes GalC and
ing mice with homogenates of the bovine corpus callosum monogalactosyldiglyceride.
(Sommer and Schachner 1981). O4 antibody labels OL lin-
eage cells at a pro-oligodendroblast stage (see Fig. 13.1), at the
2.2 M E M B R A N E P ROT E I NS
GalC-negative stage and even before the cells express sulfatide
(Bansal et al. 1992). Therefore, the antigen(s) recognized by Proteins localized at the cell surface are also frequently used to
O4 was designated as pro-oligodendroblast antigen (POA). identify OL lineage cells and judge their developmental stages.
When O4 antibody was applied to chick embryo CNS, O4 There have been lots of arguments about the specificity for
labels OPCs in a restricted region of the ventral VZ (Ono each of these proteins described in the following. It was very
et al. 1995, 1997), and also OL lineage cells at migratory and difficult to follow the fate of cells that expressed a particular

L I N E AG E A N D D E VE L O PM E N T: O L I G O D E N D R O C Y T E S • 149
marker protein at one stage in their differentiation, but then myelinating OLs). However, PLP gene transcripts, especially
downregulated it on further differentiation. However, the use its alternative-splicing product DM20, can be found from very
of the Cre-loxP system, especially tamoxifen inducible Cre, has early stages of development (Ikenaka et al. 1992; Timsit et al.
allowed precise fate mapping of cells that are expressing the 1992) even from E9.5 in the basal plate of the diencephalon
marker protein at the time of interest (Brocard et al. 1997). We (Timsit et al. 1995). However, PLP/DM20 producing cells at
now have better understanding of which kind of cells at which this very early stage have been shown to differentiate mainly
developmental stage express particular marker proteins. into neurons (Delaunay et al. 2008); thus, identifying the cell
type expressing PLP/DM20 early in development is contro-
versial. It is noteworthy that PLP/DM20 expressing cells in a
2.2.1 NG2
certain region, such as olfactory bulb, may be OPCs that are
NG2 antibody was generated using B49 cells as antigen. not expressing PDGFRα (Spassky et al. 2001).
Antisera were absorbed with neuronal cell lines, B103 and
B65 cells. Absorbed antisera recognized both pseudo-glia and
pseudo-neuron cell lines, and also tetanus toxin-positive neu- 2.3 T R A NS C R I P T I O N FAC TO R S
rons and GFAP+ astrocytes in primary culture. Therefore, the Because OL development proceeds through regulated gene
putative antigen (NG2 antigen) was believed to be expressed expression mainly carried out by transcription factors, it is not
by common progenitor cells for neurons and astrocytes surprising that several transcription factors are used to mark
(Stallcup 1981). Later, NG2 antibody was shown to react with OL lineage cells. In this chapter transcription factors com-
O-2A progenitor cells in vitro (Stallcup and Beasley 1987), monly used as OL lineage markers are described. The function
and in vivo NG2+ cells coexpress PDGFRα (see section and precise description of these factors is provided later.
2.2.2). Thus, NG2+ cells were elucidated to recognize OPCs
both in vivo and in vitro (Nishiyama et al. 1996). NG2 antigen
is a chondroitin sulfate proteoglycan (cspg4) (Stallcup et al. 2.3.1 Olig1
1983). NG2+ cells may be more immature than A2B5+ OPC, Olig1 expression commences at E9.0 in the motoneuron
because, at least in vitro, NG2+ cells appear before the expres- progenitor (pMN) domain of the mouse spinal cord, then is
sion of A2B5+ cells and generate A2B5+ cells (Baracskay et al. downregulated at around E11.0, and restarts at E12.5. From
2007). As described in chapter 10, fetal NG2+ progenitor cells this time on, it is expressed by OL lineage cells. However, early
in the rodent give rise to OLs and astrocytes in the gray matter. expressing cells generate motor neurons (Lu et al. 2002) and later
By contrast, those in newborn animals differentiate into OLs they also generate astrocytes or radial glial cells (Liu and Rao
exclusively. Therefore, NG2+ cells in early development may 2004).
be bipotential glial progenitor cells, whereas postnatal NG2+
cells are OPC. Details of fate mapping study of NG2+ cells
are discussed in chapter 10. 2.3.2 Olig2
Olig2 also starts its expression in the pMN domain of mouse
2.2.2 Platelet-Derived Growth Factor spinal cord at E8.5 and is continuously expressed to adulthood
Alpha-Receptor by the OL lineage cells. At E9.5 30% of Olig2 expressing cells
generate motor neurons, but after E12.5 they stop producing
Platelet-derived growth factor alpha-receptor PDGFRα tran- motor neurons and only produce glial cells, including OLs
scripts first appear at the VZ of pMN domain in the cervi- and astrocytes (Masahira et al. 2006).
cal spinal cord on embryonic day 12.5 (E12.5) in the mouse
(E14 in rat, E7 in chick) (Pringle and Richardson 1993; Pringle
et al. 1996). PDGFRα+ cells proliferate and migrate away from 2.3.3 Sox10
the VZ and by E17 (in the mouse) the number of PDGFRα+ In contrast to Olig1/2, Sox10 is more specific to OL lineage
cells reaches a steady state, and they are distributed evenly cells and starts its expression in the pMN domain from E11.5
throughout the cord. The onset of PDGFRα expression coin- (Stolt et al. 2002; Tripathi et al. 2011).
cides with the initiation of OL generation and several other
lines of evidence have demonstrated that PDGFRα+ cells are
OPCs (Richardson et al. 2000). Moreover, colocalization of 3 O L I G O D E N D R O C Y T E D E VE L O PM E N T
PDGFRα and NG2 has been shown in various studies (see IN ANIMAL MODELS
the preceding).
Developmental study of OLs became more popular after
O-2A progenitors were identified in vitro (see the preceding).
2.2.3 Proteolipid Protein/DM20
A great number of researchers uncovered characteristics and
Myelin proteolipid protein (PLP) is one of the major pro- behavior of O-2A progenitor cells in vitro, some of which
teins present in the CNS myelin membrane and is found in were later elucidated to reflect, at least partly, those of OPCs
the mature OL. Therefore, immunohistochemical detec- in vivo. Here, we summarize the in vivo development of OL
tion of PLP unambiguously identifies mature OLs (mostly lineage cells in the optic nerve, spinal cord and forebrain.

150 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
3.1 O P T I C N E RV E of neural cell differentiation (Richardson et al. 2000). Such
restricted appearance of OPC was later confirmed with CNP,
O-2A progenitor cells in vitro were first identified in cul-
Sox10, PLP, Olig1, Olig2, and Nkx2.2. These origins of OPCs
tures of optic nerve glia. This finding was the start point of
may be divided into two separate groups based on slight dif-
cellular and molecular analyses of OL development. Based
ferences in dorsoventral position; namely, one group includes
on this culture system, many aspects of OPCs were under-
Olig+, PDGFRα+, and Sox10+ cells, corresponding to cells
stood, which led to finding of lineage markers and behavioral
in the pMN domain, and the other are Nkx2.2+ and O4+
characters of OPC. Oligodendrocyte progenitor cells in the
cells slightly ventral to the pMN domain (dorsal part of p3
optic nerve were believed to be immigrants from the ventral
domain; Fig. 13.3). The latter group of OPC might be avian
forebrain. This was first suggested by isolated explant culture
specific (Fu et al. 2002). OPCs go on to coexpress both Olig2
(Small et al. 1987); the optic nerve in the developing rat was
and Nkx2.2 in the parenchymal region in later development
divided into chiasmal end, middle part, and retinal end, and
(Cai et al. 2010). Oligodendrocyte progenitor cells gradually
each fragment was cultured separately for several days. GalC+
disperse from the ventral part to the dorsal spinal cord, and
OLs appeared sequentially from cultures of the chiasmal end
finally they are evenly distributed throughout the spinal cord.
to the retinal end. In vivo migration of OPCs from the fore-
The initial ventral origin and subsequent ventral-to-dorsal
brain was elucidated in the chick embryo (Ono et al. 1997).
developmental gradient seem to be conserved in the verte-
O4+ OPCs initially appear in the floor of the third ventricle
brate from fish to human spinal cord (Hajihosseini et al. 1996;
around E5 as a cell cluster, and subsequently O4+ cells gradu-
Kirby et al. 2006).
ally spread into the optic nerve from the chiasmal to retinal
Initial ventral OPCs are induced by Shh from notochord
end. When chick embryos received DiI in the third ventri-
and floor plate, which was demonstrated by notochord graft-
cle at E5 or E6, O4+ progenitor cells that had incorporated
ing (Orentas et al. 1999). Later, OPCs also rise from the dor-
DiI were observed in the optic nerve 2 days later (arrow in
sal ventricular zone in a Shh independent manner (Fig. 13.3)
Fig. 13.2). This was the first direct evidence for OPC migra-
(Cai et al. 2005; Vallstedt et al. 2005). Lineage analysis with
tion in vivo. Migratory OPCs in vivo were unipolar or asym-
Msx3-Cre mice demonstrated that approximately 20% of
metrical bipolar in shape and aligned along retinal ganglion
OLs are generated from the dorsal ventricular zone in the
cell axons (see Fig. 13.2).
spinal cord (Tripathi et al. 2011). Regulatory mechanisms
of OPC migration and proliferation are summarized in the
3.2 S P I NA L C O R D following.
Oligodendrocyte progenitor cells in the spinal cord have been
most extensively studied in vivo with respect to their sites of
origin, and regulatory mechanisms of migration and prolifera-
tion. At early stages of development, spinal cord OPCs were
reported to originate in the ventral part by split culture and
dP1
DiI tracing (Warf et al. 1991). Then, after the identification
of PDGFRα as a lineage marker for OPC, the focal restricted dP2
origin of OPCs was demonstrated in the ventral ventricu-
lar zone of the fetal rat spinal cord (Pringle and Richardson Msx3 dP3
Dorsal OPC
1993). Later, similar results were described for O4+ OPCs dP4
in chick embryo spinal cord (Ono et al. 1995). These obser-
dP5
vations strongly indicated focal restricted origin of OPCs in
the spinal cord, which led to the dorsoventral domain theory dI P6
Dbx1
p0
p1
p2
04/Dil
Olig2, Sox10 pMN
Ventral OPC
Nkx2.2 p3

Notochord

Figure 13.3 Scheme of Ventrally and Dorsally Derived Oligodendrocyte


Progenitor Cells in the Spinal Cord. Ventral OPCs originate from Olig2+/
Figure 13.2 Basal Forebrain–Derived O4+ Oligodendrocyte Progenitor Sox10+ pMN domain in the rodent spinal cord. In addition to this, the dorsal part
Cells in the Optic Nerve. DiI was injected into an E5 chick embryo, and an of the Nkx2.2+ p3 domain generates OPCs in avian embryo. Dorsal OPCs are
O4+ cell incorporating DiI (arrow) was observed in the optic nerve 2 days after generated from Dbx1+ and Msx3+ domains, although the number is much less
the injection. The asterisk indicates an O4-migratory cell. compared with ventral OPCs.

L I N E AG E A N D D E VE L O PM E N T: O L I G O D E N D R O C Y T E S • 151
3.3 F O R E B R A I N development. In addition, a high mobility group (HMG) fac-
As is the case for the spinal cord, OL generation is initiated tor, SRY-box containing gene 10 (Sox 10) and Nkx 2.2 (Liu et
in the ventral part of the forebrain in a Shh-dependent man- al. 2007; Zhou et al. 2000) are important factors for differen-
ner and later converts to the dorsal part. Using chick-quail tiation of OLs as well as specification. Another HMG factor,
chimera, it was shown that OLs have a mono-focal origin Sox 9, plays a more direct role than Sox 10 in specification of
at anterior entopeduncular area, from which OPCs migrate OLs. Ablation of Sox9 results in defects in the specification of
throughout the forebrain in the chick (Olivier et al. 2001). OLs and astrocytes, which recovers at later stages of develop-
Also, in rodents OL generation starts from a restricted ven- ment (Stolt et al. 2003).
tricular foci, the anterior entopeduncular area and medial gan- Mash1/Ascl1 is broadly expressed by brain and spinal cord
glionic eminence (starting from E12.5). At later stages (a few progenitors and is also expressed in OPCs (Kondo and Raff
days later), sources for OLs change to the lateral ganglionic 2000). Mash1 cooperates in vivo with Olig2 in OL specifica-
eminence and cerebral cortex (mainly after birth), as defined tion, demonstrating an essential role for Mash1 in the genera-
by the expression of Emx1 (Kessaris et al. 2006). This dorsal tion of a subset of OLs in the mouse forebrain (Parras et al.
induction of OL lineage cells may be Shh-independent, as in 2007; Sugimori et al. 2007).
the spinal cord.
4.1.2 Sonic Hedgehog Independent Late
3.4 OT H E R B R A I N R E G I O N S Generation of Dorsal Oligodendrocytes
Oligodendrocyte development in other brain regions, such The discovery of the dorsal origin of OLs questioned the gener-
as diencephalon and cerebellum, is still controversial. Several ality of Shh-dependent appearance of OLs. Oligodendrocyte
areas expressing Olig2 can be found but lineage tracing experi- progenitor cell emergence from the dorsal spinal cord was
ment of those Olig2 expressing cells (discriminating from shown to be Shh independent and probably FGF2 dependent
Olig2 cells in other areas) are difficult to perform with cur- (Cai et al. 2005; Chandran et al. 2003). Recently, it was shown
rently existing techniques. Recently, at least a certain fraction that even in the ventral forebrain FGF2 signaling cooperates
of OPCs in the cerebellum are reported to be derived from with Shh signaling and is equally important in OL specifica-
mesencephalon (Mecklenburg et al. 2011), suggesting multiple tion (Furusho et al. 2011; Kessaris et al. 2004). FGF2 signaling
origins of cerebellar OLs. induces expression of Olig2 and Sox9 independently of each
other in zebrafish (Esain et al. 2010).
In contrast with the inducing signal by Shh and FGF2 in
4 R E GU L AT I O N O F O L I G O D E N D R O C Y T E the ventral spinal cord, specification of OLs from neural stem
D E VE L O PM E N T cells is inhibited by factors secreted from the dorsal spinal
cord (Wada et al. 2000). Wnts (Langseth et al. 2010; Shimizu
As mentioned, OLs develop through their specification from et al. 2005; Ye et al. 2009) and bone morphogenetic proteins
multipotent neural stem cells, migration, exit from the cell cycle, (BMPs) (Miller et al. 2004; Samanta et al. 2004) are shown to
and terminal differentiation/maturation to myelin-forming be the factors inhibiting generation of OPCs. Both proteins,
OLs. Many intrinsic and extrinsic factors affect these pro- at least partially, induce expression of the inhibitor of differen-
cesses. In this chapter the regulation of OL development at tiation 2 and 4 (Id 2 and 4) that repress Olig1/2 function. It
each of these steps is discussed. Unless otherwise noted, OL is also suggested that Wnt signaling exerts its effects through
development in the rodent spinal cord is described. the BMP signaling pathway (Feigenson et al. 2011; Kasai et al.
2005).
4.1 S P E C I FI C AT I O N
4.1.1 Sonic Hedgehog Dependent Early 4.2 M I G R AT I O N
Generation of Ventral Oligodendrocytes
Cells specified for the OL lineage migrate from the VZ and
Oligodendrocyte induction in the ventral spinal cord is heav- spread out widely in the parenchyma. Molecular mechanisms
ily dependent on Shh that is secreted from the notochord. by which OPC migration is regulated have been uncovered
Addition of Shh resulted in ectopic induction of OL lineage first in the optic nerve (Sugimoto et al. 2001). Oligodendrocyte
cells and inhibition of Shh signaling blocked their emergence progenitor cell expressing unc5a or neuropilin-1 were repelled
(Orentas et al. 1999; Pringle et al. 1996). Oligodendrocyte by sema3a and netrin1, both of which are expressed in the
specification is also dependent on Shh in the telencephalon chiasmal region of the ventral forebrain (Spassky et al. 2002).
(Nery et al. 2001; Spassky et al. 2001) and therefore it seems to Migration of OPCs in the spinal cord is also regulated by
be a general phenomenon. Shh induces expression of several netrin1 secreted by floor plate cells, which inhibits pro-
genes in a dose-dependent manner. These include Nkx and cess extension by OPCs and functions as a chemo repellent
Olig, both of which positively regulate OPC specification and ( Jarjour et al. 2003; Tsai et al. 2003). Thus, these repellent
development (see Fig. 13.3) (Ericson et al. 1997; Lu et al. 2000, molecules apparently facilitate OPC migration. In addition,
2002; Takebayashi et al. 2002; Zhou and Anderson 2002). OPCs were reported to cease migration through CXCL1/
Especially, Nkx6 and Olig2 are known to be essential for OPC CXCR2 signaling (Tsai et al. 2002).

152 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Endothelin-1 (ET-1) is an astrocyte-derived signal that expressing p27 that interacted with cyclin-dependent kinase
also regulates migration and differentiation of OPCs, which 2 (CDK2) did not induce differentiation of progenitors,
express functional ET(A) and ET(B) receptors (Gadea et al. even though cells arrested at the G1/S transition (Tikoo et al.
2009). Endothelin-1 exerts both chemotactic and chemoki- 1998). Together, these studies suggested that cell cycle exit was
netic effects on OPCs to enhance cell migration. necessary but not sufficient to induce differentiation of pro-
genitors into OLs.
4 .3 C E L L P RO L I FE R AT I O N A N D E X IT
FRO M T H E C E L L C YC L E 4.4 T E R M I NA L D I FFE R E N T I AT I O N/
Platelet-derived growth factor promotes the expansion of M AT U R AT I O N/MY E L I NAT I O N
OPCs and allows their timely differentiation into OLs in vitro 4.4.1 Electrical Activity
(Noble et al. 1988). Proliferation of OPCs in vivo is mainly
regulated by PDGF-A as observed in vitro; mice lacking In mammals, myelin membrane is inhibitory for axonal exten-
PDGF-A show a lack of PDGFRα+ OPCs and a decreased sion (Fig. 13.4). Therefore, myelination has to be initiated only
number of PLP+ OL (Fruttiger et al. 1999). In addition, trans- after neuronal circuit formation has been completed. Thus, strict
genic mice over expressing PDGF-A resulted in hyperprolif- regulation of myelination by the axon is necessary. Electrical
eration of OPCs, although superfluous OPCs are eliminated activity passing through the axon could be easily considered as
by programmed cell death postnatally (Calver et al. 1998). one of the candidates because electrical activity should increase
FGF-2 up-regulates PDGFRα on OPCs and, in combination after circuit formation. Indeed, the proliferation of OPCs
with PDGF, supports their long-term proliferation (Bögler depended on electrical impulse activity in neighboring axons
et al. 1990; McKinnon et al. 1990). (Barres and Raff 1993) and myelination was induced by elec-
Oligodendrocyte progenitor cells stop proliferating before trical activity (Demerens et al. 1996). The mediator between
terminal differentiation. It was originally proposed that OPC the electrical active axon and OPCs seems to be ATP (and its
differentiation is intrinsically regulated by a “timing” mecha- degraded product, adenosine) and glutamate secreted from
nism that links the number of cell divisions to growth arrest axons through vesicular release (Ishibashi et al. 2006; Wake
and the initiation of differentiation (Temple and Raff 1986). et al. 2011). ATP first acts on astrocytes and induces release of
This mechanism was regulated by mitogens (Calver et al. leukemia inhibitory factor (LIF), which then stimulates termi-
1998) and molecularly characterized by the progressive accu- nal differentiation of OL (Ishibashi et al. 2006).
mulation of the cell cycle inhibitor, p27/Kip1 (Durand et al.
1997). It was shown that only a fraction of OPCs derived from
4.4.2 Axonal Membrane Proteins
p27-knockout mice differentiated successfully but continued
proliferation (Casaccia-Bonnefil et al. 1997; Durand et al. Several different receptor-ligand pairs that connect OPCs and
1998). However, overexpression studies with viral vectors axons are induced or reduced by the electrical activity and are

SRF Astrocyte
Neuron CTGF

LIF
ATP
IGF-1
Wnt
Wnt

OPC
β -Catenin Sox17 myelin
genes OL

Glu
NGF
Notch1

Jagged1 LINGO1 TrkA Notch1


PSA-CAM Contactin
Electrical Activity Axon

Figure 13.4 Factors Affecting the Terminal Differentiation of Oligodendrocytes After migrating into the white matter, most OPCs differentiate into
myelinating OLs. There are various environmental factors affecting this process. Factors promoting differentiation are depicted in red, whereas those
inhibiting differentiation are in blue. Various transcription factors affect this process but are not listed here. Please see chapter 43 for detail.

L I N E AG E A N D D E VE L O PM E N T: O L I G O D E N D R O C Y T E S • 153
also important for OPC/OL differentiation and/or myelina- response factor (SRF). Thus, neuron-restricted SRF ablation
tion, including ErbB2 and neuregulin, TrkA and NGF, and in mice increased the production of CTGF and elevated
Notch and Jagged or F3/contactin. OPCs, while inhibiting terminal OL differentiation (Stritt
ErbB2 functions in the transition of OPC to OL by trans- et al. 2009).
ducing a terminal differentiation signal both in vitro (Park BMP signaling through type I BMP receptors also affects
et al. 2001) and in vivo (Kim et al. 2003). the maturation/myelination steps of OLs. In the BMPR1a
Nerve growth factor (NGF) is a potent regulator of the and b double mutants, the number of OPCs and the timing
axonal signals that control myelination and reduces myelina- of their emergence was unchanged compared with wild-type;
tion by OLs (Chan et al. 2004). NGF and its cognate recep- however, myelin protein expression and mature OL num-
tor, TrkA, induce the expression of leucin-rich repeat (LRR) bers were significantly reduced (See et al. 2007). These data
and Ig domain-containing, Nogo receptor–interacting pro- indicate that BMP signaling promotes the generation of
tein (LINGO-1) (Lee et al. 2007) that inhibits OL differen- mature, myelinating OLs in vivo but does not affect OPC
tiation (Mi et al. 2005). development.
Oligodendrocytes differentiation is regulated by activation Overexpression of human epidermal growth factor recep-
of the Notch pathway as well. OPCs/OLs in the developing tor (hEGFR) in OL lineage cells contributes to the earlier
rat optic nerve express Notch1 receptors and retinal ganglion maturation of OLs in the corpus callosum. The opposite
cells express Jagged1, a ligand of the Notch1 receptor, along occurs in a mouse with reduced EGFR signaling (wa2 mouse),
their axons. Jagged1 expression decreases with a time course as demonstrated by the number of differentiated, myelinating
that parallels myelination in the optic nerve (Wang et al. oligodendrocytes and myelin basic protein (MBP) expression
1998). Notch1 also interacts with F3/contactin clustered at levels (Aguirre et al. 2007). Therefore, EGF signaling is also
the axonal paranodal junction and this interaction signals via important in maturation/myelination step.
Deltex1 to promote OL differentiation (Hu et al. 2003). Thus,
the timing of OL differentiation and myelination is controlled
4.4.4 Other Membrane Proteins/Extracellular Matrix
by the Notch pathway.
PSA-NCAM is first expressed on all growing fibers and Laminins are likely to regulate CNS myelination by interact-
negatively regulates myelin formation. Then axonal expression ing with both integrin receptors and dystroglycan receptors
is downregulated and myelin deposition occurs only on PSA- (Colognato et al. 2007), and the normal role for CNS laminin
NCAM–negative axons (Charles et al. 2000). is to promote the development of OPCs into myelin-forming
OLs via modulation of Fyn regulatory molecules (Relucio
et al. 2009).
4.4.3 Humoral Factors
G-protein–coupled receptor 17 (GPR17) is regulated
Terminal differentiation/myelination is also affected by vari- by Olig1 and its function is to oppose the action of Olig1. G
ous humoral factors that are secreted from cells surrounding protein–coupled receptor 17 is restricted to OL lineage cells,
the OPCs, such as astrocytes. but is down regulated during the peak period of myelination
Although Wnt proteins inhibit differentiation of OPC as and in adulthood. G protein–coupled receptor 17 overexpres-
well as their specification, at more mature stages, it directly sion inhibited OL differentiation and maturation. Conversely,
drives myelin gene expression (Tawk et al. 2011). Thus, Wnt Gpr17 knockout mice showed early onset of OL myelination
proteins affect multiple developmental stages of OL from (Chen et al. 2009). Thus, GPR17 orchestrates the transition
specification to myelination. The Wnt pathway itself is under between immature and myelinating oligodendrocytes via an Id
fine control by several factors, including Sox17. Sox17 is maxi- protein–mediated negative regulation.
mally expressed in pro-oligodendrocytes (see Fig. 13.1), and
its downregulation increases OPC proliferation and decreases
4.4.5 Transcription Factors
lineage progression after mitogen withdrawal (Sohn et al.
2006). It suppresses cyclin D1 expression and cell prolif- The presence of various transcription factors is required for nor-
eration by directly antagonizing β-catenin, whose activity in mal differentiation of OPCs into mature OLs and for the for-
OPCs is stimulated not only by Wnt3a, but also by PDGF mation of myelin. In the brains of Olig1-null mice, expression of
(Chew et al. 2011). myelin-specific genes is abolished and they develop widespread
Insulin-like growth factor I (IGF-1) increases brain growth progressive axonal degeneration and gliosis. It has been shown
and CNS myelination in a transgenic mouse line that over- that Olig1 regulates transcription of the major myelin-specific
expresses IGF-1 (Carson et al. 1993). On the contrary, Igf1 genes, MBP, PLP, and myelin-associated glycoprotein (MAG),
gene disruption results in reduced brain size, and CNS hypo- and suppresses expression of a major astrocyte-specific gene,
myelination in young animals (Beck et al. 1995). Insulin-like glial fibrillary acid protein (GFAP). In zebrafish, Olig1 asso-
growth factor I directly affects OLs and myelination in vivo ciates physically with another myelin-associated transcription
via type 1 IGF receptor (Zeger et al. 2007), and thus IGF-1 factor, Sox10, and the Olig1/Sox10 complex activates MBP
signaling in the cells of OL lineage is required for normal OL gene transcription via conserved DNA sequence motifs in the
development and myelination. Insulin-like growth factor I sig- MBP promoter region (Li et al. 2007). Zfp488, an OL-specific
naling is inhibited by connective tissue growth factor (CTGF) zinc-finger transcription regulator, can interact with Olig2
produced in neurons, whose expression is repressed by serum to promote precocious and ectopic OL differentiation.

154 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Knockdown of Zfp488 in an OL cell line leads to the down- REFERENCES
regulation of myelin gene expression. Thus, Zfp488 functions
as a transcription coregulator important for OL maturation Aguirre A, Dupree JL, Mangin JM, Gallo V. 2007. A functional role for
and Zfp488/Olig2 cooperation can serve as a mechanism for EGFR signaling in myelination and remyelination. Nat Neurosci
10:990–1002.
OL differentiation (Wang et al. 2006). Bansal R, Stefansson K, Pfeiffer SE. 1992. Proligodendrolast antigen
Myelin gene regulatory factor (MRF) is specifically (POA), a developmental antigen expressed by A007/O4-positive oli-
expressed by postmitotic OLs within the CNS. Overexpression godendrocyte progenitors prior to the appearance of sulfatide and
of MRF within cultured OPCs or the chick spinal cord pro- galactocerebroside. J Neurochem 58:2221–2229.
motes expression of myelin genes. In mice lacking MRF within Baracskay KL, Kidd GJ, Miller RH, Trapp BD. 2007. NG2-positive cells gen-
erate A2B5-positive oligodendrocyte precursor cells. Glia 55:1001–1010.
the OL lineage, premyelinating OLs are generated but display Barres BA, Raff MC. 1993. Proliferation of oligodendrocyte precursor
severe deficits in myelin gene expression and fail to myelinate cells depends on electrical activity in axons. Nature 361:258–260.
(Emery et al. 2009). These findings establish MRF as a criti- Beck KD, Powell-Braxton L, Widmer HR, Valverde J, Hefti F. 1995. Igf1
cal transcriptional regulator essential for OL maturation and gene disruption results in reduced brain size, CNS hypomyelination,
CNS myelination. and loss of hippocampal granule and striatal parvalbumin-containing
neurons. Neuron 14:717–730.
Yin Yang 1 (YY1) is another transcription factor that is an Bögler O, Wren D, Barnett SC, Land H, Noble M. 1990. Cooperation
essential component of the transcriptional network regulating between two growth factors promotes extended self-renewal and
the transition of OPCs from cell cycle exit to differentiation. inhibits differentiation of oligodendrocyte-type-2 astrocyte (O-2A)
Yin Yang 1 acts as a lineage-specific repressor of transcrip- progenitor cells. Proc Natl Acad Sci U S A 87:6368–6372.
tional inhibitors of myelin gene expression (Tcf4 and Id4), Brocard J, Warot X, Wendling O, Messaddeq N, Vonesch JL, Chambon
P, et al. 1997. Spatio-temporally controlled site-specific somatic muta-
by recruiting histone deacetylase-1 to their promoters during genesis in the mouse. Proc Natl Acad Sci U S A 94:14559–14563.
OL differentiation (He et al. 2007). More detailed description Cai J, Qi Y, Hu X, Tan M, Liu Z, Zhang J, et al. 2005. Generation of
of transcriptional control of OL maturation can be found in oligodendrocyte precursor cells from mouse dorsal spinal cord inde-
chapter 43. pendent of Nkx6 regulation and Shh signaling. Neuron 45:41–53.
Cai J, Zhu Q, Zheng K, Li H, Qi Y, Cao Q, et al. 2010. Co-Localization
of Nkx6.2 and Nkx2.2 homeodomain proteins in differentiated
myelinating oligodendrocytes. Glia 58:458–468.
5 S U M M A RY A N D P E R S P E C T I VE S Calver AR, Hall AC, Yu WP, Walsh FS, Heath JK, Betsholtz C, et al.
1998. Oligodendrocyte population dynamics and the role of PDGF
Several questions remain elusive in OPC development. First, in vivo. Neuron 20:869–882.
recent evidence suggests that OPCs migrate for long dis- Casaccia-Bonnefi l P, Tikoo R, Kiyokawa H, Friedrich V Jr, Chao MV,
Koff A. 1997. Oligodendrocyte precursor differentiation is perturbed
tances, sometimes across regional boundaries (Mecklenburg in the absence of the cyclin-dependent kinase inhibitor p27Kip1.
et al. 2011), and thus OLs in a certain region may have mul- Gene Dev 11:2335–2346.
tiple origins. Most of these studies were performed in the Chan JR, Watkins TA, Cosgaya JM, Zhang C, Chen L, Reichardt LF, et
chick embryo. It is of interest to examine whether such long al. 2004. NGF controls axonal receptivity to myelination by Schwann
distance migration of OPCs is observed in the developing cells or oligodendrocytes. Neuron 43:183–191.
Chandran S, Kato H, Gerreli D, Compston A, Svendsen CN, Allen
rodent brain (Lachapelle et al. 1984). Second, whereas the ND. 2003. FGF-dependent generation of oligodendrocytes by a
origin of OPC in the chick optic nerve may be a focal O4+ hedgehog-independent pathway. Development 130:6599–6609.
cell cluster in the suprachiasmal neuroepithelium (Ono et al. Charles P, Hernandez MP, Stankoff B, Aigrot MS, Colin C, Rougon G,
1997), the origin of rodent or mammalian OPCs in the ventral et al. 2000. Negative regulation of central nervous system myelina-
forebrain has not yet been elucidated. Lineage tracing stud- tion by polysialylated-neural cell adhesion molecule. Proc Natl Acad
Sci U S A 97:7585–7590.
ies with Cre–loxP system are needed to identify the origin of Chen Y, Wu H, Wang S, Koito H, Li J, Ye F, et al. 2009. The
OPC in the optic nerve. Third, the mechanisms by which dor- oligodendrocyte-specific G protein-coupled receptor GPR17 is a
sally derived OPCs are induced are not fully understood. It is cell-intrinsic timer of myelination. Nat Neurosci 12:1398–1406.
probable that dorsally derived OPCs are induced in an FGF- Chew LJ, Shen W, Ming X, Senatorov VV Jr, Chen HL, Cheng Y, et
dependent manner. However, the dorsal spinal cord releases al. 2011. SRY-box containing gene 17 regulates the Wnt/β -catenin
signaling pathway in oligodendrocyte progenitor cells. J Neurosci
factors inhibiting OPC differentiation. Wnt and BMP family 31:13921–13935.
molecules are strong candidates for these inhibitory activities Colognato H, Galvin J, Wang Z, Relucio J, Nguyen T, Harrison D,
(Mekki-Dauriac et al. 2002; Shimizu et al. 2005). How these et al. 2007. Identification of dystroglycan as a second laminin recep-
negative and positive regulators in the dorsal spinal cord coor- tor in oligodendrocytes, with a role in myelination. Development
dinate OPC differentiation may be future issues of this field. 134:1723–1736.
Delauna, D, Heydon K, Cumano A, Schwab MH, Thomas JL, Suter U,
Finally, how do OPCs regulate themselves in cell number? As et al. 2008. Early neuronal and glial fate restriction of embryonic neu-
mentioned, although PDGF-A is a strong mitogen for OPC, ral stem cells. J Neurosci 28:2551–2562.
excess OPCs are eliminated and their number becomes nor- Demerens C, Stankoff B, Logak M, Anglade P, Allinquant B, Couraud F,
mal even in PDGF-A overexpressing mice. Oligodendrocyte et al. 1996. Induction of myelination in the central nervous system by
progenitor cells might recognize their own density by some electrical activity. Proc Natl Acad Sci U S A 93:9887–9892.
form of cell–cell interaction as suggested in zebrafish (Kirby Durand B, Gao FB, Raff M. 1997. Accumulation of the cyclin-dependent
kinase inhibitor p27/Kip1 and the timing of oligodendrocyte differ-
et al. 2006). Better understanding OL development by uncov- entiation. EMBO J 16:306–317.
ering these issues is clearly important not only for glial biol- Durand B, Fero ML, Roberts JM, Raff MC. 1998. p27Kip1 alters the
ogy, but also for the translational study of OLs and myelin. response of cells to mitogen and is part of a cell-intrinsic timer

L I N E AG E A N D D E VE L O PM E N T: O L I G O D E N D R O C Y T E S • 155
that arrests the cell cycle and initiates differentiation. Current Biol Kessaris N, Fogarty M, Iannarelli P, Grist M, Wegner M, Richardson
8:431–440. WD. 2006. Competing waves of oligodendrocytes in the forebrain
Eisenbarth GS, Walsh FS, Nirenberg M. 1979. Monoclonal antibody to and postnatal elimination of an embryonic lineage. Nat Neurosci
a plasma membrane antigen of neurons. Proc Natl Acad Sci U S A 9:173–179.
76:4913–4917. Kessaris N, Jamen F, Rubin LL, Richardson WD. 2004. Cooperation
Emery B, Agalliu D, Cahoy JD, Watkins TA, Dugas JC, Mulinyawe SB, between sonic hedgehog and fibroblast growth factor/MAPK
et al. 2009. Myelin gene regulatory factor is a critical transcriptional signalling pathways in neocortical precursors. Development
regulator required for CNS myelination. Cell 138:172–185. 131:1289–1298.
Ericson J, Rashbass P, Schedl A, Brenner-Morton S, Kawakami A, Kim JY, Sun Q, Oglesbee M, Yoon SO. 2003. The role of ErbB2 signaling
van Heyningen V, et al. 1997. Pax6 controls progenitor cell iden- in the onset of terminal differentiation of oligodendrocytes in vivo.
tity and neuronal fate in response to graded Shh signaling. Cell J Neurosci 23:5561–5571.
11(90):169–180. Kirby BB, Takada N, Latimer AJ, Shin J, Carney TJ, Kelsh RN, et al.
Esain V, Postlethwai JH, Charnay P, Ghislain J. 2010. FGF-receptor 2006. In vivo tim-lapse imaging shows dynamic oligodendrocyte
signalling controls neural cell diversity in the zebrafish hindbrain by progenitor behavior during zebrafish development. Nat Neurosci
regulating olig2 and sox9. Development 137:3–42. 9:1506–1511.
Espinosa de los Monteros A, Zhang M, deVellis J. 1993. O2A progenitor Kondo T, Raff M. 2000. Basic helix-loop-helix proteins and the timing
cells transplanted into the neonatal rat brain develop into oligoden- of oligodendrocyte differentiation. Development 127:2989–2998.
drocytes but not astoryctes. Proc Natl Acad Sci U S A 90:50–54. Kundu SK, Pleatman MA, Redwine WA, Boyd AE, Marcus DM. 1983.
Fancy SPJ, Baranzini SE, Zhao C, Yuk DI, Irvine K-A, Kaing S, et al. Binding of monoclonal antibody A2B5 to gangliosides. Biochem
2009. Dysregulation of the Wnt pathway inhibits timely myeli- Biophys Res Commun 116:836–842.
nation and remyelination in the mammalian CNS. Gene Dev Lachapelle F, Gumpel M, Baulac M, Jacque C, Duc P, Baumann N. 1983–
23:1571–1585. 1984. Transplantation of CNS fragments into the brain of shiverer
Feigenson K, Reid M, See J, Crenshaw EB 3rd, Grinspan JB. 2009. Wnt mutant mice: extensive myelination by implanted oligodendrocytes.
signaling is sufficient to perturb oligodendrocyte maturation. Mol I. Immunohistochemical studies. Dev Neurosci 6:325–334.
Cell Neurosci 42:255–265. Langseth AJ, Munji RN, Choe Y, Huynh T, Pozniak CD, Pleasure
Feigenson K, Reid M, See J, Crenshaw III EB, Grinspan JB. 2011. SJ. 2010. Wnts influence the timing and efficiency of oligoden-
Canonical Wnt signalling requires the BMP pathway to inhibit oli- drocyte precursor cell generation in the telencephalon. J Neurosci
godendrocyte maturation. ASN Neuro 3:e00061. 30:13367–13372.
Finzsch M, Stolt CC, Lommes P, Wegner M. 2008. Sox9 and Sox10 Lee X, Yang Z, Shao Z, Rosenberg SS, Levesque M, Pepinsky RB,
influence survival and migration of oligodendrocyte precursors et al. 2007. NGF regulates the expression of axonal LINGO-1 to
in the spinal cord by regulating PDGF receptor alpha expression. inhibit oligodendrocyte differentiation and myelination. J Neurosci
Development 135:637–646. 27:220–225.
Fruttiger M, Karlsson L, Hall AC, Abramsson A, Calver AR, Bostrom H, Li H, Lu Y, Smith HK, Richardson WD. 2007. Olig1 and Sox10 interact
et al. 1999. Defective oligodendrocyte development and severe hypo- synergistically to drive myelin basic protein transcription in oligo-
myelination in PDGF-A knockout mice. Development 126:457–467. dendrocytes. J Neurosci 27:14375–14382.
Fu H, Qi Y, Tan M, Cai J, Takebayashi H, Nakafuku M, Richarsdon W, Li H, Paes de Faria J, Andrew P, Nitarska J, Richardson WD. 2011.
et al. 2002. Dual origin of spinal oligodendrocyte progenitors and Phosphorylation regulates OLIG2 co-factor choice and the motor
evidence for the cooperative role of Olig2 and Nkx2.2 in the control neuron—oligodendrocyte fate switch. Neuron 69:918–929.
of oligodendrocyte differentiation. Development 129:681–693. Liu R, Cai J, Hu X, Tan M, Qi Y, German M, et al. 2003. Region-specific
Furusho M, Kaga Y, Ishii A, He´bert JM, Bansal R. 2011. Fibroblast and stage-dependent regulation of Olig gene expression and oli-
growth factor signaling is required for the generation of oligo- godendrogenesis by Nkx6.1 homeodomain transcription factor.
dendrocyte progenitors from the embryonic forebrain. J Neurosci Development 130:6221–6231.
31:5055–5066. Liu Y, Rao MS. 2004. Olig genes are expressed in a heterogeneous
Gadea A, Aguirre A, Haydar TF, Gallo V. 2009. Endothelin-1 regulates population of precursor cells in the developing spinal cord. Glia 45:
oligodendrocyte development. J Neurosci 29:10047–10062. 67–74.
Gard AL, Pfeiffer SE. 1993. Glial cell mitogens bFGF and PDGF dif- Liu Z, Hu X, Cai J, Liu B, Peng X, Wegner M, et al. 2007. Induction
ferentially regulate development of O4+GalC- oligodendrocyte pro- of oligodendrocyte differentiation by Olig2 and Sox10: evidence for
genitors. Dev Biol 159:618–630. reciprocal interactions and dosage-dependent mechanisms. Dev Biol
Hajihosseini M, Tham TN, Dubois-Dalcq M. 1995. Origin of oligoden- 302:683–693.
drocytes within the huma spinal cord. J Neurosci 15:7981–7994. Lu QR, Yuk D, Alberta JA, Zhu A, Pawlitzky I, Chan J, et al. 2000.
He Y, Dupree J, Wang J, Sandoval J, Li J, Liu H, et al. 2007. The tran- Sonic hedgehog-regulated oligodendrocyte lineage genes encoding
scription factor Yin Yang 1 is essential for oligodendrocyte progeni- bHLH proteins in the mammalian central nervous system. Neuron
tor differentiation. Neuron 55:217–230. 25:317–329.
Hu QD, Ang BT, Karsak M, Hu WP, Cui XY, Duka T, et al. 2003. F3/ Lu QR, Sun T, Zhu Z, Ma N, Garcia M, Stiles CD, et al. 2002. Common
contactin acts as a functional ligand for Notch during oligodendro- developmental requirement for Olig function indicates a motor neu-
cyte maturation. Cell 115:163–175. ron/oligodendrocyte connection. Cell 109:75–86.
Ikenaka K, Kagawa T, Mikoshiba K. 1992. Selective expression of Masahira N, Takebayashi H, Ono K, Watanabe K, Ding L, Furusho M,
DM-20, an alternatively spliced myelin proteolipid protein gene prod- et al. 2006. Olig2-positive progenitors in the embryonic spinal cord
uct, in developing nervous system and in non-glial cells. J Neurochem give rise to not only to motoneurons and oligodendrocytes, but also
58:2248–2253 to a subset of astrocytes and ependymal cells. Dev Biol 293:358–369
Ishibashi T, Dakin KA, Stevens B, Lee PR, Kozlov SV, Stewart CL, McKinnon RD, Matsui T, Dubois-Dalcq M, Aaronson SA. 1990. FGF
et al. 2006. Astrocytes promote myelination in response to electrical modulates the PDGF-driven pathway of oligodendrocyte develop-
impulses. Neuron 49:823–832. ment. Neuron 5:603–614.
Jarjour AA, Manitt C, Moore SW, Thompson KM, Yuh SJ, Kennedy Mecklenburg N, Barcia-Lopez R, Puelles E, Sotelo C, Martinez S. 2011.
TE. 2003. Netrin-1 is a chemorepellent for oligodendrocyte precur- Cerebellar oligodendroglial cells have a mesencephalic origin. Glia
sor cells in the embryonic spinal cord. J Neurosci 23: 3735–3744. 59:1946–1957.
Kasai M, Satoh K, Akiyama T. 2005. Wnt signaling regulates the sequen- Mekki-Dauriac S, Agius E, Kan P, Cochard P. 2002. Bone morphoge-
tial onset of neurogenesis and gliogenesis via induction of BMPs. netic proteins negatively control oligodendrocyte precursor specifica-
Genes Cells 10:777–783. tion in the chick spinal cord. Development 129:5117–5130.

156 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Mi S, Miller RH, Lee X, Scott ML, Shulag-Morskaya S, Shao Z, et al. See J, Mamontov P, Ahn K, Wine-Lee L, Crenshaw EB 3rd, Grinspan
2005. LINGO-1 negatively regulates myelination by oligodendro- JB. 2007. BMP signaling mutant mice exhibit glial cell maturation
cytes. Nat Neurosci 8:745–751. defects. Mol Cell Neurosci 35:171–182.
Miller RH, David S, Patel R, Abney ER, Raff MC. 1985. A quantitative Shimizu T, Kagawa T, Wada T, Muroyama Y, Takada S, Ikenaka K.
immunohistochemical study of macroglial cell development in the 2005. Wnt signaling controls the timing of oligodendrocyte develop-
rat optic nerve: in vivo evidence for two distinct astrocyte lineages. ment in the spinal cord. Dev Biol 282:397–410.
Dev Biol 111:35–41. Skoff RP. 1990. Gliogenesis in rat optic nerve: astrocytes are generated in
Miller RH, Dinsio K, Wang R, Geertman R, Maier CE, Hall AK. 2004. a single wave before oligodendrocytes. Dev Biol 139:149–168.
Patterning of spinal cord oligodendrocyte development by dorsally Small RK, Riddle P, Noble M. 1987. Evidence for migration of
derived BMP4. J Neurosci Res 76:9–19. oligodendrocyte-type2 astrocyte progenitor cells into the developing
Mizuguchi R, Sugimori M, Takebayashi H, Kosako H, Nagao M, rat optic nerve. Nature 328:115–117.
Yoshida S, et al. 2001. Combinatorial roles of olig2 and neurogenin2 Sohn J, Natale J, Chew LJ, Belachew S, Cheng Y, Aguirre A, et al. 2006.
in the coordinated induction of pan-neuronal and subtype-specific Identification of Sox17 as a transcription factor that regulates oligo-
properties of motoneurons. Neuron 31:757–771. dendrocyte development. J Neurosci 26:9722–9735.
Nery S, Wichterle H, Fishell G. 2001. Sonic hedgehog contributes Sommer I, Schachner M. 1981. Monoclonal antibodies (O1 to O4) to oli-
to oligodendrocyte specification in the mammalian forebrain. godendrocyte cell surfaces: an immunocytological study in the central
Development 128:527–540. nervous system. Dev Biol 83:311–327.
Nishiyama A, Lin X-H, Giese N, Heldin C-H, Stallcup WB. 1996. Spassky N, de Castro F, Le Bras B, Heydon K, Quéraud-LeSaux F,
Co-localization of NG2 proteoglycan and PDGF α-receptor on O2A Bloch-Gallego E, et al. 2002. Directional guidance of oligoden-
progenitor cells in the developing rat brain. J Neurosci Res 43:299–314. droglial migration by class 3 semaphorins and netrin-1. J Neurosci
Noble M, Murray K, Stroobant P, Waterfield MD, Riddle P. 1988. 22:5992–6004.
Platelet-derived growth factor promotes division and motility and Spassky N, Heydon K, Mangatal A, Jankovski A, Olivier C, Queraud-Lesaux
inhibits premature differentiation of the oligodendrocyte/type-2 F, et al. 2001. Sonic hedgehog-dependent emergence of oligodendro-
astrocyte progenitor cell. Nature 333:560–562. cytes in the telencephalon: evidence for a source of oligodendrocytes
Olivier C, Cobos I, Perez Villegas EM, Spassky N, Zalc B, Martinez S, in the olfactory bulb that is independent of PDGFR alpha signaling.
et al. 2001. Monofocal origin of telencephalic oligodendrocytes in Development 128:4993–5004.
the anterior entopeduncular area of the chick embryo. Development Stallcup W. 1981. The NG2 antigen, a putative lineage marker:
128:1757–1769. Immunofluorescent localization in primary cultures of rat brain. Dev
Ono K, Bansal R, Payne J, Rutishauser U, Miller RH. 1995. Early devel- Biol 83:154–165.
opment and dispersal of oligodendrocyte precursors in the embryonic Stallcup WB, Beasley L. 1987. Bipotential glial precursor cells of the optic
chick spinal cord. Development 121:1743–1754. nerve express the NG2 proteoglycan. J Neurosci 7:2737–2744.
Ono K, Yasui Y, Rutishauser U, Miller RH. 1997. Focal ventricular ori- Stallcup WB, Beasley L, Levine J. 1983. Cell-surface molecules that char-
gin and migration of oligodendrocyte precursors into the chick optic acterize different stages in the development of cerebellar intemeurons.
nerve. Neuron 19:283–292. Cold Spring Harbor Symp Quant Biol 48:761–774.
Ono K, Tsumori T, Yokota S, Yasui Y. 2001. Extensive proliferation Stolt CC, Lommes P, Sock E, Chaboissier MC, Schedl A, Wegner M.
of oligodendrocyte precursors in the parenchyma of the embryonic 2003. The Sox9 transcription factor determines glial fate choice in
chick central nervous system. Dev Biol 231:77–86. the developing spinal cord. Gene Dev 17:1677–1689.
Orentas DM, Hayes JE, Dyer KL, Miller RH. 1999. Sonic hedgehog sig- Stolt CC, Rehberg S, Ader M, Lommes P, Riethmacher D, Schachner
naling is required during the appearance of spinal cord oligodendro- M, et al. 2002. Terminal differentiation of myelin-forming oligo-
cyte precursors. Development 126:2419–2429. dendrocytes depends on the transcription factor Sox10. Gene Dev
Park SK, Miller R, Krane I, Vartanian T. 2001. The erbB2 gene is 16:165–170.
required for the development of terminally differentiated spinal cord Stritt C, Stern S, Harting K, Manke T, Sinske D, Schwarz H, et al. 2009.
oligodendrocytes. J Cell Biol 154:1245–1258. Paracrine control of oligodendrocyte differentiation by SRF-directed
Parras CM, Hunt C, Sugimori M, Nakafuku M, Rowitch D, Guillemot neuronal gene expression. Nat Neurosci 12:418–427.
F. 2007. The proneural gene Mash1 specifies an early population of Sugimori M, Nagao M, Bertrand N, Parras CM, Guillemot F, Nakafuku M.
telencephalic oligodendrocytes. J Neurosci 27:4233–4242. 2007. Combinatorial actions of patterning and HLH transcription fac-
Pringle NP, Richardson WD. 1993. A singularity of PDGF alpha- tors in the spatiotemporal control of neurogenesis and gliogenesis in the
receptor expression in the dorsoventral axis of the neural tube may developing spinal cord. Development 134: 1617–1629.
define the origin of the oligodendrocyte lineage. Development Sugimoto Y, Taniguchi M, Yagi T, Akagi Y, Nojyo Y, Tamamaki N.
117:525–533. 2001. Guidance of glial precursor cell migration by secreted cues in
Pringle NP, Yu W, Guthrie S, Roelink H, Lumsden A, Peterson AC, the developing optic nerve. Development 128:3321–3330.
et al. 1996. Determination of neuroepithelial cell fate: induction of Su, T, Dong H, Wu L, Kane M, Rowitch DH, Stiles CD. 2003.
the oligodendrocyte lineage by ventral midline cells and sonic hedge- Cross-repressive interaction of the Olig2 and Nkx2.2 transcription
hog. Dev Biol 177:30–42. factors in developing neural tube associated with formation of a spe-
Raff M, Mille, RH, Noble M. 1983. A glial progenitor that develop in cific physical complex. J Neurosci 23:9547–9556.
vitro into an astrocyte or an oligodendrocyte depending on culture Sun T, Echelard Y, Lu R, Yuk DI, Kaing S, Stiles CD, et al. 2001. Olig
medium. Nature 303:390–396. bHLH proteins interact with homeodomain proteins to regulate cell
Rao MS, Noble M, Mayer-Proschel M. 1998. A tripotential glial precur- fate acquisition in progenitors of the ventral neural tube. Current
sor cell is present in the developing spinal cord. Proc Natl Acad Sci U Biol 11:1413–1420.
S A 95:3996–4001. Takebayashi H, Nabeshima Y, Yoshida S, Chisaka O, Ikenaka K,
Relucio J, Tzvetanova ID, Ao W, Lindquist S, Colognato H. 2009. Nabeshima Y. 2002. The basic helix-loop-helix factor Olig2 is essen-
Laminin alters fyn regulatory mechanisms and promotes oligoden- tial for development of motoneuron and oligodendrocyte lineages.
drocyte development. J Neurosci 29:11794–11806. Current Biol 12:1157–1163.
Richardson WD, Smith HK, Pringle NP, Hall A, Woodruff R. 2000. Tawk M, Makoukji J, Belle M, Fonte C, Trousson A, Hawkins T, et al.
Oligodendrocyte lineagte and the motor neuron connection. Glia 2011. Wnt/-catenin signaling is an essential and direct driver of mye-
29:136–142. lin gene expression and myelinogenesis. J Neurosci 31: 3729–3742.
Samanta J, Kessler JA. 2004. Interactions between ID and OLIG pro- Temple S, Raff MC. 1986. Clonal analysis of oligodendrocyte develop-
teins mediate the inhibitory effects of BMP4 on oligodendroglial dif- ment in culture: evidence for a developmental clock that counts cell
ferentiation. Development 131:4131–4142. divisions. Cell 44:773–779.

L I N E AG E A N D D E VE L O PM E N T: O L I G O D E N D R O C Y T E S • 157
Tikoo R, Osterhout DJ, Casaccia-Bonnefi l P, Seth P, Koff A, Warf BC, Fok-Seang J, Miller RH. 1991. Evidence for the ventral origin
Chao MV. 1998. Ectopic expression of p27Kip1 in oligodendrocyte of oligodendrocytes in the rat spinal cord. J Neurosci 11: 2477–2488.
progenitor cells results in cell-cycle growth arrest. J Neurobiol Wake H, Lee PR, Fields RD. 2011. Control of local protein synthe-
36:431–440. sis and initial events in myelination by action potentials. Science
Timsit SG, Bally-Cuif L, Colman DR, Zalc B. 1992. DM-20 mRNA is 333:1647–1651.
expressed during the embryonic development of the nervous system Wang S, Sdrulla AD, diSibio G, Bush G, Nofziger D, Hicks C, et al.
of the mouse. J Neurochem 58:1172–1175. 1998. Notch receptor activation inhibits oligodendrocyte differen-
Timsit S, Martinez S, Allinquant B, Peyron F, Puelles L, Zalc B. 1995. tiation. Neuron 21:63–75.
Oligodendrocytes originate in a restricted zone of the embryonic Wilson HC, Onischke C, Raine CS. 2003. Human oligodendrocyte
ventral neural tube defined by DM-20 mRNA expression. J Neurosci precursor cells in vitro: phonotypic analysis and differential response
15:1012–1024. to growth factors. Glia 44:153–165.
Tripathi RB, Clarke LE, Burzomato V, Kessaris N, Anderson PN, Xin M, Yue T, Ma Z, Wu FF, Gow A, Lu QR. 2005. Myelinogenesis
Attwell D, et al. 2011. Dorsally and ventrally derived oligodendro- and axonal recognition by oligodendrocytes in brain are uncoupled
cytes have similar electrical properties but myelinate preferred tracts. in Olig1-null mice. J Neurosci 25:1354–1365.
J Neurosci 31:6809–6819. Ye F, Chen Y, Hoang T, Montgomery RL, Zhao XH, Bu H, et al. 2009.
Tsai H-H, Frost E, To V, Robinson S, ffrench-Constant C, Geertman HDAC1 and HDAC2 regulate oligodendrocyte differentiation by dis-
R, et al. 2002. The chemokine receptor CXCR2 controls positioning rupting the beta-catenin-TCF interaction. Nat Neurosci 12:829–838.
of oligodendrocyte precursors in developing spinal cord by arresting Zeger M, Popken G, Zhang J, Xuan S, Lu QR, Schwab MH, et al. 2007.
their migration. Cell 110:373–383. Insulin-like growth factor type 1 receptor signaling in the cells of oli-
Tsai H-H, Tessier-Lavigne M, Miller RH. 2003. Netrin 1 mediates godendrocyte lineage is required for normal in vivo oligodendrocyte
spinal cord oligodendrocyte precursor dispersal. Development development and myelination. Glia 55:400–411.
130:2095–2105. Zhang S-C, Ge B, Duncan ID. 2000. Tracing human oligodendroglial
Vallstedt A, Klos JM, Ericson J. 2005. Multiple dorsoventral origins of development in vitro. J Neurosci Res 59:412–429.
oligodendrocyte generation in the spinal cord and hindbrain. Neuron Zhou Q, Anderson DJ. 2002. The bHLH transcription factors OLIG2
45:55–67. and OLIG1 couple neuronal and glial subtype specification. Cell
Wada T, Kagawa T, Ivanova A, Zalc B, Shirasaki R, Murakami F, et al. 109:61–73.
2000. Dorsal spinal cord inhibits oligodendrocyte development. Dev Zhou Q, Choi G, Anderson DJ. 2001. The bHLH transcription factor
Biol 227:42–55. Olig2 promotes oligodendrocyte differentiation in collaboration
Wang SZ, Dulin J, Wu H, Hurlock E, Lee SE, Jansson K, et al. 2006. with Nkx2.2. Neuron 31:791–807.
An oligodendrocyte-specific zinc-finger transcription regulator Zhou Q, Wang S, Anderson DJ. 2000. Identification of a novel family of
cooperates with Olig2 to promote oligodendrocyte differentiation. oligodendrocyte lineage-specific basic helix-loop-helix transcription
Development 133:3389–3398. factors. Neuron 25:331–343.

158 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
14.
THE SCHWANN CELL LINEAGE: CELLULAR TRANSITIONS
DURING DEVELOPMENT AND AFTER INJURY
Kristján R. Jessen and Rhona Mirsky

A B B R E VI AT I O N S 2 O VE RVI EW O F T H E L I N E AG E

ADAM A disintegrin and metalloprotease protein Schwann cell precursors are the earliest glial phenotype in devel-
BACE1 (β-secretase) beta-site APP-cleaving oping spinal nerves (Jessen et al. 1994). They derive from neu-
enzyme ral crest cells that break free from the closing neural tube and
BDNF Brain derived neurotrophic factor migrate through the extracellular matrix before they convert
CHO Chinese hamster ovary to the Schwann cell precursor phenotype as they take up posi-
CNS central nervous system tions among the axons of early nerves. In mouse sciatic nerves,
Dhh desert hedgehog Schwann cell precursors are found at embryo day (E)12–13 (rat
DRG dorsal root ganglia E14–15). In contrast to crest cells, Schwann cell precursors lie
ERK Ras-extracellular signal-regulated kinas in intimate association with neurons (axons), a feature that is a
FAK focal adhesion kinase distinguishing characteristic of glial cells throughout the unper-
GDNF Glia derived neurotrophic factor turbed central nervous system (CNS) and peripheral nervous
GFAP Glia fibrillary acidic protein system (PNS) (Fig. 14.3). Schwann cell precursors generate
Ilk integrin linked kinase immature Schwann cells that populate mouse nerves from E15–
JNK c-Jun N terminal kinase 16 (rat E17–18). Starting around birth (E20–21), the process of
Lgi leucine-rich glioma-inactivated radial sorting results in some immature Schwann cells adopt-
LIF leukemia inhibitory factor ing a 1:1 relationship with single large diameter axons, form-
LPA lysophosphatidic acid ing promyelin cells. In small mammals, the large majority of the
MAPK mitogen-activated protein kinases cells that achieve a 1:1 relationship with axons progress to form
MBP myelin basic protein myelin sheaths. Other immature Schwann cells do not take part
MCP1 Monocyte chemoattractant protein1 in radial sorting, but later associate with small-diameter axons,
NCAM Neural cell adhesion molecule accommodating each axon in a separate pocket along their sur-
NRG1 β -NEUREGULIN 1 face, thus forming mature Remak fibers; in rat nerves they are
P0 myelin protein zero first seen during the third postnatal week (Diner 1965; Jessen
p75NTR p75 neurotrophin receptor and Mirsky 2005; Webster and Favilla 1984).
PNS peripheral nervous system The development of Schwann cell precursors and immature
RBPJ recombining binding protein suppressor of Schwann cells is controlled by axon-associated signals. These sig-
hairless nals are essential for the survival and differentiation of Schwann
TACE tumor necrosis factor α-converting enzyme cell precursors, determine whether immature Schwann cells myeli-
nate or form Remak cells, and control the elaborate molecular and
morphological architecture of myelinating cells. Additionally,
1 INTRODUCTION axonal signals drive the proliferation of Schwann cell precur-
sors and immature Schwann cells throughout early development
The Schwann cell lineage in rodents exhibits four main cel- (Jessen and Mirsky 2005; Stewart et al. 1993; Yu et al. 2005).
lular transitions ( Jessen and Mirsky 2005). These are: (1) the Although Schwann cells in adult nerves are quiescent, they retain
generation of Schwann cell precursors; (2) the generation of the ability to re-enter the cell cycle following injury and in patho-
immature Schwann cells, both of which take place in embry- logical conditions (Jessen and Mirsky 2005, 2008).
onic nerves; (3) the largely postnatal differentiation of myelin Neural crest cells, Schwann cell precursors and immature
and mature nonmyelin (Remak) cells; and (4) in injured Schwann cells are each faced with a fate choice during devel-
nerves, the generation of dedicated repair Schwann cells opment. This is well established for crest cells, and also for
(Figs. 14.1 and 14.2). This chapter describes the cellular immature Schwann cells because they can form both myelin
changes that characterize these four main transitions, and dis- and Remak cells. It is only recently, however, that Schwann
cusses the signals that drive cells between differentiation states cell precursors have been shown to generate endoneurial fibro-
in developing and injured adult nerves. blasts and skin melanocytes in addition to immature Schwann

159
Main transitions in the Schwann cell lineage crest cells (see Fig. 14.2) (Buchstaller et al. 2004; D’Antonio et al.
2006b; Li et al. 2007; Mirsky et al. 2008). Second, as mentioned,
Myelin Schwann cell precursors exhibit the diagnostic glial phenotype
Axon Schwann cell of intimate association with nerve cells, because they are found
among axons within nerves, whereas crest cells migrate through
mesenchymal connective tissue. Third, Schwann cell precursors
Büngner cell and crest cells respond differently to survival signals and mitogens
(Woodhoo et al. 2004). Last, compared with crest cells Schwann
cell precursors are insensitive to the neurogenic action of BMP2
Remak cell
and strongly biased toward the generation of immature Schwann
Immature
cells (Kubu et al. 2002; Woodhoo and Sommer 2008).
Neural Schwann cell The next transition, the generation of immature Schwann
crest precursor (SCP) Schwann cell
(iSch) cells from Schwann cell precursors, also involves substantial
changes in gene expression (see Fig. 14.2) (Buchstaller et al.
E 12/13 E 15/16- ADULT 2004; D’Antonio et al. 2006b; Dong et al. 1999; Frank et al.
EARLY POSTNATAL
1999). Schwann cell precursors and immature Schwann cells
also show a marked difference in survival regulation. Schwann
Figure 14.1 Main Transitions in the Schwann Cell Lineage. Schematic cell precursors are acutely dependent on axonal survival sig-
illustration showing the main cell types and developmental and
injury-induced transitions. Black uninterrupted arrows: normal nals, chiefly β-neuregulin 1 (NRG1) (Dong et al. 1995),
development. Red arrows: the Schwann cell injury response. Stippled whereas immature Schwann cells can prevent their own death
arrows: post-repair formation of myelin and Remak cells. by secretion of autocrine survival factors, a mechanism that
Schwann cell precursors lack (Dong et al. 1999; Meier et al.
cells (Adameyko et al. 2009; Joseph et al. 2004). This unex- 1999). Because Schwann cell precursors lack a basal lamina,
pected diversity of options open to Schwann cell precursors formation of basal lamina by immature Schwann cells repre-
is reminiscent of that seen in early CNS glia such as radial sents another difference between these cells.
glia, and indicates that a broad developmental potential is a Cadherin 19 is first activated when Schwann cell precur-
characteristic of early glial cells both in the CNS and PNS sors are formed from the crest, but is downregulated in imma-
(Adameyko et al. 2009; Jessen and Mirsky 2005; Joseph et al. ture Schwann cells. It is therefore the only known gene that
2004; Mirsky et al. 2008). selectively marks the Schwann cell precursor stage.
Our detailed knowledge of Schwann cell development is
derived mainly from studies on spinal nerves and in particu-
lar the sciatic nerve. The process is not identical in dorsal and 4 GLIOGENESIS: THE APPEARANCE
ventral nerve roots, where the Schwann cells have a distinct O F S C H WA N N C E L L P R E C U R S O R S
molecular characteristics and origin, because they develop
from boundary cap cells (Coulpier et al. 2009; Maro et al. Transcription factors that specify glial cell differentiation
2004). Other distinct groups of PNS glia include satellite cells from crest cells have not been identified (Cheli et al. 2010;
in ganglia and enteric glial cells (see chapter 11). Marmigère and Ernfors 2007). Similarly, cell-extrinsic factors
A small population of progenitor/precursor cells commonly required for initiation of PNS glial differentiation in vivo have
involved in tissue regeneration and repair is found in many not been clearly determined (Woodhoo and Sommer 2008).
adult tissues. Although such “standby” cells do not appear to be Three major factors that have been shown (Sox10), or sug-
present in uninjured nerves, damaged rodent nerves regenerate gested (NRG1 and Notch), to be involved in Schwann cell
successfully. A main reason for this is that injury triggers a strik- precursor generation are discussed in the following.
ing transformation of the myelin and Remak cells to generate
denervated Schwann cells that form regeneration tracks (Bands
of Büngner) distal to the injury (Fig. 14.4). These Büngner cells 4.1 S OX10
are specialized to support neuronal survival, axon regenera- The transcription factor Sox10 is required for the generation of
tion and target reinnervation. At the completion of the repair glia from the crest, but because it is expressed by essentially all
process they are re-specified to form myelin and Remak cells migrating crest cells before the appearance of glial cells and at
around the regenerated axons ( Jessen and Mirsky 2008). the earliest stages of the melanocyte lineage, it does not specify
glial cells. An important role of Sox10 may be to uphold expres-
sion of ErbB3 receptors for NRG1, a signal with key functions
3 D I F F E R E N C E S B ET W E E N T H E N E U R A L in Schwann cell development (Britsch et al. 2001).
C R E S T S C H WA N N C E L L P R E C U R S O R S ,
A N D I M M AT U R E S C H WA N N C E L L S
4.2 E -N EU R EGU L I N 1
Schwann cell precursors within embryonic nerves differ from NRG1 does not appear to be required for glial cell formation
migrating neural crest cells, first, because they express glial dif- from the crest for the following reasons. First, in zebrafish,
ferentiation markers and other factors not found on migrating Schwann cells are generated despite inactivating mutations in

160 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Associate Associate Associate
with ECM with axons with axons
NRG1 survival NRG1 survival NRG1 survival
signaling is signaling is signaling is
ECm dependent ECM independent ECM independent

Neural crest SCP iSch


ErbB3 ErbB3 ErbB3
NO CHANGE L1 L1 L1
p75NTR p75NTR p75NTR
Sox10 Sox10 Sox10
BFABP
Dhh
Po
GAP43
PMP22
PLP
Connexin 29
PrPc
UPREGULATION Astrotactin
Serpin2 GFAP
NFIB S100
Cad19 Oct6
O4
MAL
Galectin
Desmoyokin
Reelin
Decorin

α4integrin
DOWNREGULATION AP2
Ncad
Cad19

Figure 14.2 The Phenotype of the Main Stages of the Embryonic Schwann Cell Lineage. Below the lineage drawing some of the molecular markers that
characterize the lineage are shown. Four groups are shown: (1) markers that show no significant change between the three stages; (2) markers that are
upregulated at the crest/Schwann cell precursor transition; (3) markers that are upregulated at the Schwann cell precursor/immature Schwann cell
transition; and (4) markers that are downregulated at the Schwann cell precursor/immature Schwann cell transition. For references see text and Jessen
and Mirsky 2005.

the NRG1 receptor ErbB3 (Lyons et al. 2005). Second, in rat 4.3 N OTC H
neural crest cultures, cells expressing P0 mRNA, a Schwann cell
Notch increases the number of Schwann cells in rat neural
precursor marker, appear readily without addition of NRG1
crest cultures, using the late glial marker GFAP to monitor
(Woodhoo et al. 2009), and this is also true of GFAP-positive
Schwann cell appearance (Kubu et al. 2002). Other studies
Schwann cells (Shah et al. 1994). Last, the satellite cells in
on mouse and chick crest cells using the earlier differentia-
DRG, a major category of PNS glia, appear to form normally
tion marker P0 mRNA, which detects the initial appearance
in mouse mutants in which NRG1 or functional ErbB2 or 3
of Schwann cell precursors, rather than GFAP that identifies
receptors are missing (Garratt et al. 2000). Taken together,
the subsequent generation of Schwann cells, were unable to
this shows that NRG1 signaling is unlikely to be obligatory
detect this effect. Notch activation in neural crest cells in ovo
for crest cells to adopt a glial fate.
also failed to promote Schwann cell generation (Wakamatsu
Nevertheless, NRG1 may bias crest cells toward glial dif-
et al. 2000; Woodhoo et al. 2009). In mice lacking Hes1 and
ferentiation indirectly. For instance, one of the most striking
Hes 5, the effectors of canonical Notch signaling, the appear-
effects of NRG1 on neural crest cultures is inhibition of neu-
ance of Schwann cell precursors was also unaffected (Woodhoo
rogenesis. This effect could increase gliogenesis by extend-
et al. 2009). Similarly, in mice lacking the transcription factor
ing the exposure of crest cells to gliogenic signals (Shah et al.
RBPJ, a key mediator of canonical Notch signaling, Schwann
1994). After the onset of glial development, NRG1 pro-
cells were generated, although formation of satellite glia, and to
motes the Schwann cell precursor/immature Schwann cells
a lesser extent neurons, in sensory ganglia was impaired (Taylor
transition and the generation of Schwann cells by promoting
et al. 2007). Therefore, it is unlikely that the generation of
their survival and proliferation.

T H E S C H WA N N C E L L L I N E AG E : C E L LU L A R T R A N S I T I O N S D U R I N G D E VE L O PM E N T A N D A F T E R I N J U RY • 161
Schwann cell precursors in spinal nerves is significantly regu-
lated by Notch signaling. On the other hand, Notch accelerates
the subsequent step in Schwann cell development, the Schwann
cell precursor/immature Schwann cell transition, discussed in
the following.

5 E -N E UR E GUL IN 1 A N D N OTC H
S I G N A L IN G IN S C H WA N N C E L L
P R E C UR S O R S

In mouse mutants in which NRG1 or the NRG1 receptors


ErbB2 or ErbB3 have been inactivated, there is a conspicuous
loss of Schwann cell precursors (Birchmeier 2009; Birchmeier
and Nave 2008; Garratt et al. 2000). As discussed, it is proba-
ble that crest cells can initiate glial fate without NRG1 signals.
Therefore, it is likely that the loss of Schwann cell precursors
in these embryos results from the role of NRG1 in controlling
Schwann cell precursor survival and migration.

5.1 S U RVI VA L
Unlike Schwann cells, Schwann cell precursors cannot be grown
Figure 14.3 Schwann cell precursors and immature Schwann cells in in vitro without the addition of survival factors. This is because
embryonic nerves. Upper panel shows a transverse section through E14 rat Schwann cell precursors, but not Schwann cells, depend on sur-
sciatic nerve. Schwann cell precursors are embedded among the axons and
at the surface of the nerve (big arrows). A dividing Schwann cell precursor
vival signals from axons. In cell culture, Schwann cell precursor
is also seen (small arrow). Connective tissue (turquoise) is not found inside death can be prevented by contact with axons, and the axonal
the nerve. Lower panel shows a transverse section through E18 rat sciatic signal responsible for this contact-mediated rescue of Schwann
nerve. One or a few immature Schwann cells together surround a number cell precursors has been identified as NRG1. Soluble NRG1 is
of axons, forming compact groups (families; asterisk). A dividing immature also a potent survival factor for these cells (Dong et al. 1995).
Schwann cell is seen (double arrows). Connective tissue (turquoise) contain-
ing blood vessels (large arrow) is present throughout the nerve surrounding
In vivo, the Schwann cell precursor death that results from
the families. Bracket indicates the developing perineurium. Adapted from neuronal degeneration is prevented by application of NRG1
Jessen and Mirsky 2005. (Winseck and Oppenheim 2006). NRG1 is present on axonal

Myelin cells Cells that accelerate the


conduction of action potentials

The Schwann cell


injury response

Büngner cells
(forming Büngner bands)

c-Jun-dependent Dedifferentiation
repair program program

Upregulation of Myelin breakdown


molecules that Formation of Downregulation Upregulation
regeneration tracks directly and by of myelin genes of iSch markers
promote neuronal macrophage
survival (Bands of Büngner)
recruitment
and growth

Cells that control nerve repair

Figure 14.4 The Schwann Cell Injury Response Main events during the transdifferentiation of myelin cells to form Büngner repair cells.

162 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
surfaces and is expressed in DRG and motor neurons at the 6 S C H WA N N C E L L G E N E R AT I O N
developmental stage when Schwann cell precursors are found AND THE ARCHITECTUR AL
in nerves (Birchmeier and Nave, 2008; Taveggia et al. 2005). R E O R G A N I Z AT I O N O F
Therefore, NRG1 is expressed at the right time and place to con- P E R I P H E R A L N E RVE S
trol Schwann cell precursor survival. These observations indicate
that axonal NRG1 is an essential survival factor for Schwann cell The Schwann cell precursor/immature Schwann cell transition
precursors in embryonic nerves. Therefore, a major reason for is accompanied by a striking reordering of cell and tissue relation-
the lack of Schwann cell precursors in NRG1 mutants is likely to ships within nerves (Wanner et al. 2006b). At E12/13 in mouse
be cell death owing to the absence of NRG1 signaling. (rat E14/15) embryonic nerves are compact, consisting of tightly
packed axons and flattened sheetlike processes of Schwann cell
precursors separating large groups of axons. The Schwann cell
5.2 M I G R AT I O N
precursor cell bodies are found at the nerve surface, where these
NRG1 stimulates migration of developing Schwann cells along flattened cells separate the axons from surrounding connective
zebrafish nerves in vivo and the migration of rat Schwann tissue and, in larger nerves, also embedded among the axons (see
cells in vitro (Lyons et al. 2005; Meintanis et al. 2001; Yamauchi Fig. 14.3). Blood vessels and connective tissue, including fibro-
et al. 2008). In mutants without NRG1 signaling, the crest fails blasts and extracellular spaces containing collagen, are excluded
to migrate to form sympathetic ganglia, although the forma- from these early nerves. This architecture transforms during the
tion of DRG appears relatively unaffected (Britsch et al. 1998). next 2 to 3 days, at the same time as Schwann cell precursors
Therefore, failure of cell migration in early nerves is likely to convert to immature Schwann cells. An E18 nerve is a collection
contribute to the lack of Schwann cell precursors in NRG1 of axon-immature Schwann cell bundles (“families”) (Webster
mutants (Birchmeier 2009). and Favilla 1984) lying in connective tissue spaces containing
blood vessels and collagen, whereas basal lamina is forming at
the immature Schwann cell surfaces (Wanner et al. 2006b).
5.3 N RG1 O N D EV E L O P I N G AXO N S
At this time the developing perineurial sheath can first be dis-
Most of the known effects of axonal NRG1 on Schwann cerned (Parmantier et al. 1999). The signals that control these
cell precursors or Schwann cells are mediated by isoform III. changes are unknown, although it has been suggested that they
Following proteolytic cleavage, the EGF-like NRG1 domain are promoted by the downregulation of glial N-cadherin expres-
is presented on the axonal surface and signals in a juxtacrine sion that takes place at the Schwann cell precursor/immature
fashion by binding to ErbB2/3 receptors on adjacent glial Schwann cell transition (Wanner et al. 2006b).
cells (Birchmeier 2009; Taveggia et al. 2005). The membrane- We know more about the regulation of the Schwann cell
associated protease BACE1 (β-secretase) is involved in activa- precursor/immature Schwann cell transition, in particular
tion of NRG1 signaling from the axonal surface, whereas its the positive role of Notch and NRG1 (previous section). The
opposing enzyme TACE inhibits myelination (Hu et al. 2006; Schwann cell precursor/immature Schwann cell transition is
La Marca et al. 2011; Willem et al. 2006). negatively regulated by two signals, endothelin and the tran-
scription factor AP2α. Endothelin delays immature Schwann
cell generation from Schwann cell precursors in vitro, and
5.4 β -N EU R EGU L I N 1 A N D N OTC H I N T E R AC T
inactivation of endothelin B receptors in vivo results in prema-
TO P RO MOT E S C H WA N N C E L L P R ECU R S O R
ture appearance of immature Schwann cells, confirming that
S U RVI VA L A N D I M M AT U R E S C H WA N N
endothelin signaling negatively regulates immature Schwann
C E L L G E N E R AT I O N
cell generation (Brennan et al. 2000).
Notch ligands, like NRG1, are expressed on axons. Unlike Enforced AP2α expression in Schwann cell precursors in
NRG1 signaling, activation of Notch signaling alone in vitro retards the conversion of these cells to immature Schwann
Schwann cell precursors does not promote survival. But cells, whereas in vivo this transcription factor is strongly down-
Notch promotes the survival effects of NRG1, particularly at regulated at the Schwann cell precursor/immature Schwann
low NRG1 concentrations (Woodhoo et al. 2009). Another cell transition, suggesting that AP2 α is involved in maintaining
effect of Notch on Schwann cell precursors is to accelerate the the Schwann cell precursor phenotype (Stewart et al. 2001).
generation of immature Schwann cells. Thus the appearance of
immature Schwann cells is delayed in mice with conditional
activation of Notch1 or the transcription factor RBPJ, which 7 T H E MU LT I P OT E N T S C H WA N N
transduces canonical Notch signaling. Conversely, immature CELL PRECUR SOR : A SOURCE OF
Schwann cells are generated ahead of schedule if Notch1 is MEL ANOCY TES AND FIBROBL ASTS
overexpressed (Woodhoo et al. 2009).
Both of these effects of Notch are probably because Notch Although the broad developmental potential of Schwann cell
elevates expression of the NRG1 receptor ErbB3 in Schwann precursor was first detected in vitro ( Jessen and Mirsky 2004),
cell precursors (Woodhoo et al. 2009). This and the fact that there is now evidence that Schwann cell precursors generate
NRG1 promotes Schwann cell survival and differentiation both melanocytes and fibroblasts during normal development
allows these functions to be regulated indirectly by Notch in vivo ( Joseph et al. 2004). In mice a significant number of
(Brennan et al. 2000; Woodhoo et al. 2009). skin melanocytes originate from Schwann cell precursors,

T H E S C H WA N N C E L L L I N E AG E : C E L LU L A R T R A N S I T I O N S D U R I N G D E VE L O PM E N T A N D A F T E R I N J U RY • 163
and migrate from nerves to skin. Although melanogenesis 2001; Grim et al. 1992; Woldeyesus et al. 1999). A comparable
is restricted to Schwann cell precursors during development observation has been made in zebrafish (Raphael and Talbot
(Takahashi and Osumi 2005), adult myelin Schwann cells 2011).
appear capable of melanogenesis after injury, corroborating a The early stages of synapse formation are also normal with-
previous finding (Adameyko et al. 2009; Rizvi et al. 2002). out Schwann cell precursors or Schwann cells, although the
In principle, this confirms the ability of PNS glia to generate establishment of a normal, functioning relationship between
melanocytes, and provides a striking example of the plasticity nerve and muscle ultimately fails. This is owing to neuronal
of the Schwann cell phenotype. death (discussed earlier), defective fasciculation and abnormal
Lineage tracing indicates that endoneurial fibroblasts orig- axon growth and branching within the target tissue (Lin et al.
inate from cells in the nerve that express Dhh, a gene expressed 2000; Morris et al. 1999; Woldeyesus et al. 1999; Wolpowitz
by Schwann cell precursors and immature Schwann cells, but et al. 2000).
not crest cells (Bitgood and McMahon 1995; Jaegle et al. The finding that glial cells are required for the transforma-
2003; Joseph et al. 2004; Parmantier et al. 1999). The notion tion of growing nerve tips into fully formed synapses on muscle
that both fibroblasts and immature Schwann cells originate is not surprising when the architecture of growing nerve endings
from Schwann cell precursors, accords well with the fact that is considered (Wanner et al. 2006a). Quantitative, ultrastruc-
the disappearance of Schwann cell precursors dovetails with tural analysis shows that Schwann cell precursors form com-
the appearance of both immature Schwann cell and fibroblasts plex scaffolds among the axonal growth cones, and the amount
during nerve development (previous section). of membrane contact between Schwann cell precursors and
growth cones is remarkably constant from nerve front to nerve
front (Wanner et al. 2006a). Clearly, this intricate arrangement
8 S C H WA N N C E L L P R E C U R S O R S A N D between neurons and glia is essential for normal nerve fascicu-
E A R LY S C H WA N N C E L L S C O N T R O L lation, branching, and interactions with target tissues.
N E U R O N A L S U RVI VA L , FA S C I C U L AT I O N,
A N D SY N A P S E F O R M AT I O N
9 E VE N T S L E A D I N G TO S C H WA N N C E L L
8.1 N EU RO NA L S U RV I VA L D I VE R S I F I C AT I O N : P R O L I F E R AT I O N,
D E AT H , A N D R A D I A L S O RT I N G
Studies on embryonic nerves provide compelling support
for the idea that neuronal survival depends on glia. Because In late embryonic nerves, before myelination starts, immature
Sox10, ErbB2, or 3 or NRG1(isoform III) are important for Schwann cells ensheathe bundles of axons to form axon–
glial development, mutants lacking these genes all have in com- Schwann cell families (previous section). The key developmen-
mon that early glia in embryonic nerves are absent or much tal events at this stage are: (1) cell proliferation; (2) apoptotic
reduced in numbers (Britsch et al. 2001; Garratt et al. 2000; cell death, which together with proliferation matches the
Woldeyesus et al. 1999; Wolpowitz et al. 2000). Interestingly, number of immature Schwann cell and axons during develop-
all the mutants show a striking loss of spinal cord and DRG ment; (3) radial sorting of axons from the bundles to gener-
neurons at cervical and lumbar levels late in embryonic devel- ate a 1:1 relationship between axons and Schwann cells; and
opment, although these neurons are initially generated in (4) the transition from the 1:1 stage to myelination.
normal numbers. This suggests that the survival of distinct Although these events are obviously coordinated, in prin-
of CNS and PNS neuronal populations depends on signals ciple they can be regulated independently. Notch signaling,
from Schwann cell precursors and perhaps also immature for instance, drives proliferation but does not control sur-
Schwann cell and developing satellite cells in sensory ganglia. vival (Woodhoo et al. 2009). TGFβ, through TGFβ type II
It is possible that in these situations neuronal survival is medi- receptors, controls proliferation and death, but not sorting
ated by back-signaling through the intracellular domain of or myelination (D’Antonio et al. 2006a). Conversely, the
axon-associated NRG1, cleaved as a result of binding to glial transcription factor Sox10 controls sorting and myelination,
ErbB2/B3 receptors (Birchmeier and Nave 2008). but is not required for proliferation (Finzsch et al. 2010).
Because axonal signals such as NRG1 are needed for Control of sorting and myelination can also be separated.
Schwann cell precursor survival (earlier section), neurons and This is seen in Oct6 and Krox-20 mutants, in which myeli-
glia appear to be mutually dependent on each other for sur- nation is inhibited, but sorting remains essentially normal
vival during early nerve development. (Svaren and Meijer 2008; Topilko et al. 1994). Last, prolifera-
tion and cell numbers can be controlled without impact on
sorting. This is seen in Notch mutants, in which proliferation
8.2 FA S C I CU L AT I O N A N D S Y NA P S E
is impaired, resulting in about 30% reduction in cell num-
F O R M AT I O N
bers, yet the ratio of sorted to nonsorted cells remains normal
The broad pattern of limb innervation is determined by (Woodhoo et al. 2009). Eventually, however, further reduc-
outgrowing axons, and is not significantly altered in mouse tion in the number of Schwann cells would doubtless affect
mutants lacking Schwann cell precursors. This unexpected the sorting process.
observation was first made in Splotch (Pax2) mutants, and Although these events can be independently controlled,
later confirmed in mice lacking ErbB2 receptors (Britsch et al. the same molecule often takes part in regulating more than

164 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
one event. For instance, laminin directly or indirectly stimu- potent Schwann cell mitogen, and in zebrafish ErbB2 receptor
lates immature Schwann cell proliferation, but it also binds to blockers reduce Schwann cell proliferation (Lyons et al. 2005;
β1 integrin in the Schwann cell membrane to regulate sorting Nave and Salzer 2006; Taveggia et al. 2005).
(Feltri et al. 2008; Yang et al. 2005; Yu et al. 2005). It is intriguing, however, that inactivation of ErbB2 NRG1
receptors results in increased Schwann cell proliferation.
Similarly, in Sox10 mutants, Schwann cell proliferation in
9.1 P RO L I F E R AT I O N postnatal nerves is unaffected or increased in spite of substan-
It is widely accepted that axonal signals promote Schwann cell tial reduction in mRNA levels for the Schwann cell NRG1
proliferation in perinatal nerves. Early evidence for this came receptor ErbB3. Schwann cells also proliferate normally in
from neuron-Schwann cell cocultures and the in vivo observa- cyclin D1–/– mice, although NRG1 is reported to be unable
tion that cell proliferation in neonatal nerves drops if cells are to drive proliferation of cyclin D1–/– Schwann cells in vitro
deprived of axonal contact by nerve transection (Komiyama and in regenerating nerves (Atanasoski et al. 2001; Finzsch
and Suzuki 1992; Salzer et al. 1980). At present, two cell– et al. 2010; Garratt et al. 2000; Kim et al. 2000; Syroid et al.
cell signaling systems are known to drive Schwann cell divi- 2000). In sum, the notion that axonal NRG1 is an important
sion in perinatal nerves in vivo. They are the Notch pathway Schwann cell mitogen in rodent nerves is plausible, but awaits
and transforming growth factor beta (TGFβ)/TGFβ type confirmation in vivo.
II receptor (D’Antonio et al. 2006a; Woodhoo et al. 2009).
In the case of Notch, there is direct evidence that the signaling 9.1.4 Laminin
represents axon–Schwann cell interactions, but the cellular
source of TGFβ in peripheral nerves is unclear. In vivo, basal Removal of laminin from Schwann cells in vivo (γ1
lamina components, the small Rho GTPase cdc42 and focal chain mutants or dy2J/α double mutants) decreases prolif-
adhesion kinase (FAK) also stimulate Schwann cell DNA eration but increases death in perinatal nerves (Chernousov
synthesis (Benninger et al. 2007; Grove et al. 2007; Yu et al. et al. 2008; Yu et al. 2005). In vitro laminin can promote
2005). Schwann cell survival independently of proliferation (Meier
et al. 1999), and other studies confirm that laminin increases
Schwann cell numbers, but fail to determine whether
9.1.1 Notch this is owing to proliferation or survival (McGarvey et al.
Notch ligands are expressed on axons in perinatal nerves, 1984; Yang et al. 2005; Yu et al. 2005). The mechanisms
suggesting that Notch signaling is involved in axon–Schwann underlying these effects are via the activation β1 integrin
cell communication (Woodhoo et al. 2009). Inactivation of receptors, PI3K, FAK and cdc42 (Benninger et al. 2007;
Notch in Schwann cells results in a substantial reduction in Berti et al. 2011; Grove et al. 2007; Yang et al. 2005; Yu
Schwann cell DNA synthesis and cell numbers (see the preced- et al. 2005).
ing) (Woodhoo et al. 2009). Activation of Notch signaling in
Schwann cell cultures is also strongly mitogenic. Together this 9.2 D E AT H
shows that Notch mediated signaling from axons to Schwann
cells is a major driver of Schwann cell proliferation. There is in vivo evidence for two signals that potentially kill
Schwann cells, TGFβ signaling through the TGFβ type II
receptor, and NGF signaling through p75NTR . Normal devel-
9.1.2 Transforming Growth Factor β
opmental death in perinatal nerves is mediated by TGFβ type
In vivo, conditional inactivation of TGFβ type II receptor in II receptors, not p75NTR , presumably acting in concert with
Schwann cells, substantially reduces proliferation (D’Antonio proliferation and survival signals to generate correct imma-
et al. 2006a). Because Schwann cell death is also decreased, ture Schwann cell numbers. Cell death is also seen following
Schwann cell numbers are not altered. This suggests that TGFβ injury (D’Antonio et al. 2006a; Grinspan et al. 1996). Like
stimulates both proliferation and death in perinatal Schwann developmental death, injury-induced death results from sig-
cells. This dual action is also seen in vitro (Einheber et al. naling through TGFβ type II receptors but in this case there
1995; Jessen and Mirsky 2004; Parkinson et al. 2001). Because is an equally significant contribution by p75NTR, presumably
TGFβ stimulated proliferation in the presence of NRG1 but following activation by NGF.
death in the absence of NRG1, it was suggested that TGFβ The function of laminin in supporting immature Schwann
amplifies the proliferation of cells with tight axonal contact cell survival was mentioned earlier. Evidence that NRG1,
and strong NRG1 input, while promoting the death of super- which is an indispensable survival signal for Schwann cell pre-
numerary cells (Parkinson et al. 2001). cursors, also contributes to survival of iSc (immature Schwann
cells) in neonatal nerves, comes from the observation that
9.1.3 Neuregulin exogenous application of NRG1 reduces cell death induced
by axotomy (Grispan et al. 2006).
Although it is often assumed that axonal NRG1 drives Cell culture studies show that Schwann cell numbers
Schwann cell division in developing nerves, direct in vivo evi- are also regulated by autocrine mechanisms. The survival of
dence for this is still missing. In vitro NRG1, on the surface of immature Schwann cell from E18 and older nerves is density
axons, in the membrane of CHO cells or in soluble form, is a dependent. This indicates the existence of autocrine survival

T H E S C H WA N N C E L L L I N E AG E : C E L LU L A R T R A N S I T I O N S D U R I N G D E VE L O PM E N T A N D A F T E R I N J U RY • 165
circuits that enable Schwann cells to live without axonal con- Although a major role of Lgi4/ADAM22, Rac1 and Ilk is
tact or neuronal signals. The autocrine signal consists of a to promote sorting, the data indicate that these systems also
cocktail of factors, including IGF2, NT3, PDGF-B, LIF, and promote myelination proper; that is, wrapping and the for-
LPA ( Jessen and Mirsky 2005). mation of the myelin sheath by cells that have already sorted
Autocrine survival signals are absent from Schwann cell to form the 1:1 axon–Schwann cell relationship. So far, less
precursors, representing one of the most clear-cut phenotypic attention has been paid to this aspect of their function.
differences between Schwann cell precursors and immature
Schwann cell. Because of this, Schwann cell precursors are
entirely dependent on axonal NRG1 type III. This arrange- 10 F R O M S O RT I N G TO M Y E L I N AT I O N
ment is likely to help match the number of Schwann cell pre-
cursors and axons (Dong et al. 1995; Meier et al. 1999). On In rodents, attainment of a 1:1 axon–Schwann cell relation-
the other hand, the ability of most postnatal Schwann cells to ship is usually a step on the way to myelination, although some
survive in the medium term without axons (although some 5% to 15% of sorted axons (<1–1.5 μm in diameter) do not
cells die when axons are cut as described in the preceding) is progress further and remain unmyelinated. And in humans
essential for repair, because effective regeneration depends on most unmyelinated axons have undergone radial sorting
living Schwann cells in the nerve stump distal to the injury. and exist in a 1:1 relationship with Remak Schwann cells
(Berthold et al. 2005; Sharghi-Namini et al. 2006). Radial
9.3 R A D I A L S O RT I N G
sorting to attain the 1:1 axon–Schwann cell relationship is
therefore not a component of a myelination-specific sequence
In addition to its other functions, axonal NRG1 type III is of events.
involved in radial sorting and the establishment of normal Because radial sorting is needed before Schwann cells can
axon–Schwann cell relationships. This applies both before form myelin, the proteins needed for sorting are indirectly
myelination, and during generation of mature Remak cells. required for myelination. Accordingly, the sorting mutants
This function of NRG1 is distinct from its role in activating discussed in the preceding show severe lack of myelin. In con-
the myelination program and controlling myelin thickness trast, many of the proteins that control the myelin program,
(Fricker et al. 2011; Raphael and Talbot 2011; Taveggia et al. including the transcription factors Oct6 and Krox-20, and the
2005). Signals in the opposite direction also control sorting, transcription regulators NAB1/2 do not exert significant con-
from Schwann cells to axons. This is seen in studies on Lgi4, a trol over sorting (Svaren and Meijer 2008).
protein that is secreted from Schwann cells to bind to ADAM Another regulatory mechanism that works primarily by
22, a transmembrane protein on axons (Özkaynak et al. 2010). promoting the myelin program rather than sorting, involves
The transcription factor Sox10, a protein like NRG1with micro RNAs (miRNAs) and RNA binding proteins. Mice
many functions in Schwann cells including the control of with selective deletion of Dicer1, a major miRNA process-
myelination, is also essential for normal sorting. Some of the ing enzyme, from Schwann cells primarily arrest after the 1:1
sorting defects in Sox10 mutants may be caused by reduced promyelin stage has been reached, although a relatively minor
NRG1 signaling because ErbB3 receptor expression is reduced delay in sorting is also seen (Gokey et al. 2012; Iruarrizaga-
in these mice (Britsch et al. 2001). Lejarreta et al. 2012; Pereira et al. 2010; Yun et al. 2010).
Radial sorting is profoundly affected by signaling to
Schwann cells by components of the extracellular matrix,
particularly laminin. Genetic inactivation of laminins, or 11 P O S I T I VE A N D N E G AT I VE
β1-integrin or dystroglycan laminin receptors, results in R E GU L ATO R S C O N T R O L M Y E L I N AT I O N
impaired sorting (Chernousov et al. 2008; Yu et al. 2005).
There is good evidence that laminin, signaling via β1 integrin, Schwann cells in a 1:1 relationship with larger diameter axons,
activates the small Rho GTPase Rac1, and that this controls myelinate if they are induced to do so by cell-extrinsic sig-
the formation of Schwann cell lamellipodia associated with nals (discussed in chapter 44). These extrinsic signals activate
early steps of the sorting process (Chernousov et al. 2008; intracellular pathways and transcription factors that drive the
Feltri et al. 2008; Grove et al. 2007; Nodari et al. 2007; Yu myelination program (discussed in chapter 43). The existence
et al. 2005). The Rho GTPase cdc42, activated by NRG1, also of these cell-intrinsic positive regulators of myelination is well
participates early in sorting (Benninger et al. 2007). Signaling established. This applies in particular to the transcription
through dystroglycan, however, regulates later stages of the factors Krox-20, Sox10, Oct6, NAB1/2, NFkB, and NFAT
process (Berti et al. 2011). In culture NT3 stimulates Schwann (Svaren and Meijer 2008).
cell migration by activating Rac1 and may also contribute to Recently it has become clear that Schwann cell myelina-
sorting (Yamauchi et al. 2005). tion is also subject to negative regulation ( Jessen and Mirsky
Two β1 integrin-associated kinases, FAK and Ilk, also con- 2008). For instance, in certain experimental settings, activa-
trol radial sorting (Grove et al. 2007; Pereira et al. 2009). The tion of any of the major MAPK pathways ERK, JNK, and p38
major reason for the sorting defects seen in Ilk mutants is fail- can suppress myelin gene expression. The activation of certain
ure of Schwann cell process extension. transcription factors or transcriptional regulators also inhibits

166 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
or reverses myelination. At present, the evidence is strongest namely the reprogramming of the dedifferentiated state to
for c-Jun and Notch, but related observations have been made generate a dedicated repair supportive cell. This includes: (1)
for a number of other factors, including Sox2, Id2, and Pax3 upregulation of surface molecules and trophic factors that
( Jessen and Mirsky 2008; Le et al. 2005). prevent excessive death of injured neurons and promote axon
What is the function of these novel negative myelin regu- growth, such as N-cadherin, GDNF, BDNF, artemin, and
lators? Clearly, it is possible that they help prevent premature sonic hedgehog; (2) formation of cellular regeneration tracks
myelination during development or regeneration, or counter- (Bands of Büngner) that guide axons from the injury site to
act positive regulators to ensure that myelination proceeds the reinnervation target; this involves adoption of a distinc-
at an appropriate rate. A timing role of this kind has been tive cell morphology that remains to be fully determined, but
shown in principle for Notch signaling in vivo (Woodhoo appears to include the formation of several parallel processes
et al. 2009). projecting to each side of the perikaryon; and (3) activation of
But the most important function for some of the negative a poorly characterized process of endogenous myelin break-
regulators may not be developmental, but be carried out in down, that enables Schwann cells to breakdown their own
the adult, following injury. Nerve cut or crush triggers perhaps myelin, and activation of factors, including MCP1 and LIF,
the most remarkable phenotypic transition in the Schwann that recruit blood-borne macrophages to complete the clear-
cell lineage. This is the transformation of myelin and Remak ance of myelin, believed to inhibit axon growth.
cells distal to the injury into an alternative Schwann cell phe- In this way, the Schwann cell injury response transforms
notype, the denervated Schwann cell, a cell that is specialized the myelin cells to cells that are specialized for supporting
to support regeneration. Recent studies indicate that some of repair. This represents an unambiguous change in cell func-
the signals referred to as negative regulators of myelination, in tion, brought about by the combination of dedifferentiation
particular c-Jun and Notch, are essential for one or both of the and the activation of an alternative differentiation program,
two processes that are the hallmark of this transition, namely, the Schwann cell repair program. Transformations that share
the dismantling of the myelin phenotype (dedifferentiation) this set of features have been described in other systems, where
and the activation of the alternative repair phenotype. they are often referred to as transdifferentiation ( Jopling et al.
2011).
Although the original definition of the term transdiffer-
12 N E G AT I VE R E GU L ATO R S O F entiation was narrower than that implied in much current
M Y E L I N AT I O N C O N T R O L T H E S C H WA N N usage, the broader understanding of the term is useful, because
C E L L I N J U RY R E S P O N S E A N D T H E it defines a set of biological phenomena of great interest and
G E N E R AT I O N O F A D E D I C AT E D R E PA I R importance for understanding how some tissues respond to
C E L L I N I N J U R E D N E RVE S injury and disease, namely, the reorganization of differenti-
ation programs to allow a cell to abandon one function and
adopt another. The Schwann cell injury response represents a
12.1 T H E S C H WA N N C E L L I N JU RY R E S P O NS E
clear example of an event of this kind.
Nerve cut or crush results in axon death and triggers a cas- This response can be clearly differentiated from injury-
cade of changes distal to the injury that are collectively called induced dedifferentiation/proliferation, a response that is
Wallerian degeneration (discussed in chapters 54 and 55). This characteristic of situations in which the primary need is to
process has mainly been studied in the context of myelin cells, replace lost tissue. This is not the case in nerves, because cut
and this is reflected in the discussion that follows, but it is or crush involves axon death, but very little Schwann cell
likely that the same principles apply to Remak cells. loss except at the actual injury site. Therefore, the adaptive
Central to these events is a radical alteration in the pheno- value of the Schwann cell injury response is not primarily
type of Schwann cells. This transformation, the Schwann cell to replace lost cells, but to generate a novel Schwann cell
injury response, has two components (see Fig. 14.4). One is a variant specialized for supporting neuronal survival, axon
change in gene, protein, and lipid expression by myelinating regrowth, and pathfinding. Because these cells form the clas-
cells that is the reverse of that seen when immature Schwann sical Bands of Büngner it is appropriate to refer to them as
cells myelinate. This includes downregulation of components Büngner cells.
associated with myelin cells, such as Krox-20, P0, and MBP, and
reappearance of the classical markers of immature Schwann
12.2 C-JU N, N OTC H, A N D
cells, including p75NTR, NCAM, L1, and GFAP. Schwann
M ITO G E N-AC T I VAT E D P ROT E I N K I NA S E
cells of the injured distal stump also re-enter the cell cycle and
AC T I VAT I O N D R I VE T H E S C H WA N N
apoptotic cells are again seen in the Schwann cell population.
C E L L I N J U RY R E S P O NS E
Because these changes broadly represent a reversal to a previ-
ous state that resembles immature Schwann cells before myeli- Although a number of transcriptional regulators are likely to
nation, they are appropriately referred to as dedifferentiation. control Schwann cell transdifferentiation, the evidence is most
The other component of the Schwann cell injury response, extensive for c-Jun and Notch. There is also strong evidence
in contrast, can be represented as alternative differentiation, for the involvement of MAPK pathways.

T H E S C H WA N N C E L L L I N E AG E : C E L LU L A R T R A N S I T I O N S D U R I N G D E VE L O PM E N T A N D A F T E R I N J U RY • 167
12.2.1 c-Jun 13 S U M M A RY A N D P E R S P E C T I VE S
Although the transcription factor c-Jun is expressed at very The development of Schwann cell precursors from the neural
low levels in unperturbed adult nerves, injury results in a rapid crest, the subsequent transition to immature Schwann cells and
and strong expression of this protein in the Schwann cells dis- finally to myelin Schwann cells or Remak cells are described in
tal to the injury site. If this is prevented, by genetic inactiva- terms of the major molecular and cellular changes that occur.
tion of c-Jun in Schwann cells, nerve regeneration is severely In addition, the radical response of Schwann cells to nerve
impeded, although c-Jun remains normally expressed in the injury, resulting in the generation of Büngner cells that con-
neurons and other cells in the nerve. trol nerve repair is discussed and compared with the changes
This is owing to the key role of c-Jun in controlling both in differentiation states that take place during development.
components of the Schwann cell injury response. c-Jun drives In the near future there is likely to be rapid increase in stud-
dedifferentiation, which therefore proceeds more slowly in ies on the epigenetic control of the Schwann cell lineage dur-
c-Jun–/– cells (Parkinson et al. 2008). But more importantly, ing development and after injury. There is also a need for better
c-Jun is required for the normal activation of the repair pro- integration between studies on cell extrinsic signals, including
gram. The denervated c-Jun–/– Schwann cells fail to upregulate trophic factors and matrix molecules, studies on intracellu-
key genes for trophic factors that promote neuronal survival lar signaling cascades, such as MAPkinase and cAMP path-
and regeneration, form abnormal regeneration tracks, and show ways, and studies on transcription factors, during Schwann
long-term defects in myelin clearance. The generation of func- cell development. Some fundamental instances of neuroglial
tional Büngner repair cells therefore fails in the absence of c-Jun, signaling in peripheral nerves also remain obscure in molecu-
resulting in a dramatic failure of regeneration and functional lar terms. This includes the glial-derived survival signaling to
recovery (Jessen and Mirsky 2010; Arthur-Farraj et al. 2012). developing neurons in embryonic nerves, the neuronal signals
that, following injury, inform Schwann cells that axons are
12.2.2 Notch dying, and signals that cooperate with neuregulin to control
myelination. In terms of technical advances, the development
Like c-Jun, Notch signaling is activated in Schwann cells of cut of three-dimensional electron microscopy using focussed ion
or crushed nerves. Notch is likely to have more than one func- beam or field emission techniques is likely to open up radical
tion, but the best characterized of these is the promotion of the new ways of understanding the complex interactions between
dedifferentiation component of the injury response, and myelin axons and Schwann cells during embryonic development,
clearance. Even in uninjured adult nerves, activation of Notch radial sorting, myelination, and nerve regeneration.
in myelin Schwann cells is sufficient to trigger demyelination
(Woodhoo et al. 2009). In line with this, myelin breakdown is
accelerated in genetically modified mice in which Notch signal- AC K N OW L E D G M E N T S
ing is overactivated after injury. Conversely, in mice with genetic
inactivation of Notch signaling in Schwann cells (genetic inac- The authors would like to thank the present and past mem-
tivation of Notch1 or the major downstream transcription fac- bers of their laboratory for allowing them to cite work in prog-
tor RBPJ) injury-induced demyelination is delayed (Woodhoo ress, and funding from the Wellcome Trust (program grant)
et al. 2009). Interactions between Notch and c-Jun are likely to and the EU Seventh Framework Program (FP7/2007–2013)
have a major bearing on how these factors control Schwann cell under grant agreement No. HEALTH-F2–2008–201535.
dedifferentiation (DK Wilton, RM, and KRJ, unpublished).
REFERENCES
12.2.3 The MAPK Pathways: ERK, JNK, and p38
Adameyko I, Lallemend F, Aquino JB, Pereira JA, Topilko P, Müller T,
ERK, c-Jun N-terminal kinase ( JNK), and p38 are all strongly et al. 2009. Schwann cell precursors from nerve innervation are a cel-
activated in the distal stump of injured nerves. Although ERK lular origin of melanocytes in skin. Cell 139:366–379.
signaling is required for myelination (Newbern et al. 2011), Arthur-Farraj PJ, Latouche M, Wilton DK, Quintes S, Chabrol E,
Banerjee A, et al. 2012. c-Jun reprograms Schwann cells of injured
strong activation of Ras/Raf/ERK suppresses myelin genes nerves to generate a repair cell essential for regeneration. Neuron.
and induces dedifferentiation and myelin breakdown even 75(4):633–647.
in myelinating Schwann cells in contact with axons in vitro Atanasoski S, Shumas S, Dickson C, Scherer SS, Suter U. 2001. Differential
(Harrisingh et al. 2004; Napoli et al 2012). ERK is also involved cyclin D1 requirements of proliferating Schwann cells during devel-
in another component of the Schwann cell injury response, the opment and after injury. Mol Cell Neurosci 18:581–592.
Benninger Y, Thurnherr T, Pereira JA, Krause S, Wu X, Chrostek-Grashoff
upregulation of the macrophage attractant MCP1 (Fischer A, et al. 2007. Essential and distinct roles for cdc42 and rac1 in the
et al. 2008). Activation of JNK similarly inhibits myelin gene regulation of Schwann cell biology during peripheral nervous system
expression, and reverses established myelin differentiation in development. J Cell Biol 177:1051–1061.
purified Schwann cells and in neuron-Schwann cell cocultures Berthold CH, Fraher JP, King R.HM, Rydmark, M. 2005. Microscopic
(Parkinson et al. 2008). Last, p38 activation, although involved anatomy of the peripheral nervous system. In: Dyck PJ, Thomas PK
(eds.), Peripheral neuropathy, 4th ed. Amsterdam: Elsevier, pp. 35–91.
in establishing normal axon–Schwann cell relationship early Berti C, Bartesaghi L, Ghidinelli M, Zambroni D, Figlia G, Chen ZL,
in myelination, can suppress myelin genes and drive myelin et al. 2011. Non-redundant function of dystroglycan and β1 integrins
breakdown ( Jessen and Mirsky 2008; Yang et al. 2012). in radial sorting of axons. Development 138:4025–4037.

168 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Birchmeier C. 2009. ErbB receptors and the development of the nervous Gokey NG, Srinivasan R, Lopez-Anido C, Krueger C, Svaren J. 2012.
system. Exp Cell Res 315:611–618. Developmental regulation of microRNA expression in Schwann
Birchmeier C, Nave KA. 2008. Neuregulin-1, a key axonal signal that cells. Mol Cell Biol 32:558–568.
drives Schwann cell growth and differentiation. Glia 56:1491–1497. Grim M, Halata Z, Franz T. 1992. Schwann cells are not required for
Bitgood MJ, McMahon AP. 1995. Hedgehog and Bmp genes are coex- guidance of motor nerves in the hindlimb in Splotch mutant mouse
pressed at many diverse sites of cell-cell interaction in the mouse embryos. Anat Embryol (Berl) 186:311–318.
embryo. Dev Biol 172:126–138. Grinspan JB, Marchionni MA, Reeves M, Coulaloglou M, Scherer
Brennan A, Dean CH, Zhang AL, Cass DT, Mirsky R, Jessen KR. 2000. SS. 1996. Axonal interactions regulate Schwann cell apoptosis in
Endothelins control the timing of Schwann cell generation in vitro developing peripheral nerve: neuregulin receptors and the role of
and in vivo. Dev Biol 227:545–557. neuregulins. J Neurosci 16:6107–6118.
Britsch S, Goerich DE, Riethmacher D, Peirano RI, Rossner M, Nave Grove M, Komiyama NH, Nave KA, Grant SG, Sherman DL, Brophy
KA, et al. 2001. The transcription factor Sox10 is a key regulator of PJ. 2007. FAK is required for axonal sorting by Schwann cells. J Cell
peripheral glial development. Genes Dev 15:66–78. Biol 176:277–782.
Britsch S, Li L, Kirchhoff S, Theuring F, Brinkmann V, Birchmeier Harrisingh MC, Perez-Nadales E, Parkinson DB, Malcolm DS, Mudge
C, et al. 1998. The ErbB2 and ErbB3 receptors and their ligand, AW, Lloyd AC. 2004. The Ras/Raf/ERK signaling pathway drives
neuregulin-1, are essential for development of the sympathetic ner- Schwann cell dedifferentiation. EMBO J 23:3061–3071.
vous system. Genes Dev 12:1825–1836. Hu X, Hicks CW, He W, Wong P, Macklin WB, Trapp BD, et al. 2006.
Buchstaller J, Sommer L, Bodmer M, Hoffmann R, Suter U, Mantei Bace1 modulates myelination in the central and peripheral nervous
N. 2004. Efficient isolation and gene expression profi ling of small system. Nat Neurosci 9:1520–1525.
numbers of neural crest stem cells and developing Schwann cells. Iruarrizaga-Lejarreta M, Varela-Rey M, Lozano, Fernández-Ramos D,
J Neurosci 24:2357–2365. Rodríguez-Ezpeleta N, Embade1 N, et al. 2012. The RNA-binding
Cheli Y, Ohanna M, Ballotti R, Bertolotto C. 2010. Fifteen-year quest protein HuR controls global changes in gene expression during
for microphthalmia-associated transcription factor target genes. Schwann cell development. J Neurosci 32(14):4944–58.
Pigment Cell Melanoma Res 23:27–40. Jaegle M, Ghazvini M, Mandemakers W, Piirsoo M, Driegen S,
Chernousov MA, Yu WM, Chen ZL, Carey DJ, Strickland S. 2008. Levavasseur F, et al. 2003. The POU proteins Brn-2 and Oct-6 share
Regulation of Schwann cell function by the extracellular matrix. Glia important functions in Schwann cell development. Genes Dev 17:
56:1498–1507. 1380–1391.
Coulpier F, Le Crom S, Maro GS, Manent J, Giovannini M, Maciorowski Jessen KR, Brennan A, Morgan L, Mirsky R, Kent A, Hashimoto Y,
Z, et al. 2009. Novel features of boundary cap cells revealed et al. 1994. The Schwann cell precursor and its fate: a study of cell
by the analysis of newly identified molecular markers. Glia 57: death and differentiation during gliogenesis in rat embryonic nerves.
1450–1457. Neuron 12:509–527.
D’Antonio M, Droggiti A, Feltri ML, Roes J, Wrabetz L, Mirsky R, Jessen KR, Mirsky R. 2004. Schwann cell development. In: Lazzarini
et al. 2006a. TGFbeta type II receptor signaling controls Schwann RA (ed.), Myelin biology and disorders. Amsterdam: Elsevier,
cell death and proliferation in developing nerves. J Neurosci pp. 329–370
26:8417–8427. Jessen KR, Mirsky R. 2005. The origin and development of glial cells in
D’Antonio M, Michalovich D, Paterson M, Droggiti A, Woodhoo A, Mirsky peripheral nerves. Nat Rev Neurosci 6:671–682.
R, et al. 2006b. Gene profiling and bioinformatics analysis of Schwann Jessen KR, Mirsky R. 2010 Control of Schwann cell myelination. F1000
cell embryonic development and myelination. Glia 53:501–515. Biol Rep 2:19.
Diner O. 1965. Schwann cells during mitosis and their relation to axons Jessen KR, Mirsky R. 2008. Negative regulation of myelination:
during the development of the sciatic nerve in the rat. CR Acd Sci relevance for development, injury, and demyelinating disease. Glia
Hebd Seances Acad Sci D 261:1731–1734. 56:1552–1565.
Dong Z, Brennan A, Liu N, Yarden Y, Lefkowitz G, Mirsky R, et al. 1995. Jopling C, Boue S, Belmonte JC. 2011. Dedifferentiation, transdiffer-
NDF is a neuron-glia signal and regulates survival, proliferation and entiation and reprogramming: three routes to regeneration. Nat Rev
maturation of rat Schwann cell precursors. Neuron 15:585–596. Mol Cell Biol 12:79–89.
Dong Z, Sinanan A, Parkinson D, Parmantier E, Mirsky R, Jessen Joseph NM, Mukouyama YS, Mosher JT, Jaegle M, Crone SA,
KR. 1999. Schwann cell development in embryonic mouse nerves. Dormand EL, et al. 2004. Neural crest stem cells undergo multi-
J Neurosci Res 56:334–348. lineage differentiation in developing peripheral nerves to generate
Einheber S, Hannocks MJ, Metz CN, Rifk in DB, Salzer JL. 1995. endoneurial fibroblasts in addition to Schwann cells. Development
Transforming growth factor-beta 1 regulates axon/Schwann cell 131:5599–5612.
interactions. J Cell Biol 129:443–458. Kim HA, Pomeroy SL, Whoriskey W, Pawlitzky I, Benowitz LI, Sicinski
Feltri ML, Suter U, Relvas JB. 2008. The function of RhoGTPases in P, et al. 2000. A developmentally regulated switch directs regenerative
axon ensheathment and myelination. Glia 56:1508–1517. growth of Schwann cells through cyclin D1. Neuron 26:405–416.
Finzsch M, Schreiner S, Kichko T, Reeh P, Tamm ER, B ösl MR, Komiyama A, Suzuki K. 1992. Age-related differences in proliferative
et al. 2010. Sox10 is required for Schwann cell identity and pro- responses of Schwann cells during Wallerian degeneration. Brain Res
gression beyond the immature Schwann cell stage. J Cell Biol 573:267–275.
189:701–712. Kubu CJ, Orimoto K, Morrison SJ, Weinmaster G, Anderson DJ,
Fischer S, Weishaupt A, Troppmair J, Martini R. 2008. Increase of Verdi JM. 2002. Developmental changes in Notch1 and numb
MCP-1 (CCL2) in myelin mutant Schwann cells is mediated by expression mediated by local cell-cell interactions underlie progres-
MEK-ERK signaling pathway. Glia 56:836–843. sively increasing delta sensitivity in neural crest stem cells. Dev Biol
Frank M, Schaeren-Wiemers N, Schneider R, Schwab ME. 1999. 244:199–214.
Developmental expression pattern of the myelin proteolipid MAL La Marca R, Cerri F, Horiuchi K, Bachi A, Feltri ML, Wrabetz L,
indicates different functions of MAL for immature Schwann cells et al. 2011. TACE (ADAM17) inhibits Schwann cell myelination.
and in a late step of CNS myelinogenesis. J Neurochem 73:587–597. Nat Neurosci 14:857–865.
Fricker FR, Lago N, Balarajah S, Tsantoulas C, Tanna S, Zhu N, et al. 2011. Le N, Nagarajan R, Wang JYT, Araki T, Schmidt RE, Milbrandt J.
Axonally derived neuregulin-1 is required for remyelination and regen- 2005. Analysis of congenital hypomyelinating Egr2Lo/Lo nerves
eration after nerve injury in adulthood. J Neurosci 31:3225–3233. identifies Sox2 as an inhibitor of Schwann cell differentiation and
Garratt AN, Voiculescu O, Topilko P, Charnay P, Birchmeier C. 2000. myelination. Proc Natl Acad Sci U S A 102: 2596–2601.
A dual role of erbB2 in myelination and in expansion of the Schwann Li J, Habbes H, Eiberger J, Willecke K, Dermietzel R, Meier C. 2007.
cell precursor pool. J Cell Biol 148:1035–1046. Analysis of connexin expression during mouse Schwann cell

T H E S C H WA N N C E L L L I N E AG E : C E L LU L A R T R A N S I T I O N S D U R I N G D E VE L O PM E N T A N D A F T E R I N J U RY • 169
development identifies connexin29 as a novel marker for the transi- Rizvi TA, Huang Y, Sidani A, Atit R, Largaespada DA, Boissy RE, et al.
tion of neural crest cells to precursor cells. Glia 55:93–103. 2002. A novel cytokine pathway suppresses glial cell melanogenesis
Lin W, Sanchez HB, Deerinck T, Morris JK, Ellisman M, Lee KF. 2000. after injury to adult nerve. J Neurosci 22:9831–9840.
Aberrant development of motor axons and neuromuscular synapses Salzer JL, Williams AK, Glaser L, Bunge RP. 1980. Studies of Schwann
in erbB2-deficient mice. Proc Natl Acad Sci U S A 97:1299–1304. cell proliferation. II. Characterization of the stimulation and speci-
Lyons DA, Pogoda HM, Voas MG, Woods IG, Diamond B, Nix R, et al. ficity of the response to a neurite membrane fraction. J Cell Biol
2005. Erbb3 and erbb2 are essential for Schwann cell migration and 84:753–766.
myelination in zebrafish. Curr Biol 15:513–524. Shah NM, Marchionni MA., Isaacs I, Stroobant P, Anderson DJ. 1994.
Marmigère F, Ernfors P. 2007. Specification and connectivity of neuronal Glial growth factor restricts mammalian neural crest stem cells to a
subtypes in the sensory lineage. Nat Rev Neurosci 8:114–127. glial fate. Cell 77:349–360.
Maro GS, Vermeren M, Violescu O, Melton L, Cohen J, Charnay P, et al. Sharghi-Namini S, Turmaine M, Meier C, Sahni V, Umehara F, Jessen
2004. Neural crest boundary cap cells constitute a source of neuronal KR, et al. 2006. The structural and functional integrity of periph-
and glial cells of the PNS. Nat Neurosci 7:930–938. eral nerves depends on the glial-derived signal desert hedgehog.
McGarvey ML, Baron-Van Evercooren A, Kleinman HK, Dubois-Dalcq J Neurosci 26:6364–6376.
M. 1984. Synthesis and effects of basement membrane components in Stewart HJ, Brennan A, Rahman M, Zoidl G, Mitchell PJ, Jessen KR,
cultured rat Schwann cells. Dev Biol 105:18–28. et al. 2001. Developmental regulation and overexpression of the tran-
Meier C, Parmantier E, Brennan A, Mirsky R, Jessen KR. 1999. scription factor AP-2, a potential regulator of the timing of Schwann
Developing Schwann cells acquire the ability to survive without cell generation. Eur J Neurosci 14:363–372.
axons by establishing an autocrine circuit involving IGF, NT-3 and Stewart HJ, Morgan L, Jessen KR, Mirsky R. 1993. Changes in DNA
PDGF-BB. J Neurosci 19:3847–3859. synthesis rate in the Schwann cell lineage in vivo are correlated with
Meintanis S, Thomaidou D, Jessen KR, Mirsky R, Matsas R. 2001. The the precursor-Schwann cell transition and myelination. Eur J Neurosci
neuron-glia signal beta-neuregulin promotes Schwann cell motility 5:1136–1144.
via the MAPK pathway. Glia 34:39–51. Svaren J, Meijer D. 2008. The molecular machinery of myelin gene tran-
Mirsky R, Woodhoo A, Parkinson DB, Arthur-Farraj P, Bhaskaran A, scription in Schwann cells. Glia 56:1541–1551.
Jessen KR. 2008. Novel signals controlling embryonic Schwann cell Syroid DE, Maycox PJ, Soilu-Hänninen M, Petratos S, Bucci T, Burrola
development, myelination and dedifferentiation. J Peripher Nerv Syst P, et al. 2000. Induction of postnatal Schwann cell death by the
13:122–135. low-affinity neurotrophin receptor in vitro and after axotomy.
Morris JK, Lin W, Hauser C, Marchuk Y, Getman D, Lee KF. 1999. J Neurosci 20:5741–5747.
Rescue of the cardiac defect in ErbB2 mutant mice reveals essential Takahashi M, Osumi N. 2005. Identification of a novel type II classical cad-
roles of ErbB2 in peripheral nervous system development. Neuron herin: rat cadherin19 is expressed in the cranial ganglia and Schwann
23:273–283. cell precursors during development. Dev Dyn 232: 200–208.
Napoli I, Noon LA, Ribeiro S, Kerai AP, Parrinello S, Rosenberg LH, Taveggia C, Zanazzi G, Petrylak A, Yano H, Rosenbluth J, Einheber S,
et al. 2012. A central role for the ERK-signaling pathway in control- et al. 2005. Neuregulin-1 type III determines the ensheathment fate
ling Schwann cell plasticity and peripheral nerve regeneration in vivo. of axons. Neuron 47:681–694.
Neuron 73:729–742. Taylor MK, Yeager K, Morrison SJ. 2007. Physiological Notch signaling
Nave KA, Salzer JL. 2006. Axonal regulation of myelination by neuregu- promotes gliogenesis in the developing peripheral and central ner-
lin 1. Curr Opin Neurobiol 16:492–500. vous systems. Development 134:2435–2447.
Newbern JM, Li X, Shoemaker SE, Zhou J, Zhong J, Wu Y, et al. 2011. Topilko P, Schneider-Maunoury S, Levi G, Baron-Van Evercooren A,
Specific functions for ERK/MAPK signaling during PNS develop- Chennoufi AB, Seitanidou T, et al. 1994. Krox-20 controls myelina-
ment. Neuron 69:91–105. tion in the peripheral nervous system. Nature 371:796–799.
Nodari A, Zambroni D, Quattrini A, Court FA, D’Urso A, Recchia A, Wakamatsu Y, Maynard TM, Weston JA. 2000. Fate determination
et al. 2007. β1 integrin activates Rac1 in Schwann cells to generate of neural crest cells by NOTCH-mediated lateral inhibition and
radial lamellae during axonal sorting and myelination. J Cell Biol asymmetrical cell division during gangliogenesis. Development
177:1063–1075. 127:2811–2821.
Özkaynak E, Abello A, Jaegle M, van Berge L, Hamer D, Kegel L, Wanner IB, Guerra NK, Mahoney J, Kumar A, Wood PM, Mirsky R,
et al. 2010. Adam22 is a major neuronal receptor for Lgi4-mediated et al. 2006b. Role of N-cadherin in Schwann cell precursors of grow-
Schwann cell signalling. J Neurosci 30:3857–3864. ing nerves. Glia 54:439–459.
Parkinson DB, Bhaskaran A, Arthur-Farraj P, Noon LA, Woodhoo A, Wanner IB, Mahoney J, Jessen KR, Wood PM, Bates M, Bunge MB.
Lloyd AC, et al. 2008. c-Jun is a negative regulator of myelination. 2006a. Invariant mantling of growth cones by Schwann cell precur-
J Cell Biol 181:625–637. sors characterize growing peripheral nerve fronts. Glia 54:424–438.
Parkinson DB, Dong Z, Bunting H, Whitfield J, Meier C, Marie H, et al. Webster H de F, Favilla JT. 1984. Development of peripheral nerve fibers.
2001. Transforming growth factor beta (TGFbeta) mediates Schwann In: Dyck PJ, Thomas PK, Lambert EH, Bunge RP (eds.), Peripheral
cell death in vitro and in vivo: examination of c-Jun activation, interac- neuropathy, 2nd ed. Philadelphia: Saunders, pp. 329–359.
tions with survival signals, and the relationship of TGFbeta-mediated Willem M, Garratt AN, Novak B, Citron M, Kaufmann S, Rittger A,
death to Schwann cell differentiation. J Neurosci 21:8572–8585. et al. 2006. Control of peripheral nerve myelination by the
Parmantier E, Lynn B, Lawson D, Turmaine M, Namini SS, Chakrabarti beta-secretase BACE1. Science 314:664–666.
L, et al. 1999. Schwann cell-derived Desert hedgehog controls the Winseck AK, Oppenheim RW. 2006. An in vivo analysis of Schwann cell
development of peripheral nerve sheaths. Neuron 23:713–724. programmed cell death in embryonic mice: the role of axons, glial growth
Pereira JA, Baumann R, Norrmén C, Somandin C, Miehe M, Jacob C, factor, and the pro-apoptotic gene Bax. Eur J Neurosci 24:2105–2117.
et al. 2010. Dicer in Schwann cells is required for myelination and Woldeyesus MT, Britsch S, Riethmacher D, Xu L, Sonnenberg-
axonal integrity. J Neurosci 30:6763–6775. Riethmacher E, Abou-Rebyeh F, et al. 1999. Peripheral nervous
Pereira JA, Benninger Y, Baumann R, Gonçalves AF, Ozçelik M, system defects in erbB2 mutants following genetic rescue of heart
Thurnherr T, et al. 2009. Integrin-linked kinase is required for radial development. Genes Dev 13: 2538–2548.
sorting of axons and Schwann cell remyelination in the peripheral Wolpowitz D, Mason TB, Dietrich P, Mendelsohn M, Talmage DA,
nervous system. J Cell Biol 185:47–161. Role LW. 2000. Cysteine-rich domain isoforms of the neuregulin-1
Raphael AR, Talbot WS. 2011 New insights into signaling during myeli- gene are required for maintenance of peripheral synapses. Neuron 25:
nation in zebrafish. Curr Top Dev Biol 97:1–19. 79–91.

170 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Woodhoo A, Alonso MB, Droggiti A, Turmaine M., D’Antonio M, neurotrophin-3-induced Schwann cell migration. Proc Natl Acad
Parkinson DB, et al. 2009. Notch controls embryonic Schwann Sci U S A 102:14889–14894.
cell differentiation, postnatal myelination and adult plasticity. Nat Yang D, Bierman J, Tarumi YS, Zhong YP, Rangwala R, Proctor TM,
Neurosci 12:839–847. et al. 2005. Coordinate control of axon defasciculation and myeli-
Woodhoo A, Dean CH, Droggiti A, Mirsky R, Jessen KR. 2004. The nation by laminin-2 and -8. J Cell Biol 168:655–666.
trunk neural crest and its early glial derivatives: a study of survival Yang DP, Syed N, Kim J, Bhaskaran A, Mindos T, Mirsky R, et al.
responses, developmental schedules and autocrine mechanisms. Mol 2012. p38 MAPK activation promotes the denervated Schwann
Cell Neurosci 25:30–41. cell phenotype and functions as a negative regulator of Schwann
Woodhoo A, Sommer L. 2008. Development of the Schwann cell lineage: cell differentiation and myelination. J Neurosci 32:7158–7168.
from the neural crest to the myelinated nerve. Glia 56: 1481–1490. Yu WM, Feltri M.L, Wrabetz L, Strickland S, Chen ZL. 2005.
Yamauchi J, Miyamoto Y, Chan JR, Tanoue A. 2008. ErbB2 directly Schwann cell-specific ablation of laminin gamma1 causes apoptosis
activates the exchange factor Dock7 to promote Schwann cell and prevents proliferation. J Neurosci 25:4463–4472.
migration. J Cell Biol 181:351–365. Yun B, Anderegg A, Menichella D, Wrabetz L, Feltri ML, Awatramani
Yamauchi J, Miyamoto Y, Tanoue A, Shooter EM, Chan JR. 2005. R. 2010. MicroRNA-deficient Schwann cells display congenital
Ras activation of a Rac1 exchange factor, Tiam1, mediates hypomyelination. J Neurosci 30:7722–7728.

T H E S C H WA N N C E L L L I N E AG E : C E L LU L A R T R A N S I T I O N S D U R I N G D E VE L O PM E N T A N D A F T E R I N J U RY • 171
15.
MICROGLIA LINEAGE AND DEVELOPMENT
Marco Prinz

A B B R E VI AT I O N S Priller et al. 2001; Prinz and Mildner 2011; Simard et al. 2006).
However, conflicting data have been published in recent years
AGM aorta-gonad-mesonephros that contribute to the complexity of microglia research. This
BBB blood-brain barrier chapter summarizes and discusses the latest findings on the ori-
BM bone marrow gin and developmental fate of myeloid cells in the CNS during
BM-DP Bone Marrow–Derived Phagocyte health and disease (see also chapters 8, 19, 47, and 48).
CCR2 chemokine receptor 2
CNS central nervous system
Csf-1 colony stimulating factor 2 M I C R O G L I A A S PA RT O F T H E
Csf-1R colony stimulating factor 1 receptor M O N O N U C L E A R P H AG O C Y T E FA M I LY
dpc days post conception
GFP green fluorescent protein In 1908, the Russian Elie Metchnikoff (1845–1916),
HSC hematopoietic stem cells together with the German Paul Ehrlich (1854–1915), was
MPS mononuclear phagocyte system awarded the Nobel Prize for Medicine and Physiology for
SOD superoxide dismutase his ground-breaking work on phagocytosis (Kaufmann
2008). Metchnikoff coined the term phagocytes (named
after the Greek phago, meaning devour, and cytes, meaning
1 INTRODUCTION cells) and divided them into microphages (smaller cells with
polymorphonuclear-shaped nuclei now known as granulocytes)
Several reports propose that microglia are specialized cells and macrophages (large eaters). He suggested that both types of
of the mononuclear phagocyte lineage. Indeed, it has been phagocytes played an important role in host resistance against
shown that they share many features with other myeloid cells germs such as bacteria and fungi. The scientist Metchnikoff also
in the body. For example, microglia express Fc and comple- recognized the close relationship between mononuclear phago-
ment receptors CD11b and F4/80 epitopes typically found cytic cells in the bone marrow, spleen, lymph nodes, and the
on myelomonocytes (Hanisch 2002; Perry et al. 1985). connective tissue, excluding the brain, leading him to introduce
Therefore, it appears that brain microglia, like other resident the term macrophage system (Gordon and Taylor 2005). Later
macrophages, derive from myeloid precursors that originate on, the German pathologist Karl Albert Ludwig Aschoff from
during embryonic development. Aside from these similari- Freiburg im Breisgau developed this concept further and grouped
ties, microglia also display specific molecular differences when several cell types into what he called the reticulo-endothelial
compared with their myeloid cousins. For instance, it has been system in 1924 and subsequently the reticulo-histiocyte system
shown that microglia produce significantly lower levels of (Aschoff 1924). This system included reticular cells (or fixed
superoxide dismutase (SOD) compared with splenic or bone macrophages) of the spleen and lymph nodes as well as endothe-
marrow (BM) macrophages (Enose et al. 2005; Glanzer et al. lial cells, monocytes, and histiocytes as mobile macrophages. All
2007). Furthermore, during lipopolysaccharide/interferon-γ– these cells were grouped on the basis of their ability to be labeled
induced central nervous system (CNS) inflammation micro- by a vital dye in vivo, an assay assumed to measure phagocytotic
glia behave differently than CNS-infiltrating phagocytes, activity. However, some cells with poor phagocytotic capacity,
for example, as is reflected by the expression levels of C1qA, such as endothelial cells, also stained positively because of their
Trem2, and CXCL14 (Schmid et al. 2009). ability to perform pinocytosis and were subsequently falsely
In addition to the microglia that invade the brain dur- categorized. Therefore, this method of labeling is an unreliable
ing early embryogenesis, it has been postulated that myeloid criterion for the identification of mononuclear phagocytes.
progenitors can penetrate into the brain even in normal adult Hence, a new classification was introduced in 1969 to group
mice to replace decaying microglial cells. Moreover, it has been only highly phagocytic cells and their precursors in one system
reported that during CNS diseases phagocytes with morpho- called the mononuclear phagocyte system (MPS) a term that is
logical features of endogenous microglia can be derived from still used (Geissmann et al. 2010; van Furth et al. 1972).
BM cells or circulating monocytes. These cells subsequently Nowadays this broad family comprises numerous mono-
become an integral part of the pathology and can be incorpo- nuclear cells in the body, namely, circulating monocytes in
rated into the local cellular networks (Mildner et al. 2007, 2011; the bloodstream, dendritic cells in the secondary lymphoid

172
organs, and tissue macrophages (Geissmann et al. 2010; Prinz 1991, 1999) forces the developmental neuroscientist to dive
et al. 2011). As such, macrophages are resident phagocytic deeply into the complex situation of hematopoiesis. Our cur-
cells in lymphoid (spleen, lymph nodes) and nonlymphoid rent understanding of the multifaceted process of hematopoi-
tissues such as the brain (microglia), liver (Kupffer cells), skin esis was extensively reviewed recently by Ana Cumano and
(Langerhans cells), lung (alveolar macrophages), bone (osteo- Isabelle Godin (2007).
clasts), and kidney (kidney macrophages). At these sites mac- Microglia appearance in the neuroepithelium at 9.5 days
rophages are generally believed to be involved in steady state post conception (dpc) suggests that microglia precursors may
tissue homeostasis, via the clearance of apoptotic cells and originate from the yolk sac rather than hematopoietic stem
the production of growth factors. Therefore, macrophages cells (HSC) in the fetal liver or bone marrow. As shown in
are equipped with a broad range of pathogen-recognition Figure 15.1, HSC are the founders of the hematopoietic sys-
receptors that make them efficient at phagocytosis and induce tem, responsible for blood production. Starting at 10.5 dpc
the production of inflammatory cytokines (Gordon 2002). they emerge from ventral aortic hematogenic endothelial
Although the term MPS has created a framework that helped cells in the aorta-gonad-mesonephros (AGM) region of the
our understanding of tissue phagocyte biology, it also led to embryo. At 10.5 dpc HSC are only found in the embryonic
the assumption that all tissue macrophages are identical in AGM region, and HSC engrafting activity only becomes
origin and function. However, there is still a lack of clear- detectable at 11.0 dpc (Bertrand et al. 2005; Cumano and
cut genetic studies, such as fate mapping studies, that could Godin 2007). Hematopoietic stem cell–derived myeloid cells
provide unequivocal evidence that microglia and tissue mac- are then produced abundantly in the fetal liver by 12.5 dpc.
rophages such as Kupffer cells in the liver are identical twins In contrast, microglia are present in the embryonic brain
but have grown up in distinct chambers of the body. before their production from HSC. A population of mater-
nally derived macrophages can be found in the yolk sac of the
embryo as early as 7.5 dpc (not shown). This population, how-
3 THE ORIGIN OF MICROGLIA ever, subsequently decreases in number, becomes almost unde-
F R O M T H E YO L K S AC tectable at 9.0 dpc, and is later absent in the embryo (Bertrand
et al. 2005). A second wave of extraembryonic hematopoietic
The crucial finding that the brain in the developing mouse cells of zygotic origin differentiates into anucleated red blood
embryo already contains microglia at 9.5 dpc (Alliot et al. cells and macrophages in the yolk sac (Bertrand et al. 2005).

CNS
8.5-9.0 dpc
haematopoiesis
primitive

hsc and macrophages


?
in yolk sac
7.5–8.0 dpc

hsc in
fetal liver
12.5 dpc
haematopoiesis
definitive

hsc in hsc in
AGM region bone marrow
10.5 dpc 15.5 dpc

Figure 15.1 Different Regions of Hematopoietic Stem Cell Location in the Mouse. A population of transient maternally derived macrophages enters
the yolk sac between 7.5 and 9.0 days post conception (dpc, not shown). The first hematopoietic cells form as blood islands in yolk sac tissues at 8.0
dpc and present the transient and early primitive hematopoiesis (see also Fig. 15.2). As early as 10.5 dpc the aorta-gonad-mesonephros (AGM) region
serves as the main the hematopoietic organ until 12.5 dpc, when the fetal liver contains most uncommitted hematopoietic stem cells (HSC). Around
15.5 dpc bone marrow starts its activity as the hematopoietic compartment, giving rise to long-lived and definitive hematopoiesis. The bone marrow
now becomes the principal organ that sustains an adult-type hematopoiesis. Central nervous system colonization by yolk sac progenitors begins at 9.5
dpc. It is still unclear if there is a contribution from cells of the fetal liver to the CNS. Figure by Markus Knust.

M I C R O G L I A L I N E AG E A N D D E VE L O PM E N T • 173
These yolk sac macrophages invade the developing embryo At embryonic stage 13.5 dpc, when the fetal liver is already
between 9.5 and 10.5 dpc (Figs. 15.2 and 15.3) (Bertrand et al. the primary hematopoietic organ and the main site of HSC
2005). Thus, because tissues such as the CNS contain yolk expansion and differentiation (Lichanska and Hume 2000),
sac–derived macrophages, but not HSC or maternal mac- microglial precursors can be detected in significant numbers
rophages, it is reasonable to hypothesize that microglia may within the ventricular lining of the fourth ventricle (Chan
originate from yolk sac macrophages rather than from HSC et al. 2007). However, a 20-fold increase of CD11b+ F4/80+
in the fetal liver or bone marrow. microglia cell numbers can be observed during the early post-
As stated, the first macrophage-like cells with an amoe- natal period (P0–P11) in rodents (Alliot et al. 1999). It is still
boid shape appear in the rodent neuroepithelium as early as unclear whether this increase is caused by the proliferation
day 8.5 to 9.0 of embryogenesis (Ashwell, 1990, 1991; Chan of embryonic microglial precursors, a phenomenon that has
et al. 2007) independently of a developed circulatory system frequently been observed in the developing brain (Cuadros
(Kurz and Christ 1998) (see Fig. 15.1). At this early time point, and Navascues 1998; Mander and Morris 1996), or whether
the first immature macrophages can already be found in the an additional recruitment of monocyte-derived microglial
yolk sac (Takahashi et al. 1996) and were already suggested precursors occurs. The latter hypothesis is supported by the
to be the precursors of microglial cells a long time ago (Alliot fact that the absence of microglia in mice deficient for the
et al. 1999), which then develop through a nonmonocyte transcription factor PU.1 can be rescued by the injection of
pathway (Lichanska and Hume 2000; Takahashi et al. 1996). wild-type bone marrow into newborns, leading to a complete

8.0 dpc 10.5 dpc


umbilical cord

yolk sac blood islands


yolk sac

placenta

embryo embryo

vitelline duct

Figure 15.2 Different Locations of Hematopoiesis During Rodent Embryonic Development. During early embryogenesis (8.0 dpc) first hematopoietic
cells form extraembryonically in a band of primitive erythroid cells, most often described as “blood islands.” Hematopoietic cells properly enter the
embryo via a primitive capillary plexus. At later stages of development hematopoiesis occurs within different tissues of the embryo, as depicted in
Figure 15.1. Figure by Markus Knust.

C
B
A

yolk sac 8.5 dpc 9.5 dpc P1


yolk sac precursors
A B C

meninges

neuroepithelium

Figure 15.3 Embryonic and Postnatal Development of Microglia in the Mouse. A. Early stem cells from yolk sac tissues migrate to embryonic CNS
structures and first gather on neuroectodermal surface structures in the meninges at 8.5 dpc, where they differentiate into primitive macrophage-like
cells (yellow cells) in situ. B. These early embryonic microglia (yellow cells) migrate into the neuroepithelium where they are proliferative and phago-
cytotically active. C. During postnatal (P) development, microglia lose their phagocytic activity, become quiescent, and gain their typical ramified
morphology (yellow cells). Figure by Markus Knust.

174 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
repopulation of the CNS by donor-derived microglia (Beers microglia might have an essential role in the development of
et al. 2006). However, it remains to be proved whether this the CNS throughout these species.
postnatal recruitment of microglial precursors also occurs in The phases of macrophage colonization of the neuroecto-
wild-type animals. derm in rodents and humans (Sorokin et al. 1992; Verney et al.
A recent study sheds light on the origin of microglia 2010) are similar to those observed in the chick and zebrafish
(Ginhoux et al. 2010). By inducing Cre recombinase activ- (Cuadros et al. 1993; Herbomel et al. 2001). Although there
ity in the Runx locus through injections of tamoxifen into are very few macrophages in the mouse embryo at 8.5 dpc,
pregnant mice between 7.0 to 7.5 dpc, when embryonic most are found in the mesenchyme of the head and neck.
hematopoiesis is restricted to the yolk sac, the authors iden- These macrophages are thought to enter the mesenchyme by
tified immature yolk sac macrophages as the predominant crossing the pial surface of the meninges (see Fig. 15.3). They
source of microglia. Interestingly, myeloid progenitors from subsequently migrate from the mesenchyme through the base-
the blood did not significantly contribute to the pool of ment membrane of the neuroepithelium toward the ventricle.
adult microglia after birth, at odds with previous studies and Once inside the neuroepithelium, the macrophages rapidly
strongly suggesting that the expansion of microglial numbers proliferate and colonize the brain from the dorsal to the ventral
in the postnatal period depends on the proliferation of the side and rostrally to caudally (Pont-Lezica et al. 2011). Within
resident microglia population. Indeed, local proliferation the next hours microglia move deeper into the developing
of microglia rather than the entry of circulating monocytes parenchyma and begin to differentiate, become branched, and
into the CNS during autoimmune inflammation was shown express markers of mature microglia, thereby losing features
just recently (Ajami et al. 2011). Therefore, the vast major- of immature macrophages (Fig. 15.4) (Herbomel et al. 2001).
ity of adult microglia appears to be yolk sac–derived (from In the developing mouse brain, microglia numbers increase
a remarkably restricted time period during early embryogen- steadily and reach approximately 60% of proliferating micro-
esis). It remains an open question whether adult microglia glia (Alliot et al. 1999).
could also be derived in part from the embryonic liver or Although microglia can be found throughout the brain,
other hematopoietic organs during embryogenesis, such as they are not uniformly distributed during development, but
the AGM. One limitation of this seminal work (Ginhoux et tend to preferentially dwell in specific locations (Cuadros
al. 2010) is that only one third of the yolk sac macrophages et al. 1993; Verney et al. 2010). Microglia therefore preferen-
could be labeled genetically. tially concentrate at specific sites in the growing brain such
as in: (1) areas of increased neuronal apoptosis; (2) close
proximity to sprouting vessels; (3) close relation to radial
4 PAT H WAYS O F M I C R O G L I A I N TO glial cells; and finally (4) the marginal layer that contains
T H E D E VE L O P I N G B R A I N developing axon bundles (Dalmau et al. 1997; Rezaie and
Male 1999; Rigato et al. 2011). Therefore, microglia are obvi-
It is remarkable that key features of microglia ontogeny are ously required for proper neuronal and vessel development
obviously preserved among vertebrates and quite similar in in the brain, as described in more detail in the following
humans, mouse, rat, chicken, and fish (Cuadros et al. 1993; paragraphs.
Herbomel et al. 2001; Pont-Lezica et al. 2011; Sorokin Although the kinetics of microglia engraftment into
et al. 1992; Takahashi et al. 1989). This strongly indicates that the developing brain have been described in great detail by

embryonic microglia adult resting microgila


phagocytosing non-phagocytosing
CD11b+, CD45high, CX3CR1+, Iba-1+, CD11b+, CD45low, CX3CR1+, Iba-1+,
F4/80+, LAMP-2+ F4/80low

Figure 15.4 Different Morphological Appearance of Embryonic Versus Adult Rodent Microglia. Embryonic microglia (12,5 dpc, left) exhibit a
macrophage-like appearance with round soma and no processes and are phagocytically active. In contrast, adult microglia (P60, right) lose their phago-
cytic activity and undergo dramatic morphological changes giving rise to typical microglia-like cells with roundish to fusiform cell bodies and fine
delineated processes. Typical markers expressed in the respective developmental stages are indicated. Pictures by Katrin Kierdorf.

M I C R O G L I A L I N E AG E A N D D E VE L O PM E N T • 175
several groups, we do not yet know the guidance cues that et al. 1993; Herbomel et al. 2001; Rigato et al. 2011). This
direct microglia to their final target. In addition, the poten- strongly suggests that early macrophages colonize the CNS
tially protease-mediated signals that may facilitate microglia independently of blood vessels; however, the possibility
migration into the depth of the neuroepithelium are still that, at later time points, microglia may enter via blood ves-
unknown. sels cannot be excluded.
In turn, microglia may even support angiogenesis in a chap-
erone-like manner. Recent in vivo studies elegantly showed
5 T H E I M P O RTA N C E O F M I C R O G L I A that microglia apparently facilitate tip cell fusion, thus induc-
F O R B R A I N D E VE L O PM E N T ing vessel anastomosis (Fantin et al. 2010; Rymo et al. 2011).
Communication between microglia and vascular sprouts
Why are macrophages recruited to the developing brain appears to be collaborative because microglia quickly migrate
and what might be their functional relevance for this organ? toward the developing vessels, and their angiogenic activity is
Several reports have shown that during development, the enhanced in growing vessels (Rymo et al. 2011). As a matter of
arrival of microglia in the brain is correlated with the pres- fact, mice with reduced numbers of microglia such as Csfop/op,
ence of apoptotic cells, mostly neurons (Ashwell 1991; PU.1 (Table 15.1) or after depletion using clodronate lipo-
Cuadros and Navascues 1998; Wakselman et al. 2008). somes have a sparser vascular network (Checchin et al. 2006;
In fact, association of microglia and neurons undergoing devel- Fantin et al. 2010; Rymo et al. 2011).
opmental death has been described in various brain regions The association of microglia with developing neuronal
during development such as the hippocampus (Dalmau et al. axons throughout brain development has been reported in
1997; Wakselman et al. 2008), the cerebellum (Marin-Teva several species (Ashwell 1990; Cuadros et al. 1993; Herbomel
et al. 2004), the spinal cord (Chan et al. 2007; Rigato et al. et al. 2001; Verney et al. 2010). In fact, as the first marginal
2011; Sedel et al. 2004), and the retina (Ashwell et al. 1989; zones of the brain develop, microglia are consistently found in
Cuadros et al. 1992). Transgenic zebrafish expressing GFP in close association with the developing axon fascicles (Cuadros
the macrophage-specific ApoE locus were recently used to et al. 1993).
investigate microglia interaction with dying neurons in the The remarkable and recurring association of microglia
living animal (Peri and Nusslein-Volhard 2008). Engulfment with developing axons also raises the question of whether
of apoptotic neurons by microglia occurred in compartments microglia may have a role in neurite development. Indeed,
arising from the progressive fusion of vesicles and was depen- microglia are positioned at the right place and appear at the
dent on changes of the intracellular pH. This engulfing capac- proper time to influence the setup of axon tracts, axon remod-
ity might also explain that in the rodent retina, the peak of eling, and putatively, synaptogenesis. For example, the elegant
microglia density coincides with a period of retinal ganglion combination of 2-photon imaging and high resolution trans-
cell and optic nerve axon reduction (Ashwell et al. 1989). In mission electron microscopy showed that microglia processes
addition to their phagocytic and thereby clearing role, micro- in the juvenile visual cortex of the mouse permanently con-
glia have an instructive role in developmental cell death. It tact synaptic elements and that this apposition is regulated by
has been shown that in the embryonic chick and mouse retina sensory experience (Tremblay et al. 2010). In another recent
as well as in the neonatal mouse cerebellum and hippocam- study using CX3CR1GFP/GFP mice, microglia were shown to
pus microglia can instruct cells to apoptosis by production of be able to actively engulf synapses in young postnatal animals
nerve growth factor or induction of a respiratory burst (Frade (Paolicelli et al. 2011). These effects were remarkably tran-
and Barde 1998; Marin-Teva et al. 2004; Wakselman et al. sient because microglia loss observed in microglia-specific
2008). CX3CR1GFP/GFP mice, surprisingly was restricted to a short
Therefore, the ability of microglia to phagocytose and period of time, namely postnatal days 8 to 18. In light of
induce neuronal death grant microglia an important role in recent reports that the chemokine receptor CX3CR1 also
CNS development. Interestingly, microglia can also be found essentially regulates cellular activation in microglia (Prinz et
in large numbers even in the absence of apoptotic cells (Rezaie al. 2011) it is tempting to speculate that the observed effects
and Male 1999; Wakselman et al. 2008), suggesting additional on neuronal circuit maturation in CX3CR1GFP/GFP mice might
roles during CNS development. also be partially owing to CX3CR1-mediated effects on other
It has long been recognized that during development of cells, such as astrocytes or even neurons directly. Interestingly,
the neuroepithelium, microglia are found in close proximity developing microglia express the complement receptor CR3
to the developing vasculature (Chan et al. 2007; Cuadros and it has been proposed that they eliminate unwanted
et al. 1993; Herbomel et al. 2001; Rigato et al. 2011). synapses marked by the complement protein C1q (Stevens
It was recently suggested that microglia enter the brain et al. 2007). Furthermore, an essential role for microglia in
parenchyma through blood vessels (Ginhoux et al. 2010). postnatal neurogenesis has now been demonstrated in 2- to
Indeed, microglia were absent in the brains of Ncx mutant 3-day-old postnatal mice (Sierra et al. 2010). Determining
mice (Ginhoux et al. 2010), but a complete lack of heart- whether microglia are important for adult neurogenesis that
beat in these animals could also impair vessel-independent takes place months later will need to be the objective of future
microglia engraftment into the CNS. In line with this idea, research.
early microglia were identified within the brain paren- Taken together, these data suggest that microglia are poten-
chyma already before the onset of vascularization (Cuadros tial modifiers of synapse physiology during development.

176 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Table 15.1 SOME ESSENTIAL MOLECULES REGULATING MICROGLIA DEVELOPMENT
MORPHOLOGY
MOLECULE OF MICROGLIA NUMBER OF MICROGLIA REFERENCES

PU.1 NA ppp (Beers et al. 2006; Herbomel et al. 2001)


Csf-1 p p (Wegiel et al. 1998)
Csf-1R ppp ppp (Erblich et al. 2011; Ginhoux et al. 2010)
DAP12 ND pp (Otero et al. 2009)
Thymosin α1 p p (Htain et al. 1997)
Microglia phenotypes in knockout animals lacking specific transcription factors or cytokines. The p indicates a reduction in knockout animals. The number of arrows
indicates the relative strength of the effect.
ND, not determined; NA, not allocable.

6 R E GU L AT I O N O F M I C R O G L I A highly expressed in Natural Killer (NK) cells cells and myeloid


D E VE L O PM E N T cells.
A transcription factor that is expressed exclusively in
The transcriptional program that controls macrophage–mi- hematopoietic cells and is involved in microglia development
croglia differentiation is only poorly understood. Indeed, most as well is PU.1. The PU.1 (Sfpi-1) gene is a member of the ets
studies on the transcriptional control of macrophage differen- family of transcription factors (Rosenbauer and Tenen 2007).
tiation in general were done in vitro with progenitor-enriched Its target disruption leads to multiple hematopoietic abnor-
populations. Thus, the precise role of these factors in driv- malities, including a lack of mature B cells and macrophages
ing the differentiation of yolk sac macrophages into typical (McKercher et al. 1996). In fact, PU.1-deficient mice are devoid
microglia in vivo remains to be determined. This is particularly not only of circulating monocytes and tissue macrophages
important, given that functional macrophage specialization is (McKercher et al. 1996), but also of parenchymal microglia
likely to be regulated at tissue level, such as in the brain. Some in the brain (Beers et al. 2006). Similar data have been
essential molecules regulating microglia development have obtained in zebrafish microglia. In the zebrafish, yolk sac–
already been described (see Table 15.1). A dramatic reduction derived macrophages enter the developing brain where they
of tissue macrophages including microglia has been observed develop into immature microglia with high endocytotic activ-
in Csf1r–/– mice that lack the colony stimulating factor (Csf-1) ity in a PU.1-dependent fashion because the Panther muta-
receptor (Dai et al. 2002; Erblich et al. 2011; Ginhoux et al. tion of PU.1 leads to a complete loss of brain macrophages
2010) and in Csf-1op/op mutant mice (Wegiel et al. 1998; (Herbomel et al. 2001).
Yoshida et al. 1990), that have a natural null mutation in the Further studies investigated microglia development in
Csf 1 gene, establishing the key role of Csf-1 and its receptor athymic mice and demonstrated a significant reduction of
in microglia homeostasis in vivo (Pixley and Stanley 2004). CD11b+ microglia in various brain regions (Htain et al. 1997).
The precise role of Csf-1 and its receptor in microglia com- This effect was attributed to a disturbed production of the
mitment remains controversial. One hypothesis suggests that hormone thymosin by the thymus gland, which is thought to
Csf-1 drives the differentiation of phagocytic yolk sac mac- be essential for the maturation of myeloid progenitors.
rophages entering the embryo into microglia (Metcalf 1985), In contrast, only few researchers still believe that microglia
whereas a different theory proposes that Csf-1 provides a sur- are derived from neuroectodermal matrix cells that differen-
vival signal for the differentiating macrophages and that sur- tiate locally into microglia (Chan et al. 2007). Others have
viving cells use an intrinsic developmental program to become suggested that microglia originate from pericytes (Baron and
mature microglia (Lagasse and Weissman 1997; Nakahata et Gallego 1972) or from the subependyma adjacent to the lat-
al. 1982). Interestingly, microglia are more profoundly affected eral ventricles (Lewis 1968). Although the myelomonocytic
in the absence of Csf-1R than in the absence of its ligand Csf-1 origin of microglia has now been widely accepted, the neu-
(Ginhoux et al. 2010). Of note, a second Csf-1R ligand called roectodermal hypothesis remains interesting from a historical
interleukin 34 (IL-34) has been identified recently (Lin et al. point of view.
2008). Indeed, IL-34 is highly expressed in the brain of post-
natal mice (Wei et al. 2010), but the exact role of IL-34 in
the development and homeostasis of microglia remains to be 7 B O N E M A R R OW–D E R I VE D M I C R O G L I A
determined. Therefore, we are eagerly awaiting the generation D U R I N G A D U LT H O O D
of IL-34–deficient mice. A comparable phenotype of reduced
microglia cell number could be detected in mice deficient for One of the most important questions in the field of micro-
an adaptor protein for Csf-1R, DAP12 (Otero et al. 2009). glia research during the last years has been, whether “bone
DAP12 contains an ITAM in its cytoplasmic domain and is marrow–derived microglia” in the adult brain exist and if

M I C R O G L I A L I N E AG E A N D D E VE L O PM E N T • 177
so, whether they are functional. The answer to this question However, all these studies used irradiation of the recipients fol-
could have tremendous clinical implications for the treatment lowed by whole BM transplantation to discriminate between
of many important diseases of the human CNS such as amyo- the labeled hematopoietic cells from the donor and the resi-
trophic lateral sclerosis, Alzheimer, and Parkinson disease, dent microglia in the hosts. An alternative strategy was used
because specific peripheral microglia precursors could be used by Massengale and colleagues. They investigated the fate of
as carriers to deliver neuroprotective or immune relevant genes hematopoietic cells in the brain by using parabiotic mice in
into the diseased CNS to modulate pathology. which the bloodstream of a GFP-positive partner was con-
The first seminal cell transplantation experiments in rats nected to a GFP-negative mouse (Massengale et al. 2005).
demonstrated that only perivascular macrophages, but not cells Although the comparison of data from different laboratories
with ramified microglia characteristics in the parenchyma, could using divergent protocols is problematic, the parabiosis model
be observed in the CNS after BM transplantation (Hickey and always resulted in dramatically less BM-DP in the brain com-
Kimura 1988; Hickey et al. 1992). Similar results were obtained pared with irradiated chimeras (Massengale et al. 2005). What
in humans, in female patients who underwent sex-mismatched might have been the reason for these discrepancies in microglia
BM transplantation and were examined for the engraftment of engraftment between the parabiosis model and BM transplan-
Y-chromosome–positive microglial cells (Unger et al. 1993). tation studies using irradiation protocols? Two possibilities
Importantly, only donor-derived perivascular macrophages but were conceivable. First, lethal irradiation could have significant
no parenchymal microglia could be detected in this study. It influences on the CNS tissue. Indeed, it had been reported
is important to emphasize that all these studies are based on that irradiation affected the integrity of the BBB (Diserbo
immunohistochemical approaches and therefore lack the sensi- et al. 2002; Yuan et al. 2003) and the expression of tight junc-
tivity of cell transfer experiments with genetically labeled cells. tion proteins (Kaya et al. 2004), and also induced apoptosis
Priller et al. (2001) were among the first who used green fluo- of endothelial cells (Li et al. 2004). An alternative explanation
rescent protein (GFP)–marked hematopoietic cells transduced could have been the injection of BM cells by which myeloid
with retroviral vectors to examine the long-term fate of myeloid precursors gain nonphysiological access to the circulation and
cells in the murine CNS after BM transplantation in an experi- thereby facilitate phagocyte generation from the bloodstream.
mental setting, including whole body irradiation. Similar to To clarify these questions, we and others performed experi-
other groups they were able to demonstrate GFP-expressing ments to shed light onto the mystery of BM-DP (Ajami et al.
parenchymal microglia deep in the cerebellum, striatum and 2007; Mildner et al. 2007). An experimental setup was used
hippocampus several weeks after transplantation (Eglitis and in which the recipient mice received only partial irradiation
Mezey 1997; Priller et al. 2001). Despite the differences to the by excluding the head from the irradiation field and thus cir-
other studies mentioned, the concept of bone marrow–derived cumventing any irradiation-induced changes of the brain
phagocytes in the CNS was firmly established. In the follow- (“protected” irradiation). Importantly, de novo generation of
ing years, a plethora of publications appeared which examined BM-DP from the circulation was strongly diminished in the
the assumed function and fate of BM-derived mononuclear brains of mice in which this tissue was not irradiated before
phagocytes (BM-DP) in different neurological models using transplantation and also when the chemokine receptor (CCR)
similar experimental paradigms. Remarkably, CNS infiltration 2 was absent from the BM (see Fig. 15.5) (Mildner et al. 2007,
of BM-DP was demonstrated in animal disease models with 2011). CCR2 is required for BM cells to egress from the bone
no obvious BBB damage, such as amyotrophic lateral sclerosis marrow (Mildner et al. 2009; Serbina and Pamer 2006).
(Solomon et al. 2006), Alzheimer disease (Malm et al. 2005; However, GFP-positive CCR2+ BM-DP exhibiting microglia
Mildner et al. 2011; Simard et al. 2006), scrapie (Priller et al. morphology could be detected in unprotected and thus irra-
2006), and many more (Djukic et al. 2006; Priller et al. 2001). diated parts of the CNS such as the spinal cord. These data

CCR2
B

bone marrow precursors transport via blood stream conditioning of CNS (irradiation + disease) bone marrow derived phagocytes in the CNS

Figure 15.5 Formation of Bone Marrow–Derived Phagocytes (BM-DP) in the Adult Mouse Brain. Postnatal BM-DP form only under defined host
conditions in the CNS. Bone marrow precursors (left) are released into the bloodstream in a chemokine receptor (CCR) 2–dependent fashion and
may enter the conditioned CNS. Local conditioning of the CNS can occur via irradiation and neurodegeneration that both lead to disturbances of the
blood-brain barrier allowing engraftment of BM-DP. A. Yolk sac–derived microglia (orange). B. Bone marrow–derived phagocyte (green). Figure by
Markus Knust.

178 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
clearly support the idea that irradiation itself has a significant transplantation with gene-modified HSC in nonirradiated
influence on the engraftment of microglial precursors in the humans (Cartier et al. 2009). Adrenoleukodystrophy is a fatal
CNS under physiological as well as under pathological con- demyelinating disease of the CNS caused by mutations of the
ditions. Ajami et al. (2007), on the other hand, investigated adenosine triphosphate–binding cassette transporter (ABCD1)
the recruitment of peripheral myeloid precursors to the CNS gene. Cartier et al. (2009) purified CD34+ HSC of two patients
in the parabiosis model. The bloodstream of a GFP-positive with mutated ABCD1 genes. After isolation, the cells were trans-
mouse was connected with a GFP-negative animal and the duced ex vivo with a lentivirus encoding for a functional ABCD1
presence of GFP-expressing mononuclear cells was examined gene and transplanted back into the patients after they received
during steady state as well as pathogenic conditions in the CNS myeloablative treatment with cyclophosphamide and busulfan.
of the GFP-negative recipient. Unlike Massengale et al. (2005), Surprisingly, despite low levels of ABCD1-expressing periph-
Ajami et al. (2007) failed to detect BM-DP cells in the CNS of eral blood cells (only 15% of leukocytes produced ABCD1),
the GFP-negative partner under any tested conditions. These progressive cerebral demyelination stopped in both patients
findings indicate that the engraftment of bone marrow–derived and the levels of very long-chain fatty acids in the plasma as
myeloid cells in the CNS is an extremely rare event, which is indicators of disease activity decreased. Whether the BBB was
strongly influenced by the experimental design, for example, intact in these patients even on a microscopic level remains to
irradiation. Furthermore, these experiments also point to the be determined, but at least gross measurements by magnetic
fact that endogenous microglia exhibit a high potential for resonance imaging (MRI) indicated no major disturbances of
self-renewal and proliferation that was confirmed by a recent this physiological barrier. However, the authors attributed the
publication (Ajami et al. 2011). However, there are also some beneficial outcome in the treated patients to “the recruitment of
discrepancies between the studies of Mildner and Ajami and BM-derived cells to CNS macrophages/microglia” because they
questions remain. In one experiment, Ajami et al. irradiated the were able to observe precisely this treatment effect in an animal
GFP-negative partner of a parabiotic pair and analyzed the brain model using irradiated mice. The authors therefore speculated
6 weeks after irradiation for the presence of GFP-expressing that this approach “may provide a new avenue for cell-base gene
CNS mononuclear phagocytes. However, despite the irra- therapy in . . . CNS diseases” (Cartier et al. 2009). Consequently,
diation and high levels of chimerism, they still did not detect the identification of a microglial precursor for the therapy of
any GFP-positive cells in the recipient brain. Was this because CNS diseases is of great importance.
the mice did not receive an intravenous injection of BM cells, These findings clearly indicate that the engraftment of bone
which contain potential myeloid precursors, as was the case in marrow–derived myeloid cells in the CNS is an extremely rare
BM-transplanted mice? This is an interesting possibility, but it event, which is strongly influenced by the experimental design
cannot explain the fact that we failed to detect donor-derived (e.g., cranial irradiation and intravenous transfer of femoral
cells in the brains of protected (head shielded but body irra- bone marrow–enriched for hematopoietic progenitors and
diated) animals that also received an intravenous injection of stem cells). Furthermore, these results underscore the fact that
BM cells (Mildner et al. 2007). It is most likely that irradia- endogenous microglia are of yolk sac origin and exhibit a high
tion as a precondition, together with the injection of BM cells potential for self-renewal and proliferation. Nevertheless, in
synergistically facilitated BM-DP development. One concern my personal view, bone marrow–derived phagocytes might be
that arose from these studies is the supposed low chimerism of capable of exploitation for potential therapeutic application
the examined mice (Soulet and Rivest 2008). In fact, the para- in neurodegenerative settings, although the requirement for
biosis model results in a chimerism rate of about 40% to 50%, cranial irradiation to achieve CNS engraftment might limit
which is well comparable to the rate observed in protected their utility.
(head shielded but body irradiated) animals, in which the skull
bone marrow also contributes to hematopoiesis. Nevertheless,
both studies undoubtedly support the hypothesis that the rate 8 S U M M A RY A N D P E R S P E C T I VE S
of microglia turnover from the circulation under physiological
conditions in experiments using irradiation is overestimated Although long underestimated, microglia nowadays comprise
because of irradiation-induced changes and artificial BM cell an attractive target for accessing the diseased CNS. Their
injections into the bloodstream. On the other hand, an under- presence has been confirmed extensively in countless reports
estimation of microglia turnover in parabiosis animal models describing their involvement in virtually all neuropathologies.
or head-shielded irradiation methods because of low chime- Furthermore, their role for ensuring normal brain homeostasis
rism is also possible. In any case, much of the previous work on is undoubted. However, the question as to their origin is still
chimeric mouse models which involved whole body irradiation controversial, but recent achievements shed some light on the
has to be re-evaluated under these aspects. mystery of microglia ancestry.
Despite all this complexity one point is indisputable. Under Given the fact that endogenous microglia are not replen-
specific conditions, BM-DP precursors can engraft into the ished by the periphery at all under normal CNS conditions
brain and become an integral part of the cellular network in and only sparsely during neurodegenerative disease, if the
the CNS. These specific conditions include irradiation but BBB integrity is broken, it will be a major challenge for future
might also occur in certain diseases with no obvious primary research to facilitate the use of this ambivalent Trojan horse
BBB damage, such as X-linked adrenoleukodystrophy. A recent to access the brain for putative therapeutic approaches. On
publication indeed reports of successful autologous stem cell the other hand, it is now believed that localized self-renewal

M I C R O G L I A L I N E AG E A N D D E VE L O PM E N T • 179
can sustain microglia maintenance and probably also func- Cuadros MA, Martin C, Coltey P, Almendros A, Navascues J. 1993.
tion throughout adult life as a main mechanism of micro- First appearance, distribution, and origin of macrophages in the early
development of the avian central nervous system. J Comp Neurol
gliosis under conditions of preserved BBB. Therefore, further 330:113–129.
research is necessary to understand key mechanisms and mole- Cuadros MA, Moujahid A, Martin-Partido G, Navascues J. 1992. Microglia
cules involved in microglia development, local expansion, and in the mature and developing quail brain as revealed by a monoclonal
migration to the CNS during development. antibody recognizing hemopoietic cells. Neurosci Lett 148:11–14.
Cuadros MA, Navascues J. 1998. The origin and differentiation of
microglial cells during development. Prog Neurobiol 56:173–189.
Cumano A, Godin I. 2007. Ontogeny of the hematopoietic system.
AC K N OW L E D G M E N T S Annu Rev Immunol 25:745–785.
Dai XM, Ryan GR, Hapel AJ, Dominguez MG, Russell RG, Kapp S, et
The author thanks people working at the Department of al. 2002. Targeted disruption of the mouse colony-stimulating factor
Neuropathology for the continuous and lively support of 1 receptor gene results in osteopetrosis, mononuclear phagocyte defi-
ciency, increased primitive progenitor cell frequencies, and reproduc-
“microglia,” especially Alexander (“Akki”) Mildner (now in tive defects. Blood 99:111–120.
Rehovot, Israel) and Katrin Kierdorf, who has made excep- Dalmau I, Finsen B, Tonder N, Zimmer J, Gonzalez B, Castellano B.
tional investigations in the field of microglia biology in the 1997. Development of microglia in the prenatal rat hippocampus.
last years. Moreover, the author is especially grateful to Markus J Comp Neurol 377:70–84.
Knust (“Knust”) for his gifted pictures illustrating this chapter, Diserbo M, Agin A, Lamproglou I, Mauris J, Staali F, Multon E, et al.
2002. Blood-brain barrier permeability after gamma whole-body
and thanks Stefanie Brendecke for critical reading and edit- irradiation: an in vivo microdialysis study. Can J Physiol Pharmacol
ing. Without the enthusiasm of these and other co-workers of 80:670–678.
the Department of Neuropathology the work of the last years Djukic M, Mildner A, Schmidt H, Czesnik D, Bruck W, Priller J, et al.
would not have been possible. 2006. Circulating monocytes engraft in the brain, differentiate into
microglia and contribute to the pathology following meningitis in
mice. Brain 129:2394–2403.
Eglitis MA, Mezey E. 1997. Hematopoietic cells differentiate into both
REFERENCES microglia and macroglia in the brains of adult mice. Proc Natl Acad
Sci U S A 94:4080–4085.
Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FM. 2011. Enose Y, Destache CJ, Mack AL, Anderson JR, Ullrich F, Ciborowski
Infi ltrating monocytes trigger EAE progression, but do not contrib- PS, et al. 2005. Proteomic fingerprints distinguish microglia, bone
ute to the resident microglia pool. Nat Neurosci 14:1142–1149. marrow, and spleen macrophage populations. Glia 51:161–172.
Ajami B, Bennett JL, Krieger C, Tetzlaff W, Rossi FM. 2007. Local Erblich B, Zhu L, Etgen AM, Dobrenis K, Pollard JW. 2011. Absence of
self-renewal can sustain CNS microglia maintenance and function colony stimulation factor-1 receptor results in loss of microglia, dis-
throughout adult life. Nat Neurosci 10:1538–1543. rupted brain development and olfactory deficits. PLoS One 6:e26317.
Alliot F, Godin I, Pessac B. 1999. Microglia derive from progenitors, Fantin A, Vieira JM, Gestri G, Denti L, Schwarz Q, Prykhozhij S,
originating from the yolk sac, and which proliferate in the brain. et al. 2010. Tissue macrophages act as cellular chaperones for vascu-
Brain Res Dev Brain Res 117:145–152. lar anastomosis downstream of VEGF-mediated endothelial tip cell
Alliot F, Lecain E, Grima B, Pessac B. 1991. Microglial progenitors with induction. Blood 116:829–840.
a high proliferative potential in the embryonic and adult mouse brain. Frade JM, Barde YA. 1998. Microglia-derived nerve growth factor causes
Proc Natl Acad Sci U S A 88:1541–1545. cell death in the developing retina. Neuron 20:35–41.
Aschoff L. 1924. The reticoloendothelial system. Das retikuloendotheli- Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. 2010.
ale System. Erg Inn Med Kinderheilk 26:S1–117. Development of monocytes, macrophages, and dendritic cells. Science
Ashwell K. 1990. Microglia and cell death in the developing mouse cere- 327:656–661.
bellum. Brain Res Dev Brain Res 55:219–230. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. 2010.
Ashwell K. 1991. The distribution of microglia and cell death in the fetal Fate mapping analysis reveals that adult microglia derive from primi-
rat forebrain. Brain Res Dev Brain Res 58:1–12. tive macrophages. Science 330:841–845.
Ashwell KW, Hollander H, Streit W, Stone J. 1989. The appearance Glanzer JG, Enose Y, Wang T, Kadiu I, Gong N, Rozek W, et al. 2007.
and distribution of microglia in the developing retina of the rat. Vis Genomic and proteomic microglial profi ling: pathways for neuropro-
Neurosci 2:437–448. tective inflammatory responses following nerve fragment clearance
Baron M, Gallego A. 1972. The relation of the microglia with the pericytes and activation. J Neurochem 102:627–645.
in the cat cerebral cortex. Z Zellforsch Mikrosk Anat 128:42–57. Gordon S. 2002. Pattern recognition receptors: doubling up for the
Beers DR, Henkel JS, Xiao Q, Zhao W, Wang J, Yen AA, et al. 2006. innate immune response. Cell 111:927–930.
Wild-type microglia extend survival in PU.1 knockout mice with Gordon S, Taylor PR. 2005. Monocyte and macrophage heterogeneity.
familial amyotrophic lateral sclerosis. Proc Natl Acad Sci U S A Nat Rev Immunol 5:953–964.
103:16021–16026. Hanisch UK. 2002. Microglia as a source and target of cytokines. Glia
Bertrand JY, Jalil A, Klaine M, Jung S, Cumano A, Godin I. 2005. Three 40:140–155.
pathways to mature macrophages in the early mouse yolk sac. Blood Herbomel P, Th isse B, Th isse C. 2001. Zebrafish early macrophages colo-
106:3004–3011. nize cephalic mesenchyme and developing brain, retina, and epider-
Cartier N, Hacein-Bey-Abina S, Bartholomae CC, Veres G, Schmidt M, mis through a M-CSF receptor-dependent invasive process. Dev Biol
Kutschera I, et al. 2009. Hematopoietic stem cell gene therapy with 238:274–288.
a lentiviral vector in X-linked adrenoleukodystrophy. Science 326: Hickey WF, Kimura H. 1988. Perivascular microglial cells of the
818–823. CNS are bone marrow-derived and present antigen in vivo. Science
Chan WY, Kohsaka S, Rezaie P. 2007. The origin and cell lineage of 239:290–292.
microglia: new concepts. Brain Res Rev 53:344–354. Hickey WF, Vass K, Lassmann H. 1992. Bone marrow-derived elements
Checchin D, Sennlaub F, Levavasseur E, Leduc M, Chemtob S. 2006. in the central nervous system: an immunohistochemical and
Potential role of microglia in retinal blood vessel formation. Invest ultrastructural survey of rat chimeras. J Neuropathol Exp Neurol 51:
Ophthalmol Vis Sci 47:3595–3602. 246–256.

180 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Htain WW, Leong SK, Yuen R, Ling EA. 1997. Effects of thymosin Perry VH, Hume DA, Gordon S. 1985. Immunohistochemical localiza-
alpha1 on the development of amoeboid microglial cells in the cor- tion of macrophages and microglia in the adult and developing mouse
pus callosum of neonatal BALB/c and athymic mice. Brain Res brain. Neuroscience 15:313–326.
755:63–73. Pixley FJ, Stanley ER. 2004. CSF-1 regulation of the wandering macro-
Kaufmann SH. 2008. Immunology’s foundation: the 100-year anni- phage: complexity in action. Trends Cell Biol 14:628–638.
versary of the Nobel Prize to Paul Ehrlich and Elie Metchnikoff. Pont-Lezica L, Bechade C, Belarif-Cantaut Y, Pascual O, Bessis A. 2011.
Nat Immunol 9:705–712. Physiological roles of microglia during development. J Neurochem
Kaya M, Palanduz A, Kalayci R, Kemikler G, Simsek G, Bilgic B, 119:901–908.
et al. 2004. Effects of lipopolysaccharide on the radiation-induced Priller J, Flugel A, Wehner T, Boentert M, Haas CA, Prinz M, et al.
changes in the blood-brain barrier and the astrocytes. Brain Res 2001. Targeting gene-modified hematopoietic cells to the central
1019:105–112. nervous system: use of green fluorescent protein uncovers microglial
Kurz H, Christ B. 1998. Embryonic CNS macrophages and microglia engraftment. Nat Med 7:1356–1361.
do not stem from circulating, but from extravascular precursors. Glia Priller J, Prinz M, Heikenwalder M, Zeller N, Schwarz P, Heppner FL,
22:98–102. et al. 2006. Early and rapid engraftment of bone marrow-derived
Lagasse E, Weissman IL. 1997. Enforced expression of Bcl-2 in mono- microglia in scrapie. J Neurosci 26:11753–11762.
cytes rescues macrophages and partially reverses osteopetrosis in op/ Prinz M, Mildner A. 2011. Microglia in the CNS: immigrants from
op mice. Cell 89:1021–1031. another world. Glia 59:177–187.
Lewis PD. 1968. The fate of the subependymal cell in the adult rat brain, Prinz M, Priller J, Sisodia SS, Ransohoff RM. 2011. Heterogeneity
with a note on the origin of microglia. Brain 91:721–736. of CNS myeloid cells and their roles in neurodegeneration.
Li YQ, Chen P, Jain V, Reilly RM, Wong CS. 2004. Early radiation-induced Nat Neurosci 14:1227–1235.
endothelial cell loss and blood-spinal cord barrier breakdown in the Rezaie P, Male D. 1999. Colonisation of the developing human brain and
rat spinal cord. Radiat Res 161:143–152. spinal cord by microglia: a review. Microsc Res Tech 45: 359–382.
Lichanska AM, Hume DA. 2000. Origins and functions of phagocytes Rigato C, Buckinx R, Le Corronc H, Rigo JM, Legendre P. 2011. Pattern
in the embryo. Exp Hematol 28:601–611. of invasion of the embryonic mouse spinal cord by microglial cells at the
Lin H, Lee E, Hestir K, Leo C, Huang M, Bosch E, et al. 2008. Discovery time of the onset of functional neuronal networks. Glia 59:675–695.
of a cytokine and its receptor by functional screening of the extracel- Rosenbauer F, Tenen DG. 2007. Transcription factors in myeloid devel-
lular proteome. Science 320:807–811. opment: balancing differentiation with transformation. Nat Rev
Malm TM, Koistinaho M, Parepalo M, Vatanen T, Ooka A, Karlsson S, Immunol 7:105–117.
et al. 2005. Bone-marrow-derived cells contribute to the recruitment Rymo SF, Gerhardt H, Wolfhagen SF, Lang R, Uv A, Betsholtz C. 2011.
of microglial cells in response to beta-amyloid deposition in APP/PS1 A two-way communication between microglial cells and angiogenic
double transgenic Alzheimer mice. Neurobiol Dis 18:134–142. sprouts regulates angiogenesis in aortic ring cultures. PLoS One
Mander TH, Morris JF. 1996. Development of microglia and macrophages 6:e15846.
in the postnatal rat pituitary. Cell Tissue Res 286:347–355. Schmid CD, Melchior B, Masek K, Puntambekar SS, Danielson PE,
Marin-Teva JL, Dusart I, Colin C, Gervais A, van Rooijen N, Mallat Lo DD, et al. 2009. Differential gene expression in LPS/IFNgamma
M. 2004. Microglia promote the death of developing Purkinje cells. activated microglia and macrophages: in vitro versus in vivo.
Neuron 41:535–547. J Neurochem 109 Suppl 1:117–125.
Massengale M, Wagers AJ, Vogel H, Weissman IL. 2005. Hematopoietic Sedel F, Bechade C, Vyas S, Triller A. 2004. Macrophage-derived tumor
cells maintain hematopoietic fates upon entering the brain. J Exp necrosis factor alpha, an early developmental signal for motoneuron
Med 201:1579–1589. death. J Neurosci 24:2236–2246.
McKercher SR, Torbett BE, Anderson KL, Henkel GW, Vestal DJ, Serbina NV, Pamer EG. 2006. Monocyte emigration from bone marrow
Baribault H, et al. 1996. Targeted disruption of the PU.1 gene results during bacterial infection requires signals mediated by chemokine
in multiple hematopoietic abnormalities. EMBO J 15:5647–5658. receptor CCR2. Nat Immunol 7:311–317.
Metcalf D. 1985. The granulocyte-macrophage colony stimulating fac- Sierra A, Encinas JM, Deudero JJ, Chancey JH, Enikolopov G,
tors. Cell 43:5–6. Overstreet-Wadiche LS, et al. 2010. Microglia shape adult hippocam-
Mildner A, Mack M, Schmidt H, Bruck W, Djukic M, Zabel MD, pal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem
et al. 2009. CCR2+Ly-6Chi monocytes are crucial for the effector Cell 7:483–495.
phase of autoimmunity in the central nervous system. Brain 132: Simard AR, Soulet D, Gowing G, Julien JP, Rivest S. 2006. Bone
2487–2500. marrow-derived microglia play a critical role in restricting senile
Mildner A, Schlevogt B, Kierdorf K, Bottcher C, Erny D, Kummer plaque formation in Alzheimer’s disease. Neuron 49: 489–502.
MP, et al. 2011. Distinct and non-redundant roles of microglia and Solomon JN, Lewis CA, Ajami B, Corbel SY, Rossi FM, Krieger C.
myeloid subsets in mouse models of Alzheimer’s disease. J Neurosci 2006. Origin and distribution of bone marrow-derived cells in the
31:11159–11171. central nervous system in a mouse model of amyotrophic lateral scle-
Mildner A, Schmidt H, Nitsche M, Merkler D, Hanisch UK, Mack rosis. Glia 53:744–753.
M, et al. 2007. Microglia in the adult brain arise from Ly-6C(hi) Sorokin SP, Hoyt RF, Jr., Blunt DG, McNelly NA. 1992. Macrophage
CCR2(+) monocytes only under defined host conditions. Nat development: II. Early ontogeny of macrophage populations in brain,
Neurosci 10:1544–1553. liver, and lungs of rat embryos as revealed by a lectin marker. Anat
Nakahata T, Gross AJ, Ogawa M. 1982. A stochastic model of self-renewal Rec 232:527–550.
and commitment to differentiation of the primitive hemopoietic stem Soulet D, Rivest S. 2008. Bone-marrow-derived microglia: myth or real-
cells in culture. J Cell Physiol 113:455–458. ity? Curr Opin Pharmacol 8:508–518.
Otero K, Turnbull IR, Poliani PL, Vermi W, Cerutti E, Aoshi T, et al. Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS,
2009. Macrophage colony-stimulating factor induces the prolifera- Nouri N, et al. 2007. The classical complement cascade mediates
tion and survival of macrophages via a pathway involving DAP12 and CNS synapse elimination. Cell 131:1164–1178.
beta-catenin. Nat Immunol 10:734–743. Takahashi K, Naito M, Takeya M. 1996. Development and heterogene-
Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P, ity of macrophages and their related cells through their differentia-
et al. 2011. Synaptic pruning by microglia is necessary for normal tion pathways. Pathol Int 46:473–485.
brain development. Science 333:1456–1458. Takahashi K, Yamamura F, Naito M. 1989. Differentiation, matu-
Peri F, Nusslein-Volhard C. 2008. Live imaging of neuronal degradation ration, and proliferation of macrophages in the mouse yolk sac: a
by microglia reveals a role for v0-ATPase a1 in phagosomal fusion in light-microscopic, enzyme-cytochemical, immunohistochemical,
vivo. Cell 133:916–927. and ultrastructural study. J Leukoc Biol 45:87–96.

M I C R O G L I A L I N E AG E A N D D E VE L O PM E N T • 181
Tremblay ME, Lowery RL, Majewska AK. 2010. Microglial interac- microglial CD11b integrin and DAP12 immunoreceptor. J Neurosci
tions with synapses are modulated by visual experience. PLoS Biol 28:8138–8143.
8:e1000527. Wegiel J, Wisniewski HM, Dziewiatkowski J, Tarnawski M, Kozielski R,
Unger ER, Sung JH, Manivel JC, Chenggis ML, Blazar BR, Krivit Trenkner E, et al. 1998. Reduced number and altered morphology of
W. 1993. Male donor-derived cells in the brains of female microglial cells in colony stimulating factor-1-deficient osteopetrotic
sex-mismatched bone marrow transplant recipients: a Y-chromosome op/op mice. Brain Res 804:135–139.
specific in situ hybridization study. J Neuropathol Exp Neurol 52: Wei S, Nandi S, Chitu V, Yeung YG, Yu W, Huang M, et al. 2010.
460–470. Functional overlap but differential expression of CSF-1 and IL-34 in
van Furth R, Cohn ZA, Hirsch JG, Humphrey JH, Spector WG, their CSF-1 receptor-mediated regulation of myeloid cells. J Leukoc
Langevoort HL. 1972. The mononuclear phagocyte system: a new Biol 88:495–505.
classification of macrophages, monocytes, and their precursor cells. Yoshida H, Hayashi S, Kunisada T, Ogawa M, Nishikawa S, Okamura
Bull World Health Organ 46:845–852. H, et al. 1990. The murine mutation osteopetrosis is in the coding
Verney C, Monier A, Fallet-Bianco C, Gressens P. 2010. Early microg- region of the macrophage colony stimulating factor gene. Nature
lial colonization of the human forebrain and possible involvement 345:442–444.
in periventricular white-matter injury of preterm infants. J Anat Yuan H, Gaber MW, McColgan T, Naimark MD, Kiani MF, Merchant
217:436–448. TE. 2003. Radiation-induced permeability and leukocyte adhesion
Wakselman S, Bechade C, Roumier A, Bernard D, Triller A, Bessis A. in the rat blood-brain barrier: modulation with anti-ICAM-1 anti-
2008. Developmental neuronal death in hippocampus requires the bodies. Brain Res 969:59–69.

182 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
PHYSIOLOGICAL PROPERTIES
This page intentionally left blank
16.
PHYSIOLOGY OF ASTROCY TES: ION CHANNELS
AND ION TRANSPORTER S
Christian Steinhäuser, Gerald Seifert, and Joachim W. Deitmer

A B B R E VI AT I O N S is, to act as direct communication partners of neurons. Although


various types of ion channels, including voltage-gated Na+ (Nav)
AE anion exchanger and Ca2+ (Cav) channels, have been described in the cell culture,
4-AP 4-aminopyridine clear evidence for their expression in astrocytes of acute prepa-
AQP aquaporin rations or in vivo is still lacking. This might partially be caused
ATP adenosine triphosphate by the fact that the identification of small current components
BAPTA 1,2-Bis(o-aminophenoxy)ethane- of astrocytes in situ is hampered by the cells’ very low input
N,N,N′,N′-tetraacetic acid) resistance (usually <2 M:). Moreover, earlier work describing
CA carbonic anhydrase Nav and Cav channels in presumed immature astrocytes par-
Ca2+ pump calcium ATPase tially has to be reconsidered because many of those cells might
CAAC Ca2+-activated anion channel actually have been NG2 cells that have now been identified as
cAMP cyclic adenosine monophosphate a separate glial cell type. Astrocyte current phenotypes are domi-
CCC cation-chloride cotransporter nated by K+ conductances that are important for the regulation
CCE capacitive Ca2+ entry of K+ homeostasis. In addition, water and anion channels are
CNS central nervous system expressed, allowing for the control of the volume of the extra-
EK K+ equilibrium potential cellular space and the release of transmitters from astrocytes.
ER endoplasmic reticulum Astrocytic ion transporters such as the electrogenic Na+/K+
GABA γ-aminobutyric acid pump, the electrogenic Na+/Ca2+ exchanger, or the electrogenic
HEPES 4-(2-hydroxyethyl)-1-piperazineethanesul- Na+/HCO3–-cotransporter are activated in response to changes
fonic acid in extracellular ion concentrations and/or membrane potential.
KCC potassium-chloride cotransporter Depending on the stoichiometry of the transporters, consider-
MCT monocarboxylate transporter able currents can be generated on their activation. In neurons,
Na+-K+-pump sodium-potassium ATPase this can lead to significant changes in excitability: Activation of
NBC sodium-bicarbonate cotransporter the Na+/K+-ATPase generates an outward current and hence
NCX sodium-calcium exchanger counteracts excitation, whereas activation of Na+/Ca2+ exchange
NHE sodium-hydrogen exchanger generates an inward current that depolarizes the cell membrane
NKCC sodium-potassium-2 chloride cotrans-porter and promotes excitation. Because of the low input resistance
NPPB 5-nitro-2-(3-phenylpropylamino)-benzoic of astrocytes, transporter currents have less influence on their
acid membrane potential. Ion transporters are often dependent
OGD oxygen/glucose deprivation on the transmembrane Na+ gradient, and its perturbation can
PEPT peptide transporter result in modulation of transport of various ions and metabolite
PMCA plasma membrane ATP-driven Ca2+ pump substrates. This chapter gives an update about current knowl-
RVD regulatory volume decrease edge of expression and function of astroglial ion channels and
SERCA sarco-endoplasmic reticulum Ca2+ ATPase ion transporters, and discusses its impact on the regulation of
SOCE store operated Ca2+ entry neuronal excitability. Neurotransmitter-activated channels (see
SR101 sulforhodamine 101 chapter 17) and transmitter-activated and metabolic substrate–
TEA tetraethylammonium dependent transporters (see chapters 35 and 27, respectively) are
TTX tetrodotoxin dealt with in other chapters of this book. The focus here is on
VRAC volume-regulated anion channel the properties of astrocytes in acute preparations rather than cell
VSOR volume-sensitive outwardly rectifying culture, whenever possible.
anion channel
2 ION CHANNELS
1 INTRODUCTION
2.1 VOLTAG E - G AT E D Na + A N D Ca 2+ CH A N N E L S
Ion channels and ion-coupled membrane carriers in astrocytes Nav channels represent a prerequisite for initiation and propa-
enable these cells to sense and modulate neuronal activity, that gation of action potentials. The Nav channel family comprises
185
nine different α subunits, Nav1.1 to Nav1.9, although not all and 4-AP sensitive KD and KA currents, respectively (Seifert
are expressed in the CNS. The principal, pore-forming α sub- et al. 2009; Zhou and Kimelberg 2000). KD currents were
unit associates with membrane-spanning auxiliary β-subunits also recorded from immature Bergmann glial cells, but not at
(Navβ1–4). Whether or not astrocytes in situ express functional later (several weeks) developmental stages (Müller et al. 1994).
Nav channels is still under debate. Earlier work reported tetro- Developmental changes were also observed in Müller cells of
dotoxin (TTX)-sensitive Nav currents in presumed astrocytes rabbit retina, where KA currents dominated early after birth
of rat hippocampus (“complex cells”) (Bordey and Sontheimer and declined during maturation, whereas KD currents reached
1997; Kressin et al. 1995) or in the rabbit retina (Clark and their maximum in adulthood (Bringmann et al. 2000a). Little
Mobbs 1994), but from today’s perspective many of these cells is known about the subunits encoding astrocytic Kv currents.
actually might have been NG2 cells rather than astrocytes. In Kv1.5 has been detected immunocytochemically in endfeet of
line with this assumption is a recent study that did not detect hippocampal astrocytes and Bergmann glia (Roy et al. 1996),
Nav currents in SR101-positive astrocytes of the hippocampus and its modulation by Src tyrosine phosphorylation regulates
(Kafitz et al. 2008). However, the latter study did not use TTX cell differentiation in culture (MacFarlane and Sontheimer
or K+ channel blockers for current separation so that small Nav 2000). Astrocytes in spinal cord white matter express Kv1.4
currents might have been overlooked. Nav currents have been (Edwards et al. 2002). Kv11.1 (erg1) mediated K+ currents were
recorded from Müller cells freshly isolated from mouse ret- observed in hippocampal astrocytes in situ, and the authors
ina (Pannicke et al. 2002). Immunohistochemistry detected speculated that they contribute to the clearance of extracellular
Nav1.6 in Bergmann glial cells (Schaller and Caldwell 2003), K+ (Emmi et al. 2000).
but whether these cells carry functional Nav channels remains The activity of large conductance Ca2+ activated K+ chan-
uncertain. In conclusion, in the context of the increasing evi- nels (BK channels, also termed KCa1.1), depends on membrane
dence that NG2 cells in gray matter represent a distinct glial potential and [Ca2+]i. The α subunit can co-assemble with
cell population (Bergles et al. 2010), there is need to recon- β-subunits (BKβ1–4), which changes pharmacological prop-
sider previous conclusions as to the presence of functional Nav erties and the Ca2+-sensitivity of the channels. Small conduc-
channels in astrocytes in acute preparations. tance Ca2+ activated K+ channels (SK channels; KCa2.1–2.3,
By mediating Ca2+ influx on membrane depolarization, KCa3.1) are voltage-independent and possess a smaller single
Cav channels constitute an important route for Ca2+ delivery channel conductance. BK channels are ubiquitously expressed
necessary to regulate a multitude of intracellular processes, for in astrocytes in culture. In brain slices of rat hippocampus and
example, cellular metabolism, secretion, and gene expression. cerebellum, immunohistochemistry and ultrastructural analy-
According to their functional properties, the high-voltage acti- sis revealed co-localization of BK channels with aquaporin4
vated L-type (Cav1.1–1.4) and P/Q-, N-, R-type (Cav2.1–2.3) (AQP4) water channels in perivascular astrocytic endfeet,
channels are distinguished from low-voltage activated T-type suggesting the involvement of BK channels in K+ redistribu-
(Cav3.1–3.3) channels. Cav channels are formed by a princi- tion and regulation of cerebral blood flow (Price et al. 2002).
pal α subunit and auxiliary subunits (Cavα2G1–3, Cavβ1–4, Recent work confirmed that these channels (KCa1.1) in end-
Cavγ2–8). Cultured astrocytes express Cav channels, but their feet mediate arterial dilation and constriction (Filosa et al.
functional expression in situ or in vivo is still unclear. Cav cur- 2006; Girouard et al. 2010) (Fig. 16.1). This α subunit might
rents could be activated in presumed immature hippocampal co-assemble with BKβ4 or BKβ1 (Seidel et al. 2011). In addi-
astrocytes (“complex cells”) (Akopian et al. 1996), but prob- tion, evidence was provided supporting the involvement
ably these recordings were obtained from NG2 cells (see the of KCa3.1 in neurovascular coupling (Longden et al. 2011).
preceding). Immunogold labeling detected Cav1.2 in bona The latter study did not find evidence, however, for functional
fide hippocampal astrocytes (Tippens et al. 2008), and there expression of KCa2.1–2.3 in astrocytes. Interestingly, the activ-
is some indirect hint at its functional expression, but no direct ity of BK and SK channels in perivascular astrocytes is con-
proof has yet been presented. This is in contrast to Müller trolled by metabotropic glutamate receptors (mGluRs) and
cells of the retina, which express L- and T-type Cav channels, epoxyeicosatrienoic acid release (Higashimori et al. 2010).
both in situ and after acute isolation (Bringmann et al. 2000b; Similarly, BK channel activity in Müller cells is regulated by
Newman 1985). metabotropic ATP receptors (P2Y receptors) (Bringmann
et al. 2002). Thus, although neuronal BK and SK channels
couple with Cav channels, their activity in astrocytes is regu-
2.2 VOLTAG E -ACT I VAT E D A N D Ca 2+ -AC T I VAT E D
lated by Ca2+ release from intracellular stores following stimu-
K+ CHANNELS
lation of G-protein–coupled receptors (GPCRs).
So far, 12 subfamilies of the Kv α subunits have been identi-
fied, Kv1 to Kv12, which mediate delayed rectifier (KD) and
2.3 IN WA R D LY R E CT IF Y IN G A N D T WO
transient (KA) K+ currents. Astrocytes in situ are endowed with
P OR E -D OM A IN K + CH A N N E L S
KD and KA currents, although they are masked by much larger
time- and voltage-independent K+ currents mainly mediated Inwardly rectifying K+ (Kir) channels belong to the superfam-
by Kir and K2P conductances (see section 2.3), which hampers ily of two-membrane domain channels and lack an intrinsic
analysis of their kinetics, stationary properties, and develop- voltage sensor. Seven subfamilies have been identified, Kir1 to
mental regulation. To improve voltage control, astrocytes were Kir7, which form homomeric or heteromeric tetramers. Inward
freshly isolated from the hippocampus, which revealed TEA rectification results from positively charged polyamines and

186 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
A Control
4

Current (nA)
IbTX
3
Electrode
2
Parenchymal IbTX-sensitive
arteriole current
1

20 μm

–120 –80 –40 40 80


Voltage (mV)

B Baseline channel activity

C
O

Channel activity following ES


C
O

10 pA
100 ms

Figure 16.1 BK Channels in Astrocytic Endfeet. A. I–V relationship of whole-cell currents before and 7 minutes after exposure to the specific BK chan-
nel inhibitor IbTX (200 nM). Currents were recorded in response to 200 ms voltage ramps, and the holding potential was –80 mV. The IbTX-sensitive
current is shown in gray. The inset depicts positioning of the patch pipette on an endfoot in a brain slice. B. Representative traces showing single BK
channel currents in cell-attached patches from an endfoot before (top) and after (bottom) electrical stimulation (ES). The holding potential was 0 mV,
[K+]o 6 mM. C, closed; O, open state. Reproduced with permission from Filosa et al. 2006.

Mg2+, which plug the channel from the intracellular site in a This segregated expression suggests a model of K+ siphoning
voltage-dependent manner. Kir channels are sensitive to sub- in which K+ influx is mediated by Kir2.1, whereas K+ leaves
millimolar extracellular Ba2+ concentrations, intracellular pH, via Kir4.1 channels (Kofuji et al. 2002). The significance of
and adenosine triphosphate (ATP), and are modulated by G Kir4.1 for spatial buffering and control of excitability has been
proteins. Kir6 subunits co-assemble with sulfonylurea recep- confirmed through general (Kofuji et al. 2000) or conditional
tors to form channels that couple electrical activity to the met- knockout of Kir4.1 (Chever et al. 2010). Astrocytes are het-
abolic state of the cell. Kir channels are abundantly expressed erogeneous with respect to Kir subunit expression patterns.
by astrocytes of various CNS regions and upregulated dur- In the spinal cord, the K+ buffer capacity differs between sub-
ing postnatal development (Verkhratsky and Steinhäuser regions (Olsen et al. 2007). Heteromeric Kir4.1/Kir5.1 chan-
2000), whereas decreased expression is seen in the diseased nels were identified in neocortex and olfactory bulb, whereas
CNS (Seifert et al. 2006). They are predominantly respon- in the hippocampus Kir4.1 mainly forms homomeric channels
sible to set the resting potential close to the K+ equilibrium (Hibino et al. 2004; Seifert et al. 2009) (Fig. 16.2). Highly
potential (EK). Kir channels in astrocytes represent a prereq- pH sensitive Kir4.1/Kir5.1 channels in astrocytes of the ret-
uisite for the regulation of the K+ homeostasis (Orkand et al. rotrapezoid nucleus contribute to chemoreception and the
1966). In the retina, unequal distribution of K+ conductances control of breathing (Mulkey and Wenker 2011). Antibody
along the Müller cell surface allows redistribution of locally staining identified Kir6.1 in astrocytes of almost all brain
elevated K+, a process termed K+ siphoning (Newman 1986), regions, including the retina, preferentially in perisynaptic and
and similar spatial buffering of K+ has been suggested in the peridendritic processes (Thomzig et al. 2001), and Kir6-like
cortex (Holthoff and Witte 2000). Among the Kir subunits currents were recorded from Bergmann glia (Brockhaus and
identified in the CNS, Kir4.1 is special because of its selec- Deitmer 2000). Downregulation of astrocytic Kir currents
tive expression in glia where it is mainly localized to astroglial is commonly observed after injury or in the diseased CNS
processes wrapping synapses and blood vessels. In Müller (Olsen and Sontheimer 2008; Seifert et al. 2006).
cells, the strongly rectifying Kir2.1 subunit predominates in Astrocytes display prominent passive or background cur-
direct apposition to neuronal membranes, whereas the weakly rents largely lacking time- and voltage-dependency. Recent
rectifying Kir4.1 was identified in endfeet at the capillaries. data suggest that this current pattern might result from

P H YS I O L O GY O F A S T R O C Y T E S : I O N C H A N N E L S A N D I O N T R A N S P O RT E R S • 187
A B C
control 100 μM Ba2 I
Inorm 1
100

Membrane conductance
80

at –130 mV[%]
60
V[mV]
–160 –120 –70 –40 20 40
control –Ba2+ 20
–0.5
0
0.01 0.1 1 10 100
–1
1 nA Ba2+[μM]
20 ms

D E
(39)
Relative Frequency [%]

Kir4.1 5.1 2.1 2.2 2.3 bp 100


p3 p10 p21 p42 p60
612 80
495 170 kDa
60 (33) (26) (26) (26) 130 kDa Kir4.1
392 40
297 56 kDa
20 43 kDa β-actin
210 0
Kir4.1 Kir5.1 Kir2.1 Kir2.2 Kir2.3

Figure 16.2 Astrocytes Acutely Isolated from the CA1 Region of the Hippocampus Display Kir Channels. A. Membrane currents were elicited
between −160 and +20 mV (10-mV increments; holding potential, −70 mV; dashed line, zero current). After adding 100 μm Ba2+, currents were
reduced. Ba2+-sensitive currents were determined by subtracting respective current families (control—100 μM BaCl2). B. Mean I–V relationships of
Ba2+-sensitive currents (n = 9). In each cell, data were normalized to maximum inward current. C. Mean dose–inhibition curve of membrane conduc-
tance (n = 7). Half-maximal inhibition occurred at a Ba2+ concentration of 7.1 ± 3.2 μm, with a Hill coefficient of nH = 0.7 ± 0.1. D. After recording,
the cell content was harvested, and RT-PCR was performed. Kir4.1 was coexpressed with other Kir subunits, as shown in the agarose gel. Summary
of Kir detection in single astrocytes. E. Roentgen films of Western blot analysis of Kir4.1 tetramers (160 kDa) and β-actin (42 kDa). Protein content
of lysates prepared from whole hippocampi was analyzed at the ages indicated. The β-actin signal served as a loading control (60 μg of protein each).
Data in (B,C) are given as mean ± SD; cell numbers in parentheses. Reproduced with permission from Seifert et al. 2009.

superposition of currents through Kir and two-pore–domain conductances. However, before postnatal day 10, the current
K+ (K2P) channels. Similar to Kir, K2P channels are open at patterns often show voltage- and time-dependent current
rest and drive the membrane potential toward EK Channel components (probably because of coexpression of Kv chan-
activity is modulated by pH, polyunsaturated fatty acids, nels), indicating delayed upregulation of Kir4.1 and/or K2P
temperature, volatile anesthetics, phospholipids and mechani- channels during postnatal development (Seifert et al. 2009).
cal stress. Currently, 15 members of the K2P channel family
have been cloned that are active as dimers. Antibody staining 2.4 WAT E R C H A N N E L S
localized K2P3.1 (TASK1) and K2P10.1 (TREK2) protein in
astrocytes of different brain regions, including radial-glia like Water flux across cell membranes is mediated by homote-
precursors of the subventricular zone (Kindler et al. 2000; trameric transmembrane channels called aquaporins. Eleven
Pruss et al. 2011). In acutely isolated astrocytes from mouse aquaporins, AQP0 to AQP10, are known, of which AQP1,
hippocampus, small K2P2.1 (TREK1)-mediated currents AQP4 and AQP9 are expressed in the brain. AQP4 is pre-
have been identified with combined functional and transcript dominantly found in astrocytes and Müller cells where it is
analysis, which were sensitive to polyunsaturated fatty acids, primarily localized to the endfeet contacting blood vessels,
volatile anesthetics, and quinine (Seifert et al. 2009). The low the pial surface, and the vitreous body, respectively (Nagelhus
surface expression of K2P1.1 channels might result from its et al. 2004; Oshio et al. 2004). AQP4 colocalizes with Kir4.1
G-protein–mediated endosomal internalization (Feliciangeli and TRPV4 (Benfenati et al. 2011), and water flux follows
et al. 2010). Müller cells express pH- and bupivacaine-sensitive the osmotic gradient during activity-dependent K+ redistri-
K2P3.1 (TASK1) channels that may inhibit cellular swelling bution. These channels are involved in maintaining extracel-
under ischemic conditions (Skatchkov et al. 2006). How lular ion gradients and regulation of the extracellular and
K2P channels in astrocytes influence ion homeostasis and intracellular volume. In acute cortical slices, neuronal stimu-
neuron-glia signaling has still to be elucidated. lation generated a radial water flux that was mediated through
Patch-clamp recordings from astrocytes in hippocampal astrocytic AQP4 channels and facilitated by activation of
or cortical brain slices usually reveal linear current–voltage vasopressin-1a receptors expressed by astrocytes (Niermann
relationships, because of the predominating Kir4.1 and K2P et al. 2001). Knock-out or downregulation of AQP4 entailed

188 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
reduced water permeability of astrocytes (Nicchia et al. a similar time course and is predominantly localized to glial
2003; Solenov et al. 2002). Stimulation-induced increase membranes facing the pia mater and blood vessels (Wen et al.
and recovery of extracellular K+ concentration was slower 1999).
in AQP4–/– mice, probably attributable to an increase of
the extracellular space volume and impaired astrocytic K+
2.5 A N I O N C H A N N E L S
uptake (Binder et al. 2006). On the other hand, astrocytes in
AQP4–/– mice displayed enhanced gap junction coupling, Voltage-gated anion channels of the ClC-family function
which obviously counteracted the deficits in K+ buffering as homomeric or heteromeric dimers. Up to now, nine ClC
and may explain the relatively weak phenotype of AQP4–/– subunits have been cloned. ClC1–1,2, ClC-Ka, ClC-Kb,
mice (Strohschein et al. 2011). In the hippocampus, AQP4 and the β-subunit barttin are localized to the plasma mem-
is upregulated during the first 6 postnatal weeks (Hsu et al. brane, whereas ClC3–7 together with the β-subunit Ostm1
2011), mimicking the alterations seen for Kir4.1 in the same are expressed in membranes of intracellular organelles (endo-
subregion (Seifert et al. 2009). Maximal expression of AQP4 somes, lysosomes). These proteins function as chloride chan-
was observed in the CA1 stratum lacunosum-moleculare and nels or Cl–/H+ exchangers. The voltage-sensitive inwardly
stratum moleculare of the dentate gyrus (Fig. 16.3). These rectifying chloride channel ClC-2 is broadly expressed in
layers possess the highest density of microvessels in the hip- glial cells. These channels are activated after hyperpolariza-
pocampus, and thus identify them as potential sites of K+ and tion of the cell membrane or by hypotonic stress. In astrocytes
water regulation. AQP4 expression in the cerebellum follows of the hippocampus, ClC-2 is found in endfeet contacting
small blood vessels, primarily in strata oriens, pyramidale,
and lacunosum-moleculare and in the outer molecular layer
of the DG. The coincidence of ClC-2–positive astrocytes and
GABAergic presynaptic terminals suggests a role for astro-
A cytes in Cl– redistribution and an increase in efficacy of GABA
receptor–mediated inhibition (Sik et al. 2000). Detection
SO of ClC-2–mediated transmembrane currents in astrocytes
SP in situ is difficult because they are masked by the large rest-
ing membrane conductance of these cells (Kimelberg et al.
SR
2006; Makara et al. 2003). CLC-2 has also been localized
to Bergmann glial cells in the cerebellum (Blanz et al. 2007).
SLM
ClC-2–/– mice showed astrocyte activation and neurodegen-
eration in the hippocampus (Cortez et al. 2010).
ML Outwardly rectifying Cl– currents are activated when cells
are exposed to hypotonic solutions. These currents are medi-
DGC ated through volume-regulated anion channels (VRACs),
also termed volume-sensitive outwardly rectifying (VSOR)
H
50 μm anion channels. These channels are ubiquitously expressed by
virtually all cells and are permeable to small organic anions,
B such as the excitatory amino acids glutamate and aspartate.
*
80
*
* However, the molecular identity of VRACs is still elusive.
% difference in gray value

70 * Volume-regulated anion channels and amino acid release


* are inhibited by NPPB, DCPIB, tamoxifen, and phloretin.
relative to P9 SP

60
50 *
* * A physiological role of VRACs in astrocytes is thought to be
* *
40 * *
* the regulatory volume decrease (RVD) by extrusion of Cl–,
30 and osmo-active anions such as taurine, glutamate, and aspar-
20 *
tate. However, so far analyses have been performed on cultured
3w
6w

*
P9

10
0 cells, although there is no work to date on the existence of
SO SP SR SLM ML DGC Hilus VRACs on astrocytes in acute preparations (Kimelberg et al.
Figure 16.3 AQP4 Expression in the Hippocampus. A. AQP4 immuno- 2006). A recent in vivo study found some indirect evidence for
reactivity from a 6-week-old mouse hippocampus demonstrates laminar swelling-induced VRAC-mediated release of glutamate and
specificity. Although blood vessels are labeled well in all laminae, paren- aspartate from astrocytes in rat neocortex (Haskew-Layton
chymal AQP4 immunoreactivity is strongest in CA1 stratum lacunosum
moleculare (SLM), dentate gyrus molecular layer (ML), and along the
et al. 2008). Clearly, the experimental data available so far are
hippocampal fissure. Scale bar, 50 μm. DGC, dentate granule cell layer; insufficient to draw any sound conclusions as to the existence
H, hilus; SO, stratum oriens; SP, stratum pyramidale; SR, stratum radia- and physiological role of VRACs in astrocytes.
tum. B. Developmental and laminar-specific quantitation of hippocam- Ca2+ activated Cl– channels (CAACs) are activated on
pal AQP4 immunoreactivity. Values are expressed as percent difference increases in [Ca2+]i, display outward rectification and are sen-
in gray value compared with P9 stratum pyramidale (SP). P9, black bars;
3 week, red bars; 6 week, blue bars. Higher values indicate more intense
sitive to NPPB, Zn2+ and chlorotoxin. Astrocytes acutely iso-
immunoreactivity. **P < .01 and ***P < .001 compared with SP at same lated from mouse brain have recently been shown to express
time point. With permission from Hsu et al. 2011. functional CAACs encoded by bestrophin-1, a member of

P H YS I O L O GY O F A S T R O C Y T E S : I O N C H A N N E L S A N D I O N T R A N S P O RT E R S • 189
the bestrophin family. Activation of Gq/11-protein–coupled authors also reported TRPV4 immunoreactivity in astrocytes
receptors by thrombin, ATP, and bradykinin led to increases in of the superficial layer of the cerebral cortex, particularly in
[Ca2+]i and evoked large membrane currents that were inhib- astrocytic endfeet facing the pial surface and blood vessels.
ited by the Ca2+ chelator BAPTA or depletion of internal Ca2+ Together with AQP4, TRPV4 forms a complex that func-
stores by thapsigargin. 5-Nitro-2-(3-phenylpropylamino)- tions as an osmosensor because of the mechanosensitivity of
benzoic acid and niflumic acid blocked the thrombin-activated TRPV4. Actually, astrocytes respond to hypoosmotic swell-
currents, confirming their anion selectivity (Park et al. 2009). ing with [Ca2+]i elevations and initiate regulatory volume
Bestrophin-1 was identified on the mRNA and protein level decrease only if AQP4 is coexpressed (Benfenati et al. 2011).
in hippocampal astrocytes. In the cerebellum, bestrophin-1 The relatively high Ca2+ permeability of TRPV4 channels was
channels in Bergmann glial cells mediate release of GABA suggested to induce cell death through generation of reactive
and, thereby contribute to tonic inhibition. GABA release was oxygen species (Bai and Lipski 2010). The TRPV1 channels
even observed at resting [Ca2+]i (100 nM), but was profoundly seem also to be expressed by astrocytes (Toth et al. 2005) and
enhanced on [Ca2+]i elevation (Lee et al. 2010). These data add might be activated on acidification (Huang et al. 2010).
to the increasing evidence identifying astrocytes as important
partners of neurons in brain signaling.
3 I O N T R A N S P O RT E R S
2.6 T R A NS I E N T R E C E P TO R P OT E N T I A L
3.1 Na + -K + -AT Pase
CHANNELS
The maintenance of ion gradients across plasma membranes
The transient receptor potential (TRP) channel family is
is a prerequisite for the establishment of cellular membrane
subdivided into various subfamilies of which the canoni-
potentials, electrical signaling, and metabolite transport
cal (TRPC), vanilloid (TRPV), melastatin (TRPM), and
driven by ion gradients (Fig. 16.4). Here, the Na+/K+-ATPase,
ankyrin (TRPA) TRP subfamilies are found in the nervous
which exchanges intracellular Na+ for K+ with a stoichiometry
system. Transient receptor potential channels are cationic
of 3:2, is of fundamental importance. A wealth of secondary
channels that have 6 TM regions, similar to voltage-gated
and tertiary active transporters uses the inwardly directed Na+
ion channels, but TM4 lacks a voltage sensor. In astrocytes,
gradient to generate gradients for other ions such as K+, Mg2+,
Ca2+ release from internal stores activates transmembrane
H+, bicarbonate and, at least partly, Ca2+ and Cl–, and are thus
TRPC channels, to replenish Ca2+ deficits in the endoplasmic
dependent on proper function of the Na+/K+-ATPase. Na+/
reticulum (Venkatachalam and Montell 2007). This process
K+-ATPases are ubiquitous and are composed of α, β, and γ
is termed capacitive Ca2+ entry (CCE) or store operated Ca2+
subunits. In the brain, the α1 subunit is distributed in all cells,
entry (SOCE), and probably is mediated by heteromeric
whereas the ouabain-affine isoforms α2 and α3 subunits have
TRPC1, TRPC4, and TRPC5 channels. Glutamate recep-
tor activation or depletion of internal stores lead to [Ca2+]i
increase and Ca2+ oscillations in cultured astrocytes, which are
blocked by La3+ and RT-PCR identified transcripts encoding [Na+]e = 158 mM
[K+]e = 2.5 mM
TRPC1, 3, 4, and 6 in these cells (Pizzo et al. 2001). In cul- [Cl–]e = 125 mM
tured astrocytes, SOCE is confined to microdomains in the pHe = 7.2 is [H+]e = 63 nM
plasma membrane that are located in close proximity to the (blood: pH = 7.4 or 40 nM [H+]e )
endoplasmic reticulum and involve TRPC1, as shown by an [HCO3–]e = 17 mM (blood: 25 mM]
CO2 = 1.2 mM (5%)
antisense technique (Golovina 2005). TRPC1, TRPC4, and
TRPC5 protein expression has been detected in cultured and Extracellular
freshly isolated cortical astrocytes, and TRPC1-dependent
[Ca2+]i increase was shown to regulate the release of glutamate
from these cells (Malarkey et al. 2008). Long-term administra-
[K+]i = 120 mM
tion of fluoxetine reduced TRPC1-mediated SOCE in astro-
[Na+]i = 8 – 20 mM
cytes (Li et al. 2011). [Cl–]i = 20 – 40 mM
A structural analog of DAG, oleyl-acetyl-glycerol (OAG),
induces Ca2+ oscillations in cultured astrocytes, probably
through TRPC3, but here SOCE seems not to be involved pHi = 7.2 is [H+]i = 63 nM
(Grimaldi et al. 2003). Oleyl-acetyl-glycerol-sensitive TRP [HCO3–]i CO2 (5%) = 17 mM
CO2 = 1.2 mM (5 %)
channels have also been observed in freshly isolated astro-
cytes (Beskina et al. 2007). The latter study suggested that the
inflammatory cytokine IL-1β dysregulates astroglial Ca2+ sig- Intracellular
naling through an upregulation of TRPC6 channels.
The TRPV subfamily consists of six members. In cul-
tured astrocytes, TRPV4 was activated by the specific agonist Figure 16.4 Presumed Values of Intracellular Na+, K+, pH (H+), HCO3–,
4αPDD or hypotonic cell swelling, leading to [Ca2+]i increase and CO2 During Hypothetical Steady State Conditions in Astrocytes and
and transmembrane currents (Benfenati et al. 2007). The the Extracellular Space.

190 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
been found in astrocytes and neurons, respectively ( Juhaszova may occur simultaneously (Rose and Deitmer 1995a,b). Glial
and Blaustein 1997). The α subunit is required for the cata- cells have been ascribed a special role for acid-base regulation in
lytic and transport properties of the enzyme and contains the brain, in particular of the extracellular spaces.
binding sites for cations, ATP, and digitalis-like compounds Major pH regulating systems in astrocytes and other glial
like ouabain. One of the most prominent sources of [Na+]i cells are the Na+/H+ exchanger (NHE) and the Na+-HCO3–
increase in astrocytes is linked to the high-affinity uptake of co-transporter (NBC). The NHE extrudes H+ out of the cell
glutamate at excitatory synapses. Hence, rapid glutamate clear- at the expense of Na+ influx, is electroneutral and operates
ance is dependent on a large Na+ gradient across the astrocyte similarly as described for neurons and many other cell types.
membrane, which is provided by the Na+/K+-ATPase. A well- The NBC has been described in several types of macroglia,
documented consequence of glial Na+ elevations is increased including astrocytes, oligodendrocytes, and retinal Müller cells
ATP hydrolysis by the Na+/K+-ATPase, followed by increased (Deitmer and Chesler 2009), but may also be present in neu-
aerobic glycolysis, glucose uptake, and glycogen breakdown rons (Svichar et al. 2011). The NBC in astrocytes is electrogenic
(Barros and Deitmer 2010; Magistretti 2006). Thus, astro- and cotransports one Na+ with two HCO3– (Deitmer 2007).
cyte metabolism might be linked to neuronal activity via The reversal potential of NBCs lies near the resting potential,
the ATP-consuming Na+/K+-ATPase. Spread of intracellu- and can be adjusted to the actual membrane potential by driv-
lar Na+ from one hippocampal astrocyte to another was dis- ing NBC in the inward or outward direction. Activation of the
turbed by pharmacological inhibition of gap junctions, and NBC in the inward direction results in intracellular alkaliniza-
virtually omitted in Cx30/Cx43 double-deficient mice, and tion, rise in [Na+]i, and outward currents leading to a hyperpo-
Cx30/Cx43-mediated Na+ diffusion between astrocytes was larization of the membrane, whereas activation of the NBC in
speculated to represent a signal indicating increased metabolic the outward direction acidifies the cytosol, drives Na+ out of
needs, independent of concomitant Ca2+ (Langer et al. 2012). the cell, and depolarizes the cells. In addition, when HCO3– is
Cell volume responses of astrocytes in hypo-osmotic carried outwardly, buffering capacity is added to the extracel-
medium involve the net movement of osmoles, a mechanism lular spaces at the expense of the cell interior. In Müller cells,
that is energy dependent and tightly coupled to the Na+/K+- the NBC is localized in endfoot processes and has a stoichiom-
ATPase. It is noteworthy that Kir4.1, previously found to bind etry of 3 HCO3–: 1 Na+, suggesting a role in the transport of
MLC1, is also present in the macromolecular complex of the CO2/HCO3– from the retina to the vitreous humor (Newman,
Na+/K+-ATPase, thus indicating that the two proteins may 1996). The direction in which the NBC operates across the
be functionally coupled. Syntrophin and dystrobrevin might plasma membrane depends on pHi and pHo (and [HCO3–]i/o),
mediate the link between different components of the Na+/ [Na+]i, and the cell membrane potential. Depolarizations and
K+-ATPase transmembrane complexes (Brignone et al. 2011). hyperpolarizations are expected to result in an intraglial alka-
See sections 2.3 and 2.4 for discussion of the role of Kir4.1 in linization and acidification, respectively. The cotransporter
the control of K+ homeostasis. appears to have a remarkably high affinity for HCO3–, because
it can be active even in the nominal absence of CO2/HCO3–,
when [HCO3–]o is less than 0.3 mM, attributable to air-CO2
3.2 Na + /H + E XC H A N G E R A N D
(Brookes and Turner 1994). Expressed in frog oocytes, the
Na + /H C O 3 – C OT R A NSP O RT E R
NBC significantly contributes to the apparent cytosolic buffer
The intracellular, cytosolic pH (pHi) of neurons and glial cells capacity, which becomes voltage dependent and may enhance
is actively maintained at a value usually between 7.0 and 7.4, the efficacy of other acid-base transport systems (Becker and
which is similar to the extracellular pH value (pHo) in nervous Deitmer 2004). Carbonic anhydrase activity renders activ-
tissue (7.1–7.3). Thus, “resting” pHi and pHo range between 40 ity of NBCs significantly more efficient by forming a “trans-
and 100 nM free [H+]. This means that there is often only a port metabolon” (Becker and Deitmer 2007; Schueler et al.
small chemical gradient of [H+], if any, across the membranes 2011). In situ hybridization revealed NBC mRNA expression
of “resting” neurons and astrocytes. The main driving force of throughout the CNS, with particularly high levels in the olfac-
H+ from outside to inside of a cell is the negative membrane tory bulb, dentate gyrus, and cerebellum.
potential. Any conductance of the cell membrane for H+ and/ The anion Cl–/HCO3– exchanger (AE), which is expressed
or HCO3– at negative membrane potentials would result in in most cells, has not yet been studied in astrocytes, but it is
a gain of acid or loss of base, and hence lead to intracellular likely that astrocytes are also equipped with this membrane
acidification. Therefore, in living cells, pHi regulation requires carrier (Fig. 16.5). This carrier exchanges intracellular HCO3–
extrusion of acid or uptake of base. The link between CO2 with extracellular Cl–, and is required by most cells to recover
production and pH is the CO2/HCO3– buffer system, which from intracellular alkalosis. We have observed recovery from
plays a major role for the maintenance of both pHi and pHo. To alkalosis in cultured cortical astrocytes, which is affected by
understand the mechanisms underlying acid and alkaline tran- the presence of extracellular HCO3– (Theparambil, Alt, and
sients, it is often necessary to monitor pH in neurons, astrocytes, Deitmer, unpublished observations). A detailed study of this
and the extracellular spaces under the same conditions. Net process in astrocytes, however, is still lacking.
acid flux across astroglial and neuronal membranes may occur Rapid alkalosis has been observed in astrocytes of rat cortex
in opposite directions, in different cells, however. Thus, during following adjacent neuronal activity (Chesler and Kraig 1987).
neural activity, cytosolic alkalization of astrocytes, cytosolic Numerous studies have suggested that this response results
acidification of neurons, and a transient interstitial alkalization from activation of an electrogenic Na+-HCO3– cotransporter

P H YS I O L O GY O F A S T R O C Y T E S : I O N C H A N N E L S A N D I O N T R A N S P O RT E R S • 191
K+ Monocarboxylate transporters belong to a family of trans-
porters including multiple isoforms, which carry mono-
Na+ Na-K-P Na+ carboxylate anions, such as lactate, pyruvate, and ketone
bodies, in cotransport with an H+ in an electroneutral manner.
ATP
ADP
NHE Monocarboxylate transporter isoforms 1 and 4, as found in astro-
NCX Na+
cytes, can mediate both uptake and release of lactate or other
monocarboxylates. According to the lactate shuttle hypothesis,
Ca2+ H+ monocarboxylic acids are released by astrocytes via MCT1 and
Cl– 4, and taken up by neurons via neuronal MCT2, converted to
CA CO2 pyruvate, and consumed to generate ATP (Magistretti 2006)
KCC
K+
(see chapters 27 and 36). In synaptic domains, in which energy
HCO3– AE Cl– requirements are high, astroglial lactate may substitute or com-
plement the supply of glucose to synapses (Barros and Deitmer
K+ Na+ 2010). Possible functional relationships between some of the
NKCC
Cl–
NBC Na+ and H+/HCO3––coupled carriers, as discussed, with ion-
Ca2+ coupled metabolic substrate carriers are outlined in Figure 16.6.
ADP
ATP
Na+ In this scheme, both NBC and MCTs interact with intracellu-
Ca-P lar and extracellular carbonic anhydrases (CA), and support the
flow of lactate from astrocytes to neurons, as suggested by the
astrocyte-to-neuron lactate shuttle hypothesis.
Figure 16.5 Some of the Major Ion-Transporting Membrane Carriers of The neutral amino acid transporter SNAT3, which
Astrocytes Discussed in This Chapter. The Na+-K+ pump (Na-K-P) and the exchanges Na+ and H+ and the amino acid substrate cotrans-
plasmalemmal Ca2+ pump (Ca-P) require energy from ATP degradation. ported with Na+, has been shown to mediate glutamine trans-
Because of their stoichiometry, the Na-K-P, Ca-P, NCX, and NBC are
electrogenic, that is, they depolarize or hyperpolarize the cell membrane
port in astrocytes (Deitmer et al. 2003), and may be involved
when activated. See the text for further details. in the glutamate-glutamine cycle between neurons and astro-
cytes. During extrusion of glutamine via SNAT3, Na+ would
be carried out of the cell, which would require either a large
caused by glial depolarization, presumably attributable to an H+ gradient into, or a large glutamine gradient out of the cell.
increase in [K+]o, as was first described in giant glial cells of Astrocyte uptake of peptides (glycylsarcosine) is mediated
the leech (Rose and Deitmer 1994). Glutamate can induce by the high-affinity H+-peptide transporter PEPT2 (Wada et
intracellular acidosis via the glutamate uptake carrier, which is al. 2005). Because PEPT2 seemed to cooperate with NHE,
not only driven by the inward gradient of Na+, but also takes peptide transport was not only dependent on the transmem-
up one H+ per cycle. Glial pHi decreases of several tenths of brane H+ gradient, but also on [Na+] and modulated by NHE
pH units have been reported during exposure to glutamate or inhibitors. It remains to be studied, if PEPT2 activity is also
D-aspartate. As astrocytes convert glutamate to glutamine, a modulated by direct interaction with NHE1 and/or NHE2.
reaction catalyzed by the pH-sensitive glutamine synthetase,
this may in turn stimulate glutamine export from astrocytes 3.4 Ca 2+ -AT Pases A N D Na + /Ca 2+ E XCH A N G E R
using neutral amino acid transporters, which are coupled to
H+, such as SNAT3 and SNAT5. Hence, glutamate uptake Ca2+ signaling is based on the low [Ca2+]i (50–100 nM), and
and glutamine export by astrocytes, as described by the gluta- large Ca2+ concentration gradients between the cytosol and
mate/glutamine cycle hypothesis, can cause intraglial acidosis, the extracellular space and the lumen of intracellular organ-
and can modulate each other by concomitant pHi changes. elles, respectively (e.g., endoplasmic reticulum, with [Ca2+]
reaching up to a few mM). Cytosolic Ca2+ transients have been
identified to evoke transmitter release from astrocytes (see
3.3 AC I D -BA S E – C O U P L E D M ETA B O L I T E
chapters 17, 26, and 38). It is the interplay of the different cel-
T R A N S P O RT E R S
lular mechanisms (Ca2+ influx and release, Ca2+ extrusion and
H+ and HCO3– are coupled to a variety of metabolite carrier uptake, Ca2+ buffering) that control the [Ca2+li levels, which
systems in glial cells. These are either inorganic ion transport- is crucial for the generation and shape of Ca2+ signaling in
ers, such as NHE, anion exchanger (AE) or NBC, which astrocytes and other cell types. Mechanisms that are involved
are related to pH regulation (see the preceding), or might in regulating cytosolic Ca2+ contribute to shaping kinetics
be employed to drive acid-base–coupled metabolite trans- and amplitude of Ca2+ transients. Ca2+ binding proteins such
porters, such as the monocarboxylate transporter (MCT) or as parvalbumin, calcineurin, and calmodulin, which are pres-
some amino acid and peptide transporters. In addition, cou- ent in the cytoplasm, may serve to buffer cytosolic [Ca2+], and
pling of H+, OH–, and/or HCO3– renders pH a regulator of may also be involved in Ca2+-dependent signaling. Energy-
these membrane transporters (Becker and Deitmer 2004). consuming mechanisms are involved in Ca2+ clearance from
Although the Na+ gradient plays the dominant role for most the cytosol, either by transport out of the cell or sequestering
carrier systems, some transporters are driven by, or coupled to, Ca2+ into organelle stores, such as endoplasmic reticulum (ER),
the H+/HCO3– gradient. lysosomes, or mitochondria, in astrocytes as in most other cell

192 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Figure 16.6 Acid-Base–Coupled Transporters as They May Interact Between Astrocytes and Neurons. Intracellular H+ regulation and net CO2 flux
from neurons to astrocytes can drive HCO3– and Na+ via the NBC, and lactate/pyruvate via the monocarboxylate transporter MCT isoforms 1 and
4 out of astrocytes. Neurons could then take up these substrates via MCT2 and extrude H+ primarily via NHE. Note that some of the carriers may
interact with intracellular carbonic anhydrase II (CAII) and/or with extracellular carbonic anhydrase IV (CAIV).

types. Sarcoendoplasmic reticulum Ca2+-ATPases (SERCAs) 1 (NKCC1) is expressed in neurons, oligodendrocytes, and
pump Ca2+ ions into the endoplasmic reticulum. Plasma astrocytes. NKCC1 was found in astrocytes of the cortex,
membrane ATP-driven Ca2+ pump (PMCA) and Na+/Ca2+ cerebellum, hippocampus, and perivascular glia, presumably
exchangers (NCX) use ATP or the electrochemical gradient mostly in endfeet. In astrocytes, NKCC1, mainly driven by
of Na+ across the plasma membrane, respectively, to extrude the Na+ gradient, contributes to K+ uptake, Cl– accumula-
Ca2+ out of the cell. Finally, mitochondria and lysosomes are tion, cell swelling, and swelling-induced glutamate release, for
dynamic Ca2+ stores that can also sequester and release Ca2+ example, when [K+]o is increased (Su et al. 2000; Walz and
through the activity of the Ca2+ uniporter and/or channels, Hertz 1984). High-K+ stimulation of NKCC1 was reported
respectively. In cultured astrocytes, most of the Ca2+ influx to be Ca2+-dependent, and may affect the intracellular level
was reported to be mediated by Na+/Ca2+ exchange, which of other ions, which are dependent on the Na+ gradient, such
required tyrosine kinase to operate. The NCX is linked to the as Ca2+ and H+ (see the preceding). Expression of NKCC1 is
Na+ gradient and hence to the activity of the Na+/K+ pump, upregulated in cultured astrocytes with dibutyryl cAMP (Su
the α2 subunit of which might be involved in the regulation et al. 2000), and NKCC1 activity was found to be stimulated
of NCX and hence subcellular Ca2+ domains (Golovina et al. following phosphorylation during oxygen-glucose deprivation
2003a,b). In experimental ischemia, NCX was also suggested (OGD) (Lenart et al. 2004). Inhibition or genetic ablation of
to act in concert with the Cl–-coupled Na+-K+-cotransporter NKCC1 reduced OGD-mediated Na+ load, Cl– influx, and
(NKCC1) to cause perturbation of mitochondrial Ca2+ cell swelling. It has been suggested that NKCC1 may con-
homeostasis and dysfunction (Kintner et al. 2007). tribute to cerebral ischemic injury, as for example, causing cell
swelling and glutamate release, whereas inhibition of NKCC1
was found to be neuron-protective under these conditions.
3.5 Cl – -DE PE N DE N T Na + - A N D
Activation of NKCC1 is an important player in the process
K + - C OT R A NSP ORT E R S
of astrocyte swelling during brain edema under various neu-
Cation-Cl– cotransporters (CCC), encoded by the SLC12 rological conditions, and its contribution to cell swelling may
gene family, are ubiquitous and involved in the regulation of assist in the development of new therapeutic strategies for the
[Cl–]i. [Cl–]i has been determined with ion-selective micro- treatment of brain edema after ischemia, trauma, and acute
electrodes in cultured cerebral astrocytes to be between 20 liver failure (for review, see Jayakumar and Norenberg 2010).
and 40 mM, that is, higher than expected from electrochemi- The K+-Cl– cotransporter (KCC), encoded by four genes
cal equilibrium, resulting in a Nernst potential for Cl– of (KCC1–4), is electroneutral and, in contrast to NKCC, medi-
between –30 to –50 mV, which compared well with the rever- ates Cl– efflux from cells. In the brain, several KCC isoforms
sal potential of GABA-evoked membrane response of –52 mV have been reported, including the neuron-specific KCC2. In
(Kettenmann et al. 1987). Cation-Cl– cotransporter activity astrocytes and C6 glioma cells, KCl uptake has been studied,
is therefore important for GABAergic and glycinergic inhibi- but with variable Na+ dependence (Gagnon et al. 2007; Walz
tory neurotransmission and for cell volume homeostasis in and Hinks 1985). Thus, the differential contribution of Na+-
the developing and mature nervous system. NKCC, which dependent NKKC1 and Na+-independent KCC for cell swell-
cotransports Na+, K+ and Cl– into cells, has been identified in ing and Cl– regulation has been poorly studied in astrocytes,
a large variety of cell types. In the brain, the NKCC isoform and its physiological impact has still to be defined.

P H YS I O L O GY O F A S T R O C Y T E S : I O N C H A N N E L S A N D I O N T R A N S P O RT E R S • 193
4 S U M M A RY A N D P E R S P E C T I VE S Binder DK, Yao X, Zador Z, Sick TJ, Verkman AS, Manley GT. 2006.
Increased seizure duration and slowed potassium kinetics in mice
lacking aquaporin-4 water channels. Glia 53:631–636.
Astrocytes are equipped with a large number of plasma Blanz J, Schweizer M, Auberson M, Maier H, Muenscher A, Hubner
membrane ion channels and ion carriers. Research in recent CA, et al. 2007. Leukoencephalopathy upon disruption of the chlor-
years has concentrated on their functional aspects, and their ide channel ClC-2. J Neurosci 27:6581–6589.
regulation and modulation by intracellular and extracel- Bordey A, Sontheimer H. 1997. Postnatal development of ionic currents
lular messengers. Although K+ channels are subject to, and in rat hippocampal astrocytes in situ. J Neurophysiol 78:461–477.
Brignone MS, Lanciotti A, Macioce P, Macchia G, Gaetani M, Aloisi F,
important causes of, membrane potential changes, carriers are et al. 2011. The beta1 subunit of the Na,K-ATPase pump interacts
often related to the Na+ gradient across the cell membrane. with megalencephalic leucoencephalopathy with subcortical cysts
Although the chemical gradient of K+ and Na+ are both large protein 1 (MLC1) in brain astrocytes: new insights into MLC patho-
and opposite, the electrochemical gradient of K+ is usually genesis. Hum Mol Genet 20:90–103.
maintained small by maintaining a membrane potential close Bringmann A, Francke M, Pannicke T, Biedermann B, Kodal H, Faude
F, et al. 2000a. Role of glial K+ channels in ontogeny and gliosis: a
to the K+ equilibrium potential, primarily attributable to a hypothesis based upon studies on Muller cells. Glia 29:35–44.
relatively large conductance for K+. The large Ca2+ gradients Bringmann A, Pannicke T, Weick M, Biedermann B, Uhlmann S, Kohen L,
between the cytosol and the extracellular space and intracel- et al. 2002. Activation of P2Y receptors stimulates potassium and cation
lular Ca2+-storing organelles, respectively, are widely used for currents in acutely isolated human Muller (glial) cells. Glia 37:139–152.
signaling, including the modulation of some K+ channels. In Bringmann A, Schopf S, Reichenbach A. 2000b. Developmental regula-
tion of calcium channel-mediated currents in retinal glial (Muller)
the future, an important strategy will be to study functional
cells. J Neurophysiol 84:2975–2983.
relations between ion channels or transmembrane carriers Brockhaus J, Deitmer JW. 2000. Developmental downregulation of
in astrocytes and information processing, development, and ATP-sensitive potassium conductance in astrocytes in situ. Glia 32:
metabolism in the brain. 205–213.
Brookes N, Turner RJ. 1994. K+-induced alkalinization in mouse cerebral
astrocytes mediated by reversal of electrogenic Na+-HCO3- cotrans-
AC K N OW L E D G M E N T S port. Am J Physiol 267:C1633–C1640.
Chesler M, Kraig RP. 1987. Intracellular pH of astrocytes increases rap-
idly with cortical stimulation. Am J Physiol 253:R666–670.
The work of the authors is supported by Deutsche Forschungs- Chever O, Djukic B, McCarthy KD, Amzica F. 2010. Implication of
gemeinschaft (grants SFB/TR3 C1, C9, STE 552, DE 231) Kir4.1 channel in excess potassium clearance: an in vivo study on
and European Commission (FP7–202167 NeuroGLIA). The anesthetized glial-conditional Kir4.1 knock-out mice. J Neurosci
authors thank I. Nauroth for comments on the manuscript, 30:15769–15777.
and apologize to all those whose work could not be discussed Clark BA, Mobbs P. 1994. Voltage-gated currents in rabbit retinal astro-
cytes. Eur J Neurosci 6:1406–1414.
because of space constraints. Cortez MA, Li C, Whitehead SN, Dhani SU, D’Antonio C, Huan
LJ, et al. 2010. Disruption of ClC-2 expression is associated
with progressive neurodegeneration in aging mice. Neuroscience
REFERENCES 167:154–162.
Deitmer JW. 2007. Acid-base transport and pH regulation. In:
Akopian G, Kressin K, Derouiche A, Steinhäuser C. 1996. Identified Handbook of Neurochemistry and Molecular Neurobiology. Vol 5:
glial cells in the early postnatal mouse hippocampus display different Brain Energetics. Integration of Molecular and Cellular Processes
types of Ca2+ currents. Glia 17:181–194. (eds Gibson GE, Dienel GA) Springer-Verlag, Berlin, Heidelberg:
Bai JZ, Lipski J. 2010. Differential expression of TRPM2 and TRPV4 469–486.
channels and their potential role in oxidative stress-induced cell death Deitmer JW, Broer A, Broer S. 2003. Glutamine efflux from astrocytes is
in organotypic hippocampal culture. Neurotoxicology 31:204–214. mediated by multiple pathways. J Neurochem 87:127–135.
Barros LF, Deitmer JW. 2010. Glucose and lactate supply to the synapse. Deitmer JW, Chesler M. 2009. Neuron-glia pH regulation. In: New
Brain Res Rev 63:149–159. Encyclopedia of Neuroscience (ed. Squire LR), Academic Press,
Becker HM, Deitmer JW. 2004. Voltage dependence of H+ buffering Oxford, Vol. 6:739–747.
mediated by sodium bicarbonate cotransport expressed in Xenopus Edwards L, Nashmi R, Jones O, Backx P, Ackerley C, Becker L, et al.
oocytes. J Biol Chem 279:28057–28062. 2002. Upregulation of Kv 1.4 protein and gene expression after
Becker HM, Deitmer JW. 2007. Carbonic anhydrase II increases the chronic spinal cord injury. J Comp Neurol 443:154–167.
activity of the human electrogenic Na+//HCO3- cotransporter. J Biol Emmi A, Wenzel HJ, Schwartzkroin PA, Taglialatela M, Castaldo P,
Chem 282:13508–13521. Bianchi L, et al. 2000. Do glia have heart? Expression and functional
Benfenati V, Amiry-Moghaddam M, Caprini M, Mylonakou MN, role for ether-a-go-go currents in hippocampal astrocytes. J Neurosci
Rapisarda C, Ottersen OP, et al. 2007. Expression and functional char- 20:3915–3925.
acterization of transient receptor potential vanilloid-related channel Feliciangeli S, Tardy MP, Sandoz G, Chatelain FC, Warth R, Barhanin
4 (TRPV4) in rat cortical astrocytes. Neuroscience 148:876–892. J, et al. 2010. Potassium channel silencing by constitutive endocytosis
Benfenati V, Caprini M, Dovizio M, Mylonakou MN, Ferroni S, Ottersen and intracellular sequestration. J Biol Chem 285:4798–4805.
OP, et al. 2011. An aquaporin-4/transient receptor potential vanil- Filosa JA, Bonev AD, Straub SV, Meredith AL, Wilkerson MK, Aldrich
loid 4 (AQP4/TRPV4) complex is essential for cell-volume control in RW, et al. 2006. Local potassium signaling couples neuronal activity
astrocytes. Proc Natl Acad Sci U S A 108:2563–2568. to vasodilation in the brain. Nat Neurosci 9:1397–1403.
Bergles DE, Jabs R, Steinhauser C. 2010. Neuron-glia synapses in the Gagnon KB, Adragna NC, Fyffe RE, Lauf PK. 2007. Characterization
brain. Brain Res Rev 63:130–137. of glial cell K-Cl cotransport. Cell Physiol Biochem 20:121–130.
Beskina O, Miller A, Mazzocco-Spezzia A, Pulina MV, Golovina VA. Girouard H, Bonev AD, Hannah RM, Meredith A, Aldrich RW, Nelson
2007. Mechanisms of interleukin-1beta-induced Ca 2+ signals in MT. 2010. Astrocytic endfoot Ca2+ and BK channels determine
mouse cortical astrocytes: roles of store- and receptor-operated Ca2+ both arteriolar dilation and constriction. Proc Natl Acad Sci U S A
entry. Am J Physiol Cell Physiol 293:C1103–C1111. 107:3811–3816.

194 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Golovina V, Song H, James P, Lingrel J, Blaustein M. 2003a. Regulation Lenart B, Kintner DB, Shull GE, Sun D. 2004. Na-K-Cl
of Ca 2+ signaling by Na+ pump alpha-2 subunit expression. Ann N Y cotransporter-mediated intracellular Na+ accumulation affects Ca 2+
Acad Sci 986:509–513. signaling in astrocytes in an in vitro ischemic model. J Neurosci
Golovina VA. 2005. Visualization of localized store-operated calcium 24:9585–9597.
entry in mouse astrocytes. Close proximity to the endoplasmic retic- Li B, Dong L, Fu H, Wang B, Hertz L, Peng L. 2011. Effects of chronic
ulum. J Physiol 564:737–749. treatment with fluoxetine on receptor-stimulated increase of [Ca 2+]i
Golovina VA, Song H, James PF, Lingrel JB, Blaustein MP. 2003b. Na+ in astrocytes mimic those of acute inhibition of TRPC1 channel
pump alpha 2-subunit expression modulates Ca 2+ signaling. Am J activity. Cell Calcium 50:42–53.
Physiol 284:C475–C486. Longden TA, Dunn KM, Draheim HJ, Nelson MT, Weston AH,
Grimaldi M, Maratos M, Verma A. 2003. Transient receptor potential Edwards G. 2011. Intermediate-conductance calcium-activated
channel activation causes a novel form of [Ca 2+]i oscillations and potassium channels participate in neurovascular coupling. Br J
is not involved in capacitative Ca 2+ entry in glial cells. J Neurosci Pharmacol 164:922–933.
23:4737–4745. MacFarlane SN, Sontheimer H. 2000. Modulation of Kv1.5 currents by
Haskew-Layton RE, Rudkouskaya A, Jin Y, Feustel PJ, Kimelberg Src tyrosine phosphorylation: potential role in the differentiation of
HK, Mongin AA. 2008. Two distinct modes of hypoosmotic astrocytes. J Neurosci 20:5245–5253.
medium-induced release of excitatory amino acids and taurine in the Magistretti PJ. 2006. Neuron-glia metabolic coupling and plasticity.
rat brain in vivo. PLoS ONE 3:e3543. J Exp Biol 209:2304–2311.
Hibino H, Fujita A, Iwai K, Yamada M, Kurachi Y. 2004. Differential Makara JK, Rappert A, Matthias K, Steinhäuser C, Spat A, Kettenmann
assembly of inwardly rectifying K+ channel subunits, Kir4.1 and H. 2003. Astrocytes from mouse brain slices express ClC-2-mediated
Kir5.1, in brain astrocytes. J Biol Chem 279:44065–44073. Cl- currents regulated during development and after injury. Mol Cell
Higashimori H, Blanco VM, Tuniki VR, Falck JR, Filosa JA. 2010. Neurosci 23:521–530.
Role of epoxyeicosatrienoic acids as autocrine metabolites in Malarkey EB, Ni Y, Parpura V. 2008. Ca 2+ entry through TRPC1 chan-
glutamate-mediated K+ signaling in perivascular astrocytes. Am J nels contributes to intracellular Ca2+ dynamics and consequent gluta-
Physiol 299:C1068–C1078. mate release from rat astrocytes. Glia 56:821–835.
Holthoff K, Witte OW. 2000. Directed spatial potassium redistribution Mulkey DK, Wenker IC. 2011. Astrocyte chemoreceptors: mechanisms
in rat neocortex. Glia 29:288–292. of H+ sensing by astrocytes in the retrotrapezoid nucleus and their
Hsu MS, Seldin M, Lee DJ, Seifert G, Steinhäuser C, Binder DK. 2011. possible contribution to respiratory drive. Exp Physiol 96:400–406.
Laminar-specific and developmental expression of aquaporin-4 in the Müller T, Fritschy JM, Grosche J, Pratt GD, Möhler H, Kettenmann
mouse hippocampus. Neuroscience 178:21–32. H. 1994. Developmental regulation of voltage-gated K+ channel and
Huang C, Hu ZL, Wu WN, Yu DF, Xiong Q J, Song JR, et al. 2010. GABA A receptor expression in Bergmann glial cells. J Neurosci 14:
Existence and distinction of acid-evoked currents in rat astrocytes. 2503–2514.
Glia 58: 1415–1424. Nagelhus EA, Mathiisen TM, Ottersen OP. 2004. Aquaporin-4 in the
Jayakumar AR, Norenberg MD. 2010. The Na-K-Cl co-transporter in central nervous system: cellular and subcellular distribution and
astrocyte swelling. Metab Brain Dis 25:31–38. coexpression with KIR4.1. Neuroscience 129:905–913.
Juhaszova M, Blaustein MP. 1997. Na+ pump low and high ouabain affin- Newman EA. 1985. Voltage-dependent calcium and potassium channels
ity alpha subunit isoforms are differently distributed in cells. Proc in retinal glial cells. Nature 317:809–811.
Natl Acad Sci U S A 94:1800–1805. Newman EA. 1986. High potassium conductance in astrocyte endfeet.
Kafitz KW, Meier SD, Stephan J, Rose CR. 2008. Developmental pro- Science 233:453–454.
fi le and properties of sulforhodamine 101-Labeled glial cells in acute Newman EA. 1996. Acid efflux from retinal glial cells generated by
brain slices of rat hippocampus. J Neurosci Methods 169:84–92. sodium bicarbonate cotransport. J Neurosci 16:159–168.
Kettenmann H, Backus KH, Schachner M. 1987. g-Aminobutyric acid Nicchia GP, Frigeri A, Liuzzi GM, Svelto M. 2003. Inhibition of
opens Cl-channels in cultured astrocytes. Brain Res 404:1–9. aquaporin-4 expression in astrocytes by RNAi determines alteration
Kimelberg HK, MacVicar BA, Sontheimer H. 2006. Anion chan- in cell morphology, growth, and water transport and induces changes
nels in astrocytes: biophysics, pharmacology, and function. Glia in ischemia-related genes. FASEB J 17:1508–1510.
54:747–757. Niermann H, Amiry-Moghaddam M, Holthoff K, Witte OW,
Kindler CH, Pietruck C, Yost CS, Sampson ER, Gray AT. 2000. Ottersen OP. 2001. A novel role of vasopressin in the brain: modu-
Localization of the tandem pore domain K+ channel TASK-1 in the lation of activity-dependent water flux in the neocortex. J Neurosci
rat central nervous system. Mol Brain Res 80:99–108. 21:3045–3051.
Kintner DB, Luo J, Gerdts J, Ballard AJ, Shull GE, Sun D. 2007. Role Olsen M, Sontheimer H. 2008. Functional implications for Kir4.1
of Na+-K+-Cl- cotransport and Na+/Ca 2+ exchange in mitochondrial channels in glial biology: from K+ buffering to cell differentiation.
dysfunction in astrocytes following in vitro ischemia. Am J Physiol J Neurochem 107:589–601.
292:C1113–C1122. Olsen ML, Campbell SL, Sontheimer H. 2007. Differential distribution
Kofuji P, Biedermann B, Siddharthan V, Raap M, Iandiev I, Milenkovic of Kir4.1 in spinal cord astrocytes suggests regional differences in K+
I, et al. 2002. Kir potassium channel subunit expression in reti- homeostasis. J Neurophysiol 98:786–793.
nal glial cells: implications for spatial potassium buffering. Glia Orkand RK, Nicholls JG, Kuffler SW. 1966. Effect of nerve impulses on
39:292–303. the membrane potential of glial cells in the central nervous system of
Kofuji P, Ceelen P, Zahs KR, Surbeck LW, Lester HA, Newman EA. amphibia. J Neurophysiol 29:788–806.
2000. Genetic inactivation of an inwardly rectifying potassium chan- Oshio K, Binder DK, Yang B, Schecter S, Verkman AS, Manley GT.
nel (Kir4.1 subunit) in mice: phenotypic impact in retina. J Neurosci 2004. Expression of aquaporin water channels in mouse spinal cord.
20:5733–5740. Neuroscience 127:685–693.
Kressin K, Kuprijanova E, Jabs R, Seifert G, Steinhäuser C. 1995. Pannicke T, Bringmann A, Reichenbach A. 2002. Electrophysiological
Developmental regulation of Na+ and K+ conductances in glial cells characterization of retinal Müller glial cells from mouse during post-
of mouse hippocampal brain slices. Glia 15:173–187. natal development: comparison with rabbit cells. Glia 38:268–272.
Langer J, Stephan J, Theis M, Rose CR. 2012. Gap junctions mediate Park H, Oh SJ, Han KS, Woo DH, Park H, Mannaioni G, et al. 2009.
intercellular spread of sodium between hippocampal astrocytes in Bestrophin-1 encodes for the Ca 2+-activated anion channel in hip-
situ. Glia 60:239–252. pocampal astrocytes. J Neurosci 29:13063–13073.
Lee S, Yoon BE, Berglund K, Oh SJ, Park H, Shin HS, et al. 2010. Pizzo P, Burgo A, Pozzan T, Fasolato C. 2001. Role of capacitative cal-
Channel-mediated tonic GABA release from glia. Science 330: cium entry on glutamate-induced calcium influx in type-I rat cortical
790–796. astrocytes. J Neurochem 79:98–109.

P H YS I O L O GY O F A S T R O C Y T E S : I O N C H A N N E L S A N D I O N T R A N S P O RT E R S • 195
Price DL, Ludwig JW, Mi H, Schwarz TL, Ellisman MH. 2002. Strohschein S, Hüttmann K, Gabriel S, Binder DK, Heinemann U,
Distribution of rSlo Ca 2+-activated K+ channels in rat astrocyte Steinhäuser C. 2011. Impact of aquaporin-4 channels on K+ buff-
perivascular endfeet. Brain Res 956:183–193. ering and gap junction coupling in the hippocampus. Glia 59:
Pruss H, Dewes M, Derst C, Fernandez-Klett F, Veh RW, Priller J. 2011. 973–980.
Potassium channel expression in adult murine neural progenitor Su G, Haworth RA, Dempsey RJ, Sun D. 2000. Regulation of Na+-
cells. Neuroscience 180:19–29. K+-Cl- cotransporter in primary astrocytes by dibutyryl cAMP and
Rose CR, Deitmer JW. 1994. Evidence that glial cells modulate extracel- high [K+]o. Am J Physiol 279:C1710-C1721.
lular pH transients induced by neuronal activity in the leech central Svichar N, Esquenazi S, Chen HY, Chesler M. 2011. Preemptive regula-
nervous system. J Physiol (London) 481:1–5. tion of intracellular pH in hippocampal neurons by a dual mecha-
Rose CR, Deitmer JW. 1995a. Stimulus-evoked changes of extra- and nism of depolarization-induced alkalinization. J Neurosci 31:
intracellular pH in the leech central nervous system. I. Bicarbonate 6997–7004.
dependence. J Neurophysiol 73:125–131. Thomzig A, Wenzel M, Karschin C, Eaton MJ, Skatchkov SN, Karschin
Rose CR, Deitmer JW. 1995b. Stimulus-evoked changes of extra- and A, et al. 2001. Kir6.1 is the principal pore-forming subunit of astro-
intracellular pH in the leech central nervous system. II. Mechanisms cyte but not neuronal plasma membrane K-ATP channels. Mol Cell
and maintenance of pH homeostasis. J Neurophysiol 73:132–140. Neurosci 18:671–690.
Roy ML, Saal D, Perney T, Sontheimer H, Waxman SG, Kaczmarek LK. Tippens AL, Pare JF, Langwieser N, Moosmang S, Milner TA, Smith
1996. Manipulation of the delayed rectifier Kv1.5 potassium channel Y, et al. 2008. Ultrastructural evidence for pre- and postsynaptic
in glial cells by antisense oligodeoxynucleotides. Glia 18:177–184. localization of Cav1.2 L-type Ca 2+ channels in the rat hippocampus.
Schaller KL, Caldwell JH. 2003. Expression and distribution of J Comp Neurol 506:569–583.
voltage-gated sodium channels in the cerebellum. Cerebellum 2: 2–9. Toth A, Boczan J, Kedei N, Lizanecz E, Bagi Z, Papp Z, et al. 2005.
Schueler C, Becker HM, McKenna R, Deitmer JW. 2011. Transport Expression and distribution of vanilloid receptor 1 (TRPV1) in the
activity of the sodium bicarbonate cotransporter NBCe1 is enhanced adult rat brain. Mol Brain Res 135:162–168.
by different isoforms of carbonic anhydrase. PLoS ONE 6:e27167. Venkatachalam K, Montell C. 2007. TRP channels. Annu Rev Biochem
Seidel KN, Derst C, Salzmann M, Holtje M, Priller J, Markgraf R, et al. 76:387–417.
2011. Expression of the voltage- and Ca 2+-dependent BK potassium Verkhratsky A, Steinhäuser C. 2000. Ion channels in glial cells. Brain
channel subunits BKbeta1 and BKbeta4 in rodent astrocytes. Glia Res Rev 32:380–412.
59:893–902. Wada M, Miyakawa S, Shimada A, Okada N, Yamamoto A, Fujita T.
Seifert G, Hüttmann K, Binder DK, Hartmann C, Wyczynski A, 2005. Functional linkage of H+/peptide transporter PEPT2 and
Neusch C, et al. 2009. Analysis of astroglial K+ channel expression in Na+/H+ exchanger in primary cultures of astrocytes from mouse cere-
the developing hippocampus reveals a predominant role of the Kir4.1 bral cortex. Brain Res 1044:33–41.
subunit. J Neurosci 29:7474–7488. Walz W, Hertz L. 1984. Intense furosemide-sensitive potassium accu-
Seifert G, Schilling K, Steinhäuser C. 2006. Astrocyte dysfunction in mulation in astrocytes in the presence of pathologically high extracel-
neurological disorders: a molecular perspective. Nat Rev Neurosci lular potassium levels. J Cereb Blood Flow Metab 4:301–304.
7:194–206. Walz W, Hinks EC. 1985. Carrier-mediated KCl accumulation accom-
Sik A, Smith RL, Freund TF. 2000. Distribution of chloride panied by water movements is involved in the control of physiological
channel-2-immunoreactive neuronal and astrocytic processes in the K+ levels by astrocytes. Brain Res 343:44–51.
hippocampus. Neuroscience 101:51–65. Wen H, Nagelhus EA, Amiry-Moghaddam M, Agre P, Ottersen OP,
Skatchkov SN, Eaton MJ, Shuba YM, Kucheryavykh YV, Derst C, Veh Nielsen S. 1999. Ontogeny of water transport in rat brain: post-
RW, et al. 2006. Tandem-pore domain potassium channels are func- natal expression of the aquaporin-4 water channel. Eur J Neurosci
tionally expressed in retinal (Muller) glial cells. Glia 53: 266–276. 11:935–945.
Solenov EI, Vetrivel L, Oshio K, Manley GT, Verkman AS. 2002. Zhou M, Kimelberg HK. 2000. Freshly isolated astrocytes from rat hip-
Optical measurement of swelling and water transport in spinal cord pocampus show two distinct current patterns and different [K+]o
slices from aquaporin null mice. J Neurosci Methods 113:85–90. uptake capabilities. J Neurophysiol 84:2746–2757.

196 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
17.
RELEASE OF GLIOTRANSMITTER S AND TRANSMITTER
RECEPTOR S IN ASTROCY TES
Helmut Kettenmann and Robert Zorec

A B B R E VI AT I O N S was considered an exclusive property of neurons. This view was


challenged by a series of studies on cell culture demonstrating
Ach acetylcholine that astrocytes responded to the neurotransmitters glutamate
AMPA D-amino-3-hydroxy-5-methyl-4-isoxazole- and γ-aminobutyric acid (GABA) with membrane depolariza-
propionic acid tion. Although astrocytes remove neurotransmitters from the
ANP atrial natriuretic peptide extracellular medium, they are also able to release neuroactive
ANP.emd emerald green fluorescent protein ANP.emd substances such as glutamate. This led to the concept that the
ATP adenosine triphosphate synapse consists of three elements: the presynaptic and post-
BDNF brain-derived neurotrophic factor synaptic neuronal compartments and the astrocyte compart-
cAMP cyclic adenosine monophosphate ment, first called the synaptic triad (Kettenmann et al. 1996)
cGMP cyclic guanosine monophosphate and subsequently the tripartite synapse (Araque et al. 1999).
c-NP C-type natriuretic peptide
CNS central nervous system
DMCM methyl-6,7-dimethoxy-4-ethylbeta- 2 M O D E S O F G L I OT R A N S M I T T E R
carboline-3-carboxylate RELEASE
eNOS endothelial nitric oxide synthase
GABA γ-aminobutyric acid Several mechanisms of gliotransmitter release appear to coexist
GAD glutamic acid decarboxylase in a single astrocyte (Fig. 17.1A) (Hamilton and Attwell 2010).
GDNF glial-derived neurotrophic factor In addition to the vesicle-based mechanisms, non–vesicle-based
GFAP glial fibrillary acidic protein release is mediated through channels, including volume-regulated
5HT 5-hydroxytryptamine anion channels, connexon/pannexon hemichannels; ionotropic
IL-6 interleukin 6 purinergic receptors; and reversal uptake of membrane transport-
mACh muscarinic ACh ers (Parpura and Zorec 2010). Although all of these mechanisms
MAP mitogen-activated protein require a concentration gradient along which chemical messen-
mGluR metabotropic glutamate receptor gers are transported to their targets, the vesicle-based mechanism,
NMDA N-methyl-d-aspartate currently actively debated (Smith 2010), is advantageous for cells.
NP natriuretic peptide First, relatively small structures (e.g., secretory vesicle), in
NPR natriuretic peptide receptor which chemical messengers can be stored, are more economi-
PCR polymerase chain reaction cal. Transport of molecules across the membrane against a
SNARE soluble N-ethyl maleimide-sensitive fusion concentration gradient, such as l-glutamate entering a vesi-
protein attachment protein receptor cle, consumes energy in the form of adenosine triphosphate
TCA tricarboxylic acid (ATP). It was estimated that the budget for recycling 4,000
TNF tumor necrosis factor glutamate molecules into a vesicle consists of 11,000 ATP mol-
TRH thyrotropin-releasing hormone ecules (Attwell and Laughlin 2001). In contrast, the budget
UV ultraviolet for concentrating glutamate into the whole cytosol must be
VAMP vesicle-associated membrane protein much higher, because the volume of a vesicle (a sphere 50 nm
VGLUT vesicular glutamate transporter in diameter) is at least nine orders of magnitude smaller than
VIAAT vesicular inhibitory amino acid transporter the volume of a cell (a sphere 15 μm in diameter).
VIP vasoactive intestinal peptide Second, secretory vesicles represent movable compart-
VNUT vesicular nucleotide transporter ments that can be placed in sites within the cell, where a high
concentration gradient of chemical messengers is required for
diffusional delivery to the targets. Vesicles can be considered as
1 INTRODUCTION signal driving modules, which can be strategically positioned
within the cell. Such regulation cannot be attained with the
Until about 30 years ago, the release of transmitters and the nonvesicle-based modes of messenger release unless these are
expression of neurotransmitter receptors in the nervous system integrated into the plasma membrane by targeted exocytosis.
197
A may be caused by the slow delivery of vesicles to the plasma
membrane and/or distinct (slow) vesicle–plasma membrane
Ca2+-dependent fusion mechanisms (see stages of exocytosis, Fig. 17.1B). The
channel/vesicle Ca2+-dependent increase in Cm was sensitive to tetanus toxin,
Glutamate indicating a SNARE-based vesicular mechanism; however, it
transporters
is unclear which vesicles contribute to the Cm increases.
3 Na+ + H+ As for neurotransmitters, the criteria to define a chemi-
ca2+ cal as a gliotransmitter have been reviewed (Parpura and
K+
Cystine
Glutamate Verkhratsky 2011; Parpura and Zorec 2010) and include:
(1) synthesis by and/or (vesicular) storage in glia; (2) regu-
Cell swelling lated release triggered by physiological and/or pathological
stimuli; (3) activation of paracrine or autocrine responses; and
(4) a role in (patho)physiological processes.
Volume-sensitive
organic anion channel

B
hemifusion fluctuations of full fusion
4 G L I OT R A N S M I T T E R S
a narrow fusion pore
A B C
4.1 G LU TA M AT E A S G L I OT R A NS M IT T E R
In astrocytes, glutamate is synthesized de novo as a byprod-
uct of the tricarboxylic acid (TCA) cycle, involving the
Figure 17.1 A. Mechanisms of glutamate release from astrocytes. astrocyte-specific enzyme pyruvate carboxylase. Glutamate is
Glutamate may be released in a calcium-dependent manner in response
to receptor-mediated increases in cytosolic calcium. Some studies indi-
converted from the TCA cycle intermediate, α-ketoglutarate,
cate that glutamate is released by exocytosis, by channel-mediated efflux usually via transamination of another amino acid, aspartate.
of cytosolic glutamate, through volume-sensitive channels, by a reversal All three known isoforms of vesicular glutamate transporters
of membrane glutamate transport mainly under pathological conditions. (VGLUTs) 1, 2, and 3, which use the proton gradient created
B. Stages vesicles undergo to accomplish the regulated release of chemical by vacuolar (V)-ATPases to package glutamate into vesicles,
messengers by regulated exocytosis. This process is thought to begin with
the vesicle delivery to the plasma membrane, followed by the formation
have been detected in astrocytes and stimuli that increase
of a hemi-fusion stalk (transition A), an intermediate structure con- astrocytic [Ca2+]i all evoked release of glutamate (Parpura and
necting the outer leaflets of fusing membranes. Hemi-fusion stalk then Verkhratsky 2011; Parpura and Zorec 2010).
proceeds into a fusion pore (transition B), an aqueous channel connect- Vesicle-based Ca2+-dependent release of transmitters
ing a spherical vesicle and the plasma membrane, through which cargo requires the presence of an exocytotic secretory machinery. As
molecules diffuse from the vesicle lumen into the cell exterior. Fusion
pore can reversibly vary its diameter (transitions C). During the inter-
neurons, astrocytes express the proteins of the soluble N-ethyl
mediate state (C), which is energetically stable, the fusion pore diameter maleimide-sensitive fusion protein attachment protein recep-
may be too narrow to permeate luminal cargo molecules and therefore tor (SNARE) complex: synaptobrevin 2 (Sb2), also referred to
this state could be considered release incompetent. Following the delivery as vesicle-associated membrane protein 2 (VAMP 2); syntaxin
of a stimulus, vesicles in state C may transit into a state with a wider 1, synaptosome-associated protein of 23 kDa (SNAP-23), as
fusion pore diameter, leading into a state of full fusion. (A) Modified
from Nedergaard et al. 2002; (B) modified from Jorgačevski et al. 2010.
well as several ancillary proteins to this complex, includ-
ing synaptotagmin 4 (Montana et al. 2006; Parpura and
Verkhratsky 2011). The use of clostridial, tetanus, and various
3 R E GU L AT E D E XO C Y TO S I S I N types of botulinum toxins, which cleave exocytotic SNARE
A S T R O C Y T E S I S S L OW E R T H A N proteins, caused a reduction of Ca2+-dependent glutamate
IN NEURONS release in astrocytes (Montana et al. 2006; Parpura and Zorec
2010). Similarly, the expression of mutated synaptotagmin 4,
The properties of vesicle-based release mechanisms have been acting in a dominant-negative manner, caused the reduction
studied at the cellular level by monitoring changes in the of Ca2+-dependent glutamate release from astrocytes (Zhang
plasma membrane area; merging the vesicle membrane with et al. 2004a). Often the question is asked whether glutamate
the plasma membrane affects the plasma membrane area, release from astrocytes occurs in vivo under normal conditions.
which can be monitored by measuring membrane capacitance To carry out experiments with the resolution obtained in cul-
(Cm), a parameter linearly related to the membrane area (Kreft tured cells also at the level of tissue slices or even in the whole
et al. 2004) (Fig. 17.2). In cultured astrocytes, increase in cyto- brain would be difficult, if not impossible, because glutamate
solic calcium ([Ca2+]i) by photolysis of caged calcium elicited and ATP are rather common metabolites and identifying the
an increase in Cm. Half-maximal response in Cm increase exhib- cellular source and mechanism of release with subcellular reso-
ited a similar Ca2+-sensitivity to that in neurons. However, lution would be technically most challenging.
the kinetics of this response in astrocytes was at least two Astrocytic secretory vesicles are essential morphological
orders of magnitude slower compared with the rate of regu- elements for regulated Ca2+-dependent exocytosis, and range
lated exocytosis in neurons (see Fig. 17.2A) (Kreft et al. 2003, from 30 to more than 1,000 nm in diameter; they exhibit clear
2004). The relatively slow responsiveness of glial exocytosis and dense cores (Parpura et al. 2010).

198 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
A B

–kexot
C = A(1–e )

10 μM
[Ca2+]

1 pF
1s 500 fF
Cm

k ~ 2 s–1
G 5 nS
kf ~ 400 s–1

1s
C= A1(1–e–k1exot )+A2(1–e–k2exot )

Figure 17.2 Comparison of time-dependent changes in membrane capacitance (Cm) recorded in a neuronal cell (A, photoreceptor) and an astrocyte (B).
A. Two types of Ca2+-induced increases in Cm in a photoreceptor have been recorded. Top trace was best fitted by a single exponential function (dot-
ted line). The bottom trace was best fitted to a sum of two exponential functions as shown by the equation below the horizontal line. The fastest rate
constant (kf ) was around 400 s–1. B. Top trace shows time-dependent changes in cytosolic calcium activity [Ca2+]i, elicited by ultraviolet (UV) light
flash photolysis of caged Ca2+ compound dialyzed into the cytosol of the cell (Kreft et al. 2004). UV flash was applied at the time indicated by the
arrow. Note that the rapid increase in [Ca2+]i following the UV flash application induced an exponential increase in Cm with a rate constant (k) of 2 s–1.
G denotes the real part of the admittance trace. (A) Modified from Kreft et al. 2003; (B) modified from Kreft et al. 2004.

In addition to the vesicle-based mechanisms, nonvesicle of the glutamate receptors. In Bergmann glia, an electro-
mechanisms of glutamate release are also present in astro- genic GABA efflux has been reported, activating neighbor-
cytes (Hamilton and Attwell 2010). Functional hemichannels ing GABA receptors. This release was considered because of
were confirmed by passage of extracellular Lucifer Yellow into reversed GABA transport via GAT-1, leading to a tonic eleva-
astrocytes in divalent cation-free solution and the ability to tion of GABA in the extracellular space and thereby to tonic
block this passage with gap junction blocking agents (Ye et al. inhibition (Barakat and Bordey 2002). A similar observation
2003). Orellana et al. (2011) studied the release of ATP and was made in the olfactory bulb, in which spontaneous slow
glutamate from connexion 43 hemichannels, which mediate synaptic currents were observed in mitral cells. These currents
neuronal death under inflammatory conditions. were mediated by GABA and depended on astrocyte activity
because they disappeared when astrocytes were silenced. The
GABA release from astrocytes in the olfactory bulb serves to
4.2 G A BA A S G L I OT R A N S M I T T E R
synchronize neuronal activity. These slow GABA-mediated
Recent studies indicate that astrocytes can create tonic inhi- transient currents were also observed in hippocampal pyrami-
bition by the release of GABA. Astrocytes can accumulate, dal neurons. Astrocytes can also control the frequency of min-
synthesize and release GABA (Angulo et al. 2008; Vélez-Fort iature inhibitory postsynaptic currents in pyramidal neurons.
et al. 2011). Astrocytes have the capacity to accumulate GABA Stimulation of astrocytes led to an increase in these currents,
as shown with anti-GABA antibodies in the brain stem or cere- and potentially this could be mediated by GABA release from
bellum. Astrocytes express transporters for GABA as reviewed astrocytes (Kang et al. 1998). In the barrel cortex, astrocyte
in chapter 35. GABA can be produced by two distinct path- activity resulted in a tonic- and stimulation-induced inhibi-
ways: by conversion of glutamate to GABA by the enzyme glu- tion of pyramidal cells and spiny stellate cells. Silencing astro-
tamic acid decarboxylase (GAD), or by the monoacetylation cytes by dialysis with BAPTA increased the excitability of the
of putrescine. Glutamic acid decarboxylase expression in glial neurons (Benedetti et al. 2011).
cells, however, is significantly lower than in neurons. A novel mechanism for GABA release has recently been
Gallo et al. (1986) demonstrated that GABA can be identified in the cerebellum. Astrocytes chronically release
released from astrocytes. The release could be triggered by acti- GABA through an anion channel, bestrophin-1, and thereby
vating glutamate receptors, and they used the release of GABA mediate tonic inhibition. Bestrophine-1 can be modulated
from the astrocytes as a tool to characterize the pharmacology by changes in intracellular Ca2+ and cell volume, but is even

R E L E A S E O F G L I OT R A N S M I T T E R S A N D T R A N S M I T T E R R E C E P TO R S I N A S T R O C Y T E S • 199
tonically active at resting Ca2+ levels. The permeability of Atrial natriuretic peptide (ANP) is expressed in astrocytes
GABA through the channel is considerable, about four and plays a role in cerebral blood flow regulation (McKenzie
times that of Cl–. The importance for bestrophine to main- et al. 2001) and as an autocrine regulator, because all natriu-
tain tonic inhibition was demonstrated by downregulating retic peptide receptors are expressed by astrocytes. To study
bestrophin-1 by an shRNA approach. This defect could be the discharge of ANP, Krzan et al. (2003) transfected astro-
rescued by selectively expressing bestrophin in (glial fibrillary cytes with a construct to express pro-ANP fused with
acidic protein [GFAP]-positive) astrocytes (Lee et al. 2010). the emerald green fluorescent protein (ANP.emd). The num-
Although a significant body of data shows that GABA ber of fluorescent ANP.emd puncta was reduced on stimulat-
uses non–vesicle-based modes of release, there is evidence that ing the cells by ionomycin, which strictly depended on the
the vesicular inhibitory amino acid transporter (VIAAT) is extracellular Ca2+. Vesicles containing ANP also appear to
expressed in astrocytes (Echigo and Moriyama 2004), indicat- contain ATP (Pangrsic et al. 2007), which is consistent with
ing that GABA may be stored in vesicles. However, unequivo- the report that ATP is stored in secretorgranin II–containing
cal evidence to support regulated exocytosis of GABA from vesicles (Coco et al. 2003). ATP and peptides could be
astrocytes is yet to be provided. differentially released from a single vesicle by regulating
fusion-pore diameter (Stenovec et al. 2004). The mobil-
ity of recycling ANP vesicles was one order of magnitude
4 .3 d -S E R I N E A S G L I OT R A N S M I T T E R
slower than the ANP vesicles trafficking to the plasma mem-
Astrocytes can also release d-serine (Schell et al. 1995), brane sites (Potokar et al. 2005, 2007) and was reduced on
a ligand to the glycine modulatory binding site of the stimulation of cells (Potokar et al. 2008, 2010), differing from
N-methyl-d-aspartate (NMDA) receptor. d-Serine is con- the stimulation-increased mobility of anti-VGLUT1 anti-
verted from l-serine by the action of serine racemase, an body capturing vesicles in astrocytes (Stenovec et al. 2007).
enzyme found in astrocytes (Rosenberg et al. 2010; Wolosker This indicates that vesicles with different cargo may traffic by
et al. 1999). Astrocytes are thought to be the key metabolic distinct mechanisms.
provider of l-serine, which is shuttled to neurons, likely via Recycling vesicles may not only carry lumenal cargo but
membrane transporters, for d-serine production in neurons also membrane-associated signaling molecules which partici-
(Wolosker 2011). In addition to the non–vesicle-based mecha- pate in contact cell–cell interactions (Kopan and Ilagan 2009;
nisms of release, astrocytes release d-serine by regulated exo- Soos et al. 1998) or in determining the density of plasma
cytosis in an activity-dependent manner (Henneberger et al. membrane transporters (Robinson 2002), such as the gluta-
2010; Oliet and Mothet 2006). mate transporter EAAT2 (Stenovec et al. 2008).
Mothet et al. (2005) investigated the mechanism of d-ser- Recycling vesicles may serve bidirectional communica-
ine release using an enzyme-linked assay to measure extracel- tion between neurons and glia. When studying the activity-
lular d-serine concentration and established that astrocytes dependent secretion of brain-derived neurotrophic factor
release d-serine in a Ca2+-dependent manner; extracellular Ca2+ (BDNF) and its extracellular availability, Bergami et al. (2008)
was required; it was reduced by concanamycin A, a V-ATPase provided evidence that BDNF, which is synthesized de novo
inhibitor, and tetanus toxin, implicating the involvement of in neurons, is secreted in its pro-form into the extracellular
a vesicular mechanism. How d-serine is loaded into vesicles medium, and is taken up via endocytosis into astrocytes, where
remains an open question. it is processed and released by a SNARE-dependent secretory
pathway.
4.4 P E P T I D E S A S G L I OT R A N S M I T T E R
4.5 AT P A S G L I OT R A NS M IT T E R
Unlike amino acids, which can get loaded into vesicles via
membrane transporters, peptides enter vesicles via the syn- Intracellular ATP is produced via glycolysis and oxidative
thetic secretory pathway. These peptides are typically made phosphorylation to reach more than 10 mM in the cytosol,
as pro-peptides in the endoplasmic reticulum, transit Golgi establishing a concentration gradient favoring the exit of ATP
compartments, where they are concentrated and sorted into from cells. Once released into the extracellular space, ATP can
organelles; then, they are processed in vesicles to their final be used in intercellular signaling, acting directly on purinergic
form before release. The classic view holds that vesicles car- receptors. Alternatively, on hydrolysis by membrane-bound
rying peptidergic transmitters seem to have relatively larger ecto-nucleotidases, the extracellular degradation products
diameters compared with the synaptic-like vesicles, and ADP and adenosine can activate different plasma membrane
exhibit electron-dense cores (dense-core vesicles). A diam- receptors (Fields and Burnstock 2006).
eter of approximately 100 nm was reported for secretogranin ATP can be released via non-vesicle and vesicle-based
II–positive dense-core vesicles (Calegari et al. 1999). This mechanisms (Parpura and Zorec 2010). Several lines of evi-
study also reported that astrocytes contain fewer smaller and dence support the latter mechanism. ATP seems to be present
less dense secretory granules containing secretogranin II, indi- in peptide-containing vesicles and in lysosomes (Hamilton
cating that peptidergic vesicles in astrocytes are not uniform and Attwell 2010; Parpura and Zorec 2010). ATP is loaded
in morphological appearance. Neuropetide Y was shown to into vesicles by SLC17A9 (Sawada et al. 2008), a vesicular
be contained in vesicles distinct from synaptic-like vesicles in nucleotide transporter (VNUT) that appears to be present in
astrocytes (Ramamoorthy and Whim 2008). astrocytes (Larsson et al. 2011).

200 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
To study ATP release from astrocytes, Pangrsic et al. spinal cord, brainstem, cerebellum, optic nerve, nucleus ruber,
(2007) used glutamate stimulation of astrocytes and showed and retina. In the hippocampus, AMPA receptor expression
quantal release of ATP as recorded by ATP reporter cells. is restricted to the NG2 cells; the classic astrocyte (with high
The exocytotic release of ATP stored in astrocytic lysosomes GFAP promoter activity) is characterized by high glutamate
could be detected only with unphysiologically long stimula- transporter activity (Matthias et al. 2003).
tion (Zhang et al. 2007), exhibiting different sensitivity com- Although in most brain regions astrocyte AMPA receptors
pared with other types of vesicles (Liu et al. 2011), indicating display low Ca2+ permeability, Bergmann glial cells in the cere-
that distinct exocytotic mechanisms control release from vesi- bellum lack the GluR2 subunit and hence allow passage of diva-
cle subtypes. Consistent with this, only lysosomes carrying the lent cations (Müller et al. 1992). The physiological relevance
VAMP7/TI-VAMP seem to be fusion competent (Verderio of this subunit combination has been demonstrated by experi-
et al. 2011). Distinct vesicle fusion mechanisms are consistent mentally inducing the expression of GluR2 in Bergmann glial
with distinct mobility properties of astrocytic vesicles, which cells, which results in significant neuronal rearrangement (Iino
are in part determined by the intermediate filament cytoskel- et al. 2001). The pharmacological properties and single chan-
eton (Potokar et al. 2011). nel conductances of the AMPA receptors of the hippocampal
NG2 cells mimic those of their neuronal counterparts. In the
hippocampus, fast application techniques, Ca2+ imaging, and
5 T R A N S M I T T E R R E C E P TO R S single cell reverse transcription (RT)–polymerase chain reac-
tion (PCR) revealed that the astroglial receptors possess an
intermediate Ca2+ permeability and are primarily assembled
5.1 I O N OT RO P I C G LU TA M AT E R E C E P TO R S
from the subunits GluR1, GluR2, and GluR4. Molecular and
About 28 years ago, two groups provided the first evidence that functional changes in astrocyte receptor expression occur in
not only neurons but also astrocytes in cell culture express func- epilepsy, and these alterations have been proposed to contrib-
tional glutamate receptors (Fig. 17.3) (Bowman and Kimelberg ute to the generation and spread of seizure activity in human
1984; Kettenmann et al. 1984). Cultured astrocytes express temporal lobe epilepsy (Seifert and Steinhäuser 2011). After
2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl)propanoic acid ischemic insult to the adult striatum, a novel astrocyte subtype
(AMPA) (assembled from subunits GluR1–GluR4) and kain- appears with distinct properties: Cells with high GFAP pro-
ate receptors (GluR5–GluR7, KA1, KA2), but do not respond moter activity express physiological properties of NG2 cells,
to NMDA. Functional AMPA receptors have been described namely, voltage-gated channels, complex responses to kainate,
in astrocytes in situ in various brain regions, including cortex, and a high rate of gap junction coupling (Wang et al. 2008).

Presynaptic terminal

Astrocyte Astrocyte

Glu Glu
Glu
Gln Gln Glu
Glu
Gln
Gln
ER
ER Glu Ca2+
Ca2+ Glu
Glu
Glu

Postsynaptic neurone

Metabotropic Glutamate av
GluR transporter
AMPA, NMDA Glu Glutamate
ionotropic GluRs Gln Glutamine

Figure 17.3 Glutamate-Mediated Neuronal–Glial Signaling. Synaptically released glutamate activates glial ionotropic (AMPA and NMDA) and
metabotropic receptors. Activation of group I metabotropic receptors initiates phospholipase C–dependent synthesis of InsP3, which in turn triggers
Ca2+ release from the endoplasmic reticulum (ER) Ca2+ store. The majority (~80%) of glutamate released during synaptic transmission is taken up by
astroglial Na+/glutamate transporters; subsequently, glutamate is converted into glutamine, which is transported back to neurons, where it acts as a
main source of newly synthesized glutamate (“glutamate–glutamine shuttle”). From Verkhratsky and Kirchhoff 2007.

R E L E A S E O F G L I OT R A N S M I T T E R S A N D T R A N S M I T T E R R E C E P TO R S I N A S T R O C Y T E S • 201
Although antibody staining and in situ hybridization exhibit a voltage-dependent block. There is also evidence
demonstrated widespread distribution of kainate receptors that the NG2 cell population in the hippocampus expresses
in brain, clear evidence for functional expression of these NMDA receptors (Serrano et al. 2008).
receptors in astrocytes is lacking. Recently it has been estab-
lished that astrocytes in situ can express functional NMDA
5.2 M ETA B OT RO P I C G LU TA M AT E R EC E P TO R S
receptors in contrast to cultured astrocytes (Fig. 17.4)
(Verkhratsky and Kirchhoff 2007). Responses of astrocytes The metabotropic glutamate receptor (mGluR) family con-
to NMDA have been reported for astrocytes in the spinal sists of eight members, which couple to G-proteins. mGluR3
cord and cortex and Bergmann glial cells of the cerebel- and mGluR5 are the predominant subtypes expressed by
lum. Bergmann glial cells express mRNAs for NR2B, and a astrocytes and have been described in cultured cells and in situ
physiological study on acute brain slices reported NMDA- (D’Antoni et al. 2008); these subtypes are also found in human
induced membrane currents in Bergmann glial cells. The hippocampal astrocytes. mGluR1 has been reported in hip-
responses were best studied in cortical astrocytes in acute pocampal astrocytes and the spinal cord. Activation of these
slices or in freshly isolated cells (Lalo et al. 2006; Schipke receptors led to an increase in intracellular Ca2+ and inhibition
et al. 2001). In contrast to neurons, the responses could be of cAMP accumulation, although Gs-coupled cAMP stimula-
recorded at a physiologic Mg2+ concentration of 1.3 mM. tion has also been reported. Stimulation of astroglial mGluRs
Only at concentrations of 4 mM and higher did the channels leads to intracellular Ca2+ oscillations and Ca2+ wave propaga-
tion within the astrocyte network, activates Ca2+-dependent
K+ channels, and induces prostaglandin-mediated glutamate
release from astrocytes that activates neuronal receptors
A (Bezzi et al. 1998). These responses are likely to occur under
physiological conditions because astroglial mGluR activa-
EGFP
tion and subsequent Ca2+ responses could be evoked by elec-
trical stimulation of fiber tracts, causing neuronal glutamate
release in acute brain slices. Activation of mGluRs induced
other astrocyte responses, including swelling, activation of
phospholipase D and glutamine synthetase, release of arachi-
0.1 nA donic acid, cAMP-dependent block of K+ currents, modula-
50 s
tion of proliferation, and regulation of the expression of the
PDC,CNQX glutamate transporter GLAST. Dramatic changes in mGluR
NMDA expression occur under pathophysiological conditions, and in
spinal cord injury, epilepsy, and amyotrophic lateral sclerosis.
B Because group I mGluRs induce glial Ca2+ oscillations and
Ca2+ wave propagation and influence neuronal excitability
5F/Fo (see the preceding), their upregulation as observed in epilepsy
might indicate an astroglial involvement in seizure generation
or spread (Seifert and Steinhäuser 2011; Seifert et al. 2006).

5.3 G A BA A R EC E P TO R S
Ionotropic GABAA receptors are expressed by astrocytes
CNQX, TTX, Cd2+ both in culture and in situ (Vélez-Fort et al. 2011). As in neu-
NMDA
rons, astrocytic GABAA receptors form Cl– channels with a
conductance of about 30 pS (Fig. 17.5). In contrast to mature
neurons, GABAA receptor activation leads to a large depo-
0.1 nA larization in astrocytes studied in culture (Kettenmann et al.
30 s 1984). The Cl– reversal potential can be as positive as –40
mV, indicating that GABA can lead to a substantial depo-
Figure 17.4 NMDA receptors. A. Astrocytes in the cortex respond to larization from the normal resting membrane potential of
NMDA. Astrocytes were identified in cortical slices using a transgenic about –80 mV. In astrocytes of the pituitary gland, postsyn-
mouse line that expressed the green fluorescent protein under the
control of the GFAP promoter (left). NMDA was applied (100 μM)
aptic potentials were activated by neuronally released GABA
to the same cell in the presence of 50 μM PDC and 50 μM CNQX. B. (Mudrick-Donnon et al. 1993). GABAA receptor activation
NMDA triggered larger Ca2+ responses at distal processes. On the left in astrocytes also triggers a long-lasting blockade of K+ con-
is a micrograph of an astrocyte filled with Fluo-4. Note the recording ductances, thereby augmenting depolarization, for example,
pipette approaching the cell from top. Fluorescence changes (F/Fo) were in Bergmann glial cells (Müller et al. 1994) and activating
analyzed in the areas demarked by the squares and are displayed in the
top three traces. The lower trace is the simultaneously recorded current
voltage-gated Ca2+ channels with subsequent increase in
response. NMDA (100 μM) and CNQX, TTX, Cd2+ were applied as cytosolic Ca2+ (in freshly isolated astrocytes from hippocam-
indicated by bars. From Schipke et al. 2001. pus) (Fraser et al. 1995).

202 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
A 50 μM GABA B
Astrocyte in culture
Astrocyte
20 μM GABA 20 μM GABA + 10 μM Diazepam
100 pA
1 sec

50 μM GABA 0.2 nA
1 sec

1 pA Bergmann glial cell in situ


1 sec

Vm, mV
70
50 pA
50 15 s

30 GABA GABA
0 Diazepam
–30

–50
0.25 nA
–70 GABA
30 s
1 pA
50 msec GABA
Pentobarbital

Figure 17.5 GABA receptors. A. Single GABAA receptor channel recording from astrocytes. The upper panel shows single channel activity in response
to application of GABA. Below is single channel activity displayed at different membrane potentials ranging from +70 to –70 mV. B. GABA receptor
pharmacology in cultured astrocytes and Bergmann glial cells in situ. The upper trace shows the GABA response from a cultured astrocyte that is
enhanced by coapplication of diazepam. Below is a response from a Bergmann glial cell in an acutely isolated cerebellar brain slice. Note that diazepam
did not significantly change the GABA-induced current. In contrast, pentobarbital significantly increased the GABA response. From Bormann et al.
1988; Müller et al. 1994.

The GABAA receptor has a complex pharmacology. As reported in these cells: Immunolabeling preferentially identi-
in neurons, barbiturates and steroids enhance the astrocytic fied receptors along the processes. There is evidence from both
GABA response. There is a difference between the responses cell culture and in situ studies that GABAA receptor activation
in neurons and cultured astrocytes. Although typical benzo- promotes differentiation of astrocytes. In culture, GABA trig-
diazepines such as diazepam enhanced the GABA response gers formation of processes and leads to astrocytes with a more
in both neurons and cultured astrocytes, the inverse benzo- complex morphological shape.
diazepine agonist, DMCM (methyl-6,7-dimethoxy-4-ethyl-
beta-carboline-3-carboxylate), also enhanced the response in
5.4 G A BA B R EC E P TO R S
astrocytes indicating a different subunit composition of the
GABAA receptor in astrocytes. In contrast, Bergmann glial GABAB receptors in astrocytes play an important role for sens-
cells studied in cerebellar slices showed GABA responses that ing the activity of interneurons. In the hippocampus, interneu-
were insensitive to benzodiazepine due to the expression of ronal firing elicits a GABAB-receptor–mediated increase of
δ subunits (see Fig. 17.5) (Müller et al. 1994). In freshly iso- calcium in surrounding astrocytes, which in turn potentiates
lated astrocytes from the hippocampus, the benzodiazepine inhibitory postsynaptic currents in pyramidal neurons (Kang
sensitivity was as described for neurons (Fraser et al. 1995). et al. 1998). It also mediates heterosynaptic depression by acti-
This indicates that GABAA receptor subunit composition is vating GABAB receptors in the astrocytes resulting in release
heterogeneous in astrocytes, resulting in different populations of ATP (Serrano et al. 2006). In the hippocampus, astrocytic
of astrocytes with respect to their GABAA receptor profile. In GABAB receptors are part of a functional circuit involv-
Bergmann glial cells, the GABAA receptors were prominently ing inhibitory neurons, astrocytes, and pyramidal neurons.
expressed in immature cells, but were downregulated in the All three subtypes of GABAB receptors (GABAB1a, GABAB1b,
mature cerebellum. In addition, an uneven distribution was and GABAB2) are expressed by astrocytes as revealed by

R E L E A S E O F G L I OT R A N S M I T T E R S A N D T R A N S M I T T E R R E C E P TO R S I N A S T R O C Y T E S • 203
immunohistochemistry. At the ultrastructural level, GABAB astrocytes promote neurite outgrowth mediated by acetylcho-
receptor subunits were expressed on astrocytic processes sur- line-mediated signaling. Exposure of astrocytes to carbachol,
rounding symmetrical and asymmetrical synapses in the CA1 a muscarinic agonist, increased the expression of the extra-
subregion of the hippocampus (Charles et al. 2003). Functional cellular matrix proteins fibronectin and laminin-1 indicating
GABAB receptor expression changes during development. that muscarinic stimulation of astrocytes induces the release
Only 10% of astrocytes in hippocampal slices responded at P3 of permissive factors that accelerate neuronal development
or P32–34 to the application of the GABAB receptor agonist (Guizzetti et al. 2008).
baclofen, but 60% of the astrocytes responded between P11
and P15, indicating that GABAB receptor–mediated calcium
5.7 OX Y TO C I N A N D VA S O P R E S S I N R EC E P TO R S
signaling in astrocytes occurs preferentially during postnatal
development when hippocampal networks are established Vasopressin and oxytocin can trigger Ca2+ responses in cultured
(Meier et al. 2008). astrocytes, indicating the presence of their receptors (Panatier
2009). Neuronal production and release of TGF-β led to an
increase in astroglial oxytocin receptor mRNA in astrocytes,
5.5 G LYC I N E R E C E P TO R S
indicating that neurons were able to upregulate the astrocyte
Functional strychnine-sensitive glycine receptors have been receptor (Mittaud et al. 2002). Astrocytes isolated from the
detected in astrocytes and glial precursor cells (most likely hippocampus predominantly express vasopressin1b receptors,
NG2 cells) in spinal cord (Pastor et al. 1995). A combined but astrocytes isolated from the cerebral cortex of neonatal
patch clamp and RT-PCR approach revealed the expression rats solely expressed vasopressin1a receptors. Activation of
of α1 and β subunits (Kirchhoff et al. 1996). Glycine receptor both receptors triggers glutamate release from the astrocytes
expression is not a common property of astrocytes: Neither (Syed et al. 2007).
cultured astrocytes nor Bergmann glial cells express functional
glycine receptors.
5.8 VA S OAC T I VE I N T E S T I NA L
In the supraoptic nucleus, clusters of glycine receptors are
P O LY P E P T I D E R EC E P TO R S
associated with glial fibrillary acidic protein positive astroglial
processes. This finding suggests that the astrocytic glycine Vasoactive intestinal polypeptide (VIP) receptors are
receptors could mediate a paracrine communication between expressed by astrocytes and activation leads to Ca2+ signaling
astrocytes and neurons in this brain region (Deleuze et al. (Masmoudi Kouki et al. 2007). Expression of the VIP recep-
2005). tor, VPAC2, increases in astrocytes after injury (Nishimoto
et al. 2011). Vasoactive intestinal polypeptide receptor acti-
vation induces the release of interleukin 6 (IL-6) and neu-
5.6 AC ET Y L C H O L I N E R E C E P TO R S
rotrophic factors, and stimulates proliferation. Vasoactive
There is evidence for the expression of the α7 subunit of nico- intestinal polypeptide receptor activation stimulates glutamate
tinic acetylcholine receptors in astrocytes in culture and in uptake into astrocytes by upregulation of GLT-1 and GLAST
situ. In cultured astrocytes, α-bungarotoxin–sensitive nico- expression (Figiel and Engele 2000), indicating that VIP can
tinic acetylcholine receptors containing the α7 subunit led increase the strength of glutamate-mediated neurotransmis-
to Ca2+ influx through the receptor channels and Ca2+-induced sion. In addition, VIP receptors may play an important role in
Ca2+ release from caffeine-sensitive stores (Sharma and regulating energy metabolism. Vasoactive intestinal polypep-
Vijayaraghavan 2001). This subunit was also localized to tide depletes astrocyte glycogen initially, followed by delayed
astrocytes of human cerebellum in situ (Graham et al. 2002). reaccumulation to a level beyond baseline. These effects are
In the prefrontal cortex, electron microscopic immuno- mediated by regulating a number of related genes such as
labeling of α7 nicotinic acetylcholine receptor revealed glycogen synthase via the transcription factor family C/EBP.
expression on astrocyte processes, which were frequently However, there is not yet convincing evidence of the presence
located close to terminals colocalized with vesicular acetyl- of VIP receptors in astrocytes in situ.
choline transporters.
The expression of muscarinic ACh (mACh) receptors by
5.9 A D R E N E RG I C R EC E P TO R S
astrocytes is well established (Porter and McCarthy 1997;
Verkhratsky et al. 1998). There is evidence for the expression Both α- and β-adrenergic receptors have been described in
of transcripts for the mACh receptor subtypes, M1 to M5, in astrocytes in culture and in situ (Porter and McCarthy 1997;
cultured astrocytes. Acetylcholine induces IP3 formation and Verkhratsky et al. 1998). α1 Receptors affect astrocyte func-
release of Ca2+ from cytoplasmic stores, inhibits adenylate tion by inhibiting gap junction coupling via PLC. This is in
cyclase, and induces an increase in astroglial proliferation. line with the finding that mechanically induced Ca2+ waves in
Neuronal activity can stimulate astrocytic mACh receptors as hippocampal astroglial cells are inhibited after stimulation of
shown in the hippocampus (Araque et al. 2002). In the barrel α1 receptors (Muyderman et al. 1998). Moreover, α1b receptor
cortex, muscarinic acetylcholine receptor–dependent synap- activation stimulated glutamate uptake in cultured astrocytes.
tic plasticity depends on astrocyte Ca2+ signaling. This in vivo There is also a hint that metabolic activity may be controlled
study suggests a role of astrocytes as a gate for cholinergic plas- by α receptors because receptor activation induced lactate for-
ticity in the cortex (Takata et al. 2011). During development, mation and glycogen turnover (see chapters 27 and 36). In the

204 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
hippocampus, α2 receptors are located on astroglial processes, the sst2A splice variant (Viollet et al. 1997). Receptor activa-
near terminals forming asymmetrical excitatory synapses tion is linked to inhibition of cAMP accumulation, leading
(Milner et al. 1998). Neuronal stimulation in acute cerebellar to enhanced proliferation rates and a reduction in forskolin-
slices led to the release of noradrenaline from afferent fibers induced IL-6 release.
and activation of α1 receptors in Bergmann glial cells (Kulik
et al. 1999). 5.12 S E ROTO N I N R EC E P TO R S
Both subtypes of β receptors, β1 and β2, are expressed by
astrocytes in situ; β2 are most prominently expressed under RT-PCR in human and rat brain cultures revealed that astro-
pathological conditions (Laureys et al. 2010). In the injured cytes can express various serotonin receptors (Azmitia 2001;
brain, adrenergic receptors are differentially regulated. Verkhratsky et al. 1998). Distinct expression of 5HT subtypes
Although α1 receptors decrease in areas of neuronal degen- also occurs in situ because 5HT1A, 5HT2A, and 5HT5A sub-
eration and gliosis, β receptors are upregulated. Blockade of types have been identified in different brain areas. In astrocytes
β receptors suppressed glial scar formation, indicating that of the brainstem, 5-HT triggers Ca2+ responses (Härtel et al.
adrenergic receptors are part of the cascade leading to astro- 2009). 5HT receptors have been speculated to be involved in
cyte activation (Griffith and Sutin 1996). This suggestion is pathogenesis. The 5HT5A subtype is upregulated in gliosis and
substantiated by the finding that β2 receptors are activated or the 5HT2A subtype is enhanced in schizophrenia. Stimulation
upregulated in the transected optic nerve in vivo, confirm- of 5HT receptors leads to the release of S100β from astro-
ing that β-adrenergic signaling is an important feature of the cytes, and this release was hypothesized to have an impact on
astrocyte response to injury. Further support comes from the the ongoing pathological event. 5HT5A transcripts are devel-
observation that stimulation of β receptors leads to astroglio- opmentally regulated: Receptor mRNA is detected before
sis and cell proliferation in the optic nerve in vivo. This effect birth and expression peaks at postnatal day 20 in the rat.
might be mediated via the control of growth factor expres-
sion (e.g., FGF1, FGF2, BDNF, CNTF). Stimulation of β1
5.13 NAT R IU R ET I C P E P T I D E R EC E P TO R S
receptors is also accompanied by enhanced glycogen levels,
and stimulation of β2 and β3 adrenoceptors increases glucose All three subtypes, ANP, brain natriuretic peptide, and C-type
uptake in astrocytes (Catus et al. 2011) and increases cytoso- natriuretic peptide (c-NP) are present in the brain (Potter
lic glucose concentration in astrocytes (Prebil et al. 2011). The et al. 2006). Natriuretic peptides exert their actions by bind-
observation that β adrenergic receptor stimulation leads to a ing to natriuretic peptide receptors (NPRs): ANP binds pref-
cAMP-mediated inhibition of astroglial inwardly rectifying erentially to NPR-A, and brain natriuretic peptide and c-NP
K+ channels (Roy and Sontheimer 1995) points to a role in to NPR-B (Lucas et al. 2000). All NPs bind with equal affinity
the regulation of extracellular K+ homeostasis. to NPR-C (Lucas et al. 2000). NPR-A and NPR-B are par-
ticulate, cell membrane–bound guanylyl cyclase receptors that
mediate the signal by increasing cyclic guanosine monophos-
5.10 A N G I OT E NS I N R E C E P TO R S phate (cGMP). NPR-C, earlier ascribed the role of a clearance
Expression of angiotensin receptors in astrocytes in situ receptor removing the peptides to the periphery, does not pos-
seems to be restricted to white matter (Sumners et al. 1994; sess guanylyl cyclase activity (Potter et al. 2006; Rose and Giles
Verkhratsky et al. 1998). Antibody staining identified AT1 and 2008) and appears to have a complex signaling mechanism of
AT2 receptors in astrocytes of white matter tracts in cerebel- its own. The consequence of NPR-C activation is inhibition
lum and subcortical regions, in the optic nerve and in the cor- of adenylyl cyclase (cAMP) via guanine nucleotide inhibitory
pus callosum; no immunoreactivity was found in gray matter. proteins (Gi proteins) (Potter et al. 2006). However, NPR-C
The diversity in expression is also reflected when astrocytes is also coupled to endothelial nitric oxide synthase (eNOS)–
are harvested from different brain areas: AT1 receptors were dependent signal transduction, in which NO activates a solu-
found in astrocytes from medulla oblongata and cerebellum, ble form of guanylate cyclase leading to production of cGMP
whereas astrocyte cultures from hypothalamus and cortex (Murthy et al. 1998). In astrocytes, NPs have been reported to
did not express functional receptors. Receptor activation as elicit downstream signals by interacting with NPR-A (Borán
studied in culture-stimulated proliferation, increased glucose and García 2007; Prado et al. 2010) or NPR-B (Zielińska et al.
uptake and induced prostaglandin release. Moreover, astro- 2007). The presence of NPR-C in astrocytes was identified on
cytes regulate leukocyte infiltration into the central nervous the basis of ligand displacement analysis (Sumners and Tang
system (CNS) mediated by angiotensin II signaling involving 1992) and its functional role by showing that agonists of this
AT1 receptors (Füchtbauer et al. 2011). receptor attenuate the accumulation of reactive oxygen spe-
cies and nitric oxide synthesis in ammonia-treated astrocytes
(Skowrońska et al. 2010).
5.11 S O M ATO S TAT I N R E C E P TO R S
Prominent expression of somatostatin receptors in astrocytes
5.14 TAC H Y K I N I N R EC E P TO R S
of hippocampus, amygdala, and hypothalamus in situ has
been identified in binding studies on brain slices (Porter and Astrocytes express all subtypes of tachykinin receptors, NK1 to
McCarthy 1997). The subtypes have been more stringently NK3. In cell culture, activation of NK1 receptors leads to mem-
identified in culture, and it is evident that astrocytes express brane depolarization because of blockade of the constitutive

R E L E A S E O F G L I OT R A N S M I T T E R S A N D T R A N S M I T T E R R E C E P TO R S I N A S T R O C Y T E S • 205
Table 17.1 TRANSMITTER RECEPTORS AND GLIOTRANSMITTER RELEASE MECHANISMS
NON–VESICLE-BASED
GLIOTRANSMITTER GLIOTRANSMITTER RECEPTORS VESICLE-BASED RELEASE RELEASE

l-Glutamate Ionotropic receptors: Yes Yes


AMPA (GluR1–4)
Kainate (GluR5–7)
NMDA (NR2B)
Metabotropic receptors:
mGluR 1, 3–5
γ-Amino butyric acid (GABA) GABAA, GABAB ? Yes
Glycine Not common, subunits ? ?
α1 β of strychnine-sensitive
receptor
Acetylcholine (ACh) Nicotinic AChR (α7 subunit); ? ?
muscarinic AChR (M1–M5)
Oxytocin, vasopressin Oxytocin receptor (mRNA); ? ?
vasopressin receptor 1a; vaso-
pressin receptor 1b
Vasoactive intestinal peptide (VIP) VPAC2 ? ?
Adrenaline, noradrenaline α1-, α2-, β1-, β2-, ? ?
β3-adrenoreceptors
Angiotensin AT1 and AT2 ? ?
Somatostatin Somatostatin receptor ? ?
(sst2A splice variant)
Serotonin (5HT), 5HT receptor subtypes: ? ?
5-hydroxytryptamine 5HT1A, 5HT2A, 5HT5A
Atrial natriuretic peptide (ANP) NPR-A, NPR-B, NPR-C Yes ?
Tachykinin peptides NK1 to NK3 ? ?
Bradykinin B1 and B2 ? ?
Thyrotropin-releasing hormone Receptor subtype TRH1 ?Yes ?
(TRH)
Opioid peptides μ, δ, κ ? ?
Histamine H1, H2, and H3 ? ?
Dopamine D1 to D5 ? ?
Endothelin ETB
Adenosine-5′-triphosphate (ATP) Ionotropic and metabotropic: Yes Yes
see chapter 25

206 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
K+ conductance and opening of Cl– channels (Backus et al. 5.18 H I S TA M I N E R EC E P TO R S
1991). Receptor localization at the light and electron micro-
In cultured astrocytes, binding sites for H1 and H2 subtypes of
scopic level in different species, including humans, has iden-
histamine receptors have been identified (Inagaki and Wada
tified NK2 and NK3 receptors in astrocytes of various areas
1994), which couple to Gq and Gs types of G-proteins, respec-
of the CNS, for example, spinal cord, cortex, and hippocam-
tively. H1 receptors couple to PLC leading to IP3 production
pus. NK2 receptors have been found to cluster close to axon
and Ca2+ release, whereas H2 receptor activation in astrocytes
terminals in spinal cord. Substance P, as a prominent ligand,
is linked to adenylate cyclase and intracellular cAMP accu-
enhances secretion of IL-6 and PGE2 on IL-1β stimulation in
mulation. H1 receptors were mainly found on the processes
cultured spinal cord astrocytes (Palma et al. 1997). Substance of astrocytes. Histamine acts as a mitogen in cortical astro-
P augments glutamate-triggered ATP release from cultured cytes. Activation of H1 receptors has been shown to stimu-
spinal cord astrocytes via activation of neurokinin recep-
late glycogen breakdown. Histamine triggered an increase in
tors (Werry et al. 2006). Moreover, LPS-induced secretion
[Ca2+]i in acute rat hippocampus because of an upregulation
of tumor necrosis factor α (TNFα) and IL-1 from cultured of functional H1 receptors after postnatal day 8 (Shelton and
astrocytes was augmented by the ligand, substance P (Luber-
McCarthy 2000). Based on ultrastructural studies suggest-
Narod et al. 1994). A proinflammatory function of tachykinin
ing glial apposition to histaminergic neurons, astrocytes were
receptors in reactive astrocytes has been suggested because a
believed to respond to histamine released from hippocampal
lesion-induced upregulation of the receptors was observed in
neurons. In a recent study on cultured astrocytes, all three
astrocytes of transected optic nerve in vivo.
types of histaminergic receptors (H1, H2, H3) were identi-
fied by RT-PCR and a pharmacological approach. All three
5.15 B R A DY K I N I N R E C E P TO R S receptors control the release of neurotropin-3 and converge at
the level of mitogen-activated protein (MAP) kinase activity
Functional bradykinin receptors (B1 and B2 receptors) have ( Jurič et al. 2011).
only been identified in cultured astrocytes. Receptor activation
leads to an increase in intracellular Ca2+, induced by release
from intracellular stores, and to a blockade of a constitutive 5.19 D O PA M I N E R EC E P TO R S
K+ conductance mediated by B2 receptors (Gimpl et al. 1992).
B2 receptor–induced increase in intracellular Ca2+ leads to the Cultured astrocytes from the striatum express D1 to D5 sub-
release of glutamate and aspartate from astrocytes stimulating types of dopamine receptors as shown by RT-PCR (Miyazaki
neuronal glutamate receptors; this substantiates that astro- et al. 2004). D1 receptor activation leads to PTX-sensitive
cytes can directly modulate neuronal activity (Parpura et al. cAMP formation via protein kinase A stimulation. Prolonged
1994). Bradykinin is also involved in astrocyte volume control receptor activation led to a reduction of astroglial dopamine
because it directly activates volume-sensitive outwardly recti- sensitivity (Reuss et al. 2001). D2 receptors in astrocytes are
fying anion channels, even without an increase in cell volume linked to the actin cytoskeleton via interaction with filamin
(Akita and Okada 2011). In rat brain cultured astrocytes, bra- A. Ligand binding and ultrastructural analysis in vivo found
dykinin leads to upregulation of matrix metalloproteinase-9 strong expression of D2 receptors in GFAP-positive corti-
and promotes cell migration (Hsieh et al. 2008). cal astrocyte processes surrounding interneurons. In the pre-
frontal cortex, they were found to be colocalized with alpha7
nicotinic acetylcholine receptors (Duff y et al. 2011). These
5.16 T H Y ROT RO P I N-R E L E A S I N G receptors were indeed functional because their activation
H O R MO N E R E C E P TO R S induced increases in intracellular Ca2+ in the astrocytes (Khan
Thyrotropin-releasing hormone (TRH) receptors have been et al. 2001). Activation of the classic dopamine receptor D1 or
identified in culture and in situ by immunocytochemistry D2 and the phosphatidylinositol-linked D1-like receptor elicits
and Western blot. Astrocytes express the TRH1 subtype. FGF-2 biosynthesis and secretion in striatal astrocytes, which
Expression in situ is stronger in white matter than gray mat- may play a role in neuroprotection (Zhang et al. 2009).
ter as studied in the adult rat spinal cord. The receptors are
present in TRH-synthesizing astrocytes, indicating that 5.20 E N D OT H E L I N R EC E P TO R S
this hormone acts in an autocrine and paracrine fashion
(Fernández-Agulló 2001). The predominant receptor for endothelin in astrocytes is
endothelin B (ETB). Activation of the receptors triggers an
increase in Ca2+ and opens charybdotoxin-sensitive, calcium-
5.17 O P I O I D R E C E P TO R S
activated K+ channels (Supattapone and Ashley 1991). It
There is evidence for the expression of all three subtypes of stimulates proliferation and promotes activation of astro-
the opioid receptors, μ, δ, and κ, in cell culture and in situ cytes, thus inducing reactive gliosis (Ishikawa et al. 1997). It
(Ruzicka et al. 1995). The μ and δ receptors were shown to also affects glutamate levels by downregulating the glutamate
be preferentially expressed on astrocytic processes in situ. transporter GLAST in the astrocytes (Matsuura et al. 2002).
Although μ receptors are more frequently present on imma- Endothelin stimulates the release of glial cell line–derived
ture astrocytes, the expression of δ and κ receptors is increased neurotrophic factor (GDNF) (Koyama et al. 2003), increases
in adult tissue. production of 2-arachidonoyl glycerol, the most abundant

R E L E A S E O F G L I OT R A N S M I T T E R S A N D T R A N S M I T T E R R E C E P TO R S I N A S T R O C Y T E S • 207
endocannabinoid in the CNS (Walter and Stella 2003), and Attwell D, Laughlin SB. 2001. An energy budget for signalling in the
increases mRNA and extracellular release of neurotrophin-3 grey matter of the brain. J Cereb Blood Flow Metab 21:1133–1145.
Azmitia EC. 2001. Modern views on an ancient chemical: serotonin
(Koyama et al. 2005). Endothelin also controls the commu- effects on cell proliferation, maturation, and apoptosis. Brain Res
nication between astrocytes. Both endothelin-1 and endothe- Bull 56:413–424.
lin-3 inhibit the extent of coupling between astrocytes by Backus KH, Berger T. Kettenmann H. 1991. Activation of neurokinin
decreasing the expression of phosphorylated connexin 43, receptors modulates K+ and Cl– channel activity in cultured astro-
the gap junction protein prominently expressed by astrocytes cytes from rat cortex. Brain Res 541:103–109.
Barakat L, Bordey A. 2002. GAT-1 and reversible GABA transport in
(Blomstrand et al. 2004). Bergmann glia in slices. J Neurophysiol 88:1407–1419.
Benedetti B, Matyash V, Kettenmann H. 2011. Astrocytes control
GABAergic inhibition of neurons in the mouse barrel cortex.
6 S U M M A RY A N D P E R S P E C T I VE S J Physiol 589:1159–1172.
Bergami M, Santi S, Formaggio E, Cagnoli C, Verderio C, Blum R, et
al. 2008. Uptake and recycling of pro-BDNF for transmitter-induced
It has become clear over the last years that astrocytes are an secretion by cortical astrocytes. J Cell Biol 183:213–221.
integral functional part of synapses, which led to the formu- Bezzi P, Carmignoto G, Pasti L, Vesce S, Rossi D, Rizzini BL, et al. 1998.
lation of the tripartite synapse concept (see chapter 38). As Prostaglandins stimulate calcium-dependent glutamate release in
summarized in this chapter, astrocytes have the capacity to astrocytes. Nature 391:281–285.
express essentially all transmitter and hormone receptors that Blomstrand F, Venance L, Sirén AL, Ezan P, Hanse E, Glowinski J, et al.
2004. Endothelins regulate astrocyte gap junctions in rat hippocam-
are known to be relevant for signaling in the nervous system pal slices. Eur J Neurosci 19:1005–1015.
(for summary see table 17.1). Thus, they have the capacity to Borán MS, García A. 2007. The cyclic GMP-protein kinase G path-
sense any activity in the brain, including the synaptic activity. way regulates cytoskeleton dynamics and motility in astrocytes.
Astrocytes are also able to release neuroactive substances in J Neurochem 102:216–230.
an activity-dependent manner. Most prominent is the release Bormann J, Kettenmann H. 1988. Patch-clamp study of γ-aminobutyric
of glutamate, ATP, GABA, and D-serine. Thereby, astrocytes acid receptor Cl- channels in cultured astrocytes. Proc Natl Acad Sci
U S A 85:9336–9340.
are able to modulate synaptic function and network activ- Bowman CL, Kimelberg HK. 1984. Excitatory amino acids directly depo-
ity (see chapter 38). As a consequence, astrocytes modulate larize rat brain astrocytes in primary culture. Nature 311:656–659.
behavior (see chapter 39). Moreover, by their ability to take Calegari F, Coco S, Taverna E, Bassetti M, Verderio C, Corradi N, et al.
up neurotransmitters (see chapter 35), they have a second 1999. A regulated secretory pathway in cultured hippocampal astro-
way to affect the neuronal network. Although neuronal cytes. J Biol Chem 274:22539–22547.
Catus SL, Gibbs ME, Sato M, Summers RJ, Hutchinson DS. 2011. Role of
activity is decoded in a millisecond time frame, the astrocyte β -adrenoceptors in glucose uptake in astrocytes using β -adrenoceptor
activity is slower rather in the range of seconds or tenth of knockout mice. Br J Pharmacol 162:1700–1715.
seconds. Moreover, astrocytes are more part of the volume Charles KJ, Calver AR, Jourdain S, Pangalos MN. 2003. Distribution
than the wiring transmission system; thus, extracellular space of a GABAB-like receptor protein in the rat central nervous system.
plays an important role for this type of communication (see Brain Res 989:135–146.
Coco S, Calegari F, Pravettoni E, Pozzi D, Taverna E, Rosa P, et al. 2003.
chapter 34). Thus, brain function is not solely determined by Storage and release of ATP from astrocytes in culture. J Biol Chem
neurons, but is a harmonious collaboration between these 278:1354–1362.
cells and their glial partners . D’Antoni S, Berretta A, Bonaccorso CM, Bruno V, Aronica E, Nicoletti
F, et al. 2008. Metabotropic glutamate receptors in glial cells.
Neurochem Res 33:2436–2443.
Deleuze C, Alonso G, Lefevre IA, Duvoid-Guillou A, Hussy N. 2005.
AC K N OW L E D G M E N T S Extrasynaptic localization of glycine receptors in the rat supraoptic
nucleus: further evidence for their involvement in glia-to-neuron
RZ is supported by grants (P3 310, J3 4051, J3 3632, and communication. Neuroscience 133:175–183.
J3 4146) from the Slovenian Research Agency (ARRS) and Duff y AM, Fitzgerald ML, Chan J, Robinson DC, Milner TA,
the EduGlia ITN EU grant, HK is supported by Deutsche Mackie K, et al. 2011. Acetylcholine α 7 nicotinic and dopamine
D(2) receptors are targeted to many of the same postsynaptic
Forschungsgemeinschaft (TR 43). dendrites and astrocytes in the rodent prefrontal cortex. Synapse
65:1350–1367.
Echigo N, Moriyama Y. 2004. Vesicular inhibitory amino acid trans-
REFERENCES porter is expressed in gamma-aminobutyric acid (GABA)-containing
astrocytes in rat pineal glands. Neurosci Letts 367:79–84.
Akita T, Okada Y. 2011. Regulation of bradykinin-induced activation of Fernández-Agulló T. 2001. Thyrotropin-releasing hormone and its recep-
volume-sensitive outwardly rectifying anion channels by Ca 2+ nano- tor in glia. Glia 33:267–276.
domains in mouse astrocytes. J Physiol 589:3909–3927. Fields RD, Burnstock G. 2006. Purinergic signalling in neuron-glia
Angulo MC, Le Meur K, Kozlov AS, Charpak S, Audinat E. interactions. Nat Rev Neurosci 7:423–436.
2008. GABA, a forgotten gliotransmitter. Progr Neurobiol Figiel M, Engele J. 2000. Pituitary adenylate cyclase-activating polypep-
86:297–303. tide (PACAP), a neuron-derived peptide regulating glial glutamate
Araque A, Martín ED, Perea G, Arellano JI, Buño W. 2002. Synaptically transport and metabolism. J Neurosci 20:3596–3605.
released acetylcholine evokes Ca2+ elevations in astrocytes in hip- Fraser DD, Duff y S, Angelides KJ, Perez-Velazquez JL, Kettenmann H,
pocampal slices. J Neurosci 22:2443–2450. MacVicar BA. 1995. GABA A/benzodiazepine receptors in acutely
Araque A, Parpura V, Sanzgiri RP, Haydon PG. 1999. Tripartite isolated hippocampal astrocytes. J Neurosci 15:2720–2732.
synapses: glia, the unacknowledged partner. Trend Neurosci Füchtbauer L, Groth-Rasmussen M, Holm TH, Løbner M, Toft-Hansen
22:208–215. H, Khorooshi R, et al. 2011. Angiotensin II type 1 receptor (AT1)

208 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
signaling in astrocytes regulates synaptic degeneration-induced leu- Kreft M, Krizaj D, Grilc S, Zorec R. 2003. Properties of exocytotic
kocyte entry to the central nervous system. Brain Behav Immun response in vertebrate photoreceptors. J Neurophysiol 90:218–225.
25:897–904. Kreft M, Stenovec M, Rupnik M, Grilc S, Krzan M, Potokar M, et al.
Gallo V, Suergiu R, Levi G. 1986. Kainic acid stimulates GABA release 2004. Properties of Ca(2+)-dependent exocytosis in cultured astro-
from a subpopulation of cerebellar astrocytes. Eur J Pharmacol cytes. Glia 46:437–445.
132:319–322. Krzan M, Stenovec M, Kreft M, Pangrsic T, Grilc S, Haydon PG, et al.
Gimpl G, Walz W, Ohlemeyer C, Kettenmann H. 1992. Bradykinin 2003. Calcium-dependent exocytosis of atrial natriuretic peptide
receptors in cultured astrocytes from neonatal rat brain are linked to from astrocytes. J Neurosci 23:1580–1583.
physiological responses. Neurosci Letts 144:139–142. Kulik A, Haentzsch A, Lückermann M, Reichelt W, Ballanyi K.
Graham A, Court JA, Martin-Ruiz CM, Jaros E, Perry R, Volsen SG, 1999. Neuron-glia signaling via alpha(1) adrenoceptor-mediated
et al. 2002. Immunohistochemical localisation of nicotinic ace- Ca(2+) release in Bergmann glial cells in situ. J Neurosci 19:
tylcholine receptor subunits in human cerebellum. Neuroscience 8401–8408.
113:493–507. Lalo U, Pankratov Y, Kirchhoff F, North RA, Verkhratsky A. 2006.
Griffith R, Sutin J. 1996. Reactive astrocyte formation in vivo is regu- NMDA receptors mediate neuron-to-glia signaling in mouse cortical
lated by noradrenergic axons. J Comp Neurol 371:362–375. astrocytes. J Neurosci 26:2673–2683.
Guizzetti M, Moore NH, Giordano G, Costa LG. 2008. Modulation Larsson M, Sawada K, Morland C, Hiasa M, Ormel L, Moriyama Y,
of neuritogenesis by astrocyte muscarinic receptors. J Biol Chem et al. 2011. Functional and anatomical identification of a vesicu-
283:31884–31897. lar transporter mediating neuronal ATP release. Cerebral Cortex,
Hamilton NB, Attwell D. 2010. Do astrocytes really exocytose neu- doi:10.1093/cercor/bhr203.
rotransmitters? Nat Rev Neurosci 11:227–238. Laureys G, Clinckers R, Gerlo S, Spooren A, Wilczak N, Kooijman R,
Härtel K, Schnell C, Hülsmann S. 2009. Astrocytic calcium signals et al. 2010. Astrocytic beta(2)-adrenergic receptors: from physiology
induced by neuromodulators via functional metabotropic receptors to pathology. Progr Neurobiol 91:189–199.
in the ventral respiratory group of neonatal mice. Glia 57:815–827. Lee S, Yoon BE, Berglund K, Oh SJ, Park H, Shin HS, et al. 2010.
Henneberger C, Papouin T, Oliet SH, Rusakov DA. 2010. Long-term Channel-mediated tonic GABA release from glia. Science
potentiation depends on release of D-serine from astrocytes. Nature 330:790–796.
463:232–236. Liu T, Sun L, Xiong Y, Shang S, Guo N, Teng S, et al. 2011. Calcium
Hsieh HL, Wu CY, Yang CM. 2008. Bradykinin induces matrix triggers exocytosis from two types of organelles in a single astrocyte.
metalloproteinase-9 expression and cell migration through a J Neurosci 31:10593–10601.
PKC-delta-dependent ERK/Elk-1 pathway in astrocytes. Glia 56: Luber-Narod J, Kage R, Leeman SE. 1994. Substance P enhances the
619–632. secretion of tumor necrosis factor-alpha from neuroglial cells stimu-
Iino M, Goto K, Kakegawa W, Okado H, Sudo M, Ishiuchi S, et al. 2001. lated with lipopolysaccharide. J Immunol 152:819–824.
Glia-synapse interaction through Ca2+-permeable AMPA receptors Lucas KA, Pitari GM, Kazerounian S, Ruiz-Stewart I, Park J, Schulz S,
in Bergmann glia. Science 292:926–929. et al. 2000. Guanylyl cyclases and signaling by cyclic GMP. Pharmacol
Inagaki N, Wada H. 1994. Histamine and prostanoid receptors on glial Rev 52:375–414.
cells. Glia 11:102–109. Masmoudi-Kouki O, Gandolfo P, Castel H, Leprince J, Fournier A,
Ishikawa N, Takemura M, Koyama Y, Shigenaga Y, Okada T, Baba A. Dejda A, et al. 2007. Role of PACAP and VIP in astroglial functions.
1997. Endothelins promote the activation of astrocytes in rat neo- Peptides 28:1753–1760.
striatum through ET(B) receptors. Eur J Neurosci 9:895–901. Matsuura S, Ikegaya Y, Yamada MK, Nishiyama N, Matsuki N. 2002.
Jorgačevski J, Fošnarič M, Vardjan N, Stenovec M, Potokar M, Kreft M, Endothelin downregulates the glutamate transporter GLAST in
et al. 2010. Fusion pore stability of peptidergic vesicles. Mol Membr cAMP-differentiated astrocytes in vitro. Glia 37:178–182.
Biol 27:65–80. Matthias K, Kirchhoff F, Seifert G, Hüttmann K, Matyash M,
Jurič DM, Mele T, Carman-Kržan M. 2011. Involvement of histamin- Kettenmann H, Steinhäuser C. 2003. Segregated expression of
ergic receptor mechanisms in the stimulation of NT-3 synthesis in AMPA-type glutamate receptors and glutamate transporters defines
astrocytes. Neuropharmacology 60:1309–1317. distinct astrocyte populations in the mouse hippocampus. J Neurosci
Kang J, Jiang L, Goldman SA, Nedergaard M. 1998. Astrocyte-mediated 23:1750–1758.
potentiation of inhibitory synaptic transmission. Nat Neurosci McKenzie JC, Juan YW, Thomas CR, Berman NE, Klein RM. 2001.
1:683–692. Atrial natriuretic peptide-like immunoreactivity in neurons and
Kettenmann H, Backus KH, Schachner M. 1984. Aspartate, gluta- astrocytes of human cerebellum and inferior olivary complex.
mate and gamma-aminobutyric acid depolarize cultured astrocytes. J Histochem Cytochem 49:1453–1467.
Neurosci Letts 52:25–29. Meier SD, Kafitz KW, Rose CR. 2008. Developmental profi le and mech-
Kettenmann H, Faissner A, Trotter J. 1996. Neuron-glia interactions anisms of GABA-induced calcium signaling in hippocampal astro-
in homeostasis and degeneration. In: Greger R, Windhorst U (eds.), cytes. Glia 56:1127–1137.
Comprehensive human physiology. Heidelberg: Springer-Verlag, Milner TA, Lee A, Aicher SA, Rosin DL. 1998. Hippocampal
pp. 533–543. alpha2a-adrenergic receptors are located predominantly presynapti-
Khan ZU, Koulen P, Rubinstein M, Grandy DK, Goldman-Rakic PS. cally but are also found postsynaptically and in selective astrocytes.
2001. An astroglia-linked dopamine D2-receptor action in prefron- J Comp Neurol 395:310–327.
tal cortex. Proc Natl Acad Sci U S A 98:1964–1969. Mittaud P, Labourdette G, Zingg H, Guenot-Di Scala D. 2002. Neurons
Kirchhoff F, Mü lhardt C, Pastor A, Becker CM, Kettenmann H. 1996. modulate oxytocin receptor expression in rat cultured astro-
Expression of glycine receptor subunits in glial cells of the rat spinal cytes: involvement of TGF-beta and membrane components. Glia
cord. J Neurochem 66:1383–1390. 37:169–177.
Kopan R, Ilagan MX. 2009. The canonical Notch signaling pathway: Miyazaki I, Asanuma M, Diaz-Corrales FJ, Miyoshi K, Ogawa N. 2004.
unfolding the activation mechanism. Cell 137:216–233. Direct evidence for expression of dopamine receptors in astrocytes
Koyama Y, Tsujikawa K, Matsuda T, Baba A. 2003. Endothelin-1 stimu- from basal ganglia. Brain Res 1029:120–123.
lates glial cell line-derived neurotrophic factor expression in cultured Montana V, Malarkey EB, Verderio C, Matteoli M, Parpura V. 2006.
rat astrocytes. Biochem Biophys Res Commun 303:1101–1105. Vesicular transmitter release from astrocytes. Glia 54:700–715.
Koyama Y, Tsujikawa K, Matsuda T, Baba A. 2005. Endothelin increases Mothet JP, Pollegioni L, Ouanounou G, Martineau M, Fossier P, Baux
expression of exon III- and exon IV-containing brain-derived neu- G. 2005. Glutamate receptor activation triggers a calcium-dependent
rotrophic factor transcripts in cultured astrocytes and rat brain. and SNARE protein-dependent release of the gliotransmitter
J Neurosci Res 80:809–816. D-serine. Proc Natl Acad Sci U S A 102:5606–5611.

R E L E A S E O F G L I OT R A N S M I T T E R S A N D T R A N S M I T T E R R E C E P TO R S I N A S T R O C Y T E S • 209
Mudrick-Donnon LA, Williams PJ, Pittman Q J, MacVicar BA. 1993. Potter LR, Abbey-Hosch S, Dickey DM. 2006. Natriuretic peptides,
Postsynaptic potentials mediated by GABA and dopamine evoked their receptors, and cyclic guanosine monophosphate-dependent sig-
in stellate glial cells of the pituitary pars intermedia. J Neurosci naling functions. Endocr Rev 27:47–72.
13:4660–4668. Prado J, Baltrons MA, Pifarré P, García A. 2010. Glial cells as sources and
Müller T, Fritschy JM, Grosche J, Pratt GD, Möhler H, Kettenmann targets of natriuretic peptides. Neurochem Int 57:367–374.
H. 1994. Developmental regulation of voltage-gated K+ channel Prebil M, Vardjan N, Jensen J, Zorec R, Kreft M. 2011. Dynamic moni-
and GABA A receptor expression in Bergmann glial cells. J Neurosci toring of cytosolic glucose in single astrocytes. Glia 59:903–913.
14:2503–2514. Ramamoorthy P, Whim MD. 2008. Trafficking and fusion of neuro-
Müller T, Möller T, Berger T, Schnitzer J, Kettenmann H. 1992. Calcium peptide Y-containing dense-core granules in astrocytes. J Neurosci
entry through kainate receptors and resulting potassium-channel 28:13815–13827.
blockade in Bergmann glial cells. Science 256:1563–1566. Reuss B, Lorenzen A, Unsicker K. 2001. Dopamine and epinephrine, but
Murthy KS, Teng B, Jin J, Makhlouf GM. 1998. G protein-dependent not serotonin, downregulate dopamine sensitivity in cultured corti-
activation of smooth muscle eNOS via natriuretic peptide clearance cal and striatal astroglial cells. Receptors Channels 7:441–451.
receptor. Am J Physiol 275:C1409–C1416. Robinson MB. 2002. Regulated trafficking of neurotransmitter trans-
Muyderman H, Nilsson M, Blomstrand F, Khatibi S, Olsson T, Hansson porters: common notes but different melodies. J Neurochem
E, et al. 1998. Modulation of mechanically induced calcium waves in 80:1–11.
hippocampal astroglial cells. Inhibitory effects of alpha 1-adrenergic Rose RA, Giles WR. 2008. Natriuretic peptide C receptor signalling in
stimulation. Brain Res 793:127–135. the heart and vasculature. J Physiol 586:353–366.
Nedergaard M, Takano T, Hansen AJ. 2002. Beyond the role of gluta- Rosenberg D, Kartvelishvily E, Shleper M, Klinker CM, Bowser MT,
mate as a neurotransmitter. Nat Rev Neurosci 3:748–775. Wolosker H. 2010. Neuronal release of D-serine: a physiological
Nishimoto M, Miyakawa H, Wada K, Furuta A. 2011. Activation of the pathway controlling extracellular D-serine concentration. FASEB J
VIP/VPAC2 system induces reactive astrocytosis associated with 24:2951–2961.
increased expression of glutamate transporters. Brain Res 1383:43–53. Roy ML, Sontheimer H. 1995. Beta-adrenergic modulation of
Oliet SH, Mothet JP. 2006. Molecular determinants of D-serine-mediated glial inwardly rectifying potassium channels. J Neurochem 64:
gliotransmission: from release to function. Glia 54:726–737. 1576–1584.
Orellana JA, Froger N, Ezan P, Jiang JX, Bennett MV, Naus CC, et al. Ruzicka BB, Fox CA, Thompson RC, Meng F, Watson SJ, Akil H. 1995.
2011. ATP and glutamate released via astroglial connexin 43 hemi- Primary astroglial cultures derived from several rat brain regions dif-
channels mediate neuronal death through activation of pannexin 1 ferentially express mu, delta and kappa opioid receptor mRNA. Brain
hemichannels. J Neurochem 118:826–840. Res (Mol Brain Res) 34:209–220.
Palma C, Minghetti L, Astolfi M, Ambrosini E, Silberstein FC, Manzini Sawada K, Echigo N, Juge N, Miyaji T, Otsuka M, Omote H, et al. 2008.
S, et al. 1997. Functional characterization of substance P receptors Identification of a vesicular nucleotide transporter. Proc Natl Acad
on cultured human spinal cord astrocytes: synergism of substance P Sci U S A 105:5683–5686.
with cytokines in inducing interleukin-6 and prostaglandin E2 pro- Schell MJ, Molliver ME, Snyder SH. 1995. D-serine, an endogenous syn-
duction. Glia 21:183–193. aptic modulator: localization to astrocytes and glutamate-stimulated
Panatier A. 2009. Glial cells: indispensable partners of hypothalamic release. Proc Natl Acad Sci U S A 92:3948–3952.
magnocellular neurones. J Neuroendocrin 21:665–672. Schipke CG, Ohlemeyer C, Matyash M, Nolte C, Kettenmann H,
Pangrsic T, Potokar M, Stenovec M, Kreft M, Fabbretti E, Nistri A, Kirchhoff F. 2001. Astrocytes of the mouse neocortex express func-
et al. 2007. Exocytotic release of ATP from cultured astrocytes. J Biol tional N-methyl-D-aspartate receptors. FASEB J 15:1270–1272.
Chem 282:28749–28758. Seifert G, Schilling K, Steinhäuser C. 2006. Astrocyte dysfunction in
Parpura V, Baker BJ, Jeras M, Zorec R. 2010. Regulated exocytosis in neurological disorders: a molecular perspective. Nat Rev Neurosci
astrocytic signal integration. Neurochem Int 57:451–459. 7:194–206.
Parpura V, Basarsky TA, Liu F, Jeftinija K, Jeftinija S, Haydon PG. Seifert G, Steinhäuser C. 2011. Neuron-astrocyte signaling and epilepsy.
1994. Glutamate-mediated astrocyte-neuron signalling. Nature Exp Neurol doi:10.1016/j.expneurol.2011.08.024.
369:744–747. Serrano A, Haddjeri N, Lacaille JC, Robitaille R. 2006. GABAergic net-
Parpura V, Verkhratsky A. 2011. The astrocyte excitability brief: work activation of glial cells underlies hippocampal heterosynaptic
from receptors to gliotransmission. Neurochem Int doi:10.1016/j. depression. J Neurosci 26:5370–5382.
neuint.2011.12.001. Serrano A, Robitaille R, Lacaille JC. 2008. Differential NMDA-
Parpura V, Zorec R. 2010. Gliotransmission: exocytotic release from dependent activation of glial cells in mouse hippocampus. Glia 56:
astrocytes. Brain Res Rev 63:83–92. 1648–1663.
Pastor A, Chvátal A, Syková E, Kettenmann H. 1995. Glycine- and Sharma G, Vijayaraghavan S. 2001. Nicotinic cholinergic signaling
GABA-activated currents in identified glial cells of the developing in hippocampal astrocytes involves calcium-induced calcium
rat spinal cord slice. Eur J Neurosci 7:1188–1198. release from intracellular stores. Proc Natl Acad Sci U S A 98:
Porter JT, McCarthy KD. 1997. Astrocytic neurotransmitter receptors 4148–4153.
in situ and in vivo. Progr Neurobiol 51:439–455. Shelton MK, McCarthy KD. 2000. Hippocampal astrocytes exhibit
Potokar M, Kreft M, Li L, Daniel Andersson J, Pangrsic T, Chowdhury Ca2+-elevating muscarinic cholinergic and histaminergic receptors in
HH, et al. 2007. Cytoskeleton and vesicle mobility in astrocytes. situ. J Neurochem 74:555–563.
Traffic 8:12–20. Skowrońska M, Zielińska M, Albrecht J. 2010. Stimulation of natri-
Potokar M, Kreft M, Pangrsic T, Zorec R. 2005. Vesicle mobility uretic peptide receptor C attenuates accumulation of reactive oxygen
studied in cultured astrocytes. Biochem Biophys Res Commun species and nitric oxide synthesis in ammonia-treated astrocytes.
329:678–683. J Neurochem 115:1068–1076.
Potokar M, Stenovec M, Gabrijel M, Li L, Kreft M, Grilc S, et al. 2010. Smith K. 2010. Neuroscience: settling the great glia debate. Nature
Intermediate fi laments attenuate stimulation-dependent mobility of 468:160–162.
endosomes/lysosomes in astrocytes. Glia 58:1208–1219. Soos JM, Morrow J, Ashley TA, Szente BE, Bikoff EK, Zamvil SS.
Potokar M, Stenovec M, Kreft M, Gabrijel M, Zorec R. 2011. 1998. Astrocytes express elements of the class II endocytic pathway
Physiopathologic dynamics of vesicle traffic in astrocytes. Histol and process central nervous system autoantigen for presentation to
Histopathol 26:277–284. encephalitogenic T cells. J Immunol 161:5959–5966.
Potokar M, Stenovec M, Kreft M, Kreft ME, Zorec R. 2008. Stimulation Stenovec M, Kreft M, Grilc S, Pangrsic T, Zorec R. 2008. EAAT2 den-
inhibits the mobility of recycling peptidergic vesicles in astrocytes. sity at the astrocyte plasma membrane and Ca2+-regulated exocyto-
Glia 56:135–144. sis. Mol Membr Biol 25:203–215.

210 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Stenovec M, Kreft M, Grilc S, Potokar M, Kreft ME, Pangrsic T, et al. hypothalamic neurons and astrocytes in culture. J Neurochem 68:
2007. Ca 2+-dependent mobility of vesicles capturing anti-VGLUT1 2273–2280.
antibodies. Exp Cell Res 313:3809–3818. Walter L, Stella N. 2003. Endothelin-1 increases 2-arachidonoyl glycerol
Stenovec M, Kreft M, Poberaj I, Betz WJ, Zorec R. 2004. Slow spon- (2-AG) production in astrocytes. Glia 44:85–90.
taneous secretion from single large dense-core vesicles monitored in Wang LP, Cheung G, Kronenberg G, Gertz K, Ji S, Kempermann G,
neuroendocrine cells. FASEB J 18:1270–1272. et al. 2008. Mild brain ischemia induces unique physiological proper-
Sumners C, Tang W. 1992. Atrial natriuretic peptide receptor subtypes ties in striatal astrocytes. Glia 56:925–934.
in rat neuronal and astrocyte glial cultures. American J Physiol Werry EL, Liu GJ, Bennett MR. 2006. Glutamate-stimulated ATP
262:C1134–C1143. release from spinal cord astrocytes is potentiated by substance P.
Sumners C, Tang W, Paulding W, Raizada MK. 1994. Peptide receptors J Neurochem 99:924–936.
in astroglia: focus on angiotensin II and atrial natriuretic peptide. Wolosker H. 2011. Serine racemase and the serine shuttle
Glia 11:110–116. between neurons and astrocytes. Biochim Biophys Acta 1814:
Supattapone S, Ashley CC. 1991. Endothelin opens potassium channels 1558–1566.
in glial cells. Eur J Neurosci 3:349–355. Wolosker H, Sheth KN, Takahashi M, Mothet JP, Brady Jr RO,
Syed N, Martens CA, Hsu WH. 2007. Arginine vasopressin increases Ferris CD, et al. 1999. Purification of serine racemase: biosynthe-
glutamate release and intracellular Ca 2+ concentration in hip- sis of the neuromodulator D-serine. Proc Natl Acad Sci U S A 96:
pocampal and cortical astrocytes through two distinct receptors. 721–725.
J Neurochem 103:229–237. Ye ZC, Wyeth MS, Baltan-Tekkok S, Ransom BR. 2003. Functional
Takata N, Mishima T, Hisatsune C, Nagai T, Ebisui E, Mikoshiba K, et hemichannels in astrocytes: a novel mechanism of glutamate release.
al. 2011. Astrocyte calcium signaling transforms cholinergic modula- J Neurosci 23:3588–3596.
tion to cortical plasticity in vivo. J Neurosci 31:18155–18165. Zhang Q, Fukuda M, Van Bockstaele E, Pascual O, Haydon PG. 2004a.
Vélez-Fort M, Audinat E, Angulo MC. 2011. Central role of Synaptotagmin IV regulates glial glutamate release. Proc Natl Acad
GABA in neuron-glia interactions. Neuroscientist doi:10.1177/ Sci U S A 101:9441–9446.
1073858411403317. Zhang X, Zhou Z, Wang D, Li A, Yin Y, Gu X, et al. 2009. Activation
Verderio C, Cagnoli C, Bergami M, Francolini M, Schenk U, Colombo of phosphatidylinositol-linked D1-like receptor modulates FGF-2
A, et al. 2011. TI-VAMP/VAMP7 is the snare of secretory lysosomes expression in astrocytes via IP3-dependent Ca2+ signaling. J Neurosci
contributing to ATP secretion from astrocytes. Biol Cell doi:10.1111/ 29:7766–7775.
boc.201100070. Zhang Z, Chen G, Zhou W, Song A, Xu T, Luo Q, et al. 2007. Regulated
Verkhratsky A, Kirchhoff F. 2007. NMDA receptors in glia. ATP release from astrocytes through lysosome exocytosis. Nat Cell
Neuroscientist 13:28–37. Biol 9:945–953.
Verkhratsky A, Orkand RK, Kettenmann H. 1998. Glial calcium: Zieli ń ska M, Fresko I, Konopacka A, Felipo V, Albrecht J. 2007.
homeostasis and signaling function. Physiol Rev 78:99–141. Hyperammonemia inhibits the natriuretic peptide receptor
Viollet C, Lanneau C, Faivre-Bauman A, Zhang J, Djordjijevic 2 (NPR-2)-mediated cyclic GMP synthesis in the astrocytic
D, Loudes C, et al. 1997. Distinct patterns of expression and compartment of rat cerebral cortex slices. Neurotoxicology
physiological effects of sst1 and sst2 receptor subtypes in mouse 28:1260–1263.

R E L E A S E O F G L I OT R A N S M I T T E R S A N D T R A N S M I T T E R R E C E P TO R S I N A S T R O C Y T E S • 211
18.
STORAGE AND RELEASE OF NONTRANSMITTER
SIGNALING MOLECULES FROM MACROGLIA
Oliver von Bohlen und Halbach and Klaus Unsicker

A B B R E VI AT I O N S on synaptic connectivity, myelination and support of axonal


integrity, to implications in neurode- and regeneration, just to
AAV adeno-associated vectors name a few. The vast array of functions performed by glial cells
AR amphiregulin crucially depends on signaling molecules, which they release
ART artemin and to which they respond. This chapter focuses on macroglia-
BDNF brain-derived neurotrophic factor derived protein growth factors acting on neurons and their
BMP bone morphogenetic protein processes, synapses, vascular cells, and, in both autocrine and
BTC betacellulin heterocrine modes, on glial cells. The literature selected is
CNS central nervous system mostly from the past decade, with a focus on articles that have
CNTF ciliary neurotrophic factor appeared since the second edition of Neuroglia. Each subchap-
EGF epidermal growth factor ter discussing a growth factor or growth factor family (arranged
EGFR epidermal growth factor receptor in alphabetical order), respectively, is introduced by a brief
EPR epiregulin general outline followed by specific details of their functions
FGF fibroblast growth factor related to Schwann cells, astrocytes, and oligodendrocytes.
FGFR fibroblast growth factor receptor
GDF growth differentiation factor
GDNF glial cell–derived neurotrophic factor 2 E P I D E R M A L G R OW T H FAC TO R FA M I LY
GFR growth factor receptor
HB-EGF heparin-binding EGF-like growth factor Epidermal growth factor (EGF), as well as transforming
IGF insulin-like growth factor growth factor-α (TGF-α), heparin-binding EGF-like growth
IGFBP IGF-binding protein factor (HB-EGF), amphiregulin (AR), epiregulin (EPR), epi-
IL-6 interleukin 6 gen, betacellulin (BTC), and the neuregulins 1–4 (NRG1–4)
LIF leukemia inhibitory factor belong to the EGF family. Epidermal growth factor signals via
MPTP 1-methyl-4-phenyl-1,2,3,6-tetrahydro EGF receptors, known as ErbB1–ErbB4. The ErbB protein
pyridine family or epidermal growth factor receptor (EGFR) family is
NGF nerve growth factor a family of four structurally related receptor tyrosine kinases.
NRG1–4 neuregulin 1–4 ErbB1 is activated, for example, by EGF, TGF-α, HB-EGF,
NT-3 neurotrophin 3 amphiregulin, betacellulin, epiregulin, and epigen, whereas the
NT-4 neurotrophin 4 ErbB4 receptor is mainly activated, for example, by HB-EGF,
NTN neurturin betacellulin, and the neuregulins 1–4.
PAR protease-activated receptor
PDGF platelet-derived growth factor
2.1 S C H WA N N C E L L S
PNS peripheral nervous system
PSPN persephin During development of the peripheral nervous system (PNS),
TβR TGF-β receptor Schwann cells require neuregulin (NRG1) provided by neu-
TGF-α transforming growth factor-α rons for growth, migration, and myelination; moreover, NRG
TGF-β transforming growth factor-β and ErbB also control Schwann cells during axon regeneration
(Joung et al. 2010). Neuregulin acts as a potent chemoattractive
and kinetic molecule, as revealed by analyzing Schwann cell pre-
1 INTRODUCTION cursors (Cornejo et al. 2010). Neuregulin (type III) is essential for
myelination in the PNS. Interestingly, NRG1/ErbB signaling dif-
Since the second edition of Neuroglia in 2005 our knowledge fers between Schwann cells and oligodendrocytes (Brinkmann et
on macroglial cells (Schwann cells, astrocytes, and oligoden- al. 2008). Nave et al. generated a series of conditional null mutants
drocytes) has enormously expanded. This applies to almost completely lacking NRG. Surprisingly, these mice had normal
all fields of glia biology, from developmental genetics, impact amounts of CNS myelin. Furthermore, double mutants deficient

212
for ErbB3 and ErbB4 in oligodendrocytes were myelinated with- and von Bohlen und Halbach 2003). The majority of FGFs
out stimulation by neuregulins, indicating that signaling through possess amino-terminal signal peptides; therefore, they are
NRG1 and ErbB differs between Schwann cells and oligodendro- assumed to be readily secreted from cells, whereas some FGFs,
cytes. Activation of NF-kappaB seems to be critical for Schwann including FGF-2, lack conventional signal peptides, but can be
cell myelination (Limpert and Carter 2010). Its transcriptional secreted nevertheless (Okada-Ban et al. 2000). Although most
activity can be significantly increased by forskolin. Both ErbB2 FGFs act in a paracrine and/or autocrine manner, some have
and ErbB3 serve as the transducers of the NRG1 signal. Schwann potential roles in metabolism, acting in an endocrine man-
cell–derived tumors of the neurofibromatosis type 1 express the ner. In the adult CNS, high levels of the receptors FGFR-1,
EGF-R and respond to EGF (DeClue et al. 2000), suggesting FGFR-2, and FGFR-3 are expressed within the diencephalon
that the EGF-R plays an important role in Schwann cell transfor- and telencephalon, whereas their expression levels are rela-
mation. EGF is also involved in reprogramming Schwann cells to tively low in other areas. Fibroblast growth factor-1 is widely
multipotent adult neural crest cells (Widera et al. 2011). expressed, but nevertheless confined to specific neuronal
populations in the adult CNS; however, FGFR-1 has also
been detected in astrocytes of white matter tracts. Fibroblast
2.2 A S T RO C Y T E S
growth factor-2 and -3 are primarily found on glial cells. The
Transforming growth factor-α and EGF-R immunoreactiv- fourth member of the FGF receptors (FGFR-4), shows robust
ity are found in both neurons and astrocytes in the developing expression only during early stages of development, and is not
and adult brain of humans and different species of animals. In detectable in the adult CNS outside the habenular complex.
addition, TGF-α and EGF-R colocalize in most neurons and
maturing astrocytes. Heparin-binding EGF-like growth fac-
3.1 S C H WA N N C E L L S
tor stimulates the proliferation of astrocytes and multipotent
progenitors. Neuregulin 1 seems to have a specific role for the Schwann cells express FGF-1, -2, and -5, and the FGFR-1, -2,
generation of astrocytes, because NRG1–ErbB2 signaling is and -3 (Grothe and Nikkhah 2001) expression is increased
required for the establishment of radial glia and their transfor- following nerve crush. Lack of FGF-2 leads to poor func-
mation into astrocytes in cerebral cortex (Schmid et al. 2003). tional recovery after peripheral nerve injuy (Seitz et al. 2011).
EGF-induced proliferation of astrocytes requires the transcrip- Although faster regeneration after sciatic nerve injury in mice
tion factor Egr-1 (Mayer et al. 2009). Several pharmacologically overexpressing FGF-2 has been reported ( Jungnickel et al.
relevant stimuli, as for example by adrenoceptor agonists and 2006), consistent with enhanced axonal regeneration across
angiotensin II, have been shown to transactivate the EGF-R in long gaps mediated by FGF-2 (high molecular form) overex-
astrocytes (Clark and Gonzalez 2007; Li et al. 2008). Epidermal pressing Schwann cells (Timmer et al. 2003), in a facial nerve
growth factor receptor activation also downregulates collagens paradigm FGF-2 overexpressing Schwann cells apparently
in cultured astrocytes, thereby probably providing an explana- failed to promote functional recovery (Haastert et al. 2009).
tion for the fact that astrocytes do not secrete collagens in vivo Fibroblast growth factor-1 and -2 have been implicated in
(Heck et al. 2007) and that therefore the extracellular matrix stimulation of Schwann cell proliferation and inhibition of
of the CNS lacks fibrillar elements. Similarly, EGF-R ligands myelination at the lesion site, whereas activation of FGFR-3
regulate injury-induced growth factor and matrix production likely promotes neurite formation (Grothe et al. 2006).
by astroglia, as for example chondroitin sulfate proteoglycans
(Smith and Strunz 2005) and prostaglandin (Zhang and Neufeld
3.2 A S T RO C Y T E S
2005). Astroglia-derived EGF and TGF-α also promote their
own calcium oscillations (Morita et al. 2005). Together, there is Astrocytes are a prominent source of FGF-2. Autocrine and
robust evidence now that astrocytic ErbB signaling is crucially paracrine actions of FGF-2 on astrocytes include regulation
implicated in the autocrine and paracrine regulation of astro- of gap junction (connexin43) expression and function (see
glial function and integration and processing of neuronal inputs also chapter 24), neurotransmitter sensitivity, and intermedi-
by astrocytes (Sharif and Prevot 2010). ate filament density (Unsicker et al. 2006). Similar to FGF-2,
FGF-1 is also implicated in gap junction regulation (Garré
et al. 2010). Interestingly, FGF-2 affects astrocyte differentia-
2.3 O L I G O D E N D RO C Y T E S
tion selectively in the gray matter and in a brain region-specific
Neonatal oligodendrocytes contain and secrete neuregulins manner, for example, in the hindbrain (Irmady et al. 2011), as
in vitro. Furthermore, it is known that oligodendrocytes express revealed by monitoring GFAP expression and blood-brain
ErbB-3 as well as ErbB-4; moreover, HB-EGF mRNA and protein barrier integrity. (More information concerning the blood-
are expressed by oligodendrocytes in vivo (Du and Dreyfus 2002). brain barrier is provided in chapter 33.) Mechanistically, it
was shown that methylation of histone H3 at lysine 4 resi-
due associated with the STAT binding site on the GFAP
3 F I B R O B L A S T G R OW T H promoter was significantly decreased in the gray matter of
FAC TO R FA M I LY the FGF-2 null mouse, suggesting a role for FGF-2 in the
epigenetic regulation of astrocyte differentiation in vivo.
A substantial fraction of the 23 fibroblast growth factor (FGF) Astrocyte specification as well as proliferation is medi-
family members are expressed by neurons and glial cells (Reuss ated through diverse FGFRs (Kang and Song 2010). FGF-2

S TO R AG E A N D R E L E A S E O F N O N T R A N S M I T T E R S I G N A L I N G M O L E C U L E S F R O M M AC R O G L I A • 213
selectively induces A2B5-positive astrocyte precursor cells is localized adjacent to the outermost myelin sheath and in the
to differentiate into GFAP-positive cells (Lin and Goldman endoneurium. Schwann cells contain GFRα1 in both soluble
2009). A clinically relevant aspect concerns the activation of and membrane fractions; it is localized on the outer surface
FGFR through antidepressants, an effect that also implicates of the Schwann cell plasma membrane (Hase et al. 2005).
glial cell–derived neurotrophic factor (GDNF) production In peripheral nerve and spinal cord lesions substituted with
(Hisaoka et al. 2011); however, conclusive clinical studies Schwann cell grafts, GDNF has been shown to promote axon
have not yet been done. Brain injury leads to upregulation regeneration. In lesioned nerves, an autocrine loop of BMP-2
of FGF-2 in astrocytes and of FGFR-1 in both astrocytes and upregulates GDNF mRNA in Schwann cells (Kinameri and
neurons. Association of specific heparin sulfate proteoglycans Matsuoka 2003). After long-term denervation, a decline in
to the FGF-2–FGFR-1 complex accounts for the regulation Schwann cell–derived GDNF may account for impaired
of FGF-2 storage, nuclear trafficking, and cell-specific injury regeneration (Höke et al. 2002). Specific expression patterns
responses (Leadbeater et al. 2006). of GDNF family members have been reported during periph-
eral nerve development and in cultured Schwann cells (Piirsoo
3.3 O L I G O D E N D RO C Y T E S
et al. 2010). Artemin has been shown to influence neural dam-
age in chronic pancreatitis (Ceyhan et al. 2007).
Oligodendrocytes are further important target cells for
FGF-actions. Thus, FGF signaling is required for the genera-
4.2 A S T RO C Y T E S
tion of oligodendrocyte progenitors from the embryonic fore-
brain (Furusho et al. 2011). The 120-kDa isoform of FGFR-2 Glial cell–derived neurotrophic factor is also expressed by astro-
is the most abundant tyrosine-phosphorylated protein in cytes. Levels of GDNF are increased by FGF-2 and bone mar-
oligodendrocytes and phosphorylated even in the absence of row stromal cells (Shen et al. 2010; Shin et al. 2009), and release
FGF-2. Fibroblast growth factor receptor-2, but not FGFR-1 of GDNF from astroglia is stimulated by dexmedetomidine
is associated with lipid raft domains in oligodendrocytes and (Yan et al. 2011). Fibroblast growth factor-2–mediated induc-
myelin, but not in astrocytes (Bryant et al. 2009). In addition tion requires the transcription factor Egr-1. Glial cell–derived
to FGF-2, FGF-9 is expressed by both astroglial cells and oli- neurotrophic factor is likely secreted both with and without pro-
godendrocytes. Using double immunofluorescence and in situ cessing by furin-like proteases; the prodomain and C-terminal
hybridization, specific FGF-9 signals in GFAP-positive white cysteines play important roles in GDNF processing and secre-
matter astrocytes of adult rat spinal cord and brainstem as well tion in cultured astrocytes and C6 glioma cells (Oh-hashi
as in CNPase-positive cerebellar and callosal oligodendrocytes et al. 2009). Reactive, nestin-positive astrocytes induced by
have been detected (Nakamura et al. 1999). 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) express
prominent levels of GDNF, thereby probably contributing to
the neurotrophic support of neurons (Chen et al. 2006).
4 G L I A C E L L L I N E –D E R I VE D
N E U R OT R O P H I C FAC TO R FA M I LY 4.3 O L I G O D E N D RO C Y T E S

The GDNF family, consisting of GDNF, neurturin (NTN), Glial cell–derived neurotrophic factor is expressed by oligo-
artemin (ART), and persephin (PSPN), are distant members dendroglia and decreased in alpha-synuclein transgenic oligo-
of the transforming growth factor-beta (TGF-β) superfam- dendroglial cells (Ubhi et al. 2010). Further, NTN and PSPN
ily. The members of the GDNF family differentially promote mRNAs have been reported to occur in oligodendroglial cell
survival and regulate differentiation in peripheral and central lines (Strelau and Unsicker 1999).
neuronal populations. In contrast with the other members
of the TGF-β superfamily, which signal through receptor
serine-threonine kinases, the GDNF family ligands acti- 5 I N S U L I N -L I K E G R OW T H FAC TO R S
vate intracellular signaling cascades via the receptor tyrosine
kinase Ret. The GDNF family members bind first to the The insulin-like growth factors (IGFs) are proteins with high
GDNF receptor α (GFRα), a GPI-anchored membrane pro- sequence similarity to insulin. Currently, two IGFs are known
tein, forming a heterodimer with the receptor tyrosin kinase (IGF-1 and IGF-2), along with a family of six high-affinity
c-ret. This complex activates the ras/raf and PI3 kinase sig- IGF-binding proteins (IGFBP 1–6). The IGFs are known to
naling pathways. GFRα1, GFRα2, GFRα3, and GFRα4 bind to IGF-1 and IGF-2 receptors, as well as to insulin recep-
are the co-receptors for GDNF, NT, ART, and PSPN, tors, insulin-related receptors, and possibly to further receptors.
respectively (Peterziel et al. 2007). Insulin-like growth factor-1R is a cell surface heterotetrameric
tyrosine kinase receptor coupled to numerous intracellular
second messenger pathways, including Raf/Ras-mitogen–
4.1 S C H WA N N C E L L S
activated protein kinase and phosphatidylinositol-3 kinase sig-
Glial cell–derived neurotrophic factor, GFRα1, GFRα2, naling cascades; hence, IGF-1R is thought to be crucial for cell
and the alternative Ret coreceptor NCAM are expressed by survival, whereas IGF-2 appears to have a tumor suppressor role,
Schwann cells (Iwase et al. 2005). Growth factor receptor α1 because mutated or deleted forms have been reported in several

214 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
forms of cancers (Capoluongo 2011). Expression of IGF-1 and receptors), known as p75NTR receptor, which is a member
-2, IGF-IR, and the IGFBPs is well documented during devel- of the TNF receptor/Fas/CD40 superfamily. The p75NTR
opment of the CNS (Sullivan et al. 2008). Cultured embry- receptor appears to modify Trk signaling when the two recep-
onic neurons and glia synthesize IGF-1 mRNA; however, only tor types are coexpressed. When expressed in the absence of
glia seems to produce IGF-2 mRNA (Rotwein et al. 1988). Trks, p75NTR seems to mediate responses to neurotrophins,
including promotion of apoptotic death. The neurotrophins
have long been implicated in neuronal survival during develop-
5.1 S C H WA N N C E L L S
ment, but they also play prominent roles within the postnatal
Schwann cells are a prominent source of IGF-1, which has brain. Thus, there is evidence that neurotrophins are involved
been shown to promote the survival of Schwann cells, for in the maintenance of the postnatal brain and neuronal plas-
example, after serum withdrawal (Rodrigues et al. 2010). ticity (von Bohlen und Halbach 2010).
Insulin-like growth factor-1 also stimulates the early events
in myelination; the fatty acid biosynthetic pathway is a major
6.1 S C H WA N N C E L L S
target of IGF-1 (Liang et al. 2007).
Schwann cells have long been known to promote nerve growth
and regeneration by the secretion of trophic molecules and
5.2 A S T RO C Y T E S
supportive extracellular matrix. Neurotrophin family members
Evidence is emerging for a role of the IGF system in astro- and their receptors exhibit distinct expression patterns dur-
glial cell biology. Astroglial cells express IGF-1 and IGFBP-2, ing Schwann cell development and regeneration as well as in
which are upregulated by progesterone, but downregulated cultured Schwann cells (Piirsoo et al. 2010). Moreover, recent
by dexamethasone (Chesik and De Keyser 2010). Insulin-like studies have identified neurotrophins as important regulators of
growth factor-1 reduces cAMP levels through a beta2-adrener- peripheral (and CNS!) myelination (Xiao et al. 2009). Because
gic receptor–dependent process (Chesik et al. 2008). Reactive Wallerian degeneration is now being reinvestigated with
astrocytes in the spinal cord of amyotrophic lateral sclerosis molecular perspectives on inflammatory events, neurotrophins
transgenic rats upregulate IGF-2-R (Dagvajantsan et al. 2008). secreted by Schwann and other cells are once again gaining
Insulin-like growth factor-1 signaling in astrocytes is modulated momentum (Gaudet et al. 2011). Very recently, Schwann cells
by PTEN, a phosphatase which inhibits Akt activation, and in have also been targeted by injection of adeno-associated vector
turn is positively regulated by PI3K (Fernandez et al. 2008). (AAV) pseudotypes as a gene therapy strategy for peripheral
nerve regeneration (Homs et al. 2011). Schwann cells express
the pan-neurotrophin receptor p75NTR, which is massively
5.3 O L I G O D E N D RO C Y T E S upregulated following peripheral nerve lesions, thereby limit-
Oligodendrocytes express IGF-1 receptors as well as IGF-1 ing neurotrophin availability (Scott and Ramer 2010).
mRNA and IGF-1 protein, which elicit distinct effects on
developing oligodendrocytes (Du and Dreyfus 2002). Insulin-
like growth factor-1 increases proliferation, inhibits apoptosis, 6.2 A S T RO C Y T E S
and promotes differentiation of oligodendrocytes and their Astrocytes from several brain areas, including the hippocam-
precursor cells, suggesting an important function of IGFR-1 pus, are capable of synthesizing NGF and NT-3. Developing
in myelination. Vice versa, deficits in IGF-1 cause decreased and lesioned astrocytes have been shown to express p75, but
cell proliferation in the CNS white matter and decreased sur- not TrkA. Activation of p75NTR by NGF induces cell cycle
vival of newborn oligodendrocytes (Hua et al. 2009). Hence, arrest of dividing astrocytes (Cragnolini et al. 2011). BDNF–
IGF-1 holds a therapeutic potential in demyelinating and neu- expressing astroglia cells have been detected in the spinal cord
rodegenerative diseases (Chesik et al. 2008). An additional and the brain (Du and Dreyfus 2002). In the hippocampus,
“side effect” may be the neurotrophic effect of oligodendro- BDNF has been reported to restore astroglial S100 in a rat
cyte-derived IGF-1 on neurons (Wilkins et al. 2001). model of depression (Ye et al. 2011). Likewise, treatment of
astrocytes with the antidepressants fluoxetine and paroxetine
upregulates BDNF in astrocytes (Allaman et al. 2011).
6 N E U R OT R O P H I N S
6.3 O L I G O D E N D RO C Y T E S
The family of neurotrophins consists of nerve growth factor
(NGF), brain-derived neurotrophic factor (BDNF), neu- Oligodendrocytes have been identified to express NGF,
rotrophin 3 (NT-3), and neurotrophin 4 (NT-4). The neu- BDNF, NT-3, and NT-4 mRNA. Concerning BDNF, it
rotrophins bind to specific receptors that belong to the class was shown that it is expressed by oligodendrocytes myelinat-
of the Trk family of tyrosine protein kinase receptors. NGF ing the central processes of sensory neurons. Recent studies
specifically recognizes trkA, whereas BDNF and NT-4 spe- support the concept that BDNF plays a role in demyelinat-
cifically activate trkB receptors. NT-3 primarily activates trkC ing lesions by regulating the number of oligodendrocyte pro-
receptors. In addition, all neurotrophins can signal through a genitors and the abilities of demyelinating and differentiating
low-affinity receptor (which is structurally unrelated to the trk cells to express myelin proteins (VonDran et al. 2011). In

S TO R AG E A N D R E L E A S E O F N O N T R A N S M I T T E R S I G N A L I N G M O L E C U L E S F R O M M AC R O G L I A • 215
addition to expressing neurotrophins, oligodendrocytes also inter alia, leukemia inhibitory factor (LIF), cardiotrophin-1,
express neurotrophin receptors (trkA, trkB, trkC, as well as interleukin 6 (IL-6), and oncostatin-M. In the context of this
p75NTR), suggesting possible autocrine signaling (Du and chapter, data are also incorporated on cytokines and growth
Dreyfus 2002). factors unrelated to the CNTF family, often termed inflam-
matory cytokines (see chapter 22). Ciliary neurotrophic factor
is a potent survival factor for neurons and oligodendrocytes
7 P L AT E L ET-D E R I VE D G R OW T H FAC TO R and is relevant in reducing tissue destruction during inflam-
matory attacks. LIF regulates the differentiation of myeloid
Platelet-derived growth factor (PDGF) signals through two leukemic cells, but also acts as a regulatory cytokine in the
types of the alpha- and beta-type. Currently, five different iso- CNS. Cardiotrophin-1 is associated with the pathophysiology
forms of PDGF are known that can activate the two different of heart diseases, but also relevant to neuron survival. IL-6 acts
receptors: PDGFA, PDGFB, PDGFC, PDGFD, and a PDGFAB as both a proinflammatory and antiinflammatory cytokine,
heterodimer. The different isoforms seem to be expressed dif- which is secreted by T cells and macrophages to stimulate
ferently within the brain and it is thought that many neurons immune response. However, aside from this well-known func-
express the A and/or B chains of PDGF, whereas astrocytes tion, IL-6 acts as a regulatory cytokine within the CNS. The
express the A chain. members of the family of neuroregulatory cytokines share,
in part, receptor and transduction components. CNTF, for
example, binds to a complex consisting of a glycophosphati-
7.1 S C H WA N N C E L L S
dylinositol anchored α receptor, which is not involved in sig-
Cultured Schwann cells secrete PDGF and express PDGF nal transduction, the LIF receptor β (LIFRβ) and gp130. LIF
receptors. In neonatal rats, Schwann cells in the DRGs and binds to a complex consisting of LIFRβ and gp130.
sciatic nerve synthesize PDGF, but the expression of PDGF
protein declines postnatally. In adult rats, PDGF is low in
8.1 S C H WA N N C E L L S
myelinating Schwann cells, but is upregulated in nonmyelinat-
ing Schwann cells (Eccleston et al. 1993). Responses to PDGF CNTF is a classic growth factor produced by Schwann cells
are not seen until the precursor/Schwann cell transition both in vivo and in vitro and is induced following lesion-
(Woodhoo et al. 2004). PDGFR is prominently upregulated ing. Schwann cells are also a prominent source of IL-6 and
in crushed nerves (Oya et al. 2002; Yamazaki et al. 2009). IL-6R. Interestingly, factors belonging to the class of IL-6-
related regulatory cytokines, including CNTF, LIF, car-
7.2 A S T RO C Y T E S
diotrophin-1, and oncostatin M, have been found to exert
strong promyelinating effects (Stankoff et al. 2002). Human
Information on PDGF and PDGFRs in astrocytes is scarce. fetal Schwann cells constitutively express IL-1β, -6, -8, -11,
Levels are apparently low, but can be induced, for example, by -12, -15, and the receptors for IL-1, -4, -6, -8, 13, TNFα,
the HIV protein Tat (Bethel-Brown et al. 2011). In a subpopu- and gp130 (Ozaki et al. 2008). Tumor necrosis factor has
lation of astroglia located in the optic nerve head, light expo- been shown to inhibit Schwann cell proliferation by upregu-
sure has been reported to induce PDGF (Kernt et al. 2010). lating Src-suppressed protein kinase C substrate expression
When overexpressed in brain under the control of the human (Tao et al. 2009). Moreover, TNF-α plays pivotal roles in the
GFAP promoter, PDGFB induces glioblastomas (Hede et al. interactions of Schwann cells with axons. Apparently, TNF-α
2009). For further details concerning gliomas, see chapter 59. mediates its effects by orchestrating axon guidance. Schwann
cells in TNF-deficient peripheral nerve bundles fail to envelop
axons efficiently, an effect that can be rescued by exogenous
7.3 O L I G O D E N D RO C Y T E S
TNF-α. A large number of diverse pathological conditions
In 1988, PDGF was discovered to promote division and can be affected by cytokines from Schwann cells, making these
motility and inhibit premature differentiation of the oligo- molecules potential targets for treating peripheral neuropa-
dendrocyte/type-2 astrocyte progenitor cell. Oligodendroglia thies. The gene CXCL14, for example, is exclusively expressed
express PDGFRβ protein, and PDGF is capable of stimulating by Schwann cells and very prominently upregulated in PMP22
proliferation of oligodendrocytes and their precursors, respec- overexpressing mouse mutants, which model the pathogenesis
tively. Stimulation of proliferation is accompanied by inhibi- of Charcot-Marie-Tooth disease type 1 (Barbaria et al. 2009).
tion of myelination (Wang et al. 2007b). Oligodendrocyte
precursors have been shown to migrate toward a PDGF 8.2 A S T RO C Y T E S
source (Zhang et al. 2004).
Ciliary neurotrophic factor, LIF, and IL-6 have long been
known to be expressed by astroglial cells. These factors
8 R E GU L ATO RY C Y TO K I N E S promote astroglial differentiation, and CNTF is known to
induce an activated astrocyte phenotype. Certain astrocyte
The term neuroregulatory cytokines is often used for a large phenotypes may also promote myelination under certain
number of different growth factors, including the members of conditions (Nash et al. 2011). A large number of cytok-
the ciliary neurotrophic factor (CNTF) family, comprising, ines synthesized and released by astrocytes are important

216 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
players in immunological and inflammatory events in the signaling pathways, as for example p38, Jun N-terminal
CNS. Factors include IL-1β, -6, -8, -10, -17, -27, TNF-α, kinase, and mitogen-activated protein kinase pathways (Mu
IFN-γ, IFN-β, CCL-2, -3, -5, and CXCL-10 and -12 (Qin et al. 2012).
and Benveniste 2012). These factors are expressed at low
levels in the healthy brain, but are significantly elevated in
multiple sclerosis, Alzheimer and Parkinson disease, and 9.1 S C H WA N N C E L L S
amyotrophic lateral sclerosis (for more details on neurode- Schwann cells are well-established sites for the synthesis of
generative diseases, see chapters 63–65). An understanding TGF-β2 and -β3 and also carry the TGF-βRs (D’Antonio et al.
of molecular mechanisms underlying the regulation of astro- 2006). TGF-β has been implicated in the control of myeli-
glial cytokines is gradually emerging. Apparently, astrocytes nation and response of adult nerves to injury. TGF-β1 defi-
regulate the chemokine network in a pathogen-specific man- ciency results in gross abnormalities in myelination, whereas
ner (McKimmie and Graham 2010). Bacterial-associated axons are normal, suggesting that TGF-β1 may directly act
molecules induce preferentially CCL2, CXCL1, CCL20, on Schwann cells (Day et al. 2003). Lesioning downregulates
and CCL3, whereas virus-associated RNAs preferentially TGF-β2 (Stoll et al. 2004), which is involved in the regulation
upregulate CXCL10 and CCL5. Astrocytes themselves can- of myelin phagocytosis by macrophages.
not respond to these chemokines; they lack most chemokine A landmark study has recently shown that Schwann cells
receptors, but express CXCR4, CXCR6, and CXCR7 at of nerves traversing the hematopoietic niche are responsible
rest. Astroglia-derived cytokines are also implicated in the for activating latent TGF-β, thereby maintaining hematopoi-
regulation of protease-activated receptors (PAR) on astro- etic stem cell hibernation (Yamazaki et al. 2011). With regard
glia, which play important roles during development and to further mechanisms that regulate TGF-β signaling, the
in various inflammatory and neurodegenerative disorders protooncogene Ski, an inhibitor of TGF-β signaling, is cru-
(Sokolova et al. 2012). Cytokines from astroglia, for exam- cially involved in the control of Schwann cell proliferation and
ple, IL-1β and TNF-α, also regulate the expression of the myelination (Atanasoski et al. 2004).
astroglial (and neuronal) p75 receptors (Choi and Friedman Schwann cells are also the source of a novel member of the
2009). TGF-β superfamily, GDF-15, which is required for postnatal
maintenance of motoneurons in the spinal cord and sensory
8.3 O L I G O D E N D RO C Y T E S neurons in spinal ganglia (Strelau et al. 2009). In addition,
Schwann cells produce BMP-6, which is axonally transported
Ciliary neurotrophic factor and LIF promote differentia- by motoneurons and supports their survival in vitro (Wang et
tion of oligodendrocytes and oligodendrocyte progenitors. al. 2007a). Cell-specific elimination of the TβR II on Schwann
Overexpression of CNTF in oligodendrocytes has been cells has revealed reduced proliferation and ontogenetic cell
exploited for remyelination and successful functional recovery death (D’Antonio et al. 2006).
after spinal cord injury (Cao et al. 2010). Interleukin-11 has been
described as a potent factor for promoting survival, maturation,
and myelin formation in oligodendrocytes (Zhang et al. 2006).
9.2 A S T RO C Y T E S
For inducing apoptosis of oligodendroglial progenitors, exem-
plified by the OLI-neu cell line, TNF-α and TGF-β cooperate TGF-β1, -β2, and -β3 are expressed by astrocytes, and astro-
(see TGF-β in section 9). cytes respond to TGF-β (Krieglstein 2006). Astroglia-derived
TGF-βs execute multiple functions in the healthy and dis-
eased brain. An autocrine/paracrine loop with TGF-β
9 T R A N S F O R M IN G G R OW T H FAC TO R - Β regulates one of the most prominent markers of astroglia,
GFAP, involving cooperation between canonical and non-
Transforming growth factor-β (TGF-β) comprises a group canonical TGF-β pathways (Stipursky and Gomes 2007).
of more than 30 proteins that regulate many developmental During development, TGF-β has been hypothesized to foster
processes, tissue homeostasis and repair (Krieglstein, 2006). radial glia-astrocyte transformation (Stipursky and Gomes
TGF-βs are divided into two major subgroups. The first 2007). A neurotrophic role for TGF-β is well established
group consists of the TGF-βs, activins, nodal and myosta- (Krieglstein 2006). For example, TGF-βs from astroglia
tin. The second group comprises the bone morphogenetic promote the survival of midbrain dopaminergic and other
proteins (BMPs) and growth differentiation factors (GDFs). neurons. Interestingly, astrocytes from different brain regions
Within this superfamily, TGF-βs proper build a subfamily seem to have different trophic efficacies and differ with
of three members, TGF-β1, -β2, and -β3. TGF-βs are syn- respect to their content or releasable pool of TGF-β. Release
thesized as pre-proproteins containing a signal peptide and of TGF-β from astrocytes can be markedly induced by FGF-2
a C-terminally located mature part. TGF-β1, TGF-β2, and (Dhandapani et al. 2002). The effect of FGF-2 is mediated
TGF-β3 bind to heterodimers of TGF-β receptors (TβRs) by MEK/ERK signaling and activation of AP-1. Similarly,
I and II. After activation, TβR II phosphorylates and S100B, when increased in the extracellular space, has the
recruits TβR I into a heterotetrameric complex that acti- capacity to upregulate TGF-β in astrocytes (Reali et al.
vates itself as well as downstream elements, such as Smads 2011). Angiotensin II induces TGF-β expression in astro-
(Krieglstein 2006). In addition, TGF-βs activate non-Smad cytes via activation of thrombospondin-1 (Lanz et al. 2010),

S TO R AG E A N D R E L E A S E O F N O N T R A N S M I T T E R S I G N A L I N G M O L E C U L E S F R O M M AC R O G L I A • 217
Astrocytes: 4-sulfated proteoglycans released from astrocytes. Moreover,
TGF-α, EGF, FGF-2, FGF-9, GDNF, TGF-β specifically upregulates CXCR6 expression in astro-
IGF-1, IGF-2, NGF, NT-3, BDNF,
cytes (McKimmie and Graham 2010). High glucose induces
PDGF, LIF, CNTF, interleukins, TGF-
β1, TGF-β2, TGF-β3, BMPs the expression of multiple growth factors and cytokines in
astrocytes; surprisingly, however, TGF-β does not seem to be
induced (Wang et al. 2012). TGF-β released from astrocytes
is also an important means of communication with microglia
(Liu et al. 2011). In the lesioned brain, inhibition of TGF-β,
for example, by small molecule inhibitors of the TβR-I, sup-
presses formation of the fibrotic scar and promotes regen-
eration of nigrostriatal dopaminergic axons (Yoshioka et al.
2011). TβR-I immunoreactivity has been reported to be
induced in astrocytes of patients with temporal lobe epilepsy
(Lu et al. 2009). TGF-β is increased with age, most notably
Oligodendrocytes: in astrocytes (together with activated microglia and mac-
Neuregulin, FGF-9, GDNF, rophages), and is upregulated following stroke (Doyle et al.
neurturin, persephin, IGF- 2010).
1, BDNF, NGF, NT-3, NT-4,
TGF-β1, TGF-β2, TGF-β3,
In addition to TGF-βs, BMP-2, -4, the BMP scavenger
BMPs noggin, and the BMPRs IA, IB, and II are detected on both
neurons and astrocytes. BMP-7 within the locus coeruleus is
most prominently expressed in astrocytes relative to neurons
and oligodendrocytes, and cell-specifically reduced in both
human subjects with major depression disorder and in rats
Schwann cells:
exposed to chronic social defeat (Ordway et al. 2011).
FGF-1,FGF-2, FGF-5, GDNF,
IGF-1, NGF, BDNF,
persephin, PDGF, CNTF, 9.3 O L I G O D E N D RO C Y T E S
interleukins, TGF-β2, TGF-
β3, GDF-15, BMP
All three TGF-β isoforms are expressed by oligodendrocytes
(Du and Dreyfus 2002). For the generation of new mature oli-
godendrocytes, TGF-β is crucial (Gao et al. 2006) and has an
important impact on the regulation of the balance between
Figure 18.1 Schematic Overview of the Expression of Growth Factors of oligodendrocyte progenitor proliferation and differentiation,
Macroglial Origin. A tight network of bidirectional neuron–glial and employing a Notch-Jagged1 mediated pathway and astrocytes
interglial interactions is present in central and peripheral nervous tissue.
BDNF, brain-derived neurotrophic factor; BMP, bone morphogenetic
as an intermediate (Zhang et al. 2010).
protein; CNTF, ciliary neurotrophic factor; EGF, epidermal growth TGF-β also acts as a key proapoptotic molecule in oli-
factor; FGF, fibroblast growth factor; GDF, growth differentiation fac- godendroglial progenitor cells and the oligodendroglial cell
tor; GDNF, glia cell line–derived neurotrophic factor; IGF, insulin-like line OLI-neu (Schulz et al. 2009). The underlying mecha-
growth factor; LIF, leukemia inhibitory factor; NGF, nerve growth nism implies a Bcl-xl interaction with Fractin. Interestingly,
factor; NT, neurotrophin; PDGF, platelet-derived growth factor; TGF,
transforming growth factor.
TGF-β1 and the superfamily member ActivinA induce apop-
tosis in oligodendrocytes by different pathways (Schulz et
al. 2008). ActivinA acts autonomously, without cooperating
thereby sustaining brain inflammation in a mouse model with TGF-β. In the TGF-β death pathway, downregulation
of multiple sclerosis (concerning multiple sclerosis, see also of Bcl-xl is involved, whereas Bcl-xl in the Activin A–induced
chapter 61). Consequently, inhibitors of the angiotensin II pathway is sequestered by the BH3-only protein Puma. In the
type 1 receptor (which are prescribed as antihypertensives) apoptotic TGF-β pathway, caspase-3 is activated, whereas in
can interrupt this TGF-β–mediated proinflammatory path- the ActivinA pathway, apoptosis-inducing factor is released
way. As in nonneural tissues, TGF-β regulates extracellular to trigger DNA fragmentation. An intermediate in TGF-β
matrix proteins and metalloproteinases (Hsieh et al. 2010), signaling in oligodendroglial progenitors is the zinc finger
and serves as a molecular link between vascular permeabil- protein Tieg3/Klf11, which enhances TGF-β signaling by
ity and astroglial scar formation (Schachtrup et al. 2010). transcriptional repression of the inhibitory Smad7. Loss of the
Upregulation of chondroitin sulfate proteoglycans in astro- N-terminal repression domains of Tieg3/Klf11 abrogates the
cytes, which are inhibitory to axonal regeneration, are promi- proapoptotic effect of this transcription factor and abolishes
nently upregulated by stimulation with TGF-β (Susarla et the enhancement of Smad-mediate TGF-β responses (Bender
al. 2011). There is, however, a differential dependence of the et al. 2004; Gohla et al. 2008). Following a spinal cord con-
synthetic machinery on Smad2 and Smad3: TGF-β induc- tusion, BMP-2/4 is elevated in oligodendrocytes—and astro-
tion of neurocan and xylosyl transferase 1 requires Smad2, cytes around the injury site (Matsuura et al. 2008). The BMP
but not Smad3. Smad3 knockdown selectively reduces induc- scavenger noggin permits enhanced locomotor activity and
tion of chondroitin-4-sulfotransferase 1 and amounts of regrowth of the corticospinal tract, suggesting that BMP plays

218 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
an inhibitory role during axon regrowth and limits functional Capoluongo E. 2011. Insulin-like growth factor system and sporadic
recovery. Similarly, BMP-4, 6, and 7 are upregulated in spinal malignant melanoma. Am J Pathol 178:26–31.
Ceyhan GO, Bergmann F, Kadihasanoglu M, Erkan M, Park W, Hinz
cord autoimmune encephalitis in oligodendrocytes as well as U, et al. 2007. The neurotrophic factor artemin influences the
macrophages and astrocytes (Ara et al. 2008). extent of neural damage and growth in chronic pancreatitis. Gut
56:534–544.
Chen LW, Zhang JP, Kwok-Yan Shum D, Chan YS. 2006.
10 S U M M A RY A N D P E R S P E C T I VE S Localization of nerve growth factor, neurotrophin-3, and glial
cell line-derived neurotrophic factor in nestin-expressing reac-
tive astrocytes in the caudate-putamen of 1-methyl-4-phenyl-1-
The past decade has seen a great increase in the number of ,2,3,6-tetrahydropyridine-treated C57/Bl mice. J Comp Neurol
growth factors, cytokines, and respective receptors produced 497:898–909.
by and acting on macroglial cells. Although much has been Chesik D, De Keyser J. 2010. Progesterone and dexamethasone dif-
learned about regulation of their expressions (Fig. 18.1) and ferentially regulate the IGF-system in glial cells. Neurosci Lett
468:178–182.
implications for development and disease, significant gaps Chesik D, Wilczak N, De Keyser J. 2008. IGF-1 regulates cAMP levels
remain on both molecular/cellular and systems levels. For in astrocytes through a beta2-adrenergic receptor-dependant mech-
example, modes of intracellular storage and mechanisms of anism. Int J Med Sci 5:240–243.
release are enigmatic for many of these proteins. We also have Choi S, Friedman WJ. 2009. Inflammatory cytokines IL-1beta and
to admit that our knowledge on their regulatory and inter- TNF-alpha regulate p75NTR expression in CNS neurons and
astrocytes by distinct cell-type-specific signalling mechanisms. ASN
active networks is fragmentary—it almost does not exist. Neuro 1:e00010.
Comprehensive analyses based on systems biology approaches Clark MA, Gonzalez N. 2007. Angiotensin II stimulates rat astrocyte
have hardly been conducted. And, if properly planned, the mitogen-activated protein kinase activity and growth through EGF
respective strategies would definitely have to include contri- and PDGF receptor transactivation. Regul Pept 144:115–122.
butions from neurons. Thus, hopefully the coming years will Cornejo M, Nambi D, Walheim C, Somerville M, Walker J, Kim L, et al.
2010. Effect of NRG1, GDNF, EGF and NGF in the migration of a
see an expanded perception of glial cell biology with implica- Schwann cell precursor line. Neurochem Res 35:1643–1651.
tions of glia-derived growth factors both on the molecular and Cragnolini AB, Volosin M, Huang Y, Friedman WJ. 2011. Nerve
systems level. growth factor induces cell cycle arrest of astrocytes. Dev Neurobiol
72:766–776.
Dagvajantsan B, Aoki M, Warita H, Suzuki N, Itoyama Y. 2008.
Up-regulation of insulin-like growth factor-II receptor in reactive
REFERENCES astrocytes in the spinal cord of amyotrophic lateral sclerosis trans-
genic rats. Tohoku J Exp Med 214:303–310.
Allaman I, Fiumelli H, Magistretti PJ, Martin JL. 2011. Fluoxetine D’Antonio M, Droggiti A, Feltri ML, Roes J, Wrabetz L, Mirsky R, et al.
regulates the expression of neurotrophic/growth factors and glucose 2006. TGFbeta type II receptor signaling controls Schwann cell death
metabolism in astrocytes. Psychopharmacology (Berl) 216:75–84. and proliferation in developing nerves. J Neurosci 26:8417–8427.
Ara J, See J, Mamontov P, Hahn A, Bannerman P, Pleasure D, Grinspan Day WA, Koishi K, McLennan IS. 2003. Transforming growth fac-
JB. 2008. Bone morphogenetic proteins 4, 6, and 7 are up-regulated tor beta 1 may regulate the stability of mature myelin sheaths. Exp
in mouse spinal cord during experimental autoimmune encephalo- Neurol 184:857–864.
myelitis. J Neurosci Res 86:125–135. DeClue JE, Heffelfinger S, Benvenuto G, Ling B, Li S, Rui W, et al.
Atanasoski S, Notterpek L, Lee HY, Castagner F, Young P, Ehrengruber 2000. Epidermal growth factor receptor expression in neurofibro-
MU, et al. 2004. The protooncogene Ski controls Schwann cell pro- matosis type 1-related tumors and NF1 animal models. J Clin Invest
liferation and myelination. Neuron 43:499–511. 105:1233–1241.
Barbaria EM, Kohl B, Buhren BA, Hasenpusch-Theil K, Kruse F, Kury Dhandapani KM, Wade MF, Mahesh VB, Brann DW. 2002. Basic
P, et al. 2009. The alpha-chemokine CXCL14 is up-regulated in the fibroblast growth factor induces TGF-beta release in an isoform and
sciatic nerve of a mouse model of Charcot-Marie-Tooth disease type glioma-specific manner. Neuroreport 13:239–241.
1A and alters myelin gene expression in cultured Schwann cells. Doyle KP, Cekanaviciute E, Mamer LE, Buckwalter MS. 2010. TGFbeta
Neurobiol Dis 33:448–458. signaling in the brain increases with aging and signals to astrocytes and
Bender H, Wang Z, Schuster N, Krieglstein K. 2004. TIEG1 facilitates innate immune cells in the weeks after stroke. J Neuroinflammation
transforming growth factor-beta-mediated apoptosis in the oligoden- 7:1–13.
droglial cell line OLI-neu. J Neurosci Res 75:344–352. Du Y, Dreyfus CF. 2002. Oligodendrocytes as providers of growth fac-
Bethel-Brown C, Yao H, Callen S, Lee YH, Dash PK, Kumar A, et al. tors. J Neurosci Res 68:647–654.
2011. HIV-1 Tat-mediated induction of platelet-derived growth fac- Eccleston PA, Funa K, Heldin CH. 1993. Expression of platelet-derived
tor in astrocytes: role of early growth response gene 1. J Immunol growth factor (PDGF) and PDGF alpha- and beta-receptors in the
186:4119–4129. peripheral nervous system: an analysis of sciatic nerve and dorsal root
Brinkmann BG, Agarwal A, Sereda MW, Garratt AN, Muller T, Wende ganglia. Dev Biol 155:459–470.
H, et al. 2008. Neuregulin-1/ErbB signaling serves distinct functions Fernandez S, Garcia-Garcia M, Torres-Aleman I. 2008. Modulation by
in myelination of the peripheral and central nervous system. Neuron insulin-like growth factor I of the phosphatase PTEN in astrocytes.
59:581–595. Biochim Biophys Acta 1783:803–812.
Bryant MR, Marta CB, Kim FS, Bansal R. 2009. Phosphorylation and Furusho M, Kaga Y, Ishii A, Hebert JM, Bansal R. 2011. Fibroblast growth
lipid raft association of fibroblast growth factor receptor-2 in oligo- factor signaling is required for the generation of oligodendrocyte pro-
dendrocytes. Glia 57:935–946. genitors from the embryonic forebrain. J Neurosci 31:5055–5066.
Cao Q, He Q, Wang Y, Cheng X, Howard RM, Zhang Y, DeVries WH, Gao L, Macklin W, Gerson J, Miller RH. 2006. Intrinsic and extrinsic
et al. 2010. Transplantation of ciliary neurotrophic factor-expressing inhibition of oligodendrocyte development by rat retina. Dev Biol
adult oligodendrocyte precursor cells promotes remyelina- 290:277–286.
tion and functional recovery after spinal cord injury. J Neurosci Garré JM, Retamal MA, Cassina P, Barbeito L, Bukauskas FF, Saez JC,
30:2989–3001. et al. 2010. FGF-1 induces ATP release from spinal astrocytes in

S TO R AG E A N D R E L E A S E O F N O N T R A N S M I T T E R S I G N A L I N G M O L E C U L E S F R O M M AC R O G L I A • 219
culture and opens pannexin and connexin hemichannels. Proc Natl from light-induced overexpression of vascular endothelial growth
Acad Sci U S A 107:22659–22664. factor, platelet-derived growth factor, and placenta growth factor.
Gaudet AD, Popovich PG, Ramer MS. 2011. Wallerian degeneration: Growth Factors 28:211–220.
gaining perspective on inflammatory events after peripheral nerve Kinameri E, Matsuoka I. 2003. Autocrine action of BMP2 regulates
injury. J Neuroinflammation 8:110. expression of GDNF-mRNA in sciatic Schwann cells. Brain Res Mol
Gohla G, Krieglstein K, Spittau B. 2008. Tieg3/Klf11 induces apoptosis Brain Res 117:221–227.
in OLI-neu cells and enhances the TGF-beta signaling pathway by Krieglstein K. 2006. Transforming growth factor-βs in the brain. In:
transcriptional repression of Smad7. J Cell Biochem 104:850–861. Lajtha A, Lim R (eds.), Handbook of neurochemistry and molecular
Grothe C, Haastert K, Jungnickel J. 2006. Physiological function and neurobiology. New York: Springer, pp. 123–142.
putative therapeutic impact of the FGF-2 system in peripheral nerve Lanz TV, Ding Z, Ho PP, Luo J, Agrawal AN, Srinagesh H, et al. 2010.
regeneration—lessons from in vivo studies in mice and rats. Brain Res Angiotensin II sustains brain inflammation in mice via TGF-beta.
Rev 51:293–299. J Clin Invest 120:2782–2794.
Grothe C, Nikkhah G. 2001. The role of basic fibroblast growth factor in Leadbeater WE, Gonzalez AM, Logaras N, Berry M, Turnbull JE,
peripheral nerve regeneration. Anat Embryol (Berl) 204:171–177. Logan A. 2006. Intracellular trafficking in neurones and glia of fibro-
Haastert K, Grosheva M, Angelova SK, Guntinas-Lichius O, Skouras E, blast growth factor-2, fibroblast growth factor receptor 1 and hepa-
Michael J, et al. 2009. Schwann cells overexpressing FGF-2 alone or ran sulphate proteoglycans in the injured adult rat cerebral cortex.
combined with manual stimulation do not promote functional recov- J Neurochem 96:1189–1200.
ery after facial nerve injury. J Biomed Biotechnol 2009:408794. Li B, Du T, Li H, Gu L, Zhang H, Huang J, et al. 2008. Signalling path-
Hase A, Saito F, Yamada H, Arai K, Shimizu T, Matsumura K. 2005. ways for transactivation by dexmedetomidine of epidermal growth
Characterization of glial cell line-derived neurotrophic factor fam- factor receptors in astrocytes and its paracrine effect on neurons. Br J
ily receptor alpha-1 in peripheral nerve Schwann cells. J Neurochem Pharmacol 154:191–203.
95:537–543. Liang G, Cline GW, Macica CM. 2007. IGF-1 stimulates de novo
Heck N, Garwood J, Dobbertin A, Calco V, Sirko S, Mittmann T, et al. fatty acid biosynthesis by Schwann cells during myelination. Glia
2007. Evidence for distinct leptomeningeal cell-dependent paracrine 55:632–641.
and EGF-linked autocrine regulatory pathways for suppression of Limpert AS, Carter BD. 2010. Axonal neuregulin 1 type III activates
fibrillar collagens in astrocytes. Mol Cell Neurosci 36:71–85. NF-kappaB in Schwann cells during myelin formation. J Biol Chem
Hede SM, Hansson I, Afink GB, Eriksson A, Nazarenko I, Andrae J, 285:16614–16622.
et al. 2009. GFAP promoter driven transgenic expression of PDGFB Lin G, Goldman JE. 2009. An FGF-responsive astrocyte precursor iso-
in the mouse brain leads to glioblastoma in a Trp53 null background. lated from the neonatal forebrain. Glia 57:592–603.
Glia 57:1143–1153. Liu W, Tang Y, Feng J. 2011. Cross talk between activation of microglia
Hisaoka K, Tsuchioka M, Yano R, Maeda N, Kajitani N, Morioka N, and astrocytes in pathological conditions in the central nervous sys-
et al. 2011. Tricyclic antidepressant amitriptyline activates fibro- tem. Life Sci 89:141–146.
blast growth factor receptor signaling in glial cells: involvement in Lu Y, Xue T, Yuan J, Li Y, Wu Y, Xi Z, et al. 2009. Increased expression
glial cell line-derived neurotrophic factor production. J Biol Chem of TGFbeta type I receptor in brain tissues of patients with temporal
286:21118–21128. lobe epilepsy. Clin Sci (Lond) 117:17–22.
Höke A, Gordon T, Zochodne DW, Sulaiman OA. 2002. A decline in Matsuura I, Taniguchi J, Hata K, Saeki N, Yamashita T. 2008. BMP
glial cell-line-derived neurotrophic factor expression is associated inhibition enhances axonal growth and functional recovery after spi-
with impaired regeneration after long-term Schwann cell denerva- nal cord injury. J Neurochem 105:1471–1479.
tion. Exp Neurol 173:77–85. Mayer SI, Rossler OG, Endo T, Charnay P, Th iel G. 2009.
Homs J, Ariza L, Pages G, Udina E, Navarro X, Chillon M, et al. Epidermal-growth-factor-induced proliferation of astrocytes requires
2011. Schwann cell targeting via intrasciatic injection of AAV8 as Egr transcription factors. J Cell Sci 122:3340–3350.
gene therapy strategy for peripheral nerve regeneration. Gene Ther McKimmie CS, Graham GJ. 2010. Astrocytes modulate the chemo-
18:622–630. kine network in a pathogen-specific manner. Biochem Biophys Res
Hsieh HL, Wang HH, Wu WB, Chu PJ, Yang CM. 2010. Transforming Commun 394:1006–1011.
growth factor-beta1 induces matrix metalloproteinase-9 and Morita M, Kozuka N, Itofusa R, Yukawa M, Kudo Y. 2005. Autocrine
cell migration in astrocytes: roles of ROS-dependent ERK- and activation of EGF receptor promotes oscillation of glutamate-induced
JNK-NF-kappaB pathways. J Neuroinflammation 7:88. calcium increase in astrocytes cultured in rat cerebral cortex.
Hua K, Forbes ME, Lichtenwalner RJ, Sonntag WE, Riddle DR. 2009. J Neurochem 95:871–879.
Adult-onset deficiency in growth hormone and insulin-like growth Mu Y, Gudey SK, Landstrom M. 2012. Non-Smad signaling pathways.
factor-I alters oligodendrocyte turnover in the corpus callosum. Glia Cell Tissue Res 347:11–20.
57:1062–1071. Nakamura S, Todo T, Motoi Y, Haga S, Aizawa T, Ueki A, et al. 1999.
Irmady K, Zechel S, Unsicker K. 2011. Fibroblast growth factor 2 regu- Glial expression of fibroblast growth factor-9 in rat central nervous
lates astrocyte differentiation in a region-specific manner in the hind- system. Glia 28:53–65.
brain. Glia 59:708–719. Nash B, Thomson CE, Linington C, Arthur AT, McClure JD, McBride
Iwase T, Jung CG, Bae H, Zhang M, Soliven B. 2005. Glial cell MW, et al. 2011. Functional duality of astrocytes in myelination.
line-derived neurotrophic factor-induced signaling in Schwann cells. J Neurosci 31:13028–13038.
J Neurochem 94:1488–1499. Oh-hashi K, Ito M, Tanaka T, Hirata Y, Kiuchi K. 2009. Biosynthesis,
Joung I, Yoo M, Woo JH, Chang CY, Heo H, Kwon YK. 2010. Secretion processing, and secretion of glial cell line-derived neurotrophic factor
of EGF-like domain of heregulin beta promotes axonal growth and in astroglial cells. Mol Cell Biochem 323:1–7.
functional recovery of injured sciatic nerve. Mol Cells 30:477–484. Okada-Ban M, Th iery JP, Jouanneau J. 2000. Fibroblast growth factor-2.
Jungnickel J, Haase K, Konitzer J, Timmer M, Grothe C. 2006. Faster Int J Biochem Cell Biol 32:263–267.
nerve regeneration after sciatic nerve injury in mice over-expressing Ordway GA, Szebeni A, Chandley MJ, Stockmeier CA, Xiang L,
basic fibroblast growth factor. J Neurobiol 66:940–948. Newton SS, et al. 2011. Low gene expression of bone morphoge-
Kang K, Song MR. 2010. Diverse FGF receptor signaling controls astro- netic protein 7 in brainstem astrocytes in major depression. Int J
cyte specification and proliferation. Biochem Biophys Res Commun Neuropsychopharmacol 16(6):634–646.
395:324–329. Oya T, Zhao YL, Takagawa K, Kawaguchi M, Shirakawa K, Yamauchi
Kernt M, Liegl RG, Rueping J, Neubauer AS, Haritoglou C, Lackerbauer T, et al. 2002. Platelet-derived growth factor-b expression induced
CA, et al. 2010. Sorafenib protects human optic nerve head astrocytes after rat peripheral nerve injuries. Glia 38:303–312.

220 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Ozaki A, Nagai A, Lee YB, Myong NH, Kim SU. 2008. Expression Stipursky J, Gomes FC. 2007. TGF-beta1/SMAD signaling induces
of cytokines and cytokine receptors in human Schwann cells. astrocyte fate commitment in vitro: implications for radial glia devel-
Neuroreport 19:31–35. opment. Glia 55:1023–1033.
Peterziel H, Paech T, Strelau J, Unsicker K, Krieglstein K. 2007. Specificity Stoll G, Schroeter M, Jander S, Siebert H, Wollrath A, Kleinschnitz C,
in the crosstalk of TGFbeta/GDNF family members is determined et al. 2004. Lesion-associated expression of transforming growth
by distinct GFR alpha receptors. J Neurochem 103: 2491–2504. factor-beta-2 in the rat nervous system: evidence for down-regulating
Piirsoo M, Kaljas A, Tamm K, Timmusk T. 2010. Expression of NGF the phagocytic activity of microglia and macrophages. Brain Pathol
and GDNF family members and their receptors during peripheral 14:51–58.
nerve development and differentiation of Schwann cells in vitro. Strelau J, Strzelczyk A, Rusu P, Bendner G, Wiese S, Diella F, et al. 2009.
Neurosci Lett 469:135–140. Progressive postnatal motoneuron loss in mice lacking GDF-15. J
Qin H, Benveniste EN. 2012. ELISA methodology to quantify astro- Neurosci 29:13640–13648.
cyte production of cytokines/chemokines in vitro. Methods Mol Biol Strelau J, Unsicker K. 1999. GDNF family members and their recep-
814:235–249. tors: expression and functions in two oligodendroglial cell lines
Reali C, Pillai R, Saba F, Cabras S, Michetti F, Sogos V. 2011. S100B representing distinct stages of oligodendroglial development. Glia
modulates growth factors and costimulatory molecules expression in 26:291–301.
cultured human astrocytes. J Neuroimmunol 243(1–2):95–99. Sullivan KA, Kim B, Feldman EL. 2008. Insulin-like growth factors in
Reuss B, von Bohlen und Halbach O. 2003. Fibroblast growth factors the peripheral nervous system. Endocrinology 149:5963–5971.
and their receptors in the central nervous system. Cell Tissue Res Susarla BT, Laing ED, Yu P, Katagiri Y, Geller HM, Symes AJ. 2011.
313:139–157. Smad proteins differentially regulate transforming growth factor-be-
Rodrigues LS, da Silva ME, Moreira ME, Tempone AJ, Lobato LS, ta-mediated induction of chondroitin sulfate proteoglycans. J
Ribeiro-Resende VT, et al. 2010. Mycobacterium leprae induces Neurochem 119:868–878.
insulin-like growth factor and promotes survival of Schwann cells Tao T, Ji Y, Cheng C, Yang H, Liu H, Sun L, et al. 2009. Tumor necro-
upon serum withdrawal. Cell Microbiol 12:42–54. sis factor-alpha inhibits Schwann cell proliferation by up-regulating
Rotwein P, Burgess SK, Milbrandt JD, Krause JE. 1988. Differential Src-suppressed protein kinase C substrate expression. J Neurochem
expression of insulin-like growth factor genes in rat central nervous 111:647–655.
system. Proc Natl Acad Sci U S A 85:265–269. Timmer M, Robben S, Muller-Ostermeyer F, Nikkhah G, Grothe C.
Schachtrup C, Ryu JK, Helmrick MJ, Vagena E, Galanakis DK, Degen 2003. Axonal regeneration across long gaps in silicone chambers fi lled
JL, et al. 2010. Fibrinogen triggers astrocyte scar formation by pro- with Schwann cells overexpressing high molecular weight FGF-2.
moting the availability of active TGF-beta after vascular damage. Cell Transplant 12:265–277.
J Neurosci 30:5843–5854. Ubhi K, Rockenstein E, Mante M, Inglis C, Adame A, Patrick C, et al. 2010.
Schmid RS, McGrath B, Berechid BE, Boyles B, Marchionni M, Sestan Neurodegeneration in a transgenic mouse model of multiple system
N, et al. 2003. Neuregulin 1-erbB2 signaling is required for the estab- atrophy is associated with altered expression of oligodendroglial-derived
lishment of radial glia and their transformation into astrocytes in neurotrophic factors. J Neurosci 30:6236–6246.
cerebral cortex. Proc Natl Acad Sci U S A 100:4251–4256. Unsicker K, Reuss B, von Bohlen und Halbach O. 2006. Fibroblast growth
Schulz R, Vogel T, Dressel R, Krieglstein K. 2008. TGF-beta superfam- factors in brain functions. In: Lajtha A, Lim R (eds.), Handbook of
ily members, ActivinA and TGF-beta1, induce apoptosis in oligoden- neurochemistry and molecular neurobiology. Neuroactive proteins
drocytes by different pathways. Cell Tissue Res 334:327–338. and peptides. New York: Springer, pp. 93–122.
Schulz R, Vogel T, Mashima T, Tsuruo T, Krieglstein K. 2009. von Bohlen und Halbach O. 2010. Involvement of BDNF in
Involvement of Fractin in TGF-beta-induced apoptosis in oligoden- age-dependent alterations in the hippocampus. Front Aging Neurosci
droglial progenitor cells. Glia 57:1619–1629. 2:1–11.
Scott AL, Ramer MS. 2010. Schwann cell p75NTR prevents sponta- VonDran MW, Singh H, Honeywell JZ, Dreyfus CF. 2011. Levels of
neous sensory reinnervation of the adult spinal cord. Brain 133: BDNF impact oligodendrocyte lineage cells following a cuprizone
421–432. lesion. J Neurosci 31:14182–14190.
Seitz M, Grosheva M, Skouras E, Angelova SK, Ankerne J, Jungnickel Wang J, Li G, Wang Z, Zhang X, Yao L, Wang F, et al. 2012. High
J, et al. 2011. Poor functional recovery and muscle polyinnervation glucose-induced expression of inflammatory cytokines and reactive
after facial nerve injury in fibroblast growth factor-2-/- mice can be oxygen species in cultured astrocytes. Neuroscience 202:58–68.
improved by manual stimulation of denervated vibrissal muscles. Wang PY, Koishi K, McLennan IS. 2007a. BMP6 is axonally trans-
Neuroscience 182:241–247. ported by motoneurons and supports their survival in vitro. Mol Cell
Sharif A, Prevot V. 2010. ErbB receptor signaling in astrocytes: a media- Neurosci 34:653–661.
tor of neuron-glia communication in the mature central nervous sys- Wang Z, Colognato H, ffrench-Constant C. 2007b. Contrasting effects of
tem. Neurochem Int 57:344–358. mitogenic growth factors on myelination in neuron-oligodendrocyte
Shen LH, Li Y, Chopp M. 2010. Astrocytic endogenous glial cell derived co-cultures. Glia 55:537–545.
neurotrophic factor production is enhanced by bone marrow stromal Widera D, Heimann P, Zander C, Imielski Y, Heidbreder M, Heilemann
cell transplantation in the ischemic boundary zone after stroke in M, et al. 2011. Schwann cells can be reprogrammed to multipotency
adult rats. Glia 58:1074–1081. by culture. Stem Cells Dev 20:2053–2064.
Shin SY, Song H, Kim CG, Choi YK, Lee KS, Lee SJ, et al. 2009. Egr-1 Wilkins A, Chandran S, Compston A. 2001. A role for
is necessary for fibroblast growth factor-2-induced transcriptional oligodendrocyte-derived IGF-1 in trophic support of cortical neu-
activation of the glial cell line-derived neurotrophic factor in murine rons. Glia 36:48–57.
astrocytes. J Biol Chem 284:30583–30593. Woodhoo A, Dean CH, Droggiti A, Mirsky R, Jessen KR. 2004. The
Smith GM, Strunz C. 2005. Growth factor and cytokine regulation of trunk neural crest and its early glial derivatives: a study of survival
chondroitin sulfate proteoglycans by astrocytes. Glia 52:209–218. responses, developmental schedules and autocrine mechanisms. Mol
Sokolova E, Aleshin S, Reiser G. 2012. Expression of protease-activated Cell Neurosci 25:30–41.
receptor (PAR)-2, but not other PARs, is regulated by inflammatory Xiao J, Kilpatrick TJ, Murray SS. 2009. The role of neurotrophins in the
cytokines in rat astrocytes. Neurochem Int 60(3):276–285. regulation of myelin development. Neurosignals 17:265–276.
Stankoff B, Aigrot MS, Noel F, Wattilliaux A, Zalc B, Lubetzki C. 2002. Yamazaki S, Ema H, Karlsson G, Yamaguchi T, Miyoshi H, Shioda
Ciliary neurotrophic factor (CNTF) enhances myelin formation: S, et al. 2011. Nonmyelinating Schwann cells maintain hemato-
a novel role for CNTF and CNTF-related molecules. J Neurosci poietic stem cell hibernation in the bone marrow niche. Cell 147:
22:9221–9227. 1146–1158.

S TO R AG E A N D R E L E A S E O F N O N T R A N S M I T T E R S I G N A L I N G M O L E C U L E S F R O M M AC R O G L I A • 221
Yamazaki T, Sabit H, Oya T, Ishii Y, Hamashima T, Tokunaga A, et Zhang H, Vutskits L, Calaora V, Durbec P, Kiss JZ. 2004. A role for the
al. 2009. Activation of MAP kinases, Akt and PDGF receptors in polysialic acid-neural cell adhesion molecule in PDGF-induced che-
injured peripheral nerves. J Peripher Nerv Syst 14:165–176. motaxis of oligodendrocyte precursor cells. J Cell Sci 117:93–103.
Yan M, Dai H, Ding T, Dai A, Zhang F, Yu L, et al. 2011. Effects of dex- Zhang X, Neufeld AH. 2005. Activation of the epidermal growth factor
medetomidine on the release of glial cell line-derived neurotrophic receptor in optic nerve astrocytes leads to early and transient induc-
factor from rat astrocyte cells. Neurochem Int 58:549–557. tion of cyclooxygenase-2. Invest Ophthalmol Vis Sci 46:2035–2041.
Ye Y, Wang G, Wang H, Wang X. 2011. Brain-derived neurotrophic fac- Zhang Y, Taveggia C, Melendez-Vasquez C, Einheber S, Raine CS,
tor (BDNF) infusion restored astrocytic plasticity in the hippocam- Salzer JL, et al. 2006. Interleukin-11 potentiates oligodendro-
pus of a rat model of depression. Neurosci Lett 503:15–19. cyte survival and maturation, and myelin formation. J Neurosci
Yoshioka N, Kimura-Kuroda J, Saito T, Kawamura K, Hisanaga S, 26:12174–12185.
Kawano H. 2011. Small molecule inhibitor of type I transforming Zhang Y, Zhang J, Navrazhina K, Argaw AT, Zameer A, Gurfein BT,
growth factor-beta receptor kinase ameliorates the inhibitory milieu et al. 2010. TGFbeta1 induces Jagged1 expression in astrocytes via
in injured brain and promotes regeneration of nigrostriatal dopamin- ALK5 and Smad3 and regulates the balance between oligodendro-
ergic axons. J Neurosci Res 89:381–393. cyte progenitor proliferation and differentiation. Glia 58:964–974.

222 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
19.
PHYSIOLOGY OF MICROGLIA
Mami Noda and Alexei Verkhratsky

A B B R E VI AT I O N S NFAT nuclear factor of activated T cells


NK-1 neurokinin-1
Aβ amyloid β NMDA N-methyl-d-aspartate, N-methyl-d-aspartic
ADP adenosine diphosphate acid
AMPA α-amino-3-hydroxy-5-methyl-4-isoxazole NO nitric oxide
propionic acid PAF platelet-activating factor
AQ4 aquaporin4 PPADS pyridoxal-phosphate-6-azophenyl-2′,
ATP adenosine triphosphate 4′-disulfonate
AT2 angiotensin II receptors type 2 PrP prion protein
Best bestophine RANTES regulated on activation, normal T cell
BzATP 2′,3′-(benzoyl-4-benzoyl)-ATP expressed and secreted
CaRs plasmalemmal calcium receptors RyRs ryanodine receptors
C3a and C5a receptors to complement fragments TNF-α tumor necrosis factor-α
CB cannabinoid TGF-β transforming growth factor-β
CCR chemokine (C-C motif ) receptor Trk-B1 truncated tropomyosin-related kinase B-T1
CCL C-C motif chemokine ligand TRP transient receptor potential
CD200Rs CD200 receptors TRPM transient receptor potential cation channel
CLIC-1 chloride intracellular channel-1 subfamily M
COX-2 cyclooxygenase-2 TRPV transient receptor potential vanilloid
CREB cAMP response element binding protein TTX tetrodotoxin
CSF-1R colony stimulating factor-1 receptor VIP vasoactive intestinal peptide
CXCR C-X-C chemokine receptor Xc glutamate-cystine antiporter
CysLT cysteinyl leukotriene
EGFR epidermal growth factor receptor
ET endothelin 1 INTRODUCTION
FPR formyl peptide receptor
GABA gamma-aminobutyric acid Fetal macrophages/ameboid microglia, which migrate into
GLUT5 glucose transporter 5 the central nervous system (CNS) (see chapters 8 and 15),
GPCR G-protein–coupled receptors undergo fundamental phenotypic changes and become the
HIV-1 human immunodeficiency virus-1 innate immune cells generally referred to as microglia (Del Rio-
ICRAC Ca2+-release activated Ca2+ current Hortega 1932; Hanisch and Kettenmann 2007; Kettenmann
INa sodium currents et al. 2011). These changes in phenotype include a profound
IFN-γ interferon-gamma morphological metamorphosis that turns ameboid microglia
IKDR delayed rectifying K+ currents into highly specialized surveillance cells with small somata and
IK IR inward rectifier K+ currents long highly motile processes (“ramified” microglia), which are
IL-1 interleukin-1 accompanied with similarly profound changes in cell physiol-
InsP3 inositol 1,4,5-trisphosphate ogy. Microglia residing in the nervous system acquire numer-
LPA lysophosphatidic acid ous receptors to neurotransmitters, neuromodulators, and
LPS lipopolysaccharide neurohormones that allow them to monitor the normal activ-
MAPK mitogen-activated protein kinase ity of neural cells in search for possible signs of damage; mul-
MCP-1 monocyte chemotactic protein-1 tiple chemical factors originating from neurones, astrocytes
M-CSFR macrophage colony-stimulating factor and oligodendrocytes control microglial activation (Biber et al.
receptor 2007). The physiology of microglia is surveyed in this chapter,
mGluR metabotropic glutamate receptor including their membrane properties, expression of receptors
nAChRs nicotinic acetylcholine receptor and transporters; with specific focus on receptors related to
NCX Na+/Ca2+ exchanger CNS context. For many other aspects of microglial physiology,

223
including specific expression of immune-related molecules (e.g., The membrane properties of ameboid microglial cells
pattern recognition receptors) as well as for microglial surveil- studied in corpus callosum slices from early postnatal (6- to
lance–related motility, the reader is referred to other chapters 9-day-old) mice were more similar to those obtained in cul-
in this volume (see chapters 8, 15, 47, 48, 49, 50 and 53). tured microglia: these ameboid cells had more hyperpolarized
resting membrane potential (on average approximately –42
mV) and expressed a prominent inward rectifier potassium
2 M E M B R A N E P R O P E RT I E S current (Brockhaus et al. 1993).
OF MICROGLIAL CELLS Activation of microglial cells by pathological stimuli
induces substantial changes in their membrane properties.
The general membrane properties of microglial cells very In primary cultures, exposure to bacterial lipopolysaccharide
much depend on the experimental preparation and activa- (LPS) or interferon-gamma (IFN-J) induced the expression
tion status of microglia. In primary cultures from rodents and of an outward K+ current (Norenberg et al. 1992). Similarly,
humans (in which microglial cells acquire certain activation in experiments in situ, microglial activation in facial nucleus
status) the resting membrane potential is approximately –50 following a surgical lesion to the facial nerve led to substan-
mV (Kettenmann et al. 1990; Norenberg et al. 1994b). The tial increase in both the inward rectifier and outward delayed
membrane permeability of these cultured cells is dominated rectifier K+ currents. This increase was maximal 24 hours
by inward rectifying K+ currents. In contrast, microglial cells after surgery (Boucsein et al. 2000). Ischemic insult as well as
(identified by tomato lectin vital staining) in acutely isolated experimental status epilepticus caused similar upregulation in
slices from cortex, striatum, and facial nucleus, obtained from K+ currents 24 to 48 hours after the surgery/kainate injection
8- to 12-week-old rats, have very low membrane potential (Avignone et al. 2008; Lyons et al. 2000).
(on average approximately –20 mV), high input resistance,
and very small (if any) voltage-gated currents (Boucsein et
al. 2000). Similar properties were observed in microglia in 3 ION CHANNELS IN MICROGLIA
forebrain slices from 6- to 8-week-old mice; these cells lack
voltage-gated currents and had a relatively low membrane Microglial cells express multiple sets of ion channels
potentials (with two subpopulations having an average of Vm (Fig. 19.1), and this expression is often altered after micro-
approximately –52 mV and –29 mV) (Boucsein et al. 2003). glial activation. Functional expression of voltage-gated
Incidentally, holding microglial cells in situ at hyperpolar- sodium channels in microglia is debatable. Small sodium
ized potentials (–70 mV) lead to their rapid disintegration currents (INa) with peak amplitudes of approximately 50 pA
(Boucsein et al. 2000; Brockhaus et al. 1993). were described in cultured rat and human microglial cells

Ca2+-permeable
channels
CRAC K+ channels
Inward rectifier
Orai/Stim1 Kir2.1
TRPM2, TRPM6,
TRPM7,TRCPC6, Ca2+-dependent
TRPV1 BK/KCa1.1,
KCNN1-4/SK1-4

Na+ channels? Delayed rectifier


Nav1.1 Kv1.1,Kv1.2,
Nav1.5 Kv1.3,Kv1.5
Nav1.6

Aquaporins Anion channels


AQP4 CLIC-1, Best1-4

Proton channels
Hv1

Figure 19.1 Ion Channels Expressed in Microglia.

224 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
(Korotzer and Cotman 1992; Norenberg et al. 1994a). The also recorded from rodent-cultured microglia (Beck et al.
INa in cultured microglia was TTX-sensitive (complete inhi- 2008). There is evidence that TRPV1 channels initiate micro-
bition at 2–5 PM), and had a very rapid kinetics and charac- glial cell death, whereas TRPM2 channels somehow can be
teristic voltage dependence (threshold approximately –40 mV involved in microglial responses to ischemia (see Kettenmann
and peak approximately 0 mV). Sodium currents were found et al. 2011 for further details).
only in 20% of cultured rat microglial cells. Incidentally, the Potassium channels are widely expressed by microglial cells
cells with ramified morphology had a higher incidence of INa at various activation states. The inward rectifier K+ currents
detection (Korotzer and Cotman 1992). In human microglia (IKIR) are present in all types of activated and cultured micro-
(which were cultured from explants obtained during sur- glial cells. They are absent in resting microglia in situ. At the
gery on patients suffering from brain tumors; hence, these single channel level two unitary conductances (30 and 43 pS)
cells could have been pathologically modified), the majority with inward rectification have been identified (Kettenmann
of cells (30 out of 32 recorded) expressed TTX-sensitive INa et al. 2011 and references therein). The IKIR can be consid-
(Norenberg et al. 1994b). On a molecular level expression of ered as an early hallmark of microglial activation. Activation
TTX-sensitive Nav1.1 and Nav1.6 and TTX-resistant Nav1.5 of microglia by in vivo ischemia or facial nerve lesions results
channels was identified by specific antibodies in rat microglial in upregulation of inward rectifier current accompanied with
cultures (Black et al. 2009). The sodium channel Nav1.6 was a hyperpolarizing shift in the resting membrane potential
also found in activated microglia in autoimmune encepha- (Boucsein et al. 2000; Lyons et al. 2000). Similarly, in post-
lomyelitis (Craner et al. 2005). Nonetheless, the relevance natal (5- to 7-day-old) mice approximately 75% of resting
for Na+ channels in microglial physiology remains unclear microglial cells recorded in hippocampal slice had a relatively
because many laboratories failed to detect INa in resting as well small (mean current density approximately 3.6 pA/pF) IKIR .
as in activated microglia cells (see Kettenmann et al. 2011 for In activated microglia from the same organotypic slices cul-
further details). tured for 3 to 7 days the current was expressed in 90% of cells
The functional expression of voltage-gated Ca2+ channels, and its density increased threefold (to ~9.6 pA/pF) (Schilling
which were occasionally recorded from cultured microglia, and Eder 2007).
remains similarly questionable. There are also evidence indi- Activation of microglia (both in vitro and in situ) is also
cating upregulation of L-type Ca2+ channels in microglial accompanied by an appearance of delayed rectifying K+ cur-
cultures exposed to β-amyloid Aβ25–35, prion protein PrP 106– rents (IK DR), which most likely results from de novo synthesis
126 (Silei et al. 1999), HIV-1 regulatory protein Tat, or CCR5 of channel molecules (Boucsein et al. 2000; Lyons et al. 2000;
cytokine receptor agonist RANTES (CCL5) (Shideman et al. Norenberg et al. 1992). Cultured rat microglia express mRNA
2006). However, these observations remain isolated and most specific for Kv1.2, Kv1.3, and Kv1.5 channels; LPS-induced acti-
of the laboratories have not confirmed the presence of func- vation upregulates expression of Kv1.3 (Fordyce et al. 2005).
tional voltage-gated Ca2+ channels in microglia (Kettenmann In contrast, in vivo microglial activation resulted in an increase
et al. 2011). in immunoreactivity for Kv1.5 but not for Kv1.3 channels ( Jou
Microglial cells express another type of Ca2+-selective plas- et al. 1998). Incidentally, treatment of microglial cultures
malemmal channel responsible for the Ca2+-release activated with “calming” signaling agent transforming growth factor-β
Ca2+ current, ICRAC. The ICRAC was identified in whole-cell (TGF-β) induced a fivefold increase in the Kv1.3 mRNA level
patch-clamp experiments on cultured microglia (Norenberg and a sixfold increase in delayed rectifying K+ current density
et al. 1997). Subsequently, ICRAC was measured from both (Schilling et al. 2000). Amplitudes of IKDR and expression of
resting and activated mouse microglial cells. In the resting con- Kv1.3/Kv1.5 channels were also increased following treatment
ditions ICRAC amplitude was approximately 1.2 to 1.3 pA/pF; of cultured microglia with pneumococcal cell walls, β-amyloid
whereas LPS-induced activation significantly reduced the protein fragments 25 to 45, or experimentally induced (sys-
amplitude of the current to approximately 0.5 pA/pF 48 hours temic kainate injection) status epilepticus (see Kettenmann et
after exposure to LPS (Beck et al. 2008). The molecular sub- al. 2011 for further details). Manipulation with expression or
strate for ICRAC is represented by Orai proteins, which were function of delayed rectifier K+ channels affects various aspects
identified at the transcriptional level in cultured neonatal rat of microglial function. For example, antisense or knock-out
microglia. Electrophysiological and pharmacological studies deletion of Kv1.5 channels resulted in inhibition of NO release
indicated that highly Ca2+-selective (PCa/PNa > 1,000) Orai1/ from LPS activated microglia; deletion of Kv1.3 and Kv1.5
CRAC channels are responsible for ICRAC generation in micro- channels increased microglial proliferation. Pharmacological
glia (Ohana et al. 2009). inhibition of Kv1.3 channels reduce respiratory burst in cul-
Microglial cells are in possession of several types of tran- tured microglia (see Kettenmann et al. 2011 for review).
sient receptor potential (TRP) channels. At a transcriptional As for other voltage-gated K+ channels, human ether-
level (in cultured neonatal rat microglia) the following rank a-go-go–related gene product (HERG)-like K+ channel was
order of mRNA expression was found: TRPM7 > TRPC6 > reported in microglial cell line (Pennefather et al. 1998; Zhou
TRPM2 > TRPC1 > TRPC3 ≥ TRPC4 > TRPC7 > TRPC5 et al. 1998) and was also confirmed in primary cultured rat
> TRPC2 (Ohana et al. 2009). Activation of TRPM2 by microglia (Noda et al. unpublished data) as well as the expres-
H2O2 or by cADP ribose resulted in Ca2+ influx and cationic sion of Kv7 (KCNQ) channels (Noda et al. 2007b).
currents that were significantly potentiated by LPS (Kraft et Cultured microglia express several types of Ca2+-dependent
+
al. 2004). The TRPM4- and TRPM7-mediated currents were K channels represented by: (1) high-conductance (BK)

P H YS I O L O GY O F M I C R O G L I A • 225
channels, with reported single channel conductances of 106 2007). In addition, microglial cells were reported to express
pS, 149 pS, 187 pS, and 240 pS, depending on experimen- chloride intracellular channel-1 (CLIC-1) in the plasma mem-
tal preparation (Bordey and Spencer 2003; McLarnon et al. brane (unitary conductance 6.5–8 pS). Expression and activ-
1997); and (2) small conductance Ca2+/calmodulin acti- ity of CLIC-1 channels were potentiated by Aβ1–42 or Aβ25–35
vated K+ channels of KCNN4/KCa3.1/SK4/IK1 type (pre- peptides; inhibition of CLIC-1 channels reduced neurotoxic-
dominant type) and KCNN2/SK2 and KCNN3/SK3 types ity (Kettenmann et al. 2011; Milton et al. 2008).
(Kaushal et al. 2007; Khanna et al. 2001). The importance of The proton channels, characterized by an exceptional selec-
BK channels in microglia in bradykinin- and galanin-induced tivity to H+ ions, were detected in primary microglial cultures
migration (Ifuku et al. 2007, 2011) and nerve injury–induced from mice, rats, and humans (Eder and DeCoursey 2001).
neuropathic pain (Hayashi et al. 2011) was deduced based on These channels have a very small (fS range) unitary conduc-
various pharmacological assays. The immunoreactivity for tance and their function is inhibited following LPS-induced
KCNN3/SK3 was found in microglial cells in healthy adult microglial activation. It is generally assumed that microglial
rat striatum. This immunoreactivity increased after ischemic H+ currents play a role in the “respiratory burst” associated
lesion (Schlichter et al. 2010). The SK3 and SK4 channels are with phagocytosis (Eder and DeCoursey 2001). Activated
reportedly involved in the control of microglia activation and microglial cells have also been reported to express aquaporins
contribute to the upregulation of iNOS and the production of AQP4 type as well as Cx36 and Cx43 connexins; func-
of NO and peroxynitrite, which exert neurotoxic effects. The tional role of these channels remain unknown (Kettenmann
G-protein–activated K+ currents were also described in cul- et al. 2011).
tured murine microglia (Ilschner et al. 1995).
Outwardly rectifying, voltage- and time-independent
volume-sensitive Cl– currents were detected in cultured murine 4 R E C E P TO R S I N M I C R O G L I A
microglial cells, where they were suggested to regulate prolif-
eration and to be involved in setting the resting membrane
4.1 N EU ROT R A NS M IT T E R R EC E P TO R S I N
potential. These channels have a very small unitary conduc-
M I C RO G L I A
tance (1–3.5 pS) and are possibly represented by Bestophine
(Best) channel family; expression of Best 1–4 mRNAs have Acquisition of neurotransmitter receptors (Fig. 19.2) argu-
been detected in rat microglial cultures (Ducharme et al. ably represents the most profound phenotypical digression of

Purinoceptors

P2X4, Glutamate
P2Y2
P2X7 receptors
P2Y6 GluR1-4
P2Y12 Glu R5
A1, A2A, P2Y13
A2B, A3 mGluR5

InsP3 InsP3
Dopamine K+ mGluR2,3
receptors cAMP mGluR4,6,8
D1, D2 cAMP
D3, D4 cAMP
K+
InsP3
GABAB(1a),
InsP3 GABAB(1b),
GABAB(1c),
α1A,α2A
cAMP InsP3
GABA
β1, β2 receptors

5-HT2
Adrenoreceptors 7-TM
nAChR receptors
α3,α5,α7 Serotonin
β4 Ionotropic
receptors
receptors
Cholinoreceptors
Ion
channels

Figure 19.2 Neurotransmitter Receptors in Microglia.

226 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
microglia from their fetal macrophage ancestors. By express- intrathecal injection of activated cultured microglia expressing
ing neurotransmitter receptors, microglia adapt to the CNS P2X4 receptors triggered allodynia without the need of surgi-
environment and attain the ability to sense chemical signals cal nerve lesion (see Inoue and Tsuda 2009 and Kettenmann
employed in communications between neural cells. Microglial et al. 2011, for details).
neurotransmitter receptors can either trigger microglial acti- The pore-forming P2X7 receptors represent another type of
vation (“on” signals) or promote maintenance of resting, sur- purinoceptors specifically relevant for microglial function. The
veillance state (“off ” signals) (Biber et al. 2007; van Rossum P2X7 receptors have several unique features, which include:
and Hanisch 2004). (1) exceptionally low sensitivity to ATP; several mM of ATP
Probably the most widespread microglial neurotransmitter are needed to activate the receptor; (2) modulation of ATP
receptors are purinoceptors activated by adenosine triphos- sensitivity by extracellular divalent cations; and (3) ability of
phate (ATP), adenosine, and related nucleotides. ATP acts as P2X7 receptors to form large membrane pores (permeable to
a widespread neurotransmitter and is also an ancient “danger” molecules with mw up to 900 Da) on strong stimulation. The
signal because massive ATP release invariably accompanies pore formation may result from the dilatation of the channel
cell death. Numerous observations in vitro, in situ, and in vivo permeation path or from the activation of other pore-forming
have demonstrated that ATP and its analogs trigger rapid proteins (Pelegrin and Surprenant 2009). It has to be noted
functional responses of microglial cells, comprising fast con- that 2′,3′-(benzoyl-4-benzoyl)-ATP (BzATP) often used
verging movement of microglial processes toward the lesion, as a specific activator of P2X7 receptors, activates other
membrane ruffling, the outgrowth of microglial processes, P2X subunits with high potency. P2X7 receptors are widely
and the release of various biologically active substances, such expressed in the cells of the immune system and mediate many
as cytokines and inflammatory proteins (Davalos et al. 2005; immune reactions, including the processing and the release of
Kettenmann et al. 2011; Nimmerjahn et al. 2005). Microglial various cytokines. The P2X7 mediated currents were for
cells usually express several types of purinoceptors and expres- the first time identified in ameboid microglial cells in situ
sion pattern of these receptors changes with microglial activa- (Haas et al. 1996); simultaneously the P2X7-mediated [Ca2+]i
tion (Moller et al. 2000b). increases were found in freshly isolated mouse microglia
The ionotropic P2X purinoceptors are classic cati- (Ferrari et al. 1996).
onic (Na+/K+/Ca2+) ligand-gated channels assembled as In the healthy brain P2X7 receptors are present in micro-
homo-trimers or hetero-trimers from seven different subunits glial cells in all regions; brain damage triggers massive
classified as P2X1 to P2X7. The P2X receptors have substantial upregulation of microglial P2X7 expression (see Sperlagh
Ca2+ permeability, which may vary, depending on the subunit et al. 2006, for a review). Similarly, microglial P2X7 recep-
composition (PCa/Pmonovalent), between 1 and greater than 10 tors expression is significantly upregulated in various neuro-
(Pankratov et al. 2002, 2009), and could be even greater for pathologies, including multiple sclerosis, amyotrophic lateral
the pore-forming P2X7 receptors (Egan et al. 2006). The main sclerosis, and Alzheimer disease (see Kettenmann et al. 2011
type of ionotropic purinoceptors expressed in mature microg- for a detailed review). Activation of P2X7 receptors controls
lial cells are P2X4 and P2X7 receptors. multiple microglial functions. Stimulation of P2X7 receptors
The P2X4 receptors are fundamental for microglial activa- can trigger microglial cell death via apoptosis and autophagy.
tion and in particular have been found to be instrumental for Overexpression of P2X7 receptors triggers microglial activa-
pathogenesis of neuropathic pain. The genesis of neuropathic tion in the in vitro system; similarly P2X7 receptors are nec-
pain is associated with long-lasting activation of microglia in essary to launch microglial activation in response to in vivo
the spinal cord (Inoue and Tsuda 2009). The specific role for injections of Aβ protein (Monif et al. 2009; Sanz et al. 2009).
P2X4 receptors in mediating microglial activation in neuro- Finally, activation of P2X7 receptors can induce both neuro-
pathic pain was initially suggested following experiments on toxicity and neuroprotection in a context-dependent manner.
the rat spinal nerve injury model in which L5 spinal nerve was These functional responses are mediated through a variety of
surgically severed. The tactile allodynia (the symptom of neu- intracellular transcription factors (e.g., CREB, NFAT, MAP
ropathic pain) which developed following nerve lesion was kinases, etc.) to which P2X7 receptors are linked (Kettenmann
specifically alleviated following intrathecal injection of P2X et al. 2011). In addition, P2X7 receptor signaling controls basal
receptor antagonist TNP-ATP, while being not affected by and TNF-α–stimulated glial cell proliferation by regulating
another antagonist PPADS. This peculiar sensitivity of tac- AQP4 (Zou et al. 2012).
tile allodynia to P2 receptor antagonists (TNP-ATP inhibits Microglial cells express a variety of metabotropic P2Y
P2X1–4 receptors, whereas PPADS blocks all P2X receptors receptors, among which P2Y2, P2Y6, P2Y12, and P2Y13 seems
except P2X4) indicated the role for P2X4 receptors (Tsuda et to be dominating. Activation of these receptors induces Ca2+
al. 2003). Further investigations revealed a significant increase signaling, including [Ca2+]i oscillations and often triggers sec-
in P2X4 receptors expression in the spinal cord microglia fol- ondary store–operated Ca2+ influx. In ramified microglial cells
lowing nerve lesion. Similarly, increase in P2X4 receptors in in acute slices from adult mice activation of P2Y receptors
spinal cord microglia was observed in rats with experimental trigger an activation of outward rectifying K+ conductance
autoimmune neuritis and following intraperitoneal injection (Boucsein et al. 2003). The P2Y receptors regulate micro-
of LPS (which triggered hyperalgesia and microglia activa- glial release of cytokines and expression of immediate early
tion). An increased P2X4 immunoreactivity in the spinal cord genes. The UDP-preferring P2Y6 receptors regulate micro-
was confined to OC42 positive microglial cells; moreover, glial Ca2+ signaling and induction of phagocytosis (Koizumi

P H YS I O L O GY O F M I C R O G L I A • 227
et al. 2007). The ADP-preferring P2Y12 receptors (which in receptors are linked to microglial superoxide production via
the brain are predominantly expressed in microglia) control Nox activation to promote neuroprotection or neurotoxicity
early microglial responses to injury, being critical for induc- (Mead et al. 2012).
ing the morphological activation, membrane ruffling, and
chemotaxis (Kettenmann et al. 2011). The P2Y12 receptor con- 4.2 R EC E P TO R S F O R N EU RO H O R M O N E S A N D
trol integrin-β1 signaling cascade, which regulates extension N EU RO M O D U L ATO R S I N M I C RO G L I A
of microglial processes (Ohsawa et al. 2010). In the spinal cord
P2Y12 receptors are involved in the genesis of neuropathic pain Microglial expression of receptors for neurohormones and
(Inoue and Tsuda 2009). neuromodulators is summarized in Figure 19.3; the majority
Microglia also possess adenosine receptors. All four sub- of these receptors belong to 7-transmembrane G-protein–
types (A1, A2A, A2B, and A3) were detected in microglial cells coupled receptors (7-TM GPCRs), which are linked either to
from various species. Adenosine receptors control microglial PLC/InsP3 metabolism and Ca2+ signaling or coupled (via G
proliferation, K+ currents, release of prostaglandins and neu- proteins) to adenylate cyclase, K+ channels, MAP kinases, or
roprotective cascades (Kettenmann et al. 2011). other signaling cascades.
Microglial cells contain ionotropic glutamate receptors pre- The platelet-activating factor (PAF) receptors mediate
dominantly of AMPA/Kainate type; expression of all 4 GluR strong chemotactic response of microglial cells which involves
subunits for AMPA was detected in microglial cells in vitro. A G-protein and mitogen-activated protein kinase (MAPK)
minor subpopulation of cultured spinal microglia expressed signaling pathways. PAF receptors also induce robust
functional GluR5 kainate receptors; whereas GluR6,7 recep- [Ca2+]i transients originated from both ER Ca2+ release and
tors were found only in microglial cell line (Noda et al. 2000; store-operated Ca2+ entry (Aihara et al. 2000). Bradykinin
Yamada et al. 2006). Functional expression of NMDA recep- receptors of B2 subtype are constitutively expressed in micro-
tors in microglia remains questionable. In addition microglia glia. These receptors trigger InsP3-mediated Ca2+ signaling
express several types of metabotropic glutamate receptors with subsequent activation of K+Ca channels. Activation of
including mGluR5 linked to inositol 1,4,5-trisphosphate microglia induces synthesis of B1 bradykinin receptors and
(InsP3) production and Ca2+ signaling (Biber et al. 1999) and upregulation of B2 receptors expression. The B1 receptors in
mGluR2,3 (Group II) and mGluR4,6,8 (Group III) recep- activated microglia positively modulate Na+/Ca2+ exchanger
tors linked to cAMP signaling cascade; activation of group II as well as K+Ca channels and induce chemotaxis (Kettenmann
receptors triggered microglial activation, neurotoxicity and et al. 2011; Noda et al. 2003). The role of microglial B1 recep-
promoted the release of TNF-α, whereas activation of group tor is contradictory, either harmful as reported in diabetic pain
III receptors reduced microglial neurotoxicity following treat- neuropathy (Talbot and Couture 2011; Talbot et al. 2010) or
ment with LPS or chromogranin A (Kettenmann et al. 2011; neuroprotective (Noda et al. 2007a; Yasuyoshi et al. 2000).
Taylor et al. 2002). Expression of B1 receptors in the spinal cord microglia is also
Functional GABAB receptors (in all three splice variants upregulated following activation of TRPV1 channels induced
GABAB(1a), GABAB(1b) and GABAB(1c)) were found in cul- by capsaicin (Talbot et al. 2012). The role of kinins in the CNS
tured microglia and in about 50% of tomato-lectin positive is reviewed in Noda (2011a). Cultured microglial cells have
cells in brain slices; stimulation of GABAB receptors triggered histamine receptors linked to InsP3-mediated Ca2+ signaling
Ca2+ signaling and activated an outwardly rectifying K+ cur- (Kettenmann et al. 2011). The ETB endothelin receptors were
rent (Kuhn et al. 2004). Microglial cells also expressed several identified in mouse primary microglia. Stimulation of these
subunits of nicotinic acetylcholine receptors, including neu- receptors induced InsP3-mediated Ca2+ release from the ER
ronal α7nAChR subunit. Activation of nAChRs generally with subsequent store-operated Ca2+ entry in a subpopulation
inhibits the immune response of microglial cells thus repre- of microglia (Moller et al. 1997a). Microglial cells respond to
senting an endogenous “cholinergic antiinflammatory path- cannabinoids, although activation of CB1 and CB2 receptors
way” (Shytle et al. 2004). There are also some indication that coupled to Gi/o and Gi proteins, respectively; expression of
cultured microglia may possess muscarinic cholinoreceptors these receptors is strongly upregulated following microglial
linked to InsP3-mediated Ca2+ signaling (Kettenmann et al. activation. Generally, the activation of cannabinoid recep-
2011). Furthermore, microglial cells express α1A, α2A, β1, tors increases microglial proliferation (Carrier et al. 2004)
and β2 adrenoreceptors (Kettenmann et al. 2011; Tanaka et and reduces microglial neurotoxicity and neuroinflammation
al. 2002). The β2 receptors are positively coupled to adeny- (Stella 2008).
lyl cyclase, whereas α adrenoreceptors are coupled to Ca2+ The resting microglia express angiotensin II recep-
signaling. Selective stimulation of α1 or β1 or β2 receptors sup- tors type 2 (AT2), whereas microglial activation induces
pressed the expressions of IL-6 and TNF-α Microglial cells additional expression of type 1 angiotensin receptors AT1.
also contain D1,2,3,4 dopamine receptors involved in negative Pharmacological inhibition of AT1 receptors suppresses mor-
regulation of inward rectifier and positive regulation of out- phological activation of microglia, LPS-induced activation
ward K+ currents and also involved in chemotaxis (Kettenmann of NF-NB and reduces production of nitric oxide (NO) and
et al. 2011). Finally, functional expression of 5-HT2 sero- IL-1β (Miyoshi et al. 2008). Cultured rat microglial cells were
tonin receptors modulating K+ currents and triggering Ca2+ reported to express sst2, sst3 and sst4 types of somatostatin
signaling was recently discovered in murine microglial cells receptors, stimulation of which inhibits microglial prolifera-
(Krabbe et al. 2011). Some microglial neurotransmitter tion (Feindt et al. 1998). Several types of opioid receptors were

228 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
orexin
Platelet-activating receptors
factor(PAF) OX1R
receptor

Bradykinin Neurotrophin
receptors, receptors,
B1, B2 Trk-B1
InsP3/Ca2+ cAmP,
ruffling

InsP3/Ca2+
InsP3/Ca2+ VIP
Endothelin NCX receptors,
receptors, chemotaxis – VPAC1
ETB
InsP3/Ca2+ activation

NF-κB Neurokinin
(Substance P)
InsP3/Ca2+ receptors,
Histamine
chemotaxis NK-1
receptors,
migration
ETB ?
Proliferation
+ Opioid
receptors,
Angiotensin II KOR, MOR
receptors,
AT1, At2

Cannabinoid Somatostatin
receptors, receptors,
CB1, CB2 sst2,sst3,sst4 7-TM
receptors
Tyrosine
kinase
receptors

Figure 19.3 Receptors for Neurohormones and Neuromodulators in Microglia.

identified in primary human and feline microglia. The kappa- direct evidence of the expression of specific type of benzodiaz-
opioid receptors (KOR) regulate microglial immune response epine receptor has not been reported, benzodiazepine recep-
(specifically in HIV-1 dependent encephalopathies), whereas tor agonists decreased the ATP-induced increase of COX-2
mu-opioid receptor facilitate microglial activation and nega- gene expression and release of IL-1β and TNF-α (Choi et al.
tively regulate chemotaxis (Chao et al. 1997; Dobrenis et al. 2011). Likewise, neuropeptide Y receptors (Y1 receptor) are
1995). Neurokinin-1 (NK-1 or substance P) receptors are pres- supposed to inhibit interleukin-1β–induced phagocytosis in
ent in cultured murine and fetal human microglia; they control microglial cell line (Ferreira et al. 2011).
activation of transcriptional factor NF-NB, regulate produc-
tion of cytokines, and may enhance inflammatory responses
induced by bacterial infections of CNS (Kettenmann et al. 4.3 C Y TO K I N E S A N D C H E M O K I N E
2011). The VPAC1 type of vasoactive intestinal peptide (VIP) R EC E P TO R S I N M I C RO G L I A
receptors identified in cultured rat microglia, inhibit TNF-α Microglial cells are in possession of several types of cytokine
IL-1β and NO production and release in LPS-activated cells, and chemokines receptors (Fig. 19.4), which regulate a multi-
reduce microglial neurotoxicity and suppress microglial acti- tude of immune responses. The chemokines are chemoattrac-
vation in vivo (Delgado and Ganea 2003; Kettenmann et al. tive cytokines involved in regulation of cell migration, which
2011). Cultured rat microglia also express the subtype of Trk act as diffusable messengers able to create chemotactic gradi-
neurotrophin receptors, the truncated tropomyosin-related ents for cell migration. A distinction as to inflammatory and
kinase B-T1 (Trk-B1) receptors. Activation of these receptors homeostatic chemokines is based on the inducible versus con-
by BDNF triggers sustained [Ca2+]i elevation mediated by an stitutive expression. The chemokines are small proteins with
initial PLC/InsP3-driven Ca2+ release from the ER followed mw between 8 and 12 kDa that are classified into four groups
by a long-lasting activation of the store-operated Ca2+ entry. of C, CC, CXC, and CX3C chemokines (see Laing and
In addition, exposure of activated microglial cells to BDNF Secombes 2004 for a review). Chemokines are expressed in the
decreased release of NO (Mizoguchi et al. 2009). Although CNS in neurons, macroglial cells, and microglia. Microglial

P H YS I O L O GY O F M I C R O G L I A • 229
cells in particular express CCR1, CCR2, CCR7 and CCR5 a faster cognitive decline in Alzheimer disease (Westin et al.
(rodents), and CXCR1, CXCR3, and CCR3 (humans) 2012).
(Boddeke et al. 1999a,b; Flynn et al. 2003). Chemokine recep- Receptors for cytokines include tumor necrosis factor-α
tors are 7-transmembrane domain G-protein–coupled recep- receptors, interleukin receptors, and receptors to interferon
tors linked to multiple signaling cascades that include adenyl β and J. Microglial cells express two types of TNF-α recep-
cylase, phospholipases, GTPases (Rho, Rac, and Cdc42), tors, TNFR1 and TNFR2, which positively modulate
and some kinases such as mitogen-activated protein kinase microglial activation and phagocytosis (Kettenmann et al.
(MAPK) or phosphatidyl inositol-3 kinase (PI3-K) (Biber 2011). The interleukin-1 (IL-1) receptors are represented by
et al. 2008). Stimulation of cultured naïve and LPS treated IL-1 type-I receptor (IL-1RI), IL-1 type-II receptor (IL-1RII),
microglia with β-chemokines MCP-1 (or C-C chemokine and IL-1 receptor accessory protein (IL-1RAcP). At mRNA
CCL2; agonist of CCR2 receptors) or MIP-1α and RANTES level resting microglia express IL-1RII, and microglial activa-
(agonists of CCR5 receptors) resulted in Ca2+ signals. These tion upregulates expression of IL-1RI, IL-1RII, and IL-1RacP.
[Ca2+]i responses were larger in activated cells. The CCR5- In human microglial cells mRNA transcripts for multiple
dependent MIP-1α and RANTES-induced Ca2+ signals most interleukin receptors (IL-1RI, IL-1RII, IL-5R, IL-6R, IL-8R,
likely involve InsP3-dependent Ca2+ release, whereas MCP-1 IL-9R, IL-10R, IL-12R, IL-13R, and IL-15R) were identified
induced Ca2+ signaling was also sensitive to ryanodine and (Lee et al. 2002).
plasmalemmal Ca2+ influx (Boddeke et al. 1999b). In addition,
activation of chemokine receptors stimulated Ca2+-dependent
4.4 OT H E R R EC E P TO R S Y S T E M S I N M I C RO G L I A
K+ currents and activated volume-regulated Cl– channels
linked to microglial migration (see Kettenmann et al. 2011 for Microglial cells, studied in vitro, in situ, and in vivo, express
details). Expression of chemokine receptors is modified during many other receptors systems (Fig. 19.5) (see also Kettenmann
microglial activation and during neuropathology (Kremlev et al. 2011, for a detailed description and relevant references).
et al. 2004). For example, the levels of CCR5 receptors (on Microglia are endowed with glucocorticoid and mineralocor-
transcriptional level) were increased following hypoxic-isch- ticoid receptors mediating multiple pleiotropic effects (Tanaka
emic insults and nerve injury. Microglial cells from post-mor- et al. 1997). Cultured microglia also express plasmalemmal
tem Alzheimer disease brains had increased levels of CCR3 calcium receptors CaRs (the 7-transmembrane GPCRs acti-
and CCR5 receptors and increased levels of CCR5 receptors vated by extracellular Ca2+), which are involved in regulation
were also found in biopic material from patients with early of Ca2+-dependent K+ channels (Chattopadhyay et al. 1999).
stages of multiple sclerosis. Likewise, CCL2 (MCP-1) levels Cysteinyl leukotrienes receptors of CysLT1 and CysLT2
in the cerebrospinal fluid (CSF) at baseline correlated with types induced [Ca2+]i rise and ATP release from cultured rat

TNF-α receptors Chemokine receptors

TNFR1,2 CCR1,2

CCR5

Inhibition of Ca2+
activation,
neuroprotection K+ channels CXCR3
IFNγR

Volume-regulated
CI– channels
Induction of
Interferon receptors immunoproteosome
neuroprotective Regulation of
phonotype inflammatory
phenotype, and IL-1R1/R2
production of
IFNAR proinflammatory
factors IL-2R

7-TM IL-4R
receptors

Cytokine IL-18R IL-10R


receptors IL-15Rα IL-13R

Interleukin receptors

Figure 19.4 Cytokines and Chemokines Receptors in Microglia.

230 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
microglia. Notch-1 receptors are present in ameboid micro- express the macrophage colony-stimulating factor receptors
glial cells in the early postnatal brain and contribute to regula- (M-CSFRs), also known as colony stimulating factor-1 recep-
tion of production/release of NO and inflammatory cytokines tor (CSF-1R), which stimulate release of NO and proinflam-
(Grandbarbe et al. 2007). Receptors to complement fragments matory cytokines, activate phagocytosis, uptake of Aβ and
C3a and C5a (which are 7-TM GPCRs) are present in cul- promote neuroprotection (Mitrasinovic and Murphy 2003).
tured microglial cells and in microglia in situ and are coupled More importantly, absence of M-CSFRs results in loss of
to generation of Ca2+ signals, stimulation of K+ currents and microglia and these receptors therefore were considered to be
control of microglial motility; both receptors types are linked required for the development of microglia (Erblich et al. 2012).
with pertussis-toxin sensitive G proteins (Moller et al. 1997b). On the other hand, blockade of M-CSFRs (CSF-1R) signaling
The receptors for C5a for example induce membrane ruffling, completely inhibited microglial enhancement of glioblastoma
extension of lamellipodia, and the rearrangement of the actin invasion (Coniglio et al. 2012). Receptors to epidermal growth
cytoskeleton (Nolte et al. 1996). The C5a receptors are also factor (EGFRs) are members of ErbB family of receptor protein
involved in pathogenesis of neuropathic pain. kinases. Their functional expression was identified in cultured
The receptors to thrombin or proteinase-activated recep- mouse microglia, where they positively modulated K+ chan-
tors, PARs 1–4 are detected in microglia. Exposure to throm- nels through PTX-sensitive Gi proteins (Ilschner and Brandt
bin triggers cytoplasmic Ca2+ signaling and the release of 1996). Pharmacological inhibition of EGFR strongly inhib-
NO, various cytokines, and chemokines (e.g., TNF-α IL-6, ited microglia-stimulated invasion of glioblastoma (Coniglio
IL-12[p40] CXCL, monocyte chemoattractant protein 1 et al. 2012). Microglial cells also express CD200 receptors
[MCP-1, CCL2], macrophage inflammatory protein 1α and (CD200Rs), activated by surface glycoprotein CD200, the
1β [MIP-1α/-1β, CCL3/CCL4], etc.) (Moller et al. 2000a). lysophosphatidic acid receptors LPA1 and LPA3 (which trigger
All four PAR receptor subtypes were detected in rodent Ca2+ signaling) and formyl peptide receptors FPR1 (high affin-
microglia (Moller et al. 2000a). The activation of PAR-1 ity) and FPR2 (low affinity). These latter receptors also induce
receptors was suggested to contribute to microglial inflamma- microglial Ca2+ signaling. Finally, microglial cells express intra-
tory response in Parkinson disease. Activated microglial cells ER sigma receptors that regulate Ca2+ release.

Calcium
receptors,
CAR Leukotriene
Notch-1
receptor receptors,
CysLT1, CysLT2

Glucocorticoid Ca2+-activated
mineralcorticoid Complement
Release of K+ channels
receptors NO and 2+ receptors
Ca
cytolines C3a, C5a

Ca2+
pleiotropic motility
effects
Thrombin
Sigma-1 receptors,
receptors, ER InsP3/Ca2+
PAR1,3,4
σ1R
Delayed-rectifyer
K+ channels Ca2+
Release of
NO and Formyl peptide
Epidermal Ca2+ receptors,
cytolines
growth factor
FPR1, FPR2
receptors,
EGFR/Erb
Lysophophatidic
receptors,
Macrophage colony-
CD200 LPA1, LPA3
stimulating factor
receptors, M-CSFR receptors

7-TM Colony-stimulatung Gluco/mineralo


receptors factor receptors corticoid
intracellular
Tyrosine receptors
kinase DAP12 receptors DAP12 receptors
receptors
Intracellular
Sigma-1 receptors

Figure 19.5 Other Receptor Systems in Microglia.

P H YS I O L O GY O F M I C R O G L I A • 231
5 MICROGLIAL PLASMALEMMAL acquire glutamate transporters of EAAT-1/GLAST and
T R A N S P O RT E R S EAAT2/GLT types. Rapid de novo expression of both
GLAST and GLT-1 transporters was identified in rat micro-
The summary of microglial plasmalemmal transporters is glia following controlled cortical impact injury. The EAAT-1
shown in Figure 19.6. Microglial potassium gradients are transporters were also found in microglial cells of HIV-1 posi-
mainly regulated by H+/K+ ATPase; whereas the Na+/K+ tive patients. The glutamate transporters in activated microglia
ATPase, in contrast, is not functionally active. The KD for can serve neuroprotective function participating in removal of
microglial H+/K+ pump is approximately 3.7 mM, which is toxic glutamate loads (see Kettenmann et al. 2011 for details).
within the physiological range of extracellular K+ concentra- Microglial cells specifically express glucose transporter
tions (Shirihai et al. 1998). In addition microglial cells possess 5 (GLUT5) (Sasaki et al. 2004). Activated microglia also
the proton pump. Microglial cells express all three isoforms of express monocarboxylate transporters MCT1 and MCT2,
Na+/Ca2+ exchanger, NCX1, NCX2, and NCX3, with NCX1 which may be involved in the supply of lactate (Moreira et al.
being the dominant subtype (Nagano et al. 2004). Because of 2009). The expression of ATP-binding cassette ABC trans-
low levels of microglial resting membrane potential the NCX porters was reported in cultured rat microglia, where they can
is probably always operating in a reverse mode favoring Ca2+ be involved in the release of purines and cysteinyl leukotrienes
entry. The reverse mode of the NCX is involved in microglial (Ballerini et al. 2005).
migration (Ifuku et al. 2007). Chloride transporters expressed
in microglia include the K+/Cl– cotransporters of KCC1,
KCC2, KCC3, and KCC4 types. Microglia also have opera- 6 CALCIUM SIGNALING IN MICROGLIA
tional Na+/HCO3– cotransporter and/or Na+-dependent Cl–/
HCO3– exchanger (Faff et al. 1996). Anion exchange–medi- Microglial calcium signaling, similarly to other neuroglial cells
ated chloride/bicarbonate transporter is a major component (see chapter 26) is primarily regulated by endoplasmic reticu-
in the regulation of intracellular pH. lum Ca2+ store. Microglial cells contain both InsP3 receptors
Microglial cells possess several amino acid transporters. and ryanodine receptors (RyRs). The InsP3-dependent route
In particular, microglial cells express high levels of glutamate- usually dominates, although activation of RyRs by cyclic
cystine antiporter Xc, which is involved in glutathione meta- ADP ribose was found to trigger Ca2+ release in human cul-
bolism and may provide neurotoxic glutamate release, which tured microglia (Shideman et al. 2006). The status of ER Ca2+
for example causes oligodendroglial death in rat optic nerve store can be affected in pathological conditions, for example,
preparations (Domercq et al. 2007). Activated microglia the InsP3-mediated Ca2+ release (following the stimulation of

Na+/K+ Glutamate transporters


ATPase EAAT-1/EAAT-2
+ 2+
Na /Ca exchanger, activated microglia
NCX 2K+ 3N +
a

1G
lu –
2+ 1H +
1Ca
+
a +
3N 3Na
1K +
2+
a
1C
+
a
3N erse
Rev de
1K

mo
ect Chloride
+

Dir de
1Na

Bicarbonate mo
transporters
+

3

transporters
2/3HCO

2CI

1Na +

Glucose transporter
H+ pump,
GLUT5 glucose
Na+/H+ pump
H+/K+ pump

Cys
Glu
lactate

Glutamate-cystine ABC cassette transporters


transporter Xc

Monocarboxylate transporter
MCT-1, MCT-2
activated microglia

Figure 19.6 Microglial Plasmalemmal Transporters.

232 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
purinoceptors or PAF receptors) was reduced by more than to the movement of the processes, the entire cell can migrate
50% in microglial cells obtained from the brains of patients within brain tissue, including translocation of their soma.
with Alzheimer disease, possibly indicating chronic depletion It is not yet explored whether these forms of motility,
of the ER store (McLarnon et al. 2005). The store-operated migration, and process movement are distinctly regulated.
Ca2+ entry also plays important role in shaping microglial Ca2+ Moreover, a systematic study comparing the migratory
signals being mainly responsible for plateau phase of [Ca2+]i behavior of microglia during development and in response
transients (Moller et al. 1997a, 2000b). The maximal deple- to pathology has not been performed. There are many
tion of ER Ca2+ store in cultured microglia by overstimula- candidate molecules that serve as signals for pathological
tion of P2Y2/4 metabotropic purinoceptors triggers chronic events to microglia, including ATP (Davalos et al. 2005;
activation of store-operated Ca2+ entry (Toescu et al. 1998). Honda et al. 2001; Ohsawa and Kohsaka 2011), cannabi-
This chronic activation may account for an increased resting noids (Walter et al. 2003), morphine (Takayama and Ueda
[Ca2+]i levels observed in microglial cells in various forms of 2005), the chemokine CCL21 (Rappert et al. 2002), lyso-
neuropathology, such as for example in Alzheimer disease (see phosphatidic acid (Schilling et al. 2004), bradykinin (Ifuku
Kettenmann et al. 2011 for details). et al. 2007), galanin (Ifuku et al. 2011), and other neuro-
peptides (Noda et al. 2011b). Effects of various neuropep-
tides on microglial motility are shown in Figure 19.7A,
7 M I C R O G L I A L M I G R AT I O N with additional information that neuropeptide Y inhibits
A N D M OT I L I T Y IL-1β–induced microglial motility, although this was shown
only in cell line (Ferreira et al. 2012). In addition, ion chan-
Microglial cells exhibit two types of movement activity. In nels and transporters play an important role in cell migration
the ramified form, they actively move their processes without (Fig. 19.7B). This is true in particular for K+ channels, Cl–
translocation of the cell body. In the ameboid form, in addition channels, the Na+/H+ exchanger, Cl–/HCO3– exchanger,

A 200

** **
150 **
Total distance (μm)

** **

100

50

0
n l
yn I
en mb in
lth in
1
ur nin

ox sin
ato in
bs in

P
so H
sin

P
in
ste tro

n-

VI
br sin-

bo kin
do es

m oc
su stat

ex
Va TR
ce
en

es
ne ala
eli
gio on

or
tan
so yt

pr
ot
g
An C

ad

B K(Ca)(BK,IK,SK)

Ca2+
K+
Gi

Gq/11 Ca2+
Na+
PLC CI–

HCO3 Na+
InsP3 CI–
Na+
InsP3R
DAG
HCO3– H+
PKC actin

Retraction Protrusion

Figure 19.7 Microglial Migration and Motility A. Effects of various neuropeptides on microglial motility. B. Model summarizing the role of ion
channels and transporters in controlling microglial migration. (A) Modified from Noda M, Ifuku M, Okuno Y, Beppu K, Mori Y, Naoe S. 2011b.
Neuropeptides as attractants of immune cells in the brain and their distinct signaling. Adv Neuroimmune Biol 1:53–62. (B) Reproduced with permis-
sion from Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. 2011. Physiology of microglia. Physiol Rev 9:461–553.

P H YS I O L O GY O F M I C R O G L I A • 233
and Na+/HCO3– cotransporter, which all are linked to actin not Ca2+-activated TRPM4-like currents (ICAN) in cultured mouse
cytoskeleton (Schwab 2001a,b). In addition, the reverse mode microglial cells. J Physiol 586:427–439.
Biber K, Laurie DJ, Berthele A, Sommer B, Tolle TR, Gebicke-Harter PJ,
of Na+/Ca2+ exchanger might be involved in regulation of et al. 1999. Expression and signaling of group I metabotropic glutamate
migration through inducing Ca2+ influx (Ifuku et al. 2007). receptors in astrocytes and microglia. J Neurochem 72:1671–1680.
For precise information on the control of microglial pro- Biber K, Neumann H, Inoue K, Boddeke HW. 2007. Neuronal ‘On’ and
cess motility in vivo or migration in vitro induced by various ‘Off ’ signals control microglia. Trends Neurosci 30:596–602.
signals and their inhibitors, see Kettenmann et al. (2011). Biber K, Vinet J, Boddeke HW. 2008. Neuron-microglia signaling:
chemokines as versatile messengers. J Neuroimmunol 198:69–74.
Black JA, Liu S, Waxman SG. 2009. Sodium channel activity modulates
multiple functions in microglia. Glia 57:1072–1081.
8 S U M M A RY A N D P E R S P E C T I VE S Boddeke EW, Meigel I, Frentzel S, Biber K, Renn LQ, Gebicke-Harter P. 1999a.
Functional expression of the fractalkine (CX3C) receptor and its regula-
Recent studies indicate that in the normal brain microglia have tion by lipopolysaccharide in rat microglia. Eur J Pharmacol 374:309–313.
highly motile processes by which they seem to scan their ter- Boddeke EW, Meigel I, Frentzel S, Gourmala NG, Harrison JK,
Buttini M, et al. 1999b. Cultured rat microglia express functional
ritorial domains. Microglial cells can communicate with mac- beta-chemokine receptors. J Neuroimmunol 98:176–184.
roglial cells and neurones and with cells of the immune system Bordey A, Spencer DD. 2003. Chemokine modulation of
by a large number of signaling pathways. Likewise, microglial high-conductance Ca2+-sensitive K+ currents in microglia from
cells express receptors classically described for brain-specific human hippocampi. Eur J Neurosci 18:2893–2898.
communication such as receptors for neurotransmitter and Boucsein C, Kettenmann H, Nolte C. 2000. Electrophysiological prop-
neurohormones and neuromodulators. In addition, microglial erties of microglial cells in normal and pathologic rat brain slices.
Eur J Neurosci 12:2049–2058.
cells also express ion channels, transporters, and receptors ini- Boucsein C, Zacharias R, Farber K, Pavlovic S, Hanisch UK, Kettenmann
tially discovered as immune cell-specific such as receptors for H. 2003. Purinergic receptors on microglial cells: functional expres-
cytokines. In addition, as one of the most characteristic fea- sion in acute brain slices and modulation of microglial activation in
tures of microglia, microglial migration such as motility and vitro. Eur J Neurosci 17:2267–2276.
chemotaxis is controlled by various signals. Nonetheless, we Brockhaus J, Ilschner S, Banati RB, Kettenmann H. 1993. Membrane
properties of ameboid microglial cells in the corpus callosum slice
have no physiological recordings from microglial cells in vivo from early postnatal mice. J Neurosci 13:4412–4421.
so far. The majority of studies on microglial physiology were Carrier EJ, Kearn CS, Barkmeier AJ, Breese NM, Yang W, Nithipatikom
performed on cultured microglial cells or using brain slices. K, et al. 2004. Cultured rat microglial cells synthesize the endocan-
There are also many investigations performed on various types nabinoid 2-arachidonylglycerol, which increases proliferation via a
of microglial cell lines. None of the preceding may be consid- CB2 receptor-dependent mechanism. Mol Pharmacol 65:999–1007.
Chao CC, Hu S, Shark KB, Sheng WS, Gekker G, Peterson PK. 1997.
ered as a proper model of microglial cells because of substan- Activation of mu opioid receptors inhibits microglial cell chemotaxis.
tial and inconsistent modifications in physiological properties. J Pharmacol Exp Ther 281:998–1004.
Therefore, true physiology of microglia, for example, resting Chattopadhyay N, Ye C, Yamaguchi T, Nakai M, Kifor O, Vassilev PM,
membrane properties or developmental change, requires fur- et al. 1999. The extracellular calcium-sensing receptor is expressed in
ther investigations. rat microglia and modulates an outward K+ channel. J Neurochem
72:1915–1922.
Choi J, Ifuku M, Noda M, Guilarte TR. 2011. Translocator pro-
tein (18 kDa)/peripheral benzodiazepine receptor specific ligands
induce microglia functions consistent with an activated state. Glia
AC K N OW L E D G M E N T S 59:219–230.
Coniglio SJ, Eugenin E, Dobrenis K, Stanley ER, West BL, Symons MH,
MN was supported by Grants-in Aid for Scientific Research of et al. 2012. Microglial stimulation of glioblastoma invasion involves
Japan Society for the Promotion of Science; and AV’s research EGFR and CSF-1R signaling. Mol Med. Epub ahead of print, doi:
was supported by BBSRC UK, Royal Society, Alzheimer 10.2119/molmed.2011.00217.
Craner MJ, Damarjian TG, Liu S, Hains BC, Lo AC, Black JA, et al.
Research Trust (UK) and INTAS. 2005. Sodium channels contribute to microglia/macrophage activa-
tion and function in EAE and MS. Glia 49:220–229.
Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, et al. 2005.
ATP mediates rapid microglial response to local brain injury in vivo.
REFERENCES Nat Neurosci 8:752–758.
Delgado M, Ganea D. 2003. Vasoactive intestinal peptide prevents
Aihara M, Ishii S, Kume K, Shimizu T. 2000. Interaction between neu- activated microglia-induced neurodegeneration under inflamma-
rone and microglia mediated by platelet-activating factor. Genes tory conditions: potential therapeutic role in brain trauma. Faseb J
Cells 5:397–406. 17:1922–1924.
Avignone E, Ulmann L, Levavasseur F, Rassendren F, Audinat E. Del Rio-Hortega P. 1932. Microglia. In: Penfield W (ed.), Cytology
2008. Status epilepticus induces a particular microglial activation and cellular pathology of the nervous system. New York: Hoeber,
state characterized by enhanced purinergic signaling. J Neurosci pp. 482–534.
28:9133–9144. Dobrenis K, Makman MH, Stefano GB. 1995. Occurrence of the opiate
Ballerini P, Di Iorio P, Ciccarelli R, Caciagli F, Poli A, Beraudi A, et al. alkaloid-selective mu3 receptor in mammalian microglia, astrocytes
2005. P2Y1 and cysteinyl leukotriene receptors mediate purine and and Kupffer cells. Brain Res 686:239–248.
cysteinyl leukotriene co-release in primary cultures of rat microglia. Domercq M, Sanchez-Gomez MV, Sherwin C, Etxebarria E, Fern
Int J Immunopathol Pharmacol 18:255–268. R, Matute C. 2007. System xc- and glutamate transporter inhibi-
Beck A, Penner R, Fleig A. 2008. Lipopolysaccharide-induced tion mediates microglial toxicity to oligodendrocytes. J Immunol
down-regulation of Ca 2+ release-activated Ca 2+ currents (ICRAC) but 178:6549–6556.

234 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Ducharme G, Newell EW, Pinto C, Schlichter LC. 2007. Small- Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. 2011. Physiology
conductance Cl- channels contribute to volume regulation and phag- of microglia. Physiol Rev 9:461–553.
ocytosis in microglia. Eur J Neurosci 26:2119–2130. Kettenmann H, Hoppe D, Gottmann K, Banati R, Kreutzberg G. 1990.
Eder C, DeCoursey TE. 2001. Voltage-gated proton channels in micro- Cultured microglial cells have a distinct pattern of membrane channels
glia. Prog Neurobiol 64:277–305. different from peritoneal macrophages. J Neurosci Res 26:278–287.
Egan TM, Samways DS, Li Z. 2006. Biophysics of P2X receptors. Khanna R, Roy L, Zhu X, Schlichter LC. 2001. K+ channels and the micro-
Pflügers Arch 452:501–512. glial respiratory burst. Am J Physiol Cell Physiol 280:C796–806.
Erblich B, Zhu L, Etgen AM, Dobrenis K, Pollard JW. 2012. Absence Koizumi S, Shigemoto-Mogami Y, Nasu-Tada K, Shinozaki Y, Ohsawa
of colony stimulation factor-1 receptor results in loss of microglia, K, Tsuda M, et al. 2007. UDP acting at P2Y6 receptors is a mediator
disrupted brain development and olfactory deficits. PLoS One of microglial phagocytosis. Nature 446:1091–1095.
6:e26317. Korotzer AR, Cotman CW. 1992. Voltage-gated currents expressed by
Faff L, Ohlemeyer C, Kettenmann H. 1996. Intracellular pH regula- rat microglia in culture. Glia 6:81–88.
tion in cultured microglial cells from mouse brain. J Neurosci Res Krabbe G, Matyiash V, Pannasch U, Mamer L, Boddeke HW,
46:294–304. Kettenmann H. 2011. Activation of serotonin receptors promotes
Feindt J, Schmidt A, Mentlein R. 1998. Receptors and effects of the microglial injury-induced motility but attenuates phagocytic activ-
inhibitory neuropeptide somatostatin in microglial cells. Brain Res ity. Brain Behav Immun 26:419–428.
Mol Brain Res 60:228–233. Kraft R, Grimm C, Grosse K, Hoffmann A, Sauerbruch S, Kettenmann
Ferrari D, Villalba M, Chiozzi P, Falzoni S, Ricciardi-Castagnoli P, Di H, et al. 2004. Hydrogen peroxide and ADP-ribose induce
Virgilio F. 1996. Mouse microglial cells express a plasma membrane TRPM2-mediated calcium influx and cation currents in microglia.
pore gated by extracellular ATP. J Immunol 156:1531–1539. Am J Physiol Cell Physiol 286:C129–137.
Ferreira R, Santos T, Cortes L, Cochaud S, Agasse F, Silva AP, et al. 2012. Kremlev SG, Roberts RL, Palmer C. 2004. Differential expression of
Neuropeptide Y inhibits interleukin-1 b-induced microglia motility. chemokines and chemokine receptors during microglial activation
J Neurochem 120:93–105. and inhibition. J Neuroimmunol 149:1–9.
Ferreira R, Santos T, Viegas M, Cortes L, Bernardino L, Vieira OV, et al. Kuhn SA, van Landeghem FK, Zacharias R, Farber K, Rappert A,
2011. Neuropeptide Y inhibits interleukin-1b-induced phagocytosis Pavlovic S, et al. 2004. Microglia express GABA B receptors to modu-
by microglial cells. J Neuroinflammation 8:169. late interleukin release. Mol Cell Neurosci 25:312–322.
Flynn G, Maru S, Loughlin J, Romero IA, Male D. 2003. Regulation of Laing KJ, Secombes CJ. 2004. Chemokines. Dev Comp Immunol
chemokine receptor expression in human microglia and astrocytes. 28:443–460.
J Neuroimmunol 136:84–93. Lee YB, Nagai A, Kim SU. 2002. Cytokines, chemokines, and cytokine
Fordyce CB, Jagasia R, Zhu X, Schlichter LC. 2005. Microglia Kv1.3 receptors in human microglia. J Neurosci Res 69:94–103.
channels contribute to their ability to kill neurons. J Neurosci Lyons SA, Pastor A, Ohlemeyer C, Kann O, Wiegand F, Prass K, et al.
25:7139–7149. 2000. Distinct physiologic properties of microglia and blood-borne
Grandbarbe L, Michelucci A, Heurtaux T, Hemmer K, Morga E, cells in rat brain slices after permanent middle cerebral artery occlu-
Heuschling P. 2007. Notch signaling modulates the activation of sion. J Cereb Blood Flow Metab 20:1537–1549.
microglial cells. Glia 55:1519–1530. McLarnon JG, Choi HB, Lue LF, Walker DG, Kim SU. 2005.
Haas S, Brockhaus J, Verkhratsky A, Kettenmann H. 1996. ATP-induced Perturbations in calcium-mediated signal transduction in microglia
membrane currents in ameboid microglia acutely isolated from mouse from Alzheimer’s disease patients. J Neurosci Res 81:426–435.
brain slices. Neuroscience 75:257–261. McLarnon JG, Xu R, Lee YB, Kim SU. 1997. Ion channels of human
Hanisch UK, Kettenmann H. 2007. Microglia: active sensor and versa- microglia in culture. Neuroscience 78:1217–1228.
tile effector cells in the normal and pathologic brain. Nat Neurosci Mead EL, Mosley A, Eaton S, Dobson L, Heales SJ, Pocock JM. 2012.
10:1387–1394. Microglial neurotransmitter receptors trigger superoxide produc-
Hayashi Y, Kawaji K, Sun L, Zhang X, Koyano K, Yokoyama T, et al. tion in microglia; consequences for microglial-neuronal interactions.
2011. Microglial Ca 2+-activated K+ channels are possible molecular J Neurochem doi: 10.1111/j.1471–4159.2012.07659.x.
targets for the analgesic effects of S-ketamine on neuropathic pain. Milton RH, Abeti R, Averaimo S, DeBiasi S, Vitellaro L, Jiang L, et al.
J Neurosci 31:17370–17382. 2008. CLIC1 function is required for beta-amyloid-induced generation
Honda S, Sasaki Y, Ohsawa K, Imai Y, Nakamura Y, Inoue K, et al. 2001. of reactive oxygen species by microglia. J Neurosci 28:11488–11499.
Extracellular ATP or ADP induce chemotaxis of cultured microglia Mitrasinovic OM, Murphy GM Jr. 2003. Microglial overexpression of
through Gi/o-coupled P2Y receptors. J Neurosci 21:1975–1982. the M-CSF receptor augments phagocytosis of opsonized Abeta.
Ifuku M, Farber K, Okuno Y, Yamakawa Y, Miyamoto T, Nolte C, Neurobiol Aging 24:807–815.
et al. 2007. Bradykinin-induced microglial migration mediated by Miyoshi M, Miyano K, Moriyama N, Taniguchi M, Watanabe
B1-bradykinin receptors depends on Ca2+ influx via reverse-mode T. 2008. Angiotensin type 1 receptor antagonist inhibits
activity of the Na+/Ca 2+ exchanger. J Neurosci 27:13065–13073. lipopolysaccharide-induced stimulation of rat microglial cells by sup-
Ifuku M, Okuno Y, Yamakawa Y, Izumi K, Seifert S, Kettenmann H, et al. pressing nuclear factor kB and activator protein-1 activation. Eur J
2011. Functional importance of inositol-1,4,5-triphosphate-induced Neurosci 27:343–351.
intracellular Ca 2+ mobilization in galanin-induced microglial migra- Mizoguchi Y, Monji A, Kato T, Seki Y, Gotoh L, Horikawa H, et al. 2009.
tion. J Neurochem 117:61–70. Brain-derived neurotrophic factor induces sustained elevation of
Ilschner S, Brandt R. 1996. The transition of microglia to a ramified phe- intracellular Ca 2+ in rodent microglia. J Immunol 183:7778–7786.
notype is associated with the formation of stable acetylated and dety- Moller T, Hanisch UK, Ransom BR. 2000a. Thrombin-induced activa-
rosinated microtubules. Glia 18:129–140. tion of cultured rodent microglia. J Neurochem 75:1539–1547.
Ilschner S, Ohlemeyer C, Gimpl G, Kettenmann H. 1995. Modulation Moller T, Kann O, Prinz M, Kirchhoff F, Verkhratsky A, Kettenmann
of potassium currents in cultured murine microglial cells by receptor H. 1997a. Endothelin-induced calcium signaling in cultured mouse
activation and intracellular pathways. Neuroscience 66:983–1000. microglial cells is mediated through ET B receptors. Neuroreport
Inoue K, Tsuda M. 2009. Microglia and neuropathic pain. Glia 57: 8:2127–2131.
1469–1479. Moller T, Kann O, Verkhratsky A, Kettenmann H. 2000b. Activation
Jou I, Pyo H, Chung S, Jung SY, Gwag BJ, Joe EH. 1998. Expression of of mouse microglial cells affects P2 receptor signaling. Brain Res
Kv1.5 K+ channels in activated microglia in vivo. Glia 24:408–414. 853:49–59.
Kaushal V, Koeberle PD, Wang Y, Schlichter LC. 2007. The Ca 2+-activated Moller T, Nolte C, Burger R, Verkhratsky A, Kettenmann H. 1997b.
K+ channel KCNN4/KCa3.1 contributes to microglia activation and Mechanisms of C5a and C3a complement fragment-induced [Ca 2+]i
nitric oxide-dependent neurodegeneration. J Neurosci 27:234–244. signaling in mouse microglia. J Neurosci 17:615–624.

P H YS I O L O GY O F M I C R O G L I A • 235
Monif M, Reid CA, Powell KL, Smart ML, Williams DA. 2009. The Sanz JM, Chiozzi P, Ferrari D, Colaianna M, Idzko M, Falzoni S, et al.
P2X 7 receptor drives microglial activation and proliferation: a trophic 2009. Activation of microglia by amyloid b requires P2X 7 receptor
role for P2X 7R pore. J Neurosci 29:3781–3791. expression. J Immunol 182:4378–4785.
Moreira TJ, Pierre K, Maekawa F, Repond C, Cebere A, Liljequist S, Sasaki A, Yamaguchi H, Horikoshi Y, Tanaka G, Nakazato Y. 2004.
et al. 2009. Enhanced cerebral expression of MCT1 and MCT2 in a Expression of glucose transporter 5 by microglia in human gliomas.
rat ischemia model occurs in activated microglial cells. J Cereb Blood Neuropathol Appl Neurobiol 30:447–455.
Flow Metab 29:1273–1283. Schilling T, Eder C. 2007. Ion channel expression in resting and acti-
Nagano T, Kawasaki Y, Baba A, Takemura M, Matsuda T. 2004. vated microglia of hippocampal slices from juvenile mice. Brain Res
Up-regulation of Na+-Ca 2+ exchange activity by interferon-gamma in 1186:21–28.
cultured rat microglia. J Neurochem 90:784–791. Schilling T, Quandt FN, Cherny VV, Zhou W, Heinemann U, Decoursey
Nimmerjahn A, Kirchhoff F, Helmchen F. 2005. Resting microglial cells TE, et al. 2000. Upregulation of Kv1.3 K+ channels in microglia deac-
are highly dynamic surveillants of brain parenchyma in vivo. Science tivated by TGF-beta. Am J Physiol Cell Physiol 27:C1123–1134.
308:1314–1318. Schilling T, Stock C, Schwab A, Eder C. 2004. Functional importance
Noda M. 2011a. Kallikrein-kinin system in the brain. In: Michael Bader of Ca 2+-activated K+ channels for lysophosphatidic acid-induced
(ed.), Kinins. Berlin: De Gruyter, pp. 85–102. microglial migration. Eur J Neurosci 19:1469–1474.
Noda M, Ifuku M, Okuno Y, Beppu K, Mori Y, Naoe S. 2011b. Schlichter LC, Kaushal V, Moxon-Emre I, Sivagnanam V, Vincent C.
Neuropeptides as attractants of immune cells in the brain and their 2010. The Ca 2+ activated SK3 channel is expressed in microglia in the
distinct signaling. Adv Neuroimmune Biol 1:53–62. rat striatum and contributes to microglia-mediated neurotoxicity in
Noda M, Kariura Y, Amano T, Manago Y, Nishikawa K, Aoki S, et al. vitro. J Neuroinflam 7:4.
2003. Expression and function of bradykinin receptors in microglia. Schwab A. 2001a. Function and spatial distribution of ion channels
Life Sci 72:1573–1581. and transporters in cell migration. Am J Physiol Renal Physiol
Noda M, Kariura Y, Pannasch U, Nishikawa K, Wang L, Seike T, 280:F739–747.
et al. 2007a. Neuroprotective role of bradykinin because of the Schwab A. 2001b. Ion channels and transporters on the move. News
attenuation of pro-inflammatory cytokine release from activated Physiol Sci 16:29–33.
microglia. J Neurochem 101:397–410. Shideman CR, Hu S, Peterson PK, Thayer SA. 2006. CCL5 evokes
Noda M, Nakanishi H, Nabekura J, Akaike N. 2000. AMPA-kainate calcium signals in microglia through a kinase-, phosphoinositide-,
subtypes of glutamate receptor in rat cerebral microglia. J Neurosci and nucleotide-dependent mechanism. J Neurosci Res 83:
20:251–258. 1471–1484.
Noda M, Wang B, Ishikawa E, Ooboshi H, Kido MA, Wada K. 2007b. Shirihai O, Smith P, Hammar K, Dagan D. 1998. Microglia generate
Up-regulation of KCNQ channels in activated microglia. Soc external proton and potassium ion gradients utilizing a member of
Neurosc Abs 479.3. the H/K ATPase family. Glia 23:339–348.
Nolte C, Moller T, Walter T, Kettenmann H. 1996. Complement 5a Shytle RD, Mori T, Townsend K, Vendrame M, Sun N, Zeng J, et al.
controls motility of murine microglial cells in vitro via activation of 2004. Cholinergic modulation of microglial activation by alpha 7
an inhibitory G-protein and the rearrangement of the actin cytoskel- nicotinic receptors. J Neurochem 89:337–343.
eton. Neuroscience 73:1091–1107. Silei V, Fabrizi C, Venturini G, Salmona M, Bugiani O, Tagliavini F,
Norenberg W, Cordes A, Blohbaum G, Frohlich R, Illes P. 1997. et al. 1999. Activation of microglial cells by PrP and beta-amyloid
Coexistence of purino- and pyrimidinoceptors on activated rat fragments raises intracellular calcium through L-type voltage sensi-
microglial cells. Br J Pharmacol 121:1087–1098. tive calcium channels. Brain Res 818:168–170.
Norenberg W, Gebicke-Haerter PJ, Illes P. 1992. Inflammatory stimuli Sperlagh B, Vizi ES, Wirkner K, Illes P. 2006. P2X 7 receptors in the ner-
induce a new K+ outward current in cultured rat microglia. Neurosci vous system. Prog Neurobiol 78:327–346.
Lett 147:171–174. Stella N. 2008. Endocannabinoid signaling in microglial cells.
Norenberg W, Illes P, Gebicke-Haerter PJ. 1994a. Sodium channel in Neuropharmacology 56 Suppl 1:244–253
isolated human brain macrophages (microglia). Glia 10:165–172. Takayama N, Ueda H. 2005. Morphine-induced chemotaxis and
Norenberg W, Langosch JM, Gebicke-Haerter PJ, Illes P. 1994b. brain-derived neurotrophic factor expression in microglia. J Neurosci
Characterization and possible function of adenosine 5′-triphosphate 25:430–435.
receptors in activated rat microglia. Br J Pharmacol 111:942–950. Talbot S, Chahmi E, Dias JP, Couture R. 2010. Key role for spinal dorsal
Ohana L, Newell EW, Stanley EF, Schlichter LC. 2009. The Ca2+ horn microglial kinin B1 receptor in early diabetic pain neuropathy.
release-activated Ca 2+ current (ICRAC) mediates store-operated Ca2+ J Neuroinflammation 7:36.
entry in rat microglia. Channels (Austin) 3:129–139. Talbot S, Couture R. 2011. Emerging role of microglial kinin B1 receptor
Ohsawa K, Irino Y, Sanagi T, Nakamura Y, Suzuki E, Inoue K, et al. in diabetic pain neuropathy. Exp Neurol 234(2):373–381.
2010. P2Y12 receptor-mediated integrin-beta1 activation regulates Talbot S, Dias JP, Lahjouji K, Bogo MR, Campos MM, Gaudreau P, et
microglial process extension induced by ATP. Glia 58:790–801. al. 2012. Activation of TRPV1 by capsaicin induces functional Kinin
Ohsawa K, Kohsaka S. 2011. Dynamic motility of microglia: Purinergic B1 receptor in rat spinal cord microglia. J Neuroinflam 9:16.
modulation of microglial movement in the normal and pathological Tanaka J, Fujita H, Matsuda S, Toku K, Sakanaka M, Maeda N. 1997.
brain. Glia 59:1793–1799 Glucocorticoid- and mineralocorticoid receptors in microglial cells:
Pankratov Y, Lalo U, Krishtal O, Verkhratsky A. 2002. Ionotropic P2X the two receptors mediate differential effects of corticosteroids. Glia
purinoreceptors mediate synaptic transmission in rat pyramidal neu- 20:23–37.
rones of layer II/III of somato-sensory cortex. J Physiol 542:529–936. Tanaka KF, Kashima H, Suzuki H, Ono K, Sawada M. 2002. Existence
Pankratov Y, Lalo U, Krishtal OA, Verkhratsky A. 2009. P2X receptors of functional b1- and b2-adrenergic receptors on microglia. J Neurosci
and synaptic plasticity. Neuroscience 158:137–148. Res 70:232–237.
Pelegrin P, Surprenant A. 2009. The P2X7 receptor-pannexin connection Taylor DL, Diemel LT, Cuzner ML, Pocock JM. 2002. Activation of group
to dye uptake and IL-1b release. Purinergic Signal 5:129–137. II metabotropic glutamate receptors underlies microglial reactivity
Pennefather PS, Zhou W, DeCoursey TE. 1998. Idiosyncratic gating of and neurotoxicity following stimulation with chromogranin A, a pep-
HERG-like K+ channels in microglia. J Gen Physiol 111:795–805. tide up-regulated in Alzheimer’s disease. J Neurochem 82:1179–1191.
Rappert A, Biber K, Nolte C, Lipp M, Schubel A, Lu B, et al. 2002. Toescu EC, Moller T, Kettenmann H, Verkhratsky A. 1998. Long-term
Secondary lymphoid tissue chemokine (CCL21) activates CXCR3 to activation of capacitative Ca 2+ entry in mouse microglial cells.
trigger a Cl– current and chemotaxis in murine microglia. J Immunol Neuroscience 86:925–935.
168:3221–3226.

236 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka Yamada J, Sawada M, Nakanishi H. 2006. Cell cycle-dependent regu-
S, Salter MW, et al. 2003. P2X4 receptors induced in spinal lation of kainate-induced inward currents in microglia. Biochem
microglia gate tactile allodynia after nerve injury. Nature 424: Biophys Res Commun 349:913–919.
778–783. Yasuyoshi H, Kashii S, Zhang S, Nishida A, Yamauchi T, Honda Y, et
van Rossum D, Hanisch UK. 2004. Microglia. Metab Brain Dis al. 2000. Protective effect of bradykinin against glutamate neuro-
19:393–411. toxicity in cultured rat retinal neurons. Invest Ophthalmol Vis Sci
Walter L, Franklin A, Witting A, Wade C, Xie Y, Kunos G, et al. 2003. 41:2273–2278.
Nonpsychotropic cannabinoid receptors regulate microglial cell Zhou W, Cayabyab FS, Pennefather PS, Schlichter LC, DeCoursey TE. 1998.
migration. J Neurosci 23:1398–1405. HERG-like K+ channels in microglia. J Gen Physiol 111:781–794.
Westin K, Buchhave P, Nielsen H, Minthon L, Janciauskiene S, Hansson Zou J, Vetreno RP, Crews FT. 2012. ATP-P2X 7 receptor signaling
O. 2012. CCL2 is associated with a faster rate of cognitive decline controls basal and TNFa-stimulated glial cell proliferation. Glia
during early stages of Alzheimer’s disease. PLoS One 7: e30525. 60:661–673.

P H YS I O L O GY O F M I C R O G L I A • 237
20.
PHYSIOLOGY OF OLIGODENDROCY TES
Vittorio Gallo and Jean-Marie Mangin

A B B R E VI AT I O N S 1 INTRODUCTION

AMPA 2-amino-3-(5-methyl-3-oxo-1,2- oxazol-4-yl) Oligodendrocytes (OLs) are myelinating cells of the central ner-
propanoic acid vous system (CNS), closely associated with neurons and astro-
ASIC acid sensing ion channel cytes in both developing and adult brain. Oligodendrocytes
4-AP 4-Aminopyridine wrap axons in extremely large membrane expansions, and form
CC corpus callosum direct cellular contacts among themselves and with astrocytes
CFTR cystic fibrosis transmembrane conductance through gap junctions. These contacts with other neural cell
regulator types identify OLs as active participants in cellular networks
CIC voltage-gated chloride channels of the mature CNS and raise important questions about their
CNP cyclic nucleotide phosphodiesterase physiological properties.
CNS central nervous system Oligodendrocytes are found in both gray and white mat-
CNTF ciliary neurotrophic factor ter of the CNS, and undergo significant molecular, morpho-
D3R Dopamine D3 receptor logical, and physiological changes during early postnatal brain
DTI diffusion tensor imaging development. In white matter, OLs represent a major fraction
EAE experimental autoimmune (or allergic) of all cells. Because white matter function is involved in many
encephalomyelitis cognitive and behavioral processes, defining the physiologi-
EGFP enhanced green fluorescent protein cal properties of OLs is fundamental to understanding higher
FGF fibroblast growth factor brain functions, learning disabilities, and psychiatric disorders.
GABA gamma-aminobutyric acid Oligodendrocytes express a variety of ligand- and
GFP green fluorescent protein voltage-gated ion channels, as well as G-protein–coupled
GlyR glycine receptor Cl– channels neurotransmitter receptors, maintaining this expression
HVA high voltage–activated throughout the OLs’ lives (Tables 20.1 and 20.2). However,
LVA low voltage–activated their physiological role—clearly established in excitable
MBP myelin basic protein cells such as neurons—is not yet completely defined in OLs.
MS multiple sclerosis Heterogeneous receptor and channel expression has been
nAchR Nicotinic acetylcholine ionotropic receptors demonstrated in OLs, and physiological differences between
NMDAR N-methyl-d-aspartic acid (NMDA)- gray and white matter OLs suggest that, much like neurons
sensitive iGluRs and astrocytes, distinct OL subpopulations with different
OL oligodendrocyte physiological properties may exist. Moreover, many recep-
OPC oligodendrocyte progenitor cell tors and channels in OLs display remarkable plasticity during
PDGF platelet-derived growth factor development or on exposure to different environmental sig-
PI-3 phosphoinositide-3 nals, hinting at the complexity of OL physiology.
PLP proteolipid protein The anatomical relationships of OLs with neurons and
PNS peripheral nervous system astrocytes suggest that neurotransmitter receptors and ion
RMP resting membrane potential channels are actively involved in mediating cell–cell communi-
ROMK1 renal outer medullary potassium channel cation between OLs and other neural cells, in addition to the
RT-PCR Reverse transcriptase polymerase chain direct roles in developmental myelination and myelin mainte-
reaction nance. Recent studies also reveal a likely role for OLs in regu-
STX saxitoxin lating functional activity in axons (Yamazaki et al. 2007).
SUR sulfonylurea receptor The chapter attempts to integrate current information
SVZ subventricular zone about these receptors and channels into the general scheme
TEA tetraethylammonium of OL physiology, and associate this complex array of func-
TTX tetrodotoxin tional proteins with OL functions in normal and pathologi-
VDAC voltage-dependent anion-selective channel cal brains. Because of space constraints, this chapter focuses
VOCC voltage-operated calcium channel exclusively on the molecular and functional properties of

238
neurotransmitter receptors and voltage-gated ion channels in pharmacological agents that either selectively block or acti-
OLs, although membrane transporters or specific enzyme sys- vate these channels.
tems such as carbonic anhydrase are also legitimately part of
OL physiology. Readers are referred to chapters 21 and 52 for
2.1.1 Inwardly Rectifying Potassium Channels
further considerations of OL physiology and pathology.
Oligodendrocytes have a very negative RMP (approxi-
mately –80mV) and a high resting permeability for K+ ions.
2 VO LTAG E - G AT E D I O N C H A N N E L S Pharmacological and biophysical studies suggest that the
activity of inwardly rectifying, Ba2+ sensitive, weakly rectify-
The overall electrophysiological properties of OLs drastically ing Kir channels establishes the RMP. The classical view of Kir
change during their development from oligodendrocyte pro- channels in glia is that their main function is to buffer extra-
genitor cells (OPCs) to mature, myelinating OLs (Fig. 20.1). cellular K+ ions (see also chapter 16). However, their molecu-
These changes result from modifications of their total mem- lar diversity and their distinct expression pattern in different
brane size, as well as specific regulation of voltage-gated ion types of glia suggest additional functions for these channels in
channels, including a shift from Kv to Kir expression, and gliogenesis and glial pathology. For example, Kir channels are
downregulation of Na+ and Ca2+ channels during OL matura- implicated in regulating OL proliferation, differentiation, and
tion (see Fig. 20.1 and Table 20.1). survival (Gallo et al. 1996; Knutson et al. 1997; Neusch et al.
2001; Olsen and Sontheimer 2008; Sontheimer et al. 1989).
Weakly rectifying currents associated with Kir channels have
2.1 P OTA S S I U M C H A N N E L S
been recorded in OLs (Chittajallu et al. 2005; Sontheimer
Voltage-gated K+ channels play a crucial role in repolarizing et al. 1989; Steinhauser et al. 1992). Molecular and electrophys-
the membrane and maintaining the resting membrane poten- iological studies have demonstrated heterogeneity in expression
tial (RMP). These proteins represent the largest and most of these channels in OLs from distinct brain regions and differ-
diverse class of voltage-dependent ion channels (Berkefeld ent developmental stages; that is, OPCs, immature pre-OLs,
et al. 2010; Bichet et al. 2003), and are widely expressed in and mature OLs (Chittajallu et al. 2004; Soliven et al. 1989).
CNS and peripheral nervous system (PNS) glia, including In OPCs, upregulation of Kir channels is associated with
OLs. Electrophysiological studies combined with molecu- hyperpolarization of the RMP, and short-term changes in the
lar analysis have identified expression of channel subtypes RMP are counterbalanced by activation of Kir channels (Knutson
based on single channel conductance, voltage-dependence et al. 1997). Genetic deletion studies, combined with molecular
of activation/inactivation, whole-cell current properties, and and functional identification of the channels, demonstrated that

Table 20.1 VOLTAGE-GATED CHANNELS


VOLTAGE-GATED SUBTYPES/ MODE OF DEVELOPMENTAL
CHANNELS SUBUNITS ACTIVATION PHYSIOLOGICAL ROLE REGULATION

Kir potassium channels Heterogenous but Open at Maintenance of hyperpolarized Upregulated in


Kir4.1 is predominant hyperpolarized membrane potential myelinating OLs
potentials Extracellular K+ buffering
OPC: Initiation of cell cycle exit
KD potassium channels Kv1.2, Kv1.5, Kv1.4, Depolarization OPC: Upregulated by mitogens High expression at OPC
and Kv1.6 (PDGF, FGF). Promote OPC stage. Downregulated in
proliferation and differentiation mature OLs
OLs: Unknown
KA potassium channels Unknown Depolarization Unknown Downregulated during
OL differentiation
KCa potassium channels BK Depolarization Regulation of intracellular Ca2+ Decreased during OPC
IK? + intracellular Ca2+ OLs maturation? maturation
SK?
KATP potassium channels Kir6.1, Kir6.2, Intracellular ATP Unknown Unknown
and SUR2 OLs development and
regeneration?
Sodium channels Unknown Depolarization Unknown Decreased during
OL maturation. Not
expressed in mature OLs
Calcium channels L-, N-, and R-type LVA: Low Unknown, but regulate Ca2+ entry, Expressed throughout the
depolarization thus likely to influence many processes OL lineage
HVA: High from proliferation to myelination
depolarization

P H YS I O L O GY O F O L I G O D E N D R O C Y T E S • 239
A B
OPC pre-myelinating

persistant K current (pA)


300 600

peak K current (nA)


2

Na current (pA)
200

0.1 nA

0.1 nA
300
1
0.3 ms 0.3 ms 100

0 0 0

mature
pre-myelin.

OPC

mature
pre-myelin.
OPC

mature
pre-myelin

OPC

young

young
young
*
young mature
C
300 3
–80

Cm (pF)
200 2

Rin (GΩ)
Vrest (mV)
100 –40
1

0 0 0
0.5 nA

OPC
pre-myelin.

mature

OPC
pre-myelin.

mature

OPC
pre-myelin.

mature
young

young

young
20 ms

Figure 20.1 Changes in Electrophysiological Properties of Oligodendrocytes During Their Maturation. A. Whole-cell current pattern of four different
developmental stages of oligodendroglial cells. From a holding potential of –80 mV, cells were depolarized in steps of 10 mV up to +20 mV. Only in
OPCs a transient fast inward current—resembling an Na+ current—was detected soon after the beginning of the voltage step (top row insets). Mature
oligodendrocytes show a pure ohmic current response. B. Summary bar graphs of the peak amplitudes of the three current components analyzed at
each developmental stages. Voltage-activated sodium currents are lost as soon as OPCs differentiate, whereas voltage-activated potassium currents
remain present in young oligodendrocytes and are only lost once a cell has fully matured. C. Summary of the passive membrane properties quantified
in each of the four developmental stages. Error bars indicate SEM. From Kukley and Dietrich 2009.

resting conductance in OLs is mostly caused by Kir4.1 (Butt and or transient A-type K+ channels (KA). Calcium-activated K+
Kalsi 2006; Neusch et al. 2001). Oligodendrocytes are strongly channels (KCa) are also discussed briefly. KD, KA, and KCa chan-
immunopositive for Kir4.1 in situ, suggesting that they might nels are distinguished by their molecular identity and func-
be involved in buffering K+ ions released during propagation of tional and pharmacological properties. Besides their activation
action potentials (Kalsi et al. 2004). threshold and channel conductance, these channel types differ
Kir channels—in particular Kir4.1 in OLs—were shown to act significantly in terms of channel kinetics and selective ion per-
as critical regulators of cell division: Kir function correlates with meability. However, most of these channels are sensitive to both
cell cycle exit and initiation of OL differentiation. Conversely, tertraethylammonium (TEA) and 4-amino-pyridine (4-AP).
loss of functional Kir channels is associated with cells’ re-entry
into the cell cycle. Upregulation of Kir4.1 expression and a sub- 2.1.2.1 Delayed Rectifier Potassium Channels
sequent negative shift in RMP are believed to be critical for In neurons, these channels are responsible for repolariza-
differentiation and initiation of developmental myelination in tion after action potential propagation and regulating neu-
OLs (Butt and Kalsi 2006). Therefore, Kir expression and func- rotransmitter release at synapses. Studies in OL lineage cells
tion play important roles in coupling intrinsic molecular mech- identified expression of specific Kv subunits; however, some
anisms of cell cycle regulation with myelinogenesis. discrepancies were also generated in different cell prepara-
This hypothesis has been confirmed by the generation tions. Attali et al. (1997) identified Kv1.5 as a major compo-
of mice with a null mutation in Kir4.1 (Neusch et al. 2001). nent of K+ currents in A2B5+ and O4+ OPCs and in GalC+
Kir4.1–/– OLs in culture display a more immature morphol- OLs, although expression of subunits Kv1.2, Kv1.4, and Kv1.6
ogy and mutant mice display significant OL apoptotic death was also detected. Further studies (Schmidt et al. 1999) dem-
during critical phases of oligodendrogenesis and initiation onstrated that all Kv1.1 to Kv1.6 channel transcripts were
of myelination, likely because of chronic cell depolarization. expressed in OPCs, but only Kv1.4, Kv1.5, and Kv1.6 proteins
Axonal degeneration is also observed in these mice, causing were detected. Consistent with the previous study, functional
further OL damage and death at later developmental stages. evidence was found for either homomeric Kv1.5, or hetero-
Because of these cellular defects, Kir4.1–/– mice display a severe meric Kv1.4/Kv1.5 and Kv1.5/Kv1.6 channels. Analysis of K D in
hypomyelinating phenotype. OL lineage cells in situ confirmed the findings in culture, with
KD currents found in both NG2+ and O4+ cells in subcortical
white matter (Chittajallu et al. 2005). In addition to the Kv1
2.1.2 Outwardly Rectifying Potassium Channels
subunits, a recent study revealed Kv7/KCNQ channel expres-
This section focuses on the subfamilies of these channels sion in cells of the OL lineage (Wang et al. 2011).
expressed in OLs, particularly the Kv family, including the KD channels are highly expressed at immature (NG2+ and
+
delayed rectifying K+ channels (KD) and the fast-inactivating O4 ), proliferative stages of the OL lineage, but significantly

240 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
downregulated in GalC+ OLs (Chittajallu et al. 2005; Knutson and proteolipid protein (PLP) mRNA decay was blocked by
et al. 1997; Sontheimer et al. 1989). Actively dividing NG2+ inhibiting Kv1.3 expression, and this subunit was expressed in
cells in white matter displayed a higher density of KD channels immature OLs in MS tissue. These results indicate that cyto-
than did slowly dividing/postmitotic NG2+ cells in cerebral cor- toxicity, mediated by complement via antibody-independent
tex (Chittajallu et al. 2004). Thus, indications that expression of activation of the classical pathway, regulates Kv1.3 channel
these channels tightly correlates with cell cycle activity prompted expression with concomitant activation of the cell cycle and
further molecular and electrophysiological analysis of KD chan- OL de-differentiation.
nels during OL development. Chittajallu et al. (2002) showed
that upregulation of these currents occurs in the G1 phase of the 2.1.2.2 Rapidly Inactivating A-Type Potassium
OPC cycle and is caused by RNA synthesis-dependent increase Channels
in Kv1.3 and Kv1.5 expression. In situ analysis demonstrated In neurons, the main function of KA channels is to regulate
large KD currents in proliferating OPCs of the subventricular interspike intervals. KA currents display such a broad range of
zone (SVZ) and white matter; these currents were attenuated inactivation rates that, in some cases, a clear distinction between
by Kv1.3 channel blockers. These results indicate that Kv1.3 (at KA and KD currents is challenging. The Kv channel family also
variance with Attali et al. 1997) and perhaps Kv1.5 form KD includes subunits that give rise to currents with biophysical
channels in proliferating OPCs in vivo. properties representing a continuum between classical KD and
Consistent with these findings, the OL mitogens KA currents. These channels are less sensitive to TEA and more
platelet-derived growth factor (PDGF) and fibroblast growth sensitive to 4-AP than are other outwardly rectifying channels.
factor (FGF) were found to be important in inducing KD KA channels are expressed in cells of the OL lineage at dif-
expression in OPCs, particularly in upregulating Kv1.5 and ferent developmental stages, but—like KD channels—their
Kv1.6 mRNAs and increasing the density of KD currents functional expression is downregulated as OPCs differenti-
(Soliven et al. 2003). PDGFαR-mediated upregulation of K D ate to mature OLs (Knutson et al. 1997). KA currents were
current expression in NG2+ cells of subcortical white matter detected in OL lineage cells in situ, although significant het-
was prevented by tyrosine kinase inhibition (Chittajallu et al. erogeneity was observed in the levels of KA current expression
2005), demonstrating that endogenous mitogens enhance K D in OPCs. Cortical NG2+ cells displayed twice the density of
channel expression at early stages of the OL lineage through KA channels seen in their white matter counterparts, although
tyrosine kinase activity. biophysical properties were identical (Chittajallu et al. 2004).
These findings raise questions about the functional role of Because of similarities in the functional properties of KA and
KD channels in OL development. Several studies investigated KD channels and in their developmental regulation in the OL
this issue by (1) using channel blockers in various developmen- lineage, their roles in OL maturation are still poorly defined.
tal assays, and (2) analyzing the relationship between cellular
signals that modulate cell proliferation and differentiation, and 2.1.2.3 Calcium-Activated Potassium Channels
their influence on KD channel expression. These studies found All KCa channels require Ca2+ binding for activation, but three
that all KD channel blockers strongly inhibit OP cell cycle pro- subtypes—BK (large conductance; >100 pS and iberiotoxin/
gression and proliferation (Attali et al. 1997; Chittajallu et charybdotoxin/TEA sensitive), IK (intermediate conductance;
al. 2002; Gallo et al. 1996; Ghiani et al. 1999; Soliven et al. 30–70 pS and apamin insensitive), and SK (small conductance;
2003; Tiwari-Woodruff et al. 2006). Glutamate activation of 5–20 pS and apamin sensitive)—have been identified based on
2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl)propanoic acid their biophysical properties, and sensitivity to specific pharma-
(AMPA) receptors in OPCs induced Na+-dependent blockage cological reagents and selective toxic channel blockers.
of KD channels and inhibited OPC proliferation (Gallo et al. A crucial physiological role of Ca2+-activated K+ channels
1996). KD channels formed by Kv3.1 are expressed both in OPCs is to couple Ca2+ metabolism and membrane potential to K+
and OLs and are associated with the OL-specific tight junction flux and membrane excitability. A recent study first inves-
protein (OSP)/claudin-11 (Tiwari-Woodruff et al. 2006). These tigated the expression and function of BK channels in OLs
channels appear to be involved not only in OPC proliferation (Buttigieg et al. 2011). In OPCs, outward currents blocked by
and migration, but also in myelination, because Kv3.1-null mice BK channel blocker iberiotoxin were observed together with
exhibited decreased axonal diameter and myelin thickness. immunofluorescence labeling of BK channels. Fura-2AM
KD channels may also be important in OL response to microscopy showed that these channels are directly involved
pathological insults. A recent study investigated Kv subunit in regulating intracellular Ca2+ levels. Furthermore, BK chan-
expression in an animal model of multiple sclerosis (MS) nel currents and RNA and protein levels decreased with OPC
and experimental autoimmune encephalomyelitis (EAE) development, being lower in mature OLs. Thus, BK channels
(Herrero-Herranz et al. 2007). Subunit Kv1.4 was re-expressed appear to be involved in regulating Ca2+ influx in cells of the
at high levels in proliferating immature OLs of EAE mice OL lineage, and might be involved in OL maturation.
around demyelinated lesions. Kv1.4 expression was greatly
reduced in a ciliary neurotrophic factor (CNTF)-null mouse, 2.1.2.4 Adenosine Triphosphate (ATP)–Dependent
whose myelin lesion repair is impaired. In a separate study, Potassium Channels
Kv1.3 was identified as the important subunit in comple- ATP-sensitive K+ channels (KATP) are gated by ATP and
ment-induced cell cycle re-entry of immature OLs (Tegla et formed by Kir6.x-type subunits combined with sulfonylurea
al. 2011). Complement-induced myelin basic protein (MBP) receptor (SUR) subunits and with auxiliary components of

P H YS I O L O GY O F O L I G O D E N D R O C Y T E S • 241
the channel. Channels are further classified based on their sub- postnatal ages (Chittajallu et al. 2004; Paez et al. 2009a),
cellular localization into sarcolemmal (sarcKATP), mitochon- confirming previous findings that voltage-operated Na+
drial (mitoKATP), and nuclear (nucKATP). The Kir subunits have channels are not functionally expressed in premyelinating
two transmembrane domains and form the main channel pore, OLs. Conversely, Sox9-expressing OPCs located near the
whereas SUR subunits have five transmembrane domains with lateral ventricles displayed sizable Na+ currents (Paez et al.
two nucleotide-binding domains on the cytoplasmic region of 2009a).
the channel. These domains play a crucial physiological role, In summary, mature OLs do not express functional Na+
sensing changes in a cell’s metabolic activity. channels, but these channels are expressed during early devel-
Expression of different KATP channel subtypes has been opmental stages of the OL lineage. These findings raise inter-
detected in OLs, but their characterization in OLs is still esting questions about regulation and physiological function
incomplete—particularly the relationship between molecu- of these channels. One possibility is that voltage-operated Na+
lar subtypes and channels’ functional properties. An inward channels are expressed in OPCs because these cells are devel-
rectifying K+ channel with Walker type-A ATP-binding opmentally closer to multipotential neural progenitors capa-
domain (KAB-2), predominantly expressed in glial cells, was ble of generating neurons as well as glia.
isolated and functionally characterized (Takumi et al. 1995).
Expression of KAB-2 was detected in cerebellar and forebrain
2.3 C A LC IUM C H A N N E L S
OLs using in situ hybridization.
A recent study demonstrated expression of Kir6.1 and The entry route of Ca2+ across the OL membrane and the
Kir6.2 proteins, together with SUR2, in cultured OPCs and changes occurring in intracellular Ca2+ levels are factors that
OLs (Fogal et al. 2011). When a series of KATP channel acti- determine the physiological or pathological responses of OLs.
vators, including diazoxide, was tested in cultured OPCs, all Ca2+ influx in OLs can occur through routes involving distinct
these compounds enhanced cell proliferation. Furthermore, molecular mechanisms: (1) through ligand-gated channels
diazoxide enhanced myelination in vitro and attenuated the that display differential permeability to this cation (Kastritis
effects of perinatal chronic hypoxia on white matter, as dem- and McCarthy 1993; Kirchhoff and Kettenmann 1992;
onstrated by increased myelination. Thus, KATP channels might Patneau et al. 1994); (2) through voltage-operated Ca2+ chan-
regulate OL development and regeneration, and activators of nels (VOCCs) (Paez et al. 2009a; Patneau et al. 1994); and
the channels might have therapeutic value in periventricular (3) through other channels also identified in OLs (Alberdi et
white matter injury. al. 2005; Simpson et al. 1997). Transmembrane Ca2+ influx also
occurs in OLs through acid-sensing ion channels (Feldman
et al. 2008) (see section 2.4).
2.2 S O D IU M C H A N N E L S
Voltage-operated Ca2+ channels are divided into two
Na+ channels were first demonstrated in Schwann cells and classes: (1) high-voltage–activated (HVA) Ca2+ channels,
astrocytes (Bevan et al. 1984; Chiu et al. 1984), prompting displaying an activation threshold around –30 mV; and
studies to investigate the expression and regulation of Na+ (2) low-voltage–activated (LVA), displaying a lower threshold
channels in cells of the OL lineage. of approximately –60 mV and therefore requiring a smaller
Compared with K+ channels, voltage-gated Na+ channels depolarizing stimulus for activation. Six classes of HVA
in developing OLs are less well characterized and their func- VOCCs have been characterized based on molecular, electro-
tional role is intensely debated (Fields 2008). Oligodendrocyte physiological, and pharmacological properties: the P/Q, N, L,
progenitor cells are known to express voltage-gated Na+ cur- R, and T types.
rents displaying rapid activation/inactivation kinetics and Several studies demonstrated expression of L-, N-, and
tetrodotoxin (TTX) sensitivity (Sontheimer et al. 1989; R-type VOCCs in cells of the OL lineage in culture and in
Williamson et al. 1997). Because these findings were in situ (Berger et al. 1992; Butt 2006; Williamson et al. 1997).
recordings from slices (Berger et al. 1992; Chittajallu et al. Unlike voltage-gated Na+ channels, VOCCs are expressed
2004; Paez et al. 2009a), it was concluded that voltage-oper- throughout the entire OL lineage. Although OPCs isolated
ated Na+ channels are functionally expressed in OPCs in vivo from distinct tissues produced different results depending
(Fig. 20.2). on developmental stage and type of preparation, the stud-
Several studies have shown that expression of voltage-gated ies concluded that Ca2+ currents are present in OPCs. These
Na+ channels is downregulated between the NG2+ OPC and currents have also been examined in mature OLs by electro-
the O4+ pre-OL stage, and further between the O4+ and physiological recordings in early postnatal corpus callosum,
the O1+ OL stages (Berger et al. 1992; Paez et al. 2009a). revealing expression of both HVA and LVA Ca2+ channels in
Analysis of voltage-operated Na+ channels in cortical white OLs of white matter regions (Fig. 20.3) (Fulton et al. 2010;
matter in situ using whole cell patch-clamp in OLs demon- Paez et al. 2010). More recent studies characterized these
strated functional expression only at early postnatal develop- channels in corpus callosum and demonstrated expression of
mental stages. verapamil-sensitive L-type Ca2+ channels in immature OLs
Availability of transgenic mice selectively expressing (see Fig. 20.3) (Paez et al. 2010). Functional expression of
green fluorescent protein (GFP) in OLs (PLP-GFP and VOCCs appears to be progressively attenuated as OPCs
CNP-EGFP mice) in situ demonstrated that O4+ pre-OLs mature into myelinating OLs (Fulton et al. 2010; Paez et al.
in corpus callosum lack inward Na+ currents at early 2010).

242 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
A C1 cNG2+ D cNG2+
wmNG2+

40 mV
16/16 19/54 5/19

200 ms

B cNG2+ C2 cNG2+ E cNG2+

35/54 4/4 2/56

+1μM TTX

I J K *
–25
0
wmNG2+
–200 –20 ns
INa density(pA/pF)
INa amplitude(pA)

cNG2+ no spike –400 –15

–600 –10
cNG2+ spike wmNG2+
–800 –5
500 pA

wmNG2+ no spike
cNG2+ no spike
–1000 0
5 ms –60 –40 –20 0 20 wmNG2+ cNG2+ cNG2+
Vtest(mV) no spike spike

Figure 20.2 A Subpopulation of Cortical NG2+, but Not White Matter NG2+ Cells, Display TTX-Sensitive Spikes in Response to Depolarizing
Current Pulses. A. Representative current clamp traces from a single white matter (wm) NG2+ cell in response to membrane depolarization. B,C.
From a total of 56 cortical (c) NG2+ cells tested, 35 showed no membrane response (B), and 19 displayed a single spike occurring at the beginning
of the test pulse (C1). In all four of these 19 cells tested, the spike was abolished by 1 μM TTX (C2). D. Five out of the 19 spike-producing cortical
NG2+ cells also clearly displayed an after depolarization (arrow). E. Single example of a cortical NG2+ cell that displayed multiple spikes (two cells
were found with this response). G,H. Morphology and confirmation of NG2+ expression in single and multiple spiking cells, respectively. I. Single
trace examples of INa in a white matter NG2+ cell (top trace), a cortical NG2+ cell not displaying a spike (middle trace), and a cortical NG2+ cell with
spike (bottom trace). J. Voltage/current relationships of INa for white matter NG2+ cells (open circle; n = 5), and for cortical NG2+ cells without and
with spike (filled circles and filled triangles; n = 10 and 9, respectively). K. Corresponding pooled INa densities (n = 5–10). All data were obtained from
P5-P8 CNP-EGFP mice. Scale bars = 20 μM. *P < .05, ns—not significant (i.e., P > .05), Mann-Whitney U-Test. Chittajallu et al. 2004.

P H YS I O L O GY O F O L I G O D E N D R O C Y T E S • 243
A –60 –40 –20 0 20 40
–0.0 mV
Vh-120 Vh-50

LVA pA/pF
–0.5

–1.0
LVA HVA
–1.5 OL
OPC * **
50 pA **
–2.0
20 ms
0.4 0.8
0.2
–60 –40 –20 0 20 40 –60 –40 –20 0 20 40

HVA pA/pF
–0.0 mV mV
pA/pF

–0.2
–0.2
–0.4
–1.2
–0.6 HVA OL
LVA OPC
–2.2 *** ***
***
100
B PDGF+bFGF 100

Intracellular C++ (% change)


80

Intracellular Ca++ (% change)


K+(20mM) 80
–0.18
60 60
–0.17
Fura-2 ratio

NG2+
O1+ mN2
–0.16 40 40

–0.15 20 20

0 2 4 6 8 10 0 0

α
2

BP
Time (min)

4
1
IV

IV

IV

IV

G
O
O
Fr

M
N
1D

2D

3D

4D
C

G
PD
0 min D
80
K+(20mM)
LV
SVZ Intracellular Ca++ (% change)
60 SVZ OPCs

GFP Fura-2 ratio Fura-2 ratio 7 min


40
0 min 7 min

20 ** ***
***
CC 0
p

+
l
sa

ra

ife

a+
Ba

Ve

–C

GFP Fura-2 ratio Fura-2 ratio

E F SVZ OPCs
120
Amplitude of Ca++ uptake (%)

CC Ols
0.18 K+ (20mM) 0.18 K+ (20mM)
P4 P8
0.17 SVZ OPCs 0.17 CC OLs 80
Fura-2 ratio

Fura-2 ratio

0.16 0.16 **
40 * **
0.15 0.15 *

0.14 0.14 0
0 5 10 15 0 5 10 15
au

au

au

au
Pl eak

Pl eak

Pl eak

Pl eak
ate

ate

ate

ate

Time (min) Time (min)


P

Figure 20.3 Oligodendrocyte Lineage Cells Express Different Types of Voltage-Activated Ca2+ Channels. A. Low-voltage–activated and high-voltage–
activated currents in immature oligodendrocytes. Left. Recordings of Ca2+ currents in immature OLs. Currents were activated from a holding
potential of –120 mV and –50 mV. Note inactivation of the transient component when currents are activated from –50 mV. Right. Current density/
voltage graphs for LVA and HVA currents. Both types of current are reduced in immature OLs (mean ± SEM). *P < .05, **P < .01, ***P < .001. B.
Left. NG2 and O1 staining were used after Ca2+ imaging. NG2+ OPCs responded to high K+ with large increases in intracellular Ca2+ than O1+ cells.
Center. Oligodendrocyte progenitor cells were cultured for 1 to 2 days in vitro (1–2 DIV) with PDGF and bFGF, or in mitogen-free medium (mN2)
for 2 more days (3–4DIV). Ca2+ influx amplitudes after high K+ treatment are shown. Right. Staining for PDGFrα, NG2, O4, O1, and MBP was
used after Ca2+ imaging. Average Ca2+ influx amplitude (50 cells) for each OL marker is shown. C. Time lapse series of GFP-expressing OPCs in the
dorsolateral subventricular zone (SVZ) and corpus callosum (CC) (P4). An increased Fura-2 fluorescence ratio is indicated by warmer colors. Time is
denoted in minutes. LV: lateral ventricle. Scale bar (Top) 100 μm, (Bottom) 50 μm. D–E. Voltage-operated Ca2+ channel activity in GFP-expressing
OPCs from the SVZ and in OLs from the CC. Each trace corresponds to a single cell. D. K+-induced Ca2+ uptake in SVZ OPCs was abolished in 25
μM verapamil, 25 μM nifedipine and in the absence of external Ca2+ (–Ca2+). F. Voltage-operated Ca2+ channel activity was examined in SVZ OPCs
and in CC OLs. Graphs show the average maximal peak values and plateau values, expressed as percentage of change of the emission intensities (mean
± SEM). *P < .05, **P < .01, ***P < .001 versus basal (D) and versus SVZ OPCs (F). From Fulton et al. 2010; Paez et al. 2010.
The functional role of VOCCs in the OL lineage remains 2.4 C H L O R I D E A N D OT H E R C H A N N E L S
largely unknown. In OPCs these channels are thought to
Voltage-activated Cl– channels play an important physiologi-
regulate early developmental processes, including gene expres-
cal role, because Cl– is the most abundant anion present in
sion, cell proliferation, and cell migration (Butt 2006; Paez et all physiological cellular environments. So far, four distinct
al. 2009a,b). In mature, myelinating OLs, VOCCs might be classes of Cl– channels have been cloned, including: (1) Ca2+
involved in initiating and maintaining myelination. Moreover,
activated Cl– channels; (2) the cystic fibrosis transmembrane
VOCCs are thought to regulate process extension and retrac-
conductance regulator (CFTR); (3) voltage-dependent
tion of membrane sheets in mature OLs (Kirischuk et al.
anion-selective channels (VDACs); and (4) voltage-gated
1995; Paez et al. 2009b). Consistent with this notion, and
chloride channels (CICs) ( Jentsch et al. 1999).
with the hypothesis that VOCCs might be involved in modu-
Oligodendrocytes display significant resting Cl– con-
lating initiation of myelination, Kirischuk et al. (1995) found
ductances, and Cl– currents were first recorded in excised
heterogeneous distribution of these channels in subcellular
inside-out patches from OLs (Barres et al. 1988). Further stud-
OL domains. High-voltage–activated channels were primarily
ies in culture demonstrated the presence of voltage-activated
located on cell bodies, whereas LVA channels were mostly on
Cl– channels in cells of the OL lineage by whole-cell record-
cell processes, suggesting differential activation as a signal for
ings (Williamson et al. 1997). These outwardly rectifying
initiation of myelination.
currents were identified as arising from voltage-gated rather
A study by Paez et al. (2007) revealed a functional inter-
than Ca2+-activated Cl– channels. The function and molecu-
action between golli proteins and Ca2+ channels in OLs.
lar identity of Cl– channels in OLs is still unknown, but they
Golli proteins are encoded by the myelin basic protein
might be involved in cell shape or cell volume changes that
(MBP) gene, which also encodes MBP constituents of myelin
accompany Cl– efflux through these channels.
(Campagnoni et al. 1993). Overexpression of golli proteins
Acid-sensing ion channels (ASICs) are formed by different
enhanced transmembrane Ca2+ influx and intracellular Ca2+
combinations of six molecularly distinct subunit proteins that
levels in OLs, and triggered remodeling of OL processes and
associate to form either homomeric or heteromeric channels
membrane sheets. Binding of golli to the plasma membrane is
with different physiological properties. Acid-sensing ion chan-
important for modulating Ca2+ homeostasis—supporting the
nel 1a channels are Ca2+ permeable and likely to be important
hypothesis that these myelin proteins form a protein complex
in CNS pathology, as their inactivation significantly attenu-
in membrane subdomains and modulate Ca2+ entry along OL
ates ischemic brain damage (Xiong et al. 2006).
processes. The first study identifying acid-sensing Na+ channels in
Voltage-operated Ca2+ channels are also engaged in axo-
cells of the OL lineage demonstrated large Na+ currents in
glial signaling between neurons and OLs. Propagation of
response to acidification in OPCs and in more mature cells of
action potentials and axonal activity cause enough extra-
the lineage (Sontheimer et al. 1989). Their functional proper-
cellular K + accumulation to depolarize surrounding glial
ties were similar to their neuronal counterpart. A decrease in
cells and activate their VOCCs. This could be a physi-
functional expression of these proton-activated channels was
ological mechanism that couples electrical activity in
observed from OPCs to mature OLs.
neurons with axonal myelination. In fact, enhanced extra-
Feldman et al. (2008) demonstrated ASIC expression in
cellular K + levels within the normally observed physiolog-
white matter in situ, and ASIC1a was specifically found in
ical range have been shown to induce OL depolarization
white matter OL lineage cells. ASIC1a, 2a, and 4 mRNAs
and a rise in intracellular Ca 2+ (Kirischuk et al. 1995).
were expressed in OL lineage cells in culture, but mRNA levels
Furthermore, the microstructure of white matter changes
for these channels decreased during OPC maturation to OLs
continuously during OL maturation and myelination, as
(Feldman et al. 2008). Patch-clamp recordings demonstrated
demonstrated by direct diffusion tensor imaging (DTI)
predominance of homomeric ASIC1a in OLs, whose activa-
measurements showing progressive compaction of the
tion caused robust membrane depolarization and a transient
extracellular space (Schmithorst et al. 2002). Reduced
increase in intracellular Ca2+.
volume would cause larger changes in extracellular K+
Based on their functional properties and activation owing
concentration within the local axoglial microenviron-
to acidification of the extracellular environment, ASICs in OLs
ment when K+ is released from axons, so even small axon
might play a role in pathology, particularly ischemic damage.
fibers could produce fluctuations in extracellular K + able
to depolarize surrounding OLs.
Immunohistochemical analysis of VOCC expression
in white matter in situ supports this mechanism, as robust 3 L I G A N D - G AT E D I O N C H A N N E L S
expression of R-type Ca2+ channels was transiently detected
in OL cell bodies and processes, as well as in paranodal myelin Ligand-gated ion channels are a highly important family of
wraps, during the peak of myelination (Chen et al. 2000). proteins in the brain, underlying almost all neurotransmission
R-type channel immunoreactivity was closely associated with the exception of electrical synapses. Expression of a vari-
with OL membranes in direct contact with axons, allowing ety of neurotransmitter-receptor channels and metabotropic
rapid activation of these VOCCs and initiation of myeli- receptors has been recognized in glial cells (OLs among them)
nation when extracellular K+ accumulated in the axoglial for at least 20 years, yet their function in these cells remains in
microenvironment. many cases elusive.

P H YS I O L O GY O F O L I G O D E N D R O C Y T E S • 245
Table 20.2 NEUROTRANSMITTER RECEPTORS
IONOTROPIC MODE OF PHYSIOLOGICAL PHYSIOLOGICAL DEVELOPMENTAL
RECEPTORS SUBUNITS TRANSDUCTION MODE OF ACTIVATION ROLE REGULATION

AMPAR Glur2, Glur3, and Depolarization, OPC: Synaptic OPC Inhibit prolif- Peak of expression at
GluR4 Calcium entry OL: Unknown (spillover, eration, favor differen- the OPC stage.
astrocytes) tiation, and increase Downregulated in
migration. mature OLs
OL: Unknown, but
excitotoxic at high
concentration

KAR GluR6, GluR7, KA1, and Depolarization OPC: Unknown but Unknown Unknown. Low expres-
KA2 nonsynaptic sion in OPC
OL: Unknown
(spillover, astrocytes)

NMDAR NR1, NR2A, NR2B, Depolarization, OPC: Unknown but No physiological role? Unknown. Low
NR2C, NR2D, and calcium entry nonsynaptic Absence of phenotype expression in OPC
NR3A OL: Unknown in
(spillover, astrocytes, NR1
glycine) KO but could mediate
excitotoxicity

GABAAR Unknown OPC: OPC: Synaptic and/or OPC: Increase Peak of expression at
Depolarization. extrasynaptic. proliferation. the OPC stage
OL: Unknown OL: Unknown (spillover, OL: Unknown Strongly downregulated
(hyperpolarizing ?) astrocytes) in mature OLs

Purinergic P2XR P2X7 Depolarization, Mostly unknown, but Unknown, but excito- Robust expression in
calcium entry via activated by ATP release toxic OPCs and mature OLs
pore formation from astrocytes at high concentration

GlyRs α2 and β OPC: Unknown (synapses?) Unknown Peak of expression at


Depolarization. the OPC stage.
OL: Unknown Downregulated in
mature OLs
Nicotinic AchRs α3, α4, α5, α7, β2, OPC: Unknown. Synapses Unknown Unknown
and β4 Depolarization. could not be detected in
OL: Unknown hippocampal OPCs
METABOTROPIC SUBTYPES/ MODE OF PHYSIOLOGICAL PHYSIOLOGICAL DEVELOPMENTAL
RECEPTORS SUBUNITS TRANSDUCTION MODE OF ACTIVATION ROLE REGULATION

mGluRs Group I,II and III IP3-Induced Unknown (synapses?) OPC: Regulate Ca2+- Downregulated during
calcium release permeable AMPARs OL maturation
expression. Regulate
the release of BDNF.
GABABRs B1 and B2 Adenylate cyclase Unknown (synapses?) OPC: Promote prolif- Downregulated during
Inhibition eration and migration OL maturation
Purinergic P2Y P2Y1 IP3-induced Mostly unknown, but OPC: Decrease Robust expression in
calcium release activated by ATP release proliferation, increase OPCs and mature OLs
from astrocytes migration and
differentiation
OL: Unknown
Muscarinic AchRs M3>M4>M2>M1>M5 IP3-Induced Unknown OPC: Increase prolif- Downregulated during
calcium release eration and survival, OL maturation
inhibit differentiation
Dopamine D2 and D3 Not yet defined Unknown OPC: Regulate OPC D3 expressed in OPC
in OL proliferation but absent from
OL: Unknown, but mature OL. D2/D3
protective against ratio increases during
excitotoxicity maturation

246 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Oligodendrocyte lineage cells express functional ionotro- Rat OLs in culture and in vivo are known to express KAR
pic receptors for most, if not all, known neurotransmitters subunits GluK2, GluK3, GluK4, and GluK5 (De Biase et al.
(Table 20.2). Except for 5-HT and ATP, the expression level 2010; Sánchez-Gómez and Matute 1999), but OPCs appar-
of neurotransmitter receptors appear to decrease once OPCs ently express only a small number of functional KARs. Indeed,
begin differentiating into myelinating OLs—suggesting that kainate applied to OPCs in brain slices induces only small per-
neurotransmitters may play specific roles during early stages of sistent inward currents (Kukley and Dietrich 2009).
OL lineage, particularly the OPC stage, characterized by pro- Cellular consequences of AMPAR/KAR activation in
teoglycan (NG2) expression (see chapter 21). However, the OLs are not yet clearly defined. A current hypothesis regard-
physiological function of neurotransmitter receptor remains ing transduction of GluR activation in OLs involves entry
unresolved in both OPCs and oligodendrocytes. of Ca2+, the most common intracellular second messenger;
AMPA receptors expressed by OPCs are often Ca2+-permeable
(Bergles et al. 2000). Ca2+ entry might also involve the abil-
3.1 I O N OT RO P I C G LU TA M AT E R E C E P TO R S
ity of glutamate to depolarize OL membranes enough to
Ionotropic glutamate receptors (iGluR) have a central role activate voltage-operated Ca2+ channels (Berger et al. 1992).
in brain physiology, being fundamental to most excitatory Depolarization induced by glutamate has also been shown
neurotransmission between neurons. The three main types to activate Na+ channels, inducing a large enough increase
of iGluRs are: AMPA- and kainate-sensitive iGluRs (respec- in intracellular Na+ to allow Ca2+ entry via the Na+/Ca2+
tively, AMPARs and KARs), and N-methyl-d-aspartic acid exchanger (NCX1) (Chen et al. 2007); and GluR activation
(NMDA)-sensitive iGluRs (NMDARs). In neurons, the could influence OL physiology in a Ca2+-independent man-
types of iGluRs tend to have distinct functions: AMPARs ner. Indeed, Na+ entry via GluR can block voltage-gated K+
and KARs enable fast synaptic excitatory neurotransmission, channels independently of Ca2+ and G-protein (Borges and
whereas NMDARs are crucial in synaptic plasticity—mod- Kettenmann 1995). This blockade, for example, has been
ulating the strength of excitatory synapses, notably via their shown to prevent OPC proliferation and their differentiation
Ca2+ permeability. Although NMDARs and AMPARs have into myelinating OLs (Gello et al. 1996).
complementary functions in neurons with respect to synap- As in neurons, it is now clear that OPCs receive functional
tic transmission, their functions and interactions in OLs are glutamatergic synapses from neurons, both in white and gray
largely undetermined. matter (see chapter 21); however, these synapses are transient
(De Biase et al. 2010; Etxeberria et al. 2010) and disappear as
OLs mature into myelinating OLs: KARs do not appear to
3.1.1 AMPA- and Kainate-Sensitive be recruited during neuron-to-OPC synaptic transmission
Glutamate Receptors (Kukley and Dietrich 2009). Therefore, if AMPARs/KARs
AMPARs and KARs are tetrameric, cationic, receptor expressed by mature OLs are activated at all, it is likely to occur
channels that can be composed from four distinct subunits in a nonsynaptic manner such as volume transmission, spill-
for AMPAR (GluA1–GluA4) and five subunits for KAR over, or release from astrocytes (Volterra and Meldolesi 2005).
(GluK1–GluK5). AMPARs can assemble either as homo- Because in vitro studies showed that AMPAR activation
tetramers or heterotetramers, with functional properties can regulate OPC differentiation into myelinating OLs, it
dictated by their subunit composition and the presence was proposed that axons can regulate their own myelination
of auxiliary transmembrane AMPAR regulatory proteins by releasing glutamate onto surrounding OLs (Yuan et al.
(TARPs). Most AMPARs are composed of a dimer of GluA2 1998). Axonal glutamate release could allow surround-
subunits and a dimer of GluA1, GluA3, or GluA4. KARs can ing OPCs and myelinating OLs to be constantly informed
either form homomers from subunits GluK1, GluK2, GluK3, about their relative conduction speeds and degree of
or heteromers where GluK1–3 subunits combine with GluK4 synchrony with surrounding axons, allowing myelination
or GluK5. to occur in an orderly manner (Mangin and Gallo, 2011).
Reverse transcriptase polymerase chain reaction (RT-PCR) In mature OLs, the importance of AMPAR is less certain
and Western blot experiments have detected AMPAR subunits since neuron-to-OPC synapses and AMPAR expression are
GluA2, GluA3, and GluA4 in OLs (De Biase et al. 2010; Itoh et both downregulated when OLs begin myelination. Because
al. 2002). AMPA receptors in most mature neurons include a overactivation of AMPARs can mediate OLs’ cell death in
RNA-edited GluA2 subunit, which conveys poor Ca2+ perme- pathological conditions such as ischemia (Matute 2011),
ability. By contrast, AMPARs in OPCs were shown to exhibit AMPARs might also regulate physiological cell death in
a significant fraction of Ca2+-permeable AMPARs (Bergles mature OLs occurring during normal myelin turnover in
et al. 2000). It is unclear whether this property stems from adulthood.
the presence of a nonedited GluA2 subunit or the complete
absence of GluA2 subunits in the Ca2+-permeable AMPARs
3.1.2 NMDA-Sensitive Glutamate Receptors
(Li and Stys 2000). However, biochemical evidence suggests
that Ca2+-permeable AMPARs in OLs lack the GluA2 subu- NMDARs are heterotetramers formed through combinations
nit, with immmunoprecipitation experiments demonstrating of NR1 with NR2 or NR3 subunits. The NR1 subunit con-
the presence of protein complexes in OLs formed exclusively tains the binding site for glycine/d-serine and is necessary for
of GluA3 and GluA4 (Itoh et al. 2002). forming a functional NMDAR.

P H YS I O L O GY O F O L I G O D E N D R O C Y T E S • 247
Both OPCs and mature OLs express functional NMDARs examining GABA responses in rat OPCs in culture failed to
(De Biase et al. 2011; Káradóttir et al. 2005) that can induce detect modulation by the benzodiazepine flunitrazepam or
intracellular Ca2+ transients when activated (Micu et al. the inverse agonist DMCM (Williamson et al. 1998). Recent
2006). Patch-clamp recordings on rat cerebellar OLs and experiments in brain slices showed that GABAARs expressed
co-immunoprecipitation experiments with rat purified myelin by OPCs can be activated via neuron-to-OPC synapses and
have shown that functional NMDARs in OLs are most likely the resulting synaptic currents can be modulated by applying
composed of NR1, NR2C, NR2D, and NR3A subunits benzodiazepines (Lin and Bergles 2004). Because γ2 is nec-
(Burzomato et al. 2010; Micu et al. 2006). essary for synaptic targeting of GABAARs in neurons, these
Unlike AMPARs in OPCs, NMDARs are apparently not observations support functional expression of γ2-containing
significantly activated by the synaptic release of glutamate GABAARs in mouse OPCs. Interestingly, in mouse soma-
from neurons (De Biase et al. 2010). Interestingly, a recent tosensory cortex, GABAARs expressed by OPCs switched
study showed that NMDARs composed of NR1 and from a synaptic to an extrasynaptic localization during devel-
NR3 subunits in optic nerve myelin could be activated by opment, suggesting that expression of γ2 subunits may be
non-glutamatergic ligands such as glycine and D-serine physiologically regulated in OPCs (Vélez-Fort et al. 2010).
(Piña-Crespo et al. 2010). GABAAR activation in cultivated OLs can lead to intracel-
Several years ago, some high profile studies showed lular Ca2+ increases that are inhibited by the N-type VOCC
that glutamate excitotoxicity in OLs during ischemia was blocker nifedipine (Kirchhoff and Kettenmann 1992). Recent
mediated not only by AMPAR/KAR, but also by NMDAR studies in brain slices confirmed the ability of GABAAR ago-
activation during development (Karadottir et al. 2005; nists to induce Ca2+ entry into OLs (Vélez-Fort et al. 2010).
Micu et al. 2006). However, the pathophysiological and As in immature neurons, GABAAR activation in OPCs had an
physiological importance of NMDAR expression in the excitatory effect because of a depolarized reversal potential of
OL lineage was recently undermined by two studies analyz- Cl–, between –30 and –40 mV (Lin and Bergles 2004).
ing the consequences of a specific NMDAR genetic invali- GABAergic synapses from neurons are known to contact
dation in OLs. After a specific NR1 knockout in the OL OPCs (Lin and Bergles 2004). Neuron-to-OPC GABAergic
lineage, researchers failed to detect any physiological synapses tend to be lost when OPCs start differentiating into
or pathophysiological consequences of the absence of myelinating OLs. However, GABAARs can also be activated in
functioning NMDAR in OLs (De Biase et al. 2011; Guo cortical OPCs via extrasynaptic release of GABA (Vélez-Fort
et al. 2012); only upregulation of Ca2+-permeable AMPARs et al. 2010).
was noted (De Biase et al. 2011). Moreover, it has been As to physiological function, GABAAR activation is known
shown that NMDAR blockade during white matter isch- to regulate the growth rate of different types of neural precur-
emic injury could worsen outcome (Baltan et al. 2008), sug- sors by inhibiting or promoting their proliferation, depending
gesting that NMDAR activation may not mediate glutamate on the neural cell type and/or culture conditions. GABAAR
excitotoxicity. activation by exogenous application of GABA on perina-
tal OPCs in cultured organotypic cerebellar slices inhibits
their proliferation (Yuan et al. 1998). However, treating the
3.2 I O N OT RO P I C G A M M A-A M I N O BU T Y R I C
slices with GABAAR antagonist bicuculline had no effect
AC I D R E C E P TO R S
(Yuan et al. 1998), suggesting that endogenous activation of
Gamma-aminobutyric acid (GABA) receptor Cl– channels GABAAR may not be a major control mechanism for OL cell
(GABAAR) are members of the nicotinic ligand–gated ion cycle progression, at least in the cerebellum.
channel superfamily and are important in brain function for
mediating fast synaptic inhibition between neurons.
GABAARs are pentamers usually made of two α subunits, 3.3 AT P S E NS IT I VE P U R I N E RG I C
two β subunits, and one γ subunit. They exhibit great com- R EC E P TO R S
binatorial diversity because of a large number of GABAAR ATP is often released by neurons along with other neurotrans-
subunits: six different α subunits (α1–α6), three different β mitters and contributes to their excitability. ATP responses
subunits (β1–β3), and three different γ subunits (γ1–γ3). are mediated by both ionotropic (P2X) and metabotropic
Expression of functional GABAARs in OLs was first (P2Y) receptors (see section 4.3). Seven mammalian P2X
demonstrated in spinal cord organotypic culture (Gilbert et subunits (P2X1–7) assemble as trimers to form cationic
al. 1984) and OL cell culture (Von Blankenfeld et al. 1991), homomeric and heteromeric receptor channels with diverse
and later confirmed in brain slice recordings. The exact com- properties. Much like NMDA receptors, ATP-gated P2X
position of GABAARs in OLs remains unclear. For example, channels have marked Ca2+ permeability. P2X receptors are
GABAARs containing the γ subunit are identified based on expressed in CNS neurons, where they participate in fast syn-
their sensitivity to benzodiazepines, and initial study of OLs aptic transmission and modulation; P2X7 receptors mediate
in cell culture showed that GABAAR-mediated responses immunomodulatory responses in microglia and neurodegen-
were enhanced by applications of barbiturates and benzodi- eration after ischemia.
azepines and diminished by the inverse benzodiazepine ago- Oligodendrocyte lineage cells express P2X receptors in
nist DMCM (Von Blankenfeld et al. 1991). However, a study vitro and in vivo (Agresti et al. 2005a,b; Matute et al. 2007).

248 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
The electrophysiological, pharmacological, and molecular nAChRs are pentameric transmembrane proteins and per-
profile of P2XR expressed by OPCs and mature OLs mostly meable to cations. In vertebrates, nAChR subtypes are broadly
corresponds to the P2X7 subtype. P2X7 receptors are acti- classified into muscle and neuronal subtypes. In mammalian
vated at high concentrations of ATP (0.1–1 mM range), and CNS, neuronal nAChR can consist of eight α subunits (α2,
are capable of pore formation, resulting in sustained influx α3, α4, α5, α6, α7, α9, and α10) and three β subunits (β2,
of Ca2+. ATP and adenosine released from axons during elec- β3, and β4). nAChRs are either heteromers made of two α
trical impulse activity are known to regulate OPC migra- and three β or homomers made of five α subunits. Receptors
tion, proliferation, and differentiation (Agresti et al. 2005). composed of different subunits exhibit diverse pharmacologi-
Although this effect seems mostly mediated via metabotro- cal, regulatory, and functional properties.
pic purinergic receptor (see section 4.3), P2X7R may also Oligodendrocyte progenitor cells express different sub-
be involved. ATP released by astrocytes can also evoke a types of functional nAChRs. RT-PCR analysis and immu-
rapid and transient rise in intracellular Ca2+ in OPCs involv- nocytochemistry in OPCs cultured from rat corpus callosum
ing metabotropic purinergic receptor and P2X7 receptors have detected expression of nAChR subunits α3, α4, α5,
(Hamilton et al. 2010). α7, β2, and β4. Nicotine application can induce increased
Unlike many other ligand-gated ion channels in the OL lin- intracellular Ca2+ in OPCs cultivated from rat corpus cal-
eage, the robust expression of P2x7R by mature OLs suggests losum (Rogers et al. 2001), an increase sensitive to the Ca2+
a specific physiological function in these cells, possibly related VOCC blocker nifedipine, demonstrating that nAChR
to myelin formation and preservation, and allowing mature activation can depolarize OPCs enough to activate VOCCs
OLs to sense electrical activity in the axons they insulate. (Rogers et al. 2001). Recently, the presence of Ca2+-permeable
α7-containing nAChRs has been demonstrated in OPCs
recorded from mouse hippocampus slices (Vélez-Fort et al.
3.4 G LYC I N E R E C E P TO R S
2009). Importantly, this study could not detect any nAChR-
Similar to GABAARs, glycine receptor Cl– channels (GlyRs) mediated synaptic current in OPCs.
belong to the nicotinic ligand-gated ion channel superfam-
ily. Their main known function is to mediate fast synaptic
inhibition between neurons, mostly in the spinal cord and 4 M ETA B OT R O P I C R E C E P TO R S
brainstem. Structurally, GlyRs are pentameric transmembrane
proteins that can comprise two types of subunits: a 48-kDa α Metabotropic neurotransmitter receptors are G-protein–
subunit with four variants (α1, α2, α3, and α4) and a 58-kDa coupled proteins and important in neuromodulation. Acting
β subunit. Two main types of GlyRs are distinguished: a pen- indirectly on neuronal behavior, they modulate the function
tameric configuration of five α subunits and a heteromeric of ligand- and voltage-gated ion channels or influence diverse
configuration of two α and three β subunits. intracellular processes via second messengers such as Ca2+ and
Functional GlyRs have been described in newborn rat OL cAMP.
lineage cells and OPCs in spinal cord slices (Belachew et al.
1998; Kirchhoff et al. 1996). Although the exact structure and 4.1 M ETA B OT RO P I C G LU TA M AT E R EC E P TO R S
composition of GlyRs expressed in OLs is not determined,
RT-PCR using total RNA extracted from cultivated OPCs Metabotropic glutamate receptors (mGluR) can belong to one
of newborn rat cortex demonstrated only the presence of α2 of three groups. Group I includes mGluR1 and mGlur5; their
and β subunits (Belachew et al. 1998). However, GlyR subunit activation stimulates phospholipase C (PLC) to produce inosi-
expression is known to be both developmentally regulated and tol 1,4,5-triphosphate (IP3), inducing the release of Ca2+ from
region specific, so extensive analysis of GlyR subunit expres- intracellular stores. Group II, comprising mGluR2 and mGluR3,
sion is still needed in several regions of developing mouse and Group III (mGluR4, mGluR6–8) have a stimulating influ-
brain. ence on adenylate cyclase, leading to cAMP production.
As for most other ligand-gated ion channels, GlyR expres- Although OL cells can express all three groups of mGluRs
sion seems to peak at the OPC stage and decrease thereafter (Bagayogo and Dreyfus 2009; Luyt et al. 2006), their expres-
(Belachew et al. 1998). sion level is apparently developmentally regulated, becoming
very low in mature OLs. Group I mGluRs were recently shown
to regulate expression of Ca2+-permeable AMPARs in OPCs
3.5 N I C OT I N I C AC ET Y L C H O L I N E R E C E P TO R S
via elevation of Ca2+ and release of IP3 (Zonouzi et al. 2011).
Nicotinic acetylcholine ionotropic receptors (nAchRs) Group I mGluRs may also be involved in regulating normal
have numerous functions in the brain. First, they are funda- physiological OL cell death, as they offer protection against
mental to locomotion and behavior, mediating excitatory kainate-induced excitotoxicity (Luyt et al. 2006). Although
neurotransmission between motor neurons and their target the physiological role of mGluRs in OLs has not been eluci-
muscles. In adult brain, nAChRs participate in neuromodula- dated, a recent study showed that group I mGluRs can regulate
tion by influencing release of various neurotransmitters from release of growth factor BDNF in OL cell cultures (Bagayogo
neurons. During development, nAchRs participate in early and Dreyfus 2009). By cultivating cortical neurons with OLs,
activity-dependent network patterning. the authors showed that OL-derived BDNF could regulate

P H YS I O L O GY O F O L I G O D E N D R O C Y T E S • 249
the number of glutamatergic neurons, suggesting that OL 4.4 MUS C A R I N I C AC ET Y L C H O L I N E
may release trophic factors in an activity-dependent manner R EC E P TO R S
by activating group I mGluRs, and regulating neuron survival As in other metabotropic neurotransmission, mAchRs play
and synapse formation in vivo. various neuromodulatory functions. So far, five mAchR sub-
types have been identified (M1–M5).
4.2 M ETA B OT RO P I C G A BA B R E C E P TO R S Oligodendrocytes express functional mAchRs whose acti-
GABAB receptors (GABABRs) are G-coupled metabotropic vation by muscarine, or the specific agonist carbachol, triggers
receptors known to mediate slow inhibition at central synapses. intracellular signals such as MAPK, IP3, and Ca2+ mobili-
They can be composed of two major types of subunits, B1 and zation (Kastritis and McCarthy 1993). At the RNA level,
B2, among which different splice variants (GABAB1a–g) are expression of all mAChR subtypes has been reported in both
described. Most neurons coexpress both subunits, which form OPCs and OLs (De Angelis et al. 2011; Ragheb et al. 2001).
functional GABABRs via heterodimeric assembly, although The predominant mAChR subtype expressed in OLs is M3,
GABAB1 subunits may be able to form functional homodi- followed by M4, M2, M1, and M5 (Ragheb et al. 2001). As for
mers. In neurons, presynaptic GABABRs are known to sup- most other neurotransmitter receptors, all mAchR subtypes
press neurotransmitter release by inhibiting VOCCs. At the are downregulated in mature OLs (De Angelis et al. 2011).
postsynaptic membrane, GABABR activation usually inhibits In OPCs, M3, M1, and M4 activation significantly increases
adenylyl cyclase (AC) and activates Kir3-type K+ channels, OPC proliferation and survival (De Angelis et al. 2011),
hyperpolarizing the membrane. and mAchR activation inhibits OPC differentiation into
A study reported functional expression of GABABRs in myelinating OLs.
OLs (Luyt et al. 2007). Using RT-PCR and in situ hybridiza-
tion, this study showed the presence of RNA for GABAB1 and 4.5 D O PA M I N E R EC E P TO R S
GABAB2 subunits in OPCs and, to a modest extent, in a CD4-
derived OL cell line. Reduced expression of GABAB subunits in Dopamine D3 receptor (D3R) is known to be expressed by
mature OLs is compatible with the lack of immunoreactivity for precursors and immature OLs, but is absent from mature OLs
GABAB1 in white matter myelinating OLs (Charles et al. 2003). (Bongarzone et al. 1998; Niu et al. 2010). D3R expression cor-
In a physiological context, it is unknown whether GABABRs in related with the peak of myelination in corpus callosum. The
OLs are activated by synaptic or extrasynaptic release of GABA presence of D2R was also reported in a subset of mature inter-
from neurons and/or by a glial source of agonist. fascicular OLs in rat corpus callosum (Howard et al. 1998),
suggesting a model in which OLs switch from D3 to D2 as they
mature (Niu et al. 2010). Applying the D2/D3 specific agonist
4.3 P U R I N E RG I C M ETA B OT RO P I C R E C E P TO R S
quinpirole was shown to increase the number of OL precur-
Purinergic metabotropic receptors comprise both adenosine- sors in cell culture, an effect blocked by dopamine antagonist
sensitive P1 and ATP-sensitive P2Y receptors. Four subtypes haloperidol (Bongarzone et al. 1998). However, another study
of adenosine receptors have been described: P1A1 and P1A3 showed that haloperidol increased proliferation of rat OPCs in
receptors inhibit cAMP via Gi/o, whereas P1A2A and P1A2B culture (Niu et al. 2010). These contradictory results may stem
receptors stimulate cAMP via the protein Gs. Although all from the inability of quinpirole and haloperidol, acting on both
four subtypes have been detected in OPC culture by RT-PCR D2R and D3R, to discriminate between these receptors, whose
(Stevens et al. 2002), their presence in more mature OLs is ratio may vary with culture conditions and duration. Finally,
still undetermined. D2/D3 activation has been shown to protect OLs against glu-
Eight subtypes of P2Y receptors have been cloned in mam- tamate excitotoxicity (Rosin et al. 2005), suggesting that dop-
mals; they are sensitive either to adenine nucleotides ATP/ amine receptors may also regulate OL physiological cell death.
ADP (P2Y1, 11, 12, 13), uracil nucleotides UTP/UDP (P2Y4,
6), both (P2Y2), or UDP-glucose (P2Y14). P2Y receptors can 4.6 OT H E R L I G A N D S
activate PLC and induce Ca2+ release via Galpha(q/11) (P2Y1,
P2Y2, P2Y4, P2Y6, and P2Y11). They can also stimulate or Oligodendrocytes are also reported to express receptors to
inhibit AC via Galpha(s) and Galpha(i/o) proteins (P2Y12, a variety of other neuromodulators such as adrenaline α1
P2Y13, and P2Y14). receptors (Cohen and Almazan 1993), bradykinin recep-
Oligodendrocytes in cell culture and in acute corpus tors (Stephens et al. 1993), opioid μ and κ receptors (Knapp
callosum slices respond to ATP with Ca2+ elevation. et al. 2009), and cannabinoid CB1 and CB2 receptors (Gomez
Oligodendrocytes seem to predominantly express P2Y1R, its et al. 2011; Molina-Holgado et al. 2002).
activation leading to IP3-mediated release of Ca2+ from intra-
cellular stores (Agresti et al. 2005a,b). It was suggested that
P2Y responses are developmentally regulated, because only 5 S U M M A RY A N D P E R S P E C T I VE S
late OL precursors and mature OLs appeared to exhibit sig-
nificant ATP-induced Ca2+ elevation. However, recent stud- This chapter demonstrates that the study of OL physiology is
ies showed that P2Y1R activation in OPCs can stimulate cell still a work in progress. Oligodendrocytes have been clearly
migration, inhibit mitogenic response to PDGF, and promote shown to express a broad variety of membrane proteins involved
OL differentiation (Agresti et al. 2005a,b). in cell-to-cell signaling in the CNS. The major challenge now

250 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
is to elucidate and define the functional roles of these channels Alberdi E, Sanchez-Gomez MV, Matute C. 2005. Calcium and glial cell
and receptors in: (1) OL development; (2) neuron-OL and death. Cell Calcium 38:417–425.
Attali B, Wang N, Kolot A, Sobko A, Cherepanov V, Soliven B. 1997.
astrocyte-OL signaling; (3) participation in neuronal net- Characterization of delayed rectifier Kv channels in oligodendro-
work function; and (4) OL pathology. Elucidating OL physi- cytes and progenitor cells. J Neurosci 17:8234–8245.
ology will depend on optimizing the methods by which these Bagayogo IP, Dreyfus CF. 2009. Regulated release of BDNF by corti-
cells are identified, and investigating their physiology in situ cal oligodendrocytes is mediated through metabotropic glutamate
to analyze changes related to their development, morphology, receptors and the PLC pathway. ASN Neurol 1:e00001.
Baltan S, Besancon EF, Mbow B, Ye Z, Hamner MA, Ransom BR. 2008.
and survival. Many of the crucial questions still pending will White matter vulnerability to ischemic injury increases with age
be investigated using more advanced molecular and biophysi- because of enhanced excitotoxicity. J neurosci 28:1479–89.
cal approaches to determine the subcellular location of ionic Barres BA, Chun LLY, Corey DP. 1988. Ion channel expression by white
channels and receptors. matter glia: I Type 2 astrocytes and oligodendrocytes. Glia 1:10–30.
Central to understanding OL physiology is the question Belachew S, Rogister B, Rigo JM, Malgrange B, Mazy-Servais C,
Xhauflaire G, et al. 1998. Cultured oligodendrocyte progenitors
of whether these myelinating cells have additional functions, derived from cerebral cortex express a glycine receptor which is phar-
such as maintaining axonal integrity and intercellular com- macologically distinct from the neuronal isoform. Eur J Neurosci
munication. It will be important to define the distinct roles of 10:3556–3564.
receptors and channels throughout the various stages of OL Berger T, Schnitzer J, Orkand PM, Kettenmann H. 1992. Sodium and
development, as well as during OL turnover and regeneration calcium currents in glial cells of the mouse corpus callosum slice.
Eur J Neurosci 4:1271–1284.
in the brain. A second question crucial to understanding OL Berger T, Walz W, Schnitzer J, Kettenmann H. 1992. GABA- and
pathology—as well as normal physiology—is whether these glutamate-activated currents in glial cells of the mouse corpus callo-
signaling proteins regulate myelin formation directly, and sum slice. J Neurosci Res 31:21–27.
whether their engagement in myelin maintenance continues Bergles DE, Roberts JD, Somogyi P, Jahr CE. 2000. Glutamatergic syn-
in adults. A comparative analysis of the physiological proper- apses on oligodendrocyte precursor cells in the hippocampus. Nature
405:187–191.
ties of OLs in white and gray matter would be a valuable con- Berkefeld H, Fakler B, Schulte U. 2010. Ca2+-activated K+ channels:
tribution to these efforts. Finally, researchers must determine from protein complexes to function. Physiol Rev 90:1437–1459.
when, and whether, the physiological properties of OLs are Bevan S, Chiu SY, Gray PTA, Richtie JM. 1984. Sodium channels in rat
terminally established and the degree to which OL channel cultured astrocytes. J Physiol (Lond) 361:18P.
and receptor plasticity in the adult and aging brain is respon- Bichet D, Haass FA, Jan LY. 2003. Merging functional studies with
structures of inward-rectifier K(+) channels. Nat Rev Neurosci
sive to changing patterns of neuronal activity. 4:957–967.
In the past 20 years, perception of OLs has evolved from Bongarzone ER, Howard SG, Schonmann V, Campagnoni AT 1998.
images of relatively passive insulation providers for CNS Identification of the dopamine D3 receptor in oligodendrocyte pre-
axons to excitable cells sharing many membrane and conduc- cursors: potential role in regulating differentiation and myelin for-
tive properties with the neurons they support. Now, as new mation. J Neurosci 18:5344–5353.
Borges K, Kettenmann H. 1995. Blockade of K+ channels induced
questions are asked and new technologies become available to by AMPA/kainate receptor activation in mouse oligodendro-
answer those questions, it should surprise no one if the next cyte precursor cells is mediated by Na+ entry. J Neurosci Res 42:
two decades see even more rapid gains in understanding OL 579–593.
physiology—not only piling up facts, but applying the knowl- Burzomato V, Frugier G, Pérez-Otaño I, Kittler JT, Attwell D. 2010.
edge with ingenuity to OL pathologies in the human CNS. The receptor subunits generating NMDA receptor mediated currents
in oligodendrocytes. J Physiol 588:3403–3414.
Butt AM. 2006. Neurotransmitter-mediated calcium signaling in oligo-
dendrocyte physiology and pathology. Glia 54:666–675.
AC K N OW L E D G M E N T S Butt AM, Kalsi A. 2006. Inwardly rectifying potassium channels (Kir)
in central nervous system glia: a special role in glial functions. J Cell
The authors thank Li-Jin Chew for critically reading parts of this Mol Med 10:33–44.
Buttigieg J, Eftekharpour E, Karimi-Abdolrezaee S, Fehlings MG.
chapter. They also thank Pablo Paez and Ramesh Chittajallu 2011. Molecular and electrophysiological evidence for the expression
for providing figures. VG was supported by R01NS045702, of BK channels in oligodendroglial precursor cells. Eur J Neurosci
R01NS056427, P01NS0626860, MS Society RG 4019 and 34:538–547.
P30HD40677. JMM was supported by INSERM, CNRS, Campagnoni AT, Pribyl TM, Campagnoni CW, Kampf K,
and ANR. Amur-Umarjee S, Landrv CF, et al. 1993. Structure and developmen-
tal regulation of golii-mbp, a 105 kilobase gene that encompasses the
myelin basic protein gene and is expressed in cells in the oligodendro-
cyte lineage in the brain. J Biol Chem 268:4930–4938.
REFERENCES Charles KJ, Deuchars J, Davies CH, Pangalos MN. 2003. GABA
B receptor subunit expression in glia. Mol Cell Neurosci 24:
Agresti C, Meomartini ME, Amadio S, Ambrosini E, Serafini B, 214–223.
Franchini L, et al. 2005. Metabotropic P2 receptor activation regu- Chen H, Kintner DB, Jones M, Matsuda T, Baba A, Kiedrowski L, et
lates oligodendrocyte progenitor migration and development. Glia al. 2007. AMPA-mediated excitotoxicity in oligodendrocytes: role
50:132–144. for Na(+)-K(+)-Cl(–) co-transport and reversal of Na(+)/Ca(2+)
Agresti C, Meomartini ME, Amadio S, Ambrosini E, Volonté C, Aloisi exchanger. J Neurochem 102:1783–1795.
F, et al. 2005. ATP regulates oligodendrocyte progenitor migration, Chen S, Ren YQ, Bing R, Hillman DE. 2000. Alpha 1E subunit of
proliferation, and differentiation: involvement of metabotropic P2 the R-type calcium channel is associated with myelinogenesis.
receptors. Brain Res Brain Res Rev 48:157–165. J Neurocytol 29:719–728.

P H YS I O L O GY O F O L I G O D E N D R O C Y T E S • 251
Chittajallu R, Aguirre A, Gallo V. 2004. A population of cortical Herrero-Herranz E, Pardo LA, Bunt G, Gold R, Stuhmer W, Linker RA.
NG2-expressing cells display physiological properties inconsistent 2007. Re-expression of a developmentally restricted potassium chan-
with an oligodendrocyte progenitor identity. J Physiol 561:109–122. nel in autoimmune demyelination: Kv14 is implicated in oligoden-
Chittajallu R, Aguirre A, Gallo V. 2005. Downregulation of droglial proliferation. Am J Pathol 171:589–598.
platelet-derived growth factor-alpha receptor-mediated tyrosine Howard S, Landry C, Fisher R, Bezouglaia O, Handley V, Campagnoni
kinase activity as a cellular mechanism for K+-channel regulation A. 1998. Postnatal localization and morphogenesis of cells expressing
during oligodendrocyte development in situ. J Neurosci 25: the dopaminergic D2 receptor gene in rat brain: expression in non-
8601–8610. neuronal cells. J Comp Neurol 391:87–98.
Chittajallu R, Chen Y, Wang H, Yuan X, Ghiani CA, Heckman T, Itoh T, Beesley J, Itoh A, Cohen AS, Kavanaugh B, Coulter DA,
et al. 2002. Regulation of Kv1 subunit expression in oligodendrocyte et al. 2002. AMPA glutamate receptor-mediated calcium signaling
progenitor cells and their role in G1/S phase progression of the cell is transiently enhanced during development of oligodendrocytes.
cycle. Proc Natl Acad Sci U S A 99:2350–2355. J Neurochem 81:390–402.
Chiu SY, Schrager P, Richtie JM. 1984. Neuronal-type Na+ and K+ chan- Jentsch TJ, Friedrich T, Schriever A, Yamada H. 1999. The CLC chlo-
nels in rabbit cultured Schwann cells. Nature 311:156–157. ride channel family. Pflügers Arch 437:783–795.
Cohen RI, Almazan G. 1993. Norepinephrine-stimulated PI hydro- Kalsi AS, Greenwood K, Wilkin G, Butt AM. 2004. Kir41 expression by
lysis in oligodendrocytes is mediated by alpha 1A-adrenoceptors. astrocytes and oligodendrocytesin CNS white matter: a developmen-
Neuroreport 4:1115–1118. tal study in the rat optic nerve. J Anat 204:475–485.
De Angelis F, Bernardo A, Magnaghi V, Minghetti L, Tata AM. 2011. Karadottir R, Cavelier P, Bergersen LH, Attwell D. 2005. NMDA recep-
Muscarinic receptor subtypes as potential targets to modulate oli- tors are expressed in oligodendrocytes and activated in ischaemia.
godendrocyte progenitor survival, proliferation and differentiation. Nature 438:1162–1166.
Dev Neurobiol 72(5):713–728. Kastritis CH, McCarthy KD. 1993. Oligodendroglial lineage cells
De Biase LM, Kang SH, Baxi EG, Fukaya M, Pucak ML, Mishina M, express neuroligand receptors. Glia 8:106–113.
et al. 2011. NMDA receptor signaling in oligodendrocyte progenitors Kirchhoff F, Kettenmann H. 1992. GABA triggers a [Ca2+]i increase in
is not required for oligodendrogenesis and myelination. J Neurosci murine precursor cells of the oligodendrocyte lineage. EurJ Neurosci
31:12650–12662. 4:1049–1058.
De Biase LM, Nishiyama A, Bergles DE. 2010. Excitability and synap- Kirchhoff F, Mü lhardt C, Pastor A, Becker CM, Kettenmann H. 1996.
tic communication within the oligodendrocyte lineage. J Neurosci Expression of glycine receptor subunits in glial cells of the rat spinal
30:3600–3611. cord. J Neurochem 66:1383–1390.
Etxeberria A, Mangin JM, Aguirre A, Gallo V. 2010. Adult-born SVZ Kirischuk S, Scherer J, Moller T, Verkhratsky A, Kettenmann H. 1995.
progenitors receive transient synapses during remyelination in corpus Subcellular heterogeneity of voltage-gated Ca 2+ channels in cells of
callosum. Nat Neurosci 13:287–289. the oligodendrocyte lineage. Glia 13:1–12.
Feldman DH, Horiuchi M, Keachie K, Mccauley E, Bannerman P, Itoh Knapp PE, Adjan VV, Hauser KF. 2009. Cell-specific loss of kappa-opioid
A, et al. 2008. Characterization of acid-sensing ion channel expres- receptors in oligodendrocytes of the dysmyelinating jimpy mouse.
sion in oligodendrocyte lineage cells Glia 56:1238–1249. Neurosci Lett 451:114–118.
Fields RD. 2008. Oligodendrocytes changing the rules: action poten- Knutson P, Ghiani CA, Zhou J-M, Gallo V, McBain CJ. 1997. K+ chan-
tials in glia and oligodendrocytes controlling action potentials. nel expression and cell proliferation are regulated by intracellular Na+
Neuroscientist 14:540–543. and membrane depolarization in oligodendrocyte progenitor cells.
Fogal B, McClaskey C, Yan S, Yan H, Rivkees SA. 2010. Diazoxide pro- J Neurosci 17:2669–2682.
motes oligodendrocyte precursor cell proliferation and myelination. Kukley M, Dietrich D. 2009. Kainate receptors and signal integration by
PLos ONE 5:e10906. NG2 glial cells. Neuron Glia Biol 5:13–20.
Fulton DJ, Paez PM, Fisher R, Handley V, Colwell CS, Campagnoni AT. Li S, Stys PK. 2000. Mechanisms of ionotropic glutamate
2010. Regulation of L-type Ca2+ currents and process morphology receptor-mediated excitotoxicity in isolated spinal cord white matter.
in white matter oligodendrocyte precursor cells by golli-myelin pro- J Neurosci 20:1190–1198.
teins. Glia 58:1292–1303. Lin SC, Bergles DE. 2004. Synaptic signaling between GABAergic
Gallo V, Zhou JM, McBain C, Wright P, Knutson PL, Armstrong RC. interneurons and oligodendrocyte precursor cells in the hippocam-
1996. Oligodendrocyte progenitor cell proliferation and lineage pro- pus. Nat Neurosci 7:24–32.
gression are regulated by glutamate receptor-mediated K+ channel Luyt K, Slade TP, Dorward JJ, Durant CF, Wu Y, Shigemoto R, et al.
block. J Neurosci 16:2659–2670. 2007. Developing oligodendrocytes express functional GABA(B)
Ghiani CA, Yuan X, Eisen A, Knutson P, DePinho R, McBain CJ, receptors that stimulate cell proliferation and migration. J Neurochem
et al. 1999. Voltage-activated K+ channels and membrane depo- 100:822–840.
larization regulate accumulation of the cyclin-dependent kinase Luyt K, Váradi A, Durant CF, Molnár E. 2006. Oligodendroglial
inhibitors p27Kip1 and p21CIP1 in glial progenitor cells. J Neurosci metabotropic glutamate receptors are developmentally regulated and
19:5380–5392. involved in the prevention of apoptosis. J Neurochem 99:641–656.
Gilbert P, Kettenmann H, Schachner M. 1984. Gamma-Aminobutyric Mangin JM, Gallo V. 2011. The curious case of NG2 cells: transient trend
acid directly depolarizes cultured oligodendrocytes. J Neurosci or game changer? ASN Neuro 3:e00052.
4:561–569. Matute C. 2011. Glutamate and ATP signalling in white matter pathol-
Gomez O, Sanchez-Rodriguez A, Le M, Sanchez-Caro C, Molina-Holgado ogy. J Anat 219:53–64.
F, Molina-Holgado E. 2011. Cannabinoid receptor agonists modu- Matute C, Torre I, Pérez-Cerdá F, Pérez-Samartín A, Alberdi E,
late oligodendrocyte differentiation by activating PI3K/Akt and the Etxebarria E, et al. 2007. P2X(7) receptor blockade prevents ATP
mammalian target of rapamycin (mTOR) pathways. Br J Pharmacol excitotoxicity in oligodendrocytes and ameliorates experimental
163:1520–1532. autoimmune encephalomyelitis. J Neurosci 27:9525–9533.
Guo F, Maeda Y, Ko EM, Delgado M, Horiuchi M, Soulika A, et al. Micu I, Jiang Q, Coderre E, Ridsdale A, Zhang L, Woulfe J, et al. 2006.
2012. Disruption of NMDA receptors in oligodendroglial lineage NMDA receptors mediate calcium accumulation in myelin during
cells does not alter their susceptibility to experimental autoim- chemical ischaemia. Nature 439:988–992.
mune encephalomyelitis or their normal development. J Neurosci Molina-Holgado E, Vela JM, Arévalo-Martín A, Almazán G,
32:639–645. Molina-Holgado F, Borrell J, et al. 2002. Cannabinoids promote oli-
Hamilton N, Vayro S, Wigley R, Butt AM. 2010. Axons and astrocytes godendrocyte progenitor survival: involvement of cannabinoid recep-
release ATP and glutamate to evoke calcium signals in NG2-glia. tors and phosphatidylinositol-3 kinase/Akt signaling. J Neurosci
Glia 58:66–79. 22:9742–9753.

252 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Neusch C, Rozengurt N, Jacobs RE, Lester HA, Kofuji P. 2001. Kir41 Sontheimer H, Perouansky M, Hoppe D, Lux HD, Grantyn R,
potassium channel subunit is critical for oligodendrocyte develop- Kettenmann H. 1989. Glial cells of the oligodendrocyte lineage
ment and in vivo myelination. J Neurosci 21:5429–5438. express proton-activated Na+ channels. J Neurosci Res 24:496–500.
Niu J, Mei F, Li N, Wang H, Li X, Kong J, et al. 2010. Haloperidol pro- Sontheimer H Trotter J, Schachner M, Kettenmann H. 1989. Channel
motes proliferation but inhibits differentiation in rat oligodendro- expression correlates with differentiation stage during development
cyte progenitor cell cultures. Biochem Cell Biol 88:611–620. of oligodendrocytes from their precursor cells in culture. Neuron
Olsen ML, Sontheimer H. 2008. Functional implications for Kir41 2:1135–1145.
channels in glial biology: from K+ buffering to cell differentiation. Steinhauser C, Berger T, Frotscher M, Kettenmann H. 1992.
J Neurochem 107:589–601. Heterogeneity in the membrane current pattern of identified glial
Paez PM, Fulton D, Colwell CS, Campagnoni AT. 2009a. cells in the hippocampal slice Eur J Neurosci 4:472–484.
Voltage-operated Ca2+ and Na+ channels in the oligodendrocyte lin- Stephens GJ, Marriott DR, Djamgoz MB, Wilkin GP. 1993.
eage. J Neurosci Res 87:3259–3266. Electrophysiological and biochemical evidence for bradykinin
Paez PM, Fulton DJ, Spreur V, Handley V, Campagnoni AT. 2010. receptors on cultured rat cortical oligodendrocytes. Neurosci Lett
Multiple kinase pathways regulate voltage-dependent Ca 2+ influx 153:223–226.
and migration in oligodendrocyte precursor cells. J Neurosci Stevens B, Porta S, Haak LL, Gallo V, Fields RD. 2002. Adenosine: a
30:6422–6433. neuron-glial transmitter promoting myelination in the CNS in
Paez PM, Fulton DJ, Spreuer V, Handley V, Campagnoni CW, Macklin response to action potentials. Neuron 36:855–868.
WB, et al. 2009b. Golli myelin basic proteins regulate oligodendrog- Takumi T, Ishii T, Horio Y, Morishige K-I, Takahashi N, Yamada
lial progenitor cell migration through voltage-gated Ca 2+ influx. M, et al. 1995. A novel ATP-dependent Inward rectifier potas-
J Neurosci 29:6663–6676. sium channel expressed predominantly in glial cells. J Biol Chem
Paez PM, Spreuer V, Handley V, Feng JM, Campagnoni C, Campagnoni 270:16339–16346.
AT. 2007. Increased expression of golli myelin basic protein Tegla, CA, Cudrici, C, Rozycka, M, Soloviova, K, Ito, T, Singh, AK,
enhances calcium influx into oligodendroglial cells. J Neurosci et al. 2011. C5b-9-activated, K(v)13 channels mediate oligodendro-
27:12690–12699. cyte cell cycle activation and dedifferentiation. Exp Mol Pathol 91:
Patneau DK, Wright P, Winters C, Mayer ML, Gallo V. 1994. 335–345.
Glial cells of the oligodendrocyte lineage express both kainate- Tiwari-Woodruff S, Beltran-Parrazal L, Charles A, Keck T, Vu T,
and AMPA-preferring subtypes of glutamate receptor. Neuron Bronstein J. 2006. K+ channel Kv31 associates with OSP/claudin and
12:357–371. regulates oligodendrocyte development. Am J Physiol Cell Physiol
Piña-Crespo JC, Talantova M, Micu I, States B, Chen HS, Tu S, et al. 291:C687–698.
2010. Excitatory glycine responses of CNS myelin mediated by NR1/ Vélez-Fort M, Audinat E, Angulo MC. 2009. Functional alpha
NR3 “NMDA” receptor subunits. J Neurosci 30:11501–11505. 7-containing nicotinic receptors of NG2-expressing cells in the
Ragheb F, Molina-Holgado E, Cui QL, Khorchid A, Liu HN, Larocca hippocampus. Glia 57:1104–1114.
JN, et al. 2001. Pharmacological and functional characterization Vélez-Fort M, Maldonado PP, Butt AM, Audinat E, Angulo MC. 2010.
of muscarinic receptor subtypes in developing oligodendrocytes. Postnatal switch from synaptic to extrasynaptic transmission between
J Neurochem 77:1396–1406. interneurons and NG2 cells. J Neurosci 30:6921–6929.
Rogers SW, Gregori NZ, Carlson N, Gahring LC, Noble M. 2001. Volterra A, Meldolesi J. 2005. Astrocytes, from brain glue to commu-
Neuronal nicotinic acetylcholine receptor expression by O2A/oligo- nication elements: the revolution continues. Nat Rev Neurosci
dendrocyte progenitor cells. Glia 33:306–313. 6:626–640.
Rosin C, Colombo S, Calver AA, Bates TE, Skaper SD. 2005. Dopamine Von Blankenfeld G, Trotter J, Kettenmann H. 1991. Expression and
D2 and D3 receptor agonists limit oligodendrocyte injury caused developmental regulation of a GABAA receptor in cultured murine
by glutamate oxidative stress and oxygen/glucose deprivation. Glia cells of the oligodendrocyte lineage. Eur J Neurosci 3:310–316.
52:336–343. Wang W, Gao X-F, Xiao L, Xiang Z-H, He C. 2011. Kv7/KCNQ chan-
Sánchez-Gómez MV, Matute C. 1999. AMPA and kainate receptors nels are functionally expressed in oligodendrocyte progenitor cells.
each mediate excitotoxicity in oligodendroglial cultures. Neurobiol PLoS ONE 6:e21792.
Dis 46:6475–6485. Williamson AV, Compston DA, Randall AD. 1997. Analysis of the
Schmidt K, Eulitz D, Veh RW, Kettenmann H, Kirchhoff F. 1999. ion channel complement of the rat oligodendrocyte progeni-
Heterogeneous expression of voltage-gated potassium channels of tor in a commonly studied in vitro preparation. Eur J Neurosci 9:
the shaker family (Kv1) in oligodendrocyte progenitors. Brain Res 706–720.
843:145–160. Williamson,AV, Mellor JR, Grant AL, Randall AD. 1998. Properties of
Schmithorst VJ, Wilke M, Dardzinski BJ, Holland SK. 2002. Correlation GABA(A) receptors in cultured rat oligodendrocyte progenitor cells,
of white matter diff usivity and anisotropy with age during childhood Neuropharmacology 37:859–873.
and adolescence: a cross-sectional diff usion-tensor MR imaging Xiong ZG, Chu XP, Simon RP. 2006. Ca 2+-permeable acid-sensing
study. Radiology 222:212–218. ion channels and ischemic brain injury. J Membr Biol 209:
Simpson PB, Mehotra S, Lange GD, Russell JT. 1997. High density 59–68.
distribution of endoplasmic reticulum proteins and mitochondria Yamazaki Y, Hozumi Y, Kaneko K, Sugihara T, Fujii S, Goto K, et al.
at specialized Ca 2+ release sites in oligodendrocyte processes. J Biol 2007. Modulatory effects of oligodendrocytes on the conduction
Chem 272:22654–22661. velocity of action potentials along axons in the alveus of the rat hip-
Soliven B, Ma L, Bae H, Attali B, Sobko A, Iwase T. 2003. PDGF pocampal CA1 region. Neuron Glia Biol 3:325–334.
upregulates delayed rectifier via Src family kinases and sphingosine Yuan X, Eisen AM, McBain CJ, Gallo V. 1998. A role for glutamate and
kinase in oligodendroglial progenitors. Am J Physiol Cell Physiol its receptors in the regulation of oligodendrocyte development in cer-
284:C85–93. ebellar tissue slices. Development 125:2901–2914.
Soliven B, Szuchet S, Arnason BG, Nelson DJ. 1989. Expression and Zonouzi M, Renzi M, Farrant M, Cull-Candy SG. 2011. Bidirectional
modulation of K+ currents in oligodendrocytes: possible role in plasticity of calcium-permeable AMPA receptors in oligodendrocyte
myelinogenesis. Dev Neurosci 11:118–131. lineage cells. Nat Neurosci 14:1430–1438.

P H YS I O L O GY O F O L I G O D E N D R O C Y T E S • 253
21.
PHYSIOLOGICAL PROPERTIES OF NG2 + GLIAL CELLS
Dwight E. Bergles

A B B R E VI AT I O N S is known about the physiological characteristics of NG2+ glial


cells, with a focus on their membrane properties and the changes
AMPA 2-amino-3-(5-methyl-3-oxo-1, that these cells undergo during development and differentiation.
2-oxazol-4-yl) propanoic acid The expression of a diverse array of voltage-gated ion channels
CaV voltage-gated Ca2+ channel and neurotransmitter receptors, and their interaction with neu-
CNS central nervous system rons through direct synapses, raise new questions about the role
Cx32 Connexin32 of these enigmatic glial cells in CNS physiology.
EPSCs excitatory postsynaptic currents
GABA gamma-aminobutyric acid
MNI-L-glutamate 4-methoxy-7-nitroindolinyl-caged- 2 T E R M I N O L O GY
L-glutamate
NAV voltage-gated Na+ channel The development of antibodies that recognize NG2 made it pos-
NBQX 2,3-dihydroxy-6-nitro-7-sulfamoyl- sible to identify NG2+ glial cells in fixed CNS tissue (Levine and
benzo(f )quinoxaline Card 1987), and provided the impetus to develop transgenic mice
NMDA N-methyl-d-aspartate that enable visualization of these cells in living tissue for targeted
OPC oligodendrocyte precursor cell physiological studies in different brain regions (Karram et al.
PCR polymerase chain reaction 2008; Mallon et al. 2002; Zhu et al. 2008; Ziskin et al. 2007).
pre-OL premyelinating oligodendrocytes However, pioneering investigations into the physiological prop-
RS-CPP (RS)-3-(2-carboxypiperazin-4-yl)- erties of glial cells occurred long before these tools were available,
propyl-1-phosphonic acid and thus the identity of NG2+ cells was established. Although
SR-95531 6-Imino-3-(4-methoxyphenyl)- it is difficult to achieve certainty through retrospective analysis,
1(6H)-pyridazinebutanoic acid prior studies of cells defined as “β astrocytes,” “complex astrocytes
hydrobromide (Gabazine) or complex cells,” “GluR astrocytes,” and “glial progenitors,” are
TARP transmembrane AMPAR regulatory likely to represent evaluations of NG2+ cells, based on similarities
protein of their properties to those exhibited by unambiguously defined
TBOA DL-threo-β-benzyloxyaspartic acid NG2+ cells. NG2+ glial cells have the ability to differentiate into
TEA tetraethylammonium oligodendrocytes, and therefore they are routinely referred to as
oligodendrocyte precursor cells (OPCs); the term polydendro-
cytes has more recently been adopted to avoid the implication that
1 INTRODUCTION NG2+ cells only serve as oligodendrocyte progenitors. As new
roles for these ubiquitous cells are uncovered, the terms used to
The mammalian central nervous system (CNS) contains a popu- describe these cells may expand further. Moreover, the possibility
lation of stellate-shaped glial cells that express the chondroitin exists that there is further diversity within this broad grouping, a
sulfate proteoglycan NG2 (CSPG4) and the alpha receptor for topic that is discussed in the following. The studies described in
platelet derived growth factor (PDGFDR). These NG2+ glial this chapter were performed exclusively in brain tissue isolated
cells are widely distributed throughout the gray and white matter from rats and mice, although NG2+ cells (OPCs) also appear to
regions of the adult CNS, accounting for approximately 5% to be abundant in the CNS of humans and other primates (Chang
8% of all cells, and exhibit tiling behavior, with each cell occupy- et al. 2000; Peters 2004).
ing a unique domain. Although these cells were initially classified
as a type of astrocyte, based on their similar morphology, it is
now clear that they belong to the oligodendrocyte lineage and 3 M E M B R A N E P R O P E RT I E S
are physiologically distinct from typical protoplasmic astrocytes.
These cells serve as progenitors for oligodendrocytes during 3.1 M E M B R A N E C A PAC ITA N C E , R E S T I N G
both early development and adulthood, and retain the capac- P OT E N T I A L , A N D R E S T I N G M E M B R A N E
ity to divide throughout life. The recent development of trans- C O N D U C TA N C E
genic mice in which fluorescent proteins are selectively expressed
by NG2+ cells has provided unprecedented access to this glial In the mature CNS, NG2+ cells have elongated, small somata
population for physiological studies. This chapter discusses what approximately 10 Pm in diameter from which numerous,
254
A B

Hippocampus Cerebellum (ml)

C D

Optic Nerve Corpus Callosum

Figure 21.1 Morphology of NG2+ Cells. A–D. NG2 immunostaining in brain tissue from an adult mouse. NG2+ glial cells have small cell bodies from
which numerous processes extend into the surrounding neuropil. The number of processes, their length and extent of ramification, and their orienta-
tion varies between brain regions. Scale = 20 Pm.

highly branched processes extend (Fig. 21.1). Although simi- It is also likely that NG2+ cells express members of the two-
lar, their morphology varies significantly among brain regions: pore K+ channel (K2P) families, as the membrane resistance
in the hippocampus and cortex their processes extend radially, of many cells remains low in the presence of internal Cs+ and
while in the corpus callosum their processes are closely aligned tetraethylammonium (TEA), which are effective at blocking
to the orientation of callosal axons. Within these regions, vari- most voltage-dependent K+ channels, but have limited ability
ations are seen in the number of processes, their length, and to inhibit K2P channels (Kim, 2005).
their extent of ramification within the neuropil.
Whole-cell recordings from NG2+ cells in brain slices iso-
lated from young adult mice indicate that they have a total 3.2 VO LTAG E - G AT E D I O N C H A N N E L S
cell capacitance of approximately 20 pF, consistent with their 3.2.1 Voltage-Gated K+ Channels
modest size. By comparison, most mature neurons have capac-
itances in excess of 50 pF (Tyzio et al. 1999), because of their When positive current is injected into NG2+ cells from the
larger somata and extensive dendritic arbors. Unlike neurons, resting potential during whole-cell current-clamp record-
NG2+ cells do not show obvious polarity or morphological ings, their membrane depolarizes, revealing nonlinear
specializations, such as axons or dendrites that would suggest behavior that is indicative of the presence of voltage-gated
that there are defined input and output regions of the cell. In ion channels. This behavior contrasts with that exhibited by
physiological conditions, NG2+ cells exhibit a highly negative astrocytes, which exhibit passive responses because of their
resting potential of approximately –90 to –100 mV (Bergles much higher resting conductance (see chapter 16). Notably,
et al. 2000; De Biase et al. 2010; Lin and Bergles 2004), close it was the “complex” voltage waveform exhibited by NG2+
to the calculated equilibrium potential for K+, suggesting that cells that resulted in their designation as “complex cells”
the primary ion channels open at rest flux K+. In mature tissue or “complex astrocytes”. On depolarization, the membrane
significant numbers of channels are open at rest in NG2+ cells, potential rapidly moves to a new voltage and then contin-
as the membrane resistance averages 300 MΩ, a value compa- ues to rise slowly when current is maintained (Fig. 21.2).
rable to most neurons, but approximately tenfold higher than Both A-type and delayed-rectifier K+ channels contribute
astrocytes or oligodendrocytes. Among the channels that are prominently to this behavior. In voltage-clamp conditions,
likely to contribute to the resting potential are inwardly rec- rapidly activating and inactivating A-type K+ channels can
tifying K+ channels, in particular Kir4.1, a channel that has be observed in the initial response to a voltage step, whereas
been implicated in establishing the resting potential in vari- the sustained current is mediated by delayed-rectifier chan-
ous glial cell types (Djukic et al. 2007; Neusch et al. 2001). nels, both of which are inhibited by internal Cs+ and TEA.
P H YS I O L O G I C A L P R O P E RT I E S O F N G 2 + G L I A L C E L L S • 255
A NG2+ cell B Astrocyte In Kir4.1 knock-out mice, NG2+ cells (complex glia) exhib-
ited resting membrane potentials approximately 40 mV
more depolarized and much higher membrane resistances,
(~500 MΩ), suggesting that this K+ channel plays a domi-
nant role in controlling both the resting potential and rest-
ing membrane conductance of NG2+ cells, although an
indirect effect caused by removal of Kir4.1 from surround-
10 mV 0.5 mV ing astrocytes cannot be excluded.
100 ms 100 ms

3.2.2 Voltage-Gated Na+ Channels and


C D Membrane Excitability
NG2+ cells also express voltage-gated Na+ channels (NaVs) that
are sensitive to block by tetrodotoxin. The density of these chan-
nels varies over a wide range among the population (Chittajallu
et al. 2004; De Biase et al. 2010; Tong et al. 2009); however, the
current density (current normalized to cell capacitance) is sig-
nificantly lower than the density observed in neurons, and the
kinetics of these channels is slower (Tong et al. 2009). Voltage-
500 pA 2 nA
50 ms 50 ms
gated Na+ channels perform the crucial role of generating the
current necessary for the depolarizing upstroke of the action
potential in neurons. Given that NG2+ cells express the neces-
sary repertoire of channels involved in action potential genera-
E F
tion, they may also be excitable. However, in most NG2+ cells
in the mature CNS, injection of depolarizing current does
not elicit a regenerative, Na+-dependent spike in membrane
voltage characteristic of action potentials (Chittajallu et al.
2004; De Biase et al. 2010). Action potential generation is
dependent on the relative abundance of Na+ and K+ channels,
their activation and inactivation kinetics, as well as intrinsic
membrane properties (membrane capacitance and resistance).
In NG2+ cells, the relatively low density of NaV channels rela-
tive to Kv channels, and the slower kinetics of the NaV chan-
Figure 21.2 Comparison of the Physiological Properties of NG2+ Cells
and Astrocytes. A. Current-clamp recording from an NG2+ cell located nels expressed help to limit generation of action potentials.
in the stratum radiatum region of area CA1 in a hippocampal brain However, during early postnatal periods (< postnatal day 14),
slice. Injection of positive current injection elicited a complex mem- when K+ channel density is lower, one or more Na+ spikes can
brane response due to activation of NaV, CaV, and Kv. B. Whole-cell be elicited in some NG2+ cells on depolarization (Chittajallu
current-clamp recording from an astrocyte located in the same region. et al. 2004; De Biase et al. 2010; Karadottir et al. 2008)
The same current amplitudes used as shown in (A). Note that these cur-
rents only induced small deviations of the membrane potential because (Fig. 21.3). In comparison with neurons, these spikes have
of the high resting conductance. Vm = −90 mV. C. Whole-cell voltage higher threshold for initiation, smaller amplitude, and the
clamp recording from the same cell shown in (A, B). Hyperpolarizing waveform is not as tightly stereotyped, varying with the volt-
voltage steps revealed the presence of Kir currents, while depolarizing age at which the spikes are generated. NG2+ cells also typically
voltage steps reveal the presence of A-type and delayed-rectifier type K+ cannot generate prolonged trains of Na+ spikes in response to
currents. Steps: −60 pA to 300 pA, 40 pA increments from −90 mV
in (A, B), and −130 mV to 20 mV, 20-mV increments in (C, D) from continued depolarization, perhaps because of the slow recov-
−90 mV. (E,F) Tracer injection (neurobiotin) into an NG2+ cell (E) ery of NaV channels from inactivation and the presence of a
does not lead to transfer to other cells, whereas tracer injection into persistent NaV current (but see Karadottir et al. 2008). The
an astrocyte (F) results in transfer to many neighboring cells. Scale = incidence of this rudimentary form of excitability varies among
50 Pm. Adapted from Lin et al. 2004. the population even at young ages (Chittajallu et al. 2004),
but progressively declines during the first postnatal month,
as K+ channel expression increases (De Biase et al. 2010). A
It is likely that Kv.1.3 and Kv.1.5 channels contribute to these subset of NG2+ cells may still exhibit excitable behavior in
conductances because they are expressed in NG2+ cells in adults, and the proportion of cells with this capability appears
early postnatal tissue (Chittajallu et al. 2002). In response to differ among mammalian species (Karadottir et al. 2008).
to hyperpolarizing steps, an inward current develops nega- Because of their highly negative resting potential, NG2+ cells
tive to the K+ reversal potential. This current is blocked by must experience a larger depolarization to reach threshold for
low concentrations of extracellular Ba2+, and is absent in generation of these Na+ spikes, and spontaneous spiking is
glial-specific Kir4.1 knock-out mice (Djukic et al. 2007). rarely observed.

256 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
A
Early Postnatal
B
Young Adult astrocytes assemble into a functional syncytium, enabling the
* transfer of ions, metabolites, and signaling intermediates among
connected cells. Gap junctions also facilitate heterotypic inter-
actions among different cell types (Robinson et al. 1993). It is
this coupling among distinct glial cell types that has led to the
* hypothesis that there is a “pan-glial” syncytium in the CNS to
enable signaling and homeostasis among distinct glial networks.
30 mV NG2+ cells, by contrast, do not appear to form gap junctions
CC HC CC HC
200 ms with one another: Injection of a small molecular weight marker
into one NG2+ cell does not lead to its transfer to adjacent cells
Figure 21.3 Age-Dependent Changes in the Membrane Properties of
NG2+ Cells Whole-cell current-clamp recordings from NG2+ cells in the
(Bergles et al. 2000; Wallraff et al. 2004), unlike the extensive
corpus callosum (CC) and hippocampus (stratum radiatum, area CA1, intercellular transfer that occurs when one astrocyte is loaded
HC) in brain slices prepared from early postnatal (P5–P8) or mature with tracer (see Fig. 21.1E, F). This lack of direct coupling among
mice (P40–P45). The red trace shows responses to 160 pA currents. The NG2+ cells is consistent with the physical separation often pres-
red asterisk highlights a response where depolarization triggered a small ent between the processes of adjacent cells (see Fig. 21.2E, F).
NaV-mediated spike. The black asterisk highlights a response where the
largest current injection was 70 pA. Responses to two cells are shown
However, dye coupling is not the most sensitive measure of gap
for each region to highlight the variability that is seen within brain junctional coupling, and electrical coupling can occur in the
regions. From De Biase et al. 2010. absence of observable dye transfer. Although paired recordings
from NG2+ cells and neighboring neurons have failed to detect
electrical coupling (Lin et al. 2005; Mangin et al. 2008; Muller
3.2.3 Ca2+ Channels et al. 2009), recent studies indicate that some NG2 cells are
coupled to oligodendrocytes in the corpus callosum (Maglione
The influx of Ca2+ through membrane channels is a crucial et al. 2010), indicating that they are capable of forming hetero-
step in signal transduction pathways involved in regulating typic junctions (see also chapter 24).
growth, transformation, and functional plasticity. Whole- Mutations in Connexin32 (Cx32) are responsible for
cell recordings from NG2+ cells in conditions appropriate for Charcot-Marie-Tooth type X1 disease, a peripheral neu-
isolating Ca2+ currents have revealed the presence of inward ropathy associated with demyelination (see also chapter 62).
currents sensitive to inhibition by L- and T-type Ca2+ channel Connexin32 appears during first and second week of postnatal
antagonists (Haberlandt et al. 2011). These currents are rather development and is expressed by mature oligodendrocytes (and
small (peak current = –100 pA in 5 mM Ca2+), indicating that Schwann cells), which by electron microscopy has been local-
the overall density is low. Additional evidence for expression ized to paranodal loops of myelin, Schmidt-Lanterman incisures
of different voltage-gated Ca2+ channel (CaV) isoforms has and oligodendrocyte processes, rather than at contacts with
been obtained through PCR of mRNA extracted from sin- astrocytes (Kamasawa et al. 2005), suggesting that is involved
gle cells in tissue slices, indicating that transcripts for L-type in autologous or within-cell oligodendrocyte coupling. Some
(CaV 1.2, 1.3) and T-type (CaV 3.1, 3.2) are most abundant NG2+ cells in the dentate gyrus express Cx32, and in Cx32
(Haberlandt et al. 2011). These channels appear to have con- null mice, the turnover (proliferation and apoptotic removal)
ventional voltage dependencies, with a half-maximal current of NG2+ cells in this region is enhanced (Melanson-Drapeau
activation observed near –50mV. Intracellular Ca2+ dynamics et al. 2003), suggesting that early expression of this channel in
in NG2+ cells may be modified by Ca2+ induced Ca2+ release the oligodendrocyte lineage is important for the proper differ-
(Haberlandt et al. 2011), Na+-dependent Ca2+ exchange (Tong entiation of some NG2+ cells. At present, it is unclear whether
et al. 2009), and plasma membrane store refilling channels, this channel is involved in cell-cell coupling or functions as an
such as TRPC1 (Paez et al. 2011). In vitro studies of isolated unpaired connexin hemichannel to enable signal transduction.
NG2+ cells (OPCs) indicate that they undergo spontaneous Mature oligodendrocytes and CNS myelin are formed in Cx32
Ca2+ transients while migrating, events that are inhibited by null mice (Menichella et al. 2003), indicating that global expres-
the L-type Ca2+ antagonist nifedipine. Elevation of Ca2+ in sion of this channel among the NG2+ cells is not an absolute
these cells through CaVs triggers process retraction, suggest- requirement for oligodendrogenesis and myelination.
ing that these channels are involved in regulated the directed
movement of NG2+ cells during early development. Although
NG2+ cells rest more than 30 mV more negative than the acti-
vation threshold of CaVs, the higher membrane resistance and 5 N E U R OT R A N S M I T T E R R E C E P TO R
more depolarized membrane potential of NG2+ cells at this EXPRESSION
age may promote CaV activation.
5.1 G LU TA M AT E R EC E P TO R S

4 GAP JUNCTIONAL COUPLING 5.1.1 AMPA and Kainate Receptors


In vitro studies of oligodendrocyte lineage cells, in particular
One of the hallmarks of astrocytes is their extensive coupling so-called “O-2A” progenitors that have the capacity to dif-
through gap junctions. By forming these intercellular junctions, ferentiate into either oligodendrocytes or type-2 astrocytes,

P H YS I O L O G I C A L P R O P E RT I E S O F N G 2 + G L I A L C E L L S • 257
demonstrated that these glial cells express functional iono- polyamine toxins (e.g., philanthotoxin) and inward rectifica-
tropic receptors for glutamate (Barres et al. 1990; Wyllie et al. tion in current-voltage (I–V) plots, a phenomenon caused by
1991). Concurrent analysis of glial progenitors in brain slices intracellular block by polyamines. AMPA receptor currents
revealed that the presence of glutamate-gated currents in this in NG2+ cells show some inward rectification (Fig. 21.5C)
subset of glial cells was not merely an aberration of isolation (Bergles et al. 2000; Ge et al. 2006; Lin et al. 2005; Seifert
and maintenance in culture ( Jabs et al. 1994; Steinhauser and Steinhauser 1995; Ziskin et al. 2007) and partial sensitiv-
et al. 1994). Application of glutamate or kainate, an agonist ity to polyamine toxins (see Fig. 21.5C); however, the relative
of AMPA and kainate receptors that is not a substrate for glu- proportion of these receptors varies with development and
tamate transporters, to NG2+ cells voltage clamped at their between brain regions, with the highest proportion observed
normal resting potential in brain slices triggers an inward cur- in NG2+ cells in the molecular layer of the cerebellum (Lin
rent that is blocked by selective AMPA receptor antagonists et al. 2005).
and enhanced by cyclothiazide, an inhibitor of AMPA recep- Analysis of mRNA expression by single cell PCR and gene
tor desensitization (Fig. 21.4A). Although the conclusion that array analysis (OPCs) has confirmed that all four AMPA
such responses are produced by direct effects on NG2+ cells, receptor subunits are expressed in NG2+ cells (complex astro-
this assessment is complicated by the certain activation of cytes/GluR cells) (Seifert and Steinhauser 1995; Seifert et al.
receptors on surrounding neurons. However, similar responses 1997a,b), with GluA4 relatively enriched compared to most
can be induced by focal application of agonists, through tech- mature neurons.
niques such as local photolysis of caged glutamate, and by per-
forming experiments in the presence of TTX to limit neuronal
activation and depolarization of NG2+ cells through elevation 5.1.2 NMDA Receptors
of extracellular K+ (see Fig. 21.7). A small number of kainate NMDA receptors play a central role in the induction of
receptors also appear to be expressed by NG2+ cells (Kukley activity-dependent changes in synaptic strength. Although it
and Dietrich 2009), consistent with previous in vitro studies has long been thought that NMDA receptor expression was
(Patneau et al. 1994). restricted to neurons, recent physiological studies and gene
AMPA receptors are multisubunit complexes assem- expression analysis has revealed that these receptors also are
bled from four different subunits encoded by distinct genes expressed by NG2+ cells (Karadottir et al. 2005; Ziskin et al.
(GluA1–4); alternative splicing and interaction with acces- 2007) as well as astrocytes (Lalo et al. 2006) and oligodendro-
sory subunits (TARPs) can modify their properties further. cytes (Karadottir et al. 2005; Micu et al. 2006). Bath applica-
These receptors are cation channels that enable the flux of tion of NMDA (Karadottir et al. 2005) or focal application
Na+ and K+, and when formed without the GluA2 subunit of NMDA elicits currents in voltage-clamped NG2+ cells that
also allow Ca2+ influx. The hallmark of GluA2 lacking, Ca2+ are blocked by NMDA receptor antagonists (see Fig. 21.4A)
permeable AMPA receptors is sensitivity to inhibition by (De Biase et al.; Ziskin et al. 2007). There are several aspects

A B C
(+40 mV)
+ CPP –25 mV

–45 mV
6 I

–65 mV
NMDA
4
+ Picrotoxin/SR-95531

ECI = –43mV
+ GYKI 53655

(–80 mV) 1
–120 –80 –20
Kainate THIP (10 mM) V 40
–1

Figure 21.4 AMPA, NMDA, and GABAA Receptor Currents Recorded from NG2+ Cells A. Focal application of kainate (200 mM) to an NG2+ cell
in the corpus callosum elicited an inward current (lower traces) that was blocked by the AMPA receptor selective antagonist GYKI 53655 (100 PM).
Focal application of NMDA (100 PM) resulted in an outward current (Vm = 40 mV) that was blocked by the NMDA receptor selective antagonist
D,L-CPP (10 PM). B. Focal application of the GABAA receptor agonist THIP to an NG2+ cell in a hippocampal slice elicited transient currents that
reversed near the predicted Cl– reversal potential (ECl– = ~0 mV, CsCl-based electrode solution) and were blocked by the GABAA receptor antagonists
picrotoxin (100 PM) and SR-95531 (5 PM). Voltage steps were: −30 to 20 mV, 10 mV/step. C. Plot of the peak amplitudes (normalized) of responses
to focal THIP application recorded at different voltages in perforated-patch (•) or whole-cell ({) recording configurations. The reversal potential of
GABAA receptor responses was determined to be –43 mV (◊), indicating that they maintain a high level of intracellular Cl–. Adapted from Lin et al.
2004.

258 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
of NMDA receptor responses in NG2+ cells that differ from NG2+ cells exhibit a pharmacological profile consistent with
neurons. Physiological studies suggest that these receptors the presence of the D-5 subunit, a subunit that confers slow
exhibit reduced sensitivity to block by external Mg2+. The volt- deactivation kinetics. Pyramidal neurons undergo a develop-
age-dependent block by external Mg2+ ensures that Ca2+ influx mental switch from D-5 to D-1, a transition that accelerates
occurs only when the postsynaptic partner has been sufficiently the decay of GABAA receptor-mediated synaptic currents
depolarized. Because of the lower sensitivity to Mg2+, NMDA (Fig. 21.6). Measurements of GABAA receptor-mediated cur-
receptors in NG2+ cells can contribute significant current and/ rents in perforated patch recordings, which preserve the nor-
or Ca2+ influx without the need for substantial depolarization, mal internal Cl– concentration, revealed that these currents
although this property also may vary among brain regions and reverse at –43 mV, about 30 mV more positive than GABAA
species (Hamilton et al. 2009). NMDA receptors are formed reversal potential in neurons at this age, indicating that NG2+
by the assembly of two GluN1 and two GluN2, or less often cells maintain a high intracellular Cl– concentration (see Fig.
GluN3 subunits. Receptors formed from GluN2C and 2D sub- 21.4C) (Lin and Bergles 2004; Tanaka et al. 2009; Tong et al.
units have lower Mg2+ sensitivity, and in accordance with the 2009). NG2+ cells in the cortex also express GABAA recep-
physiological studies, mRNA encoding 2C subunits have been tors, suggesting that this is a feature common to these cells
observed in NG2+ cells (OPCs) (Cahoy et al. 2008). However, (Tanaka et al. 2009; Velez-Fort et al. 2010). However, the
NMDA receptors are expressed at a much lower level than in expression of GABAA receptors appears to be much lower in
neurons, and some NG2+ cells appear to lack these receptors NG2+ cells in the corpus callosum, perhaps because there are
entirely (De Biase et al. 2010; Ziskin et al. 2007). few GABAergic fibers in this region.

5.2 G A BA A R E C E P TO R S 5.3 OT H E R N EU ROT R A NS M IT T E R


R EC E P TO R S
GABAA receptors help limit neuronal excitation by hyperpo-
larizing the membrane potential and by increasing the mem- The breadth of neurotransmitter receptor expression by NG2+
brane conductance (shunting), and have been shown to play cells is slowly being uncovered. Recent studies suggest that
important roles in regulating neuronal development. NG2+ NG2+ cells in the hippocampus express nicotinic acetylcho-
cells in the hippocampus and barrel cortex express func- line receptors (Velez-Fort et al. 2009), metabotropic glutamate
tional GABAA receptors that exhibit characteristics similar to receptors (Haberlandt et al. 2011), D-1 adrenergic receptors
those expressed by young pyramidal neurons. Currents medi- (Papay et al. 2004), and P2 purinergic receptors (Hamilton
ated by these receptors have been observed in outside-out et al. 2009). Indeed, gene expression studies (Cahoy et al.
patches removed from physiologically defined NG2+ cells (see 2008) suggest that receptors for most if not all neurotransmit-
Fig. 21.4B) (Lin and Bergles 2004), providing unequivocal ters are expressed by NG2+ cells. However, a systematic, func-
evidence for the expression of GABAA receptors by these pro- tional assessment of the effects of different neurotransmitters
genitors. In the hippocampus, GABAA–mediated responses in in different brain regions remains to be performed.

A B C
mEPSCs control
+ GYKI 53655 +spermine

Control
0.5

Vm(mV)

–80 –40 40
10 pA
τ =1.64 ms
+CTZ 500 ms –0.5
200 pA 2 pA
4 ms INORM
10 ms
–1

Figure 21.5 Glutamatergic Synaptic Currents in NG2+ Cells in the Cerebellum. A. Whole-cell voltage-clamp recording from an NG2+ cell in the
molecular layer of the cerebellum (cerebellar slice) showing EPSCs elicited by stimulation of a climbing fiber. The response was potentiated by cycloth-
iazide (CTZ; 100 PM), a compound that reduces desensitization of AMPA receptors, and blocked by GYKI 53655 (100 PM) (Vm = −80 mV). B.
Spontaneous miniature EPSCs recorded in the presence of tetrodotoxin (1 PM). C. Current-voltage plot showing the voltage dependence of climbing
fiber-evoked EPSCs recorded from NG2+ cells with or without the polyamine spermine included. Black circles and solid lines indicate responses
recorded with internal spermine; the reduced outward current in this configuration indicates the contribution of Ca2+-permeable AMPA receptors.
Error bars are ± SEM. From Lin et al. 2005.

P H YS I O L O G I C A L P R O P E RT I E S O F N G 2 + G L I A L C E L L S • 259
6 N E U R O N –N G 2 + C E L L SY N A P S E S can be activated. In neurons, rapid activation of ionotropic
receptors is achieved through exposure to high concentra-
Synaptic junctions have long been considered to be the sole tions of glutamate that are released from transmitter-laden
provenance of neurons. These structures enable rapid com- vesicles into the small volume of the synaptic cleft. The
munication between distinct elements in neural circuits and near-synchronous activation of receptors results in excitatory
provide a substrate for neural plasticity. However, a wealth of postsynaptic currents (EPSCs) that reach a peak in less than
studies now indicate that NG2+ cells represent an exception a millisecond, whereas rapid dilution through diffusion and
to this rule, as axons form functional synaptic junctions with buffering by transporters ensures that such responses are
these glial cells in both white and gray matter regions of the transient. EPSCs mediated by AMPA receptors are also vis-
CNS. The existence of these neuron–NG2+ cell synapses has ible in whole-cell recordings from NG2+ cells in brain slices
been established through a variety of physiological and ana- following stimulation of surrounding axons (see Fig. 21.5A)
tomical criteria (described in the following), and this mecha- (Bergles et al. 2000; Jabs et al. 2005). These responses reach a
nism for rapid communication appears to be one of the most peak in a few hundred microseconds and decay exponentially,
defining physiological features of these progenitors. NG2+ similar to EPSCs in neurons, occur with minimal delay after
cells are targets of innervation, that is, they are postsynaptic stimulus onset, and are blocked by AMPA receptor antago-
elements. It is not yet known if NG2+ cells also act as presyn- nists. Moreover, miniature EPSCs of varying amplitude and
aptic elements capable of transmitting signals to surrounding time of occurrence occur in these glial cells, consistent with
neurons through NG2+ cell–neuron synapses. the stochastic, spontaneous vesicle fusion events that occur at
synapses between neurons (see Fig. 21.5B). In further support
of a direct fusion of vesicles opposite NG2+ cell membranes,
6.1 G LU TA M AT E RG I C SY NA P S E S
these responses are not dramatically potentiated by cycloth-
AMPA receptors have a relatively low affinity for glutamate iazide, as would be expected if they resulted from activa-
and are desensitized by low glutamate concentrations, plac- tion by glutamate that spills over from neighboring synapses
ing tight constraints on the manner in which these receptors between neurons (Dzubay and Jahr 1999). In contrast with

A1 B
* * *
+TTX

20 pA
A2 1s
5 pA
25 ms
control
20 pA
C
20 ms

A3

τ =21.9 ms
+SR 95531
5 pA 1 mV
20 ms 200 ms

Figure 21.6 GABAergic Synaptic Currents Recorded From NG2+ Cells in the Hippocampus. A1. Spontaneous miniature GABAA receptor-mediated
currents recorded from an NG2+ cell in the presence of tetrodotoxin (1 PM) to block action potentials and NBQX (5 PM) to block AMPA receptors.
A2. Overlay of three spontaneous GABAA receptor-mediated currents (highlighted by asterisks in A1). Note the slow decay kinetics. A3. Average time
course of 64 events recorded from this NG2+ cell. These responses decay approximately 50% more slowly than IPSCs recorded from CA1 pyramidal
neurons. B. Evoked responses recorded from an NG2+ cell in voltage-clamp showing the pronounced synaptic depression with repetitive stimulation.
These responses were blocked by tetrodotoxin (red trace). C. Spontaneous GABAA receptor mediated depolarizations recorded from an NG2+ cell in
the perforated-patch configuration following stimulation of interneurons with carbachol. Note the small depolarizations produced by activation of
these synapses. Recordings were performed in 5 PM NBQX and 5 PM RS-CPP, and all events were blocked by subsequent application of the GABAA
receptor antagonist SR-95531. All recordings were from cells in the stratum radiatum region of area CA1 in the whole-cell voltage-clamp configura-
tion. From Lin et al. 2005.

260 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
most neuronal glutamatergic synapses, NMDARs contribute of AMPA receptors at neuron–NG2+ cell synapses have been
little to spontaneous or evoked EPSCs (De Biase et al. 2010), observed in response to theta-burst stimulation of axons (Ge
suggesting that they are either predominantly extrasynap- et al. 2006), group I mGluR stimulation (Zonouzi et al. 2011),
tic or there are only a small number of receptors present at and NMDA receptor deletion (De Biase et al. 2011).
each synapse. The contribution of Ca2+ permeable receptors
to these EPSCs varies, with the highest level observed in the
6.2 G A BA E RG I C SY NA P S E S
molecular layer of the cerebellum (see Fig. 21.5C).
Anatomical evidence for the existence of axon–NG2+ cell +
NG2 cells also form synaptic junctions with GABAergic
synapses has been obtained from post hoc analysis of physi- interneurons. Spontaneous, miniature, and evoked GABAA
ologically defined cells and in brain sections. Examples of receptor-mediated currents have been observed in recordings
mitochondria-rich presynaptic boutons containing small ves- from NG2+ cells in the hippocampus ( Jabs et al. 2005; Lin
icles approximately 30 nm in diameter are observed in direct and Bergles 2004; Mangin et al. 2008) and cortex (Tanaka
apposition to the processes of NG2+ cells in electron micro- et al. 2009; Velez-Fort et al. 2010) in the presence of gluta-
graphs (Bergles et al. 2000; Haberlandt et al. 2011; Kukley mate receptor antagonists (see Fig. 21.6). Paired stimulation of
et al. 2007). The membranes of the cells are rigidly aligned at these inputs with a short interval induces paired-pulse depres-
these sites, and the space between the cells is filled with elec- sion, suggesting that these inputs have a high initial release
tron dense material, as seen at neuronal synapses. However, probability, and decay kinetics are slower than events observed
the postsynaptic density is less well defined, perhaps because in neurons at the same age, in accordance with the expression
of the lower number of NMDA receptors and associated regu- of GABAA receptors containing the D-5 subunit (Lin and
latory machinery. Bergles 2004).
Neuron–NG2+ cell synapses have been observed in all brain In the cortex, there is an age-dependent decrease in the
regions that have been examined, including the hippocam- amplitude and frequency of miniature and GABAA–mediated
pus (Bergles et al. 2000), cerebellum (Lin et al. 2005), cortex responses, and a lengthening of the decay of evoked responses
(Chittajallu et al. 2004), and brainstem (Muller et al. 2009), (Velez-Fort et al. 2010), a phenomenon caused by a shift from
suggesting that this mode of communication is a conserved direct activation of these receptors at synapses to indirect acti-
property of these glial cells. Moreover, these axoglial synaptic vation through spillover from presumed neuronal synapses.
junctions are not limited to gray matter, but also occur in the Thus, direct GABAergic synapses, but not GABAergic signal-
white matter of the cerebellum (Karadottir et al. 2005) and ing entirely, are restricted to the first postnatal month in corti-
corpus callosum (Kukley et al. 2007; Ziskin et al. 2007). cal NG2+ cells, in contrast with glutamatergic inputs, which
Estimates of the connectivity of NG2+ cells with axons, remain prominent into adulthood.
measured using hypertonic solution to force the release of In the cortex, GABAergic inputs to NG2+ cells appear to
docked and primed vesicles (Fig. 21.7B), indicate that the arise from parvalbumin-containing interneurons (basket and
highest degree of connectivity occurs in the corpus callosum, chandelier cells) (Tanaka et al. 2009), whereas the identity
a region that was thought to be essentially devoid of synapses of inputs in other regions is less certain. NG2+ cells often are
(De Biase et al. 2010). However, even within a particular brain located in close proximity to neuronal somata, conforming
region, the connectivity varies over a tenfold range. Although to the satellite glial cell classification. Despite this proximity,
far fewer events are seen than in neurons, when the smaller size paired recordings from interneurons and satellite NG2+ cells
of these cells is taken into account, the overall density per unit in the dentate gyrus indicated that these satellite glia were
area is remarkably similar, in keeping with the comparable never innervated by the neighboring interneuron, although
density of expression of AMPA receptors. GABAergic synaptic inputs were visible in these NG2+ cells
NG2+ cells do not appear to be innervated by a unique (Mangin et al. 2008). These findings suggest that GABAergic
population of fibers, rather they receive inputs appropri- innervation patterns are not random or based simply on
ate to the region they are located; in the CA1 region of the proximity.
hippocampus they form synapses with Schaffer collateral–
commissural fibers, in the dentate gyrus with granule cells,
the medial nucleus of the trapezoid body with axons from the 7 C H A N G E S IN T HE P H YS I O L O G I C A L
cochlear nucleus, and the molecular layer of the cerebellum P R O P E RT IE S O F N G 2 + C E L L S W IT H
with parallel and climbing fibers. Moreover, these inputs arise D IF F E R E N T I AT I O N
from the same axons that form axodendritic synapses with
surrounding neurons, as shown through paired recordings in In vivo genetic fate tracing studies indicate that NG2+ cells
the cerebellum (Lin et al. 2005), hippocampus (Mangin et al. serve as progenitors for oligodendrocytes throughout the CNS.
2008), and brainstem (Muller et al. 2009). Although this capacity is realized most frequently during early
These inputs exhibit other characteristics of neuronal postnatal life, NG2+ cells retain the ability to differentiate into
synapses, such as facilitation and depression with repetitive oligodendrocytes in the adult CNS (Kang et al. 2010; Rivers
stimulation, and release can be altered through activation of et al. 2008), a phenomenon that is enhanced following oligo-
presynaptic receptors (Ziskin et al. 2007), indicating that dendrocyte death (Traka et al. 2010; Tripathi et al. 2010) and
these junctions have assembled many of the components following exercise (Simon et al. 2011). Thus, at any one time,
required for rapid modulation. Changes in the composition and particularly in the developing CNS when the majority of

P H YS I O L O G I C A L P R O P E RT I E S O F N G 2 + G L I A L C E L L S • 261
A1 NG2 + cell B Pre-OL C OL

20 μm 20 μm 20 μm

D E F

20 pA
2.5 s
G H I

CPP
+40 mV +40 mV
CPP CPP
+NBQX/GYKI

+NBQX/GYKI TBOA
+NBQX/GYKI
–80 mV –80 mV
+NBQX/GYKI

25 pA/ 25 pA/
100 pA
100 pA 100 pA
250 ms 250 ms 250 ms

Figure 21.7 NG2+ Cell Differentiation Is Accompanied by Synapse Removal and Receptor Downregulation A–C. Representative NG2+ cells, premyeli-
nating oligodendrocytes (pre-OL), and oligodendrocytes (OL) recorded from cells in the corpus callosum. Insets show the responses to current injec-
tion, illustrating the different membrane properties exhibited by oligodendrocyte lineage cells in different stages of maturation. The red traces show
responses to 160 pA current injection; scale bars: 25 mV/100 ms. D–F. Responses of cells in these stages to focal application of hypertonic solution, a
manipulation that induces the release of docked, primed vesicles from synapses. Note the absence of inward currents from cells in the Pre-OL and OL
stages. G. Currents recorded from an NG2+ cell in response to local photolysis of MNI-L–glutamate (black dots). Lower traces show that responses
recorded at –80 mV were blocked by AMPA receptor antagonists (NBQX/GYKI53655). In these blockers, NMDA receptor-mediated responses
recorded at +40 mV were blocked by RS-CPP (upper traces). H. Responses recorded from a pre-OL in similar conditions as in (G). Note the reduced
amplitude of the AMPA receptor current and the absence of an NMDA receptor current. I. Responses from OLs recorded at –80 mV, showing the
effects of NBQX/GYKI53655, RS-CPP (upper trace, 0 Mg2+ ACSF/30 PM D-serine), and TBOA (lower trace, normal ACSF). From De Biase et al.
2010.

physiological studies have been performed, there are oligoden- in contrast, exhibit nearly tenfold lower membrane resistance
drocyte lineage cells in different states of maturation, creat- and a lower (more positive) resting potential, suggesting that
ing challenges for defining the characteristics of this dynamic the relative K+ conductance is reduced. Thus, maturation is
population. Using genetic labeling techniques it is possible to accompanied by a transient downregulation of ion channels.
identify cells in distinct stages of differentiation (De Biase et al. These changes are accompanied by a dramatic reduction in
2010). Studies of these cells have revealed that NG2+ cells rap- connectivity with surrounding neurons, as few synaptic cur-
idly alter their physiological properties as they begin to differ- rents are visible in premyelinating oligodendrocytes, and none
entiate, consistent with the analysis of glial cells in developing are observed in oliogdendrocytes (Fig. 21.7D–F) (De Biase
white matter (Berger et al. 1991). Transition to the premyeli- et al. 2010; Kukley et al. 2010). Accompanying this decrease
nating stage is accompanied by a tripling of the membrane in innervation, there is a tenfold reduction in AMPA receptor
capacitance reflecting the elaboration of additional processes, density, and NMDA receptor–mediated currents are rarely
and a doubling of the membrane resistance caused by a reduc- observed (Fig. 21.7G–I). Gene expression analysis further
tion in K+ and Na+ channel expression (De Biase et al. 2010; substantiates these findings, as this transition is accompanied
Kukley et al. 2010) (Fig. 21.7A–C). Mature oligodendrocytes, by a reduction in abundance of mRNAs that encode AMPA

262 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
receptor subunits GluA1 to 4 and the NMDA receptor sub- AMPA and NMDA receptors, and the weak sensitivity of their
unit GluN1 (NR1) (Cahoy et al. 2008), which is required NMDA receptors to Mg2+ block, could provide the means to
for the formation of NMDA receptors that are activated by link neuronal activity to Ca2+ elevation without the need for
glutamate. These studies suggest that within the oligodendro- significant depolarization, an effect that could be potentiated
cyte lineage, NG2+ cells are uniquely positioned to engage by depolarizing GABAergic inputs. Therefore, the effects of
in rapid synaptic communication with surrounding neurons. these synaptic inputs may be very local, and perhaps influence
At present, less is known about the changes in expression of the dynamics of their processes (Haberlandt et al. 2011; Kirby
other receptors during this transition period. et al. 2006) or trigger secretion of neurotransmitters, growth
factors, or matrix molecules (Hunanyan et al. 2010).
Our ever-increasing knowledge about the characteristics
8 P OT E N T I A L R O L E S O F of NG2+ cells indicate that they express many “neural” genes,
N E UR OT R A N S M IT T E R S I G N A L IN G and in vitro studies indicate that these cells can develop into
W IT H N G 2 + C E L L S neurons in response to particular growth factors (Kondo and
Raff 2000). However, genetic fate mapping studies in vivo
Synapses enable the rapid transmission of information indicate that NG2+ cells are primarily, if not exclusively, lin-
through neural circuits, and provide a substrate for modifi- eage restricted progenitors in the normal (uninjured) CNS,
cation of these networks in response to changing patterns of that either remain in the progenitor state or differentiate into
activity. The establishment of glutamatergic synapses with oligodendrocytes. Given the large number of proteins that are
NG2+ cells early in postnatal development, the high incidence required to assemble functional synaptic junctions, it seems
of these inputs among the population in diverse regions of the likely that these structures provide some advantage for sur-
CNS, and the persistence of these connections in adults, raise vival. It is possible that rapid synaptic communication with
the possibility that they play a prominent role in promoting NG2+ cells controls aspects of their behavior that is unrelated
changes in the behavior of these cells in response to neural to their potential to develop into oligodendrocytes. Moreover,
activity. Because these cells have the capacity to differentiate although it is clear that functional receptors are present at
into oligodendrocytes, such input may be used to control their these synapses, perhaps the primary role of these junctions is
maturation. Indeed, previous in vivo studies have shown that to establish areas of contact to enable transduction through
proliferation of these progenitors and myelination are influ- other signaling pathways.
enced by neural activity (Barres and Raff 1993). In support
of this hypothesis, in vitro studies have shown that glutamate
receptor signaling has diverse effects on these progenitors, 9 S U M M A RY A N D P E R S P E C T I VE S
inhibiting proliferation and differentiation (Gallo et al. 1996;
Gudz et al. 2006; Yuan et al. 1998), enhancing migration The relatively recent discovery that the mammalian CNS is
(Gudz et al. 2006), and controlling expression of myelin pro- populated by an abundant, widely distributed population of
teins (Wake et al. 2011). GABAA receptor activation also has NG2+ glial cells that are physiologically distinct from other glia
been shown to promote NG2+ cell migration in vitro (Tong and neurons, has raised many new questions about their roles
et al. 2009). Evaluation of the role of neurotransmitter signal- in the development and function of CNS circuits. Although
ing in these glial cells in vivo will require specific manipulation these cells have the capacity to differentiate into oligodendro-
of NG2+ cells, rather than pharmacological approaches, to cytes, and thus can be considered OPCs, it is unlikely that this
exclude the possibility that effects are indirectly mediated by designation fully encompasses their role in the adult CNS.
actions on neurons or other glial cells. Recent studies in which Indeed, the presence of these cells in regions that lack oligo-
the requisite NMDA receptor subunit NR1 (GluN1) subunit dendrocytes (e.g., molecular layer of the cerebellum), their
was selectively deleted from NG2+ cells indicate that NMDA persistence throughout life, and their relatively uniform distri-
receptor signaling is not required for NG2+ cell proliferation, bution and highly ramified morphology in gray and white mat-
migration, oligodendrogenesis, or myelination (De Biase et al. ter, suggest that they may be involved in homeostasis or active
2011), suggesting that there are redundant pathways to control regulation of neural circuits. NG2+ cells exhibit a number of
NG2+ cell behavior or that in vitro conditions do not accu- intriguing physiological characteristics, including the expres-
rately reproduce in vivo conditions. sion of a diverse array of ion channels and neurotransmitter
Glutamatergic and GABAergic synaptic inputs produce receptors, the formation of synapses with axons in gray and
only minimal depolarization of the membrane of NG2+ cells white matter, and the ability to proliferate and differentiate in
(Bergles et al. 2000; Jabs et al. 2005; Ziskin et al. 2007), with the adult CNS. Among the questions that remain to be fully
unitary responses averaging less than 1 mV in amplitude. How answered are the functions of the synaptic junctions that are
then would these inputs influence behavior, given that their formed with NG2+ cells, the extent to which these cells exhibit
resting potential is 20 to 30 mV more hyperpolarized than the excitability, their fate in the context of injury and disease, and
activation range of NaV and CaV channels? As these depolariza- the pathways that control their proliferation and differentia-
tions were measured at the soma, it is possible that greater volt- tion. Investigations into these questions will help answer why
age changes occur in their fine processes, which could engage these presumed progenitor cells persist in the adult CNS.
T-type CaV channels that are activated in a more negative Because it is necessary to study these cells in the complex
voltage range. Alternatively, the expression of Ca2+ permeable environment of the CNS, it is difficult to exclude the possibility

P H YS I O L O G I C A L P R O P E RT I E S O F N G 2 + G L I A L C E L L S • 263
of indirect effects mediated by surrounding neurons and glia, Chang A, Nishiyama A, Peterson J, Prineas J, Trapp BD. 2000.
particularly when the membrane resistance of these cells is low NG2-positive oligodendrocyte progenitor cells in adult human brain
and multiple sclerosis lesions. J Neurosci 20:6404–6412.
and can be influenced by changes in extracellular ion concen- Chittajallu R, Aguirre A, Gallo V. 2004. NG2-positive cells in the mouse
trations. In addition, most physiological studies have relied on white and grey matter display distinct physiological properties.
single cell recording in tissue slices, necessitating small sample J Physiol 561:109–122.
sizes, and raising the possibility of cell perturbation and selec- Chittajallu R, Chen Y, Wang H, Yuan X, Ghiani CA, Heckman T, et al.
tion bias. Notably, there have been few physiological studies 2002. Regulation of Kv1 subunit expression in oligodendrocyte pro-
genitor cells and their role in G1/S phase progression of the cell cycle.
of NG2+ cells in vivo. It also has been difficult to obtain com- Proc Natl Acad Sci U S A 99:2350–2355.
plete, high-resolution reconstructions of these cells, because De Biase LM, Kang SH, Baxi EG, Fukaya M, Pucak ML, Mishina M,
of the poor preservation of their fine processes in tissue pro- et al. 2011. NMDA receptor signaling in oligodendrocyte progenitors
cessed for transmission electron microscopy. These data could is not required for oligodendrogenesis and myelination. J Neurosci
reveal additional morphological specializations and the struc- 31:12650–12662.
De Biase LM, Nishiyama A, Bergles DE. 2010. Excitability and synap-
ture of junctions formed with different types of neurons and tic communication within the oligodendrocyte lineage. J Neurosci
glial cells, providing further insight into their roles in CNS 30:3600–3611.
circuits. Djukic B, Casper KB, Philpot BD, Chin LS, McCarthy KD. 2007.
The next decade will undoubtedly provide answers to some Conditional knock-out of Kir4.1 leads to glial membrane depolari-
of the essential questions associated with these enigmatic cells. zation, inhibition of potassium and glutamate uptake, and enhanced
short-term synaptic potentiation. J Neurosci 27:11354–11365.
The development of new genetic tools, such as inducible Dzubay JA, Jahr CE. 1999. The concentration of synaptically released
Cre lines, that enable unambiguous identification, fate trac- glutamate outside of the climbing fiber-Purkinje cell synaptic cleft.
ing, and manipulation of gene expression, will help evaluate J Neurosci 19:5265–5274.
how particular ion channels and signaling pathways influence Gallo V, Zhou JM, McBain CJ, Wright P, Knutson PL, Armstrong RC.
the behavior of NG2+ cells in vivo. When used in combina- 1996. Oligodendrocyte progenitor cell proliferation and lineage pro-
gression are regulated by glutamate receptor-mediated K+ channel
tion with mouse strains that enable cell-specific expression block. J Neurosci 16:2659–2670.
of genetically encoded indicators of voltage and Ca2+, as well Ge WP, Yang XJ, Zhang Z, Wang HK, Shen W, Deng QD, et al.
as light activated ion channels, it will be possible to develop- 2006. Long-term potentiation of neuron-glia synapses mediated by
ing a broader understanding of the properties of NG2+ cells Ca2+-permeable AMPA receptors. Science 312:1533–1537.
in the intact CNS, the extent of diversity within the popula- Gudz TI, Komuro H, Macklin WB. 2006. Glutamate stimulates oligo-
dendrocyte progenitor migration mediated via an alphav integrin/
tion, and their interactions with other neurons and glial cells. myelin proteolipid protein complex. J Neurosci 26:2458–2466.
Information about the physiological properties and functions Haberlandt C, Derouiche A, Wyczynski A, Haseleu J, Pohle J, Karram
of NG2+ cells may lead to new approaches to enhance remy- K, et al. 2011. Gray matter NG2 cells display multiple Ca2+-signaling
elination, promote axonal regeneration, and perhaps stimulate pathways and highly motile processes. PLoS One 6:e17575.
replacement of other cell types following injury or disease. Hamilton N, Vayro S, Wigley R, Butt AM. 2009. Axons and astrocytes
release ATP and glutamate to evoke calcium signals in NG2-glia.
Glia 58:66–79.
AC K N OW L E D G M E N T S Hunanyan AS, Garcia-Alias G, Alessi V, Levine JM, Fawcett JW, Mendell
LM, et al. 2010. Role of chondroitin sulfate proteoglycans in axonal
conduction in mammalian spinal cord. J Neurosci 30:7761–7769.
The author thanks Shih-chun Lin, Jennifer Ziskin, and Lindsay Jabs R, Kirchhoff F, Kettenmann H, Steinhauser C. 1994. Kainate acti-
DeBiase for contributing many of the results illustrated in this vates Ca(2+)-permeable glutamate receptors and blocks voltage-gated
chapter, Michele Pucak for her assistance with NG2 immuno- K+ currents in glial cells of mouse hippocampal slices. Pflugers Arch
labeling, and past and present members of his laboratory for 426:310–319.
Jabs R, Pivneva T, Huttmann K, Wyczynski A, Nolte C, Kettenmann H,
their insights. These studies were supported by grants from the et al. 2005. Synaptic transmission onto hippocampal glial cells with
National Institutes of Health. hGFAP promoter activity. J Cell Sci 118:3791–3803.
Kamasawa N, Sik A, Morita M, Yasumura T, Davidson KG, Nagy
JI, et al. 2005. Connexin-47 and connexin-32 in gap junctions
REFERENCES of oligodendrocyte somata, myelin sheaths, paranodal loops and
Schmidt-Lanterman incisures: implications for ionic homeostasis
Barres BA, Koroshetz WJ, Swartz KJ, Chun LL, Corey DP. 1990. Ion and potassium siphoning. Neuroscience 136:65–86.
channel expression by white matter glia: the O-2A glial progenitor Kang SH, Fukaya M, Yang JK, Rothstein JD, Bergles DE. 2010. NG2+ CNS
cell. Neuron 4:507–524. glial progenitors remain committed to the oligodendrocyte lineage in
Barres BA, Raff MC. 1993. Proliferation of oligodendrocyte precursor postnatal life and following neurodegeneration. Neuron 68:668–681.
cells depends on electrical activity in axons. Nature 361:258–260. Karadottir R, Cavelier P, Bergersen LH, Attwell D. 2005. NMDA recep-
Berger T, Schnitzer J, Kettenmann H. 1991. Developmental changes in tors are expressed in oligodendrocytes and activated in ischaemia.
the membrane current pattern, K+ buffer capacity, and morphology Nature 438:1162–1166.
of glial cells in the corpus callosum slice. J Neurosci 11:3008–3024. Karadottir R, Hamilton NB, Bakiri Y, Attwell D. 2008. Spiking and
Bergles DE, Roberts JD, Somogyi P, Jahr CE. 2000. Glutamatergic syn- nonspiking classes of oligodendrocyte precursor glia in CNS white
apses on oligodendrocyte precursor cells in the hippocampus. Nature matter. Nat Neurosci 11:450–456.
405:187–191. Karram K, Goebbels S, Schwab M, Jennissen K, Seifert G, Steinhauser
Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson C, et al. 2008. NG2-expressing cells in the nervous system revealed by
KS, et al. 2008. A transcriptome database for astrocytes, neurons, the NG2-EYFP-knockin mouse. Genesis 46:743–757.
and oligodendrocytes: a new resource for understanding brain devel- Kim D. 2005. Physiology and pharmacology of two-pore domain potas-
opment and function. J Neurosci 28:264–278. sium channels. Curr Pharm Des 11:2717–2736.

264 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Kirby BB, Takada N, Latimer AJ, Shin J, Carney TJ, Kelsh RN, et al. and piriform projection neurons in adult mice. Nat Neurosci
2006. In vivo time-lapse imaging shows dynamic oligodendrocyte 11:1392–1401.
progenitor behavior during zebrafish development. Nat Neurosci Robinson SR, Hampson EC, Munro MN, Vaney DI. 1993.
9:1506–1511. Unidirectional coupling of gap junctions between neuroglia. Science
Kondo T, Raff M. 2000. Oligodendrocyte precursor cells reprogrammed 262:1072–1074.
to become multipotential CNS stem cells. Science 289:1754–1757. Seifert G, Rehn L, Weber M, Steinhauser C. 1997a. AMPA receptor sub-
Kukley M, Capetillo-Zarate E, Dietrich D. 2007. Vesicular glutamate units expressed by single astrocytes in the juvenile mouse hippocam-
release from axons in white matter. Nat Neurosci 10:311–320. pus. Brain Res Mol Brain Res 47:286–294.
Kukley M, Dietrich D. 2009. Kainate receptors and signal integration by Seifert G, Steinhauser C. 1995. Glial cells in the mouse hippocampus
NG2 glial cells. Neuron Glia Biol 5:13–20. express AMPA receptors with an intermediate Ca2+ permeability.
Kukley M, Nishiyama A, Dietrich D. 2010. The fate of synaptic input Eur J Neurosci 7:1872–1881.
to NG2 glial cells: neurons specifically downregulate transmit- Seifert G, Zhou M, Steinhauser C. 1997b. Analysis of AMPA recep-
ter release onto differentiating oligodendroglial cells. J Neurosci tor properties during postnatal development of mouse hippocampal
30:8320–8331. astrocytes. J Neurophysiol 78:2916–2923.
Lalo U, Pankratov Y, Kirchhoff F, North RA, Verkhratsky A. 2006. Simon C, Gotz M, Dimou L. 2011. Progenitors in the adult cerebral cor-
NMDA receptors mediate neuron-to-glia signaling in mouse cortical tex: Cell cycle properties and regulation by physiological stimuli and
astrocytes. J Neurosci 26:2673–2683. injury. Glia 59:869–881.
Levine JM, Card JP. 1987. Light and electron microscopic localization of Steinhauser C, Jabs R, Kettenmann H. 1994. Properties of GABA and
a cell surface antigen (NG2) in the rat cerebellum: association with glutamate responses in identified glial cells of the mouse hippocam-
smooth protoplasmic astrocytes. J Neurosci 7:2711–2720. pal slice. Hippocampus 4:19–35.
Lin SC, Bergles DE. 2004. Synaptic signaling between GABAergic Tanaka Y, Tozuka Y, Takata T, Shimazu N, Matsumura N, Ohta A, et al.
interneurons and oligodendrocyte precursor cells in the hippocam- 2009. Excitatory GABAergic activation of cortical dividing glial
pus. Nat Neurosci 7:24–32. cells. Cereb Cortex 19:2181–2195.
Lin SC, Huck JH, Roberts JD, Macklin WB, Somogyi P, Bergles DE. Tong XP, Li XY, Zhou B, Shen W, Zhang ZJ, Xu TL, et al. 2009. Ca(2+)
2005. Climbing fiber innervation of NG2-expressing glia in the signaling evoked by activation of Na(+) channels and Na(+)/Ca(2+)
mammalian cerebellum. Neuron 46:773–785. exchangers is required for GABA-induced NG2 cell migration. J Cell
Maglione M, Tress O, Haas B, Karram K, Trotter J, Willecke K, Biol 186:113–128.
Kettenmann H. 2010. Oligodendrocytes in mouse corpus callosum Traka M, Arasi K, Avila RL, Podojil JR, Christakos A, Miller SD,
are coupled via gap junction channels formed by connexin47 and et al. 2010. A genetic mouse model of adult-onset, pervasive central
connexin32. Glia 58:1104–1117. nervous system demyelination with robust remyelination. Brain
Mallon BS, Shick HE, Kidd GJ, Macklin WB. 2002. Proteolipid pro- 133:3017–3029.
moter activity distinguishes two populations of NG2-positive cells Tripathi RB, Rivers LE, Young KM, Jamen F, Richardson WD. 2010.
throughout neonatal cortical development. J Neurosci 22:876–885. NG2 glia generate new oligodendrocytes but few astrocytes in a
Mangin JM, Kunze A, Chittajallu R, Gallo V. 2008. Satellite NG2 pro- murine experimental autoimmune encephalomyelitis model of demy-
genitor cells share common glutamatergic inputs with associated elinating disease. J Neurosci 30:16383–16390.
interneurons in the mouse dentate gyrus. J Neurosci 28:7610–7623. Tyzio R, Represa A, Jorquera I, Ben-Ari Y, Gozlan H, Aniksztejn L.
Melanson-Drapeau L, Beyko S, Dave S, Hebb AL, Franks DJ, Sellitto 1999. The establishment of GABAergic and glutamatergic synapses
C, et al. 2003. Oligodendrocyte progenitor enrichment in the con- on CA1 pyramidal neurons is sequential and correlates with the
nexin32 null-mutant mouse. J Neurosci 23:1759–1768. development of the apical dendrite. J Neurosci 19:10372–10382.
Menichella DM, Goodenough DA, Sirkowski E, Scherer SS, Paul DL. Velez-Fort M, Audinat E, Angulo MC. 2009. Functional alpha
2003. Connexins are critical for normal myelination in the CNS. 7-containing nicotinic receptors of NG2-expressing cells in the hip-
J Neurosci 23:5963–5973. pocampus. Glia 57:1104–1114.
Micu I, Jiang Q, Coderre E, Ridsdale A, Zhang L, Woulfe J, et al. 2006. Velez-Fort M, Maldonado PP, Butt AM, Audinat E, Angulo MC. 2010.
NMDA receptors mediate calcium accumulation in myelin during Postnatal switch from synaptic to extrasynaptic transmission between
chemical ischaemia. Nature 439:988–992. interneurons and NG2 cells. J Neurosci 30:6921–6929.
Muller J, Reyes-Haro D, Pivneva T, Nolte C, Schaette R, Lubke J, et al. Wake H, Lee PR, Fields RD. 2011. Control of local protein synthe-
2009. The principal neurons of the medial nucleus of the trapezoid sis and initial events in myelination by action potentials. Science
body and NG2(+) glial cells receive coordinated excitatory synaptic 333:1647–1651.
input. J Gen Physiol 134:115–127. Wallraff A, Odermatt B, Willecke K, Steinhauser C. 2004. Distinct
Neusch C, Rozengurt N, Jacobs RE, Lester HA, Kofuji P. 2001. Kir4.1 types of astroglial cells in the hippocampus differ in gap junction
potassium channel subunit is crucial for oligodendrocyte develop- coupling. Glia 48:36–43.
ment and in vivo myelination. J Neurosci 21:5429–5438. Wyllie DJ, Mathie A, Symonds CJ, Cull-Candy SG. 1991. Activation of
Paez PM, Fulton D, Spreuer V, Handley V, Campagnoni AT. 2011. glutamate receptors and glutamate uptake in identified macroglial
Modulation of canonical transient receptor potential channel 1 in the cells in rat cerebellar cultures. J Physiol 432:235–258.
proliferation of oligodendrocyte precursor cells by the golli products Yuan X, Eisen AM, McBain CJ, Gallo V. 1998. A role for glutamate and
of the myelin basic protein gene. J Neurosci 31:3625–3637. its receptors in the regulation of oligodendrocyte development in cer-
Papay R, Gaivin R, McCune DF, Rorabaugh BR, Macklin WB, McGrath ebellar tissue slices. Development 125:2901–2914.
JC, et al. 2004. Mouse alpha1B-adrenergic receptor is expressed in Zhu X, Bergles DE, Nishiyama A. 2008. NG2 cells generate both oli-
neurons and NG2 oligodendrocytes. J Comp Neurol 478:1–10. godendrocytes and gray matter astrocytes. Development 135:
Patneau DK, Wright PW, Winters C, Mayer ML, Gallo V. 1994. Glial 145–157.
cells of the oligodendrocyte lineage express both kainate- and AMPA- Ziskin JL, Nishiyama A, Rubio M, Fukaya M, Bergles DE. 2007. Vesicular
preferring subtypes of glutamate receptor. Neuron 12:357–371. release of glutamate from unmyelinated axons in white matter. Nat
Peters A. 2004. A fourth type of neuroglial cell in the adult central nerv- Neurosci 10:321–330.
ous system. J Neurocytol 33:345–357. Zonouzi M, Renzi M, Farrant M, Cull-Candy SG. 2011. Bidirectional
Rivers LE, Young KM, Rizzi M, Jamen F, Psachoulia K, Wade A, et al. plasticity of calcium-permeable AMPA receptors in oligodendrocyte
2008. PDGFRA/NG2 glia generate myelinating oligodendrocytes lineage cells. Nat Neurosci 14:1430–1438.

P H YS I O L O G I C A L P R O P E RT I E S O F N G 2 + G L I A L C E L L S • 265
22.
CY TOKINE, CHEMOKINE, AND GROW TH FACTOR
RECEPTOR S AND SIGNALING
Erik W. G. M. Boddeke, Bart J. L. Eggen, and Knut P. H. Biber

A B B R E VI AT I O N S MyD88 myeloid differentiation primary response


gene (88)
AAF IFN-α–activated factor NADE NT-associated cell death executor
AcP accessory protein NF-κB nuclear factor κB
B7 costimulatory molecule NGF nerve growth factor
BDNF brain-derived neurotrophic factor NIK NFκβ inducing kinase
CBP CREB binding protein NNT-1 novel neurotrophin-1
CCL chemokine (C-C motif ) ligand NRAGE NT-receptor–interacting MAGE homolog
CD cluster of differentiation NRIF NT-receptor–interacting factor
Cdc42 cell division control protein 42 NT neurotrophin
CDK cyclin-dependent kinase OAZ O/E-associated zinc finger protein
CLC cardiotrophin-like cytokine OSM oncostatin-M
CNS central nervous system P38 P38 mitogen–activated protein kinase
CNTF ciliary neurotrophic factor p300 E1A binding protein p300
COX-2 cyclooxygenase-2 PI3K phosphatidylinositol-3-OH kinase
CT 1 cardiotrophin-1 PDGF platelet-derived growth factor
CrkL v-CRK avian sarcoma virus CT10- PKB protein kinase B (also known as Akt)
homolog-like PLC phospholipase C
CXCL chemokine (C-X-C motif ) ligand RIP receptor interacting protein
CX3CL1 fractalkine RUNX runt-related transcription factor
ERK extracellular signal-regulated kinases SAPK stress activated protein kinase
FADD fas-associated death domain protein Shc SH2 containing proto-oncogene
FOXH1 homolog of Xenopus forkhead activin signal sIL-6R soluble IL-6 receptor
transducer-1 SODD silencer of death domain
GAS interferon-γ activating site STAT signal transducer and activator of
GDP guanosine diphosphate transcription
Gp130 glycoprotein 130 TGF transforming growth factor
GTP guanosine triphosphate TNF tumor necrosis factor
ICAM intercellular adhesion molecule TMEV Theiler’s Murine Encephalomyelitis Virus
IFN interferon TRADD tumor necrosis factor receptor-1–associated
IFNAR interferon-α/β receptor death domain protein
IKK IκB kinase TRAF tumor necrosis factor receptor–associated
IL interleukin factor
IL-1RA IL-1 receptor antagonist Trk tropomyosin-receptor-kinase
iNOS inducible nitric oxide synthase TYK2 tyrosine kinase-2
IRAK IL-1 receptor-associated kinase VCAM vascular cell adhesion protein
IRF interferon regulatory factor (ISGF3G) WNT acronym for wingless integrated
IRS-2 insulin receptor substrate-2
ISRE IFN-stimulated regulatory elements
JAK Janus kinase 1 INTRODUCTION
JNK c-jun N-terminal kinase
LIF leukemia inhibitory factor The various types of glia cells are of paramount importance
MAPK mitogen-activated protein kinase for homeostasis and maintenance of nervous tissue. They play
MHC major histocompatibility complex an essential role in processes that support survival, growth,

266
differentiation, and repair. For these tasks glial cells make 2 T H E I N T E R F E R O N R E C E P TO R FA M I LY
abundant use of cytokines, chemokines, and growth factors.
Via a large number of different receptors these signaling mole- Interferons (IFNs) are antiviral cytokines that are released
cules convey information to specific populations of target cells, from numerous cell types in response to a variety of stimuli
which in turn switch on appropriate intracellular signaling including viral infection or Toll-like receptor activation. The
cascades that lead to adaptation of cell function and gene tran- original discovery of IFN goes back to 1957, making IFNs the
scription activity. A diverse group of cytokines, chemokines, first cytokines discovered, and has set the starting point of
and growth factors activate specific receptors expressed in glia cytokine research.
cells. Notwithstanding the diversity of cytokine-, chemokine-, Three forms of IFN (type I and II IFN and the recently dis-
and growth factor receptors, the signaling cascades that are covered type III IFN) activate different IFN receptors. Type
initiated after receptor activation are conserved. Receptor I IFN has been subdivided in different classes (INF-α, β, δ,
activation is initiated by heteromerization of receptor subunits ε, κ, τ, ϖ), which are not all found in humans. IFN-γ is the
that is induced on cytokine/growth factor binding. In many only type II IFN. The type III form is IFN-λ, of which three
cases, downstream signaling is initiated by receptor tyrosine subtypes are known: IFN-λ1 (IL-29), IFN-λ2 (IL-28A), and
phosphorylation by receptor-associated kinases, followed by IFN-λ3 (IL-28B) (Takaoka and Yanai 2006). Because little is
recruitment of subsequent signaling factors. These include known about the potential role of type I IFN-δ, ε, κ, τ, and ϖ
signal transducers and activators of transcription (STAT), and type III IFNs in brain, the biology of these factors is not
mitogen-activated protein kinase (MAPK), the transcrip- discussed further.
tion factor complex NF-κB, and phosphoinositide-3 kinase/ Type I IFNs all signal through type I IFN receptors that
protein kinase B (PI3K/Akt) (Fig. 22.1). This chapter reviews are composed of a single α-chain (IFNAR1) and a single
the specific signaling pathways of cytokines, chemokines and β-chain (IFNAR2), that are coupled to the Janus tyrosine
growth factors that are involved in glia signaling. kinases TYK2 and JAK1, respectively (Fig. 22.2A). On

Figure 22.1 Receptor Signaling Cascades. A. Receptor dimerization caused by ligand binding: Example of receptors that dimerize because of recep-
tor binding. The two kinase domains cross-phosphorylate each other and thereby activate kinase domains. B. JAK/STAT signaling pathway: After
receptor activation and dimerization the associated JAKs now transphosphorylate tyrosine residues in the cytoplasmic domain of the receptor.
Phosphorylated tyrosine residues function as docking sites for STATs, which are then phosphorylated by JAKs. This results in STAT dimerization,
nuclear translocation, and target gene activation. C. PI3K signaling pathway: Receptor activation and subsequent phosphorylation of the regulatory
subunit for PI 3-kinase. D. Receptor signaling pathway resulting in MAPK-activation through a signaling cascade involving JAK/SHP2, RAS,
and RAF.

C Y TO K I N E , C H E M O K I N E , A N D G R OW T H FAC TO R R E C E P TO R S A N D S I G N A L I N G • 267
Figure 22.2 IFN Signaling. The IFN signaling pathways differ between type I and II IFN. All type I IFNs act through a heteromerized cell-surface
receptor composed of an α-chain (IFNAR1) and a β-chain (IFNAR2) and two associated cytoplasmic tyrosine kinases, JAK1 and Tyk2. A. Type I
IFN binds as a monomer to the two-chain IFNAR complex and induces binding of STAT2 to IFNAR2; subsequently, STAT1 is recruited to STAT2.
On tyrosine phosphorylation, STAT1 and STAT2 are released and associate with IRF-9/p48/ISGF3-γ to form the heterotrimeric IFN-stimulated gene
factor 3 (ISGF3) complex. ISGF3 translocates to the nucleus and binds to IFN-stimulated regulatory elements (ISREs). At the same time, STAT1
homodimers (AAF); other STAT complexes such as STAT3 or STAT5 homodimers, and STAT1/STAT3 and STAT5/CrkL heterodimers might
also form. AAF and STAT5/CrkL translocate to the nucleus and bind to IFN-γ activated sequences (GAS). B. Homodimers of type II IFN (IFN-γ)
activate a receptor complex that consists of two IFN-γR1 and two IFN-γR1 subunits, each of which is associated with JAK1 and JAK2, respectively.
On activation, two STAT1 molecules are recruited that form a dimer, which translocates to the nucleus. The activation of GAS via STAT1/STAT1
homodimers is the major route of IFN-γ signaling. Both type I and type II IFN may also activate NF-κB, ERK/MAPK, and PI3/AKT pathways.

activation by IFN-α/β, STAT2 is recruited to IFNAR2 in a IFN may also activate other signaling routes such as NF-κB,
JAK1-dependent manner and subsequently STAT1/STAT2 PI3/Akt, and ERK/MAPK pathway (Gough et al. 2008) (see
heterodimers are formed and released from the receptor com- Fig. 22.2B).
plex (Fig. 22.2A). These heterodimers form together with Interferons are crucial for a general antiviral response.
cytoplasmic interferon regulatory factor-9 (IRF-9), the het- Consequently, also viral infections of the CNS are controlled
erotrimeric IFN-stimulated gene factor 3 (ISGF3) complex, by IFNs. This has become evident in IFN receptor knock-out
which translocates into the nucleus in order to activate gene studies that provide evidence for a vital role of IFNAR, but
transcription at IFN-stimulated regulatory elements (ISREs) not IFN-γ-R in several viral infection models, indicating a piv-
(Fig. 22.2A). Other STAT complexes such as STAT5/CrkL otal role of type I IFN in clearing CNS viral response (Paul
(Crk-like protein) or STAT1/STAT3 or STAT1/STAT1 can et al. 2007).
also be formed on IFNAR activation. These latter STAT com- The exact source of type I IFN in the CNS in vivo is cur-
plexes, however, activate distinct genes via binding to IFN- rently not very well understood (Paul et al. 2007). Most likely,
γ–activated sequences (GAS), which indicates that GAS are microglia, astrocytes, and infiltrating blood cells are the main
major targets of IFN-γ–activated signaling. Homodimers of sources given the fact that the evidence that also neurons
IFN-γ activate a receptor complex that consists of two IFN- release type I IFN is mostly based on in vitro data (Paul et al.
γRI/IFN-γRII pairs, each of which is associated with JAK1 2007). Stimulation of cultured astrocytes, oligodendrocytes,
and JAK2, respectively (type II IFN signaling) (Fig. 22.2B). neurons, and microglia with type I IFN causes induction of
The activation of GAS via STAT1/STAT1 homodimers is the several interferon-stimulated genes, indicating that all cells
major route of IFN-γ signaling. However, to a lesser extent of the CNS respond to type I IFN. Supporting this notion is
also STAT1/STAT2 heterodimers are formed that together the observation that Theiler’s murine encephalomyelitis virus
with IRF-9 constitute ISGF3, which activates ISREs in the infection in mice causes an induction of MHCI in all CNS
nucleus (see Fig. 22.2B). Thus, the signaling pathways of IFN- cell types (Paul et al. 2007). These data indicate that IFNARs
α/β and IFN-γ show major overlap (Takaoka and Yanai 2006). are expressed broadly in CNS cells, which makes it difficult to
Next to the canonical JAK-STAT pathways, both type I and II relate IFN actions to specific cell types.

268 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Because type I IFN is widely used in the treatment of mul- of stem cells, neurogenesis, gliogenesis, and cell survival (Bauer
tiple sclerosis, it is important to understand its function in the et al. 2007; Lee et al. 2008). Apart from their role in develop-
CNS. Recent data using cell type–specific IFNAR deficient ment, IL-6–type cytokines regulate a variety of proinflamma-
animals show that the beneficial effect of type I IFN signal- tory and antiinflammatory processes and therefore are widely
ing in demeylinating disease requires IFNAR expression in implicated in the pathology of the CNS.
myeloid cells (Prinz and Kalinke 2010), which puts infiltrating IL-6–type cytokines bind to their corresponding ligand
macrophages and endogenous microglia in focus. This study recognition subunit (IL-6R or respectively OSMR, CNTFR,
shows that various aspects of myeloid cell immune activity IL-11R, or LIFR) and recruit one or two gp130 subunits to
are more pronounced in the absence of type I IFN signaling, activate downstream signaling pathways (Fig 22.3A) (Heinrich
indicating an antiinflammatory function of this cytokine in et al. 2003). Activation occurs via phosphorylation events of
demyelinating disease (Prinz and Kalinke 2010). In fact, there gp130-associated tyrosine kinases ( JAK kinases: JAK1, JAK2,
is little evidence that type I IFN affects other brain cells than JAK3, and/TYK2) and subsequent phosphorylation and acti-
microglia during demyelinating disease in vivo (Prinz and vation of signal transducers and activators of transcription
Kalinke 2010). Thus despite the observation that all endog- (STAT), mainly STAT3 and STAT1 (Heinrich et al. 2003). In
enous brain cells respond to type I IFN, many details about addition, phosphorylation of gp130 by JAKs may also result in
type I IFN activity in vivo remain to be established. the activation of mitogen-activated protein kinases (MAPKs),
Astrocytes and microglia are the prime source of IFN-γ including extracellular signal regulated kinase (ERK1/2), p38
in the CNS (Dafny and Yang 2005) and both cell types are and c-jun N-terminal kinases/stress-activated protein kinases
also targets of IFN-γ signaling. In contrast with type I IFN, ( JNK/SAPK). In addition to JAK/STAT and MAPK cas-
which mainly downregulates the proinflammatory capacity cades, IL-6–type cytokines may also activate the PI3K/Akt
of microglia, IFN-γ signaling in astrocytes and microglia pre- signaling pathway (Kamimura et al. 2003) (see Fig. 22.3A).
dominantly induces the release of proinflammatory cytokines Furthermore, IL-6 and its receptor display a remarkable prop-
and chemokines (Dafny and Yang 2005). It has been shown erty called trans-signaling (Rose-John et al. 2007). IL-6R is
recently that IFN-γ stimulates CXCL10 expression in astro- not only found as a membrane bound receptor, but also can be
cytes and microglia, whereas CXCL9 is induced in microglia released and thus occurs in soluble form (sIL-6R). In stark con-
but not astrocytes (Ellis et al. 2010). This cell type–specific trast to the effect of other soluble cytokine receptors, binding
effect of IFN-γ is caused by the transcription factor PU.1 that of IL-6 to its soluble receptor is not inhibitory. Instead, binding
is specifically expressed in myeloid cells, showing that next to IL-6 to its soluble receptors forms a ligand–receptor complex
the canonical IFN-γ signaling pathway (see Fig. 22.2B), the that is able to activate gp130 in the same way as the membrane-
presence of other transcription factors may determine the bound IL-6R (Fig. 22.3B). This unique process enables cells to
outcome of IFN-γ signaling (Ellis et al. 2010). Moreover, an respond to IL-6 even in the absence of endogenous IL-6R.
important role for IFN-γ in driving microglia-activation in a The shared receptor subunit of all IL-6–type cytokines
mouse neuropathic pain model was reported, indicating that gp130 is ubiquitously expressed and is therefore found in all
this cytokine may control microglia function in vivo (Tsuda types of brain cells. In contrast to gp130, the ligand recogni-
et al. 2009). The cellular source of IFN-γ, however, has not tion subunits are expressed more selectively. Leukemia inhibi-
been elucidated in this study. tory factor receptor and CNTFR are predominantly expressed
in cells of the oligodendrocyte lineage, rendering these cells
sensitive to LIF and CNTF. Accordingly, numerous findings
of LIF and CNTF in the maturation of oligodendrocytes dur-
3 I L - 6 –T Y P E C Y TO K I N E S A N D ing development and demyelinating diseases have been pub-
G P 13 0 -M E D I AT E D S I G N A L I N G lished. It was described that exogenously administered LIF
limited cuprizone-induced demyelination and LIF-deficient
The family of IL-6–type cytokines consists of seven pleio- mice showed both potentiated demyelination and oligoden-
tropic molecules, which are structurally and functionally drocyte loss after cuprizone challenge. Both were ameliorated
related, namely, IL-6, IL-11, leukemia inhibitory factor (LIF), by exogenous LIF, arguing for a direct beneficial effect of
ciliary neurotrophic factor (CNTF), oncostatin M (OSM), endogenous LIF receptor signaling. The numbers of oligo-
cardiotrophin-1 (CT 1), and novel neurotrophin-1 (NNT-1) dendrocyte progenitor cells in cuprizone-challenged mice
or cardiotrophin-like cytokine (CLC) (Heinrich et al. 2003). were not influenced by LIF, arguing for a direct effect of this
IL-6–type cytokines exert their effects via different receptor cytokines in differentiated oligodendrocyte. Endogenous LIF
complexes that all include glycoprotein 130 (gp130) and addi- receptor signaling may thus not only be protective for oligo-
tional ligand receptor subunits (Heinrich et al. 2003). Most dendrocytes but can also enhance remyelination making LIF
ligand recognition subunits (IL-6R, IL-11R, and CNTFR) rely a potentially therapeutic cytokine for limiting oligodendro-
for their intracellular signaling function on the gp130 subunit cyte damage (Marriott et al. 2008). Oncostatin-M regulator
(Heinrich et al. 2003). Because of this fact, numerous redun- is highly expressed in astrocytes, and its stimulation controls
dant properties have been reported for IL-6–type cytokines, the expression and release of proinflammatory cytokines like
and this cytokine family is also referred to as gp130 cytokines. TNF-α and IL-1β (Tanaka and Miyajima 2003). Microglia
IL-6–type cytokines are important for normal develop- differentially respond to IL-6, OSM, and CNTF. Whereas
ment of the brain by regulating proliferation and maintenance IL-6 and OSM induce proinflammatory cytokine production

C Y TO K I N E , C H E M O K I N E , A N D G R OW T H FAC TO R R E C E P TO R S A N D S I G N A L I N G • 269
Figure 22.3 IL-6 Signaling. A. IL-6 signals via two IL-6R subunits that on binding associate with two gp130 subunits. Because IL-6 receptors lack
signal-transduction capacity, signaling occurs via phosphorylation events of gp130-associated tyrosine kinases ( JAK kinases: JAK1, JAK2, JAK3, and/
TYK2) and subsequent phosphorylation and activation of signal transducers and activators of transcription (STAT), mainly STAT3 and STAT1. In
addition, phosphorylation of gp130 by JAKs may also result in the activation of mitogen-activated protein kinases (MAPKs), including extracellular
signal regulated kinase (ERK1/2), p38, and c-jun N-terminal kinases/stress-activated protein kinases ( JNK/SAPK). In addition, PI3K/Akt signal-
ing pathway may also be activated. B. IL-6R is not only found as a membrane bound receptor, but can be released and thus occurs in soluble form
(sIL-6R). Binding IL-6 to its soluble receptors forms a ligand–receptor complex that is able to activate gp130 in the same way as the membrane-bound
IL-6R (trans-signaling).

and thus activate these cells, treatment with CNTF in micro- also involved in tissue support, neuronal development, and
glia results in downregulation of cyclooxygenase-2 (COX-2) restoration of homeostasis.
without affecting TNF-α and IL-1β expression (Baker et al. Two related receptors for TNF-α are known, TNFR1 and
2010; Krady et al. 2008). It should be noted, however, that TFNRII. TNFRI contains a death domain, which TNFRII
these results are solely based on cultured cells and that very lacks. Accordingly, TNFRI is designated as a death receptor.
little is yet known about the expression of IL-6–type cytokine Both receptors are expressed by astrocytes, microglia, and
receptors in astrocytes and microglia in vivo. oligodendrocytes (Dopp et al. 1997). In glia cells, TNFRI is
constitutively expressed, whereas TNFRII expression is trig-
gered under inflammatory conditions and is found in hemato-
4 THE TUMOR NECROSIS poietic lineage cells. It has thus been demonstrated that under
R E C E P TO R FA M I LY inflammatory conditions microglia and Schwann cells express
TNFRI and TFNRII, whereas astrocytes and oligodendro-
The tumor necrosis factor (TNF) family contains approx- cytes primarily express TNFR1 (Dopp et al. 1997; Qin et al.
imately 20 members that are involved in the regulation of 2008). Both TNRF1 and TNRFII do not contain an intracel-
inflammation, immune responses, development of lym- lular signaling domain and thus signal via intracellular adaptor
phoid organs, and tissue homeostasis. These proteins act in molecules (Aggarwal et al. 2011). Receptor activation occurs
membrane bound form and as soluble cytokines. Most by oligomerization and requires internalization of the ligand–
studies on TNF-related cytokines in the central nervous receptor complex.
system (CNS) have addressed TNF-α (and to a lesser extent As mentioned, TNFRI contains a cytoplasmic death
TNF-β/lymphotoxin). Also in the brain TNF-α is considered domain that mediates induction of apoptosis and NF-κB–
a key inflammatory regulator that induces cytokine produc- mediated signaling cascades. Thus, on activation of TNFR1,
tion, inflammation, gliosis, blood-brain barrier damage, demy- dissociation of silencer of death domain (SODD), which
elination, and neural damage (Montgomery and Bowers 2011). prevents death domain signaling, takes place and recruit-
In the CNS, TNF-α is primarily produced by microglia, astro- ment of TRADD (TNF receptor–associated death domain)
cytes, and neurons (Hanisch 2002; Montgomery and Bowers initiates downstream signaling (Fig. 22.4). This involves sub-
2011). TNF-α not only drives inflammatory responses, but is sequent binding of FADD (Fas-associated death domain),

270 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Figure 22.4 Tumor Necrosis Factor-α Receptor Signaling. Binding of the TNF-α homotrimeric ligand induces trimerization of membrane-associated
tumor necrosis factor receptor 1 (TNFR1) subunits. Subsequently, recruitment of tumor necrosis factor receptor-1–associated death domain protein
(TRADD) to the cytoplasmic domain of TNFR1 occurs. TNFR1-TRADD recruits tumor necrosis factor receptor-associated factor-2 (TRAF2)
and receptor interacting protein (RIP). This trimeric complex activates c-jun N-terminal kinase ( JNK) and mediates activation of the inhibitory
κB kinase complex (IKK) that phosphorylates IκB. Phosphorylation of IκB releases and activates NF-κB. Additionally, the TNFR1-TRADD
binds Fas-associated death domain protein (FADD), which recruits caspase-8. The subsequent signaling cascade mediated by caspase-8 cleavage and
activation leads to apoptosis.

TRAF2 (TNF-receptor–associated factor 2) and RIP (recep- neurodegenerative diseases including multiple sclerosis (MS),
tor interacting protein). This receptor–signaling complex Parkinson disease (PD), and Alzheimer disease (AD); for
induces a phosphorylation cascade, leading to activation of review see Montgomery and Bowers (2011). Generally, TNF-α
the transcription factor NF-κB, which is essentially involved is believed to have a direct cytotoxic effect on oligodendrocytes
in the regulation of expression of proinflammatory cytokines, but is also important for remyelination (Arnett et al. 2001)
chemokines, adhesion molecules, and inducible effector mol- and is a potent inducer of cytokines, chemokines, and adhe-
ecules including iNOS (inducible nitric oxide synthase) and sion molecules in microglia and astrocytes. At peripheral nerve
cyclooxygenase-2 (COX-2). TNFRII directly associates with injury TNF-α is produced by Schwann cells and may act as a
TRAF2 in conjunction with TRAF1 (see Fig. 22.4) and elic- factor involved in neuropathic pain (Campana et al. 2007).
its intracellular cross-talk with TNFRI via RIP. Furthermore,
TNFRII activates the MEKK-JNK signaling pathway.
Clearly, TNFRI and TNFRII have opposing roles in cyto- 5 I N T E R L E U K I N -1 R E C E P TO R S
toxicity and cytoprotection (Fontaine et al. 2002). TNFRI is AND SIGNALING
responsible for the majority of known biological properties of
TNF-α and involves inflammation, immune responses, and Interleukin-1 (IL-1) is a pleiotropic cytokine and is essentially
cytotoxicity, which are primarily induced through induction involved in host-defense responses to injury and infection,
of adhesion molecules, cytokines, and chemokines. TFNRII including fever, sickness behavior, and metabolic and immune
is considered protective against neurodegenerative diseases by changes, and is associated with many CNS disorders, includ-
the activation of PI3K (phosphoinositol 3 kinase), PKB/Akt ing brain trauma, stroke, and epilepsy, as well as many forms
(protein kinase B/Akt) phosphorylation, and long-term acti- of chronic neurodegenerative disorders such as Alzheimer
vation of NF-κB (Marchetti et al. 2004). As mentioned, in and Parkinson disease or multiple sclerosis (Allan et al. 2005).
various types of neurodegenerative diseases (Haase et al. 2008), IL-1 induces the expression of a large variety of proinflam-
TNF-α is expressed primarily by microglia (Hanisch 2002) matory mediators, including cytokines, cytokine receptors,
and astrocytes. Glia-derived TNF-α is involved in a variety of acute-phase reactants, growth factors, tissue remodeling

C Y TO K I N E , C H E M O K I N E , A N D G R OW T H FAC TO R R E C E P TO R S A N D S I G N A L I N G • 271
enzymes, extracellular matrix components, and adhesion mol- mediates processing of IL-1β (Trendelenburg 2008 and refer-
ecules (Pinteaux et al. 2009). ences therein). Inflammasomes are powerful sensors of patho-
The primary receptor for IL1, IL-1R1 belongs to a large logical factors such as protein aggregates including amyloid-β.
Toll/IL-1R (TIR) superfamily and is defined by the Toll/ Indeed, recent work showed in vitro evidence for activation of
IL-1 receptor (TIR) domain, which occurs in the cytosolic the NALP3 inflammasome pathway in microglia in response
region of these receptors (Bowie and O’Neill 2000). IL-1R1 to amyloid-β (A-β) (Halle et al. 2008). It has thus been sug-
has shown to be expressed in astrocytes, oligodendrocytes, gested that inappropriate oligomerization of A-β protein is
microglia, and Schwann cells (Skundric et al. 1997; Wang et sensed by the glial inflammasome complex and thus gives rise
al. 2006). The activity of IL-1 is mediated by the two agonists to neuroinflammation (Masters and O’Neill 2011).
IL-1α and IL-1β that induce activation of IL-1R1. IL-IRI con- On IL-IRI signaling IL-1R accessory protein (AcP), a struc-
tains an extracellular ligand-binding domain that comprises tural homolog of IL-IRI, forms a heterodimer with IL-IRI, that
three immunoglobulin-like domains (Ig domains). IL-IRI is essential for IL-IRI–induced signal transduction. Owing to
also contains an intracellular signal-transducing domain that IL-1 binding, the IL-lRI:IL-lRAcP heterodimer assembles
bears homology to the Toll-like receptor family members and serves as a docking site for a variety of signaling proteins.
(Bowie and O’Neill 2000). The effects of IL-1R1 are coun- These signaling proteins are primarily kinases, including IL-1
teracted by the endogenously expressed receptor antago- receptor-associated kinases 1 and 2 (IRAK-1, IRAK-2), which
nist (IL-1RA), and by a nonsignaling type 2 IL-1 receptor bind to IL-lRAcP and IL-IRI, respectively (Fig. 22.5).
(IL-1R2), a soluble-binding protein that acts as a suppres- In IL-1 signaling the transcription factor NF-kB is a cen-
sor of IL-1 signaling. Furthermore, it has become clear that tral element that expression regulation of most IL-1–induced
IL-1β is involved in the inflammasome signaling cascade. In genes. IL-1–mediated activation of NF-κB yields a hetero-
the last few years, inflammasomes (protein signaling com- meric complex that leads to phosphorylation and subsequent
plexes involved in cytokine release) have emerged as a crucial degradation of the inhibitor I-κB, thus allowing nuclear trans-
intracellular pathway mediating inflammation (Strowig et al. location of NF-κB where it affects gene transcription. In addi-
2012). Activated inflammasomes give rise to cytokine release tion, MyD88, a member of the IL-1 receptor family, associates
and inflammation. It is thus well established that caspase-1 with both IL-lRAcP and IRAK-2. Subsequently, tumor necro-
is activated by the inflammasome complex and subsequently sis factor receptor–associated factor 6 (TRAF6) binds to the

Figure 22.5 Interleukin-1 Receptor Signaling. Following binding of IL-lα or IL-1β to IL-1 receptor-I (IL-IRI), IL-lRI dimerizes with IL-lRAcP.
At this scaffold the IL-1 receptor-associated kinases IRAK-1 and IRAK-2 are recruited to IL-lRAcP and IL-IRI, respectively. Additionally, MyD88
associates with IL-lRAcP and IRAK-2. This complex recruits tumor necrosis factor receptor–associated factor-6 (TRAF6), which binds and activates
NF-κB inducing kinase (NIK). NIK activates the I-κB kinase complex, which phosphorylates I-κB. IκB is then targeted for proteosomal degradation
and the now active NF-κB enters the nucleus and affects transcription of IL-1 target genes.

272 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
IL1-RI complex and recruits NF-κB-inducing kinase (NIK) STAT5a/b molecules are activated and promote transcrip-
that activates the I-κB kinase complex. This causes phosphory- tional activation of D (2 and 3) cyclins. In addition, on IL-R2
lation of I-κB, which ultimately leads to its proteosomal deg- activation of PI3K signaling, p27, an inhibitor of cyclin-D/
radation. After disposal of I-κB, NF-κB enters the nucleus and CDK activity, is downregulated (Nourse et al. 1994). These
regulates gene transcription, as mentioned. events stimulate progression through G1 of the cell cycle and
Also as mentioned, IL-1 is essentially involved in CNS thus trigger DNA synthesis and replication. Activated micro-
disorders, including brain trauma, stroke and epilepsy, and glia express IL-2R as well as IL-2 and increase their prolifera-
chronic neurodegenerative disorders, including Alzheimer tion rate upon stimulation with IL-2 and support production
and Parkinson disease and multiple sclerosis (Allan et al. of NO (Girard et al. 2008; Hanisch 2002; Sawada et al. 1995).
2005). The mechanism underlying this effect is the activa- Also oligodendrocytes express IL-2R and respond to exposure
tion of astrocytes that produce neurotoxic, neuroprotective to IL-2 by increased proliferation (Otero & Merril, 1997).
and inflammatory mediators. It has been suggested that IL-1– Furthermore, several lines of evidence suggest that IL-2 is an
induced neurotoxicity is dependent on induced production of important neuroregulatory cytokine independent of its role in
astrocytic neurotoxic factors. IL-1–induced neurotoxicity was inflammation and immunity (Hanisch and Quirion, 1995).
found to be dependent on free radical- and caspase activity and
production of TNF-α (Pinteaux et al. 2009). Most likely, the
toxic effect of IL-1 on oligodendrocytes also occurs through 8 I N T E R L E U K I N -4
actions on astrocytes. Furthermore, it has been suggested that
the neuroprotective effect of IL-1 is mediated by NGF release The IL-4 receptor (IL-4R) is expressed by astrocytes, microg-
from astrocytes (Pinteaux et al. 2009). Thus, it is likely that lia, and oligodendrocytes (Brodie et al. 1998; Cannella and
low levels of IL-1 act on astrocytes to produce neurotoxic fac- Raine 2004; Hulshof et al. 2002; Wei and Jonakait 1999),
tors, which can be counteracted by neuroprotective agents and is commonly considered an antiinflammatory mediator in
secreted by astrocytes at higher IL-1 levels. Furthermore, IL-1 these cells. IL-4 has an immunosuppressive effect on microg-
appears to control neuronal cell death directly in response to lia by suppressing expression of factors of the immune system,
injury, which may be mediated by enhancement of NMDA- including MHC-II, CD40, and B7 costimulatory molecules
induced toxicity (Viviani et al. 2003). (Nguyen and Benveniste 2000; O’Keefe et al. 1999; Wei and
Jonakait 1999). Furthermore, IL-4 inhibits NO production,
secretion of TNF-α and expression of ICAM-1 and addi-
6 T H E I N T E R L E U K I N -2 tionally induced secretion of NGF by astrocytes (Brodie et
R E C E P TO R FA M I LY al. 1998). As mentioned, IL-4R consists of a high-affinity
α-chain and a γ-chain. In addition, another form of IL-4R
Interleukin-2 receptor (IL-2R) together with receptors for exists and contains an IL-4Rα chain and an IL-13–binding
other cytokines including IL-4, IL-7, IL-9, IL-13, IL-15, and protein denoted IL-13Ra (Liu et al. 2000).
IL-21 makes up the IL-2 receptor family (O’Shea et al. 2002). IL-4R–mediated activation of JAKI and JAK3 has been
These receptors contain a ligand-specific binding chain (the shown to occur in many cell types. Activation of JAKs results
α-chain) and a general γ-chain. Additionally, IL-2 and IL-15 in recruitment and activation of STAT6 and STAT5. After
contain a β-chain, whereas the IL-13 receptor contains the recruitment and phosphorylation, the STAT molecules
IL-4 receptor α-chain and a ligand binding chain specific dimerize, translocate to the nucleus and activate transcription
for IL-13. The receptors for IL-2, IL-7, IL-9, IL-15, and IL-21 of IL-4 inducible genes.
transduce signals through activation of STAT-5, whereas the In addition to STAT signaling, IRS-2 plays an impor-
IL-4 and IL-13 receptors activate STAT-6. tant role in IL-4–induced cellular proliferation. This protein
The IL-2 and IL-4 signal transduction cascades are discussed named insulin receptor substrate 2 (IRS-2) has been shown
as examples for signaling of the IL-2 receptor superfamily. to become activated in response to IL-4 and IL-13. Insulin
receptor substrate molecules play a role in signal transmis-
sion to downstream pathways, including PI3K/Akt- and ERK
7 I N T E R L E U K I N -2
MAP kinase cascades. Finally, activation of IL-4R induces an
As mentioned, IL-2R is a heterotrimeric protein complex that increase in PI3-kinase activity. Cell type–specific modulation
is activated by IL-2. Three receptor chains (α, β, and γ) associ- of the PI3-kinase pathway by IL-4 may differentially influence
ate to form IL-2R (Wang et al. 2005). Both α- and β-chains cell proliferation or gene induction, respectively.
are involved in binding IL-2, whereas the γ-chain together
with the β subunit is involved in signal transduction. Whereas
the β-chain is associated the tyrosine kinase Janus Kinase 1 9 CHEMOKINES AND
( JAK1), the γ-chain associates with JAK3. Consequently, C H E M O K I N E R E C E P TO R S
IL-2 activates three signaling cascades, the MAP kinase cas-
cade, the phosphoinositide kinase 3 (PI3K) cascade, and the Chemokines (chemotactic cytokines) constitute the larg-
JAK-STAT cascade (Ellery and Nicholls 2002; Moon et al. est cytokine family in humans and consist of structur-
2004). After IL-R2–induced activation of JAK1/3 kinases ally related molecules that are important regulators of the

C Y TO K I N E , C H E M O K I N E , A N D G R OW T H FAC TO R R E C E P TO R S A N D S I G N A L I N G • 273
Figure 22.6 The Complex Pharmacology of the Chemokine System. There are approximately 50 different chemokines that are classified into four
subfamilies because of conserved cysteine residues (CC, CXC, CX3C, and C-chemokines). The classification of chemokine receptors is related to
their ligands; thus, chemokine receptors are also classified into four subfamilies: CCRs (CCR1–10), CXCRs (CXCR1–6), XCR1, and CX3CR1. The
pharmacology of chemokines shows a considerable amount of promiscuity; thus, many chemokines activate more than one receptor, and numerous
receptors are activated by more than one chemokine. Only seven nonredundant chemokine–chemokine receptor pairs are known. Moreover, the
chemokine system contains four atypical receptors: DARC, D6, CXCR7, and CCX-CKR that lack classical signaling properties. These atypical recep-
tors may bind a large variety of chemokines (e.g., DARC and D6) and act as decoy and scavenger receptors.

peripheral immune response and therefore are involved classical signaling properties. These so-called atypical or silent
in numerous diseases. There are approximately 50 differ- receptors lack crucial structural domains that are required for
ent chemokines which are classified into four subfamilies G-protein activation, including the DRY motif in the second
due to conserved cysteine residues (CC, CXC, CX3C, and intracellular loop (Mantovani et al. 2010). Nevertheless, these
C-chemokines) (Fig. 22.6). Chemokines are small (8- to receptors bind a variety of chemokines and act as decoy and
14-kDa) proteins that share a highly conserved tertiary struc- scavenger receptors, thereby fulfilling multiple important
ture, the so-called “chemokine-scaffold,” despite relatively low roles in the regulation of chemokine function (Mantovani
sequence homology (Mantovani et al. 2010). Unlike other et al. 2010).
cytokines, chemokines activate specific G-protein–coupled Chemokine receptors are coupled to Gi/o-proteins, which
receptors with seven transmembrane spanning domains, of primarily inhibit adenylate cyclase activity; however, it is clear
which 18 have been described. The classification of chemo- that Gi/o-proteins additionally activate other intracellular tar-
kine receptors is related to their ligands; thus, chemokine gets, including phospholipases, GTPases such as Rho, Rac,
receptors are also classified into four subfamilies: CCRs and Cdc42, and signaling pathways of major kinases such as
(CCR1–10), CXCRs (CXCR1–6), XCR1, and CX3CR1 (see mitogen-activated protein kinase (MAPK) and phosphati-
Fig. 22.6). The pharmacology of chemokines displays a con- dyl inositol-3 kinase (PI3-K) (Fig. 22.7) (Neves et al. 2002;
siderable amount of promiscuity, thus many chemokines Rodríguez-Frade et al. 2001). This diversity in intracellu-
activate more than one receptor, and numerous receptors lar signaling shows that chemokine receptors, in addition to
are activated by more than one chemokine. Only few non- pathways involved in cell migration, also activate other path-
redundant chemokine–chemokine receptor pairs are known, ways and control a large spectrum of cellular functions.
of which the CX3CL1-CX3CR1 pair is the most established Similar to other organs, the infiltration of the inflamed brain
example for the brain (Mantovani et al. 2010). An interest- by blood leukocytes is regulated to a large extent by chemok-
ing property of the chemokine system is the existence of four ines (Prinz and Priller 2010). However, also most endogenous
receptors: DARC, D6, CXCR7, and CCX-CKR that lack brain cells (astrocytes, oligodendrocytes, NG2 cells, microglia,

274 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Figure 22.7 Chemokine Receptor Signaling. All chemokine receptors are coupled to Gi/o-proteins. On binding the GDP is exchanged for GTP and
the activated α-subunit dissociates to inhibit adenylate cyclase activity and cAMP formation. The βγ-subunit can further activate other intracellular
targets, including phospholipases, GTPases such as Rho, Rac, and Cdc42, and signaling pathways of major kinases such as mitogen-activated protein
kinase (MAPK) and phosphatidyl inositol-3 kinase (PI3-K). Moreover, there is evidence that dimerization of activated chemokine receptors may
change gene transcription via activation of the JAK/STAT pathway.

and neurons) express functional chemokine receptors and are it is a microglia-activating signal in the spinal cord (Old and
able to release chemokines (see chapter 18). This indicates Malcangio 2011). After spinal nerve injury, microglia release
that the functions of endogenous chemokines in the brain cathepsin S, which causes the shedding of neuronal CX3CL1
go beyond the control of infiltration (Biber et al. 2006). For and the subsequent further activation of microglia Src-kinase
example, chemokines regulate the migration, maturation, and pathway (Old and Malcangio 2011). Such an activating activ-
survival of oligodendrocyte precursor cells via the chemokine ity of CX3CL1 was never found in the central brain and
receptors CXCR2, CXCR4, and CXCR7 (Banisadr et al. may point toward a region-specific function of this neuron–
2011). Generally, the basal expression of chemokines in the microglia signaling system. Neuronal CCL21 is unique among
healthy brain is low, but increases considerably under patho- the neuronal chemokines, as it is expressed exclusively in dam-
logical conditions. The only exception is the membrane-bound aged neurons, where it is sorted into large dense core vesicles
chemokine CX3CL1 (previously known as fractalkine), that and transported to the axon ends (Biber et al. 2011; de Jong
is highly expressed in healthy neurons. Deficient CX3CL1- et al. 2008). Mice that lack functional CCL21 do not develop
CX3CR1 signaling has long been thought to have little impact neuropathic pain in response to peripheral nerve injury, high-
on healthy brain function; however, recent findings have lighting the functional importance of these chemokines in
shown small developmental and electrophysiological abnor- neuron–microglia communication (Biber et al. 2011). Next to
malities in the brain of CX3CR1-deficient animals that point the large number chemokine functions confined to astrocytes
toward a function of CX3CR1 in microglia in pruning of syn- (Biber et al. 2006), it was recently found that CXCR4-activation
apses during development (Paolicelli et al. 2011). Despite these by CXCL12 regulates astrocytic glutamate release and thus
recent results, CX3CL1 exerts its function mainly in pathol- influences synaptic transmission (Calì and Bezzi 2010).
ogy, where it regulates the response of microglia after neuronal
injury (Cardona et al. 2006). Next to CX3CL1, CCL21 is also
an important player in the communication between damaged 10 T R A N S F O R M IN G G R OW T H
neurons and microglia, which is crucial for the development of FAC TO R - β
neuropathic pain after peripheral nerve injury (Biber et al. 2011;
Old and Malcangio 2011). Interestingly, whereas CX3CL1 in The superfamily of transforming growth factors-β (TGF-β)
the brain is a neuronal signal that dampens microglia activity, has a broad spectrum of biological effects and comprises

C Y TO K I N E , C H E M O K I N E , A N D G R OW T H FAC TO R R E C E P TO R S A N D S I G N A L I N G • 275
Figure 22.8 The TGF-β Signaling Pathway. Members of the TGF-β superfamily bind to type I (TGF-β-RI) or type II (TGF-β-RII) receptors.
TGF-β-RI is phosphorylated by TGFβ-RII, resulting in an activated receptor complex. R-SMADS are recruited to the receptor complex and are phos-
phorylated at their C-terminus. Phosphorylated SMADs then form a trimeric complex with common SMAD4. The SMAD complex then translocates
to the nucleus, where it interacts with DNA and transcription factors, including a large variety of DNA-binding transcription factors (cofactors) in a
target gene-dependent manner. FKBP12, SARA, SMURF1/2, and SMADs 6/7 are negative regulators of TGF-β signaling.

at least 30 members that include various forms of TGF-β, sclerosis (reviewed in Pratt and McPherson 1997). In general,
bone morphogenetic protein, and the factors activin and an increase in TGF-β expression has been observed under
nodal. During embryogenesis, TGF-β family members play these conditions both in neurons and glial cells. Whether
a critical role in axis formation, left-right organization, and this elevated expression of TGF-β reflects a “wound healing”
tissue patterning and disruptions in TGF-β signaling in and antiinflammatory response or contributes to pathology
humans often leads to tumorigenesis (Massagué 2008). In is unclear. In animal models, it has been shown that TGF-β
the nervous system, all three TGF-β isoforms (β1–3) are contributes to the pathology caused by β-amyloid accumu-
ubiquitously expressed (Kitisin et al. 2007) and are impor- lation by increasing its production, but on the other hand
tant for neuronal and astrocytic differentiation (McKinnon TGF-β enhances microglial clearance of β-amyloid deposits.
et al. 1993; Stipursky and Gomes 2007; Yi et al. 2010). In the In autoimmunity studies, TGF-β has been reported to have
CNS, the effect of TGF-β is different for different cell types antiinflammatory and neuroprotective functions (Owens
and has been reported to act as a neurotrophic factor as well et al. 2001).
as an inducer of apoptosis. TGF-β inhibits proliferation of The TGF-β superfamily signals through TGF-β and
astrocytes in vitro (Vergeli et al. 1995) and induces apopto- BMP receptors that can be subdivided into type I (TGF-βI)
sis in oligodendrocyte precursors (Schulz et al. 2009), but and type II (TGF-βII) serine/threonine kinase receptors.
promotes neuronal survival. This is supported by knock-out (Fig. 22.8). After ligand binding, TGFβRII receptors phos-
studies in mice, in which a loss of TGF-β1 leads to micro- phorylate TGF-β-RI receptors, which in turn phosphorylate
gliosis and reduces neuronal survival (Brionne et al. 2003). and activate specific SMAD proteins. These SMADs form a
The neuroprotective effect of TGF-β on neurons occurs complex with common SMAD4, translocate to the nucleus,
most likely indirectly through induction of neurotrophic and activate target gene transcription. The composition of
factors in glial cells. A role for TGF-β has been suggested in these complexes is dependent on the type of TGF-β recep-
many CNS pathologies, including ischemia and excitotoxic- tor activated. In the CNS, TGF-β is constitutively expressed
ity as well as a range of neurodegenerative conditions such (isoforms 1–3) and expression of TGF-β-RI and TGF-β-RII
as Parkinson’s disease, Alzheimer’s disease, and multiple is detected on neuronal as well as glial cells.

276 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Figure 22.9 Neurotrophin Signaling. Signaling by members of the neurotrophins can be mediated by two receptor types, TrkA-D and p75. Each Trk
receptor controls three major signaling pathways. Activation of Ras results in activation of the MAP kinase–signaling cascade, activation of PLC-γ1
results in activation of Ca2+- and protein kinase C–regulated pathways and activation of the PI3-Akt pathway. In general, these signaling pathways acti-
vate the expression of genes involved in differentiation and/or survival. The p75 receptor activates three major signaling pathways. NF-κB activation
results in induction of prosurvival genes. Activation of the Jun kinase pathway results in activation of proapoptotic genes. And neurotrophin binding
to p75 regulates the activity of Rho, affecting growth cone motility.

The SMADs (an acronym for SMA [the small protein in (BDNF), neurotrophin-3 (NT-3), and NT-4. Neurotrophins
C. elegans] and MAD [mothers against decapentaplegic in D. are essential factors that play a critical role in survival, differ-
melanogaster]) mediate intracellular signaling by TGF-β fam- entiation, and myelination of neurons during neuronal devel-
ily members. The SMADs are subdivided in three functionally opment (Chao 2003). Recently, NTs have also been reported
distinct groups: (1) receptor-regulated SMADs (R-SMADs), to be important regulators of myelination, both in the periph-
(2) the co-SMAD, SMAD4, and (3) antagonistic SMADs. eral and CNS (Rosenberg et al. 2006; Xiao et al. 2009).
SMAD4 participates in signal transduction of all TGF-β Nerve growth factor and BDNF are ubiquitously expressed
family members, interacts with activated R-SMADs and, by all glial cell types. Neurotrophins can signal through two
after nuclear translocation, activates target gene transcription. classes of transmembrane receptors: receptor tyrosine kinases
R-SMAD–SMAD4 complexes have been reported to interact TrkA, -B, -C, and the P75 NT receptor (a member of the
with various cofactors, these include forkhead family members tumor necrosis factor [TNF] receptor superfamily), which
like P300, CBP, FOXH1, OAZ, RUNX family members, and all are expressed by glial cells (Fig. 22.9). Nerve growth fac-
AP1 transcription factors, and different combination bind to tor preferably binds TrkA, BDNF TrkB, NT-3 TrkC, and
different cognate binding sites (Massagué et al. 2005). NT-4 TrkB, and all NTs can bind p75. Neurotrophins bind
Interferon-γ and TNF-α regulate TGF-β signaling indi- their receptors with relatively low affinity, which is regulated
rectly: interferon-γ (via JAK/STAT1) and TNF-α (via NF-κB) by receptor dimerization resulting in the formation of a high
can induce the expression of antagonistic SMAD7 protein and affinity complex. Trk receptor dimerization leads to trans-
thereby inhibit TGF-β receptor signaling. However, TNF-α phosphorylation of tyrosine residues in the cytoplasmic por-
is not exclusively antagonistic to TGF-β, TNFα can also acti- tion of the receptor (see Fig. 22.9). These phosphorylated
vate STAT3, which has been reported to be able to coopera- tyrosine residues then function as docking sites for down-
tively with SMAD1 to induce astrocyte differentiation. stream signaling cascades. Trk receptors recruit and activate
The primary effect of TGF-β signaling on astrocytes and PLCγ and the adapter protein Shc, which in turn leads to
microglia is immune suppressive. The expression of MHC activation of phosphatidylinositol 3-kinase (PI3K) Akt and
class II, B7, CD40, ICAM-1, VCAM-1, and TNF-α is inhib- Ras/ERK1/2. Ultimately, Trk signaling mediates a plethora
ited by TGF-β (O’Keefe et al. 2002). of activities, which include neuronal survival, neurite out-
growth, and myelination and differentiation of Schwann cells.
Additionally, NTs have been reported to stimulate microglial
11 N E U R OT R O P H I N S proliferation in vitro (Cosgaya et al. 2002).
The role of p75 in NT signaling is complex and is dif-
The family of neurotrophins (NTs) consists of four members: ferent in the presence or absence of Trk receptors (Nykjaer
nerve growth factor (NGF), brain-derived neurotrophic factor et al. 2005). The affinity of p75 for NTs is similar to that of

C Y TO K I N E , C H E M O K I N E , A N D G R OW T H FAC TO R R E C E P TO R S A N D S I G N A L I N G • 277
monomeric Trk. The Trk-A receptor can dimerize with p75 Signaling through PDGFR has been implicated in
and this heterodimer has a higher affinity for NGF than both glioma-genesis and glial development. During embryonic
homodimeric receptors and allows for receptor activation at development (E15), PDGF is expressed by neurons, in which
lower NGF concentrations. Additionally, the specificity of Trk the PDGFR is expressed in glial precursors, mainly of the
receptors for NTs is affected by heterodimerization with p75. oligodendrocyte lineage. PDGF-A–deficient mice show
The intracellular domain of the p75 receptor activates various reduced oligodendrocyte numbers and display hypomyelina-
intracellular signaling pathways, different from the pathways tion, indicating a lack of trophic effect of neuronal PDGF on
activated by Trk receptors. This suggests that the signaling glial precursors. This is further supported by the observation
properties of Trk homodimers and Trk/p75 heterodimers that PDGF overexpression in neurons resulted in increased
are different with different biological activity as a result. For oligodendrocyte precursor number, but normal mature oli-
instance, TrkA dimers sustain neurite outgrowth, but TrkA/ godendrocytes, presumably owing to loss of neuronal contact.
p75 heterodimers are required for maturation and long-term Additional experiments targeting tyrosines in the cytoplas-
neuronal survival. mic domain of the PDGFR required for downstream signal-
The p75 receptor also mediates Trk-independent NT signal- ing molecules has shown that these pathways are required for
ing. Genetic studies in mice have convincingly shown that p75 proper expansion of oligodendrocyte precursors and chemo-
can promote apoptosis, most likely through the activation of taxis to migrate and populate other areas of the brain.
Jun N-terminal kinase ( JNK). The intracellular domain of p75 Aberrant PDGF signaling has also been implicated in glial
has been shown to couple to downstream signaling pathways via tumorigenesis. PDGF and PDGFR have been found to be
several adaptor proteins, including NT-receptor–interacting overexpressed in glial tumors as well as glial tumor cell lines.
factor (NRIF), NT-associated cell death executor (NADE), Enhanced expression of PDGF or PDGFR was associated
NT-receptor–interacting MAGE homolog (NRAGE) and TNF with a higher tumor grade and hence a poorer prognosis. The
receptor–associated factors (TRAFs). All these interacting pro- expression of both the ligand and its receptor by these cells
teins (alone or in complexes) have been shown to promote p75- allows for autocrine stimulation. However, a causal relation
dependent apoptosis. Additionally, p75 has been shown to be between PDGF signaling and glial tumorigenesis is unclear as
involved in Schwann cell migration (involving factor Schwann well as the mechanism of PDGF/PDGFR overexpression
cell 1 [SC1]) or survival (involving receptor-interacting protein
2 [RIP2]) by activating NF-κB signaling, resulting in reduced
NGF-p75–induced Schwann cell death. With respect to astro- 13 S U M M A RY A N D P E R S P E C T I VE S
cytes, NGF inhibits their proliferation through the p75 recep-
tor (Cragnolini et al. 2009). Oligodendrocytes express NTs as In this chapter an outline of the common signaling pathways
well as all receptors: TrkA-C and p75, suggesting autocrine sig- and functions of cytokine-, chemokine-, and growth factor
naling by NTs (Du and Dreyfus 2002). Oligodendrocytes that receptors in glia cells has been provided. This signaling gen-
myelinate sensory neurons express BDNF, and a potential role erally involves activation of membrane-associated receptors
BDNF in the myelination process is further corroborated by induced by agonist binding, followed by biochemical signal-
the observation that BDNF regulates the number of oligoden- ing cascades that result in functional responses and induced
drocyte precursors and their differentiation in demyelinating gene transcription. Because these receptor families are very
lesions (VonDran et al. 2011). extensive, this outline is far from complete, but clearly delin-
eates the known response patterns of glia and their relation-
ship to glia functions.
12 P L AT E L ET-D E R I VE D G R OW T H Clearly, the wide variety of specific receptor subtypes
FAC TO R S I G N A L I N G expressed in glia allows detailed responses to neuroimmune sig-
nals that convey stress, inflammation, or regenerative processes.
Platelet-derived growth factor (PDGF) is one of the first iden- The integration of the diverse immune signals culminates in
tified mitogens, and stimulates proliferation of cells of mes- dedicated activated glia phenotypes and unified responses to
enchymal origin, mainly smooth muscle cells and glial cells. situations of maintenance/regeneration, neural damage, and
PDGF signaling is mediated by four ligands (PGDF-A, B, C, aging. Particularly the effects of cytokines and chemokines
and D) and two receptors (PDGF-Rα and -β). Expression of range from neuroprotection to severe neurodamage and are
PDGFRs seems to be restricted to cells derived from O2A tightly regulated with respect to strength and duration.
precursors, mainly type 2 astrocytes and oligodendrocytes A better understanding of the synchronized glial response
(Hart et al. 1989). All PDGF ligands activate the PDGFRs as to these complex signal patterns may reveal basal mechanisms
disulfide-linked homodimers or as PDGF-A-B heterodimers. underlying neuropathology and neurorepair and maintenance,
PDGFR is a transmembrane receptor tyrosine kinase, and on and may provide possible targets for therapeutic intervention.
ligand binding, two receptors dimerize, cross-phosphorylate
tyrosine residues in their cytoplasmic tails, which then serve
REFERENCES
as docking sites for SH2-containing adaptors, Shc and Grb2.
Signaling pathways acting downstream of the PDGFR include Aggarwal et al. 2011. Historical perspectives on tumor necrosis factor
Ras-MAPK, PI3K, Src, STATs, and PLCγ (Heldin and and its superfamily: twenty-five years later, a golden journey. Blood
Westermark 1999). 4:325225.

278 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Allan SM, Tyrrell PJ, Rothwell NJ. 2005. Interleukin-1 and neuronal Girard S, Larouche A, Kadhim H, Rola-Pleszczynski M, Gobeil F,
injury. Nat Rev Immunol 5(8):629–640. Sebire G. 2008. Lipopolysaccharide and hypoxia/ischemia induced
Arnett HA, Mason J, Marino M, Suzuki K, Matsushima GK, Ting JP. IL-2 expression by microglia in neonatal brain. Neuroreport
2001. TNF alpha promotes proliferation of oligodendrocyte progeni- 19:997–1002.
tors and remyelination. Nat Neurosci 11:1116–1122. Gough DJ, Levy DE, Johnstone RW, Clarke CJ. 2008. IFNgamma
Baker BJ, Park KW, Qin H, Ma X, Benveniste EN. 2010. IL-27 inhibits signaling. Does it mean JAK-STAT? Cytokine Growth Factor Rev
OSM-mediated TNF-alpha and iNOS gene expression in microglia. 19(5–6):383–394.
Glia 58(9):1082–1093. Haase G, Pettmann B, Raoul C, Henderson CE. 2008. Signaling by death
Banisadr G, Frederick TJ, Freitag C, Ren D, Jung H, Miller SD, receptors in the nervous system. Curr Opin Neurobiol 18:284–291.
et al. 2011. The role of CXCR4 signaling in the migration of trans- Halle, A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel
planted oligodendrocyte progenitors into the cerebral white matter. T, et al. 2008. The NALP3 inflammasome is involved in the innate
Neurobiol Dis 44(1):19–27. immune response to amyloid-β. Nat Immunol 9:857–865.
Bauer S, Kerr BJ, Patterson PH. 2007. The neuropoietic cytokine fam- Hanisch UK. 2002. Microglia as a source and target of cytokines. Glia
ily in development, plasticity, disease and injury. Nat Rev Neurosci 40:140–155.
8(3):221–232. Hanisch UK, Quirion R. 1995. Interleukin-2 as a neuroregulatory cyto-
Biber K, de Jong EK, van Weering HR, Boddeke HW. 2006. Chemokines kine. Brain Es Rev 21:246–284.
and their receptors in central nervous system disease. Curr Drug Hart IK, Richardson WD, Heldin CH, Westermark B, Raff MC. 1989.
Targets 7(1):29–46. PDGF receptors on cells of the oligodendrocyte-type-2 astrocyte
Biber K, Tsuda M, Tozaki-Saitoh H, Tsukamoto K, Toyomitsu E, (O-2A) cell lineage. Development 105(3):595–603.
Masuda T, et al. 2011. Neuronal CCL21 up-regulates microglia Heinrich PC, Behrmann I, Haan S, Hermanns HM, Müller-Newen G,
P2X4 expression and initiates neuropathic pain development. EMBO Schaper F. 2003. Principles of interleukin (IL)-6-type cytokine sig-
30(9):1864–1873. nalling and its regulation. Biochem J 374(Pt 1):1–20.
Bowie A, O’Neill LAJ. 2000. The interleukin-1 receptor/Toll-like recep- Heldin CH, Westermark B. 1999. Mechanism of action and in vivo role
tor superfamily: signal generators for pro-inflammatory interleukins of platelet-derived growth factor. Physiol Rev 79(4):1283–1316.
and microbial products. J Leukocyte Biol 67:508–514. Hulshof S, Montagne L, De Groot CJ, Van Der Valk P. 2002. Cellular
Brionne TC, Tesseur I, Masliah E, Wyss-Coray T. 2003. Loss of localizaton and expression of interleukin-10, interleukin-4, and their
TGF-beta 1 leads to increased neuronal cell death and microgliosis in receptors in multiple sclerosis lesions. Glia 38:24–35.
mouse brain. Neuron 40(6):1133–1145. Kamimura D, Ishihara K, Hirano T. 2003. IL-6 signal transduction and
Brodie C, Goldreich N, Haiman T, Kazimirsky G. 1998. Functional its physiological roles: the signal orchestration model. Rev Physiol
IL-4 receptors on mouse astrocytes: IL-4 inhibits astrocyte activation Biochem Pharmacol 149:1–38.
and induces NGF secretion. J Neuroimmunol 81:20–30. Kitisin K, Saha T, Blake T, Golestaneh N, Deng M, Kim C, et al. 2007.
Calì C, Bezzi P. 2010. CXCR4-mediated glutamate exocytosis from Tgf-Beta signaling in development. Science STKE 399:cm1.
astrocytes. J Neuroimmunol 224(1–2):13–21. Krady JK, Lin HW, Liberto CM, Basu A, Kremlev SG, Levison SW.
Campana WM. 2007. Schwann cells: activated peripheral glia and their 2008. Ciliary neurotrophic factor and interleukin-6 differentially
role in neuropathic pain. Brain Behav Immun 21:522–527. activate microglia. J Neurosci Res 86(7):1538–1547.
Cannella B, Raine CS. 2004. Multiple sclerosis: cytokine receptors on oli- Lee N, Robitz R, Zurbrugg RJ, Karpman AM, Mahler AM, Cronier SA,
godendrocytes predict innate regulation. Ann Neurol 55(1):46–57. et al. 2008. Conditional, genetic disruption of ciliary neurotrophic
Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra factor receptors reveals a role in adult motor neuron survival. Eur J
IM, et al. 2006. Control of microglial neurotoxicity by the fractalkine Neurosci 27(11):2830–2837.
receptor. Nat Neurosci 9(7):917–924. Liu H, Jacobs BS, Liu J, Prayson RA, Estes ML, Barnett GH, et al. 2000.
Chao M. 2003. Neurotrophins and their receptors: a convergence point Interleukin-13 sensitivity and receptor phenotypes of human glial
for many signalling pathways. Nature Rev Neurosci 4:299–309. cell lines: non-neoplastic glia and low-grade astrocytoma differ from
Cosgaya JM, Chan JR, Shooter EM. 2002. The neurotrophin recep- malignant glioma. Cancer Immunol Immunother 49(6):319–324.
tor p75NTR as a positive modulator of myelination. Science Mantovani A, Savino B, Locati M, Zammataro L, Allavena P, Bonecchi
298(5596):1245–1248. R. 2010. The chemokine system in cancer biology and therapy.
Cragnolini AB, Huang Y, Gokina P, Friedman WJ. 2009. Nerve growth Cytokine Growth Factor Rev 21(1):27–39.
factor attenuates proliferation of astrocytes via the p75 neurotrophin Marchetti L, Klein M, Schlett K, Pfizenmaier K, Eisel UL. 2004.
receptor. Glia 57(13):1386–1392. Tumor necrosis factor (TNF)-mediated neuroprotection against
Dafny N, Yang PB. 2005. Interferon and the central nervous system. Eur glutamate-induced excitotoxicity is enhanced by N-methyl-D-aspartate
J Pharmacol 31;523(1–3):1–15. receptor activation. Essential role of a TNF receptor 2-mediated
De Jong EK, Vinet J, Stanulovic VS, Meijer M, Wesseling E, Sjollema phosphatidyl inositol 3-kinase-dependent NF-kappa B pathway. J
K, et al. 2008. Expression, transport, and axonal sorting of neuronal Biol Chem 279:32869–32881.
CCL21 in large dense-core vesicles. FASEB J 22(12):4136–4145. Marriott MP, Emery B, Cate HS, Binder MD, Kemper D, Wu Q,
Dopp JM, Mackenzie-Graham A, Otero GC, Merril JE. 1997. Differential et al. 2008. Leukemia inhibitory factor signaling modulates both
expression, cytokine modulation, and specific functions of type-1 and central nervous system demyelination and myelin repair. Glia
type-2 tumor necrosis factor receptors in rat glia. J Neuroimmunol 56(6):686–698.
75:104–112. Massagué J 2008. TGFβ in cancer. Cell 134(2):215–230.
Du Y, Dreyfus CF. 2002. Oligodendrocytes as providers of growth fac- Massagué J, Seoane J, Wotton D. 2005. Smad transcription factors.
tors. J Neurosci Res 68(6):647–654. Genes Dev 19(23):2783–2810.
Ellery JM, Nicholls PJ. 2002. Alternate signalling pathways from the Masters SL, O’Neill LAJ. 2011 Disease-associated amyloid and mis-
interleukin-2 receptor. Cytokine Growth Factor Rev 1:27–40. folded protein aggregates activate the inflammasome. Trends Mol
Ellis SL, Gysbers V, Manders PM, Li W, Hofer MJ, Müller M, et al. Med 17:276–282.
2010. The cell-specific induction of CXC chemokine ligand 9 medi- McKinnon RD, Piras G, Ida JA Jr, Dubois-Dalcq M. 1993. A role
ated by IFN-gamma in microglia of the central nervous system is for TGF-beta in oligodendrocyte differentiation. J Cell Biol
determined by the myeloid transcription factor PU1. J Immunol 121(6):1397–1407.
1;185(3):1864–1877. Montgomery SL, Bowers WJ. 2011. Tumor Necrosis factor-alpha and the
Fontaine V, Mohand-Said S, Hanoteau N, Fuchs C, Pfizenmaier K, Eisel roles it plays in homeostatic and degenerative processes within the cen-
U. 2002. Neurodegenerative and neuroprotective effects of tumor tral nervous system. J Neuroimmune Pharmacol 34(8):1256–1267.
necrosis factor (TNF) in retinal ischemia: opposite roles of TNF Moon JJ, Rubio ED, Martino A, Krumm A, Nelson BH. 2004. A per-
receptor 1 and TNF receptor 2. J Neurosci 22:RC126. missive role for phosphatidylinositol 3-kinase in the Stat5-mediated

C Y TO K I N E , C H E M O K I N E , A N D G R OW T H FAC TO R R E C E P TO R S A N D S I G N A L I N G • 279
expression of cyclin D2 by the interleukin-2 receptor. J Biol Chem Rose-John S, Waetzig GH, Scheller J, Grötzinger J, Seegert D. 2007. The
279(7):5520–5527. IL-6/sIL-6R complex as a novel target for therapeutic approaches.
Neves SR, Ram PT, Iyengar R. 2002. G protein pathways. Science Expert Opin Ther Targets 11(5):613–624.
296(5573):1636–1639. Rosenberg SS, Ng BK, Chan JR. 2006. The quest for remyelination:
Nguyen V, Benveniste EN. 2000. Interleukin-4 activated STAT-6 inhib- a new role for neurotrophins and their receptors. Brain Pathol
its IFN-γ induced CD40 gene expression in macro phages/microglia. 16(4):288–294.
J Immunol 165:6235–6243. Sawada M, Suzumura A, Marunouchi T. 1995. Induction of functional
Nykjaer A, Willnow TE, Petersen CM. 2005. p75NTR—live or let die. interleukin-2 receptor in mouse microglia. J Neurochem 64:1973–1979.
Curr Opin Neurobiol 15(1):49–57. Schulz R, Vogel T, Mashima T, Tsuruo T, Krieglstein K. 2009.
Nourse J, Firpo E, Flanagan WM, Coats S, Polyak K, Lee MH, Involvement of Fractin in TGF-beta-induced apoptosis in oligoden-
et al. 1994. Interleukin-2-mediated elimination of the p27Kip1 droglial progenitor cells. Glia 57(15):1619–1629.
cyclin-dependent kinase inhibitor prevented by rapamycin. Nature Skundric DS, Bealmear B, Lisak RP. 1997. Induced upregulation of
372(6506):570–573. IL-1, IL-1RA and IL-1R type I gene expression by Schwann cells.
O’Keefe GM, Nguyen VT, Benveniste EN. 1999. Class II transactiva- J Neuroimmunol 174:9–18.
tor and class II MHC gene expression in microglia: modulation Stipursky J, Gomes FC. 2007. TGF-beta1/SMAD signaling induces
by the cytokines TGF- β, IL-4, IL-13, and IL-10. Eur J Immunol astrocyte fate commitment in vitro: implications for radial glia devel-
29:1275–1285. opment. Glia 55(10):1023–1033.
O’Keefe GM, Ngyuen VT, Benveniste EN. 2002. Regulation and func- Strowig T, Henao-Mejia J, Elinrav E, Flavell R. 2012. Inflammasomes in
tion of class II major histocompatibility complex, CD40, and B7 health and disease. Nature 481:278–286.
expression in macrophages and microglia: Implications in neurologi- Takaoka A, Yanai H. 2006. Interferon signalling network in innate
cal diseases. J Neurovirol 8(6):496–512. defence. Cell Microbiol 8(6):907–922.
Old EA, Malcangio M. 2011. Chemokine mediated neuron-glia com- Tanaka M, Miyajima A. 2003. Oncostatin M, a multifunctional cyto-
munication and aberrant signalling in neuropathic pain states. Curr kine. Rev Physiol Biochem Pharmacol 149:39–52.
Opin Pharmacol 1:67–73. Trendelenburg G. 2008. Acute neurodegeneration and the inflam-
O’Shea JJ, Gadina M, Schreiber RD. 2002. Cytokine signal- masome: central processor for danger signals and the inflammatory
ing in 2002 New surprises in the Jak/STAT pathway. Cell response? J Cereb Blood Flow Metab 28:867–881.
109(Suppl):S121–S131. Tsuda M, Masuda T, Kitano J, Shimoyama H, Tozaki-Saitoh H, Inoue
Otero GC, Merril JE. 1997. Response of human oligodendrocytes to K. 2009. IFN-gamma receptor signaling mediates spinal micro-
interleukin-2. Brain Behav Immun 11:24–38. glia activation driving neuropathic pain. Proc Natl Acad Sci U S A
Owens T, Wekerle H, Antel J. 2001. Genetic models for CNS inflamma- 106(19):8032–8037.
tion. Nat Med 7(2):161–166. Vergeli M, Mazzanti B, Ballerini C, Gran B, Amaducci L, Massacesi L.
Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli Pet 1995. Transforming growth factor-beta 1 inhibits the proliferation
al. 2011. Synaptic pruning by microglia is necessary for normal brain of rat astrocytes induced by serum and growth factors. J Neurosci Res
development. Science 333(6048):1456–1458. 40(1):127–133.
Paul S, Ricour C, Sommereyns C, Sorgeloos F, Michiels T 2007. Type Viviani B, Bartesaghi S, Cardoni F, Vezzani A, Behrens MM, Bartfai T,
I interferon response in the central nervous system. Biochimie et al. 2003. Interleukin-1 beta enhances NMDA receptor-mediated
89(6–7):770–778. intracellular calcium increase through activation of the Src family of
Pinteaux E, Trotter P, Simi A. 2009. Cell-specific and concentration- kinases. J Neurosci 23:8692–8700.
dependent actions of interleukin-1 in acute brain inflammation. VonDran MW, Singh H, Honeywell JZ, Dreyfus CF. 2011. Levels of
Cytokine 45:1–7. BDNF impact oligodendrocyte lineage cells following a cuprizone
Pratt BM, McPherson JM. 1997. TGF-beta in the central nervous sys- lesion. J Neurosci 31(40):14182–14190.
tem: potential roles in ischemic injury and neurodegenerative dis- Wang X, Rickert M, Garcia KC. 2005. Structure of the quaternary
eases. Cytokine Growth Factor Rev 8(4):267–292. complex of interleukin-2 with its alpha, beta, and gammac receptors.
Prinz M, Kalinke U. 2010. New lessons about old molecules: how type Science 310(5751):1159–1163.
I interferons shape Th1/Th17-mediated autoimmunity in the CNS. Wang X, Yin L, Hu J, Huang L, Yu P, Jiang X, et al. 2006. Expression
Trends Mol Med 16(8):379–386. and localization of p80 interleukin-1 receptor protein in the rat spi-
Prinz M, Priller J. 2010. Tickets to the brain: role of CCR2 and nal cord. J Mol Neurosci 29:45–53.
CX3CR1 in myeloid cell entry in the CNS. J Neuroimmunol Wei R, Jonakait GM. 1999. Neurotrophins and the anti-inflammatory
224(1–2):80–84. agents interleukin-4 (IL-4), IL-10, IL-11 and transforming growth
Qin Y, Cheng C, Wang H, Shao X, Gao Y, Shen A. 2008. TNF-alpha as factor-β 1 (TGF- β1) downregulate T cell costimulatory molecules
an autocrine mediator and its role in the activation of Schwann cells. B7 and CD40 on cultured rat microglia. J Neuroimmunol 95:8–18.
Neurochem Res 33:1077–1084. Xiao J, Kilpatrick TJ, Murray SS. 2009. The role of neurotrophins in the
Rodríguez-Frade JM, Mellado M, Martínez-AC. 2001. Chemokine regulation of myelin development. Neurosignals 17(4):265–276.
receptor dimerization: two are better than one. Trends Immunol Yi JJ, Barnes AP, Hand R, Polleux F, Ehlers MD. 2010. TGF-beta signal-
22(11):612–617. ing specifies axons during brain development Cell 142(1):144–157.

280 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
23.
LIPIDS, LIPID MEDIATOR S, AND OTHER
SIGNALING MOLECULES
Hideki Hayashi

A B B R E VI AT I O N S 1 INTRODUCTION

AA arachidonic acid Lipids in the central nervous system (CNS) play various roles as
ABC ATP-binding cassette components of membrane structures and biological regulators
AD Alzheimer disease of cell functions. Their metabolites also act as bioactive lipid
apo apolipoprotein mediators in brain functions. The brain contains the second
ATX autotaxin highest lipid content next to adipose tissue (Leskovjan et al.
Aβ amyloid β peptide 2010) and is the most cholesterol-rich organ in the body. In con-
BBB blood-brain barrier trast with the concentration of cholesterol in most other organs
CETP cholesteryl ester transfer protein of the adult mouse (in the range of 2–6 mg/g wet weight), the
CNS central nervous system brain contains at about 15 mg/g tissue (Dietschy 2009). Because
COX cyclooxygenase the CNS is segregated from the peripheral circulation by the
CSF cerebrospinal fluid blood-brain barrier (BBB), lipid homeostasis in the CNS is dis-
DHA docosahexaenoic acid tinctively different from that in peripheral tissues.
FFA free fatty acid A lipoprotein is a soluble complex of proteins and lipids
FLAP 5-lipoxygenase–activating protein that carries lipids in the circulation and the cerebrospinal fluid
GPCR G protein–coupled receptor (CSF). In contrast with the circulation, which contains very low
HDL high density lipoproteins density lipoproteins (VLDL), low density lipoproteins (LDL),
HIV human immunodeficiency virus and high density lipoproteins (HDL), the CNS contains only
HPGDS hematopoietic prostaglandin HDL-like particles of size approximately 15 nm and density
D synthase 1.06 to 1.12 g/mL (LaDu et al. 1998). Under normal conditions
IL interleukin these lipoproteins are secreted mainly from astrocytes, but it
LCAT lecithin-cholesterol acyltransferase is known that neurons and microglia are also able to express
LDL low density lipoproteins some amounts of apolipoproteins in response to injury (Xu
LOX lipoxygenase et al. 2006). The lipoproteins secreted from glia not only trans-
LPA lysophosphatidic acid port lipids, but also transduce signals via receptors of the LDL
LPCAT acetylCoA:1-o-alkyl-2-lysophosphatidylcho- receptor family. The members of the LDL receptor family are
line acetyltransferase widely and abundantly expressed in cells of the CNS, including
LPS lipopolysaccharide neurons, astrocytes, microglia, and oligodendrocytes.
LRP low density lipoprotein receptor-related Lipid mediators are the bioactive messengers formed mostly
protein from phospholipids, sphingolipids, and cholesterol of the cell
LT leukotriene membrane. The brain is rich in long-chain polyunsaturated
MCAO middle cerebral artery occlusion fatty acids (PUFAs) such as arachidonic acid (AA) (20:4 n-6)
NF-κB nuclear factor-κB and docosahexaenoic acid (DHA) (22:6 n-3) (Wainwright
NPD1 neuroprotectin D1 2002). Hence, AA- and DHA-derived lipid mediators such
PAF platelet-activating factor as eicosanoids and docosanoids act as key endogenous regu-
PG prostaglandin lators of cell proliferation, differentiation, inflammation, and
PLA phospholipase A survival in neurons and glial cells. Because PUFAs cannot be
PS phosphatidylserine synthesized from 2-carbon fragments in mammalian cells,
PUFA polyunsaturated fatty acid the brain needs to take them up directly through the BBB.
TNF-α tumor necrosis factor-α They can be found in dietary sources such as eggs, fish, and
tPA tissue plasminogen activator meat, or can be synthesized from dietary essential fatty acid
TXA thromboxane A precursors of AA and DHA such as linoleic acid (18:2 n-6) and
VLDL very low density lipoproteins α-linolenic acid (18:3 n-3), respectively (Rapoport 2003). The

281
majority of these free fatty acids (FFAs) in the CNS are incor- The human brain contains approximately 25% of the total
porated into phospholipids of membranes and used as struc- body cholesterol, although the brain accounts for only 2% of
tural components and signaling molecules. A small amount of total body mass. It has been estimated that approximately 70% of
FFAs is β-oxidized, after activation to acyl-CoA by acyl-CoA CNS cholesterol is present in myelin, 20% is present in astrocytes
synthetases (Hamilton and Brunaldi 2007) (Fig. 23.1). and microglia, and the remainder is present in neurons. Quan
et al. (2003) demonstrated that the cholesterol concentration
in brains of LDL receptor-, Apo E-, or Apo A1-deficient mice
was not altered, although outside the brain these deficient mice
2 L I P I D H O M E O S TA S I S showed significant changes. This evidence suggests that the cho-
lesterol concentration within the brain is strictly regulated by the
The regulation of lipid homeostasis is distinct between the brain cells. Because adult neurons produce only small amounts
inside and outside of the CNS because of the separation of of cholesterol (Nieweg et al. 2009), it is thought that glial cells
these compartments by the BBB. Although lipoproteins in play a central role in lipid/cholesterol homeostasis in the CNS.
Drosophila can cross the BBB, which is functionally like that
in vertebrates (Brankatschk and Eaton 2010), lipoproteins
in the bloodstream of mouse, rat, and human do not cross 2.1 A S T RO C Y T E S
the BBB (Dietschy 2009). It was reported that the apolipo-
protein (apo) E genotype was changed to that of the donor Astrocytes play crucial roles in maintaining lipid homeostasis
in the plasma of a patient who had liver transplantation, in the CNS. In a coculture system for neurons and astrocytes,
whereas the genotype of the recipient was retained in CSF cholesterol is synthesized predominantly in astrocytes. When
(Linton et al. 1991). Furthermore, radiolabeled cholesterol astrocytes are present in the same culture environment as neu-
administrated into the circulation was not detected in the rons, the neurons downregulate squalene synthase, which cat-
CNS, and the rate of accumulation of sterols, such as choles- alyzes the biosynthesis of a key cholesterol precursor, squalene
terol and desmosterol, was similar to that of newly synthe- (Nieweg et al. 2009). Thus, neurons do not efficiently synthe-
sized sterols in the brain ( Jurevics and Morell 1995). Thus, size cholesterol and rely on the provision of cholesterol from
brain cells control their own lipid homeostasis, especially for astrocyte-derived lipoproteins associated with apo E, which is
cholesterol, in the CNS. a main apolipoprotein secreted from astrocytes.

Plasma
VLDL
Albumin LDL
HDL
DHA, AA
BBB

HDL Astrocyte HDL

Neuron CNS
CoA
HDL

HDL

Figure 23.1 Model of Fatty Acid Transport into the CNS and Lipoprotein Environment Between the Plasma and CNS. PUFAs such as arachidonic acid
(AA) and docosahexaenoic acid (DHA) in the plasma disassociate from albumin and cross the BBB. The PUFAs are activated by acyl-CoA synthetase
for making membrane phospholipids. There are several types of lipoproteins such as VLDL, LDL, and HDL in the plasma, whereas only HDL-like
particles, derived mainly from astrocytes, are in the CNS. The system of lipid metabolism and transport in the CNS is different from that in the
peripheral circulation. One of the reasons is that these lipoproteins including cholesterol are not able to cross the BBB.

282 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Although the essential PUFAs such as AA and DHA are oligodendrocytes, revealed a severe disruption of CNS myeli-
highly enriched in neurons of the brain, cultured neurons nation as well as ataxia and tremor. The studies in these mutant
synthesize almost no DHA and do not desaturate PUFA mice demonstrate that a high level of cholesterol in oligoden-
precursors (Moore et al. 1991). In contrast, astrocytes read- drocytes is essential for growth of the myelin membrane and
ily synthesize AA and DHA from their precursors. This evi- normal motor function (Saher et al. 2005).
dence indicates that astrocytes elongate and desaturate PUFA
precursors, then supply the preformed PUFAs to neurons 2.3 M I C RO G L I A
(Moore 2001). These PUFAs are an important source for lipid
mediators in neurons and glial cells. The immune defense system in the CNS is maintained pri-
Lecithin-cholesterol acyltransferase (LCAT) is local- marily by microglia (see chapter 50). Apo E has been reported
ized on the surface of HDL and converts cholesterol and to downregulate the activation of microglia and reduce the
phosphatidylcholine to cholesteryl ester and lysophosphati- secretion of inflammatory molecules such as tumor necrosis
dylcholine, respectively. In the CNS, LCAT is secreted pri- factor-α (TNF-α), interleukin (IL)-1β and IL-6 (Lynch et al.
marily from astrocytes, although neurons express LCAT 2001). The lack of apo E potentiates microglial activation and
mRNA at approximately threefold higher levels than astro- worsens neuronal death in the hippocampus of kainic acid–
cytes (Hirsch-Reinshagen et al. 2009). Apo A1 is known as treated mice (Duan et al. 2006).
the principal activator for plasma LCAT. However, although Phosphatidylserine (PS) is normally sequestered in the
astrocytes do not synthesize apo A1, apo E activates LCAT inner leaflet of the plasma membrane of cells. In the early
in conditioned medium of primary cultured astrocytes stage of apoptosis, PS becomes exposed on the outer leaflet so
(Hirsch-Reinshagen et al. 2009). This finding indicates that that the apoptotic cells are recognized and ingested by phago-
apo E secreted from astrocytes is probably the principal acti- cytes. Macrophages/microglia actively limit the production
vator of LCAT in the CSF. Cholesteryl ester transfer pro- of proinflammatory cytokines after the ingestion of apoptotic
tein (CETP), which mediates the exchange and transfer of cells. Administration of PS, as PS-containing lipoproteins, can
cholesteryl ester and triglycerides between lipoproteins, is imitate the microglial response against apoptotic cells thereby
present and active in the human CSF (Albers et al. 1992). inhibiting the secretion of TNF-α, nitric oxide and superox-
However, mice, rats, and some other species do not express ide from activated microglia (Hashioka et al. 2007). Some
CETP, and the levels of cholesteryl ester and triglycerides are clinical trials showed that oral administration of PS improved
low in the brain. CETP was detected in fibrillary astrocytes the behavior and cognitive functions of dementia patients
of white matter in normal conditions, whereas reactive astro- (McDaniel et al. 2003), but the efficacy of PS has not been
cytes were strongly positive in the white matter and also gray conclusively demonstrated.
matter of the Alzheimer diseased (AD) brain (Yamada et al.
1995). These observations suggest that astrocytes play signif-
icant roles in lipid homeostasis under physiological as well as 3 L I P O P R OT E I N S A N D
pathological conditions in the CNS. T H E I R R E C E P TO R S

Because the only lipoproteins in the CNS are HDL-like par-


2.2 O L I G O D E N D RO C Y T E S
ticles, their functions and regulatory mechanisms must differ
Because this hydrophobic membrane wraps the adjacent from those in the peripheral circulation and tissues. In addi-
axons with many layers that are rich in cholesterol, the concen- tion, the composition of apolipoproteins in the CNS is similar
tration of unesterified cholesterol in the spinal cord reaches to, but not the same as, that in plasma. The CSF contains:apo
40 mg/g, whereas the cholesterol concentration of hepato- AI, apo AII, apo CI, apo CII, apo CIII, apo D, apo E, and apo
cytes is only approximately 3 mg/g in mice (Dietschy and J, but not apo B (Ladu et al. 2000). Primary cultures of astro-
Turley 2004). Thus, the rate of cholesterol synthesis is high- cytes secrete apo E, apo J, and apo D, but not apo AI (LaDu
est during the first few weeks after birth, corresponding to et al. 1998). Thus, although apo A1 is the most abundant apo-
the period of active myelination. The cholesterol content of lipoprotein in plasma, apo E is the major apolipoprotein con-
the mouse whole brain markedly increases from 1.5 to 10.6 stituent of the CSF. Members of the LDL receptor family are
mg during myelinogenesis. This is also seen in the human abundantly expressed in neurons and glial cells of the CNS.
brain, in which cholesterol increases from approximately 360 In recent years, extensive research has demonstrated that these
g in the newborn to approximately 1,400 g in the adult. The lipoprotein receptors act not only as endocytotic receptors
rate of cholesterol accumulation then declines dramatically for lipoproteins, but also act as functionally diverse signaling
with maturation of the animals (Dietschy and Turley 2004). receptors in the CNS.
Cholesterol synthesis continues in mature animals, primarily
in astrocytes, at a very low level.
3.1 L I P O P ROT E I N F O R M AT I O N BY
Demyelination, that is, the loss of oligodendrocytes, is a
A D E N O S I N E T R I P H O S P H AT E –B I N D I N G
pathological process with implications for diseases causing
C A S S ET T E T R A NS P O RT E R S
serious disability such as multiple sclerosis (see chapter 61).
Studies with squalene synthase conditional knock-out mice, Adenosine triphosphate–binding cassette (ABC) transport-
in which cholesterol biosynthesis is inactivated specifically in ers are members of a protein superfamily that use ATP to

L I P I D S , L I P I D M E D I ATO R S , A N D OT H E R S I G N A L I N G M O L E C U L E S • 283
apoE-LP apoE-LP
apoA1 HDL

OH

OH
OH
chol PC chol SM
OH

OH
ABCA1 ABCG1/ABCG4

Figure 23.2 A Model of Lipid Transport by ABC Transporters. ABCA1 mediates the ATP-dependent efflux of cholesterol (chol) and phosphatidyl-
choline (PC) to lipid-poor apo A1 and apo E-containing lipoproteins. ABCG1 preferentially mediates the efflux of chol and sphingomyelin (SM) to
HDL and apo E-containing lipoproteins. ABCG4, expressed in the eye and brain, mediates the efflux of chol to HDL.

translocate solutes across membranes. An important function 3.2 A P O E - C O N TA I N I N G L I P O P ROT E I N S


of ABC transporters in the CNS is to mediate the efflux of cel- A N D T H E I R R EC E P TO R S
lular cholesterol and phospholipids for the formation of apo
3.2.1 Functions of Apo E-Containing Lipoproteins
E-containing lipoproteins from astrocytes. Approximately
50 ABC transporters have been identified in humans Apo E is recognized as the quantitatively and functionally
(Kimura et al. 2007). ABCA1 and ABCG1 are widely major apolipoprotein in the CNS and is a component of apo
expressed in multiple tissues, whereas ABCG4 is expressed E-containing lipoproteins secreted mainly from astrocytes
primarily in the CNS (Fig. 23.2). Because neurons do not (LaDu et al. 1998). In addition to previous studies, transgenic
efficiently produce cholesterol and apo E (Nieweg et al. knock-in mice that expressed green fluorescent protein regu-
2009; Xu et al. 2006), these ABC transporters mainly func- lated by the endogenous apo E promoter demonstrated that
tion in astrocytes. the main source of apo E in the CNS is astrocytes, although
In mice, the deficiency of ABCA1 demonstrated a remark- neurons and microglia expressed minor amounts of apo E in
able reduction of the cholesterol concentration, as well as the response to nerve injury (Xu et al. 2006).
apo E level, in the CSF and the brain, whereas the amount The expression and the secretion of apo E from glial cells
of apo J was not altered (Hirsch-Reinshagen et al. 2004). are markedly upregulated in response to nerve injury (Ignatius
Furthermore, brain-specific ABCA1 deficiency not only et al. 1986). Studies with apo E-deficient mice revealed signif-
decreased cholesterol and apo E in the CSF and the brain, but icant neurodegeneration in the brain during aging (Masliah
also resulted in abnormal motor activity and synaptic struc- et al. 1995), deficits of learning and memory (Gordon et al.
tures (Karasinska et al. 2009). These findings suggest that 1996), and enhanced susceptibility of hippocampal neurons
ABCA1 is required for the formation of apo E-, but not apo to endoplasmic reticulum stress caused by transient ischemia
J- containing lipoproteins in the CNS, and that ABCA1 can (Osada et al. 2009). Furthermore, apo E-containing lipopro-
alter the functions of the CNS. teins promote axon extension (Hayashi et al. 2004) and also
Although a deficiency of either ABCG1 or ABCG4 protect CNS neurons from apoptosis in a receptor-mediated
in mice resulted in essentially normal brain sterol levels. manner (Hayashi et al. 2007). Thus, it is thought that
A deficiency of both ABCG1 and ABCG4 significantly astrocyte-derived apo E-containing lipoproteins have impor-
increased brain sterol intermediates such as desmosterol, tant roles in the protection and/or repair of neurons.
lanosterol, and lathosterol. The brain and primary astrocytes The major role of apo E-containing lipoproteins secreted
from Abcg1–/–/Abcg4–/– mice showed a marked increase in from astrocytes is thought to be the supply of lipids to neurons
ABCA1. In addition, the deficiency of both ABCG1 and in the brain. It has been reported that cholesterol associated
ABCG4 increased the secretion of apo E from primary with apo E-containing lipoproteins stimulates synaptogenesis
astrocytes (Wang et al. 2008). These findings indicate that in CNS neurons (Mauch et al. 2001) (see chapter 31) and also
ABCG1 and ABCG4 have some overlapping functions in alters synaptic plasticity and neurotransmission in the CNS
sterol efflux from astrocytes, and that ABC transporters in (Pfrieger 2003). Apo E-containing lipoproteins are taken up
astrocytes tightly cooperate with each other to maintain the by members of the LDL receptor family and astrocyte-de-
CNS sterol environment. rived cholesterol can be used as a component of membrane

284 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
structures, a source for neurosteroids and lipid mediators in
neurons. Excess cholesterol in the CNS is converted to 24S-
hydroxycholesterol, a cholesterol-derived lipid mediator, by
cholesterol 24-hydroxylase. 24-Hydroxlase is expressed only
in certain types of neurons, such as pyramidal neurons of the
hippocampus and cerebral cortex and Purkinje neurons of Ligand binding repeat
β- propeller domain
the cerebellum (Ramirez et al. 2008). This oxysterol can cross
the neuron membrane and the BBB and diffuse into the plasma EGF repeat
for delivery to the liver and subsequent elimination from the O-linked sugar domain
body in bile. 24S-Hydroxycholesterol can also diffuse to astro- NPxY motif
cytes and act as a ligand for liver X receptors. These receptors
are the members of the nuclear hormone receptor superfamily
and regulate the expression of ABCA1, ABCG1 and apo E
(Fig. 23.3) (Abildayeva et al. 2006). Therefore, it is thought
that astrocytes contribute to brain functions by regulating
cholesterol transport and metabolism in the CNS.

3.2.2 Roles of Apo E Receptors


Receptors for apo E are members of the LDL receptor fam-
ily, which are expressed widely and abundantly in the CNS.
Apo E-containing lipoproteins can bind to all members of this
receptor family, which consists of seven core members—the
LDL receptor, the VLDL receptor, the LDL receptor–related LDLR VLDLR apoER2 LRP4 LRP1 LRP1B Megalin
protein 1 (LRP1), LRP1B, megalin/LRP2, LRP4/MEGF7, /LRP8 /MEGF7 /LRP2
and apoER2/LRP8, as well as three distant members—
Figure 23.4 The Structural Organization of Seven Core Members of the
LRP5, LRP6, and SorLA/LR11 (Bu, 2009) (Fig. 23.4). In the LDL Receptor Family. All members have common characteristic motifs
CNS, the VLDL receptor and apoER2/LRP8 play impor- such as ligand binding repeats, epidermal growth factor (EGF) repeats, and
tant roles in neuron migration during development and also β-propeller domains, and also contain NPxY motif(s) in the cytoplasmic tail.
modulate synaptic plasticity and neurite outgrowth via the

N-methyl-d-aspartate receptor (Herz et al. 2009). In addition,


the LDL receptor and LRP1 are the major receptors for lipid
metabolism in the CNS (Bu 2009). This is because knock-
out of the LDL receptor and conditional knock-out of the
forebrain neuron–specific LRP1 in mice increases the level of
apo E in the brain (Fryer et al. 2005; Liu et al. 2007). The
latter conditional knock-out mice, but not the LDL receptor
knock-out mice, also showed a decrease in brain cholesterol. In
addition, apo E-containing lipoproteins secreted from astro-
cytes have a higher affinity for the LDL receptor compared
with LRP1 (Fryer et al. 2005). Whereas HDL-like particles
isolated from the CSF prefer to bind to LRP1 than the LDL
receptor (Bu 2009; Fagan et al. 1996). Thus, the LDL recep-
tor and LRP1 might have different roles in lipid metabolism
in the brain.
The multifunctional receptor LRP1 (also known as the
α2-macroglobulin receptor and CD91) is expressed abun-
dantly in neurons, astrocytes, oligodendrocytes, and microglia
Figure 23.3 Cholesterol Homeostasis in Neurons and Astrocytes. Since (Gaultier et al. 2009). At least 40 different ligands for LRP1
neurons synthesize cholesterol (Chol) at low level in the adult brain,
astrocytes provide cholesterol associated with apo E-containing lipo- have been identified, including apo E, α2-macroglobulin, tis-
proteins (Apo E-LP) in processes mediated by ABCA1 and ABCG1 sue plasminogen activator (tPA), amyloid β peptide (Aβ),
in astrocytes. Apo E-LP bind to receptors of the LDL receptor family and amyloid precursor protein. LRP1 acts as an endocytotic
(LDLRs) in neurons, are endocytosed and used for cell structure and receptor for apo E-containing lipoproteins and other ligands
functions. Any excess cholesterol is converted to 24S-hydroxycholesterol and also functions as a transmembrane signaling receptor.
(24-OH chol), which can diffuse from neurons, cross the BBB, and go
into the bloodstream. This oxysterol also diffuses to astrocytes, stimu- In Schwann cells, LRP1 activates the major anti-apoptotic
lates liver X receptor (LXR), and enhances the expression of apo E and enzyme, phosphatidylinositol 3-kinase, and antagonizes the
ABCA1, thereby promoting cholesterol efflux. unfolded protein response-activated proapoptotic signaling

L I P I D S , L I P I D M E D I ATO R S , A N D OT H E R S I G N A L I N G M O L E C U L E S • 285
pathway after nerve injury (Mantuano et al. 2011). LRP1 inter- oligodendrocytes and astrocytes. Megalin in astrocytes mediates
acts with myelin basic protein and contributes to phagocyto- the uptake of albumin, which stimulates the synthesis of oleic
sis of degraded myelin in astrocytes and microglia (Gaultier acid; oleic acid can act as a neurotrophic factor (Bento-Abreu
et al. 2009). Ischemic brain injury increases the expression of et al. 2008). The expression of metallothionein, which is released
LRP1 in astrocytes and induces an interaction between LRP1 from astrocytes and has been shown to be a neuroprotective met-
and tPA followed by activation of nuclear factor-κB (NF-κB) al-binding protein, is markedly increased in reactive astrocytes
(Zhang et al. 2007). In primary cultured microglia, the acti- of sporadic Parkinson diseased brains, and megalin expression is
vation of LRP1 by apo E suppressed c-jun N-terminal kinase also upregulated (Michael et al. 2011). Because metallothionein
activation induced by lipopolysaccharides (LPS) (Pocivavsek is a ligand of megalin, this finding indicates that megalin might
et al. 2009), and the interaction of microglial LRP1 and tPA have a neuroprotective role in the brain.
increased matrix metalloproteinase-9 expression and activity, Apo J directly binds to Aβ and to apo E, which is also a
which was associated with the development of brain edema, ligand for megalin. Because megalin is expressed in the epithe-
after middle cerebral artery occlusion (MCAO) in mice lium and ependymal cells at the choroid plexus, the apo J- or
(Zhang et al. 2009). These data suggest that LRP1 not only apo E-Aβ complex is thought to be removed from the brain
contributes to lipid metabolism, but also plays a crucial role in into plasma by megalin and other receptors such as LRP1
inflammation, survival, and other signaling events in glia. (Nuutinen et al. 2009). Although the functions of megalin as
an apo J receptor on glial cells and in the adult CNS is still not
clearly defined, this receptor seems to be an important media-
3.3 A P O J- C O N TA I N I N G L I P O P ROT E I N S tor between the CNS and the peripheral circulation.
A N D T H E I R R E C E P TO R S
3.3.1 Functions of Apo J-Containing Lipoproteins 3.4 A P O A1- C O N TA I N I N G L I P O P ROT E I N S
Apo J (also known as clusterin) is a heterodimeric, sulfated gly- Apo A1 is the most abundant apolipoprotein in plasma, but
coprotein that is expressed at high levels in the brain, ovary, tes- in the CSF the concentration of apo A1 is only 0.5% of that in
tis, and liver (de Silva et al. 1990). Similar to apo E, apo J exists in plasma (Pitas et al. 1987). Exogenously administrated apo A1
the form of lipoproteins that are secreted from astrocytes in the can induce the translocation of newly synthesized cholesterol
CNS. The levels of apo J mRNA and protein are also increased and phospholipids to the lipid-protein particles and gener-
in astrocytes after nerve injury. Cultured astrocytes isolated ate HDL-like particles from primary cultured astrocytes (Ito
from apo E-deficient mice secrete lipoprotein particles that are et al. 2004). This result indicates that apo A1 is functional in
lipid-poor compared with the particles secreted in wild-type the CNS, although this apolipoprotein is not made within the
mice. However, the lipoprotein particles express comparable CNS. Overexpression of human apo A1 in a mouse model of
amounts of apo J. Moreover, primary cultured astrocytes from AD suppressed the activation of astrocytes and microglia and
apo J-deficient mice secrete lipoproteins similar to those from improved spatial learning and memory functions in vivo. In
wild-type mice (Ladu et al. 2000). Thus, apo J and apo E are addition, exogenous administration of human apo A1 attenu-
associated with distinct lipoproteins in the CSF. ated the activation of cultured microglia and the secretion of
The deficiency of apo J does not remarkably alter the mor- proinflammatory cytokines induced by Aβ from hippocampal
phology of the CNS during development, but worsens the slice cultures (Lewis et al. 2010). It has also been reported that
susceptibility to ischemic brain injury (Imhof et al. 2006). the concentration of serum apoA1 is inversely associated with
Cytosolic apo J can bind to Bax, a pro-apoptotic protein, and the risk of AD (Saczynski et al. 2007). Moreover, the level of
inhibit cytochrome c release from mitochondria and the sub- apo A1 is reduced in the CSF, the brain and peripheral tissues
sequent activation of a caspase cascade (Zhang et al. 2005). of subjects with schizophrenia (Huang et al. 2008). Thus, apo
These findings indicate that apo J might be a neuroprotective A1 might play important roles in brain functions. However,
molecule. However, it is also reported that the lack of apo J more investigations are required.
attenuated neurodegeneration after ischemic injury in vitro
and in vivo (Han et al. 2001). Furthermore, the translocation
of truncated apo J to the nucleus induces cell death (Yang 4 L I P I D M E D I ATO R S A N D
et al. 2000). Apo J also stimulates the proliferation of primary T H E I R R E C E P TO R S
cultured astrocytes through an extracellular signal-regulated
kinase signaling pathway. Moreover, astrocyte-derived apo J Lipid mediators are bioactive regulators that are enzymatically
promotes neuronal differentiation in human neuronal precur- formed from membrane glycerophospholipids, sphingolipids,
sor cells (Cordero-Llana et al. 2011). Thus, apo J may perform and cholesterol in response to cell stimulation or injury. Lipid
diverse roles in the CNS but further research is required to mediators are involved in inter-cell and intra-cell communi-
establish these functions. cations and responses such as proliferation, differentiation,
adhesion, migration, oxidative stress, inflammation, and cell
3.3.2 Roles of Apo J Receptor survival. Phospholipases A2 (PLA2s) belong to a superfam-
ily of lipolytic enzymes that catalyze the hydrolysis of fatty
Apo J interacts with megalin (also known as LRP2), which is a acids from the sn-2 position of glycerophospholipids to
multiligand endocytic cell-surface receptor that is expressed in produce lysophospholipids and FFAs such as AA and DHA

286 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
(Fig. 23.5). More than 30 different types of PLA2s have been The enzyme 5-LOX converts AA into 5-hydroxyeicosatetr
found in mammalian cells and have been classified into five aenoic acid and leukotriene A4 (LTA4). In humans, the expres-
major groups: cytosolic PLA2, Ca2+-independent PLA2, lyso- sion of 5-LOX was upregulated in astrocytes at damaged foci
somal PLA2, secretory PLA2, and platelet-activating factor after ischemic injury, whereas COX-2 was increased in neurons
(PAF) acetyl hydrolases (Schaloske and Dennis 2006). These in the peri-infarct area (Tomimoto et al. 2002). In a rat model
PLA2s are not only important for regulating the levels of cell of focal cerebral ischemia, a 5-LOX inhibitor reduced infarct
membrane phospholipids, but they also play crucial roles volume and dampened the increase in 5-LOX expression in
in regulating the production of a variety of lipid mediators, neurons and microglia after cerebral ischemia. Furthermore,
which have been implicated in diverse events of physiological the 5-LOX inhibitor also blocked LPS-mediated activation of
and pathological states (Liu and Xu 2010). NF-κB in primary cultured microglia ( Jatana et al. 2006).

4 .1 A R AC H I D O N I C AC I D –D E R I V E D 4.1.1 Functions of Prostaglandins and


E I C O S A N O I D S A N D T H E I R R E C E P TO R S Their Receptors
Eicosanoids are generated from AA by cyclooxygenases In the CNS, activated microglia and reactive astrocytes
(COXs), lipoxygenases (LOXs), and epoxygenases. The two secrete large amounts of proinflammatory molecules includ-
COXs (COX-1 and COX-2) are rate-limiting enzymes in the ing cytokines, chemokines and PGs in response to neuroin-
conversion of AA to prostaglandins (PGs), such as PGD2, flammation, which is a diverse response to brain injury. PGs
PGE2, PGF2α, and PGI2, and thromboxane A2 (TXA2). Both and TXA2 are critical lipid mediators of neuroinflammation
COX-1 and COX-2 are expressed in the brain. COX-1 is gen- induced by traumatic brain injury, stroke, and AD (Hein and
erally considered to be constitutively expressed in most tissues O’Banion 2009). The receptors for PGD2, PGE2, PGF2α,
and mainly responsible for homeostatic states. In contrast, PGI2, and TXA2 are named DP, EP, FP, IP, and TP, respec-
COX-2 is primarily induced in response to inflammatory stim- tively. Among these receptors, the EPs (EP1–4) are the most
uli and mitogens in numerous pathological states (Milatovic well-characterized. PGE2 plays major physiological and path-
et al. 2011). However, in the CNS, COX-2 is also constitu- ological roles in the brain (Miller 2006). EPs are expressed
tively expressed in cortical and hippocampal glutamatergic in almost all organs at various levels, and the brain expresses
neurons, which are important for learning and memory (Yang all of the EPs. Astrocytes express EP2 and EP4, whereas
et al. 2008). COX-1 is predominantly localized to microglia microglia have EP1 and EP2. EP3 is inducibly expressed in
and perivascular cells, and the expression level of COX-1 is astrocytes and microglia. Neurons also express EP1, EP2,
robustly stimulated by inflammatory stimuli (Garcia-Bueno and EP3 in multiple areas of the brain, whereas EP4 localizes
et al. 2009). Moreover, COX-1-positive microglia were found in hypothalamic nuclei (Cimino et al. 2008). It is reported
surrounding amyloid plaques in the AD brain (Yermakova that exogenous PGE2 administration to primary cultured
et al. 1999). These findings indicate that COX-1 in microglia hippocampal neurons induced caspase-dependent apop-
might play important roles in inflammation, whereas COX-2 tosis (Takadera et al. 2004). In primary cultured microglia
might contribute to neurotransmission and synaptic plasticity isolated from EP2-deficient mice, LPS-mediated paracrine
in the CNS. neurotoxicity was reduced. The presence of EP2 in microglia

Phospholipids

PLA 2s

Docosahexaenoic acid Arachidonic acid Lysophospholipids

15-LOX LPCAT ATX

Neuroprotectins Resolvins Platelet activating factor Lysophosphatidic acid

COX 5-LOX

Thromboxanes Prostaglandins Leukotrienes

Figure 23.5 Pathways for Synthesis of Lipid Mediator. Phospholipids in cell membranes are hydrolyzed by phospholipases A2 (PLA2s) to liberate
PUFAs, such as arachidonic acid and docosahexaenoic acid, and lysophospholipids. Lysophospholipids are further converted to platelet activating
factor by acetyl CoA:1-O-alkyl-2-lysophosphatidylcholine acetyltransferase (LPCAT), or lysophosphatidic acid by autotaxin (ATX). Arachidonic
acid is converted to prostaglandins and thromboxanes, and leukotrienes by cyclooxygenase (COX) and 5-lipoxygenase (5-LOX), respectively.
Docosahexaenoic acid is converted to resolvins and neuroprotectins by 15-lipoxygenase (15-LOX).

L I P I D S , L I P I D M E D I ATO R S , A N D OT H E R S I G N A L I N G M O L E C U L E S • 287
was shown to be necessary to induce COX-2 and inducible (ATX) (Savaskan et al. 2007). ATX has been detected in
nitric oxide synthase for this neurotoxicity (Shie et al. 2005). oligodendrocytes, choroid plexus epithelial cells, and lep-
In primary cultured cortical astrocytes, exogenous LPS treat- tomeningeal cells under normal conditions. However, neu-
ment upregulated the expression of various enzymes involved rotrauma led to a significant increase in the level of ATX
in PG synthesis, such as cPLA2, COX-2, and PGE synthase, specifically in reactive astrocytes (Savaskan et al. 2007). To
whereas downregulating the expression of PGE-degrading date, GPCRs for LPA have been classified into five sub-
enzymes ( Johann et al. 2008). Thus, astrocytes also partici- types: LPA1–5 (Tigyi 2010). It is possible that two or four
pate in the innate immune response by providing PGE2 in the more receptors will be recognized as LPA receptors. In the
CNS. Moreover, the level of PGE2 in the CSF was increased nervous system, LPA1 was originally identified in neuronal
in patients with neurological disorders such as AD, amyo- progenitor cells of the ventricular zone and subsequently
trophic lateral sclerosis, Creutzfeldt-Jakob disease, ischemic found in oligodendrocytes. Low expression of LPA2–5
stroke, and human immunodeficiency virus (HIV)–associated has been detected in the brain by Northern blot analy-
dementia (Cimino et al. 2008). sis. Exogenous administration of LPA increases the area
PGD2 is the most abundant prostanoid in the mamma- of oligodendrocyte’s process network and the expression
lian brain (Narumiya et al. 1982) and plays a variety of roles of myelin basic protein during oligodendrocyte matura-
in the immune system. Studies with the demyelinating mouse tion (Nogaroli et al. 2009). The expression of LPA recep-
twitcher, a model of human Krabbe disease, demonstrated tors appears to be low in a normal human brain, whereas
that activated microglia express hematopoietic prostaglandin LPA is increased on reactive astrocytes following traumatic
D synthase (HPGDS), and that reactive astrocytes expressed brain injury (Frugier et al. 2011). These findings indicate
DPs for PGD2. Deficiency of HPGDS or DP in this disease that LPA and ATX may play important roles in the brain
model markedly suppressed astrogliosis and demyelination under normal and pathological conditions, particularly in
(Mohri et al. 2006). These findings indicate that PGD2 is the oligodendrocytes.
key factor for promoting these neuroinflammatory responses
in the brain.
4.3 D O C O S A H E X A E N O I C AC I D A N D
IT S M ETA B O L IT E S
4.1.2 Functions of Leukotrienes and
DHA (22:6 n-3) is one of the most abundant PUFAs in
Their Receptors
the brain; another is AA. In the adult rat, 2% to 8% of
Leukotrienes, a family of eicosanoids, are made from AA by esterified brain DHA and 3% to 5% of esterified brain AA
5-LOX. An integral membrane protein, 5-LOX-activating are replaced daily by plasma unesterified PUFAs. In the
protein (FLAP), is required for this conversion. FLAP selec- human brain, 0.3% of AA is replaced daily (Rapoport
tively transfers AA to 5-LOX and enhances the production et al. 2001). DHA and its metabolites are antiinflam-
of LTA4. LTA4 is further converted into LTB4 or cysteinyl matory, wheres AA and its metabolites are generally
leukotrienes, LTC4, LTD4, and LTE4. These leukotrienes are proinflammatory. In vitro cell culture studies showed
proinflammatory lipid mediators. Their G protein–coupled that astrocytes readily produce DHA from their precur-
receptors (GPCRs) have been identified; BLT1 and BLT2 sors, whereas neurons synthesize little DHA and have no
are receptors for LTB4, and CysLT1, CysLT2, and CysLT3 are desaturation activity. Thus, neurons rely on the uptake
receptors for LTC4, LTD4, and LTE4. It is reported that the of preformed DHA from external sources in the CNS
production of cysteinyl leukotrienes was increased in the rat (Moore, 2001). DHA is converted to the resolvins and
brain after MCAO. Treatment of these animals with a 5-LOX the neuroprotectins by 15-LOX. Resolvins downregulate
inhibitor decreased the levels of cysteinyl leukotrienes and TNF-α–stimulated IL-1β expression in human glial
the size of infarct area (Ciceri et al. 2001). Furthermore, the cells (Hong et al. 2003). DHA stimulates neuroprotec-
expression of CysLT1 was upregulated in neurons and microg- tin D1 (NPD1) biosynthesis and attenuates the secretion
lia of the ischemic core and reactive astrocytes of the bound- of Aβ, which can cause neuronal apoptosis. NPD1 also
ary zone after MCAO in rats. Moreover, a CysLT1 antagonist upregulates the antiapoptotic proteins, Bcl-2 and Bcl-xL,
reduced infarct area, neurological deficits, and astrocyte pro- and downregulates the proapoptotic proteins, Bax and Bad,
liferation (Fang et al. 2006). These findings demonstrated that during cerebral ischemia (Bazan 2005). These findings dem-
cysteinyl leukotrienes and CysLT1 in microglia and astrocytes onstrate that DHA and its metabolites—the resolvins and
play significant roles in causing neuronal damage after cerebral NPD1—facilitate neuroprotection by inducing the expres-
ischemia. sion of neuroprotective molecules and reducing the secretion
of neurotoxic molecules.
4.2 LY S O P H O S P H O L I P I D, LYS O P H O S P H O L I P I D
D E R I VAT I VE S , A N D T H E I R R E C E P TO R S 4.4 OT H E R L I P I D M E D I ATO R S A N D
T H E I R R EC E P TO R S
Lysophosphatidic acid (LPA) is a metabolite of glycero-
phospholipids. Several potential pathways produce LPA in Platelet-activating factor (PAF), 1-O-alkyl-2-acetyl-sn-
cells. One example is the production of LPA extracellularly glycero-3-phosphorylcholine, is a potent proinflammatory
by the action of a lysophospholipase D called autotaxin phospholipid mediator in infectious and inflammatory

288 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
diseases. PAF is generated from lysophospholipids by ace- regulates apolipoprotein E-mediated cholesterol efflux. J Biol Chem
tyl CoA: 1-o-alkyl-2-lysophospatidylcholine acetyltrans- 281:12799–12808.
Aihara M, Ishii S, Kume K, Shimizu T. 2000. Interaction between neu-
ferase. PAF is released from many cell types, including rone and microglia mediated by platelet-activating factor. Genes
neurons and glia. In the CNS, sustained PAF activation Cells 5:397–406.
has been observed in ischemia, encephalitis, meningitis, Albers JJ, Tollefson JH, Wolfbauer G, Albright RE Jr. 1992.
and HIV infection. PAF synthesis and secretion in cul- Cholesteryl ester transfer protein in human brain. Int J Clin Lab Res
tured neurons were upregulated by glutamate treatment, 21:264–266.
Bazan NG. 2005. Neuroprotectin D1 (NPD1): a DHA-derived media-
and cultured microglia showed a remarkable chemotactic tor that protects brain and retina against cell injury-induced oxida-
response to PAF. Furthermore, this chemotaxis was not tive stress. Brain Pathol 15:159–166.
found in microglia isolated from PAF receptor–deficient Bennett SA, Chen J, Pappas BA, Roberts DC, Tenniswood M. 1998.
mice (Aihara et al. 2000). PAF receptors from various spe- Platelet activating factor receptor expression is associated with neu-
cies possess a structure typical of GPCRs with seven trans- ronal apoptosis in an in vivo model of excitotoxicity. Cell Death
Differ 5:867–875.
membrane helices. PAF receptors are expressed primarily in Bento-Abreu A, Velasco A, Polo-Hernandez E, Perez-Reyes PL,
neurons and microglia under normal conditions. However, Tabernero A, Medina JM. 2008. Megalin is a receptor for albumin in
the expression of the PAF receptor is markedly upregulated astrocytes and is required for the synthesis of the neurotrophic factor
in activated microglia and reactive astrocytes for targeting oleic acid. J Neurochem 106:1149–1159.
apoptotic debris after kainic acid treatment (Bennett et al. Brankatschk M, Eaton S. 2010. Lipoprotein particles cross the
blood-brain barrier in Drosophila. J Neurosci 30:10441–10447.
1998). Thus, PAF receptors potentially mediate the cere- Bu G. 2009. Apolipoprotein E and its receptors in Alzheimer’s disease:
bral phagocytic response by activated glia. pathways, pathogenesis and therapy. Nat Rev Neurosci 10:333–344.
Ciceri P, Rabuffetti M, Monopoli A, Nicosia S. 2001. Production of
leukotrienes in a model of focal cerebral ischaemia in the rat. Br J
Pharmacol 133:1323–1329.
5 S U M M A RY A N D P E R S P E C T I VE S Cimino PJ, Keene CD, Breyer RM, Montine KS, Montine TJ. 2008.
Therapeutic targets in prostaglandin E2 signaling for neurologic
Lipid homeostasis in the CNS is tightly regulated under disease. Curr Med Chem 15:1863–1869.
physiological conditions by a partnership between neurons Cordero-Llana O, Scott SA, Maslen SL, Anderson JM, Boyle J,
and glia, especially astrocytes. In recent years, critical roles Chowhdury RR, et al. 2011. Clusterin secreted by astrocytes enhances
neuronal differentiation from human neural precursor cells. Cell
have been elucidated for many players associated with lipid Death Differ 18:907–913.
metabolism in the CNS, such as ABC transporters and mem- de Silva HV, Harmony JA, Stuart WD, Gil CM, Robbins J. 1990.
bers of the LDL receptor family. However, many specific func- Apolipoprotein J: structure and tissue distribution. Biochemistry
tions of these molecules in neurons and each type of glia under 29:5380–5389.
physiological and pathological conditions have not yet been Dietschy JM. 2009. Central nervous system: cholesterol turnover, brain
development and neurodegeneration. Biol Chem 390:287–293.
defined. Dietschy JM, Turley SD. 2004. Thematic review series: brain
Dietary PUFAs, such as AA and DHA, are important Lipids. Cholesterol metabolism in the central nervous system dur-
sources for structural membrane components and lipid ing early development and in the mature animal. J Lipid Res 45:
mediators, as discussed. In many cases, pathological stimuli 1375–1397.
recruit activated microglia, reactive astrocytes, and inflamma- Duan RS, Chen Z, Dou YC, Concha Quezada H, Nennesmo I, Adem
A, et al. 2006. Apolipoprotein E deficiency increased microglial acti-
tory response; including the secretion of lipid mediators, and vation/CCR3 expression and hippocampal damage in kainic acid
induction of neuroprotective and/or neurotoxic responses by exposed mice. Exp Neurol 202:373–380.
lipid mediators dependent on how stimuli are produced. To Fagan AM, Bu G, Sun Y, Daugherty A, Holtzman DM. 1996.
define these responses in glia, further research is required. Apolipoprotein E-containing high density lipoprotein promotes
neurite outgrowth and is a ligand for the low density lipoprotein
receptor-related protein. J Biol Chem 271:30121–30125.
Fang SH, Wei EQ, Zhou Y, Wang ML, Zhang WP, Yu GL, et al. 2006.
AC K N OW L E D G M E N T S Increased expression of cysteinyl leukotriene receptor-1 in the brain
mediates neuronal damage and astrogliosis after focal cerebral ische-
The author would like to thank Jean E. Vance for critical mia in rats. Neuroscience 140:969–979.
reviewing of the manuscript during its preparation and Mie Frugier T, Crombie D, Conquest A, Tjhong F, Taylor C, Kulkarni T,
et al. 2011. Modulation of LPA receptor expression in the human
Moriyama for help in preparing the figures. This work was brain following neurotrauma. Cell Mol Neurobiol 31:569–577.
supported in part by a grant-in-aid for Young Scientists B Fryer JD, Demattos RB, McCormick LM, O’Dell MA, Spinner ML,
(No. 22790254) and the Special Coordination Funds for Bales KR, et al. 2005. The low density lipoprotein receptor regulates
Promoting Science and Technology from the Ministry of the level of central nervous system human and murine apolipoprotein
Education, Culture, Sports, Science and Technology, Japan. E but does not modify amyloid plaque pathology in PDAPP mice.
J Biol Chem 280:25754–25759.
Garcia-Bueno B, Serrats J, Sawchenko PE. 2009. Cerebrovascular
cyclooxygenase-1 expression, regulation, and role in hypothalamic-
REFERENCES pituitary-adrenal axis activation by inflammatory stimuli. J Neurosci
29:12970–12981.
Abildayeva K, Jansen PJ, Hirsch-Reinshagen V, Bloks VW, Bakker Gaultier A, Wu X, Le Moan N, Takimoto S, Mukandala G, Akassoglou
AH, Ramaekers FC, et al. 2006. 24(S)-hydroxycholesterol par- K, et al. 2009. Low-density lipoprotein receptor-related protein 1 is an
ticipates in a liver X receptor-controlled pathway in astrocytes that essential receptor for myelin phagocytosis. J Cell Sci 122:1155–1162.

L I P I D S , L I P I D M E D I ATO R S , A N D OT H E R S I G N A L I N G M O L E C U L E S • 289
Gordon I, Genis I, Grauer E, Sehayek E, Michaelson DM. 1996. Leskovjan AC, Kretlow A, Miller LM. 2010. Fourier transform infra-
Biochemical and cognitive studies of apolipoprotein-E-deficient red imaging showing reduced unsaturated lipid content in the hip-
mice. Mol Chem Neuropathol 28:97–103. pocampus of a mouse model of Alzheimer’s disease. Anal Chem
Hamilton JA, Brunaldi K. 2007. A model for fatty acid transport into 82:2711–2716.
the brain. J Mol Neurosci 33:12–17. Lewis TL, Cao D, Lu H, Mans RA, Su YR, Jungbauer L, et al. 2010.
Han BH, DeMattos RB, Dugan LL, Kim-Han JS, Brendza RP, Fryer Overexpression of human apolipoprotein A-I preserves cognitive
JD, et al. 2001. Clusterin contributes to caspase-3-independent brain function and attenuates neuroinflammation and cerebral amyloid
injury following neonatal hypoxia-ischemia. Nat Med 7:338–343. angiopathy in a mouse model of Alzheimer disease. J Biol Chem
Hashioka S, Han YH, Fujii S, Kato T, Monji A, Utsumi H, et al. 2007. 285:36958–36968.
Phosphatidylserine and phosphatidylcholine-containing liposomes Linton MF, Gish R, Hubl ST, Butler E, Esquivel C, Bry WI, et al. 1991.
inhibit amyloid beta and interferon-gamma-induced microglial acti- Phenotypes of apolipoprotein B and apolipoprotein E after liver
vation. Free Radic Biol Med 42:945–954. transplantation. J Clin Invest 88:270–281.
Hayashi H, Campenot RB, Vance DE, Vance JE. 2004. Glial lipopro- Liu NK, Xu XM. 2010. Phospholipase A2 and its molecular mechanism
teins stimulate axon growth of central nervous system neurons in after spinal cord injury. Mol Neurobiol 41:197–205.
compartmented cultures. J Biol Chem 279:14009–14015. Liu Q, Zerbinatti CV, Zhang J, Hoe HS, Wang B, Cole SL, et al. 2007.
Hayashi H, Campenot RB, Vance DE, Vance JE. 2007. Apolipoprotein Amyloid precursor protein regulates brain apolipoprotein E and
E-containing lipoproteins protect neurons from apoptosis via a sig- cholesterol metabolism through lipoprotein receptor LRP1. Neuron
naling pathway involving low-density lipoprotein receptor-related 56:66–78.
protein-1. J Neurosci 27:1933–1941. Lynch JR, Morgan D, Mance J, Matthew WD, Laskowitz DT. 2001.
Hein AM, O’Banion MK. 2009. Neuroinflammation and memory: the Apolipoprotein E modulates glial activation and the endogenous
role of prostaglandins. Mol Neurobiol 40:15–32. central nervous system inflammatory response. J Neuroimmunol
Herz J, Chen Y, Masiulis I, Zhou L. 2009. Expanding functions of lipo- 114:107–113.
protein receptors. J Lipid Res 50 Suppl:S287–292. Mantuano E, Henry K, Yamauchi T, Hiramatsu N, Yamauchi K, Orita
Hirsch-Reinshagen V, Donkin J, Stukas S, Chan J, Wilkinson A, Fan J, S, et al. 2011. The unfolded protein response is a major mechanism by
et al. 2009. LCAT synthesized by primary astrocytes esterifies choles- which LRP1 regulates Schwann cell survival after injury. J Neurosci
terol on glia-derived lipoproteins. J Lipid Res 50:885–893. 31:13376–13385.
Hirsch-Reinshagen V, Zhou S, Burgess BL, Bernier L, McIsaac SA, Masliah E, Mallory M, Ge N, Alford M, Veinbergs I, Roses AD. 1995.
Chan JY, et al. 2004. Deficiency of ABCA1 impairs apolipoprotein E Neurodegeneration in the central nervous system of apoE-deficient
metabolism in brain. J Biol Chem 279:41197–41207. mice. Exp Neurol 136:107–122.
Hong S, Gronert K, Devchand PR, Moussignac RL, Serhan CN. Mauch DH, Nagler K, Schumacher S, Goritz C, Muller EC, Otto A,
2003. Novel docosatrienes and 17S-resolvins generated from doco- et al. 2001. CNS synaptogenesis promoted by glia-derived choles-
sahexaenoic acid in murine brain, human blood, and glial cells. terol. Science 294:1354–1357.
Autacoids in anti-inflammation. J Biol Chem 278:14677–14687. McDaniel MA, Maier SF, Einstein GO. 2003. “Brain-specific” nutrients:
Huang JT, Wang L, Prabakaran S, Wengenroth M, Lockstone HE, a memory cure? Nutrition 19:957–975.
Koethe D, et al. 2008. Independent protein-profi ling studies show a Michael GJ, Esmailzadeh S, Moran LB, Christian L, Pearce RK, Graeber
decrease in apolipoprotein A1 levels in schizophrenia CSF, brain and MB. 2011. Up-regulation of metallothionein gene expression in
peripheral tissues. Mol Psychiatry 13:1118–1128. Parkinsonian astrocytes. Neurogenetics 12:295–305.
Ignatius MJ, Gebicke-Harter PJ, Skene JH, Schilling JW, Weisgraber Milatovic D, Montine TJ, Aschner M. 2011. Prostanoid signaling:
KH, Mahley RW, et al. 1986. Expression of apolipoprotein E dur- dual role for prostaglandin E2 in neurotoxicity. Neurotoxicology
ing nerve degeneration and regeneration. Proc Natl Acad Sci U S A 32:312–319.
83:1125–1129. Miller SB. 2006. Prostaglandins in health and disease: an overview.
Imhof A, Charnay Y, Vallet PG, Aronow B, Kovari E, French LE, et al. Semin Arthritis Rheum 36:37–49.
2006. Sustained astrocytic clusterin expression improves remodeling Mohri I, Taniike M, Taniguchi H, Kanekiyo T, Aritake K, Inui T, et al.
after brain ischemia. Neurobiol Dis 22:274–283. 2006. Prostaglandin D2-mediated microglia/astrocyte interaction
Ito J, Li H, Nagayasu Y, Kheirollah A, Yokoyama S. 2004. Apolipoprotein enhances astrogliosis and demyelination in twitcher. J Neurosci
A-I induces translocation of protein kinase C[alpha] to a cytosolic 26:4383–4393.
lipid-protein particle in astrocytes. J Lipid Res 45:2269–2276. Moore SA. 2001. Polyunsaturated fatty acid synthesis and release by
Jatana M, Giri S, Ansari MA, Elango C, Singh AK, Singh I, et al. 2006. brain-derived cells in vitro. J Mol Neurosci 16:195–200; discussion
Inhibition of NF-kappaB activation by 5-lipoxygenase inhibitors 215–121.
protects brain against injury in a rat model of focal cerebral ischemia. Moore SA, Yoder E, Murphy S, Dutton GR, Spector AA. 1991. Astrocytes,
J Neuroinflammation 3:12. not neurons, produce docosahexaenoic acid (22:6 omega-3) and ara-
Johann S, Kampmann E, Denecke B, Arnold S, Kipp M, Mey J, et al. chidonic acid (20:4 omega-6). J Neurochem 56:518–524.
2008. Expression of enzymes involved in the prostanoid metabo- Narumiya S, Ogorochi T, Nakao K, Hayaishi O. 1982. Prostaglandin D2
lism by cortical astrocytes after LPS-induced inflammation. J Mol in rat brain, spinal cord and pituitary: basal level and regional distri-
Neurosci 34:177–185. bution. Life Sci 31:2093–2103.
Jurevics H, Morell P. 1995. Cholesterol for synthesis of myelin is made Nieweg K, Schaller H, Pfrieger FW. 2009. Marked differences in choles-
locally, not imported into brain. J Neurochem 64:895–901. terol synthesis between neurons and glial cells from postnatal rats.
Karasinska JM, Rinninger F, Lutjohann D, Ruddle P, Franciosi S, J Neurochem 109:125–134.
Kruit JK, et al. 2009. Specific loss of brain ABCA1 increases brain Nogaroli L, Yuelling LM, Dennis J, Gorse K, Payne SG, Fuss B. 2009.
cholesterol uptake and influences neuronal structure and function. Lysophosphatidic acid can support the formation of membranous
J Neurosci 29:3579–3589. structures and an increase in MBP mRNA levels in differentiating
Kimura Y, Kodan A, Matsuo M, Ueda K. 2007. Cholesterol fi ll-in model: oligodendrocytes. Neurochem Res 34:182–193.
mechanism for substrate recognition by ABC proteins. J Bioenerg Nuutinen T, Suuronen T, Kauppinen A, Salminen A. 2009. Clusterin:
Biomembr 39:447–452. a forgotten player in Alzheimer’s disease. Brain Res Rev 61:
LaDu MJ, Gilligan SM, Lukens JR, Cabana VG, Reardon CA, Van Eldik 89–104.
LJ, et al. 1998. Nascent astrocyte particles differ from lipoproteins in Osada N, Kosuge Y, Kihara T, Ishige K, Ito Y. 2009. Apolipoprotein
CSF. J Neurochem 70:2070–2081. E-deficient mice are more vulnerable to ER stress after transient fore-
Ladu MJ, Reardon C, Van Eldik L, Fagan AM, Bu G, Holtzman D, et al. brain ischemia. Neurochem Int 54:403–409.
2000. Lipoproteins in the central nervous system. Ann N Y Acad Sci Pfrieger FW. 2003. Outsourcing in the brain: do neurons depend on cho-
903:167–175. lesterol delivery by astrocytes? Bioessays 25:72–78.

290 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Pitas RE, Boyles JK, Lee SH, Hui D, Weisgraber KH. 1987. Lipoproteins Tomimoto H, Shibata M, Ihara M, Akiguchi I, Ohtani R, Budka
and their receptors in the central nervous system. Characterization H. 2002. A comparative study on the expression of cyclooxyge-
of the lipoproteins in cerebrospinal fluid and identification of nase and 5-lipoxygenase during cerebral ischemia in humans. Acta
apolipoprotein B,E(LDL) receptors in the brain. J Biol Chem Neuropathol 104:601–607.
262:14352–14360. Wainwright PE. 2002. Dietary essential fatty acids and brain func-
Pocivavsek A, Mikhailenko I, Strickland DK, Rebeck GW. 2009. tion: a developmental perspective on mechanisms. Proc Nutr Soc 61:
Microglial low-density lipoprotein receptor-related protein 1 61–69.
modulates c-Jun N-terminal kinase activation. J Neuroimmunol Wang N, Yvan-Charvet L, Lutjohann D, Mulder M, Vanmierlo T, Kim
214:25–32. TW, et al. 2008. ATP-binding cassette transporters G1 and G4
Quan G, Xie C, Dietschy JM, Turley SD. 2003. Ontogenesis and regu- mediate cholesterol and desmosterol efflux to HDL and regulate ste-
lation of cholesterol metabolism in the central nervous system of the rol accumulation in the brain. FASEB J 22:1073–1082.
mouse. Brain Res Dev Brain Res 146:87–98. Xu Q, Bernardo A, Walker D, Kanegawa T, Mahley RW, Huang Y. 2006.
Ramirez DM, Andersson S, Russell DW. 2008. Neuronal expression and Profi le and regulation of apolipoprotein E (ApoE) expression in the
subcellular localization of cholesterol 24-hydroxylase in the mouse CNS in mice with targeting of green fluorescent protein gene to the
brain. J Comp Neurol 507:1676–1693. ApoE locus. J Neurosci 26:4985–4994.
Rapoport SI. 2003. In vivo approaches to quantifying and imaging Yamada T, Kawata M, Arai H, Fukasawa M, Inoue K, Sato T. 1995.
brain arachidonic and docosahexaenoic acid metabolism. J Pediatr Astroglial localization of cholesteryl ester transfer protein in nor-
143:S26–34. mal and Alzheimer’s disease brain tissues. Acta Neuropathol 90:
Rapoport SI, Chang MC, Spector AA. 2001. Delivery and turnover of 633–636.
plasma-derived essential PUFAs in mammalian brain. J Lipid Res Yang CR, Leskov K, Hosley-Eberlein K, Criswell T, Pink JJ, Kinsella
42:678–685. TJ, et al. 2000. Nuclear clusterin/XIP8, an x-ray-induced
Saczynski JS, White L, Peila RL, Rodriguez BL, Launer LJ. 2007. The Ku70-binding protein that signals cell death. Proc Natl Acad Sci U
relation between apolipoprotein A-I and dementia: the Honolulu-Asia S A 97:5907–5912.
aging study. Am J Epidemiol 165:985–992. Yang H, Zhang J, Andreasson K, Chen C. 2008. COX-2 oxidative
Saher G, Brugger B, Lappe-Siefke C, Mobius W, Tozawa R, Wehr MC, metabolism of endocannabinoids augments hippocampal synaptic
et al. 2005. High cholesterol level is essential for myelin membrane plasticity. Mol Cell Neurosci 37:682–695.
growth. Nat Neurosci 8:468–475. Yermakova AV, Rollins J, Callahan LM, Rogers J, O’Banion MK. 1999.
Savaskan NE, Rocha L, Kotter MR, Baer A, Lubec G, van Meeteren LA, Cyclooxygenase-1 in human Alzheimer and control brain: quantita-
et al. 2007. Autotaxin (NPP-2) in the brain: cell type-specific expres- tive analysis of expression by microglia and CA3 hippocampal neu-
sion and regulation during development and after neurotrauma. Cell rons. J Neuropathol Exp Neurol 58:1135–1146.
Mol Life Sci 64:230–243. Zhang C, An J, Haile WB, Echeverry R, Strickland DK, Yepes M.
Schaloske RH, Dennis EA. 2006. The phospholipase A2 superfam- 2009. Microglial low-density lipoprotein receptor-related protein 1
ily and its group numbering system. Biochim Biophys Acta 1761: mediates the effect of tissue-type plasminogen activator on matrix
1246–1259. metalloproteinase-9 activity in the ischemic brain. J Cereb Blood
Shie FS, Montine KS, Breyer RM, Montine TJ. 2005. Microglial EP2 Flow Metab 29:1946–1954.
is critical to neurotoxicity from activated cerebral innate immunity. Zhang H, Kim JK, Edwards CA, Xu Z, Taichman R, Wang CY. 2005.
Glia 52:70–77. Clusterin inhibits apoptosis by interacting with activated Bax. Nat
Takadera T, Shiraishi Y, Ohyashiki T. 2004. Prostaglandin E2 induced Cell Biol 7:909–915.
caspase-dependent apoptosis possibly through activation of EP2 recep- Zhang X, Polavarapu R, She H, Mao Z, Yepes M. 2007. Tissue-type plas-
tors in cultured hippocampal neurons. Neurochem Int 45:713–719. minogen activator and the low-density lipoprotein receptor-related
Tigyi G. 2010. Aiming drug discovery at lysophosphatidic acid targets. protein mediate cerebral ischemia-induced nuclear factor-kappaB
Br J Pharmacol 161:241–270. pathway activation. Am J Pathol 171:1281–1290.

L I P I D S , L I P I D M E D I ATO R S , A N D OT H E R S I G N A L I N G M O L E C U L E S • 291
24.
GAP JUNCTIONS AND HEMICHANNELS
Bruce R. Ransom and Christian Giaume

A B B R E VI AT I O N S one another, leaving a narrow “gap” of only about 2.5 nm.


Each HC is composed of six Cxs (Goodenough 1976) and a
A astrocyte GJC is formed when a HC in one cell aligns with another HC
A:O astrocyte and oligodendrocyte coupling in an adjacent cell (Fig. 24.1). A gap junction is an aggregate
AMPA D-amino-3-hydroxy-5-methylisoxazole- of many, often hundreds, of tightly packed GJCs. Unlike Cxs,
4-propionic acid Panxs (homologs to innexin proteins in invertebrates) form
ATP adenosine-triphosphate stable membrane channels but not GJCs.
FGF fibroblast growth factor Each HC consists of 24 “rods” that correspond to the
CBX Carbenoxolone transmembrane spanning, alpha-helical portions of the six
GFAP glial fibrillary acid protein constituent Cxs (see Fig. 24.1). The extracellular loops of
GFP green fluorescent protein two HCs mesh to form an extracellular docking area con-
GJC gap junction channel sistent with a structural “tight seal” guaranteeing that the
HC hemichannel extracellular portion of the channel will not leak. The limit-
O oligodendrocyte ing diameter of the central pore is about 1.5 nm, setting the
O:O oligodendrocyte and oligodendrocyte upper limit for permeable ions and small molecules at about
coupling 1 to 1.2 kDa (Harris 2007).
Panx pannexin

2.2 C O N N E X I NS A N D PA N N E X I NS
1 INTRODUCTION
Connexins are a family of proteins with 20 and 21 members in
Gap junctions are membrane specializations consisting of dense rodents and humans, respectively. In the brain, at least 11 dif-
aggregates of large pore channels that extend from one cell into ferent Cxs have been detected, including: Cx26, Cx29, Cx30,
an adjacent cell and mediate direct cytoplasm-to-cytoplasm Cx32, Cx36, Cx37, Cx40, Cx43, Cx45, Cx46, and Cx47 (Sohl
communication. Gap junction channels (GJCs) in vertebrates and Willecke 2004; Theis et al. 2005). Connexins, named on
are formed by a family of proteins, called connexins (Cxs). the basis of their molecular weight, are transmembrane pro-
These channels are poorly selective for ions and small molec- teins with four transmembrane domains, two extracellular
ular weight signaling molecules, and allow extensive ionic and loops, and two cytoplasmic tails (see Fig. 24.1). Within the Cx
biochemical exchanges between cells. In the mammalian brain, protein family, the most conserved parts are the NH2 terminal
glia express Cxs most abundantly and in the greatest variety. and the two extracellular loops. Differences in Cx molecular
Under certain conditions Cxs can also operate as “half ” a GJC, weights are mainly reflected in the length of the C-terminal
or a hemichannel (HC), that represents another functional end. The functional gating of GJCs is primarily mediated
state providing a pathway suitable for autocrine as well as para- by the cytoplasmic loop and the C-terminal segment. The
crine interactions. Membrane channels may also be formed by extracellular loops mediate the end-to-end docking of paired
proteins called pannexins (Panxs) that have some properties HCs, and this union is stabilized by hydrophobic interactions
that are similar to Cx HCs. The characteristics and functions involving conserved cysteine residues (Harris 2001). All types
of these channels in glial cells are the focus of this chapter. of glia express more than one type of Cx and HCs of differ-
ent protein composition may form functional GJCs with one
another (Table 24.1). The six Cxs forming a HC can be identi-
cal, forming a homomeric HC, or consist of more than one Cx
2 G E N E R A L F E AT U R E S O F C O N N E X I N /
isoform, forming a heteromeric HC. A GJC formed from two
PA N N E X I N C H A N N E L S
different homomeric or heteromeric HCs is called heterotypic.
Astrocytes and oligodendrocytes necessarily form heterotypic
2.1 G A P J U N C T I O N A N D H E M I C H A N N E L
GJCs because they express different Cxs (astrocytes—Cx43
S T RU C T U R E
and Cx30; oligodendrocytes—Cx29, Cx32, and Cx47) (Nagy
In electron micrographs, gap junctions appear as discrete areas et al. 2003). Not all heterotypic “pairings” lead to functional
where the cell membranes of adjacent cells closely approach GJCs (Bukauskas and Verselis 2004). For example, Cx32

292
Pannexin
Pannexin channel
Extracellular
E1 E2
Cell 1
M1 M2 M3 M4

NH2
Intracellular CL
COOH

Gap junction plaque


Intracellular 1

Extracellular

Intracellular 2

Connexin hemichannel
Connexin
Extracellular
E1 E2

M1 M2 M3 M4
Cell 2
NH2 CL
Intracellular COOH

Figure 24.1 Gap Junction and Hemichannel Molecular Organization. Diagrammatic cells show how Cx GJCs form a gap junction plaque at a point
of close contact. Each GJC is formed by two Cx HCs docked together and rotated 30° with respect to one another. The center diagrams show the
topology of a Panx channel (top, Panx channels do not form GJCs; see text) and a Cx HC (bottom) in the plasma membrane. Both have four trans-
membrane domains (M1–4) with amino (NH2) and carboxy (COOH) termini on the cytoplasmic side, two extracellular loops (E1 and E2), and one
cytoplasmic loop (CL). Modified from Orellana et al 2009.

Table 24.1 GAP JUNCTION COUPLING AND CONNEXIN EXPRESSION IN MAMMALIAN GLIAL CELLS
GLIAL CELL PAIR RELATIVE COUPLING STRENGTH CONNEXIN PAIRING

Astrocytes ++++ (Sontheimer et al. 1991) Cx43-Cx43, CX43-Cx30, Cx30-Cx30, Cx30-Cx26, Cx26-Cx26
(Dermietzel et al. 1991; Giaume et al. 1991) (Dermietzel et al. 1991; Nagy et al. 1999; see also Giaume and Theis
2010; Giaume et al. 2010; Rash 2010; Theis et al. 2005)
Small amount of Cx40, Cx45, Cx46 (Dermietzel et al. 2000)

Astro-Oligo + (Ransom and Kettenmann 1990) Cx43-Cx47, Cx30-Cx32, CX26-Cx32 (Maglione et al. 2010; Nagy
et al. 2003)

Oligodendrocytes ++ (Von Blankenfeld et al. 1993) Cx32-Cx32 (Bergoffen et al. 1993),


(Butt and Ransom 1989; Kettenmann and Cx29-Cx29 (Altevogt et al. 2002; but see Ahn et al. 2008)
Ransom 1988) Cx47 (Maglione et al. 2010)

Schwann cells ++ Cx32-Cx32 (Altevogt et al. 2002)

Microglia + (Upregulated by cytokines) Cx43-Cx43 (Eugenin et al. 2001)

Muller cells ++ (Ceelen et al. 2001) Cx43-Cx43, Cx43-Cx45, Cx45-Cx45 (Zahs et al. 2003)
(Dermietzel et al. 2000)

Muller cell-astrocyte ++ (Ceelen et al. 2001) Cx43-Cx43, Cx43-Cx45, Cx45-Cx45

Ependymal cells +++ Cx43-Cx43 (Ochalski et al. 1997)

Astrocyte-neuron + Cx43-Cx36 (Alvarez-Maubecin et al. 2000; Froes et al. 1999; but see
Rash et al. 2001)

GAP JUNCTIONS AND HEMICHANNELS • 293


HCs form functional GJCs with Cx30 and Cx26 HCs, but transjunctional voltage is 0, whereas it decreases with hyper-
not with Cx43 HCs (Elfgang et al. 1995). Coupling between polarization or depolarization of either cell (Harris 2001).
different HCs is primarily determined by the compatibility of Increases in intracellular concentrations of H+ or Ca2+ rapidly
Cx extracellular loops (Harris 2001). decrease junctional conductance and permeability to large mol-
Another class of membrane proteins, the Panxs, forms ecules (Bennett et al. 1991; Harris 2001). Gating by pH oper-
membrane channels in mammalian brain with some similarity ates in the physiological range (i.e., 7 ± 0.5 pH) and depends
to Cx channels (Phelan and Starich 2001). Pannexin channels on the C-terminal cytoplasmic tail. The increases in intracel-
are expressed primarily in neurons (Sosinsky et al. 2011), but lular Ca2+ that result in decreased junctional conductance are
glia also express them (Bruzzone et al. 2003). Pannexin chan- high (e.g., t10 μM). A site sensitive to near-millimolar calcium
nels have functional properties similar to Cx HCs, and three is located on the extracellular portion of the pore (Gomez-
Panx isoforms (1–3) have been found in mouse and human; Hernandez et al. 2003). This explains how HCs are held in the
Panx1 and Panx2 have been detected in the brain (MacVicar closed position by high extracellular calcium, which maintains
and Thompson 2010). The structural topology of Panxs and cell integrity. However, the calcium-sensitive site is blocked
Cxs is similar (see Fig. 24.1), but there is only 16% overall in GJCs because the HC extracellular loops mesh tightly and
amino acid sequence identity (Panchin et al. 2000). prevent access. Signaling molecules such as neurotransmitters,
bioactive lipids and cytokines have complex effects on GJCs,
2.3 B I O P H YS I C A L P RO P E RT I E S O F
implying plasticity in the control of events mediated by GJCs
G A P JU N C T I O N C H A N N E L S
(Bennett et al. 1991; Giaume et al. 2010).

The permeability and biophysical properties of GJCs have been


2.4 P H Y S I O L O GY O F C O N N E X I N
studied using electrophysiological and imaging techniques
H E M I C H A N N E L S A N D PA N N E X I N C H A N N E L S
(Giaume et al. 2012). The extent of GJC-coupling among cells
can be detected using low molecular weight fluorescent dyes As expected from Ohm’s law, the unitary conductance of an
injected into single cells. The electrophysiological profile of a HC is double that of a GJC (Bennett et al. 2003). Connexin
given GJC (i.e., unitary conductance and voltage-dependent HCs have been identified in astrocytes after treatment with
properties) can indicate which protein(s) form the channels of proinflammatory cytokines (Retamal et al. 2007) or in amyloid
interest. Using such techniques, GJCs formed by Cx43 were treated astrocytes and microglia (Orellana et al. 2011c). They
identified between pairs of astrocytes (Dermietzel et al. 1991; can be gated open in several ways, including reducing external
Giaume et al. 1991) and Cx36 between pairs of microglial cells divalent cations (mainly Ca2+ and Mg2+), cell depolarization,
(Dobrenis et al. 2005), respectively. Dye injections can visual- and in some cases, elevation of intracellular Ca2+ (Fig. 24.2)
ize the shape and spatial distribution of coupled cells in whole (Ye et al. 2003, 2009). Pannexin channels have much higher
tissue such as acute brain slices or in vivo (Ball et al. 2007; unitary conductance compared with Cx HCs (but see Ma et al.
Houades et al. 2006). 2012) with rather similar permeability properties. However,
Electrophysiological analysis at the single channel level Panx channels are not sensitive to extracellular divalent cations
indicates that GJC abruptly open and close in a manner sim- and may open at resting membrane potential (Scemes et al.
ilar to that of other ion channels (Bennett et al. 1991; Harris 2009) a property that is also found for Cx HCs activated by
2001). Each of the different Cx isoforms “produces GJCs proinflammatory treatment (Retamal et al. 2007).
with distinct unitary conductance, molecular permeability,
and electrical and chemical gating sensitivities” (Harris 2001).
2.5 P H A R M AC O L O GY A N D G E N ET I C TO O L S
For example, the unitary conductance of GJCs varies from
15 pS to greater than 300 pS and charge selectivity varies from Great effort has been applied to the discovery and character-
slightly anion- to highly cation-selective. The rank order of ization of molecules and pharmacological strategies to selec-
unitary conductance, highest to lowest, according to Cx com- tively block Cx and Panx channels ( Juszczak and Swiergiel
position of homomeric channels is: Cx37 > Cx40 = Cx46 > 2009; Rozental et al. 2001a), with mixed results (Ye et al.
Cx43 = Cx26 > Cx32 > Cx45. GJCs formed by distinct Cxs 2009). Long chain alcohols, such as octanol and heptanals,
have different limiting pore diameters and differ in their per- and volatile anesthetics, such as halothane, block GJCs in
meability to cytoplasmic molecules, but not in a manner con- glial cells (Dermietzel et al. 1991; Giaume et al. 1991; Mantz
sistently predicted by pore size (Harris 2007). For example, et al. 1993). However, these compounds are not specific and
Cx43 channels are much more permeable to ATP and ADP unwanted side effects have been reported including neurotox-
than Cx32 channels, but Cx43 and Cx32 channels are simi- icity (see Juszczak and Swiergiel 2009; Rozental et al. 2001b).
larly permeable to glutamate and glutathione (Harris 2001). The most extensively used GJC inhibitor is carbenoxolone
This suggests that the pore may “chemically” interact with (CBX), a water-soluble glycyrrhetinic acid derivative, that
cytoplasmic molecules in a manner that selectively affects rapidly (<1 minute) and reversibly blocks astrocyte GJCs
permeability. (Meme et al. 2009). Unfortunately, this compound alters
The permeability of GJCs is influenced by physiological neuronal properties by affecting voltage-dependent potas-
variables, including transjunctional voltage, intracellular ionic sium (Rouach et al. 2003) and calcium channels (Vessey et al.
concentrations, protein kinases and phosphatases, eicosanoids 2004), swelling activated anion channels (Ye et al. 2009) and
and nitric oxide. Junctional conductance is maximal when NMDA-evoked currents (Chepkova et al. 2008). Mefloquine

294 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
low Ca2+. +Vm, quinine, dephosphorylation
A A
increased ROS, cytoplasmic signals.
Closed Open
high Ca2+, Mg2+, Ba2+, Sr2+, Ni2+, La3+, Gd3+, H+,
–Vm, phosphorylation, most gap junction blockers.

Pannexin channel
Connexin gap junction channel
B C
1.0 4 DCFS + EGTA Connexin hemichannel
glutamate release
LY loading or

glutamate release

0.8 LY
3 Ctrl
B
0.6 CBX
Glu 2
0.4
A A
0.2
1
0.0
0.0 1 10 100 0 10 20 30 40
carbenoxolone (μM) minutes

Figure 24.2 Features of Cx Hemichannels. A. Cartoon showing some of


compact myelin
the factors that influence gating of HCs. B. Astrocytes show LY loading
and glutamate release in divalent cation free solution (DCFS), which is
blocked in a concentration dependent manner by carbenoxolone (CBX). periaxonal space
C. Glutamate is released from freshly isolated adult mouse optic nerve Cx30 Cx43 Cx32 Cx47 Cx29 juxtaparanode
paranode
in DCFS (plus EGTA). Consistent with release from HCs, release is
node
blocked by CBX, a gap junction blocker. (A) Modified from Saez
et al. 2003b; (B) Modified from Ye et al. 2003.
Figure 24.3 Summary of Cx and Panx Channels in and Between
Glial Cells and Neurons. A. The size of the cell symbol represents
the magnitude of protein expression and functional communication
between the different cell types (A: astrocytes; O: oligodendrocytes; M:
at low concentration (3 μM) completely blocks Cx36 GJCs microglial cells; N: neurons). The black symbols refer to Cx-based HCs
and has no effect on Cx26, Cx32, and Cx43 GJCs; at higher and white symbols to Panx channels. Question marks indicate where the
concentration (30 μM) it inhibits almost completely Cx43 contribution of Cx or Panx channels are either controversial or remain
to be established. B. Diagram of the distribution and Cx-composition
GJCs (Cruikshank et al. 2004). Interestingly, Panx1 channels of GJCs and HCs in brain white matter. Note the presence of “reflex-
are 1,000- to 10,000-fold more sensitive to mefloquine than ive” GJCs between the oligodendrocyte paranodal loops. From
Cx43 GJCs (Cruikshank et al. 2004; Iglesias et al. 2008). Orthmann-Murphy et al. 2007, with permission.
Not surprisingly, HCs are sensitive to many of the regula-
tory factors and compounds listed for GJCs. However, unlike
GJCs, the extracellular loops of HCs are far more accessible
pharmacological agents is the specificity for selected Cxs
to extracellular drugs. For instance, when applied extracellu-
and defined cell types. Genetically altered mice deficient for
larly La3+ blocks HCs but not GJCs in astrocytes (Contreras
all the main glial Cxs as well as Panx1 and Panx2 are pres-
et al. 2002). The sensitivity of Panx channels to CBX occurs
ently available. Because global deficiency for Cx26 and Cx43
at lower concentrations (5- to 20-fold less) than with Cx
is lethal, conditional knock-out mice have been engineered
HCs, and niflumic acid inhibits Cx HCs but does not affect
whereby cell-type restricted deletion is achieved using the
Panx channels (Bruzzone et al. 2005), whereas probenecid
Cre/loxP system. A further improvement is mouse lines
does the opposite. Panx1 forms a protein–protein associa-
permitting timed gene inactivation of Cxs and Panxs to
tion with purinergic P2X7 receptors. When astrocyte P2X7
avoid the confounding factor of developmental disturbances
receptors are activated by adenosine triphosphate (ATP), a
(Giaume and Theis 2010).
cationic channel opens within milliseconds followed seconds
later by activation of a large pore permeable to molecules up
to 900 Da (Pelegrin and Surprenant 2006; Surprenant et al. 3 EXPRESSION OF CONNEXINS AND
1996). Consequently, antagonists of P2X7 receptors such PA N N E X I N S I N G L I A L C E L L S
as brilliant blue G also affect Panx channels (Qiu and Dahl
2009). 3.1 D I S T R I BU T I O N A N D A NATO MY
The major advantage of genetically modified animals Gap junction coupling and Cx expression in mammalian
with deletion of gap junction proteins compared with glial cells is summarized in Table 24.1 (see also Fig. 24.3).

GAP JUNCTIONS AND HEMICHANNELS • 295


The ultrastructure of glial gap junctions are similar to what is could enhance coupling (Ransom and Sontheimer 1992). In
described in other cells. Astrocytes are the most robustly cou- contrast with the effect of neurons, brain macrophages reduce
pled glial cells and form homotypic GJCs with other astro- Cx43 expression in astrocytes (Faustmann et al. 2003; Rouach
cytes and heterotypic junctions with oligodendrocytes and et al. 2002). Other naturally occurring molecules, including
ependymal cells. Astrocytes, as well as other glial cells, express endothelins and proinflammatory cytokines (Duff y et al.
several different Cxs that are regulated during development 2000; Meme et al. 2006), have also been shown to downregu-
(Kunzelmann et al. 1999) and affected by injury (Saez et al. late astroglial GJC expression and function.
2003a). There are some reports indicating that astrocytes form
GJCs with neurons (Alvarez-Maubecin et al. 2000; Froes et al.
3.1.2 Oligodendrocytes
1999). Except in the case of coupling between Purkinje neu-
rons and Bergmann glia (Pakhotin and Verkhratsky 2005), Based on ultrastructural studies, few gap junctions are found
these junctions may occur only during early developmental between oligodendrocytes (O:O) in rat brain, although they
stages (Rash et al. 2001). are abundant between astrocytes and oligodendrocytes (A:O),
suggesting that GJC communication between oligodendro-
cytes is mediated by A:O connections (see Fig. 24.3) (Rash
3.1.1 Astrocytes et al. 2001) (see chapter 62). However, O:O gap junctions
Mammalian astrocytes in both gray and white matter show occur between oligodendrocytes, and coupling is seen in vitro
widespread dye-coupling with each other (Butt and Ransom (Ransom and Kettenmann 1990; Von Blankenfeld et al. 1993)
1993; Gutnick et al. 1981). Dye injected into a single corti- and in situ (Butt and Ransom 1993; Robinson et al. 1993). This
cal astrocyte stains as many as 100 adjacent cells. Astrocytes apparent conflict might be explained by recent studies show-
express Cx43, Cx30, and possibly Cx26 (Lynn et al. 2011; ing marked regional differences in O:O and O:A coupling:
Wasseff and Scherer 2011). Interestingly, Cx30/Cx30 and The former predominates in white matter and the latter in gray
Cx43/Cx43 form functional channels, whereas Cx30/Cx43 matter (Maglione et al. 2010; Wasseff and Scherer 2011). The
does not (Orthmann-Murphy et al. 2007). Connexin43 is significance of regional differences in intercellular coupling
predominant, but its expression varies by brain region and patterns is not known. In corpus callosum slices from animals
stage of development. White matter astrocytes express mini- older than 2 weeks, the majority of injected oligodendrocytes
mal or no Cx30 (Nagy et al. 1999) and are less coupled than show coupling to other glial cells, mainly oligodendrocytes,
those from the gray matter (Maglione et al. 2010). Coupling but also some astrocytes. Direct O:O coupling was confirmed
between astrocytes is reduced by half in cells lacking Cx43 by persistent O:O coupling in animals lacking the astrocyte-
and abolished when both Cx43 and Cx30 are absent (Rouach specific connexins Cx43 and Cx30 (Maglione et al. 2010).
et al. 2008; Roux et al. 2011; Wallraff et al. 2006). Connexin43 The paranodes of oligodendrocytes have Cx32 homomeric
appears immediately after birth, whereas Cx30 develops sev- reflexive junctions, that is, GJCs between compartments of
eral weeks after birth (Kunzelmann et al. 1999). Patch-clamp a given cell (see Fig. 24.3B). Oligodendrocytes express Cx29,
recordings of pairs of coupled astrocytes (Giaume et al. 1991) Cx32, and Cx47 (see Table 24.1). By electrophysiological test-
reveal a unitary conductance with similar biophysical prop- ing, Cx pairs that could mediate O:O coupling are Cx32/
erties to Cx43 GJCs described in other tissues (Spray and Cx32 and Cx47/Cx47, while the heterotypic pair Cx30/Cx47
Burt 1990). Cultured astrocytes express Cx43, whereas Cx30 is not functional (Orthmann-Murphy et al. 2007). Current
appears only after 10 weeks, however, the addition of neurons evidence is in conflict about whether Cx32/Cx32 GJCs are
induces expression of Cx30 after a week (Koulakoff et al. 2008). functionally more important than Cx47/Cx47 GJCs in medi-
Neurotransmitters, endogenous active molecules (peptides, ating O:O coupling (Maglione et al. 2010; Wasseff and Scherer
endocannabinoids), and second messenger pathways regulate 2011). There is complete agreement, however, that the absence
gap junctional communication between astrocytes (for review of Cx32 and Cx47 eliminates O:O coupling. As to A:O cou-
see Giaume et al. 2010). In cultured astrocytes dye-coupling is pling, the pairings that produce functional GJCs are Cx30/
increased by application of glutamate (Enkvist and McCarthy Cx32 and Cx43/Cx47. Interestingly, the most abundant Cxs
1994), but in acute cerebellar slices glutamate, through the in these two cell types, Cx43 and Cx32, do not pair with each
activation of D-amino-3-hydroxy-5-methylisoxazole-4- other (Elfgang et al. 1995).
propionic acid (AMPA) receptors, reduces ionic coupling
between Bergmann glial cells (Muller et al. 1996). Glutamate 3.1.3 Schwann Cells
may contribute to neuronal activity–dependent control of
Cx43 expression in astrocytes with important consequences Proliferating Schwann cells, during early development, or after
for astrocyte coupling (Rouach et al. 2000). High [K+]- peripheral nerve crush injury, primarily express Cx46 and are
induced upregulation of astroglial GJCs involves the calmod- coupled to one another. Cytokines may be involved in the
ulin kinase pathway (De Pina-Benabou et al. 2001) and also injury-related switch from Cx32 to Cx46 (Chandross 1998).
occurs in the olfactory bulb, where Cx30 GJCs are regulated Nonmyelinating Schwann cells in vitro exhibit dye-coupling
by neuronal activity (Roux et al. 2011). Glutamate or high [K+] that disappears when they begin to make myelin (Konishi
solution causes membrane depolarization, and this might be 1990). Myelinating Schwann cells stop expressing Cx46 but
linked to changes in coupling via changes in intracellular pH. increase expression of Cx32, which is confined to paranodal
Depolarization produces an alkaline shift in astrocytes that regions and Schmidt-Lanterman incisures. At these sites

296 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
“reflexive” gap junctions connect the paranodal folds, facilitat- not universally coupled; using the hGFAP-eGFP mouse, some
ing ion and small molecule movements to and from the tight, eGFP positive astrocytes located within the coupling domain
periaxonal space (Balice-Gordon et al. 1998). The X-linked are not dye coupled hinting at the possibility that astrocytes
form of the hereditary neuropathy known as Charcot-Marie- may couple in a way that is purposeful and not dictated solely
Tooth disease is associated with mutations in the Cx32 by proximity (Giaume et al. 2010; Houades et al. 2006).
gene (Bergoffen et al. 1993). In this condition, Cx32 is also Selectivity in astrocyte coupling is also supported by double
depressed in the brain but oligodendrocytes appear normal as patch-clamp recordings in hippocampal slices, in which 18%
they express Cx29 (Altevogt et al. 2002). These observations of neighboring pairs were not coupled (Meme et al. 2009).
indicate that Cxs expressed in Schwann cells play a critical role Finally, in brain areas characterized by strong anatomical-
in peripheral nerve integrity and function. functional compartments, astroglial networks may overlap
with functional units of neurons such as in the somatosensory
cortex (Houades et al. 2008) and the glomerular layer of the
3.1.4 Microglial Cells
olfactory bulb (Roux et al. 2011).
Microglia in their resting state express little or no Cx. Activated
microglia, however, express Cx43 and show dye coupling
4.2 R EGU L AT I O N O F C O N N E X I N E X P R E S S I O N
in vitro (Eugenin et al. 2001); however, so far there is no in situ
AND CHANNEL FUNCTION
evidence of GJC-mediated coupling. Interestingly, activated
microglia can downregulate Cx43 expression and functional Understanding how Cx expression and function are regu-
coupling in nearby astrocytes (Faustmann et al. 2003), suggest- lated by normal and pathological brain environments is of
ing that astrocyte communication may be reduced at sites of fundamental importance. Glial Cx channels are regulated
brain injury in which microglia would assume their activated by a number of releasable bioactive molecules, and their
phenotype. Expression of Cx43 and functional GJCs can be expression is modified by brain pathologies (Giaume et al.
induced among cultured microglia by interferon-J plus tumor 2010). Indeed, like in other tissues, in the brain Cx-based
necrosis factor-D. Induction of functional GJCs between communication is subject to long- and short-term regula-
microglia is blocked in Cx43-deficient mice (Eugenin et al. tion (Bruzzone et al. 1996). Long-term regulation occurs
2001). Amyloid E peptide induces expression of Cx43 and over hours or days, and operates at the transcriptional level.
Panx1 in microglia within 72 hours. Microglia show enhanced This is likely associated with changes in the number of junc-
HC activity with release of glutamate and ATP that initiates tional plaques and/or GJCs. Moreover, changes in the rate
a pathological sequence leading to neuronal injury (Orellana of gap junction internalization and Cx degradation may also
et al. 2011b). This is hypothesized to be an important step in occur. Short-term regulation occurs in minutes and deals
the pathogenesis of Alzheimer disease. with changes in opening probability, time of opening/clo-
sure, and/or unitary conductance of functional GJCs already
in place. Interestingly, Cx expression in astrocytes is a target
4 O R G A N I Z AT I O N O F C O U P L E D for a number of neurotransmitters, growth factors, peptides,
G L I A L AG G R E G AT E S cytokines, and endogenous bioactive lipids, most of them
associated with intracellular signaling pathways (Giaume
Astrocytes coupled together by GJCs were initially assumed et al. 2010). This indicates that glial networks are subject to
to form a nonspecific glial syncytium (Mugnaini 1986; Theis plasticity being tightly modulated by neuronal products and
et al. 2005), but recent evidence suggests they are organized in secretions from other brain cells types, such as glia (astrocytes,
a more complex and restricted manner forming networks of microglia), endothelial cells, and inflammatory cells such as T
communicating cells with rules governing their functional sta- cells (Eugenin et al. 2001). An example of activity-dependent
tus and plasticity (Giaume and McCarthy 1996; Giaume et al. plasticity of astroglial networks was demonstrated in olfac-
2010). This feature has functional consequences depending on tory glomeruli in which extracellular K+ plays an essential
the nature of the signals that are exchanged through GJCs. role (Roux et al. 2011).

4.1 G L I A L N ET WO R K S 4.2.1 What Passes Through Astroglial Gap


Dye injected into a single astrocyte in the cortex, hippocam- Junction Channels
pus, olfactory bulb, and optic nerve diffuses via GJCs to other To understand the functions of glial GJCs, we must know what
cells, 80% to 95% of which are astrocytes, identified by spe- substances pass through them. Ions clearly pass through GJCs,
cific astrocyte markers or by expression of a fluorescent pro- and they have long been thought to contribute to K+ home-
tein in genetically altered mice (Binmoller and Muller 1992; ostasis during neuronal activity (Kuffler and Nicholls 1966;
Blomstrand et al. 2004; Butt and Ransom 1993; Houades Wallraff et al. 2006), and assist in the intracellular homeostasis
et al. 2008; Rouach et al. 2008; Roux et al. 2011; Schools et al. of Na+ (Rose and Ransom 1997). Biochemical coupling also is
2006). This established the notion of astroglial “networks” likely to be very important in glial functions and is expected
(Giaume and McCarthy 1996; Giaume et al. 2010), although to depend on Cxs identity. At present, however, most of the
weak coupling to oligodendrocytes and, very rarely, to neurons information available comes from astrocytes, or cell lines
may be seen (see the preceding). Interestingly, astrocytes are selectively expressing Cx43, the most common astrocyte Cx.

GAP JUNCTIONS AND HEMICHANNELS • 297


Signaling molecules that pass through these GJCs are listed open under normal physiological conditions, or be gated open
in Table 24.2. in a controlled manner during normal activity, for instance, by
Interestingly, in astrocytes the high number of GJCs can decrease in extracellular Ca2+ (Torres et al. 2012). The implica-
be used as a tool to deliver pharmacological or molecular tions for brain physiology, ranging from synaptic transmission
agents in a specific manner to a group of communicating cells. to development, are obvious but remain largely unexplored.
Whole-cell recording of a single astrocyte allows dialysis of
molecules included in the patch pipette and these can diffuse
5.1 A S T RO C Y T E S
within the network. This strategy was used to deliver the Ca2+
chelator BAPTA, showing that calcium signaling in astro- Normally, HCs open when extracellular divalent cations are
glial networks affects hippocampal heterosynaptic depression removed (Ye et al. 2003), but opening can occur in the presence
(Serrano et al. 2006). Likewise, IP3 delivered via the coupled of divalent cations during metabolic inhibition (Contreras
astrocyte network induced glutamate release, triggering tran- et al. 2002), after oxygen and glucose deprivation (Orellana
sient depolarizations, and epileptiform discharges in CA1 et al. 2011b) or after exposure to proinflammatory cytok-
pyramidal neurons (Kang et al. 2005). ines (Karpuk et al. 2011; Orellana et al. 2011a; Retamal et al.
2007). In addition, the activity of HCs composed of Cx43 or
Cx45, but not Cx26, is enhanced by FGF-1 mainly owing to
5 HEMICHANNELS IN GLIAL CELLS an increase in the surface level of HCs mediated by a rise in
intracellular Ca2+ concentration (Schalper et al. 2008). The
Unlike GJCs, Cx HCs are directly exposed to extracellu- patterns of Cx expression and activation in glia are modified
lar ions and are gated by extracellular, as well as intracellular, by pathological conditions (Koulakoff et al. 2012; Orellana
ionic changes. A wide range of factors influence HC gating et al. 2009). These changes depend on the nature and sever-
(Contreras et al. 2002; Saez et al. 2003b; Ye et al. 2003) (see Fig. ity of insult, distance from the lesion site, and elapsed time.
24.2). Open HCs are detected by their “signature” unitary con- Both deleterious and protective effects have been reported, in
ductance and uptake or release of appropriate marker dyes, and particular after ischemia, stroke, or trauma. However, in most
both events should be blocked by GJC blockers. Hemichannels studies, the relative contribution of GJCs and HCs was not
have been primarily studied in cultured cells, but functional discriminated because, in general, the pharmacological agents
HCs are seen in acute brain slices (Karpuk et al. 2011; Orellana used block both channel types (see the preceding). In neu-
et al. 2011a) and in situ (Ye et al. 2003). Pathological condi- rodegenerative diseases, changes in astroglial Cx expression
tions may predispose HC activation (Contreras et al. 2002). It have been observed, but changes in their channel functions,
also seems likely that a small fraction of these channels may be HCs, and/or GJCs, have been tested in only a few of them

Table 24.2 WHAT PASSES THROUGH ASTROGLIAL GAP JUNCTION CHANNELS


BLOCK OF GAP
JUNCTIONAL
CELL MODELS TECHNIQUES PERMEANT MOLECULES COMMUNICATION REFERENCES

Cultured cortical astrocytes SL/DT, radiolabeled Glucose, Octanol, Tabernero et al. 1996
compounds Glucose-6-phosphate, beta-glycyrrhetinic acid,
lactate Arachidonic acid,
Endothelin-1

Cultured cortical astrocytes SL/DT, radiolabeled Glutamate, Octanol Giaume et al. 1997
compounds glutamine

Cultured cortical astrocytes Calcium imaging Inositol-triphosphate NT Leybaert et al. 1998

C6 transfected with Cx43 Cell viability Unidentified death signals Nontransfected C6 cells Lin et al. 1998
co-cultured with astrocytes (TUNEL)
ATP cell content

C6 transfected with Cx43 Capture of radiolabeled ADP, ATP, Nontransfected C6 cells Goldberg et al. 1999
compounds Glutathione
Glutamate

C6 transfected with Cx43 Layered culture system, ATP, ADP, AMP, glutathione, Nontransfected C6 cells Goldberg et al. 2002
radiolabeled compounds glutamate, glucose

Cultured hippocampal SL/DT 2-NBDG Carbenoxolone, Blomstrand and


astrocytes Endothelin-1 Giaume 2006

Hippocampal acute slices Patch-clamp loading Inositol-triphosphate NT Kang et al. 2005


Hippocampal acute slices Patch-clamp loading 2-NBDG Cx43 KO, Cx30 KO, Rouach et al. 2008
6-NBDG carbenoxolone

298 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
(Koulakoff et al. 2012). Interestingly, proinflammatory cytok- from analysis of diseases associated with Cx mutations (this
ines (i.e., IL-1E and TNF-D) increase function of Cx43 HCs large topic is beyond the scope of our review, for example, see
and simultaneously decrease function of Cx43 GJCs (Retamal Abrams and Scherer 2011; Koulakoff et al. 2011; Saez et al.
et al. 2007). Recently, an opposite effect has also been reported 2003b) and from studies on specific Cx knock-out.
following Staphylococcus aureus infection in the striatum with
uncoupling and Cx43 HC/Panx1 channel activation, depend-
6.1 [ K + ] O H O M E O STA SI S
ing on time after injection and location within the infected
area (Karpuk et al. 2011). Panx1 was found in cultured astro- Neural activity generates transient increases in [K+]o that
cytes (Bianco et al. 2009; Iwabuchi and Kawahara 2009) and would disrupt orderly information processing if there are no
in situ (Santiago et al. 2011). Activation of P2X7 receptors in mechanisms to minimize these increases. The “spatial buffer
these cells induces ATP release via Panx1 channels, but not hypothesis” states that glial cells participate in moving extra-
Cx43 HCs (Iglesias et al. 2009). In contrast, cultured astro- cellular K+ from areas in which it accumulates secondary to
cytes subjected to hypoxia-reoxygenation or treated with
proinflammatory cytokines or the amyloid peptide exhibit HC A Homeostasis
activity mediated by Cx43, but not Panx1 (Froger et al. 2010;
Orellana et al. 2010; Retamal et al. 2007). To make matters Mem. Pot. Mem. Pot. Mem. Pot.
more complicated, two recent studies report Cx43 and Panx1 [Na+] [Na+] [Na+]
HC activity in astrocytes provoked by fibroblast growth fac- [Ca2+] [Ca2+] [Ca2+]
[K+] [K+] [K+]
tor (FGF) in one case (Garre et al. 2010) and proximity to a Amino acids Amino acids Amino acids
brain abscess in the other (Karpuk et al. 2011). At present, it Metabolites Metabolites Metabolites
appears that Panxs and Cxs may both form astrocyte HCs and
channels under certain conditions, the details dependent on
the conditions triggering their activation. Further evidence of B stimulus Response amplification
Cx43 HC expression in astrocytes is provided by the obser- receptor
vation that HC-mediated ATP release persists in the Panx1/ cAMP cAMP
cAMP
Panx2 double knock-out mouse (Bargiotas et al. 2011). IP3 IP3 IP3
Ca2+ Ca2+ Ca2+

5.2 M I C RO G L I A Response Response Response


In vivo, only 5% of resting microglia express low levels of Cx43.
Brain trauma in the form of a stab wound robustly induces Cx43
expression in microglia (Eugenin et al. 2003). Cultured microglia C Signaling: astrocyte calcium waves stimulus
(e.g. glutamate)
express little or no Cx43 (Meme et al. 2006; Rouach et al. 2002),
but can be induced to do so by certain proinflammatory agents glutamate
or the amyloid peptide, leading to glutamate release via HCs receptors
(Takeuchi et al. 2006). The resultant neurotoxicity is inhibited glutamate
release PLC
by CBX (D’Hondt et al. 2009). Treatment of microglia with PLC
ATP
amyloid peptide induces Cx43 and Panx1 HCs that release ATP IP3
and glutamate (Orellana et al. 2011b). Over time, the microglia- [Ca2+]i Ca2+
altered
mediated neurotoxicity is progressive and could contribute to excitability Ca2+
the progressive nature of diverse neurodegenerative diseases. astrocyte stores
neuron

6 FUNCTIONS OF GLIAL GAP Figure 24.4 Schematic Illustration of Possible Functions of GJCs and
JUNCTION CHANNELS AND HCs. A. In strongly coupled cell aggregates, the intracellular concentra-
HEMICHANNELS tions of ions and molecules less than approximately 1,000 kDa (including
amino acids, sugars, and second messenger molecules) will equilibrate
because of free exchange across gap junctions. B. Coupling can amplify
Several functions have been proposed for GJCs based on their the physiological consequences of a chemical stimulus (i.e., hormone,
physiological properties, and this list is growing (Bennett et al. neurotransmitter) acting on a single cell. Such stimuli often cause the
1991; Harris 2001; Loewenstein 1981; Saez et al. 2003b). They production of second messenger molecules (i.e., Ca2+, IP3, cAMP) that
may serve to coordinate the electrical and metabolic activities can move easily into adjacent cells via gap junctions; the coupled cells
are then recruited to respond. C. GJCs and HCs participate in astrocyte
of cell populations, act to amplify the consequences of signal Ca2+ waves. Astrocytes can be provoked to exhibit Ca2+ waves in many
transduction, control intrinsic proliferative capacity, and help ways, such as the application of glutamate. Effective stimuli activate phos-
to orchestrate the complex events of embryonic morpho- pholipase C (PLC) and inositol (1,4,5)-triphosphate (IP3) dependent
genesis (Fig. 24.4). In addition, Cxs can have functions in pathways leading to Ca2+ release from Ca2+ stores. IP3 and Ca2+ can
astrocytes that are unrelated to channel formation such as cell diffuse to adjacent cells through GJCs. Astrocytes also release ATP on
activation, probably through HCs. In turn, ATP activates purinergic
adhesion (Elias et al. 2007), modulation of purinergic recep- receptors on nearby astrocytes and this is the predominant mode by
tor responses (Scemes 2008) and influencing cell survival (Lin which Ca2+ waves spread. Increase in astrocyte [Ca2+]i can cause gluta-
et al. 2003). Further insights about Cx functions are emerging mate release that modulates the excitability of neighboring neurons.

GAP JUNCTIONS AND HEMICHANNELS • 299


neural activity, to distant areas in which the K+ is not elevated the case of a cell stimulated to produce an intracellular sec-
(Orkand et al. 1966). Strong astrocytic coupling would extend ond messenger molecule (e.g., cAMP, IP3, Ca2+) by binding a
the distance over which this K+ distribution system could specific ligand. If the cell is not connected to its neighbors by
operate and, perhaps, increase its efficiency (Mobbs et al. GJCs, it will act in isolation, but if the cell is widely coupled to
1988). Glial cells also minimize the extent of K+ accumula- other cells, the diffusible, GJC-permeable second messenger
tion occurring with neural activity by uptake and sequestra- has the potential of affecting many cells (Loewenstein 1981).
tion of K+ (Rose and Ransom 1996). Glial coupling might Traveling waves of increased cytoplasmic [Ca2+] are elicited
benefit this process by creating a functionally larger intracel- in astrocytes by a variety of stimuli including glutamate appli-
lular volume that would tend to minimize changes in intracel- cation (Cotrina et al. 1998; Finkbeiner 1992). As the wave
lular K+ concentration that occur with K+ uptake. Coupling passes through astrocytes it can elicit glutamate release and
between astrocytes and oligodendrocytes (i.e., A:O) might also alter the behavior of adjacent neurons; in essence this event
participate in [K+]o homeostasis. In fact, K+ may accumulate is a slow form of signaling (Haydon 2001). Ca2+ wave propa-
to very high levels in the paranodal area. Astrocyte processes gation is dependent on both GJCs and extracellular signal-
that project to nodes of Ranvier, called perinodal astrocytes, ing pathways, including HC-mediated ATP release (Cotrina
are frequently connected by gap junctions to the terminal et al. 1998; Giaume and Venance 1998; Guthrie et al. 1999);
paranodal loops of nearby myelinating oligodendrocytes the relative contribution of these two mechanisms may vary
(see Fig. 24.3B). This specialized arrangement could medi- in different brain regions (Scemes and Giaume 2006) (see
ate an axon-specific type of K+ spatial buffering. The impor- Fig. 24.4C). Recent in vivo confirmation of intercellular cal-
tance of A:O coupling for the integrity of myelinated axons is cium waves in astrocytes has generated heightened interest
undeniable based on the pathological changes in myelin in this mode of signaling between glia in normal (Hoogland
seen when A:O coupling is genetically altered (Abrams and et al. 2009; Kuga et al. 2011) as well as in pathological situa-
Scherer 2011). tions (Kuchibhotla et al. 2009).

6.2 I N T R AC E L LU L A R M ETA B O L I C 6.4 P RO L I FE R AT I VE AC T I VIT Y


T R A F F I C K I N G A N D H O M E O S TA S I S A N D D EV E L O PM E N T
Glial gap junctions may be important for metabolic coordina- The role of gap junctional communication during develop-
tion within the coupled aggregate. For example, coupled astro- ment has been discussed at length (Levin 2002). Embryos
cytes tend to have similar intracellular [Na+] ([Na+]i), but [Na+]i exhibit widespread gap junctional communication, which may
diverges between neighboring cells when coupling is blocked be important in establishing developmental “fields” critical for
(Rose et al. 1997). Gap junctions, therefore, can cause a group morphogenesis. Coupling in glia, however, develops as these
of individual cells to function almost as one in terms of metab- cells mature.
olism and membrane potential (see Fig. 24.4A). This offers Considerable evidence indicates that gap junctions can
a theoretical advantage for the operation of the high-affinity restrict cell proliferation. Most primary brain tumors derive
glutamate uptake system in glia. The rate of glutamate uptake from glial cells, and the possibility that loss of gap junctions
is affected by the transmembrane gradients of glutamate and contributes to their neoplastic transformation is intriguing.
the ions that are cotransported or countertransported. Strong The absence of gap junctions in C6 glioma cells and highly
glial coupling, by providing a larger effective intracellular vol- malignant human gliomas (Tani and Higashi 1973) supports
ume, prevents large changes in the intracellular concentration this concept. Moreover, proliferation is reduced in glioma cells
of the transmitter and associated ions from occurring with induced to express Cx43 (Sanchez-Alvarez et al. 2001). How
uptake, maximizing the efficiency of this process. gap junctions suppress mitotic activity is not clear; however,
The energy substrate glucose and its metabolites, including Cx43 can act as a substrate of c-Src reducing the rate of glioma
lactate, can pass through GJCs between astrocytes and allow cell proliferation (Herrero-Gonzalez et al. 2010).
astroglial networks to serve as a supply line for neurons under
certain circumstances (Blomstrand and Giaume 2006; Rouach
6.5 R EGU L AT I O N O F SY NA P T I C AC T I VI T Y
et al. 2008). Infusion of glucose and lactate within an astroglial
population was shown to sustain neuronal activity in the absence Astrocytes dynamically interact with neurons to regulate
of external glucose (Rouach et al. 2008). These observations sup- synaptic transmission (Halassa and Haydon 2010), and astro-
port the idea that astrocytes support neuron energy metabolism cyte coupling may play a role in this process. Inactivation of
by providing lactate to them (Pellerin and Magistretti 2011) (see the Cx30 and Cx43 genes increase hippocampal synaptic
chapter 27). Aspects of this process seem to depend on GJCs transmission and impairs long-term synaptic plasticity in
and are disrupted by GJC inhibitors and in the Cx43–/–/Cx30– CA1 pyramidal neurons due to astrocyte hypertrophy (Fig.
/–
double knock-out mouse (Rouach et al. 2008). 24.5) (Pannasch et al. 2011). Indeed, in normal conditions
the astroglial network facilitates glutamate and K+ removal
during synaptic activity by regulating astrocyte contribution
6.3 S I G NA L T R A N S FE R
to the rate of clearance and extracellular space volume. Thus,
Another functional consequence of intercellular coupling is astroglial Cxs play an important role in the control of synap-
the divergence of signal transmission (see Fig. 24.4B). Consider tic strength by modulating extracellular homeostasis. These

300 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Glial Cx HCs have been implicated in paracrine and auto-
crine cellular communication in several normal and patholog-
ical conditions (Saez et al. 2010). Many brain diseases, both
acute and chronic, induce variable degrees of inflammation,
and some general features of this process are emerging. Both
C +/+ –/– D RESTING STATE astrocytes and microglia participate in this pathological cas-
TMA+-ISM cade and can respectively exhibit Cx HC and Panx1 channel
activation leading to neurotoxicity. Thus, these cells can be
Neuron
Glia
“bad neighbors” for neurons. Normalization of Cx- and Panx-
–0.5
based channel dysfunctions should confer tissue protection,
fEPSP slope (mV/ms)

ACTIVITY
–0.4 and this goal should be considered a future therapeutic target
–0.3 (Orellana et al. 2011c).
–0.2
–0.1
0.0 7 S U M M A RY A N D P E R S P E C T I VE S
0.05 0.10 0.15 0.20 0.25 0.30
Fiber volley amplitude (mV)
Connexins and the special forms of transmembrane commu-
Figure 24.5 Hippocampal Synaptic Activity Is Enhanced in the Absence
of Astrocyte Cxs. A,B. Astrocytes are hypertrophic in the hippocampal
nication that they mediate have been extensively investigated
CA1 region in Cx30 –/–/Cx43–/– mice compared with wild-type. (A) in the half-century since their discovery. This is especially so
Sulforhodamine 101 cell labeling and (B) anti-GFAP immunoreactivity in the brain, in which these channels are widely expressed in
(Scale bars: 10 mm). C. Excitatory postsynaptic field potentials, and their glial cells. Although we lack final insight about how they par-
input/output slope, are enhanced in hippocampal slices from Cx30 –/–/ ticipate in brain functions and pathologies, new information
Cx43–/– mice (upper traces, scale bars: 0.2 mV, 10 ms; empty symbols:
wild-type; filled symbols: Cx30–/–/Cx43–/–). D. Schematic representation
strongly implicates glial cell GJCs and HCs in extracellular
of the changes of extracellular concentrations owing to increased astro- ion homeostasis, neural metabolic support, and disease path-
glial morphology as the result of cell swelling in the Cx30–/–/Cx43–/– ogenesis, both acute and chronic. Research prospects in this
mouse during neuronal activity compared with resting state. Changes area have never been brighter given the powerful experimental
in extracellular space volume were detected by the monitoring of techniques currently available and the intriguing progress of
time-dependent concentration of the extracellular marker tetramethylam-
monium (TMA+) recorded in the stratum radiatum of CA1 region dur-
the last decade.
ing Schaffer collateral stimulations. Modified from Pannasch et al. 2011.

AC K N OW L E D G M E N T S
results help establish glial Cxs as critical proteins for extracel-
lular homeostasis, important for the formation of functional This work was supported by NIH grants NS 15589 (BRR),
synapses. INSERM/CNRS, and ANR grants (CG). The authors wish
to thank N. Rouach and J. A. Orellana for providing them
with illustration material and Kass H. Klemz for her help in
6.6 P O S S I B L E C O N T R I BU T I O N S O F C O N N E X I N
finalizing this chapter.
CHANNELS IN BRAIN DISEASES
In general, in most neurodegenerative diseases and in isch-
REFERENCES
emia changes in Cx43 expression were detected in reactive
astrocytes. Moreover, following brain injury, changes in the Abrams CK, Scherer SS. 2011. Gap junctions in inherited human disorders
level of Cx expression are dependent on the proximity of the of the central nervous system. Biochim Biophys Acta 1818:2030–2047.
reactive astrocytes to the damaged site. What are the conse- Ahn M, Lee J, Gustafsson A, Enriquez A, Lancaster E, Sul JY, et al.
quences of such altered Cx expression on the status of astro- 2008. Cx29 and Cx32, two connexins expressed by myelinating
glial networks? In situ models start to provide expression and glia, do not interact and are functionally distinct. J Neurosci Res
86:992–1006.
function correlation for astroglial Cxs (Koulakoff et al. 2008; Altevogt BM, Kleopa KA, Postma FR, Scherer SS, Paul DL. 2002.
Ochalski et al. 1995; Theriault et al. 1997). What are the con- Connexin29 is uniquely distributed within myelinating glial cells of the
sequences of astroglial network changes on neuronal func- central and peripheral nervous systems. J Neurosci 22:6458–6470.
tion and/or survival? For that purpose, two strategies have Alvarez-Maubecin V, Garcia-Hernandez F, Williams JT, Van Bockstaele
been adopted. One is a pharmacological approach using GJC EJ. 2000. Functional coupling between neurons and glia. J Neurosci
20:4091–4098.
blocking agents. However, the results obtained are difficult to Balice-Gordon RJ, Bone LJ, Scherer SS. 1998. Functional gap junctions
interpret because of a lack of specificity of these agents. Based in the Schwann cell myelin sheath. J Cell Biol 142:1095–1104.
on these limitations, alternative strategies using Cx knockout Ball KK, Gandhi GK, Thrash J, Cruz NF, Dienel GA. 2007. Astrocytic
mice or molecular tools were developed to examine the role connexin distributions and rapid, extensive dye transfer via gap
played by astroglial Cxs in pathological conditions. Most junctions in the inferior colliculus: implications for [(14)C]glucose
metabolite trafficking. J Neurosci Res 85:3267–3283.
of these experiments concern acute injury (ischemia and Bargiotas P, Krenz A, Hormuzdi SG, Ridder DA, Herb A, Barakat W,
trauma), but so far have led to controversial results in respect et al. 2011. Pannexins in ischemia-induced neurodegeneration. Proc
to the role of Cxs (see chapter 58). Natl Acad Sci U S A 108:20772–20777.

GAP JUNCTIONS AND HEMICHANNELS • 301


Bennett MV, Barrio LC, Bargiello TA, Spray DC, Hertzberg E, Saez JC. Dobrenis K, Chang HY, Pina-Benabou MH, Woodroffe A, Lee SC,
1991. Gap junctions: new tools, new answers, new questions. Neuron Rozental R, et al. 2005. Human and mouse microglia express con-
6:305–320. nexin36, and functional gap junctions are formed between rodent
Bennett MV, Contreras JE, Bukauskas FF, Saez JC. 2003. New roles for microglia and neurons. J Neurosci Res 82:306–315.
astrocytes: gap junction hemichannels have something to communi- Duff y HS, John GR, Lee SC, Brosnan CF, Spray DC. 2000. Reciprocal
cate. Trends Neurosci 26:610–617. regulation of the junctional proteins claudin-1 and connexin43 by
Bergoffen J, Scherer SS, Wang S, Scott MO, Bone LJ, Paul DL, et al. 1993. interleukin-1beta in primary human fetal astrocytes. J Neurosci
Connexin mutations in X-linked Charcot-Marie-Tooth disease. 20:RC114.
Science 262:2039–2042. Elfgang C, Eckert R, Lichtenberg-Frate H, Butterweck A, Traub O,
Bianco F, Colombo A, Saglietti L, Lecca D, Abbracchio MP, Matteoli M, Klein RA, et al. 1995. Specific permeability and selective formation
et al. 2009. Different properties of P2X(7) receptor in hippocampal of gap junction channels in connexin-transfected HeLa cells. J Cell
and cortical astrocytes. Purinergic Signalling 5:233–240. Biol 129:805–817.
Binmoller FJ, Muller CM. 1992. Postnatal development of dye-coupling Elias LA, Wang DD, Kriegstein AR. 2007. Gap junction adhesion is nec-
among astrocytes in rat visual cortex. Glia 6:127–137. essary for radial migration in the neocortex. Nature 448:901–907.
Blomstrand F, Giaume C. 2006. Kinetics of endothelin-induced inhibi- Enkvist MO, McCarthy KD. 1994. Astroglial gap junction communica-
tion and glucose permeability of astrocyte gap junctions. J Neurosci tion is increased by treatment with either glutamate or high K+ con-
Res 83:996–1003. centration. J Neurochem 62:489–495.
Blomstrand F, Venance L, Siren AL, Ezan P, Hanse E, Glowinski J, et al. Eugenin EA, Branes MC, Berman JW, Saez JC. 2003. TNF-alpha plus
2004. Endothelins regulate astrocyte gap junctions in rat hippocam- IFN-gamma induce connexin43 expression and formation of gap
pal slices. Eur J Neurosci 19:1005–1015. junctions between human monocytes/macrophages that enhance
Bruzzone R, Barbe MT, Jakob NJ, Monyer H. 2005. Pharmacological physiological responses. J Immunol 170:1320–1328.
properties of homomeric and heteromeric pannexin hemichannels Eugenin EA, Eckardt D, Theis M, Willecke K, Bennett MV, Saez
expressed in Xenopus oocytes. J Neurochem 92:1033–1043. JC. 2001. Microglia at brain stab wounds express connexin 43
Bruzzone R, Hormuzdi SG, Barbe MT, Herb A, Monyer H. 2003. and in vitro form functional gap junctions after treatment with
Pannexins, a family of gap junction proteins expressed in brain. Proc interferon-gamma and tumor necrosis factor-alpha. Proc Natl Acad
Natl Acad Sci U S A 100:13644–13649. Sci U S A 98:4190–4195.
Bruzzone R, White TW, Paul DL. 1996. Connections with connexins: Faustmann PM, Haase CG, Romberg S, Hinkerohe D, Szlachta D,
the molecular basis of direct intercellular signaling. Eur J Biochem Smikalla D, et al. 2003. Microglia activation influences dye coupling
238:1–27. and Cx43 expression of the astrocytic network. Glia 42:101–108.
Bukauskas FF, Verselis VK. 2004. Gap junction channel gating. Biochim Finkbeiner S. 1992. Calcium waves in astrocytes-fi lling in the gaps.
Biophys Acta 1662:42–60. Neuron 8:1101–1108.
Butt AM, Ransom BR. 1989. Visualization of oligodendrocytes and Froes MM, Correia AH, Garcia-Abreu J, Spray DC, Campos de Carvalho
astrocytes in the intact rat optic nerve by intracellular injection of AC, Neto MV. 1999. Gap-junctional coupling between neurons and
lucifer yellow and horseradish peroxidase. Glia 2:470–475. astrocytes in primary central nervous system cultures. Proc Natl
Butt AM, Ransom BR. 1993. Morphology of astrocytes and oligoden- Acad Sci U S A 96:7541–7546.
drocytes during development in the intact rat optic nerve. J Comp Froger N, Orellana JA, Calvo CF, Amigou E, Kozoriz MG, Naus CC,
Neurol 338:141–158. et al. 2010. Inhibition of cytokine-induced connexin43 hemichan-
Ceelen PW, Lockridge A, Newman EA. 2001. Electrical coupling nel activity in astrocytes is neuroprotective. Mol Cell Neurosci
between glial cells in the rat retina. Glia 35:1–13. 45:37–46.
Chandross KJ. 1998. Nerve injury and inflammatory cytokines modu- Garre JM, Retamal MA, Cassina P, Barbeito L, Bukauskas FF, Saez JC,
late gap junctions in the peripheral nervous system. Glia 24:21–31. et al. 2010. FGF-1 induces ATP release from spinal astrocytes in
Chepkova AN, Sergeeva OA, Haas HL. 2008. Carbenoxolone impairs culture and opens pannexin and connexin hemichannels. Proc Natl
LTP and blocks NMDA receptors in murine hippocampus. Acad Sci U S A 107:22659–22664.
Neuropharmacology 55:139–147. Giaume C, Fromaget C, el Aoumari A, Cordier J, Glowinski J, Gros
Contreras JE, Sanchez HA, Eugenin EA, Speidel D, Theis M, Willecke K, D. 1991. Gap junctions in cultured astrocytes: single-channel cur-
et al. 2002. Metabolic inhibition induces opening of unapposed con- rents and characterization of channel-forming protein. Neuron 6:
nexin 43 gap junction hemichannels and reduces gap junctional com- 133–143.
munication in cortical astrocytes in culture. Proc Natl Acad Sci U Giaume C, Koulakoff A, Roux L, Holcman D, Rouach N. 2010.
SA 99:495–500. Astroglial networks: a step further in neuroglial and gliovascular
Cotrina ML, Lin JH, Alves-Rodrigues A, Liu S, Li J, Azmi-Ghadimi H, interactions. Nat Rev Neurosci 11:87–99.
Kang J, Naus CC, Nedergaard M. 1998. Connexins regulate cal- Giaume C, McCarthy KD. 1996. Control of gap-junctional communica-
cium signaling by controlling ATP release. Proc Natl Acad Sci U S tion in astrocytic networks. Trends Neurosci 19:319–325.
A 95:15735–15740. Giaume C, Orellana JA, Abudara V, Saez JC. 2012. Connexin-based
Cruikshank SJ, Hopperstad M, Younger M, Connors BW, Spray DC, channels in astrocytes: how to study their properties. Meth Mol Biol
Srinivas M. 2004. Potent block of Cx36 and Cx50 gap junction chan- 814:283–303.
nels by mefloquine. Proc Natl Acad Sci U S A 101:12364–12369. Giaume C, Theis M. 2010. Pharmacological and genetic approaches to
De Pina-Benabou MH, Srinivas M, Spray DC, Scemes E. 2001. study connexin-mediated channels in glial cells of the central nervous
Calmodulin kinase pathway mediates the K+-induced increase in system. Brain Res Rev 63:160–176.
Gap junctional communication between mouse spinal cord astro- Giaume C, Venance L. 1998. Intercellular calcium signaling and gap
cytes. J Neurosci 21:6635–6643. junctional communication in astrocytes. Glia 24:50–64.
Dermietzel R, Gao Y, Scemes E, Vieira D, Urban M, Kremer M, et al. Gomez-Hernandez JM, de Miguel M, Larrosa B, Gonzalez D, Barrio LC.
2000. Connexin43 null mice reveal that astrocytes express multiple 2003. Molecular basis of calcium regulation in connexin-32 hemi-
connexins. Brain Res Brain Res Rev 32:45–56. channels. Proc Natl Acad Sci U S A 100(26):16030–16035.
Dermietzel R, Hertberg EL, Kessler JA, Spray DC. 1991. Gap junctions Goodenough DA. 1976. The structure and permeability of isolated
between cultured astrocytes: immunocytochemical, molecular, and hepatocyte gap junctions. Cold Spring Harbor Symp Quant Biol
electrophysiological analysis. J Neurosci 11:1421–1432. 40:37–43.
D’Hondt C, Ponsaerts R, De Smedt H, Bultynck G, Himpens B. 2009. Guthrie PB, Knappenberger J, Segal M, Bennett MV, Charles AC, Kater
Pannexins, distant relatives of the connexin family with specific cel- SB. 1999. ATP released from astrocytes mediates glial calcium waves.
lular functions? BioEssays 31:953–974. J Neurosci 19:520–528.

302 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Gutnick MJ, Connors BW, Ransom BR. 1981. Dye-coupling between glial Lin JH, Yang J, Liu S, Takano T, Wang X, Gao Q, et al. 2003. Connexin
cells in the guinea pig neocortical slice. Brain Res 213:486–492. mediates gap junction-independent resistance to cellular injury.
Halassa MM, Haydon PG. 2010. Integrated brain circuits: astrocytic J Neurosci 23:430–441.
networks modulate neuronal activity and behavior. Annu Rev Physiol Loewenstein WR. 1981. Junctional intercellular communication: the
72:335–355. cell-to-cell membrane channel. Physiol Rev 61:829–913.
Harris AL. 2001. Emerging issues of connexin channels: biophysics fi lls Lynn BD, Tress O, May D, Willecke K, Nagy JI. 2011. Ablation of connexin30
the gap. Q Rev Biophys 34:325–472. in transgenic mice alters expression patterns of connexin26 and con-
Harris AL. 2007. Connexin channel permeability to cytoplasmic mol- nexin32 in glial cells and leptomeninges. Eur J Neurosci 34:1783–1793.
ecules. Progr Biophys Mol Biol 94:120–143. Ma W, Compan V, Zheng W, Martin E, North RA, Verkhratsky A,
Haydon PG. 2001. GLIA: listening and talking to the synapse. Nat Rev Surprenant A. 2012. Pannexin 1 forms an anion-selective channel.
Neurosci 2:185–193. Pflugers Arch 463:585–592.
Herrero-Gonzalez S, Gangoso E, Giaume C, Naus CC, Medina JM, MacVicar BA, Thompson RJ. 2010. Non-junction functions of
Tabernero A. 2010. Connexin43 inhibits the oncogenic activity of pannexin-1 channels. Trends Neurosci 33:93–102.
c-Src in C6 glioma cells. Oncogene 29:5712–5723. Maglione M, Tress O, Haas B, Karram K, Trotter J, Willecke K, et al.
Hoogland TM, Kuhn B, Gobel W, Huang W, Nakai J, Helmchen F, 2010. Oligodendrocytes in mouse corpus callosum are coupled via
Flint J, Wang SS. 2009. Radially expanding transglial calcium waves gap junction channels formed by connexin47 and connexin32. Glia
in the intact cerebellum. PNAS 106:3496–3501. 58:1104–1117.
Houades V, Koulakoff A, Ezan P, Seif I, Giaume C. 2008. Gap Mantz J, Cordier J, Giaume C. 1993. Effects of general anesthetics on
junction-mediated astrocytic networks in the mouse barrel cortex. J intercellular communications mediated by gap junctions between
Neurosci 28:5207–5217. astrocytes in primary culture. Anesthesiology 78:892–901.
Houades V, Rouach N, Ezan P, Kirchhoff F, Koulakoff A, Giaume C. Meme W, Calvo CF, Froger N, Ezan P, Amigou E, Koulakoff A, et al.
2006. Shapes of astrocyte networks in the juvenile brain. Neuron 2006. Proinflammatory cytokines released from microglia inhibit
Glia Biol 2:3–14. gap junctions in astrocytes: potentiation by beta-amyloid. FASEB J
Iglesias R, Dahl G, Qiu F, Spray DC, Scemes E. 2009. Pannexin 1: 20:494–496.
the molecular substrate of astrocyte “hemichannels.” J Neurosci Meme W, Vandecasteele M, Giaume C, Venance L. 2009. Electrical cou-
29:7092–7097. pling between hippocampal astrocytes in rat brain slices. Neurosci
Iglesias R, Locovei S, Roque A, Alberto A, Dahl G, Spray DC, et al. Res 63:236–243.
2008. P2X7 receptor-Pannexin1 complex: pharmacology and signal- Mobbs P, Brew H, Attwell D. 1988. A quantitative analysis of glial cell
ing. Am J Physiol Cell Physiol 295:752–760. coupling in the retina of the axolotl (Ambystoma mexicanum). Brain
Iwabuchi S, Kawahara K. 2009. Oxygen-glucose deprivation-induced Res 460:235–245.
enhancement of extracellular ATP-P2Y purinoceptors signaling for Mugnaini E. 1986. Cell junctions of astrocytes, ependymal and related
the propagation of astrocytic calcium waves. Bio Syst 96:35–43. cells in the mammal central nervous system, with emphasis on
Juszczak GR, Swiergiel AH. 2009. Properties of gap junction blockers the hypothesis of a generalized syncytium of supporting cells. In:
and their behavioural, cognitive and electrophysiological effects: Fedoroff S, Vernadakis A (eds.), Astrocytes. New York: Academic
animal and human studies. Progr Neuro-psychopharmacol Biol Press, pp. 329–371.
Psychiatry 33:181–198. Muller T, Moller T, Neuhaus J, Kettenmann H. 1996. Electrical cou-
Kang N, Xu J, Xu Q, Nedergaard M, Kang J. 2005. Astrocytic gluta- pling among Bergmann glial cells and its modulation by glutamate
mate release-induced transient depolarization and epileptiform receptor activation. Glia 17:274–284.
discharges in hippocampal CA1 pyramidal neurons. J Neurophys Nagy JI, Ionescu AV, Lynn BD, Rash JE. 2003. Connexin29 and con-
94:4121–4130. nexin32 at oligodendrocyte and astrocyte gap junctions and in myelin
Karpuk N, Burkovetskaya M, Fritz T, Angle A, Kielian T. 2011. of the mouse central nervous system. J Comp Neurol 464:356–370.
Neuroinflammation leads to region-dependent alterations in Nagy JI, Ionescu AV, Lynn BD, Rash JE. 2003. Coupling of astrocyte
astrocyte gap junction communication and hemichannel activity. connexins Cx26, Cx30, Cx43 to oligodendrocyte Cx29, Cx32,
J Neurosci 31:414–425. Cx47: Implications from normal and connexin32 knockout mice.
Kettenmann H, Ransom BR. 1988. Electrical coupling between astro- Glia 44:205–218.
cytes and between oligodendrocytes studied in mammalian cell cul- Nagy JI, Patel D, Ochalski PA, Stelmack GL. 1999. Connexin30 in
tures. Glia 1:64–73. rodent, cat and human brain: selective expression in gray matter
Konishi T. 1990. Dye coupling between mouse Schwann cells. Brain Res astrocytes, co-localization with connexin43 at gap junctions and late
508:85–92. developmental appearance. Neuroscience 88:447–468.
Koulakoff A, Ezan P, Giaume C. 2008. Neurons control the expression Ochalski PA, Frankenstein UN, Hertzberg EL, Nagy JI. 1997.
of connexin 30 and connexin 43 in mouse cortical astrocytes. Glia Connexin-43 in rat spinal cord: localization in astrocytes and
56:1299–1311. identification of heterotypic astro-oligodendrocytic gap junctions.
Koulakoff A, Mei X, Orellana JA, Saez JC, Giaume C. 2012. Glial con- Neuroscience 76:931–945.
nexin expression and function in the context of Alzheimer’s disease. Ochalski PA, Sawchuk MA, Hertzberg EL, Nagy JI. 1995. Astrocytic gap
Biochim Biophys Acta 23(2):163–177. junction removal, connexin43 redistribution, and epitope masking at
Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. 2009. excitatory amino acid lesion sites in rat brain. Glia 14:279–294.
Synchronous hyperactivity and intercellular calcium waves in astro- Orellana JA, Figueroa XF, Sanchez HA, Contreras-Duarte S, Velarde V,
cytes in Alzheimer mice. Science 323:1211–1215. Saez JC. 2011a. Hemichannels in the neurovascular unit and white
Kuffler SW, Nicholls JG. 1966. The physiology of neuroglial cells. Ergeb matter under normal and inflamed conditions. CNS Neurol Disord
Physiol 57:1–90. Drug Targets 10:404–414.
Kuga N, Sasaki T, Takahara Y, Matsuki N, Ikegaya Y. 2011. Large-scale Orellana JA, Froger N, Ezan P, Jiang JX, Bennett MV, Naus CC, et al.
calcium waves traveling through astrocytic networks in vivo. 2011b. ATP and glutamate released via astroglial connexin 43 hemi-
J Neurosci 31:2607–2614. channels mediate neuronal death through activation of pannexin 1
Kunzelmann P, Schroder W, Traub O, Steinhauser C, Dermietzel R, hemichannels. J Neurochem 118:826–840.
Willecke K. 1999. Late onset and increasing expression of the gap Orellana JA, Hernandez DE, Ezan P, Velarde V, Bennett MV, Giaume C,
junction protein connexin30 in adult murine brain and long-term et al. 2010. Hypoxia in high glucose followed by reoxygenation in nor-
cultured astrocytes. Glia 25:111–119. mal glucose reduces the viability of cortical astrocytes through increased
Levin M. 2002. Isolation and community: a review of the role of permeability of connexin 43 hemichannels. Glia 58:329–343.
gap-junctional communication in embryonic patterning. Biol Membr Orellana JA, Saez PJ, Shoji KF, Schalper KA, Palacios-Prado N, Velarde V,
Biol 185:177–192. et al. 2009. Modulation of brain hemichannels and gap junction

GAP JUNCTIONS AND HEMICHANNELS • 303


channels by pro-inflammatory agents and their possible role in neu- Rouach N, Koulakoff A, Abudara V, Willecke K, Giaume C. 2008.
rodegeneration. Antioxidat Redox Signal 11:369–399. Astroglial metabolic networks sustain hippocampal synaptic trans-
Orellana JA, Shoji KF, Abudara V, Ezan P, Amigou E, Saez PJ, et al. mission. Science 322:1551–1555.
2011c. Amyloid beta-induced death in neurons involves glial and Rouach N, Segal M, Koulakoff A, Giaume C, Avignone E. 2003.
neuronal hemichannels. J Neurosci 31:4962–4977. Carbenoxolone blockade of neuronal network activity in cul-
Orkand RK, Nicholls JG, Kuffler SW. 1966. Effect of nerve impulses on ture is not mediated by an action on gap junctions. J Physiol
the membrane potential of glial cells in the central nervous system of 553:729–745.
amphibia. J Neurophysiol 29:788–806. Roux L, Benchenane K, Rothstein JD, Bonvento G, Giaume C. 2011.
Orthmann-Murphy JL, Freidin M, Fischer E, Scherer SS, Abrams CK. Plasticity of astroglial networks in olfactory glomeruli. Proc Natl
2007. Two distinct heterotypic channels mediate gap junction cou- Acad Sci U S A 108:18442–18446.
pling between astrocyte and oligodendrocyte connexins. J Neurosci Rozental R, Andrade-Rozental AF, Zheng X, Urban M, Spray DC, Chiu
27:13949–13957. FC. 2001a. Gap junction-mediated bidirectional signaling between
Pakhotin P, Verkhratsky A. 2005. Electrical synapses between Bergmann human fetal hippocampal neurons and astrocytes. Dev Neurosci
glial cells and Purkinje neurones in rat cerebellar slices. Mol Cell 23:420–431.
Neurosci 28:79–84. Rozental R, Srinivas M, Spray DC. 2001b. How to close a gap junc-
Panchin Y, Kelmanson I, Matz M, Lukyanov K, Usman N, Lukyanov S. tion channel. efficacies and potencies of uncoupling agents. In:
2000. A ubiquitous family of putative gap junction molecules. Curr Bruzzone R, Giaume C (eds.), Connexin methods and protocols.
Biol 10:R473–474. Totowa, NJ: Humana Press, pp. 447–476.
Pannasch U, Vargova L, Reingruber J, Ezan P, Holcman D, Giaume C, Saez JC, Berthoud VM, Branes MC, Martinez AD, Beyer EC. 2003a.
et al. 2011. Astroglial networks scale synaptic activity and plasticity. Plasma membrane channels formed by connexins: their regulation
Proc Natl Acad Sci U S A 108:8467–8472. and functions. Physiol Rev 83:1359–1400.
Pelegrin P, Surprenant A. 2006. Pannexin-1 mediates large pore forma- Saez JC, Contreras JE, Bukauskas FF, Retamal MA, Bennett MV. 2003b.
tion and interleukin-1beta release by the ATP-gated P2X7 receptor. Gap junction hemichannels in astrocytes of the CNS. Acta Physiol
EMBO J 25:5071–5082. Scand 179:9–22.
Pellerin L, Magistretti PJ. 2011. Sweet sixteen for ANLS. J Cereb Blood Saez JC, Schalper KA, Retamal MA, Orellana JA, Shoji KF, Bennett
Flow Metab October 26 doi: 10.1038/jcbfm.2011.149. Epub ahead MV. 2010. Cell membrane permeabilization via connexin hemichan-
of print. nels in living and dying cells. Exp Cell Res 316:2377–2389.
Phelan P, Starich TA. 2001. Innexins get into the gap. BioEssays Sanchez-Alvarez R, Tabernero A, Sanchez-Abarca LI, Orfao A, Giaume C,
23:388–396. Medina JM. 2001. Proliferation of C6 glioma cells is blunted by the
Qiu F, Dahl G. 2009. A permeant regulating its permeation pore: inhi- increase in gap junction communication caused by tolbutamide.
bition of pannexin 1 channels by ATP. Am J Physiol Cell Physiol FEBS Letts 509:202–206.
296:C250–255. Santiago MF, Veliskova J, Patel NK, Lutz SE, Caille D, Charollais A,
Ransom BR, Kettenmann H. 1990. Electrical coupling, without dye et al. 2011. Targeting pannexin1 improves seizure outcome.
coupling, between mammalian astrocytes and oligodendrocytes in PLOS 6:e25178.
cell culture. Glia 3:258–266. Scemes E. 2008. Modulation of astrocyte P2Y1 receptors by the car-
Ransom BR, Sontheimer H. 1992. Cell-cell coupling demonstrated boxyl terminal domain of the gap junction protein Cx43. Glia
by intracellular injection of the fluorescent dye lucifer yellow. In: 56:145–153.
Kettenmann H, Grantyn R (eds.), Electrophysiological methods for Scemes E, Giaume C. 2006. Astrocyte calcium waves: what they are and
in vitro studies in vertebrate neurobiology. New York: Wiley-Liss, what they do. Glia 54:716–725.
pp. 336–342. Scemes E, Spray DC, Meda P. 2009. Connexins, pannexins, innexins:
Rash JE. 2010. Molecular disruptions of the panglial syncytium block novel roles of “hemi-channels.” Pflugers Arch 457:1207–1226.
potassium siphoning and axonal saltatory conduction: pertinence to Schalper KA, Palacios-Prado N, Retamal MA, Shoji KF, Martinez AD,
neuromyelitis optica and other demyelinating diseases of the central Saez JC. 2008. Connexin hemichannel composition determines the
nervous system. Neuroscience 168:982–1008. FGF-1-induced membrane permeability and free [Ca2+]i responses.
Rash JE, Yasumura T, Dudek FE, Nagy JI. 2001. Cell-specific expres- Mol Biol Cell 19:3501–3513.
sion of connexins and evidence of restricted gap junctional cou- Schools GP, Zhou M, Kimelberg HK. 2006. Development of gap junc-
pling between glial cells and between neurons. J Neurosci 21: tions in hippocampal astrocytes: evidence that whole cell electro-
1983–2000. physiological phenotype is an intrinsic property of the individual cell.
Retamal MA, Froger N, Palacios-Prado N, Ezan P, Saez PJ, Saez JC, et al. J Neurophys 96:1383–1392.
2007. Cx43 hemichannels and gap junction channels in astrocytes Serrano A, Haddjeri N, Lacaille JC, Robitaille R. 2006. GABAergic net-
are regulated oppositely by proinflammatory cytokines released from work activation of glial cells underlies hippocampal heterosynaptic
activated microglia. J Neurosci 27:13781–13792. depression. J Neurosci 26:5370–5382.
Robinson SR, Hampson EC, Munro MN, Vaney DI. 1993. Unidire- Sohl G, Willecke K. 2004. Gap junctions and the connexin protein fam-
ctional coupling of gap junctions between neuroglia. Science 262: ily. Cardiovasc Res 62:228–232.
1072–1074. Sontheimer H, Minturn JE, Ransom BR, Black JA, Cornell-Bell AH,
Rose CR, Ransom BR. 1996. Intracellular sodium homeostasis in rat Waxman SG. 1991. Cell coupling is restricted to subpopulations of
hippocampal astrocytes. J Physiol (Lond) 491:291–305. astrocytes cultured from rat hippocampus and optic nerve. Ann N Y
Rose CR, Ransom BR. 1997. Gap junctions equalize intracellular Na+ Acad Sci 633:592–596.
concentration in astrocytes. Glia 20:299–307. Sosinsky GE, Boassa D, Dermietzel R, Duff y HS, Laird DW, MacVicar
Rose CR, Ransom BR, Waxman SG. 1997. Pharmacological character- B, et al. 2011. Pannexin channels are not gap junction hemichannels.
ization of Na+ influx via voltage-gated Na+ channels in spinal cord Channels (Austin) 5:193–197.
astrocytes. J Neurophysiol 78:3249–3258. Spray DC, Burt JM. 1990. Structure-activity relations of the cardiac gap
Rouach N, Calvo CF, Glowinski J, Giaume C. 2002. Brain macrophages junction channel. Am J Physiol 258:C195–205.
inhibit gap junctional communication and downregulate connexin Surprenant A, Rassendren F, Kawashima E, North RA, Buell G. 1996.
43 expression in cultured astrocytes. Eur J Neurosci 15:403–407. The cytolytic P2Z receptor for extracellular ATP identified as a P2X
Rouach N, Glowinski J, Giaume C. 2000. Activity-dependent neuronal receptor (P2X7). Science 272:735–738.
control of gap-junctional communication in astrocytes. J Cell Biol Takeuchi H, Jin S, Wang J, Zhang G, Kawanokuchi J, Kuno R, et al. 2006.
149:1513–1526. Tumor necrosis factor-alpha induces neurotoxicity via glutamate

304 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
release from hemichannels of activated microglia in an autocrine Von Blankenfeld G, Ransom BR, Kettenmann H. 1993. Development
manner. J Biol Chem 281(30):21362–21368. of cell-cell coupling among cells of the oligodendrocyte lineage. Glia
Tani E, Higashi N. 1973. Intercisternal structures of closely arranged 7:322–328.
endoplasmic reticulum in human meningioma. Acta Neuropathol Wallraff A, Kohling R, Heinemann U, Theis M, Willecke K, Steinhauser
23:291–299. C. 2006. The impact of astrocytic gap junctional coupling on potas-
Theis M, Sohl G, Eiberger J, Willecke K. 2005. Emerging complexi- sium buffering in the hippocampus. J Neurosci 26:5438–5447.
ties in identity and function of glial connexins. Trends Neurosci Wasseff SK, Scherer SS. 2011. Cx32 and Cx47 mediate oligodendrocyte:
28:188–195. astrocyte and oligodendrocyte:oligodendrocyte gap junction cou-
Theriault E, Frankenstein UN, Hertzberg EL, Nagy JI. 1997. Connexin43 pling. Neurobiol Dis 42:506–513.
and astrocytic gap junctions in the rat spinal cord after acute com- Ye ZC, Oberheim N, Kettenmann H, Ransom BR. 2009. Pharmacological
pression injury. J Comp Neurol 382:199–214. “cross-inhibition” of connexin hemichannels and swelling activated
Torres A, Wang F, Xu Q, Fujita T, Dobrowolski R, Willecke K, et al. anion channels. Glia 57:258–269.
2012. Extracellular Ca(2) acts as a mediator of communication from Ye ZC, Wyeth MS, Baltan-Tekkok S, Ransom BR. 2003. Functional
neurons to glia. Sci Signal 5:8. hemichannels in astrocytes: a novel mechanism of glutamate release.
Vessey JP, Lalonde MR, Mizan HA, Welch NC, Kelly ME, Barnes S. J Neurosci 23:3588–3596.
2004. Carbenoxolone inhibition of voltage-gated Ca channels and Zahs KR, Kofuji P, Meier C, Dermietzel R. 2003. Connexin immuno-
synaptic transmission in the retina. J Neurophysiol 92: 1252–1256. reactivity in glial cells of the rat retina. J Comp Neurol 455: 531–546.

GAP JUNCTIONS AND HEMICHANNELS • 305


25.
PURINERGIC MECHANISMS IN GLIAL CELLS
Margaret S. Ho and Shumin Duan

A B B R E VI AT I O N S for the first time that the synaptic currents in the brain are
mediated by ATP receptors (Edwards et al. 1992). This pio-
ABC ATP-binding cassette neering work, together with abundant others, provide clues
AC adenylyl cyclase on the physiological significance of extracellular ATP and its
ADA adenosine deaminase breakdown products in the brain, further promoting in-depth
ADK adenosine kinase investigation on purinergic mechanisms in nervous systems.
ADP adenosine diphosphate Widely recognized as neurotransmitters, neuromodula-
AMP adenosine monophosphate tors, or gliotransmitters, ATP and adenosine have been shown
AK adenylate kinase to mediate intercellular signaling in the brain. Not only does
AP alkaline phosphatase ATP promote intercellular calcium wave propagation among
ATP adenosine triphosphate astrocytes (Bowser and Khakh 2007; Butt 2011; Scemes and
cAMP cyclic adenosine monophosphate Giaume 2006), ATP and adenosine-mediated cell–cell com-
CBF cerebral blood flow munication is also crucial in modulating various neuronal
DPM dipyridamole and glial functions. Notably, ATP and adenosine are essential
E-NDPK ectonucleoside diphosphokinase components that modulate synaptic transmission and plas-
E-NPP ecto-nucleotide pyrophosphatase/ ticity in both long- and short-term scales. In this chapter, we
phosphodiesterase provide an overview on purinergic mechanisms in glial cells.
E-NTPDase ecto-nucleoside triphosphate diphospho- Following an introduction on the basic properties of purines
hydrolase and purinergic receptors, we discuss the functions and mecha-
EPSC excitatory postsynaptic current nisms of purinergic signaling in glial cells. In particular, we
ER endoplasmic reticulum focus on the roles of P2X7 receptor activation in glial cells and
GPCR G protein–coupled receptor modulation of synaptic transmission and plasticity by astro-
IL-β interleukin-1β cyte-derived purines. Finally, we briefly describe other types
IP3 inositol triphosphate of purine-mediated intercellular communications in the brain,
mGluR metabotropic glutamate receptor including astrocyte calcium signaling, gliovascular coupling,
5′-NT ecto-5′-nucleotidase axon-myelinating cell interactions, and microglial responses,
OPC oligodendrocyte progenitor cell topics that are addressed in more details in other chapters.
PLC phospholipase C purinoceptors purinergic
receptors
RB-2 reactive blue 2 2 S TO R AG E , R E L E A S E , A N D
SC Schwann cell D E G R A DAT I O N M E C H A N I S M S O F
SNARE Soluble N-ethylmaleimide-sensitive factor A D E N O S I N E T R I P H O S P H AT E
attachment protein receptor
Being a widespread route for cell–cell communication, puri-
nergic signaling mediates a variety of functions during neu-
1 INTRODUCTION rons–glia or glia–glia cross-talk. The major signaler ATP
in this pathway is known to be stored in and released from
Purines are chemical substances that have been regarded as the both neurons and glia, and its impact upon releasing has been
major intracellular energy sources and drive most of the reac- immensely studied. Previous studies revealed that in neurons,
tions in the animal body. Over the years of research, discoveries ATP is often coreleased with other neurotransmitters such as
have been made on functions of these substances in various cel- acetylcholine (Redman and Silinsky 1994) or noradrenaline
lular contexts and new ideas about how they act as extracellular (von Kugelgen et al. 1998). In glial cells, ATP was reported
signaling molecules are emerging. Although some of the earli- to be stored and released from organelles that associate with
est work showed that adenosine triphosphate (ATP) acts as a components of the soluble N-ethylmaleimide-sensitive fac-
neurotransmitter in the peripheral nervous system (Burnstock tor attachment protein receptor (SNARE) complex in cul-
1972), a plethora of evidence has suggested its importance in tured astrocytes in a Ca2+-dependent manner (Coco et al.
the central nervous system. For instance, Edwards et al. showed 2003; Maienschein et al. 1999). More intriguingly, by directly

306
targeting the expression of a dominant-negative SNARE mediating lysosome exocytosis of both ATP and cathepsin B
domain in astrocytes in transgenic mice, adenosine-mediated from astrocytes (Verderio et al. 2011). Furthermore, VAMP7-
heterosynaptic suppression was shown to depend on and Rab 27-dependent lysosomal exocytosis of myelin proteins
SNARE-mediated vesicle fusion (Pascual et al. 2005). These have also been shown to play an important role in myelin bio-
results suggest that astrocyte release of ATP, like glutamate genesis and remyelination in oligodendrocytes (Feldmann et al.
(Araque et al. 2000; Bezzi et al. 2004; Jourdain et al. 2007; 2011) and Schwann cells (SCs) (Chen et al. 2012), respectively. It
Pasti et al. 1997, 2001), is vesicular and SNARE-dependent. will be of great interest to examine whether lysosomal exocytosis
Nonetheless, other venues for astrocyte ATP release have of ATP also occurs in these types of glial cells.
been continuously uncovered. For instance, molecular com- High concentration of ATP in synaptic vesicles has been
ponents such as connexin hemichannels (Cotrina et al. 1998a; reported to depend on the activity of vesicular proton pump that
Stout et al. 2002), P2X7 receptors (Suadicani et al. 2006), and maintains an inside-positive potential of the vesicle membrane for
ATP-binding-cassette (ABC) proteins (Ballerini et al. 2002) accumulating negative-charged ATP through the nucleotide trans-
have all been reported to be involved in ATP release (for details porter (Sperlágh and Vizi 1996). Although it is not clear whether
on the mechanisms of astrocyte gliotransmitter release and lysosomes also express nucleotide transporters, expression of puta-
hemichannels, please refer to chapters 17 and 24). Surprisingly, tive ATP-binding cassette transporters and multidrug-resistant
lysosomes in astrocytes were found to exhibit Ca2+-dependent proteins permeable to ATP (Abraham et al. 1993; Ballerini et al.
exocytosis in response to various stimuli (Jaiswal et al. 2007; Li 2002) on the lysosomal membrane have been reported (Cabrita
et al. 2008; Verderio et al. 2011; Zhang et al. 2007) and serve et al. 1999; Eskelinen et al. 2003). Thus, via a similar mechanism,
as the prominent exocytosis machineries for releasing ATP in lysosomes may accumulate higher amounts of ATP because their
astrocytes (Verderio et al. 2011; Zhang et al. 2007). Similar ATP higher proton pump activity and more inside-positive potential
release mechanisms have been shown in micro-glia (Dou et al. compared with synaptic vesicles (Fig. 25.2).
2012). Multiple lines of evidence indicated that physiological Unlike classical neurotransmitters, which action is
stimulation by neurotransmitters evokes partial lysosomal exo- achieved by being released into and subsequently cleared
cytosis (kiss-and-run) that allows the release of a small amount from the synaptic cleft, ATP, once released from either neu-
ATP to mediate intercellular communication, whereas patho- rons or glia, is degraded by various enzymes into products
logical stimulation by ischemia induces full lysosomal exocytosis such as nucleoside diphosphates or adenosine. The nucleotide
that enables the release of all lysosomal content including large hydrolysis pathway includes a series of events that convert
amount of ATP together with lysosome enzymes, contribut- ATP or intermediate substrates to different end-products
ing to secondary neural injury (Fig. 25.1). Recently, the tetanus (Yegutkin 2008; Zimmermann 2000) (Fig. 25.3). The break-
neurotoxin-insensitive vesicle-associated membrane protein down of ATP into ADP (adenosine diphosphate) or ADP to
TI-VAMP/VAMP7 was identified as the vesicular SNARE AMP (adenosine monophosphate) is mediated by enzymes

A B
Physiological stimulation Pathological stimulation
(Glutamate, ATP) (Ischemia, injury)

Full lysosome exocytosis


Partial lysosome exocytosis

Release of low level ATP Release high release lysosome


levelATP enzymes

A1/P2Y receptors in P2Y receptors in Propagating P2X7 pore


presynaptic terminals astrocytes ATP-induced
ATP release

Modulation of Astrocyte glutamate and


synaptic activity Ca2+ wave Microglia ATP release
and plasticity migration

Neural network stability Neuronal damage

Figure 25.1 ATP-Accumulated Lysosomes in Astrocytes Exhibit Partial and Full Exocytosis in Response to ATP and Ischemic Stimuli, Respectively
Schematic models for lysosomal exocytosis in glial cells. (A) physiological stimulation with neurotransmitters evokes transient moderate elevation of intra-
cellular Ca2+ in glial cells and induces partial lysosomal exocytosis (kiss-and-run) that allows the release of small amount ATP to activate high affinity
P2Y or A1 receptors, resulting in the heterosynaptic suppression of neuronal activity or inter-astrocyte Ca2+ signaling propagation. (B) Pathological
stimulation with ischemia or injury induces sustained dramatic increase in intracellular Ca2+ in glial cells and evokes full lysosomal exocytosis that
enables the release of all content inside lysosomes, including large amount of ATP together with lysosome enzymes. High level of extracellular ATP
activates low affinity P2X7 receptors that may form large pores to permeate molecules as large as 900 KDa. Furthermore, ATP-induced ATP exocytosis
from lysosomes in microglia may occur under such pathological condition to evoke and maintain microglia migration toward the injury site. All these
elements may contribute to the secondary neural injury.

PURINERGIC MECHANISMS IN GLIAL CELLS • 307


ATP accumulation in synaptic vesicle ATP accumulation in lysosome
ATP (~1mM) ATP (~1mM)
H+ H+
nucleotide
ABC transporter
vATPase H+ transporter H+
proton pump

H+ H+ +
(ATP~100mM) (ATP>100mM) H
PH=5-6 + PH=4-5 +
H H
VAMP1 or 2 VAMP7

Figure 25.2 Mechanisms of ATP Accumulation in Synaptic Vesicles and Lysosomes ATP has been reported to be released or coreleased with some
classical transmitters from some synapses. The concentration of ATP in these synaptic vesicles has been estimated to be ~100mM. The intravesicular
high concentration of the positive-charged proton produced by proton pump (vATPase) activity maintains an inside-positive membrane potential
of synaptic vesicles, which provides a driving force for the negative-charged ATP to be accumulated from cytoplasm (~1mM) into synaptic vesicles
through nucleotide transporters. In lysosomes, the components responsible for accumulating ATP are vATPase and ABC transporters. The concen-
tration of ATP in lysosome is speculated to be greater than 100 mM, because lysosomes are more acidic and have a more inside positive membrane
potential than synaptic vesicles.

of the ecto-nucleoside triphosphate diphosphohydrolase et al. 2003; Stefan et al. 2006), whereas the enzyme ecto-
(E-NTPDase) family (Robson et al. 2006). Among the eight 5′-nucleotidase (5′-NT) is responsible for degrading AMP into
different NPTDase family members, NPTDase1, also known adenosine. One of the 5′-NT, CD73, is known to work in con-
as the cell activation antigen CD39, is highly expressed in cert with CD39 to stimulate the immunosuppressive adenos-
microglial cells and regulates migration behaviors (Braun ine production during inflammatory responses (Deaglio et al.
et al. 2000; Farber et al. 2008). The breakdown of ATP into 2007). Other ecto-enzymes involved in the ATP breakdown
AMP is mediated by enzymes of the ecto-nucleotide pyro- process include alkaline phosphatase (AP), which exhibits
phosphatase/phosphodiesterase (E-NPP) family (Goding broad substrate specificity and adenosine deaminase (ADA)

AP
E-NPP

Ecto-5’-NT
E-NTPDase E-NTPDase AP
Adenosine ADA Inosine
ATP ADP AMP
E-NDPK ADK
AK
AP
ATP Adenosine
Ca2+
Na+ ADP

+
K
α α
β β P2X receptors (P2X1-7)
Gs γ γ
+ -Gi Gq Gs Gi
+ + - P2Y receptors
(P2Y1,2,4,6,11-14)
AC
AC
PLC P1 receptors
+ - (A1, A2A, A2B, A3)
+ -
cAMP levels
+
cAMP levels
IP3 ER Ca2+ release

Figure 25.3 ATP Degradation, Purinergic Receptors, and Their Signaling Pathways ATP undergoes degradation and is converted into various break-
down products including ADP, AMP, adenosine, and inosine. Enzymes responsible for the conversion include: ecto-nucleoside triphosphate diphos-
phohydrolase (E-NTPDase), ecto-nucleotide pyrophosphatase/phosphodiesterase (E-NPP), adenosine deaminase (ADA), adenylate kinase (AK),
adenosine kinase (ADK), alkaline phosphatase (AP), ectonucleoside diphosphokinase (E-NDPK), and ecto-5′-nucleotidase (5′-NT). Purinergic
receptors are categorized into P1 (responded to adenosine), P2X (responded to ATP) and P2Y (responded to ATP and ADP) receptors. P1 and
P2Y receptors are seven transmembrane proteins coupled to G proteins, whereas P2X receptors are ligand-gated ion channels. Depending on the
G proteins P1 and P2Y receptors are coupled to, distinct cellular responses are activated such as intracellular endoplasmic reticulum (ER) release of
Ca2+ and the stimulation or inhibition of cAMP levels.

308 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
responsible for further inactivation of adenosine into inosine. to specific purines and pyrimidines. P2Y1, P2Y6, P2Y12, and
In addition, ectonucleoside diphosphokinase (E-NDPK) and P2Y13 receptors are activated by nucleoside diphosphates
adenylate kinase (AK) are enzymes for nucleotide rephos- (ADP or UDP), whereas P2Y2, P2Y4, and P2Y11 receptors
phorylation and extracellular ATP synthesis, respectively. are activated by ATP or UTP. Depending on the downstream
These ecto-nucleotidases and their relatives are distributed in G proteins with which they interact, P2Y receptors activate
brains and other tissues, often with overlapping expressions. either the phospholipase C (PLC)/inositol triphosphate (IP3)
Yet, among these enzymes, adenosine kinase (ADK) is promi- endoplasmic reticulum (ER) Ca2+-release pathway (Golovina
nently expressed in astrocytes (Studer et al. 2006) and serves and Blaustein 2000; Scemes 2000; Sheppard et al. 1997;
as a key regulator for adenosine metabolism. ADK eliminates Verkhratsky 2005) via coupling with Gq/11 proteins (P2Y1,
and converts adenosine into AMP and its astrocytic expres- P2Y2, P2Y4, P2Y6, and P2Y11), or modulate the activity of AC
sion has further hinted the importance of glial purinergic sig- and cAMP levels via coupling with Gs (stimulatory, P2Y11) or
naling in regulating various synaptic events. Gi (inhibitory, P2Y12, P2Y13, P2Y14) protein (Abbracchio et al.
It is very crucial to maintain a working balance between 2006) (see Fig. 25.3).
ATP and adenosine in extracellular milieu and subtle changes Purinoceptors have widespread expressions in neurons
during storing, release, and degradation processes of ATP (Illes and Ribeiro 2004) and glial cells. Broadly speaking,
could possibly affect its turnover and ambient adenosine lev- most P1, P2X, and P2Y receptors are expressed in astrocytes
els. Multiple and complex regulatory mechanisms are placed and microglia, whereas some of the purinoceptors are also
upon these processes in order to coordinate the final outcome expressed in different subtypes of glial cells like SCs or oli-
of purinergic signaling. One such regulation comes from the godendrocytes (Table 25.1). mRNAs of all four P1 receptors
ability of these ATP metabolites to differentially activate cor- have been detected in cultures of oligodendrocyte progenitor
responding purinergic receptors (purinoceptors). Diversity of cells (OPCs) by RT-PCR (Stevens et al. 2002; Verkhratsky
purinoceptors, especially their differential expressions in dis- et al. 2009) and functional assays have been done to con-
tinct neuronal and glial cell populations, adds an extra layer of firm their expressions in astrocytes and microglia (Dare et al.
complexity in their ultimate actions such as modulations on 2007; Verkhratsky et al. 2009). In particular, mRNA and pro-
synaptic transmission and plasticity, glial calcium wave propa- tein expressions of A1 adenosine receptor have been revealed
gation, and glial pathological responses. in both astrocytes and murine retina glial cells. Evidence also
implicates a function in the volume homeostasis of retina glial
cells by A1 adenosine receptor (Iandiev et al. 2007; Wurm et al.
3 P U R I N E R G I C R E C E P TO R S 2009). Activities of another adenosine receptor, A2A, have also
been implicated in SCs and microglia. Adenosine activates
On the receiving end of purinergic signaling, two major types A2A receptors and stimulates MAP kinase, thereby regulating
of purinoceptors are activated by ATP metabolites: P1 and P2 SC proliferation (Stevens et al. 2004). On the other hand, A2A
(see Fig. 25.3). P1 receptors respond to mainly adenosine and receptors are expressed in microglia and crucial for the repul-
include four subtypes: A1, A2A, A2B, and A3. Early in the nam- sive effect on microglial motility (Gyoneva et al. 2009).
ing history of purinoceptors, P1 receptors were categorized Expressions of most P2 receptors have been detected
into inhibitory (A1) and stimulatory (A2) types (van Calker in various glial cell types including astrocytes, microglia,
et al. 1979). Importantly, A1 and A3 receptors activate the Gi and OPCs in both transcriptional and protein levels (see
family of G proteins and inhibit cyclic adenosine monophos- Table 25.1). Analyses of different sample preparations revealed
phate (cAMP) formation via inhibiting the adenylyl cyclase the expressions of most P2X receptor mRNAs and proteins in
(AC), whereas A2A and A2B receptors activate the Gs family of astrocytes and Müller glia (Verkhratsky et al. 2009), whereas
G proteins and stimulate AC and cAMP formation (Ralevic immunoreactive results of P2X receptors have also been
and Burnstock 1998). shown in OPCs (Agresti et al. 2005; James and Butt 2001).
The P2 receptors respond to ATP and ADP and belong In microglia, mRNA and protein expressions of P2X1, P2X4,
to two major subfamilies, P2X and P2Y (Boison et al. 2010; and P2X7 receptors have been revealed (Xiang and Burnstock
Fredholm et al. 2001; Ralevic and Burnstock 1998). At pres- 2005). Evidence for functional expressions of P2X receptors
ent, seven P2X receptors and twelve P2Y receptors have in most glial cells, however, is not conclusive. Significantly,
been named and cloned (Brake et al. 1994; Lustig et al. 1993; P2X7 receptor-mediated currents and Ca2+ signaling events
Ralevic and Burnstock 1998; Valera et al. 1994; Webb et al. have been identified in astrocytes both in cultures and in-situ,
1993). Ionotropic P2X receptors belong to the family of cat- suggesting a functional role of P2X7 receptor during synaptic
ionic ligand-gated ion channels, whose pore openings are activity (Duan et al. 2003; Fumagalli et al. 2003; Nobile et al.
mediated by ATP binding and become permeable to Na+, K+, 2003). In addition, in mouse cortical astrocytes, assays have
and Ca2+ ions. These P2X receptors form trimers of subunits been done to detect ATP-induced currents with properties
encoded by P2X1 to P2X7 genes and express in different brain unique for heterodimers of P2X1 and P2X5 subunits (Lalo
regions (Nicke et al. 1998; North 2002; Roberts et al. 2006). et al. 2008), further demonstrating the functional existence of
P2Y receptors are classical seven transmembrane-spanning P2X1/P2X5 heterodimers.
metabotropic receptors coupled to families of G proteins On the other hand, transcriptional and protein expressions
including Gi, Go, or Gq/11 (GPCRs). Among the twelve cloned of most P2Y receptors including P2Y1, P2Y2, P2Y4, and P2Y6
P2Y receptors, eight of them have been shown to respond have been shown in astrocytes, Müller glia, and microglia

PURINERGIC MECHANISMS IN GLIAL CELLS • 309


Table 25.1 PURINERGIC RECEPTORS IN GLIAL CELLS
DOWNSTREAM
SIGNALING
RECEPTOR COMPONENTS SCHWANN CELLS OLIGODENDROCYTES

MÜLLER ENTERIC
ASTROCYTE (EYE) GLIA MYELIN NON-MYELIN TERMINAL PROGENITOR MYELIN MICROGLIA

P1(Adenosine)
A1 GipcAMP R,P,F P,F F R P,F F
A2A GsncAMP F R,P,F R F
A2B GsncAMP F R R F
A3 GipcAMP F R F
P2X(ionotropic)
P2X1 R,P,F R P R,P
P2X2 R,P P
P2X3 R,P R P
P2X4 R,P R P R,P,F
P2X5 R,F R P
P2X6 R,P P
P2X7 R,P,F R,P,F P F P,F P,F R,P,F
P2Y(metabotropic)
P2Y1 Gq/11nPLC R,P,F R,P F P,F R,F
P2Y2 Gq/11nPLC R,P,F R,P F F F F P,F F
P2Y4 Gq/11nPLC R,P,F R,P F P,F R,F
P2Y6 Gq/11nPLC R,P,F R,P F R,F
P2Y11 Gs, Gq/11 R F
nPLC
ncAMP
P2Y12 GipcAMP R,P,F F P R,F
P2Y13 GipcAMP R R F R
P2Y14 GipcAMP R,F

R, mRNA evidence; P, protein evidence; F, functional evidence. Functional evidence includes calcium imaging, protein kinase activation, responses to selective agonists and antagonists, and electrophysiological studies. The down-
stream signaling components of purinergic receptors are also integrated.
Modified with permission from Fields RD, Burnstock G. 2006. Purinergic signalling in neuron-glia interactions. Nat Rev Neurosci 7:423–436 and updated to include recent research data on purinergic receptor expressions.
(Abbracchio and Ceruti 2006; Fields and Burnstock 2006; actions on ion influx and the release of various factors. Insights
Fries et al. 2005). Immunoreactive and functional studies are gained from two hypotheses proposed. Direct dilation and
also showed the expressions of most P2Y receptors in OPCs the secretory lysosome hypothesis state that ATP-mediated
(Agresti et al. 2005). Especially, P2Y12 receptors are expressed activation of P2X7 receptors transform directly the receptor
in OPCs, myelin, and astrocytes (Amadio et al. 2006; Laitinen itself into a large pore by activating the downstream second
et al. 2001). Proper P2Y12 receptor expression is required for messenger systems and rearranging cytoskeleton elements via
pathological conditions such as multiple sclerosis and signal- the cytoplasmic tail so that the receptor is now permeable to
ing between axons and myelinating glia (Amadio et al. 2010). large molecules. In conjunction with this change, the secre-
In summary, P1 and P2 receptors are generally expressed in tory lysosomes release cytokine factors such as interleukin-1β
most glial cell types, yet differential expression patterns were (IL-β) via exocytosis. On the other hand, separate pore and
found to be associated with physiological or pathological membrane bleb hypothesis explains how an unidentified sec-
changes. Together with a precise balance of extracellular ATP ond pore is formed for the passage of large molecules instead
over adenosine, these factors diversify the downstream intrac- of transforming the P2X7 receptor itself. Also, microvesicles
ellular signaling events and pattern glial purinergic responses formed by the budding of the plasma membrane are respon-
to modulate synaptic transmission, glia-glia communication, sible for pinching off cytokines into the extracellular space in
and pathological consequences. this model (Duan and Neary 2006).
The findings that P2X7 pores may directly mediate efflux
of cytosolic glutamate and ATP in astrocytes demonstrate a
4 P 2 X 7 R E C E P TO R - M E D I AT E D R E L E A S E novel mechanism of gliotransmitter release during both physi-
O F G L I OT R A N S M IT T E R S ological intercellular communication and pathological neural
A N D C Y TO K IN E S injury (Duan et al. 2003). In addition to glutamate release,
P2X7 receptors have been reported to play a key role in medi-
Among all P2X receptors, the P2X7 subtype, also known as ating ATP-induced ATP release in astrocytes (Suadicani et al.
P2Z receptor, functions either as a cation channel or a large 2006). Surprisingly, P2X7 receptor-mediated ATP release
nonselective pore, depending on the concentration of ATP is also blocked by gap junction inhibitors (Suadicani et al.
and the duration of stimulation. In response to low extracel- 2006). Connexin hemichannels frequently have been reported
lular ATP levels, P2X7 receptors serve as channels for the per- to mediate the efflux of intracellular large molecules includ-
meabilization of small ions like Ca2+, K+, and Na+, and result ing ATP and amino acids (chapter 24) (Parpura et al. 2004;
in the elevation of intracellular Ca2+. Upon induction by high Ye et al. 2003). Interestingly, some properties of P2X7 pores
levels of ATP, P2X7 receptors are transformed into large non- resemble that reported for gap junction hemichannels, such as
selective pores for the passage of molecules up to 900 kDa. inhibition by divalent cations and some anion channel inhibi-
The opening of P2X7 pores results in the influx of extracel- tors. In this respect, it should be noted that P2X7 receptors
lular Ca2+, large cations such as N-methyl-D-glutamine, and have been suggested to interact with connexins (Fortes et al.
the uptake of fluorescent dyes. Ultimately, the sustained acti- 2004) or pannexin 1, an ortholog to invertebrate innexin gap
vation could lead to cell death (Di Virgilio et al. 1998; North junctions (Pelegrin and Surprenant 2006). In particular, acti-
2002; Surprenant et al. 1996). Although P2X7 receptors are vation of the P2X7 receptor-pannexin 1 complex has been
expressed in various neural cells including astrocytes and neu- related to the processing of the cytokine IL-β in immune cells
rons, their prominent presence in microglia and high expres- (Pelegrin and Surprenant 2006) and the release of ATP from
sion in the monocyte/macrophage cell lineage have indicated astrocytes (Scemes et al. 2007). Thus, pannexin 1 may function
their essential function during microglia-mediated inflamma- as a downstream effector of P2X7 receptor activation to medi-
tion, innate immunity, and the release of cytokines and other ate transmembrane efflux/influx of large molecules and dyes.
inflammatory factors (Ballerini et al. 1996; Brandle et al. 1998;
Deuchars et al. 2001; Ferrari et al. 1996).
In addition to exhibit amino-acid sequence homology with 5 P U R I N E R G I C S I G N A L I N G M O D U L AT E S
other members of the P2X family, P2X7 receptors contain a SY N A P T I C T R A N S M I S S I O N A N D
distinct long C-terminus tail which interacts with numerous PL ASTICIT Y DURING NEURON–GLIA
cytoskeleton and lipid proteins, leading to the activation of I N T E R AC T I O N
multiple intracellular signaling pathways. Variants of P2X7
receptors with C-terminus tail deletions have been shown to One major impact of astrocyte-released ATP is to modulate
exhibit impaired ability of large pore formation, yet are still synaptic transmission of nearby synapses. For instance, a form
capable of mediating small ion influxes (Cheewatrakoolpong of short-term plasticity, heterosynaptic suppression, is mediated
et al. 2005; Surprenant et al. 1996). Moreover, C-terminal by such ATP release (Pascual et al. 2005; Zhang et al. 2003).
deleted P2X7 receptors are expressed in different cell types, Pioneer studies by Zhang et al. have thoroughly examined the
suggesting that the cell-type specific actions of P2X7 recep- role of ATP in hippocampal cultured neurons and slices. First,
tors are in some ways associated with their cytoplasmic tail by applying different purinergic receptor antagonists, the effect
formation. of endogenous ATP on synaptic transmission was examined.
To date, it remains unclear how P2X7 receptors response to Specifically, the addition of P2Y receptor antagonist, reactive
high levels of ATP and transform into large pores for differential blue 2 (RB-2), reduced ATP-mediated synaptic suppression in

PURINERGIC MECHANISMS IN GLIAL CELLS • 311


hippocampal cultures. On the other hand, direct perfusion of experimental setting in which synaptic activities of two nearby
exogenous ATP caused a reduction in the recorded excitatory neurons with reciprocal synaptic connections were measured
postsynaptic current (EPSC) amplitudes. In the presence of an by dual patch recordings. Such recordings provide precise
ectonucleotidase inhibitor, dipyridamole (DPM), the EPSC measurements of heterosynaptic activities between two neu-
amplitudes were suppressed and subsequent RB-2 addition rons and clarify a number of points in the proposed model.
has blocked this suppression. In toto, these results suggest that First, consistent with results that ATP concentrations in the
endogenous ATP tonically suppresses synaptic transmission medium were markedly increased when supplementing the
and this suppression is mediated through P2Y receptors. In pure astrocyte cultures with exogenous glutamate, heterosyn-
addition, this ATP-mediated suppression is modulated pre- aptic suppression in two nearby neurons was only induced
synaptically, as concluded from the decrease in the frequency when synapses were glutamatergic. Moreover, in accord with
rather than the amplitude of spontaneous mini EPSCs (mEP- previous observations that P2Y receptors were implicated in
SCs) when ATP is present (Zhang et al. 2003). synaptic suppression via pharmaceutical means, both RB-2
Because of its close proximity to neurons, astrocytes are and 6,7-dinitroquinoxaline-2,3(1H,4H)-dione, the antago-
readily considered to be the source of released ATP. To sup- nist of non-NMDA receptors, blocked heterosynaptic sup-
port this hypothesis, the authors further investigated whether pression. Taken together, these results support a model that
astrocytes are required for ATP-mediated synaptic suppres- glutamate released on high-frequency stimulation activates
sion. Neither high-frequency stimulation nor RB-2 treat- astrocyte non-NMDA receptors, which in turn trigger ATP
ment increased the amplitudes of spontaneous mEPSCs in release and induce heterosynaptic suppression (Zhang et al.
hippocampal neurons grown in the absence of astrocytes 2003) (Fig. 25.4A).
(GCM cultures), indicating a requirement for astrocytes dur- Several lines of evidence suggested that ATP-mediated syn-
ing synaptic transmission. Also, perfusion of a glia-specific aptic suppression by astrocytes is physiological relevant. First,
metabolic inhibitor, fluoroacetate, or gap junction inhibitor, in hippocampal slice preparation, treatment with the adenos-
octanol, abolished heterosynaptic suppression detected in an ine A1 receptor antagonist, cyclopentyltheophylline, instead

Figure 25.4 Purinergic Signaling in Astrocytes. A. Heterosynaptic suppression is mediated by the release of ATP from the astrocyte (blue).
Non-NMDA receptors in astrocytes are activated by neuronal activity (the glutamate release, brown dots), hence triggering the release of ATP
(pink dots) to activate the presynaptic A1 receptor and suppress synaptic transmission in a nearby synapse. B. ATP released by astrocytes mediate glio-
vascular coupling and cause arteriole dilation via the activation of A2 receptor on the blood vessel (red) by the degradation product of ATP, adenosine
(green dots). On the other hand, the P2Y1 receptor on either astrocyte or blood vessel can be activated by ADP, and promotes vasodilation.
C. Calcium wave propagation is mediated by purinergic signaling in astrocytes. Calcium concentration arises when triggered by the activity-dependent
release of ATP or glutamate from presynaptic terminals. ATP is then released upon intracellular calcium levels increase and this elevation propagates as
wave from one astrocyte to another.

312 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
of RB-2, was found to block the heterosynaptic suppression 2007). These findings indicate that in hippocampus, expres-
mediated by direct perfusion of ATP. Furthermore, in the sion of the foreign receptor MrgA1 that selectively increases
presence of DPM, the addition of RB-2 significantly enhances the GPCR-mediated Ca2+ signaling fails to modulate syn-
the EPSP amplitudes, suggesting that because of high activ- aptic transmission and plasticity, raising the concern of
ity of ectonucleotidases in brain tissue (not in cultured cells), whether GPCR-mediated Ca2+ signaling is indeed involved in
heterosynaptic suppression detected in hippocampal slices is gliotransmitter release by astrocytes.
mediated by endogenous adenosine derived in part from extra- To investigate the controversies further, it is of great inter-
cellularly degraded ATP (Zhang et al. 2003). This idea was est to note that [Ca2+]i microdomain produced by Ca2+ influx
further supported by results from transgenic mice expressing is likely to be more efficient in inducing gliotransmitter release
a dominant-negative SNARE domain that blocks gliotrans- than the GPCR-coupled [Ca2+]i elevation, because the former is
mitter release selectively in astrocytes. In this experimental closer to the plasma membrane where exocytosis occurs. In sup-
paradigm, endogenous adenosine, rather than ATP, is respon- port of this idea, Ca2+ influx through TRPC channels has been
sible for heterosynaptic suppression (Pascual et al. 2005). In reported to be essential for astrocyte gliotransmitter release
summary, these results conclude that astrocyte ATP release (Malarkey et al. 2008). Other players such as gap junction
is activity-dependent and requires the activation of astrocyte hemichannels, mechanical sensitive channels, and the reverse
non-NMDA receptors. Once released, ATP gets hydrolyzed Na+/Ca2+ exchange triggered by intracellular Na+ elevation
to adenosine, which activates presynaptic A1 receptors and accompanying glutamate and GABA transporter activity or
mediates heterosynaptic suppression of nearby synapses. other stimuli also participate in mediating Ca2+ influx in astro-
On the other hand, astrocytes release ATP to activate post- cytes (Doengi et al. 2009; Rojas et al. 2007, 2008; Tong et al.
synaptic P2X7 receptors and modulate a long-lasting form of 2009). Findings that propose two distinctive forms of astrocyte
plasticity at central hypothalamic glutamate synapses (Gordon calcium excitability (Shigetomi et al. 2008), and recent work on
et al. 2005, 2009). Different from classical long-term potentia- that spontaneous Ca2+ microdomain near plasma membrane in
tion that requires the coincidence of presynaptic activity and astrocytes was blocked by extracellular Ca2+ free solution instead
postsynaptic depolarization, this excitatory form of plastic- of inhibitors for intracellular Ca2+ store release (Shigetomi et al.
ity is distributed among all synapses and synaptic strength is 2010) have further supported the above conclusion. Notably,
increased in a multiplicative and scaled fashion. Triggered by based on this model, it is worthy to reconsider the exact nature
afferent activities mimicked with electrical stimulation in phys- of the previously described metabotropic glutamate receptors’
iological patterns or direct activation of astrocytes via uncag- (mGluRs) agonist-induced responses because many agonists
ing glutamate or a membrane-permeable caged IP3 compound, also serve as substrates for glutamate transporters (Duan et al.
astrocyte [Ca2+]i is elevated; consequently, ATP is released to 1999; Harris et al. 1987; Ye et al. 2001) and might as well induce
activate postsynaptic P2X7 receptors, leading to the scaling in Ca2+ responses from such transporter activities.
synaptic responses. In summary, glial purinergic signaling plays
essential roles to modulate different forms of synaptic plastic-
ity, in short and long time scales, and in inhibitory and excit- 6 ASTROCY TE PURINERGIC SIGNALING
atory modes. (Please see chapter 38 for further details.) M E D I AT E S G L I O VA S C U L A R C O U P L I N G
In addition to the heterosynaptic suppression effects dem-
onstrated in the hippocampus, similar inhibitory effects were The importance of glial purinergic signaling is exemplified
also reported in the retina, where light stimulation of glial cells again in the scenario of gliovascular coupling. Astrocytes, as
evokes Ca2+ waves, causing the release of ATP to inhibit neuronal the core component of the neurovascular unit, are ideally situ-
activity (Newman 2003, 2004). Focal application of ATP in the ated in a setting in which their endfeet connect with blood
retina glial cells, Müller cells, hyperpolarizes the neuronal mem- vessels such as arteries/arterioles and on the other end they are
brane conductance and produces outward currents in a subset in close contact with synaptic compartments. This structural
of retinal ganglion cells, thereby reducing the firing rate of these organization allows astrocytes to couple the neuronal activity
neurons. Treatment of the A1 adenosine receptor antagonist with the cerebral blood flow (CBF) via purinergic signaling.
8-cyclopentyl-1,3-dipropylxanthine, the ecto-ATPase inhibi- (Details on how astrocytes regulate gliovascular coupling in
tor 6-N,N-diethyl-D-beta,gamma-dibromomethyleneATP, or other aspects are described in chapter 37.) Increased neuronal
the ectonucleotidase inhibitor adenosine-5′-O-(alpha,beta- activity triggers localized increases in CBF via promoting
methylene)-diphosphonate abolishes this glial-evoked inhibi- vasodilation, an action of blood vessel widening mediated by
tion, suggesting that glial released ATP is degraded into ade- the Ca2+-dependent ATP release from astrocytes (Fig. 25.4B).
nosine extracellularly, which in turn activates the A1 adenosine Increased synaptic activity, represented by the release of
receptor to inhibit neuronal activity (Newman 2003, 2004). neurotransmitters glutamate and ATP, activates the mGluRs
The activation of GPCRs, which triggers the PLC/IP3 ER and P2YRs on astrocytes respectively and triggers the tran-
Ca2+-release (Golovina and Blaustein 2000; Scemes 2000; sient elevations of astrocyte [Ca2+]I, which lead to the release
Sheppard et al. 1997; Verkhratsky 2005), has long been con- of ATP. Gliovascular coupling process mediated by the
sidered to be the major pathway mediating astrocyte Ca2+ Ca2+-dependent ATP release exhibits similar features with
signaling. However, controversies arise when genetic analy- heterosynaptic modulation on nearby synapses. Findings sug-
sis of a transgenic mice line altered in astrocyte GPCR Ca2+ gested that, the dilation of arterioles requires ATP hydroly-
signaling was reported (Agulhon et al. 2008; Fiacco et al. sis. Once released, the actions of enzymes like E-NTPDases,

PURINERGIC MECHANISMS IN GLIAL CELLS • 313


E-NPPs, or 5′-NT are required to degrade ATP into products induces glial calcium wave propagation and the addition of
such as AMP, ADP, or adenosine at the astrocyte endfeet purinergic receptor antagonists blocked the propagation of
(Zimmermann 2000, 2006). These degradation products all evoked calcium waves. Moreover, in the experimental setting
possess strong vasodilation properties. For instance, degrada- where the medium during astrocyte calcium wave propaga-
tion products AMP and adenosine are capable of activating tion can be collected, the collected medium was found to con-
the A2Rs, which causes arteriole dilation (Pelligrino et al. 2011; tain ATP. Application of purinergic receptor antagonists or
Simard et al. 2003). On the other hand, ADP activates the pretreatment with apyrase, an enzyme for ATP degradation,
P2Y1R on both glial membrane and the vascular endothelium blocked the evoked calcium wave propagation (Guthrie et al.
as an alternative pathway to dilate arterioles (Duarte-Araujo 1999). In addition, both P2X and P2Y receptors contribute
et al. 2009; Xu et al. 2005). to the calcium wave propagation; particularly, the P2Y1 recep-
tor responses to micromolar ATP levels and generates fast
and rapid Ca2+ changes, whereas the P2X7 receptor responds
7 P U R I N E R G I C S I G N A L I N G R E GU L AT E S to millimolar ATP levels and generates long and sustained
N E U R O N -M Y E L I N AT I N G G L I A Ca2+ changes ( James and Butt 2002). Together with other
I N T E R AC T I O N work, purinergic signaling has been identified as the essential
pathway mediating calcium wave propagation during glia–
One of the earliest evidence on how ATP modulates synaptic glia cross-talk (Bowser and Khakh 2007; Guthrie et al. 1999;
transmission comes from the observation of the perisynaptic Hassinger et al. 1996).
SCs at the frog neuromuscular junction (Robitaille 1995).
Purinergic receptors were detected in the perisynaptic SCs
and the release of endogenous ATP from the synaptic termi- 9 PURINERGIC SIGNALING IN
nal triggers the release of Ca2+ from intracellular stores in SCs. MICROGLIA
Intriguingly, in a nonsynaptic setting, axons firing bursts of
action potentials also cause the release of ATP, which regulates Microglia are considered the resident immune cells in the brain.
SC myelination through their trophic functions. In summary, During pathological states induced by brain injury, ischemia, or
ATP activates receptors on SCs and regulates gene expressions hypoxia, ATP and adenosine are crucial substances that possess
to inhibit SC proliferation, differentiation, and myelination. dual roles. A great body of work has suggested that in addition
For further details on activity-dependent signaling between to the neuroprotection effects provided by ATP and adenos-
axons and myelinating glia, please refer to chapter 45. ine, ATP is also an important factor mediating the inflamma-
tory action of microglia. As a danger signal, high levels of ATP
were observed in the extracellular space during inflammation
8 P U R I N E R G I C S I G N A L I N G M E D I AT E S (Idzko et al. 2007; Pellegatti et al. 2008) and upon patho-
A S T R O C Y T E C A L C I U M WAVE logical release, ATP activates different purinergic receptors on
P R O PAG AT I O N microglia, leading to morphological change and proliferation
of microglia, and the release of cytokines and growth factors
Glial cells have been considered as the silent components during inflammation (Fig. 25.5). For a general description on
until calcium wave propagation within astrocytes, a form of the physiology of microglia, please refer to chapter 19.
information processing and signaling transduction, was dis- As mentioned, the role of purinergic signaling in micro-
covered in the 1990s (Cornell-Bell et al. 1990). In addition glia is exemplified in the scenario of P2X7 receptor-mediated
to the modulation of synaptic transmission by ATP released IL-1β processing and secretion (Ferrari et al. 1996; Perregaux
from glia, ATP released from astrocyte is also important for and Gabel 1994). Over the years of research, secretions of
propagating calcium waves among astrocytes network (Fig. other cytokines, chemokines, and the activated oxygen spe-
25.4C). Activity-dependent triggers from neurons induce cies have also been associated with P2X7 receptor, suggesting
an increase in the astrocyte intracellular [Ca2+]I (Dani et al. that P2X7-mediated microglia responses in general mediate
1992), which in turn causes the release of ATP. To date, major actions during inflammation. In addition to P2X7, other
there has been great interest in understanding the intrinsic receptors such as P2X4, P2Y6, and P2Y12 are also implicated
properties of astrocyte Ca2+ signaling and how it serves as an in microglia-mediated inflammation responses. For instance,
efficient way for information processing (see chapter 26). In elevated expression of P2Y6R during brain damage triggers
addition to gap junctions, known as the common route for phagocytosis in response to exogenously added or endoge-
intercellular calcium waves propagation (Boitano et al. 1992; nously released UDP, whereas the P2Y12R expression is altered
Charles 1994; Sanderson et al. 1994; Sneyd et al. 1994), ATP during ipsilateral to peripheral nerve injury in the spinal cord
released by astrocytes was demonstrated to be the crucial (Kobayashi et al. 2008; Koizumi et al. 2007; Tozaki-Saitoh
extracellular molecule that propagate the Ca2+ rise in a wave- et al. 2008; Tsuda et al. 2003).
like fashion from one astrocyte to another (Cotrina et al. Observations on reciprocal changes in expression levels of
2000; Cotrina et al. 1998b; Guthrie et al. 1999; Wang et al. P2Y12R and P2X4R (together with A2AR) upon microglial acti-
2000). vation have linked these purinoceptors to the ATP-mediated
A plethora of evidence showed that ATP is responsible microglial process extension, migration, and activation during
for calcium wave propagation. First, focal application of ATP inflammation. In-depth analysis provided by Ohsawa et al.

314 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
A ATPγS 1 mM C ATP γ S
0 min 10 min ATP γ S+Apyrase
2.5

Oriented Migration Rate


2.0

(μm/min)
1.5

1.0 *
*

0.5
*
0.0
<70μm 70~140μm 140~250μm
B Apyrase + ATPγS 1 mM
D
0 min 10 min 1.4
1.2

Oriented Migration Rate


1.0 *
0.8

(μm/min)
0.6
0.4
*
0.2
0.0

PγS N
GP S ove
ry
AT Pγ Rec
T
+A

Figure 25.5 Lysosomal Released ATP Mediates Microglia Migration A,B. Example microscopic images showing cultured microglia migrating
toward the pipette tip (indicated by the white arrow) at the center of the field before (0 min) and 10 min (right panels) after exposure to a gradient
of non-hydrolyzable ATPγS created by pulsatile application through a pipette (at the center of the image field) in the absence (A) and presence (B)
of the ATP degradation enzyme apyrase. Scale bars, 50μm. C. Averaged migration rates of microglia located in three areas at different distances from
the tip of the pipette in conditions shown in A and B. Note that greater inhibition by apyrase was seen for the migration of remote microglia (located
between 140 and 250 μm from the pipette tip). *p < .01 compared with the corresponding control group (without apyrase). D. Averaged data showed
that the ATPγS-induced microglia migration was reversibly inhibited by treatment with 100μM GPN, a substrate of the lysosomal exopeptidase
cathepsin C that selectively induces lysosome osmodialysis. *p < .001 compared with control (before GPN treatment) or between any two groups
indicated. Modified from Dou et al. 2012.

(2007), et al. showed that low levels of extracellular ATP leaking from the local injured cells may not be able to diffuse
mediate microglial chemotaxis sufficiently and recruit cells extensively to induce migration of remote microglia, because
to the injured site. The activation of P2Y12R is also required extracellular nucleotides are rapidly degraded by ecto-ATPases
to mediate such microglial process extension (Davalos et al. in the brain. Similar to that discovered in astrocytes, lysosomes
2005). Upon microglial activation, P2Y12R expression is down- in microglia are found to contain abundant ATP and exhibit
regulated while P2X4R and A2AR expressions are upregulated. Ca2+-dependent exocytosis (Dou et al. 2012). Collective evi-
Also at this point, high levels of ATP are present because of dence indicated that microglia respond to extracellular ATP
brain damage and the ATP/adenosine balance is disturbed. by releasing ATP themselves through lysosomal exocytosis
Activations of P2X4R and A2AR by ATP and adenosine then (Dou et al. 2012). This type of regenerative ATP-induced ATP
allow subsequent microglial migration and process retrac- release from surrounding cells may thus provide a positive
tion. Additional microglial functions such as phagocytosis feedback mechanism to generate and maintain a long-range
and cytokine secretion are also triggered, correlating with extracellular ATP gradient required for the sustained chemot-
all changes. Thus, in addition to the differential activation of axis of remote microglia (Dou et al. 2012) (see Fig. 25.5).
purinoceptors, an ATP concentration gradient presents also Recent studies suggest that neuron-microglia interactions
a key mechanism to achieve such graded action of microglial mediated by purinergic signaling are essential in the pathogen-
activation (Farber et al. 2008; Haynes et al. 2006; Honda et al. esis of neuropathic pain, a chronic pain following peripheral
2001; Ohsawa et al. 2007). nerve injury. Pharmacological, genetic, and behavioral find-
Although ATP released from damaged neural cells has ings indicate that, among the multiple P2 receptor subtypes
been considered the major chemokine for inducing rapid expressed in microglia, the P2X4, P2X7, and P2Y12 recep-
process extension and cell body migration of microglia, ATP tors are responsible for the neuropathic pain after peripheral

PURINERGIC MECHANISMS IN GLIAL CELLS • 315


nerve injury. Release of brain-derived neurotrophic factor and identified. In the future, it will be of great interest to study
proinflammatory cytokines including IL-1β form microglia the distribution patterns and dynamic changes of extracellular
have been linked to the activation of P2X4 and P2X7 recep- ATP-degrading enzyme expressions in various brain regions
tors, respectively, to induce neuropathic pain. Interestingly, during developmental or pathological stages.
p38 MAPK activation has been suggested to be a common
downstream signaling pathway for P2X4, P2X7, and P2Y12
receptors in neuropathic pain. (Relevant topics are also dis- AC K N OW L E D G M E N T S
cussed in chapter 68.)
The authors apologize for not being able to include all the rel-
evant research work because of limitations on chapter length.
10 S U M M A RY A N D P E R S P E C T I VE S Thanks to Nature Publishing Group, Dr. R. Douglas Fields,
and Dr. Geoffery Burnstock for permission with Table 25.1.
Purines-mediated intercellular communication plays impor- Thanks also to Tianming Yu for help with reproducing the
tant roles in various aspects of physiological and pathological table.
conditions and is recognized in almost every type of glial cell.
Thus, astrocyte-derived ATP and its degradation products par-
ticipate not only in inter-astrocyte Ca2+ wave propagation, but REFERENCES
also heterosynaptic modification of neuronal activity and glio-
vascular coupling. Furthermore, action potential–induced ATP Abbracchio MP, Burnstock G, Boeynaems JM, Barnard EA, Boyer JL,
Kennedy C, et al. 2006. International Union of Pharmacology LVIII:
release from axons regulates myelination of both oligodendro- update on the P2Y G protein-coupled nucleotide receptors: from
cytes and peripheral SCs. Finally, purines induce complicated molecular mechanisms and pathophysiology to therapy. Pharmacol
microglial responses including chemotaxis, phagocytosis, and Rev 58:281–341.
cytokine release, functions that play critical roles in various Abbracchio MP, Ceruti S. 2006. Roles of P2 receptors in glial cells: focus
pathological activities such as inflammation and neuropathic on astrocytes. Purinergic Signal 2:595–604.
Abraham EH, Prat AG, Gerweck L, Seneveratne T, Arceci RJ, Kramer R,
pain. Altogether, glial purinergic signaling nurtures a variety of et al. 1993. The multidrug resistance (mdr1) gene product functions
functions during neurons–glia or glia–glia cross-talk. as an ATP channel. Proc Natl Acad Sci U S A 90:312–316.
The mechanisms of purines release and their potential roles Agresti C, Meomartini ME, Amadio S, Ambrosini E, Volonte C, Aloisi
in mediating various glial cell functions have just begun to be F, et al. 2005. ATP regulates oligodendrocyte progenitor migration,
explored. In particular, regulated ATP release through lyso- proliferation, and differentiation: involvement of metabotropic P2
receptors. Brain Res Brain Res Rev 48:157–165.
some exocytosis has been shown as a prominent route in astro- Agulhon C, Petravicz J, McMullen AB, Sweger EJ, Minton SK, Taves
cytes and microglia. Yet, signaling molecule release through SR, et al. 2008. What is the role of astrocyte calcium in neurophysiol-
this action may not be limited to glial cells. In fact, lysosomes ogy? Neuron 59:932–946.
in melanocytes and in cells derived from the haematopoietic Amadio S, Montilli C, Magliozzi R, Bernardi G, Reynolds R, Volonte C.
lineage have been long known as secretory lysosomes because 2010. P2Y12 receptor protein in cortical gray matter lesions in mul-
tiple sclerosis. Cereb Cortex 20:1263–1273.
of their dual functions of protein degradation and signaling Amadio S, Tramini G, Martorana A, Viscomi MT, Sancesario G,
molecules secretion (Blott and Griffiths 2002). Furthermore, Bernardi G, et al. 2006. Oligodendrocytes express P2Y12 metabotro-
lysosomes can fuse with the plasma membrane for membrane pic receptor in adult rat brain. Neuroscience 141:1171–1180.
repair in response to membrane injury (Reddy et al. 2001), Araque A, Li N, Doyle RT, Haydon PG. 2000. SNARE protein-dependent
a typical behavior of exocytosis. Thus, lysosomes seem to be glutamate release from astrocytes. J Neurosci 20:666–673.
Ballerini P, Di Iorio P, Ciccarelli R, Nargi E, D’Alimonte I, Traversa U,
conserved organelles for “general” exocytosis of signaling mol- et al. 2002. Glial cells express multiple ATP binding cassette proteins
ecules in conventional cells, whereas synaptic vesicles are newly which are involved in ATP release. Neuroreport 13:1789–1792.
evolved organelles for efficient release of neurotransmitters in Ballerini P, Rathbone MP, Di Iorio P, Renzetti A, Giuliani P, D’Alimonte I,
specialized neurons. et al. 1996. Rat astroglial P2Z (P2X7) receptors regulate intracellular
As a signaling molecule, ATP is unique for its extracellular calcium and purine release. Neuroreport 7:2533–2537.
Bezzi P, Gundersen V, Galbete JL, Seifert G, Steinhauser C, Pilati E,
degradation by different enzyme systems into products that et al. 2004. Astrocytes contain a vesicular compartment that is
activate distinct subtypes of purinergic receptors coupling to competent for regulated exocytosis of glutamate. Nat Neurosci 7:
different downstream signaling pathways. Whereas ATP acti- 613–620.
vates both ionotropic P2X receptors and metabotropic P2Y Blott EJ, Griffiths GM. 2002. Secretory lysosomes. Nat Rev Mol Cell
receptors but not P1 receptors, its degradation products ADP Biol 3:122–131.
Boison D, Chen JF, Fredholm BB. 2010. Adenosine signaling and func-
and adenosine activate only metabotropic P2Y receptors tion in glial cells. Cell Death Differ 17:1071–1082.
and P1 receptors, respectively. Thus, the degradation scheme Boitano S, Dirksen ER, Sanderson MJ. 1992. Intercellular propaga-
in extracellular space mediated by collections of enzymes tion of calcium waves mediated by inositol trisphosphate. Science
is central to the paradigm of purinergic receptor activation. 258:292–295.
To understand the functional roles of purines-mediated sig- Bowser DN, Khakh BS. 2007. Vesicular ATP is the predominant
cause of intercellular calcium waves in astrocytes. J Gen Physiol
naling in a particular brain region, the cellular source of 129:485–491.
ATP release, distribution pattern of purinergic receptor sub- Brake AJ, Wagenbach MJ, Julius D. 1994. New structural motif for
types, and the expression level of different extracellular ATP- ligand-gated ion channels defined by an ionotropic ATP receptor.
degradation enzymes in the local neural network need to be Nature 371:519–523.

316 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Brandle U, Kohler K, Wheeler-Schilling TH. 1998. Expression of the Duarte-Araujo M, Nascimento C, Timoteo MA, Magalhaes-Cardoso MT,
P2X7-receptor subunit in neurons of the rat retina. Brain Res Mol Correia-de-Sa P. 2009. Relative contribution of ecto-ATPase and
Brain Res 62:106–109. ecto-ATPDase pathways to the biphasic effect of ATP on ace-
Braun N, Sevigny J, Robson SC, Enjyoji K, Guckelberger O, Hammer K, tylcholine release from myenteric motoneurons. Br J Pharmacol
et al. 2000. Assignment of ecto-nucleoside triphosphate 156:519–533.
diphosphohydrolase-1/cd39 expression to microglia and vasculature Edwards FA, Gibb AJ, Colquhoun D. 1992. ATP receptor-mediated syn-
of the brain. Eur J Neurosci 12:4357–4366. aptic currents in the central nervous system. Nature 359:144–147.
Burnstock G. 1972. Purinergic nerves. Pharmacol Rev 24:509–581. Eskelinen EL, Tanaka Y, Saftig P. 2003. At the acidic edge: emerg-
Butt AM. 2011. ATP: a ubiquitous gliotransmitter integrating ing functions for lysosomal membrane proteins. Trends Cell Biol
neuron-glial networks. Semin Cell Dev Biol 22:205–213. 13:137–145.
Cabrita MA, Hobman TC, Hogue DL, King KM, Cass CE. 1999. Farber K, Markworth S, Pannasch U, Nolte C, Prinz V, Kronenberg G,
Mouse transporter protein, a membrane protein that regulates cel- et al. 2008. The ectonucleotidase cd39/ENTPDase1 modulates
lular multidrug resistance, is localized to lysosomes. Cancer Res purinergic-mediated microglial migration. Glia 56:331–341.
59:4890–4897. Feldmann A, Amphornrat J, Schonherr M, Winterstein C, Mobius W,
Charles AC. 1994. Glia-neuron intercellular calcium signaling. Dev Ruhwedel T, et al. Transport of the major myelin proteolipid protein
Neurosci 16:196–206. is directed by VAMP3 and VAMP7. J Neurosci 31:5659–5672.
Cheewatrakoolpong B, Gilchrest H, Anthes JC, Greenfeder S. 2005. Ferrari D, Villalba M, Chiozzi P, Falzoni S, Ricciardi-Castagnoli P, Di
Identification and characterization of splice variants of the human Virgilio F. 1996. Mouse microglial cells express a plasma membrane
P2X7 ATP channel. Biochem Biophys Res Commun 332:17–27. pore gated by extracellular ATP. J Immunol 156:1531–1539.
Chen G, Zhang Z, Wei Z, Cheng Q, Li X, Li W, et al. 2012. Lysosomal Fiacco TA, Agulhon C, Taves SR, Petravicz J, Casper KB, Dong X, et al.
exocytosis in Schwann cells contributes to axon remyelination. Glia 2007. Selective stimulation of astrocyte calcium in situ does not affect
60:295–305. neuronal excitatory synaptic activity. Neuron 54:611–626.
Coco S, Calegari F, Pravettoni E, Pozzi D, Taverna E, Rosa P, et al. 2003. Fields RD, Burnstock G. 2006. Purinergic signalling in neuron-glia
Storage and release of ATP from astrocytes in culture. J Biol Chem interactions. Nat Rev Neurosci 7:423–436.
278:1354–1362. Fortes FS, Pecora IL, Persechini PM, Hurtado S, Costa V, Coutinho-Silva R,
Cornell-Bell AH, Finkbeiner SM, Cooper MS, Smith SJ. 1990. et al. 2004. Modulation of intercellular communication in mac-
Glutamate induces calcium waves in cultured astrocytes: long-range rophages: possible interactions between GAP junctions and P2 recep-
glial signaling. Science 247:470–473. tors. J Cell Sci 117:4717–4726.
Cotrina ML, Lin JH, Alves-Rodrigues A, Liu S, Li J, Azmi-Ghadimi H, Fredholm BB, AP IJ, Jacobson KA, Klotz KN, Linden J. 2001.
Kang J, et al. 1998a. Connexins regulate calcium signaling by con- International Union of Pharmacology. XXV. Nomenclature and
trolling ATP release. Proc Natl Acad Sci U S A 95:15735–15740. classification of adenosine receptors. Pharmacol Rev 53:527–552.
Cotrina ML, Lin JH, Lopez-Garcia JC, Naus CC, Nedergaard M. 2000. Fries JE, Goczalik IM, Wheeler-Schilling TH, Kohler K, Guenther E,
ATP-mediated glia signaling. J Neurosci 20:2835–2844. Wolf S, et al. 2005. Identification of P2Y receptor subtypes in human
Cotrina ML, Lin JH, Nedergaard M. 1998b. Cytoskeletal assembly muller glial cells by physiology, single cell RT-PCR, and immunohis-
and ATP release regulate astrocytic calcium signaling. J Neurosci tochemistry. Invest Ophthalmol Vis Sci 46:3000–3007.
18:8794–8804. Fumagalli M, Brambilla R, D’Ambrosi N, Volonte C, Matteoli M,
Dani JW, Chernjavsky A, Smith SJ. 1992. Neuronal activity triggers cal- Verderio C, et al. 2003. Nucleotide-mediated calcium signaling
cium waves in hippocampal astrocyte networks. Neuron 8:429–440. in rat cortical astrocytes: role of P2X and P2Y receptors. Glia 43:
Dare E, Schulte G, Karovic O, Hammarberg C, Fredholm BB. 2007. 218–203.
Modulation of glial cell functions by adenosine receptors. Physiol Goding JW, Grobben B, Slegers H. 2003. Physiological and pathophysi-
Behav 92:15–20. ological functions of the ecto-nucleotide pyrophosphatase/phospho-
Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, et al. 2005. diesterase family. Biochim Biophys Acta 1638:1–19.
ATP mediates rapid microglial response to local brain injury in vivo. Golovina VA, Blaustein MP. 2000. Unloading and refi lling of two
Nat Neurosci 8:752–758. classes of spatially resolved endoplasmic reticulum Ca(2+) stores in
Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, et al. astrocytes. Glia 31:15–28.
2007. Adenosine generation catalyzed by CD39 and CD73 expressed Gordon GR, Baimoukhametova DV, Hewitt SA, Rajapaksha WR,
on regulatory T cells mediates immune suppression. J Exp Med Fisher TE, Bains JS. 2005. Norepinephrine triggers release of glial
204:1257–1265. ATP to increase postsynaptic efficacy. Nat Neurosci 8:1078–1086.
Deuchars SA, Atkinson L, Brooke RE, Musa H, Milligan CJ, Batten TF, Gordon GR, Iremonger KJ, Kantevari S, Ellis-Davies GC, MacVicar BA,
et al. 2001. Neuronal P2X7 receptors are targeted to presynaptic Bains JS. 2009. Astrocyte-mediated distributed plasticity at hypotha-
terminals in the central and peripheral nervous systems. J Neurosci lamic glutamate synapses. Neuron 64:391–403.
21:7143–7152. Guthrie PB, Knappenberger J, Segal M, Bennett MV, Charles AC,
Di Virgilio F, Chiozzi P, Falzoni S, Ferrari D, Sanz JM, Venketaraman V, Kater SB. 1999. ATP released from astrocytes mediates glial calcium
et al. 1998. Cytolytic P2X purinoceptors. Cell Death Differ waves. J Neurosci 19:520–528.
5:191–199. Gyoneva S, Orr AG, Traynelis SF. 2009. Differential regulation of
Doengi M, Hirnet D, Coulon P, Pape HC, Deitmer JW, Lohr C. 2009. microglial motility by ATP/ADP and adenosine. Parkinsonism Relat
GABA uptake-dependent Ca(2+) signaling in developing olfactory Disord 15(Suppl 3):S195–199.
bulb astrocytes. Proc Natl Acad Sci U S A 106:17570–17575. Harris EW, Stevens DR, Cotman CW. 1987. Hippocampal cells primed
Dou Y, Wu HJ, Li HQ, Qin S, Wang YE, Li J, et al. 2012. Microglial with quisqualate are depolarized by AP4 and AP6, ligands for a puta-
migration mediated by ATP-induced ATP release from lysosomes. tive glutamate uptake site. Brain Res 418:361–365.
Cell Res. 2012 Jun;22(6):1022–33. Hassinger TD, Guthrie PB, Atkinson PB, Bennett MV, Kater SB. 1996.
Duan S, Anderson CM, Keung EC, Chen Y, Swanson RA. 2003. P2X7 An extracellular signaling component in propagation of astrocytic
receptor-mediated release of excitatory amino acids from astrocytes. calcium waves. Proc Natl Acad Sci U S A 93:13268–13273.
J Neurosci 23:1320–1328. Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB,
Duan S, Anderson CM, Stein BA, Swanson RA. 1999. Glutamate induces et al. 2006. The P2Y12 receptor regulates microglial activation by
rapid upregulation of astrocyte glutamate transport and cell-surface extracellular nucleotides. Nat Neurosci 9:1512–1519.
expression of GLAST. J Neurosci 19:10193–10200. Honda S, Sasaki Y, Ohsawa K, Imai Y, Nakamura Y, Inoue K, et al. 2001.
Duan S, Neary JT. 2006. P2X(7) receptors: properties and relevance to Extracellular ATP or ADP induce chemotaxis of cultured microglia
CNS function. Glia 54:738–746. through Gi/o-coupled P2Y receptors. J Neurosci 21:1975–1982.

PURINERGIC MECHANISMS IN GLIAL CELLS • 317


Iandiev I, Wurm A, Pannicke T, Wiedemann P, Reichenbach A, Robson Pasti L, Volterra A, Pozzan T, Carmignoto G. 1997. Intracellular cal-
SC, et al. 2007. Ectonucleotidases in Muller glial cells of the rodent cium oscillations in astrocytes: a highly plastic, bidirectional form of
retina: Involvement in inhibition of osmotic cell swelling. Purinergic communication between neurons and astrocytes in situ. J Neurosci
Signal 3:423–433. 17:7817–7830.
Idzko M, Hammad H, van Nimwegen M, Kool M, Willart MA, Muskens F, Pasti L, Zonta M, Pozzan T, Vicini S, Carmignoto G. 2001. Cytosolic
et al. 2007. Extracellular ATP triggers and maintains asthmatic airway calcium oscillations in astrocytes may regulate exocytotic release of
inflammation by activating dendritic cells. Nat Med 13:913–919. glutamate. J Neurosci 21:477–484.
Illes P, Ribeiro JA. 2004. Neuronal P2 receptors of the central nervous Pelegrin P, Surprenant A. 2006. Pannexin-1 mediates large pore forma-
system. Curr Top Med Chem 4:831–838. tion and interleukin-1beta release by the ATP-gated P2X7 receptor.
Jaiswal JK, Fix M, Takano T, Nedergaard M, Simon SM. 2007. Resolving EMBO J 25:5071–5082.
vesicle fusion from lysis to monitor calcium-triggered lysosomal exo- Pellegatti P, Raffaghello L, Bianchi G, Piccardi F, Pistoia V, Di Virgilio F.
cytosis in astrocytes. Proc Natl Acad Sci U S A 104:14151–14156. 2008. Increased level of extracellular ATP at tumor sites: in
James G, Butt AM. 2001. P2X and P2Y purinoreceptors mediate vivo imaging with plasma membrane luciferase. PLoS One 3:
ATP-evoked calcium signalling in optic nerve glia in situ. Cell e2599.
Calcium 30:251–259. Pelligrino DA, Vetri F, Xu HL. 2011. Purinergic mechanisms in gliovas-
James G, Butt AM. 2002. P2Y and P2X purinoceptor mediated Ca2+ cular coupling. Semin Cell Dev Biol 22:229–236.
signalling in glial cell pathology in the central nervous system. Eur J Perregaux D, Gabel CA. 1994. Interleukin-1 beta maturation and release
Pharmacol 447:247–260. in response to ATP and nigericin. Evidence that potassium depletion
Jourdain P, Bergersen LH, Bhaukaurally K, Bezzi P, Santello M, mediated by these agents is a necessary and common feature of their
Domercq M, et al. 2007. Glutamate exocytosis from astrocytes con- activity. J Biol Chem 269:15195–15203.
trols synaptic strength. Nat Neurosci 10:331–339. Ralevic V, Burnstock G. 1998. Receptors for purines and pyrimidines.
Kobayashi K, Yamanaka H, Fukuoka T, Dai Y, Obata K, Noguchi K. Pharmacol Rev 50:413–492.
2008. P2Y12 receptor upregulation in activated microglia is a gateway Reddy A, Caler EV, Andrews NW. 2001. Plasma membrane repair
of p38 signaling and neuropathic pain. J Neurosci 28:2892–2902. is mediated by Ca(2+)-regulated exocytosis of lysosomes. Cell
Koizumi S, Shigemoto-Mogami Y, Nasu-Tada K, Shinozaki Y, Ohsawa K, 106:157–169.
Tsuda M, et al. 2007. UDP acting at P2Y6 receptors is a mediator of Redman RS, Silinsky EM. 1994. ATP released together with acetylcho-
microglial phagocytosis. Nature 446:1091–1095. line as the mediator of neuromuscular depression at frog motor nerve
Laitinen JT, Uri A, Raidaru G, Miettinen R. 2001. [(35)S]GTPgammaS endings. J Physiol 477(Pt 1):117–127.
autoradiography reveals a wide distribution of G(i/o)-linked ADP Roberts JA, Vial C, Digby HR, Agboh KC, Wen H, Atterbury-Thomas A,
receptors in the nervous system: close similarities with the platelet et al. 2006. Molecular properties of P2X receptors. Pflugers Archiv:
P2Y(ADP) receptor. J Neurochem 77:505–518. Eur J Physiol 452:486–500.
Lalo U, Pankratov Y, Wichert SP, Rossner MJ, North RA, Kirchhoff F, Robitaille R. 1995. Purinergic receptors and their activation by endog-
et al. 2008. P2X1 and P2X5 subunits form the functional P2X recep- enous purines at perisynaptic glial cells of the frog neuromuscular
tor in mouse cortical astrocytes. J Neurosci 28:5473–5480. junction. J Neurosci 15:7121–7131.
Li D, Ropert N, Koulakoff A, Giaume C, Oheim M. 2008. Robson SC, Sevigny J, Zimmermann H. 2006. The E-NTPDase family
Lysosomes are the major vesicular compartment undergoing Ca 2+- of ectonucleotidases: Structure function relationships and pathophys-
regulated exocytosis from cortical astrocytes. J Neurosci 28: iological significance. Purinergic Signal 2:409–430.
7648–7658. Rojas H, Colina C, Ramos M, Benaim G, Jaffe EH, Caputo C, et al.
Lustig KD, Shiau AK, Brake AJ, Julius D. 1993. Expression cloning of an 2007. Na+ entry via glutamate transporter activates the reverse Na+/
ATP receptor from mouse neuroblastoma cells. Proc Natl Acad Sci U Ca2+ exchange and triggers Ca(i)2+-induced Ca 2+ release in rat cer-
S A 90:5113–5117. ebellar Type-1 astrocytes. J Neurochem 100:1188–1202.
Maienschein V, Marxen M, Volknandt W, Zimmermann H. 1999. A Rojas H, Ramos M, Benaim G, Caputo C, DiPolo R. 2008. The activ-
plethora of presynaptic proteins associated with ATP-storing organ- ity of the Na+/Ca2+ exchanger largely modulates the Ca2+i signal
elles in cultured astrocytes. Glia 26:233–244. induced by hypo-osmotic stress in rat cerebellar astrocytes. The effect
Malarkey EB, Ni Y, Parpura V. 2008. Ca2+ entry through TRPC1 chan- of osmolarity on exchange activity. J Physiol Sci 58:277–279.
nels contributes to intracellular Ca 2+ dynamics and consequent gluta- Sanderson MJ, Charles AC, Boitano S, Dirksen ER. 1994. Mechanisms
mate release from rat astrocytes. Glia 56:821–835. and function of intercellular calcium signaling. Mol Cell Endocrinol
Newman EA. 2003. Glial cell inhibition of neurons by release of ATP. 98:173–187.
J Neurosci 23:1659–1666. Scemes E. 2000. Components of astrocytic intercellular calcium signal-
Newman EA. 2004. Glial modulation of synaptic transmission in the ing. Mol Neurobiol 22:167–179.
retina. Glia 47:268–274. Scemes E, Giaume C. 2006. Astrocyte calcium waves: what they are and
Nicke A, Baumert HG, Rettinger J, Eichele A, Lambrecht G, Mutschler E, what they do. Glia 54:716–725.
et al. 1998. P2X1 and P2X3 receptors form stable trimers: a novel Scemes E, Suadicani SO, Dahl G, Spray DC. 2007. Connexin and
structural motif of ligand-gated ion channels. EMBO J 17: pannexin mediated cell-cell communication. Neuron Glia Biol
3016–3028. 3:199–208.
Nobile M, Monaldi I, Alloisio S, Cugnoli C, Ferroni S. 2003. Sheppard CA, Simpson PB, Sharp AH, Nucifora FC, Ross CA, Lange GD,
ATP-induced, sustained calcium signalling in cultured rat cortical et al. 1997. Comparison of type 2 inositol 1,4,5-trisphosphate recep-
astrocytes: evidence for a non-capacitative, P2X7-like-mediated cal- tor distribution and subcellular Ca2+ release sites that support Ca 2+
cium entry. FEBS Lett 538:71–76. waves in cultured astrocytes. J Neurochem 68:2317–2327.
North RA. 2002. Molecular physiology of P2X receptors. Physiol Rev Shigetomi E, Bowser DN, Sofroniew MV, Khakh BS. 2008. Two forms
82:1013–1067. of astrocyte calcium excitability have distinct effects on NMDA
Ohsawa K, Irino Y, Nakamura Y, Akazawa C, Inoue K, Kohsaka S. 2007. receptor-mediated slow inward currents in pyramidal neurons.
Involvement of P2X4 and P2Y12 receptors in ATP-induced microg- J Neurosci 28:6659–6663.
lial chemotaxis. Glia 55:604–616. Shigetomi E, Kracun S, Sofroniew MV, Khakh BS. 2010. A genetically
Parpura V, Scemes E, Spray DC. 2004. Mechanisms of glutamate release targeted optical sensor to monitor calcium signals in astrocyte pro-
from astrocytes: gap junction “hemichannels,” purinergic receptors cesses. Nat Neurosci 13:759–766.
and exocytotic release. Neurochem Int 45:259–264. Simard M, Arcuino G, Takano T, Liu QS, Nedergaard M. 2003. Signaling
Pascual O, Casper KB, Kubera C, Zhang J, Revilla-Sanchez R, Sul JY, at the gliovascular interface. J Neurosci 23:9254–9262.
et al. 2005. Astrocytic purinergic signaling coordinates synaptic net- Sneyd J, Charles AC, Sanderson MJ. 1994. A model for the propagation
works. Science 310:113–116. of intercellular calcium waves. Am J Physiol 266:C293–302.

318 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Sperlágh B, Vizi SE. 1996. Neuronal synthesis, storage and release of Verkhratsky A. 2005. Physiology and pathophysiology of the calcium
ATP. Semin Neurosci 8:175–186. store in the endoplasmic reticulum of neurons. Physiol Rev 85:
Stefan C, Jansen S, Bollen M. 2006. Modulation of purinergic signaling 201–279.
by NPP-type ectophosphodiesterases. Purinergic Signal 2:361–370. Verkhratsky A, Krishtal OA, Burnstock G. 2009. Purinoceptors on neu-
Stevens B, Ishibashi T, Chen JF, Fields RD. 2004. Adenosine: an roglia. Mol Neurobiol 39:190–208.
activity-dependent axonal signal regulating MAP kinase and prolif- von Kugelgen I, Goncalves J, Driessen B, Starke K. 1998. Corelease of
eration in developing Schwann cells. Neuron Glia Biol 1:23–34. noradrenaline and adenosine triphosphate from sympathetic neu-
Stevens B, Porta S, Haak LL, Gallo V, Fields RD. 2002. Adenosine: a rones. Adv Pharmacol 42:120–125.
neuron-glial transmitter promoting myelination in the CNS in Wang Z, Haydon PG, Yeung ES. 2000. Direct observation of
response to action potentials. Neuron 36:855–868. calcium-independent intercellular ATP signaling in astrocytes. Anal
Stout CE, Costantin JL, Naus CC, Charles AC. 2002. Intercellular cal- Chem 72:2001–2007.
cium signaling in astrocytes via ATP release through connexin hemi- Webb TE, Simon J, Krishek BJ, Bateson AN, Smart TG, King BF, et al.
channels. J Biol Chem 277:10482–10488. 1993. Cloning and functional expression of a brain G-protein-coupled
Studer FE, Fedele DE, Marowsky A, Schwerdel C, Wernli K, Vogt K, ATP receptor. FEBS Lett 324:219–225.
et al. 2006. Shift of adenosine kinase expression from neurons to Wurm A, Lipp S, Pannicke T, Linnertz R, Farber K, Wiedemann P, et al.
astrocytes during postnatal development suggests dual functionality 2009. Involvement of A(1) adenosine receptors in osmotic volume
of the enzyme. Neuroscience 142:125–137. regulation of retinal glial cells in mice. Mol Vis 15:1858–1867.
Suadicani SO, Brosnan CF, Scemes E. 2006. P2X7 receptors mediate Xiang Z, Burnstock G. 2005. Expression of P2X receptors on rat microg-
ATP release and amplification of astrocytic intercellular Ca 2+ signal- lial cells during early development. Glia 52:119–126.
ing. J Neurosci 26:1378–1385. Xu HL, Ye S, Baughman VL, Feinstein DL, Pelligrino DA. 2005. The
Surprenant A, Rassendren F, Kawashima E, North RA, Buell G. 1996. role of the glia limitans in ADP-induced pial arteriolar relaxation
The cytolytic P2Z receptor for extracellular ATP identified as a P2X in intact and ovariectomized female rats. Am J Physiol Heart Circ
receptor (P2X7). Science 272:735–738. Physiol 288:H382–388.
Tong XP, Li XY, Zhou B, Shen W, Zhang ZJ, Xu TL, et al. 2009. Ca(2+) Ye ZC, Ransom BR, Sontheimer H. 2001. (1R,3S)-1-Aminocyclopentane-1,
signaling evoked by activation of Na(+) channels and Na(+)/Ca(2+) 3-dicarboxylic acid (RS-ACPD) reduces intracellular glutamate lev-
exchangers is required for GABA-induced NG2 cell migration. J Cell els in astrocytes. J Neurochem 79:756–766.
Biol 186:113–128. Ye ZC, Wyeth MS, Baltan-Tekkok S, Ransom BR. 2003. Functional
Tozaki-Saitoh H, Tsuda M, Miyata H, Ueda K, Kohsaka S, Inoue K. hemichannels in astrocytes: a novel mechanism of glutamate release.
2008. P2Y12 receptors in spinal microglia are required for neuro- J Neurosci 23:3588–3596.
pathic pain after peripheral nerve injury. J Neurosci 28: 4949–4956. Yegutkin GG. 2008. Nucleotide- and nucleoside-converting ectoen-
Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, zymes: important modulators of purinergic signalling cascade.
Salter MW, et al. 2003. P2X4 receptors induced in spinal microglia Biochim Biophys Acta 1783:673–694.
gate tactile allodynia after nerve injury. Nature 424:778–783. Zhang JM, Wang HK, Ye CQ, Ge W, Chen Y, Jiang ZL, et al. 2003. ATP
Valera S, Hussy N, Evans RJ, Adami N, North RA, Surprenant A, et al. released by astrocytes mediates glutamatergic activity-dependent het-
1994. A new class of ligand-gated ion channel defined by P2x recep- erosynaptic suppression. Neuron 40:971–982.
tor for extracellular ATP. Nature 371:516–519. Zhang Z, Chen G, Zhou W, Song A, Xu T, Luo Q, et al. 2007. Regulated
van Calker D, Muller M, Hamprecht B. 1979. Adenosine regulates via ATP release from astrocytes through lysosome exocytosis. Nat Cell
two different types of receptors, the accumulation of cyclic AMP in Biol 9:945–953.
cultured brain cells. J Neurochem 33:999–1005. Zimmermann H. 2000. Extracellular metabolism of ATP and other
Verderio C, Cagnoli C, Bergami M, Francolini M, Schenk U, Colombo A, nucleotides. Naunyn Schmiedebergs Arch Pharmacol 362: 299–309.
et al. 2012. TI-VAMP/VAMP7 is the SNARE of secretory lyso- Zimmermann H. 2006. Ectonucleotidases in the nervous system.
somes contributing to ATP secretion from astrocytes. Biol Cell Novartis Found Symp 276:113–128; discussion 128–130, 233–237,
104:213–228. 275–281.

PURINERGIC MECHANISMS IN GLIAL CELLS • 319


26.
CALCIUM SIGNALING IN NEUROGLIA
Alexei Verkhratsky and Vladimir Parpura

A B B R E VI AT I O N S activation of these receptors triggers distinct secondary path-


ways that form the basis for excitability of nervous tissue. The
AMPA α-amino-3-hydroxy-5-methyl-4-isoxazole- neuronal excitability is mainly caused by their plasma mem-
propionic acid brane, which represents an excitable media because of expres-
α7AChR α7 acetylcholine receptor sion of high densities of voltage-gated ion channels leading
[Ca2+]i cytosolic free Ca2+ concentration to generation of action potentials. Glial cells (as a rule) do
[Ca2+]l intra-ER (or intraluminal) free Ca2+ not generate plasmalemmal action potentials and hence are
concentration classified as electrically nonexcitable. The excitability of neu-
CBP Ca2+ binding proteins roglia is of a different kind, being associated with intracellular
CGP37157 7-chloro-5-(2-chlorophenyl)-1,5-dihydro- movements of ions and second messengers. The intracellular
4,1-benzothiazepin-2(3H)-one signaling system mediated through highly controlled move-
CICR Ca2+-induced Ca2+ release ments of Ca2+ ions is particularly important for regulation
ER endoplasmic reticulum of glial Ca2+ responses and is generally believed to provide a
iGluR ionotropic glutamate receptor substrate for glial excitability (Verkhratsky and Kettenmann
InsP3R inositol 1,4,5 trisphosphate (InsP3)–gated 1996).
Ca2+ channel/receptor
MCU mitochondrial Ca2+ uniporter
mGluRs metabotropic glutamate receptors
MPTP mitochondrial permeability transition pore 2 PRINCIPLES OF CALCIUM SIGNALING
NCX Na+/Ca2+ exchanger
NMDA N-methyl-d-aspartate Control over intracellular concentration of inorganic ions is
PCD paroxysmal depolarization shift the fundamental property of life; movements of ions across
PLC phospholipase C plasma membrane and membranes of organelles underlie the
PMCA plasmalemmal Ca2+ ATPase most basic processes of cellular energetics and function. The
RyR ryanodine receptor monovalent ions (Na+, K+, H+, and Cl–) generally provide for
SERCA sarco(endoplasmic) reticulum Ca2+ ATPase electrical potentials across biological membranes, which drive
SOCE store-operated Ca2+ entry electrical excitability and energy production. The divalent
SPCA secretory pathway Ca2+ ATPase cations (Ca2+, Mg2+, Zn2+) are critical for cellular biochemis-
TRP transient receptor potential try through binding to enzymes and regulating their activity.
TRPC canonical type TRP channel Calcium because of its unique biophysical properties (e.g.,
VDAC voltage-dependent anion channel flexible coordination chemistry, high affinity for carboxylate
VGCC voltage-gated Ca2+ channel oxygen, which is the most frequent motif in amino acids, rapid
binding kinetics, and effects on fluidity and fusion of cellular
membranes) ( Jaiswal 2001) is central for controlling a wide
1 I N T R O D U C T I O N : G L I A L E XC I TA B I L I T Y variety of enzymatic cascades and therefore acts as a univer-
sal signaling molecule (Heilbrunn 1943; Petersen et al. 2005;
The intricate cellular web created by excitable (neurons) and nonex- Toescu and Verkhratsky 1998). This role was assumed by Ca2+
citable (neuroglia) elements provides the structural basis for infor- from the very dawn of evolution, as specificities of ATP-based
mation processing in the nervous system. The cell-to-cell signaling energetics require tight control over Ca2+ concentrations in the
that underlies information flow within this complex network is cytosol and within cellular organelles. Excess of Ca2+ inside the
achieved either by ions fluxing through the plasma membrane, cell is toxic because high Ca2+ induces aggregation of proteins
molecules diffusing through the extracellular space (neurotrans- and nucleic acids, affects the integrity of lipid membranes,
mitters, neuromodulators, and hormones), or direct intercellular and triggers precipitation of phosphates (Case et al. 2007).
flow of ions and second messengers via gap junctions. As a result, cells developed a highly sophisticated system for
Neurons and neuroglia express multiple receptors for Ca2+ homeostasis. This system includes numerous intracellular
neurotransmitters and neuromodulators (see chapter 17) Ca2+ binding proteins and several sets of Ca2+ transporters and
that allow intercellular information exchange. However, exchangers (Berridge et al. 2003; Carafoli 2004).

320
Concentration of free Ca2+ ions in the cytosol ([Ca2+]i) between intracellular compartments and the cell and extracel-
of all cells from most primitive prokaryotes to highly special- lular space produce spatiotemporally organized fluctuations
ized eukaryotes is maintained at an exceedingly low level of of free Ca2+ in different parts of the cell and within intracel-
approximately 50 to 100 nM, which is approximately 20,000 lular organelles. These fluctuations are generally referred to as
times lower than that in the extracellular space. The Ca2+ bind- Ca2+ signals.
ing proteins (CBPs; Ca2+ buffers) present in the cytosol have Various environmental signals (physical or chemical)
very high affinity to Ca2+ (KD in a low nanomolar range) (Ikura initiate Ca2+ diffusion through several sets of plasmalemmal
et al. 2002), which limits cytosolic Ca2+ diffusion and facili- and intracellular (endomembrane) Ca2+ channels that trigger
tates localization of cytosolic Ca2+ signals, which often appear [Ca2+]i rise. It is shaped in space and time by cytoplasmic Ca2+
in a form of microdomains or even nanodomains of very high buffering (CBP) and active Ca2+ transport by plasmalem-
(in excess of 10–100 μM) [Ca2+]i. These highly localized mal and intracellular Ca2+ pumps and exchangers (see Fig.
[Ca2+]i domains are fundamental for regulation of fast local 26.1). The number of Ca2+ signaling/homeostatic molecules
cellular responses (e.g., exocytosis). Similar concentration is relatively limited; the most diverse being plasmalemmal
difference also exists between cytosol and some intracellular Ca2+-permeable channels represented by the voltage- and
organelles (e.g., the ER, Golgi complex, some acidic organelles ligand-gated channels, mechanosensitive and store-operated
such as secretory vesicles), which may contain up to 1 mM of channels, and extensive family of cationic transient recep-
free Ca2+. These Ca2+ concentration gradients always directed tor potential (TRP) channels. Transmembrane Ca2+ trans-
toward the cytosol act as a backbone for Ca2+ signaling sys- port is achieved by plasmalemmal Ca2+ ATPases (PMCAs),
tem by creating a driving force underlying Ca2+ diffusion into sarco(endoplasmic) reticulum Ca2+ ATPases (SERCAs), and
the cytosol (Fig. 26.1) (Berridge et al. 2003; Carafoli 2004; secretory pathway Ca2+ ATPases (SPCAs) of the Golgi com-
Verkhratsky 2006). The combination of Ca2+ movements plex and other acidic compartments (Brini and Carafoli 2009;

iGluR
P2XR SOC Connexins
[Ca2+] ~ 2 mM NCX α7AChR PMCA GPCR TRPC Exocytosis Pannexins

[Ca2+] ~ 100 nM ADP ATP


SOCE
Astrocyre InsP3R
Calcium gradients

InsP3R

[Ca2+] ~
0.2-1 mM
Neurone

Ca2+ microdomain
RyR InsP3R
ADP ATP
[Ca2+] ~ 100 nM

VGCC/LGCC
[Ca2+] ~ 2 mM neurotransmitter NCX PMCA iGluR GPCR
release P2XR
α7AChR

Figure 26.1 Calcium Signaling Cascades in Neurons and Glia. The force driving calcium signaling is formed by Ca2+ concentration gradients, which
in turn are created by energy-dependent Ca2+ transport across cellular membranes. Physiological stimulation opens plasmalemmal or intracellular
Ca2+ channels, thus creating Ca2+ fluxes affecting [Ca2+] in various intracellular compartments. In neural cells, calcium signaling events occur either
in the form of localized [Ca2+]i microdomains or global [Ca2+]i elevation. The Ca2+ microdomains regulate various localized functional responses, for
example, being responsible for the initiation of exocytosis. Conceptually, neuronal Ca2+ signals mainly rely on Ca2+ entry via voltage- and ligand-gated
plasmalemmal Ca2+ channels. This Ca2+ entry creates short-lived [Ca2+]i microdomains that determine fast and focal neurotransmitter release from
the synaptic terminals. In glial cells, Ca2+ signals are usually initiated by Ca2+ release from the ER that creates more widespread and longer-lasting
[Ca2+]i elevation, thus determining slower and more sustained release of gliotransmitters. The domains of elevated [Ca2+]i are sensed by mitochondria,
which are able to accumulate Ca2+ via uniporter. Mitochondrial Ca2+ entry acts as the main link between neural cell activity and energy production
by regulating mitochondrial electron transport and ATP synthesis. The Ca2+ signaling cascades are tightly integrated in overall cellular homeostatic
system. Particularly in astrocytes, the NCX is colocalized with NMDA receptors, Na+-dependent glutamate transporter, and Na+/K+ ATPase. This
arrangement allows rapid reversal of the transporter in response to [Na+]i elevation associated with ionotropic receptors and glutamate transport,
which (1) maintains Na+ gradients needed for glutamate transporter function, and (2) creates local Ca2+ signals by Ca2+ entry through the exchanger.
Abbreviations: α7AChR, α7 acetylcholine receptor; CBP, Ca2+-binding protein; ER, endoplasmic reticulum; GPCR, G protein–coupled recep-
tor; InsP3, inositol(1,4,5)trisphosphate receptor; LGCC, ligand-gated Ca2+ channel; NCX, sodium-calcium exchanger; PLC, phospholipase C;
PMCA, plasmalemmal Ca2+ ATP-ase; RyR, ryanodine receptor; SERCA, sarco(endo)plasmic reticulum Ca2+ ATP-ase; SOC, store-operated Ca2+
channel; VGCC, voltage-gated Ca2+ channel. Connexins/pannexins may create high-permeability plasmalemmal channels (which also can include
pore-forming P2X7 receptors or volume-regulated anion channels) that can act as a pathway for nonexocytotic release of neurotransmitters. The role of
Ca2+ signals in controlling the activation and permeability status of these channels remains unknown.

CALCIUM SIGNALING IN NEUROGLIA • 321


Pizzo et al. 2011), as well as by Na+/Ca2+ exchangers or NCXs 3.1.1 Voltage-Gated Ca2+ Channels
(Guerini et al. 2005). Further, cellular Ca2+ signaling involves
mitochondria that accumulate and release Ca2+ through the There is little evidence for functional expression of voltage-gated
uniporter channel, permeability transition pore, and mito- Ca2+ channels (VGCCs) in astrocytes in situ/in vivo. VGCCs
chondrial NCX (Bernardi et al. 2006; De Stefani et al. 2011). have been identified in astrocytes in vitro (see chapter 16), in
These Ca2+ signaling/homeostatic cascades are tightly regu- particular cultured astrocytes were found to express mRNA
lated by Ca2+ ions themselves, so that changes in Ca2+ concen- for L- (Cav1.2), N- (Cav2.2), P/Q- (Cav2.1), R- (Cav2.3), and T
trations in different cellular compartments immediately feed (Cav3.1)- types of calcium channels. Activation of VGCCs results
back to Ca2+ channels and Ca2+ transporters regulating their in inward Ca2+ currents and [Ca2+]i transients (Barres et al. 1990;
function (Burdakov et al. 2005). Additionally, by altering D’Ascenzo et al. 2004; Duff y and MacVicar 1994; Latour et al.
expression of various components of the Ca2+ homeostatic 2003; see also Verkhratsky and Steinhauser 2000 and references
system the cells create context-dependent “Ca2+ signaling therein). Quite often, the VGCCs expression required trophic
toolkits” that tailor Ca2+ signals to environmental challenges manipulations with astrocytes, for example, incubation with
(Berridge et al. 2003). Altogether these many levels of control dibutyryl-cyclic adenosine monophosphate, coculturing with
and regulation create a versatile and robust system that ensures neurons, or acute oxidative stress (Corvalan et al. 1990; MacVicar
the variability and adaptability of Ca2+ signaling machinery. and Tse 1988). VGCCs reported for immature hippocampal
The Ca2+ signals acquire distinct spatiotemporal parameters astrocytes in brain slices (Akopian et al. 1996) most likely repre-
depending on the cell type, the nature of the stimuli, or sent channel expression in the NG2 glial cells. The VGCCs were
the cell metabolic state. These [Ca2+]i signals can appear as considered to participate in spontaneous [Ca2+]i oscillations in
local microdomains that control neurotransmitter release astrocytes in slices from the ventrobasal thalamus. These [Ca2+]i
or regulate cell process guidance. Alternatively, global Ca2+ oscillations were inhibited by nifedipine and enhanced by Bay
signals are instrumental for excitation-contraction and K8644 (Parri and Crunelli 2003). The immunoreactivity for
-secretion coupling, gene expression, and tissue development N (Cav2.2) and R (Cav2.3) channels subtypes was detected in
can be mounted (Bolsover 2005; Bregestovski and Spitzer pituicytes (hypophyseal astrocytes) in situ; water deprivation for
2005; Parpura et al. 2011; Toescu and Verkhratsky 1998; 24 hours induced a significant increase in immunoreactivity of L
Verkhratsky 2006). In the cellular syncytia (e.g., astroglial (Cav1.2)-type channels in these cells (Wang et al. 2009). These
networks) Ca2+ signals can spread over long distances in the observations, however, remain sporadic; as a rule mature astro-
form of propagating Ca2+ waves. The Ca2+ signals also con- glial cells in situ do not demonstrate measurable voltage-gated
trol cell survival and death. Ca2+-dependent pathways initi- Ca2+ currents. There are some indications about upregulation
ate programmed cell death, which is fundamental for normal of L- and P/Q- channels in cells undergoing reactive astroglio-
tissue development and homeostasis, whereas massive Ca2+ sis induced by pilocarpine-induced status epilepticus (Xu et al.
influx can compromise Ca2+ homeostasis and induce necrosis 2007). Incidentally, Ca2+ may also enter astrocytes through Kir4.1
(Nicotera et al. 2007). inward rectifier channels after lowering extracellular K+ concen-
Ca2+ signals are targeted to “Ca2+ sensors” represented tration below 2 mM, although the mechanism of this Ca2+ influx
by Ca2+-sensitive proteins, in many cases enzymes. Binding/ remains unknown (Hartel et al. 2007).
unbinding of Ca2+ ions to “Ca2+ sensors” affects their activity.
The specificity of Ca2+ signaling is determined by variability 3.1.2 Ionotropic Ca2+-Permeable Receptors
in affinities of these proteins to Ca2+ ions and by their intra-
cellular localization. Activation/deactivation of Ca2+ sensors Astroglial cells express several families of ligand-gated Ca2+-
triggers or discontinues diverse biochemical processes, thus permeable channels (see chapter 17), which can be differentially
regulating cellular reactions; the spatio-temporal organization present in various brain regions (Lalo et al. 2011; Verkhratsky and
of Ca2+ signals determines the timing/localization of “Ca2+ Steinhauser 2000). Most of the astrocytes in vitro and in situ,
sensors” reactions, hence providing for spatial and temporal express the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic
coding of Ca2+ signaling (Burgoyne et al. 2004; Toescu and acid (AMPA) glutamate receptor (GluR). The AMPA recep-
Verkhratsky 1998). tors have been found in astrocytes from cerebellum and cor-
tex (Seifert and Steinhauser 2001). In some brain regions
(e.g., in cerebellar Bergmann glial cells) the AMPA receptors
3 CALCIUM SIGNALING IN ASTROGLIA lack GluR2 subunit, which makes them moderately perme-
able to Ca2+ (PCa/Pmonovalent ~1, with fractional Ca2+ currents
3.1 Ca 2+ PE R M E A B L E PL A SM A LE M M A L
~4%) (Burnashev et al. 1992, 1995). Entry of Ca2+ through
CHANNELS
these receptors is additionally limited by fast desensitization.
In the cortex and spinal cord protoplasmic astrocytes express
Astrocytes in vitro, in situ, and in vivo express several types of functional N-methyl-d-aspartate (NMDA) receptors, which
Ca2+ permeable cationic channels which have different lev- demonstrate weak Mg2+ block at –80 mV and thus are avail-
els of Ca2+ permeability. These channels include ionotropic able for glutamate activation at the resting membrane potential
receptors, store-operated channels, TRP channels, and pos- (Lalo et al. 2006; Verkhratsky and Kirchhoff 2007). The Ca2+
sibly mechanosensitive cationic channels. The properties of permeability of astroglial NMDA receptors is about three
store-operated Ca2+ influx are discussed in the later sections; times less compared with the neuronal form of the receptor
here the focus is on voltage- and ligand-gated channels. (PCa/Pmonovalent approximately 3 (Palygin et al. 2010) (Fig. 26.2).

322 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
A B
P2X receptors

Imem(r.u.)

Imem(r.u.)
NMDA receptors

0.5 0.5
2.5 mM [Ca2+]o 2.5 mM [Ca2+]o
10 mM [Ca2+]o 10 mM [Ca2+]o
Vm(mV) Vm(mV)
–80 –60 –40 –20 –10 10 20 30 –80 –60 –40 –20 –10 10 20 30
NMDA ATP
+20 mV
2.5 mM [Ca2+]o +20 mV 2.5 mM [Ca2+]o
–0.5 –0.5
0 mV 0 mV

–20 mV –20 mV 30 pA
–1.0 25 pA –1.0 500 ms
500 ms 2+
+20 mV 10 mM [Ca2+] +20 mV 10 mM [Ca ]o
o
0 mV 0 mV
–1.5 –1.5
–20 mV –20 mV

C D

Fura-2 ratio Fura-2 ratio


(F340/F380) image (F340/F380) image

1.2 Control Single stimulus 1.2


F340/F380 Control Single stimulus
F340/F380

1.0 1.0

0.8 0.8
50 pA 50 pA
0.6 0.6
1s 1s
Control HFS train Control HFS train
1.2 1.2
F340/F380
F340/F380

1.0 1.0

0.8 0.8

0.6 0.6

1.2 HFS train


1.2 D-AP5 HFS train NF449
F340/F380
F340/F380

1.0 1.0

0.8 0.8

0.6 0.6

0 10 20 30 0 10 20 30
time (s) time (s)

Figure 26.2 Calcium Signaling in Cortical Astrocytes Induced by Activation of Ionotropic Receptors. A,B. The I–V curves and examples of current
recordings (insets) in response to stimulation with (A) NMDA (30 μM) and (B) ATP (10 μM) at 2.5 and 10 mM [Ca2+]o. Increase in [Ca2+]o shifts the
reversal potentials for both NMDA- and ATP-induced currents, indicating their Ca2+ permeability. The I–V curves were constructed from 8 (NMDA)
and 7 (P2X1/5) independent experiments. The amplitudes of responses were normalized to the value measured at -40 mV; data are presented as mean
± SD. Solid lines show the results of the best polynomial fit (least squares routine), intersection with zero current axis gives the following values of
reversal potential in 2.5 and 10 mM [Ca2+]o: 1.9 and 5.1 mV for NMDA-evoked currents and 4.6 mV and 6.7 mV for ATP-evoked currents. The permeabil-
ity ratio PCa/PK calculated by extended Goldman-Hodgkin-Katz equation is 3.1 for NMDA receptors and 2.2 for P2X receptors. C,D. Cortical layer
II astrocytes were loaded with Fura-2 in situ via patch pipette. Fluorescent images were recorded following neuronal afferent stimulation in continuous
presence of CNQX (control) and after application of NMDA receptor blocker D-AP5, 30 μM (A) or 10 nM of NF-449, selective antagonist of P2X1
receptors (B). Representative images (pseudo-color, pipette image subtracted; warmer colors correspond to higher [Ca2+]i levels) and [Ca2+]i transients
were recorded from two different cells. [Ca2+]i-transients (middle columns) are expressed as F340/F380 ratio. Modified from Palygin et al. 2010.

CALCIUM SIGNALING IN NEUROGLIA • 323


Astroglial NMDA receptors are sensitive to memantine and The InsP3-induced Ca2+ release from the ER plays the
GluR NR2C/D subunit-selective antagonist UBP141. This leading role in astroglial Ca2+ signaling. Incidentally, in the
pharmacological profile taken together with low Ca2+ perme- astrocytes in situ the ER is localized close to plasma mem-
ability and weak Mg2+ block indicates that astroglial NMDA brane, whereas in cultured cells it tends to be closer to the
receptors can possible represent trimers assembled from NR1, nuclear envelope (Pivneva et al. 2008). Astrocytes express a
NR2C/D, and NR3 subunits (Palygin et al. 2011). wide variety of metabotropic receptors coupled to phospho-
Ionotropic purinoceptors (see also chapter 25) repre- lipase C (PLC); indeed, stimulation of astrocytes in vitro,
sent another type of Ca2+ permeable ligand-gated channels. in situ and in vivo by various neurotransmitters and neuro-
Cortical astrocytes express functional P2X1/5 heteromeric modulators routinely results in production of InsP3 and sub-
purinoceptors, which are characterized by exceptionally high sequent Ca2+ release from the ER (Fig. 26.3) (Agulhon et al.
affinity to ATP (EC50 approximately 50 nM) and slow desen- 2008; Finkbeiner 1993; Verkhratsky et al. 1998). These InsP3-
sitization (Lalo et al. 2008). The P2X1/5 receptors are Ca2+ mediated Ca2+ signals have the following common features:
permeable with P Ca/Pmonovalent approximately 2 (see Fig. 26.2) (1) astroglial Ca2+ signals often do not require extracellular
(Palygin et al. 2010). Cortical astrocytes in situ were also Ca2+; (2) they are blocked following inhibition of SERCAs
reported to express functional P2X7 receptors activated by with thapsigargin or with cyclopiazonic acid; (3) they are
millimolar ATP concentrations (Oliveira et al. 2011). These inhibited by intracellular injections of InsP3R blocker hepa-
receptors can have a substantial Ca2+ permeability, especially rin; and (4) they are absent in transgenic mice lacking type 2
after pore formation following extensive stimulation. The InsP3Rs (Kirischuk et al. 1999; Petravicz et al. 2008). These
P2X7 receptors are likely to be activated in pathological con- InsP3Rs type 2 seems to be predominantly expressed in astro-
ditions. Calcium permeable α7 nicotinic cholinoreceptors glia and they are frequently concentrated in distal processes
(P Ca/Pmonovalent ~6) were also identified in cultured hippocam- (Holtzclaw et al. 2002). Astrocytes also express types 2 and 3
pal astrocytes (Sharma and Vijayaraghavan 2001); however, RyRs, although their contribution to Ca2+ signaling genera-
their expression in situ remains to be confirmed. Stimulation tion remains unclear. The RyR-mediated CICR has not been
of astroglial NMDA and P2X1/5 receptors by appropriate detected in hippocampal astrocytes. On the contrary, [Ca2+]i
agonists or electrical stimulation of neuronal inputs resulted transients in response to caffeine (which opens RyRs in mM
in generation of Ca2+ signals in both cultured astrocytes and concentrations) were described in astrocytes in thalamus
astrocytes in brain slices (see Fig. 26.2) (Palygin et al. 2010). (Parri and Crunelli 2003).
These Ca2+ signals can be functionally relevant, especially The InsP3-mediated ER Ca2+ release in astrocytes regu-
when generated in perisynaptic processes in which they can lates several functional responses. First, ER originated Ca2+
produce local [Ca2+]i microdomains, which can be important signals were found to activate Ca2+-regulated exocytosis and
for spatially segregated functional responses. Ca2+-dependent release of glutamate from cultured astrocytes
(Malarkey and Parpura 2009; Parpura et al. 2011). Inhibition
of ER Ca2+ release by exposure of cultured astrocytes to
3.2 E N D OPL A SM I C R ET I CULUM A S A PR IM A RY
thapsigargin almost completely obliterated Ca2+-dependent
S OUR CE OF AST R O G LI A L Ca 2+ SI G NA L IN G
secretion of glutamate following activation of metabotropic
The ER possesses its own Ca2+ homeostatic machinery which purinoceptors (Innocenti et al. 2000; Jeremic et al. 2001;
determines its participation in cellular Ca2+ signaling. The Malarkey and Parpura 2009). Similarly, thapsigargin inhib-
intra-ER (or intraluminal) free Ca2+ concentration ([Ca2+]L) ited glutamate release in response to mechanical stimulation
varies between 0.2 and 1 mM (Verkhratsky 2005). Levels (Hua et al. 2004). The same inhibition of glutamate release
of [Ca2+]L this high are achieved by energy-dependent Ca2+ was observed after incubation of astroglial cultures with
uptake provided by SERCA pumps. The ER lumen also con- membrane-permeable inhibitor of InsP3Rs diphenyl boric
tains a set of specific Ca2+ binding proteins (e.g., calreticulin acid 2-aminoethyl ester (Hua et al. 2004). In this context it
and calsequestrin) characterized by low affinity to Ca2+ (K D is interesting to note that glutamate release in cultured astro-
~0.5–1 mM). This arrangement helps maintaining high [Ca2+]L glial cells was also inhibited by ryanodine and preincubation
and allows unhindered Ca2+ diffusion through the lumen of with caffeine, suggesting a certain role for RyRs in mediating
the ER, which is instrumental for formation of intra-ER “Ca2+ relevant Ca2+ signals (Hua et al. 2004). Subsequently, the ER
tunnels” that contribute to long-range Ca2+ transport in polar- Ca2+ release was shown to activate the release of glutamate and
ized cells (Verkhratsky 2005). D-serine from astrocytes in acute hippocampal slices (Angulo
The ER membrane also contains several families of intra- et al. 2004; Rusakov et al. 2011; Shigetomi et al. 2008).
cellular Ca2+ channels represented by: (1) Ca2+-gated Ca2+ However, the functional role and importance of ER Ca2+
channels or ryanodine receptors (RyRs); (2) the inositol release in astroglial physiology remains under debate. Some
1,4,5-trisphosphate (InsP3)–gated Ca2+ channels/recep- experiments in transgenic animals in which the ER release was
tors (InsP3Rs); and (3) the nicotinic acid adenine dinucle- occluded (the InsP3R knock-out mice) found no changes in
otide phosphate receptors (see Berridge et al. 2003; Petersen synaptic transmission or plasticity in hippocampus. Similarly,
et al. 2005; Verkhratsky 2005 for relevant references). targeted stimulation of astrocytes overexpressing peripheral
Activation of these intracellular Ca2+ channels results in Ca2+ (i.e., nonexistent in the CNS) metabotropic receptor did not
release from the ER lumen into the cytosol, thus creating alter synaptic transmission or plasticity in Schaffer collater-
cytoplasmic Ca2+ signals. als–CA1 neuron synapses (Agulhon et al. 2010; Fiacco et al.

324 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
A B
250

30 s 200 30 s

[Ca2+]i(nM)

[Ca2+]i(nM)
65 5 min 80 5 min

ATP ATP ATP ATP ATP ATP


2+
Ca - free 500 nM thapsigargin

C D
180
Control Heparin
>3
[Ca2+]i(nM)

50 s 25 s

F/F0
1
50

ATP ATP

Figure 26.3 ATP-Induced Ca2+ Signaling in Bergmann Glial Cells Results Exclusively from Inositol 1,4,5-Trisphosphate (InsP3)–Mediated Ca2+ Release
from ER Ca2+ Stores. A. ATP-induced [Ca2+]i transients were measured from Bergmann glial cells “bulk-loaded” by incubating cerebellar slices in fura-2
acetoxymethyl ester (AM)–containing solutions. Addition of ATP triggered an increase in [Ca2+]i that persisted in Ca2+-free extracellular solution. B.
In a similar experiment, incubation of slice with 500 nM thapsigargin completely and irreversibly blocked ATP-induced Ca2+ signaling. C. Intracellular
administration of heparin via intracellular dialysis with a patch pipette inhibited [Ca2+]i increase induced by ATP. Control [Ca2+]i transient was
recorded from fura 2-AM–loaded cells before commencing intracellular dialysis. D. Illustration of spatial distribution of [Ca2+]i at time of maximum
ATP response. Note higher levels of [Ca2+]i in Bergmann glial cell processes as compared with cell body. Modified from Kirischuk et al. 1995a.

2007; Petravicz et al. 2008). However, other authors found store (capacitative function) and contributes to the plateau
that under specific conditions, InsP3-induced Ca2+ release in phase of [Ca2+]i transients that may outlast the period of stimu-
astrocytes resulted in modulation of synaptic plasticity in neu- lation. Molecular pathways involve either specific Ca2+-release,
ronal networks (Haydon and Lee 2011). Obviously, functional activated Ca2+ channels known as CRAC (Hoth and Penner
heterogeneity and intrinsic plasticity of astroglia may account 1992) or certain types of transient receptor potential (TRP)
for these seemingly contradictory results. channels (Smyth et al. 2006). The molecular nature of CRAC
The InsP3-mediated Ca2+ release is a key factor for astro- channels has been recently deciphered. This Ca2+-permeation
glia-dependent regulation of local blood flow. Astroglial Ca2+ pathway is created by plasmalemmal pore-forming Orai pro-
signals, triggered following activation of metabotropic recep- teins, activation of which is controlled by the ER resident
tors, induce release of vasoactive substances (e.g., derivatives sensor Stim1. When the ER store is depleted, the Stim1 is
of arachidonic acid or carbon monoxide) from perivascular redistributed to the near-plasmalemmal portion of the retic-
endfeet. These substances regulate the tone of cerebral arte- ulum, where it signals to Orai proteins and opens CRAC
rioles and are the leading mechanism of functional hyper- channels (Putney 2007).
emia (Iadecola and Nedergaard 2007). Glial Ca2+ signaling The SOCE is operative in all types of neuroglial cells,
of ER origin also plays numerous trophic roles and is directly including astrocytes, oligodendrocytes, microglia, and patho-
involved in initiation of Bax translocation and astroglial logically modified gliomas (Hartmann and Verkhratsky 1998;
apoptosis (Morales et al. 2011). Kettenmann et al. 2011; Tuschick et al. 1997). Whether astro-
glial SOCE involves Stim1/Orai complex remains unknown.
The expression of Orai and Stim1 proteins was detected
3.3 T HE STOR E - OPE R AT E D Ca 2+ E N T RY:
in the astroglial cell line U373 MG (Barajas et al. 2008);
A R OLE F OR T R A NSIE N T R E CE PTOR
however, this was not yet confirmed in experiments on pri-
P OT E N T I A L CH A N N E L S
mary astrocytes. In general the Ca2+-release activated Ca2+
The ER Ca2+ store is linked to the plasmalemmal Ca2+ influx currents (ICRAC) indicative of Orai/Stim1 Ca2+ permeation
pathway generally known as a “capacitative” or a store-operated have not been detected in astrocytes. At the same time astro-
Ca2+ entry (SOCE). This link, initially proposed by Jim Putney cytes widely express canonical type TRP (TRPC) channels
(1986), has been identified in a majority of nonexcitable and tetramerically assembled from an obligatory TRPC1 subunit
some excitable cells; in all of which depletion of the ER from in combination with ancillary TRPC4 and/or TRPC5 sub-
releasable Ca2+ triggers secondary Ca2+ influx through the spe- units (Golovina 2005; Malarkey et al. 2008). The TRPC1/4/5
cific set of plasmalemmal channels (Parekh and Putney 2005). channels can act as a store-operated pathway and have rela-
Activation of the SOCE facilitates replenishment of the ER tively high Ca2+ permeability (PCa/Pmonovalent ~1–9) (Nilius

CALCIUM SIGNALING IN NEUROGLIA • 325


et al. 2007). Downregulation of TRPC1 expression by the 3.5 M ITO C H O N D R I A IN A ST R O G L I A L
antisense knock down of the TRPC1 gene or its inhibition by Ca 2+ SI G NA L IN G
a blocking antibody directed at an epitope in the pore-forming The mitochondrion is a cellular organelle directly involved
region of the TRPC1 protein significantly suppressed SOCE in many aspects of Ca2+ signaling and Ca2+ homeosta-
in cultured astrocytes (Golovina 2005; Malarkey et al. 2008). sis in all eukaryotic cells (Nicholls 2005). Mitochondrial
The antibody-induced inhibition of TRPC1-containing chan- Ca2+ accumulation is mediated by ion channels located in
nels specifically suppressed plateau phase of the ATP-induced their double membrane. The outer membrane contains the
[Ca2+]i transients in astrocytes as well as mechanically induced voltage-dependent anion channels (VDAC), which have con-
Ca2+ signals and Ca2+-dependent glutamate release (Malarkey
siderable Ca2+ permeability, whereas the inner mitochondrial
et al. 2008). membrane possesses the highly selective Ca2+ channel histori-
cally known as a Ca2+ uniporter. This comprises the channel
3.4 T HE Na + /Ca 2+ E XC H A N G E R protein of mitochondrial Ca2+ uniporter (MCU) and auxil-
IN A ST R O G L I A iary EF-hand–containing protein that regulates the uniporter
The Na+/Ca2+ exchangers, NCXs, belong to the SLC8 fam- (MICU1/CBARA1) (De Stefani et al. 2011). The driving
ily of solute carriers. The mammalian NCX family is encoded force for Ca2+ accumulation is created by mitochondrial
by three genes—NCX1, NCX2, and NCX3—all of which electron transport through inner mitochondrial membrane,
are expressed in astrocytes. The NCX proteins are mainly which makes the mitochondrial interior highly electrone-
localized in perisynaptic astroglial processes, with particu- gative. On average, mitochondrial membrane potential, the
larly prominent appearance in processes covering excitatory Δψ, is set at about –160 to –200 mV. This electronegativity
synapses, where they colocalize with NMDA receptors and provides the electric force that drives Ca2+ toward the mito-
glutamate transporters (Minelli et al. 2007). According to chondrial matrix when cytosolic Ca2+ exceeds the set point
the stoichiometry of 3 Na+: 1 Ca2+ the NCX can operate in of approximately 300 to 400 nM (Nicholls 2005). The Ca2+
both forward (Ca2+ extrusion in exchange for Na+ influx) and influx into the mitochondrial matrix depolarizes the mito-
reverse (Ca2+ entry in exchange for Na+ extrusion) modes. chondrial membrane and stimulates ATP production; thus,
The switch between forward/reverse operational modes is it is the main mechanism for excitation-energetics coupling.
controlled by Na+ and Ca2+ transmembrane ion gradients and In addition, mitochondrial Ca2+ accumulation contributes to
the level of membrane potential. The cytoplasmic Na+ con- overall cellular Ca2+ buffering and limits large cytosolic Ca2+
centration in astrocytes is relatively high, around 17 to 20 mM loads. When too much of Ca2+ is accumulated in mitochon-
(approximately twice the level of Na+ concentration in neurons) dria the pathological developments may ensue. Mitochondrial
(Reyes et al. 2012), which sets the reversal potential for NCX Ca2+ overload may trigger the opening of mitochondrial per-
(approximately –70 to –90 mV) rather close to the character- meability transition pore (MPTP) that can dissipate Δψ;thus
istic resting membrane potential of astrocytes (Vm approxi- terminating ATP production and facilitating the release of cell
mately –80 mV). As a result, the NCX in astrocytes dynamically death–promoting factors (Nicholls 2005).
fluctuates between forward and reverse modes and participates According to the Ca2+ handling described in the preced-
in both Ca2+ entry and clearance (Kirischuk et al. 1997; Reyes ing, mitochondria play a dual role in astroglial Ca2+ signaling,
et al. 2012). In cultured cortical astrocytes, NCX operates in acting either as a Ca2+ buffer or a Ca2+ source. Cessation of
reverse mode even in resting conditions (Reyes et al. 2012). mitochondrial Ca2+ accumulation by inhibiting mitochon-
The Ca2+ entry mediated by the reverse mode of NCX drial uniporter by Ruthenium 360 significantly enhanced
were detected and analyzed in primary cultured astrocytes the amplitude of mechanically stimulated [Ca2+]i transients
and in astroglial cells in situ (Goldman et al. 1994; Kirischuk in primary cultured astrocytes (Reyes and Parpura 2008). At
et al. 1997; Reyes et al. 2012). The NCX can amplify Ca2+ the same time, when mitochondrial Ca2+ release was blocked
signals following activation of astroglial ionotropic receptors. by inhibiting the mitochondrial Na+/Ca2+ exchanger with
The latter carry Na+ currents and may significantly increase the 7-chloro-5-(2-chlorophenyl)-1,5-dihydro-4,1-benzothiaze-
cytosolic Na+ concentration, which can produce additional pin-2(3H)-one (CGP37157), the mechanically induced Ca2+
Ca2+ influx by reversing the exchanger (Goldman et al. 1994; signals were reduced (Reyes and Parpura 2008). Similarly, the
Kirischuk et al. 1997; Lalo et al. 2011; Reyes et al. 2012). The mechanically induced Ca2+ signals were attenuated by treat-
same cascade can be initiated by activation of the plasmalemmal ment with cyclosporin A, which inhibits the MPTP, indicat-
glutamate transporter, which induces substantial Na+ influx ing that flickering of MPTP with subsequent Ca2+ efflux into
(the stoichiometry of transporter is 3 Na+: 1 glutamate). The the cytosol may have a physiological role in astroglial cells
resulting elevation in [Na+]i (by up to 20 mM) rapidly turns (Reyes and Parpura 2008).
NCX into the reverse mode and produces substantial Ca2+
influx (Kirischuk et al. 2007). The activation of reverse mode 3.6 A ST R O G LI A L Ca 2+ WAVE S
of the NCX was also achieved by moderate depolarization of
cultured astrocytes (Paluzzi et al. 2007). The NCX-mediated Astroglial Ca2+ signals have complex spatiotemporal and
[Ca2+]i increases can be functionally relevant and were shown, hierarchical organization, ranging from local Ca2+ micro-
for example, to trigger exocytotic glutamate release (Paluzzi domains to [Ca2+]i oscillations and propagating Ca2+ waves.
et al. 2007; Reyes et al. 2012). Highly localized microdomains of [Ca2+]i can be instrumental

326 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
in controlling various astroglial functions, such as exocytosis A Control 18 s B

(Parpura et al. 2011; Shigetomi et al. 2011). In situ the initial


[Ca2+]i rise often occurs locally in a form of Ca2+ microdo-
mains in the distal perisynaptic astrocytic processes. These
Ca2+ microdomains may stay localized or may spread toward
the soma in a form of propagating wave of ER Ca2+ release (the
intracellular Ca2+ wave) (Grosche et al. 1999; Kirischuk et al. 4.5 s 22.5 s C 0.5
Stim
1995a). At the same time, the single-cell Ca2+ signals often F/F0
30 s
do not terminate at the astrocyte cell border but spread into 1

neighboring astrocytes; thus creating an intercellular Ca2+


wave, which may convey Ca2+ signal over the long distance 2

(~300–400 μm from the site of original excitation) and can


excite many tens or even hundreds of cells within the path of 9s 27 s

its propagation (see Scemes and Giaume 2006 for details and 3
further references).
The intercellular Ca2+ waves were discovered in confluent 6
astroglial cell cultures (Cornell Bell et al. 1990). This was a
seminal discovery that demonstrated that focal stimulation
with glutamate triggers propagating Ca2+ wave in the glial 13.5 s 62.5 s
4
syncytia. These Ca2+ waves had a complex path, crossed cell
borders without appreciable delay, and propagated with a
velocity of approximately 15 to 20 μm/second (Cornell Bell 5

et al. 1990). Further experiments have found that the intercel-


7
lular Ca2+ waves in cultured astroglia could be evoked by focal
mechanical stimulation, although these waves were somewhat
different in that they demonstrated delay at cell borders. The Figure 26.4 Propagation of Ca2+ Waves in an Acute Brain Slice. A. A
series of fluorescence images just before (control) and at defined times
intercellular Ca2+ wave required functional Ca2+ store and can
(as indicated) after electrical stimulation illustrate the spread of the Ca2+
be irreversibly blocked by thapsigargin. Similarly, Ca2+ wave signal within a slice. The position of the stimulation electrode is marked
propagation in cultured cells required functional PLC and by the asterisk. The cartoon in B outlines the anatomical structures of
can be blocked with the PLC inhibitor U73122 (see Scemes the fluorescence images. Moreover, the transient changes in Ca2+ were
and Giaume 2006 and references therein). The initial observa- measured at the indicated areas (B) and are displayed in C. In area 1, close
to the stimulation pipette, the increase in fluorescence (F/F0) occurred
tions on cultured cells were subsequently confirmed in experi-
right after stimulation (vertical line). At more distant areas, the delay
ments in situ in acute brain slices and in vivo (Fig. 26.4) (Dani amounted to several seconds. Note that cells in the ventricle wall respond
et al. 1992; Scemes and Giaume 2006; Schipke et al. 2002). with an intense signal. Reproduced from Schipke et al. 2002.
Propagating Ca2+ waves, which can be blocked by thapsigargin,
were also found in astrocytes in the acutely isolated rat retina
in response to local electrical stimulation or local applications connectivity, controlling spread of astroglial Ca2+ waves, can
of ATP, carbachol or phenylephrine (but not of glutamate) be regulated by physiological stimuli, such as depolarization,
(Newman and Zahs 1997). The synchronized propagating exposure to glutamate, or neuronal firing (see Arcuino et al.
Ca2+ waves that spread through several hundreds of astrocytes 2002; Cotrina et al. 1998; Scemes and Giaume 2006 for
were also recorded in vivo in mice hippocampus. These Ca2+ details and references).
waves were sensitive to inhibition of purinoceptors and gap The alternative mechanism for intercellular Ca2+ waves
junctions (Kuga et al. 2011). involves extracellular diffusion of transmitters (usually
Astroglial Ca2+ wave induction and maintenance is ATP or glutamate) released by astrocytes (Fig. 26.5B,C).
achieved through several, often complementary, mechanisms This mechanism was initially discovered in vitro in conflu-
(Fig. 26.5). First, the propagation of the wave can result from ent astroglial cultures. In these cultures the cell-free line was
diffusion of InsP3 through gap junctions (see Fig. 26.5A). It mechanically drawn. It turned out that a Ca2+ wave can “jump”
is important to remember that heavy cytosolic Ca2+ buffering over the cell-free space of up to 120 μm in width, thus suggest-
greatly limits diffusion of Ca2+ ions themselves and propagat- ing the role of diffusible extracellular messenger (Hassinger
ing Ca2+ wave is not an actual movement of Ca2+ ions, but et al. 1997). This mechanism were subsequently confirmed and
rather a propagating wave of excitation of ER Ca2+ channels. it was found that the transmitter can be released from astro-
The importance of gap junctions in Ca2+ wave propagation glia either through Ca2+-regulated exocytosis or diffusion via
was initially demonstrated by blocking the wave propaga- plasmalemmal pores associated with unpaired connexin hemi-
tion by broad spectrum gap junction inhibitors octanol and channels, pannexins, pore-forming P2X7 receptors, or a vari-
halothane, or by inhibiting gap junctions following activation ety of ATP-permeable anion channels (Arcuino et al. 2002;
of protein kinase C. Subsequently it was shown that in rat C6 Cotrina et al. 1998; Scemes and Giaume 2006).
gliomas intercellular Ca2+ waves could be generated only in cul- These two main mechanisms of Ca2+ wave propagation
tures transfected with connexins. The gap junction–mediated may coexist, or be differentially employed in astroglial syncytia

CALCIUM SIGNALING IN NEUROGLIA • 327


A chapter 24). At the same time the extent of propagation and
exact pathways carrying Ca2+ waves in the brain tissue in vivo
InsP3 remain to be characterized.
Ca2+ InsP3
Ca2+
InsP3 Ca2+ InsP3 4 C a 2+ S I G N A L IN G IN
OLIGODENDROGLIA AND NG2 GLIA

Activity dependent Ca2+ signaling was detected in oligoden-


B
drocytes and their precursors in vivo and in vitro (see chapter
20 and Verkhratsky et al. 1998 for early works and relevant
InsP3
ATP ATP references). These oligodendroglial Ca2+ signals are medi-
Ca2+
InsP3
Ca2+
ated through both Ca2+ entry via plasmalemmal channels and
InsP3 2+
through ER Ca2+ release. Immature oligodendrocytes express
Ca
high- and low- threshold VGCCs; importantly low-threshold
ATP VGCCs (T-type) are predominantly localized at the very tips
of the processes on oligodendroglial precursors. The channels
C
can be activated by rather small (up to ~10 mM) elevations in
extracellular K+ concentration, which induced highly localized
InsP3 [Ca2+]i elevations in the tips of the processes (Kirischuk et al.
ATP
InsP3 1995b). This idiosyncratic localization of T-type Ca2+ channels
Ca2+ Ca2+ in oligodendroglial precursors may help them to identify active
InsP3 Ca 2+
axons (which electrical activity causes local increase in extracel-
lular K+ concentration) and initiate the myelination program.
Oligodendrocytes are also endowed with several families
of Ca2+-permeable ionotropic receptors. In particular mature
Gap junction ER Ca2+ release oligodendrocytes express NMDA receptors, which are quite
Ca2+
similar to astroglial receptors in having weak Mg2+ block and
therefore are fully operative at physiological resting potential
Metabotropic (P2Y) Vesicular release of
receptor neurotransmitter (ATP) (see Stys and Lipton 2007; Verkhratsky and Kirchhoff 2007 for
ATP references and details). In addition, oligodendrocytes express
Non-vesicular release of Ca2+-permeable AMPA receptors and P2X7 purinoceptors (and
neurotransmitter (ATP)
maybe other P2X types as well) (Matute et al. 2007). Finally,
ATP through connexins/pannexins all oligodendrocytes demonstrate robust InsP3-mediated Ca2+
release from the ER stores (Verkhratsky et al. 1998).
Figure 26.5 Mechanisms of Propagating Astroglial Ca2+ Waves. A. Ca2+ The NG2 glial cells (see chapter 21), besides being oligo-
waves can be maintained by diffusion of InsP3 through the gap junction
and secondary initiation of InsP3-induced Ca2+ release. B. Ca2+ waves
dendroglial progenitors and perhaps pluripotent neural pre-
can be maintained by regenerative Ca2+-dependent exocytotic release of cursors, are involved in neural networking by forming contacts
neurotransmitters (e.g., ATP or glutamate) or release of neurotransmit- with both astrocytes and neuronal afferents (from which they
ters through high permeable membrane channels (connexins, pannexins, actually receive synaptic contacts). Therefore, the NG2 glia
P2X7 receptors, or volume-regulated anion channels) acting on neighbor- (also called polydendrocytes or “contact” cells) may respond to
ing cells through extracellular diffusion. C. Ca2+ waves can result from
a focal release of a neurotransmitter, which then diffuses over a long
both neuronal and astroglial inputs. Mechanisms of Ca2+ sig-
distance. Modified from Verkhratsky and Butt 2007. naling in the NG2 glia remains mostly unexplored; however,
several experiments demonstrated that NG2 glia respond with
[Ca2+]i elevations to ATP and glutamate; these signals being
in different brain regions. In neocortex, for example, propa- mediated through Ca2+ permeable AMPA receptors (Ge et al.
gating astroglial Ca2+ waves are mediated mainly through 2006), P2X7 receptors and metabotropic P2Y1 receptors and
gap junctions and completely depend on the expression of group I metabotropic glutamate receptors (Haberlandt et al.
connexin Cx43. In hippocampus and corpus callosum, in 2011; Hamilton et al. 2010). Ca2+ entry through reversed NCX
contrast, spread of Ca2+ waves is mediated by ATP release and were reported to modulate migration of NG2 glia (Tong et al.
subsequent activation of metabotropic P2Y receptors (Haas 2009). In addition, NG2 glial cells were found to express Ca2+
et al. 2006; Schipke et al. 2002). In retina both mechanisms permeable ASIC1a channels (Lin et al. 2010).
are in operation, and gap junctions are involved in propa-
gating Ca2+ wave between astrocytes, whereas ATP release
mediates Ca2+ wave spread between astrocytes and Müller 5 C a 2+ S I G N A L IN G IN M I C R O G L I A
cells (Newman and Zahs 1997). Similarly in hippocampus in
vivo astroglial Ca2+ waves were sensitive to blockade of both In microglial cells, Ca2+ signals are ultimately important for
gap junctions and purinoceptors (Kuga et al. 2011) (see also controlling their activation and multiple functional responses

328 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
(see chapter 19) (Kettenmann et al. 2011). Ca2+ signaling in the neuronal paroxysmal depolarization shift (PCDs, which
microglia is controlled by many metabotropic receptors, is an electrophysiological correlate of synchronous neuronal
including purinoceptors, adrenoceptors, thrombin, chemo- discharge) resulted from massive Ca2+-dependent release of
kine, and complement receptors (see chapter 19), which induce glutamate from astroglia; the antiepileptic drugs suppressed
Ca2+ release from the ER. In addition, microglial Ca2+ signal- astroglial Ca2+ waves and inhibited PCDs (Nedergaard et al.
ing is controlled by Ca2+-permeable P2X4 and P2X7 receptors, 2010 and references therein). Finally, glial Ca2+ dyshomeo-
which are particularly important for microglial activation in a stasis can be involved in altered neurotransmission in psychi-
variety of pathological conditions (see Kettenmann et al. 2011 atric disorders and various neurodegenerative pathologies.
for details and all relevant references). Finally, microglial cells For example, significant elevation in resting [Ca2+]i levels
express SOCE mediated by Orai/Stim1 molecular complex. and enhanced spontaneous [Ca2+]i oscillations were found
This SOCE is important for generation of long-lasting [Ca2+]i in transgenic model of Alzheimer disease (Kuchibhotla et al.
elevations and can be constantly activated following overstim- 2009).
ulation of microglia (Toescu et al. 1998).

7 S U M M A RY A N D P E R S P E C T I VE S
6 PAT H O L O G I C A L P OT E N T I A L O F
A ST R O G L I A L C a 2 + S I G N A L IN G It is generally acknowledged that excitability of neuroglia is
mediated by intracellular Ca2+ signals resulting from Ca2+
Neuroglial cells, being the primary homeostatic and defensive movements between intracellular compartments and the
cells of the nervous system are intimately involved in all forms cell and extracellular space. Neuroglial cells express highly
of neuropathology, and their reactions determine the progres- sophisticated and intrinsically regulated molecular cascades
sion and outcome of neurological diseases to a very large extent. responsible for cellular Ca2+ homeostasis and Ca2+ signal-
Glial Ca2+ signals participate in pathological developments by ing. Stimulation of neuroglial cells triggers coordinated Ca2+
regulating both neuroprotective and neurotoxic responses. signals that can appear in a form of local Ca2+ microdomains,
In ischemia and stroke, astrocytes are specifically impor- global Ca2+ signals, [Ca2+]i oscillations, or propagating Ca2+
tant for infarction spread through the ischemic penumbra waves. This complex organization of Ca2+ signals in both
surrounding the area of pan-necrosis. Astrocytes are gener- space and time determines functional outcome, which ranges
ally resilient to the ischemic stress and they quite often outlive from local signaling at the level of single synapse (regulation
neurons in the damaged areas. Furthermore, they provide the of ion/neurotransmitter release or sequestrating, local meta-
major metabolic support to neurons through anaerobic gly- bolic support) to regulation of long-lasting adaptive responses
colysis and lactate shuttle. Aberrant astroglial [Ca2+]i waves, (myelination, cell survival, cell death, and differentiation
however, are involved in the spread of damage by inducing or defensive reactions such as microglial activation or reac-
propagating wave of astroglial glutamate release, which in turn tive astrogliosis). The main future challenge lies in detailed
triggers distant excitotoxicity (see Nedergaard et al. 2010 for description of these Ca2+ signals in the cells in vivo and in
details and references). Pathological Ca2+ signaling are espe- establishing the links between Ca2+ dynamics and function
cially important for ischemic damage to white matter (e.g., in of neuroglial cells. Furthermore, the interplay between neu-
periventricular leukomalacia or Binswanger disease), which is roglial Ca2+ dynamics and inherently linked intracellular
mainly associated with the rapid death of oligodendrocytes Na+ dynamics will have to be detailed and cross-correlated
and oligodendroglial precursors that are particularly vulner- in respect to in functional contribution of neuroglia to the
able to ischemia. The oligodendroglial death shows all hall- operation of the brain.
marks of Ca2+-dependent excitotoxicity with Ca2+ entering
oligodendrocytes through P2X7 and NMDA glutamate recep-
tors (Matute 2010; Nedergaard et al. 2010). Similarly, Ca2+ AC K N OW L E D G M E N T S
excitotoxicity resulting from enhanced Ca2+ entry through
P2X7 receptors may be involved in oligodendrocyte damage The authors’ research was supported by Alzheimer’s Research
in multiple sclerosis (Matute 2010). Trust (UK) Programme Grant (ART/PG2004A/1) to AV;
Pathological Ca2+ signaling can be also involved in the an Ikerbasque grant to A. V.; a National Science Foundation
pathogenesis of epileptic seizures (see chapter 70). Astroglial (CBET 0943343) grant to V. P.; and a Grant Agency of the
Ca2+ signaling was affected in cells from patients suffering Czech Republic (GACR 305/08/1384) grant to A. V.
from the autoimmune form of child epilepsy, Rasmussen
encephalitis. These pathologically remodeled astrocytes
demonstrated spontaneous Ca2+ oscillations, probably aris- REFERENCES
ing from antibody-induced alterations in the GluR3 subunit
of AMPA glutamate receptor. Experimental epilepsy also Agulhon C, Fiacco TA, McCarthy D. 2010. Hippocampal short- and
upregulates metabotropic glutamate receptors (mGluRs), long-term plasticity are not modulated by astrocyte Ca 2+ signaling.
Science 327:1250–1254.
particularly mGluR5 linked to PLC and InsP3-induced Ca2+ Agulhon C, Petravicz J, McMullen B, Sweger EJ, Minton SK, Taves R,
release. Pathologically increased glial Ca2+ signaling can be et al. 2008. What is the role of astrocyte calcium in neurophysiology?
involved in generating epileptiform seizures. For example, Neuron 59:932–946.

CALCIUM SIGNALING IN NEUROGLIA • 329


Akopian G, Kressin K, Derouiche A, Steinhñuser C. 1996. Identified Fiacco TA, Agulhon C, Taves SR, Petravicz J, Casper KB, Dong
glial cells in the early postnatal mouse hippocampus display different X, et al. 2007. Selective stimulation of astrocyte calcium in situ
types of Ca2+ currents. Glia 17:181–194. does not affect neuronal excitatory synaptic activity. Neuron
Angulo MC, Kozlov AS, Charpak S Audinat E. 2004. Glutamate 54:611–626.
released from glial cells synchronizes neuronal activity in the hip- Finkbeiner SM. 1993. Glial calcium. Glia 9:83–104.
pocampus. J Neurosci 24:6920–6927. Ge WP, Yang XJ, Zhang Z, Wang HK, Shen W, Deng QD, et al.
Arcuino G, Lin JH, Takano T, Liu C, Jiang L, Gao Q, et al. 2002. 2006. Long-term potentiation of neuron-glia synapses mediated by
Intercellular calcium signaling mediated by point-source burst release Ca2+-permeable AMPA receptors. Science 312:1533–1537.
of ATP. Proc Natl Acad Sci U S A 99:9840–9845. Goldman, WF, Yarowsky PJ, Juhaszova M, Krueger BK, Blaustein MP.
Barajas M, Andrade A, Hernandez-Hernandez O, Felix R, Arias-Montano 1994. Sodium/calcium exchange in rat cortical astrocytes. J Neurosci
JA. 2008. Histamine-induced Ca2+ entry in human astrocytoma 14:5834–5843.
U373 MG cells: evidence for involvement of store-operated channels. Golovina VA. 2005. Visualization of localized store-operated calcium
J Neurosci Res 86:3456–3468. entry in mouse astrocytes. Close proximity to the endoplasmic retic-
Barres BA, Koroshetz WJ, Chun LL, Corey DP. 1990. Ion chan- ulum. J Physiol 564:737–749.
nel expression by white matter glia: the type-1 astrocyte. Neuron Grosche J, Matyash V, Moller T, Verkhratsky A, Reichenbach A,
5:527–544. Kettenmann H. 1999. Microdomains for neuron-glia interaction:
Bernardi P, Krauskopf A, Basso E, Petronilli V, Blachly-Dyson E, Di Lisa parallel fiber signaling to Bergmann glial cells. Nat Neurosci 2:
F, et al. 2006. The mitochondrial permeability transition from in 139–143.
vitro artifact to disease target. FEBS J 273:2077–2099. Guerini D, Coletto L, Carafoli E. 2005. Exporting calcium from cells.
Berridge MJ, Bootman MD, Roderick HL. 2003. Calcium signalling: Cell Calcium 38:281–289.
dynamics, homeostasis and remodelling. Nat Rev Mol Cell Biol Haas B, Schipke CG, Peters O, Sohl G, Willecke K, Kettenmann H.
4:517–529. 2006. Activity-dependent ATP-waves in the mouse neocortex are
Berridge MJ, Lipp P, Bootman, MD. 2000. The versatility and universal- independent from astrocytic calcium waves. Cereb Cortex 16:
ity of calcium signalling. Nat Rev Mol Cell Biol 1:11–21. 237–246.
Bolsover SR. 2005. Calcium signalling in growth cone migration. Cell Haberlandt C, Derouiche A, Wyczynski A, Haseleu J, Pohle J, Karram K,
Calcium 37:395–402. et al. 2011. Gray matter NG2 cells display multiple Ca 2+-signaling
Bregestovski P, Spitzer N. 2005. Calcium in the function of the nervous pathways and highly motile processes. PLoS One 6:e17575.
system: new implications. Cell Calcium 37:371–374. Hamilton N, Vayro S, Wigley R, Butt AM. 2010. Axons and astrocytes
Brini M, Carafoli E. 2009. Calcium pumps in health and disease. Physiol release ATP and glutamate to evoke calcium signals in NG2-glia.
Rev 89:1341–1378. Glia 58:66–79.
Burdakov D, Petersen OH, Verkhratsky A. 2005. Intraluminal cal- Hartel K, Singaravelu K, Kaiser M, Neusch C, Hulsmann S, Deitmer
cium as a primary regulator of endoplasmic reticulum function. Cell JW. 2007. Calcium influx mediated by the inwardly rectifying K+
Calcium 38:303–310. channel K ir4.1 (KCNJ10) at low external K+ concentration. Cell
Burgoyne RD, O’Callaghan DW, Hasdemir B, Haynes LP, Tepikin AV. Calcium 42:271–280.
2004. Neuronal Ca2+-sensor proteins: multitalented regulators of Hartmann J, Verkhratsky A. 1998. Relations between intracellular Ca2+
neuronal function. Trends Neurosci 27:203–209. stores and store-operated Ca 2+ entry in primary cultured human glio-
Burnashev N, Khodorova A, Jonas P, Helm PJ, Wisden W, Monyer H, blastoma cells. J Physiol (Lond) 513:411–424.
et al. 1992. Calcium-permeable AMPA-kainate receptors in fusiform Hassinger TD, Guthrie PB, Atkinson PB, Bennet MVL, Kater SB. 1997.
cerebellar glial cells. Science 256:1566–1570. An extracellular signaling component in propagation of astrocytic
Burnashev N, Zhou,Z, Neher E, Sakmann B. 1995. Fractional calcium calcium waves. Proc Natl Acad Sci U S A 93:13268–13273.
currents through recombinant GluR channels of the NMDA, AMPA Haydon PG, Lee S. 2011. The alleged glial controversy is no more:
and kainate receptor subtypes. J Physiol 485:403–418. Conditions under which astrocytic IP3/Ca 2+ signals do modulate
Carafoli E. 2004. The ambivalent nature of the calcium signal. synaptic plasticity. Trans Amer Soc Neurochem 15:3354–3365.
J Endocrinol Invest 27:134–136. Heilbrunn LV. 1943. An outline of general physiology. Philadelphia:
Case RM, Eisner D, Gurney A, Jones O, Muallem S, Verkhratsky A. Saunders.
2007. Evolution of calcium homeostasis: from birth of the first cell to Holtzclaw LA, Pandhit S, Bare DJ, Mignery GA, Russell JT. 2002.
an omnipresent signalling system. Cell Calcium 42:345–350. Astrocytes in adult rat brain express type 2 inositol 1,4,5-trisphosphate
Cornell Bell AH, Finkbeiner SM, Cooper MS, Smith SJ. 1990. Glutamate receptors. Glia 39:69–84.
induces calcium waves in cultured astrocytes: long- range glial signal- Hoth M, Penner R. 1992. Depletion of intracellular calcium stores acti-
ing. Science 247:470–473. vates a calcium current in mast cells. Nature 355:353–356.
Corvalan V, Cole R, de Vellis J, Hagiwara S. 1990. Neuronal modulation Hua X, Malarkey EB, Sunjara V, Rosenwald SE, Li WH, Parpura V.
of calcium channel activity in cultured rat astrocytes. Proc Natl Acad 2004. Ca 2+-dependent glutamate release involves two classes of
Sci U S A 87:4345–4348. endoplasmic reticulum Ca 2+ stores in astrocytes. J Neurosci Res 76:
Cotrina ML, Lin JH, Alves-Rodrigues A, Liu S, Li J, Azmi-Ghadimi H, 86–97.
Kang J, et al. 1998. Connexins regulate calcium signaling by control- Iadecola C, Nedergaard M. 2007. Glial regulation of the cerebral micro-
ling ATP release. Proc Natl Acad Sci U S A 95:15735–15740. vasculature. Nat Neurosci 10:1369–1376.
Dani JW, Chernjavsky A, Smith SJ. 1992. Neuronal activity trig- Ikura M, Osawa M, Ames JB. 2002. The role of calcium-binding pro-
gers calcium waves in hippocampal astrocyte networks. Neuron 8: teins in the control of transcription: structure to function. Bioessays
429–440. 24:625–636.
D’Ascenzo M, Vairano M, Andreassi C, Navarra P, Azzena GB, Grassi Innocenti B, Parpura V, Haydon PG. 2000. Imaging extracellular
C. 2004 Electrophysiological and molecular evidence of L-(Cav1), waves of glutamate during calcium signaling in cultured astrocytes.
N- (Cav2.2), and R- (Cav2.3) type Ca 2+ channels in rat cortical astro- J Neurosci 20:1800–1808.
cytes. Glia 45:354–363. Jaiswal JK. 2001. Calcium—how and why? J Biosci 26:357–363.
De Stefani D, Raffaello A, Teardo E, Szabo I, Rizzuto R. 2011. A for- Jeremic A, Jeftinija K, Stevanovic J, Glavaski A, Jeftinija S. 2001. ATP
ty-kilodalton protein of the inner membrane is the mitochondrial stimulates calcium-dependent glutamate release from cultured
calcium uniporter. Nature 476:336–340. astrocytes. J Neurochem 77:664–675.
Duff y S, MacVicar BA. 1994. Potassium-dependent calcium influx in Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. 2011. Physiology
acutely isolated hippocampal astrocytes. Neuroscience 61:51–61. of microglia. Physiol Rev 91:461–553.

330 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Kirischuk S, Kettenmann H, Verkhratsky A. 1997. Na+/Ca2+ exchanger Nilius B, Owsianik G, Voets T, Peters JA. 2007. Transient receptor
modulates kainate-triggered Ca 2+ signaling in Bergmann glial cells in potential cation channels in disease. Physiol Rev 87:165–217.
situ. FASEB J 11:566–572. Oliveira JF, Riedel T, Leichsenring A, Heine C, Franke H, Krugel U,
Kirischuk S, Kettenmann H, Verkhratsky A. 2007. Membrane currents et al. 2011. Rodent cortical astroglia express in situ functional P2X 7
and cytoplasmic sodium transients generated by glutamate transport receptors sensing pathologically high ATP concentrations. Cereb
in Bergmann glial cells. Pflugers Arch 454:245–252. Cortex 21:806–820.
Kirischuk S, Kirchhoff F, Matyash V, Kettenmann H, Verkhratsky A. Paluzzi S, Alloisio S, Zappettini S, Milanese M, Raiteri L, Nobile M, et al.
1999. Glutamate-triggered calcium signalling in mouse bergmann 2007. Adult astroglia is competent for Na+/Ca 2+ exchanger-operated
glial cells in situ: role of inositol-1,4,5-trisphosphate-mediated intrac- exocytotic glutamate release triggered by mild depolarization.
ellular calcium release. Neuroscience 92:1051–1059. J Neurochem 103:1196–1207.
Kirischuk S, Moller T, Voitenko N, Kettenmann H, Verkhratsky, A. Palygin O, Lalo U, Pankratov Y. 2011. Distinct pharmacological and
1995a. ATP-induced cytoplasmic calcium mobilization in Bergmann functional properties of NMDA receptors in mouse cortical astro-
glial cells. J Neurosci 15:7861–7871. cytes. Br J Pharmacol 163:1755–1766
Kirischuk S, Scherer J, Moller T, Verkhratsky A, Kettenmann H. 1995b. Palygin O, Lalo U, Verkhratsky A, Pankratov Y. 2010. Ionotropic
Subcellular heterogeneity of voltage-gated Ca2+ channels in cells of NMDA and P2X1/5 receptors mediate synaptically induced Ca2+ sig-
the oligodendrocyte lineage. Glia 13:1–12. nalling in cortical astrocytes. Cell Calcium 48:225–231.
Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. 2009. Parekh AB, Putney JW Jr. 2005. Store-operated calcium channels.
Synchronous hyperactivity and intercellular calcium waves in astro- Physiol Rev 85:757–810.
cytes in Alzheimer mice. Science 323:1211–1215. Parpura V, Grubisic V, Verkhratsky A. 2011. Ca 2+ sources for the exo-
Kuga N, Sasaki T, Takahara Y, Matsuki N, Ikegaya Y. 2011. Large-scale cytotic release of glutamate from astrocytes. Biochim Biophys Acta
calcium waves traveling through astrocytic networks in vivo. 1813:984–991.
J Neurosci 31:2607–2614. Parri HR, Crunelli V. 2003. The role of Ca2+ in the generation of sponta-
Lalo U, Pankratov Y, Kirchhoff F, North RA, Verkhratsky A. 2006. neous astrocytic Ca 2+ oscillations. Neuroscience 120:979–992.
NMDA receptors mediate neuron-to-glia signaling in mouse cortical Petersen OH, Michalak M, Verkhratsky A. 2005. Calcium signalling:
astrocytes. J Neurosci 26:2673–2683. past, present and future. Cell Calcium 38:161–169.
Lalo U, Pankratov Y, Parpura V, Verkhratsky A. 2011. Ionotropic Petravicz J, Fiacco TA, McCarthy KD. 2008. Loss of IP3 receptor-
receptors in neuronal-astroglial signalling: What is the role of dependent Ca2+ increases in hippocampal astrocytes does not
“excitable” molecules in non-excitable cells. Biochim Biophys Acta affect baseline CA1 pyramidal neuron synaptic activity.
1813:992–1002. J Neurosci 28:4967–4973.
Lalo U, Pankratov Y, Wichert SP, Rossner MJ, North RA, Kirchhoff F, Pivneva T, Haas B, Reyes-Haro D, Laube G, Veh RW, Nolte C, et al.
et al. 2008. P2X1 and P2X5 subunits form the functional P2X recep- 2008. Store-operated Ca 2+ entry in astrocytes: different spatial
tor in mouse cortical astrocytes. J Neurosci 28:5473–5480. arrangement of endoplasmic reticulum explains functional diversity
Latour I, Hamid J, Beedle AM, Zamponi GW, Macvicar BA. 2003. in vitro and in situ. Cell Calcium 43:591–601.
Expression of voltage-gated Ca2+ channel subtypes in cultured astro- Pizzo P, Lissandron V, Capitanio P, Pozzan T. 2011. Ca2+ signalling in
cytes. Glia 41:347–353. the Golgi apparatus. Cell Calcium 50:184–192.
Lin YC, Liu YC, Huang YY, Lien CC. 2010. High-density expression of Putney JW Jr. 1986. A model for receptor-regulated calcium entry. Cell
Ca 2+-permeable ASIC1a channels in NG2 glia of rat hippocampus. Calcium 7:1–12.
PLoS One 5:e12665. Putney JW Jr. 2007. Recent breakthroughs in the molecular mechanism
MacVicar BA, Tse FW. 1988. Norepinephrine and cyclic adenosine of capacitative calcium entry (with thoughts on how we got here).
3’:5’-cyclic monophosphate enhance a nifedipine-sensitive calcium Cell Calcium 42:103–110.
current in cultured rat astrocytes. Glia 1:359–365. Reyes RC, Parpura V. 2008. Mitochondria modulate Ca 2+-dependent glu-
Malarkey EB, Ni Y, Parpura V. 2008. Ca 2+ entry through TRPC1 chan- tamate release from rat cortical astrocytes. J Neurosci 28: 9682–9691.
nels contributes to intracellular Ca 2+ dynamics and consequent gluta- Reyes RC, Verkhratsky A, Parpura, V. 2012. Plasmalemmal Na+/Ca 2+
mate release from rat astrocytes. Glia 56:821–835. exchanger modulates Ca 2+-dependent exocytotic release of glutamate
Malarkey EB, Parpura V. 2009. Mechanisms of transmitter release from from rat cortical astrocytes. ASN Neurol 4(1):pii.
astrocytes. In: Parpura V, Haydon PG (eds.), Astrocytes in (patho) Rusakov DA, Zheng K, Henneberger C. 2011. Astrocytes as regulators
physiology of the nervous system. New York: Springer, pp. 301–350. of synaptic function: a quest for the Ca 2+ master key. Neuroscientist
Matute C. 2010. Calcium dyshomeostasis in white matter pathology. 17:513–523.
Cell Calcium 47:150–157. Scemes E, Giaume C. 2006. Astrocyte calcium waves: what they are and
Matute C, Alberdi E, Domercq M, Sanchez-Gomez MV, Perez-Samartin what they do. Glia 54:716–725.
A, Rodriguez-Antiguedad A, et al. 2007. Excitotoxic damage to white Schipke CG, Boucsein C, Ohlemeyer C, Kirchhoff F, Kettenmann H.
matter. J Anat 210:693–702. 2002. Astrocyte Ca2+ waves trigger responses in microglial cells in
Minelli A, Castaldo P, Gobbi P, Salucci S, Magi S, Amoroso S. 2007. brain slices. FASEB J 16:255–257.
Cellular and subcellular localization of Na+-Ca 2+ exchanger protein Seifert G, Steinhauser C. 2001. Ionotropic glutamate receptors in astro-
isoforms, NCX1, NCX2, and NCX3 in cerebral cortex and hip- cytes. Prog Brain Res 132:287–299.
pocampus of adult rat. Cell Calcium 41:221–234. Sharma G, Vijayaraghavan S. 2001. Nicotinic cholinergic signaling in
Morales AP, Carvalho AC, Monteforte PT, Hirata H, Han SW, Hsu hippocampal astrocytes involves calcium-induced calcium release
YT, et al. 2011. Endoplasmic reticulum calcium release engages Bax from intracellular stores. Proc Natl Acad Sci U S A 98:4148–4153.
translocation in cortical astrocytes. Neurochem Res 36:829–838. Shigetomi E, Bowser DN, Sofroniew MV, Khakh BS. 2008. Two forms
Nedergaard M, Rodriguez JJ, Verkhratsky A. 2010. Glial calcium and of astrocyte calcium excitability have distinct effects on NMDA
diseases of the nervous system. Cell Calcium 47:140–149. receptor-mediated slow inward currents in pyramidal neurons.
Newman EA, Zahs KR.1997. Calcium waves in retinal glial cells. Science J Neurosci 28:6659–6663.
275:844–847. Shigetomi E, Tong X, Kwan KY, Corey DP, Khakh BS. 2011. TRPA1
Nicholls DG. 2005. Mitochondria and calcium signaling. Cell Calcium channels regulate astrocyte resting calcium and inhibitory synapse
38:311–317. efficacy through GAT-3. Nat Neurosci 15:70–80.
Nicotera P, Petersen OH, Melino G, Verkhratsky A. 2007. Janus a Smyth JT, Dehaven WI, Jones BF, Mercer JC, Trebak M, Vazquez G,
god with two faces: death and survival utilise same mechanisms et al. 2006. Emerging perspectives in store-operated Ca 2+ entry: roles
conserved by evolution. Cell Death Diff 14:1235–1236. of Orai, Stim and TRP. Biochim Biophys Acta 1763:1147–1160.

CALCIUM SIGNALING IN NEUROGLIA • 331


Stys PK, Lipton SA. 2007. White matter NMDA receptors: an unex- Verkhratsky A. 2006. Calcium ions and integration in neural circuits.
pected new therapeutic target? Trends Pharmacol Sci 28:561–566. Acta Physiol (Oxf) 187:357–369.
Toescu EC, Moller T, Kettenmann H, Verkhratsky A. 1998. Long-term Verkhratsky A, Butt A. 2007. Glial neurobiology. A textbook. Chichester,
activation of capacitative Ca2+ entry in mouse microglial cells. UK: John Wiley & Sons.
Neuroscience 86:925–935. Verkhratsky A, Kettenmann H. 1996. Calcium signalling in glial cells.
Toescu EC, Verkhratsky A. 1998. Principles of calcium signallling. Trends Neurosci 19:346–352.
In: Verkhratsky A, Toescu EC (eds.), Integrative aspects of calcium Verkhratsky A, Kirchhoff F. 2007. NMDA receptors in glia.
signalling. New York: Plenum Press, pp. 2–22. Neuroscientist 13:28–37.
Tong XP, Li XY, Zhou B, Shen W, Zhang ZJ, Xu TL, et al. 2009. Verkhratsky A, Orkand RK, Kettenmann H. 1998. Glial calcium: home-
Ca 2+ signaling evoked by activation of Na+ channels and Na+/Ca2+ ostasis and signaling function. Physiol Rev 78:99–141.
exchangers is required for GABA-induced NG2 cell migration. J Cell Verkhratsky A, Steinhauser C. 2000. Ion channels in glial cells. Brain
Biol 186:113–128. Res Brain Res Rev 32:380–412.
Tuschick S, Kirischuk S, Kirchhoff F, Liefeldt L, Paul M, Verkhratsk Wang D, Yan B, Rajapaksha WR, Fisher TE. 2009. The expression of
A, et al. 1997. Bergmann glial cells in situ express endothelinB voltage-gated ca 2+ channels in pituicytes and the up-regulation of
receptors linked to cytoplasmic calcium signals. Cell Calcium 21: L-type ca 2+ channels during water deprivation. J Neuroendocrinol
409–419. 21:858–866.
Verkhratsky A. 2005. Physiology and pathophysiology of the cal- Xu JH, Long L, Tang YC, Hu HT, Tang FR. 2007. Cav1.2, Cav1.3, and
cium store in the endoplasmic reticulum of neurons. Physiol Rev Cav2.1 in the mouse hippocampus during and after pilocarpine-
85:201–279. induced status epilepticus. Hippocampus 17:235–251.

332 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
27.
THE CENTRAL ROLE OF ASTROCY TES IN
NEUROENERGETICS
Pierre J. Magistretti and Luc Pellerin

A B B R E VI AT I O N S state that after all the term neuroglia as it has been used in the
past is well adapted to describe a type of tissue which, while
ATP adenosine-5′-triphosphate being connective in nature as it connects elements of different
BDNF brain-derived neurotrophic factor nature and subserves the function of distributing nutrients,
CBF cerebral blood flow still is different from the connective tissue because of mor-
CREB cAMP response element binding protein phological and chemical characteristics as well as because of
DAB 1,4-dideoxy-1,4-imino-d-arabinitol its fundamentally different embryonic origin” [Dichiaro anzi
EAAT excitatory amino acid transporter che, dopo tutto, la parola nevroglia adoperata nel senso passato in
GABA gamma-aminobutyric acid uso mi sembra abbia titoli di preferenza, valendo ad indicare un
GDH glutamate dehydrogenase tessuto, che sebbene sia connettivo, perch connette elementi d’altra
GLAST glutamate aspartate transporter natura e alla sua volta serve alla distribuzione del materiale
GLT-1 glutamate transporter 1 nutritizio, pure si differenzia dal connettivo comune per carar-
GLUT glucose transporter reri morfologici, chimici, e quasi certamente, come diròin seguito,
LDH lactate dehydrogenase anche pel carattere fondamentale della diversa origine embrion-
MCT monocarboxylate transporter ale.@. Despite such an early suggestion for a role of astrocytes
mGluR metabotropic glutamate receptor in metabolic control, it was not until recently that the experi-
NMR nuclear magnetic resonance mental evidence supporting this early intuition was obtained.
PGE2 prostaglandin E2 Additional features such as the presence of specific glucose
PPP pentose phosphate pathway transporters (GLUT1) at the surface of astrocytes (Morgello
TCA tricarboxylic acid cycle et al. 1995; Yu and Ding 1998) favor the notion that these cells
VIP vasoactive intestinal peptide represent an active uptake site for circulating glucose into the
brain parenchyma. Other astrocytic processes ensheath syn-
apses, and express receptors and reuptake sites for a variety of
1 INTRODUCTION neurotransmitters (see chapters 16, 17, and 35) providing astro-
cytes with the capacity of sensing synaptic activity in order to
As highlighted in other chapters of this book, astrocytes come couple it with glucose utilization (Pellerin and Magistretti
in close contact with several cellular components of the brain 1994) and, as recently revealed, with the local regulation of
parenchyma including blood vessels, pial surfaces, neurons, blood flow (see chapter 37).
and other glial cells. Although astrocytes have been consid- This chapter reviews the mechanisms that have been dis-
ered traditionally as structural elements within the central covered during the last two decades supporting a central role
nervous system with the main function of maintaining the of astrocytes in brain energy metabolism, particularly as such
nervous tissue in place, hence their designation as neuroglia regulation occurs in register with synaptic activity.
or nerve glue, studies over the last two decades have high-
lighted a much more dynamic role for these cells. Already at
the end of the 19th century anatomists noticed the strategic 2 D I F F E R E N T A N D C O M P L E M E N TA RY
position occupied by astrocytes between neurons and blood M ETA B O L I C P R O F I L E S O F
vessels. It was also realized that astrocytes were sending spe- ASTROCY TES AND NEURONS
cific processes, called endfeet, in close contact with capillaries,
covering almost their entire surface as recently unequivocally There is now considerable evidence gathered from both in vitro
documented (Kacem et al. 1998). This arrangement sug- and in vivo studies that indicate clear differences in the meta-
gested that astrocytes might dynamically regulate the entry bolic profiles of astrocytes and neurons. Early studies on enzy-
and distribution of energy substrates to neurons, providing matic analyses of individually isolated cells (Hamberger and
an early argument in favor of their role in the regulation of Hydén 1963; Hydén and Lange 1962) as well as the most recent
brain energy metabolism (Andriezen 1893). This hypothesis gene expression analyses (Cahoy et al. 2008; Lovatt et al. 2007;
was eloquently formulated by Golgi (1886): “I would like to Rossner et al. 2006), indicate a predominance of glycolytic

333
and glycogen pathways in astrocytes and of lactate utilization in addition to glutamate (Yudkoff 1997). Glutamate formed
in neurons. For example, key enzyme and transporter isoforms through this mechanism would simply reintegrate the gluta-
such as lactate dehydrogenase and monocarboxylate transport- mate/glutamine cycle via its conversion to glutamine in astro-
ers are selectively expressed in each cell type (Bittar et al. 1996; cytes and its subsequent release. Another possibility is that
Laughton et al. 2007; O’Brien et al. 2007; Pellerin et al. 2005). astrocytes could export α-ketoglutarate directly to neurons
Functional studies in each cell type have consistently supported (Peng et al. 1993; Shank and Campbell 1984). Neurons would
the view of a predominant glycolytic activity in astrocytes themselves resynthesize glutamate either via a GDH-catalyzed
and an oxidative profile in neurons (Boumezbeur et al. 2010a; reaction or by a transamination of alanine by ALAT (alanine
Bouzier-Sore et al. 2006; Itoh et al. 2003; Lebon et al. 2002.). It aminotransferase). Notice that in these last two pathways,
also appears that neurons can efficiently use lactate as an energy use of α-ketoglutarate generated by astrocytes to resynthesize
substrate (Boumezbeur et al. 2010b; Bouzier et al. 2000; Qu glutamate could eventually lead to a depletion in TCA cycle
et al. 2000; Serres et al. 2005; Schurr et al. 1997) and even pref- intermediates. In order to avoid such a situation, it is neces-
erentially oxidize lactate over glucose when both substrates are sary for the astrocyte to form new TCA cycle intermediates
present (Bouzier-Sore et al. 2006; Itoh et al. 2003). downstream of α-ketoglutarate. This is done by an anaplerotic
Recent evidence provides an explanation for the limited reaction catalyzed by the enzyme pyruvate carboxylase that is
capacity of neurons to sustain glycolysis. Indeed, in con- found specifically in astrocytes (Shank et al. 1985). In this
trast to astrocytes, the enzyme 6-phosphofructose-2-kinase/ reaction, CO2 is fixed to pyruvate in order to form oxaloac-
fructose-2,6-bisphosphatase-3, which is a key positive modu- etate. In this manner, a complete astrocytic TCA cycle can be
lator of glycolysis, is virtually absent in neurons due to its maintained despite the loss of α-ketoglutarate for glutamate
constant proteasomal degradation (Almeida et al. 2004; synthesis. Finally, neurons were also shown to have the possi-
Herrero-Mendez et al. 2009). Consequently, neurons display a bility of synthesizing glutamate via TCA cycle intermediates
slower glycolytic rate than astrocytes and are unable to upregu- and replenish their intermediate pool by an anaplerotic reac-
late this pathway (Almeida et al. 2004; Herrero-Mendez et al. tion of CO2 fixation (Hassel and Brathe 2000).
2009). It appears that in neurons, the glucose flux is predomi- The important difference in their metabolic profiles, nota-
nantly through the pentose phosphate pathway (PPP)—which bly as far as glycolysis is concerned, is reflected in a larger
is essential for the production of NADPH and therefore for glucose utilization by astrocytes than neurons (Barros et al.
the maintenance of the cellular antioxidant potential. This 2009; Bouzier-Sore et al. 2006; Waagepetersen et al. 1998),
profile is consistent with the preferential use of lactate as an in particular during activation. Indeed, an increase in glucose
oxidative substrate by neurons, because it allows the sparing of uptake into astrocytes, mainly induced by glutamate, has been
glucose for the PPP, while providing, after conversion to pyru- widely confirmed since the original description by Pellerin and
vate, the fuel for the production of adenosine-5′-triphosphate Magistretti in 1994 (Bittner et al. 2011; Chuquet et al. 2010;
(ATP) by oxidative phosphorylation (Bolanos et al. 2010). Keller et al. 1996; Takahashi et al. 1995). In contrast, glucose
In terms of their specific metabolic profile, astrocytes transport into neurons is decreased upon glutamate exposure
are endowed with the capacity for the rapid removal of (Loaiza et al. 2003; Porras et al. 2004).
synaptically-released glutamate, a process primarily achieved A final note on cell-specific metabolism: as discussed in
by the astrocyte-specific sodium-dependent high affinity gluta- chapter 36 and in paragraph of this chapter, astrocytes are the
mate transporters GLT-1 and glutamate aspartate transporter exclusive site of glycogen metabolism in the brain.
(GLAST) corresponding to human excitatory amino acid
transporter EAAT2 and EAAT1, respectively (Bak et al. 2006).
Glutamate is converted to glutamine by the astrocyte-specific 3 G LU TA M AT E -M E D I AT E D
enzyme glutamine synthetase. After release and transfer to neu- T R A N S M I S S I O N D R I VE S E N E R GY
rons, glutamine is converted back to glutamate via deamination U T I L I Z AT I O N I N T H E B R A I N
by the neuron-specific enzyme phosphate-activated glutami-
nase in a metabolic pathway known as the glutamate-glutamine Nuclear magnetic resonance (NMR) studies have suggested
cycle (Bak et al. 2006; McKenna 2007). that there is a particular mechanistic link between the recy-
In addition to the glutamate/glutamine cycle, other path- cling of glutamate that occurs essentially in astrocytes, and
ways have been proposed to replenish the neuronal glutamate glucose metabolism that provides the energy to maintain neu-
pool; these mechanisms involve de novo synthesis of glutamate. ronal activity (Hyder et al. 2006; Sibson et al. 1998). From
One of these pathways is based on the ability of astrocytes to these experiments, it was also concluded that restoration of
provide the carbon backbone for the synthesis of glutamate ion gradients that occurs in parallel with glutamate/glutamine
from glucose as α-ketoglurate. Then, α-ketoglutarate formed cycling as a consequence of glutamatergic neurotransmis-
in the glial tricarboxylic acid cycle (TCA) cycle could be con- sion is responsible for approximately 80% of oxidative glu-
verted to glutamate upon addition of an amino group. This cose consumption under normal unanesthetized conditions.
amino group would be provided by either free ammonia In other words, processes not involved in neurotransmission
brought from the circulation in a reaction catalyzed by glu- but rather in maintenance of cell structure and function like
tamate dehydrogenase (GDH) or from leucine also imported protein and membrane turnover account for a rather small
from the circulation via a transamination mediated by leucine fraction of the total energy consumption. The same can be
transaminase leading to the formation of α-ketoisocaproate said for other neurotransmitter systems. In the specific case of

334 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
gamma-aminobutyric acid (GABA), which can be included in 5 A S T R O C Y T E -N E U R O N L AC TAT E
the glutamate/glutamine cycle rate because GABA is partly S H U T T L E : T H E M A I N N E U R O M ETA B O L I C
recycled in astrocytes via glutamine synthesis by a pathway COUPLING MECHANISM
known as the GABA shunt (McGeer and McGeer 1989), the
estimate is that it would not account for more than 11% of Astrocytes have a prominent capacity to spontaneously con-
the total glutamine flux in the rat (Rothman et al. 1999) or sume glucose and produce lactate under normoxia (Walz
ten times less than for glutamate in human cortex (Shen et al. and Mukerji 1988). Such a metabolic characteristic was first
1999). The essential conclusion is that the majority of energy observed on tumor cells and was termed aerobic glycolysis
consumption (which consists almost entirely of glucose oxida- by Otto Warburg in the 1920s (Warburg et al. 1924). Since
tion) is a direct reflection of the level of glutamatergic neuro- that time, it was demonstrated that aerobic glycolysis is not
transmission and its associated recycling involving astrocytes. restricted to cancer cells but it can be exhibited by normal cells
Interestingly, using a theoretical bottom-up approach, like astrocytes. Interestingly, it can be further enhanced under
Attwell and Laughlin estimated the relative energy cost of specific conditions in this cell type. Thus, it has been shown that
different cellular processes involved in brain activity both for glutamate, via a mechanism that involves its uptake through
rodents and primates. Attwell and Laughlin (2001) came to astrocyte-specific, high affinity Na+-dependent transporters
the conclusion that the large majority of energy consumption followed by an activation of the Na+/K+ ATPase, enhances
would be caused by excitatory signaling, mostly the reestablish- both glucose utilization and lactate production (Pellerin and
ment of ion gradients following action potential propagation Magistretti 1994). Based on this observation, it was suggested
and postsynaptic currents. Their value of 81% to 84% of energy that astrocytes might be endowed with the capacity to detect
cost attributed to excitatory (essentially glutamatergic) signal- synaptic activity at glutamatergic synapses and consequently
ing is in good agreement with NMR studies mentioned in the metabolize glucose into lactate that would be provided to
preceding. Moreover, estimation of glial energy cost, including neurons (Fig. 27.1). Evidence for such a central function, that
glutamate recycling, would account for approximately 6% of would be critically dependent on glial glutamate transporters,
total energy usage. These data offer guidelines to assign energy has been provided in vivo. Injection of antisense oligonucle-
consumption to specific processes and fix certain limitations otides directed against GLAST, one of the two glial gluta-
as well as proportions. What they do not provide, however, is mate transporters, led to a decrease of the glucose utilization
the mechanism(s) linking these energy consuming processes to response triggered by whisker stimulation in the somatosen-
energy generation and they do not take into account the pos- sory cortex (Cholet et al. 2001). Similarly, a reduction of the
sibility of cooperation between neurons and astrocytes. glucose utilization response upon activation of the whisker-
to-barrel pathway in knockout mice for either GLAST and
GLT-1, the two glial glutamate transporters, was observed in
4 TOWA R D A U N I F Y I N G C O N C E P T: animals at postnatal day 10 (Voutsinos-Porche et al. 2003a),
T H E R ET I N A M O D E L whereas a persistent decrease in metabolic response was
seen in adult GLT-1 knockout mice (Voutsinos-Porche et al.
Experiments performed in the honeybee retina had already 2003b). Furthermore, such findings were confirmed in both
suggested that a marked metabolic compartmentation occurs the visual cortex (Herard et al. 2005) as well as in the olfactory
between glial cells and photoreceptors. Thus, it was observed bulb (Martin et al. 2012).
that upon exposure to light, an increase in 2-deoxyglucose Several observations made in vitro as well as in vivo have
accumulation was taking place exclusively in glial cells while an provided further insights into the intracellular mechanism
increase in oxygen consumption, indicative of increased oxi- associated with the metabolic response of astrocytes. In par-
dative metabolism, occurred in photoreceptors (Tsacopoulos allel with the increase in glucose use triggered by glutamate
et al. 1988). These data suggested two important points: (1) uptake, it was demonstrated that glutamate causes a rapid
There must exist a neuronal signal released to trigger the stimulation of glucose transport in astrocytes (Loaiza et al.
increased glucose utilization in glial cells. (2) A substrate must 2003). The observed increase developed within 10 seconds.
be released by glial cells to serve as an oxidative fuel in photo- Such a fast time course would be consistent with the concept
receptors. These questions were investigated in both honey- of a rapid uptake and conversion of glucose into a metabolic
bee and mammal retinal preparations by the group of Marcos intermediate (e.g., lactate) to be released for neuronal use after
Tsacopoulos at Geneva University. It was observed that gluta- a brief activation typically encountered in the central ner-
mate (together with ammonia) constitutes the signal released vous system. Based on the known stoichiometry of glutamate
by photoreceptors to induce the metabolic response in Müller transporters that co-transport every glutamate with three Na+
glial cells of the retina. The critical step in the mechanism of ions, it was predicted that intracellular sodium concentration
activation is the conversion of glutamate into glutamine by changes should be a critical factor involved in the metabolic
glutamine synthetase and its associated ATP consumption response of astrocytes. Indeed, it was shown that glutamate
(Poitry et al. 2000). In the honeybee retina, it was observed uptake caused a significant increase of intracellular Na+ con-
that alanine was the metabolite released by glial cells to fuel centration in astrocytes (Chatton et al. 2000), and this in turn
photoreceptor oxidative metabolism (Tsacopoulos et al. was an essential condition for enhanced aerobic glycolysis
1994). In contrast, it was shown that in mammalian retina, lac- (Voutsinos-Porche et al. 2003a). As a consequence of the ele-
tate was the most likely candidate (Poitry-Yamate et al. 1995). vation in intracellular Na+ concentrations, a direct activation

T H E C E N T R A L R O L E O F A S T R O C Y T E S I N N E U R O E N E R G ET I C S • 335
A
Glutamatergic synapse Astrocyte Capillary

GLUTAMINE
ATP
LACTATE
Synaptic vesicles
ADP
GLUCOSE
GLUTAMATE GLUCOSE
H+
K+

Neuronal Na+ Glycolysis


ATP
glutamate K+
receptors ADP
NaK
LACTATE ATPase
a2

Na+

B GLAST +/+ GLAST –/–

650 700

20 20
nCulg

Figure 27.1 Astrocyte–Neuron Metabolic Interactions. A. Schematic representation of the astrocyte-neuron lactate shuttle (ANLS). Glutamate (Glu)
released at the synapse activates glutamatergic receptors (GluR) and is associated with important energy expenditures in neuronal compartments.
A large proportion of the glutamate released at the synapse is taken up by astrocytes via excitatory amino acid transporters (EAATs, more specifically
GLT-1 and GLAST) together with 3 Na+ ions. This Na+ is extruded by the action of the Na+/K+ ATPase, consuming ATP. This triggers nonoxida-
tive glucose utilization in astrocytes and glucose uptake from the circulation through the glucose transporter GLUT1 expressed by both capillary
endothelial cells and astrocytes. Glycolytically derived pyruvate is converted to lactate by lactate dehydrogenase 5 (LDH5; mainly expressed in astro-
cytes) and shuttled to neurons through monocarboxylate transporters (mainly MCT1 and MCT4 in astrocytes and MCT2 in neurons). In neurons,
this lactate can be used as an energy substrate following its conversion to pyruvate (Pyr) by LDH1 (mainly expressed in neurons). Neurons can also
take up glucose via the neuronal glucose transporter 3 (GLUT3). Concomitantly, astrocytes participate in the recycling of synaptic glutamate via the
glutamate-glutamine cycle. Following its uptake by astrocytes, glutamate is converted to glutamine (gln) by the action of glutamine synthetase (GS)
and shuttle to neurons where it is converted back to glutamate by glutaminases (GLS). B. Representative pseudocolored digi-tized autoradiograms
were obtained either from antero-posterior coronal sections at the level of the somatosensory barrel field or from tangential sections through layer IV
of the primary somatosensory cortex. The level of 2-DG uptake is color coded according to the respective colored scales. Unilateral left or bilateral
C1C2 whisker stimulation in GLAST+/+ and GLT-1+/+ mice produced a local increase in 2-DG uptake in the right somatosensory cortex (white
square) in control, but not GLAST deficient mice. Modified from Pellerin and Magistretti 1994 and Voutsinos-Porche et al. 2003a.

of the Na+/K+ ATPase was evidenced (Pellerin and Magistretti associated metabolic response. These data clearly identify glu-
1997) together with a decrease of its association with acety- tamate uptake and the subsequent activation of a particular
lated tubulin (Casale et al. 2003), an indication of a switch subunit of the Na+/K+ ATPase as key elements in the coupling
from inactive to activated state. Moreover, it was suggested mechanism involving astrocytes, although other signals and
from in vitro experiments that a particular catalytic subunit mechanisms (e.g. potassium) could also contribute to rapid
of the Na+, K+ ATPase, akin to the α2 subunit, is specifically and reversible glycolytic activation (Bittner et al. 2011).
activated upon glutamate uptake in astrocytes (Pellerin and
Magistretti 1997). In vivo, the distribution of the α2 subunit
as observed both at the light and electron microscopic lev- 6 L AC TAT E A S T H E P R E F E R R E D E N E R GY
els revealed not only that it is expressed more specifically by S U B S T R AT E F O R N E U R O N S
astrocytes but that it is found on processes that ensheath glu-
tamatergic but not GABAergic synapses (Cholet et al. 2002). Since the pioneering work of Henry McIlwain in the 1950s,
Moreover, a strong colocalization between the Na+/K+ ATPase evidence for the use of lactate as an energy substrate by neu-
α2 subunit and both glial glutamate transporters GLAST and rons has been accumulating (for review, see Bouzier-Sore et al.
GLT-1 was observed, suggesting a close association aimed 2002; Pellerin 2003). Such evidence was provided by different
at maximizing the efficiency of glutamate transport and its approaches ranging from oxidation rates measured in cultured

336 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
cells to NMR spectroscopy in vivo. One consistent and system- by neurons in a manner analogous to glucose (Boumezbeur
atic finding is the fact that lactate is efficiently oxidized to CO2 et al. 2010b). In addition, a direct demonstration that lactate is
in different neuronal preparations and is usually preferred to a preferential neuronal energy substrate in vivo has been pro-
glucose for this purpose. Experiments performed with brain vided in rat (Wyss et al. 2011).
slices (Fernandez and Medina 1986; Ide et al. 1969), cul- Functional evidence has also been provided to support
tured telencephalic neurons (McKenna et al. 2001; Tabernero the notion that lactate is used, in conjunction with low levels
et al. 1996; Vicario et al. 1991), aggregated neuronal cultures of glucose, to support neuronal activity. Several studies per-
(Honegger et al. 2002), sympathetic ganglia (Larrabee 1983, formed on hippocampal slices have highlighted not only the
1992, 1995, 1996) as well as synaptic terminals (McKenna et al. neuroprotective action of lactate but also its ability to main-
1993, 1994, 1998) have all reached the same conclusion. Lactate tain synaptic function under conditions where glucose avail-
was also able to sustain the same respiration rate attained with ability is reduced (Cater et al. 2001; Fowler, 1993; Ivanov et al.
glucose as determined by measurement of mitochondrial dehy- 2011; Izumi et al. 1994, 1997a,b; Rouach et al. 2008; Schurr
drogenase activity (Pellerin et al. 1998a). Recent studies have et al. 1988, 1999). Similarly, it was shown that lactate restores
further confirmed and extended these findings. Sokoloff and excitability of glucoresponsive neurons from hypothalamic
colleagues have demonstrated that neurons in culture display and brainstem nuclei following glucose deprivation (Ainscow
a kinetic preference for oxidation of extracellular lactate over et al. 2002; Himmi et al. 2001; Mobbs et al. 2001). In vivo,
lactate/pyruvate produced intracellularly from glucose via gly- the usefulness of lactate to preserve cognitive functions dur-
colysis (Itoh et al. 2003). In contrast, they showed that it was ing controlled hypoglycemic episodes has been demonstrated
not the case for cultured astrocytes that exhibit an intense gly- (King et al. 1998; Maran et al. 1994, 2000), thus further
colytic activity with a predominance of lactate release rather strengthening the concept of lactate as a significant and valu-
than use. Use of NMR spectroscopy to investigate this ques- able fuel to sustain neuronal activity and brain function. As
tion has also provided unequivocal answers. Both GABAergic further discussed in paragraph 7, lactate transfer from astro-
and glutamatergic neurons in culture were shown to actively cytes to neurons is required for long-term memory (Suzuki
metabolize lactate through the TCA cycle as determined by et al. 2011).
the labeling from lactate of numerous TCA cycle intermedi-
ates and several related amino acids (Schousboe et al. 1997;
Waagepetersen et al. 2000). It was also estimated that com- 7 M O N O C A R B OXY L AT E T R A N S P O RT E R S :
pared to glucose, lactate must have an equivalent access to the M A I N G AT E S F O R L AC TAT E
TCA cycle (Waagepetersen et al. 1998). A quantitative assess- TRAFFICKING
ment of the concomitant oxidative use of lactate and glucose
was performed by NMR spectroscopy on cultured telenceph- Lactate, together with pyruvate and the ketone bodies
alic neurons (Bouzier-Sore et al. 2003, 2006). When both acetoacetate and E-hydroxybutyrate, belongs to a group of
substrates were present at an equimolar concentration (either compounds known as monocarboxylates. Because of their
1.1 or 5.5 mM), it was determined that 79% of neuronal oxi- hydrophilic nature, these substances require a transporter to
dative metabolism was supported by lactate while only 21% cross cellular membranes. A family of proton-linked mono-
relied on glucose-derived pyruvate. Such observations leave no carboxylate transporters has been described. It contains
doubt that at least in vitro, lactate represents a prominent and 14 members sharing sequence homologies and identified as
preferential oxidative energy substrate for neurons. monocarboxylate transporter (MCT)1 to 9 and MCT11
Some indications have been provided to suggest that it is to 14 as well as another member known as TAT1 (Drewes
also the case in vivo. Measurements performed using either 2003; Halestrap and Meredith, 2004). Of those nine trans-
microdialysis or lactate-sensitive microelectrodes have shown porters, only MCT1, MCT2, MCT3 and MCT4 have been
enhanced lactate utilization following either electrical or functionally characterized and proven to act as monocar-
behavioral activation (Fellows et al. 1993; Hu and Wilson boxylate carriers. Kinetically, MCT2 was found to have the
1997). Observations in humans have revealed a reduction in lowest Km and thus the highest affinity for its substrates while
cerebral glucose utilization upon raising plasma lactate levels MCT4 displayed a very high Km. In the central nervous sys-
(Smith et al. 2003). More direct evidence of lactate utilization tem, the presence of MCT1, MCT2 and MCT4 has been
by the brain was provided by NMR spectroscopy. It was shown detected both at the mRNA and protein levels (Bergersen
that following intravenous injection in rat, labeled [U-(13C)] et al. 2001; Jackson et al. 1997; Koehler-Stec et al. 1998;
lactate was readily metabolized once it had entered the brain Pellerin et al. 1998b). Studies on the cellular distribution of
in a compartment deprived of pyruvate carboxylase activity these MCTs by immunohistochemistry, both in vitro and
(Bouzier et al. 2000; Hassel and Brathe 2000). Because pyru- in vivo, revealed a specific pattern. Thus, in addition to a
vate carboxylase expression is restricted to astrocytes, it was strong expression on blood vessels (Gerhart et al. 1997, 1998)
inferred that lactate utilization must take place principally and on oligodendrocytes (Rinholm et al. 2011), MCT1
in neurons. Furthermore, from the labeling pattern of key was found to be highly expressed by astrocytes, both in cul-
compounds such as glutamine, glutamate, and GABA, it was tures (Bröer et al. 1997; Debernardi et al. 2003; Hanu et al.
concluded that lactate is prominently used in glutamatergic 2000) and in vivo (Hanu et al. 2000; Pierre et al. 2000). A
neurons (Qu et al. 2000). Similarly, it was shown that plasma small but significant expression was also found in cultured
lactate is readily taken up by the human brain and oxidized cortical neurons (Debernardi et al. 2003) and by some neurons

T H E C E N T R A L R O L E O F A S T R O C Y T E S I N N E U R O E N E R G ET I C S • 337
in vivo (Leino et al. 1999). The predominant neuronal mono- Quite importantly, it was shown that a doubling of MCT2
carboxylate transporter both in vitro and in vivo was found to expression at the cell surface leads to an increase of approxi-
be MCT2 (Bergersen et al. 2001; Bröer et al. 1997; Debernardi mately 80% in lactate transport (Pierre et al. 2009). Thus,
et al. 2003; Pierre et al. 2000, 2002). Finally, MCT4 is changes in expression and localization of MCT2 induced by
expressed by Bergmann glia in the cerebellum (Bergersen et al. neuroactive signals cause significant modifications of neuronal
2001) as well as by astrocytes in numerous brain areas includ- energetics. Since such adaptations appear to occur quite rapidly
ing the hippocampus (Pellerin et al. 2005; Rafiki et al. 2003). (<1 minute measured with static approaches), it is likely that
Such a cell-specific distribution is paralleled by an enriched they represent the most appropriate process by which neurons
expression of particular lactate dehydrogenase (LDH) iso- could adapt their substrate supply and concomitant energy
forms displaying different kinetic characteristics. Isoforms production to face changing needs associated with fluctuating
containing the LDHB subunits are more abundant in neurons activity. It is expected that in the case of synaptic plasticity,
while isoforms enriched in LDHA subunits are enriched in changes in synaptic efficacy will be accompanied by adapta-
astrocytes (Bittar et al. 1996; O’Brien et al. 2007). Association tions in energy supply. The AMPA type glutamate receptor
of the high affinity transporter MCT2 with LDH isoforms subunit GluR2 is known to participate to the mechanism of
enriched in LDHB subunits creates kinetic conditions highly synaptic plasticity at glutamatergic synapses (Isaac et al. 2007).
favorable for lactate uptake and utilization by neurons. The Observations that MCT2 not only interacts with GluR2 but
presence of LDH isoforms predominantly exhibiting LDHA modifies its subcellular distribution and expression levels sup-
subunits together with the high capacity monocarboxylate port the idea that energy supply might be tightly regulated as
transporters MCT1 and MCT4 rather provide optimal set- part of the mechanism of synaptic plasticity (Maekawa et al.
tings to favor lactate production and release from astrocytes. 2009; Pierre et al. 2009). Indeed, it was recently demonstrated
Considering the preferential metabolic profile exhibited by that lactate supply to neurons via MCT2 is critical for both
each cell type as described above, these characteristics rein- short-term and long-term memory formation (Newman et al.
force the concept that lactate is shuttled preferentially from 2011; Suzuki et al. 2011(see section 8).
astrocytes to neurons, at varying degrees depending on the
activation state.
Energy substrate utilization can be limited by the trans- 8 A S T R O C Y T I C G LYC O G E N : I T S R O L E
port capacity. The possibility of shuttling lactate between I N N E U R OT R A N S M I S S I O N A N D
brain cell types is determined by the expression of specific SY N A P T I C P L A S T I C I T Y
transporters exhibiting different kinetics. Indeed, modeling
studies have demonstrated the key role that monocarboxy- In addition to glutamate-stimulated aerobic glycolysis as
late transporters play in regulating lactate influx and efflux described by the ANLS, another astrocyte-based metabolic
(Aubert et al. 2005). It was previously shown that this could pathway can produce lactate, namely glycogenolysis. Glycogen
be the case for neuronal lactate utilization since MCT2 over- is almost exclusively localized in astrocytes (Magistretti 2008).
expression in cultured neurons using viral vectors promoted Glycogen is not found in adult neurons, except occasionally in
lactate utilization in these cells when they were stimulated large neurons in the brainstem or in the peripheral nervous sys-
with kainate (Bliss et al. 2004). It became of interest to deter- tem (Sotelo and Palay 1968) (see also chapter 36). Surprisingly,
mine whether the expression of monocarboxylate transport- however, glycogen synthase—the enzyme responsible for gly-
ers could be regulated in brain cells. In astrocytes, it was cogen synthesis—is present in many neurons; it is however
recently shown that MCT4 expression is under the control permanently degraded by proteasome-dependent mechanism,
of the neuromodulator nitric oxide (Marcillac et al. 2011). mediated by the malin–laforin complex (Vilchez et al. 2007).
In parallel, it was found that MCT2 expression in neurons This complex regulation is vital for neurons, as shown by the
is under the regulation of several neuroactive substances. fact that interventions aimed at maintaining glycogen syn-
Noradrenaline, insulin, insulin-like growth factor-1, and thase function and therefore glycogen synthesis in neurons,
brain-derived neurotrophic factor (BDNF) were all shown to lead to neuronal apoptosis. A clinical condition, progressive
enhance MCT2 expression in cultured neurons (Chenal and myoclonus epilepsy (also known as Lafora disease) is caused
Pellerin 2007; Chenal et al. 2008; Robinet and Pellerin 2010). by a loss-of-function mutation in the malin/laforin complex,
The mechanism involves a stimulation of translation via the and is characterized by accumulation of glycogen-containing
PI3K/Akt/mTOR/S6 pathway. Moreover, it was shown that deposits in neurons.
such activation occurs at the synaptic level (Robinet and Astrocytic glycogen is subject to a tight regulation
Pellerin 2010). The effect of BDNF on MCT2 expression by a restricted number of neurotransmitters/modulators
was recently confirmed in vivo after its injection into the (Magistretti et al. 1981; Magistretti and Morrison 1988). Thus
hippocampus (Robinet and Pellerin 2011). In addition to noradrenaline (NA), vasoactive intestinal peptide (VIP) and
changes in the overall expression levels, evidence was provided adenosine promote glycogenolysis in astrocytes. Because of
that the amount of MCT2 proteins present at the cell sur- the particular morphology of VIP-containing neurons in the
face can be modified. It was shown that translocation of cerebral cortex, the metabolic action of VIP is restricted to
MCT2 from an endogenous pool to the cell surface can be cortical columnar modules, while that of noradrenaline, which
induced in cultured cortical neurons by exposing them to a is released from noradrenergic intracortical horizontally-
combination of glutamate and glycine (Pierre et al. 2009). oriented fibers, can prime metabolically the neocortex globally

338 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
(Magistretti and Morrison 1988) (Fig. 27.2). Noradrenaline and a corollary, downregulation of the expression of the astrocyte-
VIP interact synergistically to produce radially-oriented corti- specific lactate transporter MCT4 also resulted in memory
cal “metabolic hot-spots” (Magistretti and Schorderet 1984). inhibition, which was rescued by local administration of lactate
Thus, the astrocyte-based glycogenolysis mediated by VIP, but not by equicaloric glucose. In contrast, inhibition of expres-
NA, or adenosine (Magistretti et al 1986) represents another sion of the neuron-specific lactate transporter MCT2 although
mechanism to couple neuronal activity to energy metabolism also leading to amnesia was not subject to rescue by exogenous
by providing additional energy substrates to active neurons lactate, indicating that lactate import into neurons is necessary
(Bélanger et al. 2011; Magistretti 2008). Consistent with this for long-term memory formation. Furthermore, in parallel
view, stimulation of the whisker-to-barrel pathway promotes with the behavioral effects observed, the in vivo induction of
glycogenolysis in the barrel field of layer IV somatosensory long-term potentiation and of genes known to be required for
cortex (Swanson et al. 1992). Maintenance of sustained axonal memory formation such as phospho-CREB, activity regulated
spiking in mouse optic nerve, depends on adequate glycogen cytoskeletal-associated protein, and phospho-cofilin was pre-
content and lactate shuttling between astrocytes and the axonal vented by inhibition of glycogenolysis and MCT4 expression
fibers (Tekkök et al. 2005). in a lactate-reversible manner (Suzuki et al. 2011). These data
Recent in vivo data have demonstrated a central role of clearly demonstrate that astrocyte to neuron lactate transfer is
glycogenolysis and the resulting release of lactate from astro- required for long-term memory formation (Suzuki et al. 2011).
cytes in synaptic plasticity in a learning paradigm (Suzuki et al. Similar results have been obtained in a different (working
2011). Indeed, inhibition of glycogenolysis in the hippocampus memory) learning paradigm (Newman et al. 2011).
by stereotaxic administration of the glycogen phosphorylase
inhibitor DAB (1,4-dideoxy-1,4-imino-d-arabinitol) (Walls
et al. 2008) prevents the establishment of long-term memory 9 ROLE OF ASTROCY TE IN THE
in an inhibitory avoidance learning paradigm. Since glycog- R E GU L AT I O N O F L O C A L C E R E B R A L
enolysis results in lactate production by astrocytes, rescue of B L O O D F L OW
the DAB effect was attempted by local administration of lactate;
under these conditions long-term memory was reestablished. As As noted in the first paragraph of this chapter, astrocytes are
ideally positioned to sense synaptic activity and provide the
appropriate metabolic supply notably by interacting with
intraparenchymal capillaries. Accordingly, several lines of
experimental evidence indicate that astrocytes play a key role
in neurovascular coupling (see also chapter 37).
I
Since the original observation made by Sherrington reveal-
ing neurovascular coupling (Roy and Sherrington 1890), a
II variety of vasoactive agents, including H+, K+, neurotransmit-
& ters, adenosine, arachidonic acid metabolites and nitric oxide
III have been implicated in the activity-dependent increase in
VIP
cerebral blood flow (CBF) (Attwell et al. 2010; Carmignoto
and Gomez-Gonzalo 2010; Gordon et al. 2008; Iadecola and
IV
Nedergaard 2007). It is now clear that several of these mecha-
? nisms entail glutamate signaling on astrocytes for vasomotor
responses, both vasoconstriction and vasodilation (Attwell
NA SA
V et al. 2010; Carmignoto and Gomez-Gonzalo 2010; Iadecola
and Nedergaard 2007; Takano et al. 2006).
VI
Glutamate receptors of the metabotropic type (mGluR)
Pyr
activate Ca2+ transients in astrocytes resulting in activation of
SP cytosolic phospholipase A2, triggering the formation of vaso-
AF ECIF dilating agents such as epoxyeicosatrienoic acids and prosta-
FE IC
(SARE glandin E2 (PGE2) from arachidonic acid (Attwell et al. 2010;
)
W.M. AFFEREN
T Takano et al. 2006; Zonta et al. 2003). Arachidonic acid per
REN ERGIC
NORAD
(NA) se, can diffuse to smooth muscle cells and cause vasoconstric-
EFFERENT tion after its conversion to 20-hydroxyeicosatetraenoic acid
(Metea and Newman 2006; Mulligan and MacVicar 2004).
Figure 27.2 Functional Domains for Glycogenolysis Induced by VIP and Energy metabolism seems to also play a modulatory role in the
NA in Astrocytes. The bipolarly oriented VIP neurons promote glycog- effects of prostanoids (Gordon et al. 2008). Thus, the direc-
enolysis locally within cortical columns, while the noradrenergic fibers tionality of the vasomotor effect of the astrocyte-derived vaso-
which have a tangentially organized trajectory promote glycogenolysis active molecule PGE2 depends on the activity of astrocytes.
globally across functionally-distinct regions of the cortex. The metabolic Indeed extracellular lactate produced by astrocytes inhibits the
effect of VIP and NA is exerted in astrocytes, the only brain cells that
contain glycogen, resulting in lactate release. From Magistretti and clearance of PGE2, thus promoting vasodilation. In contrast,
Morrison. 1988. when extracellular lactate is low vasoconstriction prevails. In

T H E C E N T R A L R O L E O F A S T R O C Y T E S I N N E U R O E N E R G ET I C S • 339
addition, adenosine released by astrocytes under low-oxygen Barros LF, Courjaret R, Jakoby P, Loaiza A, Lohr C, Deitmer JW. 2009.
inhibits astrocyte-mediated vasoconstrictions at the level of Preferential transport and metabolism of glucose in Bergmann glia
over Purkinje cells: a multiphoton study of cerebellar slices. Glia
smooth muscle cells by blocking the effect of arachidonic acid. 57:962–970.
Interestingly, it was recently observed that increased blood Bélanger M, Allaman I, Magistretti PJ. 2011. Brain energy metabo-
flow in activated brain regions is strongly correlated with lism: focus on astrocyte-neuron metabolic cooperation. Cell Metab
elevations in lactate concentrations, but negatively correlated 14:724–738.
with oxygen consumption, further suggesting that increased Bergersen L, Waerhaug O, Helm J, Thomas M, Laake P, Davies AJ, et al.
2001. A novel postsynaptic density protein: the monocarboxylate
CBF is driven by glycolytic, as opposed to oxidative metabo- transporter MCT2 is co-localized with d-glutamate receptors in
lism under normal physiological conditions (Lin et al. 2010). postsynaptic densities of parallel fiber-Purkinje cell synapses. Exp
Brain Res 136:523–534.
Bittar PG, Charnay Y, Pellerin L, Bouras C, Magistretti PJ. 1996.
10 S U M M A RY A N D P E R S P E C T I VE S Selective distribution of lactate dehydrogenase isoenzymes in neu-
rons and astrocytes of human brain. J Cereb Blood Flow Metab
16:1079–1089.
This brief overview highlights the emerging role of astrocytes Bittner CX, Valdebenito R, Ruminot I, Loaiza A, Larenas V,
not only in neurometabolic but also in neurovascular coupling Sotelo-Hitschfeld T, et al. 2011. Fast and reversible stimulation of
and strongly supports the notion that astrocytes are impor- astrocytic glycolysis by K+ and a delayed and persistent effect of glu-
tant elements in the production of functional neuroimaging tamate. J Neurosci 31:4709–4713.
Bliss TM, Ip M, Cheng E, Minami M, Pellerin L, Magistretti P, et al.
signals in relation to synaptic activity (Figley and Stroman 2004. Dual-gene, dual-cell type therapy against an excitotoxic insult
2011; Magistretti and Pellerin 1996). The main challenges for by bolstering neuroenergetics. J Neurosci 24:6202–6208.
the future concern the exploration of the importance of this Bolanos JP, Almeida A, Moncada S. 2010. Glycolysis: a bioenergetics or a
role in neuroenergetics for astrocytes in the context of vari- survival pathway ? Trends Biochem Sci 35:145–149.
ous physiological and pathological situations. Indeed, it has Boumezbeur F, Mason GF, de Graaf RA, Behar KL, Cline GW,
Shulman GI, et al. 2010a. Altered brain mitochondrial metabolism
been suggested that astrocytes via their neurometabolic cou- in healthy aging as assessed by in vivo magnetic resonance spectros-
pling role could be important for various functions of the copy. J Cereb Blood Flow Metab 30:211–221.
central nervous system such as regulation of blood glucose Boumezbeur F, Petersen KF, Cline GW, Mason GF, Behar KL, Shulman
levels (Lam et al. 2005), arousal (Parsons and Hirasawa 2010), GI, et al. 2010b. The contribution of blood lactate to brain energy
respiratory control (Erlichman et al. 2008), sodium homeo- metabolism in humans measured by dynamic 13C nuclear magnetic
resonance spectroscopy. J Neurosci 30:13983–13991.
stasis (Shimizu et al. 2007), or memory formation (Newman Bouzier AK, Th iaudiere E, Biran M, Rouland R, Canioni P, Merle M.
et al. 2011; Suzuki et al. 2011). Moreover, targeting astrocytes 2000. The metabolism of [3-(13)C]lactate in the rat brain is specific
through an enhancement of their metabolic capacities could of a pyruvate carboxylase-deprived compartment. J Neurochem
represent a valuable therapeutic strategy in various neurode- 75:480–486.
generative diseases for which a deficit in energetics has been Bouzier-Sore AK, Merle M, Magistretti PJ, Pellerin L. 2002. Feeding
active neurons: (re)emergence of a nursing role for astrocytes. J Physiol
reported. Such an approach has been shown to be effective (Paris) 96:273–282.
in the case of excitotoxicity (Bliss et al. 2004) and metabolic Bouzier-Sore AK, Voisin P, Canioni P, Magistretti PJ, Pellerin L. 2003.
insult (Escartin et al. 2007), but other possibilities exist and Lactate is a preferential oxidative energy substrate over glucose for
could be already indirectly exploited, as is the case for cogni- neurons in culture. J Cereb Blood Flow Metab 23:1298–1306.
tive enhancers (Pellerin and Magistretti 2005). Bouzier-Sore AK, Voisin P, Bouchaud V, Bezancon E, Franconi JM,
Pellerin L. 2006. Competition between glucose and lactate as oxida-
tive energy substrates in both neurons and astrocytes: a comparative
NMR study. Eur J Neurosci 24:1687–1694.
REFERENCES Bröer S, Rahman B, Pellegri G, Pellerin L, Martin JL, Verleysdonk S,
et al. 1997. Comparison of lactate transport in astroglial cells and
Ainscow EK, Mirshamsi S, Tang T, Ashford MLJ, Rutter GA. 2002. monocarboxylate transporter 1 (MCT1) expressing Xenopus laevis
Dynamic changes of free cytosolic ATP concentration during fuel oocytes: expression of two different monocarboxylate transporters in
sensing by rat hypothalamic neurones: evidence for ATP-independent astroglial cells and neurons. J Biol Chem 272:30096–30102.
control of ATP-sensitive K+ channels. J Physiol 544:429–445. Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL,
Almeida A, Moncada S, Bolaños JP. 2004. Nitric oxide switches on glyco- Christopherson KS, et al.. 2008. A transcriptome database for astro-
lysis through the AMP protein kinase and 6-phosphofructo-2-kinase cytes, neurons, and oligodendrocytes: a new resource for understand-
pathway. Nat Cell Biol 6:45–51. ing brain development and function. J Neurosci 28:264–278.
Andriezen WL. 1893. On a system of fibre-like cells surrounding the Carmignoto G, Gomez-Gonzalo M. 2010. The contribution of astrocyte
blood vessels of the brain of man and mammals, and its physiological signaling to neurovascular coupling. Brain Res Rev 63:138–148.
significance. Int Monatsschr Anat Physiol 10:532–540. Casale CH, Previtali G, Barra HS. 2003. Involvement of acetylated
Attwell D, Buchan AM, Charpak S, Lauritzen M, MacVicar BA, tubulin in the regulation of Na+, K+-ATPase activity in cultured
Newman EA. 2010. Glial and neuronal control of brain blood flow. astrocytes. FEBS Lett 534:115–118.
Nature 468:232–243. Cater HL, Benham CD, Sundstrom LE. 2001. Neuroprotective role of
Attwell D, Laughlin SB. 2001. An energy budget for signaling in the grey monocarboxylate transport during glucose deprivation in slice cul-
matter of the brain. J Cereb Blood Flow Metab 21:1133–1145. tures of rat hippocampus. J Physiol 531:459–466.
Aubert A, Costalat R, Magistretti PJ, Pellerin L. 2005. Brain lactate Chatton JY, Marquet P, Magistretti PJ. 2000. A quantitative anal-
kinetics: Modeling evidence for neuronal lactate uptake upon activa- ysis of L-glutamate-regulated Na+ dynamics in mouse cortical
tion. Proc Natl Acad Sci U S A 102:16448–16453. astrocytes: implications for cellular bioenergetics. Eur J Neurosci
Bak LK, Schousboe A, Waagepetersen HS. 2006. The glutamate/ 12:3843–3853.
GABA-glutamine cycle: aspects of transport, neurotransmitter Chenal J, Pellerin L. 2007. Noradrenaline enhances the expression of
homeostasis and ammonia transfer. J Neurochem 98:641–653. the neuronal monocarboxylate transporter MCT2 by translational

340 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
activation via stimulation of PI3K/Akt and the mTOR/S6K pathway. superior colliculus of mice lacking the glial glutamate transporter
J Neurochem 102:389–397. GLT-1. Eur J Neurosci 22:1807–1811.
Chenal J, Pierre K, Pellerin L. 2008. Insulin and IGF-1 enhance the Herrero-Mendez A, Almeida A, Fernández E, Maestre C, Moncada S,
expression of the neuronal monocarboxylate transporter MCT2 Bolaños JP. 2009. The bioenergetic and antioxidant status of neurons
by translational activation via stimulation of the phosphoinosit- is controlled by continuous degradation of a key glycolytic enzyme by
ide 3-kinase-Akt-mammalian target of rapamycin pathway. Eur J APC/C-Cdh1. Nat Cell Biol 11:747–752.
Neurosci 27:53–65. Himmi T, Perrin J, Dallaporta M, Orsini JC. 2001. Effects of lactate on
Cholet N, Pellerin L, Magistretti PJ, Hamel E. 2002. Similar perisynap- glucose-sensing neurons in the solitary tract nucleus. Physiol Behav
tic glial localization for the Na+,K+-ATPase alpha 2 subunit and the 74:391–397.
glutamate transporters GLAST and GLT-1 in the rat somatosensory Honegger P, Braissant O, Henry H, Boulat O, Bachmann C, Zurich MG,
cortex. Cereb Cortex 12:515–525. et al. 2002. Alteration of amino acid metabolism in neuronal aggre-
Cholet N, Pellerin L, Welker E, Lacombe P, Seylaz J, Magistretti P, et al. gate cultures exposed to hypoglycaemic conditions. J Neurochem
2001. Local injection of antisense oligonucleotides targeted to the glial 81:1141–1151.
glutamate transporter GLAST decreases the metabolic response to Hu Y, Wilson GS. 1997. A temporary local energy pool coupled to neu-
somatosensory activation. J Cereb Blood Flow Metab 21:404–412. ronal activity: fluctuations of extracellular lactate levels in rat brain
Chuquet J, Quilichini P, Nimchinsky EA, Buzsá ki G. 2010. Predominant monitored with rapid-response enzyme-based sensor. J Neurochem
enhancement of glucose uptake in astrocytes versus neurons during 69:1484–1490.
activation of the somatosensory cortex. J Neurosci 30:15298–15303. Hydén H, Lange PW. 1962. A kinetic study of the neuron-glia relation-
Debernardi R, Pierre K, Lengacher S, Magistretti PJ, Pellerin L. 2003. ship. J Cell Biol 13:233–237.
Cell-specific expression pattern of monocarboxylate transporters in Hyder F, Patel AB, Gjedde A, Rothman DL, Behar KL, Shulman RG.
astrocytes and neurons observed in different mouse brain cortical cell 2006. Neuronal-glial glucose oxidation and glutamatergic-GABAergic
cultures. J Neurosci Res 73:141–155. function. J Cereb Blood Flow Metab 26:865–877.
Drewes LR. 2003. Molecular features, regulation, and function of mono- Iadecola C, Nedergaard M. 2007. Glial regulation of the cerebral micro-
carboxylate transporters: implications for drug delivery. J Pharm Sci vasculature. Nat Neurosci 10:1369–1376.
92:1531–1544. Ide T, Steinke J, Cahill Jr GF. 1969. Metabolic interactions of glucose,
Erlichman JS, Hewitt A, Damon TL, Hart M, Kurascz J, Li A, Leiter JC. lactate, and E -hydroxybutyrate in rat brain slices. Am J Physiol
2008. Inhibition of monocarboxylate transporter 2 in the retrotrap- 217:784–792.
ezoid nucleus in rats : a test of the astrocyte-neuron lactate-shuttle Itoh Y, Esaki T, Shimoji K, Cook M, Law MJ, Kaufman E, et al. 2003.
hypothesis. J Neurosci 28:4888–4896. Dichloroacetate effects on glucose and lactate oxidation by neurons
Escartin C, Pierre K, Colin A, Brouillet E, Delzescaux T, Guillermier M, and astroglia in vitro and on glucose utilization by brain in vivo. Proc
et al. 2007. Activation of astrocytes by CNTF induces metabolic Natl Acad Sci U S A 100:4879–4884.
plasticity and increases resistance to metabolic insults. J Neurosci Isaac JT, Ashby M, McBain CJ. 2007. The role of the GluR2 sub-
27:7094–7104. unit in AMPA receptor function and synaptic plasticity. Neuron
Fellows LK, Boutelle MG, Fillenz M. 1993. Physiological stimulation 54:859–871.
increases nonoxidative glucose metabolism in the brain of the freely Ivanov A, Mukhtarov M, Bregestovski P, Zilberter Y. 2011. Lactate effec-
moving rat. J Neurochem 60:1258–1263. tively covers energy demands during neuronal network activity in
Fernandez E, Medina JM. 1986. Lactate utilization by the neonatal rat neonatal hippocampal slices. Front Neuroenergetics 3:2.
brain in vitro. Competition with glucose and 3-hydroxybutyrate. Izumi Y, Benz AM, Katsuki H, Zorumski CF. 1997a. Endogenous mono-
Biochem J 234:489–492. carboxylates sustain hippocampal synaptic function and morpholog-
Figley CR, Stroman PW. 2011. The role(s) of astrocytes and astrocyte ical integrity during energy deprivation. J Neurosci 17:9448–9457.
activity in neurometabolism, neurovascular coupling, and the produc- Izumi Y, Benz AM, Zorumski CF, Olney JW. 1994. Effects of lac-
tion of functional neuroimaging signals. Eur J Neurosci 33:577–588. tate and pyruvate on glucose utilization in rat hippocampal slices.
Fowler JC. 1993. Glucose deprivation results in a lactate preventable Neuroreport 5:617–620.
increase in adenosine and depression of synaptic depression in rat Izumi Y, Katsuki H, Zorumski CF. 1997b. Monocarboxylates (pyru-
hippocampal slices. J Neurochem 60:572–576. vate and lactate) as alternative energy substrates for the induction
Gerhart DZ, Enerson BE, Zhdankina OY, Leino RL, Drewes LR. of long-term potentiation in rat hippocampal slices. Neurosci Lett
1997. Expression of monocarboxylate transporter MCT1 by brain 232:17–20.
endothelium and glia in adult and suckling rats. Am J Physiol Jackson VN, Price NT, Carpenter L, Halestrap AP. 1997. Cloning of the
273:E207–E213. monocarboxylate transporter isoform MCT2 from rat testis provides
Gerhart DZ, Enerson BE, Zhdankina OY, Leino RL, Drewes LR. 1998. evidence that expression in tissues is species-specific and may involve
Expression of the monocarboxylate transporter MCT2 by rat brain post-transcriptional regulation. Biochem J 324:447–453.
glia. Glia 22:272–281. Kacem K, Lacombe P, Seylaz J, Bonvento G. 1998. Structural organiza-
Golgi C. 1886. Sulla fina anatomia degli organi centrali del sistema ner- tion of the perivascular astrocyte endfeet and their relationship with
vosa. On the fine anatomy of the central organs of the nervous system. the endothelial glucose transporter: a confocal microscopy study.
Hoepli: Milano. Glia 23:1–10.
Gordon GR, Choi HB, Rungta RL, Ellis-Davies GC, MacVicar BA. Keller JN, Steiner MR, Mattson MP, Steiner SM. 1996. Lysophosphatidic
2008. Brain metabolism dictates the polarity of astrocyte control acid decreases glutamate and glucose uptake by astrocytes.
over arterioles. Nature 456:745–749. J Neurochem 67:2300–2305.
Halestrap AP, Meredith D. 2004. The SLC16 gene family-from mono- King P, Kong MF, Parkin H, MacDonald IA, Barber C, Tattersall
carboxylate transporters (MCTs) to aromatic amino acid transport- RB. 1998. Intravenous lactate prevents cerebral dysfunction dur-
ers and beyond. Pflugers Arch 447:619–628. ing hypoglycemia in insulin-dependent diabetes mellitus. Clin Sci
Hamberger A, Hydén H. 1963. Inverse enzymatic changes in neurons and 94:157–163.
glia during increased function and hypoxia. J Cell Biol 16:521–525. Koehler-Stec EM, Simpson IA, Vannucci SJ, Landschulz KT, Landschulz
Hanu R, McKenna M, O’Neill A, Resneck WG, Bloch RJ. 2000. WH. 1998. Monocarboxylate transporter expression in mouse brain.
Monocarboxylic acid transporters, MCT1 and MCT2, in cortical Am J Physiol 275:E516-E524.
astrocytes in vitro and in vivo. Am J Physiol 278:C921–C930. Lam TK, Gutierrez-Juarez R, Pocai A, Rossetti L. 2005. Regulation
Hassel B, Brathe A. 2000. Neuronal pyruvate carboxylation supports of blood glucose by hypothalamic pyruvate metabolism. Science
formation of transmitter glutamate. J Neurosci 20:1342–1347. 309:943–947
Herard AS, Dubois A, Escartin C, Tanaka K, Delzescaux T, Hantraye P, Larrabee MG. 1983. Lactate uptake and release in the presence of glu-
et al. 2005. Decreased metabolic response to visual stimulation in the cose by sympathetic ganglia of chicken embryos and by neuronal and

T H E C E N T R A L R O L E O F A S T R O C Y T E S I N N E U R O E N E R G ET I C S • 341
nonneuronal cultures prepared from these ganglia. J Neurochem Marcillac F, Brix B, Repond C, Jöhren O, Pellerin L. 2011. Nitric oxide
40:1237–1250. induces the expression of the monocarboxylate transporter MCT4 in
Larrabee MG. 1992. Extracellular intermediates of glucose metabolism: cultured astrocytes by a cGMP-independent transcriptional activa-
fluxes of endogenous lactate and alanine through extracellular pools tion. Glia 59:1987–1995.
in embryonic sympathetic ganglia. J Neurochem 59:1041–1052. Martin C, Houitte D, Guillermier M, Petit F, Bonvento G, Gurden H.
Larrabee MG. 1995. Lactate metabolism and its effect on glucose metab- 2012. Alteration of sensory-evoked metabolic and oscillatory activi-
olism in an excised neural tissue. J Neurochem 64:1734–1741. ties in the olfactory bulb of GLAST-deficient mice. Front Neural
Larrabee MG. 1996. Partitioning of CO2 production between glucose Circuits 6:1.
and lactate in excised sympathetic ganglia, with implications for McGeer PL, McGeer EG. 1989. Amino acid neurotransmitters. In:
brain. J Neurochem 67:1726–1734. Siegel G, Agranoff B, Albers RW, Molinoff P (eds.), Basic neuro-
Laughton JD, Bittar P, Charnay Y, Pellerin L, Kovari E, Magistretti PJ, chemistry: molecular, cellular, and medical aspects, 4th ed. New
et al. 2007. Metabolic compartmentalization in the human cortex York: Raven Press, pp. 311–332.
and hippocampus: evidence for a cell- and region-specific localiza- McKenna MC. 2007. The glutamate-glutamine cycle is not stoichiomet-
tion of lactate dehydrogenase 5 and pyruvate dehydrogenase. BMC ric: fates of glutamate in brain. J Neurosci Res 85:3347–3358.
Neurosci 8:35. McKenna MC, Hopkins IB, Carey A. 2001. α-cyano-4-hydroxycinnamate
Lebon V, Petersen KF, Cline GW, Shen J, Mason GF, Dufour S, et al. decreases both glucose and lactate metabolism in neurons and astro-
2002. Astroglial contribution to brain energy metabolism in humans cytes: implications for lactate as an energy substrate for neurons.
revealed by 13C nuclear magnetic resonance spectroscopy: elucida- J Neurosci Res 66:747–754.
tion of the dominant pathway for neurotransmitter glutamate reple- McKenna MC, Tildon JT, Stevenson JH, Boatright R, Huang S. 1993.
tion and measurement of astrocytic oxidative metabolism. J Neurosci Regulation of energy metabolism in synaptic terminals and cultured
22:1523–1531. rat brain astrocytes: differences revealed using aminooxyacetate.
Leino RL, Gerhart DZ, Drewes LR. 1999. Monocarboxylate transporter Dev Neurosci 15:320–329.
(MCT1) abundance in brains of suckling and adult rats: a quanti- McKenna MC, Tildon JT, Stevenson JH, Hopkins IB. 1994. Energy
tative electron microscopic immunogold study. Dev Brain Res 113: metabolism in cortical synaptic terminals from weanling and mature
47–54. rat brain: evidence for multiple compartments of tricarboxylic acid
Lin AL, Fox PT, Hardies J, Duong TQ, Gao JH. 2010. Nonlinear cycle activity. Dev Neurosci 16:291–300.
coupling between cerebral blood flow, oxygen consumption, and McKenna MC, Tildon JT, Stevenson JH, Hopkins IB, Huang X, Couto
ATP production in human visual cortex. Proc Natl Acad Sci U S A R. 1998. Lactate transport by cortical synaptosomes from adult brain:
107:8446–8451. characterization of kinetics and inhibitor specificity. Dev Neurosci
Loaiza A, Porras OH, Barros LF. 2003. Glutamate triggers rapid glucose 20:300–309.
transport stimulation in astrocytes as evidenced by real-time confocal Metea MR, Newman EA. 2006. Glial cells dilate and constrict
microscopy. J Neurosci 23:7337–7342. blood vessels: a mechanism of neurovascular coupling. J Neurosci
Lovatt D, Sonnewald U, Waagepetersen HS, Schousboe A, He W, Lin 26:2862–2870.
JH, et al. 2007. The transcriptome and metabolic gene signature Mobbs CV, Kow LM, Yang XJ. 2001. Brain glucose-sensing mechanisms:
of protoplasmic astrocytes in the adult murine cortex. J Neurosci ubiquitous silencing by aglycemia vs. hypothalamic neuroendocrine
27:12255–12266. responses. Am J Physiol 281:E649–E654.
Maekawa F, Tsuboi T, Fukuda M, Pellerin L. 2009. Regulation of the Morgello S, Uson RR, Schwartz EJ, Haber RS. 1995. The human
intracellular distribution, cell surface expression, and protein levels of blood-brain barrier glucose transporter (GLUT1) is a glucose trans-
AMPA receptor GluR2 subunits by the monocarboxylate transporter porter of gray matter astrocytes. Glia 14:43–54.
MCT2 in neuronal cells. J Neurochem 109:1767–1778. Mulligan SJ, MacVicar BA. 2004. Calcium transients in astrocytes end-
Magistretti PJ. 2008. Brain energy metabolism. In: Squire L, Berg D, feet cause cerebrovascular constrictions. Nature 431:195–199.
Bloom FE, du Lac S, Ghosh A, Spitzer N (eds.), Fundamental neuro- Newman LA, Korol DL, Gold PE. 2011. Lactate produced by glycog-
science, 3rd ed. San Diego: Academic Press, pp. 271–293. enolysis in astrocytes regulates memory processing. PLoS One
Magistretti PJ, Hof PR, Martin JL. 1986. Adenosine stimulates gly- 6(12):e28427.
cogenolysis in mouse cerebral cortex: a possible coupling mecha- O’Brien J, Kla KM, Hopkins IB, Malecki EA, McKenna MC. 2007.
nism between neuronal activity and energy metabolism. J Neurosci Kinetic parameters and lactate dehydrogenase isozyme activities
6:2558–2562. support possible lactate utilization by neurons. Neurochem Res
Magistretti PJ, Morrison JH. 1988. Noradrenaline- and vasoactive 32:597–607.
intestinal peptide-containing neuronal systems in neocortex: func- Parsons MP, Hirasawa M. 2010. ATP-sensitive potassium channel-
tional convergence with contrasting morphology. Neuroscience mediated lactate effect on orexin neurons: implications for brain
24:367–378. energetics during arousal. J Neurosci 30:8061–8070.
Magistretti PJ, Morrison JH, Shoemaker WJ, Sapin V, Bloom FE. 1981. Pellerin L. 2003. Lactate as a pivotal element in neuron-glia metabolic
Vasoactive intestinal polypeptide induces glycogenolysis in mouse cooperation. Neurochem Int 43:331–338.
cortical slices: a possible regulatory mechanism for the local control Pellerin L, Bergersen LH, Halestrap AP, Pierre K. 2005. Cellular and
of energy metabolism. Proc Natl Acad Sci U S A 78:6535–6539. subcellular distribution of monocarboxylate transporters in cultured
Magistretti PJ, Pellerin L. 1996. The contribution of astrocytes to the brain cells and in the adult brain. J Neurosci Res 79:55–64.
18F-2-deoxyglucose signal in PET activation studies. Mol Psychiatry Pellerin L, Magistretti PJ. 1994. Glutamate uptake into astro-
1:445–452. cytes stimulates aerobic glycolysis: a mechanism coupling neu-
Magistretti PJ, Schorderet M. 1984. VIP and noradrenaline act syn- ronal activity to glucose utilization. Proc Natl Acad Sci U S A 91:
ergistically to increase cyclic AMP in cerebral cortex. Nature 10625–10629.
308:280–282. Pellerin L, Magistretti PJ. 1997. Glutamate uptake stimulates
Maran A, Cranston I, Lomas J, Macdonald I, Amiel SA. 1994. Na+,K+-ATPase activity in astrocytes via activation of a dis-
Protection by lactate of cerebral function during hypoglycemia. tinct subunit highly sensitive to ouabain. J Neurochem 69:
Lancet 343:16–20. 2132–2137.
Maran A, Crepaldi C, Trupiani S, Lucca T, Jori E, Macdonald IA, et al. Pellerin L, Magistretti PJ. 2005. Ampakine CX546 bolsters energetic
2000. Brain function rescue effect of lactate following hypoglycaemia response of astrocytes: a novel target for cognitive-enhancing drugs
is not an adaptation process in both normal and type I diabetic sub- acting as alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
jects. Diabetologia 43:733–741. (AMPA) receptor modulators. J Neurochem 92:668–677.

342 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Pellerin L, Pellegri G, Bittar PG, Charnay Y, Bouras C, Martin JL, et al. Schurr A, Miller JJ, Payne RS, Rigor BM. 1999. An increase in lac-
1998a. Evidence supporting the existence of an activity-dependent tate output by brain tissue serves to meet the energy needs of
astrocyte-neuron lactate shuttle. Dev Neurosci 20:291–299. glutamate-activated neurons. J Neurosci 19:34–39.
Pellerin L, Pellegri G, Martin JL, Magistretti PJ. 1998b. Expression of Schurr A, Payne RS, Miller JJ, Rigor BM. 1997. Brain lactate is an obliga-
monocarboxylate transporter mRNAs in mouse brain: support for a tory aerobic energy substrate for functional recovery after hypoxia:
distinct role of lactate as an energy substrate for the neonatal vs. adult further in vitro validation. J Neurochem 69:423–426.
brain. Proc Natl Acad sci USA 95:3990–3995. Schurr A, West CA, Rigor BM. 1988. Lactate-supported synap-
Peng L, Hertz, L, Huang R, Sonnewald U, Petersen SB, Westergaard N, tic function in the rat hippocampal slice preparation. Science
et al. 1993. Utilization of glutamine and of TCA cycle constituents 240:1326–1328.
as precursors for transmitter glutamate and GABA. Dev Neurosci Serres S, Bezancon E, Franconi JM, Merle M. 2005. Ex vivo NMR study
15:367–377. of lactate metabolism in rat brain under various depressed states.
Pierre K, Chatton JY, Parent A, Repond C, Gardoni F, Di Luca M, et al. J Neurosci Res 79:19–25.
2009. Linking supply to demand: the neuronal monocarboxylate Shank RP, Bennett GS, Freytag SO, Campbell GL. 1985. Pyruvate car-
transporter MCT2 and the alpha-amino-3-hydroxyl-5-methyl-4-is boxylase: an astrocyte-specific enzyme implicated in the replenish-
oxazole-propionic acid receptor GluR2/3 subunit are associated in a ment of amino acid neurotransmitter pools. Brain Res 329:364–367.
common trafficking process. Eur J Neurosci 29:1951–1963. Shank RP, Campbell GL. 1984. Alpha-ketoglutarate and malate
Pierre K, Magistretti PJ, Pellerin L. 2002. MCT2 is a major neuronal uptake and metabolism by synaptosomes: further evidence for an
monocarboxylate transporter in the adult mouse brain. J Cereb Blood astrocyte-to-neuron metabolic shuttle. J Neurochem 42:1153–1161.
Flow Metab 22:586–595. Shen J, Petersen KF, Behar KL, Brown P, Nixon TW, Mason GF, et al.
Pierre K, Pellerin L, Debernardi R, Riederer BM, Magistretti PJ. 2000. 1999. Determination of the rate of the glutamate/glutamine cycle in
Cell-specific localization of monocarboxylate transporters, MCT1 the human brain by in vivo 13C NMR. Proc Natl Acad Sci U S A
and MCT2, in the adult mouse brain revealed by double immu- 96:8235–8240.
nohistochemical labeling and confocal microscopy. Neuroscience Shimizu H, Watanabe E, Hiyama TY, Nagakura A, Fujikawa A, Okado
100:617–627. H, et al. 2007. Glial Nax channels control lactate signaling to neu-
Poitry S, Poitry-Yamate C, Ueberfeld, J, MacLeish PR, Tsacopoulos M. rons for brain [Na+] sensing. Neuron 54:59–72.
2000. Mechanisms of glutamate metabolic signaling in retinal glial Sibson NR, Dhankhar A, Mason GF, Rothman DL, Behar KL, Shulman
(Muller) cells. J Neurosci 20:1809–1821. RG. 1998. Stoichiometric coupling of brain glucose metabolism and glu-
Poitry-Yamate CL, Poitry S, Tsacopoulos M. 1995. Lactate released by tamatergic neuronal activity. Proc Natl Acad Sci U S A. 95:316–321.
Muller glial cells is metabolized by photoreceptors from mammalian Smith D, Pernet A, Hallett WA, Bingham E, Marsden PK, Amiel SA.
retina. J Neurosci 15:5179–5191. 2003. Lactate: a preferred fuel for human brain metabolism in vivo.
Porras OH, Loaiza A, Barros LF. 2004. Glutamate mediates acute J Cereb Blood Flow Metab 23:658–664.
glucose transport inhibition in hippocampal neurons. J Neurosci Sotelo C, Palay SL. 1968. The fine structure of the lateral vestibu-
24:9669–9673. lar nucleus in the rat. I. Neurons and neuroglial cells. J Cell Biol
Qu H, Haberg A, Haraldseth O, Unsgard G, Sonnewald U. 2000. (13) 36:151–179.
C MR spectroscopy study of lactate as substrate for rat brain. Dev Suzuki A, Stern SA, Bozdagi O, Huntley GW, Walker RH, Magistretti
Neurosci 22:429–436. PJ, et al. 2011. Astrocyte-neuron lactate transport is required for
Rafi ki A, Boulland JL, Halestrap AP, Ottersen OP, Bergersen L. 2003. long-term memory formation. Cell 144:810–823.
Highly differential expression of the monocarboxylate transport- Swanson RA, Morton MM, Sagar SM, Sharp FR. 1992. Sensory stimu-
ers MCT2 and MCT4 in the developing rat brain. Neuroscience lation induces local cerebral glycogenolysis: demonstration by autora-
122:677–688. diography. Neuroscience 51:451–461.
Rinholm JE, Hamilton NB, Kessaris N, Richardson WD, Bergersen Tabernero A, Vicario C, Medina JM. 1996. Lactate spares glucose as
LH, Attwell D. 2011. Regulation of oligodendrocyte develop- a metabolic fuel in neurons and astrocytes from primary culture.
ment and myelination by glucose and lactate. J Neurosci 31: Neurosci Res 26:369–376.
538–548. Takahashi S, Driscoll BF, Law MJ Sokoloff L. 1995. Role of sodium and
Robinet C, Pellerin L. 2010. Brain-derived neurotrophic factor enhances potassium ions in regulation of glucose metabolism in cultured astro-
the expression of the monocarboxylate transporter 2 through trans- glia. Proc Natl Acad Sci U S A 92:4616–4620.
lational activation in mouse cultured cortical neurons. J Cereb Blood Takano T, Tian GF, Peng W, Lou N, Libionka W, Han X, et al. 2006.
Flow Metab 30:286–298. Astrocyte-mediated control of cerebral blood flow. Nat Neurosci
Robinet C, Pellerin L. 2011. Brain-derived neurotrophic factor enhances 9:260–267.
the hippocampal expression of key postsynaptic proteins in vivo Tekkök SB, Brown AM, Westenbroek R, Pellerin L, Ransom BR. 2005.
including the monocarboxylate transporter MCT2. Neuroscience Transfer of glycogen-derived lactate from astrocytes to axons via spe-
192:155–163. cific monocarboxylate transporters supports mouse optic nerve activ-
Rossner MJ, Hirrlinger J, Wichert SP, Boehm C, Newrzella D, Hiemisch ity. J Neurosci Res 81:644–652.
H, et al. 2006. Global transcriptome analysis of genetically identified Tsacopoulos M, Evequoz-Mercier V, Perrottet P, Buchner E. 1988.
neurons in the adult cortex. J Neurosci 26:9956–9966. Honeybee retinal glial cells transform glucose and supply the neurons
Rothman DL, Sibson NR, Hyder F, Shen J, Behar KL, Shulman RG. with metabolic substrate. Proc Natl Acad Sci U S A 85:8727–8731.
1999. In vivo nuclear magnetic resonance spectroscopy studies of Tsacopoulos M, Veuthey AL, Saravelos SG, Perrottet P, Tsoupras G.
the relationship between the glutamate-glutamine neurotransmitter 1994. Glial cells transform glucose to alanine, which fuels the neu-
cycle and functional neuroenergetics. Phil Trans Roy Soc (Lond B rons in the honeybee retina. J Neurosci 14:1339–1351.
Biol Sci) 354:1165–1177. Vicario C, Arizmendi C, Malloch G, Clark JB, Medina JM. 1991.
Rouach N, Koulakoff A, Abudara V, Willecke K, Giaume C. 2008. Lactate utilization by isolated cells from early neonatal rat brain.
Astroglial metabolic networks sustain hippocampal synaptic trans- J Neurochem 57:1700–1707.
mission. Science 322:1551–1555. Vilchez D, Ros S, Cifuentes D, Pujadas L, Vallès J, García-Fojeda B,
Roy CS, Sherrington CS. 1890. On the regulation of the blood supply of Criado-García O, Fernández-Sánchez E, Medraño-Fernández I,
the brain. J Physiol 11:85–108. Domínguez J, García-Rocha M, Soriano E, Rodríguez de Córdoba
Schousboe A, Westergaard N, Waagepetersen HS, Larsson OM, Bakken S, Guinovart JJ. 2007. Mechanism suppressing glycogen synthesis
IJ, Sonnewald U. 1997. Trafficking between glia and neurons of TCA in neurons and its demise in progressive myoclonus epilepsy. Nat
cycle intermediates and related metabolites. Glia 21:99–105. Neurosci 10:1407–1413.

T H E C E N T R A L R O L E O F A S T R O C Y T E S I N N E U R O E N E R G ET I C S • 343
Voutsinos-Porche B, Bonvento G, Tanaka K, Steiner P, Welker E, (DAB) as an inhibitor of brain glycogen shunt activity. J Neurochem
Chatton JY, et al. 2003a. Glial glutamate transporters mediate a 105:1462–1470.
functional metabolic crosstalk between neurons and astrocytes in the Walz W, Mukerji S. 1988. Lactate release from cultured astrocytes and
mouse developing cortex. Neuron 37:275–286. neurons: a comparison. Glia 1:366–370.
Voutsinos-Porche B, Knott G, Tanaka K, Quairiaux C, Welker E, Warburg OH, Posener K, Negelein E. 1924. Ü ber den Stoff wechsel
Bonvento G. 2003b. Glial glutamate transporters and matu- der Tumoren [About the metabolism of tumors]. Biochem Z 152:
ration of the mouse somatosensory cortex. Cereb Cortex 319–344. German.
13:1110–1121. Wyss MT, Jolivet R, Buck A, Magistretti PJ, Weber B. 2011. In vivo evidence
Waagepetersen HS, Bakken IJ, Larsson OM, Sonnewald U, Schousboe for lactate as a neuronal energy source. J Neurosci 31:7477–7485.
A. 1998. Comparison of lactate and glucose metabolism in cultured Yu S, Ding WG. 1998. The 45 kDa form of glucose transporter 1
neocortical neurons and astrocytes using 13C-NMR spectroscopy. (GLUT1) is localized in oligodendrocyte and astrocyte but not in
Dev Neurosci 20:310–320. microglia in the rat brain. Brain Res 797:65–72.
Waagepetersen HS, Sonnewald U, Larsson OM, Schousboe A. 2000. A Yudkoff M. 1997. Brain metabolism of branched-chain amino acids. Glia
possible role of alanine for ammonia transfer between astrocytes and 21:92–98.
glutamatergic neurons. J Neurochem 75:471–479. Zonta M, Angulo MC, Gobbo S, Rosengarten B, Hossmann KA, Pozzan
Walls AB, Sickmann HM, Brown A, Bouman SD, Ransom B, Schousboe T, et al. 2003. Neuron-to-astrocyte signaling is central to the dynamic
A, et al. 2008. Characterization of 1,4-dideoxy-1,4-imino-d-arabinitol control of brain microcirculation. Nat Neurosci 6:43–50.

344 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
GENOMIC PROFILES
This page intentionally left blank
28.
THE ASTROCY TE TRANSCRIPTOME
Ditte Lovatt and Maiken Nedergaard

A B B R E VI AT I O N S in different brain regions indicate that their functions dif-


fer. Another heavily debated topic is astrocyte heterogeneity
Aldh1l1 10-formyltetrahydrofolate dehydrogenase (Matyash and Kettenmann 2010; Zhang and Barres 2010). Are
AQP aquaporin there subtypes of astrocytes within the same region, and can
FACS fluorescence-activated cell sorting these be distinguished molecularly and functionally? In the
FISH fluorescent in situ hybridization diseased brain, how do reactive astrocytes differ from healthy
GFAP glial fibrillary acid protein ones? This chapter compares existing transcriptome databases
GFP green fluorescent protein and evaluates whether these unique sets of information can be
hGFAP human glial fibrillary acid protein used as a discovery tool to gain insight into the function of
LCM laser capture microdissection astrocytes in the intact adult brain (Cahoy et al. 2008; Doyle
qPCR quantitative polymerase chain reaction et al. 2008; Lovatt et al. 2007). Specifically, it discusses the use
RNA-Seq Next Generation RNA sequencing of the astrocyte transcriptome in defining whether the physio-
TRAP translating ribosome affinity purification logical characteristics, regional variations, and transformation
in reactive gliosis can be identified in the molecular profile of
astrocytes.
1 INTRODUCTION

Although astrocytes are the most abundant cell type in the 2 M ET H O D S U S E D TO P R O D U C E


mammalian brain, surprisingly little is known with regard T R A N S C R I P TO M E S F R O M A S T R O C Y T E S
to the functional properties of astrocytes in situ (Agulhon I N I N TAC T T I S S U E
et al. 2008; Ransom et al. 2003). Studies of astrocytic energy
metabolism, receptor expression, Ca2+ signaling, release of To date, only a few studies have reported on the transcriptome
neuroactive compounds, and interactions with other cell from astrocytes in situ or in vivo (Cahoy et al. 2008; Doyle
types are traditionally based on observations of cultured cells et al. 2008; Lichter-Konecki et al. 2008; Lovatt et al. 2007;
or acute slices prepared from rodent pups younger than three Yang et al. 2011). In comparison, the transcriptomes of numer-
weeks old (Nedergaard and Verkhratsky 2012; Theodosis et al. ous neuronal subtypes from striatal, cerebellar, cortical, and
2008). The inability to selectively stimulate astrocytes and hippocampal regions have already been published (Cahoy
detect a functional output has prevented a basic analysis of the et al. 2008; Doyle et al. 2008; Dugas et al. 2008; Heiman et al.
functional properties of astrocytes in the adult intact central 2008; Lobo et al. 2006; Molyneaux et al. 2005; Sugino et al.
nervous system. The first analysis of adult astrocytes in situ 2006). The technical aspects of transcriptomics procedures,
first emerged five to six years ago, when several laboratories such as microarray and RNA-seq, have not prevented progress
pioneered the use of two-photon Ca2+ imaging combined with in astrocyte transcriptomics. Rather, it is the technical aspects
electrophysiological recordings of synaptic activity (Dombeck of harvesting enriched populations of astrocytes in vivo or in
et al. 2007; Schummers et al. 2008; Takano et al. 2006; Wang situ as a source of mRNA for transcriptomics that have pre-
et al. 2006). These in vivo imaging studies showed that cor- vented progress mainly due to the limited number of trans-
tical astrocytes display complex patterns of calcium signaling genic animals with fluorescently labeled astrocytes. To date,
in response to sensory stimulation and spontaneous calcium both microarray and RNA-seq can produce the transcriptome
spikes during locomotion in awake behaving head-restrained from minute amounts of RNA with relative ease (Tang et al.
mice. Nevertheless, it is not known whether astrocytic cal- 2009; Wang et al. 2009). The processing of RNA for microar-
cium signaling plays a functional role in processing of sensory ray or sequencing, as well as transcriptome data analysis, are
information. Similarly, it is well documented from histologi- reviewed in detail elsewhere (Wang et al. 2009). However, there
cal studies that astrocytes display remarkable regional differ- are significant technical challenges in isolating mRNA from
ences in morphology, spanning from rounded, polyhedral astrocytes in vivo. As opposed to cultured astrocytes, which
cells in cortex to elongated asymmetrical cells in the neuro- are flat epithelioid-appearing cells, astrocytes in intact tissue
genic area of the hippocampal dentate region (Bushong et al. have highly branched processes and are intermingled with
2002; Oberheim et al. 2012). However, it is not established neighboring cells, making it difficult to isolate mRNA from
whether the distinct morphological features of astrocytes astrocytes without mRNA contamination from surrounding

347
cell types (Bushong et al. 2002; Simard et al. 2003). This ana- Each method has advantages and disadvantages (Table 28.1).
tomical feature is not unique to astrocytic preparations from Although LCM is capable of isolating mRNA from a single
intact tissue. Isolating mRNA from neurons and other brain cell for which the location and morphology is known, it has
cells has been equally difficult. However, many of the recently to be done in fixed or frozen brain tissue, which may compro-
developed techniques used to isolate mRNA from cells in situ mise the quality of the mRNA. It is also technically difficult to
rely on the availability of transgenic lines with fluorescently dissect out the cells without cross-contaminating with mRNA
labeled cells of interest. To this end, far more transgenic lines from neighboring cells. Because of the small size of the astro-
with fluorescently labeled neuronal subtypes exist than lines cytic cell body, LCM is rarely used to isolate astrocytic mRNA
labeling astrocytes (Feng et al. 2000; Gong et al. 2003; Regan (Ferraiuolo et al. 2011). Refinement in LCM, including use of
et al. 2007; Sugino et al. 2006; Zhuo et al. 1997). vibrating piezoelectric driven micro-chisel, may enable isola-
Over the past decade, several experimental approaches have tion of small astrocytic cell bodies in the future. FACS sort-
been developed to harvest mRNA from single cells or single ing, panning, and manual sorting all use acutely dissociated
populations of cells isolated from intact brain tissue. These brain tissue for which the location and information about the
methods include fluorescence-activated cell sorting (FACS) morphology of the sorted cells are lost during the dissociation
(Cahoy et al. 2008; Lobo et al. 2006; Lovatt et al. 2007), procedure. The standard dissociation protocol uses enzymatic
immunopanning (panning) (Cahoy et al. 2008), translat- treatment with papain followed by mechanical dissociation
ing ribosome affinity purification (TRAP)/Ribo-tag (Doyle to achieve a single cell suspension. The cell types identified in
et al. 2008; Heiman et al. 2008; Sanz et al. 2009), laser cap- these suspensions include astrocytes, oligodendrocyte precur-
ture microdissection (LCM) (Espina et al. 2006; Tang et al. sors, microglia, neurons, and endothelial cells (Daneman et al.
2009), patch pipette aspiration (Martina et al. 1998; Surmeier 2010; Dugas et al. 2008; Lobo et al. 2006; Lovatt et al. 2007).
et al. 1996), and manual sorting of reporter labeled cells in dis- Depending on the quality of the dissociation and the age of the
sociates (Hempel et al. 2007; Sugino et al. 2006) (Fig. 28.1). animal, astrocytes account for 30% to 50% of the dissociated

Start material: Brain tissue XFP expressing brain tissue TRAP BAC-transgenic brain

Freeze & Fix Dissociate Dissociate Lyze tissue

Procedure:

Label with antibody

LCM: Immunopanning: FACS: Manual sorting: TRAP:


mRNA isolation method: Dissect cell of interest Isolate cells Isolate cells Isolate cells and Affinity purify mRNA:ribosome
and extract mRNA and extract mRNA and extract mRNA extract mRNA complexes with GFP antibody

RNA amplification
and processing

Microarray RNA-seq

Transcriptome
analysis

Figure 28.1 Overview of methods used for transcriptome profiling of cells isolated from intact tissue. The type of cells to be isolated depends upon the
availability and type of transgenic mouse lines. If no transgenic line is available, the desired cell population can be isolated either by: i) surface-labeling
with a specific antibody, and then isolating the cells by FACS or immunopanning, or ii) by dissecting single cells or brain regions out from frozen and
fixed whole brain tissue selected either by morphology or antibody-labeling. If transgenics lines that label the desired cells fluorescently, are available,
then FACS or manual sorting can isolate these labeled cells. If BAC-transgenic lines, that label the translating ribosomes, are available, then the TRAP
method can be used to isolate the desired cell type. See table 28.1 for more details on these methods. Once the desired cell or cells are isolated by any
of the above methods, mRNA is extracted and linearly amplified to achieve μg levels of cDNA. The cDNA is then either labeled for microarray or is
used for library construction for RNA-seq. The resulting transcriptome is then analyzed and mined to identify, for instance, novel biomarkers, disease
related transcripts, cell-type specific alternative splicing and signaling pathways. (LCM) Laser Capture Microdissection, (FACS) Fluorescent Activated
Cell Sorting, (TRAP) Translating Ribosome Affinity Purification, (BAC) Bacterial Artificial Chromosome.

348 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Table 28.1 mRNA COLLECTION METHODS FROM INTACT TISSUE PREPARATIONS
CELL SELECTION PARAMETERS

mRNA
COLLECTION CROSS- SINGLE CELL MOLECULAR CELL SUBREGIONAL
METHOD CONTAMINATION HETEROGENEITY TIME (H) MARKER MORPHOLOGY LOCATION

FACS-array* Low + 5–67 + – –

Immunopanning† Low – 4 + – –

Manual sorting‡ Low + 3–5 + – –

LCM§ High + 1 + + +

Patch pipette¶ Medium + 1 + + +


TRAP# High – 3 + – –

Five hours using fluorescent reporter mice and six hours using antibody labeling of cells.
*Lovatt et al. 2007; †Cahoy et al. 2008; ‡Hempel et al. 2007; §Espina et al. 2006; ¶Martina et al. 1998; Surmeier et al. 1996; #Doyle et al. 2008.

cells (Lovatt et al. 2007). Astrocytes and other cell types can when quantifying low abundance transcripts, or attempting to
be isolated from the dissociate by FACS or manual sorting identify cell specific transcripts.
based on their expression of fluorescent reporters, such as Induction of immediate early genes is an important con-
GFAP-GFP, or by either FACS or immunopanning based on cern when using time-consuming harvesting methods to iso-
their expression of surface markers that can be tagged with an late mRNA from intact tissue for transcriptome profiles. To
antibody, such as anti-GLT1. Astrocytic mRNA has also been this end, both LCM and TRAP have the advantage of using
harvested using the TRAP method, in which astrocyte-specific fixed and lysed tissue preparations in which biological activi-
expression of BAC transgenic GFP-ribosome chimeras serve as ties can be inhibited. FACS, panning, and manual sorting use
a tag for affinity purifying mRNA–ribosome complexes from live dissociated cells; therefore, timing becomes an important
tissue lysates using a GFP antibody (Doyle et al. 2008). Like factor because the majority of mature astrocytes in prepara-
the methods using cell dissociates, information about cellular tions of dissociated adolescent and adult brain tissue undergo
morphology and subcellular location is lost using the TRAP apoptosis within 40 hours (Foo et al. 2011). To generate
method. Further, some investigators have used a patch pipette genomic profiles that pass quality control, it is crucial to use
approach to aspirate cytosolic mRNA from whole-cell patched high-quality mRNA that is not derived from dying cells with
cells in intact tissue (Martina et al. 1998; Surmeier et al. 1996); increased RNAse activities. The tissue dissociation procedure
however, a global transcriptome from brain cells using this of adult tissue takes about three hours, and cell isolation and
technique remains to be published. The development of meth- subsequent mRNA extraction add an additional 2 hours. Most
ods being able to isolate purer sources of mRNA from single published dissociation protocols attempt to keep the prepara-
astrocytes in intact tissue will facilitate the correlation of the tion cold whenever possible, and some even add inhibitors to
transcriptome to other types of single cell information, such as prevent excitoxicity when isolating neurons. RNA synthesis
morphology, chemical profile and physiological response pat- or RNA degradation can also be inhibited by the addition of
terns (see Table 28.1). For instance, profiling single astrocytes pharmacological inhibitors, for instance, by storing sorted cells
of different morphologies could provide insights into the mol- in products such as RNA later (Belgard et al. 2011). An analysis
ecules facilitating such morphological differences, and profil- of inflammatory and stress-related transcripts in astrocytes and
ing single astrocytes from distinct microenvironments could other cell types did not indicate that the dissociation procedure
provide important insights into how astrocytes interact with and subsequent FACS procedure induced any such responses
the local environment. (Lobo et al. 2006; Lovatt et al. 2007), suggesting that these
A comparison of the transcriptomes produced by the procedures allow harvesting of RNA from nonstressed cells as
FACS, panning, TRAP, LCM, and manual harvesting meth- long as the procedure is carried out within a 6-hour time frame
ods showed that all of them demonstrated comparably high at low temperatures and dying cells, identified by propidium
levels of reproducibility (Okaty et al. 2011). Further com- iodide uptake, are excluded.
parison showed that the data from LCM and TRAP exhib-
ited significantly higher levels of mRNA contamination from
neighboring cells (see Table 28.1). In the TRAP transcrip- 3 T H E T R A N S C R I P TO M E O F A D U LT
tomes, contamination by other cell types was apparent for A S T R O C Y T E S F R O M I N TAC T T I S S U E
astrocytes, GABAergic cells, and oligodendrocytes, suggest-
ing that contamination may be a general phenomenon of this The use of transcriptome profiling was first applied to astro-
method. Although some degree of contamination is always cytes in situ in 2007 (Lovatt et al. 2007). Using FACS, two
present, at least at trace levels, higher levels become a problem distinct populations of astrocytes were isolated from acutely

T H E A S T R O C Y T E T R A N S C R I P TO M E • 349
dissociated adult, cortical tissue from mice based on their (Steindler and Laywell 2003). However, GFAP and vimentin
transgenic GFAP-GFP expression and surface GLT1 protein are both markers of reactive gliosis in vivo (Pekny and Nilsson
expression. Following isolation, RNA was immediately har- 2005), questioning whether cultured astrocytes is a model
vested for transcriptome profiling. Analysis of the transcrip- of reactive gliosis rather than a model of healthy astrocytes
tome of these astrocytes confirmed that they expressed several in vivo (see chapter 51). However, more recent studies using
expected astrocyte specific markers such as AQP4 (aquaporin intact tissue have led to several discoveries suggesting that the
4), GFAP, Gjb6 (connexin 30), Gja1 (connexin 43), S100b, in vivo counterpart of cultured astrocytes differed morpho-
and Slc1a2 (GLT1). In contrast, they did not express enriched logically (Bushong et al. 2002) and functionally. For example,
levels of markers characteristic of other cell types, suggesting voltage-gated Ca2+ channels in cultured astrocytes are not
that the harvested RNA was from a pure population of astro- present in vivo (MacVicar 1984). This is consistent with our
cytes. The importance of pure population becomes significant analysis of cultured astrocytes that, when compared with astro-
when considering low abundance transcripts and specificity cytes in situ, express several transcripts encoding voltage-gated
determination. Even a very low level of contaminating RNA calcium channels, including Cacna1d, Cacna2d1, Cacnb3,
can obscure the transcriptome profile. Currently the only ways and Cacnb4 (see data analysis in section 5). These discoveries
to evaluate contamination are by immunolabeling the purified have left little support, indicating that cultured astrocytes suf-
cells with an astrocyte specific antibody, and inspecting the ficiently modeled astrocytes in vivo, and pushed for the devel-
transcriptome data for expression of cell type–specific mark- opment of novel methods that can be applied to the study of
ers (Lovatt et al. 2007). Besides the expression of anticipated astrocyte function both in intact tissue and under more physi-
markers, the population of astrocytes expressed about 9,400 ological conditions than the use of cultured astrocytes.
transcripts. Among these, roughly 3,000 transcripts were astro- A comparison between in situ FACS-isolated GFAP-
cyte enriched when compared with the astrocyte-depleted GFP+ astrocytes and cultured FACS-isolated GFAP-GFP+
pool. This is in line with the number of transcripts expressed astrocytes prepared according to the classical protocols of
by populations of other cell types (Cahoy et al. 2008; Lobo McCarthy and de Vellis (1980) revealed that the two types of
et al. 2006). astrocytes are indeed very different (Lovatt and Nedergaard,
Once the in situ astrocyte transcriptome had been pro- unpublished). A simple comparison of the correlation coef-
duced, one of the first questions was, how different are in situ ficient showed a very high similarity among samples within
astrocytes from their cultured counterparts? Cultured astro- the same biological group (0.98–0.99) (Table 28.2). In con-
cytes from neonatal rodent brain tissue have been the preferred trast, the correlation coefficient between cultures and in situ
model to study the physiological properties of astrocytes since astrocytes (0.78) was in the same range as that of in situ astro-
the 1980s (McCarthy and de Villis 1980). Classically cultured cytes and the astrocyte-depleted pool (0.72), suggesting that
astrocytes are made from dissociated neonatal brain tissue cultured astrocytes are as different from astrocytes in situ, as
that is then cultured in high serum-containing media. At this astrocytes in situ are different from the average of all other
point in development, the potential for glia progenitors to dif- cells in the brain. A statistical comparison revealed that 17%
ferentiate into astrocytes is high, and cultured astrocytes rep- of the genes were significantly enriched in astrocytes in situ
resent mostly progenitor cells that are differentiated in vitro as compared with cultures, and 24% of the genes were signifi-
into a cell type expressing high of levels GFAP and vimentin cantly enriched in cultures as compared with in situ astrocytes.

Table 28.2 SIMILARITY OF IN SITU AND CULTURE ASTROCYTE TRANSCRIPTOMES


COMPARISON GROUPS R^2

Within groups GFAP-GFP+/GLT1+ in situ 0.98

GFAP-GFP–/GLT1+ in situ 0.98

GFAP-GFP+ culture 0.99

GFAP-GFP–/GLT1– in situ 0.99

Between groups GFAP-GFP+/GLT1+ in situ vs. GFAP-GFP–/GLT1+ in situ 0.93

GFAP-GFP+ culture vs. GFAP-GFP+/GLT1+ in situ 0.78

GFAP-GFP+/GLT1+ in situ vs. GFAP-GFP–/GLT1– in situ 0.72


GFAP-GFP+ culture vs. GFAP-GFP–/GLT1– in situ 0.80

In situ cell populations are FACS-purified and from Lovatt et al. (2007).
GFAP-GFP+ cultures were grown according to the procedure described by McCarthy and de Villis (1980), and then
FACS-purified for GFP after two in culture.
Lovatt et al. 2007.
McCarthy and de Vellis J. 1980.

350 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
For instance, only astrocytes in situ expressed the NMDA between them. Among the genes that differed between astro-
receptor Grin2c, while only cultured astrocytes expressed the cytes and neurons were many related to metabolic pathways.
AMPA receptor Gria3. Likewise, cultures expressed Adora2a For instance, both cell types expressed genes involved in
(adenosine receptor 2A), whereas in situ astrocytes expressed the enzymatic processes of glycolytic and oxidative glucose
Adora2b (adenosine receptor 2B). Cultures also expressed metabolism, suggesting that both astrocytes and neurons have
a number of laminin subunits, suggesting that their surface self-sufficiency with respect to anerobic and oxidative glucose
composition is different from astrocytes in situ. Other dif- metabolism. In contrast, metabolic pathways that entered or
ferences included significant enrichment of the synaptic exo- exited either glycolysis or the TCA cycle, such as those for
cytosis proteins Syt1 (synaptotagmin 1) and Syn1 (synapsin lactate or glutamate metabolism, were clearly compartmental-
1) in cultured astrocytes but not in situ astrocytes. Several ized between astrocytes and neurons (Lovatt et al. 2007). A
studies have found that astrocytes in culture possess the functional validation of these data using qPCR and mass spec-
same machinery as neurons for regulated fast exocytosis, but troscopy of the glucose metabolites confirmed the findings of
whether astrocytes in vivo also exhibit fast exocytosis is heav- the transcriptome analysis, and indicates that transcriptome
ily debated. Our findings are consistent with previous stud- profiling is a powerful tool to gain insight into biological path-
ies showing that although astrocytes in vivo do express many ways and their differences among different cell types. A simi-
proteins involved in exocytosis, they lack the ones involved lar study used a combination of FACS and immunopanning
in fast exocytosis usually observed in synapses (Cahoy et al. to purify RNA from developing astrocytes, oligodendrocyte
2008; Wilhelm et al. 2004). These and other gene expression precursors, and neurons from acutely dissociated 17-day-old
differences may elicit distinct responses to ligands present in mouse brain (Cahoy et al. 2008). This study also found that
the neuropil. Nevertheless, this finding questions the use of purification by FACS or immunopanning yielded highly
cultured astrocytes as models of astrocytes. pure populations of astrocytes, as indicated by significantly
In line with our findings, a recent study found that isolat- enriched levels of Slc1a2 (GLT1), Gja1 (connexin 43), and
ing astrocytes from P1 to P8 animals by immunopanning and AQP4 (aquaporin 4), while being devoid of markers labeling
growing them in serum-free media led to a high rate of apop- oligodendrocyte precursors and neurons.
tosis 40 hours after plating. The survival rate was improved Transcriptome profiling can also be used to identify novel
after the addition of selected growth factors that matched the biomarkers in astrocytes. This is done by comparing the astro-
receptor profile of the acutely isolated cells (Foo et al. 2011). cyte transcriptome to the transcriptome of other cell types or
Subsequent transcriptional comparison of acutely isolated the transcriptome of unsorted cells, and then extracting genes
astrocytes with classically cultured astrocytes and acutely that are significantly enriched in astrocytes above a certain
isolated astrocytes grown in serum-free media with heparin- threshold. Lovatt et al. identified by transcriptome profiling
binding EGF-like growth factor as the growth factor for 7 924 genes that were highly enriched and presumably only
days in vitro suggested that astrocytes grown in a select media expressed in GLT1+ astrocytes as compared with GLT1– cells
maintained a transcriptome profile that largely mimicked and Thy1+ neurons. These astrocyte-enriched genes included
that of acutely isolated astrocytes, whereas classically cultured Cbs (Cystathionine β-synthase), Slc14a1 (solute carrier fam-
astrocytes differed dramatically from both acutely isolated ily 14 member 1, an urea transporter), Mlc1 (Megalencephalic
astrocytes and those cultured in serum-free medium for 7 leukoencephalopathy 1), and Aldh1l1 (10-formyltetrahydro-
days. Notably, it has not yet been validated whether astrocytes folate dehydrogenase) in addition to the already-mentioned
cultured using this promising novel approach remain imma- biomarkers. Cahoy et al. also identified Aldh1l1 and validated
ture (i.e., similar to the day of harvest) or if they differentiate it as a new astrocyte-specific biomarker. Aldh1l1 is a cytosolic
and display a transcriptome profile that more closely mimics enzyme that has several advantages over the well-characterized
the transcriptome of differentiated astrocytes isolated from astrocyte marker GFAP, which is a filament protein localized
intact adult brain. mainly to larger, but not finer, astrocytic processes. In con-
trast, Aldh1l1 labels both large and fine processes, making it
a preferred marker to use for histology studies, because the
4 N O VE L G E N E S A N D PAT H WAYS entire astrocyte can be visualized using a simple staining. An
I D E N T I F I E D I N A S T R O C Y T E S BY independent study (Yang et al. 2011) followed up on the dis-
T R A N S C R I P TO M E P R O F I L I N G covery of Aldh1l1 and found that postnatal expression in cor-
tex peaks at P2 and then declines to a lower steady level for the
Transcriptome profiling serves as a discovery tool to identify remainder of the rodent’s life. However, Aldh1l1 expression
novel transcripts expressed by a target cell population as well in spinal cord appears to continuously decline from a peaked
as a tool to compare quantitative expression data among sev- level at P2. Yang et al. also found that Aldh1l1 is upregulated
eral groups of cells. To this end, significant differences were in reactive astrocytes, similarly to GFAP. Thus, the expres-
detected between distinct populations of FACS isolated cells. sion of Aldh1l1 is developmentally regulated, and pathologi-
GFAP-GFP+/GLT1+ and GFAP-GFP–/GLT1+ astrocytes cal conditions also change its expression pattern. Exploring
isolated from 10- to 12-week-old animals only displayed a the transcriptome profile of astrocytes in vivo (Table 28.3,
subtle difference of approximately 1% in significantly enriched described in the following) hopefully will lead to discovery
genes, whereas age-matched GLT1+ astrocytes and Thy1+ of other novel cell-specific markers for which expression is
neurons had 14% of all present genes significantly expressed not regulated developmentally or by pathological processes.

T H E A S T R O C Y T E T R A N S C R I P TO M E • 351
Table 28.3 Enriched Genes in Cortical Astrocytes

1110020G09Rik Appl2 Ddah1 Fgfr1 Heph Mtmr11


1200009O22Rik Aqp4 Dio2 Fgfr3 Hes5 Myo6
1700019G17Rik Aqp9 Dlg5 Fjx1 Hhatl Nat1
1810014B01Rik Arhgef26 Dmp1 Fkbp10 Hopx Nat8
2310022B05Rik Asrgl1 Dmrta2 Fzd1 Hrsp12 Ndp
2610017I09Rik Atp13a4 Dpy19l3 Fzd2 Hsd11b1 Ndrg2
2610034M16Rik Atp1a2 Dtna Gabrg1 Htra1 Nkain4
2810459M11Rik Atp1b2 E130114P18Rik Gas1 Id4 Nkx2–2
2900052N01Rik Baalc E130304F04Rik Gdf10 Igdcc4 Npas3
4933431E20Rik Bcan Ednrb GFAP Igfbp2 Nr2e1
5730469M10Rik Bmpr1b Efhd1 Ghr Il18 Nrarp
A930005H10Rik Car8 Efs Gja1 Il33 Ntrk2
Aass Cbr3 EGFR Gjb6 Itih3 Ntsr2
Abcd2 Cbs Elmod3 Gldc Kcnj16 Nwd1
Abhd3 Cdh19 Elovl2 Gli2 Kcnn2 Olfml1
Abtb2 Chi3l1 Emx2 Gli3 Kctd14 Oplah
Acad11 Chpt1 Enho Glis3 Klf15 Pacrg
Acot1 Chrdl1 Entpd2 Glud1 Lcat Padi2
Acot11 Chst2 Ephx2 Gm11627 Lgi4 Papss2
Acsbg1 Cideb Eps15 Gm2115 Lgr4 Paqr8
Acsl3 Cldn10 Eps8 Gm266 Lhx2 Pax6
Acsl6 Clmn Eya1 Gm3932 Lrig1 Pdk4
Acss1 Cml1 Ezr Gm5089 Lsamp Pdlim4
Acss2 Cml5 F3 Gm715 Lxn Pdpn
Adhfe1 Cmtm5 Fabp5 Gm7901 Maob Pex11a
Adk Crot Fabp7 Gnao1 Masp1 Pfn4
Adora2b Cspg5 Fads1 Gpam Meis1 Pgcp
Agl Cth Fads2 Gpc5 Metrn Pgm2
Agpat5 Ctso Fam107a Gpld1 Mfge8 Phgdh
Agt Cxcl14 Fam176a Gpr37l1 Mfrp Phka1
Agxt2l1 Cyp2j6 Fam181b Gprc5b Mgst1 Phkg1
AI464131 Cyp2j9 Fam20a Gramd3 Mlc1 Pla2g7
AI848258 Cyp4f14 Fam55d Grin2c Mmd2 Plcd4
Aldh1l1 Cyp4f15 Fam59b Gstm1 Mras Pld2
Aldoc Cyp7b1 Fars2 Gstm5 Msi1 Plekhb1
Aox1 Daam2 Fbxo2 Hapln1 Mt1 Pm20d1
Apoe Dbi Fgf1 Hepacam Mt2 Polr3h

352 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Table 28.3 (Cont’d )

Ppap2b Pygb S100b Slc1a3 Sox9 Tspan7

Ppil6 Pygm Sc4mol Slc25a18 Spag1 Tst

Ppp1r3c Rab30 Scara3 Slc27a1 Spag5 Ttpa

Ppp1r3d Rab34 Scd1 Slc39a12 Spire1 Ttyh1

Ppp1r3g Ranbp3l Scg3 Slc41a1 Spsb4 Tubb2b

Prdm16 Rftn2 Scrg1 Slc48a1 Srebf1 Tulp3

Prdx6 Rfx4 Sdc4 Slc4a4 Svip Ugp2

Prkd1 Rgma Selenbp1 Slc6a11 Sypl2 Vav3

Proca1 Rgs20 Sfxn5 Slc7a10 Timp4 Vcam1

Prodh Rhobtb3 Sh3pxd2b Slc7a11 Tlcd1 Vegfa

Prrx1 Rlbp1 Slc13a3 Slc9a3r1 Tmem229a Wnt7a

Psat1 Rnf182 Slc13a5 Smpd2 Tmem47 Wscd1

Psd2 Rorb Slc14a1 Smpdl3a Tom1l1 Zfp521

Ptplb Rpe65 Slc15a2 Sox2 Tprkb Zfp783

Pxmp2 S100a1 Slc1a2 Sox21 Tsc22d4


This list was produced by extracting the intersection of astrocyte-enriched genes defined by a fold change ≥log2 2 and a FDR-corrected p-value ≤.05 among three previ-
ously published datasets (Cahoy et al. 2008; Doyle et al. 2008; Lovatt et al. 2007). Astrocyte-enriched genes from Lovatt et al. were produced by comparing adult cortical
GFAP-GFP+/GLT1+ astrocytes with GFAP-GFP–/GLT1– cells (N = 3). Astrocyte-enriched genes from Cahoy et al. were produced by comparing P16–17 FACS
and panning-isolated astrocytes (N = 5) to P16–17 panning-isolated neurons (N = 3). Astrocyte-enriched genes from Doyle et al. were produced by comparing TRAP
affinity-purified mRNA from BAC-transgenic Aldh1l1 mice. From Doyle et al., each sample (N = 4) derived from several 8- to 10-week-old mice, and was compared with
unbound affinity-purified samples (N = 2). Each of the three datasets was normalized individually using the RMA algorithm in R/Bioconductor. A representative probe set
was chosen for cases where a gene had more than one probe set on the platform. This was done by choosing the probe set with the lowest p-value for that gene. When there
was a p-value tie, the probeset with the higher value fold change was chosen.

Identification of such genes are essential for objective analy- (see Table 28.3; and see legend for analysis details). Notably,
sis of disease-related changes in the astrocytic transcriptome the transcripts on this list are enriched in astrocytes, but may
because it will allow unbiased collection of mRNA across vari- also be present in other cells. In addition, among the tran-
ous stages of the disease of interest. scripts in Table 28.3 that are truly astrocyte-specific, these
might be expressed only by a subset of cortical astrocytes as
opposed to all cortical astrocytes. Therefore, validation at the
5 C O M PA R I S O N O F C O RT I C A L transcript or protein level is necessary before final conclusions
A S T R O C Y T E T R A N S C R I P TO M E S can be drawn about their expression pattern in the brain. The
list can be used as a guide to identify novel astrocyte markers
To date, three studies have produced the transcriptome from because many of them are likely to be expressed above the aver-
cortical astrocytes (Cahoy et al. 2008; Doyle et al. 2008; age mRNA and protein levels. Given higher relative expres-
Lovatt et al. 2007). Each of these studies produced the astro- sion, it is likely that the protein products of the transcripts
cyte transcriptome with slight differences in the experimental in Table 28.3 may be identified with ease using antibody
design. For instance, Cahoy et al. used adolescent brain tissue markers or the promotors of these genes to drive expression
(P17), whereas Doyle et al. and Lovatt et al. used adult brain of reporter genes, such as GFP. The list can also be used to
tissue from 6- to 8- and 10- to 12-week-old animals, respec- confirm existing functions, as well as define new tasks of astro-
tively. Also, Lovatt et al. used FACS isolation of GFAP-GFP+/ cytes. Validation of the list confirmed the expression of genes
GLT1+ cells, Cahoy et al. used a combination of FACS and expected in astrocytes, such as Aldh1l1, AQP4 (aquaporin
immunopanning, and Doyle et al. used TRAP technology 4), GFAP, Gjb6 (connexin 30), Gja1 (connexin 43), S100b,
to isolate astrocytes from the Aldh1l1 BAC transgenic line and Slc1a2 (GLT1), as well as the absence of marker genes
(see Fig. 28.1). However, all of these data sets used the same of other cell types. A gene ontology (GO) analysis of these
microarray platform, and a direct comparison of them provides 311 genes revealed that 113 were assigned the GO term cata-
important information with regard to astrocyte-enriched tran- lytic activity and 36 was assigned transporter activity, in agree-
scripts. A direct comparison resulted in a list of 311 astrocyte- ment with previous reports suggesting that astrocytes express
enriched transcripts that are shared among all three data sets many transporters. Among genes with receptor activity were,

T H E A S T R O C Y T E T R A N S C R I P TO M E • 353
for instance, EGFR (epidermal growth factor receptor), perivascular glia, and ependymal glia. The classification of
GABAARG1 (gamma-aminobutyric acid (GABA) A receptor, these cell subtypes is primarily based on location and to some
gamma 1), Fgfr1 (fibroblast growth factor receptor 1), Fgfr3 extent on morphology. How these cell types differ functionally
(fibroblast growth factor receptor 3), Rorb (RAR-related is only beginning to be deciphered, and is reviewed in detail
orphan receptor beta), Nr2e1 (nuclear receptor subfamily in other chapters and reviews (Matyash and Kettenmann
2, group E, member 1), Ntsr2 (neurotensin receptor 2), and 2010; Oberheim et al. 2012; Zhang and Barres 2010) (see
Eps15 (epidermal growth factor receptor pathway substrate also chapter 4). However, the global set of information gath-
15). Other genes found on the list included Sox2, which is a ered by transcriptome profiling makes it an ideal method to
transcription factor previously characterized in self-renewing qualitatively assess astrocytic heterogeneity. Although several
progenitor cells and has been used in directed reprogramming examples of astrocyte heterogeneity exist in the literature
of somatic cells to produce induced pluripotent stem (iPS) (Bordey and Sontheimer 2000; Emsley and Macklis 2006;
cells (Graham et al. 2003; Nakagawa et al. 2008). When Sox2 Houades et al. 2006; Matthias et al. 2003; Nimmerjahn et al.
expression ceases or is inhibited in progenitor cells, they exit 2009; Takata and Hirase 2008; Walz 2000; Walz and Lang
the cell cycle and differentiate into neuroblasts. Forced expres- 1998; Zhou and Kimelberg 2000; Zhu and Kimelberg 2004),
sion of a battery of trasncription factors, including Sox2, none of them is capable of simultaneously addressing the large
induced “stemness” and the potential to divide. However, number of genes that transcriptome profiling does. Still, stud-
radial glia cells from adult brain, which are known to express ies showing that single transcripts or proteins differ among
GFAP and stem cell–like properties, also express Sox2 (Marko astrocytes motivate global profiling studies that will establish
et al. 2011). Whether cortical protoplasmic astrocytes, or even and characterize transcriptional differences, and hopefully
a small subset of them, express Sox2 at the protein level and establish astrocytic subtypes that were previously unknown.
harbor the potential to self-renew remains to be explored. The A split-Cre technology have recently been used to enriched
finding of Sox2 expression in astrocytes is interesting because adult neural stem cells by isolating cells with coincident activ-
astrocytes have previously been suggested to maintain the ity of the hGFAP and prominin1 promoters in mouse sub-
potential to divide into adulthood (Steindler and Laywell ependymal zone in vivo (Beckervordersandforth et al. 2010.
2003). Other genes with transcription factor activity included A similar approach could be utilzed to isolaed subpopupation
Srebf1 (sterol regulatory element binding transcription factor of astrocytes. An example of heterogeneity is the expression
1), Glis3 (GLIS family zinc finger 3), Emx2 (empty spiracles of the astrocyte marker glial fibrillary acid protein (GFAP),
homolog 2), and Pax6 (paired box gene 6) just to name a which can vary significantly among astrocytes from various
few. Tables 28.3 and tables of the complete transcriptome of regions in the adult mouse brain. Glial fibrillary acid protein
cortical astrocytes from 3 published data sets can be found is a good marker of protoplasmic and fibrous astrocytes in the
on http://www.networkglia.eu/en/genomics_screens may developing neocortex, but in adulthood cortical astrocytes in
serve as tools to discover novel astrocyte functions and per- the deeper layers downregulate GFAP (Lovatt et al. 2007).
haps identify novel markers labeling subsets of cortical pro- Lovatt et al. found that cortical astrocytes that were positive
toplasmic astrocytes. In addition to Table 28.3, we have also for GLT1 could be grouped as either GFAP positive or nega-
included the entire transcriptome from the same analysis in tive. However, analysis of the transcriptome between these
http://www.networkglia.eu/en/genomics_screens. two populations only demonstrated that a subtle difference
of 1% of all the expressed genes was significantly different
between the two groups, and the majority of these were not
6 H ET E R O G E N E I T Y A M O N G astrocyte specific genes. In comparison, 14% of all expressed
ASTROCY TES IN THE genes were differentially expressed by GLT1+ astrocytes and
MAMMALIAN BRAIN Thy1+ neurons. This finding suggests that the two popula-
tions of cortical astrocytes distinguished by GFAP expression
Astrocytes can be found throughout the brain, and have been are very similar at the transcriptional level. However, one has
proposed to participate in a number of functions including to keep in mind that posttranscriptional regulation may play
metabolic support of neurons, potassium buffering, synapse a role in determining levels, location and activity of protein
development, and regulation of synaptic transmission as products, and that level of a transcript and its protein prod-
reviewed in detail in other chapters. But do all astrocytes carry uct is not always directly correlated. Moreover, heterogeneity
out the same functions? Are there differences among astro- among astrocytes also tends to decrease detectable differ-
cytes from different brain regions? And how similar are astro- ences in gene expression among the populations of cells to
cytes within the same region? To date, astrocyte heterogeneity be compared.
remains a largely unexplored area with only a few published This is supported by a gene expression study investi-
gene-profiling studies comparing astrocytes across various gating regional differences among whole tissue samples
regions (inter-regional heterogeneity) or within regions from different cortical layers. This study found that
(intra-regional heterogeneity). cortical layer-specific gene expression paired to known
Among the various regions in the mammalian brain, neuron-specific genes are 63% more likely to exhibit lay-
several types of astrocytic glia have been described, includ- ered patterning than being unpatterned. In contrast,
ing tanycytes, Müller cells, radial glia cells, Bergmann glia, astrocyte-specific genes are 17% less likely to exhibit layered
protoplasmic astrocytes, fibrous astrocytes, marginal glia, patterning than being unpatterned. Although this finding

354 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
suggests that neurons exhibit more heterogeneity with heterogeneity or single astrocyte heterogeneity remains
respect to cortical layers than astrocytes, it still demon- an unexplored area (Doyle et al. 2008; Lovatt et al. 2007).
strates that astrocytic gene expression differs across layers, Future studies and improvements in methods capable of
calling for studies that decipher subpopulations of astro- producing transcriptome profiles from single astrocytes
cytes in cortical layers (Belgard et al. 2011). will provide exciting insight into the diversity among astro-
Doyle et al. used the TRAP-technology to generate the cytes, and perhaps reveal whether intraregional heterogene-
transcriptome of cortical Aldh1l1+ astrocytes and cerebellar ity among single protoplasmic astrocytes is as great as their
Sept4+ Bergmann glia (Doyle et al. 2008). A reanalysis of this numerous neighboring neuronal subtypes. Surprisingly, only
data (Lovatt and Nedergaard, unpublished data) showed that a single publication has used transcriptome analysis to study
approximately 62% of the 2,777 Bergmann glia enriched tran- astrocytes in the diseased brain so far (Lichter-Konecki et al.
scripts were shared by cortical astrocytes, and approximately 2008). In this study, a mouse with the X-linked UCD orni-
51% of the 3,354 cortical astrocyte–enriched transcripts were thine transcarbamylase (OTC) deficiency was crossed with
shared by Bergmann glia. Among the intersecting transcripts the hGFAP-EGFP mouse, and FACS was used to purify
were several that would be expected to be found in both cell astrocytes from the brains of hyperammonemic and healthy
types, including Aldh1l1, S100b, Slc1a2 (GLT1), Slc1a3 Otcspf/GFAP-EGFP mice following microarray analyses
(GLAST), Kcnj10 (Kir4.1), Kcnj16 (Kir5.1), Gja1 (con- and qRT-PCR. The major observation was significant down-
nexin 43), Gjb6 (connexin 30), Glul (glutamine synthetase), regulation of the gap-junction channel connexin 43 and
GFAP, EGFR (epidermal growth factor receptor), AQP4 aquaporin 4 genes, concomitantly with a reduced expression
(aquaporin 4), and AQP9 (aquaporin 9). Both cell types also of astrocytic inward-rectifying potassium channels Kir4.1
expressed Adk (adenosine kinase) and Adora2b (adenosine and Kir5.1 in hyperammonemic mice (Lichter-Konecki et al.
receptor 2B), suggesting that both are capable of responding 2008). Thus, ammonia toxicity may suppress the expression
to adenosine and phosphorylating adenosine after reuptake. of proteins involved in astrocyte-mediated water and potas-
In addition, the relative high discrepancy between the corti- sium homeostasis.
cal astrocytes and Bergmann glia transcriptomes suggests that
despite the fact that both are “astrocytic glia,” they may carry
out significantly different functional roles in their respective 7 T E C H N I Q U E S U S E D TO VA L I DAT E
brain regions. For instance, Bergmann glia are known to be T R A N S C R I P TO M E F I N D I N G S
intimately associated with Purkinje cells and can respond to
both purinergic and glutaminergic signals with ion channel– Once transcriptome data have been mined and novel pat-
activated Ca2+ signals. In agreement with previous litera- terns of gene expression extracted, a necessary next step is to
ture, the transcriptome data showed that Bergmann glia, validate the findings using an independent method. For the
but not cortical astrocytes, expressed Gria1 (GluR1) and majority of the cases, validation assesses the cellular expres-
Gria4 (GluR4) ionotropic glutamate receptors (Matsui et al. sion pattern of candidate genes. First, validation can occur
2005), and the ionotropic ATP-gated P2rx7 (P2X7) receptor either at the RNA or protein level. In addition, functional
(Habbas et al. 2011). Other ligand-gated receptors expressed validation at the activity or behavioral level may also be able
exclusively by Bergmann glia included P2ry1 (P2Y1), Grin2b to add supporting evidence to a particular expression pattern
(NMDA2B), Grin3a (NMDA3A), and Grm3 (mGluR3). (Lovatt et al. 2007). To validate at the RNA level, one can per-
In contrast, cortical astrocytes expressed another set of form in situ hybridization or fluorescent in situ hybridization
receptors, including P2rx4 (P2X4), P2ry5 (P2Y5), and (FISH), which with single cell resolution can detect down to
Grin2c (NMDA2C) (Lovatt et al. 2007). Of note, BEST1, single molecules (Cahoy et al. 2008; Daneman et al. 2010;
a Ca2+-activated Cl– channel that may play a role in sus- Itzkovitz et al. 2012). If large amounts of RNA are available,
tained GABA release from Bergmann glial cells (Lee et al. then one can evaluate the candidate genes using quantitative
2010), according to the transcriptional analysis of cerebellum polymerase chain reaction (qPCR) (Lobo et al. 2006; Lovatt
(Doyle et al. 2008), is not selectively expressed by Bergmann et al. 2007). The advantage of FISH over qPCR is that FISH
glial cells because it also present in the unbound TRAP can be performed in intact tissue and thus is independent of
samples that contain both other glial types and Purkinje neu- the mRNA isolation method used for the transcriptome. On
rons. Cortical astrocytes, as opposed to Bergmann glia, also the other hand, whereas qPCR may use the same mRNA iso-
expressed enriched levels of Ldhb (lactate dehydrogenase 2), lation method employed for the transcriptome, qPCR may
suggesting that the metabolic compartmentalization of corti- be better at detecting low abundant transcripts that would be
cal astrocytes and neurons versus Bergmann glia and Purkinje difficult to detect by regular in situ hybridization. If valida-
cells may differ. Ldhb functions in cortical astrocytes to syn- tion takes place at the protein level, then immunohistochem-
thesize lactate for shuttling to neurons (Lovatt et al. 2007). istry may be an option if specific antibodies are available for
Further studies exploring the repertoire of receptors and their the candidate transcript. In addition, if information about the
functional role will deepen our understanding of astrocytes promoter region of the candidate gene is available, one can
in different brain regions, and how astrocytes interact with generate promoter-driven reporter mice, such as GFAP-GFP
their environment. or BAC-transgenic eGFP mice. Altogether, validation of tran-
Although only a few studies to date have explored scriptome data is necessary to confirm the correctness of the
interregional heterogeneity of astrocytes, intraregional data-mining methodology used.

T H E A S T R O C Y T E T R A N S C R I P TO M E • 355
8 L I N K TO T R A N S C R I P TO M E DATA Belgard TG, Marques AC, Oliver PL, Abaan HO, Sirey TM,
Hoerder-Suabedissen A, et al. 2011. A transcriptomic atlas of mouse
neocortical layers. Neuron 71(4):605–616.
Tables of the complete transcriptome of cortical astrocytes Bordey A, Sontheimer H. 2000. Ion channel expression by astrocytes in
from 3 published data sets can be found on http://www.net- situ: comparison of different CNS regions. Glia 30(1):27–38.
workglia.eu/en/genomics_screens. Bushong EA, Martone ME, Jones YZ, Ellisman MH. 2002. Protoplasmic
astrocytes in CA1 stratum radiatum occupy separate anatomical
domains. J Neurosci 22(1):183–192.
Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson
9 S U M M A RY A N D P E R S P E C T I VE S KS, et al. 2008. A transcriptome database for astrocytes, neurons,
and oligodendrocytes: a new resource for understanding brain devel-
Transcriptome profiling of astrocytes is a novel experimental opment and function. J Neurosci 28(1):264–278.
tool in glial research that has just begun to unravel the roles of Daneman R, Zhou L, Agalliu D, Cahoy JD, Kaushal A, Barres BA. 2010.
astrocytes in health and disease. Future studies will explore and The mouse blood-brain barrier transcriptome: a new resource for
understanding the development and function of brain endothelial
functionally validate the astrocyte transcriptome. Such studies cells. PLoS One 5(10):e13741.
may lead to a better classification of astrocyte subtypes and dis- Dombeck DA, Khabbaz AN, Collman F, Adelman TL, Tank DW. 2007.
covery of novel roles executed by astrocytes. Gene expression Imaging large-scale neural activity with cellular resolution in awake,
studies will also explore the transcriptome of reactive astro- mobile mice. Neuron 56(1):43–57.
cytes, which remain largely unexplored by global transcriptom- Doyle JP, Dougherty JD, Heiman M, Schmidt EF, Stevens TR, Ma G,
et al. 2008. Application of a translational profi ling approach for the
ics. Such studies could lead to a better classification of diseased comparative analysis of CNS cell types. Cell 135(4):749–762.
astrocytes and possibly to medical interventions to treat neuro- Dugas JC, Mandemakers W, Rogers M, Ibrahim A, Daneman R, Barres
logical diseases in which reactive gliosis is a hallmark. Studies are BA. 2008. A novel purification method for CNS projection neu-
also needed to explore the transcriptomes of single astrocytes to rons leads to the identification of brain vascular cells as a source
gain insight into single cell heterogeneity, and how gene expres- of trophic support for corticospinal motor neurons. J Neurosci
28(33):8294–8305.
sion differences correlate with astrocyte location, morphology, Emsley JG, Macklis JD. 2006. Astroglial heterogeneity closely reflects
and physiological characteristics. Such studies could lead to a the neuronal-defined anatomy of the adult murine CNS. Neuron
better classification of neocortical astrocytes beyond the proto- Glia Biol 2(3):175–186.
plasmic and fibrous types. Espina V, Wulfkuhle JD, Calvert VS, VanMeter A, Zhou W, Coukos G,
Although methods to isolate mRNA from cells in vivo et al. 2006. Laser-capture microdissection. Nat Protoc 1(2):586–603.
Feng G, Mellor RH, Bernstein M, Keller-Peck C, Nguyen QT, Wallace
and in situ are continuously being improved, recent develop- M, et al. 2000. Imaging neuronal subsets in transgenic mice express-
ments in RNA-seq techniques now allow an unprecedented ing multiple spectral variants of GFP. Neuron 28(1):41–51.
global view of the transcriptome and its organization, includ- Ferraiuolo L, Higginbottom A, Heath PR, Barber S, Greenald D, Kirby
ing identification of introns, and novel 5′ and 3′ boundaries. J, et al. 2011. Dysregulation of astrocyte-motoneuron cross-talk in
To date, the application of RNA-seq techniques to astrocytes mutant superoxide dismutase 1-related amyotrophic lateral sclerosis.
Brain 134(Pt 9):2627–2641.
in situ or in vivo remains unpublished, and future studies Foo LC, Allen NJ, Bushong EA, Ventura PB, Chung WS, Zhou L, et al.
are likely to reveal novel insights into gene transcription in 2011. Development of a method for the purification and culture of
astrocytes. rodent astrocytes. Neuron 71(5):799–811.
The true power of transcriptome analysis has only just Gong S, Zheng C, Doughty ML, Losos K, Didkovsky N, Schambra UB,
begun to be unraveled. Further inspection and functional vali- et al. 2003. A gene expression atlas of the central nervous system based
on bacterial artificial chromosomes. Nature 425(6961):917–925.
dation of the overwhelming amount of data in the astrocytic Graham V, Khudyakov J, Ellis P, Pevny L. 2003. SOX2 functions to
transcriptome will discover novel markers and functional roles maintain neural progenitor identity. Neuron 39(5):749–765.
of astrocytes that would have been difficult or perhaps impos- Habbas S, Ango F, Daniel H, Galante M. 2011. Purinergic signaling in
sible to discover without transcriptome profiling. the cerebellum: Bergmann glial cells express functional ionotropic
P2X(7) receptors. Glia 121(1):4–27.
Heiman M, Schaefer A, Gong S, Peterson JD, Day M, Ramsey KE, et al.
2008. A translational profi ling approach for the molecular character-
AC K N OW L E D G M E N T S ization of CNS cell types. Cell 135(4):738–748.
Hempel CM, Sugino K, Nelson SB. 2007. A manual method for the
The authors thank Adam Cornwell for technical assistance purification of fluorescently labeled neurons from the mammalian
with the bioinformatics. brain. Nat Protoc 2(11):2924–2929.
Houades V, Rouach N, Ezan P, Kirchhoff F, Koulakoff A, Giaume C.
2006. Shapes of astrocyte networks in the juvenile brain. Neuron
Glia Biol 2(1):3–14.
REFERENCES Itzkovitz S, Lyubimova A, Blat IC, Maynard M, van Es J, Lees J, et al.
2012. Single-molecule transcript counting of stem-cell markers in the
Agulhon C, Petravicz J, McMullen AB, Sweger EJ, Minton SK, Taves mouse intestine. Nat Cell Biol 14(1):106–114.
SR, et al. 2008. What is the role of astrocyte calcium in neurophysiol- Lee S, Yoon BE, Berglund K, Oh SJ, Park H, Shin HS, et al. 2010.
ogy? Neuron 59(6):932–946. Channel-mediated tonic GABA release from glia. Science
Beckervordersandforth, R., Tripathi, P., Ninkovic, J., Bayam, E., Lepier, 330(6005):790–796.
A., Stempfhuber, B., Kirchhoff, F., Hirrlinger, J., Haslinger, A., Lie, Lichter-Konecki U, Mangin JM, Gordish-Dressman H, Hoffman EP,
D.C., et al. (2010). In vivo fate mapping and expression analysis reveals Gallo V. 2008. Gene expression profi ling of astrocytes from hyper-
molecular hallmarks of prospectively isolated adult neural stem cells. ammonemic mice reveals altered pathways for water and potassium
Cell stem cell 7, 744–758. homeostasis in vivo. Glia 56(4):365–377.

356 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Lobo MK, Karsten SL, Gray M, Geschwind DH, Yang XW. 2006. myelinogenic, migration-competent and efficiently engrafting
FACS-array profi ling of striatal projection neuron subtypes in juve- human oligodendrocyte progenitor cells. Nature biotechnology 29,
nile and adult mouse brains. Nat Neurosci 9(3):443–452. 934–941.
Lovatt D, Sonnewald U, Waagepetersen HS, Schousboe A, He W, Lin Sim, F.J., Lang, J.K., Waldau, B., Roy, N.S., Schwartz, T.E., Pilcher,
JH, et al. 2007. The transcriptome and metabolic gene signature W.H., Chandross, K.J., Natesan, S., Merrill, J.E., and Goldman, S.A.
of protoplasmic astrocytes in the adult murine cortex. J Neurosci 2006. Complementary patterns of gene expression by human oligo-
27(45):12255–12266. dendrocyte progenitors and their environment predict determinants
MacVicar BA. 1984. Voltage-dependent calcium channels in glial cells. of progenitor maintenance and differentiation. Annals of neurology
Science 226(4680):1345–1347. 59, 763–779.
Marko K, Kohidi T, Hadinger N, Jelitai M, Mezo G, Madarasz E. 2011. Sim, F.J., Windrem, M.S., and Goldman, S.A. 2009. Fate determina-
Isolation of radial glia-like neural stem cells from fetal and adult mouse tion of adult human glial progenitor cells. Neuron glia biology 5,
forebrain via selective adhesion to a novel adhesive peptide-conjugate. 45–55.
PLoS One 6(12):e28538. Simard M, Arcuino G, Takano T, Liu QS, Nedergaard M. 2003. Signaling
Martina M, Schultz JH, Ehmke H, Monyer H, Jonas P. 1998. Functional at the gliovascular interface. J Neurosci 23(27):9254–9262.
and molecular differences between voltage-gated K+ channels of Steindler DA, Laywell ED. 2003. Astrocytes as stem cells: nomenclature,
fast-spiking interneurons and pyramidal neurons of rat hippocam- phenotype, and translation. Glia 43(1):62–69.
pus. J Neurosci 18(20):8111–8125. Sugino K, Hempel CM, Miller MN, Hattox AM, Shapiro P, Wu C, et al.
Matsui K, Jahr CE, Rubio ME. 2005. High-concentration rapid tran- 2006. Molecular taxonomy of major neuronal classes in the adult
sients of glutamate mediate neural-glial communication via ectopic mouse forebrain. Nat Neurosci 9(1):99–107.
release. J Neurosci 25(33):7538–7547. Surmeier DJ, Song WJ, Yan Z. 1996. Coordinated expression of dop-
Matthias K, Kirchhoff F, Seifert G, Huttmann K, Matyash M, amine receptors in neostriatal medium spiny neurons. J Neurosci
Kettenmann H, et al. 2003. Segregated expression of AMPA-type 16(20):6579–6591.
glutamate receptors and glutamate transporters defines distinct Takano T, Tian GF, Peng W, Lou N, Libionka W, Han X, et al. 2006.
astrocyte populations in the mouse hippocampus. J Neurosci Astrocyte-mediated control of cerebral blood flow. Nat Neurosci
23(5):1750–1758. 9(2):260–267.
Matyash V, Kettenmann H. 2010. Heterogeneity in astrocyte morphol- Takata N, Hirase H. 2008. Cortical layer 1 and layer 2/3 astrocytes
ogy and physiology. Brain Res Rev 63(1–2):2–10. exhibit distinct calcium dynamics in vivo. PLoS One 3(6):e2525.
McCarthy KD, de Vellis J. 1980. Preparation of separate astroglial and Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, et al. 2009.
oligodendroglial cell cultures from rat cerebral tissue. J Cell Biol mRNA-Seq whole-transcriptome analysis of a single cell. Nat
85(3):890–902. Methods 6(5):377–382.
Molyneaux BJ, Arlotta P, Hirata T, Hibi M, Macklis JD. 2005. Fezl is Theodosis DT, Poulain DA, Oliet SH. 2008. Activity-dependent struc-
required for the birth and specification of corticospinal motor neu- tural and functional plasticity of astrocyte-neuron interactions.
rons. Neuron 47(6):817–831. Physiol Rev 88(3):983–1008.
Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, Aoi Walz W. 2000. Controversy surrounding the existence of discrete
T, et al. 2008. Generation of induced pluripotent stem cells with- functional classes of astrocytes in adult gray matter. Glia 31(2):
out Myc from mouse and human fibroblasts. Nat Biotechnol 95–103.
26(1):101–106. Walz W, Lang MK. 1998. Immunocytochemical evidence for a distinct
Nedergaard M, Verkhratsky A. 2012. Artifact versus reality-How astro- GFAP-negative subpopulation of astrocytes in the adult rat hip-
cytes contribute to synaptic events? Glia 60:1013–1023. pocampus. Neurosci Lett 257(3):127–130.
Nimmerjahn A, Mukamel EA, Schnitzer MJ. 2009. Motor behavior Wang X, Lou N, Xu Q, Tian GF, Peng WG, Han X, et al. 2006.
activates Bergmann glial networks. Neuron 62(3):400–412. Astrocytic Ca(2+) signaling evoked by sensory stimulation in vivo.
Oberheim NA, Goldman SA, Nedergaard M. 2012. Heterogeneity of Nat Neurosci 9:816–823.
astrocytic form and function. Meth Mol Biol 814:23–45. Wang Z, Gerstein M, Snyder M. 2009. RNA-Seq: a revolutionary tool
Okaty BW, Sugino K, Nelson SB. 2011. A quantitative comparison of for transcriptomics. Nat Rev Genet 10(1):57–63.
cell-type-specific microarray gene expression profi ling methods in the Wilhelm A, Volknandt W, Langer D, Nolte C, Kettenmann H,
mouse brain. PLoS One 6(1):e16493. Zimmermann H. 2004. Localization of SNARE proteins and secre-
Pekny M, Nilsson M. 2005. Astrocyte activation and reactive gliosis. tory organelle proteins in astrocytes in vitro and in situ. Neurosci Res
Glia 50(4):427–434. 48(3):249–257.
Ransom B, Behar T, Nedergaard M. 2003. New roles for astrocytes (stars Yang Y, Vidensky S, Jin L, Jie C, Lorenzini I, Frankl M, et al. 2011.
at last). Trends Neurosci 26(10):520–522. Molecular comparison of GLT1+ and ALDH1L1+ astrocytes in vivo
Regan MR, Huang YH, Kim YS, Dykes-Hoberg MI, Jin L, Watkins in astroglial reporter mice. Glia 59(2):200–207.
AM, et al. 2007. Variations in promoter activity reveal a dif- Zhang Y, Barres BA. 2010. Astrocyte heterogeneity: an underappreciated
ferential expression and physiology of glutamate transporters topic in neurobiology. Curr Opin Neurobiol 20(5):588–594.
by glia in the developing and mature CNS. J Neurosci 27(25): Zhou M, Kimelberg HK. 2000. Freshly isolated astrocytes from rat hip-
6607–6619. pocampus show two distinct current patterns and different [K(+)](o)
Sanz E, Yang L, Su T, Morris DR, McKnight GS, Amieux PS. 2009. uptake capabilities. J Neurophysiol 84(6):2746–2757.
Cell-type-specific isolation of ribosome-associated mRNA from Zhu Y, Kimelberg HK. 2004. Cellular expression of P2Y and beta-AR
complex tissues. Proc Natl Acad Sci U S A 106(33):13939–13944. receptor mRNAs and proteins in freshly isolated astrocytes and tis-
Schummers J, Yu H, Sur M. 2008. Tuned responses of astrocytes and sue sections from the CA1 region of P8–12 rat hippocampus. Brain
their influence on hemodynamic signals in the visual cortex. Science Res Dev Brain Res 148(1):77–87.
320(5883):1638–1643. Zhuo L, Sun B, Zhang CL, Fine A, Chiu SY, Messing A. 1997. Live astro-
Sim, F.J., McClain, C.R., Schanz, S.J., Protack, T.L., Windrem, M.S., cytes visualized by green fluorescent protein in transgenic mice. Dev
and Goldman, S.A. 2011. CD140a identifies a population of highly Biol 187(1):36–42.

T H E A S T R O C Y T E T R A N S C R I P TO M E • 357
29.
GENE EXPRESSION PATTERNS OF OLIGODENDROCY TE
PROGENITOR CELLS AND OLIGODENDROGLIA
Fraser J. Sim and Steven A. Goldman

A B B R E VI AT I O N S MBP myelin basic protein


MERTK Mer tyrosine kinase protooncogene
BAC bacterial artificial chromosome MOG myelin oligodendrocyte glycoprotein
BAMBI BMP and activin membrane-bound inhibitor NKX2–2 NK2 homeobox 2
BMP bone morphogenic protein O-2A oligodendrocyte and type-2 astrocyte
CD11b cluster of differentiation 11b OLIG1 oligodendrocyte lineage transcription
CD133 cluster of designation 133 (encoded by factor 1
PROM1 gene) OLIG2 oligodendrocyte lineage transcription
CD9 cluster of designation 9 factor 2
CHRDL1 chordin-like 1 OPC oligodendrocyte progenitor cell
CNP 2′,3-cyclic nucleotide 3′- OTP oligodendrocyte transmembrane protein
phosphodiesterase (CLDN11)
CNS central nervous system PDGFRA platelet-derived growth factor alpha
CNTN1 contactin/F3 receptor
Cre Cre recombinase PLP1 proteolipid protein 1
CSPG4 chondroitin sulfate proteoglycan-4 PPARγ peroxisome proliferator-activated receptor-
CSRP1 cysteine and glycine-rich protein 1 gamma
DEX differentially expressed PROM1 prominin 1 (encodes the CD133 antigen)
EGF epidermal growth factor PSA-NCAM polysialylated form of neural cell adhesion
EGFR epidermal growth factor receptor molecule
FACS fluorescence-activated cell sorting PTN pleiotrophin
GalC galactocerebroside C PTPRZ1 protein-tyrosine phosphatase, receptor-
GAS6 growth arrest-specific 6 type, zeta-1
GFAP glial fibrillary acidic protein Ran-2 rat neural antigen-2
GFP green fluorescent protein RISC RNA-induced silencing complex
GLT1 glutamate transporter 1 (SLC1A2) RT-PCR reverse transcription polymerase chain
GLTP glycolipid transfer protein reaction
GPR17 G protein–coupled receptor 17 S100E S100 calcium-binding protein, beta
GRB14 growth factor receptor-bound protein 14 SEPP1 selenoprotein P
GRP glia restricted precursor shRNAi short hairpin RNA
GSEA gene set enrichment analysis siRNA small interfering RNA
HAT histone acetyltransferase SOX10 Sry-Box 10
HDAC histone deacetylase SOX2 Sry-Box 2
HES-5 hairy and enhancer of split-5 ST8SIA1 ST8 alpha-N-acetyl-neuraminidase alpha-
HMGCR HMG-CoA reductase 2,8-siayltransferase 1
ID4 inhibitor of differentiation 4 STAT signal transducer and activator of transcription
IL6 interleukin 6 T3 triiodothyronine
INSIG1 insulin-induced gene 1 TCF7L1/2 transcription Factor 7-Like 1/2
JAG1 jagged 1 TF transcription factor
JAK janus kinase 1 TGFD transforming growth factor, alpha
LNX1 ligand of numb protein X1 TMEM10 transmembrane protein 10
MAPK mitogen-activated protein kinase TSA trichostatin A
MATN4 matrilin 4 ZB4 zinc finger BED domain-containing protein 4

358
1 INTRODUCTION mechanisms, as the gene expression profiles are derived from
an admixture of different cell types, so that cell type–specific
Oligodendrocyte progenitor cells (OPCs) pervade both or autonomous regulation can rarely be inferred. Recognizing
the gray and white matter parenchyma of the brain. this issue, the majority of extant databases addressing neural
Oligodendrocyte progenitor cells have typically been noted and glial gene expression were generated from either sorted
to be intrinsically bipotential for astrocytes as well as oligo- cells profiled immediately following isolation, or from cells
dendrocytes, and have thus been interchangeably called glial cultured for extended periods of time (>2 days) (see Table 29.1
progenitor cells. Yet recent studies have found that both for a summary of studies).
during development and following injury, the principal role As such, it is important to note the differences in lineage
of these cells is to generate new myelinogenic oligodendro- potential between OPCs in tissue culture, and those in vivo,
cytes (Kang et al. 2010; Rivers et al. 2008). As a result, these as revealed by recent fate mapping studies using transgenic lin-
cells are most typically referred to as oligodendrocyte progeni- eage tracing (for review, Richardson et al. 2011) (see chapters
tor cells (OPCs), and are referred to as such in this chapter. 10 and 13). Isolated glial cells from rodent adult and neonatal
Nonetheless, it is important to note that in culture, OPCs optic nerve had previously been found to generate both oligo-
remain competent to generate astrocytes as well as oligoden- dendrocytes and astrocytes in culture (ffrench-Constant and
drocytes, and under some conditions neurons as well (Nunes Raff 1986; Wolswijk and Noble 1989). This led to the termi-
et al. 2003; Richardson et al. 2011). The molecular basis of nology of O-2A progenitor or glial restricted precursor (GRP)
their largely oligodendrocytic differentiation in vivo remains a (discussed in Noble et al. 2004). Subsequent studies both in
mystery. Indeed, it is unclear what the function of these cells is vitro and following transplantation, revealed that at least a
in the normal adult brain; it is difficult to understand why 3% fraction of these precursors could be directed to generate
to 4% of all cells in the human white matter persist as resident neurons, whether by serum exposure–associated reprogram-
progenitors, absent a need for significant turnover. As such, it ming, or culture as neurosphere in the presence of mitogens
is critical for us to better understand both the natural history (Belachew et al. 2003; Kondo and Raff 2000; Nunes et al.
and molecular regulation of these cells. It is also incumbent on 2003). As such, the more generic term glial progenitor cell was
us to assess these cells using human OPCs, because substantial introduced, to include the less-restricted nature of these cells.
differences exist in both the biology and molecular control of Indeed, some studies have suggested that on activation, that
human and rodent OPCs. Therefore, this chapter focuses on parenchymal glial progenitor cells might acquire a neural stem
the molecular regulation of the homeostatic maintenance, self- cell–like character (Costa et al. 2010; Richardson et al. 2011).
renewal, lineage specification, and differentiation of human However, data from adult human OPCs indicated that these
OPCs. Each of these topics has become a focus of investiga- cells could not undergo unlimited self-renewal, and did not
tion over the past several years, as new technologies for inves- express detectable telomerase activity, suggesting that while
tigating cell-type specific gene expression and transcriptional these cells might have multilineage competence, that do not
regulation have become available. Whole genome analysis of behave as stem cells (Nunes et al. 2003). Rather, these data sug-
gene expression, chromatin organization, and miRNA regula- gested that OPCs comprise a multipotent transit-amplifying
tion represent unbiased approaches to better understand both progenitor, whose lineage choices are profoundly affected by
the natural history and disease-associated responses of OPCs. the in vitro environment (Goldman 2003).
In particular, microarray technologies and next-generation In contrast to the multilineage potential of OPCs in
high throughput sequencing have accelerated the analysis of vitro, a number of recent lineage tracing studies in vivo, using
gene expression on a genome-wide scale, allowing us to assess Cre-lox technology with BAC-driven reporters for NG2,
OPC gene expression as a function of age, developmental PDGFRA, and OLIG2, have largely concluded that in post-
stage, and differentiation, as well as to compare the expression natal brain OPCs generate only oligodendrocytes in vivo
signatures of human OPCs to both their rodent counterparts, (Kang et al. 2010; Rivers et al. 2008) (see chapters 10 and
and their differentiated astrocytic and oligodendrocytic deriv- 13 for a detailed description). The lack of astrocytic differ-
atives. The focus of this chapter is to discuss the insights that entiation in the adult mouse stands in direct contrast to the
genomic approaches have provided to our understanding of observations made using isolated cells. What is the basis for
the signal control and environmental responsiveness of human the difference? What factors restrict cell lineage in vivo? Or
OPCs, and to our understanding of how best to mobilize and are OPCs comprised of an admixture of phenotypes of dif-
instruct human OPCs for therapeutic purposes. ferent lineage competencies? How do these differ between
the developing and adult brain? These questions have con-
siderable potential clinical importance. Might OPC trans-
2 L I N E AG E A N D FAT E P OT E N T I A L O F plants for the purpose of remyelination be associated with
G L I A L P R O G E N I TO R C E L L S ectopic astrocytic or neuronal generation, and their atten-
dant clinical complication of epileptogenesis? Do the gene
Genomic approaches of cells in tissue are subject to the same expression profiles of these cells, as obtained from sorted
limitations of other biochemical analyses in that they rely on isolates, accurately predict their responses to environmental
the specificity and homogeneity of the samples used for analy- signals in vivo? These issues are serially addressed in the con-
sis. The genomic analysis of unfractionated whole tissue rarely text of describing current knowledge of gene expression by
provides coherent information regarding cell type–specific human OPCs.

G E N E E X P R E S S I O N PAT T E R N S O F O L I G O D E N D R O C Y T E P R O G E N I TO R C E L L S A N D O L I G O D E N D R O G L I A • 359
Table 29.1 GENE AND miRNA EXPRESSION STUDIES OF OLIGODENDROCYTE PROGENITOR CELLS
METHOD OF OPC
PAPER ACCESSION PLATFORM ARRAYS SPECIES AGE PURIFICATION DESCRIPTION

Sim et al. (2011) GSE29368 Aff ymetrix 10 Human Fetal CD140a CD140a+ OPCs isolated from fetal human brain
HG-U133_Plus_2 21–22 weeks FACS
Sim et al. (2009) GSE36634 Aff ymetrix 24 Human Adult A2B5 A2B5+ adult human OPCs from white matter and cor-
HG-U133_Plus_2 30–46 years MACS tex, GLT1+ astrocytes, CD11b+ microglia
Sim et al. (2006) GSE26535 Aff ymetrix 6 Human Adult A2B5 A2B5-sorted adult human white matter-derived OPCs
HG_U95Av2 17–56 years MACS
Shankar et al. (2003) NA cDNA array Research 3 Human Fetal O4 Human O4+ immunopanned cells from spinal cord at
Genetics 18–23 weeks Immunopanning 18 and 22 gestational weeks
Cahoy et al. (2008) GSE9566 Aff ymetrix 48 Mouse Postnatal Serial Oligodendrocyte lineage cells isolated at p7 by immu-
Mouse430_2 day 7 Immunopanning nopanning and S100β-EGFP astrocytes and depleted
using PDGFαR, neurons
MOG and GalC
Gobert et al. (2009) GSE14406 Aff ymetrix 54 Mouse Cell line NA Mouse OPC cell line (Oli-neu) treated with various
Mouse430_2 compounds and profiled at 10, 24, and 72 h
Budde et al. (2010) GSE21797 Agilent mRNA 2 Mouse Embryonic Shaking method Comparison of cultured OLs and astrocytes and time
GSE21799 Exiqon miRNA 2 day 14–16 course in vitro
GSE21798 Agilent miRNA 8
Zhao et al. (2010) NA Rodentia miRNA NA Mouse Not None, tissue extracts Dicer1 conditional knock-out and Olig1 null optic
specified from Dicer1 cko mice nerve and spinal cord tissue compared with wild-type
Dugas et al. (2006) NA Aff ymetrix 96 Rat Postnatal A2B5 and GalC Isolation of A2B5+ and GalC+ cells, and time course of
day 7 (OPC) rat OPC differentiation in vitro
day 10–12 (OLs)
Lau et al. (2008) GSE11218 Aff ymetrix 8 Rat Postnatal A2B5 and GalC A2B5 and GalC-sorted cells at postnatal day 7
Rat230_2 day 7 FACS
Nielsen et al. (2006) GSE5940 Aff ymetrix 18 Rat Postnatal A2B5 and O4 FACS isolated A2B5+ OPCs and O4+ oligodendro-
RAE230A/B day 7 FACS cytes from day 7 pups
Dugas et al. (2010) NA miRNA 4 Rat Postnatal PDGFαR and GalC Immunopaned OPC and oligodendrocytes
not stated day 7 Immunopanning
Lin et al. (2009) NA Aff ymetrix 3 Rat Postnatal O4 Immunopanned O4+ cells isolated from neonatal fore-
RG-U34A day 2 Immunopanning brain (p2) and adult subcortical white matter (200–250
and adult g).
Lyssiotis et al. (2007) NA Aff ymetrix 8 Rat Postnatal Serial immunopanning Rat OPCs (p6) treated with either TSA (20 nM) or
Rat230_2 day 6 BMP-2 (20ng/ml) for 6, 12, 24, and 48 h.

This table summarizes the study design of expression studies referred to in this chapter. Several studies have examined gene expression in human, mouse, and rat development. The profile of oligodendrocyte progenitor cells has been
determined by isolation using FACS, MACS, and immunopanning with various cell-type selective markers.
A B

MBP

O4
CD140a– CD140a+
C D Chondroitin sulfate biosynthesis
Glycan structures - biosynthesis 1
Axon guidance
CD140a– cells CD140a+ cells Glycerolipid metabolism
Taste transduction
TCF7L1 Thyroid cancer
WNT pathway

Sphingolipid metabolism
TCF7L2 Cell adhesion molecules (CAMs)
Glycine, serine and threonine metabolism
PPAP2B Melanoma
Focal adhesion
CCND1 Glutamate metabolism
Vibrio cholerae infection
CNTN1 Lysine degradation
Notch pathway

ECM-receptor interaction
JAG1 TGF-beta signaling pathway
Olfactory transduction
MAML2 Fatty acid metabolism
Urea cycle and metabolism of amino groups
Glycerophos pholipid metabolism
HEY2 Propanoate metabolism
Tyrosine metabolism
EGFR Homologous recombination
EGFR pathway

Ether lipid metabolism


ERBB3 Glycolysis / Gluconeogenesis
Arachidonic acid metabolism
GRB14 Nicotinate and nicotinamide metabolism
Long-term depression
TGFA Pyruvate metabolism
Notch signaling pathway
CD9 Type II diabetes mellitus
Cell cycle
Phenylalanine metabolism
Nitrogen metabolism
−3 −1 1 3 Valine, leucine and isoleucine degradation
LogFC Ubiquitin mediated proteolysis
Glycos phingolipid biosynthesis - neo-lactoseries
GS

Figure 29.1 Determination of Fetal Human CD140a-Sorted Oligodendrocyte Progenitor Cell Expression Profile. A. Fetal human CD140a+ cells were
plated onto substrate and allowed to differentiate following removal of exogenous growth factors. Four days after FACS, CD140a+ cells had developed
characteristic immature oligodendrocyte morphology and expressed the sulfatide antigen O4. To determine myelin competence, CD140a+ cells were
transplanted into shiverer/rag2 hypomyelinating mice, which lack endogenous MBP. B. Photomicrograph of the corpus callosum and fimbria of an
engrafted shiverer mouse at 12 weeks; stained for myelin basic protein (MBP, green), showing substantial donor-derived myelin (scale, 200 μm).
C. Differential gene expression analysis of CD140a sorted cell profiles identified several functionally relevant genes. This heatmap shows significantly
differential expressed genes comprise members of WNT, Notch and EGFR pathways (>3 FC, and <5% FDR) (heatmap: red, high expression; green,
lower expression; color key indicates expression levels in log base 2). D. Parametric gene set enrichment analysis of CD140a+ OPCs identified several
KEGG cell-signaling and metabolic pathways as either enriched or depleted in human fetal OPCs. Significant pathways were selected based on a
moderated t-test statistic and 5% FDR cutoff. The results are illustrated in decreasing order of significance in a heatmap whereby each sorted cell
sample is represented by a single cell and relative enrichment compared to the CD140a– fraction is indicated by deepening red color, and depletion in
blue. Modified from Sim et al. 2011. Complete data available from NCBI GEO as GSE29368, and at www.findDB.org.
3 T H E P H E N OT Y P I C H ET E R O G E N E I T Y (the gene encoding NG2), OLIG2, SOX10, and NKX2–2.
O F G L I A L P R O G E N I TO R S Compared with A2B5+/PSA-NCAM– defined fetal OPCs,
CD140a+ myelinated the hypomyelinated shiverer brain more
Oligodendrocyte progenitor cells may arise from several rapidly and efficiently (Fig. 29.1B). Interestingly, although only
distinct sites during development in vivo (Richardson et al. a fraction of A2B5+/PSA-NCAM– defined cells expressed
2006). In the forebrain, OPCs and oligodendrocytes may CD140a antigenicity, only CD140a+ cells were capable of oli-
arise not only from classically described sources in the ventral godendrocyte differentiation in vitro.
germinal zones and ganglionic eminences, but also dorsally in To determine the whole genome expression profile of
the cortical subventricular zone (Cai et al. 2005; Levison and PDGFαR/CD140a-defined fetal OPCs, Aff ymetrix microar-
Goldman 1993; Vallstedt et al. 2005). Indeed, cortical ven- rays were performed immediately following isolation on sev-
tricular zone–derived progenitors appear to play a major role eral human brain isolates. The genomic profile of CD140a+
in the generation of oligodendrocytes in the corpus callosum cells shared many characteristics of OPCs in human and
and other cortical white matter tracts, even replacing a fraction rodent brain (Table 29.2). CD140a-sorted cells expressed sig-
of the earlier-generated medial ganglionic eminence-derived nificantly high levels of the prototypic OPC markers OLIG1
OPCs (Kessaris et al. 2006). The developmental pattern of (56-fold higher in PDGFαR+), OLIG2 (24-fold), NKX2.2
oligodendrocyte generation has yet to be determined in the (20-fold), PDGFRA (31-fold), SOX10 (31-fold), CSPG4/
human brain, but these data suggest that there may be mul- NG2 (11-fold), and ST8SIA1, the enzyme responsible for
tiple routes to oligodendrocyte fate. synthesis of the gangliosides recognized by the A2B5 anti-
Indeed, recent evidence suggests significant phenotypic het- body (3.3-fold). The high expression of PDGFRA, NG2, and
erogeneity among OPCs in embryonic rodent brain and cell cycle ST8SIA1/SIAT8A resembles the phenotype of adult human
kinetics in adult brain (discussed in chapter 10). Importantly, OPCs (Sim et al. 2006) (discussed in detail in the following).
this heterogeneity may be lost to analysis when gene expression Similarly, S100ß, claudin 11 (OTP/CLDN11) and CNP were
profiles are based on cells isolated using single antigens or from also significantly upregulated in CD140a-sorted cells. Markers
multiple tissue regions. However, one advantage of gene expres- of other neural phenotypes, including oligodendrocytes, astro-
sion profiling is that high expression of genes encoding surface cytes, neurons, and neural stem cells were not highly expressed
proteins—such as receptors—provides additional candidates by by CD140a+ cells.
which to dissect heterogeneous populations. This has now been By comparing CD140a+ cells to the rest of the tissue dis-
achieved for fetal human OPCs, as described next. sociate, more than 400 genes whose expression was relatively
restricted to human OPCs could be identified (Sim et al. 2011).
Pathway analysis was then applied to identify functionally
4 G E N E E X P R E S S I O N BY F ETA L H U M A N related genes that were coordinately regulated. We identified
O L I G O D E N D R O C Y T E P R O G E N I TO R S evidence for the differential regulation of WNT, NOTCH, and
EGFR pathways (see Figs. 29.1C,D). Active WNT signaling
Oligodendrocyte progenitor cells have been isolated from the was suggested by the presence of high levels of two TCF tran-
fetal human brain using a number of techniques. The monoclo- scription factor isoforms, TCF7L1 and TCF7L2. Furthermore,
nal antibody A2B5 was first identified as a selective marker for gene set enrichment analysis (GSEA) revealed that WNT tar-
OPCs from isolates of rodent optic nerve, which do not contain get genes were significantly induced in CD140a+ cells, suggest-
neuronal cell bodies. Although many groups have used A2B5 ing active WNT signaling. Four notch regulators were found
to identify human OPCs from fetal brain (Ruffini et al. 2004; among the CD140a+ selective genes. These included the notch
Windrem et al. 2004), A2B5 is also expressed on the surface of ligands contactin/F3 (CNTN1) and jagged 1 ( JAG1), which
neuroblasts. As a result, concurrent sorting with depletion of may have opposing effects on OPC differentiation in rodents
PSA-NCAM–defined neuroblasts is needed to enrich OPCs (Hu et al. 2003). The epidermal growth factor (EGF) recep-
form the fetal brain using A2B5 (Windrem et al. 2004). tor family EGFR and erbB3 were highly enriched in CD140a+
In contrast to the lack of cell-type selectivity exhibited by cells. In addition, the SH3-adaptor protein GRB14, which
the gangliosides recognized by the A2B5 antibody, the PDGFα interacts with EGFR and the EGF-ligand TGFα, was signifi-
receptor appears to be a more specific marker by which to iden- cantly overexpressed. Interestingly, membrane-bound TGFα
tify and isolate oligodendrocyte progenitor cells. In the mouse may be primed to activate EGFR via the tetraspanin protein
brain, nearly all PDGFαR+ cells coexpress NG2, and vice versa CD9, which was also highly enriched in CD140a+ cells. CD9
(Rivers et al. 2008). On that basis, PDGFαR+ cells were isolated can strongly enhance EGFR activation via binding TGFα sug-
from the fetal human forebrain using an antibody that recog- gesting an OPC-selective mechanism for CD9-potentiated sig-
nizes the CD140a extracellular epitope of the PDGFαR pro- naling through EGFR (Shi et al. 2000). CD9 was not expressed
tein (Sim et al. 2011). CD140a+ cells comprised less than 2.5% by all CD140+ OPCs, thus permitting their further categorical
of cells in whole brain isolates before 18 weeks gestation, and subdivision (Sim et al. 2011). Indeed, this latter observation
just under 5% of cells in 21 to 23 weeks gestation cortical and highlights the power of combining cell isolation with micro
subcortical tissue. Importantly, CD140a+ cells rapidly differen- array as a means by which to both define and immunopheno-
tiated as oligodendrocytes in vitro (Fig. 29.1A). Quantitative type progressively more discrete cell lineages.
real-time RT-PCR revealed that these cells expressed high lev- Besides A2B5- and CD140a/PDGFαR–defined OPCs,
els of rodent OPC-expressed genes, such as PDGFRA, CSPG4 later-stage progenitors and oligodendrocytes have also been

362 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Table 29.2 FETAL HUMAN OLIGODENDROCYTE PROGENITOR CELL–EXPRESSED GENES
TYPE SYMBOL DESCRIPTION FC Q-VALUE TYPE SYMBOL DESCRIPTION FC Q-VALUE

Ligand BMP8B OP2 4.92 2.34E-08 Transcription SOX10 31.08 7.80E-12


TGFA TGF-alpha 5.49 4.35E-06 Factor SIX4 SIX homeobox 4 7.12 7.80E-12
BMP7 OP-1 4.41 3.27E-05 OLIG1 Oligodendrocyte transcription factor 1 55.52 3.46E-11
JAG1 Jagged1 3.31 5.76E-05 OLIG2 Oligodendrocyte transcription factor 2 24.15 9.93E-11
PDGFA PDGF-A polypeptide 3.18 6.13E-05 NKX2–2 NK2 related, locus 2 19.74 2.86E-09
DLK1 Delta-like 1 homolog 4.53 7.15E-05 TCF7L2 TCF4 3.09 2.64E-07
PTN Pleiotrophin; HB-GAM 3.65 1.65E-04 TCF7L1 TCF3 4.66 4.04E-07
IL8 Interleukin 8 5.86 2.51E-04 RFX4 Regulatory factor X, 4 4.21 4.34E-05
FGF1 Acidic FGF 4.69 1.84E-03 SOX8 3.11 1.65E-04
BMP2 3.49 3.10E-03 Enzyme B3GNT7 UDP-GlcNAc:betaGal 14.62 3.86E-12
Receptor BAMBI BMP pseudoreceptor 15.76 2.27E-12 B4GALT6 Beta 1,4- galactosyltransferase, 6 4.65 7.24E-10
PDGFRA PDGF receptor, alpha 30.80 3.26E-10 CSGALNACT1 Beta4GalNAcT 13.01 6.52E-07
polypeptide
CHRM3 Cholinergic receptor, mus- 17.08 1.71E-09 XYLT1 Xylosyltransferase I 3.06 8.44E-07
carinic 3
SEMA5A Semaphorin 5A 9.92 1.67E-07 ST8SIA1 Responsible for A2B5 synthesis 3.25 1.28E-06
PTPRE RTPε 5.16 1.29E-06 B3GAT2 Beta-1,3-glucuronyltransferase 2 4.37 1.60E-04
ERBB3 9.74 1.59E-06 HS6ST2 HS 6-O-sulfotransferase 2 3.58 3.38E-04
NTRK2 TrkB 7.02 2.37E-06 HS3ST3A1 HS 3-O-sulfotransferase 3A1 4.29 1.99E-03
PTPRG RTPγ 4.63 3.56E-04 HAS2 Hyaluronan synthase 2 4.40 2.78E-06
GPR17 G protein–coupled 3.65 5.83E-04 Other/Novel CCND1 Cyclin D1 13.18 2.82E-09
receptor 17
EGFR EGF receptor 3.30 6.63E-03
ECM/ CNTN1 Contactin 1 30.41 4.01E-15
Cell adhesion CSPG4 NG2 11.11 2.19E-09
MMP2 4.36 2.53E-06
OMG Oligodendrocyte myelin 9.66 2.62E-05
glycoprotein
GPC6 Glypican 6 4.40 3.19E-05
PROM1 CD133/prominin 1 6.48 5.53E-05
FBLN7 Fibulin 7 4.15 1.18E-04
HAPLN1 Hyaluronan and 5.26 4.34E-04
proteoglycan link 1
CD9 CD9 antigen (p24) 7.44 1.10E-03

Human CD140a+ OPCs were isolated using FACS from fetal brain between 21 and 22 weeks gestational age (n = 5) (Sim et al. 2011). Microarray profiles of CD140a+ and CD140a– cells were generated and compared using a
moderated t-test statistic with a 5% false-discovery rate cutoff. Individual differentially expressed transcripts were annotated by Gene Ontology and selected genes in each major category are shown here. The expression ratio of
positive: negative sorted cells are shown along with the FDR-corrected q-values to indicate significance. A complete list of fetal CD140a+ specific genes can be found at www.findDB.org.
isolated from the fetal spinal cord, using O4 sulfatide expres- was then found to potentiate human oligodendrocyte survival
sion (Shankar et al. 2003). Although O4 is expressed exclu- in vitro, via an AKT-dependent pathway (Shankar 2006). On
sively by postmitotic oligodendrocytes in human cells in vitro that basis, GAS6 infusion was subsequently found to reduce
(Armstrong et al. 1992), O4 is commonly used as a marker of oligodendrocyte death, and possibly promote remyelination,
progenitor cells in rodent brain (Mason and Goldman 2002). in response to cuprizone-induced demyelination (Tsiperson
Shankar and colleagues used O4-based immunopanning to et al. 2010).
isolate O4+ cells from an 18- to 22-week gestational age spinal
cord. Microarray analysis was solely focused on the expres-
sion of receptors, and data were presented only for kinases 5 C O M P L E M E N TA RY PAT T E R N S O F
expressed by O4+ cells. Nonetheless, the resultant data were G E N E E X P R E S S I O N BY A D U LT H U M A N
illuminating; among those highly expressed transcripts, the O P C S A N D W H I T E M AT T E R
GAS6 receptors, AXL, and MERTK stood out, suggesting
the attractiveness of these kinases as targets for oligodendro- In the human brain, OPCs were first identified using
glial fate modulation and support. Indeed, the GAS6 ligand PDGFαR or A2B5 antibodies (Scolding et al. 1998). Mitotic

A2B5+ OPC CD11b+ GLT1+ unsorted B


A CD36
Microglia
CD68
CD86
A2B5 +OPC vs. GLT1+astrocyte vs.
FCGR2A
ITGAM
unso rted unso rted
CDH5
Endothelial TEK
VWF
CLDN11
MAL 552 455 6435
Mature MBP 215 3137
Oligodendrocyte MOBP 343
MOG
PLP1
CSPG4 2
OPC PDGFRA
ST8SIA1
CNP
2 137 229
MAG 45 225
Oligodendrocyte NKX2−2
Lineage OLIG1
OLIG2
SOX10
AQP4 up
GFAP 829
Astrocyte GLUL
505 down
SLC1A2
TNC
ASCL1
NPC DCX
SOX1
FUT4 CD11b +microglia
NSC HES1
HES5
−5 0 5 vs. unso rted
SOX2
LogFC
C
Type Symbol Description Ratio q-value Type Symbol Description Ratio q-value
Ligands DKK1 dickkopf 1 5.44 4.22E-05 Transcription SOX11 11.03 5.89E-09
BMP2 4.06 5.57E-05 Factors ASCL1 MASH1 18.28 1.19E-08
WNT5A 4.72 1.11E-04 SIX4 SIX homeobox 4 4.85 4.13E-08
Receptors GFRA1 GDNF family receptor alpha 1 9.63 2.04E-08 MYT1 myelin transcription factor 1 8.91 6.34E-08
PDGFRA PDGF receptor, alpha 11.70 4.72E-08 SOX6 9.18 1.33E-07
CNR1 cannabinoid receptor 1 (brain) 12.65 4.84E-07 SOX4 3.89 4.98E-07
SEMA5A semaphorin 5A 4.38 5.34E-06 MYCN 3.82 2.54E-05
BAMBI BMP pseudoreceptor 3.81 5.40E-06 Enzymes XYLT1 xylosyltransferase I 4.69 7.22E-09
EPHA5 3.36 5.71E-06 HS6ST2 HS 6-O-sulfotransferase 2 7.48 9.50E-09
SEMA5B semaphorin 5B 4.00 8.06E-06 CASK 3.06 7.37E-08
PTPRZ1 RTPβ/ζ 4.94 2.83E-05 B3GAT2 glucuronosyltransferase S 10.44 7.84E-08
PTPRE RTPε 3.33 2.45E-04 HAS2 hyaluronan synthase 2 4.23 8.34E-07
SEMA3E semaphorin 3E 6.52 2.05E-03 B4GALT6 UDP-Gal:betaGlcNAc 9.70 9.33E-05
GPR17 G protein-coupled receptor 17 3.19 1.70E-02 ST8SIA1 responsible for A2B5 synthesis 5.52 5.47E-03
ECM TNR tenascin R (restrictin, janusin) 11.59 6.07E-09 Other FHL1 four and a half LIM domains 1 3.40 1.48E-06
CNTN1 contactin 1 7.73 9.06E-09 FRZB frizzled-related protein 3.54 1.39E-04
FBLN7 fibulin 7 5.10 2.27E-08
HAPLN1 hyaluronan and proteoglycan link 1 18.31 1.32E-07 Ratio and q-value vs. unsorted dissociate
CSPG4 chondroitin sulfate proteoglycan 4 7.16 1.95E-07
MMP16 11.39 2.19E-07
COL15A1 collagen, type XV, alpha 1 6.45 2.86E-07
OMG oligodendrocyte myelin glycoprotein 3.41 3.61E-05
PROM1 CD133/prominin 1 3.42 1.72E-02

Figure 29.2 Determination of Adult Human A2B5-Sorted Oligodendrocyte Progenitor Cell Expression Profile. Adult human OPCs were isolated on
the basis of A2B5-immunoreactivity using MACS. The Aff ymetrix U133+2 microarray profiles of A2B5+ OPCs were combined with expression
profiles generated from matched unsorted white matter and gray matter dissociates, and GLT1-sorted astrocytes and CD11b-sorted microglial cells.
A. The expression of cell type–specific marker genes illustrates the relative enrichment of OPC, microglial and astrocytic genes, in A2B5, CD11b,
and GLT sorted cells, respectively (heatmap: red, high expression; green, lower expression. Color key indicates expression levels in log base 2).
B. Differential gene expression was performed using linear modeling and an empirical Bayes test statistic with more than threefold change and 5%
FDR cutoffs. The gene expression of each cell type was compared with unsorted white and gray matter dissociates. The number of individual dif-
ferentially expressed probe sets are shown in the Venn diagram. Although there is some overlap of A2B5+ OPC and GLT1+ astrocyte profiles, the
three cell populations are clearly distinct from one another. C. Differentially expressed genes were categorized according to Gene Ontology Biological
process categories and selected genes in each functionally relevant category are shown in the table (ratio vs. unsorted cells, and FDR corrected q-value).
Modified from Sim et al. 2009. Complete data available from NCBI GEO as GSE36634 and at www.findDB.org.

364 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
OPCs were first identified from human subcortical white using earlier generation Aff ymetrix U95 arrays (Sim et al.
matter using CNP promoter-driven GFP-based fluores- 2006), and subsequently using contemporary U133+2 arrays
cence–activated cell sorting (FACS) (Nunes et al. 2003; Roy (Sim et al. 2009). Statistically rigorous analysis identified a
et al. 1999). As A2B5-expression is not present in postmitotic cohort of more than 100 shared with fetal CD140a+ cells,
neurons in the adult CNS, A2B5-based sorting was found as such as the recognized OPC marker genes CSPG4 (NG2),
a surrogate, providing greater yield and allowing immediate PDGFRA, and ST8SIA1 (Fig. 29.2A).
isolation following tissue dissociation by obviating the need A striking feature of these data was the number of genes
for transfection and GFP expression (Nunes et al. 2003). encoding cell surface receptors that were expressed along
Similarly, to CD140a-sorted fetal OPCs, A2B5-defined with their cognate ligands, such as PDGF and its receptor
OPCs were abundant comprising between 3% and 4% of PDGFRA, and PTN and its receptor PTPRZ1, suggest-
all cells in both adult subcortical white and cortical gray ing a prominent pattern of homeostatic autocrine regula-
matter (Nunes et al. 2003; Sim et al. 2009). Indeed, in the tion (Sim et al. 2006). In addition, a number of paracrine
adult mammalian brain, OPCs represent the most abundant pathways supporting both the homeostatic maintenance
progenitor cell population far exceeding numbers of neural of OPCs and their oligodendrocytic or astrocytic differen-
stem cells. Adult A2B5+ OPCs retain the capacity to gener- tiation have been identified (Fig. 29.3). Bone morphogenic
ate oligodendrocytes and are capable of extensive myelina- protein (BMP) signaling appears to mediate both autocrine
tion when transplanted into a shiverer brain (Windrem et al. and paracrine pathways by which oligodendrocytic differ-
2004). entiation is regulated; adult OPCs express high relative lev-
The regulation of OPC fate and homeostasis is under els of the BMP ligands, BMP2 and BMP7, which may limit
strict control in the intact adult brain, as OPCs give rise both expansion and oligodendrocytic fate. Indeed, addition
largely to oligodendrocytes and only rarely astrocytic of the BMP antagonist noggin, which relieves OPCs from
phenotypes (see preceding discussion and chapter 10). endogenous BMP signaling, potentiated the expansion of
However, when removed from their local environment, OPC in vitro, and prevented their astrocytic commitment
adult A2B5-defined OPCs give rise to both astrocytes (Sim et al. 2006). At the same time, OPCs expressed high
and neurons, both in vitro and following transplantation levels of the BMP antagonists, BAMBI, a dominant-nega-
into embryonic rat brain (Nunes et al. 2003). This sug- tive BMP receptor 1–like protein, and CHRDL1, a secreted
gests that the human OPC retains a neurogenic capacity BMP antagonist, which together appear to serve to protect
that is repressed by the adult parenchymal environment. the OPC from the effects of BMPs, especially from BMP4,
To define the in vivo signaling pathways that thus both regu- which serves as a potent stimulus to astrocytic differentia-
late the turnover of adult OPCs while restricting their differ- tion. Thus, BMP signaling in OPCs appears to be the prod-
entiation, A2B5+ OPCs were sorted from adult human brain uct of the tightly regulated extrinsic and intrinsic regulatory
tissue and profiled, and their gene expression patterns com- pathways, the net output of which may determine whether
pared with the tissue from which they were derived, initially parenchymal OPCs are maintained as such, or whether they

RTPβ /ζ
(a)

TN-R syndecan-3
PTN
Adult glial
PTN progenitor cell
cadherin FGF
(CDH11/18) NrCAM
FGFR3
PDGF-A
Jagged1 γ-secretase
P CASK
Y PDGF α R CHRDL1 BMP4
βCatenin
Notch1 P
Y P
βCatenin P BMP2
P BMPR2 BMP7
P
Numb Y
βCatenin BMPR1
RBP-J
TCF NRCAM, JUN,
MSI1 MYC
PTN PP
BAMBI
FHL1B CASK
? HES1
RBP-J Tbr-1 target genes
unknown

Figure 29.3 Signaling Pathways Differentially Expressed by Adult Human Oligodendrocyte Progenitor Cells. The annotation and analysis of differen-
tially expressed genes by adult human A2B5-defined OPCs allow the prediction of coherent signaling pathways, which may regulate OPCs at steady
state in the adult tissue environment. The signaling pathways predominant in this model include those initiated by PTPRZ1, as modulated by pleiotro-
phin, syndecan-3, and tenascin (purple); syndecan-3 cleavage and CASK translocation (yellow); notch activation (blue); PDGFRA signaling (green);
and BMP signaling and inhibition thereof (purple). Genes shown in color were found to be selectively and significantly enriched in A2B5-defined
OPCs, relative to unsorted white matter.

G E N E E X P R E S S I O N PAT T E R N S O F O L I G O D E N D R O C Y T E P R O G E N I TO R C E L L S A N D O L I G O D E N D R O G L I A • 365
instead restrict to astrocytic or oligodendrocytic fate, and if (Kirschenbaum et al. 1994). Lin and colleagues isolated O4+
so, to which lineage they commit. cells from neonatal rat forebrain and adult subcortical white
matter using immunopanning (Lin et al. 2009). Like their
human counterparts, adult rodent O4-defined OPCs differ-
6 O L I G O D E N D R O C Y T E P R O G E N I TO R entiated as oligodendrocytes more rapidly than did neona-
C E L L P R O F I L E S R E L AT I VE TO tal O4+ cells. Using Aff ymetrix RG_U24A microarrays, Lin
ASTROCY TES AND MICROGLIA IN THE et al. identified several hundred transcripts whose expression
A D U LT W H I T E M AT T E R differed by at least twofold in neonatal and adult O4+ cells.
Adult-derived OPCs expressed markers of mature oligoden-
In gene expression analysis from microarray data, as in any drocyte lineage cells, and included several myelin protein
relative quantitation methodology, the selection of point genes, whereas neonatally derived OPCs expressed higher
of comparison is vital and has a profound effect on the levels of PDGFRA and other earlier glial lineage transcripts.
identification of differentially regulated genes. As such, by Interestingly, pathway analysis of adult-selective genes using
comparing the profile of human OPCs to other defined cel- the Ingenuity database revealed a surprising number of cell
lular phenotypes it is possible to better define those genes death–related genes. In contrast, cell cycle–related genes
and pathways that distinguish OPCs from other cells in were relatively enriched in neonatal progenitors, consistent
the human brain (Fig. 29.2B). In addition to A2B5-sorted with their greater proliferative activity index and mitotic
OPCs, profiles of CD11b+ microglia and GLT1+ astrocytes competence. Yet although these data provided some insight
were obtained following magnetic bead–based sorting. This into the transcriptional distinctions between adult and neo-
larger database included profiles obtained from both subcor- natal progenitors, the selection of OPCs using O4-based
tical white matter and cortical gray matter–derived A2B5- sorting identifies a relatively heterogeneous pool of late
sorted OPCs and from unsorted cell dissociates for control. OPCs and committed immature oligodendrocytes. As such,
Using a linear model approach to reliably detect genes spe- additional antigenic markers will be needed to isolate more
cific to each cell population, partially overlapping gene sets homogeneous pools of stage-synchronized OPCs, whose
were identified that were selectively regulated by A2B5+ expression profiles might permit more discrete and informa-
OPCs, GLT1+ astrocytes, and CD11b+ microglia. These tive investigation of the ontogeny of oligodendroglial gene
data thus included the phenotype-selective patterns of gene expression.
expression of each major human glial subpopulation, with The analogous age-related changes in gene expression by
the exception of mature oligodendrocytes (Fig. 29.2C) (Sim human OPCs have not yet been formally assessed, as pub-
et al. 2009). lished studies to data have used different marker antigens
for isolating OPCs from fetal and adult human brain tissue.
Nonetheless, the profiles thus far reported indicate that a
7 D I S T I N C T PAT T E R N S O F G E N E large number of differentially expressed genes are shared by
E X P R E S S I O N BY F ETA L A N D human fetal and adult OPCs. The prototypical OPC marker
A D U LT O L I G O D E N D R O C Y T E genes, CSPG4 (NG2) and PDGFRA, as well as the A2B5
P R O G E N I TO R C E L L S synthetic enzyme ST8SIA1, were among genes shared by
both fetal and adult OPCs. In addition, their shared expres-
Age-dependent differences in the behavior of OPCs were sion of BMP2 and BMP pseudoreceptor BAMBI indicate
first described in rodents, in whom A2B5-isolated cells that the BMP signaling cascade is actively regulated by
from postnatal and adult animals behave differently in vitro human OPCs throughout development. In addition, the
with respect to both their proliferation kinetics and dif- fetal human neural stem cell marker CD133/PROM1 was
ferentiation rate (Wolswijk and Noble 1989). Subsequent highly expressed by fetal and adult human OPCs alike, sug-
work revealed that A2B5-defined progenitors isolated from gesting that CD133 exhibits promiscuous expression by
human fetal brain and adult subcortical white matter, and progenitors of varying stages in the human brain, and is
then transplanted into the hypomyelinated shiverer brain, not a specific marker of stem cells as previously described
similarly exhibited differences in phenotype (Windrem (Uchida et al. 2000). Additional pathway analysis of shared
et al. 2004). In humans as well as rodents, fetal OPCs are transcripts revealed the enrichment of genes involved in gly-
more highly and actively proliferative, and manifest shorter cosaminoglycan biosynthesis, cell adhesion, neurogenesis,
cell cycle times and slower maturation as myelinating oli- and nervous system development.
godendrocytes than do adult-derived OPCs, which exhibit
slower proliferative expansion but markedly more rapid
myelination. 8 C E L L -AU TO N O M O U S PAT T E R N S
The differences between fetal and adult rodent OPCs O F G E N E E X P R E S S I O N BY
have been examined using O4-based sorting. Monoclonal RODENT OLIGODENDROCYTE
O4 antibody recognizes an oligodendrocyte specific sul- P R O G E N I TO R C E L L S
fatide that is expressed by late OPCs and immature oligo-
dendrocytes in rodents (Armstrong et al. 1992; Gogate et al. Initial reports of the gene expression profiles of rodent OPCs
1994), as well as by postmitotic oligodendrocytes in humans were perforce limited by the restricted coverage of the rat

366 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
A Mouse cell isolate
Neuron Astrocyte OPC GalC MOG
Microglia Cd68
Fcgr3
Cdh5
Endothelial Tek
Vwf
Cldn11
Mal
Mbp
Mature Mobp
Oligodendrocyte Mog
Nkx6−2
Plp1
Ugt8a
Cspg4
GPC Pdgf ra
St8sia1
Cnp
Mag
Oligodendrocyte Nkx2−2
Lineage Olig1
Olig2
So x10
Aqp4
Gfap
Glul
Astrocyte S100b
Slc1a2
Tnc
Ascl1
NPC Dcx
S o x1
Hes1
Hes5
NSC Nes
S o x2
Ela vl3
Ela vl4
Nefh
Nefl
Neuron Nefm
Neurod6
−5 0 5 Snap25
LogFC Tuba1a
Tubb3

B
Type Symbol Description Ratio q-value Type Symbol Description Ratio q-value
Liga nd Dll3 delta-like 3 (Drosophila) 19.86 1.03E-13 Transcription My t1 myelin transcription fa ctor 1 21.76 2.30E-16
Nxp h 1 neurexophilin 1 13.90 4.32E-14 Factor Ascl1 MAS H1 10.05 8.58E-11
Dll1 delta-like 1 (Drosophila) 8.99 2.36E-10 Sox6 S RY-box containing gene 6 9.28 7.82E-13
Inhbb inhibin beta -B 7.57 4.43E-09 Gsx1 GS homeobox 1 7.49 4.95E-15
Bm p 7 bone morphogenetic protein 7 6.46 4.62E-09 Sox5 S RY-box containing gene 5 5.07 8.50E-08
Wn t4 5.80 3.07E-10 Enzyme Chst5 GlcNAc-6-O-S ulfotransferase 14.42 8.60E-16
Receptor Sema5 b semaphorin 5B 9.80 1.35E-05 Galnt10 N-acetylgalactosaminyltransferase 10 14.08 3.37E-14
Cxcr7 chemokine (C-X-C motif) receptor 7 8.15 1.17E-04 Ccnd1 cyclin D1 10.96 1.06E-08
Ntrk3 Neurotrophin 3 receptor 7.48 9.69E-09 Chst11 carbohydrate sulfotransferase 11 10.18 5.96E-12
Lrp 1 low density lipoprotein receptor 7.35 4.90E-10 B3gnt5 beta3Gn-T5 9.39 7.00E-04
Gfra2 GDNFfamily receptor alpha 2 6.14 7.65E-09 Car8 carbonic a nhydrase 8 8.95 2.07E-08
Oprl1 opioid receptor-like 1 6.04 9.76E-11 St8sia1 responsible for A2B5 synthesis 7.36 7.14E-09
Ptprn RTPn 5.58 1.77E-09 Chst7 carbohydrate sulfotransferase 7 6.72 2.79E-08
ECM / Pcdh15 protocadherin 15 25.20 1.07E-13 Chst15 carbohydrate sulfotransferase 15 5.71 1.55E-06
Cell Adhesion Ntn 1 netrin 1 15.84 2.13E-10 Other /Novel Mki6 7 Ki67 a ntigen 25.68 8.17E-06
Mmp2 matrix metallopeptida se 2 11.22 3.60E-09 Cspg4 chondroitin sulfate proteoglyca n 4 22.98 3.22E-13
Cntn6 contactin 6 6.27 1.47E-09 Tmem100 tra nsmembrane protein 100 13.75 2.06E-11
Vcan versican 6.05 1.55E-08 Ccnb2 cyclin B2 13.67 6.97E-07
Efs embryonal Fyn-associated substrate 6.00 9.78E-08 Cspg5 chondroitin sulfate proteoglyca n 5 9.83 4.34E-11
Chl1 close homolog of L1CAM 5.10 1.58E-05
Ratio and q-value vs. mean of other cell types

Figure 29.4 Mouse Cell Type–Specific Profiles. A. Mouse cell-type enriched populations were isolated using a combination of serial immunopan-
ning and FACS techniques (see Cahoy et al. 2008 for details). Gene expression of cell-type specific marker genes was examined across each of the
five populations and shows strong relative enrichment for OPC and oligodendrocyte genes in the relevent profiles (heatmap: red = high expression;
green = lower expression). To determine OPC specific genes, raw data were reanalyzed using the same analysis pipeline described in Sim et al. (2011)
with a linear model approach. Oligodendrocyte progenitor cell genes were defined by comparison of PDGFαR+ profiles against the profiles of GalC+
and MOG+ oligodendrocytes as well as neuronal and astrocytic defined profiles (>fivefold change, 1% FDR). B. Differentially expressed genes were
categorized according to GO Biological process categories, and selected genes in functionally relevant categories are shown (enrichment relative to
other cell types, and associated FDR corrected q-values; Pdgfra and Cspg4 not shown). Data from Cahoy et al. 2008; available from NCBI GEO as
GSE9566.

genome; these early studies were able to examine less than 10% profiles of sorted A2B5+O4 – OPCs were compared with
of known genes (Cohen et al. 2003; Hu et al. 2004; Scarlato those of A2B5–O4+-sorted oligodendrocytes from 7-day-old
et al. 2000). The whole genome expression profile of rat OPCs postnatal rat forebrain. However, because the O4+ fraction
was first studied using a combination of FACS and Aff ymetrix is heterogeneous in rodents, and contains both immature
analysis (Nielsen et al. 2006). In this study, the expression GalC– and more mature GalC+ oligodendrocytes, this study

G E N E E X P R E S S I O N PAT T E R N S O F O L I G O D E N D R O C Y T E P R O G E N I TO R C E L L S A N D O L I G O D E N D R O G L I A • 367
A B

1.0
Human
Human GPC Mouse OPC

0.8
genes genes
Mouse

sensitivity
0.4 0.6
244 92 465

0.2
targets missed by mouse-specific
mouse analysis genes AUC = 0.74

0.0
pAUC = 0.09
0.0 0.2 0.4 0.6 0.8 1.0
(1 − specificity)
C
Type S ymbol De s cription Type S ymbol De s cription
Lig a nd NXP H1 ne ure xophilin 1 Tra ns cription Fa ctor AS CL1 MAS H1
S CG2 s e cre tog ra nin II (chromog ra nin C) S OX11
CHGB chromog ra nin B (s e cre tog ra nin 1) S OX6
LINGO1 le ucine rich re pe a t ne urona l 6A MYT1 mye lin tra ns cription fa ctor 1
Re ce ptor CNR1 ca nna binoid re ce ptor 1 (bra in) Enzyme HS 6S T2 HS 6-O-s ulfotra ns fe ra s e 2
P DGFRA P DGF re c e ptor, a lpha polype ptide S T8S IA1 re s ponible for A2B5 s ynthe s is
GHR g rowth hormone re ce ptor CHS T11 ca rbohydra te s ulfotra ns fe ra s e 11
S EMA5B s e ma phorin 5B Othe r /Nove l CA10 ca rbonic a nhydra s e X
EP HA5 EphA5 FAM19A2 TAFA2 prote in
EC M /C e ll a dhe s ion DS CAM Down s yndrome CAM GAP 43 growth a s s ocia te d prote in 43
HAP LN1 ca rtila g e linking prote in 1
VCAN ve rs ica n
CS P G4 NG2
P CDH15 protoca dhe rin-re la te d 15
FLRT3 fibrone ctin le ucine rich prote in 3
NTN1 ne trin 1

Figure 29.5 Human and Mouse Comparison Reveals Conserved Oligodendrocyte Progenitor Cell–Expressed Genes. A. Venn analysis of the overlap of
human (A2B5-sorted) and mouse (PDGFαR-sorted) OPC-specific genes. Five hundred fifty-seven unique human homologs of mouse OPC-specific
genes were found in the human array data. Although 92 genes were significantly expressed by human A2B5+ OPCs, there were a large number, more
than 200 genes, whose expression was significant in human OPCs but not present in mouse cells. B. Receiver Operating Characteristic (ROC) curve
analysis was used to compare human and mouse profiles. The red line indicates the line of no discrimination, representing random gene selection.
At 20% FDR (vertical dashed line), the original human U95-derived gene list (Sim et al. 2006) (accessible via NCBI GEO:GSE26535) is greater
than 80% sensitive (green line), whereas the mouse OPC profile showed only approximately 50% sensitivity (black line). These analyses suggests that
although the mouse OPC profile broadly resembled that of human OPCs, there were substantial differences between both expression profiles and
evidence for species-specific gene expression in oligodendroglial progenitors. C. To examine the genes whose expression is highly conserved between
species, we identified genes expressed in both adult human A2B5 and mouse PDGFαR-sorted cells (overlap in Venn diagram). These genes were cat-
egorized according to GO Biological process categories and selected genes in each functionally relevant category are shown. Modified from Sim
et al. 2009.

only identified OPC and mature oligodendrocyte-selective In a similar study, serial immunopanning for O4+/Ran-2–/
transcripts. Using a small sample size that precluded rigor- GalC cells was used to select later stage OPCs from 7 day -old
ous analysis, a fold-change cutoff was used to identify dif- rat forebrain (Dugas et al. 2006). Over 90% of these O4+
ferentially expressed (DEX) transcripts. More than 1,000 OPCs coexpressed NG2+, suggesting that most were still pro-
genes were differentially expressed by the two antigenically genitors. Of note, earlier-stage O4 –/A2B5+ OPCs were not
defined stages. Consistent with the cessation of proliferation examined in this study, and were hence unavailable for com-
by postmitotic oligodendrocytes, A2B5+ OPCs differentially parison. Rather, these O4+ OPCs were compared to GalC+/
expressed several genes involved in cell proliferation, whereas Ran-2–/A2B5– oligodendrocytes isolated from day 10 to 12
genes involved in the metabolism of fatty acid and cholesterol postnatal brains. By this means, Dugas and colleagues identi-
were increased in the O4+ fraction. Indeed 17 of 18 cholesterol fied several novel oligodendrocyte-expressed transcripts, such
biosynthetic genes were upregulated in O4+ cells. This con- as GLTP, TMEM10, SEPP1, CSRP1, that serve to distinguish
trasted with adult human OPCs, which exhibit high relative later stages of oligodendrocytic lineage maturation (see Dugas
expression of cholesterol regulatory and synthetic genes such et al. 2006, for a more complete listing).
as HMG-CoA reductase (Sim et al. 2006). This difference To study mouse oligodendrocyte development, the same
may represent an important species divergence, or perhaps group subsequently used serial immunopanning to isolate three
instead reflects differences in gene expression between neona- stages of oligodendrocyte lineage from postnatal day 16 juvenile
tal and adult OPCs. mouse forebrain (Cahoy et al. 2008), a time point at which all

368 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
stages of oligodendrocyte maturation coexist in vivo. These stages 1993). Dugas et al. first isolated O4+/GalC – OPCs and char-
included PDGFαR+ OPCs, PDGFαR–/MOG+ myelinating acterize their gene expression profile during oligodendro-
oligodendrocytes, and PDGFαR–/MOG–/GalC+ cells, as a cyte differentiation in vitro (Dugas et al. 2006). O4+ OPCs
presumptive intermediate oligodendrocyte population. In the isolated from rat neonatal forebrain (day 7) were cultured
same study, Cahoy et al. (2008) prospectively isolated astrocytes in medium lacking mitogens and containing T3 for up to 9
on the basis of S100ß-driven GFP-based FACS, and used a sub- days and profiled every other day. The authors found that the
tractive approach to exclude the majority of glial populations, so transcriptional processes underlying oligodendrocyte differ-
as to enrich for neurons. Aff ymetrix microarray gene expression entiation were largely complete by 7 days in vitro, with less
profiling revealed a strong relationship between the three oligo- than 10% of genes regulated at longer time points. These data
dendrocyte lineage populations. Surprisingly, the authors found may have been confounded somewhat by the concurrence of
that on a genome-wide scale, oligodendroglial lineage cells were astrocytic generation in these cultures, since several astrocytic
more closely related to neurons than astrocytes. More than 2,000 genes including GFAP appeared in the O4+GalC– gene set,
genes were identified as differentially regulated by each major lin- yet were not apparent in acutely isolated GalC+ oligodendro-
eage. Components of several canonical signaling pathways were cytes. Yet once these astroglial genes were excluded, the gene
enriched in mouse oligodendrocytes, including MAPK, AKT, expression pattern of oligodendrocyte differentiation in vitro
JAK/STAT, IL6, Wnt, Notch, and several tyrosine kinase receptor was strikingly similar to that of acutely isolated oligodendro-
pathways including PDGF, IGF, neuregulin, and EGF signaling cytes. At least two stages of oligodendrocytes differentiation
(Fig. 29.4). Interestingly, unlike rat and human cells, fatty acid were suggested, by the distinct clustering of known myelin
metabolism was not enriched in the oligodendrocyte lineage but genes into those either rapidly induced in vitro in the first 2
rather astrocytes. to 3 days, or those later-appearing transcripts first expressed
Although their data were limited by a small sample size (N after day 5. The multistep nature of oligodendrocyte differ-
= 2), Cahoy et al. (2008) identified a number of OPC expressed entiation was also reflected in the profiles of transcription
transcripts, including markers PDGFαR and NG2/CSPG4. factor expression, with both early- and late-stage regulation
The G protein–coupled receptor GPR17 was identified as a observed. Indeed, the pattern of expression of selected TFs
mouse OPC-specific gene, and likewise was found to be highly correlated with the effect of knockdown on oligodendrocyte
expressed by both fetal and adult human OPCs. GPR17 has since differentiation, such that knockdown of the early TF SOX10
been shown to negatively regulate oligodendrocyte differentia- reduced the appearance of differentiating MBP+ oligoden-
tion and myelination (Chen et al. 2009). Surprisingly several of drocytes, whereas knockdown of the later-expressed ZFP536
these genes, including MATN4, LNX1, and ZBED4, were not blocked the development of later stage MOG+ oligodendro-
identified in either fetal (Sim et al. 2011) or adult (Sim et al. 2009) cytes (Dugas et al. 2006).
human cells. In an alternative approach to assessing differential gene
expression during oligodendroglial ontogeny, Gobert et al.
characterized the response of Oli-Neu cells, a line of trans-
9 C O M PA R AT I VE D I S T I N C T I O N S formed mouse OPCs, following treatment with drugs that
B ET W E E N M O U S E A N D H U M A N trigger oligodendrocyte differentiation (Gobert et al. 2009).
O L I G O D E N D R O C Y T E P R O G E N I TO R Using the resultant gene expression profiles to identify can-
C E L L G E N E E X P R E S S I O N PAT T E R N S didate genes capable of modulating oligodendrocyte dif-
ferentiation, several siRNAs were identified that induced
The analysis of cross-species conservation and divergence via oligodendrocyte differentiation. The murine homologs of
whole genome techniques is complicated by differences in both the early and late oligodendrocyte differentiation genes
tissue processing and cell isolation techniques. Notwithstanding identified during rat OPC differentiation segregated in an
that caveat, cross-study comparison of genes identified as dif- analogous manner during Oli-Neu oligodendrocytic differ-
ferentially expressed by A2B5+ mouse (Cahoy et al. 2008) and entiation, as induced by the EGFR tyrosine kinase inhibitor
human OPCs identified at least 92 shared genes that were over- PD174265. Interestingly, the time course of oligodendro-
expressed by the OPCs of both species (Sim et al. 2009). These cytic differentiation was accelerated in PD174265-treated
shared transcripts notwithstanding, substantial differences were Oli-Neu cells, such that with PLP1 expression became
noted in the gene expression patterns of human and mouse OPCs asymptotic within days, rather than over the course of over
(Fig. 29.5). The importance of these differences remains unclear, more than a week. As such, the distinction between early
but suggests caution in the interpretation of both pharmacologi- and late expressed transcripts was severely compressed, sug-
cal and genetic data obtained in the assessment of murine OPCs, gesting the potential flexibility of the time frame of OPC
at least as extended to their human counterparts. maturation, and by extension, the potential malleability of
the “fetal” and “adult” OPC phenotypes.
Of note, Cahoy et al. (2008) similarly assessed gene
10 O N TO G E N ET I C C H A N G E S I N expression during astrocytic ontogeny, using cells selected
GLIAL GENE EXPRESSION from an S100β-driven GFP transgenic mouse. Interestingly,
the majority of genes most strongly downregulated during
Oligodendrocyte development from purified OPCs has been postnatal astrocyte development were also downregulated
studied in vitro for more than two decades (Barres et al. 1992, during oligodendrocyte progression, from OPCs to MOG+

G E N E E X P R E S S I O N PAT T E R N S O F O L I G O D E N D R O C Y T E P R O G E N I TO R C E L L S A N D O L I G O D E N D R O G L I A • 369
oligodendrocytes (Cahoy et al. 2008). In both phenotypes, 12 miR N A R E GU L AT I O N O F
a downregulation of cell cycle genes accompanied differen- T R A N S L AT I O N I N O L I G O D E N D R O C Y T E
tiation, with the expression of several G2/M phase transition P R O G E N I TO R C E L L S
and M phase progression genes initially peaking then being
rapidly downregulated as differentiation proceeded. Thus, the MicroRNAs (miRNAs) are short, noncoding RNA molecules
maturation of both glial phenotypes appears to be associated between 19 and 21 nucleotides that regulate gene expression,
with the repression of a common set of genes involved in cell typically via control of mRNA translation. miRNAs are ini-
cycle and mitogen-activated signaling pathways. tially transcribed as pre-miRNAs which fold into stem-loop
structures before processing by Drosha and Dicer1 and incor-
poration into the RNA-induced silencing complex (RISC).
11 HI STO N E D E AC ET Y L A S E – Several groups have recently assessed the expression and func-
M E D I AT E D E P I G E N ET I C tional roles of miRNAs in rodent oligodendrocyte develop-
R E GUL AT I O N O F T R A N S L AT I O N IN ment (Budde et al. 2010; Dugas et al. 2010; Lau et al. 2008;
OLIGODENDROCYTE Zhao et al. 2010). By conditionally deleting Dicer1 in Olig1,
P R O G E N ITO R C E L L S Olig2, and CNP-driven Cre transgenics, a number of authors
found that Dicer1 deleted mice developed tremor and other
Epigenetic regulation plays a critical role in the specification myelin defects (Budde et al. 2010; Dugas et al. 2010; Zhao
of oligodendrocyte fate and differentiation (for a review see et al. 2010). In addition, CNPCre mice developed peripheral
Liu and Casaccia 2010). Histone acetylation is regulated by myelin defects that prevented the analysis of older animals
the balance of histone acetyltransferase (HATs), and histone (Budde et al. 2010; Dugas et al. 2010). In the CNS, Dicer1
deacetylases (HDACs). Histone deacetylase activity may be mutant animals exhibited a delay in normal myelination
required for oligodendrocytic fate commitment, in that phar- (Dugas et al. 2010; Zhao et al. 2010), which was resolved in
macological inhibition of HDAC activity prevents oligoden- part by late-recruited OPCs, in which recombination had not
drocyte differentiation by both rat (Marin-Husstege et al. occurred (Dugas et al. 2010). The number of PLP-expressing
2002) and mouse OPCs (Shen et al. 2008), while increasing oligodendrocytes was significantly reduced in Olig2Cre/Dicer1
the multilineage competence of these cells (Shen et al. 2008). mutants, but not in CNPCre/Dicer1 animals, suggesting that
Similarly, HDAC inhibition has been correlated with the later timing of Dicer1 deletion in the latter failed to block oli-
induction of SOX2, and with increased neurogenic capacity of godendrocytic development. Together, these data suggested
treated OPCs (Kondo and Raff 2004; Lyssiotis et al. 2007). that the generation of miRNAs in OPCs is required for their
Accordingly, HDAC activity has been confirmed as required oligodendrocytic differentiation and maturation.
for oligodendrocyte differentiation in vivo; in oligodendrocyte On that basis, miRNA profiling was used to identify those
lineage-specific (Olig1Cre) HDAC1/2 double mutants, normal specific miRNAs regulated during oligodendrocyte differen-
myelin fails to form, as oligodendroglial lineage cells fail to tiation (Dugas et al. 2010; Lau et al. 2008), as well as to define
appear. These animals exhibit the complete absence of either those specific to oligodendrocytic lineage restriction relative to
PDGFRA-expressing OPCs or mature oligodendrocytes astrocytes (Budde et al. 2010). miRNA microarray analysis of
(MBP or PLP) (Ye et al. 2009). rat postnatal day 7 A2B5- and GalC-sorted cells revealed that
Class I HDACs form complexes that repress the expression the miRNAs most significantly upregulated with differentia-
of several inhibitors of oligodendrocyte differentiation such tion were miR-223, miR-338, and miR-219 (Lau et al. 2008).
as SOX2, TCF7L2, ID4, HES5 (Lyssiotis et al. 2007; Shen Similarly, the top three candidates expressed by differentiat-
et al. 2008; Ye et al. 2009). To determine additional targets of ing mouse oligodendrocytes relative to OPCs were miR-219,
HDAC regulation, whole genome microarray was performed miR-138, and miR-338 (Dugas et al. 2010), whereas each
following treatment of rat OPCs with trichostatin A (TSA) of these were expressed at higher levels in OPCs than in
(Lyssiotis et al. 2007), a potent HDAC inhibitor ( Johnstone astrocytes (Budde et al. 2010). In that same vein, Dicer1
2002). Trichostatin treatment repressed several known OPC oligodendrocyte-specific deletion yielded the significant
and oligodendrocyte-expressed transcripts while inducing downregulation of miR-219 and miR-338 in both the optic
several known markers of neural stem cells. Interestingly, the nerve and spinal cord (Zhao et al. 2010). Together, these data
expression profiles were found to closely resemble the pro- suggested that these miRNAs were most highly expressed in
file of OPCs treated with BMP-2, which has been reported oligodendroglial lineage cells.
to trigger the reversion of OPCs to a multilineage compe- Subsequent anatomical studies revealed that miR-219 is
tent neural stem cell-like phenotype in vitro (Kondo and selectively expressed by oligodendroglial lineage cells in post-
Raff 2000). Together, these data support the restriction of natal brain, whereas miR-338 was restricted to spinal cord
oligodendroglial lineage by HDACs, while highlighting the white matter by p14 (Zhao et al. 2010). In contrast, miR-138
potential therapeutic modulation of this effect, such that was also expressed by neurons or other cell types (Dugas
HDAC inhibition might act not only to preserve the pheno- et al. 2010; Zhao et al. 2010). Transfection with miR-219 or
typic plasticity of early OPCs, but also to potentially recover miR-338 induced oligodendrocytic maturation in vitro in
the neurogenic competence of yet earlier stem and progenitor the presence of PDGF (Dugas et al. 2010; Zhao et al. 2010),
phenotypes. yet interestingly did not affect maturation in the absence

370 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
of PDGF (Dugas et al. 2010). However, transfection with therapeutic modulation of endogenous progenitor cells, and
miR-219 was able to only partially rescue the effect of Dicer1 for the effects of a variety of common pharmacological agents
deletion, suggesting that multiple miRNA pathways might on these cells. The statin drugs, such as atorvastatin, simvasta-
be concurrently required for normal differentiation. In con- tin, and pravastatin act as strong inhibitors of HMGCR, and
trast, miR-138 appeared sufficient to potentiate the transition simvastatin or pravastatin were each found to induce oligoden-
to CNP and MBP-expressing cells in the presence of PDGF. drocyte differentiation in a dose-dependent manner (Miron
Interestingly, the predicted targets of these miRNAs were et al. 2007; Sim et al. 2008). Importantly, they appeared to do
coordinately downregulated during oligodendrocyte differen- so via the potentiating PPARG signaling, and PPARG antago-
tiation (Cahoy et al. 2008). Specific inhibitors of oligoden- nism prevented stain-associated oligodendrocytic differentia-
drocyte differentiation, including both Sox6 and Hes5, were tion (Sim et al. 2008). Accordingly, direct PPARG agonists
found to be targets of both miR-219 and -338, strongly sug- have also been shown to regulate oligodendrocyte differ-
gesting the direct regulation of oligodendrocyte differentia- entiation in rodents (De Nuccio et al. 2012; Leisewitz et al.
tion by these miRNAs. 2008; Saluja et al. 2001). The differentiative actions of both
Microarray analysis comparing primary cultures of OPCs the statins and PPAR agonists are of potentially great clinical
and astrocytes identified miRNAs specific to each cell type. significance, given the common use of the statins for the treat-
In addition to miR-138, miR-338, and miR-219, the miR-17– ment of hyperlipidemia, and that of PPARG agonists such as
92 cluster was identified as highly expressed by both OPCs rosiglitazone in the treatment of type II diabetes, in which
and oligodendrocytes, relative to astrocytes (Budde et al. they are used as insulin sensitizers. Indeed, we may specu-
2010). Notably, this cluster was found to contribute to oli- late that the induction of terminal differentiation by both
godendrogenesis in vivo, as CNP-Cre mediated deletion of the statins and thiazolidinediones may contribute to some of
miR-17–92flox/flox mice resulted in a reduced number of Olig2- the toxicities noted in patients on long-term treatment with
expressing cells at birth (Budde et al. 2010). miR-17–92 over- each, in whom a variety of musculoskeletal, cardiovascular,
expression and antisense respectively increased and decreased and cognitive sequelae have been noted (Evans and Golomb
proliferation in cultures of mouse primary OPCs, by a path- 2009; Friedland et al. 2012). Significantly, both the effects of
way that appeared mediated via the downregulation of PTEN these agents upon OPCs, and at least some of their toxicity
translation and hence de-repression of Akt signaling. profiles, were predictable on the basis of their gene expression
signatures, highlighting the importance of these types of data
to drug development and experimental therapeutics, as well as
13 G E N E E X P R E S S I O N –B A S E D D RU G to our further understanding of oligodendrocyte progenitor
D I S C O VE RY ( RT P Z I N H I B I TO R S , cell biology.
S TAT I N S , P PA R S )

Adult human OPCs have been found to differentially express 14 S U M M A RY A N D P E R S P E C T I VE S


several pathways able to regulate OPC fate. One of the most
highly differentially expressed transcripts of human OPCs, Several gene expression studies of isolated glial and oligoden-
PTPRZ1, encodes the receptor tyrosine phosphatase RTPβ/ζ. drocyte progenitor cells have revealed a complex series of cell
PTPRZ1 expression was at high levels along with a number signaling cascades that appear to regulate both the homeo-
of its modulators, including chondroitin-sulfate proteogly- static maintenance and differentiation of OPCs. The relative
cans, pleiotrophin, NrCAM, and tenascin R. This pattern confluence of gene expression of human, mouse, and rodent
of concurrent gene expression by a host of PTPRZ1 signal OPCs has not yet been formally assessed, but it is clear that a
modulators suggested the importance of PTPRZ1 to the number of core conserved pathways are active in the OPCs of
regulatory control of the phosphoproteomes of human OPC. each species. That said, individual components of these path-
Accordingly, both pharmacological and shRNAi inhibition of ways may prove to be distinct in each species, and it is already
PTPRZ1 promoted the expansion of oligodendroglial-biased clear that a number of species-specific transcripts are differ-
progenitors in cultures of fetal human OPCs, while potenti- entially expressed in mouse and human OPCs. Although it is
ating oligodendrocyte differentiation by adult human OPCs too early to assess the importance of these species-dependent
(McClain et al. 2012; Sim et al. 2006). differences, we can nonetheless speculate that they may
The expression profiles of oligodendrocyte progenitor cells underlie the distinctions both the rate and extent of remy-
have predicted other attractive targets for their potential ther- elination observed in rodents and humans (Franklin and
apeutic modulation. The cells express differentially high levels ffrench-Constant 2008).
of the rate-limiting enzyme in cholesterol biosynthesis, HMG- Human OPCs have been isolated and profiled from both
CoA reductase (HMGCR) (Sim et al. 2008). In addition, they the fetal and adult brain, and their expression profiles have pro-
exhibit high differential expression of INSIG1, a regulator of vided critical insight into the molecular distinctions between
intracellular cholesterol homeostasis, indicating active sterol actively myelinating and quiescent progenitors. These differ-
response pathway activity. Accordingly, adult OPCs express ences in gene expression may prove of critical importance, in
low levels of PPARγ (PPARG), which is negatively regulated that they may predict those pathways by which quiescent pro-
in a sterol-response element–dependent fashion. These obser- genitors are prevented from oligodendrocytic differentiation
vations have potentially profound significance for both the and myelinogenesis in the context of chronic demyelination.

G E N E E X P R E S S I O N PAT T E R N S O F O L I G O D E N D R O C Y T E P R O G E N I TO R C E L L S A N D O L I G O D E N D R O G L I A • 371
In addition, the advent of genome-scale studies of both Evans MA, Golomb BA. 2009. Statin-associated adverse cognitive effects:
microRNA expression by, and epigenetic regulation of human survey results from 171 patients. Pharmacotherapy 29:800–811.
ffrench-Constant C, Raff MC. 1986. Proliferating bipotential glial pro-
OPCs should provide a systems-level understanding of these genitor cells in adult rat optic nerve. Nature 319:499–502.
processes. Together, these data should provide us sufficient Franklin RJM, ffrench-Constant C. 2008. Remyelination in the CNS:
information to both predict and modulate pivotal hub genes from biology to therapy. Nat Rev Neurosci 9:839–855.
and their regulated signaling pathways, by which the mobiliza- Friedland SN, Leong A, Filion KB, Genest J, Lega IC, Mottillo S, et al.
tion, expansion, and directed differentiation of human OPCs 2012. The cardiovascular effects of peroxisome proliferator-activated
receptor agonists. Am J Med 125:126–133.
may be regulated for therapeutic ends. Gobert RP, Joubert L, Curchod ML, Salvat C, Foucault I, Jorand-Lebrun
C, et al. 2009. Convergent functional genomics of oligodendrocyte
differentiation identifies multiple autoinhibitory signaling circuits.
AC K N OW L E D G M E N T S Mol Cell Biol 29:1538–1553.
Gogate N, Verma L, Zhou JM, Milward E, Rusten R, O’Connor M,
et al. 1994. Plasticity in the adult human oligodendrocyte lineage.
Studies in the Goldman lab described in this chapter were J Neurosci 14:4571–4587.
supported by NINDS R01NS039559 and R01NS075345, Goldman S. 2003. Glia as neural progenitor cells. Trends Neurosci
the Adelson Medical Research Foundation, the Mathers 26:590–596.
Charitable Foundation, the National Multiple Sclerosis Hu JG, Fu SL, Zhang KH, Li Y, Yin L, Lu PH, et al. 2004. Differential
Society, and the New York State Stem Cell Research Board gene expression in neural stem cells and oligodendrocyte precursor
cells: a cDNA microarray analysis. J Neurosci Res 78:637–646.
(NYSTEM). Hu QD, Ang BT, Karsak M, Hu WP, Cui XY, Duka T, et al. 2003. F3/
contactin acts as a functional ligand for Notch during oligodendro-
cyte maturation. Cell 115:163–175.
REFERENCES Johnstone RW. 2002. Histone-deacetylase inhibitors: novel drugs for
the treatment of cancer. Nat Rev Drug Discov 1:287–299.
Armstrong RC, Dorn HH, Kufta CV, Friedman E, Dubois-Dalcq ME. Kang SH, Fukaya M, Yang JK, Rothstein JD, Bergles DE. 2010. NG2+
1992. Pre-oligodendrocytes from adult human CNS. J Neurosci CNS glial progenitors remain committed to the oligodendrocyte
12:1538–1547. lineage in postnatal life and following neurodegeneration. Neuron
Barres BA, Hart IK, Coles HS, Burne JF, Voyvodic JT, Richardson WD, 68:668–681.
et al. 1992. Cell death and control of cell survival in the oligodendro- Kessaris N, Fogarty M, Iannarelli P, Grist M, Wegner M, Richardson
cyte lineage. Cell 70:31–46. WD. 2006. Competing waves of oligodendrocytes in the forebrain
Barres BA, Schmid R, Sendnter M, Raff MC. 1993. Multiple extracel- and postnatal elimination of an embryonic lineage. Nat Neurosci
lular signals are required for long-term oligodendrocyte survival. 9:173–179.
Development 118:283–295. Kirschenbaum B, Nedergaard M, Preuss A, Barami K, Fraser RA,
Belachew S, Chittajallu R, Aguirre AA, Yuan X, Kirby M, Anderson S, Goldman SA. 1994. In vitro neuronal production and differentiation
et al. 2003. Postnatal NG2 proteoglycan-expressing progenitor cells by precursor cells derived from the adult human forebrain. Cerebral
are intrinsically multipotent and generate functional neurons. J Cell Cortex 4:576–589.
Biol 161:169–186. Kondo T, Raff M. 2000. Oligodendrocyte precursor cells repro-
Budde H, Schmitt S, Fitzner D, Opitz L, Salinas-Riester G, Simons M. grammed to become multipotential CNS stem cells. Science 289:
2010. Control of oligodendroglial cell number by the miR-17–92 1754–1757.
cluster. Development 137:2127–2132. Kondo T, Raff M. 2004. Chromatin remodeling and histone modifica-
Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson tion in the conversion of oligodendrocyte precursors to neural stem
KS, et al. 2008. A transcriptome database for astrocytes, neurons, cells. Genes Dev 18:2963–2972.
and oligodendrocytes: a new resource for understanding brain devel- Lau P, Verrier JD, Nielsen JA, Johnson KR, Notterpek L, Hudson
opment and function. J Neurosci 28:264–278. LD. 2008. Identification of dynamically regulated microRNA
Cai J, Qi Y, Hu X, Tan M, Liu Z, Zhang J, et al. 2005. Generation of and mRNA networks in developing oligodendrocytes. J Neurosci
oligodendrocyte precursor cells from mouse dorsal spinal cord inde- 28:11720–11730.
pendent of Nkx6 regulation and Shh signaling. Neuron 45: 41–53. Leisewitz AV, Urrutia CR, Martinez GR, Loyola G, Bronfman M. 2008.
Chen Y, Wu H, Wang S, Koito H, Li J, Ye F, et al. 2009. The A PPARs cross-talk concertedly commits C6 glioma cells to oligo-
oligodendrocyte-specific G protein-coupled receptor GPR17 is a dendrocytes and induces enzymes involved in myelin synthesis. J Cell
cell-intrinsic timer of myelination. Nat Neurosci 12:1398–1406. Physiol 217:367–376.
Cohen RI, Rottkamp DM, Maric D, Barker JL, Hudson LD. 2003. A Levison SW, Goldman JE. 1993. Both oligodendrocytes and astrocytes
role for semaphorins and neuropilins in oligodendrocyte guidance. develop from progenitors in the subventricular zone of postnatal rat
J Neurochem 85:1262–1278. forebrain. Neuron 10:201–212.
Costa MR, Gotz M, Berninger B. 2010. What determines neurogenic Lin G, Mela A, Guilfoyle EM, Goldman JE. 2009. Neonatal and adult
competence in glia? Brain Res Rev 63:47–59. O4(+) oligodendrocyte lineage cells display different growth fac-
De Nuccio C, Bernardo A, De Simone R, Mancuso E, Magnaghi V, tor responses and different gene expression patterns. J Neurosci Res
Visentin S, et al. 2012. Peroxisome proliferator-activated receptor 87:3390–3402.
gamma agonists accelerate oligodendrocyte maturation and influence Liu J, Casaccia P. 2010. Epigenetic regulation of oligodendrocyte iden-
mitochondrial functions and oscillatory Ca(2+) waves. J Neuropathol tity. Trends Neurosci 33:193–201.
Exp Neurol 70:900–912. Lyssiotis CA, Walker J, Wu C, Kondo T, Schultz PG, Wu X. 2007.
Dugas JC, Cuellar TL, Scholze A, Ason B, Ibrahim A, Emery B, et al. Inhibition of histone deacetylase activity induces developmental
2010. Dicer1 and miR-219 Are required for normal oligodendrocyte plasticity in oligodendrocyte precursor cells. Proc Natl Acad Sci U S
differentiation and myelination. Neuron 65:597–611. A 104:14982–14987.
Dugas JC, Tai YC, Speed TP, Ngai J, Barres BA. 2006. Functional Marin-Husstege M, Muggironi M, Liu A, Casaccia-Bonnefi l P. 2002.
genomic analysis of oligodendrocyte differentiation. J Neurosci Histone deacetylase activity is necessary for oligodendrocyte lineage
26:10967–10983. progression. J Neurosci 22:10333–10345.

372 • P R O P E RT I E S O F N E U R O G L I A L C E L L S
Mason JL, Goldman JE. 2002. A2B5+ and O4+ Cycling progenitors in Shankar SL, O’Guin K, Cammer M, McMorris FA, Stitt TN, Basch RS,
the adult forebrain white matter respond differentially to PDGF-AA, et al. 2003. The growth arrest-specific gene product Gas6 promotes
FGF-2, and IGF-1. Mol Cell Neurosci 20:30–42. the survival of human oligodendrocytes via a phosphatidylinositol
McClain C, Sim F, Goldman S. 2012. Pleiotrophin suppression of 3-kinase-dependent pathway. J Neurosci 23:4208–4218.
receptor tyrosine phosphatase-β/ζ maintains the self-renewal com- Shen S, Sandoval J, Swiss VA, Li J, Dupree J, Franklin RJ, et al. 2008.
petence of fetal human oligodendrocyte progenitor cells. J Neurosci Age-dependent epigenetic control of differentiation inhibitors is crit-
14:1382–1389.Pages: ical for remyelination efficiency. Nat Neurosci 11:1024–1034.
Miron V, Rajasekharan S, Jarjour A, Zamvil S, Kennedy T, Antel J. Shi W, Fan H, Shum L, Derynck R. 2000. The tetraspanin CD9
2007. Simvastatin regulates oligodendroglial process dynamics and associates with transmembrane TGF-alpha and regulates
survival. Glia 55:130–143. TGF-alpha-induced EGF receptor activation and cell proliferation.
Nielsen JA, Maric D, Lau P, Barker JL, Hudson LD. 2006. Identification J Cell Biol 148:591–602.
of a novel oligodendrocyte cell adhesion protein using gene expres- Sim FJ, Lang JK, Ali TA, Roy NS, Vates GE, Pilcher WH, et al. 2008.
sion profi ling. J Neurosci 26:9881–9891. Statin treatment of adult human glial progenitors induces PPAR
Noble M, Proschel C, Mayer-Proschel M. 2004. Getting a GR(i)P on oli- gamma-mediated oligodendrocytic differentiation. Glia 56:954–962.
godendrocyte development. Dev Biol 265:33–52. Sim FJ, Lang JK, Waldau B, Roy NS, Schwartz TE, Pilcher WH, et al.
Nunes MC, Roy NS, Keyoung HM, Goodman RR, McKhann G 2nd, 2006. Complementary patterns of gene expression by human oli-
Jiang L, et al. 2003. Identification and isolation of multipotential godendrocyte progenitors and their environment predict determi-
neural progenitor cells from the subcortical white matter of the adult nants of progenitor maintenance and differentiation. Ann Neurol
human brain. Nat Med 9:439–447. 59:763–779.
Richardson WD, Kessaris N, Pringle N. 2006. Oligodendrocyte wars. Sim FJ, McClain CR, Schanz SJ, Protack TL, Windrem MS, Goldman
Nat Rev Neurosci 7:11–18. SA. 2011. CD140a identifies a population of highly myelinogenic,
Richardson WD, Young KM, Tripathi RB, McKenzie I. 2011. migration-competent and efficiently engrafting human oligodendro-
NG2-glia as multipotent neural stem cells: fact or fantasy? Neuron cyte progenitor cells. Nat Biotechnol 29:934–941.
70:661–673. Sim FJ, Windrem MS, Goldman SA. 2009. Fate determination of adult
Rivers LE, Young KM, Rizzi M, Jamen F, Psachoulia K, Wade A, et al. human glial progenitor cells. Neuron Glia Biol 471:1–11.
2008. PDGFRA/NG2 glia generate myelinating oligodendro- Tsiperson V, Li X, Schwartz GJ, Raine CS, Shafit-Zagardo B. 2010.
cytes and piriform projection neurons in adult mice. Nat Neurosci GAS6 enhances repair following cuprizone-induced demyelination.
11:1392–1401. PloS one 5:e15748.
Roy NS, Wang S, Harrison-Restelli C, Benraiss A, Fraser RA, Gravel M, Uchida N, Buck DW, He D, Reitsma MJ, Masek M, Phan TV, et al.
et al. 1999. Identification, isolation, and promoter-defined separation 2000. Direct isolation of human central nervous system stem cells.
of mitotic oligodendrocyte progenitor cells from the adult human Proc Natl Acad Sci U S A 97:14720–14725.
subcortical white matter. J Neurosci 19:9986–9995. Vallstedt A, Klos JM, Ericson J. 2005. Multiple dorsoventral origins of
Ruffini F, Arbour N, Blain M, Olivier A, Antel JP. 2004. Distinctive oligodendrocyte generation in the spinal cord and hindbrain. Neuron
properties of human adult brain-derived myelin progenitor cells. Am 45:55–67.
J Pathol 165:2167–2175. Windrem MS, Nunes MC, Rashbaum WK, Schwartz TH, Goodman
Saluja I, Granneman JG, Skoff RP. 2001. PPAR delta agonists stimulate RA, McKhann G 2nd, et al. 2004. Fetal and adult human oligoden-
oligodendrocyte differentiation in tissue culture. Glia 33:191–204. drocyte progenitor cell isolates myelinate the congenitally dysmyeli-
Scarlato M, Beesley J, Pleasure D. 2000. Analysis of oligodendroglial dif- nated brain. Nat Med 10:93–97.
ferentiation using cDNA arrays. J Neurosci Res 59:430–435. Wolswijk G, Noble M. 1989. Identification of an adult-specific glial pro-
Scolding N, Franklin R, Stevens S, Heldin CH, Compston A, Newcombe genitor cell. Development 105:387–400.
J. 1998. Oligodendrocyte progenitors are present in the normal Ye F, Chen Y, Hoang T, Montgomery RL, Zhao XH, Bu H, et al.
adult human CNS and in the lesions of multiple sclerosis. Brain 121 2009. HDAC1 and HDAC2 regulate oligodendrocyte differentia-
(Pt 12):2221–2228. tion by disrupting the beta-catenin-TCF interaction. Nat Neurosci
Shankar SL. 2006. Gas6/Axl signaling activates the phosphatidylinosi- 12:829–838.
tol 3-kinase/Akt1 survival pathway to protect oligodendrocytes from Zhao X, He X, Han X, Yu Y, Ye F, Chen Y, et al. 2010. MicroRNA-mediated
tumor necrosis factor-induced apoptosis. J Neurosci 26:5638–5648. control of oligodendrocyte differentiation. Neuron 65:612–626.

G E N E E X P R E S S I O N PAT T E R N S O F O L I G O D E N D R O C Y T E P R O G E N I TO R C E L L S A N D O L I G O D E N D R O G L I A • 373
This page intentionally left blank
SECTION 2
FUNCTIONS OF NEUROGLIAL CELLS
This page intentionally left blank
ASTROCY TES
This page intentionally left blank
30.
NEUROGENESIS AND OUTER SUBVENTRICULAR
ZONE RADIAL GLIAL CELLS
Xiaoqun Wang and Arnold R. Kriegstein

A B B R E VI AT I O N S exist in the ferret, a nonprimate species with a gyrencephalic


brain; the agouti, a rodent with a gyrencephalic brain; and
BLBP brain lipid-binding protein lissencephalic species such as marmoset, mouse, and rat (Fietz
bRG basal radial glia et al. 2010; Garcia-Moreno et al. 2012; Kelava et al. 2012;
CNS central nervous system Martínez-Cerdeño et al. 2012; Reillo et al. 2011; Shitamukai
GFAP glial fibrillary acidic protein et al. 2011; Wang et al. 2011). This chapter reviews recent data
GLAST glutamate transporter concerning these newly described progenitor cells, their role
INM interkinetic nuclear migration in cortical development, and how they may have contributed
IP intermediate progenitor to evolutionary changes in cortical size and architecture.
ISVZ inner subventricular zone
IZ intermediate zone
MST mitotic somal translocation 2 H I S TO R I C A L P E R S P E C T I VE O F
NES Nestin N E U R A L P R O G E N I TO R S I N T H E
oRG outer radial glial-like D E VE L O P I N G N E O C O RT E X
OSVZ outer subventricular zone
Pax6 paired box gene 6 Radial glial (RG) cells and intermediate (IP) or basal progeni-
RG radial glia tor cells are the two principal subtypes of neuronal progeni-
Sox2 SRY (sex determining region Y)-box 2 tor cells that have been extensively explored in the embryonic
SVZ Subventricular zone rodent neocortex (Haubensak et al. 2004; Malatesta et al.
Tbr2 T-box transcription factor 2000; Miyata et al. 2004; Noctor et al. 2001, 2004) (see
VIM vimentin chapter 5). Approximately at the onset of neurogenesis, neu-
VZ ventricular zone roepithelial cells transform into radial glial cells, and their
numbers swell as the progenitor region known as the ven-
tricular zone (VZ) increases in thickness. Radial glial-like
1 INTRODUCTION cells are ubiquitously present during neurogenic phases in the
central nervous system (CNS) of all vertebrates. Radial glial
The subventricular zone (SVZ) is significantly expanded in cells were long known to function as a scaffold for neuronal
the developing primate cortex, and the outer subventricular migration (Kriegstein and Alvarez-Buylla 2009; Kriegstein
zone (OSVZ) in particular, had been suggested as a major and Noctor 2004; Noctor et al. 2004; Rakic 1974, 1995)
site of cortical neurogenesis (Lukaszewicv et al. 2005; Rakic (see chapter 32), but they are now thought to represent the
1974; Smart et al. 2002). Cells within the OSVZ were found major population of neural progenitor cells in the develop-
to express markers of radial glia and intermediate progenitor ing mammalian neocortex (Malatesta et al. 2000; Miyata
cells (Bayatti et al. 2008; Mo and Zecevic 2008; Zecevic et al. et al. 2001; Noctor et al. 2001). Because they produce neu-
2005), but it was not until time-lapse studies were performed rons as well as a variety of glial cell types, they have recently
that the neurogenic potential of these cells and the lineage been referred to as neural stem cells. Time-lapse imaging
relationship between them was demonstrated (Hansen et al. studies have shown that RG cells divide extensively at the
2010). In the developing human neocortex, the outer radial apical surface of the VZ and undergo interkinetic nuclear
glial-like (oRG) cells were observed to undergo asymmetrical migration (INM) as they progress through the cell cycle
division to self-renew and generate intermediate progenitor (Chenn et al. 1998; Wang et al. 2009). The cell cycle–depen-
cells that undergo subsequent rounds of symmetrical divi- dent nuclear oscillations, whereby RG nuclei in G1 phase
sions. Therefore, the oRG cells may be termed neural progen- ascend to the upper VZ to undergo S phase and then descend
itor cells, and oRG daughter cells in human brain are better to the ventricular surface during G2 to undergo mitosis (Gotz
termed transit amplifying cells (Lui et al. 2011). It has been and Huttner 2005; Miyata et al. 2004; Noctor et al. 2001)
suggested that the OSVZ may be a primate specific feature serve to pseudostratify the VZ, and is believed to maxi-
and a hallmark of primate corticogenesis. But recent studies mize the number of mitoses that can occur at the apical sur-
have shown that OSVZ progenitor cells (i.e., oRG cells) also face (Taverna and Huttner 2010). During the peak phase of

379
neurogenesis, RG cells predominantly divide asymmetrically 1981; Schmechel and Rakic 1979), but these were thought at
to self-renew while simultaneously producing either a neuron, the time to be glial restricted astrocyte precursor cells. With the
or more commonly, an IP cell (Haubensak et al. 2004; Noctor appreciation that RG cells are neuronal precursors (Malatesta
et al. 2004). et al. 2000; Miyata et al. 2001; Noctor et al. 2001), the presence
Intermediate progenitor cells reside within the VZ and often of radial glial-like cells in the primate OSVZ was additional evi-
divide at the ventricular surface at early stages of neurogenesis dence in support of the designation of the OSVZ as a major site
(Noctor et al. 2004). However, as neurogenesis proceeds, the of neuron production (Smart et al. 2002).
IP cells migrate to a distinct proliferate layer adjacent to the The characterization of specific proteins expressed by pro-
VZ, the SVZ. Retroviral labeling and time-lapse imaging in genitor cells in the developing mouse cortex has led to the use
embryonic rodent cortical slice cultures as well as neuronally of these cell markers to examine cellular expression patterns in
expressed marker expression was used to demonstrate that IP the primate OSVZ. Cells in the human OSVZ were described
cells most often undergo one round of symmetrical division to that expressed RG markers such as nestin, vimentin, Pax6,
produce two neurons (Attardo et al. 2008; Haubensak et al. BLBP, Glast, and GFAP, as well as an alternate cell type that
2004; Noctor et al. 2004, 2008). In contrast with RG cells, expressed the IP cell maker, Tbr2 (Abdel-Mannan et al. 2008;
IP cells seem to lack apical-basal polarity (Attardo et al. 2008; Bayatti et al. 2008; Howard et al. 2006; Mo et al. 2007). These
Kriegstein and Noctor 2004; Miyata et al. 2004; Noctor et al. observations supported the conclusion that two or more pop-
2004). The two-step pattern of neurogenesis, involving RG ulations of progenitor cells may be present in the developing
cells and IP cells, appears to be the predominant pathway human OSVZ (Fig. 30.1), but how OSVZ progenitor cells
for cortical neurogenesis in rodents (Haubensak et al. 2004; contributed to neurogenesis was not understood.
Miyata et al. 2004; Noctor et al. 2004). It has been suggested Recent studies have begun to illuminate the diversity of
that the emergence of the SVZ and its constituent IP cells may progenitor cell types involved in human cortical develop-
have been responsible for the evolutionary increase in cortical ment and highlight the importance of the OSVZ as a region
thickness and layering that presumably occurred in the inter- of neurogenesis. Attention has focused on a cell type found in
val between a reptile-like mammalian ancestor and early mam- the human OSVZ that expresses RG markers including Pax6
mals (Cheung et al. 2007). and Sox2. DiI-coated beads applied to the pial surface of fixed
human cortical tissue revealed radial glia-like cells with radial
basal processes but without apical processes and therefore
3 OUTER RADIAL GLIAL CELLS IN lacking contact with the ventricle (Fietz et al. 2010; Hansen
T H E D E VE L O P I N G H U M A N F ETA L et al. 2010). Unlike ventricular RG cells, the OSVZ progeni-
N E O C O RT E X tors lack apico-basal polarity in that they do not express apical
membrane proteins prominin-1 or Par3, or ZO-1 that marks
In the evolution of the mammalian neocortex, neuron number apical adherens junctions, but they do make contact with the
reaches a peak in the human brain (Herculano-Houzel 2009; basal lamina thereby establishing them as epithelial cells (Fietz
Rakic 2009). Homo sapiens distinguish themselves from other et al. 2010; Hansen et al. 2010). Like ventricular RG they also
mammalian species by the enormous expansion in cortical sur- depend on notch activation to maintain identity, as well as inte-
face area, coupled with a high density of neurons per cortical grin signaling mediated through their basal process (Fietz et al.
volume, and an associated alteration of cortical architecture, 2010; Hansen et al. 2010). These cells have been termed oRG
that together form the substrate for the unique cognitive abili- cells (OSVZ radial-glia like cells) as well as OSVZ progenitors,
ties of humans (Kriegstein and Alvarez-Buylla 2009; Rakic intermediate radial glia, and basal radial glia (Fietz et al. 2010;
2009). However, the processes regulating cortical surface area Hansen et al. 2010; Reillo et al. 2011; Wang et al. 2011). They
expansion during development and evolution are still not very are referred to as oRG cells here. Double labeling of oRG cells
clear. One way to explore for answers is to examine the pro- in slice cultures using pulses of tagged thymidine analogs sug-
cess of neurogenesis in the developing neocortex of a variety gested that oRG cells undergo self-renewing division, but direct
of mammalian species. evidence for asymmetrical self-renewing division was obtained
The substrate for neurogenesis in the developing human through time-lapse imaging of fluorescently labeled human fetal
cortex lies within the fetal progenitor zones. Autoradiographic brain slices (Hansen et al. 2010). These studies also revealed a
analysis of 3H-thymidine labeled cells in the embryonic mon- lineage relationship between oRG cells and Tbr2 cells in the
key cortex suggested that although early generated neurons arise OSVZ; namely, that the oRG cells generate IP-like, Tbr2+, cells
from the VZ, the SVZ was a more important site for neurogen- through asymmetrical division (Hansen et al. 2010), not unlike
esis at later stages, and that the SVZ was likely the major source the way ventricular RG produce IP cells in the mouse.
of cortical neurons by the end of neurogenesis (Rakic 1975). At the time OSVZ cells were characterized in the develop-
Recently, the expanded SVZ of the monkey was examined in ing human cortex, they were also described in the developing
detail and was structurally subdivided into an inner subven- cortex of the ferret (Fietz et al. 2010; Reillo et al. 2011). The
tricular zone (ISVZ) and an outer subventricular zone (OSVZ) progenitor cells in the ferret SVZ were studied by immunohis-
separated by an inner fiber layer (Fish et al. 2008; Howard et al. tochemistry and shown to fall into two classes; one that labeled
2006; Smart et al. 2002). Glial fibrillary acid protein (GFAP) with markers of RG such as Pax6, BLBP, and GLAST and were
staining revealed that RG cells are present in the OSVZ as well morphologically similar to human oRG cells, and another class
as in the VZ during periods of peak neurogenesis (Levitt et al. that were Pax6 – but Tbr2+ (Fietz et al. 2010; Reillo et al. 2011;

380 • FUNCTIONS OF NEUROGLIAL CELLS


TexasRed- GFP p-vitementin Sox2
dextran Merge Sox2 PAX6 percentrin Dil
A B C D
Pia

OSVZ
180º

OSVZ
VZ
Ventricle

Figure 30.1 The Human Outer Subventricular Zone Is Populated with Outer Radial Glial-Like Cells. A. Five OSVZ cells in gestational week (GW)
15.5 cortex labeled by focal dye electroporation followed by immunostaining for SOX2 (blue). Cell bodies were retrogradely labeled 100 μm from the
injection site through their basal processes. A three-dimensional rendering is shown. B. A representative GFP-retrovirus–labeled cell in the OSVZ
at GW14 coexpressing PAX6 (red). Scale bar, 15 μm. C. GW17 cortex stained for SOX2 (blue), phospho vimentin (p-vimentin; green, cytoplasm of
M-phase cells), and pericentrin (red, centrosomes), demonstrating the morphology and centrosome location of oRG cells during mitosis. Scale bar, 15
μm. D. Fixed GW15 cortex labeled with DiI-coated beads (DiOlistics) applied to the pia. Inset demonstrates dye diffusion along radial processes from
the pia terminating at oRG cell bodies in the OSVZ (stars) amid coexistent radial fibers that traverse the OSVZ to the ventricle (arrowheads), and
autofluorescent blood vessels (x). Scale bars, 40 μm (d) and 20 μm (inset). Reprinted from Hansen et al. 2010.

Wang et al. 2011). These two progenitor cell types were similar the developing mouse cortex. Therefore, they were identified
to the progenitor cells found in the primate OSVZ, and a close as mouse oRG cells based on their oRG-like morphology and
examination of the architecture of the ferret SVZ suggested expression of molecular markers characteristic of RG cells. The
that OSVZ and ISVZ regions could be distinguished, in part mouse oRG cells were anatomically distinct from the bipolar
owing to a radial organization of progenitor cells in the ferret RG cells located in the VZ, and from the multipolar IP cells in
OSVZ (Fietz et al. 2010; Reillo et al. 2011). Thus, oRG cells, the VZ and SVZ (Shitamukai et al. 2011; Wang et al. 2011).
also termed intermediate radial glial cells in the ferret, and an
enlarged SVZ (OSVZ) are not primate-specific features, but
are also found in nonprimates such as the ferret. 5 M I TOT I C S O M A L T R A N S L O C AT I O N

One of the defining features of oRG cells is their mitotic behav-


4 OUTER RADIAL GLIAL CELLS IN ior, in which the cell body translocates very quickly up the
T H E D E VE L O P I N G M O U S E E M B RYO N I C basal fiber before cell division, as first described in human fetal
N E O C O RT E X brain (Hansen et al. 2010). Time-lapse imaging of mouse oRG
cells demonstrated that, like human oRG cells, the mouse cells
The finding of oRG cells in the developing human and ferret also undergo mitotic somal translocation (MST) before divi-
cortex prompted a search for similar embryonic cortical pro- sion (Wang et al. 2011). In mice, the GFP-retrovirus–labeled
genitor cells in other species. GFP-labeled oRG-like cells with cell body was observed to move rapidly along the basal pro-
a long basal but no apical process were observed in developing cess, usually migrating to a swelling within the process, and
mouse cortex when low-titer GFP-expressing retrovirus was traveling an average of around 25 μm (Fig. 30.3) (Wang et al.
injected into the lateral ventricle (Fig. 30.2) (Shitamukai et al. 2011). This is a shorter distance than that of human oRG cells,
2011; Wang et al. 2011). These cells could be triple-labeled with in which the MST averages approximately 60 μm (Hansen
the mitotic marker phosphohistone H3, and the RG markers et al. 2010). The duration of MST is less than 1 hour in mouse.
Sox2 and Pax6, and were located in the IZ and outer SVZ of Thus MST appears to be a highly specific mitotic behavior

N E U R O G E N E S I S A N D O U T E R S U BVE N T R I C U L A R Z O N E R A D I A L G L I A L C E L L S • 381
A B
Adeno-GFP GFP/ DNA

E12.5-E14.5

RG Cell

ORG Cell
IZ
IP Cell

Neuron
SVZ
VZ

Figure 30.2 Mouse Outer Radial Glial Cell Unipolor Polarity. A. Labeling of mouse RG cells and oRG-like cells with Adeno-GFP. Images of E14.5
cortices transfected with Adeno-GFP (green) at E12.5. High magnification images of representative cells (1 and 2) are shown to the right. Note that
the oRG-like cell (cell 1) located at the outer (basal) side of the SVZ has a long basal process (open arrowhead) but no apical process. Pairs of cells
were seen adjacent to each other in which only one cell has a long basal process (cell 2, white arrow), whereas the other has none (cell 2, red arrow)
suggesting that the oRG-like cell might have divided. Scale bars: 50 μm and 15 μm. B. A representative GFP-retrovirus–labeled cell counterstained
with a DNA dye (DAPI, blue). Scale bars: 25 μm. C. Schematic drawing depicting the asymmetrical division of radial glial cells and oRG cells. (A,B)
Reprinted from Wang et al. 2011.

characteristic of oRG cell types in the developing cortex of possible conserved mechanism. The spatial arrangement of the
multiple species. centrosome in oRG cells is unique compared with other cor-
Apical anchoring of the centrosome within the ventricular tical precursor cells because the centrosome anchors the ven-
endfoot is required for the maintenance of RG cells in the VZ tricular endfeet of RG cells during division, and remains in the
(Chenn et al. 1998; Wang et al. 2009). The relationship of cen- cell body of dividing IP cells (Wang et al. 2011). Interestingly,
trosome position to the dynamic mitotic behavior of oRG cells the basal motion that oRG cells make with each cell division
has been examined in mouse oRG cells. In utero electropora- likely contributes to a progressive developmental expansion of
tion was used to label the centrosome with Dsredex-centrin the OSVZ (Hansen et al. 2010).
and the cell body with pCAG-GFP (Wang et al. 2011). At
interphase, the centrosome was observed to move into a vari-
cosity within the basal process, with the nucleus then following 6 OUTER RADIAL GLIAL CELL ORIGINS
(Wang et al. 2011). In migrating neurons, a similar transloca-
tion of the centrosome into the leading process ahead of the Recent time-lapse imaging of RG cells in embryonic mouse
nucleus has been considered crucial for coordinated neuronal cortex demonstrated that 5% to 10% of divisions produced a
migration (Chenn et al. 1998; Elias et al. 2007; Higginbotham self-renewed RG cell and a daughter cell with oRG cell morphol-
and Gleeson 2007), and the similar dynamic behavior of the ogy (Wang et al. 2011). Moreover, oRG-like daughter cells of
centrosome and nucleus in MST (Wang et al. 2011) suggests a RG cell divisions have been observed to undergo characteristic

382 • FUNCTIONS OF NEUROGLIAL CELLS


A B

Translocation Distance
Retrovirus-GFP
60

40

(μm)
20

Figure 30.3 Outer Radial Glial Cells Undergo Mitotic Somal Translocation. A. Outer radial glial cells undergo mitotic somal translocation, shown
here in the mouse. A GFP-labeled oRG cell imaged at 10-m intervals beginning 48 hours after infection. Arrows indicate oRG cells (white) and a
non-oRG daughter (red). An asterisk indicates the characteristic swelling in the proximal basal process. A dashed line indicates the cleavage plane.
Scale bar: 20 μm. B. Quantification of mitotic somal translocation distances. Average distance = 23.56 ± 1.56 μm. C. Time-lapse images of cen-
trosome dynamics in an oRG cell. High magnification images from the outlined regions are shown in the following. The time is indicated at the top
(in hours and minutes). Arrows indicate centrosomes. Scale bars: 10 μm and 2.5 μm. Reprinted from Wang et al. 2011.

MST-associated cell division in the SVZ (Wang et al. 2011). oRG cells similarly undergo multiple rounds of self-renewing
These observations suggest that oRG cells can be generated division (Hansen et al. 2010). Nearly all oRG cells divide with a
through the asymmetrical divisions of RG cells. Cleavage horizontal or oblique cleavage plane such that the basal daugh-
plane orientation has been suggested to govern the fate of RG ter inherits the basal fiber and maintains oRG morphology.
cell progeny (Chenn et al. 1998; Farkas and Huttner 2008; These behaviors are consistent with asymmetrical self-renewing
Konno et al. 2008; Kosodo et al. 2004; Noctor et al. 2008). divisions of the oRG cells (Hansen et al. 2010; Lui et al. 2011).
Although most RG cell divisions are parallel to the ventricular Some oRG divisions produce daughter cells that grow their
surface, a minority of divisions occur with oblique or vertical own basal processes and appear to be oRG cells, but these are
divisions in which one daughter cell inherits the basal process infrequently observed. Most often, the apical oRG daugh-
and one the apical membrane (Fishell and Kriegstein 2003; ter, which is itself a progenitor cell, initially extends an apical
Gotz and Huttner 2005; Haubensak et al. 2004; Kosodo et al. process directed toward the ventricle as well as a shorter basal
2004). By experimentally manipulating spindle orientation, process, but retracts the processes before undergoing its own
Shitamukai et al. (2011) were able to induce large numbers of division. Long-term time-lapse imaging demonstrates that
oblique RG cell divisions, and observed that the cell inheriting oRG cells undergo successive self-renewing divisions, inherit-
the basal fiber became an oRG-like cell. It thus appears that ing the radial fiber at each division, and undergoing repeated
oRG cells arise from the asymmetrical division of RG cells, MST movements. The oRG cells thus move progressively in
and that they correspond to daughter cells that inherit the the basal direction with each cell cycle (Hansen et al. 2010).
basal process but not the apical membrane. Although the parent human oRG cells inherit the basal
process and remain SOX2+, TBR2–, the daughter cells become
TBR2+ and divide, indicating that the daughters of human oRG
7 TR ANSIT AMPLIFYING CELLS cells are progenitor cells in a neurogenic lineage. Cells with
IN THE HUMAN OUTER the bipolar morphology of oRG daughter cells were observed
S U BVE N T R I C U L A R Z O N E to undergo multiple divisions. These observations have been
interpreted as evidence of asymmetrical oRG cell divisions that
An important feature of RG cells is their ability to undergo yield a self-renewed oRG cell and a neuronally committed IP
multiple self-renewing divisions. Time-lapse imaging of fluo- cell. But unlike in the rodent, in which the IP cell daughters of
rescently labeled cells in slice cultures demonstrates that human RG cells divide only once to produce two neurons, the human

N E U R O G E N E S I S A N D O U T E R S U BVE N T R I C U L A R Z O N E R A D I A L G L I A L C E L L S • 383
oRG daughter cells undergo multiple rounds of division. For 8 A SY M M ET R I C A L I N H E R I TA N C E O F
this reason they have been referred to as transit amplifying cells THE BASAL FIBER
rather than IP cells (Hansen et al. 2011; Lui et al. 2011). As
mentioned, mouse oRG cell divisions appear to generate neu- Intermediate progenitor (IP) cells in the mouse SVZ are non-
rons directly (Shitamukai et al. 2011; Wang et al. 2011), high- polarized, and their divisions appear to be symmetrical and do
lighting an important difference in terms of overall neuron not require control of cleavage plane orientation. In contrast,
production between these species (Fig. 30.4). RG cells exhibit extreme apical-basal polarity, and divide with
Evidence from mouse indicates that oRG cells arise from a cleavage plane parallel to the apical-basal axis. When RG
RG cells, and this is likely how oRG cells arise during early cell divisions produce IP cells, the cleavage furrow is essen-
stages of outer subventricular zone (OSVZ) development tially vertical, so that the self-renewed RG cell inherits both
in animals with larger cortices, such as human and ferret. the basal fiber and the apical membrane (Noctor et al. 2008;
However, in human and ferret, oRG cells themselves can pro- Shitamukai et al. 2011). Recent time-lapse imaging experi-
liferate to expand the number of ORG cells in the OSVZ. ments in the mouse indicate that when RG cells produce oRG
For example, using GFP-expressing retrovirus to label OSVZ cells, the cleavage plane is perpendicular to the apical-basal
progenitors in the neonatal ferret led to a preponderance axis or oblique, so that the oRG cell inherits the basal fiber
of labeled oRG cells in the subsequent cell cycle, suggesting but not the apical membrane (Shitamukai et al. 2011; Wang
that a majority of divisions in the OSVZ produced two oRG et al. 2011). However, even though oRG cells lack apical mem-
cells (Reillo et al. 2011). This is not the case in human cortical brane, they maintain epithelial identity through the basal fiber
development, in which time-lapse imaging studies suggest that that makes contact with the basal lamina (Fietz et al. 2010;
only a minority of oRG cell divisions lead to two oRG cells, Hansen et al. 2010). Thus the retention of the basal process in
with one daughter inheriting the basal fiber and one growing a oRG cells may convert them from dividing symmetrically (as
new one (Hansen et al. 2010). The ability of proliferating oRG rodent IP cells) to dividing asymmetrically (as RG cells do).
cells to expand oRG number may be a feature of animals with The self-renewal mechanism may involve integrin signaling via
an expanded OSVZ. the basal process (Fietz et al. 2010), as well as notch activation

oRG
Daughter

Daughter
Daughter

B C
oRG
Daughter oRG D
Daughter
oRG D D D
Daughter IP
Daughter
IP IP
D D D D
N e
+D erg
ge

A
n
x6
FP

eu

er

D E
M
Pa

M
N
G

Figure 30.4 Outer Radial Glial Cells Self-Renew and Produce Transit Amplifying Cells in Human and Generate Neurons in Mice. A. A human oRG cell
at GW15 undergoes two self-renewing divisions followed by division of the daughter cell (intermediate progenitor [IP]). Red and white arrowheads fol-
low the lineages of oRG cell and oRG daughter, respectively. Scale bar, 10 μm. B. Intermediate progenitor cell resembling an oRG daughter undergoes two
rounds of proliferative division. As a result, they are classified as transit amplifying cells. Red and white arrowheads follow the lineages of the two IP cells.
Scale bar, 20 μm. C. Lineage relationships between cells in (A) and (B). D. Asymmetrical division of a mouse oRG cell (arrows) generates a self-renewed
oRG cell (arrows) and a daughter neuron (arrowheads). E. The oRG daughter is Pax6+ (a neuronal stem cell marker, blue), and the non-oRG daughter is
NeuN+ (a neuronal marker, red) after 12 hours more in culture. Scale bar: 10 μm. (A–C) Reprinted from Hansen DV, Lui JH, Parker PR, Kriegstein AR.
2010. Neurogenic radial glia in the outer subventricular zone of human neocortex. Nature 464(7288):554–561. (D,E) Reprinted from Wang et al. 2011.

384 • FUNCTIONS OF NEUROGLIAL CELLS


(Hansen et al. 2010). For example, functional disruption of number of neuroepithelial or RG founder cells, before the
integrin signaling in ferret oRG cells markedly decreases the onset of neurogenesis, thus increasing the number of radial
OSVZ progenitor population size (Fietz et al. 2010), whereas units while keeping the number of neurons per unit of cortical
drug-induced inhibition of notch signaling drives human oRG thickness the same (Rakic 1995). Although such a mechanism
cells to a neuronal fate (Hansen et al. 2010). Additionally, may contribute to evolutionary cortical expansion, the obser-
the centrosome, which is located in the basal process during vation that the VZ, and therefore the number of founder cells,
interphase (Wang et al. 2011), may likewise be involved in becomes very small in primates at stages when neuron dense
the maintenance of oRG cell polarity. Therefore, inheritance mid to upper cortical layers are still being generated (Hansen
of the basal fiber may be a key factor contributing to RG and et al. 2010; Smart et al. 2002) suggests that an increase in
oRG self-renewal. founder cells alone is not sufficient to account for cortical
expansion. Recent studies suggest that the development of
oRG cells and their transit amplifying daughter cells (i.e.,
9 OUTER RADIAL GLIAL CELLS AND IP-like cells) may be key cellular steps underlying expansion in
T H E D E VE L O PM E N T O F GY R E N C E P H A LY primate corticogenesis (Hansen et al. 2010). Given that pro-
genitor cells located in the SVZ may contribute to the expan-
Gyrencephalic brain development is highly correlated with sion of the overlying cortex, it has been proposed that SVZ size
overall brain and body size, but not with phylogeny, as exam- may be able to predict the future locations of gyri and sulci,
ples of lissencephalic and gyrencephalic brains can be found respectively, in the developing primate cortex (Kriegstein et al.
among most mammalian orders, including marsupials that 2006). In the developing ferret, regions of future gyral forma-
separated from Eutheria 160 million years ago (Hansen et al. tion contain more proliferating SVZ cells during development
2010). Among primates, although most have a large folded than regions of sulcus formation, and in these areas prolif-
cortical surface, very small primates such as the Senegal bush eration is more pronounced in the OSVZ than in the VZ or
baby and marmoset are lissencephalic. Similarly, although ISVZ (Reillo et al. 2011). The concept that oRG cells expand
most rodents have a lissencephalic cortex, larger rodents such in number within the OSVZ, and that OSVZ proliferation
as agouti and capybara are gyrencephalic. correlates with gyrus size has been further substantiated by
It has been hypothesized that cortical expansion may have experimental evidence in the ferret. Enucleation at embryonic
occurred through evolution because of an increase in the stages reduces OSVZ proliferation in the contralateral ferret

MZ

Astrocyte
CP
Direct Transformation
Asymmetric Division
Symmetric Division
Mature
neuron

Immature IZ
neuron
Blood
vessels TAC TAC nIPC
Immature oRG
neuron oRG
astrocyte
nIPC OSVZ
MZ oRG

NE SVZ
VZ

Neuroepithelial Radial glial cells


cells

First Trimester Second Trimester Third Trimester

Figure 30.5 The Mode of Neurogenesis During Cortical Development. Radial glia (RG) in cortex generate neurons (a) directly through asymmetrical
division. that generates a neurogenic intermediate progenitor cell (nIPC) that undergoes one round of amplification. Outer radial glial cells generate
neurons indirectly through transit amplifying cells (TACs) that go through multiple divisions before terminal symmetrical neurogenic division via
nIPCs. CP, cortical plate; IZ, intermediate zone; MZ, marginal zone; SVZ, subventricular zone; VZ, ventricular zone. Modified from Kriegstein and
Alvarez-Buylla 2009.

N E U R O G E N E S I S A N D O U T E R S U BVE N T R I C U L A R Z O N E R A D I A L G L I A L C E L L S • 385
visual cortex and leads to local reduction in the size of the neo- development, and may lay the groundwork for a better under-
cortical gyrus (Reillo et al. 2011). These findings confirm the standing of neurodevelopmental disorders ranging from major
importance of oRG cell proliferation in the tangential expan- cortical malformations to more subtle disorders such as autism
sion of the neocortex, but as described, oRG cells are not only and schizophrenia.
found in animals that have gyrencephalic cortex, but are also
found in the lissencephalic mouse.
In the marmoset, a near-lissencephalic primate, oRG cells REFERENCES
expressing Pax6 and Sox2 (but not Tbr2) and retaining a
basal process at mitosis, have been found in the OSVZ and Abdel-Mannan O, Cheung AF, Molnar Z. 2008. Evolution of cortical
occur at similar relative abundance as in human and ferret neurogenesis. Brain Res Bull 75(2–4):398–404.
Attardo A, Calegari F, Haubensak W, Wilsch-Brauninger M, Huttner
(Garcia-Moreno et al. 2012; Kelava et al. 2012). The finding WB. 2008. Live imaging at the onset of cortical neurogenesis reveals
of oRG cells in both gyrencephalic mammals such as human differential appearance of the neuronal phenotype in apical versus
and ferret, as well as non-gyrencephalic mammals such as the basal progenitor progeny. PLoS One 3(6):e2388.
mouse and marmoset, suggests that oRG cells, although per- Bayatti N, Moss JA, Sun L, Ambrose P, Ward JF, Lindsay S, et al. 2008.
haps necessary, are not sufficient for gyrencephaly. Possibly, A molecular neuroanatomical study of the developing human neo-
cortex from 8 to 17 postconceptional weeks revealing the early dif-
the presence of transit amplifying daughter cells, observed in ferentiation of the subplate and subventricular zone. Cereb Cortex
large numbers in the developing human cortex but not at all 18(7):1536–1548.
in the mouse, may be a more sensitive indicator of degree of Chenn A, Zhang YA, Chang BT, McConnell SK. 1998. Intrinsic polarity
cortical expansion. It is likely that changes in multiple param- of mammalian neuroepithelial cells. Mol Cell Neurosci 11(4):
eters, including progenitor diversity and abundance as well as 183–193.
Cheung AF, Pollen AA, Tavare A, DeProto J, Molnar Z. 2007.
cell cycle kinetics and features associated with neuronal dif- Comparative aspects of cortical neurogenesis in vertebrates. J Anat
ferentiation and circuit formation that occur later, including 211(2):164–176.
the physical tension exerted by axons (Van Essen 1997), may Elias LA, Wang DD, Kriegstein AR. 2007. Gap junction adhe-
determine whether the neocortex becomes lissencephalic or sion is necessary for radial migration in the neocortex. Nature
gyrencephalic. 448(7156):901–907.
Farkas LM, Huttner WB. 2008. The cell biology of neural stem and pro-
genitor cells and its significance for their proliferation versus differen-
tiation during mammalian brain development. Curr Opin Cell Biol
20(6):707–715.
10 S U M M A RY A N D P E R S P E C T I VE S Fietz SA, Kelava I, Vogt J, Wilsch-Brauninger M, Stenzel D, Fish JL,
et al. 2010. OSVZ progenitors of human and ferret neocortex
The finding of oRG cells and transit amplifying progenitor are epithelial-like and expand by integrin signaling. Nat Neurosci
13(6):690–699.
cells in the OSVZ of the developing cortex across a range of Fish JL, Dehay C, Kennedy H, Huttner WB. 2008. Making bigger
mammalian species is changing our concepts of neurogenesis, brains-the evolution of neural-progenitor-cell division. J Cell Sci
neuronal migration, and the model of cortical expansion in 121(Pt 17):2783–2793.
neocortical development and evolution (Fig. 30.5). Further Fishell G, Kriegstein AR. 2003. Neurons from radial glia: the consequences
exploration of the number and types of progenitor cells in the of asymmetric inheritance. Curr Opin Neurobiol 13(1):34–41.
Garcia-Moreno F, Vasistha NA, Trevia N, Bourne JA, Molnar Z. 2012.
developing cortices of additional mammalian species, along Compartmentalization of cerebral cortical germinal zones in a
with analysis of their lineages, will lead to a better understand- lissencephalic primate and gyrencephalic rodent. Cereb Cortex
ing of the features that may be unique to human cortical devel- 22(2):482–492.
opment. An increase in progenitor cell diversity is consistent Gotz M, Huttner WB. 2005. The cell biology of neurogenesis. Nat Rev
Mol Cell Biol 6(10):777–788.
with the evolutionary increase in neuronal diversity between Hansen DV, Lui JH, Parker PR, Kriegstein AR. 2010. Neurogenic radial
species, and underscores the importance of developing mark- glia in the outer subventricular zone of human neocortex. Nature
ers for specific progenitor cell types, and unraveling the tran- 464(7288):554–561.
scription programs that determine the cell fate of progenitor Haubensak W, Attardo A, Denk W, Huttner WB. 2004. Neurons
cells as well as the neurons they produce. arise in the basal neuroepithelium of the early mammalian tel-
encephalon: a major site of neurogenesis. Proc Natl Acad Sci U S A
The finding that the OSVZ contains RG-like cells with 101(9):3196–3201.
basal processes that could support neuronal migration has Herculano-Houzel S. 2009. The human brain in numbers: a linearly
potential implications for our understanding of neuronal scaled-up primate brain. Front Hum Neurosci 3:31.
migration as well as migration disorders. Although the basal Higginbotham HR, Gleeson JG. 2007. The centrosome in neuronal
fibers of oRG cells can provide additional guides for radial development. Trends Neurosci 30(6):276–283.
Howard B, Chen Y, Zecevic N. 2006. Cortical progenitor cells in the
migration, evidence suggests that clonally related neurons do developing human telencephalon. Glia 53(1):57–66.
not form ontogenetic cortical columns. Lateral dispersion of Kelava I, Reillo I, Murayama AY, Kalinka AT, Stenzel D, Tomancak
clonally related neurons has been observed in rodent, ferret, P, Matsuzaki F, Lebrand C, Sasaki E, Schwamborn JC, Okano H,
and primate cortex (Reillo and Borrell 2011; Reillo et al. 2011; Huttner WB, Borrell V. 2012. Abundant occurrence of basal radial
Walsh and Cepko 1993) and may be particularly prominent glia in the subventricular zone of embryonic neocortex of a lissen-
cephalic primate, the common marmoset Callithrix jacchus. Cereb
in large-brained species. Characterizing the migration tra- Cortex 22(2):469-81.
jectories of neurons generated from RG, IP, and oRG cells Konno D, Shioi G, Shitamukai A, Mori A, Kiyonari H, Miyata T, et al.
may lead to new insights into cortical organization as well as 2008. Neuroepithelial progenitors undergo LGN-dependent planar

386 • FUNCTIONS OF NEUROGLIAL CELLS


divisions to maintain self-renewability during mammalian neurogen- Noctor SC, Martinez-Cerdeno V, Kriegstein AR. 2008. Distinct behav-
esis. Nat Cell Biol 10(1):93–101. iors of neural stem and progenitor cells underlie cortical neurogen-
Kosodo Y, Roper K, Haubensak W, Marzesco AM, Corbeil D, esis. J Comp Neurol 508(1):28–44.
Huttner WB. 2004. Asymmetric distribution of the apical plasma Rakic P. 1974. Neurons in rhesus monkey visual cortex: systematic
membrane during neurogenic divisions of mammalian neuroepithe- relation between time of origin and eventual disposition. Science
lial cells. EMBO J 23(11):2314–2324. 183(123):425–427.
Kriegstein A, Alvarez-Buylla A. 2009. The glial nature of embryonic and Rakic P. 1975. Timing of major ontogenetic events in the visual cortex of
adult neural stem cells. Annu Rev Neurosci 32:149–184. the rhesus monkey. UCLA Forum Med Sci 18:3–40.
Kriegstein A, Noctor S, Martinez-Cerdeno V. 2006. Patterns of neural Rakic P. 1995. Radial versus tangential migration of neuronal clones
stem and progenitor cell division may underlie evolutionary cortical in the developing cerebral cortex. Proc Natl Acad Sci U S A 92(25):
expansion. Nat Rev Neurosci 7(11):883–890. 11323–11327.
Kriegstein AR, Noctor SC. 2004. Patterns of neuronal migration in the Rakic P. 2009. Evolution of the neocortex: a perspective from develop-
embryonic cortex. Trends Neurosci 27(7):392–399. mental biology. Nat Rev Neurosci 10(10):724–735.
Levitt P, Cooper ML, Rakic P. 1981. Coexistence of neuronal and glial Reillo I, Borrell V. 2011. Germinal zones in the developing cerebral cor-
precursor cells in the cerebral ventricular zone of the fetal mon- tex of ferret: ontogeny, cell cycle kinetics, and diversity of progenitors.
key: an ultrastructural immunoperoxidase analysis. J Neurosci Cereb Cortex doi: 10.1093/cercor/bhr284.
1(1):27–39. Reillo I, de Juan Romero C, Garcia-Cabezas MA, Borrell V. 2011. A role
Lukaszewicz A, Savatier P, Cortay V, Giroud P, Huissoud C, Berland M, for intermediate radial glia in the tangential expansion of the mam-
Kennedy H, Dehay C. 2005. G1 phase regulation, area-specific cell malian cerebral cortex. Cereb Cortex 21(7):1674–1694.
cycle control, and cytoarchitectonics in the primate cortex. Neuron Schmechel DE, Rakic P. 1979. Arrested proliferation of radial glial cells
47(3):353–364. during midgestation in rhesus monkey. Nature 277(5694): 303–305.
Lui JH, Hansen DV, Kriegstein AR. 2011. Development and evolution Shitamukai A, Konno D, Matsuzaki F. 2011. Oblique radial glial divi-
of the human neocortex. Cell 146(1):18–36. sions in the developing mouse neocortex induce self-renewing pro-
Malatesta P, Hartfuss E, Gotz M. 2000. Isolation of radial glial cells genitors outside the germinal zone that resemble primate outer
by fluorescent-activated cell sorting reveals a neuronal lineage. subventricular zone progenitors. J Neurosci 31(10):3683–3695.
Development 127(24):5253–5263. Smart IH, Dehay C, Giroud P, Berland M, Kennedy H. 2002. Unique
Martínez-Cerdeño V, Cunningham CL, Camacho J, Antczak JL, morphological features of the proliferative zones and postmitotic
Prakash AN, Cziep ME, et al. 2012. Comparative analysis of the compartments of the neural epithelium giving rise to striate and
subventricular zone in rat, ferret and macaque: evidence for an outer extrastriate cortex in the monkey. Cereb Cortex 12(1):37–53.
subventricular zone in rodents. PLoS One 7(1):e30178. Taverna E, Huttner WB. Neural progenitor nuclei IN motion. 2010.
Miyata T, Kawaguchi A, Okano H, Ogawa M. 2001. Asymmetric Neuron 67(6):906–914.
inheritance of radial glial fibers by cortical neurons. Neuron Tsai JW, Chen Y, Kriegstein AR, Vallee RB. 2005. LIS1 RNA interfer-
31(5):727–741. ence blocks neural stem cell division, morphogenesis, and motility at
Miyata T, Kawaguchi A, Saito K, Kawano M, Muto T, Ogawa M. 2004. multiple stages. J Cell Biol 170(6):935–945.
Asymmetric production of surface-dividing and non-surface-dividing Van Essen DC. 1997. A tension-based theory of morphogenesis and com-
cortical progenitor cells. Development 131(13):3133–3145. pact wiring in the central nervous system. Nature 385(6614): 313–318.
Mo Z, Moore AR, Filipovic R, Ogawa Y, Kazuhiro I, Antic SD, et al. Walsh C, Cepko CL. 1993. Clonal dispersion in proliferative layers of
2007. Human cortical neurons originate from radial glia and developing cerebral cortex. Nature 362(6421):632–635.
neuron-restricted progenitors. J Neurosci 27(15):4132–4145. Wang X, Tsai JW, Imai JH, Lian WN, Vallee RB, Shi SH. 2009.
Mo Z, Zecevic N. 2008. Is Pax6 critical for neurogenesis in the human Asymmetric centrosome inheritance maintains neural progenitors in
fetal brain? Cereb Cortex 18(6):1455–1465. the neocortex. Nature 461(7266):947–955.
Noctor SC, Flint AC, Weissman TA, Dammerman RS, Kriegstein AR. Wang X, Tsai JW, LaMonica B, Kriegstein AR. 2011. A new subtype
2001. Neurons derived from radial glial cells establish radial units in of progenitor cell in the mouse embryonic neocortex. Nat Neurosci
neocortex. Nature 409(6821):714–720. 14(5):555–561.
Noctor SC, Martinez-Cerdeno V, Ivic L, Kriegstein AR. 2004. Cortical Zecevic N, Chen Y, Filipovic R. 2005. Contributions of cortical sub-
neurons arise in symmetric and asymmetric division zones and ventricular zone to the development of the human cerebral cortex.
migrate through specific phases. Nat Neurosci 7(2):136–144. J Comp Neurol 491(20):109–122.

N E U R O G E N E S I S A N D O U T E R S U BVE N T R I C U L A R Z O N E R A D I A L G L I A L C E L L S • 387
31.
GLIAL CONTROL OF SYNAPTOGENESIS
Nicola J. Allen

A B B R E VI AT I O N S fundamental functional units of the nervous system (Scheiffele


2003; Waites et al. 2005; Yamagata et al. 2003). During devel-
ACM astrocyte conditioned media opment, nascent synapses form between an axonal growth cone
ADNF activity-dependent neurotrophic factor and a postsynaptic target, which is normally the dendritic region
AMPA alpha-amino-3-hydroxy-5-methyl-4-isoxa of another neuron. This initial contact induces the sequential
zolepropionic acid recruitment of specialized components to both the presynaptic
ApoE apolipoprotein E and postsynaptic contact site. For example, neurotransmitter
BG Bergmann glia vesicles and the machinery to release and replenish them accu-
C1q complement component 1, q sub- mulate in the axon. In the dendrite the postsynaptic specializa-
component tion recruits scaffolding proteins and forms the postsynaptic
C3 complement component 3 density, and recruits neurotransmitter receptors to the postsyn-
CNS central nervous system aptic membrane, thus rendering the postsynaptic cell responsive
CR3 complement receptor 3 to neurotransmitter release from the presynaptic cell.
CSPG chondroitin sulfate proteoglycan There are billions of synapses in the adult mammalian
DS Down syndrome brain, but what factors control the formation of synapses and
E embryonic day ensure that the correct synapses form in the right place at the
FX fragile X syndrome right time? Astrocyte processes are closely associated with
GABA γ-aminobutyric acid synapses throughout development and in the mature brain,
GFAP glial fibrillary acidic protein and emerging evidence has demonstrated that astrocytes
GFP green fluorescent protein play an active role in synapse formation via a combination of
HNS hypothalamo-neurohypophysial system contact-mediated and secreted signals. In addition to inducing
HSPG heparan sulfate proteoglycan new synapse formation astrocytes enhance synaptic strength,
KO knock-out stabilize synaptic structure, and modulate the elimination of
mEPSC miniature excitatory postsynaptic current preexisting synapses.
NMDA N-methyl-d-aspartate This chapter reviews the evidence for an active role of glia
NMJ neuromuscular junction in synapse formation, describes the glial-derived synaptogenic
NT-3 neurotrophin 3 factors that have been identified, demonstrates that glia are
P postnatal day also involved in synaptic stabilization and elimination, dis-
PC Purkinje cell cusses how glial synaptogenic factors interact with neuronal
PKC protein kinase C pathways, and finally, looks at how glial-induced synaptogen-
PNS peripheral nervous system esis goes awry in neurodevelopmental disorders.
RGC retinal ganglion cell
SPARC secreted protein acidic and rich in cysteine
TGF-β1 transforming growth factor beta 1 2 G L I A I N D U C E SY N A P S E F O R M AT I O N
TNFα tumor necrosis factor alpha B ET W E E N N E U R O N S
TSP thrombospondin
TTX tetrodotoxin A close spatial relationship exists between astrocytes and syn-
VIP vasoactive intestinal polypeptide apses, with 60% of synapses being associated with an astrocytic
WT wild-type process in the adult rodent hippocampus (Fig. 31.1) (Ventura
and Harris 1999; Witcher et al. 2007). Indeed, it has been esti-
mated that one astrocyte interacts with as many as 140,000
1 INTRODUCTION synapses in the mature brain (Bushong et al. 2002; Kirov et al.
1999). This close spatial relationship has led to the hypothesis
Synapses are specialized asymmetrical cell adhesions that permit that astrocytes actively contribute to synapse formation and
communication between neurons and target cells, and are the modulation of synaptic function.

388
A Rouget et al. 1993). Synapses only formed when astrocytes
were present, and interestingly the astrocytes had to be from
the same brain region—when striatal neurons were cul-
tured with mesencephalic astrocytes no synaptogenesis was
observed.
These in vivo temporal observations offer a compelling
case for an active role of astrocytes in neuronal synapse
formation, but direct investigation has been hampered by
the fact that astrocytes produce trophic factors that are
necessary for neuronal survival, thus making it difficult to
study synapse formation in the absence of astrocytes. This
has been demonstrated using in vitro cultures of rodent
hippocampus—in the absence of astrocytes embryonic
hippocampal neurons die within a few days of isolation
(Banker 1980; Kaech and Banker 2006). In vivo approaches
to delete mouse astrocytes, for example, by targeting deletion
of cells expressing the astrocyte-specific protein GFAP, caused
B C
the loss of Bergmann glia in the cerebellum, which led to
the death of cerebellar granule neurons and the atrophy of
Purkinje cells, and subsequently ataxic mice (Cui et al. 2001;
Delaney et al. 1996).

2.2 A S T RO C Y T E S I N D U C E SY NA P S E
D F O R M AT I O N B ET WE E N P U R I FI E D R ET I NA L
GANGLION CELLS
The inability to dissociate the effects of astrocytes on neuronal
survival from specific effects on synapse formation have been
overcome by the development of a neuronal culture system
using retinal ganglion cells (RGCs). Retinal ganglion cells can
be purified away from all of the other cells they are normally in
Figure 31.1 Location of Perisynaptic Astrocyte in Area CA1 of contact with in the retina by immunopanning, and maintained
Hippocampus from Mature Rat. Astroglial processes at the axon–spine in culture for weeks in defined serum-free media containing
interface (astroglial process, blue); psd, postsynaptic density (red); sp,
dendritic spine head (yellow); ax, axonal bouton (green). Reproduced peptide growth factors. (Meyer-Franke et al. 1995). This
with permission from Witcher et al. 2007. culture system enables high levels of neuronal survival along
with extensive axonal and dendritic growth, all in the absence
of astrocytes. Retinal ganglion cells cultured in isolation have
little synaptic activity, as assessed by electrophysiological
recording. By contrast, RGCs cultured in the presence of
2.1 IN VI VO C LU E S O F A RO L E F O R
astrocytes possess significantly more synaptic activity (Pfrieger
A S T RO C Y T E S I N SY NA P S E F O R M AT I O N
and Barres 1997). This increase in synaptic activity is caused by
An indication that astrocytes play a role in neuronal synapse astrocytes inducing the formation of new structural synapses in
formation comes from the temporal correlation between the addition to enhancing the efficacy of existing synapses (Ullian
timing of astrocyte generation and the timing of synapse for- et al. 2001). Physical contact between astrocytes and neurons
mation. During development neurons are generated before is not required as the same effect is seen when astrocytes are
astrocytes (Miller and Gauthier 2007), yet few synapses form placed in a feeding layer above the neurons as when grown
until after astrocytes are born (see chapter 12 for more details in contact, demonstrating that a soluble signal released from
on the timing of astrocyte generation). This has been dem- astrocytes increases the number of synapses (Fig. 31.2A).
onstrated in the superior colliculus of both the possum and Addition of astrocyte conditioned media (ACM) is equally
the rodent (Correa-Gillieron and Cavalcante 1999; Ullian effective in inducing synapses as a feeder layer of astrocytes,
et al. 2001). There is a delay between target innervation and suggesting synaptogenic factors are constitutively released
synaptogenesis, during which time astrocytes are generated, from astrocytes grown in isolation in culture, and do not
which suggests that glial derived signals are required for neu- require a neuronal signal to stimulate release.
rons to form synapses. Another line of evidence comes from Extensive characterization has demonstrated that the
studies of explant cultures. Embryonic striatal neurons were synapses induced in RGCs by astrocytes are fully functional,
cultured alone or on striatal astrocytes, and their ability to showing induction and maturation of multiple aspects
form synapses with neighboring explants of mesencephalon of synapse formation. Structural synapse number can be
examined by electron microscopy (Autillo-Touati et al. 1993; analyzed by immunostaining for presynaptic and postsynaptic

G L I A L C O N T R O L O F SY N A P TO G E N E S I S • 389
proteins, and counting the clustering and colocalization (a)
RGC alone RGC + astrocyte
of these markers as evidence of synapse formation, and
demonstrated a sevenfold increase in the number of synapses
in RGCs cultured in the presence of astrocytes (Fig. 31.2B). (b)
Synaptic strength can be determined by electrophysiological
recording from RGCs. Astrocyte exposure causes an increase
in presynaptic release probability, as well as an increase in the
frequency and amplitude of mEPSCs indicative of an increase
in postsynaptic strength (Fig. 31.2C, D). In addition, FM
dye-uptake can be used to study presynaptic vesicle cycling,
which is also enhanced by astrocytes. These results have been
validated by electron microscopy analysis of the synapses
induced between RGCs by astrocytes, which showed them
to be ultrastructurally normal, exhibiting presynaptic vesicle
clusters and an electron-dense postsynaptic density. Taken Pre
together these findings demonstrate that synapses induced to
form between neurons by glial cells possess similar structural Post
and functional aspects as synapses observed in vivo.
Merge
2.3 G L I A I N D U C E SY NA P S E S I N MU LT I P L E
N EU RO N C L A S S E S (c)

200 pA
Following this initial study on RGCs numerous groups have
demonstrated a role for glial cells in inducing synaptogenesis 10 sec

in multiple types of neurons (Allen and Barres 2005, 2009;


Eroglu and Barres 2010; Pfrieger 2010). Glutamatergic RGCs
(Nägler et al. 2001; Pfrieger and Barres 1997; Ullian et al.
2001), glutamatergic and GABAergic spinal motor neurons
(Li et al. 1999), glutamatergic and GABAergic hippocampal

400 pA
neurons (Elmariah et al. 2005b; Hughes et al. 2010; Tournell 200 msec
et al. 2006), glycinergic spinal cord neurons (Cuevas et al.
2005), cortical subplate neurons (McKellar and Shatz 2009), (d)
cortical neurons (Hu et al. 2007), and cerebellar neurons
(Buard et al. 2010; Steinmetz et al. 2006) all show enhanced
40 pA

synapse formation and function in the presence of astrocytes. 5 sec


In addition, synaptogenic effects can also be induced by other
types of glial cells—oligodendrocytes and Schwann cells can
induce neuronal synaptogenesis (Peng et al. 2003; Pfrieger
40 pA

and Barres 1997; Ullian et al. 2004), as well as support cells in


5 msec
the cochlea (Gómez-Casati et al. 2010). As well as enhancing
synapse formation and function between individual neurons,
astrocytes can enhance neuronal connectivity across networks Figure 31.2 Astrocytes Increase Synapse Number and Function in
of neurons (Boehler et al. 2007; Geissler and Faissner 2012; Retinal Ganglion Cells. The left column represents data from RGCs
Pannasch et al. 2011; Pyka et al. 2011a). cultured in isolation. The right column represents RGCs cultured in
the presence of an astrocyte feeder layer. A. Schematic representation
of the experimental setup showing RGCs in the presence and absence
2.4 G L I A L-I N D U C E D SY NA P TO G E N E S I S I S of an astrocyte feeder layer. B. Immunostaining for synaptic markers
demonstrates that few synapses are present when RGCs are grown in
C O NS E RVE D AC RO S S M A N Y S P EC I E S
isolation, and that many synapses are induced by the presence of astro-
The effects of astrocytes on synaptogenesis are not restricted cytes (red, presynaptic marker bassoon; green, postsynaptic marker
homer; colocalization, synapse). C. Electrophysiological record-
to rodents, but conserved across multiple species. Glial cells ing demonstrates that RGCs grown alone have little synaptic activity,
enhance synaptogenesis in human, rodent, Xenopus, and C. whereas RGCs grown in the presence of an astrocyte feeder layer have
elegans neurons (Bacaj et al. 2008; Colón-Ramos et al. 2007; large amounts of spontaneous synaptic activity. D. Electrophysiological
Hartley et al. 1999; Peng et al. 2003) (see chapter 2 for more recording in the presence of TTX to isolate mEPSCs demonstrates
details on invertebrate glia). In addition, studies using neurons an increase in mEPSC frequency and amplitude when RGCs are
cultured in the presence of astrocytes. Example data from Allen
derived from human embryonic stem cells and rodent neural et al. 2005.
stem cells have shown that the presence of astrocytes accel-
erates synapse formation (Hartley et al. 1999; Johnson et al.
2007; Song et al. 2002).

390 • FUNCTIONS OF NEUROGLIAL CELLS


3 M E C H A N I S M S O F G L I A L -I N D U C E D 2004). Astrocyte contact induces an increase in both synapse
SY N A P TO G E N E S I S number and function, as shown by a combination of immunos-
taining and electrophysiological recording. Interestingly, even
The preceding data demonstrate that astrocytes enhance when the astrocyte only contacts a local region of the neuron,
synapse formation and function, and there have been mul- there is enhancement of synaptogenesis throughout the whole
tiple studies aimed at identifying the molecular mechanisms cell, suggesting upregulation of a global signaling cascade. The
responsible. In this section evidence is presented that astro- mechanism of this enhancement is owing to integrin-mediated
cytes induce synapse formation via a combination of con- contact, leading to a spreading enhancement of PKC signaling
tact-mediated and secreted signals, and that different signals throughout the neuron, and subsequent synapse formation.
are used depending on the class of synapse being induced.
Interestingly, contact-mediated signals appear to be required 3.1.2 Synaptic Receptivity Caused by Neuronal
for synaptogenesis between embryonic neurons, whereas post- Neurexin Relocation
natal neurons are receptive to secreted signals, suggesting dis-
tinct signals are required for each stage of development (see In contrast to the postnatal RGCs described in the previous
Table 31.1 for a summary of factors). section, when embryonic RGCs are isolated at E17 and cul-
tured in the presence of soluble astrocyte-derived factors,
they are unable to receive synapses but are capable of forming
3.1 C O N TAC T-M E D I AT E D S Y NA P TO G E N E S I S presynaptic specializations (Barker et al. 2008). This was ele-
gantly demonstrated by mixing RGCs isolated from E17 and
3.1.1 Integrin-Mediated Protein Kinase C Signaling
P5 rats in vitro, and showing that P5 RGCs received many
Autaptic embryonic hippocampal neurons cultured in the presynaptic inputs onto their dendrites from E17 RGCs,
presence of ACM (to enhance survival) form few synapses; whereas E17 RGCs received few presynaptic inputs from P5
however, addition of an astrocyte that physically contacts the RGCs. It was hypothesized that contact with another cell
neuron induces multiple synapses to be formed (Hama et al. type may be responsible for the switch to occur, as many other

Table 31.1 SUMMARY OF GLIAL-SECRETED SIGNALS THAT REGULATE SYNAPSE FORMATION


GLIA/NEURON
MOLECULE SPECIES TYPE SYNAPSE CLASS ACTION REFERENCE

Thrombospondin Rodent Astrocyte; RGC, Glutamatergic Structural synapse formation Christopherson et al.
1 and 2 hippocampal 2005; Hughes et al. 2010;
Xu et al. 2009

Cholesterol Rodent Astrocyte; RGC Glutamatergic Presynaptic differentiation Mauch et al. 2001

Hevin Rodent Astrocyte; RGC Glutamatergic Structural synapse formation Kucukdereli et al. 2011

Glypican 4 and 6 Rodent Astrocyte; RGC Glutamatergic Enhancement of AMPA receptor synapse Allen et al. 2012
delivery and postsynaptic differentiation

SPARC Rodent Astrocyte; RGC, Glutamatergic Negative regulator of synapse formation Jones et al. 2011;
hippocampal and AMPA receptor synapse delivery Kucukdereli
et al. 2011

TNFα Rodent Astrocyte; Glutamatergic, Homeostatic synaptic scaling: increases Beattie et al. 2002;
hippocampal GABAergic synaptic AMPA and decreases synaptic Stellwagen and Malenka
GABAA receptors 2006; Stellwagen et al.
2005

CSPGs Rodent Astrocyte; Glutamatergic Stabilization of synaptic AMPA receptors Frischknecht et al. 2009;
hippocampal Pyka
et al. 2011b

Estrogen Rodent Astrocyte; Glutamatergic Enhancement of synapse formation Hu et al. 2007


cortical and function

ADNF Rodent Astrocyte; Glutamatergic Enhancement of synapse formation and Blondel et al. 2000
hippocampal function, increase in NMDA receptors

TGF-β1 Xenopus, Schwann cell; spi- Cholinergic Synapse formation and acetylcholine Feng and Ko 2008
rodent nal neurons receptor clustering
BDNF Rodent Support cells; Glutamatergic Synapse formation Gomez-Casati et al. 2010
hair cells

G L I A L C O N T R O L O F SY N A P TO G E N E S I S • 391
cell types are generated in the retina during this period of increases the number of structural synapses that form, to the
embryonic development. When the embryonic neurons were same extent as that induced by astrocytes, whereas depletion
grown in contact with astrocytes, but not amacrine cells from of TSP from ACM reduces its synapse-inducing effects
which they normally receive innervation in the retina, they (Christopherson et al. 2005). Thrombospondin-induced
acquired the ability to receive synapses. Surprisingly, it was synapses are presynaptically active but postsynaptically
shown that embryonic neurons had high levels of the presynap- silent—lacking the AMPA subtype of the ionotropic
tic transmembrane molecule neurexin in their dendrites (where glutamate receptor but containing extrasynaptic NMDA
it is not normally expressed in mature neurons), and the den- receptors. In vivo studies demonstrated that mice lacking
dritic neurexin level decreased when embryonic neurons were both TSP-1 and -2 form 30% fewer excitatory synapses in
contacted by astrocytes. Thus, one explanation for the switch in the cerebral cortex after 3 weeks of development, suggesting
synaptic receptivity is that embryonic neurons express neurexin prolonged defects in synapse formation. In addition to RGCs,
in their dendrites that inhibits synapse formation, and astrocyte TSPs induce excitatory, but not inhibitory, synapse formation
contact downregulates dendritic neurexin (through an as yet to between cultured hippocampal neurons (Hughes et al. 2010;
be determined mechanism, which is not dependent on PKC Xu et al. 2009).
signaling), therefore allowing synapses to be formed.
3.2.2 Cholesterol Enhances Presynaptic Maturation
3.1.3 Gamma-Protocadherins in Perisynaptic
Astrocyte-derived cholesterol bound to ApoE enhances
Astrocyte Processes
synapse formation and function between autaptic RGCs
Gamma-protocadherins are a family of 22 cell adhesion mol- in vitro (Mauch et al. 2001). Cholesterol is necessary for
ecules that are encoded by a single gene cluster and present in synaptogenesis and can be supplied to neurons by astrocytes
both neurons and astrocytes, and in astrocytes are localized when neurons are deficient in cholesterol, such as in clonal-
to the perisynaptic astrocyte processes that enwrap the syn- density cultures. Cholesterol enhances presynaptic function
apse (Garrett and Weiner 2009). Mice globally deficient in and neurotransmitter release by increasing quantal content,
γ-protocadherins die at birth and exhibit neuronal apoptosis and also increases dendritic out growth (Christopherson et al.
and decreased synapse density in the spinal cord (Wang et al. 2005; Goritz et al. 2005).
2002; Weiner et al. 2005), raising the question of whether
neuronal or astrocytic γ-protocadherins are responsible for 3.2.3 Hevin and Secreted Protein Acidic and Rich in
synaptogenesis. This was addressed with a combination of Cysteine Are Positive and Negative Regulators
in vitro and in vivo experiments in which cell-specific deletion of Structural Synapse Formation
of γ-protocadherins was conducted (either astrocytic or neu-
ronal), and the effect on synaptogenesis assessed in the spinal Hevin and SPARC are secreted matricellular proteins that
cord (Garrett and Weiner 2009). When either astrocytes or are expressed at high levels by astrocytes both in vitro and
neurons lack γ-protocadherins then synaptogenesis is greatly in vivo, and unlike thrombospondin remain expressed
delayed, although if just the astrocytes lack γ-protocadherins in the adult brain (Kucukdereli et al. 2011). Addition of
then the number of synapses eventually catches up to the hevin to postnatal RGCs in vitro induces the formation of
WT level with a delay. This demonstrates that astrocyte structural synapses, which like synapses induced by TSP are
γ-protocadherins are involved in neuronal synapse formation, presynaptically active and postsynaptically silent. SPARC by
presumably through a contact-mediated mechanism via the itself has no effect on synapse formation between RGCs, but
perisynaptic astrocyte processes (ACM was unable to rescue SPARC is a potent and specific inhibitor of hevin-induced
synaptogenesis). This study raises the possibility that a distinct synapse formation (SPARC does not inhibit synapse
γ-protocadherin code exists, whereby the diversity enabled by formation induced by TSP). In vivo analysis of synapse
22 potential γ-protocadherins enables astrocytes to specify formation in the superior colliculus, the target region of
when and where particular synapses form. RGC synapses, demonstrated that mice lacking hevin have
significantly fewer excitatory synapses, whereas mice lacking
SPARC have significantly more excitatory synapses. These
3.2 G L I A I N D U C E SY NA P TO G E N E S I S VI A studies identify hevin as a positive and SPARC as a negative
S EC R ET E D FAC TO R S regulator of synaptogenesis.
3.2.1 Thrombospondins Induce Structural
Synapse Formation 3.2.4 Glypicans Enhance Postsynaptic Function
and AMPA Glutamate Receptor Clustering
Thrombospondins comprise a family of high molecular weight
at Synapses
secreted matricellular proteins with five known members,
that mediate both cell–cell and cell–matrix interactions. TSP-1 and Both thrombospondin and hevin induce postsynaptically silent
-2 are expressed by astrocytes in vitro and in the developing synapses that lack AMPA receptors, suggesting that astrocytes
brain during the peak period of synaptogenesis, and their secrete additional signals that can recruit AMPA receptors
levels decrease by adulthood when synaptogenesis has largely to synapses or induce fully functional synapse formation.
finished. Addition of TSP to cultured postnatal RGCs When RGCs are cultured in the presence of astrocytes

392 • FUNCTIONS OF NEUROGLIAL CELLS


there is an increase in the strength of individual excitatory TNFα KO astrocytes were cultured with WT neurons activ-
synapses which is accompanied by a threefold increase in ity blockade no longer caused synaptic scaling, showing a glial
the surface levels of all of the four different AMPA receptor source of TNFα (see chapter 38 for additional details).
subunits (GluA1–4) and increased clustering of AMPA
receptors at synaptic sites, demonstrating that astrocytes
3.2.7 Chondroitin Sulfate Proteoglycans
regulate glutamate receptors (Allen et al. 2012). Glypicans
Modulate AMPA Glutamate Receptor
4 and 6, which are gpi-linked HSPGs, have been identified
Stability at Synapses
as astrocyte-derived proteins that are sufficient to induce
functional synapses, and act by increasing surface levels of Astrocytes and neurons both produce a variety of CSPGs,
the GluA1 AMPA receptor subunit and clustering AMPA including brevican, neurocan, phosphacan, and versican
receptors at synapses. Glypican 4 is expressed by astrocytes (Faissner et al. 2010) (see chapter 32 for additional details).
in the hippocampus during early postnatal development, and Chondroitin sulfate proteoglycans contribute to the extracel-
mice lacking glypican 4 have weaker excitatory synapses in the lular matrix and are concentrated at synapses in the mature
hippocampus in vivo, shown by a decrease in the amplitude of brain forming perineuronal net structures. Chondroitin
individual excitatory currents and a decreased recruitment of sulfate proteoglycans also accumulate in perineuronal nets
GluA1 to synapses. in an astrocyte–neuron coculture system as synaptogenesis
progresses. Enzymatic digestion of these nets increases the
number of structural synapses that form and decreases the
3.2.5 SPARC Is a Negative Regulator of AMPA amplitude of excitatory synaptic currents (Pyka et al. 2011b).
Glutamate Receptor Synaptic Delivery These results suggest that the build-up of CSPGs leads to the
Interestingly, in addition to releasing glypican, which enhances stabilization of mature synapses, thus regulating the num-
synaptic AMPA receptor levels, astrocytes also release SPARC, ber of synapses that are present. Indeed, CSPGs stabilize
which acts as a negative regulator of AMPA receptor deliv- AMPA receptors at synaptic sites by limiting the diffusion
ery to synapses ( Jones et al. 2011). SPARC is expressed by of these receptors within the membrane, suggesting a mecha-
astrocytes in the hippocampus during early postnatal devel- nism for the observed decrease in synaptic current amplitude
opment and is regulated by neuronal activity—when activ- (Frischknecht et al. 2009).
ity is enhanced in hippocampal slices, then SPARC protein
is increased in astrocytes. Mice lacking SPARC have stronger 3.2.8 Estrogen Enhances Synapse Formation
excitatory synapses in the hippocampus during development, and Function
with an increase in the frequency and amplitude of mEPSCs.
Studies on cultured neurons in the presence of wild-type Cortical astrocytes enhance synapse formation and transmis-
(WT) or SPARC knock-out (KO) astrocytes demonstrated sion between cortical neurons in vitro which can be attenu-
that there is an increase in neuronal surface AMPA recep- ated by treatment with tamoxifen, an antagonist of estrogen
tor levels in the absence of SPARC, and that this difference receptors (Hu et al. 2007). Astrocytes do not normally express
can be rescued by the addition of SPARC protein to the KO aromatase enzymes in vivo, which are necessary to synthesize
astrocytes. This suggests that astrocytes sense and respond to estrogen, but these enzymes are induced in reactive astrocytes
neuronal activity levels by altering the expression of SPARC, following injury. This suggests that estrogen secretion from
with high levels of activity increasing SPARC which may act astrocytes may be induced in injury, and could contribute to
back on neurons to reduce synaptic AMPA receptor levels and the recovery from damage.
hence help maintain synaptic balance.
3.2.9 Activity-Dependent Neurotrophic Factor
3.2.6 Tumor Necrosis Factor Alpha Regulates AMPA (ADNF) and N-Methyl-d-aspartate
and GABAA Receptor Levels and Homeostatic Receptors
Synaptic Scaling
Astrocyte-neuron cocultures isolated from E16 rat hippo-
In addition to regulating synaptic strength during develop- campus have few synaptic connections, whereas those isolated
ment, astrocytes have been shown to release factors that reg- from E18 rats have numerous synaptic connections (Blondel
ulate the recruitment of AMPA and GABAA receptors to et al. 2000). Vasoactive intestinal peptide is released from
synapses in the mature brain. Glia release the cytokine TNFα, embryonic hippocampal neurons when isolated at E18, but
which modulates synaptic strength by increasing surface not E16, and its addition to E16 cultures is able to enhance
AMPA receptors and decreasing surface GABAA receptors, synaptic connectivity. Vasoactive intestinal peptide acts on
thus leading to an overall strengthening of synaptic transmis- astrocytes to cause the release of ADNF, which acts back on
sion—known as homeostatic synaptic scaling (Beattie et al. neurons to stimulate release of neurotrophic factors such as
2002; Stellwagen et al. 2005; Stellwagen and Malenka 2006). NT3. The eventual outcome following 2 weeks of culture is an
Addition of TNFα to cultured hippocampal neurons causes a increase in the number of presynaptic terminals, an increase
rapid increase (within 15 minutes) in surface GluA1 AMPA in synaptic event frequency and amplitude, and an increase
receptor levels, whereas blockade of the actions of endogenous in NMDA receptor subunits (NR2A and NR2B) within
TNFα lead to a decrease in surface AMPA receptors. When neurons.

G L I A L C O N T R O L O F SY N A P TO G E N E S I S • 393
3.2.10 Perisynaptic Schwann cells Enhance Synapse synapses. Whether a synapse matures, is stabilized and main-
Formation at the Neuromuscular Junction via tained, or even eliminated can be influenced by astrocytes.
Transforming Growth Factor-β1
Perisynaptic Schwann cells are specialized Schwann cells at the 4.1 A S T RO C Y T E –SY NA P S E P H YS I C A L
NMJ synapse, which enhance synapse formation and function I N T E R AC T I O NS
between cultured spinal neurons and target muscle in Xenopus
and rodent, and stabilize synaptic connections in vivo (Cao and Physical contact between astrocytes and neurons can have
Ko 2007; Feng et al. 2005; Peng et al. 2003; Reddy et al. 2003). multiple outcomes; for example, stabilization of synaptic
Schwann cells act to enhance synapse formation and clustering contacts, elimination of synapses, or even preventing syn-
of acetylcholine receptors in neuron–muscle cocultures (Peng apses from forming by blocking potential sites. Each of these
et al. 2003). Interestingly, it appears that Schwann cells induce outcomes has been observed, suggesting that the outcome of
synapse formation by overcoming the inhibitory effects of neu- astrocyte–neuron contact is context dependent; for example,
rotrophic factors that are necessary for neuronal survival. High based on developmental stage or neuronal activity.
levels of neuronal death are observed in the absence of neu-
rotrophic factors and Schwann cells, but the neurons that sur-
4.1.1 Astrocyte–Dendritic Spine Physical
vive make synapses. The addition of neurotrophic factors to the
Interactions Stabilize Synapses
cultures greatly enhances neuronal survival, but few synapses
form, suggesting synaptogenesis has been inhibited. The addi- Astrocyte–dendritic spine contacts have been observed to be
tion of Schwann cells to the coculture, in the continuous pres- more stable at large mature spines compared to weak filopodia,
ence of neurotrophic factors, enables synapses to form between leading to the hypothesis that astrocyte–spine interactions are
neurons and muscle cells. The molecular basis for this has been involved in stabilizing synaptic connections (Lippman and
determined—neurotrophic factors inhibit the production of Dunaevsky 2005; Lippman et al. 2008; Witcher et al. 2007).
agrin in neurons, and Schwann cells overcome this inhibition Indeed, when the actin cytoskeleton is disrupted, which dis-
and reinduce the expression of agrin. Agrin is essential for the rupts astrocyte process protrusion, there is an increase in den-
stabilization of acetylcholine receptor clusters at NMJs and dritic filopodia, a decrease in mature spines, and more synapses
hence for NMJ function and stability. Transforming growth are observed (Nishida and Okabe 2007).
factor-β1 has been identified as the factor secreted from
Schwann cells that is sufficient and necessary to upregulate
agrin levels in presynaptic neurons (Feng and Ko 2008). 4.1.2 Astrocyte Coverage Limits
Synapse Formation
3.2.11 Support Cell–Induced Synaptogenesis in In the cerebellum, climbing fibers release ectopic vesicles
the Vestibular Organ Mediated by Brain-Derived of glutamate directly onto BG (Matsui and Jahr 2003),
Neurotrophic Factor and BG sense this via calcium permeable AMPA receptors,
which lack the GluA2 subunit. Expression of GluA2 in BG
In the vestibular system, supporting cells have many of the (making AMPA receptors calcium impermeable) causes the
characteristics of astrocytes and Schwann cells; therefore, retraction of glial processes from the PC with which they
their effect on synaptogenesis between hair cells and vestibu- are normally in close association (Iino et al. 2001), lead-
lar neurons in the inner ear was investigated (Gómez-Casati ing to aberrant innervation of the PC by multiple climb-
et al. 2010). A series of in vivo experiments using a variety of ing fibers instead of the single climbing fiber that innervates
transgenic and KO mice demonstrated that neurons signal each PC in the mature cerebellum. This shows that release
to support cells via the tyrosine kinase receptor erbB, leading of glutamate directly onto the BG signals the cell to remain
to the expression of BDNF in support cells that feeds back in close association with the PC it is surrounding, thus lim-
onto neurons to induce synapse formation between hair cells iting the number of synapses that can form onto the PC. In
and vestibular neurons. Blockade of erbB signaling in sup- the HNS astrocytes respond to hormonal signals by fully
port cells, or loss of BDNF specifically from support cells, retracting from synapses in a reversible manner (Theodosis
led to severe defects in synapse number at P21. Interestingly, et al. 2008). Neurons with retracted astrocytes receive more
the defect in synapse number was less severe at early postnatal synaptic inputs suggesting astrocytes were previously block-
ages, and total synapse number actually decreased with age in ing these areas and stopping neuronal innervation (see chap-
mutant mice, suggesting a failure of synapse maintenance. ter 41 for more details).

4 G L I A I N F LU E N C E SY N A P T I C
4.1.3 Physical Coupling Between Astrocyte Processes
S T RU C T U R E , E L I M I N AT I O N,
and Dendritic Spines Via EphrinA3–EphA4
A N D S TA B I L I T Y
Interactions
In addition to inducing initial synapse formation, astrocytes Dendritic spines express the receptor tyrosine kinase EphA4,
are actively involved in determining the fate of newly formed which interacts with its ligand ephrin-A3 on neighboring

394 • FUNCTIONS OF NEUROGLIAL CELLS


Figure 31.3 Example of Physical Coupling Between an Astrocyte Process and a Synapse via Adhesion Molecules Triple labeling shows ephrin-A3 on
astrocytic processes (red) encompassing a synaptophysin-positive presynaptic terminal (blue) and YFP (green) to label the dendritic spine of a CA1
pyramidal neuron where EphA4 is localized. Scale bar, 0.5 μm. Reproduced with permission from Mura et al. 2003.

astrocytic processes (Fig. 31.3) (Klein 2009; Murai and of signaling plays a role in the localization of synapses by
Pasquale 2011; Murai et al. 2003). EphA4 levels in the determining whether a spine is eliminated or stabilized.
hippocampus are high during early postnatal development
when synapses are actively forming, and it is present in 4.2 G L I A I N FLU E N C E SY NA P S E
an inactive nonphosphorylated form in the adult brain. E L I M I NAT I O N
Reactivation of EphA4 in the adult hippocampus collapsed
20% of dendritic spines and decreased the length of other In the developing brain excess synapses are formed, and
spines, causing spine retraction and an overall reduction in elimination processes remove these synapses to generate the
spine density. On the other hand, when EphA4 was inhibited final adult number. Possible mechanisms of synapse elimina-
this caused disorganization of spines and an overall increase tion include retraction of presynaptic terminals and axons,
in spine length. These results suggest that during development degeneration of axons and subsequent phagocytosis of the
(when EphA4 is active), contact between astrocytic processes debris by surrounding cells e.g. glial cells, or active pruning
and dendritic spines may be repulsive, and that the level of excess synapses by neighbouring glia. There is evidence

Astrocyte factors Immature Mature Mature synaptic unit

Presynaptic maturation Presynaptic terminal


Cholesterol
Thrombospondin 1,2
Hevin
Glypican 4,6

Structural synaptogenesis
Thrombospondin 1,2
Hevin
Glypican 4,6
Astrocyte process

Postsynaptic maturation
Glypican 4,6 Key
Neurotransmitter vesicle
Sparc
Neurotransmitter receptor
Tumour necrosis factor alpha
Postsynaptic density
CSPGs
Postsynaptic terminal

Figure 31.4 Schematic Representation of Astrocyte Regulation of Distinct Aspects of Synapse Formation During Development, via the Secretion of
Multiple Factors. Presynaptic maturation is defined as increased neurotransmitter vesicle recruitment to presynaptic sites and increased neurotransmit-
ter release probability. Structural synaptogenesis is defined as clustering of presynaptic and postsynaptic density proteins and the alignment of presyn-
aptic and postsynaptic sites. Postsynaptic maturation is defined as insertion of postsynaptic receptors into the postsynaptic density membrane.

G L I A L C O N T R O L O F SY N A P TO G E N E S I S • 395
from studies of Drosophila that glia do indeed phagocytose side and pushing off the axon. Recent studies have shown
axonal debris. In rodents, astrocytes induce synapse elimina- an essential role for microglia in synapse elimination in the
tion pathways in neurons, and microglia phagocytose syn- healthy developing CNS (Paolicelli et al. 2011; Schafer et al.
apses, demonstrating that glia are actively involved in synapse 2012). Microglia phagocytose both presynaptic and postsyn-
elimination. aptic elements, as revealed by immunoelectron microscopy
and high resolution confocal imaging, both of which show
4.2.1 Drosophila Glia Engulf Axonal Debris synaptic elements completely within microglia and in the
During Development phagocytic degradation pathway. Notably, when the ability
of microglia to phagocytose is inhibited by genetic removal
In the developing Drosophila nervous system, glial cells of the microglia-specific phagocytic pathway CR3/C3, then
invade regions of axonal degeneration and engulf axonal more synapses are present in the adult brain, demonstrating
varicosities by phagocytosis (Awasaki and Ito 2004; (Walts that microglia are actively involved in synapse elimination in
et al. 2004). Blocking glial phagocytosis significantly per- development (Schafer et al. 2012) (see chapters 15, 47, and 53
turbs axon pruning. The glial phagocytosis receptor Draper for more details).
is essential for this process—when Draper is absent from
glial cells (and muscle) the removal of axonal debris is greatly
reduced, causing defects in synapse growth (Fuentes-Medel
et al. 2009; MacDonald et al. 2006) (for more information 4 .3 A R O L E F O R G L I A I N SY N A P S E
see chapter 2). M AINTENANCE

The preceding data demonstrate that glial signals power-


4.2.2 Axosome Shedding at the fully induce synapse formation and remodeling, but do glia
Neuromuscular Junction act as a switch to enable neurons to form synapses, or are
As axons are removed at the developing NMJ they produce glial signals constantly required so as to maintain synapses?
axosomes, which are small membrane bound particles, and Initial studies suggest that glia are indeed necessary for syn-
time-lapse imaging and EM have shown that these axosomes apse maintenance.
can be seen within neighbouring Schwann cells. (Bishop
et al. 2004). Thus, a similar process of removing axonal
debris from the developing nervous system exists in both 4.3.1 Constant Astrocyte Signals Are Required
Drosophila and mammals. for Synapse Maintenance In Vitro
Retinal ganglion cells in vitro require a continuous signal
4.2.3 C1q Induction in Neurons by Astrocytes from astrocytes to maintain the astrocyte-induced increase
in synapse formation and function. When synapses are
The immune system molecule C1q is induced in RGCs when induced by astrocytes, and the astrocytes removed, the num-
they are cultured in the presence of astrocytes (Stevens et al. ber of synapses decreases over a period of days to a similar
2007). C1q acts as an opsonin in the immune system, mark- level as that observed in RGCs that have never been exposed
ing damaged cells and debris for removal by phagocytosis to astrocytes (Ullian et al. 2001). The identity of the soluble
or lysis by activation of the complement cascade. Mice defi- maintenance signal is unknown, and it is not clear whether
cient in C1q have more functional synapses in vivo, suggest- it is the same signal that induces synapse formation (e.g.,
ing that C1q has a similar role in the developing brain and thrombospondin) or a novel signal.
plays an active role in neuronal synapse elimination. The
excess of synapses in C1q deficient mice has functional con-
sequences, as these mice are spontaneously epileptic (Chu 4.3.2 Perisynaptic Schwann Cells Maintain
et al. 2010). Neuromuscular Junction Synapses In Vivo
The effect of removing synaptic glia from a mature synaptic
4.2.4 Synapse Elimination and Synaptic
contact in vivo has been investigated at the Xenopus NMJ
Stripping by Microglia
(Reddy et al. 2003). Monoclonal antibodies were used to
A role for microglia in synapse elimination has been proposed label perisynaptic Schwann cells in an intact adult frog
in the mammalian CNS, both during development and in NMJ and the cells were lysed via the complement cascade,
injury situations. The process of synaptic stripping, whereby selectively killing Schwann cells and leaving the presynap-
microglia displace axonal inputs from damaged motoneu- tic motor neuron terminal and the postsynaptic muscle cell
rons, was first described in 1968 (Blinzinger and Kreutzberg intact. Loss of perisynaptic Schwann cells had no immedi-
1968; Cullheim and Thams 2007). Synaptic inputs onto ate effect on the structure or function of the synapse, but 1
damaged neurons are eliminated, and it is hypothesized that week after ablation presynaptic function decreased by half,
microglia actively denervate the damaged postsynaptic cell and there was a tenfold increase in the retraction of presyn-
by intervening between the presynaptic and postsynaptic aptic terminals from the muscle.

396 • FUNCTIONS OF NEUROGLIAL CELLS


5 I N T E R AC T I O N S B ET W E E N 5.2 D OWNS T R E A M S I G NA L I N G C A S C A D E S
A S T R O C Y T E FAC TO R S A N D N E U R O N A L I N D U C E D I N N EU RO NS BY A S T RO C Y T E
SY N A P TO G E N I C PAT H WAYS SY NA P TO G E N I C S I G NA L S
5.2.1 Gene Transcription
There has been much work on identifying the astrocyte-derived
signals that induce synaptogenesis, but less is known about how The question of whether astrocytes induce synapse forma-
these signals interact with neurons to induce synaptogenesis. tion via global effects on gene transcription was addressed
Neuronal receptors for astrocyte factors are beginning to be iden- using microarrays to identify genes altered in RGCs following
tified, with α2δ1 identified as the TSP receptor and TrkB shown 30 hours of exposure to ACM (Göritz et al. 2007). Surprisingly
to be involved in GABAergic synapse formation. The question very few genes were changed, with 28 genes being upregulated
of whether astrocyte synaptogenic signals act locally to induce and 46 genes downregulated, and the majority of the genes
individual synapses or globally to enhance the synaptogenic were not known to be associated with synaptogenic pathways.
state of the whole neuron (e.g., via effects on gene transcription) This suggests that regulation of gene expression does not play
is still unanswered. Although physical astrocyte contact can a major role in astrocyte-induced synaptogenesis, although it
enhance global synaptogenesis in embryonic neurons (Hama would be interesting to know whether more genes would be
et al. 2004), it is not known whether this is the case for secreted regulated following a longer exposure to astrocytes, or if the
signals. Neuronal activity plays an important role in synaptic neurons and astrocytes were grown in contact.
remodeling in the developing brain, and studies are beginning
to show that activity is able to regulate the release of some astro-
5.2.2 Neuronal Activity
cyte synaptogenic factors. Whether all factors are modulated by
activity or are constitutively released or developmentally con- Neuronal activity is involved in synaptic remodeling in the
trolled will be an important point for further study. developing nervous system, but is it required for neurons to
respond to astrocyte-derived synaptogenic factors? The effect
of activity blockade was investigated in RGCs by culturing
5.1 N EU RO NA L R E C E P TO R S F O R them alone or with ACM in the presence or absence of TTX
A S T RO C Y T E S I G NA L S to block action potential-mediated synaptic transmission
5.1.1 Thrombospondin Receptors (Nägler et al. 2001). Astrocyte conditioned media caused a
significant increase in the frequency and amplitude of synap-
The synaptically localized α2δ1, first identified as an acces- tic events, even in the presence of TTX, showing that action
sory subunit of voltage-gated calcium channels, has been potentials are not necessary for neurons to form synapses in
identified as a neuronal receptor necessary for TSP-induced response to astrocyte factors. Whether synapses would still
synapse formation (Eroglu et al. 2009). Overexpression of form if all synaptic activity was blocked, for example, by pre-
α2δ1 causes an increase in synapse formation both in vitro venting glutamate release or blocking postsynaptic glutamate
and in vivo. Conversely, inhibition of α2δ1 with gabapentin receptors, remains to be addressed.
blocks TSP-induced synapse formation in vitro and greatly
reduces developmental synapse formation in vivo. A study
in hippocampal neurons showed that TSP can interact with 5.3 T R I G G E R S F O R R E L E A S E O F
postsynaptically localized neuroligin (which interacts with SY NA P TO G E N I C FAC TO R S FRO M A S T RO C Y T E S
presynaptic neurexin to stabilize synaptic contacts), suggest-
ing another potential mechanism by which TSP can regulate 5.3.1 Developmental Stage
synapse formation (Xu et al. 2009). Many of the identified astrocyte synaptogenic factors are
developmentally regulated, being highly expressed in early
postnatal stages during the peak period of synaptogenesis, and
5.1.2 TrkB in GABAergic Synapse Formation
at low levels or absent in the adult brain when very little synap-
Hippocampal neurons undergo enhanced GABAergic synap- togenesis is occurring. Examples of this include thrombospon-
togenesis in the presence of astrocytes, and as for RGCs, the din 1 and 2, glypican 4 and 6, and SPARC (Allen et al. 2012;
astrocytic effect is via a soluble factor. Astrocytes induce an Christopherson et al. 2005; Jones et al. 2011; Kucukdereli et al.
increase in inhibitory synapse number, increase surface levels 2011). Indeed, the transplantation of immature astrocytes to
of GABAA receptors, and enhance inhibitory synaptic trans- adult cat cortex reinduces ocular dominance plasticity after
mission and GABA currents (Elmariah et al. 2005a,b; Liu the end of the critical period, suggesting that immature astro-
et al. 1996). The identity of the astrocyte signal that induces cytes release more prosynaptogenic compounds than mature
GABAergic synapse formation in hippocampal neurons is astrocytes (Hensch 2005; Muller and Best 1989). Intriguingly,
currently unknown, but the downstream effectors have been there is a correlation between the formation of perineuronal
identified. Astrocytes modulate neuronal BDNF and TrkB nets, which are composed of astrocyte and neuron-derived
signalling, which leads to increased synaptogenesis by modu- CSPGs, around synapses in the visual cortex and the end of
lating postsynaptic maturation and synaptic recruitment of the critical period. Removal of these nets in adult rat brain via
GABAA receptors (Elmariah et al., 2005b). chondroitinase treatment, which degrades the CSPGs that

G L I A L C O N T R O L O F SY N A P TO G E N E S I S • 397
make up the nets, reactivated ocular dominance plasticity, gene, which encodes a protein that modulates translation of
linking astrocyte-derived CSPGs with the closure of the criti- mRNA into protein. Culturing Fmr1 KO astrocytes with WT
cal period and thus as factors that limit synaptic plasticity (see neurons and vice versa revealed that Fmr1 KO astrocytes cause
section 3.2.7) (Pizzorusso et al. 2002). defective dendrite development in neurons—dendrites are
more highly branched and the total dendritic area is reduced.
The number of synapses appears to be decreased when the total
5.3.2 Neuronal Signals and Activity
synapse number per neuron is analyzed, and increased when
Is there feedback between neurons and astrocytes to regulate synaptic density is analyzed, perhaps because of the reduced
the release of synaptogenic factors, so generating the final cor- dendritic arbor size causing the synapses to be more closely
rect pattern of synapse number and strength? When neuronal spaced ( Jacobs and Doering 2010; Jacobs et al. 2010).
activity is enhanced or astrocytes are stimulated with glutamate,
then astrocytes increase SPARC secretion, which negatively
6.3 R ET T SY N D RO M E
regulates glutamate receptor levels at synapses, hence reducing
overall synaptic activity ( Jones et al. 2011). Conversely, stimula- Rett syndrome is an X-linked autism spectrum disorder caused
tion of astrocytes with glutamate causes a decrease in release of by a loss of function of the transcription factor MeCP2.
TNFα, which would normally increase glutamate receptor lev- Non-cell autonomous effects from mutant astrocytes are
els at synapses, so acting as a negative feedback loop to prevent responsible for some of the features of the disorder (Ballas
synaptic strength from increasing by too much (Stellwagen et al. 2009; Lioy et al. 2011). Culturing of WT neurons in the
and Malenka 2006). Treatment of cultured astrocytes with presence of ACM from KO astrocytes causes a severe stunt-
ATP increases TSP1 secretion (Tran and Neary 2006), as does ing of dendritic outgrowth, suggesting that Rett’s astrocytes
mechanical trauma, suggesting that in an injury situation TSP1 are either deficient in the secretion of a pro-dendrite growth
may contribute to new synapse formation. Astrocyte condi- factor or secreting a toxic factor. Furthermore, re-expression of
tioned media, however, is able to induce active synapse forma- Mecp2 specifically in astrocytes in vivo on an MeCP2 deficient
tion in neurons, demonstrating some constitutive release of background partially rescues the behavioral defects observed
synaptogenic factors from astrocytes (Ullian et al. 2001). in Rett syndrome, as well as rescuing dendritic defects and
increasing presynaptic terminal numbers.

6 A R O L E F O R D E F E C T I VE
A S T R O C Y T E -I N D U C E D SY N A P S E 7 S U M M A RY A N D P E R S P E C T I VE S
F O R M AT I O N I N N E U R O D E VE L O PM E N TA L
DISORDER S Glia are intimately associated with synapses at all stages of
development and adult life, and induce the formation of syn-
Many neurodevelopmental disorders are characterized by apses as well as modulating presynaptic and postsynaptic func-
defective synapse formation or function, and there is emerg- tion. Glia contribute to the maintenance of synaptic structure
ing evidence that defects in astrocyte-induced synaptogenesis and arrangement, ensuring that neurons receive the correct
can play a role in the pathogenesis of these diseases. pattern of innervation and hence maintain synaptic balance
and nervous system function.
A growing list of astrocyte-derived synaptogenic factors
6.1 D OWN SY N D RO M E
have been identified, with multiple aspects of synapse for-
Down syndrome (DS), the most common genetic form of mation and maturation being modulated by distinct signals
mental impairment, is caused by the trisomy of chromosome from astrocytes (Figure 31.4). Current evidence suggests that
21 and is characterized by a reduced number of dendritic embryonic neurons first require physical contact with an astro-
spines on neurons along with disordered spine morphology. cyte so as to become receptive to astrocyte-secreted signals
To address whether astrocytes could be contributing to this that are present during postnatal periods. Different factors
phenotype, WT rodent hippocampal neurons were cultured are required for excitatory and inhibitory synapse formation,
in contact with human astrocytes, derived either from DS in the CNS compared with the PNS, and for structural syn-
or unaffected embryonic brains (Garcia et al. 2010). apse formation compared with presynaptic differentiation and
Neurons grown on DS astrocytes have fewer spines and fewer neurotransmitter receptor clustering. Further in vitro studies
mature spines than those grown on unaffected astrocytes. examining the effects of combinations of each of these synap-
Down syndrome astrocytes secrete less TSP1, and addition togenic factors on neuronal synapse formation will greatly aid
of TSP1 protein to DS astrocytes rescues spine number and in assessing the role that each of them has to fully functional
morphology, suggesting that reduced secretion of this synap- synapse formation. Investigating the precise spatiotemporal
togenic protein from astrocytes may contribute to DS. control of when and where each astrocyte synaptogenic factor
is expressed in vivo in the developing brain, and whether there
is regional heterogeneity in expression, will be an essential part
6.2 FR AG I L E X S Y N D RO M E
in understanding how each of these factors contributes to syn-
Fragile X syndrome (FX) is the most common inherited form aptogenesis, as well as to how their dysfunction may contrib-
of mental impairment and is caused by a mutation in the Fmr1 ute to neurological disorders

398 • FUNCTIONS OF NEUROGLIAL CELLS


REFERENCES Cullheim S, Thams S. 2007. The microglial networks of the brain
and their role in neuronal network plasticity after lesion. Brain
Allen NJ, Barres BA. 2005. Signaling between glia and neurons: focus on Res Rev 55:89–96.
synaptic plasticity. Curr Opin Neurobiol 15:542–548. Delaney CL, Brenner M, Messing A. 1996. Conditional ablation
Allen NJ, Barres BA. 2009. Neuroscience: glia—more than just brain of cerebellar astrocytes in postnatal transgenic mice. J Neurosci
glue. Nature 457:675–677. 16:6908–6918.
Allen NJ, Howe ML, Foo LC, Wang GX, Chakraborty C, Smith SJ, Elmariah SB, Hughes EG, Oh EJ, Balice-Gordon RJ. 2005a.
et al. 2012. Astrocyte glypican 4 and 6 promote formation Neurotrophin signaling among neurons and glia during forma-
of excitatory synapses via GluA1 AMPA receptors. Nature. tion of tripartite synapses. Neuron Glia Biol 1:1–11.
486:410–414. Elmariah SB, Oh EJ, Hughes EG, Balice-Gordon RJ. 2005b.
Autillo-Touati A, Rouget M, Araud D, Prochiantz A, Seite R. 1993. Astrocytes regulate inhibitory synapse formation via Trk-mediated
Astrocyte-regulated GABA-ergic striatal neurons development: an modulation of postsynaptic GABAA receptors. J Neurosci
in vitro ultrastructural study. J Hirnforsch 34:291–297. 25:3638–3650.
Awasaki T, Ito K. 2004. Engulfi ng action of glial cells is required for Eroglu Ç , Allen NJ, Susman MW, O’Rourke NA, Park CY, Özkan E,
programmed axon pruning during Drosophila metamorphosis. et al. 2009. Gabapentin receptor α2δ-1 is a neuronal thrombospon-
Curr Biol 14:668–677. din receptor responsible for excitatory CNS synaptogenesis. Cell
Bacaj T, Tevlin M, Lu Y, Shaham S. 2008. Glia are essential for sen- 139:380–392.
sory organ function in C. elegans. Science 322:744–747. Eroglu C, Barres BA. 2010. Regulation of synaptic connectivity by
Ballas N, Lioy DT, Grunseich C, Mandel G. 2009. Non-cell autono- glia. Nature 468:223–231.
mous influence of MeCP2-deficient glia on neuronal dendritic Faissner A, Pyka M, Geissler M, Sobik T, Frischknecht R, Gundelfinger
morphology. Nat Neurosci 12:311–317. ED, et al. 2010. Contributions of astrocytes to synapse formation
Banker G. 1980. Trophic interactions between astroglial cells and hip- and maturation—potential functions of the perisynaptic extracel-
pocampal neurons in culture. Science 209:809–810. lular matrix. Brain Res Rev 63:26–38.
Barker AJ, Koch SM, Reed J, Barres BA, Ullian EM. 2008. Feng Z, Ko C-P. 2008. Schwann cells promote synaptogenesis at
Developmental control of synaptic receptivity. J Neurosci the neuromuscular junction via transforming growth factor- β1.
28:8150–8160. J Neurosci 28:9599–9609.
Beattie EC, Stellwagen D, Morishita W, Bresnahan JC, Ha BK, Von Feng Z, Koirala S, Ko C-P. 2005. Synapse-glia interactions at the ver-
Zastrow M, et al. 2002. Control of synaptic strength by glial tebrate neuromuscular junction. Neuroscientist 11:503–513.
TNFalpha. Science 295:2282–2285. Frischknecht R, Heine M, Perrais D, Seidenbecher CI, Choquet D,
Bishop DL, Misgeld T, Walsh MK, Gan WB, Lichtman JW. 2004. Gundelfinger ED. 2009. Brain extracellular matrix affects AMPA
Axon branch removal at developing synapses by axosome shedding. receptor lateral mobility and short-term synaptic plasticity. Nat
Neuron 44:651–661. Neurosci 12:897–904.
Blinzinger K, Kreutzberg G. 1968. Displacement of synaptic termi- Fuentes-Medel Y, Logan MA, Ashley J, Ataman B, Budnik V,
nals from regenerating motoneurons by microglial cells. Cell Tiss Freeman MR. 2009. Glia and muscle sculpt neuromuscular arbors
Res 85:145–157. by engulfing destabilized synaptic boutons and shed presynaptic
Blondel O, Collin C, McCarran WJ, Zhu S, Zamostiano R, Gozes I, debris. PLoS Biol 7:e1000184.
et al. 2000. A glia-derived signal regulating neuronal differentia- Garcia O, Torres M, Helguera P, Coskun P, Busciglio J. 2010. A role
tion. J Neurosci 20:8012–8020. for thrombospondin-1 deficits in astrocyte-mediated spine and
Boehler MD, Wheeler BC, Brewer GJ. 2007. Added astroglia promote synaptic pathology in Down’s syndrome. PLoS ONE 5:e14200.
greater synapse density and higher activity in neuronal networks. Garrett AM, Weiner JA. 2009. Control of CNS synapse develop-
Neuron Glia Biol 3:127–140. ment by γ-protocadherin-mediated astrocyte–neuron contact.
Buard I, Steinmetz CC, Claudepierre T, Pfrieger FW. 2010. Glial cells J Neurosci 29:11723–11731.
promote dendrite formation and the reception of synaptic input in Geissler M, Faissner A. 2012. A new indirect co-culture set up of
Purkinje cells from postnatal mice. Glia 58:538–545. mouse hippocampal neurons and cortical astrocytes on microelec-
Bushong EA, Martone ME, Jones YZ, Ellisman MH. 2002. trode arrays. J Neurosci Meth 204:262–272.
Protoplasmic astrocytes in CA1 stratum radiatum occupy separate G ómez-Casati ME, Murtie JC, Rio C, Stankovic K, Liberman MC,
anatomical domains. J Neurosci 22:183–192. Corfas G. 2010. Nonneuronal cells regulate synapse formation
Cao G, Ko C-P. 2007. Schwann cell-derived factors modulate syn- in the vestibular sensory epithelium via erbB-dependent BDNF
aptic activities at developing neuromuscular synapses. J Neurosci expression. Proc Natl Acad Sci U S A 107:17005–17010.
27:6712–6722. Goritz C, Mauch DH, Pfrieger FW. 2005. Multiple mechanisms
Christopherson K, Ullian E, Stokes C, Mullowney C, Hell J, Agah A, mediate cholesterol-induced synaptogenesis in a CNS neuron.
et al. 2005. Th rombospondins are astrocyte-secreted proteins that Mol Cell Neurosci 29:190–201.
promote CNS synaptogenesis. Cell 120:421–433. G öritz C, Thiebaut R, Tessier L-H, Nieweg K, Moehle C, Buard I,
Chu Y, Jin X, Parada I, Pesic A, Stevens B, Barres B, et al. 2010. et al. 2007. Glia-induced neuronal differentiation by transcrip-
Enhanced synaptic connectivity and epilepsy in C1q knockout tional regulation. Glia 55:1108–1122.
mice. Proc Natl Acad Sci U S A 107:7975–7980. Hama H, Hara C, Yamaguchi K, Miyawaki A. 2004. PKC signal-
Colón-Ramos DA, Margeta MA, Shen K. 2007. Glia promote local ing mediates global enhancement of excitatory synaptogenesis in
synaptogenesis through UNC-6 (Netrin) signaling in C. elegans. neurons triggered by local contact with astrocytes. Neuron 41:
Science 318:103–106. 405–415.
Correa-Gillieron EM, Cavalcante LA. 1999. Synaptogenesis in Hartley RS, Margulis M, Fishman PS, Lee VMY, Tang C-M. 1999.
retino-receptive layers of the superior colliculus of the opossum Functional synapses are formed between human NTera2 (NT2N,
Didelphis marsupialis. Brain Behav Evol 54:71–84. hNT) neurons grown on astrocytes. J Comp Neurol 407:1–10.
Cuevas ME, Carrasco MA, Fuentes Y, Castro P, Nualart F, Roa J, et al. Hensch TK. 2005. Critical period mechanisms in developing visual
2005. The presence of glia stimulates the appearance of glycinergic cortex. In: Gerald PS (ed.), Current topics in developmental biol-
synaptic transmission in spinal cord neurons. Mol Cell Neurosci ogy. New York: Academic Press, pp. 215–237.
28:770–778. Hu R, Cai WQ, Wu XG, Yang Z. 2007. Astrocyte-derived estrogen
Cui W, Allen ND, Skynner M, Gusterson B, Clark AJ. 2001. Inducible enhances synapse formation and synaptic transmission between
ablation of astrocytes shows that these cells are required for neuronal cultured neonatal rat cortical neurons. Neuroscience 144:
survival in the adult brain. Glia 34:272–282. 1229–1240.

G L I A L C O N T R O L O F SY N A P TO G E N E S I S • 399
Hughes EG, Elmariah SB, Balice-Gordon RJ. 2010. Astrocyte secreted Nägler K, Mauch DH, Pfrieger FW. 2001. Glia-derived signals induce
proteins selectively increase hippocampal GABAergic axon length, synapse formation in neurones of the rat central nervous system.
branching, and synaptogenesis. Mol Cell Neurosci 43:136–145. J Physiol 533:665–679.
Iino M, Goto K, Kakegawa W, Okado H, Sudo M, Ishiuchi S, et al. 2001. Nishida H, Okabe S. 2007. direct astrocytic contacts regulate local mat-
Glia-synapse interaction through Ca2+-permeable AMPA receptors uration of dendritic spines. J Neurosci 27:331–340.
in Bergmann glia. Science 292:926–929. Pannasch U, Vargová L, Reingruber J, Ezan P, Holcman D, Giaume C,
Jacobs S, Doering LC. 2010. Astrocytes prevent abnormal neuronal et al. 2011. Astroglial networks scale synaptic activity and plasticity.
development in the fragile X mouse. J Neurosci 30:4508–4514. Proc Natl Acad Sci U S A 108:8467–8472.
Jacobs S, Nathwani M, Doering L. 2010. Fragile X astrocytes induce Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P,
developmental delays in dendrite maturation and synaptic protein et al 2011. Synaptic pruning by microglia is necessary for normal
expression. BMC Neurosci 11:132. brain development. Science 333:1456–1458.
Johnson MA, Weick JP, Pearce RA, Zhang S-C. 2007. Functional neural Peng HB, Yang J-F, Dai Z, Lee CW, Hung HW, Feng ZH, et al. 2003.
development from human embryonic stem cells: accelerated synaptic Differential effects of neurotrophins and schwann cell-derived sig-
activity via astrocyte coculture. J Neurosci 27:3069–3077. nals on neuronal survival/growth and synaptogenesis. J Neurosci
Jones EV, Bernardinelli Y, Tse YC, Chierzi S, Wong TP, Murai KK. 2011. 23:5050–5060.
Astrocytes control glutamate receptor levels at developing synapses Pfrieger F. 2010. Role of glial cells in the formation and maintenance of
through SPARC–β-integrin interactions. J Neurosci 31:4154–4165. synapses. Brain Res Rev 63:39–46.
Kaech S, Banker G. 2006. Culturing hippocampal neurons. Nat Pfrieger FW, Barres BA. 1997. Synaptic efficacy enhanced by glial cells in
Protocols 1:2406–2415. vitro. Science 277:1684–1687.
Kirov SA, Sorra KE, Harris KM. 1999. Slices have more synapses than Pizzorusso T, Medini P, Berardi N, Chierzi S, Fawcett JW, Maffei L.
perfusion-fi xed hippocampus from both young and mature rats. 2002. Reactivation of ocular dominance plasticity in the adult visual
J Neurosci 19:2876–2886. cortex. Science 298:1248–1251.
Klein R. 2009. Bidirectional modulation of synaptic functions by Eph/ Pyka M, Busse C, Seidenbecher C, Gundelfinger ED, Faissner A. 2011a.
ephrin signaling. Nat Neurosci 12:15–20. Astrocytes are crucial for survival and maturation of embryonic
Kucukdereli H, Allen NJ, Lee AT, Feng A, Ozlu MI, Conatser hippocampal neurons in a neuron-glia cell-insert coculture assay.
LM, et al. 2011. Control of excitatory CNS synaptogenesis by Synapse 65:41–53.
astrocyte-secreted proteins Hevin and SPARC. Proc Natl Acad Sci Pyka M, Wetzel C, Aguado A, Geissler M, Hatt H, Faissner A. 2011b.
U S A 108:E440–E449. Chondroitin sulfate proteoglycans regulate astrocyte-dependent syn-
Li Y-X, Schaffner AE, Barker JL. 1999. Astrocytes regulate the devel- aptogenesis and modulate synaptic activity in primary embryonic hip-
opmental appearance of GABAergic and glutamatergic postsynaptic pocampal neurons. Eur J Neurosci 33: 2187–2202.
currents in cultured embryonic rat spinal neurons. Eur J Neurosci Reddy LV, Koirala S, Sugiura Y, Herrera AA, Ko CP. 2003. Glial cells
11:2537–2551. maintain synaptic structure and function and promote development
Lioy DT, Garg SK, Monaghan CE, Raber J, Foust KD, Kaspar BK, et al. of the neuromuscular junction in vivo. Neuron 40:563–580.
2011. A role for glia in the progression of Rett/’s syndrome. Nature Rouget M, Araud D, Seite R, Prochiantz A, Autillo-Touati A. 1993.
475:497–500. Astrocyte-regulated synaptogenesis: an in vitro ultrastructural study.
Lippman J, Dunaevsky A. 2005. Dendritic spine morphogenesis and Neurosci Lett 150:85–88.
plasticity. J Neurobiol 64:47–57. Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR,
Lippman JJ, Lordkipanidze T, Buell ME, Yoon SO, Dunaevsky A. 2008. Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA, Stevens B.
Morphogenesis and regulation of Bergmann glial processes during 2012. Microglia Sculpt Postnatal Neural Circuits in an Activity and
Purkinje cell dendritic spine ensheathment and synaptogenesis. Glia Complement-Dependent Manner. Neuron 74:691–705.
56:1463–1477. Schafer DP, Lehrman EK, Kautzman A, Koyama R, Mardinly AR,
Liu Q-Y, Schaffner AE, Li Y-X, Dunlap V, Barker JL. 1996. Upregulation Barres BA, et al. 2012. Microglia sculpt postnatal neural circuits in
of GABAA current by astrocytes in cultured embryonic rat hip- an activity and complement-dependent manner. Neuron 1–10.
pocampal neurons. J Neurosci 16:2912–2923. Scheiffele P. 2003. Cell-cell signaling during synapse formation in the
MacDonald JM, Beach MG, Porpiglia E, Sheehan AE, Watts RJ, Freeman CNS. Annu Rev Neurosci 26:485–508.
MR. 2006. The drosophila cell corpse engulfment receptor draper Song H-j, Stevens CF, Gage FH. 2002. Neural stem cells from adult hip-
mediates glial clearance of severed axons. Neuron 50: 869–881. pocampus develop essential properties of functional CNS neurons.
Matsui K, Jahr CE. 2003. Ectopic release of synaptic vesicles. Neuron Nat Neurosci 5:438–445.
40:1173–1183. Steinmetz CC, Buard I, Claudepierre T, Nägler K, Pfrieger FW. 2006.
Mauch DH, Nagler K, Schumacher S, Goritz C, Muller EC, Otto A, Regional variations in the glial influence on synapse development in
et al. 2001. CNS synaptogenesis promoted by glia-derived choles- the mouse CNS. J Physiol 577:249–261.
terol. Science 294:1354–1357. Stellwagen D, Beattie EC, Seo JY, Malenka RC. 2005. Differential regu-
McKellar CE, Shatz CJ. 2009. Synaptogenesis in purified cortical sub- lation of AMPA receptor and GABA receptor trafficking by tumor
plate neurons. Cerebral Cortex 19:1723–1737. necrosis factor-alpha. J Neurosci 25:3219–3228.
Meyer-Franke A, Kaplan MR, Pfieger FW, Barres BA. 1995. Stellwagen D, Malenka RC. 2006. Synaptic scaling mediated by glial
Characterization of the signaling interactions that promote the TNF-[alpha]. Nature 440:1054–1059.
survival and growth of developing retinal ganglion cells in culture. Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS,
Neuron 15:805–819. Nouri N, et al. 2007. The classical complement cascade mediates
Miller FD, Gauthier AS. 2007. Timing is everything: making neurons CNS synapse elimination. Cell 131:1164–1178.
versus glia in the developing cortex. Neuron 54:357–369. Theodosis DT, Poulain DA, Oliet SHR. 2008. Activity-dependent
Muller CM, Best J. 1989. Ocular dominance plasticity in adult cat structural and functional plasticity of astrocyte-neuron interactions.
visual cortex after transplantation of cultured astrocytes. Nature Physiol Rev 88:983–1008.
342:427–430. Tournell CE, Bergstrom RA, Ferreira A. 2006. Progesterone-induced
Murai KK, Nguyen LN, Irie F, Yamaguchi Y, Pasquale EB. 2003. Control agrin expression in astrocytes modulates glia–neuron interactions
of hippocampal dendritic spine morphology through ephrin-A3/ leading to synapse formation. Neuroscience 141:1327–1338.
EphA4 signaling. Nat Neurosci 6:153–160. Tran MD, Neary JT. 2006. Purinergic signaling induces
Murai KK, Pasquale EB. 2011. Eph receptors and ephrins in neuron– thrombospondin-1 expression in astrocytes. Proc Natl Acad Sci U S
astrocyte communication at synapses. Glia 59:1567–1578. A 103:9321–9326.

400 • FUNCTIONS OF NEUROGLIAL CELLS


Ullian EM, Harris BT, Wu A, Chan JR, Barres BA. 2004. Schwann cells Watts RJ, Schuldiner O, Perrino J, Larsen C, Luo L. 2004. Glia engulf
and astrocytes induce synapse formation by spinal motor neurons in degenerating axons during developmental axon pruning. Curr Biol
culture. Mol Cell Neurosci 25:241–251. 14:678–684.
Ullian EM, Sapperstein SK, Christopherson KS, Barres BA. 2001. Weiner JA, Wang X, Tapia JC, Sanes JR. 2005. Gamma protocadherins are
Control of synapse number by glia. Science 291:657–661. required for synaptic development in the spinal cord. Proc Natl Acad
Ventura R, Harris KM. 1999. Three-dimensional relationships Sci U S A 102:8–14.
between hippocampal synapses and astrocytes. J Neurosci Witcher MR, Kirov SA, Harris KM. 2007. Plasticity of perisynaptic
19:6897–6906. astroglia during synaptogenesis in the mature rat hippocampus. Glia
Waites CL, Craig AM, Garner CC. 2005. Mechanisms of vertebrate 55:13–23.
synaptogenesis. Annu Rev Neurosci 28:251–274. Xu J, Xiao N, Xia J. 2009. Thrombospondin 1 accelerates synaptogenesis in
Wang X, Weiner JA, Levi S, Craig AM, Bradley A, Sanes JR. 2002. hippocampal neurons through neuroligin 1. Nat Neurosci 13:22–24.
Gamma protocadherins are required for survival of spinal interneu- Yamagata M, Sanes JR, Weiner JA. 2003. Synaptic adhesion molecules.
rons. Neuron 36:843–854. Curr Opin Cell Biol 15:621–632.

G L I A L C O N T R O L O F SY N A P TO G E N E S I S • 401
32.
NEURON MIGRATION AND AXON GUIDANCE
Andreas Faissner

A B B R E VI AT I O N S SVZ subventricular zone


TN-C tenascin-C, extracellular matrix
BMPs bone morphogenetic proteins glycoprotein
CAMs cell adhesion molecules TN-R tenascin-R, extracellular matrix glycoprotein
CD44 hyaluronan receptor, many isoforms known Wnt wingless proteins, morphogens in embry-
CNTN contactin, CAM of the IgSF onic development
CSF cerebrospinal fluid
CSPGs chondroitin sulfate proteoglycans
1 E VI D E N C E F O R C O N D U C I VE
DCC “deleted in colorectal cancer,” receptor of
F U N C T I O N S O F A S T R O G L I A–N E U R O N A L
the IgSF
M I G R AT I O N
DRG dorsal root ganglion
ECM extracellular matrix
1.1 N EU RO G E N E S I S EVO LVE S I N D I S T I N C T
EGF epidermal growth factor
S T E P S D U R I N G C E N T R A L N E RVO US SYS T E M
EGL external granular layer
D EVE L O PM E N T A N D I N VO LVE S R A D I A L G L I A
FGF fibroblast growth factor
A S N EU R A L S T E M C E L L
GAG glycosaminoglycan
Gcm glial cell missing The CNS originates from a layer of neuroepithelial cells that
GPI glycosylphosphatidylinositol-anchor of CAMs expand by symmetrical division. With increasing thickness
HSPGs heparan sulfate proteoglycans of the walls of the neural tube the neuroepithelium evolves to
IGL internal granular layer radial glia (RG) (see chapter 5). These proliferate and self-renew
IgSF immunoglobulin superfamily by symmetrical division, but progressively give rise to neurons
KS keratan sulfate that are generated in an asymmetrical division mode (Kriegstein
KSPGs keratan sulfate proteoglycans and Alvarez-Buylla 2009; Merkle and Alvarez-Buylla 2006) (see
L1-CAM cell adhesion molecule L1 chapter 30). Thereby, radial glia cells do give rise to neurons in
Mab monoclonal antibody vitro (Hartfuss et al. 2001; Malatesta et al. 2000) and serve as
MAG myelin associated glycoprotein neural stem cells (NSCs), as evidenced by fate mapping stud-
N-CAM neural cell adhesion molecule ies in vivo (Anthony et al. 2004; Malatesta et al. 2003; Miyata
NG2 chondroitin sulfate (part-time) proteoglycan et al. 2001; Noctor et al. 2001, 2002). A subpopulation of neu-
Nogo regeneration inhibitory glycoprotein rons called intermediate progenitors proliferates by a restricted
NSPCs neural stem/progenitor cells number of symmetrical divisions and can be viewed as a reser-
OmGP oligodendrocyte myelin glycoprotein voir for ongoing cortical expansion. In species with substan-
PDGF platelet-derived growth factor tially enlarged cortical width, a second zone of neurogenesis
PGs proteoglycans has recently been localized in a second subventricular zone,
PNN perineuronal net closer to the cortical surface (Fietz and Huttner 2011; Hansen
PSA polysialic acid et al. 2010). Following neurogenesis, oligodendrocyte precur-
PSA-N-CAM N-CAM derivatized with PSA (formerly sors are formed and migrate to target regions to myelinate the
“embryonic” N-CAM) axonal connections (Nave 2010). Finally, the radial glia recedes
RG radial glia and transforms into astrocytes, with two well-known excep-
RPTP receptor protein tyrosine phosphatase tions, the Bergmann glia of the cerebellum (Fig. 32.1) and the
Robo “roundabout,” slit-receptors of the Ig- Müller glia of the retina. Progeny of the radial glia is thought
superfamily to reside as radial-type astrocytes in the subventricular zone
Shh sonic hedgehog of the lateral ventricle and the subgranular zone of the dentate
Slits chemorepellent proteins expressed by mid- gyrus of the hippocampus, where these cells serve as stem cells
line glia, ligands of Robo-receptors for neurogenesis in the adult CNS (Kriegstein and Alvarez-
SGZ subgranular zone Buylla 2009).

402
A Basolateral B

EGL
ML

Apical
IGL
Radial migration Tangential Cerebellar migration
migration

C Phase 3-channel Hoechst GLAST βIII-


contrast Tubulin

Figure 32.1 Modes of Glia-Guided Migration. A. Neurons (red) arise from dividing (curved green arrow) radial glia by asymmetrical division and use
the glia scaffold for migration toward the outer cortical layers, where they differentiate. An independent wave of tangential migration is orthogonally
oriented (curved yellow arrow) and conveys progenitors from the median ganglionic eminence to the telencephalic cortex (Hatten 2002). B. In the
cerebellum, neuronal progenitors (red) divide in the external granular layer (EGL). After a final round of division, a leading process and then the nucleus
follow the radially oriented process of the Bergmann glia (blue). The neurons finally lodge in the growing internal granule cell layer (IGL), leaving a trail-
ing process that forms a T-shaped axon that apposes itself to the growing molecular layer (ML). C. Under particular culture conditions, neurospheres
extend radial glia processes (open arrowheads) that express the marker GLAST (green fluorescence) and guide the migration of neurons (filled arrow-
heads; ßIII-tubulin–positive, red fluorescence) that are also visible in phase contrast (Sirko et al. 2010). Micrographs courtesy of Dr. U. Theocharidis.

1.2 S H I F T FRO M SY M M ET R I C A L TO by P. Rakic and his school. The iconic reconstruction of electron
A SY M M ET R I C A L D I VI S I O N, R A D I A L micrographs represents a neuronal cell that closely apposes the
G L I A– GU I D E D N EU RO N M I G R AT I O N radial glia fiber and follows its trail toward the surface of the grow-
ing cortex (Rakic 2007). Biological clocks determine the assign-
As has been pointed out, the transition from the symmetri-
ment of the migrating neurons to discrete layers of the cortex,
cal to the asymmetrical division mode is of crucial importance
which are formed by neurons of similar birth date (Rakic et al.
for the diversification of neural cell lineages (Kriegstein and
2009). The radial unit hypothesis postulated that these neurons
Alvarez-Buylla 2009) (see chapter 30). On theoretical grounds,
would form the columnar networks of the cortex that would be
this switch of division mode can either be caused by an asym-
patterned by the trajectories of the radial glia guiding fiber (Rakic
metrical distribution of specific cellular determinants to only
2007). Recent findings have shown that a wave of neurons migrat-
one of the daughters, as has been paradigmatically proven in the
ing tangentially from the median ganglionic eminence would be
Drosophila nervous system (Gotz and Huttner 2005); alterna-
superimposed onto the pattern of radially migrating cells (see
tively, it could be caused by dispatching the daughters to distinct
Fig. 32.1). These cells are thought to give rise to GABAergic
microenvironments that might differentially instruct their fur-
interneurons of cortical networks (Hatten 2002).
ther fate (Scadden 2006).
The neuronal progenitors that arise in the course of the asym-
metrical division in the telencephalon use the radial glia cells as
1.3 C O RT I C A L L AY E R I N G A N D
migration support to reach their final destination in the form-
R E E L I N-M E D I AT E D N EU RO N M I G R AT I O N
ing layers of the emerging cortex (see Fig. 32.1). The radial glia–
guided migration represents a paradigmatic representation of At early stages of neural development, radial glia cells span
neuron–glia interactions that has been worked out in great detail the width of the growing cortex in the telencephalon and the

N E U R O N M I G R AT I O N A N D AXO N GU I DA N C E • 403
external granular layer (EGL) in the cerebellum. In the latter proliferate extensively using mechanisms that involve sonic
case, the cell type is referred to as Bergmann glia. Evidence hedgehog (Shh)–dependent signaling and cell cycle genes
for a genetic cause of migration deficits was obtained as early such as D2-cyclin and PP2A. After a final round of division,
as in the 1970s with the description of the mouse mutant progenitors detach from the EGL and begin to grow two
reeler. In this model, a general mal-positioning of neurons neurites in opposed orientation that elongate along the form-
could be determined that leads to an inversion of cortical ing molecular layer. Thereafter, a process is extended into the
layering. The gene responsible was discovered as an unfore- depth of the developing cerebellum that is oriented orthog-
seen target of insertional mutagenesis and subsequently onally with respect to the cerebellar surface (see Fig. 32.1).
named Reelin. This glycoprotein of the extracellular matrix The specification of granule neurons involves fate determin-
(ECM) is released by the Cajal-Retzius (CR) neurons of the ing receptors such as Notch2, Jagged2, and their downstream
developing cortex that vanish after development has ceased targets HES and E(spl), as well as the transcription factors
(Frotscher 1998). On the molecular level, the glycoprotein Math1 and NeuroD (Hatten and Roussel 2011).
presents extensive egf-type repeats that are similar to those The leading process extends along a glial process that spans
detected in the ECM glycoprotein Tenascin-C (Tnc) (Feng the whole width of the cerebellar cortex and is produced by
and Walsh 2001). the so-called Bergmann glia cells. This represents another
Beyond its developmental function, Reelin seems to be famous and extensively studied model of neuron migration, as
important for the adult CNS as well. Thus, several gene screens this phase is prolonged for more than two weeks after birth in
have revealed that it is associated with or implicated in neu- the rodent. Therefore, it was amenable to experimental manip-
rological or psychiatric diseases such as temporal lobe epilepsy ulation both in vivo and in ex vivo tissue explants. When the
(TLE), schizophrenia, or bipolar disorder (Frotscher 2010). In leading process has reached the layer of the Purkinje cells,
light of its roles in the regulation of cortical neuron migration, the nucleus of the granule neuron migrates along the leading
receptor systems and downstream signaling mechanisms have process and finally trans-locates the granule cell body toward
been extensively studied. The major Reelin receptors pertain the growing internal granule cell layer, beyond the Purkinje
to the broad family of lipoprotein receptors, that is, VLDLR cell layer. The trailing process finally matures to an axon that
and ApoER2, and involve downstream signaling proteins such constitutes the growing parallel fiber layer of the cerebellum
as mDab1 and abl that were also detected in genetic screens (see Fig. 32.1). This layer develops synaptic contacts with the
aiming at the bases of cortical malformations. dendrites of the Purkinje cells.
Extensive screens in the human have meanwhile provided The analysis of the migration process has revealed many
evidence for the occurrence of cortical malformations such regulating factors, including the cytoskeletal elements
as lissencephaly, a condition characterized by a reduction of doublecortin (DCX) and Tuj1 with regard to nuclear
the volume of gyri (microgyria) and sulci that is accompanied trans-location, the cell interaction molecules TAG1 (Cntn2)
by a smoothing of the cortical surface and a smaller brain size of the Ig-superfamily and the astrotactin genes (Wilson et al.
(microcephaly). This pathology is associated with neuron 2010). A particular role has been assigned to myosin II motors
migration deficits. Linkage analysis has yielded several genes and F-actin dynamics that coordinate movements of the cen-
of interest that are involved in these processes, for example, trosome and the soma during this process (Solecki et al. 2009).
centrosome-associated proteins that are implicated in progen- Also Rho GTPases have been implicated in migration control
itor cell division, and translocation of the nucleus and soma in the cerebellum (Govek et al. 2011).
in the course of neuronal migration (Ross and Walsh 2001).
These anatomical malformations are generally causing mental
1.5 N EU RO G E N E S I S I S L I M IT E D I N T H E
retardation of varying degrees of severity (Manzini and Walsh
A D U LT C E N T R A L N E RVO US SYS T E M : T H E
2011). A recent study highlights that the small GTPase RhoA
RO S T R A L M I G R ATO RY S T R E A M
expressed by radial glia plays a crucial role in the neuron migra-
tion process (Capello et al. 2012). In the adult CNS, neurogenesis is limited to two canonical
regions, that is the subventricular zone (SVZ) of the lateral
ventricle and the subgranular zone (SGZ) of the hippocampus,
1.4 N EU RO N M I G R AT I O N I N T H E C E R E B E L LUM
where a subclass of slowly dividing astrocytes are considered as
The cerebellum is a neuroanatomical system of the CNS with stem cells (Ming and Song 2005). The radial astrocytes that
comparatively well-defined cellular composition. The neurons presumably descend from radial glia serve as neural stem/pro-
of the cerebellum are generated in two distinct developmental genitor cells (NSPCs) and sustain olfactory bulb neurogenesis
phases. In the first wave, the Purkinje cells and deep cerebel- (Ming and Song 2011) (see chapter 40). The granule neurons
lar nuclei neurons, appear at the bottom of the future fourth of the hippocampus are sequentially born from NSPCs in the
ventricle, around embryonic day (E) 12/13 in the rodent. subgranular layer of the hippocampal dentate gyrus of the
Thereafter, in a second wave the granule cell progenitors are adult forebrain (Kempermann et al. 2004) (see chapter 40).
born. These proliferate and migrate from the rhombic lip into It is widely accepted that these neurogenic territories
an archlike structure that expands over the rhombic groove harbor specialized environments that sustain NSCs and are
of the future forming fourth ventricle. There, the small diam- regarded as niches that function as integrative entities for a
eter neurons constitute the external granule cell layer (EGL) large number of physiological stimuli (Scadden 2006; Zhao
(Fig. 32.1). The granule cell neurons, as they are called, et al. 2008).

404 • FUNCTIONS OF NEUROGLIAL CELLS


The privileged growth and survival environment of the of the retinal ganglion cells, the axons forming the prospec-
adult stem cell niche is constructed by astrocytes, endothelia tive optic nerve elongate along this specialized structure. The
of neighboring blood vessels, leptomeningeal cells, and cere- growth substrate is provided by a basal lamina that lines the
brospinal fluid in the case of the subventricular zone (SVZ). endfeet and comprises the extracellular matrix (ECM) com-
A variety of morphogens, cytokines, and extracellular matrix ponents laminin-1, collagen IV, heparan-sulfate proteogly-
constituents are released into the niche environment (Ihrie can (HSPG), and nidogen (Halfter et al. 2000). The growth
and Alvarez-Buylla; Kazanis and ffrench-Constant 2011), as response of retinal ganglion cells to laminin-1 is regulated by
well as neurotransmitters (Platel et al. 2010). integrins, consistent with this expression pattern.
The SVZ is starting point of a migration pathway con- Another example of the support of axon growth by astro-
cealed by astrocytes. Sonic hedgehog contributes to the main- cytes is provided by the development of the corpus callosum.
tenance and proliferation of progenitors that migrate toward The emergence of this structure depends on the formation of
the olfactory bulb in the so-called rostral migratory stream. a transient bridge of astrocytes that connects the left and the
The neuronal progenitors express PSA-NCAM in the migra- right hemisphere of the developing telencephalon. This glial
tory chain, and colonize the olfactory glomeruli, where they bridge, also called the glial sling, supports the reciprocal growth
differentiate into interneurons and contribute to the regen- of cortical axons (Fig. 32.2). The experimental interruption of
eration of the local olfactory network (Zhao et al. 2008). On the sling leads to the elimination of the corpus callosum. In
their way to the olfactory bulb, the neurons are concealed by a this situation, the connecting cortical axons role up on either
subpopulation of specialized astrocytes that form the walls of side of the cerebral midline, forming longitudinal fascicles of
a guiding astroglial tunnel. This astroglial channel is enriched misdirected axons, designated the bundle of Probst. Growth
with extracellular matrix components, in particular tenascin-C promotion of cortical axons can be restored by the implanta-
glycoproteins. These may exert inhibitory, repellent properties tion of nitrocellulose filters that are covered with embryonic
when presented as sharp gradients (see section 2.4) (Fig. 32.5), astrocytes or membranes derived therefrom, consistent with a
and thereby contribute to prevent the evasion of neuronal promoting role in vivo (Katz et al. 1983).
progenitors ( Jankovski and Sotelo 1996). On the other hand, Along these lines, channels outlined by astrocyte surfaces
in the olfactory bulb the related gene tenascin-R is required to have been revealed in the developing spinal cord and inter-
lure the neurons toward the olfactory bulb (Saghatelyan et al. preted as growth conduits for advancing corticospinal axons.
2004). Hence, also the rostral migratory stream illustrates the The blueprint hypothesis of axon growth pledges that channels
concept of glial-guided migration. Here, however, the astro- walled by astrocyte surfaces might provide a mechanical growth
cytes seem to play more a deterrent rather than a supportive and guidance substrate for growth cones. The mechanistic con-
role (see section 2 for further examples of inhibitory astroglial cept of axon guidance has been modified and substituted by an
functions). interpretation that emphasizes molecular signals in the growth
environment, the readout by specific growth cone–based recep-
tors, and the integration of these influences by signal transduc-
1.6 A S T RO G L I A L C E L L S C O N S T I T U T E
tion cascades that eventually modulate growth cone movements.
AXO NA L G ROW T H PAT H WAY S IN VI VO
Thus, a conduit function of astroglia based on the chemorepel-
The Müller cell glia spans the width of the retina and extends lent slit-2 has recently been proposed as an additional guidance
the glial endfeet toward its inner surface. After generation principle in the corpus callosum (Lemke 2001).

Axon Guidance by Astroglia

Support of outgrowing axons Growth along and deflection by


by a glial bridge (e.g. the glial sling in a glial boundary (stimulatory abutt onto
the developing corpus callosum) Inhibitory astrocytes)

Figure 32.2 Axon Guidance by Glial Surfaces. Many examples have been provided for axon guidance by astroglial cells in the developing nervous
system. Both supportive properties—as shown for the developing corpus callosum—and boundary properties, for example in the rhombomeres, have
been discussed.

N E U R O N M I G R AT I O N A N D AXO N GU I DA N C E • 405
1.7 A S T RO C Y T E S C O N S T I T U T E A genes in wiring and synaptogenesis. In some cases, protocad-
G ROW T H-P RO MOT I N G S U B S T R AT E herins are expressed by astrocytes and contribute to synapse
F O R M A N Y N EU RO NA L C E L L T Y P E S formation (Williams et al. 2010). The cytoplasmic domains
of classical cadherins interact with catenins that are required
It is well known that astrocyte monolayers constitute an excel-
for functional activation. β-Catenin is a key component of the
lent growth substrate for axons in vitro. These growth-promoting
Wnt-signaling pathway and involved in signal transduction to
properties are consistent with the conclusions derived from
the cell nucleus, also in radial glia.
the in vivo studies and are age- and lineage dependent. Thus,
The Immunoglobulin (Ig-) superfamily (IgSF) is char-
astrocytes obtained from embryonic or perinatal CNS are
acterized by the canonical Ig-domain, a structure consist-
more efficient than those obtained from postnatal tissues, exert
ing of 90 to 100 amino acids arranged in seven antiparallel
their strongest effect after short culture periods, and tend to
beta-pleated sheets that form a globular domain. In many
lose supportive properties with time in culture. Interestingly,
members, the Ig-domain is combined with fibronectin-type
Ca2+ oscillations in astrocytes condition their neurite growth
three (FNIII-, first discovered in fibronectin) domains, and
promoting properties (Kanemaru et al. 2007).
a transmembrane domain (see Fig. 32.3). In some cases,
The neurite growth promoting properties also depend on
CAMs of the Ig-superfamily are linked to the membrane by
the spatial organization of the cells. Indeed, astrocytes sus-
a glycosyl-phosphatidyl-inositol (GPI) anchor, which con-
tain axon growth when presented as monolayers, but may
fers particular mobility within the membrane. Functionally,
inhibit neurite growth when contained in a tube. In this para-
Ig-superfamily members serve calcium-independent adhe-
digm, astrocytes are grown in a cellulose acetate tube in three
sion mechanisms of the homophilic and heterophilic type
dimensions and confronted to a dorsal root ganglion that is
(Rougon and Hobert 2003). Several neuronal adhesion mol-
apposed to one end of the tube. Axons from early postnatal
ecules with strong axonal expression such as L1/Ng-CAM/
or perinatal DRGs grow readily into tubes that are filled with
neuroglia, contactin/F11/F3 (Cntn1), and TAG-1/Axonin-1
embryonic astrocytes, but less well into tubes with aged or
(Cntn2) have been grouped as Ax-CAMs, referring to their
matured astrocytes. Also, DRGs from later stages are less effi-
prominent role in axon fasciculation. This involves the activa-
cient and do not penetrate tubes with elder astrocytes. This
tion of downstream signaling mechanisms, including modula-
model hence mimics properties of the dorsal root entry zone
tion of intracellular calcium in the growth cone, converging
that represents a stop zone for centripetal axons in the adult
with those elicited via N-cadherin, and the basic FGF-receptor
(Fawcett 1997).
(Walsh and Doherty 1997).
With regard to neuron–glia interactions, selected iso-
1.8 M E M B R A N E -BA S E D A D H E S I O N SYS T E M S forms of the homophilic adhesion molecule N-CAM medi-
M E D I AT E P RO M OT I N G I N T E R AC T I O N S O F ate the binding of neurons to astrocyte surfaces (see Fig. 32.3)
N EU R A L C E L L T Y P E S (Keilhauer et al. 1985). Concerning heterophilic interactions,
the small isoform of the phosphotyrosine phosphatase recep-
The promoting and guiding effects of astrocytes discussed pre-
tor RPTP-β/ζ is expressed by astrocytes, a transmembrane
viously are mediated by specialized components such as cell
component that can interact with the neuronal adhesion
adhesion molecules (CAMs) or constituents of the extracel-
molecule contactin/F11/F3 (Cntn1) and other CAMs of the
lular matrix (ECM). In view of their functional importance,
Ig-superfamily (Faissner et al. 2006). These examples illus-
the following sections will briefly introduce the major gene
trate the functional involvement of Ig-CAMs in supportive
families involved and discuss their roles with special refer-
neuron–astrocyte interactions.
ence to astrocytes. Cell adhesion molecules were originally
The third prominent gene family of membrane based
discovered in the course of aggregation experiments per-
CAMs is represented by the integrins. These are heterodimers
formed by Holtefreter with dissociated sponges in the 1950s.
composed of α and β-subunits, which primarily mediate the
Subsequently, a large body of evidence has been accumulated
interactions of neural cells with extracellular matrix compo-
that suggests that the concept of preferential cell adhesion is
nents (see Fig. 32.3 and section 2). In some cases, interactions
also valid in the nervous system. Operationally, it is possible
between integrins and Ig-superfamily members have also been
to distinguish calcium-dependent and -independent adhesion
reported (Denda and Reichardt 2007).
mechanisms. The calcium-dependent adhesion is mediated
by the cadherin gene family, which includes the classical cad-
herins and the protocadherins. Classical cadherins comprise 2 A S T R O G L I A AT C H O I C E A N D
five cadherin repeat motifs, calcium binding sites, and a trans- DECISION POINTS
membrane domain that result in an overall molecular mass of
about 100 kDa (Fig. 32.3). The classical N-cadherin mediates 2.1 G L I A L B O U N DA R I E S ( S U B D I VI S I O N O F
neuron-astrocyte or neuron-neuron interactions. The classical R H O M B O M E R E S , N EU RO M E R E S AT T H E M O R E
cadherins underlie a homophilic, calcium-dependent adhe- A N T E R I O R , RO S T R A L C E N T R A L N E RVO US
sion mechanism that is strong enough to induce sorting-out of S Y S T E M–T H A L A MUS )
cells, as shown for E-cadherin (Takeichi 2007). A large num-
ber of cadherin superfamily genes are expressed in the CNS. Beside their function as a supportive growth substrate, also
There, the expression patterns are confined to distinct net- the formation of tissue boundaries by astrocytes has been dis-
works, and recent concepts propose a functional role of these cussed (Steindler 1993). One extensively studied example for

406 • FUNCTIONS OF NEUROGLIAL CELLS


Structures of CAMs and Recognition Molecules

s
s
CA 1
AM 0
NC 140

CA 0

ses
L

ns
he -
NC 18

12

ns
Ta F1 M

nin

s
cad oto
M/

/A F3

ass I

rin
R

n-A
α
ial”

L
P

eca
E

ina
β

ica

ζ
“cl ype
AM

AM

CN
g1 1/
xo

-β/
Pr

h-k
yp
nd

hri
Nr

T
NC

TP
Ng

Gl
Sy

Ep
Ep

RP
Ms

s
s
ns

tin
rin
CA

eri

lec
eg
dh

Se
Int
Ca

Fibronectin type Ill domain Lectin domain


Immunoglobulin domain EGF-type domain
Cadherin ectodomain Glycosyl-Phosphatidyl-Inositol
Calcium binging site Transmembrane domain
SCR consensus motif Intracellular domain
Heparan sulfates Globular domain
EF-Hand motif Cysteine-rich region
Tyrosine phosphatase

Figure 32.3 Structures of Cell Adhesion Molecules. Many molecules have been described that mediate various kinds of intercellular adhesive interac-
tions in neural tissues. These can be grouped into large gene families according to key structural features, such as the immunoglobulin type C2 domain.

segmented systems is macro-anatomically visible in 3-day-old array and abut with their endfeet on both the pial and the
chicken embryos: the rhombencephalon of the brainstem, ventricular surfaces. Comparably, in zebrafish a serial arrange-
which is subdivided into rhombomeres. The rhombomere ment of glial cells denominated the glial curtain separates
pattern has been described in higher vertebrates, including individual rhombomeres. It has been envisaged that the spe-
mammals, and can be identified as a series of eight swellings cialized boundary cells construct a privileged pathway for
along the neuraxis, which are separated by inter-rhombomeric outgrowing axons (Faissner and Steindler 1995; Kiecker and
glial boundaries. Lumsden 2005).
Thus, in rhombomere boundary regions the interkinetic For example, studies on axonogenesis have demonstrated
nuclear migration of neuroepithelial cells seems significantly that the first neurons emerge in the even-numbered and
reduced and intercellular adhesion is probably increased, as one stage later in odd-numbered rhombomeres. The axons
deduced from the finding that PSA-NCAM is enriched within of motor neurons of the brainstem emerge from the lips of
rhombomeres, whereas the more adhesive non-sialylated vari- even-numbered rhombomeres to innervate specific branchial
ant is expressed in the boundary. Neuroepithelial cells in this arches. For example, the first branchial arch is innervated by
area form a less motile and more coherent group of cells 4 to the trigeminal nerve, whose axons originate in rhombomeres
10 cell diameters wide, which might constitute a barrier to the 2 and 3 and exit in r2 at defined exit points. Lineage tracing
movement of epithelial cells from one rhombomere to the studies have shown that the neuronal progeny of these nuclei
next. Interestingly, the cells defining the inter-rhombomeric displays lineage restriction in individual segments and are pre-
boundary display a reduced spread of currents or diffusing vented to cross to the neighboring rhombomere after the glial
dyes such as Lucifer Yellow or biocytin between themselves or boundary has formed.
from one rhombomere to the next. In contrast, the neuroepi- The segmentation of the hindbrain is mirrored by remark-
thelia within the rhombomere are extensively electrically able expression patterns of transcription factors. For example,
coupled. The boundary cells exhibit an unusual fan-shaped at embryonic day 9.5 (E9.5) the mouse genes of the Hox-B

N E U R O N M I G R AT I O N A N D AXO N GU I DA N C E • 407
cluster homologues of the Drosophila antennapedia complex 2.2 M I D L I N E G L I A AT D EC I S I O N P O I N T S
are aligned and transcribed on the mouse chromosome 2 in a F O R G ROW T H C O N E S ( T H E O P T I C C H I A S M ,
5′ to 3′ direction. The limits of expression of these genes in the RO O F P L AT E , FL O O R P L AT E , A N D M I D L I N E
mouse spinal cord progress along the rostrocaudal axis and in G L I A I N DROSOPH I L A )
register with boundaries of rhombomeric pairs in ascending In addition to the boundaries present between rhombomeres
order. To illustrate the case, Hox-a2 expression terminates at and prosomeres, a second class of boundaries associated with
the boundary between r1 and r2, Hox-b2 at the r2/3, and so glial cell types has been described in the midline of developing
forth. These expression boundaries coincide in many cases with nervous systems of vertebrates. Thus, an assembly of glial cells
those of paralogous genes of the Hox-A and Hox-C complexes. separates the left and right axonal projection systems at the
The resulting Hox-code in the hindbrain can be modified by optic chiasm. It seems that growing axons interact with this
treatment with retinoic acid, which results in the reprogram- glial structure and are directed either to the ipsilateral or the
ming of rhombomere identity. These experiments underline contralateral cortex, as required in the context of binocular
the concept that the Hox-code determines regional neural vision. Several genes have been examined as potential candi-
identities in segments. A more detailed discussion of the role dates in mediating the choice decision. Among these are the
of transcription factors in developing neural and other tissues glycoproteins L1-CAM and CD44, and also chondroitin sul-
is beyond the scope of this chapter, and is available elsewhere fate proteoglycan(s) (CSPGs), as visualized by the expression
(Kiecker and Lumsden 2005; Pasini and Wilkinson 2002). of chondroitin sulfate epitopes in the glial boundary territory
The functions of glial boundaries are illustrated by the find- (Petros et al. 2008; Rasband et al. 2003).
ing that motor fibers migrating from odd- to even-numbered Comparable decisions are also observed in the roof and
rhombomeres to their exit points preferentially elongate along the floor plates of the developing spinal cord, prominent glial
boundary structures, as do neurofilament expressing reticular midline structures (Fig. 32.4). In the rat, the emerging com-
axons. A directing influence is supported by transplantation missural axons of the dorsal horn first migrate to the ventral
experiments in which axon tracts follow ectopic glial bound- half of the cord, toward the floor plate, under the influence
aries. The mechanism of the hypothesized boundary functions of the chemo attractant netrin-1. The dorsal glial midline cells
for axon guidance most probably involves specialized recogni- of the cord form a boundary that is not traversed by the com-
tion molecules. missural axons (Faissner and Steindler 1995). This structure

Open Book Preparation

Developing spinal cord Open book preparation

Roof plate Midline


Commissural neuron
Keratansulfate TAG-1/axonin-1 (cis-midline)
Proteoglycan (KSPG) L1/Ng-CAM (trans-midline)
DCC (netrin-1 receptor)
NP1/NP2 (Sema3A receptors)
Plexins (Sema receptors) Rostral
Robos (slit-receptors)
Shh

Caudal

Attractive Floor plate


(Netrin-1) Floor plate

- Netrin-1
- Nr-CAM
- Slits
Repulsive - Ephrin B3
(Slits)

Figure 32.4 The Open Book Preparation. Commissural neurons send their axons toward the floor plate in the developing rat spinal cord, following a
netrin gradient. After crossing the midline at the floor plate, responsiveness to netrins is lost and substituted by sensitivity to other types of cues, such
as slit-proteins. In response to these, the axons turn away from the midline and grow along the rostrocaudal axis, fasciculating onto existing longitudi-
nal fiber pathways. These events can be partially monitored and perturbed in vitro in the so-called open book preparation, an ex vivo explant that is
produced by cutting the developing E13 spinal cord along the midline.

408 • FUNCTIONS OF NEUROGLIAL CELLS


expresses inhibitory components, including a keratan sulfate midline-glia prevents the separation of commissural fibers
(KS) proteoglycan, and morphogenic signaling proteins of and eliminates the longitudinal connections of the ladder-like
the Wnt- and BMP families. In this context, Bmp7 seems to nervous system in drosophila. The commissural fibers of the
exert repulsive effects on a subgroup of dorsal interneurons system begin to extend between the three pairs of midline
(Chizhikov and Millen 2005). glia cells on the one hand and the pair of MP1 neurons on
On the ventral side of the cord, the glial floor plate cells the other hand. Separation of the commissural fiber systems
separate the left and right half of the neural tube. The growth involves migration of the midline glia in the correct direction
and decision events of the ventral side can be monitored in (Silies and Klambt 2011). Recently, a genetic screen has led to
the so-called open-book in the culture dish, a preparation that the identification of a number of additional genes that con-
lends itself to antibody perturbation assays (see Fig. 32.4). The trol this migratory behavior, one of which, called klötzchen,
glial floor plate cells release the extracellular (ECM) chemoat- seems to implicate the spectrin cytoskeleton of midline-glia.
tractant netrin-1, which attracts the commissural fibers toward In all cases, deficits in midline–glia migration lead to errors
the glial midline, where they cross and subsequently turn to in the separation of the connecting commissural fiber systems
elongate along the rostrocaudal orientation within the cord. (Klambt 2009).
The netrin-1 signal is decoded by the receptor DCC (deleted
in colorectal cancer), an Ig superfamily member (Colamarino
2.3 M E M B R A N E BA S E D G E N E FA M I L I E S
and Tessier-Lavigne 1995). The commissural fibers destined
I N VO LVE D I N C H O I C E P O I N T D EC I S I O N S
to cross the midline express the GPI-linked axonin-1/TAG-1
( E P H-K I NA S E S A N D E P H R I NS , RO B O,
(also referred to as contactin-2, Cntn2), an IgSF-member that
A N D S L IT, S E M A P H O R I NS )
is downregulated after the crossing has occurred. In parallel,
the IgSF-molecule L1-CAM appears on the longitudinally ori- The investigation of neural cell interactions had first resulted
ented fibers, an Ig-CAM involved in fasciculation. Antibodies in the identification of the immunoglobulin and cadherin
to the IgSF-member axonin-1/TAG-1 inhibit the crossing step superfamilies, and of growth- and motility-promoting ECM
at the glial midline, which concomitantly expresses Nr-CAM, constituents. But within the 1980s it became clear that in addi-
a heterophilic Cntn2 ligand. Concerted interactions of these tion to growth promotion, growth inhibitory molecules also
IgSF-members hence seem required to regulate the cross- contribute to the regulation of cell migration and growth cone
ing step. Subsequently, the axons lose responsiveness to the movement (Goodman 1996; Tessier-Lavigne and Goodman
attractant netrin-1, yet do not progress to the lateral part of 1996). In particular, the phenomenon of growth cone col-
the cord. This might reflect the action of selected semaphorins lapse had been described in the context of co-culture models
of the sema3-class and the complementary neuropilin and in which growth cones from sympathetic neurons encounter
plexin receptor complexes. The re-crossing of the glial mid- retinal neurites in the culture dish, or retinal neurites engage
line is prevented by the slit-proteins of the ECM, repulsive central myelin preparations. These interactions invariably
chemodiffusible signals released by the glial midline (Dickson resulted in collapse and retraction of the growth cone, which
and Gilestro 2006). Three IgSF-proteins of the robo-type (for stayed paralyzed for 30 to 60 minutes in vitro before resum-
roundabout) interact with slits and integrate the guidance sig- ing growth and exploratory behavior (Kapfhammer and
nals in a complex choreography involving downstream signal- Schwab 1992; Raper and Mason 2010). It was realized that
ing (Ypsilanti et al. 2010). In addition, the inhibitory ephrins this inhibitory effect might as well affect guidance and inhibi-
and their Eph-kinase receptors (see section 2.3) contribute to tion of regeneration.
prevent illegitimate axonal crossings (Kullander et al. 2001). With regard to the guidance aspect, the systematic analy-
Thus, ephrin-B3 is expressed by the glial midline and prevents sis of the innervation of the tectum in the visual system led
re-crossing of these axons when they enter the gray matter to the identification of novel mechanisms of axon guidance
after having migrated to the contralateral side of the spinal based on gradients of ephrin molecules and their complemen-
cord (Quinn and Wadsworth 2006). After successful cross- tary Eph-kinase receptors. One distinguishes the GPI-linked
ing, the axons are guided in the rostrocaudal direction by the ephrins-A that interact with Eph-A type tyrosine kinases from
chemodiffusible signal Shh (sonic hedgehog) (Salinas 2003). the transmembranous ephrins-B that link to the Eph-B type
Both slit and robo, which occur as several homologues tyrosine kinases. Both groups contain several members of
in the vertebrate, have originally been discovered in droso- ephrin- and Eph-type kinase genes, and a certain degree of pro-
phila, where the glial midline of the ladder-like nervous sys- miscuity in the mutual pairing combinations has been recorded
tem involves the so-called midline-glia (see chapters 1 and 2). (Egea and Klein 2007; Pasquale 2008). In many cases, a recip-
Complex genetic analysis in drosophila has revealed a battery rocal gradient–like expression of the components in neural
of genes that are required for midline-glia generation (Klambt tissues has been documented. Thus, ephrin-A5 is expressed as
et al. 2001). Upstream, the fate determination of midline gradient in the tectum and so are the complementary recep-
cells in general is controlled by single-minded. Glial cells tors EphA3 and EphA5 in the retinal neurons (Simpson et al.
emerge under the regulatory influence of spitz, a drosophila 2009). Conversely, the ephrin-B1 and ephrin-B2 ligands of
homologue of TGF-α (transforming growth factor alpha), the retinal EphB1-kinase are expressed by radial glia of the tec-
and the transcription factor pointed, which intervenes in the tum during the period of axon ingrowth (Braisted et al. 1997).
expression of gcm (glial cell missing), a further gene required It is believed that these complementary ligand receptor combi-
for the generation of glial cells in general. Elimination of the nations encode positional information in the nervous system.

N E U R O N M I G R AT I O N A N D AXO N GU I DA N C E • 409
Meanwhile, it seems clear that Eph-kinases and ephrins are (Lai Wing Sun et al. 2011). Fibronectin has been found in
expressed in neuronal and glial lineages, respectively, and that association with blood vessels, a structure in which astrocytes
they mediate the regulatory effects of glia on synapse forma- contribute to the formation of the blood-brain barrier that
tion and plasticity, in which ephrin-A3 ligand in astrocytes is isolates the CNS from the blood stream.
found close to EphA4 receptors that are exposed on dendrites Tenascin-C has been studied to some detail because it is
(Faissner et al. 2010; Murai and Pasquale 2011; Sloniowski and transiently expressed by immature astrocytes in the developing
Ethell 2011). Furthermore, they contribute to the emergence CNS in vivo. There, it has been found in numerous glial bound-
of the rhombomeric compartments mentioned in the preced- aries, for example, in the somatosensory barrel field (Faissner
ing and are involved in the choice decision of the growing cor- and Steindler 1995). The glycoproteins of the tenascin gene
ticospinal projection, where ephrin-B3 is expressed by midline family are characterized by structural motifs that are shared
glia as a ligand and stop signal for EphA4-kinase receptors (see between tenascin-C (TN-C), tenascin-R (TN-R), tenascin-X
section 2.2) (Wilkinson 2001). (TN-X), and tenascin-W (TN-W). In TN-C, a cysteine-rich
Following the avenue of growth cone collapse induction in amino-terminus is followed by a series of egf-type repeats,
sympathetic neurons, the distinct gene family of semaphorins fibronectin type III modules, and finally, homologies to
has been identified that comprises homologous members in fibrinogen β and -γ ( Joester and Faissner 2001) (see Fig. 32.5).
mouse and human. Some semaphorins can induce growth This structural organization is maintained in most members
cone collapse mediated by the plexin and neuropilin (NP1 of the gene family, with the exception of a tenascin-like gene in
and NP2) receptor complexes in the growth cone membrane drosophila, which contains the characteristic egf-type repeats
(Raper 2000). The signal transduction pathways downstream but is devoid of other structural elements. The egf-type repeats
of receptor activation involve small GTP-binding proteins, of tenascins show a particular arrangement of cysteines that
such as Rho and Rac1 (Raper and Mason 2010). Sema3a is has also been found in the extracellular matrix molecule ree-
expressed in the midline glia and regulates axon guidance at lin (see section 1.3). This motif is distinct from the egf-type
the optic chiasm (Sakai and Halloran 2006). repeat modules present in Notch or in the laminins. The
Furthermore, several constituents of the myelin sheath amino-terminus links tenascin monomers to multimers in
that inhibit axon growth by inducing growth cone collapse several cases, for example, TN-R to trimers and TN-C to hex-
have been identified, in particular Nogo-A. Nogo-A contains amers under nonreducing conditions. The hexamer appears
a region that induces growth cone collapse by interacting under the electron microscope as hexabrachion in rotary shad-
with the complementary Nogo receptor NgR (Schwab 2004; owed preparations (Chiquet-Ehrismann and Tucker 2011).
Schwab et al. 1993). NgR is GPI anchored to the growth cone Two isoforms that are distinguished by one FNIII-motif have
membrane and part of a receptor complex. Interestingly, two been described in TN-R, a gene that is expressed in oligoden-
other myelin components inhibitory to axon growth have drocytes at later stages of development (Czopka et al. 2009,
been detected, the IgSF-member MAG (myelin-associated 2010). TN-C possesses an alternative splice site between the
glycoprotein), and OMGP (oligodendrocyte-myelin glyco- fifth and sixth FNIII-module of the basic structure. As many
protein). Both also are able to activate the Nogo–receptor as six and nine additional FNIII-repeats can be inserted at
complex, which suggests a common downstream pathway this position in mouse and human TN-C, respectively. These
of myelin-dependent inhibition (Buchli and Schwab 2005; modules are highly conserved at their respective positions, but
Rossignol et al. 2007) (see chapter 56 for detailed treatment). independently spliced. Thus, an amplicon profiling performed
in the mouse CNS has revealed up to 30 alternatively spliced
variants, about 50% of the theoretically possible number of
2.4 E X T R AC E L LU L A R M AT R I X
64 isoforms ( Joester and Faissner 1999). In the human, the
G LYC O P ROT E I NS ( T E NA S C I NS , N ET R I N S ,
number of isomers could reach up to 512 possible variants,
A N D R E C E P TO R S )
assuming that the modules are freely exchangeable. In light of
The pericellular space is structured by macromolecules of the this result, the glycoprotein seems suited to specify pericellu-
extracellular matrix (ECM), which consists of glycoproteins lar microenvironments, or to distinguish glial lineages ( Joester
and proteoglycans (Barros et al. 2011; Dityatev et al. 2010; and Faissner 2001).
Garwood et al. 2001a). It was rapidly realized that astrocytes Furthermore, TN-C is associated with various pathologi-
in vitro produce many of the ECM glycoproteins originally cal conditions, including cancers and glial tumors (Orend
described in other tissues, namely, fibronectin, laminin-1, and Chiquet-Ehrismann 2006). It represents an interesting
vitronectin, thrombospondins, and tenascin-C. Laminin-1 possibility that TN-C could serve diagnostic purposes in this
is a functional component of astroglial endfeet in limiting context.
membranes, for example, in the developing retina and forms With regard to function, the tight association of expres-
an excellent growth substrate for axon extension of many sion with neuroanatomical boundaries, developmental events
neuronal cell types (Fig. 32.5). The structurally related genes and sites of plastic changes have motivated numerous experi-
netrin-1 and netrin-2 are chemodiffusible chemoattractants, mental studies in vitro. It has been shown that the glycopro-
which guide outgrowing commissural axons toward the floor- tein is antiadhesive for a large variety of cell types and deflects
plate glia of the midline in the spinal cord. This mechanism growth cones and neuronal cell bodies at boundaries in choice
is highly conserved because it has already been evolved in situations in vitro in which TN-C alternates with laminin-1
the nematode, in which unc-5 guides circumferential axons (Faissner 1997). On the other hand, homogeneous substrates

410 • FUNCTIONS OF NEUROGLIAL CELLS


A – Neurite growth promotion B – Anti-adhesion

(a) (b) A B Fluorescence


Phase

p-Orn tenascin-C

(c) (d)

Laminin-1 Fibronectin Tnc

EGF domains Fibronectin type III domains Fibrinogen domain

NH2 1 2 3 4 5 A1 A2 A4 B C D 6 7 8 COO–

EGFR αvβ3 RPTP-β Cntn1 α8β1 α2β1


α9β1 Annexin II
Syndecan

Figure 32.5. Structure-Function Model of Tenascin-C Glycoproteins. A. A homogenous tenascin-C substrate promotes neurite outgrowth from E18
hippocampal neurons. B. When presented as stripe (green fluorescence), tenascin-C boundaries deflect both neuronal cell bodies and axons. C. The
scheme depicts the structural organization of mouse tenascin-C and details functional regions that have been worked out on the basis of bioassays
using antibody perturbation and recombinant domains. Various receptors have been mapped to distinct domains. Micrograph by courtesy of
Dr. M. Michele.

of TN-C promote neurite outgrowth of most neuronal cell chain. One distinguishes heparan sulfate (HSPGs), chon-
types studied so far (see Fig. 32.5). Several receptors have been droitin sulfate (CSPGs), and keratan sulfate proteoglycans
described (Faissner 1997), among these the IgSF-member (KSPGs) of the nervous system. Tissue fractionation studies
Cntn1 (Rigato et al. 2002), which mediates TN-C–dependent performed with rat brain revealed that most HSPGs are tightly
stimulation of neurite outgrowth in embryonic hippocam- associated with cell membranes, whereas chondroitin sulfate
pal neurons, and various integrins (Myers et al. 2011) such as proteoglycans (CSPGs), which represent the major popula-
αvβ3, α1β8, and α1β9. In the extracellular matrix, TN-C tion of PGs in the CNS, are recovered in detergent-free salt
interacts with proteoglycans, for example, phosphacan or neu- extracts (Bandtlow and Zimmermann 2000). In many cases,
rocan. Although the knock-out mice appeared viable and able the HSPGs are membrane-bound and are cofactors that syn-
to reproduce at first sight, recent studies demonstrated modi- ergize the signaling of growth factors such as FGF2, PDGF,
fied behaviors in response to stress and lesions and deviations or Wnt-proteins, which bind to specific motifs in heparan
in the generation and number of NSPCs, suggesting roles in sulfate chains (Yamaguchi 2001) in the glial stem cell com-
synaptic plasticity and the neural stem cell niche (Faissner partment. Both members of the syndecan gene family and the
et al. 2010; Karus et al. 2011). GPI-linked HSPG glypican have been detected in the CNS
(Yamaguchi et al. 2010; Yanagisawa and Yu 2007).
With regard to CSPGs, the members of the lectican family
2.5 H E PA R A N S U L FAT E - A N D C H O N D RO I T I N
brevican, neurocan, versican, and aggrecan have been identi-
S U L FAT E P ROT E O G LYC A N S
fied in the CNS (Bandtlow and Zimmermann 2000). These
Various proteoglycans are expressed by embryonic radial CSPGs, which display a specific pattern of structural motifs,
glia, and both adult and reactive astrocytes and constitute associate with distinct lineages. Thus, versican is expressed
the second class of ECM components expressed in the CNS. by mature oligodendrocytes, whereas neurocan and aggre-
Proteoglycans are characterized as glycoproteins that comprise can have been found associated with neurons (Zimmermann
at least one additional, covalently linked glycosaminoglycan and Dours-Zimmermann 2008). Interestingly, several of the

N E U R O N M I G R AT I O N A N D AXO N GU I DA N C E • 411
core glycoproteins carry the HNK-1-epitope, a carbohydrate splice variant of the transmembrane receptor protein tyrosine
structure also expressed by neural recognition molecules, phosphatase beta (RPTP-β/ζ). It corresponds to the entire
or other N-linked carbohydrates, for example, of the Lewis extracellular region of the largest isoform of RPTP-β/ζ,
X-type, which are recognized by specific monoclonal antibod- which is extensively glycosylated with chondroitin sulfate gly-
ies (Hennen et al. 2011). These reports indicate a substantial cosaminoglycan chains. The large variant and a short isoform
heterogeneity of CSPGs in the CNS. Several MAbs have been derived therefrom possess a transmembrane domain and two
described that react specifically with individual PGs, such as phosphotyrosine phosphatase modules oriented toward the
CAT 301, which identifies a variant of aggrecan expressed in cytoplasm. The different isoforms of RPTP-βζ display devel-
neurons, and NG2, which recognizes the CSPG named NG2 opmental regulation and lineage-restricted expression.
that is expressed by oligodendrocyte precursors and in wound Neural stem/progenitor cells, glial precursor cells, radial
regions of neural tissues (Morgenstern et al. 2002; Richardson glia, Golgi cells, and astrocytes of different developmental
et al. 2011). The use of MAbs specific for epitopes on keratan stages and from various parts of the CNS all express
sulfate (KS) chains has shown that this carbohydrate polymer RPTP-β/ζ isoforms. For example, the large transmembrane
is transiently detectable as a boundary in the roof plate of the variant is expressed by NSPCs in the subventricular zones of
developing spinal cord, where it displays inhibitory properties the developing and the adult CNS, and by oligodendrocyte
(see section 2.2) (Chizhikov and Millen 2005). Experiments precursors. The short transmembrane form is found in
performed with versican documented inhibitory effects on astrocytes, whereas both lineages seem to release the soluble
the migration of neural crest cells and of DRG axons in a variant phosphacan to some extent. Although the mRNA is
laminin-1-rich territory. Finally, the neuronal CSPG neurocan mostly in the neuroepithelium of the embryonic brain and
binds to CAMs of the Ig-superfamily, inhibits homophilic L1- spinal cord, the protein is more widely distributed in these
or N-CAM-mediated cell adhesion, and interferes with both tissues, presumably following transport along glial processes,
neuron adhesion to and neurite outgrowth on substrates con- local secretion, and/or redistribution as a consequence of
sisting of combinations of CAMs. In light of findings such cell migration. Expression in neuronal subpopulations has
as these, CSPGs were discussed as inhibitors of neurite out- also been observed. The spatiotemporal expression pattern of
growth, an aspect that has since then obtained considerable the RPTP-β/ζ isoforms during development, maintenance,
attention in the situation of CNS lesions (Fawcett 2009; Kwok and pathology of the CNS has been correlated with a
et al. 2008; Morgenstern et al. 2002; Properzi and Fawcett range of developmental processes, which involve cell–cell
2004). The receptor tyrosine phosphatase RPTP-σ has been signaling, cellular proliferation, migration, differentiation,
proposed as receptor of inhibitory CSPGs (Shen et al. 2009). axon outgrowth, synaptogenesis, synaptic activity, and
On the other hand, glycosaminoglycans per se have not tissue regeneration (Garwood et al. 2001b). Based on the
proved inhibitory to neurite outgrowth in each situation preponderant glial expression of phosphacan/RPTP-ζ/β, the
tested, and chondroitin sulfate epitopes have been found effects on neuronal behavior of extracellular signals presented
upregulated in the regenerating peripheral nerve. In some by RPTP-ζ/β have been considered, whether as protein
cases, chondroitin sulfate epitopes have displayed neurite sequences or domains or associated with the CS-GAG chains,
outgrowth–stimulating properties. The functional properties with which they are modified.
of axon growth hence need to be considered in the context In the adult rat brain, it has been shown that phosphacan is
of overall matrix composition and the lineage and age of the expressed by a selected subpopulation of neurons that express
neurons involved (Purushothaman et al. 2012). the calcium-binding protein parvalbumin. It has been sug-
gested that CSPGs associate with hyaluronic acid in pericel-
lular matrices designated perineuronal nets (PNNs). Different
2.6 R P T P -β/ζ, P H O S P H AC A N, A N D R E L AT E D
neuronal subsets display different complements of CSPGs
I S O F O R M S I N N EU RO N– G L I A I N T E R AC T I O NS
such that perineuronal CSPGs could regulate the extracellu-
Embryonic radial glia cells and adult neurogenic niches lar milieu of neurons in cell type–specific ways. For example,
strongly express DSD-1-PG/phosphacan, one of the more late in development the mature ECM may be an important
abundant soluble CSPGs in the postnatal mouse brain and element in limiting synaptic plasticity (Faissner et al. 2010;
mouse homolog of the CSPG phosphacan from rat tissues Galtrey and Fawcett 2007; Kwok et al. 2011).
(Faissner et al. 2006). The GAG-composition of DSD-1-PG/ With regard to function, phosphacan interacts with IgSF
phosphacan is characterized by the chondroitin sulfates CS-A members such as Cntn1 and Cntn2, Nr-CAM, and Ng-CAM,
and CS-C, a keratan sulfate moiety that has been detected and hence might intervene in both homophilic and het-
with the MAb 3H1, and the DSD-1 epitope. The latter is rec- erophilic CAM interactions. Binding of Cntn1 with the
ognized by the MAb 473HD and requires (at least) a chain of amino-terminal domain of RPTP-β/ζ expressed in eucaryotic
seven disaccharides, sulfation of the carbohydrate backbone, a cells promotes neurite outgrowth. Therefore, it has been pro-
significant proportion of CS-D dimers, and dermatan sulfate posed that the transmembrane variants expressed in glia could
motifs in its sequence. The DSD-1 epitope can be enriched serve as receptors for CAMs expressed in the neuronal growth
by affinity chromatography and promotes neurite outgrowth cone in the framework of neuron–glia interactions. Because
from E18 hippocampal neurons. Thus, it represents an exam- both RPTP-β/ζ and the Ig-CAMs are linked to signal trans-
ple of a chondroitin sulfate with neurite outgrowth–promot- duction pathways, these interactions might involve reciprocal
ing properties. The secreted proteoglycan phosphacan is a signaling mechanism pathways (Stoker 2001).

412 • FUNCTIONS OF NEUROGLIAL CELLS


Within the extracellular matrix, DSD-1-PG/phosphacan on calcium modulation and G-protein activation (Giger et al.
interacts with various ligands, for example, the ECM glyco- 2010; Schwab 2004) (see chapter 56).
protein tenascin-C. The purified proteoglycan promotes neu-
rite outgrowth by E 18 rat hippocampal and cortical neurons,
3.2 R E AC T I VE A S T RO C Y T E S U P R EGU L AT E
an effect that involves the DSD-1 epitope chondroitin sulfate
E X T R AC E L LU L A R M AT R I X C O NS T IT U E N T S
chains. On the other hand, DSD-1-PG/phosphacan blocks
the neurite outgrowth promoting effect of laminin-1, suggest- Independently of myelin, the reactive astrocytes by themselves
ing that the inhibitory effect of the CSPG is context depen- inhibit neurite outgrowth in vitro and in vivo (Asher et al.
dent (Garwood et al. 1999). A similar result has been obtained 2001). The relevance of the cellular compartment in CNS
with embryonic cortical neurons. Also in this case, blockade lesion territories is emphasized by the observation that sig-
of laminin-1-dependent neurite growth promotion was still nificant regrowth of axonal connections can be obtained by
detectable after removal of the chondroitin sulfates and, there- implanting olfactory ensheathing and other cell types to foster
fore, related to the ECM context and the lineage of the respon- axonal growth through lesion structures. Likewise, Schwann
sive cells. The integration into ECM superstructures might cells promote regeneration and are able to span bridge struc-
explain to some extent why the elimination of the phosphacan tures in the CNS. The molecular components that underlie
gene does not result in serious impairment of development in the bridge properties are currently unknown, but a functional
mice. However, on the other hand it results in delayed regen- effect of ECM is also probable in this regard (Gardiner 2011).
eration of inflammatory myelin lesions (Harroch et al. 2002). From this point of view, the ECM might exert positive effects
on axon outgrowth in some cases. The functional ambivalence
of the ECM is reflected under various circumstances. Thus, an
upregulation of CSPGs in the regenerating peripheral nerve
3 THE ASTROGLIAL SCAR has been observed. In contrast, upregulation of CSPGs in CNS
structures clearly correlates with inhibition of axon growth in
3.1 S C A R I N G I N R E S P O NS E TO L E S I O N,
some territories. Numerous studies have emphasized, that the
S T RU C T U R E O F T H E S C A R , R E AC T I V E
reactive astrocytes upregulate CSPGs, and these components
A S T RO C Y T E S , A N D L E P TO M E N I N G E A L C E L L S
are sufficient to override the beneficial influence of laminin-1,
For decades the dogma prevailed that the regeneration of thus impeding the axon growth process (Kwok et al. 2008;
severed central nervous system (CNS) axons is impossible. Morgenstern et al. 2002). Along these lines, it has been
The seminal experiment by Albert Aguayo, which showed reported that neurons implanted by a nontraumatic technique
that CNS neurons can extend vigorously growing axons into into the corpus callosum are able to regenerate long fibers in
peripheral nerve tubes, challenged this view and stirred a con- the presence of intact myelin, provided the fibers escape a ring
siderable interest in cellular and molecular aspects of wound of reactive astrocytes emerging around the implantation site
reactions in the CNS. In particular, the growth capacity of that express tenascin-C and CSPGs (Silver and Miller 2004).
neurons strongly suggested that environmental factors in the In addition, tenascin-C is detected in stab wounds of rodent
lesion were responsible for the abortion of axon regrowth, and and human CNS, the tissue of human hippocampal sclero-
not an intrinsic lack of capacity of the axon. Meanwhile, it is sis, and hippocampal structures of rodents subject to experi-
well established that the incapacity of the CNS to regenerate mental seizure. Furthermore, the glycoprotein is significantly
is at least in part caused by inhibitory factors released by glial enhanced in human glial CNS tumors. In view of the numer-
cells into the lesion environment (Schwab et al. 1993; Silver ous effects on axon growth, a functional role in the scar tissue
and Miller 2004) (see chapter 56). Following lesion, microg- seems plausible (Faissner 1997). These results have strongly
lia activation leads to the release of numerous cytokines and stimulated further investigations into the molecular identity
the removal of cellular debris. Furthermore, leptomeningeal of the CSPGs expressed by reactive astrocytes. It could be
cells invade the wound territory, as a consequence of mechan- shown that inhibitory activities are associated with NG2, a
ical penetration or migration processes. This cell lineage con- CSPG that is expressed by a subclass of glial cells, upregulated
tributes to the release of collagen deposits into the lesion site. in lesions and inhibiting neurite outgrowth in several in vitro
Concomitantly, the astrocytes in the lesion territory change assays. The functional significance of CSPGs in CNS lesions is
their shape, with strong starlike projections, cellular hyper- highlighted by the observation that axon sprouting and recov-
trophy, and upregulation of the glial fibrillary acidic pro- ery of function are enhanced following treatment with chon-
tein (GFAP). The reactive astrocytes tend to form a palisade droitinase ABC in vivo (Fawcett 2009).
of cellular constituents destined to shield the wound from
the surrounding tissue (see chapters 51 and 53). The reac- 3.3 T H E M EC H A N I S M S O F I N H I B IT I O N O F
tive gliosis constitutes a central cellular element of the scar AXO N G ROW T H M I G HT I N VO LVE T H E
that is stimulated by the lesion stimulus (Asher et al. 2001; E X T R AC E L LU L A R M AT R I X S U P E R S T RU C T U R E
Fitch and Silver 2008). Finally, myelin debris of degenerating
sheaths litters the lesion zone and exposes inhibitory factors. The mechanism of inhibition enacted by CSPGs is cur-
Thus, oligodendrocytes express the inhibitory proteins rently not well understood. One possibility is that the puri-
Nogo-A, MAG, and OmGP, which block axon growth by fied proteoglycans by themselves have inhibitory properties.
induction of growth cone collapse, in a manner dependent In this context, it is significant that indirect evidence for the

N E U R O N M I G R AT I O N A N D AXO N GU I DA N C E • 413
existence of defined structural motifs in glycosaminoglycan AC K N OW L E D G M E N T S
chains of proteoglycans has been obtained (Purushothaman
et al. 2012). Also, degradation of a CSPG by matrix metallo- The author apologizes to all colleagues whose valuable contri-
proteinase-2 neutralizes its inhibitory properties. On the other butions could not be cited because of editorial limitations.
hand, several reports suggest that the core proteins of proteo-
glycans as expressed in target cells are able to deter advanc-
ing growth cones, and a receptor has been discovered (Shen REFERENCES
et al. 2009). Alternatively, it is conceivable that inhibitory,
growth cone collapse–inducing components are associated Anthony TE, Klein C, Fishell G, Heintz N. 2004. Radial glia serve as
with proteoglycan structures. For example, semaphorins com- neuronal progenitors in all regions of the central nervous system.
prise charged carboxyterminal domains that are well suited Neuron 41(6):881–890.
Asher RA, Morgenstern DA, Moon LD, Fawcett JW. 2001. Chondroitin
to interact with proteoglycans in lesions. Evidence has been sulphate proteoglycans: inhibitory components of the glial scar. Prog
reported that netrin-1 protein binds to HSPGs, an interaction Brain Res 132:611–619.
that may restrict diffusion in vivo. Furthermore, treatment of Bandtlow CE, Zimmermann DR. 2000. Proteoglycans in the devel-
ex vivo CNS tissue slices with high salt solutions, hyaluroni- oping brain: new conceptual insights for old proteins. Physiol Rev
dase, or chondroitinase reduced the inhibitory or antiadhesive 80(4):1267–1290.
Barros CS, Franco SJ, Muller U. 2011. Extracellular matrix: functions in
properties of otherwise inhospitable sections. These findings the nervous system. Cold Spring Harb Perspect Biol 3(1):a005108.
are consistent with the view that the emergence of inhibitory Braisted JE, McLaughlin T, Wang HU, Friedman GC, Anderson DJ,
properties might result from matrix assembly. Along these O’Leary D D. 1997. Graded and lamina-specific distributions of
lines, recent experiments have documented that interference ligands of EphB receptor tyrosine kinases in the developing retino-
with basal lamina formation in vivo favors the regeneration tectal system. Dev Biol 191(1):14–28.
Buchli AD, Schwab ME. 2005. Inhibition of Nogo: a key strategy
of the transected fimbria–fornix system. In this context, col- to increase regeneration, plasticity and functional recovery of the
lagens have attracted increased attention. Indeed, collagen lesioned central nervous system. Ann Med 37(8):556–567.
type IV is the major collagenous component of basal laminae, Capello S, Böhringer CRJ, Bergami M, Conzelmann K-K, Ghanem A,
which seemed to form the obstacle to successful regeneration Srubek Tomassy G, et al. 2012. A radial glia-specific role of RhoA in
in that case. This raises the question whether also fibrillar col- double cortex formation. Neuron 73:911–924.
Chiquet-Ehrismann R, Tucker RP. 2011. Tenascins and the importance
lagens are upregulated by astrocytes, and whether collagen- of adhesion modulation. Cold Spring Harb Perspect Biol 3(5):ii.
like gene products of astroglial origin exist. Chizhikov VV, Millen KJ. 2005. Roof plate-dependent patterning of
the vertebrate dorsal central nervous system. Dev Biol 277(2):
287–295.
Colamarino SA, Tessier-Lavigne M. 1995. The role of the floor plate in
4 S U M M A RY A N D P E R S P E C T I VE S axon guidance. Annu Rev Neurosci 18:497–529.
Czopka T, von Holst A, ffrench-Constant C, Faissner A. 2010. Regulatory
Migration of neuronal progenitors and growth cones repre- mechanisms that mediate tenascin C-dependent inhibition of oligoden-
sent key processes in the developing nervous system. A sub- drocyte precursor differentiation. J Neurosci 30(37):12310–12322.
stantial body of research over the past decades has provided Czopka T, Von Holst A, Schmidt G, Ffrench-Constant C, Faissner A.
2009. Tenascin C and tenascin R similarly prevent the formation of
evidence that beyond chemotactic signaling systems also myelin membranes in a RhoA-dependent manner, but antagonisti-
membrane-mediated haptotaxis plays a pivotal regulatory cally regulate the expression of myelin basic protein via a separate
role. In this context, the immature and mature astrocytes pathway. Glia 57(16):1790–1801.
serve manifold instructive morphogenetic functions. Radial Denda S, Reichardt LF. 2007. Studies on integrins in the nervous system.
glia and astrocytes form transient spatiotemporal guidance Methods Enzymol 426:203–221.
Dickson BJ, Gilestro GF. 2006. Regulation of commissural axon path-
structures that direct neurons or growth cones to the desti- finding by slit and its Robo receptors. Annu Rev Cell Dev Biol
nation. On the other hand, the astroglial structures reveal 22:651–675.
an ambivalent character because they may also act as barriers Dityatev A, Seidenbecher CI, Schachner M. 2010. Compartmentalization
that conceal neuronal assemblies and deter advancing growth from the outside: the extracellular matrix and functional microdo-
cones. A large number of gene products that distribute into mains in the brain. Trends Neurosci 33(11):503–512.
Egea J, Klein R. 2007. Bidirectional Eph-ephrin signaling during axon
distinct gene families of CAMs, ECM, and recognition mol- guidance. Trends Cell Biol 17(5):230–238.
ecules partake in neuron–glia interactions and sculpture the Faissner A. 1997. The tenascin gene family in axon growth and guidance.
connectome of the CNS. Yet, although the wide variety of Cell Tissue Res 290(2):331–341.
neurons can be classified progressively thanks to combinato- Faissner A, Heck N, Dobbertin A, Garwood J. 2006. DSD-1-Proteoglycan/
rial arrangements of transcription factors and neurotransmit- Phosphacan and receptor protein tyrosine phosphatase-beta isoforms
during development and regeneration of neural tissues. Adv Exp Med
ter receptors, much less is known about the corresponding Biol 557:25–53.
astrocyte compartments. Much remains to be achieved both Faissner A, Pyka M, Geissler M, Sobik T, Frischknecht R, Gundelfinger
with regard to developmental markers and the identification ED, et al. 2010. Contributions of astrocytes to synapse formation
of fate-determining signaling pathways and transcriptional and maturation. Potential functions of the perisynaptic extracellular
regulators. It is conceivable that finally an extensive tem- matrix. Brain Res Rev 63(1–2):26–38.
Faissner A, Steindler D. 1995. Boundaries and inhibitory molecules in
poral and topological astroglial heterogeneity may emerge, developing neural tissues. Glia 13(4):233–254.
in which astrocyte subpopulations define functionally tailored Fawcett J. 2009. Molecular control of brain plasticity and repair. Prog
microenvironments for their neuronal counterparts. Brain Res 175:501–509.

414 • FUNCTIONS OF NEUROGLIAL CELLS


Fawcett JW. 1997. Astrocytic and neuronal factors affecting axon regen- Joester A, Faissner A. 2001. The structure and function of tenascins in the
eration in the damaged central nervous system. Cell Tissue Res nervous system. Matrix Biol 20(1):13–22.
290(2):371–377. Kanemaru K, Okubo Y, Hirose K, Iino M. 2007. Regulation of neurite
Feng Y, Walsh CA. 2001. Protein-protein interactions, cytoskeletal regu- growth by spontaneous Ca2+ oscillations in astrocytes. J Neurosci
lation and neuronal migration. Nat Rev Neurosci 2(6):408–416. 27(33):8957–8966.
Fietz SA, Huttner WB. 2011. Cortical progenitor expansion, self-renewal and Kapfhammer JP, Schwab ME. 1992. Modulators of neuronal migration
neurogenesis-a polarized perspective. Curr Opin Neurobiol 21(1):23–35. and neurite growth. Curr Opin Cell Biol 4(5):863–868.
Fitch MT, Silver J. 2008. CNS injury, glial scars, and inflamma- Karus M, Denecke B, ffrench-Constant C, Wiese S, Faissner A. 2011. The
tion: Inhibitory extracellular matrices and regeneration failure. extracellular matrix molecule tenascin C modulates expression levels
Exp Neurol 209(2):294–301. and territories of key patterning genes during spinal cord astrocyte
Frotscher M. 1998. Cajal-Retzius cells, Reelin, and the formation of lay- specification. Development 138(24):5321–5331.
ers. Curr Opin Neurobiol 8(5):570–575. Katz MJ, Lasek RJ, Silver J. 1983. Ontophyletics of the nervous system:
Frotscher M. 2010. Role for Reelin in stabilizing cortical architecture. development of the corpus callosum and evolution of axon tracts. Proc
Trends Neurosci 33(9):407–414. Natl Acad Sci U S A 80(19):5936–5940.
Galtrey CM, Fawcett JW. 2007. The role of chondroitin sulfate proteo- Kazanis I, ffrench-Constant C. 2011. Extracellular matrix and the neural
glycans in regeneration and plasticity in the central nervous system. stem cell niche. Dev Neurobiol 71(11):1006–1017.
Brain Res Rev 54(1):1–18. Keilhauer G, Faissner A, Schachner M. 1985. Differential inhibition of
Gardiner NJ. 2011. Integrins and the extracellular matrix: key mediators neurone-neurone, neurone-astrocyte and astrocyte-astrocyte adhesion
of development and regeneration of the sensory nervous system. Dev by L1, L2 and N-CAM antibodies. Nature 316(6030):728–730.
Neurobiol 71(11):1054–1072. Kempermann G, Jessberger S, Steiner B, Kronenberg G. 2004. Milestones
Garwood J, Heck N, Rigato F, Faissner A. 2001a. The extracellular of neuronal development in the adult hippocampus. Trends Neurosci
matrix in neural development, plasticity and regeneration. In: Walz 27(8):447–452.
W (ed.), Neural environment, the brain homeostasis in health and Kiecker C, Lumsden A. 2005. Compartments and their boundaries in ver-
disease. Totowa, NJ: Humana Press, pp. 109–158. tebrate brain development. Nat Rev Neurosci 6(7):553–564.
Garwood J, Rigato F, Heck N, Faissner A. 2001b. Tenascin glycoproteins Klambt C. 2009. Modes and regulation of glial migration in vertebrates
and the complementary ligand DSD-1-PG/phosphacan—structur- and invertebrates. Nat Rev Neurosci 10(11):769–779.
ing the neural extracellular matrix during development and repair. Klambt C, Hummel T, Granderath S, Schimmelpfeng K. 2001. Glial cell
Restor Neurol Neurosci 19(1–2):51–64. development in Drosophila. Int J Dev Neurosci 19(4):373–378.
Garwood J, Schnadelbach O, Clement A, Schutte K, Bach A, Faissner Kriegstein A, Alvarez-Buylla A. 2009. The glial nature of embryonic and
A. 1999. DSD-1-proteoglycan is the mouse homolog of phosphacan adult neural stem cells. Annu Rev Neurosci 32:149–184.
and displays opposing effects on neurite outgrowth dependent on Kullander K, Croll SD, Zimmer M, Pan L, McClain J, Hughes V, et al.
neuronal lineage. J Neurosci 19(10):3888–3899. 2001. Ephrin-B3 is the midline barrier that prevents corticospinal tract
Giger RJ, Hollis ER, 2nd, Tuszynski MH. 2010. Guidance molecules in axons from recrossing, allowing for unilateral motor control. Genes
axon regeneration. Cold Spring Harb Perspect Biol 2(7):a001867. Dev 15(7):877–888.
Goodman CS. 1996. Mechanisms and molecules that control growth Kwok JC, Afshari F, Garcia-Alias G, Fawcett JW. 2008. Proteoglycans
cone guidance. Annu Rev Neurosci 19:341–377. in the central nervous system: plasticity, regeneration and their
Gotz M, Huttner WB. 2005. The cell biology of neurogenesis. Nat Rev stimulation with chondroitinase ABC. Restor Neurol Neurosci
Mol Cell Biol 6(10):777–788. 26(2–3):131–145.
Govek EE, Hatten ME, Van Aelst L. 2011. The role of Rho GTPase pro- Kwok JC, Dick G, Wang D, Fawcett JW. 2011. Extracellular matrix and
teins in CNS neuronal migration. Dev Neurobiol 71(6):528–553. perineuronal nets in CNS repair. Dev Neurobiol 71(11): 1073–1089.
Halfter W, Dong S, Schurer B, Osanger A, Schneider W, Ruegg M, et al. Lai Wing Sun K, Correia JP, Kennedy TE. 2011. Netrins: versatile extracel-
2000. Composition, synthesis, and assembly of the embryonic chick lular cues with diverse functions. Development 138(11):2153–2169.
retinal basal lamina. Dev Biol 220(2):111–128. Lemke G. 2001. Glial control of neuronal development. Annu Rev
Hansen DV, Lui JH, Parker PR, Kriegstein AR. 2010. Neurogenic radial Neurosci 24:87–105.
glia in the outer subventricular zone of human neocortex. Nature Malatesta P, Hack MA, Hartfuss E, Kettenmann H, Klinkert W, Kirchhoff
464(7288):554–561. F, et al. 2003. Neuronal or glial progeny: regional differences in radial
Harroch S, Furtado GC, Brueck W, Rosenbluth J, Lafaille J, Chao M, glia fate. Neuron 37(5):751–764.
et al. 2002. A critical role for the protein tyrosine phosphatase receptor Malatesta P, Hartfuss E, Gotz M. 2000. Isolation of radial glial cells
type Z in functional recovery from demyelinating lesions. Nat Genet by fluorescent-activated cell sorting reveals a neuronal lineage.
32(3):411–414. Development 127(24):5253–5263.
Hartfuss E, Galli R, Heins N, Gotz M. 2001. Characterization of CNS Manzini MC, Walsh CA. 2011. What disorders of cortical development
precursor subtypes and radial glia. Dev Biol 229(1):15–30. tell us about the cortex: one plus one does not always make two. Curr
Hatten ME. 2002. New directions in neuronal migration. Science Opin Genet Dev 21(3):333–339.
297(5587):1660–1663. Merkle FT, Alvarez-Buylla A. 2006. Neural stem cells in mammalian
Hatten ME, Roussel MF. 2011. Development and cancer of the cerebel- development. Curr Opin Cell Biol 18(6):704–709.
lum. Trends Neurosci 34(3):134–142. Ming GL, Song H. 2005. Adult neurogenesis in the mammalian central
Hennen E, Czopka T, Faissner A. 2011. Structurally distinct LewisX nervous system. Annu Rev Neurosci 28:223–250.
glycans distinguish subpopulations of neural stem/progenitor cells. Ming GL, Song H. 2011. Adult neurogenesis in the Mammalian brain:
J Biol Chem 286(18):16321–16331. significant answers and significant questions. Neuron 70(4):687–702.
Ihrie RA, Alvarez-Buylla A. 2011. Lake-front property: a unique ger- Miyata T, Kawaguchi A, Okano H, Ogawa M. 2001. Asymmetric
minal niche by the lateral ventricles of the adult brain. Neuron inheritance of radial glial fibers by cortical neurons. Neuron
70(4):674–686. 31(5):727–741.
Jankovski A, Sotelo C. 1996. Subventricular zone-olfactory bulb migra- Morgenstern DA, Asher RA, Fawcett JW. 2002. Chondroitin sulphate pro-
tory pathway in the adult mouse: cellular composition and specific- teoglycans in the CNS injury response. Prog Brain Res 137:313–332.
ity as determined by heterochronic and heterotopic transplantation. Murai KK, Pasquale EB. 2011. Eph receptors and ephrins in neuron-astrocyte
J Comp Neurol 371(3):376–396. communication at synapses. Glia 59(11):1567–1578.
Joester A, Faissner A. 1999. Evidence for combinatorial variability of Myers JP, Santiago-Medina M, Gomez TM. 2011. Regulation of axonal
tenascin-C isoforms and developmental regulation in the mouse cen- outgrowth and pathfinding by integrin-ECM interactions. Dev
tral nervous system. J Biol Chem 274(24):17144–17151. Neurobiol 71(11):901–923.

N E U R O N M I G R AT I O N A N D AXO N GU I DA N C E • 415
Nave KA. 2010. Myelination and support of axonal integrity by glia. Scadden DT. 2006. The stem-cell niche as an entity of action. Nature
Nature 468(7321):244–252. 441(7097):1075–1079.
Noctor SC, Flint AC, Weissman TA, Dammerman RS, Kriegstein AR. Schwab ME. 2004. Nogo and axon regeneration. Curr Opin Neurobiol
2001. Neurons derived from radial glial cells establish radial units in 14(1):118–124.
neocortex. Nature 409(6821):714–720. Schwab ME, Kapfhammer JP, Bandtlow CE. 1993. Inhibitors of neurite
Noctor SC, Flint AC, Weissman TA, Wong WS, Clinton BK, Kriegstein growth. Annu Rev Neurosci 16:565–595.
AR. 2002. Dividing precursor cells of the embryonic cortical ventric- Shen Y, Tenney AP, Busch SA, Horn KP, Cuascut FX, Liu K, et al. 2009.
ular zone have morphological and molecular characteristics of radial PTPsigma is a receptor for chondroitin sulfate proteoglycan, an
glia. J Neurosci 22(8):3161–3173. inhibitor of neural regeneration. Science 326(5952):592–596.
Orend G, Chiquet-Ehrismann R. 2006. Tenascin-C induced signaling in Silies M, Klambt C. 2011. Adhesion and signaling between neurons and
cancer. Cancer Lett 244(2):143–163. glial cells in Drosophila. Curr Opin Neurobiol 21(1):11–16.
Pasini A, Wilkinson DG. 2002. Stabilizing the regionalisation Silver J, Miller JH. 2004. Regeneration beyond the glial scar. Nat Rev
of the developing vertebrate central nervous system. Bioessays Neurosci 5(2):146–156.
24(5):427–438. Simpson HD, Mortimer D, Goodhill GJ. 2009. Theoretical models of
Pasquale EB. 2008. Eph-ephrin bidirectional signaling in physiology and neural circuit development. Curr Top Dev Biol 87:1–51.
disease. Cell 133(1):38–52. Sirko S, von Holst A, Weber A, Wizenmann A, Theocharidis U, Gotz M,
Petros TJ, Rebsam A, Mason CA. 2008. Retinal axon growth at the optic et al. 2010. Chondroitin sulfates are required for fibroblast growth
chiasm: to cross or not to cross. Annu Rev Neurosci 31:295–315. factor-2-dependent proliferation and maintenance in neural stem
Platel JC, Stamboulian S, Nguyen I, Bordey A. 2010. Neurotransmitter cells and for epidermal growth factor-dependent migration of their
signaling in postnatal neurogenesis: the first leg. Brain Res Rev progeny. Stem Cells 28(4):775–787.
63(1–2):60–71. Sloniowski S, Ethell IM. 2011. Looking forward to EphB signaling in
Properzi F, Fawcett JW. 2004. Proteoglycans and brain repair. News synapses. Semin Cell Dev Biol [Epub ahead of print].
Physiol Sci 19:33–38. Solecki DJ, Trivedi N, Govek EE, Kerekes RA, Gleason SS, Hatten ME.
Purushothaman A, Sugahara K, Faissner A. 2012. Chondroitin sulfate 2009. Myosin II motors and F-actin dynamics drive the coordinated
“wobble motifs” modulate maintenance and differentiation of neural movement of the centrosome and soma during CNS glial-guided neu-
stem cells and their progeny. J Biol Chem 287(5):2935–2942. ronal migration. Neuron 63(1):63–80.
Quinn CC, Wadsworth WG. 2006. Axon guidance: ephrins at WRK Steindler DA. 1993. Glial boundaries in the developing nervous system.
on the midline. Curr Biol 16(22):R954–955. Annu Rev Neurosci 16:445–470.
Rakic P. 2007. The radial edifice of cortical architecture: from neuronal Stoker AW. 2001. Receptor tyrosine phosphatases in axon growth and
silhouettes to genetic engineering. Brain Res Rev 55(2):204–219. guidance. Curr Opin Neurobiol 11(1):95–102.
Rakic P, Ayoub AE, Breunig JJ, Dominguez MH. 2009. Decision by Takeichi M. 2007. The cadherin superfamily in neuronal connections
division: making cortical maps. Trends Neurosci 32(5):291–301. and interactions. Nat Rev Neurosci 8(1):11–20.
Raper J, Mason C. 2010. Cellular strategies of axonal pathfinding. Cold Tessier-Lavigne M, Goodman CS. 1996. The molecular biology of axon
Spring Harb Perspect Biol 2(9):a001933. guidance. Science 274(5290):1123–1133.
Raper JA. 2000. Semaphorins and their receptors in vertebrates and Walsh FS, Doherty P. 1997. Neural cell adhesion molecules of the immu-
invertebrates. Curr Opin Neurobiol 10(1):88–94. noglobulin superfamily: role in axon growth and guidance. Annu
Rasband K, Hardy M, Chien CB. 2003. Generating X: formation of the Rev Cell Dev Biol 13:425–456.
optic chiasm. Neuron 39(6):885–888. Wilkinson DG. 2001. Multiple roles of EPH receptors and ephrins in
Richardson WD, Young KM, Tripathi RB, McKenzie I. 2011. neural development. Nat Rev Neurosci 2(3):155–164.
NG2-glia as multipotent neural stem cells: fact or fantasy? Neuron Williams ME, de Wit J, Ghosh A. 2010. Molecular mechanisms of syn-
70(4):661–673. aptic specificity in developing neural circuits. Neuron 68(1):9–18.
Rigato F, Garwood J, Calco V, Heck N, Faivre-Sarrailh C, Faissner A. Wilson PM, Fryer RH, Fang Y, Hatten ME. 2010. Astn2, a novel
2002. Tenascin-C promotes neurite outgrowth of embryonic hip- member of the astrotactin gene family, regulates the trafficking of
pocampal neurons through the alternatively spliced fibronectin type ASTN1 during glial-guided neuronal migration. J Neurosci 30(25):
III BD domains via activation of the cell adhesion molecule F3/con- 8529–8540.
tactin. J Neurosci 22(15):6596–6609. Yamaguchi Y. 2001. Heparan sulfate proteoglycans in the nervous sys-
Ross ME, Walsh CA. 2001. Human brain malformations and their les- tem: their diverse roles in neurogenesis, axon guidance, and synapto-
sons for neuronal migration. Annu Rev Neurosci 24:1041–1070. genesis. Semin Cell Dev Biol 12(2):99–106.
Rossignol S, Schwab M, Schwartz M, Fehlings MG. 2007. Spinal cord Yamaguchi Y, Inatani M, Matsumoto Y, Ogawa J, Irie F. 2010. Roles
injury: time to move? J Neurosci 27(44):11782–11792. of heparan sulfate in mammalian brain development current views
Rougon G, Hobert O. 2003. New insights into the diversity and func- based on the findings from Ext1 conditional knockout studies. Prog
tion of neuronal immunoglobulin superfamily molecules. Annu Rev Mol Biol Transl Sci 93:133–152.
Neurosci 26:207–238. Yanagisawa M, Yu RK. 2007. The expression and functions of glycocon-
Saghatelyan A, de Chevigny A, Schachner M, Lledo PM. 2004. jugates in neural stem cells. Glycobiology 17(7):57R-74R.
Tenascin-R mediates activity-dependent recruitment of neuroblasts Ypsilanti AR, Zagar Y, Chedotal A. 2010. Moving away from the
in the adult mouse forebrain. Nat Neurosci 7(4):347–356. midline: new developments for Slit and Robo. Development
Sakai JA, Halloran MC. 2006. Semaphorin 3d guides laterality 137(12):1939–1952.
of retinal ganglion cell projections in zebrafish. Development Zhao C, Deng W, Gage FH. 2008. Mechanisms and functional implica-
133(6):1035–1044. tions of adult neurogenesis. Cell 132(4):645–660.
Salinas PC. 2003. The morphogen sonic hedgehog collaborates Zimmermann DR, Dours-Zimmermann MT. 2008. Extracellular matrix
with netrin-1 to guide axons in the spinal cord. Trends Neurosci of the central nervous system: from neglect to challenge. Histochem
26(12):641–643. Cell Biol 130(4):635–653.

416 • FUNCTIONS OF NEUROGLIAL CELLS


33.
THE ROLE OF GLIA IN THE FORMATION AND FUNCTION
OF THE BLOOD-BRAIN BARRIER
Istvan Krizbai, Imola Wilhelm, Hans-Christian Bauer, and Hannelore Bauer

A B B R E VI AT I O N S 2 T H E N E U R O VA S C U L A R U N I T

ABC ATP-binding cassette A large number of experimental data suggested that brain
AJ adherens junction functions are largely determined by a complex interaction
apoE apolipoprotein E of different cell types including neurons, glial cells, brain
AQP4 aquaporin-4 endothelial cells, and pericytes, which led to the development
BBB blood-brain barrier of the neurovascular unit (NVU) concept. One of the princi-
bFGF basic fibroblast growth factor pal functions of the NVU is the formation of the BBB. Key
CNS central nervous system cellular components of the BBB are cerebral endothelial cells,
CSF cerebrospinal fluid astrocytes, and pericytes (Fig. 33.1).
EC endothelial cell
GDNF glial-derived neurotrophic factor
GLUT-1 glucose transporter-1 2.1 E N D OT H E L I A L C E L L S
GFAP glial fibrillary acidic protein
IL interleukin Although the concept of a blood-brain barrier was conceived
LDL low density lipoprotein nearly a hundred years ago, the role of capillary endothe-
lial cells (ECs) representing this barrier was only appreci-
LIF leukemia inhibitory factor
ated much later. Detailed electron microscopical analysis
NVU neurovascular unit
using horseradish peroxidase as a tracer revealed that the
P-gp P-glycoprotein BBB is located in the cerebral endothelium (Reese and
SLC solute carrier Karnovsky 1967). Permeability studies of cerebral and non-
TGF-β transforming growth factor-β cerebral endothelia affirmed the special role of brain capillary
TJ tight junction endothelial cells, demonstrating that they undergo an addi-
TNF-α tumor necrosis factor-α tional step of differentiation that results in a specific cel-
VEGF vascular endothelial growth factor lular phenotype. The most prominent feature of cerebral
ZO zonula occludens endothelial cells is the occurrence of continuous tight
junctions (TJs), which seal the paracellular passage for
1 INTRODUCTION macromolecules and cells from the blood stream. Other char-
acteristics include the absence of fenestrations, few pinocytotic
Proper neuronal function within the vertebrate brain requires a vesicles, a high number of mitochondria and a variety of
strictly regulated microenvironment and a steady-state level of hor- transport systems in the luminal and abluminal cellular
mones, ions, transmitters and other biologically active substances. membrane. Tight junctions are a key structure to the barrier
In providing this “milieu intérieur” two barrier systems play impor- function of cerebral endothelial cells and have been a focus of
tant roles. The blood-cerebrospinal fluid (CSF) barrier is the site intense research throughout the last decades. This has led
between the blood in the choroid plexus and the CSF in the ven- to the elucidation of an unanticipated complexity of the
tricles. It consists of a monolayer of epithelial cells without interac- molecular architecture of TJs, together with the emer-
tion with cerebral cells. The second barrier system, the blood-brain gence of new functional aspects of TJ-associated proteins
barrier (BBB), represents an active interface between the central (reviewed in Bauer et al. 2011).
nervous capillaries and the extracellular fluid of neurons and glial However, endothelial cells are not the only cell type
cells. The BBB is formed by brain capillary endothelial cells in close involved in BBB function. There is sufficient experimental
association with pericytes and macroglial cells or astrocytes cov- evidence to suggest a relationship among endothelial cells,
ering the majority of the cerebral surface of the endothelium (see pericytes, glial cells, and neurons to create and/or support
Fig. 33.1A and B). This suggests a possible influence of astrocytes the BBB. Neurons are supposed to influence astroglial mor-
on capillary endothelial cells. The present chapter focuses on the phology, differentiation, and proliferation whereas pericytes
role of glial cells in the formation and function of the BBB. The and astrocytes, in turn, directly or indirectly contribute to
blood-CSF barrier will not be dealt with further in this survey. the maintenance of the BBB.

417
2.2 A S T RO C Y T E S 2.4 OT H E R C E L L S
Originally, astrocytes were considered to create the BBB in In the adult mammalian brain, few neurons come in contact
mammals, comparable to lower vertebrates like primitive fish with cerebral endothelial cells. However, during early devel-
(Bundgaard and Abbott 2008). Astrocytes nearly completely opment, when astrocytes are still absent, neuroblasts and
ensheath the capillary walls with their foot processes, thereby undifferentiated neurons are the only partners of endothelial
covering not only endothelial cells but also the intimately asso- cells. Thus, it is not unreasonable to suggest that early neurons
ciated pericytes. In this way, astrocytes are the main mediators may play a critical role in the initial induction of the BBB in
between endothelial cells and the surrounding neuronal tis- cerebral endothelial cells. It is unclear whether there are sig-
sue. Astrocytes contact the capillaries by specialized structures nals from endothelial cells to neurons and vice versa which
called endfeet. Astrocytic endfeet are characterized by a high could be important for the brain homeostasis or for neuronal
expression level of several specific proteins at their luminal function. Such communication could be accomplished by the
surface, like glucose transporter-1 (GLUT-1), P-glycoprotein, presence of neurotransmitter receptors on endothelial cells, as
aquaporin-4 (AQP4), connexin-43, and Kir 4.1 K+ channel. In observed previously.
addition to their role in transport, astrocytes are also required Microglia (described in chapter 8 and 47) is another cell
for structural support of the BBB—an issue discussed more in type neighboring cerebral vessels. The role of microglia in
detail in the following. the NVU is still poorly understood. Microglial cells control
immune responses in the brain and their activation has been
shown to potentiate BBB damage during neuroinflammation
2.3 P E R I C Y T E S
(Nishioku et al. 2009). Possible beneficial actions of micro-
Pericytes (PCs) (described in chapter 9)—which originate glial cells on BBB integrity have also been described, especially
from the mesoderm or from the neural crest–derived neu- in response to ischemic brain injury (Denes et al. 2007); how-
roectoderm, depending on their location in the central ner- ever, contradictory results have also been published (Yenari
vous system (CNS) (Winkler et al. 2011)—are located in close et al. 2006).
proximity to cerebral endothelial cells, separated only by a
common basement membrane (see Fig. 33.1). Migrating PCs
guide endothelial cells during angiogenesis in the develop- 3 S T RU C T U R E A N D F U N C T I O N S O F
ing brain (Virgintino et al. 2007). Interestingly, the number T H E B L O O D -B R A I N B A R R I E R
of PCs associated with cerebral barrier-forming capillaries is
higher than the number of PCs found at nonbarrier capillar- The general function of the BBB is to protect the homeostasis
ies, the pericytic coverage of brain capillaries being in the range within the brain from changes arising from the vascular sys-
of 22% to 32% (Sims 1991). Pericytes are phagocytic cells but tem and to serve the nutritional demands of the CNS. Thus,
have also been suggested to be involved in capillary contrac- the BBB has a dual role: it is a barrier for cells, solutes, and cer-
tion, regulation of endothelial proliferation, and angiogenesis. tain xenobiotics, and harbors a multitude of transporters for
The role of PCs in BBB formation is illustrated by the find- the selective transport of various substances, e.g., for nutrients
ing that absence of pericytes leads to endothelial hyperpla- essential to the brain (blood to brain direction) and poten-
sia, abnormal vasculogenesis (Hellström et al. 2001), and an tially harmful metabolic products (brain to blood direction)
increased BBB permeability (Armulik et al. 2010). (Fig. 33.2).

A B
Astrocyte

Basement
membrane

Endothelial Pericyte
cell

Nerve
ending

Tight junction

Figure 33.1 Structure of a Brain Capillary. Besides endothelial cells—which form the structural basis of the BBB—the main cellular components of
brain capillaries are astrocytes and pericytes. A. Schematic structure of a brain capillary. B. Electron micrograph of a cross-sectioned brain capillary.
The image shows an endothelial cell (EC) separated from a pericyte (PC) by the basement membrane (arrowheads) and ensheathed by astrocyte foot
processes (Ap).

418 • FUNCTIONS OF NEUROGLIAL CELLS


BBB functions
Transport function Barrier function

Endocytosis: Efflux Enzymatic Transcellular


Carrier
Receptor transporters barrier barrier
mediated
mediated
Adsorbtive
Paracellular
barrier
EC ABC TJs
SLC transporters
transporters enzymes

Pericyte
Astrocyte

Figure 33.2 Scheme of a Brain Capillary Showing the Main Functions of the Blood-Brain Barrier. Barrier functions: paracellular barrier provided by
tight junctions sealing the interendothelial cleft, transcellular barrier provided by the low level of pinocytosis, metabolic barrier represented by dif-
ferent enzymes, and efflux transporters (ABC transporters) excreting xenobiotics. Transport function provided by SLC transporters, which are often
asymmetrically distributed in the membrane, adsorbtive endocytosis and receptor-mediated endocytosis via receptors such as LDL-, transferrin- or
insulin-receptor.

3.1 BA R R I E R F U N C T I O N there are differences as well. The barrier properties of


The barrier function of the BBB is to restrict the transport of the TJs are directly linked to the four-span integral
potentially harmful substances and cellular elements from the membrane proteins. In this respect, brain endothelial
blood to the brain. This is achieved by a fourfold defense line, cells express occludin, members of the claudin family, and
as follows: MarvelD3. Moreover, junctional adhesion molecules,
which are single-span transmembrane proteins with two
extracellular Ig-like domains, are believed to mediate
1. The BBB constitutes a strong paracellular barrier that limits homotypic cell adhesion and transmigration of leukocytes.
the free movement of solutes and cellular elements between The key components of the barrier are claudins, actually
adjacent cells. The molecular basis of the paracellular a superfamily of 27 members. Brain endothelial cells
barrier is formed by intercellular junctions that seal the cleft express mainly claudin-5, which forms a selective barrier
between neighboring endothelial cells (Fig. 33.3). for molecules smaller than 800 Da (Nitta et al. 2003);
The junctional complex is formed by the most apically however, the expression of other claudins (-1, -3, -12, and
located tight junctions (TJs) or zonulae occludentes and possibly others) has also been reported in the cerebral
the adherens junctions (AJs) (zonulae adherentes), which endothelium (Ohtsuki et al. 2008). Plaque proteins of
support the formation and maintenance of TJs. Junctional the TJs include proteins containing PDZ domains like
proteins associate with each other to constitute a zonula occludens proteins ZO-1 and ZO-2, and non-PDZ
branching network of fibril-like structures (TJ strands) in proteins like cingulin or junction-associated coiled-coil
the cell membrane, which forms a continuous belt in each protein/paracingulin. In addition, a considerable number
cell. Tight junctions not only seal the intercellular way of proteins locating at tight intercellular contacts have been
of transport but they play a crucial role in maintaining discovered, the role of which has just partly been unraveled.
cellular polarity by preventing lateral diffusion of A recent study, in which a transcriptional profile of CNS
membrane proteins between the apical and basolateral endothelial cells was generated, raised the possibility of
surfaces (“fence function”). the enrichment of other TJ molecules at the BBB (e.g.,
Both TJs and AJs are composed of transmembrane tricellulin, PARD3) (Daneman et al. 2010a).
proteins and cytoplasmic plaque proteins that directly AJs consist of a single-span transmembrane protein
contact the actin cytoskeleton (see Fig. 33.3) (reviewed in belonging to the cadherin family, mainly vascular
Bauer et al. 2011). Tight junctions of brain endothelial cells endothelial cadherin, which is linked to the cytoskeleton
share structural similarities with epithelial TJs; however, through catenins (α, β, γ, and p120). The second major

T H E R O L E O F G L I A I N T H E F O R M AT I O N A N D F U N C T I O N O F T H E B L O O D -B R A I N B A R R I E R • 419
OCCLUDIN
ITCH
TJ
LIN
GU
CIN

P1
UP
JAM AF6

M
CASK
actin

CLAUDIN
ZO2
ZO1

ZO2

ZO1
AJ
tenin
β-ca
VE-cadherin α-catenin
γ-catenin

Figure 33.3 Schematic Structure of Interendothelial Junctions: Tight Junctions and Adherens Junctions. The transmembrane proteins of tight junctions
are claudins (claudin-5, presumably the main constituent of tight junction strands, but claudin-1,-3 and -12 are also present), occludin (assumed to
function as a regulatory tight junction protein), and JAMs (junctional adhesion molecules, possibly interacting with leukocytes). At the inner mem-
brane surface one can find ZO-proteins (zonula occludens proteins, especially ZO-1 is thought to recruit other tight junction proteins, and ZO-2) and
MUPP-1 (multi-PDZ protein 1, possibly involved in the formation of junctional strands).

protein complex at AJs consists of the transmembrane 4. In addition to these, the barrier function is supported
nectin proteins linked to afadin/AF6. by the expression of a large number of efflux transporters
ZO proteins are scaffolding proteins that bind directly to (ATP-binding cassette [ABC] transporters), like ABCB1
transmembrane proteins of TJs and also interact with AJs and (P-gp—P-glycoprotein, MDR—multidrug resistance
signaling molecules. Moreover, they act not only as structural protein), ABCC1, ABCC4 (MRPs—multidrug
components, but are involved in signaling pathways leading resistance-associated proteins) and ABCG2 (BCRP—breast
to alterations of gene expression and cell behavior (migration, cancer resistance protein) (reviewed in Shen and Zhang
proliferation) (reviewed in Bauer et al. 2011). 2010). These transporters are mainly expressed on the
apical membrane of endothelial cells and excrete different
2. The transcellular barrier (i.e., inhibition of the transport
xenobiotics from the endothelium into the blood stream.
of substances through the cytoplasm) is based on the low
The most important is P-gp, which contributes to the brain-to-
level of endocytosis and transcytosis characteristic for
blood transport of several potentially harmful substances such
brain endothelial cells. A reduced number of caveolae in
as amyloid-β and also many drugs including antiepileptic,
the cerebral endothelium compared with other endothelia
psychotropic, antiviral, and chemotherapeutic drugs.
has also been described (Nag 2011), whereas increase
in the level of caveolin-1 and the number of caveolae is
associated with BBB breakdown. 3.2 T R A NS P O RT F U N C T I O N
3. The enzymatic barrier is provided by a complex set Because of the relative impermeability of the BBB water solu-
of enzymes, including acetylcholinesterase, alkaline ble substances cannot pass freely from blood to the brain and
phosphatase, γ-glutamyl transpeptidase, monoamine vice versa. Different transport systems localizing at the BBB
oxidases, and other drug metabolizing enzymes capable of (carrier systems, transport ATPases, and receptor-mediated
degrading different chemical compounds. transport processes) (see Fig. 33.2) are responsible for both the

420 • FUNCTIONS OF NEUROGLIAL CELLS


uptake of substances, mediators, and regulators essential for the 4 O N TO G E N I C D E VE L O PM E N T O F T H E
brain and the release of products or factors from the brain. The B L O O D -B R A I N B A R R I E R
large majority of these transporters belong to the solute carrier
(SLC) family of membrane transporters. The tetrameric glu- Vascularization of the mammalian CNS starts from a pre-
cose transporter SLC-A2 (GLUT-1) is specific for endothelial existing perineural plexus (at embryonic day 12 in the rat)
cells and together with other glucose transporters of the CNS (Daneman et al. 2010b), from where endothelial cells invade
allows for an enormous uptake of substrate to satisfy the aero- the neurectoderm (Fig. 33.4A and B) and—by fusion and
bic energy demand of the brain (reviewed in Bauer 1999). branching—establish the first vascular system in the develop-
In addition, several other SLC transporters are also ing neocortex (Risau 1997). This angiogenic phase is regulated
expressed in brain endothelial cells including heterodimeric by VEGF-A and angiopoietin-1 and results in a leaky endothe-
amino acid transporters formed by the members of the SLC3 lium. In this process activated/angiogenic pericytes play an
and SLC7 families (LAT-1, y+L b0,+ transporters), and mem- important role by guiding endothelial cells and organizing the
bers of the SLC1 (glutamate transporters), SLC6 (sodium/ growing vessel wall (Virgintino et al. 2007). Gain of barrier
neurotransmitter symporters), SLC15 (proton/oligopeptide properties (development of complex TJs) and upregulation
transporter), SLC16 (monocarboxylate transporter), SLC21 of specific transporter systems (including GLUT-1, various
(organic anion transporter), SLC22 (organic anion/cation amino acid and efflux transporters) is induced by the brain
transporter), and SLC44 (choline transporter) families. The environment during a differentiation phase. In this process the
SLC-mediated transport processes can be passive or active, Wnt/β-catenin signaling and G-protein coupled receptor 124
unidirectional, or bidirectional. Furthermore, the BBB medi- may play an important role (reviewed in Liebner et al. 2011).
ates an active ion transport through the Na+/K+-ATPase, the In the embryonic brain astrocytes are almost absent and
Na+/K+/Cl– transporter, the Na+/H+-antiporter, and the radial glia are the only cells that have contact with cerebral
H+-ATPase as well. endothelial cells. Radial glia not only provide scaffolding for
Further transport possibilities across the BBB include migration and placement of neurons but exhibit neurogenic
adsorptive endocytosis; moreover, receptor mediated endo- properties themselves. In this light, the earlier findings by
cytosis through the insulin receptor, transferrin receptor, or Tontsch and Bauer (1991) showing that a neuronal membrane
the low density lipoprotein (LDL) receptor also have a special fraction from 14-day-old mouse embryos induces BBB proper-
role (reviewed in Abbott et al. 2010). ties in cloned cerebral capillary endothelial cells even to a higher

A C E 12.5

Figure 33.4 Developmental Aspects of the Blood-Brain Barrier: Microscopic Images of Two Stages of Ontogenic BBB-Formation. A,B. At embryonic day
10 the intraneural domain gets vascularized from an already existing perineural plexus. Mesenchymal cells (endothelial cells and pericytes) invade the
neuroectoderm (large arrows). Small arrows indicate cross-sectioned blood vessels containing blood cells. Bars = 10 Pm and 20 Pm, respectively.
C. Medially sectioned 12-day-old mouse embryo, after perfusion with 0.5% Trypan Blue solution. The dye (bound to proteins) was excluded from
most parts of the CNS. A weak blue staining is observed in the frontal cortex and in the area of the developing choroid plexus (arrow), probably
reflecting a gradual establishment of the BBB.

T H E R O L E O F G L I A I N T H E F O R M AT I O N A N D F U N C T I O N O F T H E B L O O D -B R A I N B A R R I E R • 421
extent than do glial cells, may be considered an important step is already a functional BBB at least for proteins and macromol-
toward a new understanding of the mechanism(s) underlying ecules at early stages of embryonic development (Fig. 33.4C)
BBB induction (reviewed in Bauer and Bauer 2000). (Bauer et al. 1995; reviewed in Saunders et al. 2000). Smaller
During postnatal development astroglia becomes the pre- molecules, such as inulin (5 kDa) are less rigorously excluded
dominant cell population in the CNS and astrocytic endfeet by the BBB in developing animals, which may be explained
get intimately attached to the capillary walls during the first by structural and functional differences between TJs in the
postnatal week (Daneman et al. 2010b). Several data suggest developing and in the adult brain. Moreover, BBB maturation
that the bidirectional interaction of the endothelium with during development seems to correlate with the development
developing astrocytes is crucial. In astrocytes upregulation of polar heterogeneity of astrocytic endfeet.
of Src-suppressed C-kinase substrate was observed inducing
downregulation of vascular endothelial growth factor (VEGF)
and upregulation of angiopoietin-1, resulting in blockage of 5 I N VO LVE M E N T O F G L I A L C E L L S I N
angiogenesis and increase in TJ formation in endothelial cells B L O O D -B R A I N B A R R I E R F U N C T I O N
(Lee et al. 2003). Moreover, astrocytic α(v)β8 integrin has
been shown to activate TGF-β in endothelial cells promoting Several lines of evidence suggest that glial cells have a cru-
vessel differentiation and stabilization (Cambier et al. 2005). cial role both in the induction and in the maintenance of
Recently it has been shown that Sonic hedgehog secreted the BBB. In this context, astroglia-dependent induction
by astrocytes interacts with Hedgehog receptors of brain of barrier-associated characteristics, such as increase in
endothelial cells, promoting BBB formation and integrity BBB-related marker enzyme activities (e.g., γ-glutamyl trans-
(Alvarez et al. 2011). Astrocyte-derived fibronectin was also peptidase, Na+/K+-ATPase, alkaline phosphatase), elevation
shown to promote angiogenesis; however, hitherto this has of glucose transporter (GLUT) protein levels, enhancement
been demonstrated only in the retina (Stenzel et al. 2011). On of transendothelial electrical resistance, as well as increased
the other hand, endothelial cells release leukemia inhibitory expression of LDL receptors and P-gp in cultured cerebral
factor (LIF), which facilitates differentiation of astrocytes endothelial cells has been documented (reviewed in Nag
(Mi et al. 2001). 2011). Furthermore, astrocytes suppress endothelial prolif-
Taken together, neural progenitors are the primary inducers eration and regulate tubulogenesis by reducing the number
of BBB-specific genes in cerebral endothelial cells, while BBB of tubes formed and increasing their diameter and length.
permeability is controlled by pericytes during embryonic devel- Astrocytes induce localization of junctional proteins to cell
opment and astrocytes postnatally (Daneman et al. 2010b). borders, contribute to the development of tube polarity, and
It is generally agreed that the BBB matures gradually dur- induce P-gp activity (Al Ahmad et al. 2011). Barrier properties
ing development. The exclusion of dyes from large parts of the of the capillary brain endothelium influenced by astrocytes are
embryonic and neonatal CNS is a strong indication that there summarized in Figure 33.5.

Efflux transporters
SLC

EC
Migration ABC TJs
enzymes transporters
Proliferation

Differentiation
Astrocyte Polarization

Figure 33.5 Schematic Representation of the Interaction of Endothelial Cells and Astrocytes. There is a bidirectional communication between astrocytes
and brain endothelial cells. Astrocytes promote formation of tight junctions and increase the activity of efflux transporters, SLCs, and enzymes; more-
over, they inhibit proliferation and migration of endothelial cells. Endothelial cells induce differentiation and polarization of astrocytes.

422 • FUNCTIONS OF NEUROGLIAL CELLS


The potential of astroglia to determine the barrier charac- having—as a consequence—astrocytes with impaired func-
teristics of the central nervous capillaries has manifold confir- tionality, the BBB proved to be also compromised (Liedtke
mation, although the situation is more complex than originally et al. 1996) and these astrocytes were unable to induce BBB
expected (reviewed in Abbott et al. 2006). properties in vitro (Pekny et al. 1998).
Several lines of evidence indicate that induction of BBB Astrocytes and brain endothelial cells mutually influence
features in cerebral endothelial cells depends on a close and each other. In this respect, astrocytes show a strong polariza-
long lasting contact between astrocytes and the capillary tion in adult brains where the BBB is well differentiated, but
endothelium. Moreover, a physical contact between endothe- are rather unpolarized in neonates with poorly developed
lial cells and astrocytes has been found to be essential (Tontsch BBB (Wolburg 1995). Astrocytic endfeet oriented toward
and Bauer 1991). The exact nature of this physical contact is blood vessels consist of well-defined rosette-like structures,
not known but it is unlikely that a heterotypic gap junction the so-called orthogonal arrays of particles which are rich in
between endothelial cells and astrocytes can be established AQP4 (Wolburg et al. 2011). The number of these particles
due to the presence of the basal lamina. The crucial role of can be increased by co-cultivation with brain endothelial cells
the neural environment in the induction of the BBB is also (Wolburg et al. 2011). When the glial cell membrane loses
illustrated by the finding that BBB properties are inducible in contact with the basal lamina, the number of these particles
nonneural endothelial cells by close apposition to cocultured is significantly reduced. Astrocytes in coculture with endothe-
astrocytes (Hayashi et al. 1997). lial cells exhibit a morphologically differentiated appear-
Experimental results from different in vitro studies have ance and have more developed signal transduction systems
also demonstrated that astrocyte-conditioned medium might (Hansson et al. 2008). Moreover, cerebral endothelial cells
influence cerebral endothelial function (e.g., can increase were shown to induce the expression of plasminogen activa-
γ-glutamyl transpeptidase activity), but the experimental tor inhibitor-1 mRNA in astrocytes (Hultman et al. 2010),
results were not always consistent. Further, the susceptibil- whereas endothelium-derived LIF has been shown to induce
ity of cerebral endothelial cells to astroglial induction of astrocytic differentiation (Mi et al. 2001).
blood-brain barrier enzymes was shown to depend on their The antioxidative activity of astrocytes and endothelial
proliferative state (Meyer et al. 1991). Considerable efforts cells kept in coculture has been reported to be significantly
have been made to characterize a soluble factor responsible higher than in mono-cultures of astrocytes (Schroeter et al.
for inducing junctional development; however, its nature is 1999). Moreover, endothelial cells can regulate growth
still unclear. Inhibitors of protein synthesis or trypsin were (Estrada et al. 1990), glutamate synthetase activity (Spoerri
shown to prevent astrocyte-related actions on the BBB, which et al. 1997), and laminin production (Wagner and Gardner
pointed to a protein or peptide as the inductive factor. 2000) of astrocytes.
Astrocytes are able to synthesize a large number of biologi-
cally active molecules (see chapter 18), which can contribute
to the induction of BBB phenotype. These include TGF-β 6 T H E B L O O D -B R A I N B A R R I E R U N D E R
(transforming growth factor-β), GDNF (glial-derived neu- PAT H O P H YS I O L O G I C A L C O N D I T I O N S :
rotrophic factor), bFGF (basic fibroblast growth factor), and ROLE OF GLIAL CELLS
IL-6 (Haseloff et al. 2005). More recently Sonic hedgehog
secreted by astrocytes has been shown to play a critical role in Lesions of the BBB are associated with different patholo-
the formation and integrity of the BBB (Alvarez et al. 2011). gies such as brain tumors, stroke, degenerative diseases
In addition, not only secreted factors but also the extracellular (e.g., Parkinson’s disease, Alzheimer’s disease), inflammatory
matrix produced by astrocytes can induce BBB characteristics processes (e.g., meningitis, multiple sclerosis), or even mood
in brain endothelial cells (Hartmann et al. 2007). disorders. Because astrocytes play a crucial role in the main-
Despite intense investigative efforts, the role of astrocytes tenance of the BBB, an altered endothelial-glial interaction
in the induction of barrier properties of the cerebral endothe- may significantly contribute to BBB disturbance. Although
lium is far from being clear. A recent proteomic analysis has the mechanisms are not yet completely unraveled, different
found upregulation of 55 distinct genes in cerebral endothelial factors produced by astrocytes under pathological conditions
cells following astroglial induction (Pottiez et al. 2011). These (IL-6, TNF-α, endothelin, or VEGF) may have important
quantitative changes mainly affected proteins involved in cell roles in this process, causing deterioration of endothelial and
structure and motility, protein metabolism and protein modi- BBB functions. In other cases astrocytes proved to protect the
fications, indicating a broad target spectrum. The number of integrity of the BBB. Effects of astrocytes on the BBB in dif-
affected genes may be even higher but their identification is ferent CNS disorders are summarized in Table 33.1.
difficult out of technical limitations: usually proteomic analy-
ses are less suitable for the analysis of membrane proteins,
6.1 B R A I N T UMO R S
not to mention the possible posttranslational modifications.
Tools of systems biology will help to understand the biological Malignant processes of the brain include primary and meta-
meaning of the activation of the given set of genes. static brain tumors. Among the primary brain tumors the large
A very important issue in determining the effect of astro- majority are gliomas (see chapter 59). The status of the BBB
cytes on the cerebral endothelium is their functional status. in glial tumors is of great therapeutic relevance and thus has
In glial fibrillary acidic protein (GFAP) knockout mice, attracted considerable research interest. In general, the BBB

T H E R O L E O F G L I A I N T H E F O R M AT I O N A N D F U N C T I O N O F T H E B L O O D -B R A I N B A R R I E R • 423
Table 33.1 EFFECT OF ASTROCYTES ON BBB FUNCTIONALITY IN CENTRAL NERVOUS SYSTEM DISORDERS

CNS DISORDER EFFECT OF ASTROCYTES MECHANISMS, MEDIATORS REFERENCE

Brain tumors BBB permeability increase VEGF release Machein et al. 1999

Modification of basal lamina Lee et al. 2009

Cytokine release (TNF-α, IL-1β, IL-6)? Seike et al. 2011

Cerebral ischemia/ Protective effects on BBB Induction of radical defense enzymes in ECs Schroeter et al. 1999
oxidative stress
Decrease of hypoxia-induced VEGF Fischer et al. 2000
expression in ECs

Ischemic preconditioning Gesuete et al. 2011

Maintenance of claudin-5 localization in ECs Al Ahmad et al. 2011

Increase in endothelial GLUT-1 expression Regina et al. 2001

BBB permeability increase? VEGF release Kaur et al. 2006

Cytokine release (IL-1β) Zhang et al. 2000

HIV encephalitis Induction of endothelial apoptosis, Dependent on gap junctions Eugenin et al. 2011
alteration of BBB integrity

Multiple sclerosis, BBB permeability increase Release of inflammatory mediators, loss of Wolburg-Buchholz
EAE polarized localization of AQP4 et al. 2009

Downregulation of TJ proteins in ECs VEGF release Argaw et al. 2009

Immune cell migration through the BBB Astrocytic ABC transporters Kooij et al. 2011

Alzheimer’s disease Decrease of barrier properties apoE4 release Nishitsuji et al. 2011
Subarachnoidal Protective effect Osteopontin Suzuki et al. 2011
hemorrhage

in gliomas is impaired: expression of TJ-associated proteins is not compromised in small metastatic tumors; however, in
decreased, the paracellular pathways are partially opened, and metastases larger than 0.25 mm in diameter the BBB is leaky.
edema is formed in the surrounding brain because of the failure It has been suggested that survival and proliferation of meta-
to clear excess fluid. For example, the TJ protein claudin-1 is not static cells largely depends on the organ microenvironment
found, while claudin-5 and occludin expression is extremely low (reviewed in Fidler 2011). In this respect astrocytes and tumor
in vessels of glioblastoma multiforme (Liebner et al. 2000). cells may influence each other directly or by secreting factors
In small early-stage tumors the BBB is still intact; how- affecting the vasculature as well.
ever, the microvascular network becomes more and more It has been shown that tumor cells and astrocytes stimulate
aberrant as we move to more advanced-stage tumors. In large each other through the secretion of inflammatory cytokines.
astrocytomas the tumor core contains scarce, dilated vessels Metastatic lung cancer cells secrete macrophage migration
expressing GLUT-1 and the border area displays glomeruloid inhibitory factor IL-8 and plasminogen activator inhibitor-1,
vessels. The vessels are positive for VEGF (Bulnes et al. 2009). whereas astrocytes activated by these factors produce IL-6,
Currently, it is not clear why the BBB in gliomas becomes TNF-α, and IL-1β (Seike et al. 2011). All these factors may
defunct. Apparently, glioma cells lack the signals which are influence the BBB as well; however, no direct evidence exists
necessary to maintain the BBB in cerebral endothelial cells. so far.
Moreover, excessive VEGF production may have an impor-
tant role by over-stimulating endothelial cell proliferation
6.2 B L O O D -B R A I N BA R R I E R F U N C T I O N
and simultaneously suppressing BBB properties (Bulnes et al.
D U R I N G I S C H E M I A/H Y P OX I A
2009; Machein et al. 1999). In addition, glioma cells are able
to modify the composition of the basal lamina of capillaries Hypoxia/reoxygenation and consequent oxidative stress
of advanced tumors: a decrease in laminin and agrin, and an induce severe BBB dysfunction (reviewed in Lehner et al.
increase in tenascin were described (Lee et al. 2009). 2011). Brain endothelial cells are vulnerable to oxidative stress,
The majority of cerebral tumors, however, are not primary which may result in genotoxic damage, apoptosis and—of spe-
tumors but metastases. Comparably to gliomas, the BBB is cial importance for the barrier function of the BBB—loss of

424 • FUNCTIONS OF NEUROGLIAL CELLS


TJ-related proteins and increased number of pinocytotic vesi- 6.3 OT H E R C E N T R A L N E RVO US
cles. Thus ischemia/hypoxia leads to an increased permeability SYS T E M D I S O R D E R S
that allows plasma proteins to extravasate into the brain tissue Besides cerebral ischemia and brain tumors, the BBB is
and results in brain edema (Plateel et al. 1997). involved in the pathogenesis of a large number of neurologi-
In general, astrocytes cope more readily with hypoxic cal disorders including inflammatory and neurodegenerative
insults than do endothelial cells and neurons (Bresgen et al. processes of the CNS, infections, or epilepsia. Astrocytes are
2006), which could be explained by their ability to upregu- considered to play an important role in the induction of BBB
late their glycolytic capacity, allowing anaerobic glycolysis to dysfunction in these pathologies. Activated astrocytes are
provide sufficient ATP for cell survival or glutamate able to produce and release different substances (e.g., IL-1β),
uptake during hypoxic conditions. Furthermore, astrocytes which in turn might affect the functional integrity of the BBB
show higher antioxidant activity than endothelial cells as (Didier et al. 2003).
revealed by in vitro studies using cell culture models (Schroeter
It has been shown that HIV-infected astrocytes can induce
et al. 1999).
apoptosis in cerebral endothelial cells and BBB integrity was
Astrocytes may play an important role in the regulation of
shown to be compromised by a gap-junction-mediated mech-
the BBB in response to oxidative insults. Generally, astrocytes
anism (Eugenin et al. 2011). A disturbed crosstalk between
have been considered to protect the BBB during ischemic and
brain endothelial cells and astrocytes has been suggested to
oxidative stress conditions. There are indications that cerebral
play a role in brain edema formation during experimental
endothelial cells co-cultured with glial cells are less suscep-
autoimmune encephalitis (EAE)—an animal model of mul-
tible to hypoxic insult, i.e., they restore membrane structures
tiple sclerosis. In this process loss of β-dystroglycan and—as
more quickly and do not exhibit hypoxia-induced paracellu-
lar permeability changes (Fischer et al. 2000; Schroeter et al. a consequence—loss of polarized AQP4 localization in astro-
1999). Astrocytes obviously induce the expression of radical cytic endfeet was observed (Wolburg-Buchholz et al. 2009).
defense enzymes in endothelial cells, thereby improving the Moreover in EAE, astrocyte-derived VEGF-A was shown to
overall defense against oxidative stress (Schroeter et al. 1999,). be involved in the downregulation of claudin-5 and occludin
Other results suggest that astrocytes protect the BBB from in the cerebral endothelium (Argaw et al. 2009). An unex-
hypoxia-induced paracellular permeability changes by decreas- pected role of astrocytic ABC transporters in immune cell
ing hypoxia-induced VEGF expression in microvascular migration across the BBB has been reported recently: block-
endothelial cells (Fischer et al, 2000). The protective effect of ing this transporter activity on reactive astrocytes inhibited
astrocytes is demonstrated by the finding that astrocytes play immune cell migration across the BBB, which was explained
an important role in ischemic preconditioning-induced BBB by the reduction of astrocytic release of the chemokine (C-C
protection (Gesuete et al. 2011). Furthermore, under hypoxic motif ) ligand 2 (Kooij et al. 2011).
conditions astrocytes were shown to maintain the proper local- During epileptogenesis the BBB breakdown precedes and
ization of claudin-5 in brain endothelial cells (Al Ahmad et al. induces astrocyte dysfunction: primary BBB lesion leads to
2011). During cerebral ischemia, hypoxia is accompanied by extravasation of albumin which binds to TGF-β-receptor 2 in
hypoglycemia, which aggravates BBB disruption. Astrocytes astrocytes and induces rapid transcriptional modifications, astro-
might exert a protective role against hypoglycemia as well, cytic transformation and dysfunction (Friedman et al. 2009).
because experimental data show that treatment of cerebral It has also been shown that apolipoprotein E (apoE)-
endothelial cells with conditioned medium obtained from containing particles secreted by astrocytes regulate TJ integ-
glucose-deprived astrocytes increased endothelial GLUT-1 rity in an apoE isoform-dependent manner. In this respect
expression and glucose uptake (Regina et al. 2001). apoE4—which is a major risk factor in Alzheimer disease—
However, acute traumatic injury and/or ischemia/hypoxia impairs the integrity of the BBB, and this may be involved in
induce “reactive astrocytes.” Interestingly, stem/precursor the pathogenesis of the disease (Nishitsuji et al. 2011). The
cell–related proteins such as the intermediate filament protein vascular component of Alzheimer’s disease has been character-
nestin and the RNA-binding protein Musashi-1—normally ized in details in a mouse model. Decreased GLUT-1 expres-
not present in mature astrocytes—were found to be expressed sion was observed in both endothelial cells and astrocytes,
in reactive astrocytes (Oki et al. 2010). Hypoxia was shown together with signs of neurovascular uncoupling (astrocyte
to enhance expression of VEGF and AQP4 in astrocytes, endfeet retraction and swelling), and loss of expression of the
which may contribute to increased permeability of blood ves- astrocyte endfeet β-dystroglycan (Merlini et al. 2011).
sels and edema formation (Kaur et al. 2006). Oxidative stress Astrocytes are also able to express TJ-related proteins
may cause inflammatory reactions of cerebral endothelial cells (Bauer et al. 1999) and expression of occludin, claudin-2, and
and astrocytes. Locally secreted chemokines such as IL-8 and claudin-11 was found to be significantly higher in astrocytes of
the monocyte chemoattractant protein-1 (MCP-1) are impor- Alzheimer’s disease and vascular dementia brains (Romanitan
tant signals for leukocyte recruitment to an inflammatory site. et al. 2010). However, the importance of this phenomenon has
Using in vitro systems it has been further shown that hypoxia/ not been clarified so far.
reoxygenation induced an increase in the release of IL-1β into Astrocyte-derived molecules may have neuroprotective
the culture medium of astrocytes. Endothelial cells treated effects as well. For instance, osteopontin synthesized by astro-
with that medium responded with a pronounced elevation of cytes was shown to attenuate BBB dysfunction caused by sub-
chemokine expression (Zhang et al. 2000). arachnoidal hemorrhage (Suzuki et al. 2011).

T H E R O L E O F G L I A I N T H E F O R M AT I O N A N D F U N C T I O N O F T H E B L O O D -B R A I N B A R R I E R • 425
7 I N VI T R O M O D E L S U S E D F O R The most widely used in vitro models are cocultures of
T H E S T U DY O F T H E B L O O D -B R A I N cerebral endothelial cells and glial cells. For this purpose,
B A R R I E R : C O C U LT U R E O F C E R E B R A L either primary cells or stable cell lines—such as the C6 astro-
E N D OT H E L I A L C E L L S A N D A S T R O C Y T E S cytoma cell line—are used. Syngeneic mouse or rat models are
the most widespread, but allogenic models, using cells of dif-
The enormous scientific and industrial interest in the physiology ferent origin (e.g., bovine endothelial cells and rat astrocytes)
and pathology of brain barriers led to the development of several can also be applied. Endothelial cells are cultured on semiper-
BBB models. The large number of these suggests that there is no meable filter inserts, whereas glial cells can either be cultured
“perfect” model so far and—depending on the respective pur- on the bottom of the filter (in direct contact mode) or on the
pose—one or the other model can be more advantageous. Efflux bottom of the wells (noncontact mode) (Fig. 33.6). Addition
transport of drug candidates can be easily and effectively tested on of astrocyte-conditioned medium instead of astroglial cells is
epithelial cell-based models; however, real in vitro BBB models frequently chosen.
are based on the culture of brain endothelial cells. In vivo models It is generally accepted that the presence of astrocytes
are the most complex, but low-throughput and labor-intensive, significantly improves barrier characteristics of the cerebral
and therefore are used mainly for validation of data. endothelial monolayer, resulting in high transendothelial
Cerebral endothelial cells used in in vitro BBB models are electrical resistance and low permeability values (reviewed in
especially primary cells of rat, mouse, pig, and bovine origin. The Wilhelm et al. 2011).
relatively high costs and special skills required for the isolation Coculture of brain endothelial cells with pericytes was also
of brain endothelial cells led to the development of several cell shown to enhance barrier properties (Nakagawa et al. 2009);
lines mainly of murine, rat, and human origin. Although these therefore, triple coculture models consisting of endothelial
cells maintain important BBB characteristics, their barrier prop- cells, astrocytes and pericytes are considered the most suitable
erties are significantly inferior compared with primary cells. and efficient in vitro models characterized up to the present
Although cerebral endothelial cells are the principal com- (see Fig. 33.6).
ponents of the BBB, several other cell types play important There is increasing evidence that shear stress is able to affect
regulatory roles in the induction and maintenance of a prop- endothelial function. This led to the development of dynamic
erly functioning BBB. This led to the inclusion of glial cells, in vitro models. For this purpose in general hollow fibers are
pericytes, and even neurons in different BBB models, mimick- used, which mimic capillaries and allow coculture of other cell
ing the in vivo structure of the BBB. types (astrocytes) as well.

B
Endothelial cells

green: ZO-1 staining


blue: nuclei

Pericytes

α-SMA staining

Astrocytes

red: GFAP staining


blue: nuclei

Figure 33.6 In Vitro Blood-Brain Barrier Model System. A. Schematic representation of the triple coculture system. Cerebral endothelial cells are
cultured on semipermeable filter inserts, pericytes are placed on the lower side of the filters, whereas astrocytes are seeded into the wells of the culture
plate. B. Immunofluorescence staining of markers of brain endothelial cells (ZO-1 tight junction protein), pericytes (α- smooth muscle actin), and
astrocytes (GFAP).

426 • FUNCTIONS OF NEUROGLIAL CELLS


8 S U M M A RY A N D P E R S P E C T I VE S Argaw AT, Gurfein BT, Zhang Y, Zameer A, John GR. 2009.
VEGF-mediated disruption of endothelial CLN-5 promotes
blood-brain barrier breakdown. Proc Natl Acad Sci U S A
In this chapter we describe the basic characteristics of the 106:1977–1982.
structure and function of the BBB. Although the barrier Armulik A, Genove G, Mae M, Nisancioglu MH, Wallgard E, Niaudet
itself is located at the level of brain capillary endothelial cells, C, et al. 2010. Pericytes regulate the blood-brain barrier. Nature
other cellular elements like astrocytes or pericytes also play an 468:557–561.
important role in the development and maintenance of the Bauer H. 1999. Glucose transporters in mammalian barin development.
In: Pardridge WB eds), Introduction to the blood-brain barrier.
BBB. Here special attention is given to the influence of astro- Cambridge, UK: Cambridge University Press, pp. 175–187.
cytes and astrocytic factors on BBB function under physiolog- Bauer H, Sonnleitner U, Lametschwandtner A, Steiner M, Adam H,
ical and pathological conditions as well. Furthermore, some Bauer HC. 1995. Ontogenic expression of the erythroid-type glucose
developmental aspects of the BBB and the in vitro models are transporter (Glut 1) in the telencephalon of the mouse: correlation to
also discussed. the tightening of the blood-brain barrier. Brain Res Dev Brain Res
86:317–325.
In the last few years important scientific developments and Bauer H, Stelzhammer W, Fuchs R, Weiger TM, Danninger C, Probst
discoveries have been made with respect to the mutual inter- Get al. 1999. Astrocytes and neurons express the tight junction-specific
action of brain endothelial cells and astrocytes in physiologi- protein occludin in vitro. Exp Cell Res 250:434–438.
cal and pathological conditions. However, the picture is still Bauer HC, Bauer H. 2000. Neural induction of the blood-brain barrier:
incomplete. Despite many known details, the mechanisms of still an enigma. Cell Mol Neurobiol 20:13–28.
Bauer HC, Traweger A, Zweimueller-Mayer J, Lehner C, Tempfer H,
how glial cells may intervene with BBB properties of endothe- Krizbai I, et al. 2011. New aspects of the molecular constituents of
lial cells are far from being clear. Recent proteomic analyses tissue barriers. J Neural Transm 118:7–21.
have opened new directions, and tools of systems biology are Bresgen N, Jaksch H, Bauer HC, Eckl P, Krizbai I, Tempfer H. 2006.
expected to help in understanding the role of different genes Astrocytes are more resistant than cerebral endothelial cells toward
and molecules that seem to be involved in the induction of geno- and cytotoxicity mediated by short-term oxidative stress.
J Neurosci Res 84:1821–1828.
endothelial barrier characteristics by astrocytes. Bulnes S, Bilbao J, Lafuente JV. 2009. Microvascular adaptive changes
Despite a significant progress made in the elucidation of in experimental endogenous brain gliomas. Histol Histopathol
BBB physiology and function during the last decades, two 24:693–706.
major problems—concerning its clinical relevance—have Bundgaard M, Abbott NJ. 2008. All vertebrates started out with a glial
remained unresolved. First, it is still unknown whether BBB blood-brain barrier 4–500 million years ago. Glia 56:699–708.
Cambier S, Gline S, Mu D, Collins R, Araya J, Dolganov G, et al.
dysfunction—which is observed under neuropathological 2005. Integrin alpha(v)beta8-mediated activation of transforming
conditions—is the cause or the consequence of the disease. growth factor-beta by perivascular astrocytes: an angiogenic con-
Second, because of its relative impermeability, the BBB repre- trol switch. Am J Pathol 166:1883–1894.
sents a major impediment in the therapy of a large number of Daneman R, Zhou L, Agalliu D, Cahoy JD, Kaushal A, Barres BA.
disorders affecting the brain. At present, drug delivery strate- 2010. The mouse blood-brain barrier transcriptome: a new resource
for understanding the development and function of brain endothe-
gies are far from satisfying all therapeutic needs. Elucidation lial cells. PLoS One 5:e13741.
of the molecular mechanisms underlying BBB function is an Daneman R, Zhou L, Kebede AA, Barres BA. 2010. Pericytes are
indispensable need to help to design successful therapies and required for blood-brain barrier integrity during embryogenesis.
prophylactic strategies. Nature 468:562–566.
Denes A, Vidyasagar R, Feng J, Narvainen J, McColl BW, Kauppinen
RA, et al. 2007. Proliferating resident microglia after focal cerebral
ischaemia in mice. J Cereb Blood Flow Metab 27:1941–1953.
AC K N OW L E D G M E N T S Didier N, Romero IA, Creminon C, Wijkhuisen A, Grassi J, Mabondzo
A. 2003. Secretion of interleukin-1beta by astrocytes mediates
The work of I. K. and I. W. was supported by the Hungarian endothelin-1 and tumour necrosis factor-alpha effects on human
brain microvascular endothelial cell permeability. J Neurochem
Scientific Research Fund (OTKA K-100807 and PD-100958, 86:246–254.
respectively). H. C. B. and H. B. are supported by the European Estrada C, Bready JV, Berliner JA, Pardridge WM, Cancilla PA.
Union (HEALTH-F2–2009–241778, Neurobid). 1990. Astrocyte growth stimulation by a soluble factor produced
by cerebral endothelial cells in vitro. J Neuropathol Exp Neurol
49:539–549.
Eugenin EA, Clements JE, Zink MC, Berman JW. 2011. Human
REFERENCES immunodeficiency virus infection of human astrocytes disrupts
blood-brain barrier integrity by a gap junction-dependent mecha-
Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ. 2010. nism. J Neurosci 31:9456–9465.
Structure and function of the blood-brain barrier. Neurobiol Dis Fidler IJ. 2011. The role of the organ microenvironment in brain metas-
37:13–25. tasis. Semin Cancer Biol 21:107–112.
Abbott NJ, Ronnback L, Hansson E. 2006. Astrocyte-endothelial inter- Fischer S, Wobben M, Kleinstück J, Renz D, Schaper W. 2000. Effect of
actions at the blood-brain barrier. Nat Rev Neurosci 7:41–53. astroglial cells on hypoxia-induced permeability in PBMEC cells. Am
Al Ahmad A, Taboada CB, Gassmann M, Ogunshola OO. 2011. J Physiol Cell Physiol 279:C935–944.
Astrocytes and pericytes differentially modulate blood-brain barrier Friedman A, Kaufer D, Heinemann U. 2009. Blood-brain barrier
characteristics during development and hypoxic insult. J Cereb Blood breakdown-inducing astrocytic transformation: novel targets for the
Flow Metab 31:693–705. prevention of epilepsy. Epilepsy Res 85:142–149.
Alvarez JI, Dodelet-Devillers A, Kebir H, Ifergan I, Fabre PJ, Terouz Gesuete R, Orsini F, Zanier ER, Albani D, Deli MA, Bazzoni G, et al.
S, et al. 2011. The Hedgehog pathway promotes blood-brain barrier 2011. Glial cells drive preconditioning-induced blood-brain barrier
integrity and CNS immune quiescence. Science 334:1727–1731. protection. Stroke 42:1445–1453.

T H E R O L E O F G L I A I N T H E F O R M AT I O N A N D F U N C T I O N O F T H E B L O O D -B R A I N B A R R I E R • 427
Hansson E, Westerlund A, Bjorklund U, Olsson T. 2008. mu-Opioid rat brain endothelial cells, pericytes and astrocytes. Neurochem Int
agonists inhibit the enhanced intracellular Ca(2+) responses in 54:253–263.
inflammatory activated astrocytes co-cultured with brain endothe- Nishioku T, Dohgu S, Takata F, Eto T, Ishikawa N, Kodama KB,
lial cells. Neuroscience 155:1237–1249. et al. 2009. Detachment of brain pericytes from the basal lam-
Hartmann C, Zozulya A, Wegener J, Galla HJ. 2007. The impact of ina is involved in disruption of the blood-brain barrier caused by
glia-derived extracellular matrices on the barrier function of cerebral lipopolysaccharide-induced sepsis in mice. Cell Mol Neurobiol
endothelial cells: an in vitro study. Exp Cell Res 313:1318–1325. 29:309–316.
Haseloff RF, Blasig IE, Bauer HC, Bauer H. 2005. In search of the astro- Nishitsuji K, Hosono T, Nakamura T, Bu G, Michikawa M. 2011.
cytic factor(s) modulating blood-brain barrier functions in brain cap- Apolipoprotein E regulates the integrity of tight junctions in an
illary endothelial cells in vitro. Cell Mol Neurobiol 25:25–39. isoform-dependent manner in an in vitro blood-brain barrier model.
Hayashi Y, Nomura M, Yamagishi S, Harada S, Yamashita J, Yamamoto J Biol Chem 286:17536–17542.
H. 1997. Induction of various blood-brain barrier properties in Nitta T, Hata M, Gotoh S, Seo Y, Sasaki H, Hashimoto N, et al.
non-neural endothelial cells by close apposition to co-cultured astro- 2003. Size-selective loosening of the blood-brain barrier in
cytes. Glia 19:13–26. claudin-5-deficient mice. J Cell Biol 161:653–660.
Hellström M, Gerhardt H, Kalen M, Li X, Eriksson U, Wolburg H, et al. Ohtsuki S, Yamaguchi H, Katsukura Y, Asashima T, Terasaki T. 2008.
2001. Lack of pericytes leads to endothelial hyperplasia and abnor- mRNA expression levels of tight junction protein genes in mouse
mal vascular morphogenesis. J Cell Biol 153:543–553. brain capillary endothelial cells highly purified by magnetic cell sort-
Hultman K, Bjorklund U, Hansson E, Jern C. 2010. Potentiating effect ing. J Neurochem 104:147–154.
of endothelial cells on astrocytic plasminogen activator inhibitor Oki K, Kaneko N, Kanki H, Imai T, Suzuki N, Sawamoto K, et al. 2010.
type-1 gene expression in an in vitro model of the blood-brain barrier. Musashi1 as a marker of reactive astrocytes after transient focal brain
Neuroscience 166:408–415. ischemia. Neurosci Res 66:390–395.
Kaur C, Sivakumar V, Zhang Y, Ling EA. 2006. Hypoxia-induced astro- Pekny M, Stanness KA, Eliasson C, Betsholtz C, Janigro D. 1998.
cytic reaction and increased vascular permeability in the rat cerebel- Impaired induction of blood-brain barrier properties in aortic
lum. Glia 54:826–839. endothelial cells by astrocytes from GFAP-deficient mice. Glia
Kooij G, Mizee MR, van Horssen J, Reijerkerk A, Witte ME, Drexhage 22:390–400.
JA, et al. 2011. Adenosine triphosphate-binding cassette transport- Plateel M, Teissier E, Cecchelli R. 1997. Hypoxia dramatically increases
ers mediate chemokine (C-C motif) ligand 2 secretion from reac- the nonspecific transport of blood-borne proteins to the brain.
tive astrocytes: relevance to multiple sclerosis pathogenesis. Brain J Neurochem 68:874–877.
134:555–570. Pottiez G, Duban-Deweer S, Deracinois B, Gosselet F, Camoin L,
Lee J, Lund-Smith C, Borboa A, Gonzalez AM, Baird A, Eliceiri BP. Hachani J, et al. 2011. A differential proteomic approach identifies
2009. Glioma-induced remodeling of the neurovascular unit. Brain structural and functional components that contribute to the dif-
Res 1288:125–134. ferentiation of brain capillary endothelial cells. J Proteomics 75:
Lee SW, Kim WJ, Choi YK, Song HS, Son MJ, Gelman IH, et al. 2003. 628–641.
SSeCKS regulates angiogenesis and tight junction formation in Reese TS, Karnovsky MJ. 1967. Fine structural localization of a
blood-brain barrier. Nat Med 9:900–906. blood-brain barrier to exogenous peroxidase. J Cell Biol 34:207–217.
Lehner C, Gehwolf R, Tempfer H, Krizbai I, Hennig B, Bauer HC, et al. Regina A, Morchoisne S, Borson ND, McCall AL, Drewes LR, Roux F.
2011. Oxidative stress and blood-brain barrier dysfunction under 2001. Factor(s) released by glucose-deprived astrocytes enhance glu-
particular consideration of matrix metalloproteinases. Antioxid cose transporter expression and activity in rat brain endothelial cells.
Redox Signal 15:1305–1323. Biochim Biophys Acta 1540:233–242.
Liebner S, Czupalla CJ, Wolburg H. 2011. Current concepts of Risau W. 1997. Mechanisms of angiogenesis. Nature 386:671–674.
blood-brain barrier development. Int J Dev Biol 55:467–476. Romanitan MO, Popescu BO, Spulber S, Bajenaru O, Popescu LM,
Liebner S, Fischmann A, Rascher G, Duffner F, Grote EH, Kalbacher Winblad B, et al. 2010. Altered expression of claudin family proteins
H, et al. 2000. Claudin-1 and claudin-5 expression and tight junc- in Alzheimer’s disease and vascular dementia brains. J Cell Mol Med
tion morphology are altered in blood vessels of human glioblastoma 14:1088–1100.
multiforme. Acta Neuropathol 100:323–331. Saunders NR, Knott GW, Dziegielewska KM. 2000. Barriers in the
Liedtke W, Edelmann W, Bieri PL, Chiu FC, Cowan NJ, Kucherlapati immature brain. Cell Mol Neurobiol 20:29–40.
R, et al. 1996. GFAP is necessary for the integrity of CNS white mat- Schroeter ML, Mertsch K, Giese H, Muller S, Sporbert A, Hickel B,
ter architecture and long-term maintenance of myelination. Neuron et al. 1999. Astrocytes enhance radical defence in capillary
17:607–615. endothelial cells constituting the blood-brain barrier. FEBS Lett 449:
Machein MR, Kullmer J, Fiebich BL, Plate KH, Warnke PC. 1999. 241–244.
Vascular endothelial growth factor expression, vascular volume, Seike T, Fujita K, Yamakawa Y, Kido MA, Takiguchi S, Teramoto N,
and, capillary permeability in human brain tumors. Neurosurgery et al. 2011. Interaction between lung cancer cells and astrocytes via
44:732–740; discussion 740–741. specific inflammatory cytokines in the microenvironment of brain
Merlini M, Meyer EP, Ulmann-Schuler A, Nitsch RM. 2011. Vascular metastasis. Clin Exp Metastasis 28:13–25.
beta-amyloid and early astrocyte alterations impair cerebrovascular Shen S, Zhang W. 2010. ABC transporters and drug efflux at the
function and cerebral metabolism in transgenic arcAbeta mice. Acta blood-brain barrier. Rev Neurosci 21:29–53.
Neuropathol 122:293–311. Sims DE. 1991. Recent advances in pericyte biology—implications for
Meyer J, Rauh J, Galla HJ. 1991. The susceptibility of cerebral endothelial health and disease. Can J Cardiol 7:431–443.
cells to astroglial induction of blood-brain barrier enzymes depends Spoerri PE, Grant MB, Gomez J, Vernadakis A. 1997. Endothelial cell
on their proliferative state. J Neurochem 57:1971–1977. conditioned media mediated regulation of glutamine synthetase
Mi H, Haeberle H, Barres BA. 2001. Induction of astrocyte differentia- activity in glial cells. Brain Res Dev Brain Res 104:205–208.
tion by endothelial cells. J Neurosci 21:1538–1547. Stenzel D, Lundkvist A, Sauvaget D, Busse M, Graupera M, van der
Nag S. 2011. Morphology and properties of astrocytes. In: Nag S (ed,), Flier A, et al. 2011. Integrin-dependent and -independent func-
The blood-brain and other neural barriers: reviews and protocols. tions of astrocytic fibronectin in retinal angiogenesis. Development
New York: Springer, pp. 69–100. 138:4451–4463.
Nakagawa S, Deli MA, Kawaguchi H, Shimizudani T, Shimono T, Suzuki H, Hasegawa Y, Ayer R, Sugawara T, Chen W, Sozen T, et al.
Kittel A, et al. 2009. A new blood-brain barrier model using primary 2011. Effects of recombinant osteopontin on blood-brain barrier

428 • FUNCTIONS OF NEUROGLIAL CELLS


disruption after subarachnoid hemorrhage in rats. Acta Neurochir Wolburg H. 1995. Orthogonal arrays of intramembranous particles: a
Suppl 111:231–236. review with special reference to astrocytes. J Hirnforsch 36:239–258.
Tontsch U, Bauer HC. 1991. Glial cells and neurons induce blood-brain Wolburg H, Wolburg-Buchholz K, Fallier-Becker P, Noell S, Mack AF.
barrier related enzymes in cultured cerebral endothelial cells. Brain 2011. Structure and functions of aquaporin-4-based orthogonal
Res 539:247–253. arrays of particles. Int Rev Cell Mol Biol 287:1–41.
Virgintino D, Girolamo F, Errede M, Capobianco C, Robertson D, Wolburg-Buchholz K, Mack AF, Steiner E, Pfeiffer F, Engelhardt B, et al.
Stallcup WB, et al. 2007. An intimate interplay between preco- 2009. Loss of astrocyte polarity marks blood-brain barrier impairment
cious, migrating pericytes and endothelial cells governs human during experimental autoimmune encephalomyelitis. Acta Neuropathol
fetal brain angiogenesis. Angiogenesis 10:35–45. 118:219–233.
Wagner S, Gardner H. 2000. Modes of regulation of laminin-5 produc- Yenari MA, Xu L, Tang XN, Qiao Y, Giffard RG. 2006. Microglia potenti-
tion by rat astrocytes. Neurosci Lett 284:105–108. ate damage to blood-brain barrier constituents: improvement by mino-
Wilhelm I, Fazakas C, Krizbai IA. 2011. models of the blood-brain bar- cycline in vivo and in vitro. Stroke 37:1087–1093.
rier. Acta Neurobiol Exp (Wars) 71:113–128. Zhang W, Smith C, Howlett C, Stanimirovic D. 2000. Inflammatory acti-
Winkler EA, Bell RD, Zlokovic BV. 2011. Central nervous system peri- vation of human brain endothelial cells by hypoxic astrocytes is medi-
cytes in health and disease. Nat Neurosci 14:1398–1405. ated by IL-1beta. J Cereb Blood Flow Metab 20:967–978.

T H E R O L E O F G L I A I N T H E F O R M AT I O N A N D F U N C T I O N O F T H E B L O O D -B R A I N B A R R I E R • 429
34.
CONTROL OF THE EXTRACELLULAR IONIC
ENVIRONMENT AND VOLUME
Eva Syková

A B B R E VI AT I O N S brain tissue volume. Later, it was shown by a number of differ-


ent techniques (rapid freezing, which preserves the ECS for
ADC apparent diffusion coefficient electron microscopy, the use of radiotracers and, in particular,
ADCW apparent diffusion coefficient of water studying the diffusion of ions using ion-selective microelec-
APP amyloid precursor protein trodes) that in the adult brain, the average ECS volume under
CNS central nervous system physiological conditions is about 18% to 24% of the total brain
CSPG chondroitin sulfate proteoglycan volume (Nicholson and Sykova 1998). Nevertheless, the ECS
Cx30 connexin 30 volume and composition change dynamically every second
Cx43 connexin 43 throughout one’s entire life during neuronal activity, develop-
DW-MRI diffusion-weighted magnetic resonance ment and aging as well as in pathological states. The size and
imaging geometry of the ECS affect the concentration, movement, and
ECM extracellular matrix clearance of substances that are released into it. The ECS is
ECS extracellular space also an important communication channel. Substances that
GABA gamma-aminobutyric acid are released by neurons and glia diffuse through the ECS and
ISM ion-selective microelectrode thus reach receptors located on nearby cells or synapses. This
[K+]e extracellular potassium concentration means of communication is called extrasynaptic or volume
mM millimolar transmission because the substances are diffusing through the
pHe extracellular pH volume of the ECS.
pHi intracellular pH
RVD regulatory volume decrease
TEA tetraethylammonium 2 E C S I O N I C A N D VO LU M E
TMA tetramethylammonium H O M E O S TA S I S
VT volume transmission
WHO World Health Organization Increases in the extracellular concentration of K+ ([K+]e),
decreases in extracellular Ca2+ and alkaline-acid shifts in extra-
cellular pH (pHe) accompany neuronal activity in different
1 T H E E X T R AC E L LU L A R S PAC E brain regions (Sykova, 1992). Glial cells play an important role
in the maintenance of ionic homeostasis.
The extracellular space (ECS) is the microenvironment of
neurons and glia, and its chemical and biophysical properties
2.1 T H E F U N C T I O N O F G L I A I N
are crucial for the proper functioning of neurons and also for
E X T R AC E L LU L A R P OTA S S IUM H O M EO S TA S I S
signal transmission. It is a porous structure with uneven pore
sizes (Sykova and Nicholson 2008). It includes ions, trans- Measurements with K+-selective microelectrodes revealed
mitters, metabolites, peptides, neurohormones, and other the local changes in extracellular K+ activity during sponta-
neuroactive molecules as well as various macromolecules of neous discharges (Sykova et al. 1974) as well as after stimula-
the extracellular matrix. Neurons, astrocytes, and oligoden- tion (Kriz et al. 1975; Svoboda and Sykova 1991). However,
drocytes release ions, neurotransmitters, and many other neu- even during intensive stimulation, [K+]e does not exceed a
roactive substances into the ECS, which then diffuse to the certain steady state, the so-called “ceiling” level, which is
nearby neuronal elements. The actual structure and volume of about 8 to 10 mM in the adult mammalian cortex and spinal
the ECS are therefore of crucial importance. cord (Czeh et al. 1981; Heinemann and Lux 1977; Kriz et al.
The first estimates of ECS volume were made in the sec- 1974; Sykova and Svoboda 1990). After the activity ends, the
ond half of the last century from data obtained by electron original [K+]e is quickly reestablished through redistribution
microscopy. However, the true ECS volume was altered by by several mechanisms localized particularly in neighboring
conventional preparation procedures, leading to the erroneous astrocytes (Fig. 34.1). Potassium ions are cleared from the ECS
conclusion that the ECS represents less than 5% of the total by glial cells through: (1) KCl uptake through Cl- channels

430
Ref.
– MRF
+ K+

ISM [K+]e
ion exchanger

action potentials

striatum spinal cord

5
+
[K ]
+ mM
+ K
K 3

7.3
pH
pH pH
7.4

light 60 s 100 Hz

Figure 34.1 Schema of a double-barreled potassium-selective microelectrode for measuring extracellular neuronal activity together with K+ changes
in the extracellular space. MRF: Spontaneous neuronal action potential bursts are accompanied by an increase in [K+]e in the rat mesencephalic
reticular formation. Striatum: Increases in [K+]e and pHe in the chick striatum as a result of light stimulation of the contralateral eye. Spinal cord:
Stimulation-evoked changes in [K+]e and pHe in the adult rat spinal cord. The change in pH was biphasic: the fast “initial” alkaline shift was followed
by a dominant acid shift. Note the poststimulation undershoots in both [K+]e and pHe. Adapted from Sykova and Chvatal 2000, with permission from
Elsevier.

activated by membrane depolarization (Walz and Hertz 1983; 2.2 T H E RO L E O F G L I A I N E X T R AC E L LU L A R


Walz and Hinks 1986); (2) the opening of Ca2+-activated K+ P H H O M EO S TA S I S
channels (MacVicar 1984); (3) Na+/K+-ATPase transport, Extracellular space pH changes—alkaline as well as acid
present on the membranes of both neurons and glial cells; shifts—result from both neuronal and glial cell activity. It was
and (4) K+ spatial buffering (Orkand et al. 1966) (Fig. 34.2). demonstrated that activity-related alkaline shifts are of neu-
Potassium spatial buffering is based on the high permeability ronal origin, whereas acid shifts are of glial origin ( Jendelova
of the glial membrane for K+ and astrocytic coupling into and Sykova 1991; Sykova 1992). During early development
the syncytium, wherein cells are interconnected by gap junc- when glia precursor cells dominate, glial homeostatic function
tions, allowing for the movement of K+, Ca2+, and other is incomplete, and activity-related pHe and [K+]e changes are
ions (Coles and Orkand 1983; Kettenmann et al. 1983). The substantially different from those in the mature central ner-
part of the syncytium that is depolarized by a local increase vous system (CNS) (Chvatal et al. 1995). Alkaline shifts dom-
in [K+]e acts as a cathode, whereas the distant regions with a inate in newborn rats with incomplete gliogenesis. During
normal membrane potential represent the anode. A current is development they become smaller and are finally overtaken
carried by K+ inside the glial syncytium and by Na+ and Cl- by the acid shifts characteristic of mature glial homeostatic
outside the syncytium in the ECS. In the regions with a nor- function ( Jendelova and Sykova 1991). Stimulation-evoked
mal extracellular K+ concentration and membrane potential, alkaline shifts are of neuronal origin because they are blocked
K+ returns back to the ECS. This redistribution of K+ does by the synaptic transmission blockers Mn2+ and Mg2+, whereas
not require energy and depends only on the existence of a K+ acid shifts are unaffected.
gradient. Gap junction proteins, connexin 30 (Cx30) and Changes of pHe and pHi (intracellular pH) are caused
connexin 43 (Cx43), mediate the extensive network of astro- by the movement of H+, OH- and their equivalents NH4+
cytes (see chapter 24). Gap junctional networking facilitates and HCO3- through the membranes of neurons and glia.
extracellular potassium and glutamate removal during synap- Some of the membrane transport mechanisms regulat-
tic activity through the modulation of the astroglial clearance ing pHe and pHi, such as Na+/H+ exchange (Astion et al.
rate and ECS volume (Pannasch et al. 2011). It has been shown 1989; Chesler 1987; Deitmer and Schlue 1987) or Na+/H+/
that the inactivation of the Cx30 and Cx43 genes in mice Cl–/HCO3– cotransport (Thomas 1977), are common to
increases hippocampal synaptic transmission and impairs both neurons and glial cells, whereas others are specific for
long-term synaptic plasticity. either neurons, for example voltage-dependent H+ channels

C O N T R O L O F T H E E X T R AC E L LU L A R I O N I C E N VI R O N M E N T A N D VO LU M E • 431
K+ homeostasis pH homeostasis
H2O
Na+
Na+ K+ Na+ Na+
K+ H+ H+
ATP
K+
ATP
Na-K pump
Na+ H+
Na cycle Cl– CO
Na+ H+ Na+ HCO3– 2
+
glia 1 Cl– HCO3– H2O
Na+
K+ spatial HCO3–
buffering Cl–
neuron mechanism HCO3– HCO3–
H2CO3
K+ K+
Cl–
neuron glia
Ca2+ Ca2+
H+
K+ H+
Lac–
Cl– KCI uptake
H+
Cl– +
HCO3–
NT Cl– HCO3–
glia 2 GABA GABA
H2O

extracellular space extracellular space

Figure 34.2 Schema of ECS K+ and pH Homeostasis. Note the differences in the membrane transport processes controlling ionic homeostasis in neu-
rons and glia. Potassium ions are cleared from the ECS by glial cells owing to KCl uptake through Cl– channels activated by membrane depolarization,
the opening of Ca2+-activated K+ channels, Na+/K+-ATPase transport, and K+ spatial buffering. Glial membrane transport mechanisms regulating pHe
and pHi are Na+/H+ exchange, Na+/H+/Cl–/HCO3– cotransport, voltage-dependent Na+- HCO3– cotransport, and H+-lactate extrusion.

(Meech and Thomas 1987; Sykova and Svoboda 1990), or decrease (RVD) (Mongin and Orlov 2001; Pasantes-Morales
for glia, such as voltage-dependent Na+- HCO3– cotransport et al. 2000) and is dependent on stretch-activated channels.
(Astion et al. 1991) or H+-lactate extrusion (Siesjo et al. 1985) The mechanisms of cell volume regulation are modulated
(see Fig. 34.2). The membrane transporters resulting in extra- by the intracellular concentration of Ca2+ (McCarthy and
cellular alkaline shifts (acid loaders) predominate in neurons, O’Neil 1992) and intracellular pH (Kempski et al. 1990).
whereas those resulting in extracellular acid shifts (acid extrud- In addition, the release of glutamate, taurine, aspartate, and
ers) prevail in glial membranes ( Jendelova and Sykova 1991). other amino acids from cells into the ECS is also involved in
Active neurons release K+, which accumulates in the ECS RVD (Kimelberg et al. 1990; Pasantes-Morales and Schousboe
and is taken up by glia. An alkaline shift is evoked in depolar- 1988). Recent data indicate an important role for aquaporin
ized glial cells, for example by the activation of Na+- HCO3– 4 in sensing osmolarity changes and in regulatory volume
cotransport. This alkaline shift in glial pHi causes an acid shift decrease. Aquaporin 4 is mainly expressed by astrocytes and
in pHe. Extracellular acidosis suppresses neuronal activity. may affect ECS homeostasis during pathological states such as
Glial swelling resulting in an ECS volume decrease leads to a the outcome of brain edema (Benfenati et al. 2011).
greater accumulation of ions and to a further decrease of neu- Astrocytic swelling is a major clinical problem after trau-
ronal excitability (see Fig. 34.4). This phenomenon has been matic brain injury. Moreover, various studies have demon-
termed a nonspecific feedback mechanism suppressing neu- strated that astrocytic swelling is an early event in numerous
ronal activity (Sykova, 1997). pathological states, accompanied by an elevation of [K+]e
and a decrease of extracellular osmolality (Kimelberg 1991;
Kimelberg et al. 1990). In isolated spinal cords of 4 to 21-day-
2.3 T H E RO L E O F A S T RO C Y T E S I N old rats, the application of an isotonic solution containing
E X T R AC E L LU L A R VO LU M E H O M E O S TA S I S
50 mM K+ or hypotonic solution (235 mmol/kg) evoked an
All ions cross the cell membrane in a hydrated form. This initial decrease in ECS volume of about 50% (Sykova et al.
transmembrane water movement leads to cellular (particu- 1999). Because the total water content remained stable, the
larly astrocytic) swelling and to compensatory ECS volume changes were attributed to cell swelling. The observed changes
shrinkage. An increased cell volume activates transport mech- in ECS volume were blocked in Cl– free solution and slowed
anisms that decrease the concentration of osmotically active down by furosemide and bumetanide, blockers of KCl uptake,
substances in cells, which results in a decrease in the volume suggesting the involvement of glial swelling. In animals older
of the swollen cells. This process is called regulatory volume than 10 days, during the continuous application of 50 mM K+

432 • FUNCTIONS OF NEUROGLIAL CELLS


or hypotonic solution, the ECS volume started to return to underlying mechanism of extrasynaptic volume transmis-
control values because of the shrinkage of previously swollen sion (VT). It has been shown that VT is far more common
cells caused by RVD (Cserr et al. 1991; Gullans and Verbalis than classical synaptic transmission. Synaptic transmission is
1993). The mechanisms of RVD are almost exclusively related a precise communication through fixed synapses, for exam-
to the homeostatic regulation of volume by glial function, ple cell to cell or one to one communication, whereas VT
because RVD is not observed in immature animals with communication involves a population of cells at the same
incomplete gliogenesis and is also blocked by the gliotoxin time (long-distance communication), that is, one to many
fluoroacetate (Sykova et al. 1999). (Fig. 34.3). This type of communication is the only way in
which nerve cells can communicate with glial cells, because
glia have no synapses. The existence of functional interac-
3 E X T R AC E L LU L A R ( VO LU M E ) tions between nerve cells without any synaptic contacts, “mis-
TR ANSMISSION matches” between release sites and the location of receptors,
and the widespread existence of high-affinity nonsynaptic
Neuroactive substances such as ions, neurotransmitters (see receptors led to the conclusion that VT is a distinct and impor-
chapter 17), neurohormones, neurotrophic factors and regu- tant alternative mode of signal transmission (Agnati et al.
latory cytokines (see chapter 22) and metabolites diffuse 1995; Fuxe and Agnati 1991; Kiss and Vizi 2001; Nicholson
through the ECS and affect nerve and glial cells distant from and Sykova 1998; Sykova 1997, 2001, 2003, 2004, Sykova and
their release site (Sykova, 1997). Diffusion in the ECS is the Nicholson 2008; Vizi 1980, 1984, 2000; Zoli et al. 1999).

EXTRASYNAPTIC “VOLUME” TRANSMISSION (VT)

A LONG-DISTANCE B SHORT-DISTANCE

neu presynaptic
g ron

volume fraction
tortuosity
uptake g
Glu K+
g
glia
g Ca2+
glia glia
GLT1/GLAST
glia
ATP

extracellular
space
postsynaptic cell
postsynaptic

C D
SPILLOVER SYNAPTIC PLASTICITY
Presynaptic
terminal New
synapse

Glutamate

Dendritic
spine Glutamate
Astrocyte receptor

Astrocyte
Extrasynaptic
receptors

Figure 34.3 Concept of Long- and Short-Distance Communication by Diffusion (i.e., Extrasynaptic Volume Transmission). A. The CNS architecture is
composed of neurons, axons, glial cells (glia), cellular processes (G), molecules of the extracellular matrix, and intercellular channels between cells. This
architecture slows the long-distance movement (diffusion) of substances in the brain, which is critically dependent on the ECS diffusion parameters
volume fraction (α), tortuosity (λ) and nonspecific uptake (k'). B. This synapse is tightly ensheathed by glial processes and the extracellular matrix,
forming perisynaptic nets. C. An open synapse is typical of volume transmission. It allows the escape of transmitters (e.g., glutamate, GABA) from
the synaptic cleft (spillover), diffusion through the ECS and binding to receptors on nearby synapses. This phenomenon is known as “cross-talk”
between synapses. D. The spillover may also lead to plastic changes, inducing the formation of new synapses or eliciting the rearrangement of astro-
cytic processes around the synapse. Adapted from Sykova, 2004; and Sykova and Nicholson, 2008, with permission from Elsevier and The American
Physiological Society.

C O N T R O L O F T H E E X T R AC E L LU L A R I O N I C E N VI R O N M E N T A N D VO LU M E • 433
Diffusion is characterized by the random Brownian motion changes in the ECS volume fraction, or detect changes that
of molecules. In comparison with diffusion in a free medium, are only partially related to ECS volume changes (Nicholson
described by Fick´s laws, diffusion in the ECS is restricted and Sykova 1998). An important noninvasive method that
by: (1) the size of the extracellular pores; (2) diffusion barri- can also be used in humans is measuring the ADC of water
ers such as membrane infoldings, fine neuronal and glial pro- (ADCW ) by diffusion-weighted magnetic resonance imaging
cesses, macromolecules of the extracellular matrix (ECM), and (DW-MRI), which reveals the inhomogeneous and aniso-
charged molecules; and (3) nonspecific cellular uptake. The tropic diffusion of water, similar to the diffusion of TMA+
diffusion parameters in the CNS were described by Nicholson (Mamata et al. 2002; Pierpaoli et al. 1996; Vorisek et al.
and Phillips (1981), who modified the original diffusion equa- 2002). It has been found that under some experimental con-
tions by introducing three diffusion parameters: (1) extra- ditions, a decrease in ADCW (i.e., lower diffusibility) can be
cellular volume fraction (α), sometimes also called porosity; related either to a decrease in α, typically during fast acute
(2) tortuosity (λ), representing the hindrances to the move- changes such as ischemia (Van der Toorn et al. 1996), or to
ment of molecules in the ECS that are absent in a free an increase in the number of diffusion barriers, represented
medium; and (3) nonspecific concentration-dependent or by an increase in λ without substantial changes in α such as
independent uptake, (k´) (see Fig. 34.3). The ECS volume occur during chronic states after injury (Vorisek et al. 2002).
fraction α is a dimensionless quantity defined as the ratio
between the volume of the ECS and the total volume of the
3.1 D I FF US I O N I N T H E EC S I S
tissue:
I N H O M O G E N EO US A N D A N I S OT RO P I C
α VECS/V Total
The ECS has a complicated and uneven geometry, which
It is now well established that the ECS of adult brain tissue depends on the type and number of cells in the local tissue,
represents about 20% to 25% of the total tissue volume, which the density and orientation of cellular processes, and the
means that α = 0.20–0.25. It is also evident that the ECS is actual macromolecular content of the tissue, particularly
not uniform; nevertheless, a study using quantum dot nano- that of the extracellular matrix. The ECS therefore cannot be
crystals estimated the average width of the ECS in the rat cor- homogeneous; its properties vary around different cells and
tex to be between 38nm and 64nm (Thorne and Nicholson also in different brain regions (Sykova and Nicholson 2008).
2006). Inhomogeneity as a result of different tissue structures affects
The free diffusion coefficient in the brain is reduced by both αandλfor example in the hippocampus inhomogene-
the presence of diffusion barriers by the tortuosity factor λ ity exists between the CA1 and CA3 layers and the dentate
(Nicholson and Sykova, 1998), which is defined as: gyrus (Vorisek and Sykova 1997a).
λ  D/ADC Whereas the volume fraction is a scalar quantity and
therefore has only one value in a given tissue, tortuosity is
where D is the diffusion coefficient in free medium and ADC a vector, so its values differ in anisotropic regions; that is,
is the apparent diffusion coefficient of a substance in the ner-
they differ along different axes in the tissue as a consequence
vous tissue. The tortuosity value, which reflects the number
of differences in the local structure (Fig. 34.5). Diffusion
and extent of diffusion barriers, is about 1.5 in healthy tissue,
anisotropy has been found in the white matter such as the
which means that diffusion in the brain is about 2.5× slower
than in a free medium. corpus callosum, as well as in the gray matter of the cerebel-
The advent of ion-selective microelectrodes (ISMs) made lum, striatum, hippocampus, and supraoptic nucleus (for
it possible to introduce a technique for diffusion measure- review, see Sykova and Nicholson 2008). Substances diffuse
ments in situ. Both real-time iontophoretic and also real-time more freely along axon bundles than across them, for example
pressure ejection methods use micropipettes to deliver mol- in the myelinated corpus callosum (Vorisek and Sykova 1997a)
ecules to which cell membranes are relatively impermeable or along astrocytic processes that are organized in parallel,
(tetramethyl- or tetraethylammonium—TMA+ or TEA+) such as those in the hippocampal dentate gyrus (Mazel
into the ECS. The concentration changes of these mol- et al. 1998; Sykova et al. 1998) (see Fig. 34.5). Glial cells can
ecules are sensed by ISMs at a known distance (Sykova and influence both α and λ under physiological as well as
Nicholson 2008), and the recorded diffusion curves serve for pathological conditions, a fact that is sometimes used for
mathematical analysis (see Figs. 34.4, 34.5, 34.6, and 34.7). diagnostic purposes, for example in demyelinating dis-
The values of α, ADCTMA, λ and k´ are extracted by a nonlin- eases. Besides α and λ diffusion in the ECS is affected by
ear curve-fitting simplex algorithm operating on the diffusion nonspecific, concentration-dependent uptake (kc), bet-
curve (Nicholson and Phillips 1981). This is the most suitable ter characterized as the irreversible loss of a given sub-
method for measuring the absolute values of the ECS diffu- stance across the blood-brain-barrier or owing to its
sion parameters and their dynamic changes in nervous tissue binding to receptors, enzymatic degradation or cellular
in vitro as well as in vivo. uptake, for example to astrocytes. In many cases substances
The other methods used to study ECS volume fraction released extrasynaptically such as glutamate and GABA
and tortuosity are less comprehensive because they can either (see chapters 38 and 41) are transported by energy-dependent
measure only one of the parameters, determine only relative uptake systems that obey nonlinear kinetics.

434 • FUNCTIONS OF NEUROGLIAL CELLS


A Resting state C Δ[K+]e
7
5 [K+]
Diffusion curves mM
Iontophoresis TMA+-ISM [TEA+]e 3
α = 0.20 10
λ = 1.55 I = 40 nA 30 Hz
+
5 [TEA ]
mM
0
[TMA+]
TMA+ 7.2
pH
80 nA pHe 7.4
5 min
30 Hz 60 s

B Neuronal activity D

neuron pHe
α = 0.12
glia λ = 1.70

pHi excitability

[TMA+] HCO3–
TMA + Na+ –
Cl +
K
H2O K+
80 nA
GLIA NEURON

Figure 34.4 A,B. Experimental setup, TMA+ diffusion curves and typical ECS diffusion parameters α (volume fraction) and λ (tortuosity) obtained
in the brain before (A, resting state) and during (B, activity) neuronal activity, evoked by the same iontophoretic current of 80 nA. A TMA+-selective
double-barreled ISM was glued to a bent iontophoresis microelectrode. The ECS in the unstimulated brain is 20% (volume fraction α = 0.20) and λ is
1.55. The ECS is smaller because of cell swelling during stimulation-evoked neuronal activity, therefore the diffusion curves are larger. The ECS volume
decreased to about 12% (α = 0.12), whereas λ increased to 1.70. C. The effect of repetitive stimulation of afferent input on TEA+ diffusion curves,
extracellular K+ and pH in the isolated frog spinal cord. Higher diffusion curves indicate a decrease in α due to cell swelling outlasting the 1 min
repetitive stimulation (30 Hz) of the sciatic nerve for 30 minutes. Note the extracellular K+ increase and the acid shift evoked by stimulation. The time
course of the acid shift correlates with the decrease in α. D. Schematic of the mechanism of nonspecific feedback suppressing neuronal excitability.
Active neurons release K+, which accumulates in the ECS and depolarizes glial cells. This causes an alkaline shift in glial pHi and an acid shift in pHe.
Extracellular acidosis further suppresses neuronal activity. Transmembrane ionic movements result in glial swelling, an ECS volume decrease and the
increased accumulation of ions and neuroactive substances in the ECS. From Sykova 2003, with permission from John Wiley and Sons.

4 THE ECS AROUND ASTROCY TES hypotonic solution resulted in a 306% increase in K+ accu-
AND OLIGODENDROCYTES mulation around astrocytes but only a 110% increase around
oligodendrocytes. The different effects of an osmotic chal-
The existence of regional differences in the extracellular space lenge on tail currents in astrocytes and oligodendrocytes can
volume around glial cells affects the membrane currents gener- be explained by a more “compact” space around oligodendro-
ated in response to voltage steps applied through a patch-clamp cytes. As tail currents appear in oligodendrocytes during their
pipette, because of K+ accumulation just outside the cell. In maturation and correspond temporally with the myelination
spinal cord oligodendrocytes the currents decay and large tail of the tissue (Chvatal et al. 1997), we can assume that the dif-
currents appear after the offset of the voltage command (Berger fusion barriers around oligodendrocytes are formed mainly by
et al. 1991; Chvatal et al. 1999). The current decay and the tail myelin sheaths or by extracellular matrix molecules produced
currents are related to the accumulation of K+ in the vicinity by mature oligodendrocytes.
of oligodendrocytes because of barriers that prevent the fur-
ther diffusion of K+ that has escaped from the cell. Because
5 D I F F U S I O N PA R A M ET E R S I N
glial cells are exclusively permeable for K+, knowing the val-
P H YS I O L O G I C A L S TAT E S
ues of the tail current and the reversal potential, the Nernst
equation can be used to calculate the value of [K+]e in the
5.1 D EV E L O PM E N T
vicinity of the cell membrane. Osmotically induced cell swell-
ing or cell shrinkage has revealed that the extracellular space During development, the diffusion parameters in the CNS
in the close vicinity of astrocytes is larger than that around are different from those found in adulthood. In rats, the
oligodendrocytes (Vargova et al. 2001). The application of a volume fraction decreases throughout the animal’s entire

C O N T R O L O F T H E E X T R AC E L LU L A R I O N I C E N VI R O N M E N T A N D VO LU M E • 435
A Young Aged

GFAP GFAP 200 μm

C D

α = 0.27 α = 0.16
Δ [TMA+] = 1 mM

λx = 1.50 λx = 1.53
λy = 1.60 λy = 1.55
λz = 1.70 λz = 1.59

60 s 60 s
(I = 80 nA) (I = 80 nA)

E F >1000
900

800
700
600
500
400
ADCW = 631 μm s 2 –1 2 –1
ADCW = 452 μm s <300
ADCW = [μm2s–1]

Figure 34.5 Structural Changes in the Hippocampus Gyrus Dentatus Region of Young and Aged Rats. A. Astrocytes stained for GFAP in a young adult
rat; note the radial organization of the astrocytic processes between the pyramidal cells (not stained). B. In an aged rat, the radial organization of the
astrocytic processes is lost. C. Anisotropic diffusion in the dentate gyrus of a young adult rat. TMA+ diffusion curves (concentration-time profiles)
were measured along three orthogonal axes: x—mediolateral, y—rostrocaudal, z—dorsoventral. The slower rise in the z than in the y direction and in
the y than in the x direction indicates a higher tortuosity and more restricted diffusion. The amplitude of the curves shows that the TMA+ concentra-
tion, at approximately the same distance from the tip of the iontophoresis electrode, is much higher along the x-axis than along the y-axis and even
higher than along the z-axis (lx, ly, lz). Note that the actual ECS volume fraction α can be calculated only when measurements are done along the x, y,
and z axes. D. Anisotropy is almost lost in an aged rat. E,F. Pseudocolor images showing typical ADCW maps of a young (E) and an aged
(F) rat. The scale at right shows the relation between the intervals of the ADCW values and the colors used for visualization Adapted from Sykova
2003, with permission from John Wiley and Sons.

life. A steep fall occurs in the first 2 postnatal weeks, during The connection between gliogenesis and the ECS diffusion
which α is reduced by approximately half in the rat cortex parameters is probably most evident in the white matter.
and corpus callosum (Lehmenkuhler et al. 1993; Prokopova The period of the gradual myelination of the corpus callo-
et al. 1997; Vorisek and Sykova 1997a). This decrease can sum closely corresponds with the decrease in the extracellu-
be attributed to changes in ECM composition, neuronal lar volume fraction. Moreover, the tortuosity values, which
migration, the development of dendritic arborization, rapid during the first postnatal week are the same along all three
myelination, and the proliferation of glia. The larger volume orthogonal axes (i.e., diffusion is isotropic) start to differ
fraction in immature tissue (0.35 to 0.40) slows down any during the second week—a period of intensive myelination.
increase in the concentration of neuroactive substances to After myelination is fully completed at the end of the third
neurotoxic levels and represents a protective factor in the postnatal week, diffusion in the corpus callosum is strictly
immature brain during pathological states such as ischemia anisotropic with preferential diffusion along the myelinated
(Vorisek and Sykova 1997b) or epilepsy (Kilb et al. 2006). fibers (Vorisek and Sykova 1997a).

436 • FUNCTIONS OF NEUROGLIAL CELLS


5.2 AC T I VIT Y-R E L AT E D C H A N G E S Neurons interact both by synapses as well as by the diffusion
of molecules and neuroactive substances in the ECS. Neurons
Because the ions crossing the cell membrane during neuronal
also release ions and other substances in synapses and from the
activity are accompanied by water molecules, their transmem-
extrasynaptic regions of their membranes. Since glial cells have
brane transport causes cell swelling and compensatory ECS
no synapses, neuron–glia communication is only achieved by
shrinkage. Repetitive electrical stimulation of the dorsal root
diffusion in the ECS, and glia release substances only nonsyn-
of the rat or frog spinal cord causes a substantial increase in
aptically. This type of extrasynaptic communication acts over
[K+]e and a decrease in α from about 0.24 to between 0.17
both short and long distances (see Fig. 34.3) and provides a
and 0.12, that is, a decrease of 30% to 50% (see Fig. 34.4).
mechanism for synchronizing the activity of a population of
A 20% to 50% decrease in ECS volume also occurs after
neurons and long-range information processing in functions
injury of the ipsilateral hind paw evoked by a subcutaneous
such as vigilance, hunger, sleep, chronic pain, depression, lac-
injection of turpentine or after a thermal injury that produces
tation, long-term potentiation and depression, and memory
chronic pain and long-lasting interneuronal activity (Svoboda
formation.
and Sykova 1991). The changes in the ECS diffusion param-
Synaptic transmission, which is characterized by fixed syn-
eters persist after stimulation ends (for 30 minutes after 1
aptic contacts, usually allows only one to one cell communi-
minute of electrical stimulation or even 120 minutes after 3
cation. The classical concept of a tripartite synapse (Haydon
minutes of stimulation), suggesting long-term glial swell-
2001) involves a presynaptic terminal, a postsynaptic element,
ing and subsequent long-term changes in neuronal excitabil-
and astrocytes (see chapter 39). Certain synapses (“private”
ity, neuron–glia communication, and extrasynaptic volume
synapses) or even whole neurons are tightly ensheathed by
transmission.
astrocytic processes and by extracellular matrix molecules
produced by astrocytes; these components form perineuronal
5.3 L AC TAT I O N nets (Celio et al. 1998). Other synapses are left more “open,”
and those can be easily reached by diffusible signals in the ECS
In rat males and virgin females, the hypothalamic supraoptic
(Sykova 2001). Transmitters such as glutamate and GABA
nucleus is an anisotropic structure with a relatively large extra-
can thus escape from a nearby synapse and reach the neigh-
cellular volume of 32% (Piet et al. 2004). The retraction of
boring one. This frequent phenomenon is called transmitter
glial processes toward the basal lamina under specific physi-
spillover and results in synaptic “cross-talk” (Asztely et al.
ological conditions, such as lactation or dehydration, leads to a
1997; Kullmann et al. 1996). Cross-talk could be involved
significant reduction in the astrocytic coverage of the magno-
in long-term potentiation and long-term depression, which
cellular neurons (Hatton 1997; Theodosis and Poulain 1993)
involve both ionotropic and metabotropic receptors (Isaacson
(see also chapter 41). A relative absence of glial processes
et al. 1993; Piet et al. 2004) (see Fig. 34.3).
around glutamatergic synapses results in the insufficient clear-
ance of glutamate and thus increases its level in the ECS (Oliet
et al. 2001). During lactation, the ECS volume decreases to
about 20%, and the retraction of astrocytic processes results 7 E C S D I F F U S I O N PA R A M ET E R S I N
in the disappearance of diffusion barriers. Facilitated diffu- PAT H O L O G I C A L S TAT E S
sion accompanied by the retraction of astrocytic processes
results in glutamate-mediated cross-talk between synapses and Many pathological states that are accompanied by structural
enhanced heterosynaptic depression of GABA-ergic transmis- changes in the neuronal and astrocytic populations, changes
sion (Piet et al. 2004). Depressing the inhibitory effect of in the ECM, or the deposition of macromolecules such as
GABA-ergic neurons can increase hormone release during βamyloid substantially affect the diffusion parameters in the
suckling and thus represents one of the mechanisms regulat- brain and therefore information processing as well. Astrocytic
ing milk ejection in the rat supraoptic nucleus. rebuilding and nerve cell loss are typical features of damaged
tissue. Brain injury of any kind elicits reactive gliosis, involv-
ing both the hyperplasia and hypertrophy of astrocytes. Active
6 A S T R O C Y T E S , T H E T R I PA RT I T E astrocytes change their morphology; their processes become
SY N A P S E A N D D I F F U S I O N PA R A M ET E R S shorter and thicker, and qualitative and quantitative changes
in their production of extracellular matrix molecules can be
The depolarization of astrocytes leads to the activation of ion detected. Changes in the ECS diffusion parameters have been
channels, second messengers, and intercellular metabolic path- found during pathological states such as ischemia, epilepsy,
ways and subsequently to changes in glial volume, structural injury, hydrocephalus, multiple sclerosis, Parkinson’s disease,
changes of the astrocytic processes and, eventually, to the pro- Alzheimer’s disease, and tumors.
duction of extracellular matrix molecules. Therefore, in addi-
tion to their role in the maintenance of extracellular ionic and 7.1 I S C H E M I A/A N OX I A , B R A I N I N J U RY
volume homeostasis, glial cells may play an important role in A N D A S T RO G L I O S I S
regulating synaptic as well as extrasynaptic volume transmis-
sion. Signal transmission in the brain results from the activation CNS ischemia, anoxia or hypoxia is accompanied by large
of receptors located in synapses or situated extrasynaptically. ionic shifts in the ECS, for example an increase in [K+]e to 50

C O N T R O L O F T H E E X T R AC E L LU L A R I O N I C E N VI R O N M E N T A N D VO LU M E • 437
to70 mM and an acid shift to 6.4 to 6.6. The ionic changes are processes including a decreased number and efficacy of syn-
accompanied by the redistribution of water from the ECS to apses, a decrease in transmitter release, neuronal loss, astro-
intracellular compartments. Therefore, both neurons and glia gliosis, demyelination, deposits of βamyloid, changes in the
swell, and ECS volume decreases from about 20% (α = 0.2) to extracellular matrix and others, resulting in behavioral changes
as low as 5% of total tissue volume (α = 0.05), while tortuosity and a cognitive deficit, particularly memory and learning
(λ increases fromabout 1.5 to 2.0 to 2.2 (Homola et al. 2006; impairment. These changes affect the biophysical and diffu-
Sykova et al. 1994; Vorisek and Sykova 1997b). A decrease sion properties of the ESC in the cortex, corpus callosum and
in ADCW is revealed by DW-MRI (Sykova and Nicholson hippocampus of senescent rats (CA1, CA3 and dentate gyrus).
2008). In experiments in which the animals are resuscitated In all three brain regions, the mean ECS volume fraction α is
and recover to normoxic conditions, the diffusion parameters significantly reduced in aged rats compared to young adults,
return to control values, and the volume fraction often reaches and the anisotropy typical of the hippocampus decreases
25% to 30% owing to the formation of postischemic edema. (Mazel et al., 1998). Morphological changes observed in the
In models of brain injury, changes in α and λ are often inde- hippocampal tissue include the disorganization of astrocytic
pendent and result from the structural remodeling of the tissue processes (Fig. 34.5) and a loss of the extracellular matrix mol-
(cell death, astrogliosis) (Roitbak and Sykova 1999; Vorisek et al. ecules that form perineuronal nets, for example fibronectin
2002). The formation of gliotic tissue caused by local brain dam- and chondroitin sulfate proteoglycan (CSPG). This accounts
age is responsible for a long-lasting increase of diffusion barriers for both the decrease in α and the disappearance of anisot-
in the ECS, preventing or slowing down the diffusion of neu- ropy. Rats with severe morphological changes also display a
roactive substances and growth factors and leading to impaired significant learning impairment that closely correlates with
extrasynaptic transmission, cell-to-cell communication, and changes in the ECS diffusion parameters (Sykova et al., 2002).
regenerative processes. In the formation of diffusion barriers, It is therefore reasonable to assume that diffusion anisotropy,
not only are hypertrophied astrocytic processes involved, but which leads to a certain degree of specificity in extrasynaptic
also the enhanced production of extracellular matrix molecules. communication by channeling the flux of substances in a pref-
Changes in the diffusion parameters in areas remote from the erential direction, may also play an important role in memory
site of injury possibly lead to functional deficits. formation and learning (Mazel et al., 1998; Sykova et al., 2002;
Wieshmann et al., 1999). The decrease in ECS volume during
aging not only impairs extrasynaptic transmission in the cor-
7.2 D E MY E L I NAT I N G D I S E A S E S
tex and hippocampus, but is also responsible for the greater
Changes in the ECS diffusion parameters, including a loss susceptibility of the aged brain to pathological insults, the
of the anisotropy typical of white matter, have been found poorer outcome of clinical therapy and limited regeneration.
in patients with demyelinating diseases such as multiple scle- Changes in the ECS diffusion parameters have also been
rosis. In an animal model of multiple sclerosis, experimental found in transgenic APP23 mice—a model of Alzheimer’s
autoimmune encephalomyelitis, typical morphological fea- disease. The deposition of β-amyloid is associated with an
tures of multiple sclerosis can be observed in the CNS tissue, increase in ECS volume fraction and tortuosity (Fig. 34.6).
including demyelination, an inflammatory reaction, astroglio- It is assumed that the volume fraction occupied by extracel-
sis, blood-brain-barrier damage, and clinical signs of paraly- lular amyloid plaques is not included in the value of α. The
sis (see chapter 61). Diffusion measurements have revealed deposition of macromolecules or ECS matrix can thus prevent
an increase in ECS volume fraction α and a decrease of the ECS volume fraction decrease normally observed in aging
tortuosity in both the white and the gray matter of the spinal animals (Sykova et al. 2005a). Changes in the ECS diffusion
cord. There was a close correlation between the changes in the parameters have been found in mice lacking tenascin-R, a large
ECS diffusion parameters and the manifestation of clinical extracellular glycoprotein (Sykova et al. 2005b). Decreases in
signs (paraparesis, paraplegia), which were preceded and out- both α and λ were found in the entire cortex and hippocam-
lasted by astrogliosis and an inflammatory reaction (Simonova pus in the knockout mice as compared to wild-type animals.
et al. 1996). Measurements using DW-MRI showed a reduction in ADCW
Changes in ECS diffusion have also been found during in the cortex, hippocampus, and thalamus in the knockouts
acute inflammation. In an experimental model, inflammation (see Fig. 34.6).
was evoked by intracerebral inoculation with a weakly patho- Extracellular matrix molecules play a role in tissue cyto-
genic strain of Staphyloccocus aureus. The evoked acute inflam- architecture by maintaining the optimal size of the intercel-
mation and an increase in blood-brain barrier permeability in lular spaces (Sykova et al. 2005b). This role of the ECM in
the abscess region resulted in a rather mild increase in ECS determining the ECS volume can be demonstrated by the
volume fraction and a lower tortuosity (Lo et al. 1993). changes seen during aging, when a decrease in the amount of
CSPG and fibronectin in the hippocampus of old rats with
7.3 AG I N G A N D A L Z H E I M E R’S D I S E A S E
a severe learning disability coincides with a decrease in ECS
volume (Sykova et al. 2002). The explanation for the rela-
Aging is a normal physiological process of the organism that tionship between the size of the ECS and the ECM content
has many features in common with degenerative diseases of the is simple: the side chains of ECM macromolecules, especially
CNS, including Alzheimer’s disease. Both aging and degener- those of glycosaminoglycans, carry a large number of nega-
ative brain diseases are accompanied by various pathological tively charged groups that repel one another. Concomitantly,

438 • FUNCTIONS OF NEUROGLIAL CELLS


Tenascin R-/- control - aged

α = 0.12 α = 0.11
λ = 1.53 λ = 1.43

1 mM TMA+
α = 0.19

1 mM TMA+
λ = 1.63 α = 0.23
λ = 1.60
APP23
Tenascin R+/+

60 s 60 s
(I = 180 nA) (I = 180 nA)

Tenascin R-/- aged

ADCW = 0.61 ADCW = 0.45

Tenascin R+/+ APP23

ADCW = 0.52 ADCW = 0.55

<0.3 0.4 0.5 0.6 0.7 0.8 0.9 >1.0


ADCW × 105cm2s–1

Figure 34.6 (Left) TMA+ diffusion curves with the corresponding values of volume fraction (α) and tortuosity (λ) obtained in the cortex and
pseudocolor ADCW maps obtained throughout the whole brain of tenascin-R knockout (-/-) and control (+/+) mice. (Right) TMA+ diffusion curves
with the corresponding values of volume fraction (α) and tortuosity (λ) obtained in the cortex and pseudocolor ADCW maps obtained throughout
the whole brain of aged control mice and aged APP23 mice (17–25 months old); the APP23 mice showed excessive extracellular amyloid deposition.
The larger ECS volume fraction demonstrates that diffusion from any given source will lead to a lower concentration of the diffusing substance in the
surrounding tissue and a smaller action radius in APP23 mice than in age-matched controls. The scale at the bottom shows the relation between the
intervals of the ADCW values and the colors used for visualization. Adapted from Sykova et al., 2005a and Sykova et al., 2005b, with permission from
the National Academy of Sciences USA and John Wiley and Sons.

they attract osmotically active cations such as Na+, causing a manifested in glial tumors. Studies using human glioma sam-
large amount of water to be drawn into the matrix. This cre- ples obtained from patients during surgery showed that the
ates a turgor that enables the matrix to withstand compres- migratory abilities of tumor cells might be strongly affected by
sive forces and leads to the expansion of the ECS (Alberts changes in the ECS volume fraction and the content of ECM
et al. 1994). molecules in tumors (Vargova et al. 2003; Zamecnik et al.
2004). The tumor slices were histopathologically classified
according to WHO grading (WHO grade I-IV) following
7.4 G L I A L T U MO R S
diffusion measurements, and the changes in the values of the
The impact of glial remodeling and rebuilding on the biophys- ECS diffusion parameters correlated with tumor malignancy.
ical properties and diffusion parameters of the ECS is also The proliferative activity of the tumors was assessed by the

C O N T R O L O F T H E E X T R AC E L LU L A R I O N I C E N VI R O N M E N T A N D VO LU M E • 439
labeling indices MIB-1 and anti-topo-II α. The measurements and a number of transport mechanisms on the glial mem-
showed that the proliferative activity and malignancy grade brane (Sontheimer et al. 1989) allow for the maintenance of
of astrocytomas are directly proportional to increasing val- ionic and volume homeostasis, which in turn affect neuronal
ues of α and λ (Fig. 34.7). In the cellular regions of the most function. A link between ionic and volume changes has been
malignant glioblastomas (WHO grade IV), tortuosity was proposed in a model of a nonspecific feedback mechanism
about 1.8 and α was twice as high (0.44) as in normal cortex suppressing neuronal activity (Sykova 1997). Glial cells con-
(Vargova et al. 2003). The increase in α and λ in high-grade trol not only the ionic composition of the ECS, but also its
tumors strongly correlated with an increased accumulation biophysical properties that determine the movement of ions,
of ECM molecules, particularly of tenascin and vitronectin neuroactive substances, neurohormones, growth factors, and
(Zamecnik et al. 2004). It has been shown that the overex- metabolites. Changes in the ECS diffusion parameters have
pression of certain types of ECM molecules corresponds with a strong impact on modulating both synaptic and extrasyn-
tumor malignancy (Camby et al. 2002; Hayen et al. 1999; aptic volume transmission, the synchronization of neuronal
Zhang et al. 1998), as these molecules may serve as “ropes for activity in complex functions, and neuron–glia communica-
climbing” for migrating tumor cells and thus facilitate their tion. Astrocytes are highly sensitive to stimuli accompanying
infiltration into the surrounding tissue. In addition, a large both physiological and pathological states such as increased
ECM content holds cells apart and thus creates a larger space potassium levels, osmolality changes, neuromediators, or sig-
for migrating cells. nals from damaged tissue, and promptly react by cell swelling,
proliferation, and hypertrophy. The production of extracellu-
lar matrix molecules by activated astrocytes further hinders
8 S U M M A RY A N D P E R S P E C T I VE S already impaired diffusion and contributes to an increase in
tortuosity. Astrocytes can thus be viewed as active elements
It is evident that glial cells play an important role in differ- in signal transmission as they affect both synaptic as well as
ent brain functions. Voltage- and ligand-activated channels extrasynaptic volume transmission.

TMA+ - iontophoresis
Human cortex:
α = 0.24
[TMA+]e λ = 1.45
k′ = 3.2 × 10–3 s–1
Δ [TMA+] = 0.5 mM

Glioblastoma:
α = 0.37
TMA+ - ISM λ = 1.86
k′ = 9.3 × 10–3 s–1

60 s
(I = 200 nA)

Cortex Glioblastoma

100 μm

Figure 34.7 Schema of the Experimental Arrangement for Measuring the ECS Diffusion Parameters in a Glioblastoma. A TMA+-selective
double-barreled microelectrode was glued to a bent iontophoresis microelectrode. Diff usion curves were recorded in acute brain slices obtained
during neurosurgery. Typical diffusion curves in the normal cortex and in the glioblastoma show that the volume fraction (α) and tortuosity
(λ) were increased in the tumor. This increase correlated with the increased deposition of extracellular matrix and with the mitotic index of the
tumor cells.

440 • FUNCTIONS OF NEUROGLIAL CELLS


AC K N OW L E D G M E N T S Hatton GI. 1997. Function-related plasticity in hypothalamus. Annu
Rev Neurosci 20:375–397.
Haydon PG. 2001. GLIA: listening and talking to the synapse. Nature
The author is grateful for the support provided by the Grant Rev Neurosci 2:185–193.
Agency of the Czech Republic (309/09/1597). Hayen W, Goebeler M, Kumar S, Riessen R, Nehls V. 1999. Hyaluronan
stimulates tumor cell migration by modulating the fibrin fiber archi-
tecture. J Cell Sci 112:2241–2251.
Heinemann U, Lux HD. 1977. Ceiling of stimulus induced rises in extra-
REFERENCES cellular potassium concentration in the cerebral cortex of cat. Brain
Res 120:231–249.
Agnati LF, Zoli M, Stromberg I, Fuxe K. 1995. Intercellular communica- Homola A, Zoremba N, Slais K, Kuhlen R, Sykova E. 2006. Changes in dif-
tion in the brain: Wiring versus volume transmission. Neuroscience fusion parameters, energy-related metabolites and glutamate in the rat
69:711–726. cortex after transient hypoxia/ischemia. Neurosci Lett 404:137–142.
Alberts B, Bray D, Lewis J, Raff M, Roberts K, Watson JD. 1994. Molecular Isaacson JS, Solis JM, Nicoll RA. 1993. Local and diff use synaptic actions
biology of the cell. New York: Garland Science Publishing. of GABA in the hippocampus. Neuron 10:165–175.
Astion ML, Chvatal A, Orkand RK. 1989. Na+/H+ exchange in glial Jendelova P, Sykova E. 1991. Role of glia in K+ and pH homeostasis in the
cells of Necturus optic nerve. Neurosci Lett 107:167–172. neonatal rat spinal cord. Glia 4:56–63.
Astion ML, Chvatal A, Orkand RK. 1991. Further studies of electro- Kempski O, Staub F, Jansen M, Baethmann A. 1990. Molecular mecha-
genic Na+/HCO3- cotransport in glial cells of Necturus optic nerve: nisms of glial cell swelling in acidosis. Adv Neurol 52:39–45.
regulation of pHi. Glia 4:461–468. Kettenmann H, Orkand RK, Schachner M. 1983. Coupling among
Asztely F, Erdemli G, Kullmann DM. 1997. Extrasynaptic glutamate identified cells in mammalian nervous system cultures. J Neurosci
spillover in the hippocampus: dependence on temperature and the 3:506–516.
role of active glutamate uptake. Neuron 18:281–293. Kilb W, Dierkes PW, Sykova E, Vargova L, Luhmann HJ. 2006.
Benfenati V, Caprini M, Dovizio M, Mylonakou MN, Ferroni S, Ottersen Hypoosmolar conditions reduce extracellular volume fraction and
OP, et al. 2011. An aquaporin-4/transient receptor potential vanil- enhance epileptiform activity in the CA3 region of the immature rat
loid 4 (AQP4/TRPV4) complex is essential for cell-volume control in hippocampus. J Neurosci Res 84:119–129.
astrocytes. Proc Natl Acad Sci U S A 108:2563–2568. Kimelberg HK. 1991. Swelling and volume control in brain astroglial
Berger T, Schnitzer J, Kettenmann H. 1991. Developmental changes cells. In: Giles RA (ed.), Advances in comparative and environmental
in the membrane current pattern, K+ buffer capacity, and physiology. Berlin: Springer-Verlag.
morphology of glial cells in the corpus callosum slice. J Neurosci 11: Kimelberg HK, Goderie SK, Higman S, Pang S, Waniewski RA. 1990.
3008–3024. Swelling-induced release of glutamate, aspartate, and taurine from
Camby I, Belot N, Lefranc F, Sadeghi N, de Launoit Y, Kaltner H, et al. astrocyte cultures. J Neurosci 10:1583–1591.
2002. Galectin-1 modulates human glioblastoma cell migration Kiss JP, Vizi ES. 2001. Nitric oxide: a novel link between synaptic and
into the brain through modifications to the actin cytoskeleton and nonsynaptic transmission. Trends Neurosci 24:211–215.
levels of expression of small GTPases. J Neuropathol Exp Neurol Kriz N, Sykova E, Ujec E, Vyklicky L. 1974. Changes of extracellular
61:585–596. potassium concentration induced by neuronal activity in the spinal
Celio MR, Spreafico R, De Biasi S, Vitellaro-Zuccarello L. 1998. cord of the cat. J Physiol (Lond) 238:1–15.
Perineuronal nets: past and present. Trends Neurosci 21:510–515. Kriz N, Sykova E, Vyklicky L. 1975. Extracellular potassium changes in
Chesler M. 1987. pH regulation in the vertebrate central nervous system: the spinal cord of the cat and their relation to slow potentials,
microelectrode studies in the brain stem of the lamprey. Can J Physiol active transport and impulse transmission. J Physiol (Lond) 249:
Pharmacol 65:986–993. 167–182.
Chvatal A, Anderova M, Ziak D, Sykova E. 1999. Glial depolariza- Kullmann DM, Erdemli G, Asztely F. 1996. LTP of AMPA and NMDA
tion evokes a larger potassium accumulation around oligodendro- receptor-mediated signals: evidence for presynaptic expression and
cytes than around astrocytes in gray matter of rat spinal cord slices. extrasynaptic glutamate spill-over. Neuron 17:461–474.
J Neurosci Res 56:493–505. Lehmenkuhler A, Sykova E, Svoboda J, Zilles K, Nicholson C. 1993.
Chvatal A, Berger T, Vorisek I, Orkand RK, Kettenmann H, Sykova E. Extracellular space parameters in the rat neocortex and subcortical
1997. Changes in glial K+ currents with decreased extracellular vol- white matter during postnatal development determined by diff usion
ume in developing rat white matter. J Neurosci Res 49:98–106. analysis. Neuroscience 55:339–351.
Chvatal A, Pastor A, Mauch M, Sykova E, Kettenmann H. 1995. Distinct Lo WD, Wolny AC, Timan C, Shin D, Hinkle GH. 1993. Blood-brain
populations of identified glial cells in the developing rat spinal cord barrier permeability and the brain extracellular space in acute cere-
slice: ion channel properties and cell morphology. Eur J Neurosci bral inflammation. J Neurol Sci 118:188–193.
7:129–142. MacVicar BA. 1984. Voltage-dependent calcium channels in glial cells.
Coles JA, Orkand RK. 1983. Modification of potassium movement Science 226:1345–1347.
through the retina of the drone (Apis mellifera male) by glial uptake. Mamata H, Mamata Y, Westin CF, Shenton ME, Kikinis R, Jolesz FA,
J Physiol (Lond) 340:157–174. et al. 2002. High-resolution line scan diff usion tensor MR imaging of
Cserr HF, DePasquale M, Nicholson C, Patlak CS, Pettigrew KD, white matter fiber tract anatomy. Am J Neuroradiol 23:67–75.
Rice ME. 1991. Extracellular volume decreases while cell volume is Mazel T, Simonova Z, Sykova E. 1998. Diff usion heterogeneity and
maintained by ion uptake in rat brain during acute hypernatremia. anisotropy in rat hippocampus. Neuroreport 9:1299–1304.
J Physiol (Lond) 442:277–295. McCarthy NA, O’Neil RG. 1992. Calcium signaling in cell volume reg-
Czeh G, Kriz N, Sykova E. 1981. Extracellular potassium accumulation ulation. Physiol Rev 72:1037–1061.
in the frog spinal cord induced by stimulation of the skin and ventro- Meech RW, Thomas RC. 1987. Voltage-dependent intracellular pH in
lateral columns. J Physiol (Lond) 320:57–72. Helix aspersa neurones. J Physiol (Lond) 390:433–452.
Deitmer JW, Schlue WR. 1987. The regulation of intracellular pH by Mongin AA, Orlov SN. 2001. Mechanisms of cell volume regulation and
identified glial cells and neurones in the central nervous system of the possible nature of the cell volume sensor. Pathophysiology 8:77–88.
leech. J Physiol (Lond) 388:261–283. Nicholson C, Phillips JM. 1981. Ion diff usion modified by tortuosity
Fuxe K, Agnati LF. 1991. Volume transmission in the brain: novel mech- and volume fraction in the extracellular microenvironment of the rat
anisms for neural transmission. New York: Raven Press. cerebellum. J Physiol (Lond) 321:225–257.
Gullans SR, Verbalis JG. 1993. Control of brain volume during hyperos- Nicholson C, Sykova E. 1998. Extracellular space structure revealed by
molar and hypoosmolar conditions. Annu Rev Med 44:289–301. diff usion analysis. Trends Neurosci 21:207–215.

C O N T R O L O F T H E E X T R AC E L LU L A R I O N I C E N VI R O N M E N T A N D VO LU M E • 441
Oliet SH, Piet R, Poulain DA. 2001. Control of glutamate clearance and Sykova E, Svoboda J, Polak J, Chvatal A. 1994. Extracellular volume
synaptic efficacy by glial coverage of neurons. Science 292:923–926. fraction and diff usion characteristics during progressive ischemia
Orkand RK, Nicholls JG, Kuffler SW. 1966. Effect of nerve impulses on and terminal anoxia in the spinal cord of the rat. J Cereb Blood Flow
the membrane potential of glial cells in the central nervous system of Metab 14:301–311.
amphibia. J Neurophysiol 29:788–806. Sykova E, Vargova L, Prokopova S, Simonova Z. 1999. Glial swelling
Pannasch U, Vargova L, Reingruber J, Ezan P, Holcman D, Giaume C, and astrogliosis produce diff usion barriers in the rat spinal cord. Glia
et al. 2011. Astroglial networks scale synaptic activity and plasticity. 25:56–70.
Proc Natl Acad Sci U S A 108:8467–8472. Sykova E, Vorisek I, Antonova T, Mazel T, Meyer-Luehmann M, Jucker
Pasantes-Morales H, Franco R, Torres-Marquez ME, Hernandez-Fonseca M, et al. 2005a. Changes in extracellular space size and geometry in
K, Ortega A. 2000. Amino acid osmolytes in regulatory volume APP23 transgenic mice: a model of Alzheimer’s disease. Proc Natl
decrease and isovolumetric regulation in brain cells: contribution and Acad Sci U S A 102:479–484.
mechanisms. Cell Physiol Biochem 10:361–370. Sykova E, Vorisek I, Mazel T, Antonova T, Schachner M. (2005b). Reduced
Pasantes-Morales H, Schousboe A. 1988. Volume regulation in astrocytes: extracellularspaceinthebrainoftenascin-R-andHNK-1-sulphotransferase
a role for taurine as an osmoeffector. J Neurosci Res 20:503–509. deficient mice. Eur J Neurosci, 22, 1873–1880.
Pierpaoli C, Jezzard P, Basser PJ, Barnett A, Di Chiro G. 1996. Diff usion Theodosis DT, Poulain DA. 1993. Activity-dependent neuronal-glial
tensor MR imaging of the human brain. Radiology 201:637–648. and synaptic plasticity in the adult mammalian hypothalamus.
Piet R, Vargova L, Sykova E, Poulain DA, Oliet SH. 2004. Physiological Neuroscience 57:501–535.
contribution of the astrocytic environment of neurons to intersynap- Thomas RC. 1977. The role of bicarbonate, chloride and sodium ions in
tic crosstalk. Proc Natl Acad Sci U S A 101:2151–2155. the regulation of intracellular pH in snail neurones. J Physiol (Lond)
Prokopova S, Vargova L, Sykova E. 1997. Heterogeneous and aniso- 273:317–338.
tropic diff usion in the developing rat spinal cord. Neuroreport Thorne RG, Nicholson C. 2006. In vivo diff usion analysis with quan-
8:3527–3532. tum dots and dextrans predicts the width of brain extracellular space.
Roitbak T, Sykova E. 1999. Diff usion barriers evoked in the rat cortex by Proc Natl Acad Sci U S A 103:5567–5572.
reactive astrogliosis. Glia 28:40–48. Van der Toorn A, Sykova E, Dijkhuizen RM, Vorisek I, Vargova L,
Siesjo BK, von Hanwehr R, Nergelius G, Nevander G, Ingvar M. 1985. Skobisova E, et al. 1996. Dynamic changes in water ADC, energy
Extra- and intracellular pH in the brain during seizures and in the metabolism, extracellular space volume, and tortuosity in neonatal
recovery period following the arrest of seizure activity. J Cereb Blood rat brain during global ischemia. Magn Reson Med 36:52–60.
Flow Metab 5:47–57. Vargova L, Chvatal A, Anderova M, Kubinova S, Ziak D, Sykova E. 2001.
Simonova Z, Svoboda J, Orkand P, Bernard CC, Lassmann H, Sykova Effect of osmotic stress on potassium accumulation around glial cells
E. 1996. Changes of extracellular space volume and tortuosity in the and extracellular space volume in rat spinal cord slices. J Neurosci Res
spinal cord of Lewis rats with experimental autoimmune encephalo- 65:129–138.
myelitis. Physiol Res 45:11–22. Vargova L, Homola A, Zamecnik J, Tichy M, Benes V, Sykova E. 2003.
Sontheimer H, Trotter J, Schachner M, Kettenmann H. 1989. Channel Diff usion parameters of the extracellular space in human gliomas.
expression correlates with differentiation stage during the devel- Glia 42:77–88.
opment of oligodendrocytes from their precursor cells in culture. Vizi ES. 1980. Nonsynaptic modulation of transmitter release:
Neuron 2:1135–1145. Pharmacological implication. Trends Pharmacol Sci 1:172–175.
Svoboda J, Sykova E. 1991. Extracellular space volume changes in the Vizi ES. 1984. Non-synaptic interaction between neurones: Modulation
rat spinal cord produced by nerve stimulation and peripheral injury. of neurochemical transmission. New York: Wiley and Sons.
Brain Res 560:216–224. Vizi ES. 2000. Role of high-affinity receptors and membrane transport-
Sykova E. 1992. Ionic and volume changes in the microenvironment of ers in nonsynaptic communication and drug action in the central
nerve and receptor cells. Berlin: Springer-Verlag 1–167. nervous system. Pharmacol Rev 52:63–89.
Sykova E. 1997. Extracellular space in the CNS: its regulation, vol- Vorisek I, Hajek M, Tintera J, Nicolay K, Sykova E. 2002. Water ADC,
ume and geometry in normal and pathological neuronal function. extracellular space volume, and tortuosity in the rat cortex after trau-
Neuroscientist 3:28–41. matic injury. Magn Reson Med 48:994–1003.
Sykova E. 2001. Glial diff usion barriers during aging and pathological Vorisek I, Sykova E. 1997a. Evolution of anisotropic diff usion in the
states. Prog Brain Res 132:339–363. developing rat corpus callosum. J Neurophysiol 78:912–919.
Sykova E. 2003. Diff usion parameters of the extracellular space. Isr J Vorisek I, Sykova E. 1997b. Ischemia-induced changes in the extracellu-
Chem 43:55–69. lar space diff usion parameters, K+, and pH in the developing rat cor-
Sykova E. 2004. Diff usion properties of the brain in health and disease. tex and corpus callosum. J Cereb Blood Flow Metab 17:191–203.
Neurochem Int 45:453–466. Walz W, Hertz L. 1983. Intracellular ion changes of astrocytes in
Sykova E, Chvatal A. 2000. Glial cells and volume transmission in the response to extracellular potassium. J Neurosci Res 10:411–423.
CNS. Neurochem Int 36:397–409. Walz W, Hinks EC. 1986. A transmembrane sodium cycle in astrocytes.
Sykova E, Mazel T, Hasenohrl RU, Harvey AR, Simonova Z, Mulders Brain Res 368:226–232.
WH, et al. 2002. Learning deficits in aged rats related to decrease in Wieshmann UC, Clark CA, Symms MR, Barker GJ, Birnie KD, Shorvon
extracellular volume and loss of diff usion anisotropy in hippocam- SD. 1999. Water diff usion in the human hippocampus in epilepsy.
pus. Hippocampus 12:269–279. Magn Reson Imaging 17:29–36.
Sykova E, Mazel T, Simonova Z. 1998. Diff usion constraints and Zamecnik J, Vargova L, Homola A, Kodet R, Sykova E. 2004.
neuron-glia interaction during aging. Exp Geront 33:837–851. Extracellular matrix glycoproteins and diff usion barriers in human
Sykova E, Nicholson C. 2008. Diff usion in brain extracellular space. astrocytic tumours. Neuropathol Appl Neurobiol 30:338–350.
Physiol Rev 88:1277–1340. Zhang H, Kelly G, Zerillo C, Jaworski DM, Hockfield S. 1998.
Sykova E, Rothenberg S, Krekule I. 1974. Changes of extracellular potas- Expression of a cleaved brain-specific extracellular matrix protein
sium concentration during spontaneous activity in the mesencephalic mediates glioma cell invasion in vivo. J Neurosci 18:2370–2376.
reticular formation of the rat. Brain Res 79:333–337. Zoli M, Jansson A, Sykova E, Agnati LF, Fuxe K. 1999. Intercellular
Sykova E, Svoboda J. 1990. Extracellular alkaline-acid-alkaline tran- communication in the central nervous system. The emergence of the
sients in the rat spinal cord evoked by peripheral stimulation. Brain volume transmission concept and its relevance for neuropsychophar-
Res 512:181–189. macology. Trends Pharmacol Sci 20:142–150.

442 • FUNCTIONS OF NEUROGLIAL CELLS


35.
AMINO ACID NEUROTRANSMITTER SYNTHESIS
AND REMOVAL
Arne Schousboe, Lasse K. Bak, Karsten K. Madsen, and Helle S. Waagepetersen

A B B R E VI AT I O N S analog, taurine, which is present in the brain in large amounts


(Huxtable 1992), is sometimes referred to as a neurotransmitter
AAT aspartate aminotransferase or neuromodulator because it can activate both GABA and gly-
ALAT alanine aminotransferase cine receptors (Oja and Saransaari 2007), but this amino acid
AOAA aminooxyacetic acid will not be dealt with in this chapter. Interestingly, the amino
BCAA branched chain amino acid acid neurotransmitters include a D-amino acid namely D-serine
BCAT branched chain aminotransferase which is functionally closely related to glycine (Wolosker et al.
BGT betaine-GABA transporter 2008). The amino acid neurotransmitters mediate via activa-
CIT citrate tion of specific receptors either excitatory or inhibitory signals.
EAAC excitatory amino acid carrier Hence, glutamate and aspartate function as excitatory neu-
EAAT excitatory amino acid transporter rotransmitters (Danbolt 2001) whereas glycine and GABA
GABA gamma-aminobutyric acid function as inhibitory transmitters (Curtis and Johnston 1974;
GABA-T GABA transaminase Schousboe and Waagepetersen 2008). Glycine, however, has
GAD glutamate decarboxylase a more complex mode of action because it, in addition to its
GAT GABA transporter action on specific glycine receptors, also acts as a cotransmitter
GC glutamate hydroxyl carrier (coagonist) with glutamate on a separate glycine receptor site
GDH glutamate dehydrogenase on the N-methyl-d-aspartate (NMDA) subtype of the iono-
GLAST glutamate aspartate transporter tropic glutamate receptors (Zafra and Gimenez 2008). This
GLN glutamine way glycine can be regarded as an excitatory neurotransmitter
GLU glutamate as well. However, because this action is strictly dependent on
GlyT glial glycine transporter glutamate binding to the NMDA receptor, glycine is normally
GS glutamine synthase considered to be inhibitory in its own right. In relation to the
GLT-1 glutamate transporter 1 glycine site on the NMDA receptors, D-serine is of interest
ME malic enzyme because it turns out that it binds to the glycine site, thereby
NMDA N-methyl-d-aspartate replacing glycine as the coagonist. This aspect will be discussed
OAA oxaloacetate in a separate section because astrocytes appear to be directly
PAG phosphate activated glutaminase involved in this action of D-serine (Wolosker et al. 2008).
PC pyruvate carboxylase The major excitatory amino acid neurotransmitter glutamate
PDH pyruvate dehydrogenase exhibits a complex metabolism involving an obligatory neu-
PKC protein kinase C ronal–glial interaction (Waagepetersen et al. 2009). The neu-
SLC solute carrier rotransmitter activity of glutamate is terminated by binding to
SSA succinic semialdehyde and transport by a number of specific, high affinity transporters,
SSADH SSA dehydrogenase the majority of which in terms of transport capacity are located
TCA tricarboxylic acid on astroglial cells (Danbolt 2001; Gegelashvili and Schousboe
TMD transmembrane spanning domain 1997). The metabolism of the inhibitory neurotransmitter
GABA is also complex, involving both neuronal and astrocytic
elements and, in analogy to glutamate, its inactivation is medi-
1 INTRODUCTION ated by a number of high affinity transporters in neurons and
astrocytes (Schousboe and Waagepetersen 2008). However,
Among the amino acids present in the central nervous system, contrary to glutamate, the GABA transporter capacity has a
only glutamate, aspartate, and glycine are normally considered more prominent expression in GABAergic neurons compared
to act as neurotransmitters in addition to being part of meta- with that in astrocytes (Schousboe and Waagepetersen 2008).
bolic processes and protein synthesis. In addition to these, gam- This chapter will deal in detail with the previously men-
ma-aminobutyric acid (GABA) also acts as a transmitter and tioned aspects of glutamatergic, GABAergic, and glycinergic
a metabolite but it is not found in proteins. Its closely related neurotransmission, emphasizing the role of astroglial cells.

443
2 G LU TA M AT E Glutamate neurotransmission (Fig. 35.1) is terminated
by diffusion of glutamate away from the receptor, binding to
2.1 B I O SY N T H E S I S A N D M ETA B O L I S M high affinity glutamate transporters primarily in the astrocyte
plasma membrane and eventually uptake (see section 2.3). In
Astrocytes are key elements in the maintenance of glutamater- the cytosol of the astrocyte, glutamate may be converted to
gic neurotransmission because only astrocytes express the glutamine by the enzyme GS, which is exclusively expressed
enzymes that are mandatory for homeostasis of glutamate, in astrocytes (Norenberg and Martinez-Hernandez 1979).
such as glutamine synthetase (GS) and pyruvate carboxylase Glutamate is irreversibly amidated using free ammonium
(Norenberg and Martinez-Hernandez 1979; Yu et al. 1983). and energy is provided by the cleavage of ATP to ADP. The
In contrast, neurons are metabolically handicapped and are Km values for GS in brain have been reported to be 0.2 mM
not equipped to perform de novo synthesis of their own neu- for ammonia, 2.5 mM for glutamate, and 2.3 mM for ATP
rotransmitter. The concept of metabolic interaction between (Pamiljans et al. 1962). Intracellular ATP levels in brain are
neurons and astrocytes necessary to sustain glutamate home- approximately 2.4 mM, and the ammonia concentration in
ostasis was founded on studies of glutamate and glutamine the astrocyte compartment is estimated to be less than 0.2
metabolism in brain tissue preparations. The early studies mM (Cooper and Plum 1987). The glutamate concentra-
indicated distinct cellular compartments of glutamate and tion in astrocytes is low because of rapid metabolism, and
glutamine with different turnover rates (van den Berg and the microenvironment around the glutamate transporter is
Garfinkel 1971). probably of great functional importance. A phenomenon

Glucose

PYR
PDH
PC
ME
OAA
TCA CIT
Cyele
MAL

Glutamatergic α–KG
neuron ASP
Gln Gln
NH4+ AAT
+
SNAT3 + GDH OAA
NH4+ Na H GS
GLU
Gln Gln
GLU GLN SNAT1 LAT2 GLN
Na+ AA
PAG
Gln Gln
ASCT2
Na+ GS Astrocyte
NH4+

GLU GLU
GLUTAMATE

Figure 35.1 Neurotransmitter glutamate is to a large extent taken up by the glial transporters, GLT-1 or GLAST, subsequent to postsynaptic receptor
interaction. Glutamate (GLU) may be amidated to glutamine (GLN) via glutamine synthetase (GS) and glutamine is subsequently released from the
astrocyte via specific transporters (i.e., SNAT3, LAT2, or ASCT2), and taken up by the neuron via SNAT1. In the neuron, glutamine is deamidated by
phosphate activated glutaminase (PAG) to glutamate, completing the glutamate–glutamine cycle. Alternatively, glutamate may be oxidatively metabo-
lized after entrance into the mitochondria. In this organelle glutamate is deamidated by glutamate dehydrogenase (GDH) or converted by aspartate
aminotransferase to α-ketoglutarate (α-KG). For complete oxidation of the carbon skeleton, pyruvate (PYR) recycling is required via the concerted
action of malic enzyme (ME) and pyruvate dehydrogenase (PDH). Glutamate and glutamine can be de novo synthesized from glucose in astrocytes
via the carboxylation of pyruvate catalyzed by the mitochondrial enzyme pyruvate carboxylase (PC). CIT, Citrate; MAL, malate; OAA, oxaloacetate;
TCA, tricarboxylic acid.

444 • FUNCTIONS OF NEUROGLIAL CELLS


that is only sparsely investigated but worth noting is the rapid removal of glutamate away from the microenvironment
recent finding that GS was not one of the enzymes immu- around the transporter may continuously preserve the plasma
noaffinity isolated with the glutamate transporter GLT-1 membrane gradient of glutamate and efficient uptake (see
(Genda et al. 2011). section 2.3.3).
Glutamine is released from astrocytes and taken up by Glutamate is to a considerable extent metabolized oxida-
nearby neurons via specific transporters as described later (see tively (Fig. 35.2) and the significance of this pathway is appar-
section 2.2). The subsequent deamidation of glutamine form- ently increasing with the extracellular glutamate concentration,
ing glutamate by phosphate activated glutaminase, an enzyme whereas the flux of glutamate converted into glutamine is inde-
enriched in neurons, completes the glutamate–glutamine pendent of this (McKenna et al. 1996). Moreover, the synthesis
cycle (see Fig. 35.1). Phosphate activated glutaminase is asso-
ciated with the inner mitochondrial membrane, a localization
that may have functional consequences facilitating oxidative
metabolism of glutamine in neurons (Waagepetersen et al.
2005). As an alternative to the simple amidation of glutamate
into glutamine in astrocytes, glutamate may enter the mito-
chondria for oxidative metabolism, which is likely promoted Glucose
by a close association of the glutamate transporter, GLT-1,
to mitochondria (Genda et al. 2011). Oxidative metabolism PYR
LDH
LAC
of glutamate in astrocytes and that of glutamine in neurons
PC PDH
disrupts a simple stoichiometric view of the glutamate–glu-
tamine cycle but provides the cells with a dynamic system ME
OAA
ready to respond to changes in neurotransmission as well as TCA CIT
energy requirements. cycle
MAL
One of the ways of glutamate entrance into the mitochon-
α–KG
dria is the glutamate hydroxyl carrier (GC). Another entry via
the aspartate/glutamate carrier Aralar1 was found plausible NH4+ ASP
AAT
from the results of a transcriptomic analysis of acutely iso- GDH OAA
GLU
lated astrocytes (Lovatt et al. 2007) but translation into the
protein level has been questioned in astrocytes (Ramos et al.
2003). Aralar1 is an antiporter transporting aspartate out Astrocyte
accompanied by the simultaneous import of glutamate to the
mitochondria, which has to be taken into account considering
the purpose or metabolic fate of glutamate inside the mito- GLU
chondria. In contrast to Aralar1, GC enables transfer of gluta-
mate either into or out of the mitochondria dependent on the
concentration gradient of glutamate and protons. This allows
coupling of the transport to the activity of the electron trans-
port chain and subsequent ATP yield. Two distinct isoforms
of the glutamate hydroxyl carrier, GC1 and GC2, have been
identified with equal expression level in the brain (Fiermonte
et al. 2002). This is in contrast to other organs, which pre-
Figure 35.2 The extent to which glutamate (GLU) is oxidized in astrocytes
dominantly express GC1. Primarily, GC1 has been found in seems to increase particularly during higher glutamate concentrations. A
an enriched glial fraction (Berkich et al. 2007) whereas no net synthesis of tricarboxylic acid (TCA) cycle intermediates occurs
information is available on the cellular localization of GC2 when the initial reaction is catalyzed by glutamate dehydrogenase
in brain. Distinct kinetic characteristics are exhibited by (GDH), which paves the way for the complete oxidation of the carbon
GC1 and 2, the Km value for GC1 being 5 mM glutamate and skeleton of glutamate. This requires pyruvate recycling via the concerted
action of malic enzyme (ME) and pyruvate dehydrogenase (PDH)
that for GC2 only approximately 0.26 mM. The capacity for converting malate into acetyl CoA producing NADPH. Acetyl CoA is
transport, that is the Vmax, which is dependent on pH, ranges oxidized completely in one turn of the TCA cycle. A partial oxidation of
from 12 to 63 μmol/minute per gram for GC1 and from 4 glutamate is acquired when pyruvate (PYR) is reduced to lactate instead
to 16 μmol/minute per gram for GC2. For comparison the of being oxidized to acetyl CoA. The redox state of the cell is likely
glutamate-glutamine cycling rate has been estimated to be 0.5 important in the regulation of the destiny of the glutamate molecule.
Alternative to the activity of GDH, aspartate aminotransferase (AAT)
μmol/minute per gram in awake animals (Oz et al. 2004). facilitates the formation of α-ketoglutarate (α-KG) from glutamate at
Interestingly, Aralar1 as well as GC1 were immunoaffinity the expense of oxaloacetate (OAA); thus no net synthesis of TCA cycle
isolated with GLT-1 (Genda et al. 2011), strongly indicating intermediates is obtained. In contrast to the complete oxidation initiated
that Aralar1 is translated in astrocytes. A close association of by the activity of GDH, AAT enables the truncated TCA cycle, which
mitochondria to plasma membrane glutamate uptake may refers to the net synthesis of aspartate from glutamate, a pathway shown
to accelerate during hypoglycemic conditions. All enzymes except ME
support the energy requirement of the linked Na+/K+ ATPase and LDH are located in the mitochondria. CIT, Citrate; PC, pyruvate
activity as suggested by Genda et al. (2011). Additionally, a carboxylase.

A M I N O AC I D N E U R OT R A N S M I T T E R SY N T H E S I S A N D R E M O VA L • 445
of glutamine seems resistant to energy deprivation in spite of and GDH2, but likely especially GDH2, may have a particu-
the energy requirement of the reaction (Morland et al. 2004). lar role in glutamate oxidation when astrocytes are experienc-
The concentration dependency can on one side reduce a poten- ing either a global or local elevation of ADP.
tial excitotoxic level of glutamate in the synaptic area because of Carboxylation of pyruvate to oxaloacetate is catalyzed by
oxidation of glutamate and reduction of glutamate-glutamine pyruvate carboxylase (see Fig. 35.2), the most important ana-
cycle activity. On the other hand, it may lead to depletion of plerotic enzyme in brain and it is like GS confined to astrocytes
the neuronal pool of tricarboxylic acid (TCA) cycle intermedi- (Yu et al., 1983). Anaplerosis, for example de novo synthesis
ates and compromise neuronal energy metabolism. The initial of TCA cycle intermediates, is the opposite of pyruvate recy-
reaction for glutamate oxidation is either an oxidative deami- cling, but the activity of anaplerosis, which has been estimated
nation by glutamate dehydrogenase (GDH) or a transamina- to account for 25% of glutamate–glutamine cycle activity is
tion by an aminotransferase. Of the transaminases, aspartate high in contrast to that of pyruvate recycling (Oz et al. 2004).
aminotransferase (AAT) is the one exhibiting the highest Pyruvate carboxylation increases in response to hyperammone-
activity in brain, although the branched chain aminotrans- mia, providing the carbon skeleton necessary for assimilation
ferase (BCAT) and alanine aminotransferase (ALAT) also of ammonia into glutamine (Sibson et al. 2001).
have to be taken into account. Only the mitochondrial iso- The glutamate–glutamine cycle does not provide any spe-
form of BCAT seems to be expressed in astrocytes (Bixel et al. cific mechanism by which ammonia generated in the gluta-
2001). The relative importance of GDH and AAT has been matergic neurons reaches the astroglial compartment to allow
debated and GDH seems to be primarily active in the deami- amidation of glutamate catalyzed by GS. Although some
nation of glutamate whereas AAT catalyzes the conversion of ammonia will be cycled from neurons to astrocytes by passive
α-ketoglutarate to glutamate, concomitantly forming oxaloac- diffusion as suggested by Provent et al. (2007), an amino acid
etate from aspartate (Westergaard et al. 1996). It is important shuttle mechanism would decrease the concentration of free
to realize the metabolic consequences of the different enzymes ammonia (Fig. 35.3). Two different mechanisms have been
that can catalyze the formation of α-ketoglutarate from gluta- proposed based on nitrogen transfer via alanine or one of
mate, such as AAT, ALAT, BCAT, and GDH. Glutamate is the branched chain amino acids (BCAAs) (Bak et al. 2006).
converted to α-ketoglutarate by AAT at the expense of oxalo- Glutamine is deamidated in the neuron and the liberated
acetate conversion to aspartate. It should be noted, however, ammonia is suggested to be used in the reductive amination
that the activity of AAT enables the truncated TCA cycle, of α-ketoglutarate by glutamate dehydrogenase that works in
that is, the net conversion of glutamate to aspartate, forming concert with either BCAT or ALAT, transferring the nitrogen
75% of the ATP that is generated from oxidation of acetylCoA into one of the BCAAs or alanine, respectively. Alanine or the
(Yudkoff et al. 1994). In contrast, the activity of the three latter BCAA is released from the neuron and taken up by the astro-
enzymes ALAT, BCAT, and GDH leads to a net synthesis of cyte, thereby transferring the nitrogen, which through the
TCA cycle intermediates, and potentially a complete oxida- concerted action of the respective transaminases and GDH
tion of glutamate. Pyruvate recycling, which is the concerted is dissimilated and available for GS. The alanine shuttle was
action of malic enzyme and pyruvate dehydrogenase catalyz- shown to operate in co-cultures of glutamatergic neurons and
ing the oxidative decarboxylations of malate into acetyl CoA cerebellar astrocytes; however, the activity was not increasing
via pyruvate, is required for such complete oxidation of gluta- in parallel with the glutamate-glutamine cycle as would have
mate (see Fig. 35.2). Alternatively, pyruvate may be reduced to been expected (Bak et al. 2005). The GDH reaction has to
lactate for release. The extent to which this occurs is probably operate in both directions for these shuttles to operate, that
regulated by the energy and redox state of the cell (McKenna is, reductive amination in neurons and oxidative deamina-
et al. 1996; Sonnewald et al. 1996). Pyruvate recycling activity tion in astrocytes; however, GDH is predominantly located
has been demonstrated in cultured astrocytes repeatedly but in astrocytes and the deamination is favored except during
based on in vivo data the activity is limited. high levels of ammonia (Zaganas et al. 2009). Additionally,
Several studies have shown that GDH is predominantly the BCAAs have been suggested to provide the amino nitro-
expressed in astrocytes, especially in regions exhibiting high gen for de novo synthesis of glutamate via pyruvate carboxy-
glutamatergic activity (Aoki et al. 1987). There are two iso- lation in astrocytes followed by amidation of glutamate with
forms of GDH, GDH1 and GDH2. Only great apes and free ammonia by GS and transfer of glutamine to the neurons.
humans possess the gene, GLUD2, from which GDH2 is The BCAA is thought to be regenerated in the neuron via
expressed, mainly in retina, brain, and testis (Zaganas et al. BCAT using glutamate formed via GDH catalyzed reductive
2009). In brain GDH is mainly active in the direction of amination of α-ketoglutarate and returned to the astrocyte.
oxidative deamination although the thermodynamic equilib- It should be noted that this is the unfavored direction of the
rium favors reductive amination. The reason for this is a high GDH catalyzed reaction (Lieth et al. 2001).
NAD+/NADH ratio in astrocytes and a high Km value (in
the range of 10 mM) of ammonia (Zaganas et al. 2009). Both
2.2 RO L E O F G LU TA M I N E T R A NS P O RT E R S
GDH1 and 2 are activated by ADP and leucine. However,
I N G LU TA M AT E H O M EO S TA S I S
the effect is 10-fold higher for GDH2 than for GDH1 and
GDH2 is barely active in the absence of ADP. In contrast to Efflux of glutamine from astrocytes must be met by influx in
GDH1, GDH2 is insensitive to an inhibitory effect of GTP neurons for the glutamate/GABA-glutamine cycle to func-
(Plaitakis et al. 2003; Zaganas et al. 2009). Thus, both GDH1 tion. To achieve this, one would need two sets of transport

446 • FUNCTIONS OF NEUROGLIAL CELLS


Glutamatergic
neuron Astrocyte
ka ka
Glu Glu

AT AT
α-KG α-KG
aa aa
GDH
GDH
NH4+

GLU GLN GLN


PAG

GS NH4+

GLU GLU
GLUTAMATE

Figure 35.3 Neurotransmitter glutamate is primarily taken up by the glial transporters, GLT-1 or GLAST, subsequent to postsynaptic receptor interaction.
Glutamate (GLU) may be amidated to glutamine (GLN) via glutamine synthetase (GS) and glutamine is subsequently released from the astrocyte via
specific transporters followed by uptake into the neuron. In the neuron glutamine is deamidated by phosphate activated glutaminase (PAG) to gluta-
mate, completing the glutamate–glutamine cycle. Although often neglected, the cycling of glutamate and glutamine is accompanied by a net transfer
of nitrogen from the astrocyte to the neuron carried by glutamine. We have suggested that this nitrogen transfer is counteracted by transport of a
neuroinactive amino acid (aa) likely either alanine or one of the branched chain amino acids. The glutamate–glutamine cycle is coupled to the aa–keto
acid (ka) cycle via the concerted action of glutamate dehydrogenase (GDH) and the relevant aminotransferase (AT).

systems, one in astrocytes and one in neurons that may or may Table 35.1 NOMENCLATURE OF CLONED GLUTAMINE
not be the same (see Fig. 35.1). It would intuitively make sense TRANSPORTERS
if the astrocytic transport system is coupled closely to the syn- SYSTEMATIC NAME ALTERNATIVE NAMES
thesis of glutamine in astrocytes and if the neuronal transport
system is concentrative; this would create ideal conditions System A SNAT1 ATA1, GInT, SA2, SAT1,
for a transcellular flux of glutamine. Indeed, the current con- (neurons) SNAT2 NAT2
sensus is that the neuronal transporter is the sodium-coupled SNAT4 ATA2, SA1, SAT2
ATA3, NAT3
system A, whereas the astrocytic transport is mediated by sys-
tem N, a glutamine/Na+/H+ antiporter, albeit systems L and System N SNAT3 SN1, NAT1
ASC also may be involved as discussed below (see Fig. 35.1). (astrocytes) SNAT5 SN2
The system A transporter has been suggested by a number of
Nomenclature for systems A and N, two of the transport systems suggested as
authors as the neuronal transporter (see Bak et al. 2006 and being involved in neuronal and astrocytic glutamine cycling, respectively.
references) and it is comprised of three isoforms (Table 35.1):
SNAT1 (ATA1, GlnT, SA2, SAT1, NAT2), SNAT2 (ATA2, See text and Bak et al. 2006.
SA1, SAT2), and SNAT4 (ATA3, NAT3). Both SNAT1 and
2 are expressed in the brain, with SNAT1 being fairly brain-
and neuron-specific and found in the membrane of both glu- 230 μM compared with 1.65 mM for SNAT2 (references in
tamatergic and GABAergic neurons (see Bak et al. 2006 for Bak et al. 2006); the extracellular concentration of glutamine
references). On the other hand, SNAT2 seems to be more in rat brain is about 400 μM (Kanamori and Ross 2004).
widely distributed and is found on both neurons and astro- However, some controversy remains since Rae et al. (2003)
cytes (references in Bak et al. 2006). The previously men- found that a transport system different from system A is
tioned requirements for concentrative glutamine transport involved in glutamine cycling and Conti and Melone (2006)
into neurons are exhibited by SNAT1, being a Na+-glutamine were not able to localize system A transport molecules to syn-
symporter (i.e., electrogenic) with a Km for glutamine of aptic regions employing immunocytochemistry. System N

A M I N O AC I D N E U R OT R A N S M I T T E R SY N T H E S I S A N D R E M O VA L • 447
(see Table 35.1) comprising SNAT3 (SN1, NAT1) and to decreased glutamate uptake (Divac et al. 1977), glutamate
SNAT5 (SN2) has been suggested to mediate the glutamine transport was originally believed to be associated primarily
efflux from astrocytes, as reviewed by Bak et al. (2006). with glutamatergic neurons. The demonstration of a high affin-
System N is a Na+-glutamine symporter/H+ antiporter, mak- ity glutamate uptake in bulk-prepared glial cells (Schousboe
ing it sodium-dependent, yet electroneutral (see Bak et al. 1981) and primary cultures of astrocytes (Schousboe 1981)
2006 for references). Interestingly, this transporter mediates showed, however, that astrocytes must play an important role
glutamine flux in both directions under physiological condi- in glutamate transport.
tions probably governed by fluctuations in intracellular Na+
levels and pH as shown in Xenopus laevis oocytes expressing 2.3.1 Cloning of Transporters
SNAT3; Na+ levels and pH are parameters that are likely to
vary at glial microdomains during neurotransmission because The purification of a glutamate transporter from a synapto-
glutamate uptake may affect both (see section 2.3). The Km of somal preparation (Danbolt et al. 1990) paved the way for the
system N (SNAT3) transport was lowered in cultured astro- cloning of a series of glutamate transporters (for references, see
cytes by incubation in the presence of glutamate (30–60 Danbolt 2001; Gegelashvili and Schousboe 1997). The names
minutes, 10–100 μM; Bröer et al. 2004) further substantiat- of the transporters have been somewhat confusing because two
ing the possible involvement of this transport system in efflux different systems have been used. The original acronyms and
of neurotransmitter glutamate-derived glutamine. However, the more systematic names of the transporters as well as a list
the possibility that efflux of glutamine from astrocytes is of selective synthetic substrates and inhibitors are provided in
mediated by other transport systems in addition to system N Table 35.2 as well as in a recent review by Gether et al. (2006).
should not be neglected. Thus, Deitmer et al. (2003) found
that glutamine efflux was mediated by no less than four dif- 2.3.2 Cellular Localization of Transporters
ferent transport systems, namely system N (SNAT3), system
The transporters GLAST (EAAT-1) and GLT-1 (EAAT-2) are
L (LAT2), system ASC (ASCT2), and an unidentified trans-
by far the most highly expressed glutamate transporters in the
port system mediating as much as 40% of the efflux. System L
brain, with GLAST being more highly expressed in cerebel-
is an amino acid antiporter comprising two isoforms, LAT1
lum compared with forebrain and GLT-1 comparatively more
and 2, of which LAT2 shows affinity for glutamine (see Bak
highly expressed in forebrain (Danbolt 2001; Gegelashvili
et al. 2006 for references). System L mediates glutamine release
and Schousboe 1998). The transporter EAAC-1 excitatory
in the presence of extracellular amino acid substrates such as
amino acid carrier (EAAT-3), on the other hand, is primarily
alanine and leucine, which might be important for shuttling
expressed in neuronal spines and EAAT-4 seems to be located
nitrogen from neurons to astrocytes. System ASC is a sodium-
in spines of particularly Purkinje cells (Danbolt 2001). The
dependent antiporter, which exists as two different isoforms
present review therefore will deal only with the transporters
termed ASCT1 and ASCT2, with ASCT2 having affinity
GLT-1 and GLAST.
for glutamine, although this transport activity is probably of
minor importance in the adult mammalian brain (Bak et al. 2.3.2.1 GLT-1
2006 and references therein). Several regulatory mechanisms Using highly specific antibodies against GLT-1, Danbolt
have been proposed for astroglial glutamine transport activ- et al. (1992), Levy, (1993), and Rothstein et al. (1994) all have
ity including protein kinase C-mediated phosphorylation of found this glutamate transporter to be expressed only in astro-
SNAT3 and ASCT2 and subsequent ubiquitin-mediated deg- cytic processes (see Figs. 35.1 and 35.2). However, the mRNA
radation as well as a possible rapid caveolin-dependent inter- for GLT-1 has been found in neurons (Danbolt 2001) but if
nalization of SNAT3 protein induced by protein kinase C GLT-1 protein is expressed in neurons, it must be at a very low
(PKC) activity (Balkrishna et al. 2010; Sidoryk-Wegrzynowicz level or alternatively it may be a variant that is not recognized
et al. 2011). However, much work is still needed to ascertain by the currently available antibodies (Danbolt 2001). A recent
which amino acid transporters are involved in the glutamate/ immunocytochemical study using immunogold labeling at the
GABA-glutamine cycle and their regulation. EM level and a D-aspartate specific antibody has convincingly
shown that D-aspartate is taken up into presynaptic nerve end-
2.3 H I G H A FFI N IT Y T R A NS P O RT E R S ings (Furness et al. 2008) as indicated in Figure 35.1. It should
also be noted that in early development, GLT-1 appears to
The concept of an efficient and concentrative, energy- be expressed in neuronal populations and also tissue culture
dependent transport system for glutamate in the brain origi-
nates from studies by sir Hans Krebs and coworkers of brain Table 35.2 NOMENCLATURE OF CLONED GLUTAMATE
slices (Stern et al. 1949). These and numerous subsequent TRANSPORTERS
studies using brain slice preparations have convincingly pro-
vided evidence for high affinity glutamate transporters, a dem- TRANSPORTER SUBTYPE
onstration instrumental for the concept of glutamate acting as
Systematic EAAT-1 EAAT-2 EAAT-3 EAAT-4 EAAT-5
a neurotransmitter to be accepted (for references, see Danbolt name
2001; Schousboe 1981)). Based on the demonstration of high
affinity glutamate transport in synaptosomes (Danbolt, 2001) Original GLAST GLT-1 EAAC-1 EAAT-4 EAAT-5
name
and on the finding that degeneration of neuronal pathways led

448 • FUNCTIONS OF NEUROGLIAL CELLS


preparations of neurons have been reported to express GLT-1 transporters mediate changes in the intracellular concentra-
(Danbolt 2001). It may be of interest that in spite of the fact tions of Na+ and protons (Rose and Ransom 1996). Because
that GLT-1 is the most prevalent glutamate transporter in fore- a tight functional coupling appears to exist between the
brain (Danbolt 2001), cultured astrocytes from cerebral cortex glutamate transporters and the Na+/K+-ATPase in the astro-
generally have a higher expression of GLAST compared with cytic membrane (Bauer et al. 2012; Cholet et al. 2002; Genda
that of GLT-1 (Gegelashvili et al. 1997; Skytt et al. 2010). This et al. 2011; Rose et al. 2009; Schousboe et al. 2011), it may be
likely reflects the finding that the expression of GLT-1 in cul- assumed that the dissipation of the sodium gradient associated
tured astrocytes, and perhaps also in the brain, is dependent with glutamate transport is rapidly recovered.
on neuronal signaling (Gegelashvili et al. 1997; Schlag et al.
1998; Swanson et al. 1997) as well as other signaling molecules
2.3.4 Regulation of Expression of Glutamate
(O’Shea et al. 2006; Schlag et al. 1998). It should also be noted
Aspartate Transporter and Glutamate Transporter 1
that the surface expression of the glutamate transporters is reg-
ulated by trafficking from the cytoplasm to the membrane and The first indication that expression of glutamate transporters
vice versa (for references, see Robinson 2002). in astrocytes may be regulated by neuronal stimuli came from
a study by Drejer et al. (1983) demonstrating that exposure of
2.3.2.2 GLAST cultured cerebellar astrocytes to a conditioned medium from
As mentioned (section 2.3.2), GLAST exhibits a higher cultured cerebral cortical neurons led to a dramatic increase
expression level in cerebellum than in forebrain, that is, the in glutamate transport activity. It was subsequently shown
opposite of that of GLT-1 but it should be emphasized that by several investigators that this upregulation of transport
astrocytes throughout the brain express both GLAST pro- capacity in astrocyte cultures induced either by coculturing
tein and mRNA (for references, see Danbolt 2001). Cultured with neurons or by exposure to neuronal conditioned cul-
embryonic neurons seem to express GLAST in analogy to ture medium is explained by selective upregulation of the
their expression of GLT-1 (Plachez et al. 2000). It should be GLT-1 transporter (Gegelashvili et al. 1997; Schlag et al.
noted that the highest expression level of GLAST has been 1998; Swanson et al. 1997). Other factors such as arachidonic
found in the cerebellar Bergmann glia (Danbolt 2001), which acid, the redox state, the pH, and availability of zinc ions also
also have a high density of Ca2+-permeable AMPA receptors seem to play important roles (Levy et al. 2002). Agents that
(Müller et al. 1992; Parpura et al. 2012). affect the phosphorylation state have repeatedly been shown
to enhance the transport capacity of astrocytes, and it appears
2.3.3 Role of GLAST and GLT-1 in Termination that both astrocytic transporters are regulated by phosphory-
of the Neurotransmission Process lation (Levy et al. 2002). It may be of interest that GLAST
and GLT-1 are expressed in microglial cells after cortical injury
Because of the extremely high density of glutamate trans- (van Landeghem et al. 2001). It is important to note that there
porters in the astrocytic plasma membrane (Danbolt 2001), is a rapid and highly regulated trafficking of the transporters
it is clear that termination of the interaction of glutamate between the cytoplasm and the plasma membrane because this
with its receptors is functionally related to these transporters can lead to rapid changes in the transport capacity in response
(Danbolt 2001; Robinson and Dowd 1997). It is, however, to changes in the cellular milieu (Robinson 2002).
important to note that the cycling time for the transporters
of 50 to 100 milliseconds (Wadiche et al. 1995) is too slow
to effectively terminate the transmission process which should 2.3.5 Failure of Glial Glutamate Uptake Results
operate at a micro-to-millisecond scale (Tong and Jahr 1994). in Neurodegeneration
The high density of the transporters does, however, allow effi- As mentioned previously (section 2.3.3) it has been known
cient binding of a large number of glutamate molecules to take for a long time that high extracellular concentrations of the
place prior to transport, thereby securing the process to occur excitatory transmitters glutamate and aspartate almost cer-
within the required time scale (Tong and Jahr 1994). Hence, tainly result in excitotoxic neuronal damage (Choi 1988).
the concerted action of binding to and transport by the glial Under physiological conditions the capacity of the glutamate
transporters leads to an extremely efficient clearance of gluta- transporters expressed in astrocytes is high enough to clear
mate from the synaptic cleft, preventing accumulation of neu- the extracellular or extrasynaptic space for glutamate and thus
rotoxic extracellular concentrations to occur (Choi 1988). prevent any neuronal damage (Choi 1988; Danbolt 2001).
It may be interesting to note that because of the cotrans- During energy failure, which is associated with reduced oxy-
port of three sodium ions and possibly a proton and the gen and/or glucose supply, the ATP level is reduced leading to
counter-transport of one potassium ion, which makes the dramatic reduction of the activity of the Na+/K+-ATPase and
transporters highly electrogenic (Levy et al. 1998), the trans- dissipation of the sodium gradient needed for maintenance of
port process leads to a depolarization of the astrocytes which, the glutamate transporter efficiency. This results in reversal of
without a pharmacological characterization, may be indistin- the transporters and an increase in the extracellular glutamate
guishable from that seen after activation of ionotropic gluta- concentration (Phillis et al. 2000; Rossi et al. 2000). It should,
mate receptors (Bowman and Kimelberg 1984; Kettenmann however, be noted that the glycogen store in astrocytes (see
et al. 1984). Interestingly, it was shown that in cultured hip- chapter 36) can sustain sodium homeostasis in the absence of
pocampal astrocytes, kainate receptors as well as glutamate glucose and oxygen at least for a limited period of time (Rose

A M I N O AC I D N E U R OT R A N S M I T T E R SY N T H E S I S A N D R E M O VA L • 449
et al. 1998). This is in keeping with the demonstration that enzyme glutamate decarboxylase (GAD; Schousboe and
glycogen metabolism and turnover is important for astrocytic Waagepetersen 2008). Degradation of GABA requires GABA-
glutamate transport (Schousboe et al. 2011). Inhibition of the transaminase (GABA-T) to convert GABA to succinic semial-
glutamate transporters using a nontransportable inhibitor dehyde (SSA) by transamination with the cosubstrates glutamate
such as DL-threo-beta-benzyloxyaspartate (Shimamoto et al. and α-ketoglutarate (Schousboe and Waagepetersen 2008).
1998) has been shown to effectively prevent neurodegenera- Succinic semialdehyde is subsequently oxidized by SSA dehy-
tion in hippocampal slices induced by oxygen-glucose depriva- drogenase (SSADH) to succinate, a constituent of the TCA
tion (Bonde et al. 2003) clearly demonstrating the importance cycle. These latter processes can take place both in neurons and
of glutamate transporters. This latter notion is underlined by astrocytes because the enzymes involved are expressed in both
the finding that exposure of hippocampal slices to a variety cell types (Schousboe and Waagepetersen 2008). As discussed
of glutamate transporter inhibitors under physiological con- (sections 2.1 and 2.2), glutamate homeostasis involves an active
ditions will lead to extensive neurodeneration (Bonde et al. exchange of substrates between neurons and astrocytes. The
2003; O’Shea et al. 2002). same is the case for GABA because its de novo synthesis requires
glutamine (see Fig. 35.4), which is generated in astrocytes and
transferred to GABAergic neurons (Schousboe and
3 GABA Waagepetersen 2008).

3.1 B I O SY N T H E S I S A N D M ETA B O L I S M 3.2 M ETA B O L I S M A N D E P I L E P SY


The metabolism of GABA (Fig. 35.4) may be less complex than GABA-T is a pyridoxal phosphate dependent enzyme and
that of glutamate (see section 2.1) and its synthesis is restricted thus, GABA-T is sensitive to carbonyl trapping agents such
to GABAergic neurons expressing the GABA synthesizing as aminooxyacetic acid (AOAA), which potently inhibits the

Glucose

PYR
PDH
PC
NH4+
GLU GLN OAA
GLN TCA CIT
PAG cycle
GS suc
NH4+ SSADH
GAD GABAergic α–KG
neuron SSA
CO2 GLU GABA-T

GABA
GABA
GABA
Na+

Astrocyte
GABA
GAT1
Na+
GAT1

GABA
GAT3
GABA GABA Na+

Figure 35.4 Neurotransmitter GABA is to a large extent taken up by the neuronal GAT1 and to a minor extent by glial GAT1 and GAT3, subsequent to
postsynaptic receptor interaction. GABA taken up by neurons is primarily reused as neurotransmitter whereas GABA entering astrocytes is metabolized
via succinic semialdehyde (SSA) to succinate by the concerted action of GABA-transaminase (GABA-T) and SSA dehydrogenase (SSADH). The
astrocytic part of the synapse provides a net synthesis of glutamine (gln) via the concerted action of pyruvate carboxylase (PC) and pyruvate dehydro-
genase (PDH) generating OAA and acetyl-CoA, the combination of which leads to synthesis of CIT. The presynaptic neuron shows the biosynthetic
machinery for GABA comprising conversion of glutamine (gln) to GABA via glutamate (glu) catalyzed by the two enzymes phosphate activated
glutaminase (PAG) and glutamate decarboxylase (GAD), respectively. CIT, Citrate; GS, glutamine synthetase; OAA, oxaloacetate.

450 • FUNCTIONS OF NEUROGLIAL CELLS


enzyme (Schousboe et al. 1974). In addition, AOAA increases acting as a proconvulsant, it was hypothesized that selective
GABA levels and displays anticonvulsant activity in a variety inhibitors of the glial GAT would be promising agents for sei-
of seizure models (Sarup et al. 2003), but since it is nonselec- zure management (Schousboe 2000).
tive it is not a valuable tool in this regard. To circumvent this
problem the GABA analog, γ-vinyl GABA was synthesized 3.4 C L O N I N G O F T R A NS P O RT E R S
(Lippert et al. 1977). This compound is a GABA-T substrate
that displays irreversible covalent binding to the active site of Radian et al. (1986) isolated from rat brain what was believed
GABA-T upon transamination and thus acts as a suicide, cat- to be the neuronal GAT. Subsequently, 4 years later this trans-
alytic site-directed inhibitor (Lippert et al. 1977). This action porter was cloned revealing a 67 kDa protein consisting of
explains the remarkable specificity displayed by γ-vinyl GABA. 599 amino acids with an apparent Km for GABA of 7μM
Because of its high selectivity toward GABA-T it was devel- (Guastella et al. 1990) compared with 32 and 8 μM, which
oped as a clinically active antiepileptic drug named vigabatrin is the apparent Km for GABA for the astrocytic and neuronal
(Gram 1990). It currently represents the only antiepileptic GAT, respectively (Schousboe 2000). The pharmacological
agent whose mechanism of action specifically relies on inhibi- studies conducted confirmed this to be comparable to the
tion of GABA-T. It may not be clear to what extent inhibition neuronal subtype and it was designated GAT-1 (Guastella
of the astroglial GABA-T may be involved in the anticonvul- et al. 1990). Following this initial discovery a total of four
sant activity of vigabatrin, but from a study of the potency of GATs were cloned in several species which unfortunately has
vigabatrin as a GABA-T inhibitor in neurons and astrocytes resulted in a rather confusing nomenclature. The nomencla-
it appears that the action on the neuronal enzyme is the more ture in rat, canine, and human is identical, namely GAT-1,
important in this regard (Larsson et al. 1986). In GABAergic BGT-1, GAT-2, and GAT-3, whereas the nomenclature in
neurons, glutamine released from astrocytes appears to be an mouse is mGAT1, mGAT2, mGAT3, and mGAT4, respec-
important precursor for GABA (Walls et al. 2011 and refer- tively. BGT-1 (rat)/mGAT2 (mouse) is the betaine-GABA
ences therein) via formation of glutamate that is subsequently transporter that transports GABA with an apparent Km of 79
decarboxylated. It should be noted that recently a novel spe- μM and betaine with an apparent Km of 398 μM and encodes
cific inhibitor of GABA-T has been developed and because a protein of 614 amino acids. GAT-2 (rat)/mGAT3 (mouse)
this compound appears to have fewer side effects than vigaba- encodes a protein of 602 amino acids with an apparent Km for
trin it could serve as a model compound for development of a GABA of 18 μM and an apparent Km for β-alanine of 28 μM.
new antiepileptic drug (Pan et al. 2012). For a further discus- Last, GAT-3 (rat)/GAT4 (mouse) encodes a protein of 627
sion of this topic, please see chapter 70. amino acids that also transports GABA and β-alanine with an
apparent Km of 0.7 and 99 μM, respectively (Liu et al. 1993).
To avoid confusion, the nomenclature GAT1, BGT1, GAT2,
3.3 T R A NS P O RT E R S , T H E I R L O C A L I Z AT I O N and GAT3 as proposed by the HUGO gene nomenclature
A N D F U N C T I O NA L A N D P H A R M AC O L O G I C A L committee (Table 35.3) will be used hence forth (please note
C H A R AC T E R I Z AT I O N that it corresponds to the rat nomenclature without hyphen.
Termination of GABAergic neurotransmission is facilitated GABA transport by GAT2 and GAT3 is strongly inhibited
via high affinity GABA transporters (GATs) embedded in the by β-alanine and certain taurine analogs such as hypotaurine,
plasma membrane of both neurons and astrocytes (Iversen and suggesting that these two transporters most likely resemble
Neal 1968). However, this discovery was preceded by Elliott the astroglial GAT. On the other hand, BGT-1 seems not to
and van Gelder (1958) who were the first to show that GABA resemble either the neuronal or astroglial GAT but it is, how-
in the incubation media accumulated in slices of cerebral corti- ever, the only GAT inhibited by betaine (Liu et al. 1993).
ces. Much attention has been devoted to describe the neuronal
and glial transport systems and it was shown that most of the 3.5 S T RU C T U R E A N D F U N C T I O N
exogenous [3H]GABA applied to slices and synaptosomes of O F T R A NS P O RT E R S
rat cerebral cortices accumulated into nerve terminals; using
The GABA transporters belong to the Solute Carrier 6 (SLC6)
[3H]β-alanine it was demonstrated that this is a substrate for
superfamily, which is also comprised of the monoamine
the glial GAT, but not for the neuronal GAT (Schousboe 1981,
2000). Investigations using glial preparations have demon-
strated that uptake of GABA into these cell types could account Table 35.3 NOMENCLATURE OF CLONED GABA
for a substantial portion of the vesicular released GABA esti- TRANSPORTERS
mated to approximately 20% (Schousboe 2000). As described TRANSPORTER SUBTYPE
above the concerted action of GABA-T and SSADH degrades
GABA to succinate in both neurons and astrocytes. Only a Systematic GAT1 BGT1 GAT2 GAT3
name
minor fraction of GABA degradation takes place in neurons
due to recycling of neurotransmitter, whereas all GABA taken Mouse mGAT1 mGAT2 mGAT3 mGAT4
up by astrocytes is degraded (see Fig. 35.4). Because of a signif- nomenclature
icant loss of neurotransmitter via GABA uptake in astrocytes
and subsequent degradation, along with the discovery that Rat/human GAT-1 BGT-1 GAT-2 GAT-3
nomenclature
the neuronal GABA uptake inhibitor diaminobutyric acid is

A M I N O AC I D N E U R OT R A N S M I T T E R SY N T H E S I S A N D R E M O VA L • 451
transporters for serotonin, dopamine, and noradrenaline and et al. 2007). Compared with GAT1, the expression of GAT3
transporters for amino acids like proline, glycine, and tau- is restricted to distal astrocytic processes (see Fig. 35.4) that
rine (Kristensen et al. 2011; Madsen et al. 2007). The energy are in direct contact with GABAergic synapses and perhaps
demanding uptake of these transmitters is coupled to an to a much lesser degree on neuronal elements (Madsen et al.
inward cotransport of sodium ions which fuels the process. 2007). The highest degree of expression of GAT2 is in the
The four GABA transporters show an absolute requirement neonatal brain suggesting a role in fetal development. In the
for two sodium and one chloride ion per transported GABA adult brain GAT2 is strongly expressed in the leptomeninges
molecule (Kristensen et al. 2011). The sodium driving force is and ependyma. However, GAT2 is only weakly expressed on
capable of generating a concentration gradient in the order of cortical neurons close to both symmetric and asymmetric syn-
magnitude of 105 between the intracellular and extracellular apses but away from areas with synaptic specialization, and on
GABA (Schousboe 1981). The GATs share similar structural distal and proximal astrocytic processes and cell body (Madsen
features with the other transporters of the SLC6 gene fam- et al. 2007). BGT1 has also been found in the leptomeninges
ily in that they have twelve transmembrane spanning domains (Takanaga et al. 2001) and on pyramidal neurons in the cere-
(TMD) with a large extracellular loop between TMD3 and bral cortex and in the CA field of the hippocampus. It local-
4 containing three glycosylation sites and intracellular facing izes to small diameter dendrites or dendritic spines making
carboxy and amino terminals (Kristensen et al. 2011). Recently, asymmetric synapses but to an extrasynaptic region away from
a crystal structure of a bacterial homologue of a sodium- and synaptic specialization (Zhu and Ong 2004). It should be
chloride-dependent neurotransmitter transporter has brought noted, however, that a recent study by Lehre et al. (2011) has
about critical knowledge regarding the structure and function demonstrated a low expression level of BGT1 in the brain.
of these transporters (Yamashita et al. 2005). As seen with the
pharmacological profile of the GATs, GAT1 shares the least
3.8 P H A R M AC O L O GY O F T R A NS P O RT E R S
amount ~50% of amino acid sequence homology with the
other GATs whereas BGT1, GAT2, and GAT3 have an iden- The development of the GAT1 selective inhibitor tiagabine by
tity of 65% to 70% (Miller et al. 2002). Novo Nordisk underlines that inhibitors of GABA transporters
can be developed as anticonvulsants (Nielsen et al. 1991). Many
compounds were developed inhibiting GABA transporters;
3.6 R EGU L AT I O N O F T R A N S P O RT E R AC T I VIT Y however, only a few preferentially inhibited the glial GAT. In
The regulation of GAT surface expression has been shown a series of compounds based on exo-THPO the most selective
to be influenced by PKC, tyrosine kinase/phosphatase activ- compound to this date is N-methyl-exo-THPO, displaying a
ity and the presence of extracellular substrate all of which 10-fold selectivity toward the glial carrier compared to the neu-
can exert changes in the rates of endocytosis and exocytosis ronal GAT (Madsen et al. 2007). The knowledge obtained from
and/or the number of readily available transporters for recy- the cloning of the four GATs and their localization suggests
cling (Hu and Quick 2008; Madsen et al. 2007). Activation that GAT1 should be responsible for the majority of neuronal
of PKC-dependent phosphorylation decreases the function reuptake, whereas GAT3 should primarily be responsible for
of GAT1 by an increase in the rate of endocytosis and direct the uptake into astrocytes. Therefore, the finding that the glial
tyrosine phosphorylation of GAT1 decreases the rate of endo- preferring inhibitors selectively inhibited GAT1 was surpris-
cytosis whether it is activated by brain derived neurotrophic ing. However, as indicated by Vaz et al. (2011) as much as 40%
factor or short exposures to carrier substrates (Hu and Quick of GABA uptake in astrocyte cultures is facilitated by GAT1,
2008; Madsen et al. 2007; Vaz et al. 2011). Brain-derived neu- offering an explanation to the GAT1 and glial selective inhibi-
rotrophic factor affects the function of GAT1 in both neurons tors. A question remains as to what mechanism is responsible
and astrocytes but not the function of GAT3 (Vaz et al. 2011). for the glial selectivity when considering that the majority of
Prolonged exposure for hours rather than minutes to higher GAT1 is expressed on neurons? To obtain glial selective GAT
concentrations of GAT substrates causes an increase in the inhibitors rational drug design approaches also suggest target-
rate of internalization and a decrease in the recycling pool of ing the GAT3 subtype. Unfortunately, such an endeavor has
GAT1 mediated via a GABA receptor effect through serine not yet been successful. The compound SNAP-5114 is almost
phosphorylation (Hu and Quick 2008). equipotent at GAT2, GAT3, and BGT1 (Dalby 2000), which
makes this drug less optimal to investigate the importance of
the GAT3 subtype in relation to seizure management.
3.7 C E L LU L A R L O C A L I Z AT I O N
O F T R A N S P O RT E R S
The GABA transporters GAT1 and GAT3 which are respon- 4 G LYC I N E A N D D -S E R I N E A S
sible for the majority of GABA uptake in the brain are also TR ANSMITTER S AND THE ROLE
restricted to this organ, whereas BGT1 and GAT2 are found OF ASTROCY TES
in multiple other organs like liver and kidney. Expression of
GAT1 is along GABAergic pathways primarily on presynaptic Glycine serves as the major inhibitory transmitter in the spi-
GABAergic neurons but also on distal astrocytic processes (see nal cord (Curtis and Johnston 1974) and in keeping with this,
Fig. 35.4) adjacent to GABAergic synapse formations (Madsen a high affinity transport system was described in spinal cord

452 • FUNCTIONS OF NEUROGLIAL CELLS


( Johnston and Iversen 1971). It is also clear that glycine trans- transporters for glutamate and GABA are intimately associ-
port is associated with astrocytes and the cloning and struc- ated with the metabolic processes and the exchange between
tural characterization of the high affinity glycine transporters neurons and astrocytes of the transmitters and their precursor.
have identified specific transporters in neurons and astrocytes As the transport processes are also coupled to energy metabo-
(for references, see Eulenburg and Gomeza 2010; Zafra and lism due to the dependency of ATP supply it will be important
Gimenez 2008). It appears that the glial glycine transporter 1 to establish a better understanding of the mechanisms involved
(GlyT1) is of vital importance because GlyT1 knockout mice in regulation of this coupling between transport and energy
die shortly after birth (Gomeza et al. 2003; Eulenburg et al. metabolism. In this context it appears that astrocytic glycogen
2010). This glycine transporter also appears to be an interesting metabolism is particularly interesting (Obel et al. 2012).
pharmacological target because it plays a role in neuropsychi-
atric disorders and dementia (Gether et al. 2006; Kristensen
et al. 2011). It is of particular interest that glycine in addition REFERENCES
to acting as an inhibitory neurotransmitter in its own right
Aoki C, Milner TA, Berger SB, Sheu KF, Blass JP, Pickel VM. 1987. Glial
functions as a coagonist at NMDA receptors (Eulenburg and glutamate dehydrogenase: ultrastructural localization and regional
Gomeza 2010) and the glycine transporters therefore have a distribution in relation to the mitochondrial enzyme, cytochrome
functional role in the regulation of the activity of excitatory oxidase. J Neurosci Res 18:305–318.
neurotransmission (Eulenburg and Gomeza 2010; Zafra and Bak LK, Schousboe A, Waagepetersen HS. 2006. The glutamate/
Gimenez 2008). GABA-glutamine cycle: aspects of transport, neurotransmitter
homeostasis and ammonia transfer. J Neurochem 98:641–653.
It is of considerable interest that the D-amino acid, D-serine
Bak LK, Sickmann HM, Schousboe A, Waagepetersen HS. 2005. Activity
has been identified as a coagonist at NMDA receptors (for of the lactate-alanine shuttle is independent of glutamate-glutamine
references, see Oliet and Mothet 2009; Pollegioni and Sacchi cycle activity in cerebellar neuronal-astrocytic cultures. J Neurosci
2010; Wolosker 2007; Wolosker et al. 2008;). Originally, Res 79:88–96.
D-serine was believed to be primarily if not exclusively synthe- Balkrishna S, Broer A, Kingsland A, Broer S. 2010. Rapid downregula-
sized in astroglia by the enzyme serine racemase (Pollegioni and tion of the rat glutamine transporter SNAT3 by a caveolin-dependent
trafficking mechanism in Xenopus laevis oocytes. Am J Physiol-Cell
Sacchi 2010), but there is considerable evidence that neurons Physiol 299:C1047–C1057.
also play a role in this context (Wolosker 2007) and the origi- Bauer DE, Jackson JG, Genda EN, Montoya MM, Yudkoff M, Robinson
nal term characterizing D-serine as a gliotransmitter probably MB. 2012 The glutamate transporter, GLAST, participates in a
needs to be modified (Wolosker 2007). Nevertheless, there is macromolecular complex that supports glutamate metabolism.
no question that astrocytes play an important role in regulat- Neurochem Int Jan 28 [Epub ahead of print].
Berkich DA, Ola MS, Cole J, Sweatt AJ, Hutson SM, LaNoue KF.
ing NMDA receptor activity via the two coagonists glycine 2007. Mitochondrial transport proteins of the brain. Journal of
and D-serine, albeit the release mechanism for the two amino Neuroscience Research 85:3367–3377.
acids from astrocytes may not be fully understood and char- Bixel M, Shimomura Y, Hutson S, Hamprecht B. 2001. Distribution
acterized (Hamilton and Attwell 2010). It may be of interest of key enzymes of branched-chain amino acid metabolism in glial
to note that although glycine can be effectively removed from and neuronal cells in culture. J Histochem Cytochem 49:407–418.
Bonde C, Sarup A, Schousboe A, Gegelashvili G, Zimmer J, Noraberg J.
the synaptic cleft by the high affinity transporters, no such sys- 2003. Neurotoxic and neuroprotective effects of the glutamate trans-
tems have been identified for D-serine (Wolosker et al. 2008). porter inhibitor DL-threo-beta-benzyloxyaspartate (DL-TBOA)
For this reason, it is believed that D-serine more efficiently during physiological and ischemia-like conditions. Neurochem Int
compared to glycine can reach the synaptic receptors be evad- 43:371–380.
ing the transport systems (Wolosker et al. 2008). Bowman CL, Kimelberg HK. 1984. Excitatory amino acids directly depo-
larize rat brain astrocytes in primary culture. Nature 311:656–659.
Further discussions of gliotransmitters may be found in Bröer A, Deitmer JW, Bröer S. 2004. Astroglial glutamine transport by
chapter 17. system N is upregulated by glutamate. Glia 48:298–310.
Choi DW. 1988. Glutamate neurotoxicity and diseases of the nervous
system. Neuron 1:623–634.
Cholet N, Pellerin L, Magistretti PJ, Hamel E. 2002. Similar perisynap-
5 S U M M A RY A N D P E R S P E C T I VE S tic glial localization for the Na+,K+-ATPase alpha 2 subunit and the
glutamate transporters GLAST and GLT-1 in the rat somatosensory
Because neurons are incapable of performing a de novo synthe- cortex. Cereb Cortex 12:515–525.
sis of glutamate and GABA from glucose because of the lack of Conti F, Melone M. 2006. The glutamine commute: lost in the tube?
pyruvate carboxylase (PC), a symbiotic relationship between Neurochem Int 48:459–464.
Cooper AJ, Plum F. 1987. Biochemistry and physiology of brain ammo-
neurons and astrocytes exists allowing transfer of glutamine, nia. Physiol Rev 67:440–519.
the main precursor for these amino acids, from astrocytes to Curtis DR, Johnston GA. 1974. Amino acid transmitters in the mam-
neurons. Astrocytes, which express PC as well as glutamine malian central nervous system. Ergeb Physiol 69:97–188.
synthetase, that is, the key enzymes allowing de novo synthesis Dalby NO. 2000. GABA-level increasing and anticonvulsant effects
of glutamine from glucose, are therefore of paramount impor- of three different GABA uptake inhibitors. Neuropharmacology
39:2399–2407.
tance for the maintenance of excitatory and inhibitory neu-
Danbolt NC. 2001. Glutamate uptake. Prog Neurobiol 65:1–105.
rotransmission. Regulatory mechanisms related to this will Danbolt NC, Pines G, Kanner BI. 1990. Purification and reconstitution
be in focus for future studies of the metabolism of glutamate of the sodium- and potassium-coupled glutamate transport glycopro-
and GABA. It should be kept in mind that the high affinity tein from rat brain. Biochemistry 29:6734–6740.

A M I N O AC I D N E U R OT R A N S M I T T E R SY N T H E S I S A N D R E M O VA L • 453
Danbolt NC, Storm-Mathisen J, Kanner BI. 1992. An [Na+ + K+] Kristensen AS, Andersen J, Jorgensen TN, Sorensen L, Eriksen J, Loland
coupled L-glutamate transporter purified from rat brain is located in CJ, et al. SLC6 neurotransmitter transporters: structure, function,
glial cell processes. Neuroscience 51:295–310. and regulation. Pharmacol Rev 63:585–640.
Deitmer JW, Broer A, Broer S. 2003. Glutamine efflux from astrocytes is Larsson OM, Gram L, Schousboe I, Schousboe A. 1986. Differential
mediated by multiple pathways. J Neurochem 87:127–135. effect of gamma-vinyl GABA and valproate on GABA-transaminase
Divac I, Fonnum F, Storm-Mathisen J. 1977. High affinity uptake of glu- from cultured neurones and astrocytes. Neuropharmacology
tamate in terminals of corticostriatal axons. Nature 266:377–378. 25:617–625.
Drejer J, Meier E, Schousboe A. 1983. Novel neuron-related regulatory Lehre AC, Rowley NM, Zhou Y, Holmseth S, Guo C, Holen T, et al.
mechanisms for astrocytic glutamate and GABA high affi nity uptake. 2011. Deletion of the betaine-GABA transporter (BGT1; slc6a12)
Neurosci Lett 37:301–306. gene does not affect seizure thresholds of adult mice. Epilepsy Res
Elliott KA, van Gelder NM. 1958. Occlusion and metabolism of gam- 95:70–81.
ma-aminobutyric acid by brain tissue. J Neurochem 3:28–40. Levy LM. 2002. Structure, function and regulation of glutamate trans-
Eulenburg V, Gomeza J. 2010. Neurotransmitter transporters expressed porters. In: Egebjerg J, Schousboe A, Krogsgaard-Larsen P (eds.),
in glial cells as regulators of synapse function. Brain Res Rev Glutamate and GABA receptors and transporters: structure, func-
63:103–112. tion and pharmacology. London: Taylor and Francis 307–336.
Eulenburg V, Retiounskaia M, Papadopoulos T, Gomeza J, Betz H. 2010. Levy LM, Lehre KP, Rolstad B, Danbolt NC. 1993. A monoclonal anti-
Glial glycine transporter 1 function is essential for early postnatal body raised against an [Na(+)+K+]coupled L-glutamate transporter
survival but dispensable in adult mice. Glia 58:1066–1073. purified from rat brain confirms glial cell localization. FEBS Lett
Fiermonte G, Palmieri L, Todisco S, Agrimi G, Palmieri F, Walker 317:79–84.
JE. 2002. Identification of the mitochondrial glutamate trans- Levy LM, Warr O, Attwell D. 1998. Stoichiometry of the glial gluta-
porter. Bacterial expression, reconstitution, functional characteri- mate transporter GLT-1 expressed inducibly in a Chinese hamster
zation, and tissue distribution of two human isoforms. J Biol Chem ovary cell line selected for low endogenous Na+-dependent glutamate
277:19289–19294. uptake. J Neurosci 18:9620–9628.
Furness DN, Dehnes Y, Akhtar AQ, Rossi DJ, Hamann M, Grutle Lieth E, LaNoue KF, Berkich DA, Xu B, Ratz M, Taylor C, et al. 2001.
NJ, et al. 2008. A quantitative assessment of glutamate uptake into Nitrogen shuttling between neurons and glial cells during glutamate
hippocampal synaptic terminals and astrocytes: new insights into synthesis. J Neurochem 76:1712–1723.
a neuronal role for excitatory amino acid transporter 2 (EAAT2). Lippert B, Metcalf BW, Jung MJ, Casara P. 1977. 4-amino-hex-5-enoic
Neuroscience 157:80–94. acid, a selective catalytic inhibitor of 4-aminobutyric-acid amin-
Gegelashvili G, Danbolt NC, Schousboe A. 1997. Neuronal soluble factors otransferase in mammalian brain. Eur J Biochem 74:441–445.
differentially regulate the expression of the GLT1 and GLAST gluta- Liu QR, Lopez-Corcuera B, Mandiyan S, Nelson H, Nelson N. 1993.
mate transporters in cultured astroglia. J Neurochem 69:2612–2615. Molecular characterization of four pharmacologically distinct gam-
Gegelashvili G, Schousboe A. 1997. High affi nity glutamate transport- ma-aminobutyric acid transporters in mouse brain [corrected]. J Biol
ers: regulation of expression and activity. Mol Pharmacol 52:6–15. Chem 268:2106–2112.
Gegelashvili G, Schousboe A. 1998. Cellular distribution and kinetic Lovatt D, Sonnewald U, Waagepetersen HS, Schousboe A, He W, Lin
properties of high-affinity glutamate transporters. Brain Res Bull JH, et al. 2007. The transcriptome and metabolic gene signature
45:233–238. of protoplasmic astrocytes in the adult murine cortex. J Neurosci
Genda EN, Jackson JG, Sheldon AL, Locke SF, Greco TM, O’Donnell 27:12255–12266.
JC, et al. 2011. Co-compartmentalization of the astroglial glutamate Madsen KK, White HS, Clausen RP, Frølund B, Larsson OM,
transporter, GLT-1, with glycolytic enzymes and mitochondria. J Krogsgaard-Larsen P, et al. 2007. Functional and pharmacologi-
Neurosci 31:18275–18288. cal aspects of GABA transporters. In: Lajtha A (ed.), Handbook of
Gether U, Andersen PH, Larsson OM, Schousboe A. 2006. neurochemistry and molecular neurobiology. Berlin: Springer Verlag
Neurotransmitter transporters: molecular function of important 285–303.
drug targets. Trends Pharmacol Sci 27:375–383. McKenna MC, Sonnewald U, Huang X, Stevenson J, Zielke HR. 1996.
Gomeza J, Hulsmann S, Ohno K, Eulenburg V, Szoke K, Richter D, et al. Exogenous glutamate concentration regulates the metabolic fate of
2003. Inactivation of the glycine transporter 1 gene discloses vital role glutamate in astrocytes. J Neurochem 66:386–393.
of glial glycine uptake in glycinergic inhibition. Neuron 40:785–796. Miller JW, Kleven DT, Domin BA, Fremeau RT, Jr. 2002. Cloned
Gram L. 1990. Vigabatrin. In: Dam M, Gram L (eds.), Comprehensive Sodium- (and Chloride-) Dependent high-affinity transporters for
epileptology. New York: Raven Press, pp. 631–640. gaba, glycine, proline, betaine, taurine, and creatine. In: Reith MEA
Guastella J, Nelson N, Nelson H, Czyzyk L, Keynan S, Miedel MC, (ed.), Neurotransmitter transporters: structure, function, and regula-
et al. 1990. Cloning and expression of a rat brain GABA transporter. tion. Totowa, NJ: Humana Press, pp. 101–150.
Science 249:1303–1306. Morland C, Boldingh KA, Iversen EG, Hassel B. 2004. Valproate is neu-
Hamilton NB, Attwell D. 2010. Do astrocytes really exocytose neu- roprotective against malonate toxicity in rat striatum: an association
rotransmitters? Nat Rev Neurosci 11:227–238. with augmentation of high-affinity glutamate uptake. J Cereb Blood
Hu J, Quick MW. 2008. Substrate-mediated regulation of gam- Flow Metab 24:1226–1234.
ma-aminobutyric acid transporter 1 in rat brain. Neuropharmacology Müller T, Moller T, Berger T, Schnitzer J, Kettenmann H. 1992. Calcium
54:309–318. entry through kainate receptors and resulting potassium-channel
Huxtable RJ. 1992. Physiological actions of taurine. Physiol Rev blockade in Bergmann glial cells. Science 256:1563–1566.
72:101–163. Nielsen EB, Suzdak PD, Andersen KE, Knutsen LJ, Sonnewald U,
Iversen LL, Neal MJ. 1968. The uptake of [3H]GABA by slices of rat Braestrup C. 1991. Characterization of tiagabine (NO-328), a new
cerebral cortex. J Neurochem 15:1141–1149. potent and selective GABA uptake inhibitor. Eur J Pharmacol
Johnston GA, Iversen LL. 1971. Glycine uptake in rat central nervous 196:257–266.
system slices and homogenates: evidence for different uptake systems Norenberg MD, Martinez-Hernandez A. 1979. Fine structural locali-
in spinal cord and cerebral cortex. J Neurochem 18:1951–1961. zation of glutamine synthetase in astrocytes of rat brain. Brain Res
Kanamori K, Ross BD. 2004. Quantitative determination of extracel- 161:303–310.
lular glutamine concentration in rat brain, and its elevation in vivo Obel LF, Muller MS, Walls AB, Sickmann HM, Bak LK, Waagepetersen
by system A transport inhibitor, alpha-(methylamino)isobutyrate. HS, et al. 2012. Brain glycogen-new perspectives on its metabolic
J Neurochem 90:203–210. function and regulation at the subcellular level. Front Neuro-
Kettenmann H, Backus KH, Schachner M. 1984. Aspartate, gluta- energetics 4:3.
mate and gamma-aminobutyric acid depolarize cultured astrocytes. Oja SS, Saransaari P. 2007. Taurine. In: Oja SS, Schousboe A, Saransaari
Neurosci Lett 52:25–29. P (eds.), Handbook of neurochemistry and molecular neurobiology,

454 • FUNCTIONS OF NEUROGLIAL CELLS


3rd ed.: Amino acids and peptides in the nervous system. Berlin: Rose EM, Koo JCP, Antflick JE, Ahmed SM, Angers S, Hampson DR.
Springer-Verlag 155–206. 2009. Glutamate Transporter Coupling to Na,K-ATPase. Journal of
Oliet SH, Mothet JP. 2009. Regulation of N-methyl-D-aspartate recep- Neuroscience 29:8143–8155.
tors by astrocytic D-serine. Neuroscience 158:275–283. Rossi DJ, Oshima T, Attwell D. 2000. Glutamate release in severe brain
O’Shea RD, Fodera MV, Aprico K, Dehnes Y, Danbolt NC, Crawford ischaemia is mainly by reversed uptake. Nature 403:316–321.
D, et al. 2002. Evaluation of drugs acting at glutamate transporters in Rothstein JD, Martin L, Levey AI, Dykes-Hoberg M, Jin L, Wu D, et al.
organotypic hippocampal cultures: new evidence on substrates and 1994. Localization of neuronal and glial glutamate transporters.
blockers in excitotoxicity. Neurochem Res 27:5–13. Neuron 13:713–725.
O’Shea RD, Lau CL, Farso MC, Diwakarla S, Zagami CJ, Svendsen Sarup A, Larsson OM, Schousboe A. 2003. GABA transporters and
BB, et al. 2006. Effects of lipopolysaccharide on glial phenotype and GABA-transaminase as drug targets. Curr Drug Targets CNS Neurol
activity of glutamate transporters: evidence for delayed up-regulation Disord 2:269–277.
and redistribution of GLT-1. Neurochem Int 48:604–610. Schlag BD, Vondrasek JR, Munir M, Kalandadze A, Zelenaia OA,
Oz G, Berkich DA, Henry PG, Xu Y, LaNoue K, Hutson SM, et al. Rothstein JD, et al. 1998. Regulation of the glial Na+-dependent
2004. Neuroglial metabolism in the awake rat brain: CO2 fi xation glutamate transporters by cyclic AMP analogs and neurons. Mol
increases with brain activity. J Neurosci 24:11273–11279. Pharmacol 53:355–369.
Pamiljans V, Krishnaswamy PR, Dumville G, Meister A. 1962. Studies Schousboe A. 1981. Transport and metabolism of glutamate and GABA
on the mechanism of glutamine synthesis; isolation and properties of in neurons are glial cells. Int Rev Neurobiol 22:1–45.
the enzyme from sheep brain. Biochemistry 1:153–158. Schousboe A. 2000. Pharmacological and functional characteriza-
Pan Y, Gerasimov MR, Kvist T, Wellendorph P, Madsen KK, Pera E, tion of astrocytic GABA transport: a short review. Neurochem Res
et al. 2012. (1S, 3S)-3-amino-4-difluoromethylenyl-1-cyclopentanoic 25:1241–1244.
acid (CPP-115), a potent gamma-aminobutyric acid aminotransferase Schousboe A, Sickmann HM, Bak LK, Schousboe I, Jajo FS, Faek SAA,
inactivator for the treatment of cocaine addiction. J Med Chem et al. 2011. Neuron-glia interactions in glutamatergic neurotrans-
55:357–366. mission: roles of oxidative and glycolytic adenosine triphosphate as
Parpura V, Heneka MT, Montana V, Oliet SH, Schousboe A, Haydon energy source. J Neurosci Res 89:1926–1934.
PG, et al. 2012. Glial cells in (patho)physiology. J Neurochem Schousboe A, Waagepetersen HS. 2008. GABA neurotransmission:
121:4–27. an overview. In: Lajtha A (ed.), Handbook of neurochemistry and
Phillis JW, Ren J, O’Regan MH. 2000. Transporter reversal as a molecular neurobiology. New York: Springer. pp. 213–226.
mechanism of glutamate release from the ischemic rat cerebral Schousboe A, Wu JY, Roberts E. 1974. Subunit structure and kinetic
cortex: studies with DL-threo-beta-benzyloxyaspartate. Brain Res properties of 4-aminobutyrate-2-ketoglutarate transaminase purified
868:105–112. from mouse brain. J Neurochem 23:1189–1195.
Plachez C, Danbolt NC, Recasens M. 2000. Transient expression of the Shimamoto K, Lebrun B, Yasuda-Kamatani Y, Sakaitani M, Shigeri Y,
glial glutamate transporters GLAST and GLT in hippocampal neu- Yumoto N, et al. 1998. DL-threo-beta-benzyloxyaspartate, a potent
rons in primary culture. J Neurosci Res 59:587–593. blocker of excitatory amino acid transporters. Mol Pharmacol
Plaitakis A, Spanaki C, Mastorodemos V, Zaganas I. 2003. Study 53:195–201.
of structure-function relationships in human glutamate dehydro- Sibson NR, Mason GF, Shen J, Cline GW, Herskovits AZ, Wall JE, et al.
genases reveals novel molecular mechanisms for the regulation of 2001. In vivo (13)C NMR measurement of neurotransmitter glu-
the nerve tissue-specific (GLUD2) isoenzyme. Neurochem Int 43: tamate cycling, anaplerosis and TCA cycle flux in rat brain during.
401–410. J Neurochem 76:975–989.
Pollegioni L, Sacchi S. 2010. Metabolism of the neuromodulator Sidoryk-Wegrzynowicz M, Lee E, Ni MW, Aschner M. 2011. Disruption
D-serine. Cell Mol Life Sci 67:2387–2404. of Astrocytic Glutamine Turnover by Manganese Is Mediated by the
Provent P, Kickler N, Barbier EL, Bergerot A, Farion R, Goury S, et al. Protein Kinase C Pathway. Glia 59:1732–1743.
2007. The ammonium-induced increase in rat brain lactate con- Skytt DM, Madsen KK, Pajecka K, Schousboe A, Waagepetersen
centration is rapid and reversible and is compatible with traffick- HS. 2010. Characterization of primary and secondary cultures of
ing and signaling roles for ammonium. J Cereb Blood Flow Metab astrocytes prepared from mouse cerebral cortex. Neurochem Res
27:1830–1840. 35:2043–2052.
Radian R, Bendahan A, Kanner BI. 1986. Purification and identi- Sonnewald U, Westergaard N, Jones P, Taylor A, Bachelard HS,
fication of the functional sodium- and chloride-coupled gam- Schousboe A. 1996. Metabolism of [U-13C5] glutamine in cultured
ma-aminobutyric acid transport glycoprotein from rat brain. J Biol astrocytes studied by NMR spectroscopy: first evidence of astrocytic
Chem 261:15437–15441. pyruvate recycling. J Neurochem 67:2566–2572.
Rae C, Hare N, Bubb WA, McEwan SR, Broer A, McQuillan JA, et al. Stern JR, Eggleston LV, Hems R, Krebs HA. 1949. Accumulation of glu-
2003. Inhibition of glutamine transport depletes glutamate and tamic acid in isolated brain tissue. Biochem J 44:410–418.
GABA neurotransmitter pools: further evidence for metabolic com- Swanson RA, Liu J, Miller JW, Rothstein JD, Farrell K, Stein BA, et al.
partmentation. J Neurochem 85:503–514. 1997. Neuronal regulation of glutamate transporter subtype expres-
Ramos M, del AA, Pardo B, Martinez-Serrano A, Martinez-Morales sion in astrocytes. J Neurosci 17:932–940.
JR, Kobayashi K, et al. 2003. Developmental changes in the Takanaga H, Ohtsuki S, Hosoya K, Terasaki T. 2001. GAT2/BGT-1
Ca2+-regulated mitochondrial aspartate-glutamate carrier aralar1 as a system responsible for the transport of gamma-aminobutyric
in brain and prominent expression in the spinal cord. Brain Res Dev acid at the mouse blood-brain barrier. J Cereb Blood Flow Metab
Brain Res 143:33–46. 21:1232–1239.
Robinson MB. 2002. Regulated trafficking of neurotransmitter trans- Tong G, Jahr CE. 1994. Block of glutamate transporters potentiates
porters: common notes but different melodies. J Neurochem postsynaptic excitation. Neuron 13:1195–1203.
80:1–11. van den Berg CJ, Garfinkel D. 1971. A stimulation study of brain com-
Robinson MB, Dowd LA. 1997. Heterogeneity and functional proper- partments. Metabolism of glutamate and related substances in mouse
ties of subtypes of sodium-dependent glutamate transporters in the brain. Biochem J 123:211–218.
mammalian central nervous system. Adv Pharmacol 37:69–115. van Landeghem FK, Stover JF, Bechmann I, Bruck W, Unterberg A,
Rose CR, Ransom BR. 1996. Mechanisms of H+ and Na+ changes Buhrer C, et al. 2001. Early expression of glutamate transporter pro-
induced by glutamate, kainate, and D-aspartate in rat hippocampal teins in ramified microglia after controlled cortical impact injury in
astrocytes. J Neurosci 16:5393–5404. the rat. Glia 35:167–179.
Rose CR, Waxman SG, Ransom BR. 1998. Effects of glucose depriva- Vaz SH, Jorgensen TN, Cristovao-Ferreira S, Duflot S, Ribeiro JA,
tion, chemical hypoxia, and simulated ischemia on Na+ homeostasis Gether U, et al. 2011. Brain-derived neurotrophic factor (BDNF)
in rat spinal cord astrocytes. J Neurosci 18:3554–3562. enhances GABA transport by modulating the trafficking of GABA

A M I N O AC I D N E U R OT R A N S M I T T E R SY N T H E S I S A N D R E M O VA L • 455
transporter-1 (GAT-1) from the plasma membrane of rat cortical Wolosker H, Dumin E, Balan L, Foltyn VN. 2008. D-amino acids in the
astrocytes. J Biol Chem 286:40464–40476. brain: D-serine in neurotransmission and neurodegeneration. FEBS
Waagepetersen HS, Qu H, Sonnewald U, Shimamoto K, Schousboe A. J 275:3514–3526.
2005. Role of glutamine and neuronal glutamate uptake in glutamate Yamashita A, Singh SK, Kawate T, Jin Y, Gouaux E. 2005. Crystal struc-
homeostasis and synthesis during vesicular release in cultured gluta- ture of a bacterial homologue of Na+/Cl–dependent neurotransmit-
matergic neurons. Neurochem Int 47:92–102. ter transporters. Nature 437:215–223.
Waagepetersen HS, Sonnewald U, Schousboe A. 2009. Energy and Yu AC, Drejer J, Hertz L, Schousboe A. 1983. Pyruvate carboxylase
amino acid neurotransmitter metabolism in astrocytes. In: Parpura activity in primary cultures of astrocytes and neurons. J Neurochem
V, Haydon PG (eds.), Astrocytes in (patho)physiology of the nervous 41:1484–1487.
system. Boston: Springer 177–200. Yudkoff M, Nelson D, Daikhin Y, Erecinska M. 1994. Tricarboxylic acid
Wadiche JI, Arriza JL, Amara SG, Kavanaugh MP. 1995. Kinetics of a cycle in rat brain synaptosomes. Fluxes and interactions with aspar-
human glutamate transporter. Neuron 14:1019–1027. tate aminotransferase and malate/aspartate shuttle. J Biol Chem
Walls AB, Eyjolfsson EM, Smeland OB, Nilsen LH, Schousboe I, 269:27414–27420.
Schousboe A, et al. 2011. Knockout of GAD65 has major impact Zafra F, Gimenez C. 2008. Glycine transporters and synaptic function.
on synaptic GABA synthesized from astrocyte-derived glutamine. IUBMB Life 60:810–817.
J Cereb Blood Flow Metab 31:494–503. Zaganas I, Kanavouras K, Mastorodemos V, Latsoudis H, Spanaki C,
Westergaard N, Drejer J, Schousboe A, Sonnewald U. 1996. Evaluation Plaitakis A. 2009. The human GLUD2 glutamate dehydrogenase:
of the importance of transamination versus deamination in astrocytic localization and functional aspects. Neurochem Int 55:52–63.
metabolism of [U-13C]glutamate. Glia 17:160–168. Zhu XM, Ong WY. 2004. A light and electron microscopic study of
Wolosker H. 2007. NMDA receptor regulation by D-serine: new find- betaine/GABA transporter distribution in the monkey cerebral neo-
ings and perspectives. Mol Neurobiol 36:152–164. cortex and hippocampus. J Neurocytol 33:233–240.

456 • FUNCTIONS OF NEUROGLIAL CELLS


36.
GLYCOGEN AND ENERGY METAB OLISM
Angus Brown

A B B R E VI AT I O N S in both plants and higher organisms developing mechanisms


of storing glucose by polymerization of this ubiquitous car-
aCSF artificial cerebrospinal fluid bohydrate into polysaccharides. In plants this polysaccharide
AMP adenosine monophosphate is amylose and amylopectin, whereas in higher animals it is
ATP adenosine triphosphate the more complex molecule, glycogen (Voet and Voet 2011).
CAP compound action potential Glycogen evolved to act as protection against the unreliable
cAMP cyclic adenosine monophosphate food sources encountered by our hunter-gatherer ancestors.
CNS central nervous system Fat deposits serve a similar purpose in storing excess fat in sub-
DAB 1,4-dideoxy-1,4-imino-D-arabinitol cutaneous depots, whereas glycogen is stored primarily in the
EPSP excitatory postsynaptic potential liver and skeletal muscle.
Hz Hertz The glycogen stores in the body are far exceeded by the
LDH lactate dehydrogenase fat stores (rough estimates are that fat stores provide energy
MCT monocarboxylate transporter for ~35 days during starvation, whereas total body glyco-
mM milli moles per liter gen provides <1 days worth of energy). Glycogen does not
MON mouse optic nerve contain as much energy as the more reduced fat; therefore,
mRNA messenger ribonucleic acid why does the body store glycogen at all? The answer is that:
ms millisecond (1) glycogen is much more rapidly metabolized than fat,
NMR nuclear magnetic resonance and can thus provide energy substrate at very short notice;
OGI oxygen uptake ratio (2) glycogen can be metabolized in the absence of oxygen,
Pa active phosphorylase a a key consideration for exercising muscle, whereas fat can-
Pb inactive phosphorylase b not; and (3) animals cannot convert fat to glucose; thus, for
PNS peripheral nervous system glucose-dependent organs such as the brain, fat cannot pro-
PP1 protein phosphatase 1 vide essential glucose (Voet and Voet 2011). The dependence
TCA tricarboxylic acid of the brain on glucose provides an explanation of the advan-
T2D type 2 diabetes tages of glycogen as an energy store. As the body’s most vora-
UDP uridine diphosphate cious glucose consumer, per unit weight, the brain takes up
UTP uridine triphosphate 20% of available glucose, although it represents only 2% of
ZDF Zucker diabetic fatty body weight (McKenna et al. 2006).
ZL Zucker lean The brain can receive glucose from three main sources:
ZOb Zucker obese (1) blood-borne glucose provided by dietary intake of carbo-
hydrates, (2) gluconeogenesis, whereby nonglucose energy
substrates are converted to glucose in the liver and kidney, and
(3) metabolism of glycogen. Given our ancestors’ unpredict-
1 INTRODUCTION able supply of food, and the slow rate of substrate conversion
associated with gluconeogenesis in response to falling blood
The main energy support of the brain is glucose, which must glucose levels, brain glycogen is the ideal energy reserve to
be delivered continuously and in excess of demand for the provide for a sudden shortfall in glucose. A curious feature
brain to function normally. Interruption of blood-borne glu- of brain glycogen is its exclusive localization to astrocytes in
cose and oxygen results in brain dysfunction within seconds, adult brain (Cataldo and Broadwell 1986). This feature, and
highlighting the extreme dependency of the mammalian cen- the astrocyte’s inability to enzymatically cleave phosphate
tral nervous system (CNS) on these energy substrates (Rossen from glucose residues, could well have contributed to the
et al. 1943). Drastic reduction of glucose alone, in the con- early prejudice that brain glycogen was not important. The
tinued presence of oxygen, induces neural dysfunction more importance of astrocyte glycogen and the unique fashion of
slowly, and the presence of brain glycogen is a major reason its metabolism in support of brain energy needs are the sub-
for this, as we shall see. Evolutionary pressures have resulted ject of this chapter.

457
2 G LYC O G E N B I O C H E M I S T RY formation of branch points via D(1→6) glycosidic bonds
(Voet and Voet 2011). Glycogen synthesis is initiated by an
2.1 G LYC O G E N A S A C A R B O H Y D R AT E existing glycogen molecule, or by glycogenin, a protein that
S TO R AG E D E P OT can accept glucose molecules, but only after tyrosine binds to
Glycogen is formed from molecules of D-glucose, a hexose, the glycogenin and activates the site of glucose attachment.
and is a branched homopolysaccharide. D-glucose is a pyranose The process of glucose attachment liberates 1 molecule of H2O
closed structure with the six carbon atoms arranged in a hex- per glucose molecule, and thus the MW of each glucosyl resi-
agonal formation (Berg et al. 2006). The main bonds formed due is 162 compared with 180 for glucose. After attachment of
between glucose molecules in glycogen are D(1→4) glycosidic a few glucosyl residues to glycogenin, the structure can act as a
bonds, which result in a linear structure. In plants, the primary primer that glycogen synthase can act on. A first step in glyco-
storage form of glucose, amylose, is formed entirely of glucose gen formation is the combination of glucose-1-phosphate with
molecules connected by D(1→4) bonds. This results in a linear uridine triphosphate (UTP), to form UDP-glucose. This mol-
structure with the disadvantages of being poorly water-soluble ecule is the substrate for glycogen synthase, which can only
and having only a limited number of locations onto which glu- act on existing glycogen molecules. Increasing the size of the
cose molecules can be attached. Glycogen has D(1→6) branch glycogen molecule requires the movement of glucose from
points that defeat these problems. Glycogen molecules can be the UDP-glucose molecule to the growing chain, forming an
up to 108 Daltons and contain 120,000 glucose units (Voet D(1→4) glycosidic bond between the hydroxyl group on car-
and Voet 2011), their structure a discrete self-contained gran- bon 4 of the terminal glucosyl residue, and the carbon 1 of
ule, between 10 and 40 nm in diameter, located in the cyto- the new glucosyl residue to be added. The UDP released in
plasm. The enzymes responsible for metabolism and synthesis this action is converted back to UTP, an adenosine triphos-
of glycogen are also present in the granule (Voet and Voet phate (ATP)–requiring step. Repetition of this reaction adds
2011; Wender et al. 2000) (Fig. 36.1). increasing numbers of glucosyl residues to the glycogen mol-
ecule. If this were the only reaction that occurred, then the
linear molecule amylose would be formed. However, in mam-
2.1.1 Synthesis and Degradation mals at about every eighth to tenth glucosyl residue a D(1→6)
Glycogen synthesis is a three step process, requiring: bond is inserted that results in branching of the molecule.
(1) formation of UDP-glucose via the combination of The branches are formed by the action of the enzyme amylo-
glucose-1-phosphate and uridine triphosphate (UTP); (2) (1,4→1,6)-transglycolase, which attaches a branch of up to a
transfer of glucosyl residues from UDP-glucose to the gly- dozen glucosyl residues to the end of the chain by breaking
cogen molecule forming D(1→4) glycosidic bonds; and (3) an D(1→4) bond. Glycogen synthase then adds glucosyl res-
idues to each of the ends of the chain. A key action of this
branching process is to create multiple ends to which, or from
which, glucosyl residues can be attached or removed, respec-
tively. This makes glycogen a very dynamic molecule, which
in time of need can release multiple glucosyl residues simulta-
neously, or to which multiple glucosyl residues can be added
simultaneously.
Glycogen metabolism requires the coordinated action of
three enzymes to complete the following steps: (1) release of
glucose-1-phosphate from glycogen; (2) restructuring of the
glycogen molecule so it is available for further metabolism; and
(3) conversion of glucose-6-phosphate to glucose-1-phosphate
(Berg et al. 2006). Glycogen is degraded, or metabolized,
by the enzyme glycogen phosphorylase. A key aspect of
glycogen metabolism is that degradation is not simply a rever-
sal of synthesis using the same enzymes, but rather is a com-
pletely separate biochemical pathway using a separate set of
enzymes. Thus, degradation and synthesis can occur simul-
taneously endowing glycogen with an increased flexibility
depending on prevailing conditions. The product of glycogen
breakdown in the periphery is either glucose-1-phosphate,
the result of breakdown of D(1→4) bonds, or free glucose,
the result of breakdown of D(1→6) bonds. In the brain each of
these compounds will form glucose-6-phosphate via the actions
of phosphoglucomutase or hexokinase, respectively. Given that
astrocytes do not possess the enzyme glucose-6-phosphatase
Figure 36.1 Glycogen Granules Are Present in Astrocytes in Mouse Optic (Wiesinger et al. 1997) glucose cannot be formed, a simi-
Nerve. From Wender et al., 2000. lar situation occurs in skeletal muscle (Voet and Voet 2011),

458 • FUNCTIONS OF NEUROGLIAL CELLS


but in the liver free glucose is formed as a result of glyco- (a) Glycogen
gen metabolism, for subsequent liberation into the systemic Glycogen phosphorylase
circulation. In astrocytes the glucose-6-phosphate is Glycogen(n=1)
glycolytically metabolized ultimately to lactate (see later) Glucose-1-phosphate
(Dringen et al. 1993). Glycogen phosphorylase breaks the Phosphoglucomutase
D(1→4) bonds until only four glucosyl residues extend past
the nearest D(1→6) bond. This short four-residue sec- Glucose-6-phosphate

tion is called limit dextran and is the substrate for


oligo-D(1→4)→D(1→4) glucan transferase, which trans- Pyruvate
Muscle Liver
fers the last three residues to the end of the chain, where brain Glucose-6-phosphatase
glycogen phosphorylase removes the individual residues as
glucose-1-phosphate. The single residue remaining at the Lactate H2O+CO2 Glucose
branch point is removed by amylo-D(1→6)-glucosidase
and released as free glucose. The relative proportion of
glucose-1-phosphate to glucose released as a result of glyco- (b)
gen metabolism depends on the number of glucosyl residues
Adrenaline Glucagon Insulin
between each branch point, but a widely accepted value is
90%. Details of glycogen biochemistry are covered in detail in
standard biochemistry textbooks, of which the following are
recommended (Berg et al. 2006; Garrett and Grisham 1999;
Voet and Voet 2011).
ATP cAMP
Once glucose enters cells it is phosphorylated by hexoki-
nase to glucose-6-phosphate, a step that requires ATP. As PKAb PKAa GSK GSK
previously described, glycogen breakdown ultimately releases
glucose-6-phosphate; thus, it is natural to ask what is the cost PKb PKa
of storing excess glucose as glycogen? Only 1 ATP is required G-1-P
because, depending on the frequency of branch points, about GPb GPa GSa GSb
90% of glycogen is released as glucose-1-phosphate, which is
converted to glucose-6-phosphate by phosphoglucomutase Glycogen
in a reaction that does not require ATP. Conversion of UDP
PP1
to UTP requires ATP, thus oxidation of glucose-6-phosphate
produces 31 molecules of ATP, whereas storage as glycogen Figure 36.2 Metabolism of Glycogen. A. Pathways of glycogen metabo-
only requires 1 ATP per molecule of glucose-6-phosphate, an lism highlighting the tissue-dependent fate of glycogen-derived metabo-
efficiency of about 97% (Fig. 36.2). lites. Key enzymes are in blue. Adapted from Figure 21.3 (Berg et al.
2006). B. Regulation of glycogen metabolism indicating the reciprocal
modulation of glycogen phosphorylase and glycogen synthase via (de)
2.1.2 RO L E O F G LYC O G E N I N L I VE R A N D phosphorylation of key enzymes. Glycogen metabolism is initiated by
S K E L ETA L MUS C L E adrenaline or glucagon increasing cAMP levels via binding to metabotro-
pic receptors. cAMP phosphorylates protein kinase A (PKA), which
The two main depots of glycogen stores in the body are the in turn phosphorylates phosphorylase kinase (PK) to the active a form.
liver and skeletal muscle. Glycogen is present at higher con- PKa phosphorylates glycogen phosphorylase (GP) to its active a form,
centrations in the liver compared with skeletal muscle (10% and additionally phosphorylates glycogen synthase (GS) to its inactive
phosphorylated b form. Phosphorylated GPa promotes glycogen break-
vs 1%–2%), but given the greater mass of skeletal muscle in down to glucose-1-phosphate (G-1-P). Glycogen synthesis is promoted by
the body skeletal muscle contains about 400 g of glycogen and insulin via the action of insulin receptor substrates converting glycogen
the liver 100 g (Champe and Harvey 2008). Limits on the synthase kinase (GSK) from its dephosphorylated to its inactive phos-
amount of glycogen stored are set such that during extended phorylated state. Dephosphorylated GSK maintains GS in its phosphory-
periods of plenty glycogen content remains fixed within this lated inactive state; thus, insulin acts to release GS from this resting level
of inhibition and promotes incorporation of glucosyl residues into the
range. However, pathology resulting from the family of gly- glycogen molecule. Protein phosphatase 1 (PP1) promotes conversion of
cogen storage diseases can result in excess glycogen stored in GS to its active a form, whereas dephosphorylating PK inhibits glycogen
both liver and muscle (Voet and Voet 2011). The role of liver metabolism. Glycogen phosphorylase is also allosterically modulated by
glycogen is to buffer fluctuations in blood glucose levels such AMP, ATP, glucose-6-phosphate, and glucose (not shown).
that during periods of excess, glucose is stored as glycogen
in the liver, but during periods of falling glucose (systemic
blood glucose < 5 mM) glycogen metabolism is mobilized to (cAMP) in the hepatocytes, causing glycogen breakdown,
release glucose into the systemic circulation to maintain eug- which leads to elevated glucose-6-phosphate, the substrate
lycemia (Voet and Voet 2011). Glucose inhibits pancreatic D for glucose-6-phosphatase and ultimately released as glucose.
cells from releasing glucagon; thus, when blood glucose lev- In periods of excess glucose insulin is released, which acts
els fall, this inhibition is released. The glucagon acts on liver on liver cells to trans-locate the Glut-4 transporter from the
cells, resulting in increased cyclic adenosine monophosphate cytosol to the cell membrane to facilitate influx of glucose

G LYC O G E N A N D E N E R GY M ETA B O L I S M • 459


and subsequent storage as glycogen (Voet and Voet 2011). In in which glycogen is metabolized to provide ATP to sustain
the liver, glucose and glucose-6-phosphate act to inhibit the muscle contraction. In the third phase, the intercontraction
action of glycogen phosphorylase. phase, glycogen is resynthesized. Stoichiometric calculations
In muscle, glycogen content is controlled by insulin and (Shulman and Rothman 2001) indicate that metabolism
adrenaline. Adrenaline acts to promote glycogen breakdown of glucose to fuel muscle contraction is more efficient than
by activating a cAMP-dependent cascade involving phospho- the glycogen shunt, producing 2 molecules of ATP versus
rylation, thus resulting in inhibition of glycogen synthesis 1. However, it is the time scale that is important, with the
but promoting glycogen metabolism. Insulin acts to stimulate very rapid enzyme glycogen phosphorylase producing ATP
glycogen synthesis via action on protein phosphatase 1 (PP1). more rapidly than glucose metabolism, with glycogen resyn-
In muscle, glycogen is metabolized to glucose-6-phosphate, thesized between periods of muscle contraction (Shulman
which is subsequently glycolytically metabolized to lactate and Rothman 2001). The glycogen shunt also has been pro-
because muscle cells lack glucose-6-phosphatase. In liver gly- posed for metabolism in the brain. The resting OGI in the
cogen phosphorylase is sensitive to glucose, which deacti- brain is about 5.5, indicative of incomplete glucose oxidation
vates the enzyme. There is 90% homology between liver and (Fox and Raichle 1986). Sensory stimulation of various brain
skeletal muscle phosphorylase, with a key difference being areas leads to an increase in glucose metabolism not matched
that liver glycogen phosphorylase is not sensitive to adenos- but oxygen uptake (Fox and Raichle 1986), with excess lac-
ine monophosphate (AMP), because AMP does not exhibit tate produced (Prichard et al. 1991). These results, as in mus-
the large concentration fluctuations in liver that occur in cle, were explained as glycolytic metabolism of glucose with
skeletal muscle. lactate produced as a waste product. These data suggest that
An emerging aspect of muscle glycogen metabolism activated brain requires little energy above the basal needs of
is that the excess lactate liberated during this process, which was the tissue, which is in disagreement with data showing the
previously considered to be a waste product and converted by majority of energy in the nonstimulated state supports func-
gluconeogenesis in the liver or kidneys, now may have a more tional processes (Shulman et al. 2001). The glycogen shunt
interesting role. Recent human in vivo experiments showed proposes that glycogen, as in muscle, acts to provide energy
that during severe exercise the systemic concentrations of lac- substrate to fuel rapidly firing actions potentials, that is, in
tate can rise tenfold, from 1 to 10 mM, whereas blood glucose the millisecond timescale, and that glycogen is replenished
levels fall to hypoglycemic levels of less than 4 mM. This blood during the interspike interval (Shulman et al. 2001). The
glucose concentration would normally result in symptoms of glycogen-derived energy substrate is lactate, which is shut-
hypoglycemia, but this does not occur. Evidence suggests that tled out of the astrocyte to the neuron where it is oxidatively
under these circumstances the brain takes up and metabolizes metabolized. However, lactate in excess of requirements is
systemic lactate, sparing glucose for muscle metabolism. This produced, and is recorded as an increase in the extracellular
metabolic cooperation suggests that the brain may be more lactate concentration. Based on the stoichiometric analysis
versatile in its use of energy substrates, and the muscle may (Shulman et al. 2001) the net ATP produced relative to the
be more altruistic in providing usable energy substrate than proportion of glucose metabolized via the glycogen shunt is
previously thought. illustrated in Figure 36.3 (Shulman et al. 2001), indicating
that in terms of ATP production, efficiency decreases as the
glycogen shunt contribution increases. However, the same
2.1.3 I N C R E A S E D AWA R E N E S S O F G LYC O G E N
amount of lactate is produced irrespective of its source. This
I N T H E C E N T R A L N E RVO US SYS T E M :
“glycogen shunt” bears many similarities to the astrocyte
T H E G LYC O G E N S H U N T
neuron lactate shuttle hypothesis proposed in 1994 (Pellerin
An unexplained phenomenon in muscle physiology is that and Magistretti 1994), in that astrocyte production of lac-
under conditions of increased tissue energy demand the oxy- tate fuels neural elements in response to increase neuronal
gen usage does not match the glucose uptake; thus, the glu- activity.
cose uptake:oxygen uptake ratio (OGI) falls below the value
of 6 expected for total oxidation of glucose to CO2 and H2O
(Shulman and Rothman 2001). In addition to the OGI falling 3 G LYC O G E N I N T H E C E N T R A L
as low as 3, excess lactate is produced. This was conventionally N E RVO U S SYS T E M
explained as insufficient oxygen availability in muscle during
3.1 A S T RO C Y T I C L O C AT I O N O F
exercise, resulting in incomplete oxidative metabolism of glu-
G LYC O G E N I N A D U LT B R A I N
cose with excess lactate produced as the waste product. The
“glycogen shunt” model proposed by Shulman and Rothman It has been known for many years that the brain does con-
seeks to explain the experimental contradictions obtained in tain glycogen (Nelson et al. 1968). However, it is present in
muscle by suggesting a key role for glycogen during muscle such low quantities relative to liver and skeletal muscle glyco-
contraction (Shulman and Rothman 2001). In this scenario gen, that a role as an energy reserve has been dismissed. This
glycogen is required to provide energy substrate very rapidly is primarily owing to calculations demonstrating that in the
(on the order of milliseconds) to fuel muscle contraction. The absence of glucose brain glycogen could only support func-
first phase of muscle contraction is fueled by phosphocrea- tion for several minutes (Dienel 2009). Recent interest has
tine breakdown to supply ATP, followed by a second phase been rekindled in brain glycogen for reasons described in this

460 • FUNCTIONS OF NEUROGLIAL CELLS


(a) confused picture strongly suggests establishing baseline levels
2 of glycogen in distinct brain regions in commonly used rodent
ATP produced strains, a priority that would clarify the variation of glycogen
expression. In addition it is likely that glycogen content varies
1 with both age and disease state (see later). It has been found
that glycogen content is lowered in T1 and T2 diabetes, but
the effects of other metabolic and neurological conditions are
0
0 0.5 1
mostly unknown. Aside from a handful of studies (Dalsgaard
Fraction to glycogen
et al. 2006), this is an entirely neglected field and worthy of
exploration. Indeed, our recent result of robust glycogen con-
(b) tent in peripheral nerve Schwann cells is the first account of
the presence of glycogen in this tissue.
2
Lactate produced

3.2 E X P R E S S I O N O F R E L EVA N T E N ZY M E S
1
The presence of glycogen can be deduced by the presence
of the enzymes responsible for its metabolism and synthe-
0 sis, glycogen phosphorylase, and glycogen synthase, respec-
0 0.5 1 tively, given the absence of a reliable antibody for glycogen.
Fraction to glycogen Glycogen synthase is present in three isoforms, associated
with muscle, liver, or brain (Pellegri et al. 1996). The brain
Figure 36.3 The ATP and Lactate Production via the Brain Glycogen and liver isoforms are specific for those tissues, but the mus-
Shunt Is Dependent of the Proportion of Glucose Metabolized via Glycogen
(Fraction to Glycogen) A. As the net fraction of glucose metabolized cle isoform is present in most tissues. Brain tissue labeling was
via the glycogen shunt (x) increases the net ATP production (solid line) highest in the cerebellum, hippocampus, and olfactory bulb,
decreases. Net ATP production from glycolysis (dashed line) is 2(1-x) and and at the cellular level, although both astrocytes and neurons
via the glycogen shunt (dotted line) is x. B. The total lactate produced expressed the mRNA, expression was greatest in astrocytes
(solid line) is unaffected by glucose flux via the glycogen shunt as lactate (Pellegri et al. 1996). As previously discussed, glycogen is pre-
production via glycolysis is 2(1-x) and via the glycogen shunt (dotted line)
is 2x. Data adapted from Shulman et al. 2001. dominantly expressed in astrocytes; thus, its expression in
neurons is puzzling. The expression of glycogen synthase does
not necessarily predict glycogen expression, and indeed only
chapter. It is generally accepted that glycogen is present exclu- under pathological conditions is robust neuronal glycogen
sively in astrocytes in the adult brain (Cataldo and Broadwell expression found (Vilchez et al. 2007). Glycogen phospho-
1986), but this must be qualified. It is certainly true that gly- rylase exists as liver, muscle, and brain isoforms. Astrocytes
cogen is expressed in neurons in embryologic tissue, but this express only the muscle and brain isoform, but not the liver
expression decreases with maturity. Glycogen is expressed isoform (Pfeiffer-Guglielmi et al. 2003), suggesting brain gly-
in some neurons in the brainstem, ependyma, and choroid cogen phosphorylase is regulated in a manner similar to mus-
plexus cells (Bloom and Fawcett 1968), and our recent experi- cle (see section 3.3); that is, it is regulated allosterically and
ence (see later) suggests that it may well be expressed in other, requires AMP for full activation, thus it is sensitive to energy
as yet unidentified cell types. A key area worthy of investiga- changes within the cell.
tion is the hypothalamus, given its role in energy regulation.
3.3 R EGU L AT I O N O F G LYC O G E N C O N T E N T
It is interesting to note that glycogen is expressed in tanycytes
in Siberian hamster (Nilaweera et al. 2011), but its expres- Glycogen metabolism is strictly regulated and involves mul-
sion varies depending on the duration of photoperiod the tiple pathways. Although the details of glycogen regulation
hamsters were exposed to, suggesting a specialized role that have not yet been fully described for the brain, given the simi-
does not apply to astrocytic glycogen located elsewhere in the larities of glycogen structure and the homology between the
brain. That glycogen is located predominantly in astrocytes is isoforms of glycogen synthase (Pellegri et al. 1996) and glyco-
beyond question, although this broad description of cell types gen phosphorylase (Pfeiffer-Guglielmi et al. 2003) it is a fairly
based on morphology gives no clue as to any specific function safe assumption that the mechanisms of glycogen regulation
carried out by glycogen in particular subtypes of astrocytes. in the periphery also apply to the brain. Glycogen content is
Different brain areas have different concentrations of glyco- regulated by the hormones glucagon, adrenaline, and insu-
gen, but whether this reflects astrocyte density is unknown. lin, but also allosterically by glucose, glucose-6-phosphate,
Even among different strains of rat glycogen content of the AMP, and ATP, indicators of the cellular energy status.
same brain area varies, illustrated in a recent study, which Broadly speaking, the factors that promote glycogen metabo-
showed similar values for glycogen in the cortex, hippocam- lism also inhibit glycogen synthesis and vice versa, highlight-
pus, and cerebellum of Zucker Lean and Zucker Diabetic ing the advantages of two separate pathways for metabolism
fatty rats, but with much lower glycogen levels measured and synthesis. Glucagon and adrenaline activate glycogen
in Sprague-Dawley rats (Sickmann et al. 2010). This rather phosphorylase and promote glycogen metabolism, whereas

G LYC O G E N A N D E N E R GY M ETA B O L I S M • 461


insulin activates glycogen synthase and promotes storage of changes, resulting in phosphorylase kinase activation. Thus
glucose-6-phosphate as glycogen. muscle contraction, which causes increases in intracellular
Glycogen metabolism requires the combined actions of Ca2+ via release of Ca2+ from the sarcoplasmic reticulum, thus
several enzymes, which (1) degrade glycogen, (2) remodel leads to glycogen metabolism, providing ATP for the energy-
glycogen so it remains viable for degradation, and (3) con- dependent contraction process (Berg et al. 2006; Garrett and
vert glycogen breakdown product into a suitable form for fur- Grisham 1999; Voet and Voet 2011).
ther metabolism. Glycogen phosphorylase exists as an active
phosphorylase, a (Pa) and an inactive phosphorylase b (Pb).
Phosphorylase kinase phosphorylates the inactive Pb to active 4 FUNCTION IN THE CENTR AL
Pa. Phosphorylase kinase itself is converted to its active form N E RVO U S SYS T E M
(phosphorylase kinase a) from phosphorylase kinase b by
phosphorylation by protein kinase. The active form of protein 4 .1 WH I T E M AT T E R : G LYC O G E N S U P P O RT S
kinase a is produced by phosphorylation of protein kinase b by AXO N C O N D U C T I O N I N AG LYC E M I A A N D
cAMP. Protein kinase a also acts to covert the active a form I N C R E A S E D E N E RGY D E M A N D
of glycogen synthase to the inactive b form. Thus, a cascade
of phosphorylation initiated by production of cAMP results Initial studies that drew attention to a potential functional
in glycogen phosphorylase activation and glycogen metabo- role for glycogen suggested that active neurons utilized
lism. The cAMP is produced by the binding of glucagon and astrocytic glycogen. In a tissue culture study, the reliance of
adrenaline to metabotropic receptors and activation of G neuronal survival on the presence of astrocytes was clearly
proteins. Thus, a signaling cascade mediated by hormone bind- demonstrated. More detailed analysis revealed that the key
ing initiates glycogen metabolism (Berg et al. 2006; Voet and factor that sustained the neurons was the glycogen that was
Voet 2011). present in the astrocytes; the greater the glycogen content the
Glycogen synthesis is catalyzed by glycogen synthase, longer the duration of neuronal survival (Swanson and Choi
which is converted from its inactive b form to the active a form 1993). This was an important study for several reasons. First,
by protein phosphatase 1 (PP1). PP1 binds to the glycogen– it corroborated an earlier study, which had demonstrated
glycogen synthase complex and results in production of gly- that cultured neurons incubated in the presence of glial cells
cogen synthase a by dephosphorylating glycogen synthase a, survived longer than neurons grown in the absence of glia
promoting glycogen synthesis. PP1 also dephosphorylates Pa cells, although no mechanism was proposed (Whatley et al.
to Pb, thereby inhibiting glycogen metabolism, and addition- 1981). Second, it directly correlated neuronal survival with
ally dephosphorylates phosphorylase kinase a to phosphorylase the metabolic status of astrocytes. For more than 100 years
kinase b, thereby inhibiting activation of glycogen phosphory- it has been hypothesized that there was an intimate meta-
lase (Berg et al. 2006; Voet and Voet 2011). bolic link between neurons and astrocytes, in which neurons
Thus, the general pattern of regulation is that cAMP were dependent on astrocytes for the supply of metabolic
results in a phosphorylation cascade mediated by kinases, substrate (Andriezen 1893). Third, it suggested an intimate
which results in glycogen metabolism, whereas PP1 causes cell-to-cell metabolic signaling pathway between astrocytes
dephosphorylation of key enzymes promoting glycogen syn- and neurons, in which astrocytes supplied an energy sub-
thesis. Insulin and adrenergic receptors are highly expressed strate to neurons. This implies a great degree of functional
in liver and muscle cells, whereas glucagon receptors are more cooperation between the two cells types, such that the appro-
prevalent in the liver than in muscle. priate transporters and enzymes must be present in each cell
Allosteric modulation of glycogen metabolism is mediated type. In addition, appropriate signaling systems must be pre-
by compounds that are indicative of the energy status of the sent for the neuron to signal the astrocyte when substrate is
cell, namely, glucose, glucose-6-phosphate, AMP, and ATP. In required (see also chapter 27).
muscle cells, AMP activates glycogen phosphorylase, and ATP In a study using autoradiographic techniques, a link was
and glucose-6-phosphate inhibit glycogen phosphorylase, made between sensory stimulation and glycogen use in the
whereas glycogen synthase is activated by glucose-6-phosphate. associated brain area. Under in vivo conditions, stimulation
This demonstrates that indicators of increased energy statues of whiskers in the rat resulted in a decrease in glycogen in
(glucose-6-phosphate and ATP) inhibit glycogen phosphory- the barrel cortex (Swanson et al. 1992). This demonstrated a
lase and activate glycogen synthase; conversely, indicators of metabolic relationship between neuronal activity and glyco-
decreased energy status (AMP) promote glycogen metabolism. gen content under in vivo conditions, but more importantly
In liver, glucose inhibits glycogen phosphorylase and activates showed that there was a relationship between physiological
glycogen synthase, because liver cells express glucose-6-phos- function, in this case afferent sensory input, and glycogen
phatase and convert glycogen-derived glucose-6-phosphate breakdown in the associated cortical area. The contemporary
to glucose. However, muscle cells do not express glucose-6- experiments in the Magistretti lab demonstrating the regula-
phosphatase; thus, glucose-6-phosphate is used as the muscle tion of glycogen content by neurotransmitters (Hof et al. 1988;
energy substrate. In muscle cells, Ca2+ is an important modula- Magistretti 1988; Magistretti et al. 1986; Sorg and Magistretti
tor of phosphorylase kinase. The muscle phosphorylase kinase 1991), suggested CNS glycogen was not a vestigial throwback,
consists of four subunits, one of which, calmodulin, is sensitive but rather a dynamic compound intimately involved in brain
to Ca2+. Binding of Ca2+ to calmodulin causes conformational function.

462 • FUNCTIONS OF NEUROGLIAL CELLS


We took these experiments as the basis for studies we car- A 30
ried out in rat, and subsequently in mouse optic nerve, as the
rodent optic nerve offered a simplified structure, a represen-

Failure latency
20
tative central white matter tract that was composed of myeli-

(mins)
nated axons, astrocytes, and oligodendrocytes, but that lacked
the complications of glutamatergic synapses and neuronal 10
cell bodies (Ransom et al. 1994). The optic nerve affords the
opportunity to probe cell-to-cell metabolic communication 0
in a reduced, simplified model, but that is physiologically rel- 0 2 4 6 8 10
evant, because white matter comprises 50% of human adult Glycogen (pmoles/μg protein)
brain volume (Wang et al. 2008). The tissue is minimally
disrupted during dissection and can be maintained for many B
hours in a superfusion bath while recording axon conduction. 1
Initial studies in rat optic nerve showed that in the absence of
oxygen the CAP area, an index of axon conduction, falls to

CAP area
zero within a few minutes (Stys et al. 1992), but in the absence 0.5 1 2 3
of glucose, with oxygen present, the CAP area took more than
30 minutes before it began to fail (Ransom and Fern 1997).
These results suggested that the tissue contained an endoge- 0
nous energy reserve that could support axon conduction in 0 60 120 180 240
the absence of exogenously supplied glucose. Upregulation or Latency (mins)
downregulation of glycogen content was achieved by incubat-
ing the nerves at various concentrations of glucose, or in the Figure 36.4 Glycogen Prolongs Latency to CAP Failure During Glucose
Withdrawal. A. Latency to stimulus-evoked CAP failure in MON dur-
presence of noradrenaline, indicative of glycogen as a labile ing aglycemia is linearly related to the glycogen content of the tissue at
energy reserve (Wender et al. 2000). Because glycogen is onset of aglycemia according to the relationship Latency = 3.87*
stored in astrocytes in the adult, a transportable conduit must [Glycogen] – 8.32. B. Latency to CAP failure of the A peak (3) and C
be present for axons to benefit from astrocyte glycogen. The peak (1) of the sciatic nerve during aglycemia showing the A peak failure
ability of blockers of monocarboxylate transporters (MCTs) is significantly delayed relative to C peak failure, an effect that is lost
when glycogen metabolism is inhibited by DAB (2). (A) Data adapted
to impede the ability of glycogen to support axon conduction from Brown et al. 2003.
is strong evidence that the transportable conduit was lactate
(Wender et al. 2000). Subsequent rodent optic nerve studies
were carried out on the mouse optic nerve for compatibility glucose decrease (although this does suggest that glycogen
with future transgenic models, and for the decreased diffusion might be targeted as a clinically relevant therapeutic target
distance (Baltan Tekkök et al. 2003) in studies of this sort in to stave off the injury that results from iatrogenic systemic
which energy substrates and drugs are applied exogenously in insulin-induced hypoglycemia) (Suh et al. 2007). We sought
the superfusate. to determine if glycogen played a role in more physiological
Complementary data from the mouse optic nerve (MON) function in the form of high frequency firing, which is an
reinforced the capacity of rodent optic nerve to conduct experimental correlate for increased tissue energy demand
action potentials in the absence of glucose (Brown et al. resulting from increased frequency of axon action poten-
2003), although only for about 20 minutes compared to 30 tials. High frequency stimulus (up to 100 Hz) was imposed
minutes in the rat. Biochemical assays of glycogen carried out on the MONs under euglycemic conditions and the result-
in parallel with electrophysiological recordings showed that ing CAPs recorded. Parallel experiments measuring glycogen
the onset of aglycemia coincided with a decrease in glycogen content at the end of the stimulus period showed that there
content, and once glycogen levels reached their nadir CAP was a significant decrease in glycogen during the period of
conduction failed, establishing a direct relationship between stimulus (Brown et al. 2003). This indicated that glycogen was
glycogen and axon conduction (Brown et al. 2003). Elevating metabolized, even though the tissue was perfused with 10 mM
glycogen content before aglycemia prolonged latency to CAP glucose aCSF, implying that euglycemic concentrations of glu-
failure; conversely, decreasing glycogen content before agly- cose are insufficient to support function, and glycogen must
cemia accelerated CAP failure (Brown et al. 2003). Indeed, be metabolized to provide a supplemental energy substrate for
there was a linear relationship between glycogen content at axon conduction to be fully maintained (Brown et al. 2003).
the onset of aglycemia and the latency to CAP failure, such However, an alternate explanation is that the glucose supplied
that accurate predictions about latency to CAP failure could to the nerve partly supports axon conduction and partly sup-
be made based on the glycogen content of the tissue (Brown ports glycogen synthesis. During periods of increased tissue
et al. 2003) (Fig. 36.4). energy demand, the glucose is preferentially used to support
Given the rarity of spontaneous hypoglycemia, and the axon conduction and thus less or none is used to support gly-
role of liver glycogen in maintaining euglycemia during sys- cogen synthesis. However, the experiment lasted 4 minutes, a
temic glucose decrease, it seems unlikely that the primary time period considered too rapid for the effect to result from
role for glycogen is supporting function during periods of reduction in glycogen turnover.

G LYC O G E N A N D E N E R GY M ETA B O L I S M • 463


The ability of glycogen to support function implies a unstimulated baseline conditions. These results imply that lac-
metabolic pathway from astrocytes to axons that is capable of tate is constantly produced in the tissue and exported to the
metabolizing glycogen to lactate in the astrocyte, transporting extracellular fluid. Given the rate of our perfusion system and
the lactate out of the astrocyte, followed by the axon having the superfusion of the tissue with aCSF, this lactate is washed
the ability to take up the lactate and metabolize it oxidatively. out of the tissue and into the bath. These data certainly suggest
The enzyme lactate dehydrogenase, which interconverts lac- that under resting conditions an excess of lactate is produced
tate and pyruvate, comprises several isoforms. The isoform relative to the energy requirements of the tissue, but more
that preferentially converts lactate to pyruvate (LDH1) is importantly suggests that lactate is present in the extracel-
found in neural elements, whereas the isoform that converts lular fluid in sufficiently large concentrations to be used as a
pyruvate to lactate (LDH5) is found in astrocytes, consistent source of energy during periods of need. The cellular source
with lactate production in astrocytes and metabolism in axons (s) of lactate has not been identified, but astrocyte metabo-
(Bittar et al. 1996). For lactate transport to occur between the lism is almost certain to contribute. Whether glycogen
two cell types, the appropriate transporter proteins must also metabolism contributes to this lactate signal was investigated
be present. Lactate is cotransported with H+ via monocarbox- by adding 1 mM 1,4-dideoxy-1,4-imino-D-arabinitol (DAB),
ylate transporters (MCTs). There are four isoforms of MCTs a compound that inhibits the action of glycogen phosphory-
that can transport lactate in mammals (Halestrap 2012), but lase (Walls et al. 2008). The lactate signal fell by 30% in the
the isoform that exports lactate (MCT1) is found on astro- presence of DAB and was reversible on DAB washout, dem-
cytes, and the isoform that imports lactate (MCT2) is located onstrating that under baseline resting conditions glycogenoly-
on neural elements (Halestrap and Price 1999; Koehler-Stec sis contributes to the lactate production of the tissue, implying
et al. 1998; Pierre et al. 2000). Immunohistochemical studies that glycogen is constantly being metabolized, and thus syn-
in MON confirm the appropriate cellular location for MCT thesized, indicating that glycogen turnover is very active in
subtypes (Baltan Tekkök et al. 2005). Experiments with pH- this tissue. This flies in the face of conventional attitudes
sensitive microelectrodes showed that the onset of aglycemia that glycogen is a minor energy reserve that is only called on
resulted in alkalization of the extracellular fluid consistent in times of glucose depletion or periods of increased energy
with decreased lactate transport out of astrocytes into the demand (Fig. 36.5) (Wender et al, 2000).
ECF in the face of decreased glucose availability, succeeded
by a second stage in which extracellular pH increased to bath
4.2 G R AY M AT T E R : G LYC O G E N S U P P O RT S
pH of 7.45, indicative of a total loss of lactate efflux (Brown
SY NA P T I C F U N C T I O N I N AG LYC E M I A
et al. 2001). Recent experiments with lactate biosensors have
illuminated these data. Recording lactate in the vicinity of the Pioneering studies seeking the location of glycogen noted a
nerve registers a lactate concentration of about 400 μM under high expression in synaptic regions (Koizumi 1974; Koizumi

Blood Astrocyte Neural element

cap
astrocyte glycogen
end foot
pyruvate
e
tat

glucose glucose ATP


lac

endfeet
E CO2 & H2O
n
d glucose
o
t
h
e
l
i
u
m

Figure 36.5 Model of Metabolic Substrate Relationship Between Astrocytes and Neural Elements. Astrocytic endfeet almost completely ensheathe
capillary walls. Glucose present in the blood can be transported into the extracellular fluid for uptake into neural elements, or alternatively, may be
taken up into astrocytes, where it can be converted to glycogen. Glycogen metabolism yields lactate, which is transported into the extracellular fluid
for uptake and metabolism by neural elements. Under baseline conditions in which glucose is available in excess of demand, lactate accumulates in the
extracellular fluid because not all is taken up by axons. In the absence of glucose, glycogen is metabolized to lactate, which is shuttled out of astrocytes
and taken up by neural elements as their sole metabolic substrate. Because lactate would not accumulate in the extracellular fluid under these
conditions, its concentration is low.

464 • FUNCTIONS OF NEUROGLIAL CELLS


and Shiraishi 1970a,b; Phelps 1972). Given the high meta- 4.4 G LYC O G E N A N D L E A R N I N G : G LYC O G E N
bolic cost of synaptic activity (Attwell and Laughlin 2001), S U P P O RTS M E M O RY AC Q U I S IT I O N
such a location seems logical. Indeed, a central role for glyco- I N T H E A D U LT RO D E N T
gen is likely given the recent proposal of the tripartite synapse
Recently Suzuki et al. (2011) have used the model described
(Araque et al. 1999; Perea et al. 2009) in which the astro-
in the preceding section as a template to examine the role of
cytes play a key role as sensors for neurotransmitter release.
glycogen in memory formation in the rodent. Using an in vivo
In hippocampal slice preparations, in which stimulus-evoked
rat preparation in which field potentials were recorded from
CAPs recorded from the CA1 region are monitors of
the CA1 region of the hippocampus, along with a dialysis
synaptic efficacy, removal of glucose from perfusing aCSF
probe for introduction of DAB or lactate, they investigated
leads to a delayed fall in the response. Depleting glycogen
the effect of DAB on the ability of rats to retain memory in
before aglycemia leads to an accelerated fall in the excitatory
the form of aversion therapy, which serves as a proxy for mem-
postsynaptic potential (EPSP) (Izumi et al. 1997), whereas
ory. Rats were housed in a white room with a door leading
preincubation in 30 mM glucose, a maneuver that leads to
to a dark room. When the rats entered the dark room they
increased glycogen content, resulted in prolonged latency to
received an electric shock. Rats were then re-housed in the
EPSP failure (Cater et al. 2001). These results strongly sug-
white room and the duration before they re-entered the dark
gested that hippocampal glycogen supports synaptic activity
room was taken as an index of memory, the longer they took
during aglycemia.
to enter the room, the better the memory was consolidated.
The study showed that DAB was only effective in abolishing
4.3 G LYC O G E N A N D L E A R N I N G : memory when administered 15 minutes before the training
G LYC O G E N S U P P O RT S L E A R N I N G D U R I N G period. DAB injection after the training had no effect on the
D EV E L O PM E N T I N C H I C K memory. After DAB injection the memory was absent for up
to 7 days after the training period, showing that DAB had effi-
Studies carried out on memory acquisition and consolidation ciently prevented memories being formed. Injecting lactate in
in 1-day-old chicks demonstrates the importance of glycogen the region around the CA1 in the presence of DAB restored
in this higher brain function. DAB was injected either focally the ability to lay down memories, convincingly showing
into the brain or systemically to inhibit glycogenolysis. A train- that glycogen-derived lactate is the metabolic conduit that sup-
ing paradigm using avoidance techniques were used as a proxy ports laying down of memory (Suzuki et al. 2011). Interestingly,
for memory. In the chick there are several stages in the laying the study found that glucose was not as effective as lactate in
down of memory. Phase A lasts for 10 to 25 minutes, and phase laying down memories. This appears initially puzzling, but
B lasts from 30 to 55 minutes after the training event. DAB if the memory mechanism requires an immediate energy
was found to affect memory when applied 5 minutes before supply, then perhaps glycogen is more suited to deliver this
training, with testing of memory at 120 minutes after train- than an increase in glucose delivery mediated by increase
ing. The effects of DAB were dependent on when DAB was blood flow, and only lactate rather than glucose can act as the
applied. The greatest effects were seen 5 minutes before train- preferred energy substrate. These results support a physiologi-
ing or between 25 and 40 minutes after training, implying that cal role for glycogen predicted by previous experiments in
glycogen is involved in the memory processing at these two white matter (Brown et al. 2003), namely, that it is an energy
distinct time periods (Gibbs et al. 2006). Additional experi- buffer to provide energy substrate during periods of increased
ments determined that injection of DAB 5 minutes before tissue energy demand that cannot be met by ambient glu-
training caused deficits in memory when tested 5 to 10 min- cose. These experiments predict that glycogen is a vital energy
utes after training and for 40 minutes onward after training. source in the brain, supporting a critical aspect of higher brain
Similarly, DAB injected 25 minutes after training with mem- function.
ory tested at 35 to 60 minutes after training, showed deficits
in memory. However, memory could be restored if glutamine,
or acetate and aspartate, were injected immediately after 5 G LYC O G E N I N T H E P E R I P H E R A L
training in DAB-treated chicks. These experiments showed N E RVO U S SYS T E M : G LYC O G E N I S
for the first time that glycogen is actively involved in higher L O C A L I Z E D TO S C H WA N N C E L L S
brain function in vivo (Gibbs et al. 2006), albeit in chick. The A N D S E L E C T I VE LY S U P P O RT S A
experiments suggested that glycogen is important in memory FIBER CONDUCTION
consolidation at several stages of the process. As glycogen is
metabolized to pyruvate, which can then be converted to glu- The previous data have described glia in the CNS as possessing
tamate via TCA cycling, supplemental glutamine, presumably glycogen and supplying glycogen derived substrate to supply
a precursor to glutamate, circumvents the inhibitory effects neural elements, via astrocyte release of lactate into the extra-
of DAB. Fueling the astrocyte TCA cycle with acetate and cellular fluid. Immunohistochemical studies in the periph-
aspartate also restored memory, probably because aspartate eral nervous system (PNS) suggested that this might not be
compensates for pyruvate carboxylation and acetate compen- the case in peripheral nerve (Pfeiffer-Guglielmi et al. 2007).
sates for pyruvate dehydrogenase to supply glutamate (Gibbs In sciatic nerve, antibodies to glycogen phosphorylase revealed
et al. 2006). its presence exclusively in axons with none apparent in the

G LYC O G E N A N D E N E R GY M ETA B O L I S M • 465


Schwann cells (Pfeiffer-Guglielmi et al. 2007). This was a tan- axons. Small myelinated axons have a higher energy demand
talizing result because it suggested that if glycogen was pres- per unit volume than do large axons (Nicholls et al. 2001).
ent in peripheral nerves it was located in the axons rather than Thus it could well be that the axons do not benefit from what-
the glial cells, in this case the Schwann cell, and its function ever glycogen is available. We carried out experiments in which
must be different from that in the CNS. The authors under- we upregulated glycogen by 2 hours before incubation in 30
took a series of experiments to determine the cellular location mM glucose and found that the latency to C fiber CAP failure
of glycogen in the sciatic nerve, a peripheral nerve. Previously was delayed during aglycemia relative to control, results that
there had been no reports of glycogen in peripheral nerve, appear to highlight that C axons are not incapable of benefit-
only the appearance of glycogen granules as a result of pathol- ing from glycogen-derived substrate, but that under physio-
ogy (Kalichman et al. 1998; Katsuragi et al. 1988; Powell et al. logical conditions, insufficient substrate is available. Given the
1985). Our initial studies using biochemical assay detected parallels between the optic and sciatic nerve, we determined
glycogen in the sciatic nerve at concentrations about double whether the transportable conduit was likely to be lactate. We
that of optic nerve; thus, our next objective was to locate showed that lactate can support both the A and C fibers in
the cellular domain of the glycogen. Immunohistochemical the absence of glucose, and additionally can rescue A and C
studies identified glycogen phosphorylase in both axons and fibers during aglycemia; thus, lactate is capable of supporting
Schwann cells. It is important to note at this point that the axon conduction, and is most likely the conduit. Experiments
presence of glycogen phosphorylase does not necessarily sig- with lactate-sensitive biosensors showed that although the
nify the presence of glycogen. We used electron microscopy to tissue does release lactate, the concentration recorded was
localize glycogen predominantly in the Schwann cells myeli- much lower than that recorded from the optic nerve (120 vs
nating large A fibers, but found no evidence of glycogen in the 400 μM). The reasons for this are unclear, but may be related
Schwann cells bundling unmyelinated C fibers. We also found to the decreased metabolic demand of peripheral versus cen-
no evidence of glycogen in axons. As such these data par- tral tissue. As was the case in optic nerve, DAB reversibly
tially refute the initial data from the PNS (Pfeiffer-Guglielmi decreased lactate concentration by about 25%, indicating that
et al. 2007) and demonstrate selective glial expression of gly- glycogen-derived lactate is constantly produced by the tissue,
cogen. Experiments were then carried out to identify a role(s) and this lactate is surplus to the demands of the tissue under
for glycogen. resting conditions. Thus, it appears peripheral and central
We investigated the ability of glycogen to support the sci- nerve share a similar debt to glycogen in providing lactate to
atic nerve action potential conduction. The stimulus-evoked the extracellular fluid that can be taken up and metabolized by
CAP in sciatic nerve is comprised of two separate elements: axons as required.
a fast unipolar peak that occurs within a millisecond of the
stimulus artifact and is contributed to by large myelinated
axons (A peak), followed by a smaller bipolar peak 5 to 15 ms
6 G LYC O G E N A N D D I S E A S E
later that is caused by small unmyelinated C axons (C peak).
On exposure to aglycemia the A peak was sustained for 2.5
6.1 D I A B ET E S
hours, whereas the C peak lasted only 0.5 hour. Measurement
of glycogen during aglycemia showed that it falls and reaches For glycogen to play a role in diabetes in humans, the presence
its nadir temporally correlated with the A peak failure, con- of glycogen in the human must be established. This has been
sistent with glycogen metabolism during aglycemia, but once done using nuclear magnetic resonance (NMR) spectroscopy
the glycogen is exhausted the A peak fails. The C peak did not (Oz et al. 2003, 2007) and biochemical assay of resected human
appear correlated with glycogen content. These data suggest brain tissue (Dalsgaard et al. 2006). The effects of type-2 diabe-
that glycogen does not afford the C axons any particular pro- tes (T2D) were studied in Zucker obese rat (ZOb) and Zucker
tection during aglycemia, consistent with the apparent lack of Diabetic fatty rat (ZDF) models of T2D, and the general trend
glycogen in Schwann cells surrounding C fibers. To test this was that in models of T2D model, glycogen levels were lower
further we bathed the sciatic nerve in DAB before exposure to than in the cerebellum and hippocampus of control mice.
aglycemia. Under these conditions the A peak fell at the same However, this was not found in the cortex (Sickmann et al.
time as the C peak, increasing the evidence that glycogen sup- 2010). The results were confused by the differences in brain
ports the A axons, but not the C axons, and in the absence of glycogen levels among the control animals, namely, that in the
glycogen-derived substrate there is no difference between the control rats, Sprague-Dawley, Zucker lean (ZL), and Zucker
A and C peaks. Diabetic lean (ZDL), there was considerable variation in gly-
These data differ from the CNS optic nerve, in which glyco- cogen levels among the cortex, cerebellum, and hippocampus.
gen appears to support all axons equitably. The obvious reason The Zucker lean (ZL) is the genetic control for the ZOb, and
for this difference is in the morphology of the tissue, with clear had a higher glycogen content in cortex; however, in the other
differences between the large A fibers, which are myelinated two brain regions the glycogen was lower in ZOb than in ZL.
by single Schwann cells, and the C fibers, which are bundled This could be caused by a variety of factors. In these animal
with several others such that multiple C axons are myelinated models of diabetes there is insensitivity to insulin accompa-
by a single Schwann cell. Thus, Schwann cells myelinating C nied by ambient hyperglycemia. It is known that CNS glyco-
axons that lack glycogen are not protected by glycogen. An gen is sensitive to ambient glucose (Brown et al. 2003), and
additional consideration is the energy requirements of the also to insulin (Banks et al. 1997), but the balance between

466 • FUNCTIONS OF NEUROGLIAL CELLS


the two is unknown under control or pathological conditions. of hereditary glycogen storage disease associated with deficient
The situation is different from the periphery in which the expression of glucose-6-phosphatase (type I), D(1→4) glucosi-
insulin-sensitive Glut-4 transporter is the major route of cel- dase (type II), glycogen debranching enzyme (type 3), glycogen
lular glucose entry (Voet and Voet 2011). In the CNS the main branching enzyme (type 4), glycogen phosphorylase (types V
routes of cellular glucose access are via the insulin-insensitive and VI), phosphofructokinase (type VII), phosphorylase kinase
Glut-1 or Glut-3 receptors (Vannucci et al. 1997); thus, insu- (types VIII and IX) and glycogen synthase (type 0) (Voet and
lin does not play a key role in glucose uptake in CNS tissue, Voet 2011). All of these diseases result in an overabundance of
as it does in the periphery. It is interesting to note that in the glycogen in liver and/or muscle, except type 0, which results in
cortex the glycogen levels are higher than control, as would be decreased glycogen content, suggesting that the presence of gly-
expected if ambient glucose was the sole factor in determining cogen is a prerequisite for life. The effects of glycogen storage
glycogen content. That glycogen levels were lower in the cere- diseases on brain glycogen have been little studied, and there
bellum and hippocampus in T2D suggests that diabetes may are only a few scattered accounts in the literature, reporting
affect brain regions selectively. A possible explanation is the pathology associated with the disease, rather than any puta-
selective expression of the insulin-sensitive Glut-4 transporter tive effect of the disease on brain glycogen content (Garty
predominantly in neurons in the olfactory bulb, hippocam- et al. 1992; Lamperti et al. 2009; Martin et al. 1973; Melis et al.
pus, and cerebellum (El Messari et al. 1998, 2002; Vannucci 2004; Middleditch et al. 1998; Weghuber et al. 2007).
et al. 1998); however, these issues require further study.
The first measurement of glycogen in the brains of patients
with T1D has recently been published (Oz et al. 2011). When 7 S U M M A RY A N D P E R S P E C T I VE S
compared to control subjects, newly synthesized glycogen
was greater in control subjects than T1D patients, results that In conclusion, interest in glycogen has recently been reawak-
could be explained as higher glycogen content or increased ened after a period when interest in its physiological impor-
glycogen turnover. Subsequent metabolic modeling of the tance was neglected. Recent advances have seen in vivo
data, when taking into account prevailing glucose and insu- measurements of glycogen in human brain (Oz et al. 2003,
lin levels, indicated that patients with T1D did indeed have 2007), demonstration of the ability of glycogen to support
lower glycogen levels. The issue is complex, however, because neural function in the absence of glucose in both gray (Cater
if glucose uptake is both glucose and insulin dependent, then et al. 2001; Izumi et al. 1997) and white matter (Wender et al.
the contribution of each may lead to unpredictable values, 2000), and its ability to provide supplemental energy sub-
depending on prevailing insulin and glucose levels. In addi- strate during increased tissue energy demand in white matter
tion, Glut-1 glucose transporters in the blood brain barrier are (Brown et al. 2003). Glycogen has also been shown to support
upregulated relative to control in T1D patients, which would higher brain function under in vivo conditions (Suzuki et al.
augment glucose influx into the brain. The method of mea- 2011), and recent reports suggest diabetes is associated with
suring glycogen could also lead to problems of interpretation brain glycogen control dysfunction (Oz et al. 2011; Sickmann
(Oz et al. 2003), because the method requires long periods of et al. 2010). Thus, these are exciting times for brain glycogen
infusion of 13C glucose; however, the results for control sub- research, in which a multitude of experimental approaches in
jects were consistent with biochemical assay of glycogen from both animal models and humans are gradually elucidating key
biopsied human brain tissue (Dalsgaard et al. 2006). A second roles in fundamental brain function.
aim of this study was to determine if glycogen played a role in
hypoglycemia unawareness. This is a condition in which expo-
sure to hypoglycemia results in a decrease in the autonomic REFERENCES
warning signals of impending hypoglycemia (Frier and Fisher Andriezen WL. 1893. On a system of fibre-like cells surrounding the
2007). The argument for glycogen involvement proposes that blood vessels of the brain of man and mammals, and its physiological
post-hypoglycemia levels of glycogen super-compensate to significance. Int Monatsschr Anat Physiol 10:532–540.
higher levels than pre-hypoglycemia (Oz et al. 2009). It has Araque A, Parpura V, Sanzgiri RP, Haydon PG. 1999. Tripartite synapses:
thus been hypothesized that this increased glycogen, and the glia, the unacknowledged partner. Trends Neurosci 22(5):208–215.
Attwell D, Laughlin SB. 2001. An energy budget for signaling in the grey
release of glycogen-derived substrates in the brain leads to the matter of the brain. J Cereb Blood Flow Metab 21(10):1133–1145.
brain being fooled into thinking there is sufficient energy sub- Baltan Tekkök S, Brown AM, Ransom BR. 2003. Axon function per-
strate in the face of hypoglycemia. However, the time course of sists during anoxia in mammalian white matter. J Cereb Blood Flow
the glycogen concentration changes did not correlate with the Metab 23:1340–1348.
time course of the faulty signaling, which argues against post Baltan Tekkök S, Brown AM, Westenbroek RE, Pellerin L, Ransom BR.
2005. Transfer of glycogen-derived lactate from astrocytes to axons
hypoglycemia super-compensation playing a role in hypogly- via specific monocarboxylate transporters supports mouse optic
cemia unawareness. nerve activity. J Neurosci Res 81:644–652.
Banks WA, Jaspan JB, Kastin AJ. 1997. Selective, physiological transport
of insulin across the blood-brain barrier: novel demonstration by
6.2 G LYC O G E N S TO R AG E D I S E A S E S species-specific radioimmunoassays. Peptides 18(8):1257–1262.
Berg JM, Tymoczko JL, Stryer L. 2006. Biochemistry. New York:
Glycogen storage diseases are a family of conditions resulting Freeman.
from deficient expression of a variety of enzymes involved in Bittar PG, Charnay Y, Pellerin L, Bouras C, Magistretti PJ. 1996.
glycogen metabolism and synthesis. There are at least 10 types Selective distribution of lactate dehydrogenase isoenzymes in

G LYC O G E N A N D E N E R GY M ETA B O L I S M • 467


neurons and astrocytes of human brain. J Cereb Blood Flow Metab Koizumi J. 1974. Glycogen in the central nervous system. Prog Histochem
16(6):1079–1089. Cytochem 6(4):1–37.
Bloom W, Fawcett DW. 1968. A textbook of histology. Philadelphia: Koizumi J, Shiraishi H. 1970a. Glycogen accumulation in dendrites of
Saunders. the rabbit pallidum following trifluoperazine administration. Exp
Brown AM, Baltan Tekkök S, Ransom BR. 2003. Glycogen regulation and Brain Res 11(4):387–391.
functional role in mouse white matter. J Physiol 549(2):501–512. Koizumi J, Shiraishi H. 1970b. Ultrastructural appearance of glycogen
Brown AM, Wender R, Ransom BR. 2001. Ionic mechanisms of aglyce- in the hypothalamus of the rabbit following chlorpromazine admin-
mic axon injury in mammalian central white matter. J Cereb Blood istration. Exp Brain Res 10(3):276–282.
Flow Metab 21:385–395. Lamperti C, Salani S, Lucchiari S, Bordoni A, Ripolone M, Fagiolari
Cataldo AM, Broadwell RD. 1986. Cytochemical identification of cere- G, et al. 2009. Neuropathological study of skeletal muscle, heart,
bral glycogen and glucose-6-phosphatase activity under normal and liver, and brain in a neonatal form of glycogen storage disease
experimental conditions. I. Neurons and glia. J Electron Microsp type IV associated with a new mutation in GBE1 gene. J Inherit
Techn 3:413–437. Metab Dis. http://dx.doi.org/10.1007/s10545-009-1134–8.
Cater HL, Benham CD, Sundstrom LE. 2001. Neuroprotective role of Magistretti PJ. 1988. Regulation of glycogenolysis by neurotransmitters
monocarboxylate transport during glucose deprivation in slice cul- in the central nervous system. Diabetes Metab 14(3):237–246.
tures of rat hippocampus. J Physiol 531(Pt 2):459–466. Magistretti PJ, Hof PR, Martin JL. 1986. Adenosine stimulates glycog-
Champe PC, Harvey RA. 2008. Biochemistry. Baltimore: Lippincott enolysis in mouse cerebral cortex: a possible coupling mechanism
Williams & Wilkins. between neuronal activity and energy metabolism. J Neurosci Res
Dalsgaard MK, Madsen FF, Secher NH, Laursen H, Quistorff B. 2006. 6(9):2558–2562.
High glycogen levels in the hippocampus of patients with epilepsy. Martin JJ, de Barsy T, van Hoof F, Palladini G. 1973. Pompe’s disease: an
J Cereb Blood Flow Metab 27:1–5. inborn lysosomal disorder with storage of glycogen. A study of brain
Dienel GA. 2009. Energy metabolism in the brain. In: Byrne JH, Roberts and striated muscle. Acta Neuropathol 23(3):229–244.
JL (eds.), From molecules to networks: an introduction to cellular and McKenna MC, Gruetter R, Sonnewald U, Waagepetersen HS,
molecular neuroscience. New York: Academic Press, pp. 49–110. Schousboe A. 2006. Energy metabolism of the brain. In: Siegel GJ,
Dringen R, Gebhardt R, Hamprecht B. 1993. Glycogen in astrocytes: Albers RW, Brady ST, Price DL (eds.), Basic neurochemistry. San
possible function as lactate supply for neighboring cells. Brain Res Diego: Academic Press, pp. 531–558.
623(2):208–214. Melis D, Parenti G, Della Casa R, Sibilio M, Romano A, Di Salle F,
El Messari S, Ait-Ikhlef A, Ambroise DH, Penicaud L, Arluison M. 2002. et al. 2004. Brain damage in glycogen storage disease type I. J Pediatr
Expression of insulin-responsive glucose transporter GLUT4 mRNA 144(5):637–642.
in the rat brain and spinal cord: an in situ hybridization study. J Chem Middleditch C, Clottes E, Burchell A. 1998. A different isoform of
Neuroanat 24(4):225–242. the transport protein mutated in the glycogen storage disease 1b is
El Messari S, Leloup C, Quignon M, Brisorgueil MJ, Penicaud L, Arluison expressed in brain. FEBS Lett 433(1–2):33–36.
M. 1998. Immunocytochemical localization of the insulin-responsive Nelson SR, Schulz DW, Passonneau JV, Lowry OH. 1968. Control of
glucose transporter 4 (Glut-4) in the rat central nervous system. glycogen levels in brain. J Neurochem 15(11):1271–1279.
J Comp Neurol 399(4):492–512. Nicholls JG, Martin AR, Wallace BG, Fuchs PA. 2001. From neuron to
Fox PT, Raichle ME. 1986. Focal physiological uncoupling of cerebral brain. Sunderland, MA: Sinauer Associates.
blood flow and oxidative metabolism during somatosensory stimula- Nilaweera K, Herwig A, Bolborea M, Campbell G, Mayer CD, Morgan
tion in human subjects. Proc Natl Acad Sci U S A 83(4):1140–1144. PJ, et al. 2011. Photoperiodic regulation of glycogen metabolism,
Frier BM, Fisher BM. 2007. Hypoglycaemia in clinical diabetes. New glycolysis, and glutamine synthesis in tanycytes of the Siberian ham-
York: Wiley. ster suggests novel roles of tanycytes in hypothalamic function. Glia
Garrett RH, Grisham CM. 1999. Biochemistry. Fort Worth, TX: 59(11):1695–1705.
Saunders College Publishing. Oz G, Henry PG, Seaquist ER, Gruetter R. 2003. Direct, noninvasive
Garty BZ, Rachmel A, Mandel H, Nitzan M. 1992. Brain abscesses in measurement of brain glycogen metabolism in humans. Neurochem
glycogen-storage disease-type 1b. Acta Paediatr 81(11):948. Int 43(4–5):323–329.
Gibbs ME, Anderson DG, Hertz L. 2006. Inhibition of glycogenolysis in Oz G, Kumar A, Rao JP, Kodl CT, Chow L, Eberly LE, et al. 2009.
astrocytes interrupts memory consolidation in young chickens. Glia Human brain glycogen metabolism during and after hypoglycemia.
54(3):214–222. Diabetes 58(9):1978–1985.
Halestrap AP. 2012. The monocarboxylate transporter family: structure Oz G, Seaquist ER, Kumar A, Criego AB, Benedict LE, Rao JP, et al.
and functional characterization. IUBMB Life 64(1):1–9. 2007. Human brain glycogen content and metabolism: implications
Halestrap AP, Price NT. 1999. The proton-linked monocarboxylate on its role in brain energy metabolism. Am J Physiol Endocrinol
transporter (MCT) family: structure, function and regulation. Metab 292(3):E946–951.
Biochem J 343:281–299. Oz G, Tesfaye N, Kumar A, Deelchand DK, Eberly LE, Seaquist ER.
Hof PR, Pascale E, Magistretti PJ. 1988. K+ at concentrations reached 2011. Brain glycogen content and metabolism in subjects with type 1
in the extracellular space during neuronal activity promotes a diabetes and hypoglycemia unawareness. J Cereb Blood Flow Metab
Ca 2+-dependent glycogen hydrolysis in mouse cerebral cortex. 32(2):256–263.
J Neurosci 8(6):1922–1928. Pellegri G, Rossier C, Magistretti PJ, Martin JL. 1996. Cloning, localiza-
Izumi Y, Benz AM, Katsuki H, Zorumski CF. 1997. Endogenous tion and induction of mouse brain glycogen synthase. Brain Res Mol
monocarboxylates sustain hippocampal synaptic function and Brain Res 38(2):191–199.
morphological integrity during energy deprivation. J Neurosci Pellerin L, Magistretti PJ. 1994. Glutamate uptake into astrocytes
17(24):9448–9457. stimulates aerobic glycolysis: a mechanism coupling neuronal
Kalichman MW, Powell HC, Mizisin AP. 1998. Reactive, degenerative, activity to glucose utilization. Proc Natl Acad Sci U S A 91(22):
and proliferative Schwann cell responses in experimental galactose 10625–10629.
and human diabetic neuropathy. Acta Neuropathol 95(1):47–56. Perea G, Navarrete M, Araque A. 2009. Tripartite synapses: astro-
Katsuragi S, Shimoji A, Watanabe K, Miyakawa T. 1988. Pathological cytes process and control synaptic information. Trends Neurosci
findings of the sural nerve in mitochondrial encephalomyopathy. 32(8):421–431.
Jpn J Psychiatry Neurol 42(2):307–313. Pfeiffer-Guglielmi B, Fleckenstein B, Jung G, Hamprecht B. 2003.
Koehler-Stec EM, Simpson IA, Vannucci SJ, Landschulz KT, Landschulz Immunocytochemical localization of glycogen phosphorylase
WH. 1998. Monocarboxylate transporter expression in mouse brain. isozymes in rat nervous tissues by using isozyme-specific antibodies.
Am J Physiol 275(3 Pt 1):E516–E524. J Neurochem 85(1):73–81.

468 • FUNCTIONS OF NEUROGLIAL CELLS


Pfeiffer-Guglielmi B, Francke M, Reichenbach A, Hamprecht B. 2007. Suh SW, Bergher JP, Anderson CM, Treadway JL, Fosgerau K, Swanson
Glycogen phosphorylase isozymes and energy metabolism in the rat RA. 2007. Astrocyte glycogen sustains neuronal activity during hypogly-
peripheral nervous system—an immunocytochemical study. Brain Res cemia: studies with the glycogen phosphorylase inhibitor CP-316,819
1136(1):20–27. ([R-R*,S*]-5-chloro-N-[2-hydroxy-3-(methoxymethylamino)-3-oxo-1-
Phelps CH. 1972. Barbiturate-induced glycogen accumulation in brain. (phenylmet hyl)propyl]-1H-indole-2-carboxamide). J Pharmacol Exp
An electron microscopic study. Brain Res 39(1):225–234. Ther 321(1):45–50.
Pierre K, Pellerin L, Debernardi R, Riederer BM, Magistretti PJ. 2000. Suzuki A, Stern SA, Bozdagi O, Huntley GW, Walker RH, Magistretti
Cell-specific localization of monocarboxylate transporters, MCT1 PJ, et al. 2011. Astrocyte-neuron lactate transport is required for
and MCT2, in the adult mouse brain revealed by double immu- long-term memory formation. Cell 144(5):810–823.
nohistochemical labeling and confocal microscopy. Neuroscience Swanson RA, Choi DW. 1993. Glial glycogen stores affect neuronal sur-
100(3):617–627. vival during glucose deprivation in vitro. J Cereb Blood Flow Metab
Powell HC, Haas R, Hall CL, Wolff JA, Nyhan W, Brown BI. 1985. 13(1):162–169.
Peripheral nerve in type III glycogenosis: selective involvement of Swanson RA, Morton MM, Sagar SM, Sharp FR. 1992. Sensory stimula-
unmyelinated fiber Schwann cells. Muscle Nerve 8(8):667–671. tion induces local cerebral glycogenolysis: demonstration by autora-
Prichard J, Rothman D, Novotny E, Petroff O, Kuwabara T, Avison diography. Neuroscience 51(2):451–461.
M, et al. 1991. Lactate rise detected by 1H NMR in human visual Vannucci SJ, Koehler-Stec EM, Li K, Reynolds TH, Clark R, Simpson IA.
cortex during physiologic stimulation. Proc Natl Acad Sci U S A 1998. GLUT4 glucose transporter expression in rodent brain: effect of
88(13):5829–5831. diabetes. Brain Res 797(1):1–11.
Ransom BR, Fern R. 1997. Does astrocytic glycogen benefit axon func- Vannucci SJ, Maher F, Simpson IA. 1997. Glucose transporter proteins in
tion and survival in CNS white matter during glucose deprivation? brain: delivery of glucose to neurons and glia. Glia 21(1):2–21.
Glia 21(1):134–141. Vilchez D, Ros S, Cifuentes D, Pujadas L, Valles J, Garcia-Fojeda B, et al. 2007.
Ransom BR, Waxman SG, Stys PK. 1994. Anoxic injury of central myeli- Mechanism suppressing glycogen synthesis in neurons and its demise in
nated axons: nonsynaptic ionic mechanisms. In: Hartman A, Yatsu progressive myoclonus epilepsy. Nat Neurosci 10(11):1407–1413.
F, Kuschinsky W (eds.), Cerebral ischemia and basic mechanisms. Voet D, Voet JG. 2011. Biochemistry. Hoboken, NJ: Wiley.
Heidelberg: Springer-Verlag, pp. 77–90. Walls AB, Sickmann HM, Brown A, Bouman SD, Ransom B, Schousboe
Rossen R, Kabat H, Anderson JP. 1943. Acute arrest of cerebral circula- A, et al. 2008. Characterization of 1,4-dideoxy-1,4-imino-d-arabinitol
tion in man. Arch Neurol Psychiatry 50:510–528. (DAB) as an inhibitor of brain glycogen shunt activity. J Neurochem
Shulman RG, Hyder F, Rothman DL. 2001. Cerebral energetics and the 105(4):1462–1470.
glycogen shunt: neurochemical basis of functional imaging. Proc Natl Wang SS, Shultz JR, Burish MJ, Harrison KH, Hof PR, Towns LC,
Acad Sci U S A 98(11):6417–6422. et al. 2008. Functional trade-offs in white matter axonal scaling.
Shulman RG, Rothman DL. 2001. The “glycogen shunt” in exercising J Neurosci 28(15):4047–4056.
muscle: a role for glycogen in muscle energetics and fatigue. Proc Natl Weghuber D, Mandl M, Krssak M, Roden M, Nowotny P, Brehm A, et al.
Acad Sci U S A 98(2):457–461. 2007. Characterization of hepatic and brain metabolism in young
Sickmann HM, Waagepetersen HS, Schousboe A, Benie AJ, Bouman SD. adults with glycogen storage disease type 1: a magnetic resonance spec-
2010. Obesity and type 2 diabetes in rats are associated with altered troscopy study. Am J Physiol Endocrinol Metab 293(5):E1378–1384.
brain glycogen and amino-acid homeostasis. J Cereb Blood Flow Wender R, Brown AM, Fern R, Swanson RA, Farrell K, Ransom BR.
Metab 30(8):1527–1537. 2000. Astrocytic glycogen influences axon function and survival
Sorg O, Magistretti PJ. 1991. Characterization of the glycogenolysis elic- during glucose deprivation in central white matter. J Neurosci 20:
ited by vasoactive intestinal peptide, noradrenaline and adenosine 6804–6810.
in primary cultures of mouse cerebral cortical astrocytes. Brain Res Whatley SA, Hall C, Lim L. 1981. Hypothalamic neurons in dissociated
563(1–2):227–233. cell culture: the mechanism of increased survival times in the presence
Stys PK, Waxman SG, Ransom BR. 1992. Ionic mechanisms of anoxic of non-neuronal cells. J Neurochem 36(6):2052–2056.
injury in mammalian CNS white matter: role of Na+ channels and Wiesinger H, Hamprecht B, Dringen R. 1997. Metabolic pathways for
Na+-Ca2+ exchanger. J Neurosci 12(2):430–439. glucose in astrocytes. Glia 21(1):22–34.

G LYC O G E N A N D E N E R GY M ETA B O L I S M • 469


37.
ASTROCY TE REGULATION OF NEUROVASCULAR
CONTROL
Clare Howarth, Grant R. J. Gordon, and Brian A. MacVicar

A B B R E VI AT I O N S 1 INTRODUCTION

AA arachidonic acid It is critical for the maintenance of normal brain function that
ASL arterial spin labeling cerebral blood flow (CBF) is matched to neuronal metabolic
ATP adenosine triphosphate needs. Blood flow is increased to areas in which neurons are
BK channels big potassium channels more active (a response termed functional hyperemia), and it
BOLD blood oxygen level dependent is this blood flow increase that forms the basis of functional
[Ca2+]i intracellular calcium concentration imaging techniques such as blood oxygen level dependent
(BOLD) functional magnetic resonance imaging (fMRI).
CBF cerebral blood flow
Although a coupling between cerebral energy consumption
cGMP cyclic guanosine monophosphate and neuronal activity was originally suggested more than 100
COX cyclooxygenase years ago (Roy and Sherrington 1890), the exact relationship
CSD cortical spreading depression remains an active area of research. Over the past two decades
CYP4A cytochrome P450 4A there has been extensive research into the role astrocyte Ca2+
EETs epoxyeicosatrienoic acids signals play in CBF regulation. Astrocytes, which are the most
EP4 receptor prostaglandin 4 receptor numerous cells in the central nervous system (CNS), are situ-
FITC fluorescein isothiocyanate ated at the synaptic cleft, and every astrocyte has at least one
fMRI functional magnetic resonance endfoot, which is in contact with the vasculature. Therefore,
imaging astrocytes are ideally positioned to relay information regard-
GABA gamma-aminobutyric acid ing changes of synaptic activity to blood vessels so that neu-
GFAP glial fibrillary acidic protein ronal energy demands can be met. In response to glutamate
20-HETE 20-hydroxyeicosatetraenoic acid release by active neurons, astrocyte [Ca2+]i increases and dif-
fusible messengers are released from astrocytic endfeet, which
IOS intrinsic optical signals
are opposed to smooth muscle cells. In this way, astrocytes
[K+]o extracellular potassium communicate with the smooth muscle cells, evoking changes
concentration in arteriole diameter and thus regulating CBF. This chapter
Kir channel inward rectifier potassium channel focuses on the specializations of astrocytes, which make these
LDF laser Doppler flowmetry cells well suited to vascular control, the evidence for synap-
mGluRs metabotropic glutamate receptors tic activity–evoked Ca2+ signals in astrocytes, how such Ca2+
MPEP 2-methyl-6-(phenylethynyl)pyridine signals may evoke changes in vascular diameter, thereby regu-
MRI magnetic resonance imaging lating blood flow, and how this neurovascular coupling can
NADH nicotinamide adenine dinucleotide change in pathology.
NMR nuclear magnetic resonance
nNOS neuronal nitric oxide synthase
NO nitric oxide 2 T H E I M P O RTA N C E O F R E GU L AT I N G
PET positron emission tomography C E R E B R A L B L O O D F L OW
PGE2 prostaglandin E2
2.1 O VE RVI EW: T H E I M P O RTA N C E
PLA2 phospholipase A2 O F S U P P LY I N G S U F F I C I E N T E N E RGY
pO2 oxygen partial pressure FOR THE BRAIN
RBC red blood cells
siRNA small interfering RNA The human brain accounts for only 2% of the body’s weight,
TBOA threo-β-benzyloxyaspartic acid yet uses 20% of the body’s resting energy use (Clarke and
Sokoloff 1999). This high energy demand occurs largely
UV ultraviolet
because the neural processing of information is metabolically

470
expensive. Most of the energy is expended on signaling, with in a fluorescent serum containing nonfluorescent (dark)
the majority being used on the reversal of ion fluxes underlying RBCs. The passage of RBCs within an individual capillary can
synaptic potentials and action potential propagation (Attwell be imaged, allowing determination of RBC velocity, flux, and
and Laughlin 2001; Clarke and Sokoloff 1999). When neural linear density (Kleinfeld et al. 1998). Alternatively, capillary
activity increases, the demand for oxygen and glucose by those diameter changes can be measured to estimate the impact on
neurons increases. Because the total brain energy supply is flow using Poiseuille’s law.
essentially constant, energy resources must be allocated flexi-
bly among regions according to neuronal demand. Therefore, a
tight spatial and temporal coupling exists between local blood 3 T H E R E L AT I O N S H I P O F T H E
flow regulation and neuronal metabolic need. Understanding A S T R O C Y T E E N D F E ET W I T H C E R E B R A L
the mechanisms of neurovascular coupling is essential for B L O O D VE S S E L S
the development of therapies to correct problems with CBF
that occur during disorders such as ischemic stroke, subarach- 3.1 D E S C R I P T I O N O F T H E E N D F O OT
noid hemorrhage, and cerebral palsy after perinatal asphyxia, M O R P H O L O GY A N D C A P I L L A R I E S ,
in which CBF and neuronal metabolic demands can be A RT E R I O L E S , A N D VE I N S T RU C T U R E S
mismatched, leading to neuron and glia damage and death.
Furthermore, it is essential so as to correctly interpret data More than 150 years ago (1858), Virchow commented on the
from experiments applying high resolution functional imag- presence of glia around blood vessels, and almost a century
ing techniques such as BOLD fMRI. ago (1913), Ramon y Cajal suggested that astrocytes are ide-
ally situated to relay information between neurons and the
microvasculature. Single astrocytes extend processes that con-
2.2 R EVI EW O F S T R AT E G I E S TO M E A S U R E tact a large number of synapses (Ventura and Harris 1999),
C E R E B R A L B L O O D F L OW I N H U M A N S A N D other neighboring astrocytes (Massa and Mugnaini 1982),
F U N C T I O NA L H Y P E R E M I A and the cerebrovasculature (Simard et al. 2003). Contacts
Positron emission tomography (PET) can be used to quanti- between astrocytes and brain microvessels are made by end-
tatively measure CBF. A radioactive tracer isotope is injected feet, enlarged compartments at the distal end of astrocytic
into or inhaled by the subject. The isotope decays by emitting processes, which ensheathe microvessels. Collectively, all
a positron, which travels a short distance before encountering astrocyte endfeet completely circumscribe the brain microvas-
an electron. As the positron and electron annihilate each other, culature. The thin barrier of astrocyte endfeet enclosing the
a pair of photons is created. These photons move in opposite blood vessels within the brain is known as the glia limitans
directions and are detected by a coincidence detector. perivascularis. Structural studies have suggested that astrocyte
A noninvasive technique commonly used for brain imaging endfeet could allow direct exchanges of signaling molecules
is magnetic resonance imaging (MRI). In arterial spin labeling and metabolites between neurons and microvessels (Kacem
(ASL), endogenous water proton nuclear spins are labeled in et al. 1998).
the arterial blood by inverting them in the neck region, before The microvasculature consists of arterioles, veins, and cap-
entering the brain. The difference between images acquired illaries. Arterioles are covered by a continuous layer of smooth
with and without ASL can be modeled, allowing calculation muscle formed of annuli around the vessel. The constriction
of a quantitative blood flow image (Williams et al. 1992). and dilation of these smooth muscle cells cause alterations
Alternatively, BOLD contrast fMRI uses changes in deoxy- in the diameter of arterioles and, thereby, blood flow. Veins
hemoglobin levels as an indirect measure of CBF and neural have only a thin layer of smooth muscle cells surrounding the
activity (Ogawa et al. 1990). The iron in deoxyhemoglobin is endothelial cell layer. By contrast, capillaries lack a continuous
paramagnetic, creating local inhomogeneities in the magnetic layer of smooth muscle; instead, contractile cells called peri-
field and a faster decay of the MRI signal. During an increase cytes are located along the capillary wall. Pericytes have recently
in neuronal activity, the oxygen use increases followed within been shown to alter capillary diameter in vitro (Peppiatt et al.
a few seconds by a larger fractional increase in blood flow, 2006) and to constrict brain capillaries in response to ischemia
resulting in a net decrease in the concentration of deoxyhemo- in vivo (Yemisci et al. 2009). However, their role in functional
globin (Fox and Raichle 1986). hyperemia remains unclear because in vivo studies failed to
Laser Doppler flowmetry (LDF) allows continuous measure- find evidence of pericyte-mediated increases in blood flow in
ment of local, relative, CBF. Laser Doppler flowmetry is based on response to neural activity (Fernandez-Klett et al. 2010).
the principle of the Doppler shift: When laser light is reflected
from moving blood cells, the light undergoes a frequency shift;
3.2 K N OWN S P EC I A L I Z AT I O NS O F E N D FE ET
therefore, this reflected light can be used to measure the move-
T H AT A R E R E L AT E D TO VA S C U L A R C O N T RO L
ment of red blood cells (Frerichs and Feuerstein 1990).
Blood flow within capillaries can be visualized and One of the first suggestions that astrocytes have a dynamic
even quantified by observing the passage of red blood cells and active role in brain function, rather than simply being
(RBCs) using two photon laser scanning microscopy in vivo structural support, came from evidence that glutamate could
(Kleinfeld et al. 1998). Injection of a fluorescein isothiocya- induce [Ca2+]i oscillations in cultured astrocytes that propa-
nate (FITC)–conjugated dextran into the bloodstream results gated as waves between adjacent astrocytes (Cornell-Bell et al.

A S T R O C Y T E R E GU L AT I O N O F N E U R O VA S C U L A R C O N T R O L • 471
1990). [Ca2+]i waves in brain slices were also elicited by neu- of adjacent astrocytes and their endfeet. Although both these
ronally released glutamate that acted via astrocytic metabotro- groups evoked a [Ca2+]i increase in astrocytes, opposite vas-
pic glutamate receptors (mGluRs), suggesting that astrocyte cular responses were observed with Zonta et al. (2003a)
networks could act as a signaling system in response to syn- observing vasodilation, whereas Mulligan and MacVicar
aptic activity (Dani et al. 1992; Pasti et al. 1997; Porter and (2004) observed vasoconstriction. Pharmacological inhibi-
McCarthy 1995). tion of 20-hydroxyeicosatetraenoic acid (20-HETE) synthesis
Astrocytic endfeet have many specializations thought to implicated the arachidonic acid (AA) metabolite, 20-HETE
be important for vascular control, including ion channels (see in the arteriolar constriction. By using ultraviolet (UV) light
chapter 16), receptors (see chapter 17), and signaling path- to uncage Ca2+ in astrocyte endfeet, Takano et al. (2006)
ways, allowing astrocytes to detect changes in neural activity examined the role of astrocyte signals in arteriole diameter
and convey this information to the vasculature. The gap junc- control in vivo. Similar to the in vitro findings of Mulligan
tion protein connexin-43 and the purinergic receptors P2Y2 and MacVicar (2004), when Ca2+ was uncaged in the astro-
and P2Y4 are highly expressed in astrocyte vascular endfeet cyte endfoot, a [Ca2+]i rise was triggered within the astrocyte;
in situ (Simard et al. 2003). Calcium signaling is mediated by however, in this case the [Ca2+]i rise was followed by dilation
ATP release from astrocytes via the activation of astrocytic of the adjacent arteriole.
purinergic receptors (Arcuino et al. 2002; Guthrie et al. 1999), Combining all these data, it seems that astrocytes relay
whereas connexin-43 is thought to regulate Ca2+ signaling by information to the vasculature via a Ca2+-evoked AA pathway
controlling ATP release (Cotrina et al. 1998). Hence, vascular and there may be other factors that determine whether vaso-
endfeet are well equipped for the generation and propagation dilation or vasoconstriction occurs. These issues are discussed
of Ca2+ waves. in greater detail later.
Therefore, astrocytes are able to respond with an increase There is a growing body of evidence that astrocytic glu-
in [Ca2+]i to various neurotransmitters released by synaptic tamate uptake may provide an alternative pathway by which
activity. How this information is communicated to the vascu- astrocytes can control CBF. In the olfactory bulb in vivo,
lature, and results in arteriole diameter change is discussed in odor-evoked intrinsic optical signals (IOS, used as a proxy for
section 5. CBF changes) were found to be coupled to action potential–
evoked glutamate release, but surprisingly, were independent
of ionotropic or metabotropic glutamate receptors (Gurden
et al. 2006). However, a broad-spectrum glutamate trans-
4 SY N A P T I C M O D U L AT I O N O F
porter blocker, threo-β-benzyloxyaspartic acid (TBOA),
C E R E B R A L B L O O D F L OW VI A
inhibited the odor-evoked increase in IOS, suggesting that
ASTROCY TES
glial uptake of glutamate mediates the increase in CBF. More
recently, Schummers et al. (2008) combined 2-photon imag-
4.1 I N VI VO EVI D E N C E T H AT A S T RO C Y T E
ing with IOS to investigate the link between the response of
C A L C IUM S I G NA L S A LT E R C E R E B R A L B L O O D
astrocytes to a visual stimulus and hemodynamic signals in the
FL OW A N D/O R A RT E R I O L E D I A M ET E R
visual cortex. In agreement with the earlier study in the olfac-
In vivo work into the role that astrocyte [Ca2+]i signals play tory bulb, 2-photon imaging of cells loaded with a calcium
in functional hyperemia was sparked by early in vitro find- indicator dye revealed that both the increase in astrocytic
ings suggesting that astrocytes were critically involved in the [Ca2+]i and the IOS response evoked by a visual stimulus were
dynamic matching of CBF to neuronal energy demands. significantly reduced in the presence of TBOA. Although
One of the first pieces of evidence that astrocytes may the astrocytic [Ca2+]i transients observed here demonstrated
participate in neurovascular coupling was presented by the a longer latency than the neuronal responses, Ca2+ signals
Carmignoto lab in 2003. In acutely isolated cortical slices, fol- within endfeet were not examined; hence, no conclusion can
lowing mGluR activation or synaptic stimulation, Zonta et al. be drawn about their timescale. Intrinsic optical signals are
(2003a) observed astrocyte soma and endfeet [Ca2+]i signals commonly assumed to reflect a change in hemoglobin oxygen-
that were correlated with the time course of vasodilation. ation and/or blood volume; however, changes in IOS can also
Similar responses were seen when individual astrocytes were be caused by changes in light scattering owing to astrocyte cell
stimulated with patch pipettes. Pharmacologically inhibiting volume changes. Nonetheless, further evidence supporting a
cyclooxygenase (COX) blocked the vasodilation, implying role for astrocyte uptake of glutamate in the control of CBF
that COX products (e.g., prostaglandin E2 [PGE2]) played a was presented by Petzold et al. (2008), who used multipho-
role in the vasodilation. Zonta et al. (2003a) extended their ton microscopy to reveal that odor-evoked [Ca2+]i elevations
findings to the in vivo case, using LDF to demonstrate that in astrocytic endfeet were strongly correlated, both tempo-
the CBF increase in the contralateral somatosensory cortex rally and spatially, with increases in arteriolar cross-sectional
following forepaw stimulation was reduced by an mGluR area. Pharmacological experiments implicated two indepen-
antagonist. This suggested that mGluRs play a key role in dent pathways by which glutamate could mediate functional
functional hyperemia in vivo. A year later, in hippocampal hyperemia. The first was via mGluR5, which in the olfactory
slices, the MacVicar lab used two-photon fluorescence micros- glomerulus is exclusively expressed by astrocytes. Injection
copy and uncaging to evoke [Ca2+]i transients directly within of 2-methyl-6-(phenylethynyl)pyridine (MPEP), a selective
an astrocyte soma. [Ca2+]i waves spread through the network mGluR5 antagonist, into the bulb resulted in a large reduction

472 • FUNCTIONS OF NEUROGLIAL CELLS


in both odor-evoked astrocytic [Ca2+]i increases and functional Membrane phopholipids
hyperemia. The second was via astrocytic glutamate uptake.
Blocking glial glutamate transporters reduced odor-evoked Phospholipase A2 Ca2+
increases in blood flow, but in contrast to the results in the
visual cortex (Schummers et al. 2008), had no effect on the Arachidonic Acid
astrocyte calcium response. These studies imply that astrocytic
glutamate uptake is an important pathway contributing to the

Cyclooxygenases
regulation of arteriole diameter. However, the cellular mecha-

Lipoxygenase
nisms underlying this pathway are as yet unresolved.
After a decade of research investigating astrocytes as poten-

Epoxygenases
tial mediators of functional hyperemia, the idea remains contro-

ω–hydroxylase
versial. The presence, prevalence, and timing of astrocyte [Ca2+]i

(CYP4A)
signaling in response to neural activity and its role in vivo with HPETE
PgH2
respect to control of CBF is still hotly debated. Using in vivo
2-photon imaging of astrocytes within the mouse whisker bar- EETs
rel cortex, Wang et al. (2006) reported only delayed increases
in astrocyte [Ca2+]i in response to whisker stimulation. It has
20-HETE Leukotrienes
been suggested that these [Ca2+]i transients, which peak at 9
seconds, are too slow to mediate functional hyperemia, which
can occur within hundreds of milliseconds after the onset of
neuronal activity (Kleinfeld et al. 1998; Zonta et al. 2003a).

Thromboxane
Prostacyclin
However, in contrast to these observations, other groups have

synthase

synthase

synthase

synthase

synthase
PgD
PgE

PgF
found evidence consistent with a role for astrocytes in func-
tional hyperemia. Using two-photon in vivo imaging, Winship
et al. (2007) reported that approximately 5% of astrocytes in
the hindlimb area of the mouse primary somatosensory cortex
displayed short-latency, limb-specific [Ca2+]i transients in their PgI2 PgE2 PgD2 PgF2a TxA2
soma and endfeet. The kinetics of these Ca2+ signals are on a
similar timescale to neuronal activity and are correlated with Figure 37.1 Following the activation of PLA2, arachidonic acid is formed
the onset of functional hyperemia, measured by IOS. from membrane phospholipids. Arachidonic acid is metabolized by the
enzymes shown to form vasodilators (EETs, PGE2, PgD2, and PgI2) or
vasoconstrictors (PgH2, 20-HETE, PgF2α, TxA2, and leukotrienes).

5 BIDIRECTIONAL CONTROL OF
A RT E R I O L E D I A M ET E R BY A S T R O C Y T E
CALCIUM 5.2 P RO S TAG L A N D I N E2 A N D
E P OX Y E I C O S AT R I E N O I C AC I D S
5.1 A R AC H I D O N I C AC I D M ETA B O L I T E S C AUS E D I L AT I O NS
A N D VA S C U L A R C H A N G E S
Immunohistochemistry using antibodies against COX-1 and
Increases in astrocyte [Ca2+]i have been reported to evoke both GFAP revealed that astrocytes in situ express COX-1 in their
vasodilation (Petzold et al. 2008; Takano et al. 2006; Zonta endfeet (Gordon et al. 2008; Takano et al. 2006). Notably,
et al. 2003a) and vasoconstriction (Chuquet et al. 2007; astrocyte processes far from vessels are negative for COX-1
Mulligan and MacVicar 2004). Both responses can result from (Takano et al. 2006), suggesting that COX-1 is localized to
neuronally released glutamate acting on astrocyte mGluRs, the vascular region. In cultured astrocytes, COX activity
increasing astrocyte [Ca2+]i (Pasti et al. 1997). Elevations triggered by glutamate agonists resulted in the generation of
in astrocyte [Ca2+]i activate phospholipase A2 (PLA2), a PGE2 (Zonta et al. 2003b), a vasodilator (Niwa et al. 2000;
Ca2+-sensitive enzyme that is highly expressed in astrocytes Takano et al. 2006; Zonta et al. 2003a). Following its release
(Cahoy et al. 2008), resulting in arachidonic acid (AA) from astrocyte endfeet, PGE2 can bind to EP4 receptors on
release from the plasma membrane. Arachidonic acid can be smooth muscle cells, evoking hyperpolarization and vasodi-
metabolized within the astrocyte to produce COX-generated lation. Based on results using COX inhibitors (Takano et al.
derivatives such as PGE2 and cytochrome P450 (CYP) epox- 2006; Zonta et al. 2003a), PGE2 was suggested to mediate
ygenase-derived epoxyeicosatrienoic acids (EETs) (Fig. 37.1). a significant component of the dilatory response to raised
Arachidonic acid is highly diffusible and can be released from astrocyte [Ca2+]i. However, controversy remains regarding
the astrocyte and metabolized within the smooth muscle cell the role of COX-1 in astrocyte-mediated vasodilation in
to form 20-HETE. Hence, astrocytes are capable of produc- response to increased neural activity in vivo. Although a
ing and releasing several vasoactive compounds in response high dose of the COX-1 specific inhibitor SC560 (500μM)
to synaptic activity, evoking vasodilation or vasoconstriction can inhibit the increase in CBF in response to odorant
depending on which AA metabolism pathway dominates. stimulation of the olfactory bulb (Petzold et al. 2008) or

A S T R O C Y T E R E GU L AT I O N O F N E U R O VA S C U L A R C O N T R O L • 473
uncaging of Ca2+ in astrocytes (Takano et al. 2006), several observed in response to either light stimulation or uncaging
groups have found that lower doses have no effect on the of Ca2+ within glial cells (Metea and Newman 2006), whereas
CBF response to whisker stimulation (Liu et al. 2011a; Niwa inhibition of COX activity had no effect, suggesting that
et al. 2001). In addition, using genetic knock-out of COX-1 glial-evoked vasodilations in the retina are mediated by EETs
had no effect on functional hyperemia, whereas attenuating rather than COX products.
hypercapnia-evoked CBF increases (Niwa et al. 2001). In
contrast, pharmacological inhibition or genetic knock-out of
5.3 20-H Y D ROX Y E I C O S AT ET R A E N O I C
COX-2 leads to a reduction in the CBF response to neuronal
AC I D C AUS E S C O NS T R I C T I O NS
activation (Liu et al. 2011a; Niwa et al. 2000). As COX-2 is
more highly expressed in neurons than astrocytes, neuronal Downstream of Ca2+-evoked activation of PLA2, AA can dif-
COX activity and release of COX products may underlie fuse from the astrocyte endfoot to the smooth muscle layer
functional hyperemia. surrounding the arteriole, producing 20-HETE (Lange et al.
Astrocytes express CYP epoxygenase, CYP 2C11, which 1997; Mulligan and MacVicar 2004) via ω-hydroxylases
can metabolize AA to form vasodilatory epoxyeicosa- (Roman 2002). 20-hydroxyeicosatetraenoic acid inhibits K+
trienoic acids (EETs) (Alkayed et al. 1996; Ellis et al. 1990). channels, thus evoking more depolarization activated Ca2+
Epoxyeicosatrienoic acids activate K+ channels and hyper- entry, and hence, arteriole constriction (Lange et al. 1997)
polarize smooth muscle cells, thus inhibiting voltage-gated (Fig. 37.2).
Ca2+ channels and evoking vasodilation (Alkayed et al. 1997;
Gebremedhin et al. 1992; Hu and Kim 1993). In cultured
5.4 RO L E S F O R P OTA S S IUM E FFLUX FRO M
astrocytes, the production of EETs can be stimulated by gluta-
A S T RO C Y T E S I N C H A N G I N G A RT E R I O L E TO N E
mate agonists (Alkayed et al. 1997). In vivo evidence has sug-
gested that the cortical increase in CBF in response to both Increases in extracellular potassium concentration ([K+]o)
mGluR activation (Liu et al. 2011b) and whisker stimulation evoke vasodilation (Kuschinsky and Wahl 1978) by hyperpo-
(Liu et al. 2011a) is largely dependent on EET production. larizing smooth muscle cells through the action of inward rec-
Furthermore, in ex vivo retina, pharmacological inhibition tifier K+ (Kir) channels, leading to a decrease in calcium entry
of EET synthesis reduced the magnitude of vasodilations through voltage-gated channels and vasodilation (Knot et al.

Synapse
glu
mGluR
NMDAR
Ca2+ Astrocyte

Ca2+ PLA2*

AA
Neuron nNOS

NO

EETs PgE2
NO

Smooth NO AA 20HETE Basement


muscle cell cGMP* dilation dilation membrane
constriction

Red
blood cell
Endothelial
cells

Figure 37.2 Glutamate-Mediated Pathways of Cerebral Blood Flow Regulation. Synaptically released glutamate can act on neuronal NMDA recep-
tors (NMDAR), increasing [Ca2+]i, activating neuronal nitric oxide synthase (nNOS) and leading to the release of nitric oxide (NO). Nitric oxide can
act directly on the smooth muscle cells, evoking vasodilation via cGMP activation. Glutamate can also act on astrocytic mGluRs, increasing astrocyte
[Ca2+]i, activating PLA2 resulting in the release of AA from the plasma membrane. Arachidonic acid can be metabolized within the astrocyte to form
PGE2 or EETs, which are released and act on the smooth muscle cell, evoking vasodilation. Alternatively, AA can be released and act on smooth
muscle cells, where it is metabolized to the vasoconstrictor 20-HETE.

474 • FUNCTIONS OF NEUROGLIAL CELLS


1996). The high potassium conductance of astrocyte endfeet lactate and adenosine. Under these conditions, vasodilation by
(Newman 1986) led to the hypothesis that astrocytic “siphon- PGE2 was facilitated because extracellular lactate can inhibit
ing” of potassium in response to neuronal activity plays a role prostaglandin uptake (Chan et al. 2002), whereas adenosine
in local blood flow regulation (Paulson and Newman 1987). can act directly on smooth muscle A2A receptors, blocking
Following neuronal activity, a local increase in [K+]o leads to Ca2+ channels (Murphy et al. 2003) and preventing vaso-
an influx of K+ into glial cells, depolarizing the glial cells and, constriction. These findings are supported by several studies
in addition, causing K+ efflux from quiescent regions of the in vivo in animals and humans, demonstrating that changing
cell. It was hypothesized that most K+ would be “siphoned” the external lactate/pyruvate ratio, and hence directly con-
through the endfeet (where the highest density of Kir 4.1 trolling the intracellular NADH/NAD+ ratio, regulates CBF
occurs) (Brew et al. 1986; Newman 1986) onto blood vessels. (Ido et al. 2004; Mintun et al. 2004; Vlassenko et al. 2006).
However, recent work by the Newman lab found that direct More recently, Lin et al. (2010) used NMR spectroscopy to
depolarization of astrocytes with patch pipettes, which should demonstrate a strong correlation between the lactate signal
provide driving force for K+ efflux, did not cause vasodilation, and CBF increases. However, the role of O2 levels in deter-
thus concluding that glial K+ siphoning does not contribute mining functional hyperemia remains controversial. In one
significantly to neurovascular coupling in the retina (Metea study, changing tissue pO2 by altering the level of O2 breathed
et al. 2007). in resulted in a change in basal CBF, the direction of which
An alternative pathway involving the efflux of K+ through matched the predictions of the in vitro work (Lu et al. 2009).
BK channels (expressed in astrocytes) (Price et al. 2002) has However, in more recent work the functional hyperemia
been proposed (Filosa et al. 2006), although it is difficult to response to sensory stimulation was not altered in animals
imagine how such a scenario would occur because glial mem- in a hyperbaric chamber breathing high O2 (Lindauer et al.
brane potentials are close to the equilibrium potential for 2010). It is interesting to note that although forepaw stimu-
potassium (Kuffler et al. 1966), meaning that increasing K+ lation evokes an increase in CBF and glucose metabolism in
conductance would not increase the net efflux of K+. the contralateral hemisphere as expected, there is a decrease
in CBF but an increase in glucose metabolism in the ipsilat-
eral hemisphere (Devor et al. 2008), indicating that a complex
5.5 R E L AT I O NS H I P TO OT H E R K N OWN interaction occurs between metabolic and neurogenic CBF
FAC TO R S T H AT H AVE VA S CU L A R AC T I O NS signaling factors.
Adenosine triphosphate (ATP), which acts as an extracellular Blanco et al. (2008) hypothesized that there is a vascu-
messenger to propagate astrocytic [Ca2+]i waves in response to lar tone set point that modulates the polarity of the arte-
stimulation (Guthrie et al. 1999), is also a potent vasomodu- riole’s response to astrocyte signals. As vascular tone was
lator (Peppiatt et al. 2006). Adenosine, produced when ATP increased, larger vasodilation responses were evoked by
is broken down by ectoenzymes during neuronal activation, increased [K+]o, whereas the polarity of the response to
has been shown to be vasodilatory in both the cerebral cortex mGluR activation was found to be dependent on initial levels
and cerebellum, and is thought to be involved in the coupling of tone. The importance of this pathway in vivo remains to be
of CBF to neuronal activity (Akgoren et al. 1997; Dirnagl determined.
et al. 1994). Not only does adenosine play a significant role It is likely that all the elements described in this chapter act
in the functional hyperemia response in vivo (Shi et al. 2008), together to ensure that information about increased synaptic
but also it can potentiate the [Ca2+]i increase in both retina activity is relayed to the vasculature via the astrocyte network
(Newman 2003) and cerebellum ( Jimenez et al. 1998). Hence, so as to match blood flow with neuronal energy demands.
the fast and transient ATP-evoked increase in astrocyte [Ca2+]i This feed forward model allows for faster alterations of CBF
is potentiated by the ectoenzymatic breakdown product of in response to neural activity compared with the previously
ATP, adenosine. suggested negative feedback models that rely on the buildup
In the parenchyma, both the partial pressure of oxygen of metabolic products such as CO2 or an increase in the use
(pO2) (Offenhauser et al. 2005), and extracellular lactate of O2 or glucose.
concentration (Hu and Wilson 1997), change during neural
activity. Recently, Gordon et al. (2008) demonstrated that
the metabolic state of brain tissue determines the polarity of
the response to increased astrocyte [Ca2+]i. Clamping O2 at a 6 I N F LU E N C E O F N E U R O N S O N
high or low level evoked vasoconstrictions and vasodilations, A S T R O C Y T E –VA S C U L A R C O U P L I N G
respectively, in response to uncaging Ca2+ within astrocytes.
6.1 T R A NS M IT T E R AC T I VAT I O N O F
Low O2 was achieved by bubbling ACSF with 20% O2, 5%
A S T RO C Y T E C A L C IUM S I G NA L S L E A D I N G TO
CO2, resulting in a pO2 of approximately 19 mmHg at 50
C A L C IU M WAVE S A N D VA S C U L A R C H A N G E S
μm below the surface, where many imaging and electrophysi-
ology experiments are performed, which is similar to physi- Local blood flow control is achieved by fast neurotransmitter-
ological pO2 levels (12.7–64.4 mmHg in vivo) (Offenhauser mediated signaling (Attwell and Iadecola 2002). Both glu-
et al. 2005). At these lower O2 levels, the metabolic tamate and NMDA elicit vasodilatory responses in neocor-
state of the tissue was altered, resulting in raised levels of tical and hippocampal microvessels (Fergus and Lee 1997b).

A S T R O C Y T E R E GU L AT I O N O F N E U R O VA S C U L A R C O N T R O L • 475
Astrocytes are ideally located because of their close proximity on the vasculature, many enzymes responsible for the produc-
to synaptic clefts, to receive synaptic information via released tion of vasoactive molecules from AA are sensitive to NO.
neurotransmitters. Appropriately, astrocytes in situ and in vivo Pharmacological experiments in vivo suggested that increased
express several neurotransmitter receptors, including gluta- NO production by nNOS during functional activation in the
matergic, adrenergic, GABAergic, and purinergic receptors whisker barrel cortex may act to suppress 20-HETE synthesis
(see chapters 17 and 25) (see review by Porter and McCarthy or its downstream signaling and so promote EETs-dependent
1997, and references therein). These receptors are coupled to vasodilation (Liu et al. 2008). In contrast to the cortex, in the
intracellular signaling pathways, which can lead to changes in cerebellum NO donors do not reverse the effect of inhibiting
vascular diameter and/or cerebral blood flow. Astrocytes in situ nNOS; hence, NO is a mediator of the functional hyperemia
express mGluRs, in particular mGluR5 (van den Pol et al. 1995). response in cerebellum (Akgoren et al. 1996).
During neuronal activity, glutamate is released, which can act Nitric oxide levels have been hypothesized to dictate
on astrocyte mGluRs, raising astrocyte [Ca2+]i (Pasti et al. 1997) whether vasodilation or vasoconstriction occurs in response
resulting in AA-mediated changes in arteriole diameter. to increased glial [Ca2+]i in the retina (Metea and Newman
In the brain, astrocytes are a major target for noradrenergic 2006). Constrictions within the retina were caused by AA
innervations. Astrocytes in situ have shown immunoreactiv- conversion to 20-HETE, in agreement with work in brain
ity for β-adrenergic receptors in the cortex and neostriatum slices (Mulligan and MacVicar 2004), whereas vasodila-
(Aoki et al. 1987; Liu et al. 1992), whereas astrocytes in the tions were caused by conversion of AA into EETS. Nitric
hippocampus, trigeminal motor nucleus, cerebral cortex, oxide can inactivate CYP2C11 (Roman 2002), thus inhibit-
striatum, and cerebellum express α1-adrenoceptors (Duff y ing synthesis of EETs and vasodilation. As NO levels in the
and MacVicar 1995; Shao and Sutin 1992). In vivo, CBF is retina rose, the frequency of vasodilations diminished, reveal-
decreased following activation by noradrenaline (Raichle ing only vasoconstrictions (Metea and Newman 2006). The
et al. 1975). In addition to its direct effects on smooth muscle MacVicar group had also observed that vasoconstrictions in
cells, noradrenaline can evoke Ca2+ transients in astrocytes in response to astrocyte stimulation were switched to vasodila-
hippocampal slices (Duff y and MacVicar 1995) and the cortex tions in the presence of the NO synthase inhibitor, L-NAME
in vivo (Bekar et al. 2008). This increase in astrocyte [Ca2+]i (Mulligan and MacVicar 2004). Initially this was thought to
can result in the release of AA from astrocyte endfeet and its be caused by changes in resting vessel tone, because L-NAME
conversion to 20-HETE. causes preconstriction of vessels, thereby predisposing them to
Functional GABA receptors are expressed by astrocytes in dilate (Blanco et al. 2008). Differences in the basal level of the
the hippocampus (Fraser et al. 1995) and Bergmann glia in the NO of the two preparations may account for the differences
cerebellum (Muller et al. 1994). The inhibitory neurotransmit- seen, that is, the NO level of brain slices may be higher than
ter, GABA, has been implicated in neurovascular signaling in in the retina, explaining the prevalence of vasoconstrictions
the hippocampus (Fergus and Lee 1997a), where activation of observed by the MacVicar group, as the EET pathway could
GABAA receptors (Fraser et al. 1995) results in an efflux of Cl– be blocked by the basal level of NO. In addition to the effect
and depolarization of astrocytes, activating voltage-gated Ca2+ of NO on the CYP2C11 enzyme, the enzymes responsible for
channels and increasing astrocyte [Ca2+]i. In addition, GABA producing 20-HETE are also sensitive to NO (Roman 2002),
released from hippocampal interneurons causes GABAB suggesting that a complex relationship may exist between NO
receptor–mediated elevations of [Ca2+]i in astrocytes (Kang levels and vasomotor responses. Several groups have demon-
et al. 1998), in a mechanism analogous to that proposed for strated that interactions exist among the NO, cyclooxygenase,
glutamate (Zonta et al. 2003a), and dilates arterioles (Fergus and epoxygenase pathways. Combining inhibition of these
and Lee 1997a). pathways does not result in a greater block of CBF increase
than inhibiting them individually (Liu et al. 2011a; Peng et al.
2004) (Fig. 37.3).
6.2 R E L E A S E O F N I T R I C OX I D E F RO M N I T R I C
OX I D E SY N T H A S E AC T I VAT I O N
Synaptically released glutamate can act on NMDA/AMPA 7 A LT E R AT I O N S O F A S T R O C Y T E A N D
receptors to raise neuronal [Ca2+]i, activating neuronal nitric N E U R O VA S C U L A R C O U P L I N G I N
oxide synthase (nNOS) (Fergus and Lee 1997b). The release PAT H O L O G I C A L C O N D I T I O N S
of nitric oxide (NO) activates guanylate cyclase in vascular
smooth muscle, producing cyclic guanosine monophosphate Failure to provide sufficient blood flow to match the energy
(cGMP). Cyclic guanosine monophosphate activates K+ demands of neurons can lead to cell damage or death follow-
channels, producing a hyperpolarization that reduces calcium ing several pathological conditions, including ischemic stroke,
entry into the muscle, resulting in vasodilation and increased secondary ischemia after spinal cord injury, and vasospasm
blood flow (Akgoren et al. 1994; Robertson et al. 1993). after subarachnoid hemorrhage. Hence, alterations in neu-
In the cortex, inhibition of nNOS reduced the increase in rovascular coupling may underlie neurological deficits that
blood flow evoked by neuronal activity (Ma et al. 1996). The occur following such conditions.
response can be restored by NO donors, suggesting that NO is Ischemic stroke occurs when there is a failure of the brain
a modulator rather than a mediator of the blood flow response blood flow because of a blockage (e.g., a blood clot in an arte-
(Lindauer et al. 1999). In addition to the direct effects of NO riole). Following clearance of the blockage there is an initial

476 • FUNCTIONS OF NEUROGLIAL CELLS


Synapse
glu
mGluR
NMDAR
Ca2+ Muller cell
or
Ca 2+ PLA2* Astrocyte
AA
Neuron nNOS

NO

EETs
NO

Smooth NO 20HETE AA Basement


cGMP* dilation dilation
muscle cell constriction membrane

Red
blood cell
Endothelial
cells

Figure 37.3 Nitric Oxide Effects on Glutamate-Mediated Retinal Blood Flow Changes. As seen in Figure 37.2, glutamate can lead to the release of NO
from neurons. Nitric oxide can evoke vasodilation via cGMP activation. Alternatively, NO can inhibit the conversion of AA to 20-HETE by the
enzyme ω-hydroxylase, hence promoting vasodilation. Nitric oxide can also inhibit dilation by inhibiting CYP450 epoxygenase, which converts AA to
EETs. These enzymes have different sensitivities to NO.

transient increase in blood flow followed by a decrease that subarachnoid hemorrhage (Dreier et al. 2006). The transient
can last for several hours (Ames et al. 1968; Hauck et al. 2004; increase in blood flow is thought to be caused by the release
Nelson et al. 1992). It was originally hypothesized that this of calcitonin gene–related peptide (CGRP) by trigeminal
long-lasting decrease in blood flow, which may result in fur- neurons (Wahl et al. 1994), although the target cell of CGRP
ther damage to cells after the initial stroke damage, was caused remains to be confirmed. In addition, vasodilation may, in part,
by astrocyte endfeet swelling, causing capillary diameters to be caused by the release of NO (Fabricius et al. 1995; Wahl
be reduced and capillaries to become blocked by blood cells et al. 1994), possibly because of NO inhibition of the synthesis
(Ames et al. 1968). However, the importance of this effect is of 20-HETE. In vivo, two-photon imaging of astrocytes loaded
debated (Theilen et al. 1994), and it has been suggested that with a calcium indicator dye combined with vascular labeling
a failure of arteriole vasodilatory mechanisms underlies the with FITC dextran revealed that vasoconstrictions of penetrat-
CBF decrease (Leffler et al. 1989; Nelson et al. 1992). Because ing cortical arterioles occurred during CSD at the onset of a
it is difficult to tell these two situations apart experimentally, it fast astrocyte [Ca2+]i wave (Chuquet et al. 2007). The evoked
remains unclear which pathway is more important. Recently, vasoconstriction was inhibited in the presence of thapsigargin,
pericyte constrictions were proposed to play a key role in which inhibits the refilling of internal calcium stores, and
the long-lasting decrease in blood flow (Peppiatt et al. 2006; Methyl arachidonyl fluorophosphonate (MAFP), which inhib-
Yemisci et al. 2009). its PLA2, suggesting that astrocytes mediate the CSD-induced
Cortical spreading depression (CSD) is a phenomenon that vasoconstriction that occurs via the PLA2-mediated release of
occurs during migraine and following stroke, brain trauma or AA, in agreement with previous in vitro findings (Mulligan and
subarachnoid hemorrhage. During CSD, there is a transient rise MacVicar 2004). However, this study only investigated the ini-
in [K+]o from approximately 3 mM to approximately 50 mM, tial vasoconstriction that occurs before the transient increase in
leading to depolarization of neurons and astrocytes (resulting CBF. Impairments in the NO system (Fabricius et al. 1995) are
in increased [Ca2+]i) and a rise in glutamate (Vanharreveld partly responsible for the disruption in functional hyperemia
and Kooiman 1965) because of release by neurons and a fail- that occurs after CSD (Wahl et al. 1987). A decrease in NO
ure of uptake by glia (Barbour et al. 1988). Although energy levels could result in either less dilation of vessels because of a
use by the brain increases during CSD, after an initial transi- direct effect of NO on smooth muscle cells or less inhibition of
ent increase, CBF is decreased for hours (Fabricius et al. 1995; 20-HETE production by NO.
Takano et al. 2007). This mismatch between energy supply and Finally, in Alzheimer disease, which is characterized by the
demand may result in increased neuronal damage following deposition of amyloid-β peptide in the neuropil and blood

A S T R O C Y T E R E GU L AT I O N O F N E U R O VA S C U L A R C O N T R O L • 477
Akgoren N, Fabricius M, Lauritzen M. 1994. Importance of nitric oxide
vessels and the accumulation of neurofibrillary tangles in for local increases of blood flow in rat cerebellar cortex during electri-
neurons, resting CBF is reduced and functional hyperemia is cal stimulation. Proc Natl Acad Sci U S A 91(13):5903–5907.
impaired (Girouard and Iadecola 2006). Amyloid-β peptide Akgoren N, Mathiesen C, Rubin I, Lauritzen M. 1997. Laminar analysis
promotes oxidative stress, which inhibits the production of of activity-dependent increases of CBF in rat cerebellar cortex: depen-
vasodilating messengers; hence, decreasing functional hyper- dence on synaptic strength. Am J Physiol 273(3 Pt 2): H1166–1176.
Alkayed NJ, Birks EK, Narayanan J, Petrie KA, Kohler-Cabot AE,
emia (Sun et al. 2008; Zou et al. 1999). Furthermore, mice Harder DR. 1997. Role of P-450 arachidonic acid epoxygenase
with amyloid-β plaques had an enhanced occurrence of spon- in the response of cerebral blood flow to glutamate in rats. Stroke
taneous astrocyte [Ca2+]i waves (Kuchibhotla et al. 2009) and 28(5):1066–1072.
increased contractility of vascular smooth muscle cells (Chow Alkayed NJ, Narayanan J, Gebremedhin D, Medhora M, Roman RJ,
et al. 2007), these alterations could underlie changes in neuro- Harder DR. 1996. Molecular characterization of an arachidonic acid
epoxygenase in rat brain astrocytes. Stroke 27(5):971–979.
vascular coupling. Ames A 3rd, Wright RL, Kowada M, Thurston JM, Majno G. 1968.
Cerebral ischemia. II. The no-reflow phenomenon. Am J Pathol
52(2):437–453.
8 S U M M A RY A N D P E R S P E C T I VE S Aoki C, Joh TH, Pickel VM. 1987. Ultrastructural localization of
beta-adrenergic receptor-like immunoreactivity in the cortex and
Over the past two decades much progress has been made in neostriatum of rat brain. Brain Res 437(2):264–282.
Arcuino G, Lin JH, Takano T, Liu C, Jiang L, Gao Q, et al. 2002.
understanding the role of astrocytes and their Ca2+ signals in Intercellular calcium signaling mediated by point-source burst release
the regulation of arteriole diameter, and thereby blood flow in of ATP. Proc Natl Acad Sci U S A 99(15):9840–9845.
response to changes in synaptic activity. It is clear that astro- Attwell D, Iadecola C. 2002. The neural basis of functional brain imag-
cytes are ideally positioned to sense changes in synaptic activity ing signals. Trends Neurosci 25(12):621–625.
and relay this information to the cerebrovasculature, releasing Attwell D, Laughlin SB. 2001. An energy budget for signaling in the grey
matter of the brain. J Cereb Blood Flow Metab 21(10):1133–1145.
gliotransmitters from their endfeet onto smooth muscle cells Barbour B, Brew H, Attwell D. 1988. Electrogenic glutamate uptake
and thus evoking changes in arteriole diameter. However, sev- in glial cells is activated by intracellular potassium. Nature
eral questions remain. Do astrocyte Ca2+ signals reliably occur 335(6189):433–435.
under physiological conditions and, if so, is their timing relevant Bekar LK, He W, Nedergaard M. 2008. Locus coeruleus
to the regulation of CBF? What determines whether a vaso- alpha-adrenergic-mediated activation of cortical astrocytes in vivo.
Cereb Cortex 18(12):2789–2795.
constriction or vasodilation response dominates in vivo? Does Blanco VM, Stern JE, Filosa JA. 2008. Tone-dependent vascular
this factor depend on the physiological conditions or stimulus? responses to astrocyte-derived signals. Am J Physiol Heart Circ
What role does glial glutamate uptake play in the regulation Physiol 294(6):H2855–2863.
of CBF and how is this effect mediated? The use of new tech- Brew H, Gray PT, Mobbs P, Attwell D. 1986. Endfeet of retinal glial
niques such as genetic manipulation (including optogenetics, cells have higher densities of ion channels that mediate K+ buffering.
Nature 324(6096):466–468.
gene knock-out, and siRNA techniques), which enable specific Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL,
manipulation of astrocytes, their Ca2+ signaling, and the down- Christopherson KS, et al. 2008. A transcriptome database for astro-
stream pathways, which result in changes in arteriole diameter, cytes, neurons, and oligodendrocytes: a new resource for understand-
will allow researchers to address these questions. The combina- ing brain development and function. J Neurosci 28(1):264–278.
tion of optogenetics with BOLD fMRI will be a powerful tool Chan BS, Endo S, Kanai N, Schuster VL. 2002. Identification of lactate
as a driving force for prostanoid transport by prostaglandin trans-
to allow researchers to specifically target activation of astro- porter PGT. Am J Physiol Renal Physiol 282(6):F1097–1102.
cytes while using genetic or pharmacological manipulation to Chow N, Bell RD, Deane R, Streb JW, Chen J, Brooks A, et al. 2007.
alter the pathways underlying functional hyperemia. Serum response factor and myocardin mediate arterial hypercontrac-
tility and cerebral blood flow dysregulation in Alzheimer’s pheno-
type. Proc Natl Acad Sci U S A 104(3):823–828.
AC K N OW L E D G M E N T S Chuquet J, Hollender L, Nimchinsky EA. 2007. High-resolution in
vivo imaging of the neurovascular unit during spreading depression.
J Neurosci 27(15):4036–4044.
CH is a Sir Henry Wellcome Postdoctoral Fellow, a Clarke DD, Sokoloff L. 1999. Basic neurochemistry: molecular, cellular
Government of Canada Postdoctoral Fellow, and a Michael and medical aspects. Philadelphia: Lippincott.
Smith Foundation for Health Research Postdoctoral Fellow. Cornell-Bell AH, Finkbeiner SM, Cooper MS, Smith SJ. 1990.
BAM is a Canada Research Chair Tier 1 in Neuroscience. Glutamate induces calcium waves in cultured astrocytes: long-range
GRJG is a Canada Research Chair Tier 2 and a Heart and glial signaling. Science 247(4941):470–473.
Cotrina ML, Lin JH, Alves-Rodrigues A, Liu S, Li J, Azmi-Ghadimi H,
Stroke Foundation of Canada McDonald Scholar. BAM is et al. 1998. Connexins regulate calcium signaling by controlling ATP
funded by CIHR, the Fondation Leducq, Human Frontiers release. Proc Natl Acad Sci U S A 95(26):15735–15740.
Science Program, and Heart and Stroke Foundation. Dani JW, Chernjavsky A, Smith SJ. 1992. Neuronal activity trig-
gers calcium waves in hippocampal astrocyte networks. Neuron
8(3):429–440.
Devor A, Hillman EM, Tian P, Waeber C, Teng IC, Ruvinskaya L, et al.
REFERENCES 2008. Stimulus-induced changes in blood flow and 2-deoxyglucose
uptake dissociate in ipsilateral somatosensory cortex. J Neurosci
Akgoren N, Dalgaard P, Lauritzen M. 1996. Cerebral blood flow 28(53):14347–14357.
increases evoked by electrical stimulation of rat cerebellar cortex: Dirnagl U, Niwa K, Lindauer U, Villringer A. 1994. Coupling of cere-
relation to excitatory synaptic activity and nitric oxide synthesis. bral blood flow to neuronal activation: role of adenosine and nitric
Brain Res 710(1–2):204–214. oxide. Am J Physiol 267(1 Pt 2):H296–301.

478 • FUNCTIONS OF NEUROGLIAL CELLS


Dreier JP, Woitzik J, Fabricius M, Bhatia R, Major S, Drenckhahn C, Kang J, Jiang L, Goldman SA, Nedergaard M. 1998. Astrocyte-mediated
et al. 2006. Delayed ischaemic neurological deficits after subarach- potentiation of inhibitory synaptic transmission. Nat Neurosci
noid hemorrhage are associated with clusters of spreading depolariza- 1(8):683–692.
tions. Brain 129(Pt 12):3224–3237. Kleinfeld D, Mitra PP, Helmchen F, Denk W. 1998. Fluctuations and
Duff y S, MacVicar BA. 1995. Adrenergic calcium signaling in astrocyte stimulus-induced changes in blood flow observed in individual capil-
networks within the hippocampal slice. J Neurosci 15(8):5535–5550. laries in layers 2 through 4 of rat neocortex. Proc Natl Acad Sci U S
Ellis EF, Police RJ, Yancey L, McKinney JS, Amruthesh SC. 1990. A 95(26):15741–15746.
Dilation of cerebral arterioles by cytochrome P-450 metabolites of Knot HJ, Zimmermann PA, Nelson MT. 1996. Extracellular K+-induced
arachidonic acid. Am J Physiol 259(4 Pt 2):H1171–1177. hyperpolarizations and dilatations of rat coronary and cerebral
Fabricius M, Akgoren N, Lauritzen M. 1995. Arginine-nitric oxide path- arteries involve inward rectifier K+ channels. J Physiol 492 (Pt 2):
way and cerebrovascular regulation in cortical spreading depression. 419–430.
Am J Physiol 269(1 Pt 2):H23–29. Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. 2009.
Fergus A, Lee KS. 1997a. GABAergic regulation of cerebral microvascu- Synchronous hyperactivity and intercellular calcium waves in astro-
lar tone in the rat. J Cereb Blood Flow Metab 17(9):992–1003. cytes in Alzheimer mice. Science 323(5918):1211–1215.
Fergus A, Lee KS. 1997b. Regulation of cerebral microvessels by gluta- Kuffler SW, Nicholls JG, Orkand RK. 1966. Physiological properties of
matergic mechanisms. Brain Res 754(1–2):35–45. glial cells in the central nervous system of amphibia. J Neurophysiol
Fernandez-Klett F, Offenhauser N, Dirnagl U, Priller J, Lindauer U. 29(4):768–787.
2010. Pericytes in capillaries are contractile in vivo, but arterioles Kuschinsky W, Wahl M. 1978. Local chemical and neurogenic regula-
mediate functional hyperemia in the mouse brain. Proc Natl Acad Sci tion of cerebral vascular resistance. Physiol Rev 58(3):656–689.
U S A 107(51):22290–2295. Lange A, Gebremedhin D, Narayanan J, Harder D. 1997.
Filosa JA, Bonev AD, Straub SV, Meredith AL, Wilkerson MK, Aldrich 20-Hydroxyeicosatetraenoic acid-induced vasoconstriction and
RW, et al. 2006. Local potassium signaling couples neuronal activity inhibition of potassium current in cerebral vascular smooth mus-
to vasodilation in the brain. Nat Neurosci 9(11):1397–1403. cle is dependent on activation of protein kinase C. J Biol Chem
Fox PT, Raichle ME. 1986. Focal physiological uncoupling of cerebral 272(43):27345–27352.
blood flow and oxidative metabolism during somatosensory stimula- Leffler CW, Busija DW, Mirro R, Armstead WM, Beasley DG. 1989.
tion in human subjects. Proc Natl Acad Sci U S A 83(4):1140–1144. Effects of ischemia on brain blood flow and oxygen consumption of
Fraser DD, Duff y S, Angelides KJ, Perez-Velazquez JL, Kettenmann H, newborn pigs. Am J Physiol 257(6 Pt 2):H1917–1926.
MacVicar BA. 1995. GABAA/benzodiazepine receptors in acutely Lin AL, Fox PT, Hardies J, Duong TQ, Gao JH. 2010. Nonlinear
isolated hippocampal astrocytes. J Neurosci 15(4):2720–2732. coupling between cerebral blood flow, oxygen consumption, and
Frerichs KU, Feuerstein GZ. 1990. Laser-Doppler flowmetry. A review ATP production in human visual cortex. Proc Natl Acad Sci U S A
of its application for measuring cerebral and spinal cord blood flow. 107(18):8446–8451.
Mol Chem Neuropathol 12(1):55–70. Lindauer U, Leithner C, Kaasch H, Rohrer B, Foddis M, Fuchtemeier
Gebremedhin D, Ma YH, Falck JR, Roman RJ, VanRollins M, Harder M, et al. 2010. Neurovascular coupling in rat brain operates inde-
DR. 1992. Mechanism of action of cerebral epoxyeicosatrienoic pendent of hemoglobin deoxygenation. J Cereb Blood Flow Metab
acids on cerebral arterial smooth muscle. Am J Physiol 263(2 Pt 30(4):757–768.
2):H519–525. Lindauer U, Megow D, Matsuda H, Dirnagl U. 1999. Nitric oxide:
Girouard H, Iadecola C. 2006. Neurovascular coupling in the normal a modulator, but not a mediator, of neurovascular coupling in rat
brain and in hypertension, stroke, and Alzheimer disease. J Appl somatosensory cortex. Am J Physiol 277(2 Pt 2):H799–811.
Physiol 100(1):328–335. Liu X, Li C, Falck JR, Harder DR, Koehler RC. 2011a. Relative contri-
Gordon GR, Choi HB, Rungta RL, Ellis-Davies GC, MacVicar BA. bution of cyclooxygenases, epoxyeicosatrienoic acids, and pH to the
2008. Brain metabolism dictates the polarity of astrocyte control cerebral blood flow response to vibrissal stimulation. Am J Physiol
over arterioles. Nature 456(7223):745–749. Heart Circ Physiol 302(5):H1075–1085.
Gurden H, Uchida N, Mainen ZF. 2006. Sensory-evoked intrinsic opti- Liu X, Li C, Falck JR, Roman RJ, Harder DR, Koehler RC. 2008.
cal signals in the olfactory bulb are coupled to glutamate release and Interaction of nitric oxide, 20-HETE, and EETs during functional
uptake. Neuron 52(2):335–345. hyperemia in whisker barrel cortex. Am J Physiol Heart Circ Physiol
Guthrie PB, Knappenberger J, Segal M, Bennett MV, Charles AC, Kater 295(2):H619–631.
SB. 1999. ATP released from astrocytes mediates glial calcium waves. Liu X, Li C, Gebremedhin D, Hwang SH, Hammock BD, Falck JR,
J Neurosci 19(2):520–528. Roman RJ, et al. 2011b. Epoxyeicosatrienoic acid-dependent cerebral
Hauck EF, Apostel S, Hoffmann JF, Heimann A, Kempski O. 2004. vasodilation evoked by metabotropic glutamate receptor activation in
Capillary flow and diameter changes during reperfusion after global vivo. Am J Physiol Heart Circ Physiol 301(2):H373–381.
cerebral ischemia studied by intravital video microscopy. J Cereb Liu Y, Jia WG, Strosberg AD, Cynader M. 1992. Morphology and dis-
Blood Flow Metab 24(4):383–391. tribution of neurons and glial cells expressing beta-adrenergic recep-
Hu S, Kim HS. 1993. Activation of K+ channel in vascular smooth tors in developing kitten visual cortex. Brain Res Dev Brain Res
muscles by cytochrome P450 metabolites of arachidonic acid. Eur J 65(2):269–273.
Pharmacol 230(2):215–221. Lu J, Dai G, Egi Y, Huang S, Kwon SJ, Lo EH, et al. 2009. Characterization
Hu Y, Wilson GS. 1997. A temporary local energy pool coupled to neu- of cerebrovascular responses to hyperoxia and hypercapnia using MRI
ronal activity: fluctuations of extracellular lactate levels in rat brain in rat. Neuroimage 45(4):1126–1134.
monitored with rapid-response enzyme-based sensor. J Neurochem Ma J, Ayata C, Huang PL, Fishman MC, Moskowitz MA. 1996.
69(4):1484–1490. Regional cerebral blood flow response to vibrissal stimulation in mice
Ido Y, Chang K, Williamson JR. 2004. NADH augments blood flow in lacking type I NOS gene expression. Am J Physiol 270(3 Pt 2):
physiologically activated retina and visual cortex. Proc Natl Acad Sci H1085–1090.
U S A 101(2):653–658. Massa PT, Mugnaini E. 1982. Cell junctions and intramembrane par-
Jimenez AI, Castro E, Delicado EG, Miras-Portugal MT. 1998. ticles of astrocytes and oligodendrocytes: a freeze-fracture study.
Potentiation of adenosine 5’-triphosphate calcium responses by dia- Neuroscience 7(2):523–538.
denosine pentaphosphate in individual rat cerebellar astrocytes. Metea MR, Kofuji P, Newman EA. 2007. Neurovascular coupling is
Neurosci Lett 246(2):109–111. not mediated by potassium siphoning from glial cells. J Neurosci
Kacem K, Lacombe P, Seylaz J, Bonvento G. 1998. Structural organiza- 27(10):2468–2471.
tion of the perivascular astrocyte endfeet and their relationship with Metea MR, Newman EA. 2006. Glial cells dilate and constrict
the endothelial glucose transporter: a confocal microscopy study. blood vessels: a mechanism of neurovascular coupling. J Neurosci
Glia 23(1):1–10. 26(11):2862–2870.

A S T R O C Y T E R E GU L AT I O N O F N E U R O VA S C U L A R C O N T R O L • 479
Mintun MA, Vlassenko AG, Rundle MM, Raichle ME. 2004. Increased Schummers J, Yu H, Sur M. 2008. Tuned responses of astrocytes and
lactate/pyruvate ratio augments blood flow in physiologically acti- their influence on hemodynamic signals in the visual cortex. Science
vated human brain. Proc Natl Acad Sci U S A 101(2):659–664. 320(5883):1638–1643.
Muller T, Fritschy JM, Grosche J, Pratt GD, Mohler H, Kettenmann H. Shao Y, Sutin J. 1992. Expression of adrenergic receptors in individual
1994. Developmental regulation of voltage-gated K+ channel and astrocytes and motor neurons isolated from the adult rat brain. Glia
GABAA receptor expression in Bergmann glial cells. J Neurosci 14(5 6(2):108–117.
Pt 1):2503–2514. Shi Y, Liu X, Gebremedhin D, Falck JR, Harder DR, Koehler RC. 2008.
Mulligan SJ, MacVicar BA. 2004. Calcium transients in astrocyte end- Interaction of mechanisms involving epoxyeicosatrienoic acids,
feet cause cerebrovascular constrictions. Nature 431(7005):195–199. adenosine receptors, and metabotropic glutamate receptors in neu-
Murphy K, Gerzanich V, Zhou H, Ivanova S, Dong Y, Hoffman G, et al. rovascular coupling in rat whisker barrel cortex. J Cereb Blood Flow
2003. Adenosine-A2a receptor down-regulates cerebral smooth mus- Metab 28(1):111–125.
cle L-type Ca2+ channel activity via protein tyrosine phosphatase, not Simard M, Arcuino G, Takano T, Liu QS, Nedergaard M. 2003. Signaling
cAMP-dependent protein kinase. Mol Pharmacol 64(3):640–649. at the gliovascular interface. J Neurosci 23(27):9254–9262.
Nelson CW, Wei EP, Povlishock JT, Kontos HA, Moskowitz MA. 1992. Sun J, Druhan LJ, Zweier JL. 2008. Dose dependent effects of reactive
Oxygen radicals in cerebral ischemia. Am J Physiol 263(5 Pt 2): oxygen and nitrogen species on the function of neuronal nitric oxide
H1356–1362. synthase. Arch Biochem Biophys 471(2):126–133.
Newman EA. 1986. High potassium conductance in astrocyte endfeet. Takano T, Tian GF, Peng W, Lou N, Libionka W, Han X, et al. 2006.
Science 233(4762):453–454. Astrocyte-mediated control of cerebral blood flow. Nat Neurosci
Newman EA. 2003. Glial cell inhibition of neurons by release of ATP. 9(2):260–267.
J Neurosci 23(5):1659–1666. Takano T, Tian GF, Peng W, Lou N, Lovatt D, Hansen AJ, et al. 2007.
Niwa K, Araki E, Morham SG, Ross ME, Iadecola C. 2000. Cortical spreading depression causes and coincides with tissue hypoxia.
Cyclooxygenase-2 contributes to functional hyperemia in Nat Neurosci 10(6):754–762.
whisker-barrel cortex. J Neurosci 20(2):763–770. Theilen H, Schrock H, Kuschinsky W. 1994. Gross persistence of capillary
Niwa K, Haensel C, Ross ME, Iadecola C. 2001. Cyclooxygenase-1 par- plasma perfusion after middle cerebral artery occlusion in the rat brain.
ticipates in selected vasodilator responses of the cerebral circulation. J Cereb Blood Flow Metab 14(6):1055–1061.
Circ Res 88(6):600–608. van den Pol AN, Romano C, Ghosh P. 1995. Metabotropic glutamate
Offenhauser N, Thomsen K, Caesar K, Lauritzen M. 2005. receptor mGluR5 subcellular distribution and developmental expres-
Activity-induced tissue oxygenation changes in rat cerebellar cortex: sion in hypothalamus. J Comp Neurol 362(1):134–150.
interplay of postsynaptic activation and blood flow. J Physiol 565 Vanharreveld A, Kooiman M. 1965. Amino acid release from the cerebral
(Pt 1):279–294. cortex during spreading depression and asphyxiation. J Neurochem
Ogawa S, Lee TM, Kay AR, Tank DW. 1990. Brain magnetic resonance 12:431–439.
imaging with contrast dependent on blood oxygenation. Proc Natl
Ventura R, Harris KM. 1999. Three-dimensional relationships between
Acad Sci U S A 87(24):9868–9872.
hippocampal synapses and astrocytes. J Neurosci 19(16): 6897–6906.
Pasti L, Volterra A, Pozzan T, Carmignoto G. 1997. Intracellular cal-
Virchow RL. 1858. Cellular pathology as based upon physiological and
cium oscillations in astrocytes: a highly plastic, bidirectional form of
pathological histology. London: John Churchill.
communication between neurons and astrocytes in situ. J Neurosci
17(20):7817–7830. Vlassenko AG, Rundle MM, Raichle ME, Mintun MA. 2006. Regulation
Paulson OB, Newman EA. 1987. Does the release of potassium from astro- of blood flow in activated human brain by cytosolic NADH/NAD+
cyte endfeet regulate cerebral blood flow? Science 237(4817):896–898. ratio. Proc Natl Acad Sci U S A 103(6):1964–1969.
Peng X, Zhang C, Alkayed NJ, Harder DR, Koehler RC. 2004. Wahl M, Lauritzen M, Schilling L. 1987. Change of cerebrovascular
Dependency of cortical functional hyperemia to forepaw stimulation reactivity after cortical spreading depression in cats and rats. Brain
on epoxygenase and nitric oxide synthase activities in rats. J Cereb Res 411(1):72–80.
Blood Flow Metab 24(5):509–517. Wahl M, Schilling L, Parsons AA, Kaumann A. 1994. Involvement of
Peppiatt CM, Howarth C, Mobbs P, Attwell D. 2006. Bidirectional calcitonin gene-related peptide (CGRP) and nitric oxide (NO) in the
control of CNS capillary diameter by pericytes. Nature pial artery dilatation elicited by cortical spreading depression. Brain
443(7112):700–704. Res 637(1–2):204–210.
Petzold GC, Albeanu DF, Sato TF, Murthy VN. 2008. Coupling of neu- Wang X, Lou N, Xu Q, Tian GF, Peng WG, Han X, et al. 2006. Astrocytic
ral activity to blood flow in olfactory glomeruli is mediated by astro- Ca2+ signaling evoked by sensory stimulation in vivo. Nat Neurosci
cytic pathways. Neuron 58(6):897–910. 9(6):816–823.
Porter JT, McCarthy KD. 1995. GFAP-positive hippocampal astro- Williams DS, Detre JA, Leigh JS, Koretsky AP. 1992. Magnetic reso-
cytes in situ respond to glutamatergic neuroligands with increases in nance imaging of perfusion using spin inversion of arterial water.
[Ca2+]i. Glia 13(2):101–112. Proc Natl Acad Sci U S A 89(1):212–216.
Porter JT, McCarthy KD. 1997. Astrocytic neurotransmitter receptors Winship IR, Plaa N, Murphy TH. 2007. Rapid astrocyte calcium sig-
in situ and in vivo. Prog Neurobiol 51(4):439–455. nals correlate with neuronal activity and onset of the hemodynamic
Price DL, Ludwig JW, Mi H, Schwarz TL, Ellisman MH. 2002. response in vivo. J Neurosci 27(23):6268–6272.
Distribution of rSlo Ca2+-activated K+ channels in rat astrocyte Yemisci M, Gursoy-Ozdemir Y, Vural A, Can A, Topalkara K, Dalkara
perivascular endfeet. Brain Res 956(2):183–193. T. 2009. Pericyte contraction induced by oxidative-nitrative stress
Raichle ME, Hartman BK, Eichling JO, Sharpe LG. 1975. Central nora- impairs capillary reflow despite successful opening of an occluded
drenergic regulation of cerebral blood flow and vascular permeability. cerebral artery. Nat Med 15(9):1031–1037.
Proc Natl Acad Sci U S A 72(9):3726–3730. Zonta M, Angulo MC, Gobbo S, Rosengarten B, Hossmann KA,
Ramon y Cajal S. 1913. Contribucion al conocimento de la neuroglia del Pozzan T, et al. 2003a. Neuron-to-astrocyte signaling is central to
cerebro humano. Trab Lab Invest Biol Univ Madrid 11:255–315. the dynamic control of brain microcirculation. Nat Neurosci 6(1):
Robertson BE, Schubert R, Hescheler J, Nelson MT. 1993. 43–50.
cGMP-dependent protein kinase activates Ca-activated K channels Zonta M, Sebelin A, Gobbo S, Fellin T, Pozzan T, Carmignoto G.
in cerebral artery smooth muscle cells. Am J Physiol 265(1 Pt 1): 2003b. Glutamate-mediated cytosolic calcium oscillations regulate a
C299–303. pulsatile prostaglandin release from cultured rat astrocytes. J Physiol
Roman RJ. 2002. P-450 metabolites of arachidonic acid in the control of 553(Pt 2):407–414.
cardiovascular function. Physiol Rev 82(1):131–185. Zou MH, Leist M, Ullrich V. 1999. Selective nitration of prostacyclin
Roy CS, Sherrington CS. 1890. On the regulation of the blood-supply of synthase and defective vasorelaxation in atherosclerotic bovine coro-
the brain. J Physiol 11(1–2):85–158 17. nary arteries. Am J Pathol 154(5):1359–1365.

480 • FUNCTIONS OF NEUROGLIAL CELLS


38.
ASTROCY TES: MODULATION OF SYNAPTIC FUNCTION
AND NET WORK ACTIVIT Y
Andrea Volterra

A B B R E VI AT I O N S the previously unknown roles of these cells in the control of


synapse formation, function, and plasticity. The communi-
AMPA 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl) cation properties of astrocytes were discovered only recently
propanoic acid because astrocytes differ in their excitation mechanisms from
BAPTA 1,2-bis(o-aminophenoxy)ethane- neurons: They do not generate action potentials and have
N,N,N′,N′-tetraacetic acid “passive” membrane properties, which for many years led
CB Cannabinoid researchers to consider them nonexcitable cells, and there-
DHPG (S)-3,5-dihydroxyphenylglycine fore unable to communicate. However, in the 1990s, the
dnSNARE dominant negative SNAP (soluble NSF development of cellular imaging approaches changed this
attachment protein) receptor protein view. Application of Ca2+ imaging to the study of astrocytes
GABA gamma-aminobutyric acid revealed that these cells display intracellular Ca2+ ([Ca2+]i)
GFAP glial fibrillary acidic protein elevations in response to chemical signals coming from
GPCR G protein–coupled neighboring cells, including from synapses (Dani et al. 1992;
HFS high-frequency stimulation Pasti et al. 1997; Porter and McCarthy 1996). In turn, [Ca2+]i
LTD long-term depression rises were found to trigger release of transmitters from astro-
LTP long-term potentiation receptors cytes able to modulate neuronal functions (Bezzi et al. 1998;
IP3R2 inositol 1,4,5-trisphosphate receptor type 2 Kang et al. 1998; Parpura et al. 1994). Initial data suggested
mEPSC miniature excitatory postsynaptic current the existence of communication loops between synapses and
2MeSADP 2-methylthio-adenosine-5′-diphosphate astrocytes, by which astrocytes can feedback break/accel-
mGluR metabotropic glutamate receptor erator inputs to synapses. This gave rise to the concept of
MrgA1 mas-related gene receptor type A1 gliotransmission (Bezzi and Volterra 2001), subsequently
NMDA N-methyl-d-aspartate expanded to encompass feedforward astrocytic actions in
NPY neuropeptide Y which an astrocyte responds to synaptic activation by releas-
NR2B NMDA receptor subtype 2B ing a transmitter on a synaptic circuit distinct from the one
PP-GC perforant path-granule cell responsible for its activation. The next studies then clarified
SIC slow inward currents that astrocytes are able to produce multiple synaptic effects
SNAP25 synaptosomal-associated protein 25kDa and exert both inhibitory and stimulatory influences. Several
SOC slow outward current gliotransmitters were identified, together with multiple
TNFα tumor necrosis factor-alpha release pathways and a variety of synaptic targets, providing a
TNF-R1 tumor necrosis factor receptor type 1 first hint of the complexity of gliotransmission, governed by
TTX tetrodotoxin astrocytic Ca2+ encoding, whose properties still are not well
VAMP vesicle-associated membrane protein understood. Based on all the evidence, a provocative para-
v-SNARE vesicle SNAP (soluble NSF attachment digm shift in synaptic function was proposed at the end of the
protein) receptor protein 1990s, summarized as the tripartite synapse concept (Araque
et al. 1999; Kettenmann et al. 1996; Volterra et al. 2002). This
suggests that communication at the synapse is not limited to
1 INTRODUCTION presynaptic and postsynaptic compartments, but extends to
neighboring astrocytic processes endowed with receptors for
The last 20 years have brought about a potential conceptual synaptic mediators. It must be emphasized that the tripartite
revolution about the role of astrocytes in synaptic and net- synapse concept, although vividly illustrating the inclusion
work function. In the early 1990s, it was discovered that astro- of astrocytes in synaptic communication, does not define a
cytes possess active properties—notably a competence for specific anatomical unit, because the morphological relations
releasing transmitters, dubbed gliotransmitters as opposed to between astrocytic and synaptic elements differ significantly
neurotransmitters of neuronal origin. Recognizing that astro- in different brain circuits (Ventura and Harris 1999) and are
cytes communicate has been instrumental for understanding subject to dynamic changes (Oliet et al. 2001).

481
As a counterpart to the advances in the functional under- from “ectopic” presynaptic release sites of climbing fibers
standing of astrocytes, parallel new observations were made (Matsui and Jahr 2006). Moreover, astrocytes can be acti-
on their structural organization. Dye injection of neighboring vated by postsynaptic retrograde messengers such as endocan-
astrocytes revealed that these cells have an ordered arrange- nabinoids (Navarrete and Araque 2008, 2010). Synaptically
ment with minimal overlap (Bushong et al. 2002), which induced astrocytic [Ca2+]i elevations are thought to be evoked
allows each astrocyte to cover a specific territory comprising in most cases by binding of mediators to G protein–coupled
a few neuronal cell bodies, hundreds of dendrites, and several receptors (GPCR) linked to inositol 1,4,5-trisphosphate (IP3)
thousands of synapses (Halassa et al. 2007). Consequently, it signaling and Ca2+ release from the endoplasmic reticulum. In
was proposed that an individual astrocyte might oversee all the some circuits, activation of Ca2+-permeated ligand-gated recep-
activities undergoing in its territory, providing an extra layer tors, such as AMPAR, was also reported, with direct influx of
of intercellular coordination. However, an emerging concept Ca2+ from the extracellular medium. Moreover, other types of
in parallel is that astrocytes possess specialized microdomains Ca2+ channels or exchangers could operate to increase [Ca2+]i
in their perisynaptic processes able to control fractions of the in astrocytes in response to synaptic activity (see chapter 26).
astrocytic territory autonomously. [Ca2+]i elevation in astrocytes is not an “on-off ” type of
signal. On the contrary, there are multiple and varied patterns
of [Ca2+]i elevation, which probably underlie different types of
2 A S T R O C Y T E S A R E AC T I VAT E D function, including generation of diverse output signals to syn-
BY SY N A P T I C F U N C T I O N apses. The relevance of differences in amplitude, frequency, and
spatiotemporal properties of [Ca2+]i events generated in astro-
Astrocyte activation during synaptic transmission was first cytes by neuronal activity is under intense investigation. Initial
observed in brain slices. Tetrodotoxin (TTX)-sensitive astro- studies suggested a relation between neuronal input intensity
cytic [Ca2+]i elevations were reported to occur in several brain and astrocytic Ca2+ response (Matyash et al. 2001; Pasti et al.
areas, including the hippocampus, cerebral cortex, cerebellum, 1997). For example, in the cerebellum, low-frequency firing of
nucleus accumbens, olfactory bulb, and retina, in response to parallel fibers generated [Ca2+]i rises in Bergmann glial cells
release of a variety of transmitters and factors, such as gluta- restricted to small peripheral cell protrusions, possibly rep-
mate, adenosine triphosphate (ATP), gamma-aminobutyric resenting independent functional microdomains (Grosche
acid (GABA), acetylcholine, noradrenaline, nitric oxide, and et al. 1999). However, when parallel fibers discharged at high
endocannabinoids (Perea et al. 2009; Volterra and Meldolesi frequency, several such microdomains were activated and
2005). Most recently, studies were extended to living ani- coordinated, and the ensuing [Ca2+]i rise became generalized
mals and confirmed that astrocytes show neuronal activity– and propagated up to the cell body (Matyash et al. 2001).
dependent [Ca2+]i elevations triggered by physiological sen- The functional difference between the two types of Ca2+ sig-
sory stimuli (Petzold et al. 2008; Schummers et al. 2008; nal was not defined, but the fact that neuronal activity can
Wang et al. 2006) or locomotor behavior (Nimmerjahn et al. generate highly localized Ca2+ responses in glial processes
2009) (see section 11). Transfer of information from neurons suggests that astrocytes contain privileged zones for local
to astrocytes is thought to occur mostly by spillover of synap- communication with neurons (Grosche et al. 1999), allowing
tically released transmitters, particularly during intense activ- them to distinguish and integrate incoming activity gener-
ity. However, examples of direct synapse–glia communication ated by different axonal pathways (Perea and Araque 2005).
do exist, such as the one received by cerebellar Bergmann glia Recent work (Di Castro et al. 2011; Panatier et al. 2011)

WS
0.5
∆F/F

10 s
2MeSADP
0.3
∆G/R
2s

Figure 38.1 Ca2+ Signals in Astrocytic Processes and Cell Body Have Different Properties and May Underlie Distinct Biological Phenomena. (Blue)
Cell body Ca2+ signal, recorded in the somatosensory cortex in vivo following whisker stimulation (Wang et al. 2006). (Red) Ca2+ signal in an astro-
cytic process, recorded in hippocampal slices in response to local P2Y1R stimulation with 2MeSADP (Santello et al. 2011). Both signals were recorded
using Fluo-4 Ca2+ dye and are scaled in time and amplitude for comparison. Note that the Ca2+ signal in the process is spatially confined (∼4 μm),
occurs immediately on stimulation, is relatively fast (time-to-peak: hundreds milliseconds), short-lasting (∼3 seconds), and of small amplitude,
whereas the signal in the cell body is spatially larger, occurs with a few-second delay from stimulation, is slower (time-to-peak: ∼6 seconds),
longer-lasting (∼30 seconds), and of larger amplitude. Adapted from Wang et al. 2006. Santello et al. 2011.

482 • FUNCTIONS OF NEUROGLIAL CELLS


has strengthened this idea, by showing that individual random adult hippocampus, in which immunogold labeling indicates
synaptic events or single-action potentials induce highly con- that they contain glutamate (Bergersen et al. 2011) and express
fined, small, and fast local Ca2+ responses in astrocytic pro- vesicular glutamate transporters (VGLUTs) and proteins sus-
cesses, distinct from somatic [Ca2+]i rises (Fig. 38.1). These taining regulated exocytosis, such as the v-SNARE VAMP3/
local phenomena are capable of triggering gliotransmission cellubrevin (Bezzi et al. 2004). The presence of VGLUT
and modulating basal transmission at individual synapses. transcripts in adult hippocampal astrocytes is confirmed by
single-cell RT-PCR experiments, and a third, functional line of
evidence supporting SNARE-dependent astrocytic glutamate
3 A S T R O C Y T E S R E L E A S E N E U R OAC T I VE release in situ comes from experiments in which tetanus toxin
G L I OT R A N S M I T T E R S (TeNT), a protease that specifically inactivates VAMP2 and
VAMP3, was selectively infused in hippocampal astrocytes (see
Evidence exists for the release of several neuroactive gliotrans- section 4), leading to the abolishment of glutamate-dependent
mitters from astrocytes in situ, including glutamate, D-serine, synaptic modulation ( Jourdain et al. 2007; Perea and Araque
ATP (often rapidly metabolized to adenosine), and GABA, as 2007). The same effect was obtained with TeNT when study-
well as eicosanoids, including arachidonic acid, prostaglandins, ing astrocyte D-serine (Henneberger et al. 2010) and ATP/
epoxyeicosatrienoic acids (EETs), and the cytokine TNFα (see adenosine release (Panatier et al. 2011). At variance with
chapters 17 and 18). Concerning the mechanisms of release, this evidence, a transcriptome study of forebrain astrocytes
Ca2+-dependent exocytosis is the best characterized. There is from juvenile mice failed to find transcripts for VGLUTs
direct evidence for Ca2+-dependent secretion of glutamatergic and SNARE proteins such as VAMP2 and SNAP25 (Cahoy
vesicles in cultured astrocytes (Bezzi et al. 2004; Marchaland et al. 2008). However, VGLUT transcripts in astrocytes are
et al. 2008). Such organelles at the EM level appear small expected to be much less abundant than in synaptic terminals
(40- to 100-nm diameter), clear, and round, resembling synap- (and, therefore, elusive for transcriptomics), given that astro-
tic vesicles in nerve terminals; for this reason they were dubbed cytes lack the large “reserve pool” that accounts for approxi-
synaptic-like microvesicles (SLMV). There is also evidence for mately 90% of synaptic vesicles in neurons. Moreover, adult
secretion of D-serine from a similar vesicle population, as well hippocampal astrocytes express SNARE protein isoforms
as for release of ATP from lysosomes and peptides (NPY) distinct from canonical synaptic ones (Schubert et al. 2011).
from secretory granules (reviewed in Hamilton and Attwell Finally, astrocytic exocytosis seems to have peculiar prop-
2010). Synaptic-like microvesicles have been ultrastructur- erties distinguishing it from synaptic exocytosis, such as
ally identified also in perisynaptic astrocytic processes in the TNFα-dependent regulation (Santello et al. 2011). Of course,

Blocking gliotransmission

Ins(1,4,5)P3R
Inhibit TCA cycle No release BAPTA [Ca2+]i knockout
Fluoroacetate or fluorocitrate

Cleaves No Binds No
TeNT VAMP2 dn SNARE
and VAMP3 exocytosis SNARES exocytosis

Stimulating gliotransmission

+ ++
[Ca2+]i I +mV [Ca2+]i

Light

[Ca2+]i Ca2+ [Ca2+]i


P2Y1 MRGA1
2MeSADP RF amide

Figure 38.2 Experimental Strategies Used to Reveal the Role of Gliotransmission in Synaptic Function. (Upper) Methods and genetic models used
to selectively block gliotransmission, including metabolic poisoning of astrocytes, chelation of internal Ca2+, deletion of key astrocyte Ca2+ signal-
ing proteins (e.g., IP3R2), and pharmacological (TeNT) or transgenic (dnSNARE) blockade of exocytosis. (Lower) Methods and genetic models to
selectively stimulate [Ca2+]i elevation and gliotransmission, including mechanical or electrical astrocytic stimulation, activation of endogenous (P2Y1)
or transgenic (MrgA1) GPCR, and Ca2+ uncaging in astrocytes. Adapted from Hamilton and Attwell 2010.

A S T R O C Y T E S : M O D U L AT I O N O F SY N A P T I C F U N C T I O N A N D N ET WO R K AC T I VI T Y • 483
much more work is needed to clarify the many pending aspects In contrast, an astrocyte-specific pharmacological approach
concerning astrocytic gliotransmitter exocytosis (Hamilton consists in infusing nonpermeant agents in an individual
and Attwell 2010). astrocyte or a gap junction–connected network of astrocytes
Adenosine triphosphate, glutamate, GABA, and other via the patch-pipette used for whole-cell clamping the astro-
gliotransmitters are apparently released by astrocytes also cyte. This approach was successfully used to block astrocytic
via additional Ca2+-dependent and -independent processes, [Ca2+]i elevations via infusion of the Ca2+ chelator BAPTA
mediated by large-pore ion channels, such as gap-junction ( Jourdain et al. 2007; Serrano et al. 2006), the Ca2+ clamp
hemichannels, P2X7 receptor-channels, and transmitter car- solution (Henneberger et al. 2010), or the G protein inhibitor,
riers or exchangers (Volterra and Meldolesi 2005). In several GDP-beta-S, which blocks signal transduction downstream
cases operation of these release pathways in physiological con- of GPCR (Robitaille 1998). Infusion of the activated TeNT
ditions remains to be demonstrated. However, recent work light-chain, which blocks SNARE protein–dependent vesicle
has provided evidence that Bestrophin-1 anion channels in fusion involved in gliotransmitter release (see section 3), also
cerebellar glia are a physiological source of GABA release has been used successfully ( Jourdain et al. 2007; Perea and
underlying tonic inhibitory control of cerebellar granule cells Araque 2007). The internal dialysis approach is significantly
(Lee et al. 2010) (see section 10). more selective than use of gliotoxins because it affects only spe-
cific pathways, just for the limited time of the experiment and
only in the patched astrocyte or in those directly connected to
4 E X P E R I M E N TA L PA R A D I G M S TO it. However, only low MW agents (<1.2 kDa) can diffuse within
S T U DY G L I OT R A N S M I S S I O N minutes via gap junctions to a network of connected astrocytes
and block large communication domains. Selective enrich-
One of the main challenges associated with the emerging idea ment in astrocytes over other brain cells of key signaling mol-
that astrocytes play active roles at synapses concerns the meth- ecules such as IP3R2 (Holtzclaw et al. 2002), one of the three
odological approaches to study integrated synapse-astrocyte IP3 receptor isoforms, has led to the use of IP3R2ko mice in
functioning and the design of experiments appropriate for gliotransmission studies (Di Castro et al. 2011; Navarrete et al.
revealing specific astrocytic contributions within established 2012; Petravicz et al. 2008; Takata et al. 2011). These mice repre-
paradigms of synaptic function. Two complementary strate- sent a powerful tool, particularly for bringing analyses from the
gies have been used to date, based on either selectively acti- cellular/network level to the behavioral level. However, correct
vating or inactivating astrocyte signaling (Fig. 38.2). They are interpretation of the results requires accurate verification that
presented here, and their advantages and limitations are dis- IP3R2 is knocked-out only in astrocytes and that its permanent
cussed so as to provide readers with a critical appraisal of the deletion does not trigger compensatory mechanisms. A more
quality of the evidence presented by different studies. refined genetic approach consists in the conditional, cell-specific
inactivation of gliotransmission signaling steps. In principle, this
was achieved with the creation of the so-called dnSNARE mice,
4.1 S U BT R AC T I VE S T R AT EGY: A P P ROAC H E S in which the SNARE domain of SNARE proteins is condition-
TO S E L EC T I VE LY I NAC T I VAT E ally expressed under the astrocytic GFAP promoter (Pascual
G L I OT R A NS M I S S I O N et al. 2005). Expression of this domain has a dominant negative
These approaches aim at perturbing and abolishing signaling effect, trapping endogenous SNARE partners and preventing
steps involved in gliotransmission, while in parallel measuring formation of SNARE complexes necessary for vesicle fusion. An
the consequences on synaptic or network function. Multiple expected consequence is blockade of gliotransmitter exocytosis,
methods are used to achieve this goal. Some of them rely on although the selective occurrence of this event in dnSNARE
broadly perturbing astrocyte physiology, thereby secondarily mice has not yet been unequivocally demonstrated.
affecting gliotransmission pathways. This can be achieved
with toxins that are selectively taken up by astrocytes, such
4.2 A D D IT I VE S T R AT EGY: A P P ROAC H E S TO
as α-amino adipate (Khurgel et al. 1996), or with metabolic
S E L EC T I VE LY AC T I VAT E G L I OT R A NS M I S S I O N
poisons such as fluorocitrate and fluoroacetate, which target
metabolic enzymes selectively present in astrocytes (Swanson The opposite approach, selective stimulation of gliotransmis-
and Graham 1994). These are quite crude approaches that end sion, has also been used, verifying in parallel its impact on syn-
up by intoxicating the cells and fully compromising their func- aptic and network functions. One conceptual problem with
tion. However, more selective and preferable approaches exist. this approach is that it may overlap with ongoing endogenous
In some cases, one can use pharmacological blockers of recep- gliotransmission. Thus, subtractive approaches often show that
tors whose activation triggers gliotransmission, or of enzymes blockade of endogenous gliotransmission leads to synaptic
involved in the synthesis of specific gliotransmitters (e.g., changes. Therefore, by adding exogenous astrocyte stimulation,
D-serine), but only if parallel evidence exists that the target one may not simply amplify the endogenous mechanisms, but
receptors or enzymes are selectively expressed in astrocytes in also produce complex interactions with them, which compli-
the brain region investigated ( Jourdain et al. 2007; Panatier cates the interpretation of the results. The approaches initially
et al. 2006). It must be emphasized that extracellular applica- used privileged selectivity of astrocyte stimulation over repro-
tion of pharmacological agents in brain slices or in vivo does not duction of physiological stimuli. Quite harsh approaches such
provide per se evidence that they act specifically on astrocytes. as mechanical stimulation or strong electrical depolarization of

484 • FUNCTIONS OF NEUROGLIAL CELLS


patched astrocytes were shown to produce [Ca2+]i elevations [Ca2+]i elevations driving gliotransmission. In practice, MrgA1
that trigger gliotransmission (Araque et al. 1998; Kang et al. activation leads to massive astrocytic [Ca2+]i elevations last-
1998). Pharmacological stimulation of GPCR expressed in ing several minutes (Agulhon et al. 2010; Fiacco et al. 2007),
astrocytes was also used, although the cell selectivity of these which are far from mimicking the spatiotemporal patterns of
agents remains an issue. In some cases, use of a GPCR agonist [Ca2+]i elevations elicited by physiological stimuli (see Fig.
was coupled to a subtractive approach, showing that the agent 38.1) (see section 2). This may be caused by overexpression
lost its activity when a Ca2+ chelator was selectively introduced of MrgA1 receptors, possibly at sites different than those of
in astrocytes (Santello et al. 2011). A much more targeted endogenous GPCR. Another elegant approach recently used
approach consists in locally uncaging glutamate or other astro- to activate astrocytic signaling in vivo is optogenetics, via selec-
cyte receptor agonists to stimulate gliotransmission (Panatier tive viral transfection of channel rhodopsin (ChR) in astro-
et al. 2011; Poskanzer and Yuste 2011). Even if one can restrict cytes (Gourine et al. 2010). UV light–evoked ChR activation
the target area to approximately 200 nm by using two-photon is used in neurons to trigger physiological patterns of action
uncaging, this area is still too large to assure astrocyte speci- potentials on demand. In astrocytes, ChR activation was found
ficity. Thus, a tight contiguity (often only a few nm) exists to induce [Ca2+]i elevations, but the underlying mechanism
between perisynaptic astrocytic processes and synaptic ele- and its physiological relevance remain uncertain, given that
ments expressing glutamate receptors. This observation is also these cells normally lack Na+ channels and do not fire action
valid in the opposite direction, and in interpreting their results, potentials. In conclusion, critical analysis indicates that sev-
studies using glutamate uncaging to evaluate the role of presyn- eral clever approaches have been developed to study the role of
aptic or postsynaptic glutamate receptors should not exclude gliotransmission in synaptic and network function. However,
the involvement of astrocytic mechanisms. A more physiolog- no perfect approach exists. Therefore, with the present tools,
ical and cell-selective approach consists in introducing caged researchers should strengthen their conclusions by using more
Ca2+ (or IP3) in the astrocyte via the patch pipette (Fiacco than one approach in their studies.
and McCarthy 2004; Perea and Araque 2007). Although this
approach can generate [Ca2+]i elevations of physiological ampli-
tude, it hardly reproduces the spatiotemporal features of [Ca2+]i 5 C O N T R O L O F P R E SY N A P T I C
elevations induced by physiological stimuli, features that may TR ANSMITTER RELE ASE
be important in shaping the downstream gliotransmission (see
section 2). A genetic model was created to overcome this issue, Multiple observations indicate that gliotransmission can
in which a GPCR not endogenously present in brain, MrgA1, modulate presynaptic excitability by controlling the probabil-
is conditionally expressed only in astrocytes using the GFAP ity of synaptic transmitter release directly in nerve terminals
promoter, and activated on demand by applying its natural (Fiacco and McCarthy 2004; Jourdain et al. 2007; Navarrete
agonists, RF amides (Fiacco et al. 2007). Theoretically, this ele- and Araque 2010; Perea and Araque 2007). Surprisingly,
gant approach should mimic with high-fidelity the endogenous recent data indicate that astrocytic inputs can also modulate

5
1

4b

NR2B
[Ca 2+]i
3
NMDAR 2
AMPAR 1

GluT Glu
P2Y1R ATP
TNFR TNFα

Figure 38.3 Mechanism and Ultrastructural Correlate of Gliotransmission Inducing Presynaptic Modulation at Perforant Path–Granule Cell
Hippocampal Synapses. (Left) Adenosine triphosphate released from the synapse or astrocytes as a result of synaptic transmission (1) activates P2Y1R (2),
raising astrocyte [Ca2+]i (3), and triggering glutamate release from SLMV (4) facing NR2B-containing pre-NMDAR. N-methyl-d-aspartate receptor
activation increases synaptic transmitter release (5). Ambient TNFα is permissive on astrocyte glutamate release (4b) ( Jourdain et al. 2007; Santello
et al. 2011). (Right) Immunogold EM showing preferential NR2B expression (arrows) in the extrasynaptic region of an excitatory nerve terminal (ter)
facing an astrocytic process (ast) carrying SLMV (arrowheads, magnified in inset). Adapted from Jourdain et al. 2007.

A S T R O C Y T E S : M O D U L AT I O N O F SY N A P T I C F U N C T I O N A N D N ET WO R K AC T I VI T Y • 485
action potentials (Sasaki et al. 2011). Thus, astrocytes would antagonists, not by NMDAR antagonists as seen at PP-GC
send inputs to CA3 pyramidal cell axons (e.g., by releasing glu- synapses, suggesting the existence of presynaptic stimulatory
tamate acting on axonal AMPA/kainate receptors), thereby mGluR on CA3 afferents. However, no ultrastructural corre-
causing action potentials to broaden and resulting in enhanced late of the functional data has been provided to date.
synaptic transmitter release. A direct local modulatory action Studies at CA3-CA1 synapses also identified a puriner-
at the level of excitatory nerve terminals was described in the gic inhibitory presynaptic control by astrocytes, via adenos-
hippocampal dentate gyrus (Di Castro et al. 2011; Jourdain ine acting on A1 receptors. Thus, use of dnSNARE mice (see
et al. 2007; Santello et al. 2011). There, astrocytes sense syn- section 4), in which ATP/adenosine release from astrocytes
aptic activity at perforant path-granule cell (PP-GC) syn- is abolished, demonstrated that astrocyte adenosine is largely
apses, respond with local [Ca2+]i elevations in their processes, responsible for tonic suppression of transmission at these syn-
mediated by GPCR such as P2Y1 purinergic receptors, and apses (Pascual et al. 2005). Intriguingly, a recent study, in iden-
increase the probability of transmitter release at individual GC tifying local synapse–astrocyte communication at individual
synapses. This effect depends on astrocyte glutamate release CA3-CA1 synapses, found that synaptic glutamate stimulated
activating ifenprodil-sensitive NMDA receptors present on Ca2+-dependent and TeNT-sensitive ATP/adenosine release.
the excitatory terminals (pre-NMDAR). Pre-NMDAR acti- This astrocyte adenosine, however, was found to activate A2,
vation increases synaptic release probability via an unknown not A1 receptors, and increase the probability of synaptic glu-
mechanism, as indicated by the observed effects: An increase tamate release (Panatier et al. 2011). The conditions leading
in mEPSC frequency (without parallel change in amplitude TeNT-sensitive ATP/adenosine release from astrocytes to
and kinetics of the events), and an increase in the amplitude privilege activation of A2 versus A1 receptors (or vice versa)
of evoked EPSCs accompanied by changed paired-pulse ratio. remain to be understood.
This neuromodulatory action of astrocytes is the only one for Astrocyte-dependent presynaptic modulation is not
which a precise ultrastructural correlate has been established restricted to excitatory circuits. Evidence for such a con-
(Fig. 38.3). Thus, Jourdain et al. found that excitatory nerve ter- trol exists also for inhibitory synapses made by GABAergic
minals in the dentate molecular layer express NR2B subunits, interneurons onto CA1 pyramidal cells. The astrocytic
which are particularly abundant in the extrasynaptic terminal mechanism is responsible for physiological potentiation of
membrane apposed to astrocytic processes, where SLMVs can the inhibitory input to pyramidal cells observed in response
be observed within tens of microns from the NR2B subunits. to repetitive firing of the interneurons (Kang et al. 1998).
Intriguingly, the presence of ambient levels of TNFα was Interneuron firing activates GABAB receptors on astrocytes,
found to be a critical factor controlling pre-NMDAR activa- inducing elevation of astrocytic [Ca2+]i and glutamate release.
tion by astrocyte glutamate (Santello et al. 2011). It is likely that astrocytic glutamate induces feedback potenti-
A similar stimulatory action on synaptic transmitter ation by targeting excitatory GluR5-containing kainate recep-
release by astrocyte glutamate was reported at CA3 Schaffer tors present on interneurons (Liu et al. 2004).
collateral–CA1 pyramidal cell (CA3-CA1) synapses. The glu-
tamate release was found to be Ca2+ dependent and blocked
by infusion of TeNT in the astrocytes (Perea and Araque 6 C O N T R O L O F P O S T SY N A P T I C
2007), such as the release at PP-GC synapses. However, the E XC I TA B I L I T Y
astrocytic control at CA1 synapses was initially observed only
in artificial conditions, that is, on uncaging IP3 (Fiacco and Another proposed consequence of gliotransmission is stim-
McCarthy 2004) or Ca2+ (Perea and Araque 2007) in stratum ulation of postsynaptic neuronal excitability (Araque et al.
radiatum astrocytes. Actually, when the McCarthy group sub- 1998). In hippocampal slices, peculiar transient inward
sequently used MrgA1 and IP3R2ko mice (see section 4) to currents, often of very large amplitude (up to 1 nA) and
stimulate and block gliotransmission, respectively, they failed much slower than synaptic currents, were observed in CA1
to reproduce their own initial observations, and argued that pyramidal neurons (Angulo et al. 2004; Fellin et al. 2004)
the neuromodulatory gliotransmission previously observed (Fig. 38.4). Such slow inward currents (SICs) occur at low
was a methodological artifact resulting from the unphysiolog- frequency, particularly in Mg2+-containing medium, are
ical stimuli used (Fiacco et al. 2007). However, more recent TTX-insensitive, sustained by NMDAR activation, and
data by the Araque group demonstrate that this is not the case, have been attributed to glutamate released from astrocytes in
showing that astrocyte glutamate is released following physi- response to evoked or intrinsic [Ca2+]i elevations. Like its pre-
ological stimuli that activate endogenous astrocyte GPCR. synaptic effects, astrocyte-released glutamate directly activates
Thus, this occurs when astrocyte CB1 receptors are stimulated ifenprodil-sensitive NMDAR, this time located extrasynapti-
by endocannabinoids released as result of CA1 pyramidal cell cally on dendrites. Consequently, the large SICs evoked can
activity (Navarrete and Araque 2010) or when astrocytic mus- significantly depolarize the postsynaptic cell and even trigger
carinic acetylcholine receptors are stimulated by activity of its firing. Importantly, astrocyte-evoked SICs have been found
cholinergic afferents coming from the septum. Moreover, in to occur in strict temporal correlation in two or more neigh-
the latter case, the synaptic consequences of astrocyte gluta- boring CA1 pyramidal cells, which could lead the cells to fire
mate release are abolished in IP3R2ko mice (Navarrete et al. synchronously. Generation of synchronous SICs has been
2012). The increase in EPSC frequency induced by astrocyte proposed to depend on a single episode of glutamate release
glutamate at CA3-CA1 synapses is blocked by group I mGluR in an individual astrocyte, sensed simultaneously by different

486 • FUNCTIONS OF NEUROGLIAL CELLS


Figure 38.4 Astrocytic Glutamate Release Triggers Synchronous Slow Inward Currents in Neighboring Neurons, Coordinately Increasing Their
Excitability. (Left) Proposed mechanism for synchronous slow inward current (SIC) generation. Glutamate released from an astrocyte in a single
event is sensed by receptors on dendrites belonging to two CA1 pyramidal neurons sitting within the astrocytic release territory. (Right) Recordings
from two neighboring CA1 pyramidal cells show occurrence of synchronous SICs (enlarged in insets). Slow inward currents occur both spontaneously
and on mGluR agonist (DHPG) stimulation. Scale: 200 pA, 50 s; insets: 200 pA, 400 ms. Adapted from (left) Volterra and Meldolesi 2005. (right)
Fellin et al. 2004.

pyramidal cells sitting in its territory. Thereby, astrocytes cause de novo formation of large-diameter (>1-μm) organelles
would contribute to synchronization of neuronal activity. in astrocytes, compatible with ensuing release of large gluta-
Astrocyte-evoked SICs have been observed in several brain mate amounts that induce large-amplitude SICs, as observed
areas (D’Ascenzo et al. 2007; Parri et al. 2001), but it is unclear by most authors. However, the Araque group reported SICs of
whether they represent a relevant phenomenon at all or only at much smaller amplitude (<20 pA) (Perea and Araque 2005),
some synapses. For instance, SICs were not observed at PP-GC compatible with glutamate release from SLMV, raising the
hippocampal synapses ( Jourdain et al. 2007). This could be question of whether “small” and “large” SICs represent the
explained by differences existing between the excitatory cir- same phenomenon mechanistically.
cuits in the CA1 region and dentate gyrus. For instance, at Another phenomenon similar to SIC generation, but with
PP-GC synapses, pre-NMDAR activation seems to prevail opposite functional consequences, has been described in mitral
over extrasynaptic NMDAR activation (Dalby and Mody cells of the olfactory bulb, where low-frequency, slow, and large
2003; Jourdain et al. 2007). Expression of NR2B subunits is outward currents (SOCs) occur spontaneously in response to
fourfold higher in presynaptic than dendritic membrane–fac- mechanical stimulation of astrocytes. Such currents have been
ing astrocytes ( Jourdain et al. 2007). Several additional aspects attributed to release of GABA from the astrocytes and shown
about SICs remain to be clarified; for example, the conditions to occur synchronously in adjacent mitral cells, leading to
under which they occur. Slow inward currents were found to blockade of their firing activity (Kozlov et al. 2006).
be generated on activation of GPCR that induce astrocytic Another form of postsynaptic inhibition by astrocytes has
[Ca2+]i elevation such as mGluR5 (D’Ascenzo et al. 2007), been reported in the retina, where Müller cells release ATP, then
CB1 (Navarrete and Araque 2008), or PAR1 (Shigetomi transformed to adenosine, which causes postsynaptic hyperpo-
et al. 2008). However, other GPCR that produce astrocytic larization of ganglion cells via A1 receptors, an effect that reduces
[Ca2+]i elevations, such as P2Y1R, were shown not to gener- their action potential firing frequency (Newman 2003).
ate SICs ( Jourdain et al. 2007; Shigetomi et al. 2008), sug- Astrocyte-released transmitters seem to act mainly at extra-
gesting that GPCR-induced astrocytic [Ca2+]i elevations need synaptic receptors on dendrites. The case of astrocyte-released
to have specific characteristics to produce SICs. On the other D-serine is different, because most of the effects reported to
hand, some authors reported that they could produce SICs date for this gliotransmitter appear to be caused by a direct
in conditions not involving GPCR-evoked [Ca2+]i elevation, action at synaptic NMDAR (Henneberger et al. 2010;
such as hypoosmotic conditions (Fiacco et al. 2007), or could Panatier et al. 2006; Stevens et al. 2003; Yang et al. 2003). In
trigger spontaneously occurring SICs in CA1 pyramidal cells several brain circuits, D-serine, not glycine, was shown to be
on repeatedly applying high concentrations (50 mM) of exog- the endogenous ligand of the coagonist binding sites of synap-
enous glutamate (Xu et al. 2007). The latter were found to tic NMDAR. Such coagonist binding sites were found to be

A S T R O C Y T E S : M O D U L AT I O N O F SY N A P T I C F U N C T I O N A N D N ET WO R K AC T I VI T Y • 487
not tonically saturated, allowing phasic D-serine release from postsynaptic depolarization (Perea and Araque 2007). This
astrocytes to exert important physiological controls on synap- implies that coincident astrocytic and synaptic activity, like
tic transmission and plasticity (see section 8). The reason why coincident presynaptic and postsynaptic activity, can induce
D-serine activates mainly synaptic instead of extrasynaptic long-term synaptic plasticity. Recently, astrocyte-dependent
NMDAR is presently unclear. Possibly, the coagonist binding LTP of CA3-CA1 synapses was shown to occur physiologi-
sites on extrasynaptic NMDAR are tonically saturated, and cally in situ and in vivo when cholinergic afferents from the
diffusion of D-serine from its astrocytic release sites to the septum coincidentally stimulate stratum radiatum astrocytes
cleft is not restricted by potent uptake systems, as is probably and CA1 pyramidal cells (Navarrete et al. 2012). A similar
the case for glutamate diffusion. finding was made in the barrel cortex in vivo, where combined
stimulation of mouse whiskers and cholinergic afferents from
the nucleus basalis of Meynert produced NMDAR-mediated
7 H ET E R O SY N A P T I C R E GU L AT I O N LTP, requiring muscarinic receptor–evoked [Ca2+]i eleva-
tion and D-serine release from astrocytes (Fig. 38.5) (Takata
Astrocyte signaling can functionally interconnect synaptic cir- et al. 2011). An involvement of astrocyte D-serine in the
cuits. This feedforward action was first described as part of the modulation of NMDAR-dependent LTP was initially pro-
phenomenon known as heterosynaptic depression, a form of posed via pharmacological studies at CA3-CA1 synapses
intersynaptic communication in which active synapses decrease (Yang et al. 2003). More recent work (Henneberger et al.
the efficacy of neighboring, inactive synapses. At CA3-CA1 2010) confirmed that astrocytes respond to the CA3-CA1
synapses high-frequency activity of a Schaffer collateral fiber LTP induction protocol with transient [Ca2+]i elevation
can trigger heterosynaptic depression of another, adjacent and D-serine release, which boosts NMDAR activation.
fiber. This process is probably not caused by direct intersyn- The D-serine–dependent component of NMDAR activa-
aptic cross-talk via neurotransmitter spillover, as originally tion is apparently necessary for LTP induction because its
thought, but by the feedforward modulation of an intermedi- selective blockade is sufficient to prevent LTP. Interestingly,
ate astrocyte that senses the level of activity in the first fiber and astrocyte-released D-serine contributes to LTP within a
consequently turns down the activity of a second one by releas- defined spatial range. Thus, it appears that any individual
ing inhibitory adenosine (Zhang et al. 2003). An intermedi- astrocyte provides D-serine for inducing LTP at all the stim-
ary GABAergic interneuron, stimulated by glutamate released ulated synapses lying within or near its anatomical volume,
from the first fiber, is apparently responsible for astrocyte acti- whereas it is unable to influence synapses that are greater than
vation and enhanced astrocytic [Ca2+]i via GABAB receptors or equal to 200 μm away. Availability of astrocytic D-serine
(Serrano et al. 2006). Heterosynaptic depression is abolished crucially also controls NMDAR-dependent synaptic plastic-
in dnSNARE mice lacking astrocyte ATP/adenosine release, ity in other circuits, notably in the supraoptic hypothalamic
confirming that this form of synaptic adaptation requires nucleus (SON) (Panatier et al. 2006). There, astrocytes
astrocytic signaling (Pascual et al. 2005). Gliotransmission retract from synapses on SON neurons during lactation,
also was recently found to mediate the opposite phenomenon, thereby reducing D-serine availability for NMDAR activa-
heterosynaptic facilitation. At CA3-CA1 synapses, release of tion. Consequently, stimulations that induce LTP in virgin
endocannabinoids from CA1 pyramidal neurons was shown animals instead induce LTD in lactating rats. This metaplas-
to have a dual action on proximal and more distant synapses. ticity is likely explained by the fact that reduced coagonist
Although proximal synapses are homosynaptically depressed site occupancy by D-serine reduces NMDAR activation to
via the classical direct action of cannabinoids on presynaptic levels sufficient to induce LTD but not LTP (see chapter 41).
CB1 receptors, more distant ones are heterosynaptically poten- Another astrocyte-released mediator, lactate, was recently
tiated via activation of astrocytic CB1 receptors, followed by found to be critical for the maintenance of hippocampal LTP
[Ca2+]i elevation in the astrocyte and glutamate release at dis- and the formation of long-term memory in vivo (Suzuki et al.
tal sites, resulting in the activation of presynaptic stimulatory 2011). However, it is unclear if lactate sustains memory for-
group I mGluRs (Navarrete and Araque 2010). mation metabolically or by stimulating relevant neuronal sig-
naling. On the other hand, very recent work has implicated
astrocyte CB1 signaling in an opposite in vivo effect, that is,
8 ASTROCY TES IN induction of LTD reducing spatial working memory (Han
AC T I VI T Y-D E P E N D E N T SY N A P T I C et al. 2012). This effect was mediated by glutamate release
PL ASTICIT Y from astrocytes. Consistently, CB1-dependent astrocyte
glutamate exocytosis was recently found to underlie spike
Astrocyte signaling participates to the establishment of timing–dependent depression (t-LTD) in cortical slices via
Hebbian forms of synaptic plasticity such as long-term poten- activation of pre-NMDAR (Min and Nevian 2012).
tiation (LTP) and long-term depression (LTD), although this Despite the preceding evidence supporting the involve-
has been disputed by some studies (discussed at the end of ment of gliotransmission in activity-dependent synaptic plas-
this section). An initial indication came from the observation ticity, the work of the McCarthy group provided negative
that Ca2+ uncaging in a stratum radiatum astrocyte, which per results (Agulhon et al. 2010). This was particularly surprising
se produces short-lasting synaptic facilitation at CA3-CA1 because these authors obtained results opposite to those of
synapses, produced long-lasting potentiation when paired to Henneberger et al. (2010), although the two groups studied

488 • FUNCTIONS OF NEUROGLIAL CELLS


Air Puff+NBM
160

140

Relative LFP slope (%)


Air Puff
120

NBM 100

80

0
–20 0 20 40 60
Time (min)

Air Puff+NBM Air Puff+NBM+D-Ser


160 160
IP3R2-KO IP3R2-KO
140 140
Relative LFP slope (%)

Relative LFP slope (%)


120 120

100 100

80 80

0 0
–20 0 20 40 60 –20 0 20 40 60
Time (min) Time (min)

NBM axon

Astrocyte mAchR Ach

NMDAR Glu
[Ca2+]i
D-Ser

[Ca2+]i LTP

Presynapse Postsynapse

Figure 38.5 Cholinergic Long-Term Potentiation in the Somatosensory Cortex In Vivo Requires Gliotransmission. (Upper Panels, Left) Experimental
setup for paired air puff whisker stimulation and electrical stimulation of nucleus basalis of Meynert (NBM). (Right) Paired stimulation induces
long-term potentiation of the local field potentials (LFP). (Middle Panels, Left) Long-term potentiation is abolished in IP3R2ko mice lacking Ca2+
signaling in astrocytes. (Right) Long-term potentiation is rescued by D-serine infusion in the same mice. (Lower Panel) Proposed mechanism for the
astrocytic involvement. Acethylcholine released from NBM axonal terminals activates muscarinic receptors in astrocytes, increasing [Ca2+]i and induc-
ing D-serine release necessary for LTP induction at the cortical excitatory synapses. Adapted from Takata et al. 2011.

LTP in the same circuit (CA3-CA1 synapses) and used the but in this case too HFS-evoked LTP was unaffected, which
same induction protocol (classical high-frequency stimulation led the authors to conclude that astrocyte Ca2+ signaling and
[HFS]). However, the experimental strategies were totally dif- gliotransmission do not modulate LTP. More recently, how-
ferent. In the Henneberger et al. study, HFS of afferent fibers ever, other authors observed an astrocytic modulation of LTP
was delivered after having dyalized astrocytes with Ca2+ chela- in MrgA1 mice using the same protocol as Agulhon et al.
tors or exocytosis blockers to abolish endogenous signaling. (2010). The only difference was that they induced LTP by
In this condition, HFS failed to induce LTP because astro- using theta-burst stimulation, a milder stimulation than HFS
cytes were unable to release D-serine. In the Agulhon et al. (Lee et al. 2010). In complement, two recent studies reported
(2010) study, the authors used MrgA1 mice (see section 4), disruption of cholinergic LTP in hippocampus and cerebral
and via stimulation of MrgA1 receptors, induced large astro- cortex in vivo in IP3R2ko mice (Navarrete et al. 2012; Takata
cytic [Ca2+]i elevation just before HFS. However, the resulting et al. 2011). Overall, these data suggest that gliotransmission
LTP was identical to that produced by HFS alone. They also participates in synaptic plasticity in defined conditions that
tested HFS in IP3R2ko mice lacking GPCR-mediated Ca2+ depend on specific types of activity and/or particular circuits
signaling in astrocytes (Petravicz et al. 2008) (see section 4), and are not reproduced by any type of stimulus in any circuit.

A S T R O C Y T E S : M O D U L AT I O N O F SY N A P T I C F U N C T I O N A N D N ET WO R K AC T I VI T Y • 489
9 A S T R O C Y T E S I N H O M E O S TAT I C for tonic glutamate release is via astrocytic cystine–glutamate
SY N A P T I C P L A S T I C I T Y exchange (Baker et al. 2002), although the available evidence
relies on use of rather unselective pharmacological inhibi-
Astrocyte-released factors such as TNFα in the hippocampus tors. Recent work has provided more compelling evidence for
(Beattie et al. 2002; Stellwagen and Malenka 2006; Stellwagen tonic GABA release from cerebellar Bergmann glia and astro-
et al. 2005) and ATP in the hypothalamus (Gordon et al. 2005, cytes via bestrophin-1 channels (Lee et al. 2010). Such chan-
2009) are involved in synaptic scaling, a form of homeostatic nels have the intriguing property of being both leakage anion
plasticity independent of coincident changes in presynap- channels, therefore tonically active, and channels regulated by
tic and postsynaptic activity. Thus, unlike LTP or LTD, syn- [Ca2+]i or cell swelling. However, it is unclear when and how
aptic scaling affects all the synapses in a neuron and consists the two functional modes take place. Bestrophin-1–dependent
in a proportional adaptation of their strength to changes in tonic (Ca2+ independent) GABA release from glia is the main
network activity with the goal of stabilizing the neuron’s fir- source of GABA responsible for tonic inhibition of cerebellar
ing. Postsynaptic scaling is classically observed in response granule cells.
to persistent blockade (e.g., with TTX) of network activ-
ity in vitro, or to sensory deprivation in vivo, although other
adaptive mechanisms probably exist that operate over differ- 11 OT H E R F O R M S O F A S T R O C Y T E
ent temporal and spatial scales (Pozo and Goda 2010). Both SY N A P T I C C O N T R O L : G LU TA M AT E
TNFα, acting via TNF-R1, and ATP, acting via P2X7, induce U P TA K E A N D M O R P H O L O G I C A L
synaptic potentiation (scaling up) by promoting trafficking of PL ASTICIT Y
postsynaptic AMPAR subunits and increasing their insertion
at the postsynaptic membrane. Tumor necrosis factor alpha Forms of astrocyte–synapse interaction other than direct
also decreases postsynaptic GABAA receptors. According to release of neuroactive gliotransmitters have important modu-
the mechanistic model initially proposed, astrocytes sense latory influences on synaptic functions. A well-recognized
the reduction in presynaptic activity and neuronal glutamate mode is via the activity of glutamate transporters expressed on
release and react by increasing TNFα release. Tumor necrosis the plasma membrane of perisynaptic astrocytic processes. By
factor alpha then acts on neurons to scale excitatory activity binding and taking up synaptic glutamate, such transporters
up and inhibitory activity down via its effects on receptor traf- dynamically control the extracellular level of the transmitter
ficking. Recent data suggest, however, that TNFα is not the and therefore determine its capacity to activate or desensitize
active signal mediating synaptic scaling; rather it is a permis- synaptic and extrasynaptic glutamate receptors, as well as to
sive factor for expression or maintenance of the phenomenon spillover to neighboring synapses and glial cells (Bergles and
(Steinmetz and Turrigiano 2010). Initially described in cell Jahr 1998; Kullmann and Asztely 1998). Because the extent
cultures, TNFα-dependent scaling may take place also in vivo, of astrocytic coverage differs from synapse to synapse and not
as a component of the visual cortex plasticity that follows mon- all synapses, even in the same area, are directly contacted by
ocular deprivation in the critical period (Kaneko et al. 2008). astrocytic processes (Ventura and Harris 1999), this mode of
Synaptic scaling induced in the hypothalamus by astrocyte ATP control is synapse specific. Thus, its functional consequences
is different from TNFα-dependent scaling: It is triggered by depend on the reciprocal distribution of glutamate transport-
increased (not decreased) presynaptic activity, develops rapidly ers and receptors that, in turn, depends on the morphological
(minutes), and is mediated by glutamate activating mGluRs on arrangement of the astrocyte around the synapse (Rusakov
a local astrocyte. This triggers a [Ca2+]i elevation in the astro- and Lehre 2002). It is noteworthy that such an arrangement
cyte, spreading to its processes and resulting in a release of ATP is not static but plastic, and specific physiological conditions
that invests the dendritic arbor of a neighboring magnocellular induce synaptic modulation via a dynamic change of the astro-
neuron and induces strengthening of all its synapses. cytic coverage of synapses. For instance, in the hypothalamic
SON, lactation triggers retraction of astrocytic processes sur-
rounding magnocellular neurons, and causes repositioning of
10 TO N I C SY N A P T I C C O N T R O L S glutamate transporters far away from the synapses (see section
BY A S T R O C Y T E S 8). Consequently, synaptically released glutamate can diffuse
to presynaptic metabotropic receptors at the same excitatory
Tonic activation of high-affinity glutamate (NMDAR) and or neighboring inhibitory synapses, inducing homosynaptic or
GABA (α6 and δ subunit-containing GABAA) receptors has heterosynaptic depression, a phenomenon not seen in nonlac-
been described at cerebellar and hippocampal synapses and tating animals (Oliet et al. 2001; Piet et al. 2004) (see chapter
appears to play an important developmental role. Moreover, 41). Increased sensory stimulation has an opposite effect in the
there is a clear role for tonic activation of GABA receptors in barrel cortex, enhancing coverage of synapses by astrocytic pro-
the adult nervous system, where they contribute to informa- cesses and increasing expression of glutamate transporters which
tion processing in the cerebellum and probably in the hip- most likely increase clearance and reduce spillover of synaptic
pocampus. The importance of tonic activation of glutamate glutamate out of the cleft (Genoud et al. 2006). Morphological
receptors in the adult is less certain (Cavelier et al. 2005). plasticity of astrocytic processes also influences the positioning
There are indications that astrocytes are the source of tonic of gliotransmitter release sites, as already discussed for D-serine
glutamate and GABA release. A main mechanism proposed release in the hypothalamus (see section 8).

490 • FUNCTIONS OF NEUROGLIAL CELLS


12 C O O R D I N AT E AC T I VI T Y O F 13 S U M M A RY A N D P E R S P E C T I VE S
ASTROCY TIC AND NEURONAL
N ET WO R K S Twenty years of research outline a role for astrocytes in the
control of synaptic circuits, providing a new conceptual frame
Increasing evidence suggests that astrocytic signaling plays for understanding brain function. A variety of neuromodula-
modulatory roles not only at the level of individual syn- tory effects of astrocytes have been reported, forming a com-
apse, but also of macroscopic networks. In vivo studies plex and sometimes contradictory preliminary picture. A
show the occurrence of coordinated [Ca2+]i elevations in number of experimental paradigms and genetic models have
vast astrocytic populations. This is particularly evident in been developed to study astrocyte–synapse interactions and
unanesthetized, behaving animals because anesthesia dra- define their relevance. Given the incomplete understanding
matically suppresses astrocytic Ca2+ signaling (Schummers of the underlying biology, such methods and models are not
et al. 2008). During locomotor behavior [Ca2+]i elevations fully validated and the resulting data need to be interpreted
involving hundreds of Bergmann glial cells are seen in the with some caution. For instance, if it is well established that
cerebellum (Nimmerjahn et al. 2009). Such [Ca2+]i eleva- astrocytes respond to physiological neuronal activity with
tions, never observed in resting animals, strictly depend [Ca2+]i elevations triggering astrocytic responses, the full
on neuronal excitatory activity. Their function is unclear: range of synaptically evoked astrocytic Ca2+ signals and their
They arise at the same time when blood flow increases in the individual roles remain at present undefined, although it is
same region, but the overall time courses of the two events likely that different types of signal have distinct biological
do not match. Moreover, glial [Ca2+]i elevations are sup- function. Recent work suggests that fast local signals confined
pressed by anesthetics, whereas the blood response is not or to astrocytic processes are relevant for neuromodulation, but
to a lower extent. Population [Ca2+]i elevations are observed most astrocyte studies, notably in vivo studies, mainly record
also in visual cortex astrocytes in response to visual stimuli the larger and slower cell body signals, whose functional role
(Schummers et al. 2008). Importantly, Ca2+ responses in and neuromodulatory relevance are unclear. The other side
astrocytes occur according to specific functional maps over- of the same issue is that distinct paradigms used to exper-
lapping with neuronal ones and share many receptive-field imentally increase [Ca2+]i in astrocytes most likely trigger
characteristics with neuronal responses. Functional organ- biologically nonequivalent responses. Moreover, the effect
ization of astrocytic networks according to maps reflecting produced by increasing astrocytic [Ca2+]i can be different at
the neuronal maps is confirmed by dye-coupling experi- different brain locations depending on the synaptic circuit
ments showing that astrocytes within an olfactory glomer- to which the astrocyte is apposed, and on local differences in
ulus are gap junction–coupled among them, but not with the structural and molecular determinants of astrocyte–syn-
astrocytes present in neighboring glomeruli (Roux et al. apse communication. Moreover, even at the same brain site,
2011). The appearance of coordinated waves of [Ca2+]i ele- such determinants can change depending on the animal’s
vation in hundreds of astrocytes is another interesting phe- age; therefore, data obtained in developing animals do not
nomenon recently described in the hippocampus in vivo necessarily match those in adults. Studies to date have not
(Kuga et al. 2011). These responses, called glissandi, depend carefully considered all these emerging complexities, which
on neuronal activity and have been proposed to specifically are probably the cause of discrepant results. Future studies
emerge in response to network state changes. In this con- need to take all of these points into full account, and address
text, a recent study provided the first evidence that astrocyte critical pending issues, such as the specific mechanisms by
Ca2+ signaling actively controls neuronal networks, notably which gliotransmission modulates neuronal functions and
the number of their UP states (Poskanzer and Yuste 2011). the time-course of astrocytic actions. Future studies need to
UP states consist of membrane potential depolarizations also bring more clarity to the existence of different modes
occurring synchronously in an entire neuronal network, of gliotransmission that sustain distinct astrocytic actions,
and are thought to underlie behaviorally relevant cortical for example, some modes that are activity dependent and in
states. According to Poskanzer and Yuste, when Ca2+ activ- register with synaptic transmission, and others that are tem-
ity is triggered in the astrocytic network, the frequency of porally uncorrelated, and set the background of neuronal
neuronal UP states increases, and when astrocytic Ca2+ sig- circuit function or its long-term modulation. Indeed, there
naling is blocked, neuronal UP state frequency decreases. is need of a better understanding of the many and appar-
Although these fascinating observations in cortical slices ently contradictory modes of astrocytic action reported to
await confirmation in the living animal, other in vivo stud- date. How do astrocytes control individual synapses inde-
ies already have shown that blocking Ca2+ signaling in astro- pendently, and all the synapses in their territory collectively,
cytes affects complex network activity–sustaining behavior. and all the synapses in a neuron independent of astrocytic
This is the case for circadian rhythms in Drosophila (Ng territories?
et al. 2011) and respiratory activity in rodents (Gourine
et al. 2010). In the latter study, brainstem astrocytes were
found to be chemosensitive and respond to decreased blood AC K N OW L E D G M E N T S
pH/increased Pco2 with [Ca2+]i elevations expanding in the
astrocytic network to reach respiratory neurons and modify Research by AV is supported by the Swiss National Science
breathing patterns. Foundation, grants 3100A0 120398 and 31003A 140999

A S T R O C Y T E S : M O D U L AT I O N O F SY N A P T I C F U N C T I O N A N D N ET WO R K AC T I VI T Y • 491
NCCR “Synapsy” and NCCR “Transcure.” The author is Fiacco TA, McCarthy KD. 2004. Intracellular astrocyte calcium waves
grateful to Nicolas Liaudet for the artwork and to Hajime in situ increase the frequency of spontaneous AMPA receptor cur-
rents in CA1 pyramidal neurons. J Neurosci 24:722–732.
Hirase and the Journal of Neuroscience for granting permission Genoud C, Quairiaux C, Steiner P, Hirling H, Welker E, Knott GW.
to reproduce figures from the 2011 Takata et al. study. 2006. Plasticity of astrocytic coverage and glutamate transporter
expression in adult mouse cortex. PLoS Biol 4:e343.
Gordon GR, Baimoukhametova DV, Hewitt SA, Rajapaksha WR, Fisher
TE, Bains JS. 2005. Norepinephrine triggers release of glial ATP to
REFERENCES increase postsynaptic efficacy. Nat Neurosci 8:1078–1086.
Gordon GR, Iremonger KJ, Kantevari S, Ellis-Davies GC, MacVicar BA,
Agulhon C, Fiacco TA, McCarthy KD. 2010. Hippocampal short- and Bains JS. 2009. Astrocyte-mediated distributed plasticity at hypotha-
long-term plasticity are not modulated by astrocyte Ca2+ signaling. lamic glutamate synapses. Neuron 64:391–403.
Science 327:1250–1254. Gourine AV, Kasymov V, Marina N, Tang F, Figueiredo MF, Lane S,
Angulo MC, Kozlov AS, Charpak S, Audinat E. 2004. Glutamate et al. 2010. Astrocytes control breathing through pH-dependent
released from glial cells synchronizes neuronal activity in the hip- release of ATP. Science 329:571–575.
pocampus. J Neurosci 24:6920–6927. Grosche J, Matyash V, Möller T, Verkhratsky A, Reichenbach A,
Araque A, Parpura V, Sanzgiri RP, Haydon PG. 1998. Glutamate-dependent Kettenmann H. 1999. Microdomains for neuron-glia interac-
astrocyte modulation of synaptic transmission between cultured hip- tion: parallel fiber signaling to Bergmann glial cells. Nat Neurosci
pocampal neurons. Eur J Neurosci 10:2129–2142. 2:139–143.
Araque A, Parpura V, Sanzgiri RP, Haydon PG. 1999. Tripartite synapses: Halassa MM, Fellin T, Takano H, Dong JH, Haydon PG. 2007. Synaptic
glia, the unacknowledged partner. Trends Neurosci 22:208–215. islands defined by the territory of a single astrocyte. J Neurosci
Baker DA, Xi ZX, Shen H, Swanson CJ, Kalivas PW. 2002. The origin 27:6473–6477.
and neuronal function of in vivo nonsynaptic glutamate. J Neurosci Hamilton NB, Attwell D. 2010. Do astrocytes really exocytose neu-
22:9134–9141. rotransmitters? Nat Rev Neurosci 11:227–238.
Beattie EC, Stellwagen D, Morishita W, Bresnahan JC, Ha BK, Von Han J, Kesner P, Metna-Laurent M, Duan T, Xu L, Georges F, et al. 2012.
Zastrow M, et al. 2002. Control of synaptic strength by glial TNFα . Acute cannabinoids impair working memory through astroglial CB1
Science 295:2282–2285 receptor modulation of hippocampal LTD. Cell 148:1039–1050.
Bergersen LH, Morland C, Ormel L, Rinholm JE, Larsson M, Wold JF, Henneberger C, Papouin T, Oliet SH, Rusakov DA. 2010. Long-term
et al. 2011. Immunogold detection of L-glutamate and D-serine in potentiation depends on release of D-serine from astrocytes. Nature
small synaptic like microvesicles in adult hippocampal astrocytes. 463:232–236.
Cereb Cortex, in press. Holtzclaw LA, Pandhit S, Bare DJ, Mignery GA, Russell JT. 2002.
Bergles DE, Jahr CE. 1998. Glial contribution to glutamate uptake Astrocytes in adult rat brain express type 2 inositol 1,4,5-trisphosphate
at Schaffer collateral-commissural synapses in the hippocampus. receptors. Glia 39:69–84.
J Neurosci 18:7709–7716. Jourdain P, Bergersen LH, Bhaukaurally K, Bezzi P, Santello M, Domercq
Bezzi P, Carmignoto G, Pasti L, Vesce S, Rossi D, Lodi Rizzini B, et al. M, et al. 2007. Glutamate exocytosis from astrocytes controls synap-
1998. Prostaglandins stimulate calcium-dependent glutamate release tic strength. Nat Neurosci 10:331–339.
in astrocytes. Nature 391:281–285. Kaneko M, Stellwagen D, Malenka RC, Stryker MP. 2008. Tumor
Bezzi P, Gundersen V, Galbete JL, Seifert G, Steinhäuser C, Pilati E, necrosis factor-alpha mediates one component of competitive,
et al. 2004. Astrocytes contain a vesicular compartment that is experience-dependent plasticity in developing visual cortex. Neuron
competent for regulated exocytosis of glutamate. Nat Neurosci 7: 58:673–680.
613–620. Kang J, Jiang L, Goldman SA, Nedergaard M. 1998. Astrocyte-mediated
Bezzi P, Volterra A. 2001. A neuron-glia signalling network in the active potentiation of inhibitory synaptic transmission. Nat Neurosci
brain. Curr Opin Neurobiol 11:387–394. 1:683–692.
Bushong EA, Martone ME, Jones YZ, Ellisman MH. 2002. Protoplasmic Kettenmann H, Faissner A, Trotter J. 1996. Neuron-glia interactions
astrocytes in CA1 stratum radiatum occupy separate anatomical in homeostasis and degeneration. In: Greger R, Windhorst U (eds.),
domains. J Neurosci 22:183–192. Human physiology. Heidelberg: Springer-Verlag, pp. 533–543.
Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson Khurgel M, Koo AC, Ivy GO. 1996. Selective ablation of astrocytes by
KS, et al. 2008. A transcriptome database for astrocytes, neurons, intracerebral injections of alpha-aminoadipate. Glia 16:351–358.
and oligodendrocytes: a new resource for understanding brain devel- Kozlov AS, Angulo MC, Audinat E, Charpak S. 2006. Target cell-specific
opment and function. J Neurosci 28:264–278. modulation of neuronal activity by astrocytes. Proc Natl Acad Sci U
Cavelier P, Hamann M, Rossi D, Mobbs P, Attwell D. 2005. Tonic exci- S A 103:10058–10063.
tation and inhibition of neurons: ambient transmitter sources and Kuga N, Sasaki T, Takahara Y, Matsuki N, Ikegaya Y. 2011. Large-scale
computational consequences. Prog Biophys Mol Biol 87:3–16. calcium waves traveling through astrocytic networks in vivo.
Dalby NO, Mody I. 2003. Activation of NMDA receptors in rat dentate J Neurosci 31:2607–2614
gyrus granule cells by spontaneous and evoked transmitter release. Kullmann DM, Asztely F. 1998. Extrasynaptic glutamate spillover in the
J Neurophysiol 90:786–797 hippocampus: evidence and implications. Trends Neurosci 21:8–14.
Dani JW, Chernjavsky A, Smith SH. 1992. Neuronal activity triggers cal- Lee S, Yoon BE, Berglund K, Oh SJ, Park H, Shin HS, et al. 2010a.
cium waves in hippocampal astrocyte networks. Neuron 8:429–440 Channel-mediated tonic GABA release from glia. Science
D’Ascenzo M, Fellin T, Terunuma M, Revilla-Sanchez R, Meaney DF, 330:790–796.
Auberson YP, et al. 2007. mGluR5 stimulates gliotransmission in the Lee SH, Halassa MM, Hines D, Gainey L, Deng Q, Pascual O, et al. 2010b.
nucleus accumbens. Proc Natl Acad Sci U S A 104:1995–2000. Astrocytic IP3/Ca2+ signals are required for mGluR5-dependent
Di Castro MA, Chuquet J, Liaudet N, Bhaukaurally K, Santello M, enhancement of synaptic plasticity and learning. San Diego: Society
Bouvier D, et al. 2011. Local Ca 2+ detection and modulation of syn- for Neuroscience.
aptic release by astrocytes. Nat Neurosci 14:1276–1284. Liu QS, Xu Q, Arcuino G, Kang J, Nedergaard M. 2004.
Fellin T, Pascual O, Gobbo S, Pozzan T, Haydon PG, Carmignoto G. Astrocyte-mediated activation of neuronal kainate receptors. Proc
2004. Neuronal synchrony mediated by astrocytic glutamate through Natl Acad Sci U S A 101:3172–3177.
activation of extrasynaptic NMDA receptors. Neuron 43:729–743. Marchaland J, Calì C, Voglmaier SM, Li H, Regazzi R, Edwards RH,
Fiacco TA, Agulhon C, Taves SR, Petravicz J, Casper KB, Dong X, et al. et al. 2008. Fast subplasma membrane Ca2+ transients con-
2007. Selective stimulation of astrocyte calcium in situ does not affect trol exo-endocytosis of synaptic-like microvesicles in astrocytes.
neuronal excitatory synaptic activity. Neuron 54:611–626 J Neurosci 28:9122–9132.

492 • FUNCTIONS OF NEUROGLIAL CELLS


Matsui K, Jahr CE. 2006. Exocytosis unbound. Curr Opin Neurobiol Robitaille R. 1998. Modulation of synaptic efficacy and synaptic depres-
16:305–311. sion by glial cells at the frog neuromuscular junction. Neuron
Matyash V, Filippov V, Mohrhagen K, Kettenmann H. 2001. Nitric 21:847–855.
oxide signals parallel fiber activity to Bergmann glial cells in the Roux L, Benchenane K, Rothstein JD, Bonvento G, Giaume C. 2011.
mouse cerebellar slice. Mol Cell Neurosci 18:664–670. Plasticity of astroglial networks in olfactory glomeruli. Proc Natl
Min R, Nevian T. 2012. Astrocyte signalling controls spike Acad Sci U S A 108:18442–18446.
timing-dependent plasticity at neocortical synapses. Nat Neurosci Rusakov DA, Lehre KP. 2002. Perisynaptic asymmetry of glia: new
15(5):746–753. insights into glutamate signalling. Trends Neurosci 25:492–494.
Navarrete M, Araque A. 2008. Endocannabinoids mediate Santello M, Bezzi P, Volterra A. 2011. TNFα controls glutamatergic gliotrans-
neuron-astrocyte communication. Neuron 57:883–893. mission in the hippocampal dentate gyrus. Neuron 69:988–1001.
Navarrete M, Araque A. 2010. Endocannabinoids potentiate synap- Sasaki T, Matsuki N, Ikegaya Y. 2011. Action-potential modulation dur-
tic transmission through stimulation of astrocytes. Neuron 68: ing axonal conduction. Science 331:599–601.
113–126. Schubert V, Bouvier D, Volterra A. 2011. SNARE protein expression in
Navarrete M, Perea G, Fernandez de Sevilla D, Gomez-Gonzalo M, synaptic terminals and astrocytes in the adult hippocampus: a com-
Nunez A, Martin ED, et al. 2012. Astrocytes mediate in vivo parative analysis. Glia 59:1472–1488.
cholinergic-induced synaptic plasticity. Plos Biol 10:e1001259. Schummers J, Yu H, Sur M. 2008. Tuned responses of astrocytes and
Newman EA. 2003. Glial cell inhibition of neurons by release of ATP. J their influence on hemodynamic signals in the visual cortex. Science
Neurosci 23:1659–1666. 320:1638–1643.
Ng FS, Tangredi MM, Jackson FR. 2011. Glial cells physiologically mod- Serrano A, Haddjeri N, Lacaille JC, Robitaille R. 2006. GABAergic net-
ulate clock neurons and circadian behavior in a calcium-dependent work activation of glial cells underlies hippocampal heterosynaptic
manner. Curr Biol 21:625–634. depression. J Neurosci 26:5370–5382.
Nimmerjahn A, Mukamel EA, Schnitzer MJ. 2009. Motor behavior Shigetomi E, Bowser DN, Sofroniew MV, Khakh BS. 2008. Two forms
activates Bergmann glial networks. Neuron 62:400–412. of astrocyte calcium excitability have distinct effects on NMDA
Oliet SH, Piet R, Poulain DA. 2001. Control of glutamate clearance receptor-mediated slow inward currents in pyramidal neurons.
and synaptic efficacy by glial coverage of neurons. Science 292: J Neurosci 28:6659–6663.
923–926. Steinmetz CC, Turrigiano GG. 2010. Tumor necrosis factor-alpha sig-
Panatier A, Theodosis DT, Mothet JP, Touquet B, Pollegioni L, Poulain naling maintains the ability of cortical synapses to express synaptic
DA, et al. 2006. Glia-derived D-serine controls NMDA receptor scaling. J Neurosci 30:14685–14690.
activity and synaptic memory. Cell 125:775–784 Stellwagen D, Beattie EC, Seo JY, Malenka RC. 2005. Differential regu-
Panatier A, Vallée J, Haber M, Murai KK, Lacaille JC, Robitaille R. lation of AMPA receptor and GABA receptor trafficking by tumor
2011. Astrocytes are endogenous regulators of Basal transmission at necrosis factor-alpha. J Neurosci 25:3219–3228
central synapses. Cell 146:785–798. Stellwagen D, Malenka RC. 2006. Synaptic scaling mediated by glial
Parpura V, Basarsky TA, Liu F, Jeftinija K, Jeftinija S, Haydon PG. TNF-alpha. Nature 440:1054–1059.
1994. Glutamate-mediated astrocyte-neuron signalling. Nature Stevens ER, Esguerra M, Kim PM, Newman EA, Snyder SH, Zahs KR,
369:744–747. et al. 2003. D-serine and serine racemase are present in the vertebrate
Parri HR, Gould TM, Crunelli V. 2001. Spontaneous astrocytic Ca 2+ retina and contribute to the physiological activation of NMDA recep-
oscillations in situ drive NMDAR-mediated neuronal excitation. Nat tors. Proc Natl Acad Sci U S A 100:6789–6794.
Neurosci 4:803–812. Suzuki A, Stern SA, Bozdagi O, Huntley GW, Walker RH, Magistretti
Pascual O, Casper KB, Kubera C, Zhang J, Revilla-Sanchez R, Sul JY, PJ, et al. 2011. Astrocyte-neuron lactate transport is required for
et al. 2005. Astrocytic purinergic signaling coordinates synaptic net- long-term memory formation. Cell 144:810–823.
works. Science 310:113–116. Swanson RA, Graham SH. 1994. Fluorocitrate and fluoroacetate effects
Pasti L, Volterra A, Pozzan T, Carmignoto G. 1997. Intracellular calcium on astrocyte metabolism in vitro. Brain Res 664:94–100.
oscillations in astrocytes: a highly plastic, bi-directional form of communi- Takata N, Mishima T, Hisatsune C, Nagai T, Ebisui E, Mikoshiba K,
cation between neurons and astrocytes in situ. J Neurosci 17:7817–7830. et al. 2011. Astrocyte calcium signaling transforms cholinergic mod-
Perea G, Araque A. 2005. Properties of synaptically evoked astrocyte ulation to cortical plasticity in vivo. J Neurosci 31:18155–18165.
calcium signal reveal synaptic information processing by astrocytes. Ventura R, Harris KM. 1999. Three-dimensional relationships between
J Neurosci 25:2192–2203. hippocampal synapses and astrocytes. J Neurosci 19:6897–6906.
Perea G, Araque A. 2007. Astrocytes potentiate transmitter release at Volterra A, Magistretti PJ, Haydon PG (eds.) 2002. The tripartite synapse:
single hippocampal synapses. Science 317:1083–1086. glia in synaptic transmission. New York: Oxford University Press.
Perea G, Navarrete M, Araque A. 2009. Tripartite synapses: astrocytes pro- Volterra A, Meldolesi J. 2005. Astrocytes, from brain glue to com-
cess and control synaptic information. Trends Neurosci 32:421–431. munication elements: the revolution continues. Nat Rev Neurosci
Petravicz J, Fiacco TA, McCarthy KD. 2008. Loss of IP3 receptor-dependent 6:626–640.
Ca 2+ increases in hippocampal astrocytes does not affect baseline CA1 Wang X, Lou N, Xu Q, Tian GF, Peng WG, Han X, et al. 2006.
pyramidal neuron synaptic activity. J Neurosci 28:4967–4973. Astrocytic Ca 2+ signaling evoked by sensory stimulation in vivo. Nat
Petzold GC, Albeanu DF, Sato TF, Murthy VN. 2008. Coupling of neu- Neurosci 9:816–823.
ral activity to blood flow in olfactory glomeruli is mediated by astro- Xu J, Peng H, Kang N, Zhao Z, Lin JH, Stanton PK, et al. 2007.
cytic pathways. Neuron 58:897–910. Glutamate-induced exocytosis of glutamate from astrocytes. J Biol
Piet R, Vargova L, Sykova E, Poulain DA, Oliet SH. 2004. Physiological Chem 282:24185–24197.
contribution of the astrocytic environment of neurons to intersynap- Yang Y, Ge W, Chen Y, Zhang Z, Shen W, Wu C, et al. 2003. Contribution
tic crosstalk. Proc Natl Acad Sci U S A 101:2151–2155. of astrocytes to hippocampal long-term potentiation through release
Porter JT, McCarthy KD. 1996. Hippocampal astrocytes in situ of D-serine. Proc Natl Acad Sci U S A 100:15194–15199.
respond to glutamate released from synaptic terminals. J Neurosci Zhang JM, Wang HK, Ye CQ, Ge W, Chen Y, Jiang ZL, et al. 2003. ATP
16:5073–5081. released by astrocytes mediates glutamatergic activity-dependent het-
Poskanzer KE, Yuste R. 2011. Astrocytic regulation of cortical UP states. erosynaptic suppression. Neuron 40:971–982.
Proc Natl Acad Sci U S A 108:18453–18458. Zonta M, Angulo MC, Gobbo S, Rosengarten B, Hossmann KA, Pozzan
Pozo K, Goda Y. 2010. Unraveling mechanisms of homeostatic synaptic T, et al. 2003. Neuron-to-astrocyte signaling is central to the dynamic
plasticity. Neuron 66:337–351. control of brain microcirculation. Nat Neurosci 6:43–50.

A S T R O C Y T E S : M O D U L AT I O N O F SY N A P T I C F U N C T I O N A N D N ET WO R K AC T I VI T Y • 493
39.
ASTROCY TIC MODULATION OF MAMMALIAN
SYNAPSES: CIRCUITS AND BEHAVIOR S
Michael M. Halassa and Philip G. Haydon

A B B R E VI AT I O N S transmission as well as for the normal control of sleep-wake


cycles. Because sleep disorders are prevalent and comorbid
A1R adenosine 1 receptor with many other neurological and psychiatric disorders, it is
ATP adenosine triphosphate hypothesized that astrocytes play important roles in the gen-
cAMP cyclic adenosine monophosphate esis and expression of these conditions.
CPT cyclopentyl theophylline
EEG electroencephalograph
EMG electromyograph 2 ASTROCY TIC TR ANSMITTER
GABA gamma-aminobutyric acid R E C YC L I N G I S E S S E N T I A L F O R
GLT-1 glutamate transporter 1 NORMAL COGNITION
GS glutamine synthetase
HFS high frequency stimulation Synapses are now recognized to be tripartite structures. In
L-LTP late long-term potentiation addition to the presynaptic and postsynaptic terminals, the
LTP long-term potentiation surrounding astrocytic process is considered to be an integral
NREM non–rapid eye movement part of the synapse (Araque 2006; Haydon, 2001; Kettenmann
REM rapid eye movement et al. 1996). In contrast to the neural elements of the synapses,
SCN suprachiasmatic nucleus however, astrocytes operate on a slower timescale (>six orders
SNARE soluble NSF attachment protein receptor of magnitude) to modulate synaptic transmission (Halassa
TNF-α tumor necrosis factor alpha et al. 2007a). For example, astrocytes are known to express
TTX tetrodotoxin inward rectifier potassium channels (e.g., Kir4.1) (Olsen and
VAMP2 vesicle-associated membrane protein 2 Sontheimer 2008) whose function is essential for maintain-
ing normal ionic homeostasis at the synapse. Loss of astrocytic
potassium buffering is associated with a number of neuro-
1 INTRODUCTION logical illnesses, including epilepsy (Hinterkeuser et al. 2000;
Steinhauser and Seifert 2002) and tuberous sclerosis ( Jansen
Synapses are tripartite structures in which the astrocyte is a et al. 2005).
third element along with the presynaptic and postsynaptic ter- One of the most recognized roles of astrocytes at the syn-
minal. The tripartite synapse is recognized as playing essential apse is neurotransmitter uptake and recycling (Sattler and
roles to sustain synaptic transmission and as a consequence Rothstein 2006) (see chapter 35). Astrocytes express high
the function of neuronal circuits that underlie behavior. This levels of the glutamate transporter GLT-1, which constitutes
chapter highlights how the astrocytic component can influ- about 1% of total brain protein (Rothstein et al. 1994). This
ence synapses through the supply of neurotransmitter precur- transporter uses the sodium concentration gradient to drive
sors, cytokines as well as gliotransmitters, which modulate the influx of glutamate into astrocytes following synaptic
synaptic transmission. In particular, this chapter pays special transmission. Astrocytes subsequently convert glutamate to
attention to the intact animal, in which it is critical to under- glutamine, which they release back onto presynaptic termi-
stand how sensory experiences influence astrocytes and as a nals. In excitatory terminals, glutamine is converted to gluta-
consequence how the astrocyte provides feedback regulation mate, completing the “glutamate-glutamine” cycle, whereas
of the behavioral state. This chapter also pays special attention it is used in inhibitory terminals for the synthesis of GABA
to the authors’ work, which provided the first demonstra- (Schousboe et al. 1991, 1992). Interestingly, GABAergic ter-
tion of behavioral roles for astrocytes in the control of sleep minals have a limited stored pool of glutamine and thus are
homeostasis through the regulation of the level of the extracel- highly dependent on astrocytic glutamine synthesis com-
lular purine adenosine. Although astrocytes are not electrically pared with glutamatergic terminals (Liang et al. 2006). In
excitable and are thus relatively slow signaling systems, it is fact, acute inhibition of astrocytic glutamine synthetase (GS)
now known that they exert powerful actions in brain function in rodents precipitates seizures (Eid et al. 2008; Wang et al.
and are essential for the maintenance of GABAergic synaptic 2009), likely because of a failure of inhibition. Furthermore,

494
astrocytes express the GABAergic transporter GAT-3 that example of major depressive disorder, in which parallels have
has been recently shown to be regulated by submembrane been found between decreases in cortical astrocytes (Ongur
Ca2+ changes mediated by TRPA1 channels (Shigetomi et al. et al. 1998) and decreases in neocortical GABA (Sanacora
2011). Inhibition of these ion channels results in a significant et al. 1999). Further research is needed to determine whether
decrease in the surface expression of GAT-3 in astrocytes and these processes are causally linked.
a subsequent attenuation of miniature inhibitory postsynaptic One area in which such a linkage has been found concerns
potentials of nearby neurons. epilepsy (see chapter 70). In the epileptic temporal lobe, astro-
Astrocytic impact on synaptic inhibition is critical to cytes enter a state of reactive astrocytosis with an associated
understanding systems-level processes given the emerging reduction in the expression of the astrocyte-enriched enzyme
roles of inhibition in neural circuit function. In addition to glutamine synthetase. Given that acute pharmacological inhi-
other functions, fast synaptic inhibition is critical for sensory bition of this enzyme leads to an attenuation of GABAergic
function (Swadlow 2003), gating of modulatory input (Friedel inhibition and seizures, it would not be surprising if reactive
and van Hemmen 2008), and the emergence of particular neu- astrocytosis initiates certain neuronal dysfunctions. In agree-
ronal oscillations (Bazhenov et al. 2000; Hajos et al. 2004). ment with this possibility, selective transduction of astrocytes
Neocortical gamma oscillations, for example, are thought to with adeno-associated virus serotype 5 leads to reactive astro-
be generated by rhythmic activity of parvalbumin-positive fast cytosis and a loss of glutamine synthetase (Ortinski et al. 2011).
spiking interneurons (Cardin et al. 2009). Gamma oscillations Coincident with this change is a reduction in synaptic inhibi-
are thought to be essential for normal cognitive function, tion, which is restored by addition of exogenous glutamine,
likely because of the impact it has on regulating the timing of and enhanced excitation viewed with voltage-sensitive dyes
pyramidal cell firing (Cardin et al. 2009; Sohal et al. 2009). throughout brain slices (Fig. 39.1).
As a result, failure of gamma oscillations is seen in a number of
different neuropsychiatric diseases, including schizophrenia
and autism (Uhlhaas and Singer 2006, 2007; Uhlhaas et al. 3 A S T R O C Y T E -D E R I VE D
2008), and is often linked to a primary dysfunction in inhibi- C Y TO K I N E S M O D U L AT E C O RT I C A L
tory interneurons. Given the important modulatory impact D E VE L O PM E N TA L P L A S T I C I T Y
astrocytes have on inhibition at multiple levels (neurotrans-
mitter synthesis and uptake), a disruption in these processes Tumor necrosis factor alpha (TNFα) is a proinflammatory
is likely to lead to major pathology. This is supported by the cytokine that is known to mediate crucial signaling functions

A B +10 ms +100 ms +600 ms

CA1
Control

CA2
SC
EC
TA
DG 0.2
dF/F (%)
AAV2/5

CA3 0.1
stim
0

–0.1

C
AAV2/5
Control
SO

SO

0.12 0.12
dF/F (%)

dF/F (%)

0.08 0.08
SR

SR

0.04 0.04
SLM

SLM

0 0
0 0.3 0.7 0 0.3 0.7 0 0.3 0.7 0 0.3 0.7
Time (s) Time (s) Time (s) Time (s)

Figure 39.1 Selective Induction of Astrocytic Gliosis Results in Hippocampal Hyperexcitability. A. Schematic of the hippocampal slice preparation.
B. Voltage-sensitive dye imaging at different time points following the stimulation of entorhinal cortex input to CA1. C. Induction of astrogliosis via
selective transduction of astrocytes with high-titer AAV2/5 harboring green fluorescent protein results in enhanced network activity in response to
input at the EC to CA1 synapses. From Ortinski et al. 2011.

A S T R O C Y T I C M O D U L AT I O N O F M A M M A L I A N SY N A P S E S : C I R C U I T S A N D B E H AVI O R S • 495
across multiple organs (Heller and Kronke 1994). In the brain, 3.1 B E H AVI O R M O D U L AT E S A S T RO C Y T I C
this molecule is thought to be released by astrocytes, based S T RU C T U R E A N D F U N C T I O N
on cell culture experiments in which astrocyte-enriched glial
Astrocytic morphology is known to be modulated by neu-
cultures released this molecule, and recently has been shown
ronal activity resulting from behavioral changes. Because the
to mediate excitatory synaptic scaling (Stellwagen and
greatest majority of astrocytic volume is in tertiary processes
Malenka 2006). This form of non-Hebbian plasticity occurs
that contact synapses (Bushong et al. 2002; Halassa et al.
after prolonged periods of neuronal inactivity, such as experi-
2007b), astrocytic morphological changes have profound
mental incubation of cell or slice cultures with an activ-
impact on synaptic physiology. This process has been observed
ity blocker such as the voltage-sensitive channel blocker
in multiple brain regions, including the hypothalamus, supra-
tetrodotoxin (TTX). After a 24-hour incubation with
chiasmatic nucleus (SCN), and sensory cortex. In the hypo-
TTX, cultured neurons exhibit an increase in the miniature
thalamus, astrocytic coverage of oxytocin neurons has been
excitatory postsynaptic currents (mEPSCs), a process that
shown to be modulated by lactation, parturition, and chronic
requires release of TNFα from nearby astrocytes. Co-culturing
neurons derived from wild-type mice with astrocytes dehydration, conditions associated with massive neurohypo-
derived from TNFα knockout mice failed to result in syn- physial hormone secretion (Oliet et al. 2008; Panatier and
aptic scaling when neuronal activity was blocked, providing Oliet 2006) (see chapter 41). Although the functional conse-
direct mechanistic support for an astrocytic source of TNFα quence of this process is unclear, it is associated with increased
in synaptic scaling. Interestingly, astrocytic TNFα does not juxtaposition of neuronal processes and a rise in extracellular
appear to be essential for Hebbian forms of plasticity (LTP glutamate because of a decrease in glutamate transport (Oliet
and LTD), at least under a limited number of stimulation et al. 2001). In the suprachiasmatic nucleus, astrocytic mor-
paradigms. phology changes as a function of the circadian phase (Gerics
Experience-dependent plasticity in primary sensory neo- et al. 2006). Recent molecular investigations have shown
cortical areas is critical for the development of normal percep- that expression of the cytoskeletal astrocytic protein ezrin is
tion (Buonomano and Merzenich 1998). In the visual system, modulated by the circadian cycle in the SCN (Lavialle et al.
monocular deprivation during a critical period (28 days 2011). Ezrin expression is linked to astrocytic filopodia motil-
postnatal in mice) results in rapid cortical plasticity (4 days ity and activity of astrocytic metabotropic glutamate recep-
in mice) (Gordon and Stryker 1996). Recordings in the bin- tors, suggesting that astrocytic volumetric changes in the SCN
ocular region of mouse primary visual cortex (V1) normally are driven by photic stimulation. The impact of this process
show a predominant representation of the contralateral eye on circadian rhythms is unclear, although a general role for
(Gordon et al. 1996). This representation shifts to a primar- astrocytes in regulating circadian rhythms is emerging. In pri-
ily ipsilateral presentation on closure of the contralateral eye. mary somatosensory cortex, chronic vibrissal stimulation by
Recent studies have shown that two dissociable processes may remotely controlled piezo stimulation in mice results in a two-
underlie this rapid shift; a weakening of visual responsiveness fold increase of astrocytic coverage of synapses and upregula-
to the deprived eye and a delayed strengthening of the nonde- tion of GLT-1 expression (Genoud et al. 2006).
prived eye (Frenkel and Bear 2004). Although the first pro-
cess is thought to be mediated by Hebbian plasticity rules that 4 S E N S O RY S T I MU L AT I O N S E VO K E
mediate synaptic depression, non-Hebbian synaptic scaling C A L C I U M S I G N A L S I N C O RT I C A L
processes are thought to underlie the second process (Desai ASTROCY TES
et al. 2002). Guided by the discovery that astrocytic TNFα
mediates synaptic scaling, Kaneko et al. (2008) made intrigu- Astrocytes respond to transmitter spillover from nearby syn-
ing observations on the role of this process in developmental apses by intracellular Ca2+ elevation (see chapter 26). In the
visual plasticity. Tumor necrosis factor alpha knock-out mice anesthetized mouse, repetitive 10-Hz single vibrissal stimu-
showed overall attenuated monocular deprivation plastic- lation results in astrocytic Ca2+ signals in the corresponding
ity response compared with wild-type littermates. However, cortical barrel. This response, which has a latency of approx-
intrinsic optical imaging of the cortical response to visual imately 3 seconds, is dependent on activation of astrocytic
stimulation of the deprived eye 3 days after lid closure revealed metabotropic glutamate receptors (Wang et al. 2006). In
an equal attenuation in TNFα knock-out and wild-type ani- the anesthetized ferret, visual gratings that trigger neuronal
mals. After 5 days of lid closure, wild-type animals showed an activity results in similar activation of nearby astrocytes,
increased response to the nondeprived eye, whereas TNFα suggesting a spatial visual alignment between these two cell
knockouts failed to show an increase, concluding that a syn- types (Schummers et al. 2008). Interestingly, astrocytes show
aptic scaling–like process in vivo is essential for mediating the sharper tuning to orientation and spatial frequency than
later phase of monocular deprivation plasticity. Although an neurons, and a group of two to fewer than ten astrocytes
astrocyte-specific manipulation was not performed in these responds to a particular one-stimulus orientation, suggesting
experiments, it is possible that astrocytes control this process a network organization of visual cortical astrocytes. Because
based on aforementioned culture data. Overall, this finding sensory tuning of cortical neurons is known to be shaped by
strengthens the concept that astrocytes act as modulators the interplay between excitation and inhibition (Higley and
of neuronal activity, mediating processes over a timescale of Contreras 2006; Liu et al. 2010), this experiment suggests that
hours to days. astrocytes may be tracking the output rather than the input

496 • FUNCTIONS OF NEUROGLIAL CELLS


of neurons. Inhibiting the astrocytic visual response inhibits One of the first demonstrations that ATP is a gliotransmit-
the hemodynamic response in visual cortex (Schummers et al. ter was made by Kater’s laboratory when they performed cell
2008). Similarly, mouse olfactory bulb astrocytes respond to culture experiments examining the mechanism underlying the
odor-evoked synaptic input, and their response coincides with propagation of Ca2+ waves between astrocytes in cell culture
the vascular response (Petzold et al. 2008). These converging (Guthrie et al. 1999). In culture an elevation of astrocytic Ca2+
findings from different brain regions and species strongly link in one cell can propagate to neighbors as a wave of elevated
astrocytic Ca2+ signaling to activity-dependent neurovascular Ca2+. Although the relevance in vivo is unclear, at the time
coupling. Consistent with this conclusion are recent findings these studies were performed there was great interest in identi-
showing decoupling of cerebral blood flow from local neu- fying the intercellular signaling pathways in culture to identify
ronal activity (Mishra et al. 2011) under certain conditions, the potential mechanisms that may operate in vivo. Kater’s lab-
suggesting a non-neuronal mediator of the cerebrovascular oratory made two critical observations that initiated a series of
signal, and that under such conditions the vascular response studies on the role of purinergic signaling by astrocytes. First,
might play an anticipatory role in brain state changes, rather they asked whether Ca2+ waves propagate between neighbor-
than act as a follower for neuronal activity. This suggests that ing cells via a cell contact–mediated mechanism, such as gap
one function of astrocytic sensory drive is mediating an antic- junctions, or whether there was an extracellular route of signal
ipatory hemodynamic response to alter local circuit function propagation. To achieve this objective, they physically iso-
(Moore and Cao 2008). lated astrocytes by making a scratch in the culture. Strikingly,
they observed that when a Ca2+ wave approached the edge of
a monolayer of astrocytes, after a brief pause, the Ca2+ wave
5 P H C H A N G E S E VO K E C A 2 + S I G N A L S could continue at a distance in physically isolated astrocytes
IN M E D UL L A RY A ST R O C Y T E S (Fig. 39.2). This clearly pointed to the presence of an extra-
cellular signal in Ca2+ wave propagation. Further support was
Ca2+ imaging using the genetically encoded indicator Case12 provided by a clever experiment in which they sucked the
revealed that astrocytes of the medulla chemosensitive zone are extracellular saline from around astrocytes when their Ca2+
sensitive to changes in pH (Gourine et al. 2010). A pH drop level elevated. They then moved the pipette to a different cel-
from 7.4 to 7.2 resulted in a robust increase in intracellular Ca2+ lular region with low Ca2+ and were able to evoke a Ca2+ signal
of several astrocytes. These Ca2+ signals were independent of on ejection. To identify the nature of the extracellular signal
neuronal activity as application of TTX or muscimol (a potent they used apyrase, an enzyme that metabolizes ATP as well as
GABA receptor agonist known to silence local neural activity) purinergic antagonists that both blocked the ability of the col-
had no impact on astrocytic Ca2+ response to changes in pH. lected medium to evoke a Ca2+ signal. These elegant studies
Furthermore, pH sensitivity appeared to be a feature of astro- clearly identified ATP as a potential extracellular signal in the
cytes of the ventral medulla because cortical or dorsal brain- CNS that is used by astrocytes.
stem astrocytes failed to show a Ca2+ response to a pH drop. Making the transition from cell culture studies to in situ
Although the precise mechanism by which changes in pH are and in vivo approaches has been more challenging, largely
linked to astrocytic Ca2+ signals is unknown, hydrolyzing extra- because of the difficulty of providing selective stimulation and
cellular ATP by application of apyrase results in abolishment inhibition of astrocytes. However, the advent of cell-specific
of the astrocytic Ca2+ response. Further, optogenetic elevation molecular genetics has provided a powerful tool to investi-
of astrocytic Ca2+ in medullary astrocytes augments breathing gate the regulation of gliotransmission in vivo. In 2005 we
rate, causally demonstrating the astrocytic drive of behavior. developed a conditional astrocyte-selective tool that impaired
the astrocyte-derived accumulation of extracellular purines
(Pascual et al. 2005). There has been much debate about how
5.1 T H E S L E E P -WA K E C YC L E I S A S S O C I AT E D
gliotransmitters are released from astrocytes. Although space
WI T H C H A N G E S I N A S T RO C Y T E -
will not be used here to rehash the pros and cons of different
D E R I VE D A D E N O S I N E
release mechanisms, the authors used a powerful and highly
Since 1994 when we first observed that gliotransmitters can selective tool to impair regulated exocytosis (Fig. 39.3).
be released from astrocytes (Parpura et al. 1994), there has Vesicle fusion requires the formation of a SNARE com-
been a constant pursuit of identifying the conditions that plex. The authors interfered with SNARE complex formation
control when these gliotransmitters are released. Although using a well-characterized approach of expressing the SNARE
this initial observation identified glutamate as a gliotransmit- domain of a vesicle protein: The expressed protein prevents
ter, understanding the regulation of this gliotransmitter has the formation of the endogenous complex and thereby pre-
been the most difficult, probably because of the presence of vents that particular SNARE-mediated membrane fusion
avid glutamate transporters, which clamp extracellular gluta- event. It should be noted that there is a high degree of speci-
mate at around 1 μM. In contrast to glutamate, the regula- ficity between different SNARES. For example, the SNARE
tion of the gliotransmitter adenosine has yielded much more domain of VAMP2 does not inhibit VAMP8 events and visa
readily, probably because it is less tightly controlled. Much of versa (Scales et al. 2000a,b).
the uptake is regulated by equilibrative transporters rather The authors used the tetracycline-off system together
than concentrative Na+-dependent uptake, as is the case for with an astrocyte-selective promoter to conditionally con-
glutamate. trol the expression of the SNARE domain of VAMP2 only in

A S T R O C Y T I C M O D U L AT I O N O F M A M M A L I A N SY N A P S E S : C I R C U I T S A N D B E H AVI O R S • 497
A B

Figure 39.2 Astrocytic Ca2+ Waves Spread Through an Extracellular Messenger. Pioneering work by Kater’s group using fluo-3 AM imaging of Ca2+
in cultured astrocytes. Astrocytic Ca2+ wave can spread throughout the culture despite physical discontinuity. From Guthrie et al. 1999.

astrocytes. Independent studies have shown that this pertur- that astrocytes supply the source of adenosine that tonically
bation impairs regulated membrane cycling, but expression of suppresses excitatory synaptic transmission (Fig. 39.4C).
the SNARE domain does not affect trafficking of channels or There are two prominent mechanisms that could give rise
receptors, for example (Fellin et al. 2009). That this is a highly to extracellular adenosine: the release through equilibrative
selective approach is also provided by the observation that transporters or the release of ATP that is metabolized in the
astrocytes maintain their physiological integrity even follow- extracellular space to adenosine. Data supported the concept
ing months of continued expression of the SNARE domain. that this arises from the latter because luminescence imaging
Once the authors developed mice expressing dominant demonstrated that dnSNARE expression also reduced extra-
negative SNARE only in astrocytes (termed dnSNARE mice), cellular ATP and the inhibition of equilibrative nucleoside
it was asked whether they changed the extracellular concen- transporters did not lead to a depletion of adenosine, but
tration of chemical transmitters. Initially neurons were used rather to an increase in adenosine. Together with other data
as sensors of gliotransmitters, but more recently these results it was concluded that astrocytes release ATP that is hydro-
have been validated using biosensors of purines, which directly lyzed in the extracellular space to adenosine. Because of the
measure purines in the extracellular space. Initial studies were selectivity of the dnSNARE manipulation for inhibiting
performed using hippocampal brain slices in which the effect of exocytosis, the authors conclude that the most likely mecha-
astrocytic expression of dnSNARE on basal synaptic transmis- nism underlying this release is through vesicle fusion with the
sion was monitored (Pascual et al. 2005). Input/output curves plasma membrane. However, there is also evidence indicating
of Schaffer collateral CA1 synaptic transmission was moni- that connexins can contribute to ATP release. Whether these
tored in the presence and absence of dnSNARE, and a strik- two pathways are linked or independent parallel mechanisms
ing result was observed: When dnSNARE was expressed in remains to be revealed.
astrocytes, the magnitude of basal excitatory transmission was Perhaps the most surprising aspect of this dnSNARE study
augmented (Fig. 39.4A,B). It is well known that there is a tonic was that much of the adenosine was found to be derived from
adenosine-mediated presynaptic inhibition of excitatory trans- a glial source. Given that neurons can release ATP during syn-
mission in hippocampus and cortex. Therefore, the authors aptic transmission, one might have thought that adenosine
asked whether this source of adenosine was derived from the would accumulate from neuronal release of purines. Clearly,
astrocyte. Initial studies determined the magnitude of enhance- neurons can provide purines. However, they release adenosine
ment of synaptic transmission caused by addition of adenosine in a pulsatile, activity-dependent manner providing fast neu-
1 receptor (A1R) antagonists. In dnSNARE mice there was a romodulation, while astrocytes control the tonic accumula-
significant attenuation of the basal adenosine tone, indicating tion of adenosine.

498 • FUNCTIONS OF NEUROGLIAL CELLS


A Doxycycline B
hGFAP tTA tTA

Doxycycline No Doxycycline

TetO Lac-Z TetO Lac-Z

C D
TetO EGFP TetO EGFP

TetO SNARE TetO SNARE


Astrocyte-specific
No expression
expression dn-SNARE + Dox dn-SNARE – Dox

E F G

β-Gal/EGFP GFAP Merge

H I J

β-Gal/EGFP NeuN Merge

Figure 39.3 Temporally Regulated, Astrocyte-Specific Expression of a Dominant Negative SNARE Domain. A. Outline of the genetic strategy used:
two lines of transgenic mice are made in which the first uses the human GFAP promoter to drive the expression of the tetracycline transactivator
(tTA), the second harbors the tetracycline operator (tetO) driving the expression of the dominant negative SNARE (dnSNARE), and two reporter
proteins, GFP and LacZ. When the two mice are mated, astrocyte specific expression of these transgene is achieved and can be inhibited by including
doxycycline in the mice’s diet. B–E. Astrocyte-specific expression that is suppressed by doxycycline. From Pascual et al. 2005.

Adenosine is known to be regulated during sleep-wake synaptic transmission. Striking diurnal rhythms of adenosine
cycles. In basal forebrain, for example, microdialysis and tone were revealed. When brain slices were isolated at the end
HPLC have revealed that during wakefulness adenosine rises of the dark phase (the animals’ subjective daytime) adenosine
and during sleep adenosine falls (Porkka-Heiskanen et al. tone was high. When isolated during the light phase (subjective
1997). The source of adenosine has been presumed to be neu- night time) adenosine tone was low. Importantly, when mice
ronal. However, given that astrocytes have been demonstrated were sleep deprived during the light phase, they maintained an
to powerfully control extracellular adenosine, the question elevated level of adenosine. When dnSNARE and wild-type
that begs to be answered is whether these sleep wake cycles mice were compared, the dark phase and wakefulness depen-
of adenosine depend on the astrocyte source of adenosine. To dent elevation of adenosine was shown to be derived from an
address this question two levels of experimentation have been astrocytic source. In addition to using synaptic transmission to
performed. First, the authors monitored adenosine in brain monitor extracellular adenosine, enzymatic/amperometric bio-
slices that are isolated at different times of day during the sleep sensors of adenosine were also used that directly measured ade-
cycle (Schmitt et al. 2012). Second, a time of day experiment nosine and validated all of the synaptic bioassay results. These
was performed in vivo in which the astrocytic contribution results provide striking insight into the control of adenosine.
to the neuromodulatory purine pool could be assessed. Brain Wakefulness regulates the extracellular level of adenosine from
slices were isolated at 4-hour intervals throughout the 24-hour an astrocyte-dependent dnSNARE sensitive pathway.
day and the basal adenosine tone was monitored using A1R One concern about these studies is that they were per-
antagonists to relieve the basal presynaptic inhibition of formed in situ. Could they be reproduced in vivo? To ask this

A S T R O C Y T I C M O D U L AT I O N O F M A M M A L I A N SY N A P S E S : C I R C U I T S A N D B E H AVI O R S • 499
A 3 B 2.5 C **
** 100
dn-SNARE
2.5 2 80
wt

Δ fEPSP slope (% of control)


fEPSP slope (-mV/ms)

fEPSP slope (-mV/ms)


dn-SNARE
2 60 wt
1.5
40
1.5
1 20
1
0
0.5 0.5
–20
0 0 –40 D-AP5 Bicuculline RB-2 DPCPX
0 5 10 15 20 25 wt dn-SNARE wt dn-SNARE (n=4) (n=5) (n=4) (n=4)
Stim (V) Dox + + – – –60 Ifenprodil PPADS DPCPX
(n=9) (n=5) (n=6)

Figure 39.4 Astrocytic dnSNARE Expression Attenuates Tonic Adenosine in Hippocampal Slices. A. dnSNARE-derived slices show an increased input/
output relation of CA3-CA1 synaptic transmission. B. This effect is inhibited by doxycycline-mediated suppression of dnSNARE expression. C.
Enhanced synaptic transmission can be explained by an absent suppressive adenosine tone, evident by a blunted impact of A1-R antagonist on synaptic
transmission in the dnSNARE-derived slices. From Pascual et al. 2005.

question local field potential recordings in anesthetized mice promote wakefulness (Basheer et al. 2000). To examine this
were performed. Under urethane anesthesia, the cortex exhib- issue, the dnSNARE mouse was used in EEG and EMG stud-
its slow oscillations of network activity that are modulated by ies of sleep-wake cycles to determine whether their patterns
adenosine. Therefore, slow oscillations were recorded in vivo were perturbed. Initially it was asked whether the amount of
and the magnitude of A1R modulation was determined. Just time spent in the three vigilance states—wake, rapid eye move-
as observed in brain slice studies, it was found that adenos- ment sleep (REM), and non-REM (NREM) sleep—were dif-
ine was controlled by wakefulness and from an astrocytic ferent in dnSNARE mice. Initial results showed no distinction
source. When dnSNARE was expressed in astrocytes, sleep between wild-type and dnSNARE mice (Halassa et al. 2009).
deprivation no longer elevated extracellular adenosine Sleep is controlled by at least two processes: the circadian
(Schmitt et al. 2012). oscillator, which controls the timing of sleep and wakefulness,
Taken together, these results provide a powerful example and the sleep homeostat, which integrates the amount of time
of how behavior regulates via an astrocyte intermediate the one has been awake and delivers the drive to sleep (Borbely and
extracellular level of a gliotransmitter that in turn modulates Achermann 1999). To discriminate between the two a simple
synaptic transmission and neuronal network activity (slow test is performed in which one sleep deprives mice for 4 to 6
oscillations). During wakefulness, astrocytes lead to elevated hours and determines whether there is a homeostatic increase
adenosine, but during sleep this source of gliotransmitter in the delta power of NREM sleep. In dnSNARE mice there
declines to low levels. These results also provide an important is an attenuation of the sleep deprivation–dependent increase
warning to scientists in the field. The time at which the brain in delta power, indicating that the astrocyte modulates the
is studied is an extremely important variable that affects the sleep hemostat (Fig. 39.5A–C). Further support is provided
ability to observe astrocyte-dependent events. Most animal by the observation that the compensatory increase in sleep
facilities have their light phase coincident with our light phase. time that normally follows sleep deprivation is no longer
However, this means that in vivo studies or brain slice experi- present in dnSNARE mice (Fig. 39.5D–G). Using osmotic
ments performed during the light phase (subjective nighttime minipumps to deliver A1R antagonists, it was also found
for mice) will have minimal activation of this gliotransmitter that the dnSNARE phenotype was phenocopied by inhibit-
pathway. Thus, it becomes imperative to record zeitgeber time ing A1R. Consequently, these data indicate that wakefulness
when studies are performed so that clarity can return to a field stimulates the extracellular accumulation of astrocyte-derived
in which there has recently been conflicting results. adenosine, and that this adenosine, acting through A1R, pro-
vides the pressure to sleep that normally follows a day of wake-
fulness (Halassa et al. 2009).
5.2 G L I OT R A N S M I S S I O N MO D U L AT E S
B E H AVI O R—S L E E P H O M E O S TA S I S
5.3 A S T RO C Y T E S A FFEC T T H E CO G N IT I V E
Given that sleep-wake cycles regulate astrocyte-derived ade-
CONSEQUENCES OF SLEEP LOSS
nosine, a natural resulting question is whether this nucleoside
provides a feedback regulation of behavior given that it exerts It is well known that sleep is critical for the consolidation of
powerful modulatory actions on synaptic transmission and long-term memories. Sleep deprivation can prevent memory
neural networks. The first hint that astrocytes might affect formation. Because astrocyte-derived adenosine is necessary
sleep was provided by a review of the literature that showed for compensatory responses to sleep loss (increased sleep time
that adenosine rises during wakefulness and falls during sleep, and NREM bout duration), it is intriguing to ask if the astro-
and that adenosine promotes sleep and adenosine antagonists cyte actively prevents memory consolidation. Therefore, the

500 • FUNCTIONS OF NEUROGLIAL CELLS


A 140 B 160 * Wildtype C 260 Recovery after SD
Wildtype
* dnSNARE Wildtype
dnSNARE

Normalized Power
Normalized Power

Normalized Power
140 * * 220 * * dnSNARE
*

(0.5-1.5 Hz)
(0.5-0.4 Hz)

(0.5-1.5 Hz)
120
* *
120 180

100 140
100

80 100
0 4 8 12 0 4 8 12 6 8 10 12
Time (Zeitgeber) Time (Zeitgeber) Time (Zeitgeber)

Baseline Wildtype Baseline Wildtype


D Recovery E dnSNARE F Recovery G dnSNARE
** * *
60 N.S. 10 300 N.S. 120
(% Recording Time)

(% Recording Time)
** ΔTotal Sleep Time
Total Sleep Time

ΔNREM bouts
NREM bouts

(seconds)
(seconds)
40 200 80
5
20 100 40

0 0 0 0
Wildtype dnSNARE Wildtype dnSNARE

Figure 39.5 Astrocytes Control Sleep Homeostasis. A. dnSNARE mice show an attenuated expression of slow wave activity under baseline conditions.
B. This effect is amplified if the low-frequency component of SWA activity is examined. C. The impact of the dnSNARE on SWA is accentuated by
sleep deprivation. D,E. Although wild-type mice respond to sleep deprivation with an increase in recovery sleep time, dnSNARE mice do not. F,G.
A similar effect is seen when examining the increased NREM bout duration following sleep deprivation. From Halassa et al. 2009.

authors performed studies in which a period of sleep depriva- potentiation (LTP) transitions to a Late-LTP that is protein
tion was interjected between a training session and probe test. synthesis dependent. Late long-term potentiation is thought
Initially, novel object recognition and later spatial object rec- to be a cellular correlate of long-term memory. dnSNARE
ognition was studied with similar results. In the novel object mice and wild-type littermate mice exhibit similar L-LTP.
recognition task mice were allowed to explore two identical However, a striking result is observed when mice are sleep
objects at the beginning of the light phase, and then were deprived before testing L-LTP. Sleep deprivation prevents
allowed to sleep and were re-exposed to objects 24 hours later. the induction of L-LTP unless brain slices are obtained from
However, one of the familiar objects was replaced by a novel dnSNARE mice (Florian et al. 2011) (Fig. 39.6). Moreover,
object. If mice have a recognition memory of the object, they the infusion of the A1R antagonist CPT similarly protects
were exposed to in the training session they will spend more synapses from the negative consequences of sleep deprivation.
time exploring the novel object. Indeed, this was the case in Thus during sleep deprivation the wakefulness dependent
both dnSNARE mice and wild-type littermates. However, if source of astrocyte-derived adenosine acts on A1R to pre-
a 6-hour period of sleep deprivation followed the training ses- vent cellular (L-LTP) and behavioral memory consolidation.
sion, wild-type mice did not show a preferential exploration Because an elevation of cAMP is necessary for memory con-
of the novel object on the next day: They had no recognition solidation and A1R couple through the G protein Gi, which
memory. In contrast, dnSNARE mice had equivalent recogni- negatively regulates adenylyl cyclase, it is presumed that during
tion memory regardless of whether they were left undisturbed enforced wakefulness (sleep deprivation) astrocytes attenuate
or were sleep deprived (Halassa et al. 2009). neuronal elevations of cAMP that are required for memory
Because it is known that dnSNARE prevents the accumu- consolidation (Vecsey et al. 2009).
lation of extracellular adenosine, it was then asked whether
this sleep deprivation–dependent cognitive impairment was
phenocopied by A1R antagonist. In wild-type mice introduc- 6 S U M M A RY A N D P E R S P E C T I VE S
tion of the A1R antagonist CPT prevented the negative con-
sequences of sleep deprivation. Since the first demonstration of gliotransmission in 1994,
How could an astrocyte actively inhibit memory consoli- the field has made considerable progress toward appreciat-
dation? To answer this question use was made of the fact that ing roles that gliotransmitters can play in behavior. During
brain slices maintain the adenosine tone present at the time development gliotransmitters play influential developmen-
the animal is euthanized, and it was asked whether cellular tal roles in synaptic scaling that is important for the for-
long-term memory was affected by prior sleep deprivation. mation of experience-dependent sensory maps. Later in
Tetanic high frequency stimulation (HFS) of the Schaffer col- adulthood, the environment modulates astrocytic structure
laterals leads to long-term potentiation of synaptic transmis- and function. In addition to structural change the astrocyte
sion. When the HFS is repeated four times, this long-term responds to sensory inputs with Ca2+ signals that operate on

A S T R O C Y T I C M O D U L AT I O N O F M A M M A L I A N SY N A P S E S : C I R C U I T S A N D B E H AVI O R S • 501
450 450
NSD SD NSD SD
400 400
fEPSP Slope (% baseline)

fEPSP Slope (% baseline)


350 350
300 300
250 250
200 200
150 150
100 100
WT-NSD (n=5) dnSNARE-NSD (n=6)
50 50
WT-NSD (n=7) dnSNARE-SD (n=6)
0 0
0 25 50 75 100 120 150 0 25 50 75 100 120 150
Time (min) Time (min)

Figure 39.6 Gliotransmission Is Required for the Effect of Sleep Deprivation on the Late Phase of Hippocampal Long-Term Potentiation. Although sleep
deprivation abolishes L-LTP in wild-type slices, it has no impact on this process in slices derived from dnSNARE mice.

the timescale of seconds. On one hand, these signals modu- Bushong EA, Martone ME, Jones YZ, Ellisman MH. 2002. Protoplasmic
late blood flow with the potential to promote anticipatory astrocytes in CA1 stratum radiatum occupy separate anatomical
hemodynamic changes that may impact neural response. On domains. J Neurosci 22:183–192.
Cardin JA, Carlen M, Meletis K, Knoblich U, Zhang F, Deisseroth K,
the other hand, sensory activation of the astrocyte influences et al. 2009. Driving fast-spiking cells induces gamma rhythm and
feedback control of neural networks and behavior. This is controls sensory responses. Nature 459:663–667.
particularly well highlighted by the observation that wake- Desai NS, Cudmore RH, Nelson SB, Turrigiano GG. 2002. Critical
fulness signals to astrocytes to in turn regulate the level of periods for experience-dependent synaptic scaling in visual cortex.
extracellular adenosine. As a consequence of this elevated Nat Neurosci 5:783–789.
Eid T, Ghosh A, Wang Y, Beckstrom H, Zaveri HP, Lee TS, et al. 2008.
adenosine, an astrocyte and adenosine-dependent pressure Recurrent seizures and brain pathology after inhibition of glutamine
to sleep accumulates during the daytime and contributes to synthetase in the hippocampus in rats. Brain 131:2061–2070.
the drive to sleep. Because sleep disorders are comorbid with Fellin T, Halassa MM, Terunuma M, Succol F, Takano H, Frank M,
so many neurological and psychiatric conditions, it will be et al. 2009. Endogenous nonneuronal modulators of synaptic trans-
intriguing to determine whether there exist primary dysfunc- mission control cortical slow oscillations in vivo. Proc Natl Acad Sci
U S A 106:15037–15042.
tions in astrocytes that initiate these disorders of brain func- Florian C, Vecsey CG, Halassa MM, Haydon PG, Abel T. 2011.
tion. Two areas of compelling interest are: major depressive Astrocyte-derived adenosine and A1 receptor activity contribute to
disorder, in which there is a correlated change in astrocyte sleep loss-induced deficits in hippocampal synaptic plasticity and
structure together with sleep dysfunction; and epilepsy, in memory in mice. J Neurosci 31:6956–6962.
which changes in glutamine synthetase expression in astro- Frenkel MY, Bear MF. 2004. How monocular deprivation shifts ocular
dominance in visual cortex of young mice. Neuron 44:917–923.
cytes contribute to reduced inhibition and epilepsy. As one Friedel P, van Hemmen JL. 2008. Inhibition, not excitation, is the key to
looks back on the progress in the field of gliotransmission of multimodal sensory integration. Biol Cybern 98:597–618.
the past 18 years and how its pervasive nature and dominant Genoud C, Quairiaux C, Steiner P, Hirling H, Welker E, Knott GW.
influence on synapses, circuits, and behavior is now appre- 2006. Plasticity of astrocytic coverage and glutamate transporter
ciated, one cannot resist looking forward to what is in the expression in adult mouse cortex. PLoS Biol 4:e343.
Gerics B, Szalay F, Hajos F. 2006. Glial fibrillary acidic protein immuno-
future. Perhaps in the next 18 years the first treatment for a reactivity in the rat suprachiasmatic nucleus: circadian changes and
major brain dysfunction will be through small molecules or their seasonal dependence. J Anat 209:231–237.
biologics targeted to glia! Gordon JA, Cioffi D, Silva AJ, Stryker MP. 1996. Deficient plasticity in
the primary visual cortex of alpha-calcium/calmodulin-dependent
protein kinase II mutant mice. Neuron 17:491–499.
Gordon JA, Stryker MP. 1996. Experience-dependent plasticity of bin-
REFERENCES ocular responses in the primary visual cortex of the mouse. J Neurosci
16:3274–3286.
Araque A. 2006. Astrocyte-neuron signaling in the brain—implications Gourine AV, Kasymov V, Marina N, Tang F, Figueiredo MF, Lane S,
for disease. Curr Opin Investig Drugs 7:619–624. et al. 2010. Astrocytes control breathing through pH-dependent
Basheer R, Porkka-Heiskanen T, Strecker RE, Thakkar MM, McCarley release of ATP. Science 329:571–575.
RW. 2000. Adenosine as a biological signal mediating sleepiness fol- Guthrie PB, Knappenberger J, Segal M, Bennett MV, Charles AC, Kater
lowing prolonged wakefulness. Biol Signals Recept 9:319–327. SB. 1999. ATP released from astrocytes mediates glial calcium waves.
Bazhenov M, Timofeev I, Steriade M, Sejnowski T. 2000. Spiking-bursting J Neurosci 19:520–528.
activity in the thalamic reticular nucleus initiates sequences of spindle Hajos N, Palhalmi J, Mann EO, Nemeth B, Paulsen O, Freund TF. 2004.
oscillations in thalamic networks. J Neurophysiol 84:1076–1087. Spike timing of distinct types of GABAergic interneuron during hip-
Borbely AA, Achermann P. 1999. Sleep homeostasis and models of sleep pocampal gamma oscillations in vitro. J Neurosci 24:9127–9137.
regulation. J Biol Rhythms 14:557–568. Halassa MM, Fellin T, Haydon PG. 2007a. The tripartite synapse:
Buonomano DV, Merzenich MM. 1998. Cortical plasticity: from syn- roles for gliotransmission in health and disease. Trends Mol Med
apses to maps. Annu Rev Neurosci 21:149–186. 13:54–63.

502 • FUNCTIONS OF NEUROGLIAL CELLS


Halassa MM, Fellin T, Takano H, Dong JH, Haydon PG. 2007b. Petzold GC, Albeanu DF, Sato TF, Murthy VN. 2008. Coupling of neu-
Synaptic islands defined by the territory of a single astrocyte. ral activity to blood flow in olfactory glomeruli is mediated by astro-
J Neurosci 27:6473–6477. cytic pathways. Neuron 58:897–910.
Halassa MM, Florian C, Fellin T, Munoz JR, Lee SY, Abel T, et al. 2009. Porkka-Heiskanen T, Strecker RE, Thakkar M, Bjorkum AA, Greene
Astrocytic modulation of sleep homeostasis and cognitive conse- RW, McCarley RW. 1997. Adenosine: a mediator of the sleep-inducing
quences of sleep loss. Neuron 61:213–219. effects of prolonged wakefulness. Science 276:1265–1268.
Haydon PG. 2001. GLIA: listening and talking to the synapse. Nat Rev Rothstein JD, Martin L, Levey AI, Dykes-Hoberg M, Jin L, Wu D, et al.
Neurosci 2:185–193. 1994. Localization of neuronal and glial glutamate transporters.
Heller RA, Kronke M. 1994. Tumor necrosis factor receptor-mediated Neuron 13:713–725.
signaling pathways. J Cell Biol 126:5–9. Sanacora G, Mason GF, Rothman DL, Behar KL, Hyder F, Petroff OA,
Higley MJ, Contreras D. 2006. Balanced excitation and inhibition et al. 1999. Reduced cortical gamma-aminobutyric acid levels in
determine spike timing during frequency adaptation. J Neurosci depressed patients determined by proton magnetic resonance spec-
26:448–457. troscopy. Arch Gen Psychiatry 56:1043–1047.
Hinterkeuser S, Schroder W, Hager G, Seifert G, Blumcke I, Elger CE, Sattler R, Rothstein JD. 2006. Regulation and dysregulation of gluta-
et al. 2000. Astrocytes in the hippocampus of patients with tempo- mate transporters. Handb Exp Pharmacol:277–303.
ral lobe epilepsy display changes in potassium conductances. Eur J Scales SJ, Bock JB, Scheller RH. 2000a. The specifics of membrane
Neurosci 12:2087–2096. fusion. Nature 407:144–146.
Jansen LA, Uhlmann EJ, Crino PB, Gutmann DH, Wong M. 2005. Scales SJ, Chen YA, Yoo BY, Patel SM, Doung YC, Scheller RH. 2000b.
Epileptogenesis and reduced inward rectifier potassium current SNAREs contribute to the specificity of membrane fusion. Neuron
in tuberous sclerosis complex-1-deficient astrocytes. Epilepsia 26:457–464.
46:1871–1880. Schmitt Sims RE, Dale N, Haydon PG. 2012. Wakefulness affects synap-
Kaneko M, Stellwagen D, Malenka RC, Stryker MP. 2008. Tumor tic and network activity by increasing extracellular astrocyte-derived
necrosis factor-alpha mediates one component of competitive, adenosine. J Neurosci 32:4417–4425.
experience-dependent plasticity in developing visual cortex. Neuron Schousboe A, Larsson OM, Frandsen A, Belhage B, Pasantes-Morales H,
58:673–680. Krogsgaard-Larsen P. 1991. Neuromodulatory actions of glutamate,
Kettenmann H, Faissner A, Trotter J. 1996. Neuron-glia interactions GABA and taurine: regulatory role of astrocytes. Adv Exp Med Biol
in homeostasis and degeneration. In: Greger R, Windhorst U (eds.), 296:165–180.
Human physiology. Heidelberg: Springer-Verlag, pp. 533–543. Schousboe A, Westergaard N, Sonnewald U, Petersen SB, Yu AC,
Lavialle M, Aumann G, Anlauf E, Prols F, Arpin M, Derouiche A. 2011. Hertz L. 1992. Regulatory role of astrocytes for neuronal biosyn-
Structural plasticity of perisynaptic astrocyte processes involves ezrin thesis and homeostasis of glutamate and GABA. Prog Brain Res
and metabotropic glutamate receptors. Proc Natl Acad Sci U S A 94:199–211.
108:12915–12919. Schummers J, Yu H, Sur M. 2008. Tuned responses of astrocytes and
Liang SL, Carlson GC, Coulter DA. 2006. Dynamic regulation of syn- their influence on hemodynamic signals in the visual cortex. Science
aptic GABA release by the glutamate-glutamine cycle in hippocam- 320:1638–1643.
pal area CA1. J Neurosci 26:8537–8548. Shigetomi E, Tong X, Kwan KY, Corey DP, Khakh BS. 2011. TRPA1
Liu BH, Li P, Sun YJ, Li YT, Zhang LI, Tao HW. 2011. Intervening inhi- channels regulate astrocyte resting calcium and inhibitory synapse
bition underlies simple-cell receptive field structure in visual cortex. efficacy through GAT-3. Nat Neurosci 15:70–80.
Nat Neurosci 13:89–96. Sohal VS, Zhang F, Yizhar O, Deisseroth K. 2009. Parvalbumin neurons
Mishra AM, Ellens DJ, Schridde U, Motelow JE, Purcaro MJ, and gamma rhythms enhance cortical circuit performance. Nature
DeSalvo MN, et al. 2011. Where fMRI and electrophysiology agree 459:698–702.
to disagree: corticothalamic and striatal activity patterns in the Steinhauser C, Seifert G. 2002. Glial membrane channels and receptors
WAG/Rij rat. J Neurosci 31:15053–15064. in epilepsy: impact for generation and spread of seizure activity. Eur J
Moore CI, Cao R. 2008. The hemo-neural hypothesis: on the role of blood Pharmacol 447:227–237.
flow in information processing. J Neurophysiol 99:2035–2047. Stellwagen D, Malenka RC. 2006. Synaptic scaling mediated by glial
Oliet SH, Panatier A, Piet R, Mothet JP, Poulain DA, Theodosis DT. TNF-alpha. Nature 440:1054–1059.
2008. Neuron-glia interactions in the rat supraoptic nucleus. Prog Swadlow HA. 2003. Fast-spike interneurons and feedforward inhibition
Brain Res 170:109–117. in awake sensory neocortex. Cereb Cortex 13:25–32.
Oliet SH, Piet R, Poulain DA. 2001. Control of glutamate clearance and Uhlhaas PJ, Singer W. 2006. Neural synchrony in brain disorders: rel-
synaptic efficacy by glial coverage of neurons. Science 292: 923–926. evance for cognitive dysfunctions and pathophysiology. Neuron
Olsen ML, Sontheimer H. 2008. Functional implications for Kir4.1 52:155–168.
channels in glial biology: from K+ buffering to cell differentiation. Uhlhaas PJ, Singer W. 2007. What do disturbances in neural synchrony
J Neurochem 107:589–601. tell us about autism? Biol Psychiatry 62:190–191.
Ongur D, Drevets WC, Price JL. 1998. Glial reduction in the subgen- Uhlhaas PJ, Haenschel C, Nikolic D, Singer W. 2008. The role of oscil-
ual prefrontal cortex in mood disorders. Proc Natl Acad Sci U S A lations and synchrony in cortical networks and their putative rel-
95:13290–13295. evance for the pathophysiology of schizophrenia. Schizophr Bull 34:
Ortinski PI, Dong J, Mungenast A, Yue C, Takano H, Watson DJ, et al. 927–943.
2011. Selective induction of astrocytic gliosis generates deficits in Vecsey CG, Baillie GS, Jaganath D, Havekes R, Daniels A, Wimmer M,
neuronal inhibition. Nat Neurosci 13:584–591. et al. 2009. Sleep deprivation impairs cAMP signalling in the hip-
Panatier A, Oliet SH. 2006. Neuron-glia interactions in the hypothala- pocampus. Nature 461:1122–1125.
mus. Neuron Glia Biol 2:51–58. Wang X, Lou N, Xu Q, Tian GF, Peng WG, Han X, et al. 2006. Astrocytic
Parpura V, Basarsky TA, Liu F, Jeftinija K, Jeftinija S, Haydon PG. Ca2+ signaling evoked by sensory stimulation in vivo. Nat Neurosci
1994. Glutamate-mediated astrocyte-neuron signalling. Nature 9:816–823.
369:744–747. Wang Y, Zaveri HP, Lee TS, Eid T. 2009. The development of recurrent
Pascual O, Casper KB, Kubera C, Zhang J, Revilla-Sanchez R, Sul JY, seizures after continuous intrahippocampal infusion of methionine
et al. 2005. Astrocytic purinergic signaling coordinates synaptic net- sulfoximine in rats: a video-intracranial electroencephalographic
works. Science 310:113–116. study. Exp Neurol 220:293–302.

A S T R O C Y T I C M O D U L AT I O N O F M A M M A L I A N SY N A P S E S : C I R C U I T S A N D B E H AVI O R S • 503
40.
ADULT NEUROGENESIS
Gerd Kempermann

A B B R E VI AT I O N S process that is regulated at numerous levels (Kempermann


2011d). As such it allows particularly sensitive adaptations,
BMP bone morphogenic protein but adult neurogenesis comes at high cost if the regulation is
DCX doublecortin going to be specific. In many “lower” animals in contrast, adult
GFAP glial fibrillary acidic protein neurogenesis might be widespread and not specific to particu-
LTP long-term potentiation lar functional and structural contexts.
NG2 neuron-glia 2 (proteoglycan) Neurogenesis, like all development, originates from stem
RGL radial glia-like (cells) cells, which are defined as cells with the ability to self-renew.
SEZ subependymal zone Dividing stem cells always produce one (in an asymmetrical
SGZ subgranular zone division) or two (in a symmetrical division) new stem cells.
shh sonic hedgehog Whereas symmetrical divisions allow expansion of the stem
SVZ subventricular zone cell pool, only asymmetrical divisions generate differentiating
progeny, while still maintaining the stem cell pool. The differ-
entiating daughter cell of a stem cell is called a progenitor cell,
1 I N T R O D U C T I O N : W H AT I S whose ability to self-renew is limited, and that may undergo
A D U LT N E U R O G E N E S I S ? a terminal “neurogenic symmetrical division” in which both
progeny are differentiating cells and the precursor cell is thus
Adult neurogenesis is the developmental process of generating consumed. Although stem cells are usually rare, progenitor
new neurons under the conditions of the adult brain. Adult cells are numerous and highly reactive to regulation. Expansion
usually stands for sexually mature, but when exactly and how can occur at the progenitor cell stage. The term precursor cell
postnatal neurogenesis transgresses into adult neurogenesis is used here as an umbrella term for stem and progenitor cells
remains a matter of discussion (Rakic 2002a). Despite many as well as neuroblasts (an ill-defined and problematic category
similarities, adult neurogenesis differs from fetal and postna- of more advanced progenitor cells), if details are not known or
tal neurogenesis in a number of issues. Significantly, although all cells are meant. Strictly speaking, the term stem cell should
the early forms of neurogenesis are primarily dependent on be reserved for cases in which self-renewal and multipotency
genetic programs, adult neurogenesis is characterized by mas- have been demonstrated, but the common use of the term is
sive regulatory influence of extrinsic factors, whether physio- much looser, and this has infiltrated the scientific literature.
logical activity or pathology. Adult neurogenesis is a particular Besides self-renewal, stem cells are defined by their ability
form of brain plasticity in which structural and functional to differentiate along at least two different lineages (known as
adaptation to functional demands occurs at the level of cells multipotency). Different stem cells differ in the width of their
rather than neurites and synapses. This is an exceptional situa- potential to produce differentiated cells. Throughout this
tion and in adult rodents and primates is essentially limited to book (see chapters 5 and 30), slightly different nomenclatures
two “canonical” neurogenic regions, the subventricular zone for stem and progenitor cells are used, which reflects the state
(SVZ) of the lateral ventricles that produces several types of of the different subfields of neurogenesis research. The termi-
interneurons in the olfactory bulb, and the subgranular zone nology used here is widely used but not commonly accepted
(SGZ) of the dentate gyrus in the hippocampus, in which new and many details remain unresolved.
excitatory granule cells are generated (Fig. 40.1) (Kempermann
2011a). Other species have more or fewer neurogenic zones.
Zebrafish, for example, have 16 (Grandel et al. 2006; Kaslin 2 R E L AT I O N S H I P W I T H F ETA L A N D
et al. 2008), drosophila none (Siegrist et al. 2010), most bats E A R LY P O S T N ATA L N E U R O G E N E S I S
only one in the olfactory system (Amrein et al. 2007), and in
humans neurogenesis in the olfactory bulb appears to dry out Among the key similarities between embryonic and adult
in late childhood or adolescence (Curtis et al. 2007; Sanai et al. neurogenesis, the fact stands out that neurogenesis is a glial
2011), whereas neurogenesis in the adult human hippocampus process to a large degree (Morrens et al. 2012). Radial glial
is maintained into old age (Eriksson et al. 1998; Knoth et al. cells serve as stem cells both in the embryo and the adult
2010). Adult neurogenesis is an extremely complex, polygenic (Chojnacki et al. 2009; Doetsch et al. 1999a; Kriegstein and

504
Subgranular zone (SGZ)
of the hippocampal dentate gyrus
Immature
neuron
Type-3 Immature
neuronally
and
determined
neuroblasts mature,
new
granule cell
Type-1
radial glia-like Type-2b postmitotic
stem cells Type-2a neuronally maturation stages
glia-like determined CA3
in the granule cell
intermediate progenitor cells
Glia-like progenitor cells layer
Neuronal
precursor cells
precursor cells
in the SGZ in the SGZ

3.

1E
leehwniP
1E lacipa :buh
1E sessecorp
2.
Ventricle

sllec 1B fo
1E B1
1E
B1
New neurons in the
B1 granule cell layer and
1.
Migratory neuroblasts periglomerular zones
eht no elcirtnev eht morf weiV
llaw ralucirtnev eht fo ecafrus Precursor cells (A cells) of the olfactory bulb
B1 cell C cell A cell in the SVZ in the rostral migratory stream

E1
E1
Subventricular zone (SVZ) / Olfactory bulb
Pinwheel pattern
(at ventricular surface)

Figure 40.1 The Neurogenic Regions of the Adult Rodent Brain. Most mammals have two “canonical” neurogenic regions, the dentate gyrus in the
hippocampus and the subventricular zone (SVZ)/olfactory bulb, sharing certain features of neurogenesis.

Alvarez-Buylla 2009; Noctor et al. 2001). However, several Neurogenesis in the adult SVZ and olfactory bulb, in
definitional and semantic issues arise if it comes to describing contrast, essentially follows a ventral pattern and produces
radial glia in a way that satisfies the particular demands of the interneurons and oligodendrocytes. Here, neurogenesis is in
conditions of both the fetal and the adult brain. Therefore, continuation of embryonic neurogenesis originating from
the stem cells of the adult brain are often referred to as radial the ganglionic eminences. Because of the different types of
glia-like (Bonaguidi et al. 2011). The alternative has been a interneurons that are produced, the pattern of transcription
neutral nomenclature with letters in the SVZ (Doetsch et al. factors is less uniform. There is even one lineage showing the
1997) and numbers in the SGZ (Kempermann et al. 2004). dorsal sequence from Pax6 to Tbr1 intermingled with the
Adult hippocampal neurogenesis is a process in the dorsal otherwise ventral process (Brill et al. 2009).
brain; therefore, the same or similar patterns of transcription fac- Despite the many similarities, adult neurogenesis is also
tors and regulatory principles that govern dorsal neurogenesis in fundamentally different from embryonic and fetal neurogen-
the embryo also appear in adult hippocampal neurogenesis. For esis in that it represents an exception in an environment that
example, there is a signature sequence of transcription factors is otherwise fully developed and has turned nonneurogenic.
from Pax6 in the stem cells over Neurogenin2 and Tbr2 in the Other than in the fetal brain, adult neurogenesis is also not
intermediates to Tbr1 in the neurons that is shared by fetal cor- as much synchronized as it is during embryonic brain devel-
tical neurogenesis and adult hippocampal neurogenesis (Hodge opment, but all stages of development are found next to each
et al. 2008). Excitatory neurons and astrocytes are generated. other at any given time (Fig. 40.2).

A D U LT N E U R O G E N E S I S • 505
Figure 40.2 Adult-Generated Neurons and Glia in the Granule Cells Layer (GCL) of the Murine Dentate Gyrus. Within each panel the antigens
investigated are listed in the order red, green, and blue. A,B. Two types of astrocytes can be distinguished in the granule cell layer of the adult dentate
gyrus. One type has a radial or vertical morphology and expresses GFAP (blue) and Nestin (Nestin-GFP, green) and represents the radial glia-like stem
cell (type-1 cells; left panel in B). The second type is more horizontally oriented and expresses GFAP (blue) together with S100β (red), is postmitotic
and does not have precursor cell functions. Scale bar (in M for all images) 40 μm for (A). C. Four hours after a single BrdU injection newly generated
BrdU-positive cells (green) in the SGZ express doublecortin (DCX), which here identifies neuroblasts and immature postmitotic neurons (blue). Scale
bar 25 μm. D. One week after BrdU an intermediate stage of neuronal maturation in the SGZ is detected in most newborn neurons. Newly generated
BrdU-positive cells (green) coexpress DCX (blue) and postmitotic neuronal marker NeuN (red). Scale bar 30 μm. E. DCX (blue) expressing “neu-
roblast” (type-3 cells) in the SGZ co-express Ki67 (green) a marker detecting cells during active cell cycle. Scale bar 25 μm. F. Four weeks after BrdU
almost all BrdU-positive cells (green) coexpressed the mature neuronal marker NeuN (red) and have lost DCX expression (blue). Scale bar 60 μm. G.
Four hours after BrdU, a subset of the labeled cells (red) coexpress GFAP (blue), indicating the proliferative activity of the stem cells. Scale bar 25 μm.
H. Three days after BrdU GFAP-positive (blue) cells coexpressed BrdU (red) and Ki67 (green), indicating these cells were still mitotically active in this
phase of maturation or have re-entered the cell cycle. Scale bar 25 μm. I. Four weeks after BrdU almost all GFAP-positive (blue)/BrdU-positive (red)
cells coexpressed S100β (green), indicating these cells were classical astrocytes in terms of antigen expression. Scale bar 25 μm. K. At all investigated
time points newly generated BrdU-positive (red) ramified microglia cells expressing Iba-1 (blue) were detected in the SGZ. Scale bar 40 μm.
L. NG2 cells (green) were detected in the SGZ and hilus. Only the cells in the hilus incorporated BrdU. No BrdU-positive (red) NG2-positive cells
were found in the SGZ. Scale bar 30 μm. M. CNPase-positive (green) mature oligodendrocytes did not incorporate BrdU (red) at any investigated
time point. Scale bar 30 μm. Reprinted from Steiner et al. 2006.

3 A S T R O C Y T E -L I K E O R R A D I A L et al. 2003). The stem cells that are found in the adult SVZ
G L I A-L I K E S T E M C E L L S ? and SGZ share many glial properties besides GFAP expres-
sion (see Fig. 40.2A,B). Their basic morphology and marker
Accordingly, the fact justifying that adult neurogenesis profile (GLAST, BLBP, Pax6) is highly similar to radial glia
is featured in a book on neuroglia is that a population of in the developing brain, even though they do not span out
astrocyte- or radial glia-like cells produces the new neurons between the ventricular and pial surface and do not serve as
of the adult brain. On the basis of GFAP expression, stem guidance structure to radially migrating newborn neurons
cells can be isolated from the brain and genetic ablation of (Merkle et al. 2004). In the hippocampus, they also have lost
GFAP-expressing cells eliminated adult neurogenesis (Garcia contact with the ventricular surface, whereas in the SVZ their
et al. 2004; Giachino et al. 2005; Imura et al. 2003; Morshead apical process reaches the ventricular lumen through a central

506 • NEUROGLIA
opening of ependymal cells that is ordered in a rosette-like different contributions to niche functionality, maintenance of
arrangement (Mirzadeh et al. 2008). In the SGZ they are the precursor cells, and regulation of adult neurogenesis.
referred to as type-1 cells or radial glia-like (RGL) stem cells; Astrocytes play a dual role, as stem cells and niche cells,
in the SVZ as B cells. In analogy to the role of radial glia dur- and at present it is not fully clear whether the astrocytes per-
ing cortical development (see chapters 5 and 30) they serve as forming these tasks are always identical or subpopulations
the stem cell of the two neurogenic zones. Consequently, they exist (Fig. 40.3). In the SGZ a population of nonproliferating,
express stem cell markers such as Sox2 and Nestin (Komitova S100E-positive astrocytes exists (and is produced in the course
and Eriksson 2004; Steiner et al. 2006; Suh et al. 2007). In of adult hippocampal neurogenesis) (Steiner et al. 2004),
the SGZ they generate one lineage of granule cells plus which has a “horizontal” morphology, as opposed to the radial
astrocytes (Fig. 40.2C–F), in the SVZ there are at least appearance of stem cells (Seri et al. 2001). How much they
seven (probably more) distinct types of interneurons in the contribute to niche functions is not quite clear. Niche astro-
olfactory bulb (Lledo et al. 2008). A separate lineage, prob- cytes from neurogenic regions are different from astrocytes
ably originating from a specific population of B cells is respon- in nonneurogenic zones (Song et al. 2002), which supports
sible for producing NG2 cells and oligodendrocytes. These do the idea that the niche provides a critical neurogenic permis-
not migrate to the olfactory bulb as do the neurons, but below siveness. Astrocytes exert numerous niche functions (besides
the corpus callosum and into the cortex. The radial glia- their role as precursor cells) in that they are a main source of
like stem cells have low proliferative activity but produce secreted factors. Despite concrete evidence for a number of
fast dividing intermediate progenitor cells, referred to as factors and plausible hypotheses for several others (Morrens
type-2 cells in the hippocampus and C cells in the SVZ. et al. 2012), it is still unresolved what exactly renders a neuro-
At the type-2 cell stage, the progenitor cells go through a genic region neurogenic.
transition from glial to early neuronal marker expression. Key examples of soluble niche signals are the paracrine fac-
A characteristic marker that is expressed at this stage is tors Notch, Wnt, Shh, and BMP. All of these have well-studied
doublecortin (DCX) (Fig. 40.2C–F), a cytoskeleton-associated functions in embryonic neurogenesis and play similar but
protein involved in migration and neurite extension adjusted roles in adult neurogenesis (Bonaguidi et al. 2005;
(Brown et al. 2003; Couillard-Despres et al. 2005; Rao and Breunig et al. 2008; Lie et al. 2005; Lim et al. 2000; Lugert
Shetty 2004). et al. 2010; Palma et al. 2005). By and large, they are all
Quiescent subpopulations exist at the stem cell level that involved in maintaining the stem cell pool and balancing this
can be recruited into the process of neurogenesis. Studies maintenance function with effects on later stages of develop-
using different methods to study the mode of division of the ment. We still know little about how the four factors interact
stem cells in the hippocampus came to opposing conclusions and how far their signaling is combinatorial, but much para-
(Bonaguidi et al. 2011; Encinas et al. 2011). Possibly, in the crine signaling originates from astrocytes, supporting their
absence of activating stimuli, the stem cells exhaust them- role as key orchestrators.
selves, ultimately turning into astrocytes (Encinas et al. 2011).
However, both symmetrical and asymmetrical divisions seem
principally possible (Bonaguidi et al. 2011), so that activating 5 OT H E R G L I A L C O N T R I B U T I O N S
situations are conceivable, in which exhaustion is prevented TO A D U LT N E U R O G E N E S I S
(Kempermann 2011c).
Glial cells play numerous roles related to adult neurogen-
esis (Fig. 40.4). Microglia are prime candidates as source of
4 ASTROCY TES AS NICHE CELLS cytokine signaling and mediators of immune effects on adult
neurogenesis. However, astrocytes have certain immunologi-
The stem cell niche is the functional unit of the stem cells and cal functions, too, so that the precise individual contributions
their microenvironment. Stem cell cultures have to provide have not yet been identified. Microglia have a double-edged
the particular niche factors and often still do so insufficiently. influence on adult neurogenesis in that microglia might both
Cell culture conditions are biased toward secreted humoral support and damage adult neurogenesis, presumably depend-
factors and extracellular matrix, but generally do not faithfully ing on their state of activation. Evidence for this dual role,
represent the cellular composition of the niche. The so-called however, so far largely comes from studies on ischemia-in-
neurospheres agglomerate cultures of neural precursor cells duced reactive neurogenesis in the striatum. An initial inflam-
in which spontaneous clusters of cells develop, and probably matory response directly following the injury downregulated
represent intrinsic attempts for self-organizing niches that in precursor cell activity in the SVZ, but was prevented when
this case drive the cells toward differentiation (Reynolds and microglia were inhibited with the drug minocycline (Ekdahl
Rietze 2005). Monolayer cultures are more homogenous, et al. 2003). At the same time, the very low but long-lasting
but are essentially devoid of cell–cell contacts, which are an neurogenic response in the striatum also seems to be a con-
important component of the niche. sequence of the immunological response (Ekdahl et al. 2003;
Niche cells comprise the different precursor cells, endothe- Thored et al. 2009). Microglia in physiological neurogenesis
lial cells, astrocytes, and microglia (Fig. 40.2K). In addition, have been far less studied, but it is assumed that even in a
the SVZ niche also contains ependymal cells, whereas in the resting state they contribute to niche effects, for example, by
SGZ neurons are also found. The different niche cells make phagocytosis of cells produced in excess (Sierra et al. 2010).

A D U LT N E U R O G E N E S I S • 507
astrocyte-like
precursor cell other astrocyte

generates

NICHE
• cell-cell contact
influences • paracrine/autocrine (short-range)
• modulation of ambient
neurotransmitters (mid-range)
generates • growth factors, hormones,
cytokines (long-range)

Figure 40.3 Astrocytes and the Concept of the Stem Cell Niche. Astrocytes play a particular role in the “stem cell niches” of the adult brain.
Astrocyte-like precursor cells give rise to new neurons and produce other astrocytes without precursor cell functions (see Fig. 40.1). Both types of
astrocytes are constituents of the niche, but their relative contribution has not yet been elucidated. The depicted morphologies reflect the situation
in the hippocampus. Reprinted from Morrens et al. 2012.

6 N E U R O G E N E S I S I N T H E A D U LT number (at least seven) of different populations of interneu-


S U BVE N T R I C U L A R Z O N E A N D rons in the olfactory bulb (Merkle et al. 2007). These are func-
O L FAC TO RY B U L B tionally distinct, and it is unclear whether specific functional
demand might trigger the production of the subtypes.
In neurogenesis of the adult SVZ and olfactory bulb, neuronal
development is spread out over a long distance between the
ventricular wall and periphery of the olfactory bulb, and hence 7 N E U R O G E N E S I S I N T H E A D U LT
involves extensive migration (see Fig. 40.1). Proliferation HIPPOCAMPUS
of SVZ precursor cells is much more prominent than in the
hippocampus. Adult hippocampal neurogenesis differs from neurogenesis
The radial glia-like stem cells (B cells) give rise to tran- in the adult olfactory system in numerous ways: Only one
siently amplifying C cells, which in turn generate the migra- type of neuron is produced and this is an excitatory principal
tory neuroblasts (A cells), which embark on the long journey cell, not an interneuron. The entire process of hippocampal
to the olfactory bulb (Doetsch et al. 1999a,b). For this migra- neurogenesis takes place in a very confined space, with migra-
tion through the rostral migratory stream, they use a partic- tion from the SGZ into the granule cell layer amounting
ular type of migration, called chain migration, in which the to a few hundred microns at most. The new neurons in the
migrating cells use each other as guidance structures (Lois hippocampus feature a very elaborate morphology with a
and Alvarez-Buylla 1994). The rostral migratory stream, at large dendritic tree and a long axon that becomes part of
least in rodents, is lined by a tube of astrocytes, which serve the mossy fiber projection between the dentate gyrus and
as a guidance structure to the migratory neuroblasts and also CA3 region. The mossy fiber tract shows an amazing degree
have precursor cell functions. New migratory A cells and of plasticity, and adult neurogenesis explains part of this
thus new neurons can arise from the stem cells of the RMS as adaptiveness.
well (Gritti et al. 2002). However, migration can also occur In the SGZ, the radial glia-like stem cells (type-1 cells)
without this glial tube ( Jankovski et al. 1998). Blood vessels have many astrocytic features (Filippov et al. 2003; Seri et al.
and possible the obliterated olfactory ventricles seem to be 2001) and give rise to transient amplifying type-2 cells, which
involved in route finding (Whitman et al. 2009). In the bulb, are highly proliferative and respond to many external regula-
the cells switch to a radial mode of migration and reach their tory influences (Kronenberg et al. 2003) (see Fig. 40.1). At the
final destination in the granule cell layer of the periglomerular level of type-2 cells, a transition from a glia-like (type-2a) to an
zones. early neuronal phenotype (type-2b) takes place (Steiner et al.
Precursor cells in the SVZ are subregionalized, and differ- 2006). The following “neuroblast” stage is referred to as type-3.
ent parts of the walls of the lateral ventricles give rise to a large Type-3 cells might still proliferate but their rate of proliferation

508 • NEUROGLIA
Subgranular zone Subventricular zone

Precursor cells
• self-renewal, expansion
• gap-junctional coupling
• guidance structure
Type-1 • auto- and paracrine signals
• ECM production B1 cell
Type-2a
Type-2b C cell
Type-3 A cell
Non-precursor cell
astrocytes
• modulation of
neurotransmitter effects B2 cell
• ECM production
Horizontal astrocyte • secretion of trophic factors Other astrocyte (?)
• paracrine signals

NG2 cells
• modulation of
neurotransmitter effects (?) NG2+ C cell

Microglia
• cytokine signaling
• phagocytosis
Microglial cell Ependyma Microglial cell
• paracrine and
juxtacrine signaling (?)
• back-up precursor cell
function Ependymal cell

Vasculature
• secretion of trophic factors
Capillaries • general nutrient supply Capillaries
• guidance structure (not in SVZ proper but
reached by B cell processes)

Neurons
• secretion of trophic factors
• ambient (and synaptic)
neurotransmitters
• guidance structure
Granule cells
and No neurons in the
interneurons SVZ proper

Figure 40.4 The Cellular Components of the Neurogenic Niches. Reprinted from Morrens et al. 2012.

is much lower than at the type-2 level. Type-3 cells also migrate 8 R E GU L AT I O N O F A D U LT
their short distance into the granule cell layer (but the major- NEUROGENESIS
ity stays in the lower third), and at this stage also the first
neurites might appear. After exit from the cell cycle, a wave Adult neurogenesis responds to a very large number of factors
of cell death occurs while the new neurons mature. Within and stimuli at diverse conceptual levels, from behavioral down
about 10 days (in rodents) the axon has reached CA3 (Hastings to genetic and epigenetic (see discussion in Kempermann
and Gould 1999; Stanfield and Trice 1988), where the new 2011d). This hierarchy is immensely complex. Because adult
terminals compete with existing synapses at the mossy neurogenesis is a process involving many stages, regulation is
fiber boutons, the characteristic input structures to CA3 also spread out over time. In the course of development, late
(Toni et al. 2008). In the following period, the new neurons regulatory events build on the preceding events.
go through a phase of increased synaptic plasticity (Garthe Importantly, neurogenesis in the adult hippocampus is
et al. 2009; Saxe et al. 2006; Schmidt-Hieber et al. 2004; strongly regulated by behavioral activity and learning (Gould
Snyder et al. 2001), which not only results in a particular et al. 1999a; Kee et al. 2007; Kronenberg et al. 2003; Tashiro
functionality during this time, but is also instrumental for et al. 2007). This aspect seems less prominent in the olfactory
their ultimate recruitment and lasting integration into the bulb (Lledo et al. 2006; Rochefort et al. 2002), but in both
network. situations adult neurogenesis is part of adaptive plastic events.

A D U LT N E U R O G E N E S I S • 509
Adult hippocampal neurogenesis appears to be the brain’s way memory has been proposed, but this is likely to be a down-
to cope with novelty and complexity in learning experiences stream effect of some functionality in the olfactory bulb itself.
and is a way to master the tradeoff between stability and flex- Adult-generated olfactory interneurons might help to opti-
ibility in a neuronal network that is supposed to learn. In this mize the network (Lledo and Lagier 2006), also with respect
sense, regulation of neurogenesis and its function are closely to the challenge that occurs, because owing to massive lifelong
linked. neurogenesis in the olfactory epithelium that constantly alters
At the cellular level the regulatory factors converge on the the input to the olfactory bulb.
niche. Cell–cell contacts, extracellular matrix, and humoral
factors make up a complex machinery. A large number of
secreted molecules including autocrine and paracrine signals, 10 N E U R O G E N E S I S O U T S I D E T H E
growth and neurotrophic factors, and neurotransmitters have NEUROGENIC REGIONS OF THE
been studied and yield the picture of a process that is sensitive A D U LT B R A I N ?
to a broad range of extrinsic stimuli and builds on the integra-
tion of numerous intrinsic signaling pathways. Numerous studies have dealt with the question, whether new
neurons might also be constitutively found in additional
regions of the rodent (and primate) brain. Adult cortical neu-
9 F U N C T I O N A L R E L E VA N C E O F rogenesis received most attention and controversy (Gould
A D U LT N E U R O G E N E S I S et al. 1999b; Rakic 2002b). Examples of the two imaginable
cases of neurogenesis originating from the SVZ and local
In the hippocampus, the new neurons contribute to basic resident precursor cells within the cortical parenchyma have
functionality of the dentate gyrus. All detectable enhanced been reported (Dayer et al. 2005; Gould et al. 1999b; Ohira
postsynaptic potentials (long-term potentiation) originate et al. 2010). But ultimately most claims about constitutive
from the newborn neurons, which go through a postmitotic adult cortical neurogenesis have not withstood the most rig-
period of enhanced synaptic plasticity. Mature granule cells orous testing or at least still await independent confirmation.
are heavily inhibited by the local interneurons. If one elimi- Mostly methodological issues caused fundamental concern
nates adult neurogenesis, dentate gyrus LTP disappears, unless (Nowakowski and Hayes 2000; Rakic 2002b). One exception
one also blocks the inhibition (Garthe et al. 2009; Saxe et al. is the case of an extremely low level of neurogenesis in corti-
2006). Then, a strong LTP from the mature cells can be seen. cal layer VI at least in young-adult animals (Dayer et al. 2005;
This plasticity provided by the new neurons seems to be Inta et al. 2008), but the numbers are extremely low. The case is
important for the functions of the dentate gyrus, especially different for reactive neurogenesis in the case of ischemia and
pattern separation (Aimone et al. 2006; Clelland et al. 2009) after boosting with growth factor infusions. The best known
and the avoidance of catastrophic interference (Wiskott et al. example is neurogenesis in a very precise neuronal ablation
2006). Pattern separation is the ability to memorize two stim- model, in which no inflammatory response is evoked (Magavi
uli as distinct and is a cardinal feature underlying higher-or- et al. 2000). However, for region CA1 of the hippocampus a
der cognitive functions. New neurons, one theory goes, are case of massive regenerative neurogenesis after ischemia and
involved in providing a “time-stamp” to incoming information growth factor infusion has been claimed but never been con-
that needs to be learned (Aimone et al. 2009). Related to this firmed and substantial questions about the proposed mecha-
is the finding that adult-born neurons are required to form the nism remain (Nakatomi et al. 2002). Nevertheless, it cannot
association between a shock stimulus and the environment, in be excluded that pathological situations might uncover a neu-
which this shock was applied (contextual fear conditioning) rogenic potential outside the “canonical” neurogenic regions.
(Imayoshi et al. 2008). This function also bridges between the Possibly many examples of “neurogenesis” actually repre-
cognitive and affective functions of the hippocampus (Revest sent signs of neuronal differentiation in NG2 cells (see chapter 10).
et al. 2009). The hypothesis that adult neurogenesis might play The problem is that, interestingly, neuronal markers can also
a role in the development and maintenance of major depres- appear in NG2-expressing glia, even though these cells never
sion is supported by this involvement (Sahay and Hen 2007). turn into neurons devoid of coexisting glial features and signs
Route finding and spatial memory are a particular case of of neuronal maturity. Especially the expression of doublecor-
episodic memory in which a sequence of combinations of land- tin (DCX) in NG2 cells can be misleading (Ehninger et al.
marks has to be memorized. Consequently, there is also a mea- 2011; Guo et al. 2010; Tamura et al. 2007). Doublecortin is
surable effect of adult neurogenesis on the acquisition of the characteristically expressed in precursor cells and new neurons
classical water maze task, a test for hippocampus-dependent of SGZ and SVZ; it is often used as a surrogate marker of neu-
learning (Garthe et al. 2009). In addition, however, and more rogenesis, even though the generalization from neurogenesis
importantly, new neurons are critically involved in perfor- in SGZ and SVZ is not justified.
mance in the “reversal” situation, in which the target to which The hypothalamus stands out among noncortical regions
the animals have to navigate, is secretly moved to a new posi- for which neurogenesis has been reported (Kokoeva et al.
tion and the flexibility of the animals to adjust to the new situ- 2005; Migaud et al. 2010; Perez-Martin et al. 2010). Here,
ation and learn the new target location is assessed. radial glia-like precursor cells of the wall of the third ventricle
The function of new neurons in the adult olfactory bulb is are thought to be the origins of adult-generated neuroendo-
less clear. An involvement of adult neurogenesis in olfactory crine cells. The appropriate developmental potential of these

510 • NEUROGLIA
precursor cells has demonstrated ex vivo (Markakis et al. the conditions of the adult brain. Second, adult neurogenesis
2004). Apparently, neurogenesis can be elicited by CNTF itself has medical relevance if it fails to deliver the particular
(Kokoeva et al. 2005) or IGF1 (Perez-Martin et al. 2010), but type of plasticity for which it is responsible. Several specific
methodological concerns remain. hypotheses have been raised linking adult hippocampal neu-
Generally, the absence of a phenomenon is difficult to rogenesis to age-related cognitive decline and dementia, major
prove, and interesting observations should not be prema- depression and posttraumatic stress disorder, and schizophre-
turely dismissed, but should be rigorously tested before they nia and temporal lobe epilepsy (Kempermann 2011b). These
are accepted. It might turn out that a too-rigid definition of theories can obviously only relate to the contribution that
neurogenesis, excluding the neuronlike properties of NG-cells hippocampal function makes to the disease. Therefore, failing
(see chapter 10) is not justified. On the other hand, departure adult neurogenesis does not explain major depression in its
from very stringent criteria of what a neuron is in relation to a entirety, but might represent an important facet at the inter-
glial cell will enhance confusion rather than reducing it. After face of affective functions and cognition as it takes place in
all, adult neurogenesis in the hippocampus and olfactory bulb, the hippocampus. In the case of temporal lobe epilepsy, the
in which neurons lifelong originate from astrocyte-like cells, central idea is that seizure induces proliferation of precursor
calls many classical distinctions into question. cells, including those at later stages of development, leading
to increased numbers of immature neurons with ectopic posi-
tions and aberrant neurites (Bengzon et al. 1997; Jessberger
11 G L I O G E N E S I S I N T H E C O N T E X T et al. 2005; Parent et al. 2006). These might contribute to the
O F A D U LT N E U R O G E N E S I S propagation of the disease and the transition from individual
seizures to epilepsy. Seizure activity also affects the SVZ, but
Gliogenesis and neurogenesis are intricately linked, not only the pathological consequences of this activation are not yet
because cells with glial properties produce new neurons. Both clear (Young et al. 2011).
neurogenic regions produce glia alongside neurons: oligoden- Failing neurogenesis in the olfactory bulb has been brought
drocytes and interneurons in the SVZ, and astrocytes and into connection with the anosmia found in neurodegenerative
excitatory granule cells in the SGZ. These astrocytes are not disease (Galvan and Bredesen 2007) and depression (Negoias
precursor cells, but have a distinct morphology and antigen et al. 2010), but given the limited importance of the sense of
profile. They are found throughout the granule cell layer. In the smell for greater function of the human brain, neurogenesis in
hippocampus, gliogenesis and neurogenesis are coregulated to the adult olfactory bulb has received less medical attention.
a certain extent (Steiner et al. 2004). One of the population The potential contribution to SVZ-derived precursor cells to
models describing the dynamics of the different precursor cells regenerative processes throughout the brain, whether regener-
and developmental stages in the adult hippocampus suggests ative neurogenesis or other forms of cellular plasticity, has had
an even tighter coupling, ultimately leading to an exhaustion a significant impact on regenerative neurobiology (Lindvall
of the precursor cell pool and their conversion into astrocytes and Kokaia 2006). Much of this interest has been sparked by
(Encinas et al. 2011). Other studies suggest more flexibility in the observation of regenerative neurogenesis after ischemia in
self-renewal and the proportions within the lineage tree, but the striatum. Here, after experimental stroke, neural precur-
similarly suggest a very close interdependency (Bonaguidi sor cells migrated from the SVZ into the striatum, where they
et al. 2011; Suh et al. 2007). differentiated into neurons, albeit at extremely low numbers
Self-renewal of the astrocyte-like stem cells itself is also (Arvidsson et al. 2002). However, the effect was very long last-
a particular form of gliogenesis present in both neurogenic ing (Thored et al. 2006).
regions. The adult SGZ produces very few oligodendrocytes The presence of neural precursor cells in the adult brain
(Kempermann et al. 2003), and it is not fully clear whether and their astrocyte-like properties have also affected con-
these originate from the same population of precursor cells cepts about the origin of brain tumors (Park and Rich 2009;
as the astrocytes and neurons. Ex vivo, the precursor cells are Siebzehnrubl et al. 2011; Stiles and Rowitch 2008). Generally,
indeed tripotent, and overexpression of transcription fac- the stem cell hypothesis of cancerogenesis has fundamentally
tor Ascl1 (Mash1) in precursor cells of the hippocampus in changed oncology, but the influence on neurooncology has
vivo induced oligodendrogenesis ( Jessberger et al. 2008). been particularly strong. If gliomas originate from precursor
Conversely, a low number of astrocytes without involvement cells rather than differentiated glial cells, then several features
in neurogenesis might be produced in the SVZ, but there is of gliomas, especially the patterns of differentiation within
limited information on this aspect. them, can be explained. This particularly applies to features of
neuronal differentiation in gliomas, which have always been
difficult to understand, and to neuronal tumors, such as gangli-
12 A D U LT N E U R O G E N E S I S I N ocytomas and neurocytomas. Thus, the differences among the
B R A I N PAT H O L O GY known types of brain tumors might reflect their origin from
different precursor cell types in the neurogenic zones or brain
Adult neurogenesis is relevant for medicine for several reasons. parenchyma. At the same time, tumors seem to attract precur-
First, adult neurogenesis inspires fundamental research on sor cells, and precursor cells exert a certain antitumorigenic
regenerative medicine because here nature demonstrates what activity that could be consistent with their role in the niche to
bioengineering tries to achieve: how to make neurons under keep proliferative activity controlled (see also chapter 30).

A D U LT N E U R O G E N E S I S • 511
Anticancer treatments aiming at proliferative tumor cells Breunig JJ, Sarkisian MR, Arellano JI, Morozov YM, Ayoub AE, Sojitra
might do collateral damage to proliferating precursor cells, S, et al. 2008. Primary cilia regulate hippocampal neurogenesis
by mediating sonic hedgehog signaling. Proc Natl Acad Sci U S A
possibly explaining cognitive deficits after chemotherapy and 105:13127–13132.
irradiation. Brill MS, Ninkovic J, Winpenny E, Hodge RD, Ozen I, Yang R, et al.
2009. Adult generation of glutamatergic olfactory bulb interneurons.
Nat Neurosci 12:1524–1533.
13 S U M M A RY A N D P E R S P E C T I VE S Brown JP, Couillard-Despres S, Cooper-Kuhn CM, Winkler J, Aigner
L, Kuhn HG. 2003. Transient expression of doublecortin during
adult neurogenesis. J Comp Neurol 467:1–10.
Adult neurogenesis is a process that adds a particular type of Chojnacki AK, Mak GK, Weiss S. 2009. Identity crisis for adult periven-
functionality and plasticity to the adult brain. Rather than tricular neural stem cells: subventricular zone astrocytes ependymal
being an atavism, as was presumed for some time after the cells or both? Nat Rev Neurosci 10:153–163.
initial description, adult neurogenesis is a highly specialized Clelland CD, Choi M, Romberg C, Clemenson GD Jr, Fragniere A, Tyers
P, et al. 2009. A functional role for adult hippocampal neurogenesis
mechanism that adds flexibility and adaptation to very par- in spatial pattern separation. Science 325:210–213.
ticular types of networks. Outside the two canonical neuro- Couillard-Despres S, Winner B, Schaubeck S, Aigner R, Vroemen M,
genic regions in the hippocampus and olfactory system, the Weidner N, et al. 2005. Doublecortin expression levels in adult brain
interpretation of the often-conflicting results and potential reflect neurogenesis. Eur J Neurosci 21:1–14.
functional benefit is more difficult. However, there might be Curtis MA, Kam M, Nannmark U, Anderson MF, Axell MZ, Wikkelso
C, et al. 2007. Human neuroblasts migrate to the olfactory bulb via a
more examples of neurogenesis than hippocampus and olfac- lateral ventricular extension. Science 315:1243–1249.
tory system, even in mammals, but under physiological con- Dayer AG, Cleaver KM, Abouantoun T, Cameron HA. 2005.
ditions they seem to range on a minute scale, if they exist at New Gabaergic interneurons in the adult neocortex and striatum are
all. Nevertheless, some neurogenesis might be inducible under generated from different precursors. J Cell Biol 168:415–427.
pathological conditions, although if and how this reactive Doetsch F, Caille I, Lim DA, Garcia-Verdugo JM, Alvarez-Buylla A.
1999a. Subventricular zone astrocytes are neural stem cells in the
neurogenesis contributes to regeneration is not known. adult mammalian brain. Cell 97:703–716.
At the cellular level, the fact that glia makes neurons and Doetsch F, Garcia-Verdugo JM, Alvarez-Buylla A. 1997. Cellular
even normal astrocytes can be experimentally reprogrammed composition and three-dimensional organization of the subven-
to become neurogenic has overturned many traditional con- tricular germinal zone in the adult mammalian brain. J Neurosci
cepts about the relationship between the cell lineages of the 17:5046–5061.
Doetsch F, Garcia-Verdugo JM, Alvarez-Buylla A. 1999b. Regeneration
brain. But below this deceiving simplicity lies an immensely of a germinal layer in the adult mammalian brain. Proc Natl Acad Sci
complex process, controlled by numerous genes and regulated U S A 96:11619–11624.
by behavioral activity and a large number of extrinsic factors. Ehninger D, Wang LP, Klempin F, Romer B, Kettenmann H,
The key questions in the field that await answers in the Kempermann G. 2011. Enriched environment and physical activity
years to come are the following. How does adult neurogenesis reduce microglia and influence the fate of Ng2 cells in the amygdala
of adult mice. Cell Tissue Res 345:69–86.
contribute to brain plasticity in humans? How is the recipro-
Ekdahl CT, Claasen JH, Bonde S, Kokaia Z, Lindvall O. 2003.
cal interaction of regulation and function in adult neurogen- Inflammation is detrimental for neurogenesis in adult brain. Proc
esis established? What exactly is the specific function of new Natl Acad Sci U S A 100:13632–13637.
neurons? And what exactly is the nature of the glia-like stem Encinas JM, Michurina TV, Peunova N, Park JH, Tordo J, Peterson
cells that drive adult neurogenesis? DA, et al. 2011. Division-coupled astrocytic differentiation and
age-related depletion of neural stem cells in the adult hippocampus.
Cell Stem Cell 8:566–579.
Eriksson PS, Perfi lieva E, Björk-Eriksson T, Alborn AM, Nordborg
REFERENCES C, Peterson DA, et al. 1998. Neurogenesis in the adult human hip-
pocampus. Nat Med 4:1313–1317.
Aimone JB, Wiles J, Gage FH. 2006. Potential role for adult neurogenesis Filippov V, Kronenberg G, Pivneva T, Reuter K, Steiner B, Wang LP,
in the encoding of time in new memories. Nat Neurosci 9:723–727. et al. 2003. Subpopulation of nestin-expressing progenitor cells in the
Aimone JB, Wiles J, Gage FH. 2009. Computational influence of adult adult murine hippocampus shows electrophysiological and morpho-
neurogenesis on memory encoding. Neuron 61:187–202. logical characteristics of astrocytes. Mol Cell Neurosci 23:373–382.
Amrein I, Dechmann DK, Winter Y, Lipp HP. 2007. Absent or low rate Galvan V, Bredesen DE. 2007. Neurogenesis In the adult brain: impli-
of adult neurogenesis in the hippocampus of bats (chiroptera). Plos cations for alzheimer’s disease. CNS Neurol Disord Drug Targets
One 2:E455. 6:303–310.
Arvidsson A, Collin T, Kirik D, Kokaia Z, Lindvall O. 2002. Neuronal Garcia AD, Doan NB, Imura T, Bush TG, Sofroniew MV. 2004.
replacement from endogenous precursors in the adult brain after Gfap-expressing progenitors are the principal source of constitutive
stroke. Nat Med 8:963–970. neurogenesis in adult mouse forebrain. Nat Neurosci 7:1233–1241.
Bengzon J, Kokaia Z, Elmér E, Nanobashvili A, Kokaia M, Lindvall Garthe A, Behr J, Kempermann G. 2009. Adult-generated hippocampal
O. 1997. Apoptosis and proliferation of dentate gyrus neurons after neurons allow the flexible use of spatially precise learning strategies.
single and intermittent limbic seizures. Proc Natl Acad Sci U S A Plos One 4:E5464.
94:10432–10437. Giachino C, De Marchis S, Giampietro C, Parlato R, Perroteau I, Schutz
Bonaguidi MA, Mcguire T, Hu M, Kan L, Samanta J, Kessler JA. G, et al. 2005. Camp response element-binding protein regulates dif-
2005. Lif and bmp signaling generate separate and discrete types of ferentiation and survival of newborn neurons in the olfactory bulb.
gfap-expressing cells. Development 132:5503–5514. J Neurosci 25:10105–10118.
Bonaguidi MA, Wheeler MA, Shapiro JS, Stadel RP, Sun GJ, Ming GL, Gould E, Beylin A, Tanapat P, Reeves A, Shors TJ. 1999a. Learning
et al. 2011. In vivo clonal analysis reveals self-renewing and multipo- enhances adult neurogenesis in the hippocampal formation. Nat
tent adult neural stem cell characteristics. Cell 145:1142–1155. Neurosci 2:260–265.

512 • NEUROGLIA
Gould E, Reeves AJ, Graziano MS, Gross CG. 1999b. Neurogenesis in hippocampus respond differently to physiologic neurogenic stimuli.
the neocortex of adult primates. Science 286:548–552. J Comp Neurol 467:455–463.
Grandel H, Kaslin J, Ganz J, Wenzel I, Brand M. 2006. Neural stem cells Lie DC, Colamarino SA, Song HJ, Desire L, Mira H, Consiglio A, et al.
and neurogenesis in the adult zebrafish brain: origin proliferation 2005. Wnt signalling regulates adult hippocampal neurogenesis.
dynamics migration and cell fate. Dev Biol 295:263–277. Nature 437:1370–1375.
Gritti A, Bonfanti L, Doetsch F, Caille I, Alvarez-Buylla A, Lim DA, Lim DA, Tramontin AD, Trevejo JM, Herrera DG, Garcia-Verdugo JM,
et al. 2002. Multipotent neural stem cells reside into the rostral exten- Alvarez-Buylla A. 2000. Noggin antagonizes Bmp signaling to create
sion and olfactory bulb of adult rodents. J Neurosci 22:437–445. a niche for adult neurogenesis. Neuron 28:713–726.
Guo F, Maeda Y, Ma J, Xu J, Horiuchi M, Miers L, et al. 2010. Pyramidal Lindvall O, Kokaia Z. 2006. Stem cells for the treatment of neurological
neurons are generated from oligodendroglial progenitor cells in adult disorders. Nature 441:1094–1096.
piriform cortex. J Neurosci 30:12036–12049. Lledo PM, Alonso M, Grubb MS. 2006. Adult neurogenesis and func-
Hastings NB, Gould E. 1999. Rapid extension of axons into the ca3 region tional plasticity in neuronal circuits. Nat Rev Neurosci 7:179–193.
by adult-generated granule cells. J Comp Neurol 413:146–154. Lledo PM, Lagier S. 2006. Adjusting neurophysiological computations
Hodge RD, Kowalczyk TD, Wolf SA, Encinas JM, Rippey C, in the adult olfactory bulb. Semin Cell Dev Biol 17:443–453.
Enikolopov G, et al. 2008. Intermediate progenitors in adult hip- Lledo PM, Merkle FT, Alvarez-Buylla A. 2008. Origin and function
pocampal neurogenesis: tbr2 expression and coordinate regulation of of olfactory bulb interneuron diversity. Trends Neurosci 31:
neuronal output. J Neurosci 28:3707–3717. 392–400.
Imayoshi I, Sakamoto M, Ohtsuka T, Takao K, Miyakawa T, Lois C, Alvarez-Buylla A. 1994. Long-distance neuronal migration in
Yamaguchi M, et al. 2008. Roles of continuous neurogenesis in the adult mammalian brain. Science 264:1145–1148.
the structural and functional integrity of the adult forebrain. Nat Lugert S, Basak O, Knuckles P, Haussler U, Fabel K, Gotz M, et al. 2010.
Neurosci 11:1153–1161. Quiescent and active hippocampal neural stem cells with distinct
Imura T, Kornblum HI, Sofroniew MV. 2003. The predominant neural morphologies respond selectively to physiological and pathological
stem cell isolated from postnatal and adult forebrain but not early stimuli and aging. Cell Stem Cell 6:445–456.
embryonic forebrain expresses Gfap. J Neurosci 23:2824–2832. Magavi S, Leavitt B, Macklis J. 2000. Induction of neurogenesis in the
Inta D, Alfonso J, Von Engelhardt J, Kreuzberg MM, Meyer AH, Van neocortex of adult mice. Nature 405:951–955.
Hooft JA, et al. 2008. Neurogenesis and widespread forebrain migra- Markakis EA, Palmer TD, Randolph-Moore L, Rakic P, Gage FH. 2004.
tion of distinct gabaergic neurons from the postnatal subventricular Novel neuronal phenotypes from neural progenitor cells. J Neurosci
zone. Proc Natl Acad Sci U S A 105:20994–20999. 24:2886–2897.
Jankovski A, Garcia C, Soriano E, Sotelo C. 1998. Proliferation migra- Merkle FT, Mirzadeh Z, Alvarez-Buylla A. 2007. Mosaic organization of
tion and differentiation of neuronal progenitor cells in the adult neural stem cells in the adult brain. Science 317:381–384.
mouse subventricular zone surgically separated from its olfactory Merkle FT, Tramontin AD, Garcia-Verdugo JM, Alvarez-Buylla A.
bulb. Eur J Neurosci 10:3853–3868. 2004. Radial glia give rise to adult neural stem cells in the subven-
Jessberger S, Romer B, Babu H, Kempermann G. 2005. Seizures induce tricular zone. Proc Natl Acad Sci U S A 101:17528–17532.
proliferation and dispersion of doublecortin-positive hippocampal Migaud M, Batailler M, Segura S, Duittoz A, Franceschini I, Pillon D.
progenitor cells. Exp Neurol 196:342–351. 2010. Emerging new sites for adult neurogenesis in the mammalian
Jessberger S, Toni N, Clemenson GD Jr, Ray J, Gage FH. 2008. Directed brain: a comparative study between the hypothalamus and the classi-
differentiation of hippocampal stem/progenitor cells in the adult cal neurogenic zones. Eur J Neurosci 32:2042–2052.
brain. Nat Neurosci 11:888–893. Mirzadeh Z, Merkle FT, Soriano-Navarro M, Garcia-Verdugo JM,
Kaslin J, Ganz J, Brand M. 2008. Proliferation neurogenesis and regen- Alvarez-Buylla A. 2008. Neural stem cells confer unique pinwheel
eration in the non-mammalian vertebrate brain. Philos Trans R Soc architecture to the ventricular surface in neurogenic regions of the
Lond B Biol Sci 363:101–122. adult brain cell. Stem Cell 3:265–278.
Kee N, Teixeira CM, Wang AH, Frankland PW. 2007. Preferential Morrens J, Van Den Broeck W, Kempermann G. 2012. Glial cells in
incorporation of adult-generated granule cells into spatial memory adult neurogenesis. Glia 60:159–174.
networks in the dentate gyrus. Nat Neurosci 10:355–362. Morshead CM, Garcia AD, Sofroniew MV, Van Der Kooy D. 2003. The
Kempermann G. 2011a. Adult neurogenesis 2. Stem cells and neuronal ablation of glial fibrillary acidic protein-positive cells from the adult
development in the adult brain. New York: Oxford University Press. central nervous system results in the loss of forebrain neural stem cells
Kempermann G. 2011b. Medicine. In: Kempermann G (ed.), Adult neu- but not retinal stem cells. Eur J Neurosci 18:76–84.
rogenesis 2. Stem cells and neuronal development in the adult brain. Nakatomi H, Kuriu T, Okabe S, Yamamoto S, Hatano O, Kawahara N,
New York: Oxford University Press. et al. 2002. Regeneration of hippocampal pyramidal neurons after
Kempermann G. 2011c. The pessimist’s and optimist’s views of adult ischemic brain injury by recruitment of endogenous neural progeni-
neurogenesis. Cell 145:1009–1011. tors. Cell 110:429–441.
Kempermann G. 2011d. Seven principles in the regulation of adult neu- Negoias S, Croy I, Gerber J, Puschmann S, Petrowski K, Joraschky P,
rogenesis. Eur J Neurosci 33:1018–1024. et al. 2010. Reduced olfactory bulb volume and olfactory sensitivity
Kempermann G, Gast D, Kronenberg G, Yamaguchi M, Gage FH. 2003. in patients with acute major depression. Neuroscience 169:415–421.
Early determination and long-term persistence of adult-generated new Noctor SC, Flint AC, Weissman TA, Dammerman RS, Kriegstein AR.
neurons in the hippocampus of mice. Development 130:391–399. 2001. Neurons derived from radial glial cells establish radial units in
Kempermann G, Jessberger S, Steiner B, Kronenberg G. 2004. Milestones neocortex. Nature 409:714–720.
of neuronal development in the adult hippocampus. Trends Neurosci Nowakowski RS, Hayes NL. 2000. New neurons: extraordinary evi-
27:447–452. dence or extraordinary conclusion? Science 288:771.
Knoth R, Singec I, Ditter M, Pantazis G, Capetian P, Meyer RP, et al. Ohira K, Furuta T, Hioki H, Nakamura KC, Kuramoto E, Tanaka Y,
2010. Murine features of neurogenesis in the human hippocampus et al. 2010. Ischemia-induced neurogenesis of neocortical layer 1 pro-
across the lifespan from 0 to 100 years. Plos One 5:E8809. genitor cells. Nat Neurosci 13:173–179.
Kokoeva MV, Yin H, Flier JS. 2005. Neurogenesis in the hypothalamus of Palma V, Lim DA, Dahmane N, Sanchez P, Brionne TC, Herzberg CD,
adult mice: potential role in energy balance. Science 310:679–683. et al. 2005. Sonic hedgehog controls stem cell behavior in the postna-
Komitova M, Eriksson PS. 2004. Sox-2 is expressed by neural progenitors tal and adult brain. Development 132:335–344.
and astroglia in the adult rat brain. Neurosci Lett 369:24–27. Parent JM, Elliott RC, Pleasure SJ, Barbaro NM, Lowenstein DH. 2006.
Kriegstein A, Alvarez-Buylla A. 2009. the glial nature of embryonic and Aberrant seizure-induced neurogenesis in experimental temporal
adult neural stem cells. Annu Rev Neurosci 32:149–184. lobe epilepsy. Ann Neurol 59:81–91.
Kronenberg G, Reuter K, Steiner B, Brandt MD, Jessberger S, Yamaguchi Park DM, Rich JN. 2009. Biology of glioma cancer stem cells. Mol Cell
M, et al. 2003. Subpopulations of proliferating cells of the adult 28:7–12.

A D U LT N E U R O G E N E S I S • 513
Perez-Martin M, Cifuentes M, Grondona JM, Lopez-Avalos MD, Snyder JS, Kee N, Wojtowicz JM. 2001. Effects of adult neurogen-
Gomez-Pinedo U, Garcia-Verdugo JM, et al. 2010 Igf-I stimulates esis on synaptic plasticity in the rat dentate gyrus. J Neurophysiol
neurogenesis in the hypothalamus of adult rats. Eur J Neurosci 85:2423–2431.
31:1533–1548. Song H, Stevens CF, Gage FH. 2002. Astroglia induce neurogenesis
Rakic P. 2002a. Adult neurogenesis in mammals: an identity crisis. J from adult neural stem cells. Nature 417:39–44.
Neurosci 22:614–618. Stanfield BB, Trice JE. 1988. Evidence that granule cells generated in the
Rakic P. 2002b. Neurogenesis in adult primate neocortex: an evaluation dentate gyrus of adult rats extend axonal projections. Exp Brain Res
of the evidence. Nat Rev Neurosci 3:65–71. 72:399–406.
Rao MS, Shetty AK. 2004. Efficacy of doublecortin as a marker to Steiner B, Klempin F, Wang L, Kott M, Kettenmann H, Kempermann
analyse the absolute number and dendritic growth of newly gen- G. 2006. Type-2 cells as link between glial and neuronal lineage in
erated neurons in the adult dentate gyrus. Eur J Neurosci 19: adult hippocampal neurogenesis. Glia 54:805–814.
234–246. Steiner B, Kronenberg G, Jessberger S, Brandt MD, Reuter K,
Revest JM, Dupret D, Koehl M, Funk-Reiter C, Grosjean N, Piazza Kempermann G. 2004. Differential regulation of gliogenesis in the
PV, et al. 2009. Adult hippocampal neurogenesis is involved in context of adult hippocampal neurogenesis in mice. Glia 46:41–52.
anxiety-related behaviors. Mol Psychiatry 14:959–967. Stiles CD, Rowitch DH. 2008. Glioma stem cells: a midterm exam.
Reynolds BA, Rietze RL. 2005. Neural stem cells and neurospheres— Neuron 58:832–846.
re-evaluating the relationship. Nat Methods 2:333–336. Suh H, Consiglio A, Ray J, Sawai T, D’amour KA, Gage FH. 2007. In vivo
Rochefort C, Gheusi G, Vincent JD, Lledo PM. 2002. Enriched fate analysis reveals the multipotent and self-renewal capacities of Sox2+
odor exposure increases the number of newborn neurons in the neural stem cells in the adult hippocampus. Cell Stem Cell 1:515–528.
adult olfactory bulb and improves odor memory. J Neurosci Tamura Y, Kataoka Y, Cui Y, Takamori Y, Watanabe Y, Yamada
22:2679–2689. H. 2007. Multi-directional differentiation of doublecortin- and
Sahay A, Hen R. 2007. Adult hippocampal neurogenesis in depression. ng2-immunopositive progenitor cells in the adult rat neocortex in
Nat Neurosci 10:1110–1115. vivo. Eur J Neurosci 25:3489–3498.
Sanai N, Nguyen T, Ihrie RA, Mirzadeh Z, Tsai HH, Wong M, et al. Tashiro A, Makino H, Gage FH. 2007. Experience-specific functional
2011. Corridors of migrating neurons in the human brain and their modification of the dentate gyrus through adult neurogenesis: a crit-
decline during infancy. Nature 478:382–386. ical period during an immature stage. J Neurosci 27:3252–3259.
Saxe MD, Battaglia F, Wang JW, Malleret G, David DJ, Monckton JE, Thored P, Arvidsson A, Cacci E, Ahlenius H, Kallur T, Darsalia V. 2006.
et al. 2006. Ablation of hippocampal neurogenesis impairs contex- Persistent production of neurons from adult brain stem cells during
tual fear conditioning and synaptic plasticity in the dentate gyrus. recovery after stroke. Stem Cells 24:739–747.
Proc Natl Acad Sci U S A 103:17501–17506. Thored P, Heldmann U, Gomes-Leal W, Gisler R, Darsalia V, Taneera
Schmidt-Hieber C, Jonas P, Bischofberger J. 2004. Enhanced synaptic J, et al. 2009. Long-term accumulation of microglia with proneuro-
plasticity in newly generated granule cells of the adult hippocampus. genic phenotype concomitant with persistent neurogenesis in adult
Nature 429:184–187. subventricular zone after stroke. Glia 57:835–849.
Seri B, Garcia-Verdugo JM, Mcewen BS, Alvarez-Buylla A. 2001. Toni N, Laplagne DA, Zhao C, Lombardi G, Ribak CE, Gage FH, et al.
Astrocytes give rise to new neurons in the adult mammalian hip- 2008. Neurons born in the adult dentate gyrus form functional syn-
pocampus. J Neurosci 21:7153–7160. apses with target cells. Nat Neurosci 11:901–907.
Siebzehnrubl FA, Reynolds BA, Vescovi A, Steindler DA, Deleyrolle LP. Whitman MC, Fan W, Rela L, Rodriguez-Gil DJ, Greer CA. 2009.
2011. The origins of glioma: e pluribus unum? Glia 59:1135–1147. Blood vessels form a migratory scaffold in the rostral migratory
Siegrist SE, Haque NS, Chen CH, Hay BA, Hariharan IK. 2010. stream. J Comp Neurol 516:94–104.
Inactivation Of both foxo and reaper promotes long-term adult neu- Wiskott L, Rasch MJ, Kempermann G. 2006. A functional hypothesis
rogenesis in Drosophila. Curr Biol 20:643–648. for adult hippocampal neurogenesis: avoidance of catastrophic inter-
Sierra A, Encinas JM, Deudero JJ, Chancey JH, Enikolopov G, ference in the dentate gyrus. Hippocampus 16:329–343.
Overstreet-Wadiche L, S et al. 2010. Microglia shape adult hippocam- Young SZ, Taylor MM, Bordey A. 2011. Neurotransmitters couple
pal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem brain activity to subventricular zone neurogenesis. Eur J Neurosci
Cell 7:483–495. 33:1123–1132.

514 • NEUROGLIA
41.
MODULATION OF NEUROENDOCRINE SYSTEMS
Stéphane H. R. Oliet

A B B R E VI AT I O N S within the PVN and SON (Ludwig 1998). Peripheral and cen-
tral secretion of these hormones is dependent on the electrical
AMPA D-amino-3-hydroxy-5-méthylisoazol-4- activity of magnocellular neurons that is itself under the influ-
propionate ence of afferent synaptic inputs. The SON is a very homog-
ATP adenosine triphosphate enous nucleus, composed essentially of the soma and dendrites
ECM extracellular matrix of OT and VP neurons, astrocytes, and vascular elements. The
ECS extracellular space majority of glial cells in this nucleus are radial astrocytes whose
GABA J-aminobutyric acid cell bodies are located in the ventral portion of the SON, or
GnRH gonadotropin-releasing hormone ventral glia limitans, from which arise long processes oriented
HNS hypothalamo-neurohypophysial system ventrodorsally that enwrap neuronal elements (Bonfanti et al.
LTD long-term depression 1993; Israel et al. 2003b). These astrocytes are reminiscent of
LTP long-term potentiation radial glia in the developing central nervous system. The main
mGluR metabotropic glutamate receptor afferents to the SON and the PVN use GABA and glutamate
NCAM Neural cell adhesion molecule as inhibitory and excitatory transmitters, respectively (Wuarin
NMDA N-methyl-d-aspartate and Dudek 1993). In the SON, about 40% of all synapses are
NO nitric oxide GABAergic, whereas 25% are glutamatergic (Theodosis 2002).
OT oxytocin At the level of the neurohypophysis, at which axon terminals of
PSA polysialic acid OT and VP neurons contact the blood vessels together, one can
PVN paraventricular nucleus find specialized astrocytes known as pituicytes.
SON supraoptic nucleus Another hypothalamic area that has received a lot of
VP vasopressin attention in the context of neuronal–glial interactions is the
gonadotropin-releasing hormone (GnRH) system that is made
of GnRH neurons whose cell bodies are located in the arcuate
1 INTRODUCTION nucleus and which project their axon terminals in the median
eminence, where GnRH can be directly released in the portal
The hypothalamus is a brain structure controlling several vital system to mediate key reproductive functions. Astrocytes can be
functions such as reproduction, lactation, food intake, drink- found in both the arcuate nucleus and the median eminence. In
ing, circadian rhythms, autonomic functions, body fluid, and the latter, specialized glial cells known as tanycytes are also pres-
temperature homeostasis. It is comprised of many heteroge- ent. These radial-like glial cells extend from the third ventricle to
neous nuclei that are themselves composed of neurons and the portal system where they enwrap GnRH axon terminals.
glial cells. Hypothalamic neurons are neuroendocrine cells that One of the most remarkable properties of the
synthesize and release peptides as a function of their electrical hypothalamo-neurohypophysial system is its ability to
activity. Such peptides can be released locally, where they serve undergo a reversible anatomical remodeling that is character-
as signaling molecules, and/or secreted in the bloodstream, ized by a reduction of the glial coverage of magnocellular neu-
where they act as neurohormones to regulate distant targets. rons and synapses (Bobak and Salm 1996; Hatton et al. 1984;
One of the most extensively studied hypothalamic areas is the Theodosis and Poulain 1993). This occurs during physiologi-
hypothalamo-neurohypophysial system (HNS). It is made of cal conditions such as parturition, lactation, and dehydration.
magnocellular neurons that synthesize the hormones oxytocin A similar process takes place at the level of the neurohypo-
(OT) and vasopressin (VP) and whose cell bodies are located in physis, where the pituicytes retract from between the axon
the supraoptic (SON) and paraventricular (PVN) nuclei of the terminals (Hatton 1999). This phenomenon has permitted
hypothalamus. These neurons project their axons in the neuro- the study of neuronal–glial interactions by comparing cel-
hypophysis where their hormonal content is released directly in lular processes under different conditions of glial environ-
the blood stream. OT is essential for vital functions such as lac- ment. As such, it was proved to be a unique model to establish
tation and parturition, whereas VP is the antidiuretic hormone the contribution of glial cells to neuronal functions. Similar
critical to body fluid and cardiovascular homeostasis. In addi- neuronal–glial anatomical changes have since been reported
tion to their peripheral release, it is now clear that OT and VP in other hypothalamic centers of rodents and nonhuman pri-
can also be released centrally in different brain regions, including mates, as well as in nonhypothalamic structures.

515
2 CONTRIBUTION UNDER that does not affect d-serine levels has no effect on NMDAR
BASAL CONDITIONS activity in the SON. Thus, it appears that astrocytes are essen-
tial for all the processes that depend on NMDA receptor activ-
Because of their position in the neuropile, astrocytic processes ity. Similarly, adenosine triphosphate (ATP) originating from
represent a physical barrier restricting spillover and diffusion of astrocytes has been shown to induce the insertion in the mem-
signaling molecules that are released locally (Piet et al. 2004). brane of D-amino-3-hydroxy-5-méthylisoazol-4-propionate
Moreover, because of their position, these glial cells are known (AMPA) receptors in PVN magnocellular neurons (Gordon
to control the local environment in which neurons develop and et al. 2005). Such a release is triggered by noradrenaline, and
function. In particular, they maintain a tight control on local ATP action is mediated by P2X7 subtype of receptors. This
ion concentrations and pH. Through their coupling with blood phenomenon is believed to be important for controlling syn-
vessels, astrocytes provide metabolic substrates to neurons aptic strength under conditions in which adrenaline-releasing
while clearing away toxic waste. The concentrations of extracel- A1 inputs onto PVN are recruited.
lular K+ or glutamate, that are produced as a result of neuronal In the neurohypophysis, as well as the external layer of the
activity for example, need to be tightly regulated because their median eminence, processes of astrocytic-like cells (pituicytes,
accumulation in the extracellular space (ECS) can alter neu- tanycytes) enclose neurosecretory axons, thereby limiting the
ronal excitability dramatically (Boudaba et al. 2003; Oliet et al. release of their products of secretion and their possible paracrine
2001). The clearance of glutamate, the major excitatory neu- and/or autocrine actions. In addition, these glial processes lie
rotransmitter in the brain, is ensured by GLT-1 and GLAST, alongside perivascular spaces, thereby physically restricting the
two glutamate transporter subtypes expressed at the glial mem- access of neurosecretory terminals to the perivascular basal lam-
brane (see chapter 35). This control is essential to preserve the ina. Such a physical barrier affects neurosecretion by limiting dif-
spatiotemporal profile of the glutamate transient and regulate fusion of neuropeptides released at the terminals into the general
the synaptic efficacy through the activation of presynaptic glu- circulation. In the median eminence, where capillaries are fenes-
tamate receptors coupled to transmitter release. This includes trated, enwrapping of blood vessels by tanycyte processes appears
metabotropic glutamate (mGluR), as well as kainate receptors, to contribute to the blood-brain barrier (Prevot et al. 2010).
whose activation inhibits and facilitates synaptic transmission Under control conditions, these tanycytes also prevent access of
in the SON, respectively (Bonfardin et al. 2010; Boudaba et al. most neuroendocrine terminals to the pericapillary space.
2003; Oliet et al. 2001).
Glial cells also release active substances termed gliotrans-
mitters by analogy to neurotransmitters (see chapter 17). 3 G L I A L S T RU C T U R A L P L A S T I C I T Y
In the hypothalamus, several gliotransmitters have been identi-
fied, including taurine, d-serine, and ATP (Gordon et al. 2005;
3.1 C H A N G E I N G L I A L C OVE R AG E
Hussy et al. 2000; Panatier et al. 2006). Taurine is released
O F N EU RO NS
through volume-sensitive ion channels in response to hypo-
tonic stimulation. It activates strychnine-sensitive glycinergic It is now more than two decades ago that evidence was
receptors expressed by magnocellular neurons. These receptors reported for the first time that the morphology of glial cells
are permeable to chloride ions so that they mediate membrane in specific neuroendocrine systems undergoes remarkable
hyperpolarization when they are stimulated at resting mem- dynamic changes in relation to the activity of adjacent neu-
brane potential. This hypotonic-induced release of taurine is rons. It is the case of the HNS during conditions associated
believed to be part of the osmoregulatory process that enables with enhanced secretion of VP and OT, such as chronic dehy-
the control of osmolality by VP cells (Hussy et al. 2000). dration, lactation, or parturition (Theodosis 2002). These
When osmolality is reduced, glial cells swell, causing the release changes are completely reversible on cessation of stimulation
of taurine. Glycinergic receptors are in turn activated, leading and are characterized by a progressive hypertrophy of the neu-
to hyperpolarization of VP cells and inhibition of their firing. rons, an increased number of excitatory and inhibitory syn-
This ultimately leads to reduced secretion of VP in the general apses, and a pronounced reduction in the astrocytic coverage
circulation. As a consequence, diuresis is increased, thereby of magnocellular neurons (Fig. 41.1). As a result, neuronal
contributing to the recovery of normal osmolality. surfaces no longer separated by glial processes become directly
d-Serine is also released from astrocytes in the SON juxtaposed. Similar changes occur at the level of the neurohy-
(Panatier et al. 2006). It serves as an endogenous coagonist pophysis, in which neuro-hemal contacts increase as a result
of N-methyl-d-aspartate receptors (NMDARs). These glu- of pituicyte retraction. These changes are similar to those
tamate receptors contribute to excitatory neurotransmission that were reported later in the suprachiasmatic (Gerics et al.
and mediate the most common forms of activity-dependent 2006; Lavialle and Serviere 1993; Lavialle et al. 2001), arcu-
synaptic plasticity, long-term potentiation, and long-term ate (Garcia-Segura et al. 1994b), and preoptic (Gerhold and
depression. In the SON, NMDARs are also responsible for Wise 2006; Jansen et al. 2003) nuclei in response to circadian
generating oscillatory activity (Hu and Bourque 1992). In the rhythms and fluctuating levels of sexual steroid, respectively.
absence of such a coagonist, NMDA receptors cannot operate, In the hypothalamus, OT neurons usually occur in tightly
as revealed by the use of D-amino acid oxidase, an enzyme that packed clusters intermingled with VP neurons. Electron
selectively degrades d-serine without affecting glycine (Panatier microscopy shows unambiguously that in spite of such close
et al. 2006). Conversely, degrading glycine with glycine oxidase packing, OT cells remain separated by fine astrocytic processes

516 • NEUROGLIA
unstimulated neurosecretory terminals and about 60% by pituicytes. These
proportions are reversed during conditions that stimulate
dendrite
VP and/or OT secretion (Hatton et al. 1984; Monlezun
soma1 et al. 2005).
Glial–neuronal changes occur within a few hours of
the onset of parturition or osmotic stimulation. They can
be reproduced in vitro, in acute slices of adult hypothala-
mus that include the SON (Langle et al. 2003; Theodosis
et al. 2006), or isolated neurohypophysis (Monlezun et al.
2005; Perlmutter et al. 1984). In such preparations, the glial
soma2
remodeling takes place within 1 hour after the onset of stimu-
soma3 lated neurosecretion. When the stimulation of OT neurons
ends and circulating OT concentrations come back to basal
values, the astrocytic coverage of neurons returns to its ini-
stimulated
tial level, a process that can be very rapid (Langle et al. 2003)
depending on how long the system has remained activated
(Theodosis 2002).
~10 nm 0.1 μm Rapid activity-dependent astrocytic plasticity is detectable
in other hypothalamic areas. In the rat arcuate nucleus, com-
soma2 posed of neurons that control growth hormone, luteinizing
hormone, and prolactin secretion, the proportion of neuronal
soma1 somatic surface covered by astrocytic processes fluctuates with
changing sexual steroid levels. This proportion is high when
plasma estrogen is elevated, as in the afternoon of proestrus
and morning of estrus, and is low 24 hours later at metestrus
soma3 when estrogen levels have diminished (Garcia-Segura et al.
1994a). Administration of estrogen to ovariectomized animals
mimics the effects of the estrous cycle on the glial coverage of
neurons (Garcia-Segura et al. 1994a). A similar type of struc-
Figure 41.1 Anatomical Glial Remodeling in the Supraoptic Nucleus. tural reorganization has been reported in the infundibulum
Electron microscopy pictures taken in the SON under control conditions and preoptic area of nonhuman primates (Garcia-Segura et al.
(top; unstimulated) and in lactating rats (bottom; stimulated). Thin astro-
cytic processes (blue) separate neuronal somatic and dendritic elements 1994a; Witkin et al. 1991), in which fluctuation of steroid
in unstimulated conditions. The glial withdrawal that accompanies the levels causes a variation of the glial coverage of the neurons
structural remodeling of the SON in stimulated conditions such as synthesizing GnRH, a hormone that controls the secretion of
lactation results in directly juxtaposed neuronal elements. Adapted gonadotropin from the pituitary. Such variations can be mim-
from Montagnese et al. 1988. icked when estrogen is given to ovariectomized animals.
The axons of GnRH neurons project to the external layer
that also enwrap synapses. In contrast, during parturition, of the median eminence, where changing steroid levels result
lactation, chronic dehydration, or in response to elevated in glial changes similar to those observed in the neurohypo-
ambient levels of OT, there is a significant reduction in the physis (De Serrano et al. 2004; King and Rubin 1995), includ-
astrocytic coverage of the neurons and their synapses. Under ing retraction of tanycyte processes from the perivascular
normal conditions, astrocytic processes cover about 90% of zone and access of GnRH terminals to fenestrated capillar-
any OT somata, which is reduced to 70% during lactation or ies. Therefore, in neurohemal structures glial rearrangements
chronic dehydration (Chapman et al. 1986; Theodosis et al. ultimately promote the access of neurosecretory terminals
1986a). It is of note that the astrocytic coverage of VP neurons to perivascular zones. This is likely to favor the secretion of
is not altered and remains around 90% even after strong and neurohormone into the bloodstream, while at the same time
persistent stimulation, such as chronic salt loading (Chapman allowing exposition of neuroendocrine terminals to signaling
et al. 1986). At the same time, part of the membrane surface molecules circulating in the general circulation.
of OT somata and dendrites become directly juxtaposed (see
Fig. 41.1). Although VP neurons display similar juxtapositions,
3.2 P E R M I S S I VE FAC TO R S
their incidence and extent are low and show no variations with
conditions that modify VP secretion (Chapman et al. 1986; The structural plasticity that occurs in different neuroendocrine
Theodosis et al. 1986a). Concomitantly, glial remodeling in structures must involve several permissive and inductive factors,
the neurohypophysis results in a reduced glial ensheathment including cell surface and extracellular matrix (ECM) molecules,
of neurosecretory terminals, leading to reduced gliovascular as well as soluble agents, some of which have been identified in
and increased neurovascular contact zones (Theodosis and developing systems undergoing similar glial and neuronal struc-
MacVicar 1996). Under basal conditions of neurosecretion, tural remodeling. It is not surprising then that neural systems
about 40% of the perivascular basal lamina is contacted by displaying remodeling properties in the adult continue to express

M O D U L AT I O N O F N E U R O E N D O C R I N E SYS T E M S • 517
many of these embryonic factors. This includes cell adhesion infusion of this peptide induces morphological neuronal–
molecules such as cadherins, neurexins, and Neural cell adhe- glial changes in the SON similar to those detected during lac-
sion molecule (NCAM). Complex ECM glycoproteins such tation (Theodosis et al. 1986b). These effects are also observed
as chondroitin sulfate proteoglycans and tenascins also appear in vitro in acute hypothalamic slices obtained from gestating
to participate (Kleene and Schachner 2004; Theodosis et al. female rats. That this neuropeptide acts specifically via its
2004a; Waites et al. 2005). NCAM, and in particular its highly receptors was confirmed by using close analogs that mimicked
sialylated isoform PSA-NCAM (polysialic acid-NCAM), seem OT action on plasticity, whereas specific antagonists inhibited
to play an important role in glial–neuronal structural plastic- it (Langle et al. 2003). Furthermore, VP, which is structurally
ity. Whereas PSA-NCAM is abundant in developing tissues, related to OT, had no effect. The receptor-mediated action of
it is not the case in adult structures that express NCAM with the neuropeptide may explain why morphological changes in
little PSA. However, it continues to be expressed strongly in the HNS are restricted to its OT system because OT receptors
adult systems that are able to undergo morphological plastic- occur on OT but not VP neurons in the HNS (Barberis and
ity (Bonfanti 2006; Bonfanti et al. 1992; Seki and Arai 1993). Tribollet 1996). Interestingly, OT receptors are also expressed
In the HNS, PSA-NCAM is made by both astrocytes and neu- by astrocytes (Guenot-Di Scala and Strosser 1992).
rons throughout life (Kiss et al. 1993; Theodosis et al. 1991, It is likely that OT does not act alone when considering the
1999). Whereas in some systems, such as the hippocampus, induction of the structural plasticity of the HNS. Among the
PSA-NCAM expression depends on synaptic activity (Kiss and cofactors that could be involved are estrogens. When given
Muller 2001), its expression in the HNS is constitutive and not alone or together with OT, estrogens facilitate morphological
very sensitive to neuronal activity. plasticity in vivo and in vitro (Langle et al. 2003; Montagnese
PSA-NCAM appears to be a prerequisite for the struc- et al. 1990; Theodosis et al. 2006). The action of these steroids
tural plasticity of the HNS. The specific enzymatic removal appears to be rapid, in agreement with the involvement of a
of PSA from NCAM in situ in the SON inhibits glial and membrane receptor. It is of note that steroids have a facilitat-
neuronal remodeling associated with lactation and chronic ing effect on the electrical activity of OT neurons (Israel and
dehydration (Theodosis et al. 1999). Similarly, morphological Poulain 2000), their secretory activity, and their gene expres-
changes occurring at the levels of axon and glia in the neuro- sion (Crowley and Amico 1993). Whether this is related to
hypophysis are compromised by the PSA enzymatic removal their reported effect on HNS morphological neuronal–glial
(Monlezun et al. 2005). This manipulation, however, has no remodeling remains to be elucidated. Although OT, with or
effect once the remodeling has already occurred. PSA-NCAM without steroids, appears essential for inducing the structural
is also very highly expressed in the GnRH system of rodents plasticity in the HNS, it is also possible that other factors are
(Bonfanti et al. 1992; Hoyk et al. 2001), sheep (Viguié et al. involved in such signaling. One of them is glutamate, which
2001), and primates (Perera et al. 1993). The mechanism by can serve as a bidirectional signal to transfer information
which PSA-NCAM plays a role in morphological plasticity between excited neurons and adjacent astrocytes. In vitro, the
is unknown, but it is very likely that large quantities of car- remodeling induced in the SON by OT and estrogen is indeed
bohydrate on cell membranes reduces adhesion (Rutishauser inhibited with a cocktail of metabotropic and ionotropic glu-
and Landmesser 1996), thereby favoring, if not enabling, tamate receptor antagonists (Langle et al. 2003), suggesting a
cells’ detachment from each other or the ECM. PSA-NCAM role for the excitatory amino acid in mediating hypothalamic
is then considered a permissive factor for morphological activity–dependent structural changes. Another candidate
plasticity because its expression is required for the changes signaling molecule is nitric oxide (NO). Electron micro-
to happen. scopic analysis of the external layer of the median eminence
indicates that NO regulates neuronal–glial structural plastic-
ity (De Serrano et al. 2004). Activation of endogenous NO
3.3 I N D U C T I V E FAC TO R S
release induced rapid structural remodeling, resulting in the
There must be other factors, specific to each plastic system, withdrawal of tanycyte endfeet and favoring access of GnRH
that act as specific stimuli, triggering cascades of intracellular terminals to the basal lamina. Other putative signaling can-
events and ultimately resulting in glial and neuronal reorga- didates for hypothalamic structural plasticity are purines,
nization. There are many potential candidates for such func- especially in the neurohypophysis, in which ATP is packed in
tions, including peptides, transmitters, steroids, and trophic secretory granules (Sperlagh et al. 1999). Once released from
factors that are released by neurons and/or glial cells. Presently, neurohypophysial terminals, ATP could then act on pituicytes
however, not much is known about these signaling pathways that express purinergic receptors (Loesch and Burnstock
and how they could operate to affect neural cell morphol- 2001; Rosso et al. 2002). Furthermore, adenosine, which is
ogy. In the HNS, the conditions in which plasticity is most produced from the metabolic breakdown of ATP, has been
striking, lactation and parturition, are associated with the shown to affect glial morphology, triggering stellation of cul-
massive peripheral release of OT, whose secretion within the tured pituicyte (Miyata et al. 1999; Rosso et al. 2002) (see also
hypothalamic nuclei is also augmented. Central OT acts as a chapter 25).
local modulator, affecting presynaptic and postsynaptic activ- Finally, there is strong evidence indicating that steroids
ity of OT neurons (de Kock et al. 2003; Israel et al. 2003a; themselves trigger the onset of morphological changes.
Jourdain et al. 1998). Most important, intracerebroventricular As noted, estrogen can substitute for OT, at least in vitro,

518 • NEUROGLIA
to induce neuronal–glial changes in the SON (Langle et al. 4.1 D I FF US I O N I N T H E
2003; Theodosis et al. 2006). In the arcuate nucleus, admin- E X T R AC E L LU L A R S PAC E
istration of estradiol to match plasma levels measured at Modification of the glial environment of neurons results in
proestrus causes glial rearrangements similar to those detected important changes in the ECS volume and geometry. As a
at that specific period of the estrous cycle (Garcia-Segura consequence, extrasynaptic transmission mediated by the
et al. 1994a). Because GnRH neurons do not express estrogen diffusion of molecules is profoundly altered. Similarly, the
receptors, such a steroid action has to be mediated either by absence of fine astrocytic processes around neuronal elements
presynaptic terminals and/or astrocytes (Witkin et al. 1991). and synapses in the SON of lactating rats produces changes
Indeed, in this area of the hypothalamus, astrocytes are not in ECS properties. This hypothesis was tested in the SON
only affected by estrogen action, but they also release factors of virgin and lactating rats (Piet et al. 2004) using the real-
that could affect neuronal–glial interactions. Thus, gonadal time tetramethylammonium (TMA+) iontophoretic method
steroids facilitate the astrocytic expression of growth fac-
(Nicholson and Sykova 1998). This approach permits mea-
tors of the epidermal growth factor family and their tyrosine
surement of diffusion parameters such as tortuosity, which is a
kinase receptors, thereby contributing to neuronal–glial sig-
measure of restriction on diffusion in the tissue in comparison
naling occurring at the beginning of puberty (Ojeda and Ma
with an obstacle-free medium, volume fraction which corre-
1999). Furthermore, astrocytes and tanycytes of the median
sponds to the volume of tissue occupied by the ECS, and non-
eminence release factors such as PGE2, which contribute to
specific uptake. In virgin rats, diffusion in the SON was not
the estrogen-induced structural plasticity of GnRH terminals
equivalent in all directions, a condition known as anisotropy,
in the external layer of the median eminence ( Garcia-Segura
with tortuosity being hindered less along the ventrodorsal
and McCarthy 2004; Ojeda and Ma 1999).
axis (Fig. 41.2). This situation probably reflects the particular
ventrodorsal orientation of most astrocytic processes in this
structure (Bonfanti et al. 1993; Israel et al. 2003b). Glial with-
4 CONSEQUENCES OF THE GLIAL drawal in the SON of lactating animals caused a significant
S T RU C T U R A L P L A S T I C I T Y reduction of volume fraction and tortuosity. As a result, dif-
fusion became equivalent in all the planes (isotropy), indicat-
Whereas some factors involved in the anatomical remodeling ing that the glial remodeling not only facilitated diffusion in
of the SON have been identified (Theodosis 2002), the func- the ECS, but also modified the geometry of the nucleus. The
tional consequences of such changes have remained hypotheti- reduction in volume fraction can be explained by the hypertro-
cal for a long time. It was proposed that the increased number of phy of the neurons filling the empty space resulting from glial
synapses represented a compensatory mechanism to maintain withdrawal. Such modifications of the diffusion parameters of
a stable level of synaptic density for hypertrophied neurons. the SON should enhance largely the range of action of diffus-
Electrophysiological recordings performed in the SON at dif- ing substances and their local concentrations (Fig. 41.3).
ferent stages of the reproductive cycle confirmed this hypoth-
esis (Brussaard et al. 1999). Regarding the relative absence of
4.2 G LU TA M AT E U P TA K E
astrocytic processes in the vicinity of neuronal elements and
synapses, this has important functional consequences in view Because glial cells play a key role in glutamate clearance, with-
of the various roles played by glia in the CNS. drawal of astrocytic processes in the vicinity of glutamatergic

real time iontophoretic TMA+ method

Virgin rat Lactating rat

TMA+
[TMA+]e λx = 1.39 λx = 1.35
λy = 1.49 λy = 1.38
Δ [TMA+] = 1 mM

λz = 1.53 λz = 1.36

SON α = 0.30 α = 0.20

optic chiasm

x z
60 s
y (I = 200 nA)

Figure 41.2 Real-Time Measurements of Diffusion Parameters. Real-time measurements of SON diffusion properties can be obtained through ionto-
phoresis applications of TMA+ in acute slices (left panel) and its detection by an ion-sensitive electrode. In virgin rats (middle panel), the concentration
profiles varies according to the axis (x, y, and z), yielding different tortuosity (O) values. Such anisotropy disappears in the SON of lactating rats, in
which tortuosity become equivalent in all direction (right panel). The volume fraction (D) is reduced once structural neuronal–glial remodeling has
occurred. Adapted from Piet et al. 2004.

M O D U L AT I O N O F N E U R O E N D O C R I N E SYS T E M S • 519
unstimulated Piet et al. 2003) and chronically dehydrated (Boudaba et al.
2003) animals in which a similar neuroglial reorganization
SON neuron occurs (Theodosis 2002). As expected from delayed glutamate
clearance, mGluR tonic activity is significantly enhanced under
glutamate conditions of reduced glial coverage, resulting in reduced
GLT-1
terminal synaptic efficacy at glutamatergic inputs.
astrocytic process
Conversely, the elevation in glutamate concentrations
associated with anatomical remodeling is apparently not suffi-
mGluR GABA
terminal
cient to modulate mGluRs at GABAergic synapses (Piet et al.
2003). At GABA synapses, tonic activation of mGluRs by
ambient glutamate was detected neither in virgin rats nor lac-
SON neuron
tating animals. It seems then that basal glutamate release is not
sufficient to activate glutamate receptors located at a distance
from excitatory inputs. This is consistent with previous stud-
stimulated
ies describing the regulation of GABAergic transmission by
synaptically released glutamate, a process known as heterosyn-
SON neuron
aptic modulation. Such modulation depends on the activity
of glutamatergic inputs, so that glutamate levels in the ECS
glutamate have to reach sufficient concentrations to saturate glutamate
terminal
transporters, thereby enabling diffusion to remote receptors.
In the SON of virgin rats, such heterosynaptic depression
GABA of GABAergic transmission occurs when short condition-
terminal
ing trains of stimulation were applied to glutamatergic fibers
(Oliet et al. 2004). This inhibition has a presynaptic origin
SON neuron and is caused by the activation of mGluRs present on inhibi-
tory terminals. Because this type of modulation relies on the
diffusion of glutamate in the ECS, it should be sensitive to
Figure 41.3 Structural Plasticity in the Supraoptic Nucleus and Its anatomical remodeling of the SON in lactating animals that
Consequences On Transmitter Concentrations and Diffusion. Diagrams
illustrating the glial structural remodeling in the SON of stimulated
impairs glutamate uptake. In agreement with this hypothesis,
rats and its consequences on synaptic and extrasynaptic transmission. intersynaptic cross-talk between glutamate and GABA syn-
In unstimulated animals (virgin or normally hydrated rats), astrocytic apses is greatly facilitated in the SON of lactating rats (see
processes are enwrapping synapses and neurons, thereby preventing Fig. 41.3) under conditions of reduced glial coverage of neu-
neurotransmitters such as glutamate to diffuse in the ECS. In stimulated
animals (lactating or chronically dehydrated), glial withdrawal promotes rons and synapses (Oliet et al. 2004).
an elevation of glutamate levels around glutamatergic synapses and
enhanced activation of presynaptic autoreceptors (mGluRs). Under con-
ditions of intense glutamatergic fiber stimulation, glutamate could diffuse 4.3 G L I OT R A NS M I S S I O N
away from its release sites and affect remote receptors such the mGluRs Anatomical changes in the neuronal–glial relationship mod-
located on GABAergic terminals. This extrasynaptic form of communi-
cation is likely to be favored by the facilitated diffusion resulting from ify the concentration and diffusion in the ECS of the various
morphological remodeling. Adapted from Oliet and Piet 2004. gliotransmitters released from astrocytes (see chapters 34 and
35). The retraction of glial processes occurring in the SON
and paraventricular (PVN) nuclei of the hypothalamus during
lactation and dehydration, respectively, results in remarkable
release sites may have important consequences on the extra- changes in astrocyte–neuron communication. In the SON of
cellular levels of the excitatory amino acid. This in turn may lactating rats, the concentration of d-serine within the synap-
cause change in synaptic transmission and neuronal excitabil- tic cleft is dramatically reduced (Fig. 41.4), as revealed by the
ity. Because astrocytes play a major role in the uptake of glu- reduced activity of NMDA receptors (Panatier et al. 2006).
tamate (see chapter 35), removal of fine astrocytic processes This has severe consequences on the NMDA receptor-depen-
should result in deficient or delayed glutamate clearance that dent forms of synaptic plasticity such as long-term potentiation
could have a tremendous impact on synaptic transmission. (LTP) and depression (LTD). In the SON of lactating rats,
Accordingly, inhibition of glutamate transporters in the SON the activity dependence of these processes is shifted toward
of virgin rats yielded augmented levels of glutamate in the higher values, so it takes a much stronger synaptic activity to
ECS, enhanced tonic activation of presynaptic mGluRs and induce LTP in lactating rats compared with virgin animals (see
reduced synaptic efficacy (Boudaba et al. 2003; Oliet et al. Fig. 41.4). This is because fewer NMDA receptors are available
2001). To assess whether the removal of fine glial processes for activation and, consequently, less Ca2+ enters the postsyn-
enveloping synapses and neurons indeed affected the local aptic neurons. Both NMDA receptor activity and the ability
concentration of glutamate, the tonic activity of presynaptic to induce LTP and/or LTD in lactating rats are completely res-
mGluRs was evaluated under conditions of reduced astrocytic cued when exogenous d-serine is applied to the tissue. Because
coverage of neurons in the SON of lactating (Oliet et al. 2001; d-serine release from astrocytes requires the activation of glial

520 • NEUROGLIA
UNSTIMULATED STIMULATED

synaptic terminal synaptic terminal

astrocyte astrocyte

glutamate glutamate

D-serine D-serine

NMDAR NMDAR

OT neuron OT neuron

Figure 41.4 Gliotransmission in Lactating Rats. Under unstimulated conditions (left panel), astrocytes provide d-serine to the glutamatergic
synapses of OT neurons, enabling NMDA receptor activation. In lactating rats (stimulated; right panel), the glial withdrawal causes a deficiency in
d-serine supply, impairing NMDA receptor activation.

glutamatergic receptors (Mothet et al. 2005), it is likely that analyses demonstrated that GABAergic, glutamatergic, and
such release is less stimulated under these conditions because noradrenergic inputs undergo remodeling in relation to stim-
of the increased distance between glial and neuronal elements. ulation of OT secretion, with the most important increases
Because of this anatomical change, even if d-serine release occurring for GABAergic inputs (El Majdoubi et al. 1997;
occurs, concentrations of the d-amino acid reaching the syn- Gies and Theodosis 1994). In the SON of lactating rats, 50%
aptic cleft would be largely reduced compared with control of all axosomatic and axodendritic synapses are GABAergic,
conditions. As a result, the activity dependence of long-term compared with 35% in virgin animals. Such synaptogen-
synaptic changes is shifted toward higher values (Fig. 41.5). esis was reproduced in vitro in acute hypothalamic slices
A similar observation has been made in the PVN of dehy- that include the SON. In this model, synapse formation was
drated rats in which the release of ATP from glial cells no found to be very rapid, occurring within 1 hour and resulting
longer activates P2X receptors on magnocellular neurons, in the formation of inhibitory synapses that were functional
thereby compromising synapse strengthening through AMPA (Theodosis et al. 2006). In the arcuate nucleus of adult female
receptor insertion (Gordon et al. 2005). Here again, this form rats, there are also important synaptic changes accompanying
of synaptic plasticity is rescued by providing exogenous ATP the astrocytic remodeling (Garcia-Segura et al. 1994a). The
to the tissue. Taken together, these data indicate that remodel- number of axosomatic GABAergic synapses on arcuate neu-
ing of glial processes has tremendous consequences on excit- rons falls significantly between the morning and afternoon
atory transmission and long-term synaptic plasticity (Bains of proestrus, remains low throughout estrus, and comes back
and Oliet 2007). to baseline levels on metestrus, to fall again at proestrus. The
inverse correlation between the level of glial ensheathment and
the number of synaptic inputs in relation to steroid levels has
4.4 SY NA P TO G E N E S I S
been reported not only in rats but in nonhuman primates as
In the magnocellular nuclei of rodents, variations in astro- well (Garcia-Segura et al. 1994a; Witkin et al. 1991). Whether
cytic coverage of neuronal somata and dendrites are always such synapse turnover is directly related to the glial remodel-
accompanied by synapse turnover. The most obvious change is ing occurring in those different hypothalamic areas remains to
an increased number of terminals impinging onto more than be determined.
one postsynaptic element simultaneously. They are referred
to as multiple or shared synapses. In the magnocellular nuclei,
their incidence increases during conditions of enhanced OT 5 C O N C LU S I O N S
release coincident with the reduction of astrocytic coverage of
neuronal elements (Theodosis et al. 2005). In the OT system, The hypothalamus is made of several neuroendocrine sys-
augmentation of synaptic densities also concerns terminals tems that intervene in the regulation of many vital processes.
making single synaptic contact. Ultrastructural morphometric Their morphological reorganization is now a well-established

M O D U L AT I O N O F N E U R O E N D O C R I N E SYS T E M S • 521
A Virgin Lactating B
Control Control D-serine
DAAO D-serine LTP + –
250 250 Pairing
Pairing
200 200
amplitude (%)

amplitude (%)
Virgin
150 150

100 100
Lactating
50 50

0 0 LTD
–10 0 10 20 30 –10 0 10 20 30 low high
time (min) time (min) synaptic stimulation

Figure 41.5 Glia-Dependent Plasticity. A. Pairing synaptic stimulation with membrane depolarization causes long-term potentiation (LTP) in
the supraoptic nucleus of virgin rats (left panel; control; black dots), whereas the same protocol applied in lactating animals causes long-term depres-
sion (LTD) (right panel, control; black dots). Long-term potentiation can be restored in lactating rats by supplying d-serine to the slices (right panel;
d-serine; empty dots) and is switched into LTD in virgin animals when d-serine is degraded with D-amino acid oxidase (left panel; DAAO; empty
dots). B. At these synapses, the induction of plasticity and its magnitude depends on the rate of synaptic stimulation according to the model described
by Biennenstock, Cooper, and Munro (black curve; virgin). Glial withdrawal in the SON (red curve, lactating) causes a rightward shift of the activity
dependence of synaptic plasticity. This relationship between plasticity and synaptic stimulation is governed by the availability of d-serine to NMDA
receptors, a process that depends on the glial environment of synapses. From Bains and Oliet 2007.

phenomenon. Because these systems, especially the HNS and accumulating glutamate in the extracellular environment,
median eminence, are easily accessible in vivo and in vitro, via its heterosynaptic actions, can inhibit GABAergic trans-
they have been used as model to analyze the importance of mission in the vicinity of glutamatergic inputs (Piet et al. 2003,
neuronal–glial remodeling not only at the level of synaptic 2004), leading to a local disinhibition of the somatodendritic
function, as discussed, but in the more general context of sys- compartment (Mitchell and Silver 2000).
tem physiology as well. This is particularly clear in the case Also as described, the presence or absence of astrocytic
of the OT system, during its highly characteristic activity at processes has important consequences on the concentration
lactation. of gliotransmitters such as d-serine, which affect NMDA
Peripheral information during suckling is transmitted to receptor activation, intracellular Ca2+ concentrations, and
OT neurons through proximal glutamatergic afferents. During ultimately phenomena of synaptic plasticity such as LTP and
the milk ejection reflex, for example, OT neurons display high LTD (Panatier et al. 2006). During lactation, when fewer
frequency bursts of action potentials (Poulain and Wakerley NMDA receptors are occupied by d-serine, the threshold for
1982) that are triggered by bursts of glutamatergic postsynap- inducing LTP is elevated, making it more difficult for syn-
tic potentials (Israel et al. 2003a; Jourdain et al. 1998). As dis- apses to become potentiated. Making potentiation harder to
cussed, patch clamp recordings provided convincing evidence achieve may favor very active synapses, such those communi-
that excitatory information transmitted by glutamatergic cating the suckling stimulus to OT neurons, whereas at others
inputs is affected by glial remodeling (Oliet et al. 2004). Thus, less active, synaptic strength would rather be decreased or left
the reduced glial coverage of OT neurons and their synapses unchanged by the afferent drive activity. It should also be kept
during lactation results in increased levels of glutamate in the in mind that the increased number of synapses associated with
ECS, which in turn augment the tonic inhibition exerted by reduced glial coverage in the magnocellular nuclei at lactation
presynaptic mGluRs on glutamate release (Oliet et al. 2001). (El Majdoubi et al. 1997) facilitate enhanced OT firing as well.
On first sight, it seems paradoxical that these inputs exhibit It is obvious that an increased number of glutamatergic inputs
a lower probability of release at a time when OT neurons are potentiate such firing, just like the additional noradrenergic
strongly activated. However, presynaptic inhibition is not a synapses (Michaloudi et al. 1997) that provide an excitatory
linear process in the sense that its efficiency depends on pre- input to OT neurons, both on their own (Day et al. 1984) and
synaptic activity. A tonic presynaptic inhibition mediated by through their synergistic action with glutamate (Daftary et al.
mGluR activation is likely to be overcome by the facilitation 1998; Parker and Crowley 1993). As for the highly significant
of probability of release associated with the intraterminal increase in the number of inhibitory synapses, their action,
Ca2+ strong increase that occurs during high-frequency trains at first glance, might appear incompatible with enhanced
of action potentials in glutamatergic terminals at the time of excitability. A likely possibility is that the increase number
milk ejection. Information transmitted via high-frequency of inhibitory inputs may serve to prevent the neurons from
excitatory synaptic activity, therefore, would be less affected being stimulated by factors other than those relevant to partu-
by a negative glutamate feedback than information transmit- rition and lactation, attenuating the response of OT neurons
ted by low or moderate frequency activities. This may serve as to stimuli other than suckling (Fénelon et al. 1994; Lightman
a high-pass filter to increase signal-to-noise ratio for informa- and Young 1989). This mechanism could also contribute to
tion carried by high-frequency activities. On the other hand, maintain membrane potential of OT neurons at a depolarized

522 • NEUROGLIA
level, thereby facilitating their subsequent synchronous activa- AC K N OW L E D G M E N T S
tion just before milk ejection (Moos 1995; Voisin et al. 1995).
In addition to these phenomena in the hypothalamus, The author wishes to thank Dionysia Theodosis and
glial–axonal changes in the neurohypophysis and the median Dominique Poulain for their continual support, wise advice,
eminence appear to favor neurosecretion. Retraction of glial and persistent encouragement throughout the last 15 years.
processes from around neurosecretory axons and the perivas-
cular space promote paracrine actions of the secreted peptides.
Moreover, as noted, by removing a physical barrier from the REFERENCES
perivascular spaces, such plasticity facilitates diffusion of the
secreted hormones into fenestrated capillaries and thus the Bains JS, Oliet SH. 2007. Glia: they make your memories stick! Trends
general circulation. Neurosci 30:417–424.
Barberis C, Tribollet E. 1996. Vasopressin and oxytocin receptors in the
Although the consequences of the glial remodelings central nervous system. Crit Rev Neurobiol 10:119–154.
occurring in different neuroendocrine centers have not been Bobak JB, Salm AK. 1996. Plasticity of astrocytes of the ventral glial
completely determined, and the detailed molecular processes limitans subjacent to the supraoptic nucleus. J Comp Neurol
underlying such forms of structural plasticity remain to be 376:188–197.
identified, these phenomena have served as remarkable mod- Bonfanti L. 2006. PSA-NCAM in mammalian structural plasticity and
neurogenesis. Prog Neurobiol 80:129–164.
els to improve our understanding of glial cells and their contri- Bonfanti L, Olive S, Poulain DA, Theodosis DT. 1992. Mapping of the
bution to neuronal functions. distribution of polysialylated neural cell adhesion molecule through-
out the central nervous system of the adult rat: an immunohistochem-
ical study. Neuroscience 49:419–436.
Bonfanti L, Poulain DA, Theodosis DT. 1993. Radial glia-like cells in
6 S U M M A RY A N D P E R S P E C T I VE S the supraoptic nucleus of the adult rat. J Neuroendocrinol 5:1–6.
Bonfardin VD, Fossat P, Theodosis DT, Oliet SHR. 2010.
Investigations on neuronal–glial interactions carried Glia-dependent switch of kainate receptor presynaptic action.
out in the hypothalamus have provided very impor- J Neurosci 30:985–995.
tant insights regarding the contribution of glial cells Boudaba C, Linn DM, Halmos KC, Tasker JG. 2003. Increased tonic
activation of presynaptic metabotropic glutamate receptor in the rat
to the physiology of neurons and their dynamic inter- supraoptic nucleus. J Physiol 551:815–823.
actions. The hypothalamo-neurohypophysial sys- Brussaard AB, Devay P, Leyting-Vermeulen JL, Kits KS. 1999. Changes
tem has proved to be an excellent model to study these in properties and neurosteroid regulation of GABAergic synapses in
processes because of the remarkable activity-dependent the supraoptic nucleus during the mammalian female reproductive
remodeling that occurs during physiological responses such cycle. J Physiol 516:513–524.
Chapman DB, Theodosis DT, Montagnese C, Poulain DA, Morris JF.
as lactation, parturition, and dehydration. The magnitude of 1986. Osmotic stimulation causes structural plasticity of neurone-glia
this remodeling is so important that it was possible to study relationships of the oxytocin but not vasopressin secreting neurones
it a time when structural analysis was limited to electron in the hypothalamic supraoptic nucleus. Neuroscience 1:679–686.
microscopy. The physiological changes that affect the glial Crowley RS, Amico JA. 1993. Gonadal steroid modulation of oxytocin
environment of neurons during such structural plasticity and vasopressin gene expression in the hypothalamus of the osmoti-
cally stimulated rat. Endocrinology 133:2711–2718.
has revealed the crucial roles played by astrocytes, including Daftary SS, Boudaba C, Szabó K, Tasker JG. 1998. Noradrenergic exci-
the regulation of glutamate concentration and diffusion in the tation of magnocellular neurons in the rat hypothalamic paraven-
extracellular space and the supply of key signaling molecules tricular nucleus via intranuclear glutamatergic circuits. J Neurosci
such as ATP and serine for controlling synaptic strength. 18:10619–10628.
Day TA, Ferguson AV, Renaud LP. 1984. Facilitatory influence of nora-
The ongoing development of imaging techniques has per- drenergic afferents on the excitability of rat paraventricular nucleus
mitted to observe similar types of structural neuronal–glial neurosecretory cells. J Physiol 355:237–249.
remodeling, although of smaller extent, in other major de Kock CPJ, Wierda KDB, Bosman LWJ, Min R, Koksma JJ,
brain areas such as the brainstem, hippocampus, or cortex. Mansvelder HD, et al. 2003. Somatodendritic secretion in oxyto-
Availability techniques such as super resolution microscopy cin neurons is upregulated during the female reproductive cycle.
J Neurosci 23:2726–2734.
permitting the visualization of thin glial processes should De Serrano S, Estrella C, Loyens A, Cornea A, Ojeda SR, Beauvillain
allow in the near future the study of the morphofunctional JC, et al. 2004. Vascular endothelial cells promote acute plastic-
plasticity of the tripartite synapse at the nanoscopic level. This ity in ependymoglial cells of the neuroendocrine brain. J Neurosci
should provide unexpected details of the intricate and dynamic 24:10353–10363.
association among glia, neurons, and the extracellular matrix. El Majdoubi M, Poulain DA, Theodosis DT. 1997. Lactation-induced
plasticity in the supraoptic nucleus augments axodendritic and axoso-
The time courses of these remodelings as well as the intracel- matic gabaergic and glutamatergic synapses: an ultrastructural analy-
lular cascades involved await description and identification. sis using the dissector method. Neuroscience 80:1137–1147.
At a more macroscopic level, the functional consequences Fénelon VS, Theodosis DT, Poulain DA. 1994. Fos synthesis in
of the glial remodeling on the physiology of the neuroen- identified magnocellular neurons varies with phenotype, stimulus,
docrine centers are still debated. Whether such forms of location in the hypothalamus and reproductive state. Brain Res 662:
165–177.
structural plasticity are essential processes for lactation, dehy- Garcia-Segura LM, Chowen JA, Parducz A, Naftolin F. 1994a. Gonadal
dration, the estrous cycle, or the circadian rhythm remains to hormones as promoters of structural synaptic plasticity: cellular
be answered. mechanisms. Prog Neurobiol 44:279–307.

M O D U L AT I O N O F N E U R O E N D O C R I N E SYS T E M S • 523
Garcia-Segura LM, Luquin S, Parducz A, Naftolin F. 1994b. Gonadal Langle SL, Poulain DA, Theodosis DT. 2003. Induction of rapid,
hormone regulation of glial fibrillary acidic protein immunoreactiv- activity-dependent neuronal-glial remodeling in the adult hypothala-
ity and glial ultrastructure in the rat neuroendocrine hypothalamus. mus in vitro. Eur J Neurosci 18:206–214.
Glia 10:59–69. Lavialle M, Begue A, Papillon C, Vilaplana J. 2001. Modifications or
Garcia-Segura LM, McCarthy MM. 2004. Role of glia in neuroendo- retinal afferent activity induce changes in astroglial plasticity in the
crine function. Endocrinology 145:1082–1086 hamster circadian clock. Glia 34:88–100.
Gerhold LM, Wise PM. 2006. Vasoactive intestinal polypeptide reg- Lavialle M, Serviere J. 1993. Circadian fluctuations in GFAP distribu-
ulates dynamic changes in astrocyte morphometry: impact on tion in the Syrian hamster suprachiasmatic nucleus. Neuroreport
gonadotropin-releasing hormone neurons. Endocrinology 147: 4:1243–1246.
2197–2202. Lightman SL, Young WS 3rd. 1989. Lactation inhibits stress-mediated
Gerics B, Szalay F, Hajos F. 2006. Glial fibrillary acidic protein immuno- secretion of corticosterone and oxytocin and hypothalamic accumu-
reactivity in the rat suprachiasmatic nucleus: circadian changes and lation of corticotropin-releasing factor and enkephalin messenger
their seasonal dependence. J Anat 209:231–237. ribonucleic acids. Endocrinology 124:2358–2364.
Gies U, Theodosis DT. 1994. Synaptic plasticity in the rat supraoptic Loesch A, Burnstock G. 2001. Immunoreactivity to P2X6 receptors in
nucleus during lactation involves GABA innervation and oxytocin the rat hypothalamo-neurohypophysial system: an ultrastructural
neurons: a quantitative immunocytochemical analysis. J Neurosci study with extravidin and colloidal gold-silver labelling. Neuroscience
14:2861–2869. 106:621–631.
Gordon GR, Baimoukhametova DV, Hewitt SA, Rajapaksha WR, Fisher Ludwig M. 1998. Dendritic release of vasopressin and oxytocin.
TE, Bains JS. 2005. Norepinephrine triggers release of glial ATP to J Neuroendocrinol 10:881–895.
increase postsynaptic efficacy. Nat Neurosci 8:1078–1086. Michaloudi HC, el Majdoubi M, Poulain DA, Papadopoulos GC,
Guenot-Di Scala D, Strosser MT. 1992. Oxytocin receptors on cultured Theodosis DT. 1997 The noradrenergic innervation of identi-
astroglial cells. Kinetic and pharmacological characterization of fied hypothalamic magnocellular somata and its contribution to
oxytocin-binding sites on intact hypothalamic and hippocampic cells lactation-induced synaptic plasticity. J Neuroendocrinol 9:17–23.
from foetal rat brain. Biochem J 284:491–497. Mitchell SJ, Silver RA. 2000. Glutamate spillover suppresses inhibition
Hatton GI, Perlmutter LS, Salm AK, Tweedle CD. 1984. Dynamic by activating presynaptic mGluRs. Nature 404:498–502.
neuronal-glial interactions in hypothalamus and pituitary: impli- Miyata S, Furuya K, Nakai S, Bun H, Kiyohara T. 1999. Morphological
cations for control of hormone synthesis and release. Peptides plasticity and rearrangement of cytoskeletons in pituicytes cultured
5:121–118. from adult rat neurohypophyses. Neurosci Res 33:299–306.
Hatton GI. 1999. Astroglial modulation of neurotransmitter/peptide Monlezun S, Ouali S, Poulain DA, Theodosis DT. 2005. Polysialic acid
release from the neurohypophysis: present status. J Chem Neuroanat is required for active phases of morphological plasticity of neurosecre-
16:203–222. tory axons and their glia. Mol Cell Neurosci 29:516–524.
Hoyk Z, Parducz A, Theodosis DT. 2001. The highly sialylated isoform Montagnese C, Poulain DA, Vincent JD, Theodosis DT. 1988. Synaptic
of the neural cell adhesion molecule is required for estradiol-induced and neuronal-glial plasticity in the adult oxytocinergic system in
morphological synaptic plasticity in the adult arcuate nucleus. Eur J response to physiological stimuli. Brain Res Bull 20:681–692.
Neurosci 13:649–656. Montagnese C, Poulain DA, Theodosis DT. 1990. Influence of ovarian
Hu B, Bourque CW. 1992. NMDA receptor-mediated rhythmic burst- steroids on the ultrastructural plasticity of the adult supraoptic nucleus
ing activity in rat supraoptic nucleus neurones in vitro. J Physiol induced by central administration of oxytocin. J Neuroendocrinol
458:667–687. 2:225–231.
Hussy N, Deleuze C, Brès V, Moos FC. 2000. New role of taurine as an Moos FC. 1995. GABA-induced facilitation of the periodic bursting
osmomediator between glial cells and neurons in the rat supraoptic activity of oxytocin neurones in suckled rats. J Physiol 488:103–114.
nucleus. Adv Exp Med Biol 483:227–237. Mothet JP, Pollegioni L, Ouanounou G, Martineau M, Fossier P,
Israel JM, Le Masson G, Theodosis DT, Poulain DA. 2003a. Glutamate Baux G. 2005. Glutamate receptor activation triggers a
afferent input governs periodicity and synchronisation of bursting elec- calcium-dependent and SNARE protein-dependent release of the
trical activity in oxytocin neurons. Eur J Neurosci 17:2619–2629. gliotransmitter D-serine. Proc Natl Acad Sci U S A 102:5606–5611.
Israel JM, Poulain DA. 2000. 17 beta-oestradiol modulates in vitro elec- Nicholson C, Sykova E. 1998. Extracellular space structure revealed by
trical properties and responses to kainate of oxytocin neurones in lac- diff usion analysis. Trends Neurosci 21:207–215.
tating rats. J Physiol 524:457–470. Ojeda SR, Ma YJ. 1999. Glial-neuronal interactions in the neuroendo-
Israel JM, Schipke CG, Ohlemeyer C, Theodosis DT, Kettenmann crine control of mammalian puberty: facilitatory effects of gonadal
H. 2003b. GABA-A receptor-expressing astrocytes in the supra- steroids. J Neurobiol 40:528–540.
optic nucleus lack glutamate uptake and receptor currents. Glia Oliet SHR, Piet R. 2004. Glial modulation of synaptic transmission:
44:102–110. insights from the hypothalamic supraoptic nucleus. J Neuroendocrinol
Jansen HT, Cutter C, Hardy S, Lehman MN, Goodman RL. 2003. 16:1–5.
Seasonal plasticity within the gonadotropin-releasing hormone Oliet SHR, Piet R, Poulain DA. 2001. Control of glutamate clearance and
(GnRH) system of the ewe: changes in identified GnRH inputs and synaptic efficacy by glial coverage of neurons. Science 292:923–926.
glial association. Endocrinology 144:3663–3676. Oliet SHR, Piet R, Poulain DA, Theodosis DT. 2004. Glial modulation
Jourdain P, Israel JM, Dupouy B, Oliet SHR, Allard M, Vitiello S, of synaptic transmission: insights from the hypothalamic supraoptic
et al. 1998. Evidence for a hypothalamic oxytocin-sensitive nucleus. Glia 47:258–267.
pattern-generating network governing oxytocin neurons in vitro. Panatier A, Theodosis DT, Mothet JP, Touquet B, Pollegioni L, Poulain
J Neurosci 18:6641–6649. DA, et al. 2006. Glial control of NMDA receptor activity contributes
King JC, Rubin BS. 1995. Dynamic alterations in luteinizing to synaptic memory. Cell 125:775–784
hormone-releasing hormone (LHRH) neuronal cell bodies and ter- Parker SL, Crowley WR. 1993. Stimulation of oxytocin release
minals of adult rats. Cell Mol Neurobiol 15:89–106. in the lactating rat by a central interaction of alpha 1-adrenergic
Kiss JZ, Muller D. 2001. Contribution of the neural cell adhesion mole- and alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid-
cule to neuronal and synaptic plasticity. Rev Neurosci 12:297–310. sensitive excitatory amino acid mechanisms. Endocrinology 133:
Kiss, JZ, Wang C, Rougon G. 1993. Nerve-dependent expression of high 2855–2860.
polysialic acid neural cell adhesion molecule in neurohypophysial Perera AD, Lagenaur CF, Plant TM. 1993. Postnatal expression of
astrocytes of adult rats. Neuroscience 53:213–222. polysialic acid-neural cell adhesion molecule in the hypothala-
Kleene R, Schachner M. 2004. Glycans and neural cell interactions. Nat mus of the male rhesus monkey (Macaca mulatta). Endocrinology
Rev Neurosci 5:195–208. 133:2729–2735.

524 • NEUROGLIA
Perlmutter LS, Hatton GI, Tweedle CD. 1984. Plasticity in the in Theodosis DT, Koksma JJ, Trailin A, Langle SL, Piet R, Lodder JC, et al.
vitro neurohypophysis: effects of osmotic changes on pituicytes. 2006. Oxytocin and estrogen promote rapid synapse formation of
Neuroscience 12:503–511. functional GABA synapses in the adult supraoptic nucleus. Mol Cell
Piet R, Bonhomme R, Theodosis DT, Poulain DA, Oliet SHR. 2003. Neurosci 31:785–794.
Modulation of GABAergic transmission by endogenous glutamate in Theodosis DT, MacVicar BA. 1996. Neuron-glia interactions in the
the rat supraoptic nucleus. Eur J Neurosci 17:1777–1785. hypothalamus and pituitary. Trends Neurosci 19:363–367.
Piet R, Vargova L, Sykova E, Poulain DA, Oliet SHR. 2004. Physiological Theodosis DT, Montagnese C, Rodriguez F, Vincent JD, Poulain DA.
contribution of the astrocytic environment of neurons to intersynap- 1986b. Oxytocin induces morphological plasticity in the adult
tic crosstalk. Proc Natl Acad Sci U S A 101:2151–2155. hypothalamo-neurohypophysial system. Nature 322:738–740.
Poulain DA, Wakerley JB. 1982. Electrophysiology of hypotha- Theodosis DT, Piet R, Poulain DA, Oliet SHR. 2004a. Neuronal,glial
lamic magnocellular neurones secreting oxytocin and vasopressin. and synaptic remodeling in the adult hypothalamus: functional con-
Neuroscience 7:773–808. sequences and role of cell surface and extracellular matrix adhesion
Prevot V, Hanchate NK, Bellefontaine N, Sharif A, Parkash J, Estrella molecules. Neurochem Int 45:491–501.
C, et al. 2010. Function-related structural plasticity of the GnRH Theodosis DT, Poulain DA. 1993. Activity-dependent neuronal-glial
system: a role for neuronal-glial-endothelial interactions. Front and synaptic plasticity in the adult mammalian hypothalamus.
Neuroendocrinol 31:241–258. Neuroscience 57:501–535.
Rosso L, Peteri-Brunbäck B, Vouret-Craviari V, Deroanne C, Troadec Theodosis DT, Rougon G, Poulain DA. 1991. Retention of embryonic fea-
JD, Th irion S, et al. 2002. Rho A inhibition is a key step in pituicyte tures by an adult neuronal system capable of plasticity: Polysialylated
stellation induced by A1-type adenosine receptor activation. Glia N-CAM in the hypothalamo-neurohypophysial system. Proc Natl
38:351–362. Acad Sci U S A 88:5494–5498.
Rutishauser U, Landmesser LT. 1996. Polysialic acid in the vertebrate Theodosis DT, Trailin A, Poulain DA. 2005. Remodeling of astro-
nervous system: a promoter of plasticity in cell-cell interactions. cytes, a prerequisite for synapse turnover in th adult brain? Insights
Trends Neurosci 19:422–427. from the oxytocin system of the hypothalamus. Am J Physiol
Seki T, Arai Y. 1993. Distribution and possible roles of the highly poly- 290:R1175–R1182.
sialylated neural cell adhesion molecule (NCAM-H) in the develop- Viguié C, Jansen HT, Glass JD, Watanabe M, Billings HJ, Coolen L,
ing and adult central nervous system. Neurosci Res 17:265–290. et al. 2001. Potential for polysialylated form of neural cell adhesio
Sperlagh B, Mergl Z, Juranyi Z, Vizi ES, Makara GB. 1999. Local reg- molecule-mediated neuroplasticity within the gonadotropin-releasing
ulation of vasopressin and oxytocin secretion by extracellular ATP hormone neurosecretory system of the ewe. Endocrinology
in the isolated posterior lobe of the rat hypophysis. J Endocrinol 142:1317–1324.
160:343–350. Voisin DL, Herbison AE, Poulain DA. 1995. Central inhibitory effects
Theodosis DT. 2002. Oxytocin-secreting neurons: a physiological model of muscimol and bicuculline on the milk ejection reflex in the anaes-
of morphological neuronal and glial plasticity in the adult hypothala- thetized rat. J Physiol 483:211–224.
mus. Front Neuroendocrinol 23:101–135. Waites CL, Craig AM, Garner CC. 2005. Mechanisms of vertebrate
Theodosis DT, Bonhomme R, Vitiello S, Rougon G, Poulain DA. 1999. synaptogenesis. Ann Rev Neurosci 28:251–274.
Cell surface expression of polysialic acid on NCAM is a prerequisite Witkin JW, Ferin M, Popilskis SJ, Silverman AJ. 1991. Effects of
for activity-dependent morphological neuronal and glial plasticity. gonadal steroids on the ultrastructure of GnRH neurons in the
J Neurosci 19:10228–102336. rhesus monkey: synaptic input and glial apposition. Endocrinology
Theodosis DT, Chapman DB, Montagnese C, Poulain DA, Morris JF. 129:1083–1092.
1986a. Structural plasticity in the hypothalamic supraoptic nucleus Wuarin JP, Dudek FE. 1993. Patch-clamp analysis of spontaneous synap-
at lactation affects oxytocin- but not vasopressin-secreting neurones. tic currents in supraoptic neuroendocrine cells of the rat hypothala-
Neuroscience 17:661–678. mus. J Neurosci 13:2323–2331.

M O D U L AT I O N O F N E U R O E N D O C R I N E SYS T E M S • 525
This page intentionally left blank
OLIGODENDROCY TES/SCHWANN CELLS

527
This page intentionally left blank
42.
MYELIN, IMPULSE CONDUCTION, AND THE
PATHOPHYSIOLOGY OF DEMYELINATION
Lakshmi Bangalore and Stephen G. Waxman

A B B R E VI AT I O N S 2 S T RU C T U R A L C O M P O N E N T S O F
T H E M Y E L I N AT E D AXO N
CNS central nervous system
EAE experimental autoimmune A myelinated nerve fiber consists of an axon ensheathed in seg-
encephalomyelitis ments of compact extensions of the cell membranes of myeli-
FGF fibroblast growth factor nating glia. In the peripheral nervous system (PNS), where
FHF FGF homologous factor axoglial interactions are much simpler than in the central ner-
GFAP glial fibrillary acidic protein vous system (CNS), myelinated fibers derive their myelin from
hESC human embryonic stem cells myelin-forming Schwann cells. A single Schwann cell can form
Kv voltage-gated potassium channels only one myelin segment, thus it takes multiple Schwann cells
md myelin-deficient to form successive segments of myelin sheath along a peripheral
Nav voltage-gated sodium channels axon (see also chapters 7, 44 and 45). In contrast, myelin in the
NCX sodium-calcium exchanger CNS is produced by oligodendrocytes that extend numerous
PLP proteolipid protein processes, each of which forms a segment of myelin around mul-
PNS peripheral nervous system tiple axons (Bjartmar et al. 1994; Hildebrand 1971). The oligo-
MS multiple sclerosis dendrocyte cell body typically does not circumferentially hug
its myelin sheaths but instead, maintains contact with its myelin
sheaths through thin cytoplasmic bridges (Bunge et al. 1961;
Hirano et al. 1968). This topographically tenuous connection
1 INTRODUCTION between the genomic and biosynthetic machinery within the
oligodendrocyte cell body and the myelin has been suggested to
Myelinated axons are essential for nervous system function underlie the scarcity of remyelination within the CNS. Studies
and represent examples of exquisite biological design. In have suggested that there is local biosynthesis of myelin in dis-
this chapter, we build upon our chapter in a prior edition tal parts of the oligodendroglial processes close to the regions
and discuss implications of the biological design of myeli- in which polyribosomes are present (Waxman 1984; Waxman
nated axons in normal and pathological conditions. Rapid et al. 1988). This is functionally important because it permits
transmission of nerve impulses confers a selective advan- a single oligodendrocyte to produce myelin sheaths of differ-
tage to species. Rapid impulse transmission can be achieved ent thickness around axons of different diameter in its vicinity
either by increasing axonal diameter or by myelination. In (Waxman et al. 1988).
axons without myelin, the speed of impulse conduction is The advantages of myelination are twofold. First, compact
proportional to the square root of axonal inner diameter. lipid-rich myelin provides high electrical resistance and low
Species such as cephalopods that lack myelin enlarged capacitance, which prevents current loss and enables rapid and
their axons substantially in order to achieve a faster rate efficient action potential conduction. Second, increased con-
of impulse conduction. The squid giant axons evolved to duction velocity allows for computational complexity within
be as large as 400 to 900Pm in diameter to support rapid a compact nervous system. The nodes of Ranvier that punctu-
conduction velocities—this specialization enabled early ate myelinated regions are the primary sites of ionic exchange
electrophysiologists to study the nerve impulse through between the interior and exterior of the nerve fiber. Internodal
recordings on single nerve fibers. The price of axonal distances that range from less than 100 Pm (small-diameter
gigantism, however, is spatial expansion, unaffordable fibers) to slightly over 1 mm (larger-diameter fibers) optimize
in higher species that have more axons within the same conduction velocity. Myelin, nodes of Ranvier, and internodal
cross-sectional area of one squid giant axon. Higher species distance specializations enable action potentials generated in
thus evolved another mechanism for increasing conduction the axon initial segment to propagate from node of Ranvier to
velocity by wrapping small-diameter axons in multiple lay- node of Ranvier in a saltatory manner, resulting in rapid and effi-
ers of myelin that enable nerve impulses to transmit in a cient impulse conduction at a conduction velocity that is pro-
rapid “saltatory” manner. portional to fiber diameter (Huxley and Stämpfli 1949; Tasaki

529
1959). Moreover, whereas conduction velocity in myelinated studies using a highly sensitive, quantitative electron micro-
fibers has a linear dependence on axon diameter, nonmyelinated scopic immunogold technique (SDS-digested freeze-fracture
fibers conduct with slower velocity that varies with the square replica-labeling) estimated the density of Nav1.6 at the nodes
root of axon diameter (Waxman and Bennett 1972). Therefore, to be approximately 350/Pm2, nearly twice that observed at the
above a critical diameter wherein the conduction velocity– axon initial segment (Lorincz and Nusser 2008).
diameter relationships intersect, myelinated fibers conduct Myelinated axons also express several proteins that modu-
impulses more rapidly than nonmyelinated fibers of the same late the availability and activity of Nav channels at the nodes.
diameter. This critical diameter of intersection is approximately A member of the fibroblast growth factor (FGF) homologous
0.2 Pm. Within the CNS, this is the diameter at which myelina- factor (FHF) subfamily, FHF2B, colocalizes with Nav1.6
tion is first seen (Waxman and Bennett 1972) (Fig. 42.1). at mature nodes of Ranvier in myelinated sensory fibers in
the dorsal root of the sciatic nerve (Wittmack et al. 2004).
However, retinal ganglion cells and spinal ventral horn motor
3 M O L E C U L A R C H A R AC T E R I S T I C S neurons have low expression of FHF2B, and no nodal FHF2B
O F T H E M Y E L I N AT E D AXO N staining within the optic nerve and ventral root, respectively
(Wittmack et al. 2004). Electrophysiological studies show
Although nonmyelinated axons generally display a uniform that the association of FHF2B with Nav1.6 results in increased
membrane structure that does not vary from one region to current density and a small depolarizing shift in channel avail-
another (Black et al. 1981), the myelinated axon membrane is ability (Rush et al. 2006; Wittmack et al. 2004). Wittmack
highly specialized with a spatially heterogeneous distribution et al. (2005) show association of p38 MAP kinase with Nav1.6
of voltage-sensitive ion channels and other proteins (Fig. 42.2). in rat brain tissue and demonstrate modulation of Nav1.6 cur-
High densities (approximately 1,000/Pm2) of voltage-gated Na+ rent in vitro by activated p38 MAP kinase via phosphoryla-
channels (Nav) cluster in the axon membrane at the nodes of tion of the L1 loop of Nav1.6 at serine 553. Recent studies
Ranvier (Ritchie and Rogart 1977; Waxman and Quick 1977). have reported that the ubiquitin ligase Nedd4–2 interacts
The internodal axon membrane beneath the myelin sheath has with a Pro-Ser-Tyr motif and a Pro-Gly-Ser-Pro motif in the
a much lower density of Na+ channels (<25/Pm2) (Ritchie and C terminus of Nav1.6, and reduces Nav1.6 current density in
Rogart 1977; Waxman and Quick 1977), and thus is unable to a p38-MAP kinase dependent manner. Interestingly, block-
support conduction at high frequencies. Nine different sodium ing Nedd4–2 from binding to the Pro-Ser-Tyr motif in the
channel subtypes have been discovered to date, with distinct C terminus results in stress-mediated increase in Nav1.6 cur-
molecular structures superimposed on an invariant overall motif rent density. Thus, Nav1.6 can be differentially modulated by
(four similar domains, each containing six membrane-spanning Nedd4–2 depending on the availability of the C-terminal
segments). The identities of sodium channel subtypes within Pro-Ser-Tyr motif to bind Nedd4–2 (Gasser et al. 2010).
various compartments of the myelinated axon are well under- Myelinated axons display a heterogeneous distribu-
stood. Sodium channel Nav 1.6 is the predominant sodium tion of several types of voltage-dependent K+ channels (Kv).
channel at the nodes of Ranvier (Caldwell et al. 2000). Recent Electrophysiological studies have revealed the expression of at
least three distinct K+ channel currents in myelinated axons—
8
the “fast” K+ current, a “slow” K+ current, and an inward rec-
tifier current (Röper and Schwarz 1989; Waxman and Ritchie
myelinated 1993). Fast K+ channels, which can be blocked by exogenous
6 4-aminopyridine (4-AP), have a reciprocal distribution to that
Conduction velocity (m/sec)

of the clustered nodal Na+ channels. They are present at rela-


tively low densities along the nodal axon membrane and high-
est density in the axon membrane beneath the myelin (Chiu
4 and Ritchie 1980, 1981; Foster et al. 1982; Kocsis et al. 1982).
Voltage-clamped experiments suggest that the density of fast K+
non-myelinated channels is maximal in the paranode, decreasing to one-sixth of
2 this paranodal density within the node and internode (Röper
and Schwarz 1989), but this study may not have accurately dif-
ferentiated between the paranodal and juxtaparanodal regions.
Immunocytochemical studies have shown high-density
0 1 2
clusters of Kv1 channels (Kv1.1 and Kv1.2; Shaker/KCNA
3 4
subfamily of voltage-dependent K+ channels) at the juxtapara-
Fiber diameter (μm)
nodes of myelinated axons (Rasband et al. 1998; Vabnick and
Figure 42.1 The relationships between conduction velocity (Y-axis) and Shrager 1998; Wang et al. 1993). More recently, studies have
diameter (X-axis), for myelinated (solid line) and nonmyelinated (dashed identified Kv1 channels enriched at the axon initial segments
line) axons are superimposed. Above a diameter of 0.2Pm, at which the wherein they form macromolecular complexes with a variety
two relationships cross, myelinated axons conduct more rapidly than
nonmyelinated axons of the same size. 0.2 Pm is, in fact, the diameter of of cell-adhesion molecules and cytoskeletal proteins (Goldberg
the smallest myelinated axon within the CNS. Modified from Waxman et al. 2008; Kole et al. 2007; Ogawa and Rasband 2008). Recent
and Bennett 1972. studies have also identified Kv channels KCNQ2 and KCNQ3

530 •
g g
Na K
AXON

OLIGO./S.C

Figure 42.2 Schematic model of ion channel organization of the myelinated fiber. gNa+ sodium conductance; gK fast potassium conductance. Sodium
channels gNa are clustered at the node of Ranvier. In contrast, fast potassium channels, responsible for repolarization of the action potential,
are present in the internodal axon membrane.

of the Kv7 (KCNQ) subfamily, which are slowly activated by demyelinated (former internodal) axon regions may restore the
depolarization, are present at axon initial segments and nodes capability to support action potential conduction in the absence
of Ranvier (Devaux et al. 2004; Pan et al. 2006). The seques- of myelin (Black et al. 1987; Dugandzija-Novakovic et al.
tration of KCNQ2 and KCNQ3 to the axon initial segments 1995; England et al. 1990; Foster et al. 1980). Unfortunately,
and nodes of Ranvier is dependent on their binding to the this may have deleterious consequences in terms of placing an
cytoskeletal scaffolding protein ankyrin G, which is essential increased metabolic load on axons. Pharmacological blockade
for the assembly of the axon initial segment and maintenance of fast K+ channels that are unmasked by demyelination is also
of neuronal polarity (Pan et al. 2006). Another subfamily expected to increase the safety factor (i.e., reliability) for con-
of Kv channels, the Kv3 channels also present within myeli- duction in demyelinated axons (Bowe et al. 1987; Kocsis et al.
nated axons and contribute to high-frequency repetitive firing 1987).
(Devaux et al. 2003; Rudy and McBain 2001). A unique splice
variant of the Kv3.1 gene, Kv3.1b, is found in a subset of CNS
and PNS nodes of Ranvier although it is more highly expressed 4 AC T I O N P OT E N T I A L
in CNS nodes wherein it colocalizes with Nav channels, partic- E L E C T R O G E N E S I S I N M Y E L I N AT E D
ularly in large myelinated axons (Devaux et al. 2003). In con- A N D D E M Y E L I N AT E D AXO N S
trast to other nodal proteins that have a presence at the axon
initial segments, Kv3.1b is not present at axon initial segments In contrast to nonmyelinated axons, in which action poten-
and many smaller nodes unlike other nodal proteins that also tials are conducted in a continuous manner, myelinated fibers
have a presence at the axon initial segments, and it does not exhibit saltatory conduction. In mammalian myelinated axons
account for the effects of 4-AP on compound action potentials at 370C, the time between excitation of one node and the
recorded from central myelinated tracts (Devaux et al. 2003). next (internodal conduction time) is approximately 20Psec
Furthermore, nodal localization of Kv3.1b is maintained, (Rasminsky and Sears 1972a, 1972b). Action potential con-
at least temporarily, even in the absence of myelin sheath, as duction is usually unidirectional in nature because sodium
shown in myelin-deficient (md) rats with severe CNS demyeli- channels close and remain inactive for a short period soon
nation (Devaux et al. 2003) and paralleling the persistence of after activation. This refractory period, during which the axon
node-like clusters of Nav channels after oligodendrocyte death is unable to support a second action potential, has a duration
in md rats (Arroyo et al. 2002). of milliseconds.
The specialized organization of myelinated axonal elements Because myelin functions as an insulator, the action cur-
poses severe problems in demyelinated axons. Damage to myelin rent from each active node of Ranvier is shunted to subsequent
disrupts action potential electrogenesis because of loss of capaci- nodes via the relatively low-resistance axoplasm (Fig. 42.3A).
tative shield and low density of inward Na+ current within the The first few wraps of tight myelin seem to be the most impor-
exposed intermodal axon membrane, which is normally scarce tant because they provide a capacitative shield (Funch and
in Na+ channels (Waxman 1982). Loss of myelin also unmasks Faber 1984). Safety factor (the ratio between current avail-
fast K+ channels (normally covered by the myelin) that clamp able to stimulate a node of Ranvier and current required to
the demyelinated resting membrane potential close to the K+ stimulate the node) is 5 to 7 in normal myelinated fibers, so
equilibrium potential Ek, opposing depolarization and further that there is a high degree of reliability for impulse conduction
impeding action potential conduction (Chiu and Ritchie 1981). (Tasaki 1959).
Demyelination also imposes a significant energy burden as Na+, Capacitative and resistive shunting causes a fall in density
K+–ATPases attempt to restore the ionic gradient and maintain of action current, following damage to myelin (Fig. 42.3B).
neuronal excitability (Ames 2000; Benarroch 2011). If the safety factor is decreased but still greater than 1.0, the
Molecular plasticity of the axon membrane contributes to charging time for the nodal membrane will be increased
the recovery of conduction following demyelination. Expression so that it will take longer than normal for the axon to reach
of higher-than-normal density of Na+ channels in some threshold, and conduction will continue but conduction

M Y E L I N, I M P U L S E C O N D U C T I O N, A N D T H E PAT H O P H YS I O L O GY O F D E M Y E L I N AT I O N • 531
A

Figure 42.3 Diagrammatic representation of current flow associated with conduction through normally (A) myelinated and (B) demyelinated regions
of an axon. The action potential is conducted from left to right (arrow). This idealized diagram represents the myelin as a perfect insulation. Dashed
arrows illustrate current flow resulting from an action potential that is located at the cross-hatched node. Current is lost in demyelinated regions as a
result of capacitative and resistive shunting.

velocity will be reduced. In demyelinated spinal root fibers, demyelinated membrane close to the potassium equilibrium
internodal conduction time can be increased nearly 20-fold, Ek, further impeding action potential electrogenesis (Bostock
to about 500Psec (Rasminsky and Sears 1972a). Thus conduc- et al. 1981; Ritchie et al. 1981).
tion velocity is reduced. In more severely demyelinated axons, Conduction failure need not be all-or-none. It can be
the safety factor can fall to less than 1.0, reaching threshold, frequency-related; with high-frequency impulse trains fail-
and thus lead to conduction failure (Waxman 1982). ing to propagate while low-frequency trains conduct reliably
From a descriptive point of view, demyelinated axons (see Fig. 42.4D). In more severely affected fibers, conduction
can display a spectrum of conduction abnormalities under- failure can be complete, with even single action potentials
lying signs and symptoms of pathology. Conduction abnor- failing to propagate beyond the region of demyelination (see
malities that are negative in the clinical or Jacksonian sense Fig. 42.4E). It has been hypothesized that hyperpolarization
include slowed, desynchronized, or blocked conduction of the axon membrane, owing to electrogenic pump (Na+/
Figure 42.4B-E. Such abnormalities are confined to the K+-ATPase) activity, may contribute to conduction block
zone of demyelination, with normal conduction proximal of high-frequency impulse trains (Bostock and Grafe 1985).
and distal to the zone (Fig. 42.5). Slowed conduction appears There is also evidence suggesting that increased intracellular
to be less important than conduction block in producing Na+ at the “driving node” (Rasminsky and Sears 1972a) and
clinical deficits (McDonald 1963; McDonald and Sears depolarization of demyelinated axons because of increases
1970). Loss of synchrony in tracts, in which different in extracellular K+ concentration may also contribute to
fibers exhibit unequal degrees of conduction slowing, also high-frequency block.
known as temporal dispersion, can occur as a consequence Conduction abnormalities that are positive in a Jacksonian
of decreased conduction velocity and reflects the variabil- sense are also important from a clinical perspective, and are
ity in internodal conduction times in demyelinated axons illustrated in Figure 42.4F-I. Ectopic impulse generation (Fig.
(see Fig. 42.4C). Temporal dispersion can interfere with 42.4F) has been observed, for example, in demyelinated dor-
functions such as the stretch reflex that require synchronous sal column axons (Smith and McDonald 1980). Abnormal
discharge. cross-talk, also termed ephaptic interaction, may occur between
Changes in passive and active characteristics of demyeli- abnormally myelinated axons (see Fig. 42.4H). Increased
nated fibers both contribute to conduction block. In addition mechanosensitivity (Fig. 42.4G) has been experimentally
to capacitative current loss through injured myelin sheaths, observed in demyelinated axons and probably accounts for
conduction in focally demyelinated axons can also fail because clinical phenomena such as Tinel’s and Lhermitte’s sign
of impedance mismatch between the normally myelinated (Smith and McDonald 1980). Impulse reflection may occur
and demyelinated regions (Sears et al. 1978; Waxman and in some focally demyelinated fibers (Burchiel 1980) and may
Brill 1978). Low Na+ channel density in the demyelinated result in extraneous activity (e.g., paresthesia, pain, or tonic
axon membrane also contributes to conduction block. In spasms). Because reflected (antidromic) impulses can collide
addition, fast K+ channels (normally covered by the myelin) with and abolish orthodromic impulses, impulse reflection
are unmasked after demyelination and tend to clamp the may also interfere with normal impulse traffic.

532 • NEUROGLIA
A NORMAL CONDUCTION
F ECTOPIC IMPULSE GENERATION

B DECREASED CONDUCTION VELOCITY

G INCREASED MECHANOSENSITIVITY

C TEMPORAL DISPERSION

H CROSS-TALK

D FREQUENCY-RELATED BLOCK

I IMPULSE REFLECTION

E TOTAL CONDUCTION BLOCK

Figure 42.4 Conduction Abnormalities in Demyelinated Axons. Demyelinated regions of the axon are diagrammatically shown as dashed lines. Cell bodies
are located to the left, and axon terminals to the right. The direction of normal conduction is indicated by the arrow; see text for further explanation.

A
1 2

2.0
1.0 mV
msec
prox. demyel. dist.

1 2

0.2 mV

C
1 2

0.5 mV

Figure 42.5 Recording obtained proximal to (A), across (B), and distal (C) to a focally demyelinated region (injected with lysophosphatidylcholine) from
rat sciatic nerve. Conduction is relatively normal in proximal and distal nerve segments in which myelin is intact (A2, C2). However, conduction
slowing, block, and temporal dispersion (B2) are present when action potentials are conducted through the lesion site. Modified from Kocsis and
Waxman 1985.
4 .1 C O N T I NU O US C O N D U C T I O N excitability and suggest that this occurs in a time-dependent
F O L L OWI N G D E MY E L I NAT I O N manner after axons are covered by myelin. From a functional
standpoint, this insures that conduction is not compromised
Continuous action potential conduction, similar to that of
owing to premature loss of sodium channels during develop-
normal unmyelinated axons, has been observed in some demy-
ment (Black and Waxman 1986).
elinated axons (Bostock and Sears 1976). Some demyelinated
Immunocytochemical studies utilizing subtype specific
axons can conduct with a velocity that can fall to as low as 5% of
antibodies to sodium channels have provided evidence that
normal saltatory conduction velocity, over continuous lengths
supports the suggestion of dedifferentiation of the axon
of demyelination exceeding 2 mm (several internodes) (Felts
membrane in some demyelinated axons. Craner et al. (2003,
et al., 1977). Sequestration of sodium channels at nodes along
2004a) observed a switch from Nav1.6 to Nav1.2 expression
normal myelinated axons raises the question of how continuous
at nodes (Fig. 42.6) along demyelinated CNS axons in experi-
conduction can occur along previously internodal parts of the
mental allergic encephalomyelitis and multiple sclerosis (MS)
axon following demyelination. Computer simulations provide
tissue. Some demyelinated axons exhibited continuous immu-
a partial answer in demonstrating that, in some small-caliber
nostaining for Nav1.2 and less frequently for Nav1.6 chan-
demyelinated axons, the density of preexisting Na+ channels in
nels, while extending for tens of micrometers along the fiber
the demyelinated region may approach the density required to
trajectory (i.e., as far as the axons could be followed within
support conduction (Hines and Shrager 1991; Waxman and
sections). A similar pattern of continuous Nav1.2 immunos-
Brill 1978). In small-diameter premyelinated axons (diameter
taining has been reported in premyelinated axons (Boiko et al.
<0.25Pm) in the optic nerve, the conduction of single action
2001), which may provide a substrate for continuous conduc-
potentials can be supported by Na+ channel densities of less
tion of impulses. The functional implications of a reversion to
than 10/Pm2 (Waxman et al. 1989). The diameter of some
Nav1.2 expression (rather than Nav1.6) are not fully under-
demyelinated axons is reduced (Prineas and Connell 1978;
stood. Evidence from patch-clamp studies (Rush et al. 2005)
Smith et al. 1983), possibly as a result of decreased neurofila-
indicates that different functional properties may permit
ment phosphorylation and increased neurofilament packing
Nav1.6 channels to support higher firing rates. Substitution
density (De Waegh et al. 1992). These results are applicable to
of Nav1.2 for Nav1.6 may permit conduction to continue in
demyelinated axons with small diameters but may not apply to
demyelinated axons even if they are depolarized, because the
larger axons, because of their lower input impedance. Because
voltage dependence of inactivation of Nav1.2 is depolarized
of this, the acquisition of a higher-than-normal Na+ channel
compared with that of Nav1.6 (Rush et al., 2005).
density appears to be required for restoration of conduction.
Experimental evidence indicates that this does in fact occur in
some demyelinated axons. 4.2 N O NU N I F O R M C O N D U C T I O N F O L L OWI N G
Electron microscopy studies on experimentally demy- D E MY E L I NAT I O N
elinated axons demonstrated the development of regions
of axon membrane with cytochemical (Foster et al. 1980) Smith et al. (1982) have argued that nonuniform impulse prop-
and freeze-fracture (Black et al. 1987) characteristics similar agation may also contribute to recovery of conduction in some
to those of nodal membrane, suggesting the acquisition of a demyelinated axons. In this mode of conduction, action poten-
higher-than-normal sodium channel density. Early immuno- tials propagate nonuniformly between isolated foci of inward
cytochemical studies also provided evidence for the acquisi- current generation (termed phi-nodes). These seem to consist of
tion of increased Na+ channel densities in the demyelinated focal aggregations of Na+ channels that are established before
parts of the axon membrane. These studies, on fish lateral remyelination (Smith et al. 1982). Phi-nodes develop several
line nerves, demonstrated the development of relatively high days before remyelination, and it has been suggested (Smith
densities of Na+ channels in previously internodal regions et al. 1982) that they are the precursors of nodes.
(England et al. 1990). Immunocytochemical observations
2 to 3 weeks following injection of the demyelinating toxin
doxorubicin also suggested the expression of sodium channels 5 BASIS FOR MOLECULAR REMODELING
at newly formed nodes along mammalian remyelinated axons O F T H E D E M Y E L I N AT E D AXO N
(Dugandzija-Novakovic et al. 1995). These early investiga-
tions, however, utilized pan-sodium channel antibodies that It has been clearly established that, in some demyelinated
did not differentiate between channel subtypes. axons, sodium channel density increases so that it can support
It is possible that acquisition of higher-than-normal densi- impulse invasion into the demyelinated region and continuous
ties of Na+ channels in demyelinated axon regions is because of conduction through it (Bostock and Sears 1978; Foster et al.
dedifferentiation of the axon membrane (see later). Although 1980). There is evidence (Hines and Shrager 1991) that chan-
the mature internodal membrane is incapable of secure con- nel diffusion from nearby nodes is not sufficient to support
duction (Ritchie and Rogart 1977; Waxman 1977), the pre- action potential conduction through demyelinated regions.
myelinated axon membrane, (including regions that will Former nodes of Ranvier in demyelinated axons, when stud-
develop into internodal membrane), is electrically excitable ied by patch-clamp, display sharp gradients in channel density,
(Foster et al. 1982; Waxman et al. 1989). Studies on the devel- which have been interpreted as suggesting that Na+ channels
oping internodal axon during normal development indicate do not diffuse away from the node in large numbers follow-
that suppression of Na+ channel expression reduces axonal ing demyelination (Shrager 1989). Radioimmunoassay studies

534 • NEUROGLIA
Figure 42.6 E-APP-positive spinal cord axons coexpress NCX and Nav1.6 over extensive regions in acute MS lesions. Digital images demonstrate axons
in MS spinal cord white matter immunostained for E-APP (E and F; blue), Nav1.6 (A; red) or Nav1.2 (B; red), and NCX (C and D; green). G and H
correspond to merged images. A, C, E, and G show coexpression of Nav1.6 and NCX within axons displaying E-APP, a marker of axonal injury. In
contrast, B, D, F, and H demonstrate NCX-immunopositive staining but an absence of Nav1.2 immunostaining within E-APP-positive axons, and
coexpression of NCX and Nav1.2 within E-APP-negative axons. From Craner et al. 2004b.

demonstrate a significant increase in Na+ channel concentra- these glial-synthesized channels to the axon (Bevan et al. 1985;
tion per weight of tissue (approximately three-fold at 21 to Gray and Ritchie 1985). Astrocyte processes and Schwann cell
28 days after peripheral nerve demyelination), supporting the microvilli contact the axon membrane at the node in a highly
idea that new Na+ channels are inserted into the demyelinated specific manner (Hildebrand 1971; Waxman and Black 1984).
axon membrane (England et al. 1991). Quantitative autora- Similar glial processes are apposed to demyelinated axons at
diography reveals a fourfold increase in STX-binding sites in sites of Na+ channel clustering (Black et al. 1984; Rosenbluth
demyelinated white matter in MS patients, compared with 1985; Rosenbluth and Blakemore 1984). The two special-
normal white matter (Moll et al. 1991). The site of produc- izations (glial contact and Na+ channel clustering) usually
tion of the Na+ channels in demyelinated axons has not been are juxtaposed—an observation that has been interpreted
firmly identified. Synthesis of channels in the neuronal cell as suggesting that glial cell contact may contribute to the
body, with translocation by the axonal transport, is a clear pos- development of Na+ channel clusters in the axon membrane.
sibility. Electrophysiological experiments have demonstrated Voltage-sensitive Na+ currents are present in astrocytes (Bevan
the production of new, functional Na+ channels by neurons et al. 1985) and in some cases the density of sodium channels in
after axonal transection, and there is evidence for axonal astrocytes is sufficient to support production of over-shooting
transport of Na+ channels in peripheral nerves (Lombet et al. action potentials, if resting inactivation is removed by hyperpo-
1985). Craner et al. (2003) noted upregulated transcription of larizing the cell. Black et al. (2010) have recently demonstrated
Nav1.2 mRNA in cell bodies of demyelinated axons, implying a striking upregulation of sodium channel Nav1.5 within
that new channels continue to be produced by the neuron. reactive astrocytes in MS plaques (Fig. 42.7). Physiological
Another potential source of sodium channels that has been properties of tetrodotoxin-resistant sodium channels in astro-
proposed is synthesis in glial cells, with subsequent transfer of cytes are consistent with these channels being of the Nav1.5

M Y E L I N, I M P U L S E C O N D U C T I O N, A N D T H E PAT H O P H YS I O L O GY O F D E M Y E L I N AT I O N • 535
subtype. However, there is no evidence for expression of Nav1.5
within either normal myelinated or demyelinated axons. The
functional role of Nav1.5 channels in reactive astrocytes is under
investigation but, at this time, there is no evidence that these
astrocyte channels are transferred to axons. Although the glial
sodium channel synthesis hypothesis attracted much attention,
and more than 20 years have passed since Ritchie and collabo-
rators proposed this hypothesis, there is no demonstration of
channel transfer from glial cells to axons. Alternative functions
for astrocyte Na+ channels have been proposed (e.g., providing
a return pathway for Na+ ions that maintains astrocyte Na+ and
K+ -ATPase activity) (Sontheimer et al. 1994). Astrocytes may
also secrete extracellular molecules that target or anchor Na+
channels at sites of aggregation (Waxman et al. 1992).

6 S O D I U M C H A N N E L S A N D AXO N A L
D E G E N E R AT I O N

Axonal degeneration occurs early in the course of MS


and is a strong contributor to acquisition of persistent
(non-remitting) deficits (Trapp et al. 1999). Studies suggest
that sodium channels are crucial to this degeneration process.
The sodium-calcium exchanger (NCX) is present along mam-
malian myelinated axons within the CNS (Steffensen et al.
1997), consistent with the presence of Nav1.6 at nodes. Stys
et al. (1993) showed that, in energetically stressed axons, a
persistent Na+ influx, via sodium channels, can drive reverse
Na+-Ca2+ exchange which, in turn, imports deleterious lev- Figure 42.7 Nav1.5 Expression in Astrocytes Within MS Tissue.
els of Ca2+ into the axonal cytosol, triggering axonal injury. A montage of grayscale images of R. communis agglutinin I (RCA1)
Craner et al. showed that Nav1.6 sodium channels and NCX positive macrophages (white) throughout a portion of a section of
MS tissue was constructed to provide orientation for localization of
are both present, and are colocalized, along injured axons in astrocytes within the tissue. Two regions of substantial macrophage
experimental allergic encephalomyelitis (EAE) (Craner et al. infiltration (top and bottom), are separated by white matter that exhibits
2004a) and in MS (Craner et al. 2004b) (see Fig. 42.6). It has limited macrophage infiltration. In a serial section to that stained for
been shown that Nav1.6 produces substantial persistent cur- RCA I, labeling for Nav1.5 (red), GFAP (green) and PLP (blue) was
rent (Rush et al. 2005) and it appears likely that high levels performed and high-magnification images acquired (right-hand panels).
An astrocyte within a normal-appearing white matter region of the sec-
of expression of Nav1.6 along demyelinated axons, in regions tion not included in the montage and which exhibits strong PLP labeling
that also express NCX, can trigger axonal degeneration. displayed minimal Nav1.5 labeling (top right panel). In contrast, an astro-
The expression of Nav1.2 along demyelinated axons in EAE cyte in the white matter between two areas of macrophage infiltration
(Craner et al. 2004a) and MS (Craner et al. 2004b) is not (panel 2) is hypertrophic and also shows an upregulation of Nav1.5. Note
colocalized with NCX, suggesting that expression of Nav1.2 that the PLP labeling (blue) is more disrupted compared with that in
normal-appearing white matter (top right panel). Astrocytes at the border
is an adaptive change, supporting conduction along demy- of (panel 3) and within (panel 4) the active lesion display intense Nav1.5
elinated axons while not contributing to their degeneration immunolabeling. Note the substantial disruption of PLP labeling at the
(Waxman 2006). border of the lesion (panel 3) and the absence of PLP labeling within the
lesion (panel 4). PLP, blue; GFAP, green; Nav1.5, red; co-localization of
GFAP and Nav1.5, yellow. From Black et al. 2010.
7 I M P E DA N C E M I S M ATC H I N
D E M Y E L I N AT E D AXO N S

A high Na+ channel density in demyelinated axon regions does developed a high Na+ channel density (similar to that at
not, in itself, ensure reliable conduction. Electrical loading, nodes) show that despite the high Na+ channel density in
largely as a result of increased membrane capacitance, imposes the demyelinated area, conduction block occurs at the junc-
impedance mismatch that can cause conduction block at tion between normal and demyelinated axon region because
sites of axonal inhomogeneity such as the junction between of impedance mismatch which prevents threshold from being
myelinated and demyelinated regions (Waxman 1978). reached (Waxman and Brill, 1978).
Computer simulation studies of action potentials (Waxman Impedance matching can be overcome through the devel-
and Brill 1978) in a fiber with a single focally demyelinated opment of relatively short myelinated segments proximal to the
internode, in which the demyelinated axon membrane has demyelinated area (Waxman and Brill 1978), decrease in axon

536 • NEUROGLIA
diameter within the demyelinated region (Sears and Bostock of Ranvier (Koles and Rasminsky 1972; Waxman and Brill
1981), the development of increased Na+ channel density at 1978). Conduction velocity approaches normal in demyeli-
the node proximal to the demyelinated area, or the develop- nated peripheral nerves, and the refractory period and the
ment of a specialized transition zone (with relatively high Na+ ability to conduct high-frequency impulse trains is improved
channel densities or relatively low K+ channel densities) at as remyelination occurs (Smith et al, 1980; Smith et al. 1981,
the edge of the region of demyelination (Waxman and Wood 1983). Moreover, clinical recovery in rats with EAE is cor-
1984). Remyelinated myelin segments are known to be shorter related with remyelination and restoration of conduction
than normal internodes, and this serves as an adaptive func- (Stanley and Pender, 1991). Reduced internode distances in
tion to facilitate impedance matching. remyelinated fibers result in conduction velocities that, while
higher than in demyelinated axons, are lower than in normally
myelinated axons (Brill et al. 1977). However, reduced con-
8 E N E R G ET I C A S P E C T S O F duction velocity, in itself, does not necessarily produce clini-
D E M Y E L I N AT I O N cal deficits. Restoration of conduction due to remyelination is
expected to lead to clinical improvement. Both oligodendro-
As a result of larger capacitative load and increased overall cyte and Schwann cell-mediated remyelination of CNS axons
number of sodium channels per unit length, the energetic can enhance conduction. Remyelination of dorsal column
load imposed by impulse conduction is substantially higher axons by Schwann cells, for example, has been shown to restore
in non-myelinated compared to myelinated axons. Wang et al. secure action potential conduction with a normal refractory
(2008) suggest that energetic cost per action potential per period of transmission (but with decreased conduction veloc-
centimeter of axon is 10-fold higher in nonmyelinated, com- ity) and the capability to conduct the high-frequency impulse
pared with myelinated axons. Similar considerations appear trains in these fibers (Blight and Young 1989; Felts and Smith
to apply to demyelinated axons. Thus, the observation of 1991).
increased number and activity of mitochondria along axons in
MS lesions (Witte et al. 2009) might be viewed to be a com-
pensatory change. Energetically stressed mitochondria, how- 10 T R A N S P L A N TAT I O N O F
ever, produce ROS, which can injure axons and, moreover, M Y E L I N -F O R M I N G C E L L S
show increased susceptibility to mutations within mitochon-
drial DNA. Demyelinating diseases with a genetic etiology, such as leu-
Campbell et al. (2011) observed an increased number of kodystrophies, can cause dysmyelination (abnormal myelin
respiratory deficient neurons within brain tissue from patients formation), hypomyelination (insufficient myelin), and demy-
with secondary progressive MS and noted high levels of clon- elination (myelin degradation) (Biffi et al. 2011; Kohlschutter
ally expanded mitochondrial DNA deletions within these et al. 2010; Nave 2010; Pastores 2009). Inflammatory
neurons. Dutta et al. (2006) observed reduced mitochon- demyelinating diseases, exemplified by MS in the CNS
drial gene expression within axons in MS lesions and sug- and Guillain-Barre syndrome in the PNS, involve immune
gested that reduced ATP production might contribute to system-mediated attack of components of myelin, which leads
impaired ionic homeostasis, thereby playing a role in axonal to the progressive loss of axonal integrity (Makowska et al.
degeneration in MS. Studies also show a reduced number of 2008; Steinman et al. 2002). Despite the inherent capacity of
Na+/K+-ATPase positive axons in chronic MS lesions (Young the nervous system to remyelinate, persistent demyelination
et al. 2008) and impaired mitochondrial complex IV activity is a major problem in leukodystrophies, inflammatory demy-
within Na+/K+-ATPase positive chronic MS lesions (Mahad elination, and spinal cord trauma, and results in a spectrum
et al. 2009), suggesting that mitochondrial dysfunction may of pathologies including irreversible neurological damage.
contribute to axonal degeneration in active demyelinating Potential approaches to myelin restoration include methods
lesions. Interestingly, chronic inactive areas of demyelination to improve endogenous remyelination as well as methods to
from the same tissue display increased mitochondrial mass deliver exogenous sources of myelin to demyelinated lesions.
and complex IV activity, possibly reflecting a compensatory Numerous studies have demonstrated the feasibility of remy-
response to increased energy demand (Mahad et al. 2009). elination through transplantation of central or peripheral
Current studies are exploring the detailed nature of the mito- derived myelin-forming cells in experimental animal models
chondrial contribution to pathophysiology of axonal injury in of demyelination (Einstein et al. 2006; Learish et al. 1999;
MS (see, e.g., Campbell and Mahad 2011). Pluchino 2003).
Some of the earliest proof-of-principle studies of
transplantation-based myelin repair utilized Schwann cells
9 R E M Y E L I N AT I O N (Aguayo et al. 1977; Blakemore 1977) and grafts containing
oligodendrocyte precursor cells (Blakemore and Crang 1988;
Computer simulations predict that remyelination with even Duncan et al. 1988; Gansmuller et al. 1986; Lachapelle et al.
thin, or short, myelin sheaths should provide a capacitative 1983). Subsequent studies demonstrated the ability of a variety
shield that promotes conduction through previously demy- of cell types including O2A+ progenitor cells from optic nerve
elinated fibers, as long as sodium channel densities similar cultures (Groves et al. 1993), olfactory ensheathing cells (Sasaki
to those in normal fiber develop at the remyelinated nodes et al. 2006), cultured neurospheres (Pluchino et al. 2003),

M Y E L I N, I M P U L S E C O N D U C T I O N, A N D T H E PAT H O P H YS I O L O GY O F D E M Y E L I N AT I O N • 537
A2B5+ glial progenitor cells (Windrem et al. 2004), and oli- Several studies have provided evidence for transplantation-
godendrocyte precursor cells (Crang et al. 2004; Franklin based restoration of myelin and improved action potential
and Blakemore 1997) to differentiate into myelin forming conduction in demyelinated and dysmyelinated axons in ani-
cells upon transplantation. However, the ability of these cell mal models. Human embryonic stem cells (hESC) committed
types to restore myelin varies widely and a direct compar- to an oligodendrocyte lineage (induced pluripotent stem cells)
ison of their efficiency to remyelinate has not been studied. are among the cell-types currently being studied (Keirstead
For example, fetal progenitor cells myelinate more extensively et al. 2005; McDonald et al. 1999). Induced pluripotent stem
than adult cells, although with somewhat delayed onset of cells, in addition to providing a robust supply of oligodendro-
myelin production (Windrem et al. 2004), whereas oligoden- cyte precursor cells with a uniform differentiation capacity,
drocyte precursor cells from higher order mammals migrate avoid the possibility of immunological rejection when gen-
more extensively and exhibit more robust remyelination com- erated from autologous tissue (see review by Hu et al. 2009;
pared to their rodent counterparts (Windrem et al. 2008). Potter et al. 2011). Highly purified populations of oligoden-
Furthermore, the highly migratory nature of oligodendrocyte drocyte precursor cells have been generated and demonstrated
precursor cells enables them to expand and terminally differen- to be capable of differentiating into myelinating oligodendro-
tiate differently in response to varying regional cues (Windrem cytes upon transplantation into the spinal cord of shiverer
et al. 2008). Although the primary assay for the outcome of mice (Nistor et al. 2005). Encouraged by these results, Geron
transplantation studies is structural, that is, whether or not Corporation initiated a phase 1 clinical trial in 2009 to evalu-
morphologically normal myelin is formed, does not, in itself, ate the safety of transplanting oligodendrocyte precursor cells
ensure improved conduction, because impedance mismatch derived from hESC into people with acute spinal cord injury.
or failure to form nodes with adequate Na+ channel densities Two years later, however, Geron announced that it will not
can lower the safety factor. pursue its spinal cord injury trial using oligodendrocyte pre-
In the first physiological study of conduction properties cursor cells derived from hESC. Thus the safety and efficacy
in axons myelinated by transplanted cells, Utzschneider et al. of transplantation-based remyelinating therapies remains to
1994) examined myelin-deficient axons in the md rat spinal cord be determined. A recent report of tumor development many
after transplantation of oligodendrocytes, and demonstrated a years after transplantation of poorly characterized human
fourfold increase in conduction velocity, which approached fetal brain-derived cells into a patient with ataxia telangiecta-
myelinated control values, after formation of myelin by sia has increased awareness about the potentially malignant
transplanted glial cells. The ability to follow high-frequency side-effects of stem cell transplants (Amariglio et al. 2009).
stimulation returned to almost normal. Moreover, action
potentials initiated outside the transplant region could invade
and propagate into the region of demyelination, and could 11 S U M M A RY A N D P E R S P E C T I VE
then propagate beyond the demyelinated region. Similar
improvements in action potential conduction have been Rapid action potential conduction in vertebrates relies heav-
observed following transplantation of Schwann cells ily on the functional architecture of the myelinated axon. A
(Honmou et al. 1996), olfactory ensheathing cells (Imaizumi myelinated axon is more than just a bare axon with myelin
et al. 1998),”humanized” olfactory ensheathing cells from the wrapped around it. It is a complex and intimate interaction
pig (Imaizumi et al. 2000) and bone marrow stromal cells between two cells that defines the regional heterogeneity
(Akiyama et al. 2002a,, 2002b) to demyelinated lesions within characteristic of a myelinated axon. The high resistance and
the spinal cord of adult rats. low capacitance of myelin, combined with nodes of Ranvier
More recent studies of transplanted olfactory ensheathing between segments of myelinated internodes support saltatory
cells from green fluorescent protein-expressing donor rats into conduction while restricting action potential and ion currents
a region of spinal cord demyelination demonstrated exten- to less than 0.5% of the axon’s surface and reducing energy con-
sive remyelination, including development of mature nodes, sumption. Strategic subdomain localization of voltage-gated
paranodes, and juxtaparanodes, and restoration of impulse ion channels and associated molecules in the myelinated axon
conduction, as assessed by ultrastructural, immunocytochem- membrane governs action potential conduction. The specific
ical, and electrophysiological analyses (Sasaki et al. 2006). assortment of molecules present within subsets of neurons,
Furthermore, remyelinated axons display clusters of Nav1.6 further adds to the complexity of action potential initiation
at the nodes and Kv1.2 at the juxtaparanodes, demonstrat- and conduction. The complexity of the myelinated fiber is
ing that in addition to forming compact myelin, transplanted matched by the complexity of demyelination. Passive and
cells provide an environment that supports the restoration active properties of the axon interact to shape the conduction
of the molecular organization of a myelinated axon (Sasaki properties of a demyelinated axon and a variety of cellular and
et al. 2006). Black et al. (2006) demonstrate that spontaneous molecular mechanisms contribute to these properties. The
remyelination of central axons by endogenous Schwann cells multifactorial determinants of altered axonal conduction after
recapitulates the nodal clustering of Nav1.6 and juxtaparan- demyelination have made it more difficult to study, but also
odal distribution of Kv1.2, consistent with findings by Schafer have provided multiple therapeutic opportunities that may
et al. (2006) that Nav1.6, but not Nav1.2, is detected at newly improve function in demyelinated axons. These include phar-
formed nodes in remyelinated axons in a lysolecithin-induced macological manipulation of K+ channels, promotion of Na+
model of PNS demyelination. channel expression, manipulation of cellular characteristics to

538 • NEUROGLIA
correct impedance mismatch at the edge of the plaque, promo- demyelinating lesions in adult central nervous system. Dev Neurosci
tion of endogenous remyelination, and remyelination through 10:1–11.
Blight A, Young W. 1989. Central axons in injured spinal cord recover
transplantation of myelin-forming cells. electrophysiological function following remyelination by Schwann
cells. J Neurol Sci 91:15–34.
Boiko T, Rasband MN, Levinson SR, Caldwell JH, Mandel G, Trimmer
AC K N OW L E D G M E N T S JS, et al. 2001. Compact myelin dictates the differential targeting of
two sodium channel isoforms in the same axon. Neuron 30:91–104.
Bostock H, Grafe P. 1985. Activity–dependent excitability changes in
The authors’ research has been supported in part by grants normal and demyelinated rat spinal root axons. J Physiol (Lond)
from the Rehabilitation Research Service and Medical 365:239–257.
Research Service, Department of Veterans Affairs, and by gifts Bostock H, Sears TA. 1976. Continuous conduction in demyelinated
from the Paralyzed Veterans of America. mammalian nerve fibres. Nature 263:786–787.
Bostock H, Sears TA. 1978. The internodal axon membrane: electrical
excitability and continuous conduction in segmental demyelination.
J Physiol (Lond) 280:273–301.
REFERENCES Bostock H, Sears TA, Sherratt RM. 1981. The effects of 4–
aminopyridine and tetraethylammonium ions on normal and demy-
Aguayo AJ, Kasarjian J, Skamene E, Kongshavn P, Bray GM. 1977. elinated mammalian nerve fibers. J Physiol (Lond). 313:301–315.
Myelination of mouse axons by Schwann cells transplanted from nor- Bowe CM, Kocsis JD, Targ EF, Waxman SG. 1987. Physiological effects
mal and abnormal human nerves. Nature 268:753–755. of 4-aminopyridine on demyelinated mammalian motor and sensory
Akiyama Y, Radke C, Honmou O, Kocsis JD. 2002a. Remyelination of fibers. Ann Neurol 22:264–268.
the spinal cord following intravenous delivery of bone marrow cells. Brill MH, Waxman SG, Moore JW, Joyner RW. 1977. Conduction
Glia 39:227–234. velocity and spike configuration in myelinated fibers: computed
Akiyama Y, Radtke C, Kocsis JD. 2002b. Remyelination of the rat spi- dependence on internode distance. J Neurol Neurosurg Psychiatry
nal cord by transplantation of indentified bone marrow stromal cells. 40:769–774.
J Neurosci 22:6623–6630. Bunge MB, Bunge RP, Pappas GD. 1961. Electron microscope demon-
Amariglio N, Hirshberg A, Scheithauer BW, Cohen Y, Loewenthal R, stration of connections between glia and myelin sheaths in the devel-
Trakhtenbrot L, et al. 2009. Donor-derived brain tumor following oping mammalian central nervous system. J Cell Biol 12:448–453.
neural stem cell transplantation in an ataxia telangiectasia patient. Burchiel K. 1980. Abnormal impuse generation in focally demyelinated
PLoS Med 6:e1000029. trigeminal roots. J Neurosurg 53:674–683.
Ames A, 3rd. 2000. CNS energy metabolism as related to function. Brain Caldwell JH, Schaller KL, Lasher RS, Peles E., Levinson SR. 2000.
Res Brain Res Rev 34:42–68. Sodium channel Na(v)1.6 is localized at nodes of Ranvier, dendrites,
Arroyo EJ, Xu T, Grinspan J, Lambert S, Levinson SR, Brophy PJ, et al. and synapses. Proc Natl Acad Sci U S A 97:5616–5620.
2002. Genetic dysmyelination alters the molecular architecture of Campbell GR, Mahad DJ. 2011. Mitochondria as crucial players in
the nodal region. J Neurosci 22:1726–1737. demyelinated axons: lessons from neuropathology and experimental
Benarroch EE. 2011. Na+, K+-ATPase: functions in the nervous system demyelination. Autoimmune Dis 2011:262847.
and involvement in neurologic disease. Neurology 76:287–293. Campbell GR, Ziabreva I, Reeve AK, Krishnan KJ, Reynolds R, Howell
Bevan S, Chiu SY, Gray PTA, Ritchie JM. 1985. The presence of O, et al. 2011. Mitochondrial DNA deletions and neurodegeneration
voltage-gated sodium, potassium and chloride channels in rat cul- in multiple sclerosis. Ann Neurol 69:481–492.
tured astrocytes. Proc Roy Soc Lond B 225:229–313. Chiu SY, Ritchie JM. 1980. Potassium channels in nodal and inter-
Biffi A, Aubourg P, Cartier N. 2011. Gene therapy for leukodystrophies. nodal axonal membrane in mammalian myelinated fibers. Nature
Hum Mol Genet 20:R42–53. 284:170–171.
Bjartmar C, Hildebrand C, Loinder K. 1994. Morphological heteroge- Chiu SY, Ritchie JM. 1981. Evidence for the presence of potassium chan-
neity of rat oligodendrocytes: electron microscopic studies on serial nels in the paranodal region of acutely demyelinated mammalian
sections. Glia 11:235–244. nerve fibres. J Physiol (Lond) 313:415–437.
Black JA, Foster RE, Waxman SG. 1981. Freeze-fracture ultrastructure Craner MJ, Hains BC, Lo AC, Black JA, Waxman SG. 2004a.
of rat C.N.S. and P.N.S. nonmyelinated axolemma. J Neurocytol Co-localization of sodium channel Nav1.6 and the sodium-calcium
10:981–993. exchanger at sites of axonal injury in the spinal cord in EAE. Brain
Black JA, Newcombe J, Waxman SG. 2010. Astrocytes within mul- 127:294–303.
tiple sclerosis lesions upregulate sodium channel Nav1.5. Brain Craner MJ, Lo AC, Black JA, Waxman SG. 2003. Abnormal sodium
133:835–846. channel distribution in optic nerve axons in a model of inflammatory
Black JA, Waxman SG. 1986. Molecular structure of the axolemma of demyelination. Brain 126:1552–1561.
developing axons following altered gliogenesis in rat optic nerve. Craner MJ, Newcombe J, Black JA, Hartle C, Cuzner ML, Waxman SG.
Devel Biol 115:301–312. 2004b. Molecular changes in neurons in multiple sclerosis: altered
Black JA, Waxman SG, Hildebrand C. 1984. Membrane specializa- axonal expression of Nav1.2 and Nav1.6 sodium channels and Na+/
tion and axo-glial association in the rat retinal nerve fibre layer: Ca2+ exchanger. Proc Natl Acad Sci U S A. 101:8168–8173.
freeze-fracture observations. J Neurocytol 13:417–430. Crang AJ, Gilson JM, Li WW, Blakemore WF. 2004. The remyelinating
Black JA, Waxman SG, Smith KJ. 2006. Remyelination of dorsal col- potential and in vitro differentiation of MOG-expressing oligoden-
umn axons by endogenous Schwann cells restores the normal pattern drocyte precursors isolated from the adult rat CNS. Eur J Neurosci
of Nav1.6 and Kv1.2 at nodes of Ranvier Brain 129:1319–1329. 20:1445–1460.
Black JA, Waxman SG, Smith ME. 1987. Macromolecular structure Devaux J, Alcaraz G, Grinspan J, Bennett V, Joho R, Crest M, et al.
of axonal membrane during acute experimental allergic encephalo- 2003. Kv3.1b is a novel component of CNS nodes. J Neurosci
myelitis in rat and guinea pig spinal cord. J Neuropath Exp Neurol 23:4509–4518.
46:167–184. Devaux JJ, Kleopa KA, Cooper EC, Scherer SS. 2004. KCNQ2 is a
Blakemore WF. 1977. Remyelination of CNS axons by Schwann cells nodal K+ channel. J Neurosci 24:1236–1244.
transplanted from the sciatic nerve. Nature 266:68–69. De Waegh SM, Lee VM, Brady ST. 1992. Local modulation of neurofi la-
Blakemore WF, Crang AJ. 1988. Extensive oligodendrocyte remyelina- ment phosphorylation, axonal caliber, and slow axonal transport by
tion following injection of cultured central nervous system cells into myelinating Schwann cells. Cell 68:451–463.

M Y E L I N, I M P U L S E C O N D U C T I O N, A N D T H E PAT H O P H YS I O L O GY O F D E M Y E L I N AT I O N • 539
Dugandzija-Novakovic S, Koszowski AG, Levinson SR, Shrager P. 1995. promotes axonal regeneration in rat spinal cord. Nat Biotechnol
Clustering of Na+ channels and node of Ranvier formation in remy- 18:949–953.
elinating axons. J Neurosci 15:492–503. Imaizumi T, Lankford KL, Waxman SG, Green CA, Kocsis JD. 1998.
Duncan ID, Hammang JP, Jackson KF, Wood PM, Bunge RP, Langford Transplantated olfacotry ensheathing cells remyelinate and enhance
L. 1988. Transplantation of oligodendrocytes and Schwann cells into axonal conduction in the demyelinated dorsal columns of the rat spi-
the spinal cord of the myelin-deficient rat. J Neurocytol 17:351–360. nal cord. J Neurosci 18:6176–6185.
Dutta R, McDonough J, Yin X, Peterson J, Chang A., Torres T, et al. Keirstead HS, Nistor G, Bernal G, Totoiu M, Cloutier F, Sharp K, et al.
2006. Mitochondrial dysfunction as a cause of axonal degeneration 2005. Human embryonic stem cell-derived oligodendrocyte progeni-
in multiple sclerosis patients. Ann Neurol 59:478–489. tor cell transplants remyelinate and restore locomotion after spinal
Einstein O, Grigoriadis N, Mizrachi-Kol R, Reinhartz E, Polyzoidou cord injury. J Neurosci 25:4694–4705.
E, Lavon I, et al. 2006. Transplanted neural precursor cells reduce Kocsis JD, Eng DL, Gordon TR, Waxman SG. 1987. Functional differ-
brain inflammation to attenuate chronic experimental autoimmune ences between 4-aminopyridine and tetraethylammonium-sensitive
encephalomyelitis. Exp Neurol 198:275–284. potassium channels in myelinated axons. Neurosci Lett 75:193–198.
England JD, Gamboni F, Levinson SR. 1991. Increased numbers Kocsis JD, Ruiz JA, Cummins KL. 1982. Modulation of axonal excit-
of sodium channels form along demyelinated axons. Brain Res ability mediated by surround electrical activity: an intra-axonal study.
548:334–337. Exp Brain Res 47:151–153.
England JD, Gamboni F, Levinson SR, Finger TE. 1990. Changed dis- Kocsis JD, Waxman SG. 1985. Demyelination: causes and mechanisms
tribution of sodium channels along demyelinated axons. Proc Natl of clinical abnormality and functional recovery. In: Koetsier JC (ed.),
Acad Sci U S A 87:6777–6780. Handbook of clinical neurology, vol. 3. The demyelinating diseases.
Felts P, Baker TA, Smith KJ. 1977. Conduction in segmentally demyeli- Amsterdam: Elsevier, pp. 29–47.
nated mammalian central axons. J Neurosci 17:7267–7277. Kohlschutter A, Bley A, Brockmann K, Gartner J, Krageloh-Mann I,
Felts P, Smith K. 1991. Conduction properties of central nerve fibers Rolfs A, et al. 2010. Leukodystrophies and other genetic metabolic
remyelinated by Schwann cells. Brain Res 574:178–192. leukoencephalopathies in children and adults. Brain Dev 32:82–89.
Foster RE, Connors BW, Waxman SG. 1982. Rat optic nerve: Kole MH, Letzkus JJ, Stuart GJ. 2007. Axon initial segment Kv1 chan-
Electrophysiological, pharmacological, and anatomical studies dur- nels control axonal action potential waveform and synaptic efficacy.
ing development. Dev. Brain Res 3:361–376. Neuron 55:633–647.
Foster RE, Whalen CC, Waxman SG. 1980. Reorganization of the Koles ZJ, Rasminsky M. 1972. A computer simulation of conduction in
axonal membrane of demyelinated nerve fibers: morphological evi- demyelinated nerve fibres. J Physiol (Lond) 227:351–364.
dence. Science 210:661–663. Lachapelle F, Gumpel M, Baulac M, Jacque C, Duc P, Baumann N.
Franklin RJ, Blakemore WF. 1997. To what extent is oligodendrocyte 1983. Transplantation of CNS fragments into the brain of shiverer
progenitor migration a limiting factor in the remyelination of multi- mutant mice: extensive myelination by implanted oligodendrocytes.
ple sclerosis lesions? Mult Scler 3:84–87. I. Immunohistochemical studies. Dev Neurosci 6:325–334.
Funch PG, Faber DS. 1984. Measurement of myelin sheath resistances: Learish RD, Brustle O, Zhang SC, Duncan ID. 1999. Intraventricular
implications for axonal conduction and pathophysiology. Science transplantation of oligodendrocyte progenitors into a fetal mye-
225:538–540. lin mutant results in widespread formation of myelin. Ann Neurol
Gansmuller A, Lachapelle F, Baron-Van Evercooren A, Hauw JJ, 46:716–722.
Baumann N, Gumpel M. 1986. Transplantations of newborn CNS Lombet A, Laduron P, Mourre C, Jacomet Y, Lazdunski M. 1985.
fragments into the brain of shiverer mutant mice: extensive myeli- Axonal transport of the voltage–dependent Na channel protein iden-
nation by transplanted oligodendrocytes. II. Electron microscopic tified by its tetrodotoxin–binding site in rat sciatic nerves. Brain Res
study. Dev Neurosci 8:197–207. 345:153–158.
Gasser A, Cheng X, Gilmore ES, Tyrrell L, Waxman SG, Dib-Hajj SD. Lorincz A, Nusser Z. 2008. Cell-type-dependent molecular composition
2010. Two Nedd4-binding motifs underlie modulation of sodium of the axon initial segment. J Neurosci 28:14329–14340.
channel Nav1.6 by p38 MAPK. J Biol Chem 285:26149–26161. Mahad DJ, Ziabreva I, Campbell G, Lax N, White K, Hanson PS, et al.
Goldberg EM, Clark BD, Zagha E, Nahmani M, Erisir A, Rudy B. 2008. 2009. Mitochondrial changes within axons in multiple sclerosis.
K+ channels at the axon initial segment dampen near-threshold excit- Brain 132:1161–1174.
ability of neocortical fast-spiking GABAergic interneurons. Neuron Makowska A, Pritchard J, Sanvito L, Gregson N, Peakman M, Hayday A,
58:387–400. et al. 2008. Immune responses to myelin proteins in Guillain-Barre
Gray PT, Ritchie JM. 1985. Ion channels in Schwann and glial cells. syndrome. J Neurol Neurosurg Psychiatry 79:664–671.
Trends Neurosci 8:411–415. McDonald JW, Liu XZ, Qu Y, Liu S, Mickey SK, Turetsky D, et al. 1999.
Groves AK, Barnett SC, Franklin RJ, Crang AJ, Mayer M, Blakemore Transplanted embryonic stem cells survive, differentiate and promote
WF, et al. 1993. Repair of demyelinated lesions by transplantation of recovery in injured rat spinal cord. Nat Med 5:1410–1412.
purified O-2A progenitor cells. Nature 362:453–455. McDonald WI. 1963. The effects of experimental demyelination on con-
Hildebrand C. 1971. Ultrastructural and light-microscopic studies of the duction in peripheral nerve: A histological and electrophysiological
developing feline spinal cord white matter. I. The nodes of Ranvier. study. Electrophysiological observations. Brain 86:501–524.
Acta Physiol Scand Suppl 364:81–101. McDonald WI. Sears TA. 1970. The effects of experimental demy-
Hines M, Shrager P. 1991. A computational test of the requirements for elination on conduction in the central nervous system. Brain
conduction in demyelinated axons. Resto Neurol Neurosci 3:81–93. 93:583–598.
Hirano A, Levine S, Zimmerman HM. 1968. Remyelination in the cen- Moll C, Mourre C, Lazdunski M, Ulrich J. 1991. Increase of sodium
tral nervous system after cyanide intoxication. J Neuropath Exper channels in demyelinated lesions of multiple sclerosis. Brain Res
Neurol 27:234–245. 556:311–316.
Honmou O, Felts P, Waxman S, Kocsis J. 1996. Restoration of normal Nave KA. 2010. Myelination and support of axonal integrity by glia.
conduction properties in demyelinated spinal cord axons in the Nature 468:244–252.
adult rat by transplantation of exogenous Schwann cells. J Neurosci Nistor GI, Totoiu MO, Haque N, Carpenter MK, Keirstead HS. 2005.
16:3199–3208. Human embryonic stem cells differentiate into oligodendrocytes
Hu BY, Du ZW, Zhang SC. 2009. Differentiation of human oligoden- in high purity and myelinate after spinal cord transplantation. Glia
drocytes from pluripotent stem cells. Nat Protoc 4:1614–1622. 49:385–396.
Huxley AF, Stämpfli R. 1949. Evidence for saltatory conduction in Ogawa Y, Rasband MN. 2008. The functional organization and assem-
peripheral myelinated nerve fibres. J Physiol (London) 108:315–339. bly of the axon initial segment. Curr Opin Neurobiol 18:307–313.
Imaizumi T, Lankford KL, Burton WV, Fodor WL, Kocsis JD. 2000. Pan Z, Kao T, Horvath Z, Lemos J, Sul JY, Cranstoun SD, et al. 2006.
Xenotransplantation of transgenic pig olfactory ensheathing cells A common ankyrin-G-based mechanism retains KCNQ and NaV

540 • NEUROGLIA
channels at electrically active domains of the axon. J Neurosci Sontheimer H, Fernandez-Marques E, Ullrich N, Pappas CA, Waxman
26:2599–2613. SG. 1994. Astrocyte Na+ channels are required for maintenance of
Pastores GM. 2009. Krabbe disease: an overview. Int J Clin Pharmacol Na+/K(+)-ATPase activity. J Neurosci 14:2464–2475.
Ther 47(Suppl 1):S75–81. Stanley GP, Pender MP. 1991. Pathophysiology of chronic relapsing
Pluchino S, Quattrini A, Brambilla E, Gritti A, Salani G, Dina G, et al. experimental allergic encephalomyelitis. Brain 114:1827–1853.
2003. Injection of adult neurospheres induces recovery in a chronic Steffensen I, Waxman SG, Mills L, Stys PK. 1997. Immunolocalization
model of multiple sclerosis. Nature 422:688–694. of the Na(+)-Ca2+ exchanger in mammalian myelinated axons.
Potter GB, Rowitch DH, Petryniak MA. 2011. Myelin restoration: Brain Res 776:1–9.
progress and prospects for human cell replacement therapies. Arch Steinman L, Martin R, Bernard C, Conlon P, Oksenberg JR. 2002.
Immunol Ther Exp (Warsz) 59:179–193. Multiple sclerosis: deeper understanding of its pathogenesis reveals
Prineas J, Connell F. 1978. The fine structure of chronically active multi- new targets for therapy. Annu Rev Neurosci 25:491–505.
ple sclerosis plaques. Neurology 28:68–75. Stys P, Sontheimer H, Ransom B, Waxman S. 1993. Noninactivating,
Rasband MN, Trimmer JS, Schwarz TL, Levinson SR, Ellisman MH, tetrodotoxin-sensitive Na+ conductance in optic nerve axons. Proc
Schachner M, et al. 1998. Potassium channel distribution, clustering, Natl Acad Sci U S A 90:6976–6980.
and function in remyelinating rat axons. J Neurosci 18:36–47. Tasaki I. 1959. Conduction of the nerve impulse: In: Handbook of
Rasminsky M, Sears TA. 1972a. Internal conduction in undissected Physiology. Sect. 1 Neurophysiology. Bethesda, MD: American
demyelinated nerve fibers. J Physiol 227:323–350. Physiological Society.
Rasminsky M, Sears TA. 1972b. Internodal conduction in undissected Trapp BD, Bo L, Mork S, Chang A. 1999. Pathogenesis of tissue injury in
demyelinated nerve fibers. J Physiol (London) 227:323–350. MS lesions. J Neuroimmunol 98:49–56.
Ritchie JM, Rogart RB. 1977. The density of sodium channels in mamma- Utzschneider DA, Archer DR, Kocsis JD, Waxman SG, Duncan ID.
lian myelinated nerve fibers and the nature of the axonal membrane 1994. Transplantation of glial cells enhances action potential con-
under the myelin sheath. Proc Natl Acad. Sci U S A 74:211–215. duction of amyelinated spinal cord axons in the myelin-deficient rat.
Ritchie JM, Waxman SG, Waksman BH. 1981. Basic and clinical elec- Proc Natl Acad Sci U S A 91:53–57.
trophysiology of demyelinating diseases. Neurology 31:1308–1310. VabnickI, Shrager P. 1998. Ion channel redistribution and function dur-
Röper J, Schwarz JR. 1989. Heterogeneous distribution of fast and slow ing development of the myelinated axon. J Neurobiol 37:80–96.
potassium channels in myelinated rat nerve fibers. J Physiol (London). Wang H, Kunkel DD, Martin TM, Schwartzkroin PA, Tempel BL.
416:93–110. 1993. Heteromultimeric K+ channels in terminal and juxtaparan-
Rosenbluth J. 1985. Intramembranous particle patches in myelin-deficient odal regions of neurons. Nature 365:75–79.
rat mutant. Neurosci Letts 62:19–24. Wang SS, Shultz JR, Burish MJ, Harrison KH, Hof PR, Towns LC,
Rosenbluth J, Blakemore WF. 1984. Structural specializations in cat of et al. 2008. Functional trade-offs in white matter axonal scaling.
chronically demyelinated spinal cord axons as seen in freeze-fracture J Neurosci 28:4047–4056.
replicas. Neurosci Letts 48:171–177. Waxman SG. 1977. Conduction in myelinated, unmyelinated, and
Rudy B, McBain CJ. 2001. Kv3 channels: voltage-gated K+ chan- demyelinated fibers. Arch Neurol 34:585–590.
nels designed for high-frequency repetitive firing. Trends Neurosci Waxman SG. 1978. Prerequisites for conduction in demyelinated fibers.
24:517–526. In: Neurology. Vol. 28. pp. 27–34.
Rush AM, Dib-Hajj SD, Waxman SG. 2005. Electrophysiological prop- Waxman SG. 1982. Current concepts in neurology: membranes, mye-
erties of two axonal sodium channels, Nav1.2 and Nav1.6, expressed lin and the pathophysiology of multiple sclerosis. N Engl J Med
in mouse spinal sensory neurones. J Physiol. 564:803–815. 306:1529–1533.
Rush AM, Wittmack EK, Tyrrell L, Black JA, Dib-Hajj SD, Waxman Waxman SG. 2006. Axonal conduction and injury in multiple sclerosis:
SG. 2006. Differential modulation of sodium channel Na(v)1.6 by the role of sodium channels. Nat Rev Neurosci 7:932–941.
two members of the fibroblast growth factor homologous factor 2 Waxman SG, Bennett MVL. 1972. Relative conduction velocities of
subfamily. Eur J Neurosci 23:2551–2562. small myelinated and non- myelinated fibers in the central nervous
Sasaki M, Black JA, Lankford KL, Tokuno HA, Waxman SG, Kocsis system. Nat New Biol 238:217–219.
JD. 2006. Molecular reconstruction of nodes of Ranvier after remy- Waxman SG, Black JA, Kocsis JA, Ritchie JM. 1989. Low density of
elination by transplanted olfactory ensheathing cells in the demyeli- sodium channels supports action potential conduction in axons of
nated spinal cord. J Neurosci 26:1803–1812. neonatal rat optic nerve. Proc Natl Acad Sci 86:1406–1410.
Schafer DP, Custer AW, Shrager P, Rasband MN. 2006. Early events in Waxman SG, Brill MH. 1978. Conduction through demyelinated
node of Ranvier formation during myelination and remyelination in plaques in multiple sclerosis: computer simulations of facilitation by
the PNS. Neuron Glia Biol 2:69–79. short internodes. J Neurol Neurosurg Psychiatry 41:408–417.
Sears TA, Bostock H. 1981. Conduction failure in demyelination: Is it Waxman SG, Quick DC. 1977. Cytochemical differentiation of the
inevitable? In: Wxman SG, Ritchie JM (eds.), Demyelinating dis- axon membrane in A- and C-fibers. J Neurol Neurosurg Psychiatry
eases: basic and clinical electrophysiology. New York: Raven Press, 40:769–774.
pp. 357–375. Waxman SG, Ritchie JM. 1993. Molecular dissection of the myelinated
Sears TA, Bostock H, Sherratt M. 1978. The pathophysiology of demy- axon. Ann Neurol 33:121–136.
elination and its implications for the symptomatic treatment of mul- Waxman SG, Sims TJ. 1984. Specificity in central myelination: evidence
tiple sclerosis. Neurology 28:21–26. for local regulation of myelin thickness. Brain Res 292:179–185.
Shrager P. 1989. Sodium channels in single demyelinated mammalian Waxman SG, Sims TJ, Gilmore SA. 1988. Cytoplasmic membrane elab-
axons. Brain Res 483:149–154. orations in oligodendrocytes during myelination of spinal motoneu-
Smith KJ, Blakemore WF, McDonald WI. 1981. The restoration of con- ron axons. Glia 1:286–291.
duction by central remyelination. Brain 104:383–404. Waxman SG, Sontheimer H, Black JA, Minturn JE, Ransom BR. 1993.
Smith KJ, Bostock H, Hall SM. 1982. Saltatory conduction precedes Dynamic aspects of sodium channel expression in astrocytes. Adv
remyelination in axons demyelinated with lysophosphatidyl choline. Neurol. 59:135–155.
J Neurol Sci54:13–31. Waxman SG, Wood SL. 1984. Impulse conduction in inhomogeneous
Smith KJ, Hall SM. 1980. Nerve conduction during peripheral demyeli- axons: Effects of variation in voltage-sensitive ionic conductances
nation and remyelination. J Neurol Sci 104:383–404. on invasion of demyelinated axon segments and preterminal fibers.
Smith KJ, McDonald WI. 1980. Spontaneous and mechanically evoked Brain Res 294:111–122.
activity due to central demyelinating lesions. Nature 286:154–155. Windrem MS, Nunes MC, Rashbaum WK, Schwartz TH, Goodman
Smith ME, Kocsis JD, Waxman SG. 1983. Myelin protein metabolism RA, McKhann G 2nd, et al. 2004. Fetal and adult human oligoden-
in demyelination and remyelination in the sciatic nerve. Brain Res drocyte progenitor cell isolates myelinate the congenitally dysmyeli-
270:37–44. nated brain. Nat Med 10:93–97.

M Y E L I N, I M P U L S E C O N D U C T I O N, A N D T H E PAT H O P H YS I O L O GY O F D E M Y E L I N AT I O N • 541
Windrem MS, Schanz SJ, Guo M, Tian GF, Washco V, Stanwood N, 2B: association with Nav1.6 and selective colocalization at nodes of
et al. 2008. Neonatal chimerization with human glial progenitor cells Ranvier of dorsal root axons. J Neurosci 24:6765–6775.
can both remyelinate and rescue the otherwise lethally hypomyeli- Wittmack EK, Rush AM, Hudmon A, Waxman SG, Dib-Hajj SD.
nated shiverer mouse. Cell Stem Cell 2:553–565. 2005. Voltage-gated sodium channel Nav1.6 is modulated by p38
Witte ME, Bo L, Rodenburg RJ, Belien JA, Musters R, Hazes T, et al. mitogen-activated protein kinase. J Neurosci 25:6621–6630.
2009. Enhanced number and activity of mitochondria in multiple Young EA, Fowler CD, Kidd GJ, Chang A, Rudick R, Fisher E,
sclerosis lesions. J Pathol 219:193–204. et al. 2008. Imaging correlates of decreased axonal Na+/K+
Wittmack EK, Rush AM, Craner MJ, Goldfarb M, Waxman SG, ATPase in chronic multiple sclerosis lesions. Ann Neurol 63:
Dib-Hajj SD. 2004. Fibroblast growth factor homologous factor 428–435.

542 • NEUROGLIA
43.
TRANSCRIPTION FACTOR S IN MYELINATING CELLS
Michael Wegner

A B B R E VI AT I O N S network that exceed its buffering capacity, it will move from its
old metastable state to a different one and thereby establish a new,
BMP bone morphogenetic proteins characteristic pattern of gene expression. In other words: a gene
cAMP 3c,5ccyclic adenosine monophosphate regulatory network is robust enough to guarantee the expression
CNS central nervous system pattern that is characteristic for a cell in a particular stage of its
Gpr G-protein–coupled receptor development, but also allows the ordered changes in gene expres-
HDAC histone deacetylase sion that underlie lineage progression and maturation.
HMG high mobility group To satisfy the need for stability and at the same time allow
MRF myogenic regulatory factor change, gene regulatory networks usually contain transcrip-
MSE myelinating Schwann cell enhancer tion factors that are present at all times and those that are only
NFAT nuclear factor of activated T cells recruited at specific times or under certain circumstances.
OPC oligodendrocyte precursor Those that are constitutively present function as determinants
PDGFRα platelet-derived growth factor receptor α of cell type identity and lineage, whereas the facultative and
PNS peripheral nervous system transiently present ones mark a specific stage. The network
POU Pit1-Oct1/2-Unc86 also needs checks and balances. Reinforcing and antagonizing
SCAN Srezbp, Ctfin51, AW-1, and number 18 activities, stabilizing and destabilizing activities, transcriptional
YY Yin Yang activators and repressors, and pro- and anti-differentiation
Zfp zinc finger protein factors are present at all times. This means that transcription
factors are expected in myelinating glia with the following
functions: (1) factors that establish competence for myelina-
tion and impart cell identity, (2) factors that initiate the myeli-
1 INTRODUCTION nation program and maintain it, and (3) factors that actively
counteract this program at various stages of cell development.
Transcription factors are the immediate effectors of gene Transcription factors have two main modes of action.
expression. Considering the special gene expression profile They promote or suppress recruitment of the basal transcrip-
of myelinating glia (see chapter 29), it has long been assumed tion machinery to genes by binding to their regulatory regions
that the transcription factors in charge are also unique and (i.e. promoter, enhancer, and silencer elements). Contacts
restricted in their occurrence to Schwann cells and oligoden- with components of the basal transcription machinery can be
drocytes. However, such factors are few, and none is completely direct or indirect through transcriptional cofactors such as the
restricted in occurrence and function to myelinating glia. Gene Mediator complex. Alternatively, transcription factors recruit
expression in myelinating glia is thus caused to a large extent by chromatin modifying enzymes and chromatin remodeling
transcription factors with limited specificity. Conceptionally, complexes, and by modifying the epigenetic state of chromatin
this is feasible because gene expression is not a readout of the change accessibility of their target genes for the transcriptional
action of single transcription factors, but of transcription factor apparatus. It follows that many transcription-related proteins
combinations and their synergistic, additive, and antagonistic including components of the basal transcription machinery,
interactions in the context of a gene regulatory network. chromatin modifiers, and remodeling complexes are tightly
The advantages of regulating gene expression through such linked to the gene regulatory network.
networks are manifold. Considering that many different gene Various types of small RNAs are also part of the network.
regulatory networks with unique characteristics can be gen- MicroRNAs in particular play a role in fine tuning network activ-
erated from relatively few building blocks in a combinatorial ity. Considering the multitude of transcription factors involved,
manner, a network is resource-efficient in evolutionary terms. the intricate relationships among them and the many interactions
Additionally, a gene regulatory network has substantial homeo- between transcription factors, basal transcription machinery,
static capacity because of the many redundant, reinforcing, syn- chromatin modifiers, remodelers, and microRNAs, this chapter
ergistic, and antagonistic relationships among its constituting focuses on key transcription factors in myelinating glia and their
transcription factors. As a consequence, it is optimally suited to interactions within the network. Other comprehensive accounts
maintain expression patterns. At the same time, a gene regulatory of transcription factors in myelinating glia have recently been
network remains receptive to input. If changes are induced in the published (e.g., Li et al. 2009; Svaren and Meijer 2008).

543
2 TR ANSCRIPTIONAL CONTROL et al. 2011). Although required for Schwann cell specification,
O F P E R I P H E R A L N E RVO U S SYS T E M Sox10 alone cannot be sufficient because it is expressed in neu-
M Y E L I N AT I O N BY S C H WA N N C E L L S ral crest precursors long before, and because it is also involved
in other neural crest specification events including melanocyte
specification. This is not surprising because HMG-domain
2.1 S C H WA N N C E L L S P E C I FI C AT I O N
containing, minor groove binding transcription factors gen-
Schwann cells as the myelinating glia of the Peripheral erally cooperate in gene activation with other transcription
Nervous System (PNS) are neural crest derivatives (see chap- factors that bind the major groove of DNA (Wegner 2010).
ter 14). Therefore signals and regulatory mechanisms must The identity of these cooperating transcription factors during
exist that commit neural crest progenitor cells to the Schwann Schwann cell specification is currently unknown.
cell lineage. This first step in lineage development is still
poorly understood on the transcriptional level. Although
2.2 T H E S C H WA N N C E L L O N T H E WAY
loss of several transcription factors leads to a reduction in
TO MY E L I NAT I O N
Schwann cell numbers, it has rarely been analyzed whether
Schwann cell development is directly influenced or indirectly Sox10 expression does not extinguish after specification (see
through effects on early neural crest development. To date the Fig. 43.1). Instead it continues in Schwann cells at all times
best studied example of a transcription factor with influence (Kuhlbrodt et al. 1998). This expression is ensured by mul-
on Schwann cell specification is Sox10 (Fig. 43.1). This fac- tiple enhancers in the vicinity of the Sox10 gene that respond
tor belongs to the SoxE subgroup of the Sox protein family to different sets of transcription factors and autoregulation
and contains a DNA-binding high mobility group (HMG)- (Wahlbuhl et al. 2011; Werner et al. 2007). Conditional
domain as defining feature (Wegner 2010). It has been shown deletion of Sox10 at various stages of Schwann cell develop-
in Sox10-deficient mice and zebrafish that neural crest cells ment and the analysis of hypomorphic mouse mutants has
find their way to the nerve roots, but then fail to convert confirmed that Sox10 remains functional and contributes
into Schwann cell precursors that express the ErbB2/ErbB3 to Schwann cell development during the consecutive stages
receptor, which in turn establishes neuregulin dependence of the immature Schwann cell, the promyelinating and
of this cell type (Britsch et al. 2001; Dutton et al. 2001) (see the myelinating Schwann cell (see chapter 14) and is even
also chapter 14). In fact ErbB3 expression in Schwann cells is required in the mature Schwann cell for myelin maintenance
under direct control of a Sox10-responsive enhancer (Prasad (Bremer et al. 2011; Finzsch et al. 2010; Fröb et al. 2012;

Schwann cell development

NC cell SCP iSC pSC mSC

Oct6 (& Brn2) Krox20


Sox10

Sox10
Nkx2.2 MRF

Olig2 (& Olig1)

NEP cell OPC pOL mOL

Oligodendroglial development

Figure 43.1 Overview of Transcription Factors with Restricted Occurrence and Prodifferentiation Function in Myelinating Glia. Expression during
Schwann cell development (upper part) is indicated by red bars, expression during oligodendroglial development (lower part) by green bars. Axons
are in blue. NC, Neural crest cell; SCP, Schwann cell precursor; iSC, immature Schwann cell; pSC, promyelinating Schwann cell; mSC, myelinating
Schwann cell; NEP, neuroepithelial precursor; OPC, oligodendrocyte precursor; pOL, pro-oligodendrocyte; mOL, myelinating oligodendrocyte.

544 • NEUROGLIA
Schreiner et al. 2007). Sox10 can therefore be regarded as a transcription factor Oct6 (also known as Pou3f1, Scip, or
marker for Schwann cell lineage and identity. It represents one Tst-1) is the key effector at the downstream end of this signal-
of the invariant building blocks of the gene regulatory network ing pathway (Monuki et al. 1989) (see also chapters 7 and 14).
in this cell type. Intriguingly, Sox10 activates different target In fact, Oct6 expression cannot be detected in zebrafish in
genes during various developmental stages. Sox10-dependent the absence of Gpr126 (Monk et al. 2009). Another fac-
ErbB3 activation occurs already in the Schwann cell precur- tor required for Oct6 expression in Schwann cells is Sox10
sor (Britsch et al. 2001; Prasad et al. 2011), whereas activa- (Fig. 43.2A), which binds to and activates the Schwann
tion of Desert Hedgehog starts in the immature Schwann cell cell-specific enhancer downstream of the Oct6 gene ( Jagalur
(Finzsch et al. 2010) and activation of myelin genes such as et al. 2011; Schreiner et al. 2007). It is unknown whether
Myelin Associated Glycoprotein, Connexin-32, Myelin Protein Sox10 activity is itself regulated by cAMP or rather cooperates
Zero, Peripheral Myelin Protein 22, Periaxin, and Myelin Basic with a cAMP-responsive transcription factor to regulate Oct6
Protein (see chapter 7) is delayed until the onset of myelina- expression. One such cAMP-responsive factor in Schwann
tion (Bondurand et al. 2001; Denarier et al. 2005; Jones et al. cells is the broadly expressed p65 subunit of NF-NB (Yoon
2007, 2011, 2012; LeBlanc et al. 2007; Peirano et al. 2000). et al. 2008). PKA-dependent phosphorylation of NF-NB in
This further emphasizes the need for cooperating tran- response to high cAMP concentrations increases Oct6 levels.
scriptions factors that modulate the function of Sox10 in a Expression of Oct6 initiates late during the immature
stage-specific manner. One way of identifying these factors Schwann cell stage and peaks during the promyelinating
is the detailed dissection of Schwann cell–specific regulatory stage in which Schwann cells prepare for myelination and
regions which have been identified for most of the mentioned are in a one-to-one relationship with large caliber axons (see
Sox10-responsive genes. Fig. 43.1) (see chapter 14). In fact, Oct6 is one of the major
At least some of the cooperating transcription factors should driving forces in the transition from immature into myelinat-
be responsive to signals that drive Schwann cell development ing Schwann cells. This is documented by the transient arrest
and myelination such as increased intracellular cAMP levels of Schwann cell development in the promyelinating stage in
(Monuki et al. 1989). In zebrafish and mammals the rise in Oct6-deficient mice (Bermingham et al. 1996; Jaegle et al.
cAMP levels is caused by activation of the G-protein–coupled 1996). This relatively mild phenotype is caused by functional
orphan receptor Gpr126 (Monk et al. 2009, 2011). The axonal redundancy between Oct6 and the closely related and coex-
ligand for the receptor is unknown, but the POU-domain pressed POU-domain transcription factor Brn2 (also known

A Oct6
Krox20
Sox10 myelin
genes

iSC pSC mSC


B
MSE
N
FA
T

YY

AC Sox10
1

Oct6
HD

Pol ll
P Krox20

Figure 43.2 Essential Regulatory Events During Schwann Cell Differentiation. A. Genetic and functional interactions of the prodifferentiation
factors Sox10, Oct6, and Krox20 in Schwann cells. B. Model of MSE-dependent Krox20 gene activation in myelinating Schwann cells. P, Promoter;
PolII, RNA polymerase II.

T R A N S C R I P T I O N FAC TO R S I N M Y E L I N AT I N G C E L L S • 545
as Pou3f2). In accord, mice with deficiency for both Oct6 biosynthetic enzymes are directly bound by Krox20 in vivo
and Brn2 exhibit a dramatically enhanced arrest at the pro- (Denarier et al. 2005; Jones et al. 2007, 2011, 2012; LeBlanc
myelinating stage and severe hypomyelination ( Jaegle et al. et al. 2006, 2007), and expression of the genes depends on
2003). This is just one example for the frequently existing Krox20 (Nagarajan et al. 2001) and its cofactors Nab1 and
functional redundancy between transcription factors in myeli- Nab2 (Le et al. 2005b). Additionally, there is support from
nating cells. SREBP transcription factors in the regulation of lipid produc-
In addition to the cAMP pathway, there is a strong influence tion (Verheijen et al. 2003) and from Sox10 in the regulation
on Schwann cell development by neuregulins through activa- of myelin genes. In fact, Krox20 and Sox10 often target the
tion of the ErbB2/ErbB3 receptor. Downstream signaling same regulatory regions in myelinating Schwann cells and
involves PI3 kinase activation, increased intracellular calcium, cooperate to induce high levels of myelin gene expression.
and the MAP kinase pathway (see chapter 14). These pathways This cooperativity is further strengthened by direct interac-
in turn influence the function of a number of transcriptional tions between the two proteins and cross-stimulatory effects
regulators that are present in Schwann cells around the time of on their respective expression (Ghislain and Charnay 2006;
myelination. The DNA-binding activity of the p65 subunit of Reiprich et al. 2010; Wahlbuhl et al. 2011; Wissmüller et al.
NF-NB is enhanced (Limpert and Carter 2010). Additionally, 2006). The multiple links between Sox10 and Krox20 are
the broadly expressed transcription factor YY1 becomes quite remarkable and likely indicate the central role of this
phosphorylated (He et al. 2010), and calcineurin-dependent pair of transcription factors in the gene regulatory network of
dephosphorylation of NFATc3/c4 leads to nuclear transloca- myelinating Schwann cells.
tion and activation (Kao et al. 2009).
All these signal-dependent changes in the gene regula-
2.3 C O U N T E R AC T I N G MY E L I NAT I O N
tory network contribute to the initiation of the myelination
I N S C H WA N N C E L L S
program in Schwann cells. This is closely connected with the
induction of the zinc-finger transcription factor Krox20 (also Krox20 and Sox10 are functional both during induction and
known as Egr2), which is often referred to as the master regula- maintenance of myelin gene expression. This is not the case for
tor of peripheral myelination (see Fig. 43.1) because Schwann Oct6. In fact Oct6 has to be downregulated after the initial
cell development is stalled permanently at the promyelinat- phases for myelination to proceed properly. Experimentally
ing stage in Krox20-deficient mice (Topilko et al. 1994) and prolonged Oct6 expression leads to hypomyelination and loss
Schwann cells fail to form myelin. of axons in the PNS (Ryu et al. 2007). It has been proposed
Activation of Krox20 requires the Oct6 protein (and/ that Krox20 plays a role in the eventual shut-down of Oct6
or its relative Brn2) to bind to the myelinating Schwann cell expression (Zorick et al. 1999). It is known that Oct6 itself
enhancer (MSE) of the Krox20 gene (Ghislain and Charnay is also a negative regulator of its own expression ( Jaegle et al.
2006) (see Fig. 43.2B). The presence of Oct6 alone is, how- 2003). Why Oct6 affects early and late phase of myelination
ever, not sufficient to activate the MSE. It rather depends on differently is not yet clear. It shows however, that Oct6 has two
the synergistic support of Sox10 (Ghislain and Charnay 2006; distinct functions: it promotes initiation and the early steps of
Reiprich et al. 2010), the very same transcription factor that has the myelination program, but at the same time prevents transi-
already been involved in activating Oct6 expression (see Fig. tion into the fully mature state.
43.2A,B). Additionally, the MSE is targeted and activated by Such a behavior is typical for many transcription fac-
dephosphorylated NFATc3/c4 and phosphorylated YY1 (He tors with transient expression during Schwann cell develop-
et al. 2010; Kao et al. 2009) (see Fig. 43.2B). At least NFATc3/ ment. They function both as positive and negative regulators.
c4 again requires the synergistic support of Sox10 for Krox20 Immature Schwann cells, for instance, express the transcrip-
activation (Kao et al. 2009). A picture thus emerges in which tion factors cJun and Sox2, and contain nuclear complexes
several signaling pathways converge through their downstream of the Notch intracellular domain and RBPJ as a result of
effectors on the MSE and combine with Schwann cell identity active Notch signaling (Le et al. 2005a; Parkinson et al.
factors such as Sox10 to synergistically activate Krox20 and 2008; Woodhoo et al. 2009). These factors establish and
kick-start the myelination program. define the immature Schwann cell state. Active Notch sig-
Myelination not only requires transcription factor activity naling, for instance promotes the generation of immature
but also relies on microRNAs. This is evident from deletion of Schwann cells from Schwann cell precursors, controls prolif-
the microRNA-processing enzyme Dicer in Schwann cells and eration and regulates the size of the Schwann cell pool (see
the resulting myelination block (e.g., Dugas et al. 2010; Pereira chapter 14). Additionally, however, these factors also actively
et al. 2010; Yun et al. 2010). Numerous microRNAs are differ- repress the myelination program by inhibiting Krox20 expres-
entially expressed during Schwann cell development and thus sion, and thereby prevent precocious Schwann cell differentia-
represent candidates for stage-specific regulators (Gokey et al. tion. As a consequence, prolonged expression leads to delayed
2012). Intriguingly, at least nine of these differentially regu- myelination. Their deletion, in contrast, causes premature myeli-
lated microRNAs are under transcriptional control of Sox10. nation. Once inhibition is overcome in developing Schwann
While microRNAs participate in the regulation of myeli- cells and the myelinated state is established, Krox20 similarly
nation, Krox20 is the decisive regulator. Schwann cell-specific suppresses cJun and Sox2 expression. These transcription factors
regulatory regions of myelin genes and genes for lipid are therefore not only functionally antagonistic and expressed

546 • NEUROGLIA
cJun

iSC Sox2 Krox20

RBPJ/NICD

mSC

Figure 43.3 Cross-Regulatory Interactions Between the Differentiation Inhibitors cJun, Sox2, and RBPJ, and the Prodifferentiation Factor Krox20 in
Schwann Cells.

in a mutually exclusive manner, they also cross-inhibit each A PNS


other’s expression (Fig. 43.3). This implies that even mature
Schwann cells will start to re-express cJun, Sox2 and acti-

HD
vate Notch signaling if Krox20 levels drop to sufficiently low CBP

AC
levels. It grants Schwann cells a certain degree of plasticity and Ac p65
permits them to dedifferentiate in cases of nerve injury as a p65
precondition for the ensuing demyelination and remyelina-
tion processes (see chapter 14).
There is also evidence that genes coding for activators and
inhibitors of the myelination program carry opposite histone
marks in their chromatin at any given time, with trimethylated myelination
H3K4 being found on the active, and trimethylated H3K9
being found on the repressed gene (Chen et al. 2011). In myeli-
nating Schwann cells, prodifferentiation genes are marked by
trimethlyated H3K4, whereas antidifferentiation genes prefer-
entially carry H3K9. In immature Schwann cells, the situation
is reversed. Ordered changes in histone marks and chromatin
thus contribute to Schwann cell differentiation and myelina- TCF4
βcat

H
tion. So far, the effects of histone methylases and demethylases TCF4

DA
Groucho
C
on Schwann cell development have not been studied. The
only group of chromatin modifying enzymes studied B CNS
are histone deacetylases (HDACs), in particular HDAC1
and HDAC2. Their deletion stalls Schwann cell development Figure 43.4 Cofactor Dependency of Transcription Factors in
in vivo (Chen et al. 2011; Jacob et al. 2011). The mechanism Myelinating Glia. A. By switching CBP for HDAC, the p65 subunit
is, however, less straightforward than expected. The histone of NF-NB turns from a differentiation inhibitor in Schwann cells into a
pro-differentiation factor. B. Similarly, TCF4 changes activity in prooli-
deacetylases seem to predominantly target transcription fac- godendrocytes by exchanging E-catenin for Groucho and HDACs. Ac,
tors. One prime target is the p65 subunit of NF-NB, which by Acetylated lysine residue.
exchanging histone acetylase CBP for HDACs as interaction
partners, becomes deacetylated (Fig. 43.4A). This in turn causes
the replacement of inhibitory by activating histone marks on 2.4 T R A NS C R I P T I O NA L C O N T RO L O F
prodifferentiation genes such as Sox10 and the reverse on C E N T R A L N E RVO US SYS T E M MY E L I NAT I O N BY
anti-differentiation genes (Chen et al. 2011). A second O L I G O D E N D RO C Y T E S
HDAC target appears to be the Sox10 protein whose prod-
2.4.1 Oligodendroglial Specification
ifferentiation activity is increased in complex with HDAC2
( Jacob et al. 2011). These results show that chromatin changes In contrast to Schwann cells, oligodendrocytes are central
occur during differentiation and that the machinery involved nervous system (CNS) cells. Oligodendroglial cells therefore
in these changes is part of the gene regulatory network in have to be specified from pluripotent neuroepithelial pre-
Schwann cells. cursor cells in the ventricular zone of the embryonic CNS.

T R A N S C R I P T I O N FAC TO R S I N M Y E L I N AT I N G C E L L S • 547
The oligodendroglial specification process is much better This corresponds to the pMN domain (see Fig. 43.5). Olig2
understood than Schwann cell specification (see chapters 13, is thus a specific marker for the pMN domain and its pluri-
14). Oligodendrocyte precursors (OPCs) are generated in potent neuroepithelial precursor cells. It is required for the
defined, frequently ventrally located areas of the ventricular production of both motoneurons and OPCs (Lu et al. 2000,
zone, usually after local neurogenesis has already been com- 2002; Zhou and Anderson 2002; Zhou et al. 2000). The pro-
pleted (Rowitch 2004) (see also chapter 13). In the spinal cord, duction of both from the pMN domain is severely impaired
which serves as a simple model system, OPCs are first gener- in Olig2-deficient mice. However, OPCs do not completely
ated ventrally in the so-called pMN domain, and only later disappear in all CNS regions. This is because of the remain-
in the more dorsal dP3-dP5 domains (Fogarty et al. 2005). ing activity of the closely related Olig1 protein. A complete
The pMN domain first gives rise to motoneurons, and then to OPC loss is only observed in mice deficient for both Olig1
the majority of spinal cord OPCs (Fig. 43.5). This argues that and Olig2.
oligodendrocyte specification is dependent on at least two Considering that neurons and glia have to be pro-
different sets of transcription factors: one that defines pMN duced at different times, there must be factors that promote
domain identity and a second that allows a switch from neu- Olig2-dependent motoneuron production and prevent OPC
ron to glia production. production at early times. These include the bHLH transcrip-
The pMN domain identity depends on the sonic hedge- tion factors Neurogenin-1 and Neurogenin-2 (Mizuguchi
hog (Shh) gradient along the dorsoventral axis of the spinal et al. 2001; Nieto et al. 2001; Sun et al. 2001) (see Fig. 43.5). At
cord and the resulting concentration-dependent induction later stages, however, Olig2 function has to be modulated such
of different transcription factors (see chapter 13). Some tran- that glia instead of neurons are produced. This is achieved by
scription factors are only induced in a narrow window of Shh induction of gliogenic transcription factors including NFIA,
concentrations; others have a wider window. The Nk home- Sox9, and activated Notch (Deneen et al. 2006; Park and
odomain transcription factor Nkx2.2 is, for instance, only Appel 2003; Stolt et al. 2003) (see Fig. 43.5). The Neurogenins
expressed under highest concentrations in the most ventral are simultaneously down-regulated (Mizuguchi et al. 2001;
region, whereas the Iroquois homeodomain transcription fac- Nieto et al. 2001; Sun et al. 2001). As a consequence of the
tor Irx3 requires lower Shh concentrations (see Fig. 43.5). The combined action of Olig2 and gliogenic factors, OPCs
closely related Nkx6.1 and Nkx6.2 proteins in contrast can be are specified. Intriguingly, Olig2 undergoes different phos-
induced over a wider range of Shh concentrations so that they phorylation events at multiple sites (Li et al. 2011; Sun et al.
are coexpressed with both Nkx2.2 and Irx3 in their respec- 2011). One of these phosphorylations changes its ability
tive domain as well as in the domain between them (Cai et al. to interact with neurogenins (Li et al. 2011) arguing that
2005; Vallstedt et al. 2005). One of the target genes of Nkx6.1 Olig2 function is not only determined by the availability of
and Nkx6.2 is the gene for bHLH transcription factor Olig2. interacting transcription factors. Posttranslational modifica-
Considering that Nkx2.2 and Irx3 both repress Olig2 expres- tions additionally modulate the process. Such modifications
sion, Nkx6.1 and Nkx6.2 will activate Olig2 expression spe- may also determine interplay with transcriptional cofactors
cifically in the region between the Nkx2.2 and Irx3 domains. and chromatin modifying enzymes such as HDACs that

Neurogenesis Gliogenesis
(early) (late)

ventricular zone
mantle mantle

Irx3

Nkx6.1
MN Olig2 Nkx6.2 pMN Olig2 OPC

+ Neuro- Shh Nkx2.2 + Sox9


genin NFIA
Notch

ventral spinal cord

Figure 43.5 Expression of signaling molecules and transcription factors in the ventricular zone of the ventral spinal cord that lead to formation of the
pMN domain, and consecutive production of motoneurons and oligodendrocyte precursors in the mantle zone. MN, Motoneurons; OPC oligodendrocyte
precursor; Shh, sonic hedgehog.

548 • NEUROGLIA
are required for oligodendroglial specification and as much to maintain expression of the PDGFRD receptor (Finzsch
involved in oligodendrocyte as in Schwann cell development et al. 2008), the main oligodendroglial receptor for PDGF as
(Cunliffe and Casaccia-Bonnefil 2006; Shen et al. 2005; an important survival factor, mitogen and migratory cue (see
Ye et al. 2009). chapters 13, 20, and 22).
Oligodendroglial specification is marked by the induction Similarly, Notch signaling is active in OPCs (Genoud
of Sox10 expression (see Fig. 43.1). Analysis of the regulatory et al. 2002). One of the genes induced by Notch in OPCs
regions of the Sox10 gene furthermore indicates that Sox10 is is the bHLH transcription factor Hes5 (Liu et al. 2006),
a direct target gene of Olig2 in OPCs (Küspert et al. 2011). which has been mainly described as a differentiation inhibi-
Whereas motoneurons lose Olig2 expression after specifica- tor (Fig. 43.6). It prevents premature terminal differentiation
tion, cells of the oligodendrocyte lineage stay positive for and myelination by binding directly to the regulatory region
Olig2 throughout development and in the mature state (Lu of myelin genes and by repressing their expression following
et al. 2000; Zhou et al. 2000). The same holds for Sox10 (Stolt HDAC recruitment, histone deacetylation, and changes in
et al. 2002). In areas of the ventricular zone where Olig2 is not chromatin structure. Additionally, Hes5 interacts with Sox10
expressed in the pluripotent precursors, and OPCs arise inde- and thereby sequesters it in a complex that prevents Sox10
pendent of Olig2 such as the dP3-dP5 domains of the spinal from stimulating myelin gene expression.
cord, Olig2 comes on as soon as the cells are specified (Cai In this inhibitory function, Hes5 is not alone. The OPCs
et al. 2005; Vallstedt et al. 2005). Again, Olig2 is immediately also express high levels of the Id2 and Id4 inhibitors (see
followed by Sox10. Considering the expression of Olig2 in a Fig. 43.6) as downstream effectors of BMP signaling. Part of
subset of neuroepithelial precursors, motoneurons and some the Id proteins’ inhibitory function is exerted by heterodi-
astrocytes, Olig2 is not an unambiguous marker of oligoden- merization with Olig proteins (Kondo and Raff 2000; Samanta
drocyte lineage and identity. Rather it is the combination of and Kessler 2004; Wang et al. 2001). Additionally, high lev-
Olig2 and Sox10. Both the similarities and the differences to els of Sox6 and its close relative Sox5 can be found in OPCs
Schwann cells are striking. Only oligodendrocyte specifica- (Stolt et al. 2006). These members of the SoxD subgroup of
tion depends on Olig2, and only Schwann cell specification the Sox protein family also counteract premature differen-
depends on Sox10. Both types of glia, however, depend for tiation (see Fig. 43.6). They bind to the regulatory regions
their identity at all times on Sox10. of myelin genes, recruit HDACs and block the binding
of Sox10. In addition to these anti-differentiation activi-
2.4.2 Maintaining the Oligodendroglial Precursor ties, many of the mentioned transcription factors probably
State and Preventing Differentiation also have genuine functions in actively maintaining the pro-
genitor state of OPCs, although these aspects are less well
Whereas Schwann cells express Sox10 as the only SoxE pro- studied. For instance, Id2 and Id4 do not only inhibit myelin
tein, oligodendroglial cells additionally contain the closely gene expression but also promote proliferation (Kondo and
related Sox9 and Sox8 for most of their development (Stolt Raff 2000; Marin-Husstege et al. 2006; Samanta and Kessler
et al. 2005). The ensuing functional redundance makes it 2004; Wang et al. 2001). Similarly, evidence from chondro-
much more difficult to determine SoxE function during oli- cyte development indicates that Sox5 and Sox6 do not only
godendrocyte than Schwann cell development. Considering counteract SoxE activity but can also enhance it (Wegner
that both SoxE and Olig proteins are expressed in OPCs 2010). Thus it is utterly plausible that Sox5 and Sox6 prevent
as they proliferate, migrate, and distribute throughout the Sox10 from activating myelin genes prematurely, and at the
parenchyma, it is expected that these factors are relevant for same time help it to activate genes whose products are required
OPC development. In agreement, SoxE factors are required in OPCs.

CNS myelination
Zfp488 MRF activators

Sox10
Olig1/2 YY1

Nkx2.2 Mash1

Promoter Myelin Basic Protein Gene

Id2/4 Sox5/6

Hes5 repressors

Figure 43.6 Summary of Transcription Factors That Influence Oligodendroglial Myelin Gene Expression and CNS Myelination. Activating factors are
in dark green if their direct influence on myelin gene expression was shown, and in light green if postulated. Inhibiting factors are in red.

T R A N S C R I P T I O N FAC TO R S I N M Y E L I N AT I N G C E L L S • 549
2.4.3 Converting Precursors to Oligodendrocytes Myelination inhibitors are not only removed by the action
of antagonistic transcription factors. MicroRNAs also play a
Around the time when OPCs start to enter the final phase
role. As in Schwann cells, there is a plethora of microRNAs
of differentiation that leads to a myelinating phenotype,
with differential expression during oligodendrocyte develop-
a group of novel transcription factors appears in these
ment (Lau et al. 2008) and oligodendrocyte-specific dele-
pro-oligodendrocytes. These include Sox17 (Sohn et al. 2006).
tion of the microRNA processing Dicer enzyme proves the
This member of the SoxF subgroup of the Sox protein fam-
requirement of microRNAs for terminal differentiation of
ily is only distantly related to either SoxE or SoxD proteins.
oligodendrocytes and myelination (Dugas et al. 2010; Zhao
Sox17 appears to induce cell cycle exit by cyclin D upregula-
tion and by counteracting E-catenin (Chew et al. 2011), which et al. 2010). Among these differentially regulated microRNA
as the downstream effector of the Wnt signaling pathway are miR-219 and miR-338. As YY1, they are upregulated dur-
promotes OPC maintenance and proliferation (Fancy et al. ing the transition from late OPC to pro-oligodendrocyte and,
2009; Shimizu et al. 2005). Another modulator of Wnt and among other targets, recognize the 3c-UTR of Sox6 and Hes5
E-catenin activity at the transition from OPC to oligoden- transcripts. By inhibiting the translation of transcripts for
drocyte is the Tcf/Lef family transcription factor TCF4 (also myelination inhibitors, these microRNAs help cells to enter
known as TCF7L2). This E-catenin interacting transcription the final phase of differentiation.
factor has been reported to function as an inhibitor of myeli- Additionally, activators of the myelination program need
nation (He et al. 2007). Its deletion in the mouse, however, to be induced as late OPCs turn into prooligodendrocytes.
leads to a delay in myelination (Ye et al. 2009), arguing that Among them are the bHLH transcription factor Mash1 (also
it functions also as a prodifferentiation factor. Likely, this is known as Ascl1) and the Nk homeodomain transcription
due to an exchange of interacting cofactors from E-catenin to factor Nkx2.2 as indicated by the severe delay of terminal
Groucho corepressors and HDACs at this critical time of oli- oligodendrocyte differentiation and myelination in the cor-
godendroglial development (Ye et al. 2009) (see Fig. 43.4B). responding mouse mutants (Qi et al. 2001; Sugimori et al.
Another transcription factor with essential functions dur- 2008) (see Fig. 43.1). These factors combine with Sox10
ing this transition period is YY1 (He et al. 2007). Shortly and the Olig proteins to establish the gene regulatory net-
after cell cycle exit, it starts to repress factors such as Hes5 and work that initiates terminal differentiation and myelination
Id4 that maintain the progenitor state and counteract differ- (Fig. 43.7). Many of the transcription factors that are part of
entiation. Also, YY1 recruits HDAC-containing repressor this network influence each other’s expression. For instance,
complexes to the corresponding genes, which lead to tran- Nkx2.2 is downstream of Olig2, Sox10 (Liu et al. 2007), and
scriptional inhibition. In addition to its function as a repressor Mash1 (Sugimori et al. 2008). In return, Nkx2.2 positively
of myelination inhibitors, YY1 may also function as a direct influences both Olig2 and Sox10 expression (Liu et al. 2007).
activator of the myelination program as indicated by its ability At least in case of Sox10, this effect may be direct (Küspert
to stimulate PLP expression (see Fig.43.6). et al. 2011).

NEP cell OPC pOL mOL

Sox10
Nkx2.2

Olig2 Maxh1 myelin


genes
Zfp488

MRF

Figure 43.7 Summary of the Regulatory Network in Myelinating Oligodendrocytes with Focus on the Genetic Interactions Among Participating
Transcription Factors and Their Effect on Myelin Gene Expression. Proposed interactions are marked by stippled lines.

550 • NEUROGLIA
2.4.4 Terminal Differentiation and Myelination The role of Nkx2.2 is currently unclear, but likely different
in Oligodendrocytes from its function in the initiation phase of myelination.
Both Nkx2.2 and Mash1 are transiently expressed and dis-
appear during later phases of the myelination process (see 3 S U M M A RY A N D P E R S P E C T I VE S
Fig. 43.1). Their prodifferentiation function must therefore
be restricted to the initial phase of myelination. This is differ- Recent years have seen the identification and characteriza-
ent for the myogenic regulatory factor (MRF) transcription tion of many transcription factors with regulatory functions
factor (see Fig. 43.1), which appears in prooligodendrocytes in myelinating glia. It has become increasingly clear that com-
shortly before the onset of myelin gene expression, but con- mon principles govern the development of Schwann cells and
tinues to be expressed from then on (Emery et al. 2009). oligodendrocytes. However, the emerging picture also suggests
Prooligodendrocytes undergo apoptosis in the absence of that similar concepts have been implemented differently in the
MRF, thus arguing that MRF is required for survival at two cell types with few of the major players being conserved
this stage. Expression profiling furthermore showed that at their position between the two gene regulatory networks.
there is no myelin gene expression in MRF-deficient oli- This includes Oct6 and Krox20 as much as MRF and the Olig
godendrocytes. Its function may therefore be analogous proteins. This may be attributed to the fact that Schwann cells
to the role of Krox20 in myelinating Schwann cells (see and oligodendrocytes are of different developmental origins
Figs. 43.6 and 43.7). Similar to Krox20 in the PNS, MRF and likely acquired their capacity to myelinate independently.
seems to activate CNS myelin gene expression in synergy with Myelination of the CNS and PNS clearly represent convergent
Sox10 (Emery et al. 2009). Considering that Sox10 regulates processes. On this background, the central comparable role of
expression of many myelin genes in the CNS (Bondurand et al. Sox10 in both gene regulatory networks is striking and identi-
2001; Schlierf et al. 2006; Stolt et al. 2002), that Olig proteins fies Sox10 as a key factor and cornerstone in myelination.
do the same, and that Sox10 not only cooperates with MRF, The concept of gene regulatory networks has opened up
but also with Olig proteins (Li et al. 2007; Wissmüller et al. new perspectives on the processes of transcriptional regula-
2006), MRF, Sox10, and Olig proteins seem to jointly acti- tion in myelinating glia. Interactions between participating
vate and maintain the myelination program (see chapter 13). transcription factors allow for cross-regulation, cooperativ-
It also deserves mentioning that Olig1-deficient mice exhibit ity, and antagonism and explain how specificity is generated
severe myelination defects throughout the CNS. Specification and how transcription factors can acquire new functions or
defects in the same mice were minimal. This argues that the loose previous ones depending on the context of the network.
function of Olig1—different from Olig2—is much more It is probably fair to assume that most of the key transcrip-
prominent during terminal differentiation and myelination tional regulators in myelinating glia have been identified by
than during oligodendroglial specification (Arnett et al. 2004; now. Thus, it will be the next challenge to understand how
Lu et al. 2002; Xin et al. 2005; Zhou and Anderson 2002). they interact and influence each other, and how this will be
In their activation of the myelination program MRF, Sox10 modulated by posttranslational modifications. Several critical
and Olig proteins receive support from additional transcription circuits between key factors have already been identified in the
factors. An example is the zinc finger protein Zfp488 (Wang gene regulatory networks of Schwann cells and oligodendro-
et al. 2006). In the CNS, Zfp488 is selectively induced in the oli- cytes. Nevertheless, it is still a long way to a complete picture.
godendrocyte lineage at the same time as the major myelin genes The more pieces are added to the puzzle, the better will be
(see Fig. 43.1). Its expression depends on the Olig proteins with the understanding of the myelination process and of myelin
which it then interacts to maintain high levels of myelin gene diseases, including those that are caused by mutations in glial
expression in differentiating oligodendrocytes (see Figs. 43.6 transcription factors such as Krox20 and Sox10 (Inoue et al.
and 43.7). A similar effect on myelin gene expression and the 2004; Warner et al. 1998) (see also chapter 62). It will also be
myelination program has also been observed for the SCAN zinc easier to selectively and systematically manipulate the system
finger transcription factor Zfp191 (Howng et al. 2010). In con- for treatment of myelin diseases and traumatic injuries.
trast to Zfp488, it is also present in other CNS cell types and ear- Finally there is no doubt that for a full understanding of
lier stages of oligodendrocyte development so that its functions these gene regulatory networks it will be necessary to integrate
are likely not restricted to differentiating oligodendrocytes. the action of small RNAs, in particular microRNAs, and of
As mentioned for Schwann cells, gene expression in oli- chromatin modifiying and remodeling machineries. Although
godendrocytes has to be adjusted once cells transit from the the few existing studies prove the relevance of these factors
active state of myelination into the steady state of myelin main- in myelinating glia, the current picture is at best patchy with
tenance. This again requires changes of the gene regulatory many areas waiting to be filled in.
network. In mature oligodendrocytes the majority of Olig1 is,
for instance, removed from the nucleus (Arnett et al. 2004).
Additionally, Nkx2.2 and Nkx6.2 become reexpressed (Cai AC K N OW L E D G M E N T S
et al. 2010). Analysis of Nkx6.2-deficient mice reveals subtle
myelin abnormalities and altered expression of paranodal pro- Work in the author’s laboratory on the subject is sup-
teins, arguing that regulation of paranodal axon–glia interac- ported by grants from the Fonds der Chemischen Industrie,
tions may be part of its function (Southwood et al. 2004). ForNeuroCell and the Deutsche Forschungsgemeinschaft.

T R A N S C R I P T I O N FAC TO R S I N M Y E L I N AT I N G C E L L S • 551
REFERENCES Genoud S, Lappe-Siefke C, Goebbels S, Radtke F, Aguet M, Scherer SS,
et al. 2002. Notch1 control of oligodendrocyte differentiation in the
Arnett HA, Fancy SP, Alberta JA, Zhao C, Plant SR, Kaing S, et al. spinal cord. J Cell Biol 158:709–718.
2004. bHLH transcription factor Olig1 is required to repair demy- Ghislain J, Charnay P. 2006. Control of myelination in Schwann cells:
elinated lesions in the CNS. Science 306:2111–2115. a Krox20 cis-regulatory element integrates Oct6, Brn2 and Sox10
Bermingham JR, Scherer SS, O’Connell S, Arroyo E, Kalla KA, Powell activities. Embo Rep 7:52–58.
FL, et al. 1996. Tst-1/Oct-6/SCIP regulates a unique step in periph- Gokey NG, Srinivasan R, Lopez-Anido C, Krueger C, Svaren J. 2012.
eral myelination and is required for normal respiration. Genes Dev Developmental regulation of MicroRNA expression in Schwann
10:1751–1762. cells. Mol Cell Biol 32:558–568.
Bondurand N, Girard M, Pingault V, Lemort N, Dubourg O, Goossens He Y, Dupree J, Wang J, Sandoval J, Li J, Liu H, et al. 2007. The tran-
M. 2001. Human Connexin 32, a gap junction protein altered in scription factor Yin Yang 1 is essential for oligodendrocyte progeni-
the X-linked form of Charcot-Marie-Tooth disease, is directly tor differentiation. Neuron 55:217–230.
regulated by the transcription factor SOX10. Hum Mol Genet He Y, Kim JY, Dupree J, Tewari A, Melendez-Vasquez C, Svaren J, et al.
10:2783–2795. 2010. Yy1 as a molecular link between neuregulin and transcriptional
Bremer M, Fröb F, Kichko T, Reeh P, Tamm ER, Suter U, et al. 2011. modulation of peripheral myelination. Nat Neurosci 13:1472–1480.
Sox10 is required for Schwann cell homeostasis and myelin mainte- Howng SY, Avila RL, Emery B, Traka M, Lin W, Watkins T, et al. 2010.
nance in the adult peripheral nerve. Glia 59:1022–1032. ZFP191 is required by oligodendrocytes for CNS myelination. Genes
Britsch S, Goerich DE, Riethmacher D, Peirano RI, Rossner M, Nave Dev 24:301–311.
KA, et al. 2001. The transcription factor Sox10 is a key regulator of Inoue K, Khajavi M, Ohyama T, Hirabayashi S-i, Wilson J, Reggin JD,
peripheral glial development. Genes Dev 15:66–78. et al. 2004. Molecular mechanism for distinct neurological phenotypes
Cai J, Qi Y, Hu X, Tan M, Liu Z, Zhang J, et al. 2005. Generation conveyed by allelic truncating mutations. Nat Genet 36:361–369.
of oligodendrocyte precursor cells from mouse dorsal spinal cord Jacob C, Christen CN, Pereira JA, Somandin C, Baggiolini A, Lotscher P,
independent of Nkx6 regulation and Shh signaling. Neuron et al. 2011. HDAC1 and HDAC2 control the transcriptional pro-
45:41–53. gram of myelination and the survival of Schwann cells. Nat Neurosci
14:429–436.
Cai J, Zhu Q, Zheng K, Li H, Qi Y, Cao Q, et al. 2010. Co-localization of
Jaegle M, Ghazvini M, Mandemakers W, Piirsoo M, Driegen S,
Nkx6.2 and Nkx2.2 homeodomain proteins in differentiated myeli-
Levavasseur F, et al. 2003. The POU proteins Brn-2 and Oct-6 share
nating oligodendrocytes. Glia 58:458–468.
important functions in Schwann cell development. Genes Dev
Chen Y, Wang H, Yoon SO, Xu X, Hottiger MO, Svaren J, et al. 2011.
17:1380–1391.
HDAC-mediated deacetylation of NF-kappaB is critical for Schwann
Jaegle M, Mandemakers W, Broos L, Zwart R, Karis A, Visser P, et al.
cell myelination. Nat Neurosci 14:437–441.
1996. The POU factor Oct-6 and Schwann cell differentiation.
Chew LJ, Shen W, Ming X, Senatorov VV Jr., Chen HL, Cheng Y, et al.
Science 273:507–510.
2011. SRY-box containing gene 17 regulates the Wnt/beta-catenin
Jagalur NB, Ghazvini M, Mandemakers W, Driegen S, Maas A, Jones
signaling pathway in oligodendrocyte progenitor cells. J Neurosci
EA, et al. 2011. Functional dissection of the Oct6 Schwann cell
31:13921–13935.
enhancer reveals an essential role for dimeric Sox10 binding.
Cunliffe VT, Casaccia-Bonnefi l P. 2006. Histone deacetylase 1 is essen-
J Neurosci 31:8585–8594.
tial for oligodendrocyte specification in the zebrafish CNS. Mech
Jones EA, Brewer MH, Srinivasan R, Krueger C, Sun G, Charney KN,
Dev 123:24–30.
et al. 2012. Distal enhancers upstream of the Charcot-Marie-Tooth
Denarier E, Forghani R, Farhadi HF, Dib S, Dionne N, Friedman HC, type 1A disease gene PMP22. Hum Mol Genet 21:1581–1591.
et al. 2005. Functional organization of a Schwann cell enhancer. J Jones EA, Jang SW, Mager GM, Chang LW, Srinivasan R, Gokey NG,
Neurosci 25:11210–11217. et al. 2007. Interactions of Sox10 and Egr2 in myelin gene regulation.
Deneen B, Ho R, Lukaszewicz A, Hochstim CJ, Gronostajski RM, Neuron Glia Biol 3:377–387.
Anderson DJ. 2006. The transcription factor NFIA controls the onset Jones EA, Lopez-Anido C, Srinivasan R, Krueger C, Chang LW,
of gliogenesis in the developing spinal cord. Neuron 52:953–968. Nagarajan R, et al. 2011. Regulation of the PMP22 Gene through an
Dugas JC, Cuellar TL, Scholze A, Ason B, Ibrahim A, Emery B, et al. Intronic Enhancer. J Neurosci 31:4242–4250.
2010. Dicer1 and miR-219 Are required for normal oligodendrocyte Kao SC, Wu H, Xie J, Chang CP, Ranish JA, Graef IA, et al. 2009.
differentiation and myelination. Neuron 65:597–611. Calcineurin/NFAT signaling is required for neuregulin-regulated
Dutton KA, Pauliny A, Lopes SS, Elworthy S, Carney TJ, Rauch J, et al. 2001. Schwann cell differentiation. Science 323:651–654.
Zebrafish colourless encodes sox10 and specifies non-ectomesenchymal Kondo T, Raff MC. 2000. The Id4 HLH protein and the timing of oligo-
neural crest fates. Development 128: 4113–4125. dendrocyte differentiation. EMBO J 19:1998–2007.
Emery B, Agalliu D, Cahoy JD, Watkins TA, Dugas JC, Mulinyawe SB, Kuhlbrodt K, Herbarth B, Sock E, Hermans-Borgmeyer I, Wegner
et al. 2009. Myelin gene regulatory factor is a critical transcriptional M. 1998. Sox10, a novel transcriptional modulator in glial cells.
regulator required for CNS myelination. Cell 138:172–185. J Neurosci 18:237–250.
Fancy SP, Baranzini SE, Zhao C, Yuk DI, Irvine KA, Kaing S, et al. 2009. Küspert M, Hammer A, Bosl MR, Wegner M. 2011. Olig2 regulates
Dysregulation of the Wnt pathway inhibits timely myelination and Sox10 expression in oligodendrocyte precursors through an evolution-
remyelination in the mammalian CNS. Genes Dev 23:1571–1585. ary conserved distal enhancer. Nucleic Acids Res 39:1280–1293.
Finzsch M, Schreiner S, Kichko T, Reeh P, Tamm ER, Bosl MR, et al. Lau P, Verrier JD, Nielsen JA, Johnson KR, Notterpek L, Hudson LD.
2010. Sox10 is required for Schwann cell identity and progression 2008. Identification of dynamically regulated microRNA and
beyond the immature Schwann cell stage. J Cell Biol 189:701–712. mRNA networks in developing oligodendrocytes. J Neurosci
Finzsch M, Stolt CC, Lommes P, Wegner M. 2008. Sox9 and Sox10 28:11720–11730.
influence survival and migration of oligodendrocyte precursors in Le N, Nagarajan R, Wang JY, Araki T, Schmidt RE, Milbrandt J. 2005a.
the spinal cord by regulating PDGF receptor {alpha} expression. Analysis of congenital hypomyelinating Egr2Lo/Lo nerves identifies
Development 135:637–646. Sox2 as an inhibitor of Schwann cell differentiation and myelination.
Fogarty M, Richardson WD, Kessaris N. 2005. A subset of oligodendro- Proc Natl Acad Sci U S A 102:2596–2601.
cytes generated from radial glia in the dorsal spinal cord. Development Le N, Nagarajan R, Wang JY, Svaren J, LaPash C, Araki T, et al. 2005b.
132:1951–1959. Nab proteins are essential for peripheral nervous system myelination.
Fröb F, Bremer M, Finzsch M, Kichko T, Reeh P, Tamm ER, et al. 2012. Nat Neurosci 8:932–940.
Establishment of myelinating Schwann cells and barrier integrity LeBlanc SE, Jang SW, Ward RM, Wrabetz L, Svaren J. 2006. Direct reg-
between central and peripheral nervous systems depend on Sox10. Glia ulation of myelin protein zero expression by the Egr2 transactivator.
60:806–819. J Biol Chem 281:5453–5460.

552 • NEUROGLIA
LeBlanc SE, Ward RM, Svaren J. 2007. Neuropathy-associated Egr2 Rowitch DH. 2004. Glial specification in the vertebrate neural tube. Nat
mutants disrupt cooperative activation of myelin protein zero by Rev Neurosci 5:409–419.
Egr2 and Sox10. Mol Cell Biol 27:3521–3529. Ryu EJ, Wang JY, Le N, Baloh RH, Gustin JA, Schmidt RE, et al. 2007.
Li H, de Faria JP, Andrew P, Nitarska J, Richardson WD. 2011. Misexpression of Pou3f1 results in peripheral nerve hypomyelination
Phosphorylation regulates OLIG2 cofactor choice and the motor and axonal loss. J Neurosci 27:11552–11559.
neuron-oligodendrocyte fate switch. Neuron 69:918–929. Samanta J, Kessler JA. 2004. Interactions between ID and OLIG pro-
Li H, He Y, Richardson WD, Casaccia P. 2009. Two-tier transcriptional teins mediate the inhibitory effects of BMP4 on oligodendroglial dif-
control of oligodendrocyte differentiation. Curr Opin Neurobiol ferentiation. Development 131:4131–4142.
19:479–485. Schlierf B, Werner T, Glaser G, Wegner M. 2006. Expression of
Li H, Lu Y, Smith HK, Richardson WD. 2007. Olig1 and Sox10 interact Connexin47 in oligodendrocytes is regulated by the Sox10 transcrip-
synergistically to drive myelin basic protein transcription in oligo- tion factor. J Mol Biol 361:11–21.
dendrocytes. J Neurosci 27:14375–14382. Schreiner S, Cossais F, Fischer K, Scholz S, Bösl MR, Holtmann B, et al.
Limpert AS, Carter BD. 2010. Axonal neuregulin 1 type III activates 2007. Hypomorphic Sox10 alleles reveal novel protein functions and
NF-kappaB in Schwann cells during myelin formation. J Biol Chem unravel developmental differences in glial lineages. Development
285:16614–16622. 134:3271–3281.
Liu A, Li J, Marin-Husstege M, Kageyama R, Fan Y, Gelinas C, et al. 2006. Shen S, Li J, Casaccia-Bonnefi l P. 2005. Histone modifications affect
A molecular insight of Hes5-dependent inhibition of myelin gene timing of oligodendrocyte progenitor differentiation in the develop-
expression: old partners and new players. EMBO J 25:4833–4842. ing rat brain. J Cell Biol 169:577–589.
Liu Z, Hu X, Cai J, Liu B, Peng X, Wegner M, et al. 2007. Induction Shimizu T, Kagawa T, Wada T, Muroyama Y, Takada S, Ikenaka K.
of oligodendrocyte differentiation by Olig2 and Sox10: evidence for 2005. Wnt signaling controls the timing of oligodendrocyte develop-
reciprocal interactions and dosage-dependent mechanisms. Dev Biol ment in the spinal cord. Dev Biol 282:397–410.
302:683–693. Sohn J, Natale J, Chew LJ, Belachew S, Cheng Y, Aguirre A, et al. 2006.
Lu QR, Sun T, Zhu Z, Ma N, Garcia M, Stiles CDet al. 2002. Common Identification of Sox17 as a transcription factor that regulates oligo-
developmental requirement for olig function indicates a motor neu- dendrocyte development. J Neurosci 26:9722–9735.
ron/oligodendrocyte connection. Cell 109:75–86. Southwood C, He C, Garbern J, Kamholz J, Arroyo E, Gow A. 2004.
Lu QR, Yuk D, Alberta JA, Zhu Z, Pawlitzky I, Chan J, et al. 2000. CNS myelin paranodes require Nkx6–2 homeoprotein transcrip-
Sonic hedgehog-regulated oligodendrocyte lineage genes encoding tional activity for normal structure. J Neurosci 24:11215–11225.
bHLH proteins in the mammalian central nervous system. Neuron Stolt CC, Lommes P, Sock E, Chaboissier M-C, Schedl A, Wegner M.
25:317–329. 2003. The Sox9 transcription factor determines glial fate choice in
Marin-Husstege M, He Y, Li J, Kondo T, Sablitzky F, Casaccia-Bonnefi l P. the developing spinal cord. Genes Dev 17:1677–1689.
2006. Multiple roles of Id4 in developmental myelination: predicted Stolt CC, Rehberg S, Ader M, Lommes P, Riethmacher D, Schachner M,
outcomes and unexpected findings. Glia 54:285–296. et al. 2002. Terminal differentiation of myelin-forming oligoden-
Mizuguchi R, Sugimori M, Takebayashi H, Kosako H, Nagao M, drocytes depends on the transcription factor Sox10. Genes Dev
Yoshida S, et al. 2001. Combinatorial roles of olig2 and neurogenin2 16:165–170.
in the coordinated induction of panneuronal and subtype-specific Stolt CC, Schlierf A, Lommes P, Hillgärtner S, Werner T, Kosian T, et al.
properties of motoneurons. Neuron 31:757–771. 2006. SoxD proteins influence multiple stages of oligodendrocyte devel-
Monk KR, Naylor SG, Glenn TD, Mercurio S, Perlin JR, Dominguez C, opment and modulate SoxE protein function. Dev Cell 11:697–710.
et al. 2009. A G protein-coupled receptor is essential for Schwann Stolt CC, Schmitt S, Lommes P, Sock E, Wegner M. 2005. Impact of
cells to initiate myelination. Science 325:1402–1405. transcription factor Sox8 on oligodendrocyte specification in the
Monk KR, Oshima K, Jors S, Heller S, Talbot WS. 2011. Gpr126 is mouse embryonic spinal cord. Dev Biol 281:323–331.
essential for peripheral nerve development and myelination in mam- Sugimori M, Nagao M, Parras CM, Nakatani H, Lebel M, Guillemot F,
mals. Development 138:2673–2680. et al. 2008. Ascl1 is required for oligodendrocyte development in the
Monuki ES, Weinmaster G, Kuhn R, Lemke G. 1989. SCIP: a glial POU spinal cord. Development 135:1271–1281.
domain gene regulated by cyclic AMP. Neuron 3:783–793. Sun T, Echelard Y, Lu R, Yuk DI, Kaing S, Stiles CD, et al. 2001. Olig
Nagarajan R, Svaren J, Le N, Araki T, Watson M, Milbrandt J. 2001. bHLH proteins interact with homeodomain proteins to regulate cell
EGR2 mutations in inherited neuropathies dominant-negatively fate acquisition in progenitors of the ventral neural tube. Curr Biol
inhibit myelin gene expression. Neuron 30:355–368. 11:1413–1420.
Nieto M, Schuurmans C, Britz O, Guillemot F. 2001. Neural bHLH Sun Y, Meijer DH, Alberta JA, Mehta S, Kane MF, Tien AC, et al. 2011.
genes control the neuronal versus glial fate decision in cortical pro- Phosphorylation state of Olig2 regulates proliferation of neural pro-
genitors. Neuron 29:401–413. genitors. Neuron 69:906–917.
Park HC, Appel B. 2003. Delta-notch signaling regulates oligodendro- Svaren J, Meijer D. 2008. The molecular machinery of myelin gene tran-
cyte specification. Development 130:3747–3755. scription in Schwann cells. Glia 56(14):1541–1551.
Parkinson DB, Bhaskaran A, Arthur-Farraj P, Noon LA, Woodhoo A, Topilko P, Schneider MS, Levi G, Baron VEA, Chennoufi AB,
Lloyd AC, et al. 2008. c-Jun is a negative regulator of myelination. Seitanidou T, et al. 1994. Krox-20 controls myelination in the periph-
J Cell Biol 181:625–637. eral nervous system. Nature 371(6500):796–799.
Peirano RI, Goerich DE, Riethmacher D, Wegner M. 2000. Protein zero Vallstedt A, Klos JM, Ericson J. 2005. Multiple dorsoventral origins of
expression is regulated by the glial transcription factor Sox10. Mol oligodendrocyte generation in the spinal cord and hindbrain. Neuron
Cell Biol 20:3198–3209. 45:55–67.
Pereira JA, Baumann R, Norrmen C, Somandin C, Miehe M, Jacob C, Verheijen MH, Chrast R, Burrola P, Lemke G. 2003. Local regulation of
et al. 2010. Dicer in Schwann cells is required for myelination and fat metabolism in peripheral nerves. Genes Dev 17:2450–2464.
axonal integrity. J Neurosci 30:6763–6775. Wahlbuhl M, Reiprich S, Vogl MR, Bosl MR, Wegner M. 2011.
Prasad MK, Reed X, Gorkin DU, Cronin JC, McAdow AR, Chain K, Transcription factor Sox10 orchestrates activity of a neural crest-specific
et al. 2011. SOX10 directly modulates ERBB3 transcription via an enhancer in the vicinity of its gene. Nucleic Acids Res 40:88–101.
intronic neural crest enhancer. BMC Dev Biol 11:40. Wang S, Sdrulla A, Johnson JE, Yokota Y, Barres BA. 2001. A role for the
Qi Y, Cai J, Wu Y, Wu R, Lee J, Fu H, et al. 2001. Control of oligoden- helix-loop-helix protein Id2 in the control of oligodendrocyte devel-
drocyte differentiation by the Nkx2.2 homeodomain transcription opment. Neuron 29:603–614.
factor. Development 128:2723–2733. Wang SZ, Dulin J, Wu H, Hurlock E, Lee SE, Jansson K, et al. 2006.
Reiprich S, Kriesch J, Schreiner S, Wegner M. 2010. Activation of Krox20 An oligodendrocyte-specific zinc-finger transcription regulator
gene expression by Sox10 in myelinating schwann cells. J Neurochem cooperates with Olig2 to promote oligodendrocyte differentiation.
112:744–754. Development 133:3389–3398.

T R A N S C R I P T I O N FAC TO R S I N M Y E L I N AT I N G C E L L S • 553
Warner LE, Mancias P, Butler IJ, McDonald CM, Keppen L, Koob KG, by disrupting the beta-catenin-TCF interaction. Nat Neurosci
et al. 1998. Mutations in the early growth response 2 (EGR2) gene are 12:829–838.
associated with hereditary myelinopathies. Nature Genet 18:382–384. Yoon C, Korade Z, Carter BD. 2008. Protein kinase A-induced phos-
Wegner M. 2010. All purpose Sox: The many roles of Sox proteins in phorylation of the p65 subunit of nuclear factor-kappaB promotes
gene expression. Int J Biochem Cell Biol 42:381–390. Schwann cell differentiation into a myelinating phenotype. J Neurosci
Werner T, Hammer A, Wahlbuhl M, Bösl MR, Wegner M. 2007. 28:3738–3746.
Multiple conserved regulatory elements with overlapping functions Yun B, Anderegg A, Menichella D, Wrabetz L, Feltri ML, Awatramani R.
determine Sox10 expression in mouse embryogenesis. Nucleic Acids 2010. MicroRNA-deficient Schwann cells display congenital hypo-
Res 35:6526–6538. myelination. J Neurosci 30:7722–7728.
Wissmüller S, Kosian T, Wolf M, Finzsch M, Wegner M. 2006. The Zhao X, He X, Han X, Yu Y, Ye F, Chen Y, et al. 2010. MicroRNA-
High-mobility-group domain of Sox proteins interacts with the mediated control of oligodendrocyte differentiation. Neuron 65:
DNA-binding domains of many transcription factors. Nucleic Acids 612–626.
Res 34:1735–1744. Zhou Q, Anderson DJ. 2002. The bHLH transcription factors olig2
Woodhoo A, Alonso MB, Droggiti A, Turmaine M, D’Antonio M, and olig1 couple neuronal and glial subtype specification. Cell
Parkinson DB, et al. 2009. Notch controls embryonic Schwann cell dif- 109:61–73.
ferentiation, postnatal myelination and adult plasticity. Nat Neurosci Zhou Q, Wang S, Anderson DJ. 2000. Identification of a novel family of
12:839–847. oligodendrocyte lineage-specific basic helix-loop-helix transcription
Xin M, Yue T, Ma Z, Wu FF, Gow A, Lu QR. 2005. Myelinogenesis factors. Neuron 25:331–343.
and axonal recognition by oligodendrocytes in brain are uncoupled Zorick TS, Syroid DE, Brown A, Gridley T, Lemke G. 1999. Krox-20
in Olig1-null mice. J Neurosci 25:1354–1365. controls SCIP expression, cell cycle exit and susceptibility to apop-
Ye F, Chen Y, Hoang T, Montgomery RL, Zhao XH, Bu H, et al. 2009. tosis in developing myelinating Schwann cells. Development
HDAC1 and HDAC2 regulate oligodendrocyte differentiation 126:1397–1406.

554 • NEUROGLIA
44.
FACTOR S CONTROLLING MYELIN FORMATION
Ruth Stassart, Sandra Goebbels, and Klaus-Armin Nave

A B B R E VI AT I O N S MAPK mitogen-activated protein kinase


MBP myelin basic protein
ADAM A disintegrin and metalloprotease MTMR2 myotubularin-related protein 2
AKT V-Akt murine thymoma viral oncogene mTOR mammalian target of rapamycin
homolog 1 NCAM neural cell adhesion molecule
ATP adenosine triphosphate NECL nectin-like protein
BACE1 E-site amyloid precursor protein cleaving NFAT nuclear factor of activated T cells
enzyme 1 NGF nerve growth factor
BDNF brain-derived neurotrophic factor NgR1 nogo receptor 1
cAMP cyclic adenosine monophosphate NICD notch intracellular domain
Cdc42 cell division control protein 42 homolog NMDA N-methyl-d-aspartate
CnB1 calcineurin NRD1 N-arginine dibasic convertase/Nardilysin
CNS central nervous system NRG1 neuregulin-1
CNTF ciliary neurotrophic factor NT3/NT4/5 neurotrophin-3/4/5
CRB crumbs complex N-WASP neuronal Wiskott–Aldrich syndrome
CREB cAMP response element-binding protein
Dcc deleted in colorectal carcinoma Oct6 octamer transcription factor 6
Dlg1 disk-large homolog 1 Olig1/Olig2 oligodendrocyte transcription factor 1/2
DRG dorsal root ganglia OPC oligodendrocyte precursor cell
EAE experimental autoimmune encephalomyelitis p75NTR P75 neurotrophin receptor
EGF-like domain epidermal growth factor–like domain Pals1 protein associated with lin seven 1
EGR-2 early growth response protein 2 Par complex partitioning-defective protein polarity
ErbB erythroblastic leukemia viral oncogene complex
homolog PDGF platelet-derived growth factor
ERK extracellular signal-regulated kinase PH domain pleckstrin homology domain
FGF fibroblast growth factor PI3K phosphatidylinositol 3-kinase
FGFR FGF receptor tyrosine kinase PIP2 phosphatidylinositol (4,5)-bishosphate
GDNF glial cell line derived neurotrophic factor PIP3 phosphatidylinositol (3,4,5)-trisphosphate
GH growth hormone PLC phospholipase C
GPCR/GPR G protein–coupled receptor PLP proteolipid protein
GPI glycosylphosphatidylinositol Pmp22 peripheral myelin protein of 22kDa
HDAC histone deacetylase PNS peripheral nervous system
hnRNP heterogeneous nuclear ribonucleoprotein PrPc prion protein cellular
ID2/ID4 inhibitor of DNA-binding protein PSA-NCAM polysialylated form of the neural cell
L1 cell adhesion molecule L1 adhesion molecule
Ig immunoglobulin PTEN phosphatase and tensin homolog
IGF-1 insulin-like growth factor 1 P2Y purinergic receptors, G protein–coupled
IGFR1 IGF receptor tyrosine kinase receptor
ILK integrin linked kinase Rac1 Ras-related C3 botulinum toxin substrate 1
Kif13b kinesin family member 13B Rho Ras homolog gene family
Lgi4 leucine-rich repeat LGI family, member 4 SCRIB scribble complex
LIF leukemia inhibitory factor SHP2/PTPN11 protein tyrosine phosphatase, non-receptor
Lingo1 leucine rich repeat and Ig domain type 11
containing NOGOreceptor interacting SREBP sterol responsive element binding proteins
protein TACE tumor necrosis factor-D–converting
MAG myelin-associated glycoprotein enzyme (ADAM17)

555
TCF T-cell factor/lymphoid enhancer factor or 2009), and hormones (Calzà et al. 2010; Schumacher et al.
TCF/LEF 2012).
TGF-E transforming growth factor beta
TGN trans Golgi network
TrkA/B/C tropomyosin-related kinase receptors A/B/C 2 F O R M AT I O N O F T H E
AXO N –M Y E L I N U N I T

1 INTRODUCTION Myelination in the peripheral nervous system (PNS) is in


many ways simpler than in the central nervous system (CNS),
The last steps in the cellular evolution of the nervous system although there is no evidence that it originated here in ner-
included the invention of myelin, some 300 million years vous system evolution. As detailed in chapter 14, Schwann
ago in ancestors of lower vertebrates. This was a prerequi- cells that are derived from neural crest are associated with
site for the subsequent evolution of complex yet compact, axons throughout PNS development. However, before myeli-
quickly operating, and thus powerful nervous systems. The nation, myelinating Schwann cells sort out a single axon into
importance of myelinated fibers becomes obvious in the con- a stable one-to-one relationship. Enwrapping this axon with
text of severe neurological diseases, in which the formation a myelin membrane establishes the axon–myelin unit that
or maintenance of myelin is perturbed (see chapters 54, 61, then grows in size, becoming up to 1 mm or more in inter-
62, and 71). nodal length (Hildebrand et al. 1994). In contrast, myelinat-
This chapter gives a brief overview of the factors that ing oligodendrocytes of the CNS develop from multipolar
have been identified as regulators of myelin formation, oligodendrocyte progenitor cells (OPC). These precursors
mostly non–cell autonomous protein ligands and their glial have no such tight association with axonal membranes before
receptors (Table 44.1), and intracellular second messengers. myelination. In contrast, oligodendrocytes interact with mul-
Other systemic factors that influence myelin biogenesis (and tiple axons that they individually ensheath. Internodal length
remyelination) cannot be adequately covered here, and the in the CNS is significantly shorter but measures up to sev-
reader is referred to articles on the role of nutrition and vita- eral hundred micrometers (Remahl and Hildebrand 1990).
mins (Steinfeld et al. 2009), trace elements (Todorich et al. A combination of imaging and recording techniques led to

Table 44.1 NON–CELL AUTONOMOUS FACTORS CONTROLLING MYELINATION


SIGNAL, LIGAND,
AND/OR REFERENCES AND FURTHER
RECEPTOR PNS CNS READING

Direct Axonal electrical Inhibits Schwann cell Promotes differentiation and myeli- Barres et al. 1993
axon–glia activity proliferation/differentia- nation in vitro Demerens et al. 1996
signaling tion in vitro Stevens et al. 2002
Wake et al. 2011
Fields 2008 (Rev)

NRG1 typeIII Promotes Schwann cell Dispensable for Michailov et al. 2004
ErbB2/3/4 survival, differentiation, myelination in vivo Taveggia et al. 2005,
myelin growth control Brinkmann et al. 2008
Taveggia et al. 2010 (Rev)

Nectin-like Required for Schwann Contribute to the initiation of Maurel et al. 2007
proteins cell myelination in vitro myelination Spiegel et al. 2007
Park et al. 2008
Pereira et al. 2012 (Rev)

Prion protein Required for myelin Dispensable in vivo Bremer et al. 2010
(axonal) maintenance Pereira et al. 2012 (Rev)

Lingo1 Inhibits oligodendrocyte differentia- Mi et al. 2004, 2005


tion and myelination Lee et al. 2007
Kremer et al. 2011 (Rev)

Jagged Inhibits myelin Inhibits oligodendrocyte differentia- Givogri et al. 2002


Notch1 formation tion and myelination Woodhoo et al. 2009
Taveggia et al. 2010 (Rev)
Piaton G et al. 2010 (Rev)
PSA-NCAM Inhibits myelination Charles et al. 2000
in vitro Fewou et al. 2007
Piaton G et al. 2010 (Rev)

(Continued)

556 • NEUROGLIA
Table 44.1 (Cont’d)
SIGNAL, LIGAND,
AND/OR REFERENCES AND FURTHER
RECEPTOR PNS CNS READING

Orphan receptors GPR126 Required for Schwann Dispensable for Monk et al. 2009, 2011
cell differentiation and myelination in vivo Pereira et al. 2012 (Rev)
myelination Taveggia et al. 2010 (Rev)

GPR17 Not expressed Inhibits oligodendrocyte Chen et al. 2009


differentiation Taveggia et al. 2010 (Rev)
Neurotrophins NGF Promotes myelination Inhibits myelin formation Chan et al. 2004
in vitro in vitro Lee et al. 2007
Rosenberg et al. 2006 (Rev)
Xiao et al. 2009 (Rev)
BDNF Promotes myelination via Promotes myelination, Xiao et al. 2010
p75NTR, inhibits it via but controversial reports Vondran et al. 2010
TrkB receptors Rauskolb et al. 2010
Taveggia et al. 2010 (Rev)
NT-3 Inhibits myelination Cosgaya et al. 2002
in vitro; controversial Xiao et al. 2009 (Rev)
in vivo data
Other IGF-1 Promotes myelination Important for oligodendrocyte Carson et al. 1993
extracellular differentiation/myelination in Ye et al. 1995, 2002
factors development Russel et al. 2000
Zeger et al. 2007
Taveggia et al. 2010 (Rev)
FGF Promotes myelin formation in Bansal et al. 2002
vivo; FGF2 inhibits differentiation Fortin et al. 2005
in vitro Mason et al. 2007
Furusho et al. 2012
Wnt Promotes myelination Inhibits oligodendrocyte Fancy et al. 2009
differentiation Feigenson et al. 2009
Promotes myelination Ye et al. 2009
Tawk et al. 2011
Rosenberg and Chan 2009 (Rev)
GDNF Positive role for myelina- Höke et al. 2003
tion suggested Xiao et al. 2009
Extracellular Laminin Required for Schwann Important for oligodendrocyte Previtali et al. 2003
matrix Dystroglycan cell differentiation, basal differentiation Benninger et al. 2006
Integrin lamina production, Barros et al. 2009
myelination Camara et al. 2009
Colognato and Tzvetanova 2011
(Rev)
Hormones Progesterone Promotes myelination Promotes oligodendrocyte Schumacher et al. 2012 (Rev)
differentiation
Thyroid hormone Promotes oligodendrocyte Calza et al. 2010 (Rev)
differentiation/myelination Taveggia et al. 2010 (Rev)

Compilation of signaling molecules that either promote or inhibit myelination in the peripheral nervous system and central nervous system, or are dispensable based on
genetic experiments.
For details see references (right, reviews indicated as ‘rev’) and text.

the discovery that in the developing corpus callosum many Glial cell processes and their membranes that interact with
OPC receive a transient synaptic input from unmyelinated myelination-competent axons in trans become polarized, which
axons before myelination (Ziskin et al. 2007). Whether these is required for the initial ensheathment and determines the site of
glutamatergic synapses induce (or alternatively prevent) membrane growth (Bauer and ffrench-Constant 2009; Simons
subsequent maturation of OPC and myelin assembly is not and Trotter 2007). The assembly of large quantities of myelin
known. components occurs in the secretory path and is regulated within

FAC TO R S C O N T R O L L I N G M Y E L I N F O R M AT I O N • 557
distinct subcellular compartments. It involves both high-level can differentiate into myelinating cells in culture without
lipid biosynthesis and translation and proper trafficking of direct contact with neurons or axons (Dubois-Dalcq et al.
myelin proteins. The generation of myelin membranes further 1986; Mirsky et al. 1980). Indeed, oligodendrocytes are capa-
requires a tight coordination of gene expression and membrane ble of ensheathing paraformaldehyde-fixed axons or synthetic
growth control (Emery 2010a; Nave 2010) to maintain the fibers in vitro, indicating that only limited bidirectional sig-
unique stoichiometry of myelin proteins and lipids (<30% pro- naling is required (Lee et al. 2012; Rosenberg et al. 2008). On
teins and >70% lipids, with about 50% cholesterol). Even when the other hand, oligodendrocytes only target axonal processes,
developmental myelination has come to a halt, maintaining the never dendrites (Lubetzki et al. 1993), and synthesize myelin
fine balance of continued exocytosis and endocytosis of myelin appropriate to axon calibre (although g-ratios in the CNS
membranes seems essential. Minor deviations of the steady state vary considerably more than in the PNS). This suggests that
can theoretically lead to significant demyelination over time. oligodendrocytes also myelinate in response to axonal signals.
However, these signals are not necessarily “instructive,” and an
alternative hypothesis holds that oligodendrocyte differentia-
3 AXO N – G L I A I N T E R AC T I O N I N tion and myelin membrane synthesis is ultimately by default,
THE PERIPHERAL AND CENTRAL determined by an intrinsic transcriptional network (see chap-
N E RVO U S SYS T E M ter 43) and guided by inhibitory extracellular factors. Indeed,
potent inhibitory axonally expressed signals, including ligands
Historically, the demonstration that glial cell membranes were such as Lingo-1, Jagged, and PSA-NCAM, have been impli-
the source of myelin (Bunge 1968; Geren and Raskind 1953) cated in either inhibiting OPC differentiation or preventing
was a starting point for the investigation of interactions between myelination by oligodendrocytes (Kremer et al. 2011; Piaton
axons and their ensheathing glial cells. Studies involving the et al. 2010).
addition of purified neurite membrane fractions to cultured Growth factors and cytokines (e.g., PDGF, FGF-2, IGF-1,
Schwann cells were the first to demonstrate that axonal sig- NT3, CNTF, BMP and LIF) have been studied in detail as
nals stimulate Schwann cells to survive and proliferate (Salzer potent regulators of OPC proliferation and oligodendrocyte
et al. 1980a,b; Wood and Bunge 1975). In turn, in the absence differentiation however, the source of these factors is not nec-
of axonal signals Schwann cells are unable to differentiate into essarily axonal (see chapters 6 and 13). The specific require-
myelinating cells (Mirsky et al. 1980). Finally, determining the ment of axons for oligodendrocyte survival was indicated by
axon caliber of cultured dorsal root ganglia (DRG) neurons lesion studies, in which optic nerve transections reduced the
revealed that the addition of Schwann cells triggers myelina- number of oligodendrocytes (Barres et al. 1993). By contrast,
tion, but also an increase of the axon diameter (Windebank optic nerves of Bcl2-transgenic mice, in which the number of
et al. 1985), establishing bidirectional axon–glial signaling. ganglion cell axons was increased, harbored correspondingly
In the PNS only axons greater than a diameter of about 1 more oligodendrocytes (Burne et al. 1996). In other experi-
μm are myelinated, and the axonal caliber maintains a rather ments, it was the axonal electrical activity that promoted
constant ratio to the myelin sheath thickness (Friede and OPC proliferation and supported myelination (Barres and
Bischhausen 1982), also quantified as the g-ratio (Fig. 44.1A). Raff 1993; Demerens et al. 1996), possibly as a response to the
Experimental support for the idea that axons regulate myeli- axonal release of adenosine (Stevens et al. 2002). However,
nation came from nerve graft experiments. Schwann cells unlike in the PNS, no specific axonally presented signaling
originating from unmyelinated fibers can differentiate into molecule has been identified to be essential for CNS myelina-
myelinating Schwann cells once transplanted into a myeli- tion, as is axonal Neuregulin-1 for the myelination of periph-
nated nerve with larger axons (Aguayo et al. 1976). Thus, eral nerves.
Schwann cells change their myelination fate depending on
axonal signals. Axon caliber itself poses a crucial variable as
demonstrated by Voyvodic (1989), who found that normally 4 N E U R E GU L I N 1/ E R B B R E C E P TO R
unmyelinated sympathetic fibers become (de novo) myeli- S I G N A L I N G I N P E R I P H E R A L N E RVO U S
nated by resident Remak cells following an increase of the SYS T E M M Y E L I N AT I O N
axon caliber (experimentally induced here by a nerve hemisec-
tion, which increased the size of the neurotrophic target tis- Neuregulin-1 (NRG1) comprises a family of growth factors,
sue, resulting in radial axonal growth) (Voyvodic 1989). encoded by one of the largest mammalian genes (~1.6 Mb).
This response of Schwann cells to the diameter of the axon Expressed in many cell types, more than 16 NRG1 isoforms are
is compatible with a model of an axon-resident myelination sig- generated by different promoter usage and alternative RNA
nal that is integrated by Schwann cells as a measure of axon size, splicing (Nave and Salzer 2006). By their different N-termini,
and that controls myelination quantitatively once a threshold neuregulins are currently subdivided into six subtypes (Nave
level has been reached. In the PNS, this signal is provided by and Salzer 2006), with NRG1 types I, II, and III most widely
the axonal growth factor Neuregulin 1 (Michailov et al. 2004; studied. All isoforms are synthesized as transmembrane pro-
Taveggia et al. 2005). Myelination in the CNS is less well teins and further processed by proteases; however, NRG1 type
understood, although the same axons are often myelinated by III (with a C-terminal cystein-rich domain) is anchored twice
Schwann cells and oligodendrocytes as they course from the in the membrane and therefore not shed from the membrane
PNS into the CNS. Unlike Schwann cells, oligodendrocytes by a single processing step (Fig. 44.1B).

558 • NEUROGLIA
A a NRG1 type III +/– wildtype NRG1 type III tg
g-ratio =
b

a Axonal NRG1
b

NRG1 type III +/- NRG1 +/+ NRG1 type III tg


(viable) (wildtype) (viable)

Figure 44.1 Neuregulin-1 Is an Axonal Growth Factor That Regulates Myelination by Schwann Cells. A. Myelin sheath thickness is roughly propor-
tional to the axon diameter, a relationship quantified as the “g-ratio” between the axonal caliber (a) and the total fiber diameter (b), as illustrated on
the electron micrograph of a myelinated axon (left). Schwann cells “measure” the axon caliber by integrating NRG1 type III signaling at the axonal
surface as a surrogate marker, so as to wrap the corresponding numbers of myelin membranes and reach the normal g-ratio of about 0.67 (in the
PNS). Neuregulin-1 type III–dependent control of myelination can be demonstrated genetically (Michailov et al. 2004; Taveggia et al. 2005) with
heterozygous Nrg1 mouse mutants, in which axons expressing 50% NRG1 are thinly myelinated (left) compared with wild-type (middle). In contrast,
transgenic mice overexpressing NRG1 type III in projection neurons exhibit hypermyelinated axons caused by elevated NRG1 density (right).
B. Activation of axonal NRG1 type III by proteolytic cleavage. The major NRG1 isoforms (depicted are types I, II, and III) differ at their N-terminus
as a result of alternative transcription and RNA splicing, but share the same EGF-like signaling domain necessary for ErbB receptor activation. NRG1
type III is anchored by a second membrane spanning region, the cysteine rich domain (CRD). Proteolytic processing of NRG1 by the protease
BACE1 activates this growth factor, but only NRG1 types I and II are also released as paracrine signaling molecules. BACE1 cleavage of type III cre-
ates a paracrine signal that promotes myelination (Hu et al. 2006; Velanac et al. 2012; Willem et al. 2006). In contrast, the protease TACE/ADAM17
cleaves NRG1 in the EGF-like domain, thereby inhibiting NRG1 signaling (La Marca et al. 2011). The conserved intracellular domain of NRG1 is
processed by gamma-secretases that thereby create a “retrograde” signaling molecule within neurons. The temporal order of these cleavage steps and
their intracellular localization are not known. (A) Adapted from ffrench-Constant et al. 2004. (B) Adapted from Nave and Salzer 2006.

All NRG1 isoforms share an epidermal growth factor heterodimerization, with autophosphorylation of tyrosine
(EGF)–like domain, which is required and sufficient to acti- residues for the recruitment of adaptor proteins and the acti-
vate erythroblastic leukemia viral oncogene homolog (ErbB) vation of various downstream kinases. For NRG1/ErbB sig-
receptor tyrosine kinases. Binding of axonal NRG1 to glial naling in Schwann cells, the PI3K/AKT pathway, the Ras/
ErbB proteins induces receptor homodimerization and MAPK/ERK pathway, and the NFAT/Calcineurin signaling

FAC TO R S C O N T R O L L I N G M Y E L I N F O R M AT I O N • 559
pathway are most important (Newbern and Birchmeier 2010). The role of NRG1 in the CNS is more complex and pos-
Because ErbB3 lacks a potent kinase domain and ErbB2 lacks sibly more relevant for neuron-to-neuron signaling. Although
receptor functions, NRG1 signaling to Schwann cells requires some experiments suggested that NRG1/ErbB signaling stim-
ErbB2/3 heterodimerization. In oligodendrocytes also ErbB4 ulates the proliferation of OPC and oligodendrocyte survival,
is abundantly expressed. and possibly myelination, the detailed analysis of mice lack-
Because NRG1 is involved in heart development, mouse ing NRG1 from cortical neurons, or both ErbB3 and ErbB4
mutants completely lacking NRG1 die embryonically. To receptors from oligodendrocytes, revealed normal oligoden-
genetically analyze this growth factor in nervous system devel- drocyte numbers and intact myelination of subcortical white
opment, NRG1 isoform-specific conditional mouse mutants matter tracts (Brinkmann et al. 2008). In contrast, trans-
and transgenic mice were instrumental (Birchmeier and genic overexpression of NRG1 type III (as well as NRG1
Nave 2008). In the PNS, axonal NRG1 type III emerged as type I) in cortical neurons induces some hypermyelination
a key regulator of virtually all steps of Schwann cell develop- (Brinkmann et al. 2008). Thus, oligodendrocytes are respon-
ment and myelination (Birchmeier and Nave 2008; Newbern sive to NRG1, but NRG1/ErbB signaling is not required for
and Birchmeier 2010) (see also chapter 14). At early devel- CNS myelination.
opmental stages, NRG1 promotes Schwann cell precursor
proliferation and migration, and ablation of NRG1/ErbB3
signaling results in the near complete absence of Schwann 5 P O L A R I Z AT I O N A S A P R E R E Q U I S I T E
cell progenitors in the nerve. Later, NRG1 switches from a F O R M Y E L I N AT I O N
pro-proliferation to a pro-differentiation signal (Birchmeier
and Nave 2008), which is at least partially mediated by the ele- NRG1/ErbB signaling activates glial PI3K, and elevated
vation of cAMP in Schwann cells (Arthur-Farraj et al. 2011). phosphatidylinositol (3,4,5)-trisphosphate (PIP3) has been
In general, NRG1 modulates the Schwann cell phenotype in recognized as a key regulator of cellular polarity in other sys-
a dose-dependent fashion, at least in vitro. Low concentra- tems. This strongly suggests that altered glial cell polarity is
tions promote myelin formation, whereas high concentra- an important step in initiating axonal ensheathment, which is
tions inhibit differentiation, possibly caused by an altered supported by morphological data. Myelin sheaths assembled by
balance between the activated second messenger pathways oligodendrocytes and Schwann cells are polarized, longitudi-
(Syed et al. 2010). nally from node to node, and radially from the inner adaxonal
NRG1 serves as an instructive signal for myelination and membrane (and compact myelin) to the outer abaxonal mem-
provides Schwann cells with the information about axons brane. In analogy to polarized transport in epithelial cells, the
that have reached radial size (~1 μm) to be myelination biogenesis of myelin requires correct spatiotemporal sorting
competent. This information is encoded as a threshold level of cargo proteins and the recruitment of proteins, lipids, and
of NRG1 type III on the axonal surface. Indeed, overex- mRNA to subcellular domains, which requires a tightly regu-
pression of NRG1 type III in normally unmyelinated fibers lated intracellular trafficking machinery (Simons and Trotter
results in their de novo myelination in vitro (Taveggia et al. 2007). For example, major myelin lipids and structural pro-
2005) and in vivo (Schwab and Nave in preparation). First teins are delivered in vesicules from the trans Golgi network
evidence that NRG1/ErbB signaling regulates myelination (TGN) to the plasma membrane, and are later internalized by
came from hypomyelinated mouse mutants lacking ErbB2 cholesterol-dependent and clathrin-independent endocyto-
selectively from EGR2-positive Schwann cells (Garratt sis, transported to a late endosome/lysosomal compartment
et al. 2000). Direct proof that axonal NRG1 type III reg- (Trajkovic et al. 2006) or directly targeted to the growing
ulates myelin sheath thickness involved mouse mutants myelin compartment. This balance of endocytosis and exocy-
with different NRG1 gene dosages (Michailov et al. 2004). tosis at the cell membrane is thought to regulate myelin bio-
Heterozygous mice exhibit a reduced content and presum- genesis (Simons and Trotter 2007).
ably density of NRG1 in PNS axons, which are only thinly The key principles of establishing the basolateral-apical
myelinated (see Fig. 44.1A). In contrast, transgenic mice polarity in epithelial cells are also found in myelinating glial
that overexpress NRG1 type III in neurons exhibit a hyper- cells (Baron and Hoekstra 2010; Pereira et al. 2012). In epi-
myelination phenotype (see Fig. 44.1A) (Michailov et al. thelial cells, apical and basolateral membrane domains exhibit
2004; Taveggia et al. 2005). This increase in myelin sheath different compositions of membrane proteins and lipids. The
thickness is specific for NRG1 type III, because no periph- major regulatory protein complexes are the crumbs (CRB)
eral hypermyelination is induced by neuronal overexpres- complex localizing to the apical domain, the scribble (SCRIB)
sion of NRG1 type I (Michailov et al. 2004). Interestingly, complex defining the basolateral membrane domain, and the
mice heterozygous for the ErbB2 gene are fully myelinated, partitioning-defective protein (Par) polarity complex that pro-
demonstrating that only the ligand, not the receptor, is motes the formation of the apical-basolateal membrane bor-
rate-limiting for myelination (Michailov et al. 2004). Once der (Martin-Belmonte and Perez-Moreno 2012; Mellman and
myelination has been completed, continued NRG1/ErbB Nelson 2008). Also, the signaling lipids phosphatidylinositol
signaling is mostly dispensable for myelin maintenance (4,5)-bishosphate (PIP2) and PIP3 are asymmetrically distrib-
in adult life (Atanasoski et al. 2006), and would only be uted, with PIP3 (and its synthesizing enzyme PI3Kinase) being
required for remyelination, for example after peripheral enriched in the basolateral domain and PIP2 (and the lipid
nerve injury (Fricker et al. 2011). phosphatase and tensin homolog [PTEN]) predominantly

560 • NEUROGLIA
found in the apical domain (Mellman and Nelson 2008). N EC T I N-L I K E P ROT E I NS
How does this cellular architecture translate to myelinating
Nectin-like proteins (NECL1–NECL4) comprise a family of
glial cells?
immunoglobulin-like adherens junction proteins, localized at
In peripheral myelin, the epithelial basolateral markers
both sides of the internodal axon–glia interface, which con-
are enriched in the abaxonal domain, whereas apical mark-
tribute to Schwann cell–axon adhesion during myelin forma-
ers define the adaxonal domain and Schmidt-Lanterman
tion (Maurel et al. 2007; Spiegel et al. 2007) (see Fig. 44.2).
incisures. These observations suggest that the control of
They predominantly undergo heterophilic binding, for exam-
Schwann cell polarization is similar to that of epithelial
ple, NECL1 (on axons) with NECL4 (on Schwann cells). In
cells (Pereira et al. 2012). Indeed, the polarity protein Par3,
myelinating DRG cocultures, NECL4 expression in Schwann
by directly interacting with p75NTR , is required for the
cells increases upon axonal contact, and is required for normal
polarization of Schwann cells before myelination (Chan
Schwann cell differentiation and in vitro myelination (Maurel
et al. 2006). Moreover, interfering with the uneven distri-
et al. 2007; Spiegel et al. 2007). However, NECL function is
bution of PIP2 and PIP3 in Schwann cells induces strik-
mechanistically distinct from that of signaling proteins. When
ing nerve pathology, with myelin outfoldings and focal
axonal NECL1 was ablated in mice, mutants were still nor-
hypermyelination in mutant mice (Goebbels et al. 2012).
mally myelinated in sciatic nerves, and only a delayed myeli-
Apical-basolateral polarity of epithelial cells contributes to
nation of optic nerve and spinal cord was noted (Park et al.
directional transport. Correspondingly, the polarity pro-
2008). These discrepancies of in vitro and in vivo findings are
teins Dlg1 (in concert with MTMR2, Sec8, and the kinesin
not well explained, but point to functional redundancy of
Kif13b) and Pals1 (required for the polarized distribution
NECL isoforms, or differences between acute and chronic
of Sec8 and syntaxin4) affect myelin formation by regulat-
perturbation experiments.
ing polarized vesicle trafficking and thus myelin membrane
formation (Pereira et al. 2012).
Also, basal lamina components, such as laminins, are AXO NA L P R I O N P ROT E I N
required for normal Schwann cell polarization (Feltri and
Wrabetz 2005) (see section 9). Schwann cells express various The widely expressed prion protein, a membrane-anchored
types of laminin receptors exerting distinct roles (Previtali protein known for its detrimental role in transmissible
et al. 2003). Integrin signaling often converges onto the regu- Creutzfeldt-Jakob disease, plays an unexpected role in the long-
lation of small Rho GTPases, such as Rac1, Rho, and Cdc42. term maintenance of peripheral myelin (see Fig. 44.2). Mice
Indeed, Cdc42 and Rac1 are crucial for early stages of myelin lacking prion protein cellular (PrPc) develop an adult-onset
formation in the CNS (Thurnherr et al. 2006). A critical role demyelinating neuropathy, predominantly affecting large-cal-
for Rac-dependent actin regulation has also been suggested iber fibers. Surprisingly, it is the transgenic ablation of axonal
for Schwann cells during axonal sorting and myelination (Park (not glial) PrPc that can prevent that demyelinating pheno-
and Feltri 2011). Recently it was further demonstrated that type in PrPc mutant mice (Bremer et al. 2010). The underlying
another actin regulator, neuronal Wiskott–Aldrich syndrome molecular mechanisms are not understood, and CNS myelin
protein (N-WASP), a downstream effector of RhoGTPases appears unaffected by the absence of axonal PrPc.
including Rac1, is similarly required for axonal sorting and
myelination (Park and Feltri 2011). Together, these findings
strongly indicate that extracellular signals affecting the actin 7 AXO N A L E L E C T R I C A L AC T I VI T Y
cytoskeleton are required for myelination.
That myelination is influenced by action potentials of the
axon was first suggested in the 1960s, when it was noted that
6 D I R E C T AXO N – G L I A I N T E R AC T I O N S optic nerve myelination is decreased in animals raised in the
T H AT P R O M OT E M Y E L I N AT I O N A N D dark (Gyllensten and Malmfors 1963) (see also chapter 45).
MYELIN M AINTENANCE With the help of neurotoxins, electrical activity was shown
to promote oligodendrocyte survival (Barres and Raff 1993)
Compared with Schwann cells, oligodendrocyte lineage and enhance myelination (Demerens et al. 1996). In CNS
cells respond to a much larger number of stimulatory signals preparations, the release of adenosine, adenosine triphos-
(Taveggia et al. 2010), many of which also affect myelination, phate, and glutamate by electrically active axons promotes
but they are not as well studied in vivo. Moreover, the cellular oligodendrocyte differentiation (Stevens et al. 2002; Wake
source of soluble factors in the CNS is not always clear and et al. 2011). Furthermore, in vitro, Adenosine inhibits OPC
can include astrocytes. Some of the myelin structural pro- proliferation via adenosine receptors (Stevens et al. 2002;
teins are not essential for myelin assembly, but for maintain- Wake et al. 2011). Glutamate, released from vesicular sources
ing long-term function and stability of the axon–myelin unit along the axonal membrane, was shown to induce myelina-
(e.g., PLP/DM20 or MAG, Fig. 44.2), which is discussed in tion in DRG neuron/oligodendrocyte cocultures, mediated
more detail in chapter 62. Only few myelination-promoting by N-methyl-d-aspartate (NMDA) receptors and metabotro-
factors are clearly axonal in nature, and these have been pic glutamate receptors (Wake et al. 2011). Here, glutamate
mostly studied in the PNS (as detailed for NRG1 above (see induces the phosphorylation of Fyn, leading to the local trans-
section 4)). lation of transcripts for myelin basic protein (MBP), that is,

FAC TO R S C O N T R O L L I N G M Y E L I N F O R M AT I O N • 561
Axon NRG1 type III
COOH

TACE/ NH2 Jagged


NECL1 ADAM22 ADAM19 ADAM17 Bace1

EGF
PrPc
Lgi4

EGF
? ATP

PIP3 PIP2 P P MAG


GPR 126 P2Y
P P p75NTR
NECL4 PI3K Myelin
PTEN P P maintenace PAR3
DIg1 Notch
ErbB2 - ErbB3
DIg1
Initiation of
P P myelination/
cAMP ? ? AKT PLCγ MEK SHP2
polarity

?
Ca2+ Calcineurin ?

SREBPs
P P P
mTOR NFATc4 NFATc4 Erk1/2 (+ cAMP)

Lipid/ P
Choresterol NFATc4 SOX10 YY1
biosynthesis

EGR2

Schwann cell MYELINATION

Figure 44.2 Multiple Factors Controlling Peripheral Nervous System Myelin Formation. In the peripheral nervous system, myelination by Schwann
cells depends on instructive cues from the underlying axon. The schema lists different ligand–receptor systems of axon–glia interaction and their
downstream signaling cascades that have been implicated in peripheral myelination. Question marks denote interactions that are still poorly under-
stood. Black lines mark positive myelination-promoting effects, orange lines inhibitory effects. At present, the best-studied axonal growth factor is
NRG1 type III. After proteolytic activation by BACE1 and other secretases, this NRG isoform remains membrane-tethered (via a second transmem-
brane domain), with the epidermal growth factor domain (EGF)–like domain at the new C-terminus becoming a juxtracrine signal. However, the
release of this EGF-like domain following a secondary proteolytic cleavage step is also possible and could lead to paracrine signaling underneath the
myelin sheath (not shown). In contrast to BACE1, protease TACE/ADAM17 cleaves NRG1 type III within the EGF-like domain, thereby inactivat-
ing it. Binding of NRG1 to ErbB3 induces ErbB2-ErbB3 heterodimerization, autophosphorylation and transphosphorylation of specific tyrosine
residues, and recruitment of adaptor proteins. This results in the activation of several signaling cascades, including the AKT/mTOR, RAS/ERK1/2,
and Calcineurin/NFAT pathways, which collectively control myelination at the transcriptional and posttranslational level (downstream transcription
factors are highlighted in light brown). For details and further references see text Modified from Pereira et al. 2012.

in compartments adjacent to electrical active axons (Wake 2000) (see Fig. 44.2). In Schwann cell/DRG neuron cocul-
et al. 2011). The in vivo requirement for this type of regula- tures, action potentials increase the ATP concentration and
tion is unclear, however, as NMDA receptors are dispensable activate Ca2+/calmodulin and MAPK signaling pathways via
for oligodendrocyte development and myelination (De Biase purinergic P2 receptors. Subsequently, phosphorylation of the
et al. 2011). In vitro, astrocytes respond to axonal electrical transcription factor CREB and expression of c-fos and Krox24
activity (and the associated release of ATP) with secretion of are induced (Stevens and Fields 2000). Whether these imme-
leukemia inhibitory factor (LIF), a cytokine that promotes diate early genes antagonize maturation of Schwann cells in
myelination by oligodendrocytes. response to ATP remains to be established. Different puri-
Interestingly, neuronal activity inhibits Schwann cell nergic receptors in Schwann cells and oligodendrocytes may
proliferation and differentiation in vitro (Stevens and Fields account for different effects of electrically active axons.

562 • NEUROGLIA
8 I N H I B I TO R S O F M Y E L I N G R OW T H (Givogri et al. 2002). Significantly, the function of Notch1 in
oligodendrocytes may depend on its interaction with different
In the CNS, factors that negatively regulate myelin forma- ligands and downstream signaling pathways. For example, the
tion exceed myelination-promoting factors. This may reflect GPI-anchored neural cell recognition protein F3/Contactin1,
a strong intrinsic drive of oligodendrocyte to myelinate target localized at the paranodal region, is a functional, noncanonical
structures at least in part by default. Thus, negative signals may ligand of Notch1. Interaction of Notch1 with F3/Contactin1
be required to control the spatiotemporal development of oli- promotes oligodendrocyte differentiation, although in vivo data
godendrocytes and inappropriate ensheathment of neuronal are still lacking. In the PNS, inverse functions of Notch have
and glial processes. Indeed, axon-derived inhibitory signals, been observed. The canonical Notch1 pathway induces Schwann
including Lingo1 or Jagged, can prevent precocious oligo- cell proliferation and promotes the differentiation from precur-
dendrocyte differentiation and myelination. Clearly, the dys- sors into mature Schwann cells, possibly via Notch1-mediated
regulation of such inhibitory signals might contribute to the upregulation of ErbB2 receptors (Woodhoo et al. 2009). Before
failure of efficient myelin repair in diseases, such as multiple myelination, the transcription factor Egr2 induces a decrease of
sclerosis (see section 9.2). the NICD protein. This repression of Notch signaling during
myelination is required because Notch1 turns into an inhibitory
factor that delays myelin formation at later developmental stages
L I N G O1
(see Fig. 44.2), most probably via noncanonical Notch signal-
The leucine rich repeat and Ig domain containing NOGO ing and posttranslationally targeting Wnt/E-catenin (Woodhoo
receptor interacting protein 1 (Lingo1) is a transmembrane et al. 2009).
protein, originally described as a neuronal coreceptor for
Nogo-66 and other myelin-associated neurite outgrowth
P S A-N C A M
inhibitors (Mi et al. 2004). Ablation of the Lingo1 gene in
mice results in an enhanced maturation of oligodendrocytes The polysialylated form of the neural cell adhesion molecule
and a premature onset of myelination (Mi et al. 2005). Lingo1 NCAM (PSA-NCAM) is highly expressed on the axonal mem-
is expressed on axons and oligodendrocytes, and is thought to brane before myelination and later downregulated (Charles
exert its function via cis interactions with Nogo Receptor 1 et al. 2000). PSA-NCAM acts as a negative regulator of myeli-
(NgR1) and p75NTR . Lingo1 induces a downregulation of Fyn nation in cell culture, possibly by preventing myelin-forming
kinase and subsequent activation of RhoA kinase in oligoden- cells to attach the axonal surface, and removal of PSA-NCAM
drocytes, at least in vitro. Likewise, disruption of Fyn inhibits from axons is required for the initiation of myelination in vitro
oligodendrocyte differentiation and myelination in vitro (Mi (Charles et al. 2000). A transgenic mouse line lacking the normal
et al. 2004, 2005). Also, axonal Lingo1 emerged as an inhibi- downregulation of PSA-NCAM exhibits myelin abnormalities
tor of oligodendrocyte differentiation and myelination in vitro and axonal degeneration (Fewou et al. 2007). However, an effi-
(Lee et al. 2007). Transgenic mice that overexpress Lingo1 in cient inhibition of myelination by PSA-NCAM, as observed in
neurons show an impairment of CNS myelination, as demon- cell culture, has not yet been observed in vivo.
strated by a reduced number of myelinated axons in the spinal
cord. Expression of axonal Lingo1 is regulated, at least in vitro, In summary, the “positive” regulation of myelination
by neurotrophins and the TrkA receptor (Lee et al. 2007). The by direct axo–glial signaling is most important in the PNS,
expression of Lingo1 has no obvious effect on Schwann cell whereas the correct timing of myelination in the CNS involves
myelination (Lee et al. 2007; Mi et al. 2005). inhibitory signals that may also prevent aberrant myelination
of cellular processes.
N OTC H1
Different effects on PNS and CNS myelination have also been 9 R O L E O F T H E E X T R AC E L LU L A R
reported for the receptor protein Notch1 expressed by glial M AT R I X I N M Y E L I N AT I O N
cells. At early developmental stages, the canonical Notch1
ligands Delta and Jagged are highly expressed in neurons of The contact of a myelinating glial cell to its extracellular matrix
the CNS, whereas in the PNS both Schwann cells and axons constitutes an important prerequisite for the correct forma-
only express the ligand Jagged1. Upon ligand binding, Notch tion of a myelin sheath. The extracellular matrix mediates
is cleaved by gamma-secretase, and the notch intracellular the integration of other signaling molecules, such as growth
domain (NICD) translocates to the cell nucleus and activates factors, and the embedding of cytoskeletal dynamics (Pereira
gene transcription (Taveggia et al. 2010). Notch1 signaling et al. 2012; Taveggia et al. 2010). Among the extracellular
inhibits oligodendrocyte differentiation and myelination in matrix molecules, laminins, integrins, and dystroglycans are
vitro (Givogri et al. 2002; Taveggia et al. 2010). In vivo, abla- functionally best characterized.
tion of Notch1 induces premature expression of myelin pro-
teins (Genoud et al. 2002). Interestingly, some myelinated
L A M I N I NS
axons were also observed in the molecular layer of the cerebel-
lum, which normally remains unmyelinated. Thus, Jagged1 Laminins are heterotrimeric proteins comprising at least 15
expression on axons may also prevent aberrant myelination different isoforms. They are essential components of the basal

FAC TO R S C O N T R O L L I N G M Y E L I N F O R M AT I O N • 563
lamina (Chernousov et al. 2008), which is a specific feature Integrin interacts with NRG1/ErbB signaling in the reg-
of Schwann cells. The natural dystrophic mouse, defined by a ulation of oligodendrocyte differentiation, at least in vitro
mutation in the gene for the laminin alpha 2 subunit, exhib- (Barros et al. 2009; Colognato et al. 2002). Here, the binding
its a peripheral neuropathy (Helbling-Leclerc et al. 1995). A of laminin to E1 integrin switches NRG1 from a proliferation
conditional null mutant of the laminin gamma-1 gene (lack- signal to a differentiation signal of oligodendrocytes, which is
ing all laminin expression) shows a severe defect of the radial mediated by shifting the downstream response from PI3K to
sorting of axons and an impaired formation of the basal MAPK signaling (Barros et al. 2009; Colognato et al. 2002).
lamina, with mutant Schwann cells arrested at the premy- Since laminin is localized on the axonal surface, laminin can
elinating stage (Chernousov et al. 2008). Laminins are the be considered an axonal signal that promotes oligodendrocyte
ligands for integrin and dystroglycan receptors that activate differentiation upon axonal contact (Colognato et al. 2002).
different downstream signaling pathways. Laminin-deficient The dystroglycan receptor of laminin contributes to the
Schwann cells display a reduction of ErbB2 phosphoryla- terminal steps of oligodendrocyte differentiation and myelina-
tion and impaired PI3K signaling (Yang et al. 2005; Yu et al. tion. Blocking dystroglycan in vitro impairs myelin formation
2005), suggesting that ErbB and laminin/integrin signaling without affecting oligodendrocyte survival (Colognato et al.
are coupled (see the following). 2007). Because dystroglycan influences the ability of insulin-
like growth factor 1 (IGF1) to promote oligodendrocyte dif-
ferentiation, also laminin may act via a modulation of IGF1
I N T EG R I NS A N D DY S T RO G LYC A N
signaling (Galvin et al. 2010).
Laminin binds to the E1 integrin and the dystroglycan recep-
tor. Ablation of either receptor in Schwann cells results in a
severe defect of axonal segregation, leading to hypomyelina- 10 E X T R AC E L LU L A R FAC TO R S A N D
tion and defective myelin compaction (Berti et al. 2011). O R P H A N R E C E P TO R S
Importantly, E1 integrin and dystroglycan have sequential,
nonredundant roles for myelination. E1 Integrin is required
I NS U L I N-L I K E G ROW T H FAC TO R 1
early in development for Schwann cell proliferation and sur-
vival (Berti et al. 2011). In contrast, dystroglycan mutant mice Insulin-like growth factor 1 (IGF1) acts mainly through the
develop extensive foldings of the myelin sheath, disorganized type I IGF receptor tyrosine kinase (IGFR1), and both IGF1
nodal structures, and an abnormal distribution of sodium and IGFR1 are expressed in neurons and glial cells (Anlar et al.
channels at the axon surface (Berti et al. 2011). Laminin 2 and 1999). Insulin-like growth factor signaling is also thought to
dystroglycan have also been implicated in the compartmental- mediate the effect of growth hormone (GH) on myelination.
ization and the elongation of the myelin sheath, presumably A positive role of IGF1 in myelination was convincingly dem-
by promoting Schwann cell polarization (Court et al. 2009). onstrated in IGF1-overexpressing transgenic mice that are
Indeed, dystroglycan forms a complex with other proteins of hypermyelinated throughout the CNS (Carson et al. 1993;
the dystrophin family in specific subcellular domains (Court Ye et al. 1995). In turn, ablation of the IGF1 gene impairs
et al. 2009). The cleavage by matrix metalloproteinases 2 and oligodendrocyte proliferation and myelination. However,
9 modulates the composition of this complex and the size of no persistant myelin abnormalities can be observed in adult
cytoplasmic Schwann cell compartements (Court et al. 2011). mutants, suggesting that the upregulation of IGF2 compen-
Additionally, E4 integrin is polarized in Schwann cells, with sates functionally (Beck et al. 1995; Ye et al. 2002). The con-
predominant expression at the abaxonal site. This protein is ditional deletion of the receptor IGFR1 in oligodendrocytes
important for myelin stability since abnormal myelin infold- confirmed the role of IGF1 in myelin formation (Zeger et al.
ings have been observed in aged E4 integrin knockout mice 2007). In vitro, IGF1 also promotes Schwann cell myelina-
(Feltri et al. 1994; Nodari et al. 2008). tion (Russell et al. 2000) and induces fatty acid biosynthesis
In the CNS, the laminin mutation of dystrophic mice by activating the PI3K/AKT pathway (Liang et al. 2007).
causes a regional hypomyelination, involving abnormal PI3K
and integrin-linked kinase (ILK) signaling (Chun et al.
FI B RO B L A S T G ROW T H FAC TO R S
2003). Laminin/integrin signaling induces oligodendrocyte
differentiation, at least in part, by modulating Fyn kinase Fibroblast growth factors (FGF) and their receptor tyrosine
(Buttery and ffrench-Constant 1999; Relucio et al. 2009). kinases (FGFR1–4) comprise complex protein families with
Also the integrin/contactin complex promotes myelination widespread functions in proliferation, differentiation, and
by stimulating Fyn activity in vitro (Laursen et al. 2009). organogenesis (Mason 2007). Neurons and astrocytes express
Whether E1 integrin itself is required for CNS myelina- FGF1 and FGF2 at the time of myelination, and FGFRs are
tion is controversial. In two studies, mouse mutants with developmentally regulated in the oligodendrocyte lineage
targeted ablation of E1 integrin expression in oligoden- (Fortin et al. 2005). Because FGF2 stimulates the prolifera-
drocytes exhibited an impaired initiation of myelination, tion of OPCs (in vitro), it appears inhibitory to differentia-
whereas in a third study E1 integrin was required for oligo- tion (Bansal, 2002). FGFR1 is expressed in both OPCs and
dendrocyte survival but not for CNS myelination or remy- differentiated oligodendrocytes, whereas FGFR2 is more spe-
elination (Barros et al. 2009; Benninger et al. 2006; Camara cific to the latter, enriched in membrane lipid-rafts, and associ-
et al. 2009). ated with paranodes (Bryant et al. 2009). Fibroblast growth

564 • NEUROGLIA
factor receptor activation of mature oligodendrocytes stimu- 2011; Monk et al. 2009). Indeed, pharmacological elevation
lates process-outgrowth (Fortin et al. 2005). A mild CNS of cAMP levels in zebrafish GPR126 mutants can rescue the
hypomyelination phenotype in mice with targeted disruption Schwann cell differentiation arrest (Monk et al. 2009). The
of both FGFR1 and FGFR2 in oligodendrocytes suggests that mouse mutant of GPR126 develops a severe dysmyelination
FGF signaling regulates myelin membrane growth and myelin and peripheral neuropathy, with defects of axonal sorting and
sheath thickness, perhaps similar to NRG1/ErbB signaling altered Remak bundles, leading to premature death at 16 days
in the PNS (Furusho et al. 2012). However, the responsible (Monk et al. 2011).
ligand(s) are not well defined and their association with the GPR17 is a P2Y purinergic receptor of nucleotides as well as
axonal membrane would only be indirect, for example, by a receptor for cysteinyl-leukotrienes (Ciana et al. 2006), and is
binding to extracellular matrix proteins. transiently expressed during oligodendrocyte differentiation.
GPR17 has been implicated in CNS myelination, because it
inhibits oligodendrocyte differentiation when transgenically
WN T S I G NA L I N G
overexpressed (Chen et al. 2009). In turn, lack of this receptor
The canonical Wnt/E-catenin pathway regulates glia cell dif- causes premature differentiation of oligodendrocytes. These
ferentiation and myelination via a complex transcriptional and effects are in part mediated by the inhibitory transcription fac-
epigenetic modulation of gene expression (Fancy et al. 2009; tors Id2 and Id4 that translocate to the nucleus upon GPR17
Feigenson et al. 2009; Tawk et al. 2011; Ye et al. 2009) (see activation (see chapter 43) (Chen et al. 2009). Since Id2 and
chapter 43). However, the cell type that provides Wnt during Id4 physically interact and sequester the differentiation factors
development is not known. Classically, the binding of Wnt Olig1 and Olig2, it is thought that GPR17 counteracts their
to its receptor frizzled at the cell membrane stabilizes cytoso- myelination-promoting functions (Chen et al. 2009; Emery
lic E-catenin, which subsequently forms a complex with the 2010b).
transcription factors TCFs (T-cell factor/lymphoid enhancer
factor or TCF/LEF) to activate (or repress) target genes. In
transgenic mice, constitutive overexpression of E-catenin in 11 N E U R OT R O P H I N S
oligodendrocytes inhibits oligodendrocyte differentiation and
delays myelination (Fancy et al. 2009). That continuous Wnt Neurotrophins are growth and survival factors that are widely
signaling has only a transient effect may be caused by its bind- expressed by neurons and glial cells, comprising NGF, BDNF,
ing partner TCF4 (TCF7L2). Since TCF4 is normally down- NT-3, and NT-4/5. The vital function of neurotrophins in
regulated in development, its absence at later stages is likely to early development has hampered their in vivo analysis during
prevent efficient Wnt signaling, and thereby to allow myelin postnatal myelination, but insight came from in vitro systems
gene transcription to resume (Rosenberg and Chan 2009). The (Rosenberg et al. 2006; Xiao et al. 2009). All neurotrophins are
histone deacetylases HDAC1 and 2 antagonize Wnt signal- ligands for the low-affinity receptor p75NTR and one or more
ing and stimulate oligodendrocyte differentiation by prevent- high-affinity Trk (tropomyosin-related kinase) receptors.
ing the complex formation of TCF4 with E-catenin (Ye et al.
2009). However, exogeneous Wnt has been shown to promote
N E RVE G ROW T H FAC TO R
myelin gene expression in Schwann cells and oligodendrocytes
in vitro, and ablation of Tcf3 in zebrafish inhibits myelinogen- Nerve growth factor (NGF) promotes myelination in vitro by
esis by Schwann cells (Tawk et al. 2011). These discrepancies activating TrkA on the axonal surface, potentially mimicking
may be explained by a change of Wnt/E catenin function in glia-to-axon signaling (Chan et al. 2004). However, in vivo
different developmental stages. In addition to its role during only a subpopulation of small-caliber nociceptive axons that
myelination, Wnt signaling is also required for remyelination, remain largely unmyelinated expresses TrkA receptors (Xiao
and a dysregulation of the Wnt pathway has been implicated in et al. 2009). How axonal TrkA receptors influence myelina-
remyelination failure of MS (Fancy et al. 2009). tion has not been clarified, but an interaction with NRG1
signaling has been suggested. For example, soluble NRG1 iso-
forms have been suggested to be locally released in response to
G P ROT E I N– C O U P L E D R E C E P TO R S
Schwann cell–derived NGF, BDNF, and GDNF (Esper and
G protein–coupled receptors (GPCR) have been identified Loeb 2004, 2009). In CNS-derived cultures, binding of NGF
as crucial for glial cell differentiation and myelination. In a to axonal TrkA receptors does not promote but rather inhibits
mutant screen of zebrafish, GPR126 was identified as essen- oligodendrocyte differentiation and myelination (Chan et al.
tial for Schwann cell differentiation and myelination (Monk 2004), potentially by inducing the inhibitor Lingo on the
et al. 2009) (see Fig. 44.2). Mutant Schwann cells are arrested axonal surface (Lee et al. 2007; Mi et al. 2005).
at the promyelin stage and fail to express the transcription
factors Oct6/SCIP and Egr2, which are essential for myeli-
B R A I N-D E R I VE D N EU ROT RO P H I C FAC TO R
nation (Monk et al. 2009). However, GPR126 has remained
an “orphan receptor” because its natural ligand is unknown. Brain-derived neurotrophic factor (BDNF) has been impli-
Because Schwann cells normally respond to elevated levels of cated in axon-to-glia and glia-to-axon signaling. A neuronal
cAMP, it has been suggested that GPR126 promotes myelina- increase of BDNF expression enhances myelin formation in
tion via the activation of adenylate cyclase (Arthur-Farraj et al. DRG neuron/Schwann cell cocultures. This is presumably

FAC TO R S C O N T R O L L I N G M Y E L I N F O R M AT I O N • 565
by interaction with p75NTR and p75NTR mutant mice display mice revealed a mild hypomyelination in several CNS regions
impaired peripheral myelination (Cosgaya et al. 2002). One (Vondran et al. 2010; Xiao et al. 2010). However, whether this
suggested mechanism is the interaction of p75NTR with the myelination defect is transient or persists in adulthood is less
polarity protein Par3, regulating the initial polarization of clear and may be regionally different (Du et al. 2003; Vondran
Schwann cells that is required for myelination (Fig. 44.2) et al. 2010; Xiao et al. 2010). When BDNF expression is con-
(Chan et al. 2006). Whereas BDNF-mediated activation of ditionally ablated selectively in postmitotic neurons, mutant
p75NTR on axons enhances myelin formation by Schwann cells mice show normal oligodendrocyte differentiation and unal-
in vitro (Xiao et al. 2009), BDNF binding to the axonally tered myelination in the optic nerve (Rauskolb et al. 2010).
bound TrkB inhibits myelination in vivo, and the expres- Thus, other cellular sources of BDNF (possibly astrocytes)
sion level of this neurotrophin varies widely, for example, may play a role and again myelination needs to be analyzed in
between different subpopulations of DRG neurons (Xiao different brain regions.
et al. 2009).
In oligodendrocyte cultures (in contrast to Schwann cells),
N EU ROT RO P H I N-3
BDNF exerts a promyelinating effect by stimulating glial
TrkB receptors (Xiao et al. 2010). Different loss-of-function Neurotrophin-3 (NT3) signaling to Schwann cells for myelin
mutants have been investigated. Heterozygous BDNF mutant formation is still debated, with most in vitro data suggesting
that NT3 acts as an inhibitory factor of myelination by acti-
vating TrkC receptors. In contrast, ablation of NT3 expres-
A sion in mice is associated with mildly impaired myelination,
control PTEN mutant but this myelination defect may be secondary to a massive
neuronal loss (Xiao et al. 2009).

In conclusion, neurotrophins act as modulators of myelin


formation. Their role in myelination can be both inhibitory
and promoting, depending on the receptor composition pre-
sented on glial cells and axons. Finally, Glial cell line–derived
neurotrophic factor (GDNF), is not a “neurotrophin,” but a
member of the TGF-E family of growth factors. GDNF has
been shown to trigger myelination of normally unmyelinated
axons in the sciatic nerve, which is most probably secondary
to a massive increase of Schwann cell proliferation (Höke
et al. 2003).

12 D I VE R S I F I C AT I O N O F
G R OW T H FAC TO R F U N C T I O N BY
B
P O S T T R A N S L AT I O N A L P R O C E S S I N G
control AKT mutant
Proteolytic processing is a posttranslational mechanism by
which growth factor signaling is modified, for example, by
determining whether the growth factor or its receptor is pre-
sented as a soluble or membrane-bound protein. This deci-
sion may alter downstream signaling pathways in axons and
myelinating glial cells. “Secretases,” such as ADAM proteins,
BACE1, and the J-secretase complex have all been implicated
in myelination. ADAM17, a D-secretase also known as TACE,
and BACE1 both cleave NRG1 type III at distinct but closely
spaced sites, leading to opposite effects on myelination (see
Figs. 44.1B and 44.2). While BACE1 activates NRG1 type
Figure 44.3 Elevation of PI3K Signaling in Oligodendrocytes Induces III and promotes the myelination by Schwann cells (Hu et al.
Hypermyelination. A. Increasing the signaling lipid PIP3 in oligodendro-
cytes by cell-specific ablation of the lipid phosphatase PTEN. When ana- 2006; Velanac et al. 2012; Willem et al. 2006) and also remy-
lyzed by magnetic resonance imaging (T2 weighted images; top panel) the elination after nerve injury (Hu et al. 2008), TACE process-
subcortical white matter (arrow: corpus callosum) has a larger volume in ing negatively regulates peripheral myelination, most likely by
PTEN mutant mice compared with controls. This phenotype is associated cutting within the EGF domain and thus reducing the level
with increased myelin sheath thickness. B. Transgenic overexpression of a of functional NRG1 type III (La Marca et al. 2011). It is yet
constitutively active isoform of AKT kinase in oligodendrocytes also causes
a similar pattern of mTOR-dependent hypermyelination (Flores et al. 2008; to be determined in which subcellular neuronal/axonal com-
Goebbels et al. 2010; Narayanan et al. 2009). (A) Taken from Goebbels partment these processing steps occur. That BACE1 activity
et al. 2010. (B) and Flores et al. 2008. exerts a similar effect on CNS myelination (Hu et al. 2006)

566 • NEUROGLIA
has been debated (Treiber et al. 2012). Nardilysin (NRD1, proteins raptor and rictor, respectively (Tyler et al. 2009).
N-arginine dibasic convertase) is a metalloendopeptidase and Independent evidence for the role of PI3K signaling in CNS
enhancer of protein ectodomain shedding that has been asso- myelination comes from mice lacking PTEN (phosphatase and
ciated with NRG1 cleavage (Ohno et al. 2009). Whether its tensin homolog) in oligodendrocytes. This enzyme antagonizes
myelination-promoting function in PNS and CNS is related PI3K signaling by dephosphorylating PIP3 at the D3 position
to the activation of TACE, BACE1, or other proteases is not of the inositol and recreating PIP2. Consequently, PTEN
known. Two other ADAMs have been involved in periph- ablation in oligodendrocytes leads to elevated PIP3 levels and
eral myelination (see Fig. 44.2): ADAM19/Meltrin-beta activation of mTOR and its downstream kinases, resulting in
deficient mice exhibit a delayed remyelination upon injury enhanced myelin gene expression and widespread hypermyeli-
(Wakatsuki et al. 2009). Mice devoid of the catalytically nation (see Fig. 44. 3) (Goebbels et al. 2010).
inactive ADAM22 are hypomyelinated, most likely because In Schwann cells, PI3K is activated when axonal NRG1
this neuronal membrane protein is the axonal receptor for type III binds to ErbB2/ErbB3 receptor heterodimers
the secreted soluble Schwann cell protein Lgi4 (leucine-rich (Taveggia et al. 2005), but PI3K is also coupled to receptors
repeat LGI family, member 4) (Ozkaynak et al. 2010; Sagane of other ligands, such as IGF-1 (insulin-like growth factor 1)
et al. 2005). Lgi4 itself is an essential regulator of myelination that stimulate myelination (Liang et al. 2007). Activation of
in the PNS, because a mutation of this gene causes a severe PI3K promotes myelination also in an AKT-dependent fash-
congenital dysmyelination in the spontanous mouse mutant ion, at least in vitro. In cultured Schwann cells, the activation
claw paw (Bermingham et al. 2005). The broadly expressed of AKT after IGF-1 stimulation is thought to mediate the
J-secretase complex cleaves and codetermines the turnover of EGR2-dependent upregulation of P0/MPZ gene expression
a range of transmembrane protein substrates, including those (Wakatsuki et al. 2009). Phosphorylated AKT also promotes
that regulate glial development and myelination, i.e., NRG1 the expression of SREBPs (sterol responsive element binding
(Bao et al. 2003), ErbB4 (Ni et al. 2001), p75NTR (Zampieri proteins), the key transcription factors that regulate the biosyn-
et al. 2005), and Notch1 (Fortini 2002). Oligodendroglial thesis of cholesterol, which is by itself crucial for myelination
J-secretase activity is important for the ensheathment of (Porstmann et al. 2005; Saher et al. 2009). Similar to the CNS,
retinal ganglion cell axons when maintained in a CNS cocul- mTOR downstream signaling is suggested to play a central role
ture system (Watkins et al. 2008). In summary, proteolysis in regulating the growth of the myelin sheath in the PNS in
comprises posttranslational and probably rapid regulation vivo (Sherman et al. 2012). However, transgenic mice that
of axonal myelination signals, and some of the enzymes are overexpress a constitutively active AKT isoform in Schwann
potential targets in pharmacological attempts to boost myelin cells reportedly show normal peripheral myelination (Flores
repair in demyelinating diseases. et al. 2008), in marked difference to the CNS phenotype (see
the preceding). Moreover, the elevation of PIP3 in Schwann
cells following the ablation of PTEN does not cause the same
13 G L I A L R E C E P TO R AC T I VAT I O N A N D hypermyelination phenotype in mice as the neuronal (axonal)
D OW N S T R E A M S I G N A L I N G C A S C A D E S overexpression of NRG1 type III. Instead, isolated elevation of
PIP3 triggers progressive myelin pathology with focal myelin
Despite striking differences of axon–glia signaling in CNS outfoldings and tomacula, similar to findings in some patients
and PNS at the level of axonal ligands and their glial receptors, with demyelinating peripheral neuropathy (Goebbels et al.
essential downstream mechanisms in oligodendrocytes and 2012). Although abnormal mTOR signaling may be causative
Schwann cells appear to be conserved. For example, activated of abnormal myelin growth, the specific pathology is probably
by glial receptors, the enzymes phosphatidylinositol 3-kinase the loss of PTEN as a local “brake,” which limits the insertion
(PI3K) and AKT1 (v-Akt murine thymoma viral oncogene of new membranes into the myelin sheath (Goebbels et al.
homolog 1) are critical for myelination in the entire nervous 2012). This brake is formed by PTEN in conjunction with
system. PI3K phosphorylates the membrane lipid PIP2 to the mammalian Disk-large homolog Dlg1 and stabilized by
produce the key second messenger PIP3. This in turn leads to NRG1 stimulation, likely to prevent ‘overmyelination’ during
the local recruitment and activation of various effector pro- myelin sheath assembly (Cotter et al. 2010).
teins that can bind to PIP3 at the cell membrane with their In addition to PI3K/AKT/mTOR signaling, myelina-
pleckstrin homology (PH) domain. Among those AKT1, tion control also involves other second messenger pathways
a serine-threonine protein kinase, has been studied in most that transduce axonal signaling to Schwann cells, for exam-
detail; however, mostly in the context of CNS myelination. For ple, downstream of the activation of ErbB2/ErbB3 het-
example, transgenic expression of a constitutively active AKT1 erodimers by NRG1 type III. For example, the extracellular
isoform in oligodendrocytes enhances myelin growth by acti- signal regulated kinase 1/2 (Erk1/2) and the non–recep-
vating the serine-threonin protein kinase mTOR (mammalian tor tyrosine phosphatase SHP2/PTPM11 are critical for
target of rapamycin) and its further downstream substrates Schwann cells to fully respond to NRG1. In the absence of
(e.g., S6 kinase and the ribosomal protein S6) (see Fig. 44.3) ERK1/2 from cultured Schwann cells, NRG1 no longer has
(Flores et al. 2008; Narayanan et al. 2009). In addition, mTOR a survival-promoting effect (Newbern et al. 2011). Likewise,
regulates mRNA expression (specifically of MBP, PLP, and normal proliferative and migratory responses to NRG1 can-
enzymes for lipid synthesis) by forming one of two complexes, not be triggered in SHP2-mutant cultured Schwann cells
mTORC1 or mTORC2, which are defined by the adaptor (Grossmann et al. 2009). Conditional inactivation of SHP2

FAC TO R S C O N T R O L L I N G M Y E L I N F O R M AT I O N • 567
in murine Schwann cells causes marked hypomyelination of that axonal electrical activity and the release of glutamate
peripheral nerves associated with a persistent impairment of stimulates myelination by locally increasing Fyn-dependent
ERK1/2 signaling, whereas PI3K/AKT activation is unaf- MBP synthesis (Wake et al. 2011).
fected (Grossmann et al. 2009). Moreover, Schwann cell–
specific inactivation of ERK1/2 inhibits peripheral myelina-
tion (Newbern et al. 2011). 14 S U M M M A RY A N D P E R S P E C T I VE S
Finally, NFATc3/4 (nuclear factor of activated T cells) is
a transcription factor that is critical for NRG1 signaling to The widely held model that neuronal signaling molecules trig-
Schwann cells (Kao et al. 2009). NRG1 binding to ErbB2/ ger the proliferation of axon-associated glial cells and control
ErbB3 receptors induces a phospholipase C (PLC-J)– the synthesis of myelin, originally derived from classical cell
dependent increase in cytosolic calcium levels, which activates culture and nerve graft experiments, has been largely confirmed
calcineurin, a phosphatase that dephosphorylates this tran- at the molecular level and by in vivo experiments. However,
scription factor for subsequent nuclear translocation (Kao in its simplest form the model only applies to Schwann cells
et al. 2009). Mouse mutants that lack an essential regulatory responding to the axonal growth factor NRG1 type III. In
subunit of calcineurin (CnB1) in Schwann cells display severe the peripheral nervous system, this axonal membrane protein
defects in Schwann cell differentiation (Kao et al. 2009). has emerged with an unexpected broad range of develop-
Interestingly, calcineurin mutant mice also show decreased mental functions, from promoting Schwann cell survival in
expression of the transcription factor EGR2, indicating that embryonic development to regulating myelin sheath thickness
EGR2 requires calcineurin for its activity. In addition, NFAT according to axon size in postnatal life. In the CNS, oligoden-
facilitates the binding of Sox10 to the regulatory region of drocytes and their precursor cells respond to a large number of
the Egr2 gene. Thus, a synergistic function NFAT and Sox10 signals, derived mostly as soluble factors from different cellular
upregulates myelin gene expression (Kao et al. 2009). sources, including astrocytes, and including factors associated
For oligodendrocyte maturation, Fyn kinase plays a cen- with electrical activity of axons. However, oligodendrocytes
tral role as an integrator and mediator of axon–glia signaling. appear not to depend on NRG1 or other single growth fac-
Mice lacking functional Fyn kinase depict a marked hypomy- tors to become myelination competent, and the ensheathment
elination in the forebrain and have fewer myelinated fibers in of their axons appears to be triggered by a battery of signals
optic nerve and corpus callosum (Krämer-Albers and White received by oligodendrocytes and a transcriptional program,
2011). Fyn kinase is activated by various axonal signals and and ultimately to occur synchronously “by default.” Thus,
their respective oligodendroglial surface receptors, includ- inhibitory cues that help restrict myelination to appropriate
ing D6E1-integrin, Dcc (deleted in colorectal carcinoma), axonal targets become developmentally equally important.
Lingo-1, and the GPI-anchored protein F3/contactin. The On the down side, these negative factors of CNS myelina-
latter binds the axonal cell adhesion molecule L1 and activates tion appear to also limit myelin repair in neurological diseases,
Fyn specifically in lipid-raft microdomains. Fyn itself relays which is required to protect axonal integrity. Therefore, recent
into three major downstream signaling pathways, all of which research activities have concentrated also on the intracellular
affect the spatiotemporal control of oligodendrocyte differ- second messenger systems that appear to be largely shared by
entiation and myelination (Krämer-Albers and White 2011). my elinating glial cells in PNS and CNS, and that serve to inte-
First, activated Fyn alters oligodendrocyte morphology by grate the plethora of non–cell autonomous signals received.
regulating the Rho-family GTPases RhoA, Cdc42, and Rac1, It is anticipated that a better understanding of bidirectional
known modulators of actin cytoskeleton dynamics and oli- axon–glia interactions, and the even more complex neuron–
godendrocyte process formation (Liang et al. 2004). Second, glia signaling in the CNS, will allow to identify rate limiting
active Fyn recruits microtubuli (via its SH2 and SH3 protein steps of remyelination, which can then be pharmacologically
binding domains) and the microtubule-associated protein targeted for the future treatment of demyelinating diseases.
Tau (Klein et al. 2002), which stabilizes the glial cytoskeleton,
contributes to oligodendrocyte polarization and facilitates
(microtubule-dependent) transport of myelin-cargo toward AC K N OW L E D G M E N T S
the axon–glia contact site. Finally, Fyn affects transport and
stability of MBP mRNA and activates translation of MBP, the The authors thank members of the Nave lab for helpful com-
only known protein that is rate-limiting of CNS myelination ments. Work in the authors’ lab was supported by the Max
(Popko et al. 1987; Readhead et al. 1987). Specifically, MBP Planck Society and grants from the DFG (CMPB, SFB/
mRNA is transported in RNA granules (mediated by hnRNP TR43), BMBF (Leukonet), EU-FP7 (Ngidd, Leukotreat),
A2) from the nucleus to distal oligodendroglial processes in ELA, and the MS Society (USA). KAN holds an ERC
a translationally silenced state. Release of this repression is Advanced Grant (Axoglia).
thought to occur on contact between axonal L1 with the oli-
godendroglial cell process and subsequent Fyn activation and
phosphorylation of hnRNPA2 (White et al. 2008). Hereby, REFERENCES
the local dissociation of the transport granule allows for local-
Aguayo AJ, Charron L, Bray GM. 1976. Potential of Schwann cells from
ized MBP synthesis at the site of axon–glia contact where unmyelinated nerves to produce myelin: a quantitative ultrastruc-
myelin deposition is required. Further, it has been proposed tural and radiographic study. J Neurocytol 5:565–573.

568 • NEUROGLIA
Anlar B, Sullivan KA, d Feldman EL. 1999. Insulin-like growth promote myelin repair via endogenous stem and precursor cells. J Mol
factor-I and central nervous system development. Horm Metab Res Endocrinol 44:13–23.
31:120–125. Camara J, Wang Z, Nunes-Fonseca C, Friedman HC, Grove M, Sherman
Arthur-Farraj P, Wanek K, Hantke J, Davis CM, Jayakar A, Parkinson DL, et al. 2009. Integrin-mediated axoglial interactions initiate
DB, et al. 2011. Mouse schwann cells need both NRG1 and cyclic myelination in the central nervous system. J Cell Biol 185:699–712.
AMP to myelinate. Glia 59:720–733. Carson MJ, Behringer RR, Brinster RL, McMorris FA. 1993. Insulin-like
Atanasoski S, Scherer SS, Sirkowski E, Leone D, Garratt AN, Birchmeier growth factor I increases brain growth and central nervous system
C, et al. 2006. ErbB2 signaling in Schwann cells is mostly dispensable myelination in transgenic mice. Neuron 10:729–740.
for maintenance of myelinated peripheral nerves and proliferation of Chan JR, Cosgaya JM, Wu YJ, Shooter EM. 2001. Neurotrophins are
adult Schwann cells after Injury. J Neurosci 26:2124–2131. key mediators of the myelination program in the peripheral nervous
Bansal R. 2002. Fibroblast growth factors and their receptors in oligo- system. Proc Natl Acad Sci U S A 98:14661–14668
dendrocyte development: implications for demyelination and remy- Chan JR, Jolicoeur C, Yamauchi J, Elliott J, Fawcett JP, Ng BK, et al.
elination. Dev Neurosci 24:35–46. 2006. The polarity protein Par-3 directly interacts with p75NTR to
Bao J, Wolpowitz D, Role LW, Talmage DA. 2003. Back signaling by the regulate myelination. Science 314:832–836.
Nrg-1 intracellular domain. J Cell Biol 161:1133–1141. Chan JR, Watkins TA, Cosgaya JM, Zhang C, Chen L, Reichardt
Baron W, Hoekstra D. 2010. On the biogenesis of myelin membranes: LF, et al. 2004. NGF controls axonal receptivity to myelination by
sorting, trafficking and cell polarity. FEBS Lett 584:1760–1770. Schwann cells or oligodendrocytes. Neuron 43:183–191.
Barres BA, Jacobson MD, Schmid R, Sendtner M, Raff MC. 1993. Does Charles P, Hernandez MP, Stankoff B, Aigrot MS, Colin C, Rougon G,
oligodendrocyte survival depend on axons? Curr Biol 3:489–497. et al. 2000. Negative regulation of central nervous system myelina-
Barres BA, Raff MC. 1993. Proliferation of oligodendrocyte pre- tion by polysialylated-neural cell adhesion molecule. Proc Natl Acad
cursor cells depends on electrical activity in axons. Nature 361: Sci U S A 97:7585–7590.
258–260. Chen Y, Wu H, Wang S, Koito H, Li J, Ye F, et al. 2009. The
Barros CS, Nguyen T, Spencer KSR, Nishiyama A, Colognato H, Muller oligodendrocyte-specific G protein-coupled receptor GPR17 is a
U. 2009. {beta}1 integrins are required for normal CNS myelination cell-intrinsic timer of myelination. Nat Neurosci 12:1398–1406.
and promote AKT-dependent myelin outgrowth. Development Chernousov MA, Yu WM, Chen ZL, Carey DJ, Strickland S. 2008.
136:2717–2724. Regulation of Schwann cell function by the extracellular matrix. Glia
Bauer NG, ffrench-Constant C. 2009. Physical forces in myelination and 56:1498–1507.
repair: a question of balance? J Biol 8:78. Chun SJ, Rasband MN, Sidman RL, Habib AA, Vartanian T. 2003.
Beck KD, Powell-Braxton L, Widmer HR, Valverde J, Hefti F. 1995. Igf1 Integrin-linked kinase is required for laminin-2-induced oligodendro-
gene disruption results in reduced brain size, CNS hypomyelination, cyte cell spreading and CNS myelination. J Cell Biol 163:397–408.
and loss of hippocampal granule and striatal parvalbumin-containing Ciana P, Fumagalli M, Trincavelli ML, Verderio C, Rosa P, Lecca D,
neurons. Neuron 14:717–730. et al. 2006. The orphan receptor GPR17 identified as a new dual
Benninger Y, Colognato H, Thurnherr T, Franklin RJM, Leone DP, uracil nucleotides/cysteinyl-leukotrienes receptor. EMBO J 25:
Atanasoski S, et al. 2006. Beta1-integrin signaling mediates premy- 4615–4627.
elinating oligodendrocyte survival but is not required for CNS myeli- Colognato H, Baron W, Avellana-Adalid V, Relvas JB, Baron-Van
nation and remyelination. J Neurosci 26:7665–7673. Evercooren A, Georges-Labouesse E, et al. 2002. CNS integrins
Bermingham JR, Shearin H, Pennington J, O’Moore J, Jaegle M, Driegen switch growth factor signalling to promote target-dependent sur-
S, et al. 2005. The claw paw mutation reveals a role for Lgi4 in periph- vival. Nat Cell Biol 4:833–841.
eral nerve development. Nat Neurosci 9:76–84. Colognato H, Galvin J, Wang Z, Relucio J, Nguyen T, Harrison D,
Berti C, Bartesaghi L, Ghidinelli M, Zambroni D, Figlia G, Chen Z-L, et al. 2007. Identification of dystroglycan as a second laminin recep-
et al. 2011. Non-redundant function of dystroglycan and E1 integrins tor in oligodendrocytes, with a role in myelination. Development
in radial sorting of axons. Development 138:4025–4037. 134:1723–1736.
De Biase LM, Kang SH, Baxi EG, Fukaya M, Pucak ML, Mishina M, Colognato H, Tzvetanova ID. 2011. Glia unglued: how signals from the
et al. 2011. NMDA receptor signaling in oligodendrocyte progenitors extracellular matrix regulate the development of myelinating glia.
is not required for oligodendrogenesis and myelination. J Neurosci Dev Neurobiol 71:924–955
31:12650–12662. Cosgaya JM, Chan JR, Shooter EM. 2002. The neurotrophin recep-
Birchmeier C, Nave KA. 2008. Neuregulin-1, a key axonal signal tor p75NTR as a positive modulator of myelination. Science
that drives Schwann cell growth and differentiation. Glia 56: 298:1245–1248.
1491–1497. Cotter L, Ozçelik M, Jacob C, Pereira JA, Locher V, Baumann R,
Bremer J, Baumann F, Tiberi C, Wessig C, Fischer H, Schwarz P, et al. et al. 2010. Dlg1-PTEN interaction regulates myelin thickness
2010. Axonal prion protein is required for peripheral myelin mainte- to prevent damaging peripheral nerve overmyelination. Science
nance. Nat Neurosci 13:310–318. 328:1415–1418.
Brinkmann BG, Agarwal A, Sereda MW, Garratt AN, Müller T, Wende Court FA, Hewitt JE, Davies K, Patton BL, Uncini A, Wrabetz L, et al.
H, et al. 2008. Neuregulin-1/ErbB signaling serves distinct functions 2009. A laminin-2, dystroglycan, utrophin axis is required for com-
in myelination of the peripheral and central nervous system. Neuron partmentalization and elongation of myelin segments. J Neurosci
59:581–595. 29:3908–3919.
Bryant MR, Marta CB, Kim FS, Bansal R. 2009. Phosphorylation and Court FA, Zambroni D, Pavoni E, Colombelli C, Baragli C, Figlia
lipid raft association of fibroblast growth factor receptor-2 in oligo- G, et al. 2011. MMP2–9 cleavage of dystroglycan alters the size
dendrocytes. Glia 57:935–946. and molecular composition of Schwann cell domains. J Neurosci
Bunge RP. 1968. Glial cells and the central myelin sheath. Physiol Rev 31:12208–12217.
48:197–251. Demerens C, Stankoff B, Logak M, Anglade P, Allinquant B, Couraud F,
Burne JF, Staple JK, Raff MC. 1996. Glial cells are increased propor- et al. 1996. Induction of myelination in the central nervous system by
tionally in transgenic optic nerves with increased numbers of axons. electrical activity. Proc Natl Acad Sci U S A 93:9887–9892.
J Neurosci 16:2064–2073. Du Y, Fischer TZ, Lee LN, Lercher LD, Dreyfus CF. 2003. Regionally
Buttery PC, ffrench-Constant C. 1999. Laminin-2/integrin interactions specific effects of BDNF on oligodendrocytes. Dev Neurosci
enhance myelin membrane formation by oligodendrocytes. Mol Cell 25:116–126.
Neurosci 14:199–212. Dubois-Dalcq M, Behar T, Hudson L, Lazzarini RA. 1986. Emergence
Calzà L, Fernandez M, Giardino L. 2010. Cellular approaches to cen- of three myelin proteins in oligodendrocytes cultured without neu-
tral nervous system remyelination stimulation: thyroid hormone to rons. J Cell Biol 102:384–392.

FAC TO R S C O N T R O L L I N G M Y E L I N F O R M AT I O N • 569
Emery B. 2010a. Regulation of oligodendrocyte differentiation and Grossmann KS, Wende H, Paul FE, Cheret C, Garratt AN, Zurborg
myelination. Science 330:779–782. S, et al. 2009. The tyrosine phosphatase Shp2 (PTPN11) directs
Emery B. 2010b. Transcriptional and post-transcriptional control of Neuregulin-1/ErbB signaling throughout Schwann cell develop-
CNS myelination. Curr Opin Neurobiol 20:601–607. ment. Proc Natl Acad Sci U S A 106:16704–16709.
Esper RM, Loeb JA. 2004. Rapid axoglial signaling mediated by neu- Gyllenstein L, Malmfors T. 1963. Myelinization of the optic nerve and
regulin and neurotrophic factors. J Neurosci 24:6218–6227. its dependence on visual function—a quantitative investigation in
Esper RM, Loeb JA. 2009. Neurotrophins induce neuregulin mice. J Embryol Exp Morphol 11:255–266.
release through protein kinase Cdelta activation. J Biol Chem Helbling-Leclerc A, Zhang X, Topaloglu H, Cruaud C, Tesson F, et al.
284:26251–26260. 1995. Mutations in the laminin alpha 2-chain gene (LAMA2) cause
Fancy SPJ, Baranzini SE, Zhao C, Yuk D-I, Irvine K-A, Kaing S, et al. 2009. merosin-deficient congenital muscular dystrophy. Nat Genet 11:216–218.
Dysregulation of the Wnt pathway inhibits timely myelination and Hildebrand C, Bowe CM, Remahl IN. 1994. Myelination and myelin
remyelination in the mammalian CNS. Genes Dev 23:1571–1585. sheath remodelling in normal and pathological PNS nerve fibres.
Feigenson K, Reid M, See J, Crenshaw EB 3rd, Grinspan JB. 2009. Wnt Prog Neurobiol 43:85–141.
signaling is sufficient to perturb oligodendrocyte maturation. Mol Höke A, Ho T, Crawford TO, LeBel C, Hilt D, Griffi n JW. 2003. Glial
Cell Neurosci 42:255–265. cell line-derived neurotrophic factor alters axon Schwann cell units
Feltri ML, Scherer SS, Nemni R, Kamholz J, Vogelbacker H, Scott MO, and promotes myelination in unmyelinated nerve fibers. J Neurosci
et al. 1994. Beta 4 integrin expression in myelinating Schwann cells 23:561–567.
is polarized, developmentally regulated and axonally dependent. Hu X, He W, Diaconu C, Tang X, Kidd GJ, Macklin WB, et al. 2008.
Development 120:1287–1301. Genetic deletion of BACE1 in mice affects remyelination of sciatic
Feltri ML, Wrabetz L. 2005. Laminins and their receptors in Schwann nerves. Faseb J 22:2970–2980.
cells and hereditary neuropathies. J Peripher Nerv Syst 10:128–143 Hu X, Hicks CW, He W, Wong P, Macklin WB, Trapp BD, et al. 2006.
Fewou SN, Ramakrishnan H, Büssow H, Gieselmann V, Eckhardt M. Bace1 modulates myelination in the central and peripheral nervous
2007. Down-regulation of polysialic acid is required for efficient system. Nat Neurosci 9:1520–1525.
myelin formation. J Biol Chem 282:16700–16711. Kao S-C, Wu H, Xie J, Chang C-P, Ranish JA, Graef IA, et al. 2009.
Fields RD. 2008. Oligodendrocytes changing the rules: action poten- Calcineurin/NFAT signaling is required for neuregulin-regulated
tials in glia and oligodendrocytes controlling action potentials. Schwann cell differentiation. Science 323:651–654.
Neuroscientist 6:540–543 Klein C, Kramer E-M, Cardine A-M, Schraven B, Brandt R, Trotter J.
Flores AI, Narayanan SP, Morse EN Shick HE, Yin X, Kidd G, et al. 2002. Process outgrowth of oligodendrocytes is promoted by inter-
2008. Constitutively active Akt induces enhanced myelination in the action of fyn kinase with the cytoskeletal protein tau. J. Neurosci.
CNS. J Neurosci 28:7174–7183. 22:698–707.
Fortin D, Rom E, Sun H, Yayon A, Bansal R. 2005. Distinct fibro- Krämer-Albers E-M, White R. 2011. From axon–glial signalling to
blast growth factor (FGF)/FGF receptor signaling pairs initiate myelination: the integrating role of oligodendroglial Fyn kinase. Cell
diverse cellular responses in the oligodendrocyte lineage. J Neurosci Mol Life Sci 68:2003–2012.
25:7470–7479. Kremer D, Aktas O, Hartung H-P, Küry P. 2011. The complex
Fortini ME. 2002. Gamma-Secretase-mediated proteolysis in world of oligodendroglial differentiation inhibitors. Ann Neurol
cell-surface-receptor signalling. Nat Rev Mol Cell Biol 3:673–684. 69:602–618.
Ffrench-Constant C, Colognato H, Franklin RJ. 2004. Neuroscience. Laursen LS, Chan CW, ffrench-Constant C. 2009. An integrin-contactin
The mysteries of myelin unwrapped. Science 304:688–689 complex regulates CNS myelination by differential Fyn phosphoryla-
Fricker FR, Lago N, Balarajah S, Tsantoulas C, Tanna S, Zhu N, et al. tion. J Neurosci 29:9174–9185.
2011. Axonally derived neuregulin-1 is required for remyelina- Lee S, Chong SYC, Leack MK, Mellon SH, Tuck SJ, Fenk ZQ, et al.
tion and regeneration after nerve injury in adulthood. J Neurosci 2012. Engineering nanofibers for oligodendrocyte myelination: fiber
31:3225–3233. diameter is sufficient to initiate wrapping. Nat Methods, in press.
Friede RL, Bischhausen R. 1982. How are sheath dimensions affected by Lee X, Yang Z, Shao Z, Rosenberg SS, Levesque M, Pepinsky RB,
axon caliber and internode length? Brain Res 235:335–350. et al. 2007. NGF regulates the expression of axonal LINGO-1 to
Furusho M, Dupree JL, Nave K-A, Bansal R. 2012. Fibroblast growth inhibit oligodendrocyte differentiation and myelination. J Neurosci
factor receptor signaling in oligodendrocytes regulates myelin sheath 27:220–225.
thickness. J Neurosci 32(19):6631–6641. Liang G, Cline GW, Macica CM. 2007. IGF-1 stimulates de novo
Galvin J, Eyermann C, Colognato H. 2010. Dystroglycan modulates the fatty acid biosynthesis by Schwann cells during myelination. Glia
ability of insulin-like growth factor-1 to promote oligodendrocyte 55:632–641.
differentiation. J Neurosci Res 88:3295–3307. Liang X, Draghi NA, Resh MD. 2004. Signaling from integrins to Fyn
Garratt AN, Voiculescu O, Topilko P, Charnay P, Birchmeier C. 2000. to Rho family GTPases regulates morphologic differentiation of oli-
A dual role of erbB2 in myelination and in expansion of the Schwann godendrocytes. J Neurosci 24:7140–7149.
cell precursor pool. J Cell Biol 148:1035–1046. Lubetzki C, Demerens C, Anglade P, Villarroya H, Frankfurter A, Lee
Genoud S, Lappe-Siefke C, Goebbels S, Radtke F, Aguet M, Scherer SS, VM, et al. 1993. Even in culture, oligodendrocytes myelinate solely
et al. 2002. Notch1 control of oligodendrocyte differentiation in the axons. Proc Natl Acad Sci U S A 90:6820–6824.
spinal cord. J Cell Biol 158:709–718. La Marca R, Cerri F, Horiuchi K, Bachi A, Feltri ML, Wrabetz L, et al.
Geren BB, Raskind J. 1953. Development of the fine structure of the 2011. TACE (ADAM17) inhibits Schwann cell myelination. Nat
myelin sheath in sciatic nerves of chick embryos. Proc Natl Acad Sci Neurosci 14:857–865.
U S A 39:880–884. Martin-Belmonte F, Perez-Moreno M. 2011. Epithelial cell polarity, stem
Givogri MI, Costa RM, Schonmann V, Silva AJ, Campagnoni AT, cells and cancer. Nat Rev Cancer 12:23–38
Bongarzone ER. 2002. Central nervous system myelination in Mason I. 2007. Initiation to end point: the multiple roles of fibro-
mice with deficient expression of Notch1 receptor. J Neurosci Res blast growth factors in neural development. Nat Rev Neurosci
67:309–320. 8:583–596.
Goebbels S, Oltrogge JH, Kemper R, Heilmann I, Bormuth I, Wolfer Maurel P, Einheber S, Galinska J, Thaker P, Lam I, Rubin MB, et al.
S, et al. 2010. Elevated phosphatidylinositol 3,4,5-trisphosphate in 2007. Nectin-like proteins mediate axon–Schwann cell interactions
glia triggers cell-autonomous membrane wrapping and myelination. along the internode and are essential for myelination. J Cell Biol
J Neurosci 30:8953–8964. 178:861–874.
Goebbels S, Oltrogge JH, Wolfer S, Wieser GL, Nientiedt T, Pieper A, Mellman I, Nelson WJ. 2008. Coordinated protein sorting, targent-
et al. 2012. Genetic disruption of Pten in a novel mouse model of ing and distribution in polarized cells. Nat Rev Mol Cell Biol
tomaculous neuropathy. EMBO Mol Med, in press. 9:833–845

570 • NEUROGLIA
Mi S, Lee X, Shao Z, Th ill G, Ji B, Relton J, et al. 2004. LINGO-1 is Previtali SC, Nodari A, Taveggia C, Pardini C, Dina G, Villa A, et al.
a component of the Nogo-66 receptor/p75 signaling complex. Nat 2003. Expression of laminin receptors in Schwann cell differentia-
Neurosci 7:221–228. tion: evidence for distinct roles. J Neurosci 23:5520–5530.
Mi S, Miller RH, Lee X, Scott ML, Shulag-Morskaya S, Shao Z, et al. Rauskolb S, Zagrebelsky M, Dreznjak A, Deogracias R, Matsumoto T,
2005. LINGO-1 negatively regulates myelination by oligodendro- Wiese S, et al. 2010. Global deprivation of brain-derived neurotrophic
cytes. Nat Neurosci 8:745–751. factor in the CNS reveals an area-specific requirement for dendritic
Michailov GV, Sereda MW, Brinkmann BG, Fischer TM, Haug B, growth. J Neurosci 30:1739–1749.
Birchmeier C, et al. 2004. Axonal neuregulin-1 regulates myelin Readhead C, Popko B, Takahashi N, Shine HD, Saavedra RA, Sidman
sheath thickness. Science 304:700–703. RL, et al. 1987. Expression of a myelin basic protein gene in trans-
Mirsky R, Winter J, Abney ER, Pruss RM, Gavrilovic J, Raff MC. 1980. genic shiverer mice: correction of the dysmyelinating phenotype. Cell
Myelin-specific proteins and glycolipids in rat Schwann cells and oli- 48:703–712.
godendrocytes in culture. J Cell Biol 84:483–494. Relucio J, Tzvetanova ID, Ao W, Lindquist S, Colognato H. 2009.
Monk KR, Naylor SG, Glenn TD, Mercurio S, Perlin JR, Dominguez Laminin alters fyn regulatory mechanisms and promotes oligoden-
C, et al. 2009. A G protein-coupled receptor is essential for Schwann drocyte development. J Neurosci 29:11794–11806.
cells to initiate myelination. Science 325:1402–1405. Remahl S, Hildebrand C. 1990. Relations between axons and oligo-
Monk KR, Oshima K, Jörs S, Heller S, Talbot WS. 2011. Gpr126 is dendroglial cells during initial myelination. II. The individual axon.
essential for peripheral nerve development and myelination in mam- J Neurocytol 19:883–898.
mals. Development 138:2673–2680. Rosenberg SS, Chan JR. 2009. Modulating myelination: knowing when
Narayanan SP, Flores AI, Wang F, Macklin WB. 2009. Akt signals to say Wnt. Genes Dev 23:1487–1493.
through the mammalian target of rapamycin pathway to regulate Rosenberg SS, Kelland EE, Tokar E, De la Torre AR, Chan JR. 2008.
CNS myelination. J Neurosci 29:6860–6870. The geometric and spatial constraints of the microenvironment
Nave K-A. 2010. Myelination and support of axonal integrity by glia. induce oligodendrocyte differentiation. Proc Natl Acad Sci U S A
Nature 468:244–252. 105:14662–14667.
Nave K-A, Salzer JL. 2006. Axonal regulation of myelination by neu- Rosenberg SS, Ng BK, Chan JR. 2006. The quest for remyelination:
regulin 1. Curr Opin Neurobiol 16:492–500. a new role for neurotrophins and their receptors. Brain Pathol
Newbern J, Birchmeier C. 2010. Nrg1/ErbB signaling networks in 16:288–294.
Schwann cell development and myelination. Semin Cell Dev Biol Russell JW, Cheng HL, Golovoy D. 2000. Insulin-like growth factor-I
21:922–928. promotes myelination of peripheral sensory axons. J Neuropathol
Newbern JM, Li X, Shoemaker SE, Zhou J, Zhong J, Wu Y, et al. Exp Neurol 59:575–584.
2011. Specific Functions for ERK/MAPK Signaling during PNS Sagane K, Hayakawa K, Kai J, Hirohashi T, Takahashi E, Miyamoto
Development. Neuron 69:91–105. N, et al. 2005. Ataxia and peripheral nerve hypomyelination in
Ni CY, Murphy MP, Golde TE, Carpenter G. 2001. gamma -Secretase ADAM22-deficient mice. BMC Neurosci 6:33.
cleavage and nuclear localization of ErbB-4 receptor tyrosine kinase. Saher G, Quinte S, Möbius W, Wehr MC, Krämer-Albers E-M, Brügger
Science 294:2179–2181. B, et al. 2009. Cholesterol regulates the endoplasmic reticulum exit of
Nodari A, Previtali SC, Dati G, Occhi S, Court FA, Colombelli C, et al. the major membrane protein P0 required for peripheral myelin com-
2008. Alpha6beta4 integrin and dystroglycan cooperate to stabilize paction. J Neurosci 29:6094–6104.
the myelin sheath. J Neurosci 28:6714–6719. Salzer JL, Bunge RP, Glaser L. 1980a. Studies of Schwann cell prolifera-
Occhi S, Zambroni D, Del Carro U, Amadio S, Sirkowski EE, Scherer tion. III. Evidence for the surface localization of the neurite mitogen.
SS, et al. 2005. Both laminin and Schwann cell dystroglycan are nec- J Cell Biol 84:767–778.
essary for proper clustering of sodium channels at nodes of Ranvier. Salzer JL, Williams AK, Glaser L, Bunge RP. 1980b. Studies of Schwann
J Neurosci 25:9418–9427. cell proliferation. II. Characterization of the stimulation and spec-
Ohno M, Hiraoka Y, Matsuoka T, Tomimoto H, Takao K, Miyakawa ificity of the response to a neurite membrane fraction. J Cell Biol
T, et al. 2009. Nardilysin regulates axonal maturation and myeli- 84:753–766.
nation in the central and peripheral nervous system. Nat Neurosci Schumacher M, Hussain R, Gago N, Oudinet J-P, Mattern C, Ghoumari
12:1506–1513. AM. 2012. Progesterone synthesis in the nervous system: implica-
Ozkaynak E, Abello G, Jaegle M, van Berge L, Hamer D, Kegel L, tions for myelination and myelin repair. Front Neurosci 6:10.
et al. 2010. Adam22 is a major neuronal receptor for Lgi4-mediated Sherman DL, Krols M, Wu L-MN, Grove M, Nave K-A, Gangloff Y-G,
Schwann cell signaling. J Neurosci 30:3857–3864. et al. 2012. Arrest of myelination and reduced axon growth when
Park HT, Feltri ML. 2011. Rac1 GTPase controls myelination and demy- Schwann cells lack mTOR. J Neurosci 32:1817–1825.
elination. Bioarchitecture 3:110–113 Simons M, Trajkovic K. 2006. Neuron-glia communication in the con-
Park J, Liu B, Chen T, Li H, Hu X, Gao J, et al. 2008. Disruption of trol of oligodendrocyte function and myelin biogenesis. J Cell Sci
Nectin-like 1 cell adhesion molecule leads to delayed axonal myelina- 119:4381–4389.
tion in the CNS. J Neurosci 28:12815–12819. Simons M, Trotter J. 2007. Wrapping it up: the cell biology of myelina-
Pedraza L, Huang JK, Colman D. 2009. Disposition of axonal caspr with tion. Curr Opin Neurobiol 17:533–540.
respect to glial cell membranes: implications for the process of myeli- Spiegel I, Adamsky K, Eshed Y, Milo R, Sabanay H, Sarig-Nadir O, et al.
nation. J Neurosci Res 87:3480–3491. 2007. A central role for Necl4 (SynCAM4) in Schwann cell-axon
Pereira JA, Lebrun-Julien F, Suter U. 2012. Molecular mechanisms regu- interaction and myelination. Nat Neurosci 10:861–869.
lating myelination in the peripheral nervous system. Trends Neurosci Steinfeld R, Grapp M, Kraetzner R, Dreha-Kulaczewski S, Helms G,
35:123–134 Dechent P, et al. 2009. Folate receptor alpha defect causes cerebral
Piaton G, Gould RM, Lubetzki C. 2010. Axon-oligodendrocyte interac- folate transport deficiency: a treatable neurodegenerative disorder
tions during developmental myelination, demyelination and repair. associated with disturbed myelin metabolism. Am J Hum Genet
J Neurochem 114:1243–1260. 85:354–363.
Popko B, Puckett C, Lai E, Shine HD, Readhead C, Takahashi N, Stevens B, Fields RD. 2000. Response of Schwann cells to action poten-
et al. 1987. Myelin deficient mice: expression of myelin basic pro- tials in development. Science 287:2267–2271.
tein and generation of mice with varying levels of myelin. Cell Stevens B, Porta S, Haak LL, Gallo V, Fields RD. 2002. Adenosine: a
48:713–721. neuron-glial transmitter promoting myelination in the CNS in
Porstmann T, Griffiths B, Chung Y-L, Delpuech O, Griffiths JR, response to action potentials. Neuron 36:855–868.
Downward J, et al. 2005. PKB/Akt induces transcription of enzymes Syed N, Reddy K, Yang DP, Taveggia C, Salzer JL, Maurel P, et al. 2010.
involved in cholesterol and fatty acid biosynthesis via activation of Soluble neuregulin-1 has bifunctional, concentration-dependent
SREBP. Oncogene 24:6465–6481. effects on Schwann cell myelination. J Neurosci 30:6122–6131.

FAC TO R S C O N T R O L L I N G M Y E L I N F O R M AT I O N • 571
Taveggia C, Feltri ML, Wrabetz L. 2010. Signals to promote myelin for- White R, Gonsior C, Krämer-Albers E-M, Stöhr N, Hüttelmaier S,
mation and repair. Nat Rev Neurol 6:276–287. Trotter J. 2008. Activation of oligodendroglial Fyn kinase enhances
Taveggia C, Zanazzi G, Petrylak A, Yano H, Rosenbluth J, Einheber S, translation of mRNAs transported in hnRNP A2–dependent RNA
et al. 2005. Neuregulin-1 type III determines the ensheathment fate granules. J Cell Biol 181:579–586.
of axons. Neuron 47:681–694. Willem M, Garratt AN, Novak B, Citron M, Kaufmann S, Rittger
Tawk M, Makoukji, Belle M, Fonte C, Trousson A, Hawkins T, et al. 2011. A, et al. 2006. Control of peripheral nerve myelination by the
Wnt/beta-catenin signaling is an essential and direct driver of myelin beta-secretase BACE1. Science 314:664–666.
gene expression and myelinogenesis. J Neurosci 31:3729–3742. Windebank AJ, Wood P, Bunge RP, Dyck PJ. 1985. Myelination
Thurnherr T, Benninger Y, Wu X, Chrostek A, Krause SM, Nave KA, determines the caliber of dorsal root ganglion neurons in culture.
et al. 2006. Cdc42 and Rac1 signaling are both required for and act J Neurosci 5:1563–1569.
synergistically in the correct formation of myelin sheaths in the CNS. Wood PM, Bunge RP. 1975. Evidence that sensory axons are mitogenic
J Neurosci 26:10110–10119. for Schwann cells. Nature 256:662–664.
Todorich B, Pasquini JM, Garcia CI, Paez PM, Connor JR. 2009. Woodhoo A, Alonso MBD, Droggiti A, Turmaine M, D’Antonio M,
Oligodendrocytes and myelination: the role of iron. Glia 57: Parkinson DB, et al. 2009. Notch controls embryonic Schwann
467–478. cell differentiation, postnatal myelination and adult plasticity. Nat
Trajkovic K, Dhaunchak AS, Goncalves JT, Wenzel D, Schneider A, Bunt Neurosci 12:839–847.
G, Nave KA, et al. Neuron to glia signaling triggers myelin membrane Xiao J, Kilpatrick TJ, Murray SS. 2009. The role of neurotrophins in the
exocytosis from endosomal storage sites. J Cell Biol 172:937–948. regulation of myelin development. Neurosignals 17:265–276.
Treiber H, Hagemeyer N, Ehrenreich H, Simons M. 2012. BACE1 Xiao J, Wong AW, Willingham MM, van den Buuse M, Kilpatrick TJ,
in central nervous system myelination revisited. Mol Psychiatry Murray SS. 2010. Brain-derived neurotrophic factor promotes cen-
17:237–239. tral nervous system myelination via a direct effect upon oligodendro-
Tyler WA, Gangoli N, Gokina P, Kim HA, Covey M, Levison SW, cytes. Neurosignals 18:186–202.
et al. 2009. Activation of the mammalian target of rapamycin Yang D, Bierman J, Tarumi YS, Zhong Y-P, Rangwala R, Proctor TM,
(mTOR) is essential for oligodendrocyte differentiation. J Neurosci et al. 2005. Coordinate control of axon defasciculation and myelina-
29:6367–6378. tion by laminin-2 and -8. J Cell Biol 168:655–666.
Velanac V, Unterbarnscheidt T, Hinrichs W, Gummert MN, Fischer Ye F, Chen Y, Hoang T, Montgomery RL, Zhao X, Bu H, et al. 2009.
TM, Rossner MJ, et al. 2012. Bace1 processing of NRG1 type III pro- HDAC1 and HDAC2 regulate oligodendrocyte differentiation by dis-
duces a myelin-inducing signal but is not essential for the stimulation rupting the beta-catenin-TCF interaction. Nat Neurosci 12:829–838.
of myelination. Glia 60:203–217. Ye P, Carson J, D’Ercole AJ. 1995. In vivo actions of insulin-like growth
Vondran MW, Clinton-Luke P, Honeywell JZ, Dreyfus CF. 2010. factor-I (IGF-I) on brain myelination: studies of IGF-I and IGF bind-
BDNF+/- mice exhibit deficits in oligodendrocyte lineage cells of the ing protein-1 (IGFBP-1) transgenic mice. J Neurosci 15: 7344–7356.
basal forebrain. Glia 58:848–856. Ye P, Li L, Richards RG, DiAugustine RP, D’Ercole AJ. 2002. Myelination
Voyvodic JT. 1989. Target size regulates calibre and myelination of sym- is altered in insulin-like growth factor-I null mutant mice. J Neurosci
pathetic axons. Nature 342:430–433. 22:6041–6051.
Wakatsuki S, Yumoto N, Komatsu K, Araki T, Sehara-Fujisawa A. 2009. Yu W-M, Feltri ML, Wrabetz L, Strickland S, Chen Z-L. 2005. Schwann
Roles of meltrin-beta/ADAM19 in progression of Schwann cell dif- cell-specific ablation of laminin gamma1 causes apoptosis and pre-
ferentiation and myelination during sciatic nerve regeneration. J Biol vents proliferation. J Neurosci 25:4463–4472.
Chem 284:2957–2966. Zampieri N, Xu C-F, Neubert TA, Chao MV. 2005. Cleavage of p75
Wake H, Lee PR, Fields RD. 2011. Control of local protein synthe- neurotrophin receptor by D-secretase and J-secretase requires specific
sis and initial events in myelination by action potentials. Science receptor domains. J Biol Chem 280:14563–14571.
333:1647–1651. Zeger M, Popken G, Zhang J, Xuan S, Lu QR, Schwab MH, et al. 2007.
Watkins TA, Emery B, Mulinyawe S, Barres BA. 2008. Distinct stages of Insulin-like growth factor type 1 receptor signaling in the cells of oli-
myelination regulated by J-secretase and astrocytes in a rapidly myeli- godendrocyte lineage is required for normal in vivo oligodendrocyte
nating CNS coculture system. Neuron 60:555–569. development and myelination. Glia 55:400–411.
Weinberg HJ, Spencer PS. 1976. Studies on the control of myelinogen- Ziskin JL, Nishiyama A, Rubio M, Fukaya M, Bergles DE. 2007. Vesicular
esis. II. Evidence for neuronal regulation of myelin production. Brain release of glutamate from unmyelinated axons in white matter.
Res 113:363–378. Nat Neurosci 10:321–330.

572 • NEUROGLIA
45.
REGULATION OF MYELINATION BY
FUNCTIONAL ACTIVIT Y
R. Douglas Fields

A B B R E VI AT I O N S of neurons, producing a quantum leap in cognitive ability of


vertebrates.
ATP adenosine triphosphate When myelin is damaged through disease or injury, loss
CNS central nervous system of sensory and motor function causes severe debilitation.
DRG dorsal root ganglion Relegation of myelin to within the context of disease was
DTI diffusion tensor imaging responsible for the longstanding concept that myelin was
ERK/MAPK extracellular signal–regulated kinase/ static insulation and irrelevant to nervous system plasticity,
mitogen-activated protein kinase except after injury. This view has changed rapidly (Fields 2005,
Fyn Fyn kinase 2010). New cellular and molecular information on modes of
GABA gamma aminobutyric acid axon–glial communication regulated by electrical activity has
GFP green fluorescent protein emerged from the development of new methods of studying
L1-CAM L1 cell adhesion molecule myelination in cell culture. Specialized magnetic resonance
LIF leukemia inhibitory factor imaging (MRI) brain imaging techniques enable visualizing
MAG myelin-associated glycoprotein microstructural changes in white matter in the human brain in
MBP myelin basic protein response to environmental experience (Fields 2011a; Zatorre
mGluR metabotropic glutamate receptor et al. 2012). Lineage tracing in transgenic animals shows that
MRI magnetic resonance imaging immature oligodendrocytes (OPCs) persist in abundance in
NCAM neural cell adhesion molecule the adult brain (Paschoula et al. 2009; Rivers et al. 2008).
NMDA N-methyl-d-aspartate Myelination is no longer regarded strictly as an early develop-
OPC oligodendrocyte progenitor cell mental process; myelination continues well into adult life.
PDGF platelet-derived growth factor Myelination is now known to be influenced by functional
PNS peripheral nervous system activity in axons. Several mechanisms have been identified for
TTX tetrodotoxin activity-dependent effects on myelination and many more are
certain to be discovered. The consequences are wide-ranging
and profound, from illuminating how environmental experi-
1 INTRODUCTION ence modifies brain development through early life; to how
cognitive abilities decline with age; to understanding a new
A compelling argument can be made that more than any other aspect of mental illnesses and developmental disorders; and
development, the evolution of myelin was responsible for the expanding the cellular mechanisms of learning beyond neuro-
success of vertebrates. The multilayered wrapping of insulation transmitter signaling at synapses.
around axons and the formation of nodes of Ranvier funda-
mentally changed the way electrical impulses are conducted
1.1 H I S TO R I C A L P E R S P EC T I VE O N MY E L I N
through nerve axons, increasing impulse propagation speed by
A N D F U N C T I O NA L AC T I VIT Y
at least 50 times faster than in unmyelinated axons (see also
chapter 42). The ability to transmit impulses rapidly over long The concept that myelination is associated with development
distances permitted increased body size, rapid sensation and of functional activity in nerve fibers originated with the earli-
movement, and the ability to concentrate neurons into a single est histological studies of the brain, but much confusion and
organ, the brain, rather than the necessity of having isolated uncertainty persisted because the true nature and function of
ganglia distributed throughout the body and situated close to myelin was mysterious from the time it was first seen in the late
the structures they control, as they are in invertebrates, which 19th century. Electron microscopy in the mid-1950s to early
lack myelin (Bullock et al. 1984). The concentration of neurons 1960s finally revealed myelin to be a multi-laminar wrapping
into one organ (cephalization) and the miniaturization that of compacted membrane around an axon (Geren 1954), which
myelin enabled by allowing extremely slender axons to con- is formed by Schwann cells in the peripheral nervous system
duct impulses as rapidly as the largest axons in invertebrates, (PNS) and oligodendrocytes in the central nervous system
enabled complex integration among many different types (CNS) (Fig. 45.1). Originally, myelin was believed to be a fatty

573
A C

Figure 45.1 Myelinating Glia A. Spindle-shaped Schwann cells form myelin in the peripheral nervous system. B. Multipolar oligodendrocytes form
myelin in the central nervous system. Individual processes of the oligodendrocyte (green) form contacts with axons (purple) to begin wrapping myelin
membrane around them. Electrical activity promotes myelination by increasing survival of oligodendrocytes, promoting differentiation of oligoden-
drocytes, and stimulating the local synthesis of myelin basic protein in oligodendrocyte processes associated with electrically active axons.
C. Cross-section of axons in optic nerve of an adult mouse showing the multilaminar compacted membranes of myelin wrapped around the axons.
(A,B) are from cell culture. Bar = 10 um (A,B), 100 nm (C).

substance secreted by the axon. In the CNS this was the most animal series, it would seem to us that myelin repre-
reasonable explanation because the slender processes of oligo- sents nothing more than a refinement of the insulating
dendrocytes forming myelin on axons were not revealed by the function assumed for so long by neuroepithelial (i.e.,
histological stains used by early anatomists. Thus, there was no ependymal) cells and glia (i.e., astrocytes). S. Ramón
evidence linking the thick white coating on axons to any cel- y Cajal Histology of the Nervous System of Man and
lular source other than the axon. In the PNS, Louis-Antoine Vertebrates, p. 206. 1995.
Ranvier suggested that myelin resided inside the Schwann cell
like a fat droplet clinging to the axon (Ranvier 1878). The func- Although the function of the nodes of Ranvier in salta-
tion of myelin as an electrical insulator arose naturally with the tory conduction was suggested by Lillie in 1925 and shown
understanding that communication in the nervous system is experimentally by Tasaki in 1939, the function in saltatory
electrical, but it was also clear that conduction could proceed conduction was not universally appreciated. “The function of
perfectly well in the absence of myelin, as many fibers in the the nodes [of Ranvier] remains obscure,” wrote JZ Young in an
brain and peripheral nerves are unmyelinated. Thus myelin article published in Nature in 1944. “It may be that they serve
was not regarded as essential or as having any unique prop- to prevent movement of the fluid within the fibres. Fibres of
erties in serving as an electrical insulator, as expressed by the the central nervous system are usually without nodes, and lack
renowned neuroanatomist Ramón y Cajal in the early twenti- the definite tubes which are present in peripheral nerve, no
eth century: doubt to meet the stresses imposed by movement” (Young
1944, p. 522).
Myelin does not enjoy a monopoly on insulation, even This uncertainty accounts for the slow progress in under-
if one acknowledges that it does indeed insulate, which standing how functional activity could affect myelination.
has not yet been demonstrated conclusively. Because Synapses are understandably the focus of studies on functional
axons in invertebrates, the autonomic system of ver- plasticity in the nervous system. Nevertheless, there has always
tebrates, the olfactory nerve, and most parts of the been considerable interest in studying myelination in correla-
cerebrum, cerebellum, and optic lobe of fish, reptiles, tion with development of various nervous system functions,
and amphibians lack myelin sheaths entirely, their because unlike synapses, which were invisible through light
only protection against the lateral escape of current, or microscopes, myelination of fibers was readily apparent to his-
short circuit, appears to be a glial plexus [of astrocytes] tologists. Although the correlation between development of
between fibers . . . because of its late appearance in the functional activity in fibers with the onset of myelination was

574 • NEUROGLIA
grasped by most early anatomists, the correlation was usually 2 C O N T R O VE R SY A N D E A R LY E VI D E N C E
perceived as providing an index of neuronal maturity, not that O N T H E R O L E O F I M P U L S E AC T I VI T Y
myelination was necessarily stimulated by electrical activity in O N M Y E L I N AT I O N
the axon.
Paul Flechsig (1876), first observed that tracts become Experimental studies on the effects of functional activity on
myelinated in a definite sequence so that fibers belonging to myelination have yielded a confusing and contradictory set
related functional systems become myelinated at the same time of data. Environmental experience during the period when
(see Langworthy 1932). Birth was seen as a great stimulus to myelination is taking place has been shown to affect oligoden-
myelination in human development. “Probably it is the result drocyte development and myelination (Fields 2008; Zalc and
of increased stimulation of the sensory organs” (Langworthy Fields 2000). The number of oligodendrocytes increases 27%
1932, p. 1379). The vestibular portion of the acoustic nerve to 33% in the visual cortex of rats raised in environments that
is myelinated early in development, but the olfactory nerve is are enriched by additional play objects and social interaction
unmyelinated at birth and the optic nerve is just beginning to (Bennett et al. 1964; Sirevaag and Greenough 1987; Szeligo
myelinate at birth. Motor roots are myelinated before sensory and Leblond 1997). Enriched environments increase the num-
roots, and sensory pathways begin to myelinate at the time ber of myelinated axons in the corpus callosum connecting the
when sensory endings become functional. Sensory endings two cerebral hemispheres of rats ( Juraska and Kopocik 1988;
in the skin, muscle, and vestibular and cochlear organs would Markham and Greenough 2004), and the corpus callosum
be stimulated in utero, but optic and olfactory nerves are not increases in size in rhesus monkeys raised in enriched envi-
strongly activated until after birth. ronments in parallel with improved performance in cognitive
The suggestion that axon pathways become myelinated tests (Sanchez et al. 1998).
when they become functional was also provided by animal Sensory input to the visual system has been used to address
studies. The right eyelid of the rabbit opens slightly earlier activity-dependent effects on myelination in several studies,
than the left and myelination begins in the retina of the right but results differ among studies. Mice reared in dark develop
eye of the rabbit before the left eye (Narang 1977), suggest- fewer myelinated axons in the optic nerve compared with nor-
ing that the onset of vision stimulates myelination of visual mally reared mice (Gyllensten and Malmfors 1964), and pre-
circuits. In the minority of cases in which the left eye of rab- mature eye opening increases the level of myelin protein in the
bits opened first, that eye developed more myelinated fibers. optic nerve of rabbit (Tauber et al. 1980).
Similar asymmetry in myelination was seen in the optic These findings from experimental manipulation of visual
nerves. experience are consistent with effects of disrupting impulse
Typically myelination begins after the axon has reached its activity by severing axons. There is marked reduction in myeli-
appropriate target, formed synapses, and begun firing action nation of the chick optic tectum following deafferentation
potentials, even though the glial cells have been closely associ- (Bondy and Madsen 1972), and sciatic neurectomy in the rat
ated with the axons and fully matured from much earlier in inhibits myelination in the spinal cord (Kingsley et al. 1970),
development. This suggests that myelination is influenced by suggesting myelination is related to physiological activity. The
axonal factors, not simply determined by maturation of oli- axons are also slightly smaller in diameter after deafferenta-
godendrocyte progenitor cells to a mature myelinating stage. tion, however, which could in turn affect myelination, because
Migration and differentiation of oligodendrocytes in the optic there is a minimum threshold of axon diameter for myelina-
nerve of rat and most species progresses in a gradient from the tion, and the thickness of the myelin sheath is proportional to
brain (optic chiasm) to the retina, but myelination in most spe- axon diameter.
cies proceeds in the reverse manner, from the cell body (near In contrast, other studies have reported that intraocu-
the retina) to the distal axon (chiasma). In human cerebral lar injections of the sodium channel blocker tetrodotoxin
white matter, oligodendrocytes are present in some regions (TTX), which blocks action potential activity, had no effect
for at least 3 months before the cells commit to myelino- on the number of myelinated fibers or the time of myelination
genesis, supporting dissociation between events regulating onset in optic nerves of rat (Colello and Pott 1997; Colello
oligodendrocyte maturation and their commitment to et al. 1995; Crespo et al. 1995). In experiments on goldfish,
myelinogenesis (Back et al. 2002). action potential blockade by intraocular injection of TTX
Myelination of appropriate brain regions coincides with during optic nerve regeneration had no effect on myelination
the development of specific behaviors and cognitive func- of regenerated fibers (Hayes and Meyer 1989). Dark-rearing
tions (Mabbott et al. 2006; Nagy et al. 2004; Yakolev and of kittens (Moor et al. 1976) and rats (Fukui et al. 1991)
Lecours 1967), such as reading (Kraft et al. 1980), develop- has been reported to have no effect on initiation of myeli-
ment of vocabulary (Pujol et al. 2006) and proficiency in nation of the visual pathway during early postnatal develop-
executive decision making (Giedd 2004; Liston et al. 2006). ment. Mouse spinal cord explants in vitro undergo normal
Incomplete myelination of the forebrain until the early twen- oligodendrocyte development and myelination in the pres-
ties has been suggested as a neurological basis for weaker ence of TTX (Shrager and Novakovic 1995); however,
decision-making skills in adolescence (Beckman 2004). blocking potassium channels with tetraethylammonium ion
Whether these correlations reflect developmental limitations eliminated myelination in these experiments, suggesting pos-
on behavior or behavioral influences on development is the sible involvement of ion channels in axons or oligodendro-
intriguing question. cytes in myelination.

R E GU L AT I O N O F M Y E L I N AT I O N BY F U N C T I O N A L AC T I VI T Y • 575
In apparent contradiction to the studies using TTX in spi- treatments were given at narrowly defined points in develop-
nal cord explant cultures, in which no effects on myelination ment. These results suggest that electrical activity affects the
were evident, dissociated cultures of embryonic mouse brain myelination process only within a narrow time frame, and
hemispheres treated 2 to 4 days with TTX starting 5 to 7 days around the time of eye opening when electrical activity in reti-
before myelination normally begins, decreased the number nal ganglion cells changes from a transient to a repetitive pat-
of myelinated fibers by 80% at 18 to 21 days in vitro (DIV) tern of firing (Rorig and Grantyn 1993; Skaliora et al. 1993).
(Demerens et al. 1996). The number of oligodendrocytes and Much of this apparent discrepancy in different studies,
neurons was not affected. Results consistent with this were and the sharp developmental windows for the effects, may be
also obtained after TTX injection into the eye to block action explained in part by an expectation that functional activity in
potentials in the optic nerve, and increasing action potential axons could affect multiple processes involved in oligoden-
firing with the sodium channel activator alpha-scorpion toxin drocyte development and myelination (Fig. 45.2). This could
increased myelination by 2.4 times without affecting the num- include regulating cell proliferation, survival, migration, dif-
ber of oligodendrocytes (Demerens et al. 1996). However, in ferentiation, and myelination. Depending on when functional
both in vitro and in vivo studies the effects were seen only when activity is modified in different experimental studies and when

A Early
development Cell survival?
Cell proliferation?
Cell migration?
OPC

Axon

B Differentiation Adenosine
Promotes differentiation of
OPCs to myelinating
ATP oligodendrocytes

ATP

Acts on promyelinating
oligodendrocytes to increase
LIF myelination
Astrocyte

C Myelination
Regulation of cell adhesion Formation of cholesterol-rich
molecules on axons signalling domain
Stimulates local translation
of MBP
CAMs

Oligodendrocyte

Glu

Myelinated axon

Figure 45.2 Electrical Activity in Axons Can Affect Multiple Steps in the Myelination Process. A. Activity-dependent release of growth factors, ions,
other diffusible cell–cell signaling molecules, or changes in cell adhesion molecules on the surface of axons can affect cell survival, proliferation, and
migration of myelinating glia. B. Differentiation of oligodendrocyte progenitor cells and myelination by promyelinating oligodendrocytes can be
stimulated by release of diffusible substances, including neurotransmitters (adenosine triphosphate and adenosine) released from axons, and cytokines
released from astrocytes responding to the neurotransmitter ATP liberated from electrically active axons. C. Initiation of myelination can be affected
by activity-dependent regulation of cell adhesion molecules on axons, and the vesicular release of glutamate from axons firing action potentials.

576 • NEUROGLIA
in development the results are evaluated, myelination could be diffusion imaging often shows increased resistance to diffu-
differentially affected or unaffected by the confounding pro- sion radially from axon tracts, which would be consistent with
cesses that could be involved. In addition, the effects on oligo- an increase in myelin. Rats trained in the Morris water maze
dendrocytes and myelination could be secondary to a primary showed changes in diffusivity or anisotropy in several brain
effect on neurons. A major confound is that oligodendrocytes regions, including the corpus callosum (Blumenfeld-Katzir
and their precursors have many neurotransmitter receptors et al. 2011). Histological analysis confirmed that the increased
and ion channels that could be directly affected by experi- fractional anisotropy was associated with increased staining
ments using neurotoxins applied with the intention of modu- for myelin basic protein, suggesting that the learning task had
lating electrical activity in axons. Moreover, both positive and stimulated myelination.
negative factors may participate in controlling myelination, Independent evidence is supportive of brain imaging
and the pattern of impulse activity may be an important cri- data, suggesting that white matter structure can change.
terion that would not be reproduced by using neurotoxins or Oligodendrocyte progenitor cells remain resident in substan-
pharmacological treatments. These problems were eliminated tial numbers in the adult brain (Psachoulia et al. 2009). These
by cell culture studies in dishes equipped with electrodes for cells could participate in repair after myelin damage, but they
electrical stimulation of axons in coculture with Schwann cells could in theory participate in learning-dependent changes in
or oligodendrocytes, as described in section 4. white matter if myelination of unmyelinated axons is stimu-
lated by functional activity. Internodal lengths decrease in
visual cortex of rhesus monkeys during normal aging (Peters
2.1 HUM A N B R A I N I M AG I N G O F WH IT E
et al. 2008), suggesting active remyelination throughout life.
M AT T E R P L A S T I C I T Y
Changes in physical activity, such as bed rest or outer space
Magnetic resonance imaging, and in particular diffusion ten- missions, temporarily reduce conduction velocities (Ruegg
sor imaging (DTI), which is highly sensitive to microstructural et al. 2003), and hind-limb unweighting to reduce activity in
changes in brain tissue, has revealed many examples of differ- muscle fibers causes activity-dependent changes in myelin in
ences in white matter structure that correlate with specific cog- the PNS of adult rat (Canu et al. 2009).
nitive abilities (Fields 2011a). This includes difference in white
matter structure associated with differences in performance
within the normal range of variation, not simply dysfunction 3 AC T I VI T Y-D E P E N D E N T AXO N – G L I A L
resulting from white matter defects. Individual differences in C O M MU N I C AT I O N
normal cognitive development (Casey et al. 2000; Liston et al.
2006; Walhovd and Fjell 2007), IQ (Miller 1994; Schmithorst A broad range of signaling molecules and transcription factors
et al. 2005), normal variation in reading skill (Gold et al. 2007; are involved in regulating glial development and myelination,
Klingberg et al. 2000; Niogi and McCandliss 2006), work- many of which could be influenced by electrical activity in
ing memory (Nestor et al. 2007), and musical proficiency axons. Glia express a wide variety of neurotransmitter-gated and
(Bengtsson et al. 2005; Hyde et al. 2006) are correlated with voltage-sensitive ion channels (Karadottir et al. 2005; Ransom
differences in white matter structure in specific brain regions and Orkand 1996; Sontheimer et al. 1996). Modulation of ion
mediating these tasks. channel activity in oligodendrocytes can influence their pro-
Recently, multiple MRI studies have revealed experience- liferation (Chiu and Wilson, 1989; Gallo et al. 1996; Knutson
or learning-dependent changes in white matter in the human et al. 1997; MacFarlane and Sontheimer 1997; Pappas et al.
brain. These studies indicate changes in white matter struc- 1994). Inhibition of glial potassium channel function by tet-
ture, rather than differences in structure that could predispose raethylammonium ion eliminated myelination in spinal cord
proficiency in certain cognitive tasks. Complex skills, such as explants (Shrager and Novakovic 1995). Blocking impulse
playing the piano, are accompanied by increased organization activity in optic nerve using TTX can reduce the proliferation
of white matter structure in appropriate brain tracts involved rate of oligodendrocyte progenitor cells, but exposure to the
in musical performance (Bengtsson et al. 2005). Significantly, mitogen PDGF (platelet-derived growth factor) can restore
the level of white matter structure increased proportionately the cell numbers to normal (Barres and Raff 1993).
to the number of hours each subject had practiced the instru-
ment, indicating white matter changes in acquiring the skill
3.1 VE S I CU L A R A N D N O N VE S I CU L A R R E L E A S E
rather than performance being predetermined by a limitation
O F N EU ROT R A NS M IT T E R FRO M AXO N S
on white matter development. Learning to read (Carreiras
et al. 2009), juggle (Scholz et al. 2009), and many other skills It is not immediately obvious how myelinating glia, far
(Fields 2011a), alters white matter structure in the human removed from synaptic endings, could monitor functional
brain. activity in axons. However, vesicular release of neurotransmit-
Magnetic resonance imaging does not provide cellular reso- ters from axons can occur at focal regions and from ectopic
lution, however, and the effects seen by MRI could result from sites along the axon (Fields 2011b; Kriegler and Chiu 1993;
various cellular changes, including effects on axon branching, Wake et al. 2011). In addition, synapses can form transiently on
sprouting, crossing, or diameter, and responses in astrocytes, some OPCs in white matter (DeBase et al. 2010; Kukley et al.
neurons, or vascular tissue, in addition to the possibility of 2007; Ziskin et al. 2007). Both glutamatergic and GABAergic
effects on myelination (Zatorre et al. 2012). In some studies, currents have been recorded in OPCs (see also chapter 21).

R E GU L AT I O N O F M Y E L I N AT I O N BY F U N C T I O N A L AC T I VI T Y • 577
Neurotransmitters also can be released from axons in an potentials. L1-CAM is downregulated by 0.1-Hz stimulation
activity-dependent manner in the absence of functional syn- for 5 days, but not by 0.3- or 1-Hz stimulation in the same
apses and SNARE-dependent release of synaptic vesicles experiments (Itoh et al. 1997). Neural cell adhesion molecule
(Fields and Ni 2010). Activity-dependent release of the neu- (NCAM) was not downregulated by any stimulus frequency
rotransmitter ATP from axons has been shown to take place tested, and N-cadherin was downregulated in proportion to
through volume-regulated anion channels in axons that stimulus frequency between 0.1 and 10 Hz.
become activated by trains of action potentials (Fields and Ni L1-CAM is known to be essential for the initial wrapping
2010), and these channels are permeable to small amino acids, of Schwann cell membrane around the axon, after which the
including glutamate. Inhibiting the activation of these chan- expression of L1-CAM decreases in both the axon and Schwann
nels disrupts calcium responses in astrocytes seen in response cell (Seilheimer et al. 1989; Wood et al. 1990). After stimulat-
to action potential stimulation in axons. Thus, myelinating ing neurons at different frequencies for 5 days, Schwann cells
glia along axons can have access to activity-dependent release in coculture were stimulated to form myelin by addition of
of neurotransmitters through nonvesicular release from axons ascorbic acid. One-third the normal number of myelin seg-
as well as vesicular release. ments formed on axons stimulated at 0.1 Hz compared with
unstimulated controls. Stimulation at other frequencies had
no effect on myelination in these experiments, and myelina-
3.2 OT H E R P O S S I B L E M E C H A N I S M S O F AXO N
tion was not reduced when the activity-dependent downregu-
C O M MU N I C AT I O N WI T H S C H WA N N C E L L S
lation in L1-CAM mRNA was prevented by addition of NGF
A N D O L I G O D E N D RO C Y T E S
during stimulation. No effects on the number of Schwann
Myelin formation requires cell recognition to myelinate the cells were detected in these experiments, consistent with the
appropriate axons, the formation of adhesive contacts, elabo- involvement of L1-CAM in the initial stages of myelination.
ration of vast areas of cell membrane to form myelin sheets, In this case, 0.1 Hz electrical impulse activity acts as a negative
wrapping many layers of membrane around axons, and the factor on myelin initiation.
removal of cytoplasm between the wraps of myelin to form Myelination in the CNS is known to be influenced by
compact stacks of lipid membrane, all of which might be L1-CAM (Barbin et al. 2004), but whether this cell adhe-
influenced by signaling from electrical activity in axons. sion molecule contributes to activity-dependent regulation
A large number of signaling molecules are involved in regu- of myelination in the CNS has not been reported. Electrical
lating development of myelinating glia and myelination, stimulation of mixed cortical cultures of oligodendrocytes
including growth factors, cytokines, ions (K+, pH, Ca2+), neu- and neurons increases myelination through an unknown
rotransmitters, cell adhesion, and extracellular matrix mol- mechanism increasing oligodendrocyte survival (Gary et al.
ecules, for reviews see (Emery 2010; Nave 2010; Taveggia et al. 2012). The effects depended on the frequency of electrical
2010), but few of these have been investigated in the context stimulation and require contact between axons and OPCs,
of activity-dependent regulation of myelination. suggesting possible activity-dependent regulation of unidenti-
fied cell adhesion molecules on axons that promote survival of
oligodendrocytes.
4 C E L LU L A R A N D M O L E C U L A R
M E C H A N I S M S O F AC T I VI T Y-D E P E N D E N T
4.2 D I FF US I B L E S I G NA L I N G MO L ECU L E S I N
M Y E L I N AT I O N
AC T I VIT Y-D E P E N D E N T MY E L I NAT I O N
Five cellular–molecular mechanisms for activity-dependent Both glutamate and ATP released from axons have been
regulation of myelination have been identified; all of them shown to regulate myelination in an activity-dependent man-
from studies in cell cultures in which electrodes were used to ner. Adenosine triphosphate released from axons firing action
stimulate axons in specific patterns. These mechanisms can potentials acts on Schwann cells to inhibit their proliferation
be categorized as involving: (1) cell surface molecules, (2) and development to a myelinating stage (Stevens and Fields
diffusible signals from axons, and (3) signals from astrocytes 2000). Adenosine, generated by dephosphorylation of ATP
responding to action potentials in axons. released from axons firing action potentials, inhibits prolif-
eration of Schwann cells via a different intracellular signaling
pathway involving ERK/MAPK activation through stimula-
4.1 C E L L SU R FAC E MO L ECU L E S I N
tion of cAMP-linked A2A adenosine receptors, but unlike
AC T I VI T Y-D E P E N D E N T MY E L I NAT I O N
ATP, adenosine does not inhibit differentiation of Schwann
The first mechanism to be identified for effects of impulse cells (Stevens et al. 2004).
activity on myelination involved regulation of a cell adhe- In the CNS, adenosine generated by electrically active
sion molecule on axons (Stevens et al. 1998). Neural impulse axons stimulates myelination by inhibiting OPC prolifera-
activity was found to lower L1-CAM expression on dorsal tion and stimulating differentiation. Nonhydrolyzable P2
root ganglion (DRG) axons in cell culture (Itoh et al. 1995). receptor agonist 2MeS-ATP is not effective (Stevens et al.
Interestingly, specific frequencies of stimulation were required 2002), indicating involvement of P1 adenosine receptors in
to downregulate the gene, and other cell adhesion molecules the activity-dependent increase in myelination (Stevens et al.
showed different responses to different frequencies of action 2002).

578 • NEUROGLIA
Initiation of myelination is regulated by axonal signals Even though the newly synthesized myelin basic protein
beyond those that affect cell proliferation or differentiation. is generated locally in the OCP cell process in contact with
Experiments using botulinum toxin to block vesicular release electrically active axons, if the myelin protein were free to
from axons show that electrical stimulation promotes differ- migrate within the lipid membrane, the effects of electrical
entiation of OPCs to a myelinating stage, even in the absence activity would not be confined specifically to the axon firing
of SNARE-dependent vesicular release from axons (Wake et action potentials. Studies using photoactivated GFP-labeled
al. 2011). However, myelination is strongly inhibited on axons myelin basic protein show that the diffusion of MBP in the
in which vesicular release has been blocked by prior treatment OPC membrane becomes highly restricted to discrete regions
with botulinum or tetanus toxin, indicating that vesicular adjacent to axons firing action potentials unless vesicular
release of substances from axons promotes myelination in a release is blocked by botulinum toxin. This suggests that vesic-
manner distinct from signals promoting differentiation of ular release of glutamate from axons promotes formation of
OPCs to a myelinating stage (Fig. 45.3). Calcium imaging a cytoskeletal membrane complex in oligodendrocytes that
shows that nonvesicular release of ATP from axons induces restrict MBP to appropriate membrane regions adjacent to
calcium responses in the cell soma of OPCs, but responses the axon being myelinated.
in the slender cell processes of OPCs closely associated with
axons are inhibited when vesicular release from axons is
4.3 A S T RO C Y T E S I N AC T I VIT Y-D E P E N D E N T
blocked by botulinum toxin (Wake et al. 2011). This is consis-
MY E L I NAT I O N
tent with vesicular release of glutamate from axons controlling
initiation of myelination locally in individual cell processes of The fourth mechanism known for activity-dependent myeli-
oligodendrocytes. nation involves activation of astrocytes by ATP released from
Glutamate release from synaptic vesicles along axons of electrically active axons (Ishibashi et al. 2006). This stimu-
mouse DRG neurons in culture promotes myelin induction lates release of the cytokine LIF from astrocytes, which acts
by stimulating the formation of cholesterol-rich signaling to promote myelination by mature oligodendrocytes. This
domains between oligodendrocytes and axons, and increasing activity-dependent pathway acts on mature oligodendrocytes,
the local synthesis of the major protein in the myelin sheath, in contrast with the effects of adenosine, which inhibits pro-
myelin basic protein, through Fyn kinase-dependent signal- liferation at the progenitor stage and promotes differentiation
ing (Wake et al. 2011) (Fig. 45.4). This axon–oligodendrocyte (Stevens et al. 2002).
signaling would promote myelination of electrically active
axons to regulate neural development and function accord-
ing to environmental experience. Pharmacological studies 5 SIGNIFICANCE OF
show that these effects depend on activation of mGluR and AC T I VI T Y-D E P E N D E N T M Y E L I N AT I O N
NMDA receptors on OPCs, but not AMPA or purinergic
receptors. Block of SNARE-dependent vesicular release from In theory, activity-dependent effects on myelination
axons could also impede activity-dependent delivery of other would have profound effects on information process-
proteins to the axonal membrane that may be involved in ing in the nervous system, but this is only beginning to be
myelination. explored. Environmental experience, physical therapy, and

A B

Figure 45.3 Vesicular Release from Axons Promotes Myelination. A. Compact myelin (green) forms on mouse dorsal root ganglion axons (purple)
stimulated to fire action potentials in cell culture. B. When oligodendrocyte progenitor cells are plated onto axons that have been previously treated
with botulinum toxin to inhibit vesicular release from axons, electrical stimulation promotes differentiation of oligodendrocytes, but the formation of
myelin is greatly inhibited. Myelin basic protein (green), neurofilament (purple), nuclei (blue). Adapted from Wake et al. 2011.

R E GU L AT I O N O F M Y E L I N AT I O N BY F U N C T I O N A L AC T I VI T Y • 579
GluR +
axon

lam
+

ini
n
L1
integrin
F3

NMDA

mGluR

M
+ +

BP
cholesterol rich micro domain Fyn
P P translation

M
A2

+ BP

AAAAAA
mic

A2 MBP mRNA
rot
ubl
e

Figure 45.4 Vesicular release of glutamate from axons firing action potentials stimulates the formation of cholesterol-rich signaling domains between oligo-
dendrocytes and axons, and induces local protein synthesis of myelin basic protein from mRNA in OPC cell processes in contact with electrically active axons.
This signaling is dependent upon NMDA and mGluR receptor activation and Fyn kinase (Fyn) intracellular signaling. From Wake et al. 2011.

electrical stimulation can promote recovery from injury, and change is only 2 to 4 milliseconds in duration. Thus, axonal
activity-dependent myelination could contribute. Electrical conduction time is a critical variable in information pro-
stimulation in vivo has been used to promote recovery from cessing and synaptic function. The human corpus callosum
spinal cord injury by increasing the pool of oligodendrocytes is largely unmyelinated at birth, for example, and in adults
in adult rats (Becker et al. 2010), for example. Myelination of 30% of the fibers remain unmyelinated. The conduction time
regenerated axons is required to restore function. between the left and right hemispheres is 30 milliseconds
through myelinated callosal fibers and 150 to 300 millisec-
onds through unmyelinated fibers (Swadlow et al. 1978).
5.1 EFFEC T O F MY EL I N P L A S T I C I T Y O N
Synaptic integration is affected profoundly by whether an
I N F O R M AT I O N P RO C E S S I N G
intercallosal axon becomes myelinated or remains unmyeli-
Activity-dependent myelination could contribute to nervous nated. Considering the many variables affecting conduction
system plasticity by optimizing the speed of impulse conduc- delays, genetic instruction alone would seem inadequate to
tion. The speed and synchrony of impulse traffic between specify the optimal conduction velocity in every axon, sug-
distant cortical regions is critical for optimal mental perfor- gesting the possibility of regulating impulse conduction
mance and learning, and myelin is one of the most important velocity and myelin by functional activity. Consistent with
factors affecting impulse conduction velocity. Millisecond this theory, conduction velocity varies widely among differ-
precision is necessary for the coincident arrival and summa- ent axons (over 100-fold) and many axons are slowly con-
tion of synaptic signals, because the postsynaptic potential ducting and ummyelinated.

580 • NEUROGLIA
Rushton (1951) noted that along peripheral nerve trunks, children suffering severe childhood neglect have a 17% reduc-
myelin thickness, and intermodal distance are adjusted so as tion in corpus callosum area (Teicher et al. 2004). A recent
to maximize conduction velocity and thereby optimize func- study shows that environmental influences during the period
tion. Interestingly, many cases are known in which myelin when the corpus callosum is undergoing myelination can have
is structured not to maximize the speed of conduction of lasting effects on white matter structure and behavior. In this
action potentials, but instead to ensure simultaneous arrival MRI study, individuals who reported experiencing verbal
of impulses from multiple points that are separated by large abuse as middle school children had abnormal white mat-
differences in distance (Waxman 1997). The olivocerebel- ter development in the corpus callosum and they exhibited
lar projections carrying information to Purkinje cells in the increased psychiatric problems as adults (Teicher et al. 2010).
cerebellum (Sugihara et al. 1993), the electromotor axons of
certain electric fish (Waxman et al. 1972), and the axons of
5.3 WH IT E M AT T E R P L A S T I C IT Y A N D
retinal ganglion cells located at different eccentricities within
P SYC H I AT R I C I L L N E S S
the retina (Stanford 1987) show differences in conduction
times that are adjusted by the structure of myelin to provide Although synaptic dysfunction is the cellular basis for most
simultaneous arrival of impulses within millisecond preci- mental illnesses, disruptions in functional connectivity between
sion. There is an optimal spacing of nodes of Ranvier in the distant brain regions can impair information processing in
auditory brain stem circuits of the barn owl to adjust conduc- association with a range of neurological processes. Defects
tion velocities appropriately to detect interaural time differ- in myelin insulation can lead to impaired cognitive function
ences (Carr and Konishi 1990). These examples demonstrate in 40% of multiple sclerosis patients, for example (Kujala et al.
that the relationship between axons and myelinating glia are 1997). Cognitive decline in aging also parallels subtle changes
in some way regulated to meet the precise functional require- in the integrity of white matter (Gootjes et al. 2004). This sug-
ments of individual axons. gests that impaired cognitive ability, disorganized thinking,
Long-term electrophysiological recording shows that con- mood disorders or hallucinations, and accompanying psychiat-
duction velocity through axons can change. The conduction ric illness might result from slowed or desynchronized impulse
velocity of one-third of callosal axons changes (increases or conduction between distant cortical regions.
decreases) over 1 year of chronic recordings in rabbit (Swadlow A diverse range of psychiatric disorders are accompa-
1985), and conduction velocity and neural synchrony increase nied by changes in white matter structure evident by MRI
in auditory circuits 1 year after congenitally deaf children or abnormalities in myelin genes. Polymorphisms for sev-
receive cochlear implants to restore hearing (Gordon et al. eral myelin genes have emerged as unexpected risk factors
2003). One of the largest categories of genes in which expres- for schizophrenia (Hakak et al. 2001; Kubicki et al. 2005;
sion changes during sleep are genes that control oligodendro- Tkachev et al. 2003), depression (Tkachev et al. 2003),
cyte development and myelination (Cirelli et al. 2004). The and obsessive-compulsive disorder (Stewart et al. 2007).
reason for this is unclear, but sleep is linked to consolidat- Post-mortem examination of brain tissue from patients suf-
ing memory. As the human body grows, somatosensory and fering schizophrenia (Georgieva et al. 2006; Steingard et al.
motor conduction delays in CNS pathways remain constant 2002; Tkachev et al. 2003), major depression (Aston et al.
after 2 years of age despite substantial increases in axon length 2005), and bipolar disorder (Tkachev et al. 2003) reveals
with body growth, reflecting a compensatory increase in con- reduced abundance of several mRNA transcripts of myelin
duction velocity in CNS axons (Eyre et al. 1991). Interestingly, genes or genes regulating differentiation and survival of
this compensation does not occur in the peripheral nervous myelin-forming cells. Disrupting Neuregulin 1 signaling in
system, and conduction delays increase in proportion to the transgenic mice that express a dominant negative erbB recep-
length of a growing limb. Conduction delays in the PNS are tor in oligodendrocytes, results in white matter defects and
relatively minor relative to the time required for integration in behavioral changes resembling schizophrenia in humans (Roy
the CNS, perhaps lessening the need for activity-dependent et al. 2007).
regulation of PNS myelination.

6 FUTURE DIRECTIONS
5.2 E A R LY C H I L D H O O D E X P E R I E N C E
The human brain continues to undergo myelination until at The cellular mechanisms that have been identified thus far
least the third decade of age, and the frontal regions of the for activity-dependent myelination all affect myelination
cerebral cortex, which carry out higher-level executive func- of unmyelinated axons, but it is unclear whether functional
tions, are the last to become myelinated. This prolonged activity can influence myelin or nodal structure once formed.
period of postnatal brain development may allow myelination Because many axons remain unmyelinated throughout life, it
to be influenced by the specific environment that an indi- is possible that functional activity may promote myelination of
vidual experiences during rearing rather than being specified unmyelinated axons throughout life, but this is not adequately
exclusively by genetic instruction. Early experience increases verified experimentally. Histological analysis after brain imag-
white mater structure in the internal capsule and frontal ing in experiments studying environmentally induced white
lobes in newborn human infants in parallel with improved matter changes should provide answers to these critical ques-
performance in behavioral tests (Als et al. 2007). Conversely, tions in forthcoming years.

R E GU L AT I O N O F M Y E L I N AT I O N BY F U N C T I O N A L AC T I VI T Y • 581
In theory there are many structural modifications of mye- 7 S U M M A RY A N D P E R S P E C T I VE S
linated fibers that could affect impulse conduction, and there
is some circumstantial evidence in support of this possibility. White matter is essential for proper impulse conduction, and
Myelin can influence conduction velocity by regulating axon connectivity between distant cortical regions with precise
diameter, thickness of the myelin sheath, the number and timing of transmission is critical for higher-level cognitive
spacing of nodes of Ranvier, and nodal structure and molec- functions. The new evidence showing white matter plas-
ular composition of ion channels in the node and paranodal ticity in response to electrical activity widens the scope of
regions. investigation into learning and functional plasticity beyond
Large-caliber fibers conduct impulses at higher speeds the synapse to include the transmission of information through
because the resistance to electrical current is reduced as the cal- neural networks. Changes in white matter have been seen
iber of the fibers increases. However, myelin can also regulate in humans in response to environmental experience and
axon diameter (Sanchez et al. 1996). This is obvious at nodes learning, and extrapolating from cellular and animal stud-
of Ranvier, in which the axon often shows marked changes in ies, myelination could have a role. White matter differences
diameter in the unmyelinated nodal regions. Axon diameter that correlate with the scoring on intelligence quotient
is reduced in myelin-deficient mutants (Cole et al. 1994), and tests (Schmithorst et al. 2005) and certain psychiatric
signaling from myelin proteins, such as myelin-associated conditions (Fields 2008) may be attributed in part to a direct
glycoprotein (MAG) has been implicated in the signaling role for white matter plasticity in learning and cognitive
cascade controlling neurofilament phosphorylation which function.
in turn affects axon caliber and axoplasmic transport (Hsieh
et al. 1994; Lunn et al. 2002; Yin et al. 1998).
The thickness of the myelin sheath can have a dominant AC K N OW L E D G M E N T S
influence on conduction velocity. Myelin sheaths thicker or
thinner than the theoretical optimal g-ratio of 0.65 will reduce This work is supported by funds for intramural research from
conduction velocity (Rushton 1951). (The g-ratio is the ratio NICHD.
of axon diameter divided by total fiber diameter, including the
myelin sheath.) There is, however, large variation in g-ratio
around this optimal mean among different myelinated fibers. REFERENCES
Even along the same fiber the conduction velocity is not always
constant along the length of an axon (Baker et al. 1990; Traub Als H, et al. 2007. Early experience alters brain function and structure.
and Mendell 1988). Pediatrics 113, 846–857.
Aston C, et al. 2005. Transcriptional profi ling reveals evidence for
The number of wraps of myelin increases as animals grow signaling and oligodendroglial abnormalities in the temporal cor-
and their axons lengthen, showing that myelin production is tex from patients with major depressive disorder. Mol Psychiatry
a perpetual process and that the g-ratio can change (Berthold 10:309–322.
et al. 1983). Increased myelin thickness can compensate for Back SA, Luo NL, Borenstein NS, Bolpe JJ, Kinney HC. 2002. Arrested
increased internodal distance during body growth, which oligodendrocyte lineage progression during human cerebral white mat-
ter development: dissociation between the timing of progenitor differ-
would otherwise reduce conduction velocity (Friede, et al. entiation and myelinogenesis. J Neuropathol Exp Neurol 61:197–211.
1985; Hara et al. 2003; Schro¨der et al. 1978). Experimentally Baker GE, Stryker MP. 1990. Retinofugal fibres change conduction
increasing nodal distance, by lengthening the femur of a rat, velocity and diameter between the optic nerve and tract in ferrets.
increases myelin protein expression 160% (Hara et al. 2003), Nature 344:342–345.
suggesting a feedback signaling between changes in internodal Barbin G, Aigrot MS, Charles P, Foucher A, Grumet M, Schachner M,
et al. 2004. Axonal cell-adhesion molecule L1 in CNS myelination.
distance and myelin thickness to regulate conduction velocity. Neuron Glia Biol 1:65–72.
Dimensions of the node or the morphology of the para- Barres BA, Raff MC. 1993. Proliferation of oligodendrocyte precursor
nodal loops of myelin flanking the node may affect con- cells depends on electrical activity in axons. Nature 361:258–260.
duction velocity, but little is known about whether these Becker D, Gary DS, Rosenzweig ES, Grill WM, McDonald JW. 2010.
change. Swelling of paranodal region after intense repetitive Functional electrical stimulation helps replenish progenitor cells in
the injured spinal cord of adult rats. Exp Neurol 222:211–218.
activation has been observed (Wurtz and Ellisman 1986). Beckman M. 2004. Crime, culpability, and the adolescent brain. Science
Myelinating glia have a major influence on localization 305:596–599.
of ion channels into concentrated domains at the node Bengtsson SL, Nagy Z, Skare S, Forsman L, Forssbert H, Ullén F. 2005.
during development through contact and diffusible fac- Extensive piano practicing has regionally specific effects on white
tors (Dupree et al. 2004). The types of ion channels and the matter development. Nat Neurosci 8:1148–1150.
Bennett EL, et al. 1964 Chemical and anatomical plasticity brain.
spatial localization of channels in the axon, most prominently Science 146:610–619.
concentration of sodium channels in the node and potas- Berthold CH, et al. 1983 Axon diameter and myelin sheath thickness in
sium channels clustered in the paranodal region, affect the nerve fibres of the ventral spinal root of the seventh lumbar nerve of
excitability, frequency of impulse firing, refractory period, the adult and developing cat. J Anat 136:483–509.
and velocity of impulse conduction. In addition to affect- Blumenfeld-Katzir T, Pasternak O, Dagan M, Assaf Y. 2001. Diff usion
MRI of structural brain plasticity induced by a learning and memory
ing conduction velocity, myelin also decreases the refractory task. PLoS ONE 6:e20678.
period, thus enabling transmission of spike trains at a higher Bondy SC, Madsen CJ. 1972. Arrested development of myelin in chick
frequency. optic tectum following deaferrentiation. Brain Res 47:177–184.

582 • NEUROGLIA
Bullock TH, Moore JK, Fields RD. 1984. Evolution of myelin sheaths: Gary DS, Malone M, Capestany P, Houdayer T, McDonald JW. 2012.
both lamprey and hagfish lack myelin. Neurosci Lett 48:145–148. Electrical stimulation promotes the survival of oligodendrocytes in
Canu MH, Carnaud M, Picquet F, Goutebroze L. 2009. mixed cortical cultures. J Neurosci Res 90:72–83.
Activity-dependent regulation of myelin maintenance in the adult Georgieva L, et al. 2006. Convergent evidence that oligodendrocyte
rat. Brain Res 1252:45–51. lineage transcription factor 2 OLIG2 and interacting genes influ-
Carr CE, Konishi M. 1990. A circuit for detection of interaural time dif- ence susceptibility to schizophrenia. Proc Natl Acad Sci U S A
ferences in the brain stem of the barn owl. J Neurosci 10:3227–3246. 103:12469–12474.
Carreiras M, Seghier ML, Baquero S, Estevez A, Lozano A, Devlin JT, Geren BB. 1954. The formation from the Schwann cell surface of
et al. 2009. An anatomical signature for literacy. Nature 461:983–986. myelin in the peripheral nerves of chick embryos. Exp Cell Res
Casey BJ, et al. 2000. Structural and functional brain development and 7:558–562.
its relation to cognitive development. Biol Psychol 54:241–257. Giedd JN. 2004. Structural magnetic resonance imaging of the adoles-
Chiu SY, Wilson GF. 1989. The role of potassium channels in Schwann cent brain. Ann N Y Acad Sci 1021:105–109.
cell proliferation in Wallerian degeneration of explant rabbit sciatic Gold BT, et al. 2007. Speed of lexical decision correlates with diff usion
nerves. J Physiol Lond 408:199–122. anisotropy in left parietal and frontal white matter: evidence from
Cirelli C, et al. 2004. Extensive and divergent effects of sleep and wake- diff usion tensor imaging. Neuropsychologia 45:2439–2446.
fulness on brain gene expression. Neuron 41:35–43. Gootjes L, et al. 2004. Regional distribution of white matter hyperin-
Cole JS, et al. 1994. Modulation of axon diameter and neurofi laments tensities in vascular dementia Alzheimer’s disease and healthy aging.
by hypomyelinating Schwann cells in transgenic mice. J Neurosci Dement Geriatr Cogn Disord 18:180–188.
14:6956–6966. Gordon KA, et al. 2003. Activity-dependent developmental plasticity of
Colello, RJ, Devey,LR Imperato,E, and Pott, U 1995. The chronology of oli- the auditory brain stem in children who use cochlear implants. Ear
godendrocye differentiation in the rat optic nerve: evidence for a signal- Hear 24:485–500.
ing step initiating myelination in the CNS. J Neurosci 15:7665–7672. Gyllensten L, Malmfors T. 1964. Myelinization of the optic nerve and its
Colello RJ, Pott U. 1997. Signals that initiate myelination in the develop- dependence on visual function: a quantitative investigation in mice. J
ing mammalian nervous system. Mol Neurobiol 15:83–100. Embryo Exp Morphol 11:255–256.
Crespo D, Verduga R, Villegas F, Fernandez-Viadera C. 1995. Dimorphic Hakak Y, et al. 2001. Genome-wide expression analysis reveals dysregula-
myelin in the rat optic nerve as a result of retinal activity blockage tion of myelination-related genes in chronic schizophrenia. Proc Natl
by tetrodotoxin during early postnatal period. Histol Histopathol Acad Sci U S A 98:4746–4751.
10:289–299. Hara Y, et al. 2003. P0 mRNA expression increases during gradual nerve
De Biase LM, Nishiyama A, Bergles DE. 2010. Excitability and synap- elongation in adult rats. Exp Neurol 184:428–435.
tic communication within the oligodendrocyte lineage. J Neurosci Hayes WP, Meyer RL. 1989. Impulse blockade by intraocular tet-
30:3600–3611. rodotoxin during optic regeneration in goldfish: HRP-EM evi-
Demerens C, Stankoff B, Logak M, Anglade P, Allinquant B, Couraud F, dence that the formation of normal numbers of optic synapses and
et al. 1996. Induction of myelination in the central nervous system by the elimination of exuberant optic fibers is activity independent.
electrical activity Proc Natl Acad Sci U S A 93:9887–9892. J Neurosci 9:1414–1423.
Dupree JL, et al. 2004. Oligodendrocytes assist in the maintenance of Hsieh S-T, et al. 1994. Regional modulation of neurofi lament organiza-
sodium channel clusters independent of the myelin sheath. Neuron tion by myelination in normal axons. J Neurosci 14:6392–6401.
Glia Biol 1:1–14. Hyde KL, et al. 2006. Morphometry of the amusic brain: a two-site
Emery B. 2010. Regulation of oligodendrocyte differentiation and mye- study. Brain 129:2562–2570.
lination. Science 330:779–782. Ishibashi T, Dakin KA, Stevens B, Lee PR, Zozlov SV, Stewart CL, et
Eyre JA, Miller S, Ramesh V. 1991. Constancy of central conduction al. 2006. Astrocytes promote myelination in response to electrical
delays during development in man: investigation of motor and soma- impulses. Neuron 49:823–832.
tosensory pathways. J Physiol 343:441–452. Itoh K, Stevens B, Schachner M, Fields RD. 1995. Regulation of the neu-
Fields RD. 2005. Myelination: an overlooked mechanism of synaptic ral cell adhesion molecule L1 by specific patterns of neural impulses.
plasticity? Neuroscientist 11:528–531. Science 270:1369–1372.
Fields RD. 2008. White matter in learning, cognition and psychiatric Itoh K, Ozaki M, Stevens B, Fields RD. 1997. Activity-dependent reg-
disorders. Trends Neurosci 31:361–370. ulation of N-cadherin in DRG neurons: differential regulation of
Fields RD. 2010. Neuroscience change in the brain’s white matter. N-cadherin, NCAM and L1 by distinct patterns of action potentials.
Science 330:768–769. J Neurobiol 33:735–748.
Fields RD. 2011a. Imaging learning: the search for a memory trace. Juraska JM, Kopcik JR. 1988. Sex and environmental influences on
Neuroscientist 17:185–196. the size and ultrastructure of the rat corpus callosum. Brain Res
Fields RD. 2011b. Imaging single photons and intrinsic optical signals 450:1–8.
for studies of vesicular and non-vesicular ATP release form axons. Karadottir R, Cavelier P, Bergersen LH, Attwell D. 2005. NMDA recep-
Front Neuroanat 5:32. tors are expressed in oligodendrocytes and activated in ischaemia.
Fields RD, Ni Y. 2010. Nonsynaptic communication through ATP release Nature 438:1162–1166.
from volume-activated anion channels in axons. Sci Signal 3:73. Kingsley JR, Collins GH, Converse WK. 1970. Effect of sciatic neurectomy
Flechsig P. 1876. Die Leitungsbahnen im Gehirn und Rűckenmark des on myelinogenesis in the rat spinal cord. Exp Neurol 26:498–508.
Menschen auf Grund entwicklungsgeschichtlicher Untersuchungen. Klingberg T, et al. 2000. Microstructure of temporo-parietal white mat-
Leipzig: W Englmann, 1987. ter as a basis for reading ability: evidence from diff usion tensor mag-
Friede RL. et al. 1985. Changes in myelin sheath thickness and inter- netic resonance imaging. Neuron 25:493–500.
node geometry in rabbit phrenic nerve during growth. J Anat Knutson P, Ghiani CA, Zhou JM, Gallo V, McBain CJ. 1997. K+ channel
143:103–113. expression and cell proliferation are regulated by intracellular sodium
Fukui Y, Hayashaka S, Bedi KS, Ozaki HS, Takeuchi Y. 1991. and membrane depolarization in oligodendrocyte progenitor cells.
Quantitative study of the development of the optic nerve in rats J Neurosci 17:2669–2682.
reared in the dark during early postnatal life. J Anat 174:37–47. Kraft RH, et al. 1980. Hemispheric asymmetries during six-to
Gallo V, Zhou JM, McBain CJ, Wright CJ, Knutson PL, Armstrong RC. eight-year-olds performance of Piagetian conservation and reading
1997. Oligodendrocyte progenitor cell proliferation and lineage pro- tasks. Neuropsychologia 18:637–643.
gression are regulated by glutamate receptor-mediated K+ channel Kriegler S, Chiu SY. 1993. Calcium signaling of glial cells along mamma-
block. J Neurosci 16:2659–2670. lian axons. J Neurosci 13:4229–4245.

R E GU L AT I O N O F M Y E L I N AT I O N BY F U N C T I O N A L AC T I VI T Y • 583
Kubicki M, Park H, Westin CF, Nestor PG, Mulkern RV, Maier SE, Ruegg DG, Kakebeeke TH, Gabriel JP, Bennefeld M. 2003. Conduction
et al. 2005. DTI and MTR abnormalities in schizophrenia: analysis velocity of nerve and muscle fiber action potentials after a space mis-
of white matter integrity. Neuroimage 26:1109–1118. sion or a bed rest. Clin Neurophysiol 114:86–93.
Kujala P, et al. 1997. The progress of cognitive decline in multiple sclero- Rushton WA. 1951. A theory of the effects of fibre size in medullated
sis. A controlled 3-year follow-up. Brain 120:289–297. nerve. J Physiol 115:101–122.
Kukley M, Capetillo-Zarate E, Dietrich D. 2007. Vesicular glutamate Sanchez I, et al. 1996. Oligodendroglia regulate the regional expansion of
release from axons in white matter. Nat Neurosci 10:311–320. axon caliber and local accumulation of neurofi laments during develop-
Langworthy OR. 1932. Development of behavior patterns and ment independently of myelin formation. J Neurosci 16:5095–5105.
myelinization of tracts in the nervous system. Arch Neurol Psychiatry Sanchez MM, Hearn EF, Do D, Rilling JK, Herndon JG. 1998.
28:136–182. Differential rearing effects corpus callosum size and cognitive func-
Lillie RS. 1925. Factors affecting transmission and recovery in the pas- tion of rhesus monkeys. Brain Res 812:38–49.
sive iron nerve model. J Gen Physiol 7:473–507. Schmithorst VJ, et al. 2005. Cognitive functions correlate with white
Liston C, et al. 2006. Frontostriatal microstructure modulates efficient matter architecture in normal pediatric population: a diff usion ten-
recruitment of cognitive control. Cerebral Cortex 16:553–560. sor MRI study. Hum Brain Map 26:239–147.
Lunn MPT, et al. 2002. Anti-myelin-associated glycoprotein antibodies Scholz J, Klein MC, Behrens TE, Johansen-Berg H. 2009. Training
alter neurofi lament spacing. Brain 125:904–911. induces changes in white-matter architecture. Nat Neurosci
Mabbott DJ, et al. 2006. White matter growth as a mechanism of cogni- 12:1370–1371.
tive development in children. Neuroimage 33:936–946. Schro¨der JM, et al. 1978 Changes of the ratio between myelin thick-
MacFarlane SN, Sontheimer H. 1997. Electrophysiological changes that ness and axon diameter in the human developing sural nerve. Acta
accompany reactive gliosis in vitro. J Neurosci 17:7316–7329. Neuropathol 43:169–178.
Markham MA, Greenough WT. 2004. Experience-driven brain plastic- Seilheimer B, Persohn E, Schachner M. 1989. Antibodies to the L1 adhe-
ity: beyond the synapse. Neuron Glia Biol 1:351–364. sion molecule inhibit Schwann cell ensheathment of neurons in vitro.
Miller EM. 1994. Intelligence and brain myelination: a hypothesis. J Cell Biol 107:341–351.
Person Indiv Diff 17:803–832. Shrager P, Novakovic SD. 1995. Control of myelination, axonal growth,
Moor CL, Kalil R, Richards W. 1976. Development of myelination in and synapse formation in spinal cord explants by ion channels and
optic tract of the cat. J Comp Neurol 165:125–136. electrical activity. Dev Brain Res 88:86–87.
Nagy Z, et al. 2004. Maturation of white matter is associated with Sirevaag AM, Greenough WT. 1987. Differential rearing effects on rat
the development of cognitive functions during childhood. J Cogn visual cortex synapses III Neuronal and glial nuclei, boutons, den-
Neurosci 16:1227–1233. drites, and capillaries. Brain Res 424:320–332.
Narang HK. 1977. Right-left asymmetry of myelin development in Skaliora I, Scobey RP, Chalupa LM. 1993. Prenatal development of
epiretinal portion of rabbit optic nerve. Nature 266:855–856. excitability in cat retinal ganglion cells: action potentials and sodium
Nave KA. 2010. Myelination and support of axonal integrity by glia. currents. J Neurosci 13:313–323.
Nature 468:244–252. Sontheimer H, Black JA, Waxman SG. 1996. Voltage-gated Na+ chan-
Nestor PG, et al. 2007. Episodic memory and neuroimaging of hippocam- nels in glia: properties and possible functions. Trends Neurosci
pus and fornix in chronic schizophrenia. Psychiatry Res 155:21–28. 19:325–331.
Niogi SN, McCandliss BD. 2006. Left lateralized white matter micro- Stanford LR. 1987. Conduction velocity variations minimize conduc-
structure accounts for individual differences in reading ability and tion time differences among retinal ganglion cell axons. Science
disability. Neuropsychologia 44:2178–2188. 238:358–360.
Pappas CA, Ulrich N, Sontheimer H 1994. Reduction of glial pro- Steingard RJ, Renmshaw PF, Hennen J, Lenox M, Cintron CB, et al.
liferation by K+ channel blockers is mediated by changes in pH. 2002. Smaller frontal lobe white matter volumes in depressed adoles-
Neuroreport 6:193–196. cents. Biol Psychiatry 52:413–417.
Peters A, Verderosa A, Sethares C. 2008. The neuroglial popula- Stevens B, Fields RD. 2000. Response of Schwann cells to action poten-
tion in the primary visual cortex of the aging rhesus monkey. Glia tials in development. Science 287:2267–2271.
56:1151–1161. Stevens B, Ishibashi T, Chen J-F, Fields RD. 2004. Adenosine: an
Psachoulia K, Jamen F, Young KM, Richardson WD. 2009. Cell cycle activity-dependent axonal signal regulating MAP kinase and prolif-
dynamics of NG2 cells in the postnatal and ageing brain. Neuron eration in developing Schwann cells. Neuron Glia Biol 1:23–34.
Glia Biol 5:57–67. Stevens B, Porta S, Haak LL, Gallo V, Fields RD. 2002. Adenosine: a
Pujol J et al. 2006. Myelination of language-related areas in the develop- neuron-glial transmitter promoting myelination in the CNS in
ing brain. Neurology 66:339–343. response to action potentials. Neuron 36:855–868.
Ramón y Cajal S. 1995. Histology of the nervous system of man and ver- Stevens B, Tanner S, Fields RD. 1998. Control of myelination by specific
tebrates. New York: Oxford University Press. patterns of neural impulses. J Neurosci 15:9303–9311.
Ransom BR, Orkand RK. 1996. Glial-neuronal interactions in Stewart SE, et al. 2007. A genetic family-based association study of
non-synaptic areas of the brain: studies in the optic nerve. Trends OLIG2 in obsessive-compulsive disorder. Arch Gen Psychiatry
Neurosci 19:352–358. 64:209–206.
Ranvier ML. 1878. Lecons sur l’histologie du système nerveux. Paris, Sugihara I, Lang EJ, Llinas R. 1993. Uniform olivocerebellar conduction
1878. Quoted in Young JZ 1944. time underlies Purkinje cell complex spike synchronicity in the rat
Rivers LE, Young KM, Rizzi M, Jamen F, Psachoulia K, Wade A, et cerebellum. J Physiol Lond 470:243–271.
al. 2008. PDGFa/NG2 glia generate myelinating oligodendro- Swadlow HA. 1985. Physiological properties of individual cere-
cytes and piriform projection neurons in adult mice. Nat Neurosci bral axons studied in vivo for as long as one year. J Neurophysiol
11:1392–1401. 54:1346–1362.
Rorig B, Grantyn R. 1993. Glutamatergic and GABAergic synaptic cur- Swadlow HA, Rosene DL, Waxman SG. 1978. Characteristics of inter-
rents in ganglion cells from isolated retinae of pigmented rats during hemispheric impulse conduction between prelunate gyri of the rhesus
postnatal development. Brain Res Dev Brain Res 74:989–110. monkey. Exp Brain Res 33:455–467.
Roy K, Murtie JC, El-Khodor BF, Edgar N, Sardi SP, Hooks BM, et al. Szeligo F, Leblond CP. 1997. Response of the tree main types of glial
2007. Loss of erbB signaling in oligodendrocytes alters myelin and cells of cortex and corpus callosum in rats handled during suckling or
dopaminergic function, a potential mechanism for neuropsychiatric exposed to enriched, control and impoverished environments follow-
disorders. Proc Natl Acad Sci U S A 104:8131–8136. ing weaning. J Comp Neurol 172:247–263.

584 • NEUROGLIA
Tasaki I. 1939. The electro-saltatory transmission of the nerve impulse Waxman SG, Pappas GD, Bennett MVL. 1972. Morphological corre-
and the effect of narcosis upon the nerve fiber. Am J Physiol lated functional differentiation of nodes of Ranvier along single fibers
127:211–227. in the neurogenic electric organ of the knife fish Stenarchus. J Cell
Tauber H, Waehneidt TV, Neuhoff V. 1980. Myelination in rabbit optic nerves Biol 53:210–224.
is accelerated by artificial eye opening. Neurosci Lett 16:235–238. Wood PM, Schachner M, Bunge RP. 1990. Inhibition of Schwann
Taveggia C, Feltri ML, Wrabetz L. 2010. Signals to promote myelin for- cell myelination in vitro by antibody to the L1 adhesion molecule.
mation and repair. Nat Rev Neurol 6:276–287. J Neurosci 10:3635–3645.
Teicher MH, Dumont NL, Ito Y, Vaituzis C, Giedd JN, Andersen SL. Wurtz CC, Ellisman MH. 1986. Alterations in the ultrastructure of
2004. Childhood neglect is associated with reduced corpus callosum peripheral nodes of Ranvier associated with repetitive action poten-
area. Biol Psychiatry 56:80–85. tial propagation. J Neurosci 6:3133–3145.
Teicher MH, Samson JA, Sheu TS, Polcari A, McGreenery CE. 2010. Yakolev PI, Lecours AR. 1967. In: Minkowski A (ed.), Regional develop-
Hurtful words: association of exposure to peer verbal abuse with ele- ment of the brain in early life. New York: Oxford University Press,
vated psychiatric symptom scores and corpus callosum abnormalities. pp. 3–70.
Am J Psychiatry 167:1464–1471. Yin X, et al. 1998. Myelin-associated glycoprotein is a myelin signal that
Tkachev D, et al. 2003. Oligodendrocyte dysfunction in schizophrenia modulates the caliber of myelinated axons. J Neurosci 18:1953–1962.
and bipolar disorder. Lancet 362:798–805. Young JZ. 1944. Surface tension and the degeneration of nerve fibres.
Traub RJ, Mendell LM. 1988. The spinal projection of individual identi- Nature 154:521–522.
fied A-d- and C-fibres. J Neurophysiol 59:41–55. Zalc B, Fields RD. 2000. Do action potentials regulate myelination?
Wake H, Lee PR, Fields RD. 2011. Control of local protein synthesis Neuroscientist 6:5–12.
and the initial events in myelination by action potentials. Science Zatorre RJ, Fields RD, Johansen-Berg H. 2012. Plasticity in gray and
333:1647–1651. white: neuroimaging changes in brain structure during learning. Nat
Walhovd KB, Fjell AM. 2007. White matter volume predicts reaction Neurosci 15:528–536.
time instability. Neuropsychologia 45:2277–2284. Ziskin JL, Nishiyama A, Rubio M, Fukaya M, Bergles DE. 2007. Vesicular
Waxman SG 1997. Axon-glia interactions: building a smart nerve fiber. release of glutamate from unmyelinated axons in white matter.
Curr Biol 7:R406–410. Nat Neurosci 10:321–330.

R E GU L AT I O N O F M Y E L I N AT I O N BY F U N C T I O N A L AC T I VI T Y • 585
46.
IRON AND GLIA
James R. Connor

A B B R E VI AT I O N S transport and uptake proteins in the context of their specific


contributions to glial cell biology.
ATP Adenosine triphosphate
BBB blood-brain barrier
CSF cerebrospinal fluid 2 IRON AND GLIA
DAB 3-3´diaminobenzidine
DMT1 divalent metal transporter 1 Iron and Glia has both a distinctive regional and cellular distri-
HFE human hemochromatosis bution. Iron is quantifiable using atomic absorption spectros-
HMG-CoA 3-hydroxy-3-methylglutaryl-coenzyme A copy or can be visualized using a histochemical stain known as
IFN interferon the Perl reaction. At the regional level, iron is most prominent
IL interleukin in areas associated with motor functions such as basal ganglia,
IRE Iron Responsive Element substantia nigra (pars reticulata), and deep cerebellar nuclei.
LPS lipopolysaccharid Indeed the iron concentration in the globus pallidus is report-
MAG myelin-associated glycoprotein edly similar to that found in the liver (Hallgren and Sourander
MBP myelin basic protein 1958). The liver is considered the main iron storage organ in
MS multiple sclerosis the body. This proclivity for iron in regions associated with
NF- κB nuclear factor kappa-light-chain-enhancer the motor system is consistent among all animal species inves-
of activated B cells tigated (Guizzetti 1915; Spatz 1922) from rodents to monkeys
OPC oligodendrocyte precursor cell to humans. Moreover, there is also a high concentration of
PND postnatal day iron in white matter (Connor and Menzies 1990; Hallgren
RBC red blood cell and Sourander 1958; Rajan et al. 1976).
sla sex-linked anemia At the cellular level, the cells that most prominently stain
SVZ subventricular zone for iron are oligodendrocytes. Iron enriched oligodendro-
Tf transferrin cytes are found in white matter and even in the gray matter
TfR transferrin receptor regions where iron is enriched in select brain nuclei such as
Tim-2 T-cell immunoglobulin mucin domain the substantia nigra (Fig. 46.1A) and striatum (Fig. 46.1B).
2 protein Iron-positive oligodendrocytes are also found in lower animal
TNF tumor necrosis factor forms such as chickens but not frogs or fish (Erb et al. 1996),
WM white matter suggesting that the function of iron in oligodendrocytes is
WML white matter lesion not limited to myelin. The latter comment is perhaps consis-
tent with reports of perivascular and perineuronal oligoden-
drocytes staining for iron in rodents and humans (Fig. 46.2).
1 INTRODUCTION Nonetheless, peak uptake of iron in the rodent brain occurs
in concert with peak times of myelination (Taylor et al. 1991;
Iron is an essential metal in biological systems. It is necessary Taylor and Morgan 1990) and is consistent with oligodendro-
for the consumption of oxygen and production of adenos- cytes in white matter staining robustly for iron. Iron is report-
ine triphosphate (ATP). It is also an essential cofactor for edly associated with myelin sheaths (Francois 1981). Not all
many enzymes including those involved in cholesterol and oligodendrocytes in the white matter can be seen following an
neurotransmitter synthesis, and in degradation of proteins iron stain.1 In the human brain it is especially obvious that the
that require ubiquitination. Because of the ability of iron to iron-positive oligodendrocytes in white matter are organized
interact with oxygen, it can also be a potent generator of free
radicals. Thus, cells have developed an elegant system for keep- 1
There was confusion and controversy when iron staining in the brain was first
ing iron bound to proteins during delivery to cells and stor- reported in the white matter. Iron staining in the brain is significantly affected by
age within cells. The discovery of iron regulatory proteins fixation, storage in formalin where the formalin breaks down into formic acid and
including those involved in export of iron is an evolving story iron leeches out of the tissue. Moreover, iron staining in paraffin and frozen sec-
tions can be difficult to detect. The modification of 3-3’diaminobenzidine (DAB)
and the data to date are summarized in Table 46.1. Thus, this intensification to iron staining first described in 1980 was a significant advance to
chapter will focus on iron but will include discussions of iron the study of brain iron histology. Nguyen-Legros et al. 1980.

586
Table 46.1 PROTEINS INVOLVED IN IRON HOMEOSTASIS FOR GLIA
CELL TYPE PROTEIN FUNCTION REFERENCE

Oligodendrocytes Tf Nonsecreted form transports Fe within cell and is Bloch et al. 1985, Espinosa de los Monteros
required for maturation and myelinogenesis; taken et al. 1994, Bartlett, Li and Connor 1991,
up by OPCs de Arriba Zerpa et al. 2000, Møllgård et al.
1988

Tf R1 Expressed only by OPCs to take up iron via Tf Moos et al. 2007


endocytosis

Tim2 Ferritin receptor that may allow for iron to be Todorich et al. 2008, Todorich, Zhang and
taken up and incorporated in endogenous Tf Connor 2011

CXCR4 Interacts with ferritin; is important for Patel et al. 2010, Dziembowska et al. 2005,
oligodendrocyte migration and maturation Carbajal et al. 2011, Li et al. 2006

Hephastin Ferroxidase that mediates iron efflux in mature Schulz et al. 2011
oligodendrocytes
No ceruloplasmin Schulz et al. 2011
2+ +
DMT1 Fe and H symporter into cytosol; low expression Burdo et al. 2001
in subcortical white matter
Ferroportin Fe2+ efflux into cytosol; associated with hephaestin Burdo et al. 2001
Ferritin Iron storage and ferroxidase Connor and Menzies 1996, Blissman et al.
1996
IRP1 Iron sensor that modulates the translation of Hentze et al. 1987, Casey et al. 1989
ferritin and Tf R by binding to an IRE; cytoplasmic
aconitase activity
IRP2 Iron sensor that modulates the translation of ferritin Magaki et al. 2007
and Tf R by binding to an IRE

Microglia No Tf Benkovic and Connor 1993

No Tf R1 Lin and Connor 1989

Ferritin Iron storage and ferroxidase; secreted ferritin can Todorich et al. 2008, Todorich et al. 2011,
associate with receptors on oligodendrocytes Zhang et al. 2006, Cheepsunthorn, Palmer
and Connor 1998

CXCR4 Interacts with ferritin but iron related function is Lavi et al. 1997
unknown

HFE Low expression in CD68+ activated microglia in Gebril et al. 2011, Roy et al. 2004
white matter; associates with Tf R1 to reduce iron
uptake and is involved in innate immunity

IRP1 Iron sensor that modulates the translation of Hentze et al. 1987, Casey et al. 1989,
ferritin and Tf R by binding to an IRE; cytoplasmic Rathnasamy, Ling and Kaur 2011
aconitase activity

IRP2 Iron sensor that modulates the translation of ferritin Rathnasamy et al. 2011
and Tf R by binding to an IRE

Astrocytes Ceruloplasmin Ferroxidase that converts Fe2+ to Fe3+ so that it Patel and David 1997, Patel, Dunn and
can be exported by ferroportin David 2000

APP Ferroxidase that converts Fe2+ to Fe3+ so that it Rogers et al. 1999, Rogers et al. 2002, Duce
can be exported by ferroportin; increased protein et al. 2010
expression in response to IL-1

Ferroportin Fe2+ efflux into cytosol; associated with Wu et al. 2004


ceruloplasmin
Hepcidin Modulates iron export by binding to ferroportin Nemeth and Ganz 2006, Zechel,
and inducing internalization Huber-Wittmer and von Bohlen und
Halbach 2006

(continued)

IRON AND GLIA • 587


Table 46.1 (Cont’d)
CELL TYPE PROTEIN FUNCTION REFERENCE

DMT1 Fe2+ and H+ symporter into cytosol Burdo et al. 2001

Ferritin Iron storage and ferroxidase Connor et al. 2003

No Tf Connor and Benkovic 1992

No Tf R1 Moos 1996

HFE Low expression in white matter; associates with Gebril et al. 2011
Tf R1 to reduce iron uptake

IRP1 Iron sensor that modulates the translation of ferritin Hentze et al. 1987, Casey et al. 1989
and Tf R by binding to an IRE; cytoplasmic aconitase
activity
IRP2 Iron sensor that modulates the translation of ferritin Magaki et al. 2007
and Tf R by binding to an IRE

into patches (Todorich et al. 2009) (Fig. 46.3) but the patches inflammatory responses in diseases such as multiple sclerosis
of iron-positive oligodendrocytes are also seen in rodents; to (MS) ( see chapter 61). The relationship between the patches
visualize the patches in the rodents usually requires thicker of iron-positive oligodendrocytes and plaque formation in
histological sections than standard paraffin or cryostat sec- MS or white matter (WM) lesions in the aging brain in which
tions. The patches oligodendrocytes are only seen following iron distribution is altered (Bagnato et al. 2011; Gebril 2011)
staining for iron. Those oligodendrocytes in the white mat- has not been investigated and would be difficult to do so, but
ter that stain for transferrin, the iron mobilization protein, or the idea that patches of iron enriched oligodendrocytes are the
ferritin (iron storage protein) are not organized in discern- preferred site for initiation of MS or WM lesions with aging
ible patches (Connor and Menzies 1996a; Dwork et al. 1988) is intriguing. Perhaps, as resolution with neuroimaging tech-
(Fig. 46.4A,B). It appears, then, that some oligodendrocytes niques improves, the relationship between iron and WM his-
contain much more stainable iron than others; this could be a topathology can be interrogated (Bagnato et al. 2011).
clinically meaningful observation because iron enriched oligo- The patches of iron-positive oligodendrocytes appear to
dendrocytes are more vulnerable to oxidative stress (Thorburne form early in development and are located around blood vessels.
and Juurlink 1996) and thus could be more susceptible to The circumference of the patch increases with age and reaches

A B

Figure 46.1 Iron in the Brain. A. Visualization of iron in the substantia nigra of the mouse. Oligodendrocytes of the substantia nigra reticulata
contain most of the iron of the nigra and appear as small, dark, round cells. The substantia nigra pars compacta (bounded by the dashed line), in which
dopaminergic neurons that require iron for catecholamine production are located, contains comparatively less iron than the reticulata. The inset
provides higher magnification of oligodendrocytes in the reticulata. B. In the mouse striatum, iron-positive oligodendrocytes are robustly stained in
the white matter striations. These cells have few processes and appear “capped” at ends because of the eccentric position of the nucleus within the cell.
Other oligodendrocytes with the same capped morphology appear outside of the white matter patches.

588 • NEUROGLIA
of auditory and visual evoked potentials, which is interpreted
bv as insufficient myelination (Lozoff et al. 2006). In rodents,
in which myelin can be directly measured, restricting iron in
the diet during development results in decreased expression
of myelin proteins, lipids, and cholesterol (Beard et al. 2003;
Ortiz et al. 2004a; Yu et al. 1986). In contrast, it has recently
been reported that a mutation in the human hemochromato-
sis (HFE) protein that normally would limit iron uptake into
cells is associated with higher myelination ( Jahanshad et al.
2012). Because loss of myelin with age has been linked to cog-
nitive decline with aging (Bartzokis 2004), this mutation in
the HFE protein is argued to preserve cognitive function with
age; an intriguing concept that should be explored.
The accumulation of iron by oligodendrocytes is not depen-
dent of the structural integrity of myelin. In rodent models
with abnormal myelin, as long as oligodendrocytes develop
50 μm normally, they accumulate iron. (Connor and Menzies 1990;
LeVine 1991). On the other hand, if oligodendrocyte develop-
Figure 46.2 DAB enhanced Perl reaction was used to stain iron in the stria- ment is compromised, the iron accumulates in astrocytes and
tum of mouse brain. Iron-positive oligodendrocytes were located in the white microglia in the white matter (Connor and Menzies 1990).
matter tract (arrows) and near blood vessels (rectangular box). The insert is a
representative of perivascular oligodendrocytes. bv, Blood vessel.
2.1 D EV E L O PM E N T
adult patterns by postnatal day (PND) 28 in the rat (Connor As mentioned, peak iron uptake into the brain coincides
et al. 1988). The appearance of transferrin in oligodendrocytes, with peak myelinogenesis in the rat brain (Taylor and
an early marker of oligodendrocyte differentiation (Espinosa Morgan 1990). However, iron-positive cells in the rat brain
de los Monteros et al. 1988), in the developing optic nerve can be detected as early as PND 3 (Connor et al. 1995a). The
has a similar relationship to blood vessels (Lin and Connor iron-positive cells at this time period are primarily found in
1989b). This early relationship between iron accumulation by the subventricular zone (SVZ) and subcortical white matter
oligodendrocytes and blood vessels is consistent with histori- tracts. The morphological appearance and apparent migra-
cal studies on the initiation sites of myelinogenesis in white tion of the iron-positive cells from SVZ to subcortical white
matter tracts, which were thought to occur near blood vessels. matter mirrors that of GD3+ oligodendrocyte precursor
The concept of iron accumulation by oligodendrocytes occur- cells (LeVine and Goldman 1988). The iron-positive cells
ring coincident with onset of myelin is consistent with numer- in the SVZ are large and round and non–process-bearing
ous reports of hypomyelination in iron deficiency. For example, (Connor et al. 1995a; Rathnasamy et al. 2011), whereas those
in studies on iron deficient children there is increased latency farther away from the SVZ have more processes and a smaller
soma. Over time, the cellular processes become shorter and
thinner until the cells become aligned with other iron-posi-
tive cells in the white matter and lose their processes. Many
iron-positive oligodendrocytes are seen in clusters around
blood vessels. It is not known if the oligodendrocytes around
the blood vessels migrated to that site with iron or began to
accumulate iron when they came in proximity to the blood
vessels. Oligodendrocytes from the SVZ do migrate to sites
near blood vessels (Levison et al. 1993; Levison et al. 1993).
The circumference of the patch expands from the blood vessel
with age in developing animals and in the Belgrade rat model
in which iron uptake is compromised, the circumference of
the patch of oligodendrocytes around the blood vessel is
smaller (Burdo et al. 1999). These data strongly suggest that
the oligodendrocytes are accumulating iron in situ as opposed
100 μm to migrating into the subcoritical white matter with iron.
The functional significance of the iron accumulation near
blood vessels is not clear but could relate to myelinogenic
Figure 46.3 Iron visualization in the brain using the DAB-Perl stain. This foci near blood vessels (Skoff et al. 1980). The contribu-
image from the human white matter tract demonstrates the “patchy” nature
tion to iron management in the brain by the different glial
of iron staining in white matter. The inset shows oligodendrocytes aligning
in a classic row-like pattern, which is a hallmark characteristic of these cell types and oligodendrocyte development is shown in
cells. Schematic 46.1

IRON AND GLIA • 589


A B

LV

50 μm 100 μm
10 μm

Figure 46.4 Transferrin and Ferritin in Oligodendrocytes. A. Immunohistochemical staining for Transferrin in the corpus callosum of mouse brain.
Transferrin-positive oligodendrocytes appear in a row in the corpus callosum. Very few of the cells are process bearing. The inset shows a representative
image of two transferrin-positive oligodendrocytes. The ependymal cells lining the lateral ventricles are also stained for transferrin (arrows). LV, Lateral
ventricle. B. Ferritin immunostaining of oligodendrocytes in human putamen. In contrast to iron distribution in white matter, ferritin immunostained
cells are homogeneously spread throughout the tissue. Arrowheads denote examples of oligodendrocytes aligned in rows; the inset provides higher
magnification of these cells.

The need for iron for oligodendrocytic maturation and is extensive (Table 46.2). For example, glucose-6-phosphate
function is established. Although no direct regulatory roles dehydrogenase is a key enzyme in the pentose-phosphate
for iron in oligodendrocytes have been identified (i.e., there shunt pathway used by oligodendrocytes for metabolism of
are no known iron regulatory elements on the mRNAs of glucose (Cammer et al. 1982; Kugler 1994; Sanchez-Abarca
myelin proteins) the list of enzymes and metabolic processes et al. 2001; Warringa et al. 1987). There are two steps in the
that are unique to or highly expressed in oligodendrocytes synthesis of cholesterol, a key component of myelin, that

OPC Oligodendrocyte Microglia

Key

Fe2+ Tim2

Fe3+
Endolysosome
Astrocyte DMT1
Ferritin/iron

TF
Ceruloplasmin

TFR Ascorbate

Schematic 46.1 Oligodendrocyte progenitor cells (OPCs) express transferrin receptors (TfR) and the ferritin receptor Tim2, which allow for iron uptake
from transferrin and ferritin. The iron is released from these two proteins while they are within an endolysosome. As the OPC matures into an oligo-
dendrocyte, TfR expression is reduced and Tim2 expression remains. Microglia secrete ferritin, which can bind to Tim2 that is expressed by OPCs
and mature oligodendrocytes, allowing for continuous iron uptake. Indeed this expression of Tim2 and the amount of iron deliverable via ferritin
may be a necessary step for oligodendrocyte maturation. It has been proposed that astrocytes release ascorbate, which can associate with ferrous iron.
Ferrous iron can then be taken up by the astrocyte and be used within the cell. Astrocytes also express ceruloplasmin, which is a ferroxidase that con-
verts ferrous iron into ferric iron. Ferric iron binds to transferrin, which can then bind to TfR and be taken up by a cell.

590 • NEUROGLIA
Table 46.2 IRON DEPENDENT ENZYMES IN GLIA et al. 2009). The association between myelinogenesis and iron
ENZYME FUNCTION REFERENCES
and the cells involved are discussed later in this chapter, but
we have proposed that there are windows of opportunity that
Ribonucleotide DNA synthesis that Wrigglesworth and Baum need to be identified for rescuing a myelin deficit in the brain.
reductase catalyzes rate-limiting 1988 Moreover, a key question is whether iron deficiency impacts
step the development and migration of oligodendrocytes or just
Cytochrome Electron transport chain Wrigglesworth and Baum their ability to make myelin. This remains a key biological and
oxidases 1988 clinically important question—is there a sufficient number
of oligodendrocytes available in the white matter to rescue a
Succinate Citric acid cycle and Friede 1962
dehydrogenase electron transport chain developmental hypomyelination caused by iron deficiency?

NADH Electron transport chain Wrigglesworth and Baum


dehydrogenase 1988 2.2 T R A NS F E R R I N

Aconitase Citric acid cycle Wrigglesworth and Baum The early studies reporting on transferrin in oligodendrocytes
1988 considered that iron could be released from oligodendro-
cytes via transferrin because transferrin is an iron mobiliza-
HMG-CoA Cholesterol syn- Cammer 1984
reductase thesis: enriched in tion protein and is secreted by the liver and by the choroid
oligodendrocytes plexus (Watanabe et al. 1990; Zakin et al. 2002). Thus it was
logical to think that transferrin should also be secreted by
Lipooxygenases Lipid synthesis: Shimizu and Wolfe 1990 oligodendroyctes until a study by Zakin and colleagues (de
enriched in
oligodendrocytes Arriba Zerpa et al. 2000) demonstrated that the mRNA for
transferrin lacks a secretory signal which distinguishes it from
liver transferrin. Thus, transferrin in oligodendrocytes was
not secreted, making them the only cells that synthesize but
requires iron including 3-hydroxy-3-methylglutaryl-coenzyme do not secrete transferrin. The lack of secretion of transfer-
A (HMG-CoA) reductase, which is the rate limiting step in rin emphasizes the importance of iron in oligodendrocytes.
cholesterol synthesis and this enzyme is enriched in oligo- Moreover, the transferrin receptor is found primarily on neu-
dendrocytes (Nelissen et al. 2012; Pleasure et al. 1977). Lipid rons in gray matter thus transferrin bound iron would have to
synthesis and breakdown is also iron dependent (Bourre et al. leave the oligodendrocytes in the white matter and make its
1984;). Iron is essential for cytochrome oxidase activity and way to neurons in the gray matter, which is not the optimal
the production of ATP. Thus, in addition to a direct impact design for iron delivery to neurons. The current paradigm is
on myelin synthesis, iron is also essential to support the meta- that the transferrin in the extracellular space in the brain is
bolic activity of oligodendrocytes. The relatively high content likely from cerebrospinal fluid (CSF) and transport across the
of iron in the oligodendrocytes may reflect their reported blood-brain barrier (BBB).
relatively high metabolic rate compared to other cells (Hyden What is the role of transferrin in oligodendrocytes?
1960; Sanchez-Abarca et al. 2001). In support of the concept Transferrin is expressed very early in oligodendrocyte matu-
that iron is metabolically essential to oligodendrocytes is the ration in culture, colocalizing with myelin basic protein but
consistent observation in studies that there is a general loss appearing before the expression of galactocerebroside in cul-
of myelin and myelin proteins as opposed to loss of a specific ture (Connor et al. 1993; Espinosa de los Monteros et al. 1988;
myelin component following iron deficiency. Espinosa de los Monteros and de Vellis 1988; Espinosa de los
That iron is required for myelin production was clev- Monteros et al. 1999). In vivo, transferrin (Tf ) expression
erly shown in a study using gastrostomy-reared rats that had in oligodendrocytes preceded both galactocerebroside and
only the amount of iron changed in the milk substitutes that myelin basic protein (Lin and Connor 1989). Overexpression
the animals were fed. This approach eliminated concerns of of transferrin in oligodendrocytes will accelerate their differ-
undernutrition that may accompany studies on dietary iron entiation and is associated with more myelin in vivo (Saleh
deficiency. In this study, there were not extensive growth defi- et al. 2003; Sow et al. 2006). The expression of transferrin in
cits in the body or brain but the iron deficiency was associated oligodendrocytes occurs even when myelin integrity is com-
with a dramatic delay in the development of myelination (de promised as in the shiverer mouse mutant (Connor et al. 1993)
los Monteros et al. 2000). and the myelin deficient rat model (Lin and Connor 1989). If
The question of delay in myelination associated with oligodendrocyte development is compromised, however, the
dietary iron deficiency is significant from a standpoint of expression of transferrin at both the protein and mRNA is
clinical intervention. The World Health Organization has decreased (Bartlett et al. 1991). Studies on transferrin mobi-
identified iron deficiency as the leading nutritional problem lization within oligodendrocytes suggest that it is transported
in the world based on the cognitive and behavioral deficits in clathrin coated vesicles from cell bodies and into the oligo-
that are sustained into adulthood in children who were iron dendrocytic processes wherein it promotes stabilization of the
deficient. Recent data has suggested that attention deficit dis- processes (Ortiz et al. 2005). Indeed, in transgenic mice over-
order could be associated with iron deficiency during develop- expressing transferrin, there is an increase in several microtu-
ment (Cortese et al. 2009) and myelin impairment (Luders bule-associated proteins in the white matter particularly in

IRON AND GLIA • 591


the stable tubule only peptide supporting the argument that these cells rather than internalized from extracellular ferritin.
transferrin stabilizes processes in oligodendrocytes (Marta Indeed in cell cultures, H-ferritin mRNA is increased in oligo-
et al. 2002). Thus, it appears that there is trafficking of endo- dendrocytes when they first adhere to culture plates (Sanyal
somes in oligodendrocytes for iron delivery throughout the et al. 1996), indicating ferritin synthesis is an early event in
cell and that trafficking into the oligodendrocyte processes oligodendrocyte maturation. An alternative, but not mutually
may be directly related to formation of myelin membranes and exclusive, interpretation is that ferritin could be released by
perhaps consistent with the observations of iron in the myelin the microglia and taken up by nearby oligodendrocytes. This
processes that was noted by electron microscopy. idea is consistent with our finding that ferritin could be taken
up by oligodendrocytes (Hulet et al. 2000). To test this idea,
we took conditioned media from microglial cells in culture
2.3 I RO N AC Q U I S I T I O N
and demonstrated that the media has a trophic effect on oligo-
Iron acquisition by developing oligodendrocytes has received dendrocytes and that the trophic effect can be eliminated by
considerable attention in both cell culture and in vivo studies. reducing the expression of H-ferritin in the microglia (Zhang
Transferrin was considered an obligatory protein for oligo- et al. 2006a). This indirect evidence that ferritin was a trophic
dendrocytes in culture until recently when we demonstrated factor for oligodendrocytes was subsequently confirmed using
that transferrin could be replaced by H-ferritin (Todorich direct exposure of ferritin to oligodendrocytes in cultures
et al. 2011). The requirement of transferrin for oligodendro- (Todorich et al. 2011). Moreover, a receptor for H-ferritin
cyte survival in culture was incongruent with studies on the was discovered on oligodendrocytes (Todorich et al. 2008)
distribution of transferrin receptors in the brain that consis- (Fig. 46.5), which is consistent with our reports of ferritin bind-
tently failed to detect transferrin receptor expression in white ing to white matter tracts in rodent and human brains (Hulet
matter (Hill et al. 1985; Hulet et al. 1999a,b; Mash et al. 1990) et al. 1999a). The receptor for H-ferritin is Tim-2 (T-cell immu-
except during very early developmental periods (Lin and noglobulin mucin domain 2) protein (Chen et al. 2005).
Connor 1989b). Although one could argue that the lack of Tf An iron-dependent relationship between macrophage acti-
receptor expression on oligodendrocytes was a result of down- vation and oligodendrocyte was further shown in a spinal cord
regulation of the receptor on these cells because they had high injury model (Schonberg and McTigue 2009). This study was
intracellular iron stores, there was no detectable expression of based on two findings: (1) lipopolysaccharide (LPS) injections
Tf receptors even under conditions of dietary iron deficiency into the spinal cord results initially in a loss of oligodendrocytes
that were sufficient to cause hypomyelination (Han et al. followed by significant oligodendrogenesis (Schonberg et al.
2003). The data strongly argue that transferrin receptors 2007) and (2) iron-positive microglia are upregulated in the
are not expressed on mature oligodendrocytes in vivo. We brain following LPS injection (Zhang et al. 2005a). Following
demonstrated that receptors for H-ferritin were selectively the LPS injection there were numerous ferritin-positive
expressed in the white matter (Hulet et al. 1999a) and thus macrophages suggesting these cells were accumulating iron.
proposed that H-ferritin was the major iron delivery protein Around the lesion, NG2-positive cells were detected and these
for oligodendrocytes. cells contain ferritin. One week after the LPS injection, there
The first suggestion that H-ferritin may have a trophic
relationship to oligodendrocytes, came from a developmen-
tal study in rats (Cheepsunthorn et al. 1998). In this study, it
was reported that ferritin was found predominantly in micro-
glia in white matter at PND 5 and by PND 30 the ferritin
staining was predominantly in oligodendrocytes. The salient
finding in this report was the temporal and spatial relation-
ship between the ferritin staining of microglia and the onset
of ferritin expression in oligodendrocytes. At PND 5, the
ferritin-positive microglia are numerous and robust in subcor-
tical white matter and corpus callosum. Very few oligodendro-
cytes are present and they are not ferritin positive. With age,
the number of ferritin-positive oligodendrocytes increase and
the number of ferritin-positive microglia decrease. Moreover,
this negative correlation occurs in microglia and oligoden-
drocytes that are within close proximity in specific regions of
the subcortical white matter. By PND 30, the ferritin posi-
tive oligodendrocytes almost completely replaced the micro-
glia and myelination of the callosum is homogeneous. This
developmental pattern of ferritin in microglia giving way to
ferritin-positive oligodendrocytes in the white matter is simi-
lar to that reported for cellular staining of iron in white mat- Figure 46.5 Tim-2 in Rodent Oligodendrocytes in Cell Culture. In this
ter during development (Connor et al. 1995). It is possible confocal image, Tim-2 immunostaining appears in the green channel and
that the ferritin in the oligodendrocytes was synthesized by DAPI, a nuclear stain, is in the blue channel.

592 • NEUROGLIA
was a significant increase in ferritin-positive oligodendroyctes and mechanism of delivery has been demonstrated in vivo as
in the lesions. Similar to the reports in the developmental well. In studies that inject apo-transferrin (iron poor trans-
study in the white matter, there was a shift from iron-positive ferrin) into the cerebral cortex as a mechanism to improve
macrophages to iron/ferritin-positive oligodendrocytes. The myelination an effect is only seen between 2 and 7 days of age
oligodendrocyte response to the injury can be blocked by (Marta et al. 2000). The data indicate that there is a window
treating the animals with an iron chelator. This effect of iron when transferrin can enhance myelination, which is concur-
chelation on oligodendrocytes is consistent with cell culture rent with the presence of prooligodendrocytes. The uptake of
studies that showed the trophic potential of the macrophages transferrin into the oligodendrocytes could be mediated by
on oligodendrocytes is lost when the cells are treated with a transferrin receptor at this age because there are reports of
an iron chelator (Zhang et al. 2006a). The iron in these acti- transferrin binding to or being expressed by developing oligo-
vated macrophages could be from a number of sources, but dendrocytes (Espinosa de los Monteros and Foucaud 1987;
one intriguing possibility is that the injured oligodendrocytes Giometto et al. 1990; Lin and Connor 1989b).
released iron that was taken up by the macrophages and subse- Whether the effect of the apo-transferrin is iron indepen-
quently returned to oligodendrocyte progenitor cells once the dent or if the apo-transferrin bound iron once it was injected
insult had subsided to allow oligodendrogenesis and myelina- into the brain has not been resolved. There are cell culture
tion to proceed. studies that show oligodendrocytes exposed to apo-transferrin
In the spinal cord injury model, there was very little demy- develop a multipolar morphology and increase expres-
elination (Schonberg et al. 2007). Thus, although this injury sion of MBP and myelin-associated glycoprotein (MAG).
model provides compelling evidence for an interrelation- Apo-transferrin exposure also inhibits the migration of oli-
ship among macrophages, iron, and oligodendrogenesis, the godendrocyte precursor cells and this effect is blocked by the
importance of this relationship for myelin production could transferrin receptor (Paez et al. 2002). Thus these data suggest
not be demonstrated. In the cuprizone-induced demyelina- that apo-transferrin supports maturation of oligodendrocyte
tion model, macrophages in the lesions expressed ferritin, but precursor cells. The data would also be consistent with the
because myelination proceeded when cuprizone was stopped, notion that transferrin can promote stabilization of oligoden-
the ferritin shifted to oligodendrocytes, which was followed drocytic processes (Ortiz et al. 2005). In the cuprizone model
by the appearance of myelin basic protein (MBP) (Adamo of demyelination, rats treated with apo-transferrin at the time
et al. 2006). These data strongly support a converse functional of cuprizone withdrawal had a significant improvement in
and temporal relationship between ferritin in macrophages remyelination compared to spontaneous recovery (Adamo et
and ferritin in oligodendrocytes and myelination. Why such al. 2006). That apo-transferrin injections could be effective in
a relationship could not exist and support remyelination in a a disease such as multiple sclerosis is supported by the observa-
demyelinating disease such as MS is not known but perhaps tions of transferrin receptor-positive oligodendrocytes in the
supports the argument for an immune response that destroys periplaque regions (Hulet et al. 1999b). Overall, the data sug-
new oligodendrocytes before they can remyelinate. One gest that transferrin is an effective nutrient in oligodendrocyte
hypothesis under investigation is that the Tim-2 receptor, the differentiation and maturation.
receptor for H-ferritin on oligodendrocytes, may interact with A major question, however, regarding the role of transferrin
semaphorin A from the immune system. Before the discovery in the development of oligodendrocytes is whether or not the
that Tim-2 binds H-ferritin, Sema4A was the only reported trophic influence of this protein is dependent or independent
ligand for Tim-2 (Chen et al. 2005; Kumanogoh et al. of iron. The injections of apo-transferrin into the brain paren-
2002). Semaphorins, such as class IV semaphorins (Sema4A chyma would surely encounter iron and given that the associa-
and Sema4D), are an example of shared signaling molecules tion constant of transferrin for iron is 1022M-1 apo-transferrin
between the immune and nervous systems (Kikutani and would acquire iron even if the iron were bound to many other
Kumanogoh 2003; Kumanogoh and Kikutani 2003). The proteins. Indeed when apo-transferrin was injected into the
hypothesis is that when oligodendrocytes reach a stage of cerebrospinal fluid, 52% of a 2-mg injection of apo-transferrin
maturation that they express Tim-2 for ferritin uptake, they was bound to iron within 4.5 hours (Ueda et al. 1993). That the
become targets for Semaphorin A. This notion is supported by effect of apo-transferrin is iron dependent is supported by the
our observations that immature oligodendrocytes surround- iron deficiency model where apo-transferrin injections were
ing MS lesions are transferrin receptor-positive but there is no not able to completely rescue the myelin deficit suggesting the
ferritin binding in the same region (Hulet et al. 1999b). iron was needed for the apo-transferrin effect (Badaracco et al.
Much more investigation is needed into the question of res- 2008). It cannot be ruled out, however, that there were insuf-
cuing myelin following injury or subsequent to developmental ficient numbers of oligodendrocytes to completely rescue the
iron deficiency. These investigations must focus on both the myelin.
timing of the rescue attempt as well as the mode of delivery The relationship between transferrin and oligodendro-
of the iron. For example, in a cell culture study, providing iron cytes was also investigated using hypotransferrinemic mice.
(ferric citrate) to glial-restricted precursor cells can increase the These mice have a splicing defect in the transferrin gene and
differentiation of these cells into galactocerebroside-expressing do not make transferrin. Hypotransferrinemic mice require
oligodendrocytes but providing iron to O2A progenitor cells transferrin injections to survive and the injected transferrin
resulted in increased proliferation but not differentiation is detected in oligodendrocytes of these mice (Dickinson and
(Morath and Mayer-Proschel 2001). The timing of delivery Connor 1995). Moreover, the hypotransferrinemic mice have

IRON AND GLIA • 593


increased expression of integral myelin proteins and higher surprising and inconsistent with some of the other findings in
than normal iron concentration in the white matter (Ortiz this study; for example there was a dramatic downregulation
et al. 2004b). These data clearly argue for a transferrin uptake of Tim-2, the receptor for ferritin in the white matter oligo-
mechanism in the oligodendrocytes, yet transferrin receptors dendrocytes in the sla mice, clearly suggesting significant iron
were not detectable on the oligodendrocytes (Dickinson and accumulation by these cells. Nonetheless, these studies dem-
Connor 1998). We cannot rule out that iron had accumulated onstrate a dynamic system for iron uptake and release from
in these cells early in development; but the injected transfer- oligodendrocytes.
rin from those early time points would have been degraded
over time. Assuming the uptake of transferrin into the oligo-
dendrocytes was a receptor-mediated endocytotic process, the 3 THE ROLE OF IRON IN
injected transferrin in the oligodendrocytes of the hypotrans- OLIGODENDROCY TIC VULNER ABILIT Y
ferrinemic mice should not have entered the cytosol because TO OX I DAT I VE S T R E S S
there is no endocytic export mechanism known for transfer-
rin, only iron. Thus, the transferrin in these cells should not Because oligodendrocytes are enriched in iron, have a high
have had an intracellular effect (e.g., stabilization of processes) rate of oxidative metabolism, and reside in the rich lipid envi-
beyond that of iron delivery. The hypotransferrinemic mice ronment of myelin, these cells can be expected to be highly
are an underutilized model for intracellular effects of transfer- vulnerable to oxidative damage. In addition, oligodendrocytes
rin in oligodendrocytes. have lower levels of endogenous antioxidant protection sys-
tems such as glutathione (Back et al. 1998). Oxidative damage
results from the interaction of iron with oxygen, which is nec-
2.4 I RO N E F F LUX
essary for the transport of electrons in the production of ATP.
Although much effort has been placed on understanding iron The presence of a relatively high concentration of catalase-rich
uptake by oligodendrocytes, recent data have shown that there peroxisomes in oligodendrocytes (Kim and Kim 1991) is tes-
is an iron export mechanism in place as well; and this mecha- timony to both the high rate of oxygen consumption in the
nism may differ between gray and white matter oligodendro- oligodendrocytes and the need to detoxify H2O2, a normal
cytes. The proteins of interest for iron export are hephastin by-product of oxygen metabolism that can interact with iron
and ferroportin. Hephastin is a copper-dependent ferroxidase to generate the free hydroxyl radical, a reaction known as the
initially identified in the duodenum wherein it was shown Fenton Reaction.
to be involved in iron efflux by enterocytes. In a cell culture The vulnerability of oligodendrocytes to oxidative stress
model from the cerebellum, all of the oligodendrocytes but has been clearly and consistently demonstrated (for two recent
none of the astrocytes expressed hephastin (Schulz et al. 2011). reviews see (Benarroch 2009; McTigue and Tripathi 2008;
In the sex-linked anemia (sla) mouse, there is a partial dele- chapter 52). In vivo, two particular mechanisms that involve
tion of the hephastin protein resulting in limited ferroxidase oligodendrocyte cell death and oxidative stress are hypoxia
activity (Chen et al. 2004; Vulpe et al. 1999). Ferroportin is and exposure to proinflammatory cytokines. Overexpression
a membrane protein that couples with ferroxidases to export of tumor necrosis factor (TNF)-D or interferon (INF)-J in
iron from cells (Vulpe et al. 1999). Ferroportin is more ubiq- the brains of transgenic mice results in severe central nervous
uitously expressed in astrocytes and oligodendrocytes and system (CNS) hypomyelination during development (Corbin
microglia. The expression of hephastin or ferroportin does et al. 1996; Probert et al. 1995). Both direct and indirect
not appear in culture until the oligodendrocytes have matured mechanisms can be identified for oligodendrocyte cell death
to express galactocerbroside. The appearance of hephastin and following exposure to cytokines. An example of the direct
ferroportin on oligodendrocytes has also been demonstrated mechanism is TNFD binding to p55 TNF receptor on oli-
in vivo (Schulz et al. 2011). godendrocytes followed by induction of apoptotsis ( Jurewicz
The role of hephastin in iron efflux by oligodendrocytes et al. 2005). Although this mechanism is direct, it can also
could be demonstrated using cells from that sla mice in cul- involve iron because TNFD is secreted from activated micro-
ture. In this study, in addition to the novel observations around glia. Production of this cytokine and others is influenced by the
hephastin, it is important to note the almost two-thirds of the iron content of the microglia and the degree of vulnerability
iron that is taken up by oligodendrocytes is released within of the oligodendrocytes is influenced by their iron content.
24 hours; a novel observation that there is iron release and In cell culture studies, TNFD and IFN-J are toxic to oli-
thus a dynamic iron concentration in oligodendrocytes. In godendrocytes in a dose-dependent manner but not interleu-
the oligodendrocytes from the sla mice almost no iron was kin (IL)-1E (Zhang et al. 2005b). The toxic concentration of
released, indicating the role of hephastin in the dynamics of TNFD and IFN-J can be decreased by exposing the cells to
regulation of cellular iron content in oligodendrocytes. The iron. Iron status of the oligodendrocytes had no impact on the
lack of iron release in the absence of hephastin by oligoden- lack of susceptibility to IL-1E. Interestingly, treatment with
drocytes was also shown in vivo, wherein more intense iron deferoxamine (an iron chelator) did not protect the oligoden-
staining was also seen in vivo in the oligodendrocytes from drocytes from the cytokines, perhaps supporting the evidence
the sla mice compared with control (Schulz et al. 2011). The that there are direct as well as indirect effects of the cytok-
increased iron-positive oligodendrocytes were more apparent ines on oligodendrocytes. The data also support the relative
in the gray matter than the white matter. These findings are vulnerability of oligodendrocytes to oxidative stress because

594 • NEUROGLIA
astrocytes and microglia actually accumulate iron in response ischemic insult during the first week of life in a developing
to activation through various injurious conditions including rat pup results in a marked increase in iron-positive micro-
inflammation (see later). The mechanism of action for toxicity glia (Palmer et al. 1999a; Rathnasamy et al. 2011). Likewise
of the cytokines to oligodendrocytes appeared to be through hypoxia/ischemia in the PND 7 rat pup results in an increase
loss of mitochondrial integrity because the anti-oxidant TPPB in ferritin-positive microglia and a delay in expression of
that targets mitochondria was protective. This observation is myelin markers. As myelin markers begin to appear, they are
consistent with in vivo studies in MS wherein mitochondrial accompanied by the appearance of ferritin-positive oligoden-
dysfunction is thought to underlie oligodendrocyte cell death drocytes and the disappearance of ferritin-positive microglia
(Kalman 2006). recapitulating the normal developmental pattern.
A curious observation in the cytokine exposure studies is The trophic relationship between the iron-enriched micro-
that the cytokines did not increase the expression of ferritin in glia in development and following injury or in demyelinating
the oligodendrocytes (Zhang et al. 2005b). Ferritin is typically diseases also presents potential problems. Microglia are able
increased following cytokine exposure, including in astrocytes to accumulate higher levels of iron and withstand higher
and microglia. The lack of ferritin increase is puzzling and amounts of oxidative stress than most cells (Robb and Connor
potentially important because it would be expected that such 1998; Zhang et al. 2006b). Indeed, microglia use iron to gen-
an increase would be part of the cellular protective mechanism. erate free radicals as part of a microbicidal defense mecha-
The lack of ferritin increase may couple with the relatively low nism (Fang 2011) but the generation of these free radicals
glutathione expression as part of the vulnerability profile of in the vicinity of the lipid rich myelin environment and the
the oligodendrocytes. The possible deleterious relationship iron rich oligodendrocytes could result in oxidative stress and
between microglia and oligodendrocytes involving iron and death of the oligodendrocytes. We have discussed earlier the
inflammation was investigated in a cell culture model (Zhang relationship between microglia, iron and oligodendrocytes
et al. 2006a). As discussed earlier, when conditioned media in the developing brain. The periventricular white matter, a
from iron-loaded microglia is placed on oligodendrocytes the region particularly susceptible to injury in preterm infants, is
media has a trophic effect and that trophic effect can be elimi- enriched in iron-positive oligodendrocytes and microglia and
nated by treating the microglia with siRNA for H-ferritin it has been argued that the vulnerability of this region is owing
(Zhang et al. 2006a). However, when the iron-loaded micro- to oxidative stress (French et al. 2009). Similarly, a potential
glia are treated by LPS, there is a decrease in H-ferritin release mechanism for oligodendrocyte death in multiple sclerosis
from the microglia and an increase in pro-inflammatory could be through production of free radicals by macrophages
cytokines. The conditioned-media from the activated micro- in the vicinity of the MS lesions (Fisher et al. 1988; Haider
glial cells is toxic to the oligodendrocytes. Treatment of the et al. 2012) (see chapter 61). Indeed, there is substantial histo-
microglia with an iron chelator blocks the LPS-induced acti- logical evidence of oxidative stress in MS (Haider et al. 2012).
vation of nuclear factor kappa-light-chain-enhancer of acti- In support of a role for iron and oxidative stress in promotion
vated B cells (NF-κB) in microglia and decreases the amount of demyelination is the finding of iron deposits in the white
of cytokines that are released. matter in MS (LeVine 1997) and that treatment with an iron
Another physiological event that has been associated with chelator or an antioxidant can limit the amount of demyeli-
damage to the white matter, in particular the periventricular nation and behavioral abnormalities that are associated with
white matter, is hypoxia. The hypoxic insult is associated with experimental allergic encephalomyelitis (Bowern et al. 1984;
increased levels of iron in serum and CSF (Shouman et al. Hartung et al. 1988; LeVine 1997). The relationship among
2008) and elevated iron staining in the periventricular white inflammation, hypoxia, iron, and microglia and oligodendro-
matter (Kaur and Ling 1999; Palmer et al. 1999b; Qi et al. cytes has been studied in cell culture models. In response to
1995; Rathnasamy et al. 2011). The iron in the periventricular hypoxia, microglia become activated and accumulate iron. In
white matter is found in both oligodendrocytes and micro- cell culture, exposure of microglia to hypoxia was coupled with
glia. Hypoxia leads to ferritin synthesis in oligodendrocytes in increased cellular iron consistent with the in vivo findings but
culture but the ferritin synthesis appears secondary to a release also production of reactive oxygen species, reactive nitrogen
of iron in the oligodendrocytes (Qi et al. 1995). The release species, and pro-inflammatory cytokines, all of which could
of iron in the hypoxic oligodendrocytes is associated with a be reversed by treating the microglia with an iron chelator.
decrease in intracellular pH. Thus, it appears that an increase When medium from the hypoxic microglia was transferred
in iron in the cytosol is an early event following hypoxia in to oligodendrocyte cultures, there was a decrease in expres-
oligodendrocytes. Ferritin in the oligodendrocytes may serve sion of glutathione in the oligodendrocytes. Glutathione
to sequester the iron and limit damage from hypoxia but this was increased in the oligodendrocytes, however, only if the
potentially protective ferritin reaction may be limited in vivo microglia-conditioned hypoxic medium contained deferox-
because of the interaction of iron and microglia and hypoxia. amine, an iron chelator. Deferoxamine alone had no effect on
Ferritin expression in oligodendrocytes and microglia in glutathione expression by oligodendrocytes. Lipid peroxida-
the periventricular white matter occurs in a spatiotemporal tion in oligodendrocytes was elevated following exposure to
relationship similar to that reported for iron (Cheepsunthorn the medium conditioned by hypoxic microglia and this effect
et al. 1998). The switch from ferritin-positive microglia to is reduced if the microglia were also treated with deferoxam-
ferritin-positive oligodendrocytes correlates temporally and ine in the media at the time of hypoxia. Exposure to TNFD
spatially with the appearance of myelin. Exposure to hypoxic/ or exposure to hypoxic-conditioned medium from microglia

IRON AND GLIA • 595


will increase caspase 3 activation in oligodendrocytes and this the blood-brain-barrier regarding brain iron status (Dringen
activation is reversed by deferoxamine or antiserum to TNFD et al. 2007).
(Rathnasamy et al. 2011). These data strongly implicate iron in The mechanism of iron uptake into astrocytes does not
the oligodendrocyte damage seen in hypoxia. appear to involve transferrin receptors because these are
Another area that involves oxidative stress damage to rarely seen on astrocytes in vivo, although they are expressed
oligodendrocytes is radiation used to treat brain tumors. on these cells in culture (Dringen et al. 2007). An interest-
Oligodendrocytes are the most radiosensitive of the glial cells ing model has been proposed that suggests astrocytes release
(Kim et al. 2008). No direct evidence could be found in the ascorbate that binds the ferrous (soluble) form of iron in the
literature for the contribution of iron to the radiosensitivity extracellular space and then this ascorbate-iron complex is
of oligodendrocytes. However, exposing oligodendrocytes imported into the astrocytes (Lane et al. 2011). Once inside
to blue light increases oxidative stress sixfold in these cells the cells the iron could be released from endosomes via diva-
(Thorburne and Juurlink 1996) and the increase in oxidative lent metal transporter 1 (DMT1) protein (Burdo et al. 2001).
stress can be prevented by iron chelation. This protein is required to move iron from the endosomes
into the cytosol. Astrocytes express amyloid precursor pro-
tein, which has recently been described as having ferroxidase
4 ASTROCY TES activity (Duce et al. 2010), which will likely result in evi-
dence that astrocytes and their iron status play a direct role
The role of astrocytes in iron neurobiology has mostly focused in plaque formation and possibly Alzheimer disease (Zhao
on the accumulation of iron in these cells following damage et al. 2011). As mentioned earlier, ferritin-positive astrocytes
or alterations in development of myelin associated with muta- are rare, which would indicate that little iron is being stored in
tions in myelin proteins that altered the health of oligoden- astrocytes. There is a unique release mechanism for iron from
drocytes (Connor and Menzies 1996b). Normally there is no astrocytes, ceruloplasmin (GPI-linked). It has been shown
detectable iron staining in astrocytes and rarely can ferritin- that ceruloplasmin-defective astrocytes will load iron but
positive astrocytes be found (Fig. 46.6). Recently, however, not release it (Patel and David 1997). Moreover, in individu-
compelling evidence has begun to emerge that astrocytes als or mouse models of aceruloplasminemia, astrocytes stain
have a role in maintaining iron homeostasis in the normal strongly for iron. This last comment deserves a caveat because
brain. Specifically, astrocytes may regulate extracellular iron the images of iron staining in these samples have the morpho-
availability in the brain and may be important for signaling logical appearance of oligodendrocytes but there is no question
that there are also ferritin-positive astrocytes in the acerulo-
plasmic brains (Oide et al. 2006; Texel et al. 2008). Astrocytes
release hepcidin, which is a protein that decreases iron release
from cells by binding to ferroportin. Thus, astrocytes through
secretion of hepcidin can regulate iron release from the BBB,
microglia (Du et al. 2012), and oligodendrocytes. Astrocytic
uptake of iron may be accelerated by hypoxia providing a
neuroprotective role (Yang et al. 2012). Thus, astrocytes may
have a critical role in brain iron homeostasis not only during
damage by accumulating iron but also by regulating brain iron
uptake from the BBB and release from other cells.
One additional possible source of iron to astrocytes, most
likely occurring only during damage, is hemin (Dang et al.
2012). Following injury, astrocytes strongly express heme-
oxygenase-1, which will degrade heme and release the iron.
Depending on the availability of ferritin, this iron could either
be stored or be a source of oxidative stress. The iron status of
the astrocytes will enhance their sensitivity to oxidative stres-
sors and treatment of astrocytes with an iron chelator will
minimize oxidative stress (Robb and Connor 1998). Iron
accumulation appears to cause damage to the mitochondria of
the astrocytes (Robb et al. 1999). Peroxide-induced death of
10 μm
astrocytes involves generation of superoxide radicals and loss
of mitochondrial membrane potential as well as depletion of
ATP (Robb et al. 1999; Song et al. 2006). Iron chelation will
enable astrocytes to maintain membrane potential and pre-
Figure 46.6 Ferritin Immunostaining of Glia in Human Putamen. serve production of ATP. However, because ATP production
Multiple glial cell-types express ferritin. The arrows indicate astrocytes requires iron, longer-term exposure of astrocytes to an iron
with multiple emanating projections protruding from the cell body.
The single arrowhead denotes a round oligodendrocyte, which is typical chelator is associated with a decline in ATP production. These
morphology for this cell-type. cell culture observations are directly relevant to histological

596 • NEUROGLIA
observations of increased peroxidase inclusions in astrocytes IgG secondary antibodies and thus the immunostaining may
of the substantia nigra with age and evidence of iron accu- have resulted from the interaction of the secondary antibody
mulation in mitochondria of astrocytes in Parkinson Disease to the Fc receptor on microglia (Kaur and Ling 1995). To date
(Dringen et al. 2007; Schipper et al. 2009) (also see chapters we have not seen transferrin receptors on microglia, resting or
51 and 65). activated.

5 MICROGLIA 6 S U M M A RY A N D P E R S P E C T I VE S

A common observation in activated microglia is that these Iron management in the brain may start with regulation of
cells accumulate iron (Berg et al. 2001; Gorter et al. 2005). iron transport and release from the BBB that is regulated by
Microglia can accumulate twice the amount of iron that an astrocytes through their ability to release hepcidin. The iron
oligodendrocyte can (Zhang et al. 2005c, 2006a). entering the brain, although not obligated to pass through
Iron enriched microglia are seen in demyelinating disor- glial endfeet, seems to require a ceruloplasmin-mediated pro-
ders such as MS (Adams 1988; Craelius et al. 1982; LeVine cess that is also associated with astrocytes. Astrocytes, under
1997) and in demyelination associated with Human immuno- normal conditions, appear to have little stored iron. The glial
deficiency virus (HIV) (Gelman et al. 1992), and excess iron cells most in need of iron for normal function appear to be
in the MS white matter is supported by neuroimaging studies oligodendrocytes. These cells may acquire iron via transferrin
(Haacke et al. 2009, 2010; Williams et al. 2012). For an excel- in earliest stages of development but appear to have their own
lent review on iron and MS, see Williams et al. (2012) and acquisition system: uptake of H-ferritin. One source of ferri-
chapter 61. When the microglia are iron laden, there are few tin in the brain is microglia although it is also possible for fer-
to no iron-enriched oligodendrocytes seen in the white mat- ritin to be transported across the BBB. Microglia accumulate
ter tracts consistent with our observations in development. iron in both early stages of normal development and following
Iron-laden microglia are not limited to degenerative processes injury and thus iron appears critical to functions associated
in the white matter but are also seen in many neurodegen- with these cells as well. Moreover, the iron in microglia can
erative diseases such as Alzheimer disease in the vicinity of be released to oligodendrocytes bound in ferritin and be used
neuritic plaques (Connor et al. 1992a) and in Parkinson dis- by the oligodendrocytes for maturation and myelin produc-
ease ( Jellinger et al. 1993; Kienzl et al. 1995) including animal tion (see Schematic 46.1). Oligodendrocytes are also the most
models of Parkinson disease (Goto et al. 1996), demyelination sensitive to low iron because hypomyelination is a consistent
(Forge et al. 1998; Pedchenko and LeVine 1998), and stroke finding of decreased iron availability both in development
(Kondo et al. 1995). and adults. There is some suggestion in the literature that iron
We have already mentioned the animal models of hypoxia/ changes in white matter tracts may be a biomarker for demy-
ischemia, where iron-enriched microglia are present in the elination, which could be clinically meaningful information
damaged area (Bidmon et al. 2001; Cheepsunthorn et al. to guide timing of treatment interventions. An exciting new
2001; Rathnasamy et al. 2011). Moreover, when oligodendro- area is the role of genetic variations that impact brain iron sta-
cyte development is compromised, iron is found predomi- tus and hence myelination—leading to alterations in rates of
nantly in microglia (Connor and Menzies 1990). Therefore, cognitive decline with age or degree of damage with disease.
microglia appear to play an important role in iron homoeosta- The balance of iron, however, is clearly critical to glial func-
sis in the injured brain. It is not known at this time if the iron tion because proinflammatory cytokines, hypoxia, and other
taken up by the microglia is part of the protective function means of damage to the brain will use the ability of iron to
of the microglia or if the iron is also used by the microglia. generate oxidative stress to promote cell death in all of the glial
Certainly, the metabolic activity of the activated microglial subtypes but the most sensitive are oligodendrocytes followed
cells is increased. The amount of proinflammatory cytokines by astrocytes and then microglia. The ability of iron chelation
released by microglia is increased by iron loading (Sindrilaru to protect oligodendrocytes not only from direct exposure to
et al. 2011; Zhang et al. 2006a) as well as increased release of proinflammatory cytokines but also from the toxic effects of
matrix metalloproteinase-9 (Mairuae et al. 2011) and activa- activated microglia suggest that attempts to treat hypoxic/
tion of NF-NB (Crichton et al. 2002). One way to minimize ischemic injury (Sorond and Ratan 2000) and demyelinating
the impact of iron on damage to the brain following insult diseases (Pedchenko and LeVine 1998) with iron chelators are
may be to limit iron in the diet. Rats fed an iron-deficient diet well founded. There is clear evidence that iron chelation will
in the kainite model of epilepsy suffered less damage through- decrease inflammatory reactions. A challenge will be in the
out the brain and had less microgliosis when compared with delivery of the iron chelator and the timing of delivery. The
those on a control diet. In the same model, rats fed an iron chelating compound or the mechanism of delivery must be
supplement diet had increased brain damage and microgliosis able to traverse the BBB and in adequate amounts. Moreover,
(Shoham and Youdim 2000). the chelating compound has to discern between “good and
Iron acquisition by macrophages occurs primarily through bad” iron so as not to limit energy production that may be
phagocytosis of damaged cells, myelin, and even red blood needed for remyelination. For example, we have shown (Robb
cells (RBC)s. Although one group has reported expression and Connor, 1998) that deferoxamine will protect astrocytes,
of transferrin receptors on microglia, these studies use whole specifically maintaining ATP production, from oxidative

IRON AND GLIA • 597


stressors but after a few hours of exposure ATP production Berg D, Gerlach M, Youdim MB, Double KL, Zecca L, Riederer P, et al.
declines presumably because iron needed for cytochrome oxi- 2001. Brain iron pathways and their relevance to Parkinson’s disease.
J Neurochem 79(2):225–236.
dase activity is diminishing. Thus, iron chelation is a viable Bidmon HJ, Emde B, Oermann E, Kubitz R, Witte OW, Zilles K.
clinical option to limit damage from hypoxia, ischemia, and 2001. Heme oxygenase-1 (HSP-32) and heme oxygenase-2 induc-
traumatic brain injury but mechanisms to regulate the chelat- tion in neurons and glial cells of cerebral regions and its relation to
ing compound both in terms of amount of iron being chelated iron accumulation after focal cortical photothrombosis. Exp Neurol
and the distribution of the chelator within the brain (perhaps 168(1):1–22.
Blissman G, Menzies S, Beard J, Palmer C, Connor J. 1996. The expres-
limiting it to the injury site) may be critical to its function. sion of ferritin subunits and iron in oligodendrocytes in neonatal
Lowering dietary iron may be one way to limit iron available porcine brains. Dev Neurosci 18:274–281.
to exacerbate damage associated with neurological trauma, Bloch B, Popovici T, Levin MJ, Tuil D, Kahn A. 1985. Transferrin gene
but again, iron must be reintroduced for remyelination and expression visualized in oligodendrocytes of the rat brain by using in
return to normal function. situ hybridization and immunohistochemistry. Proc Natl Acad Sci U
S A, 82:6706–6710.
Bourre JM, Pascal G, Durand G, Masson M, DumontO, Picotti MJ.
1984. Alterations in the fatty acid composition of rat brain cells
AC K N OW L E D G M E N T S (neurons, astrocytes and oligodendrocytes) and subcellular fractions
(myelin and synaptosomes) induced by a diet devoid of n-3 fatty acids.
I am grateful to the many outstanding students and postdoc- J Neurochem 43:342–348.
Bowern N, Ramshaw IA, Clark IA, Doherty PC. 1984. Inhibition
toral fellows who worked with me in generating the data over of autoimmune neuropathological process by treatment with an
25 years from our first observation of transferrin in oligo- iron-chelating agent. J Exp Med 160(5):1532–1543.
dendrocytes into an exciting field of research involving brain Burdo JR, Martin J, Menzies SL, Dolan KG, Romano MA, Fletcher RJ,
iron regulation and its importance in glial function. I am par- et al. 1999. Cellular distribution of iron in the brain of the Belgrade
ticularly grateful to current students Dominique Leitner and rat. Neuroscience 93(3):1189–1196.
Burdo JR, Menzies SL, Simpson IA, Garrick LM, Garrick MD,
Wint Nandar and postdoctoral fellow Dr. Amanda Snyder Dolan KG, et al. 2001. Distribution of divalent metal transporter
who helped generate the micrographs, schematics, and tables 1 and metal transport protein 1 in the normal and Belgrade rat.
in this chapter. Dominique and Dr. Snyder also provided J Neurosci Res 66(6):1198–1207.
valuable suggestions and editing of this manuscript. I am also Cammer W. 1984. Oligodendrocyte associated enzymes. New York:
grateful to the funding agencies that allowed us to pursue Plenum.
Cammer W, Snyder DS, Zimmerman TR Jr, Farooq M, Norton WT.
this work, primarily the National Institutes of Health, and 1982. Glycerol phosphate dehydrogenase, glucose-6-phosphate dehy-
also other funding agencies including the National Multiple drogenase, and lactate dehydrogenase: activities in oligodendrocytes,
Sclerosis Society. neurons, astrocytes, and myelin isolated from developing rat brains.
J Neurochem 38(2):360–367.
Carbajal KS, Miranda JL, Tsukamoto MR, Lane TE. 2011. CXCR4 sig-
naling regulates remyelination by endogenous oligodendrocyte pro-
REFERENCES genitor cells in a viral model of demyelination. Glia.
Casey JL, Koeller DM, Ramin VC, Klausner RD, Harford JB. 1989. Iron
Adamo AM, Paez PM, Escobar Cabrera OE, Wolfson M, Franco PG, regulation of transferrin receptor mRNA levels requires iron-respon-
Pasquini JM, et al. 2006. Remyelination after cuprizone-induced sive elements and a rapid turnover determinant in the 3’ untranslated
demyelination in the rat is stimulated by apotransferrin. Exp Neurol region of the mRNA. EMBO J 8:3693–3699.
198:519–529. Cheepsunthorn P, Palmer C, Connor JR. 1998. Cellular distribu-
Adams CW. 1988. Perivascular iron deposition and other vascu- tion of ferritin subunits in postnatal rat brain. J Comp Neurol
lar damage in multiple sclerosis. J Neurol Neurosurg Psychiatry 400(1):73–86.
51(2):260–265. Cheepsunthorn P, Palmer C, Menzies S, Roberts RL, Connor JR. 2001.
Back SA, Gan X, Li Y, Rosenberg PA, Volpe JJ. 1998. Maturation-dependent Hypoxic/ischemic insult alters ferritin expression and myelination in
vulnerability of oligodendrocytes to oxidative stress-induced death neonatal rat brains. J Comp Neurol 431(4):382–396.
caused by glutathione depletion. J Neurosci 18(16):6241–6253. Chen H AZ, Su T, Syed BA, Gao H, Alaeddine RM, Fox TC, et al.
Badaracco ME, Ortiz EH, Soto EF, Connor J, Pasquini JM. 2008. 2004. Hephaestin is a ferroxidase that maintains partial activity in
Effect of transferrin on hypomyelination induced by iron deficiency. sex-linked anemia mice. Blood 103:3933–3939.
J Neurosci Res 86(12):2663–2673. Chen TT, Li L, Chung DH, Allen CD, Torti SV, Torti FM, et al. 2005.
Bagnato F, Hametner S, Yao B, van Gelderen P, Merkle H, Cantor FK, TIM-2 is expressed on B cells and in liver and kidney and is a receptor
et al. Tracking iron in multiple sclerosis: a combined imaging and his- for H-ferritin endocytosis. J Exp Med 202(7):955–965.
topathological study at 7 Tesla. Brain 134(Pt 12):3602–3615. Connor JR. Benkovic SA. 1992. Iron regulation in the brain: histochem-
Bartlett WP, Li XS, Connor JR. 1991. Expression of transferrin ical, biochemical, and molecular considerations. Ann Neurol 32
mRNA in the CNS of normal and jimpy mice. J Neurochem Suppl:S51–61.
57(1):318–322. Connor JR, Boyer PJ, Menzies SL, Dellinger B, Allen RP, Ondo WG, Earley
Bartzokis G. 2004. Age-related myelin breakdown: a developmental CJ. 2003. Neuropathological examination suggests impaired brain
model of cognitive decline and Alzheimer’s disease. Neurobiol Aging iron acquisition in restless legs syndrome. Neurology 61:304–309.
25(1):5–18; author reply 49–62. Connor JR, Menzies SL. 1990. Altered cellular distribution of iron in
Beard JL, Wiesinger JA, Connor JR. 2003. Pre- and postweaning iron the central nervous system of myelin deficient rats. Neuroscience
deficiency alters myelination in Sprague-Dawley rats. Dev Neurosci 34(1):265–271.
25(5):308–315. Connor JR, Menzies SL. 1996. Relationship of iron to oligodendrocytes
Benarroch EE. 2009. Oligodendrocytes: susceptibility to injury and and myelination. Glia 17(2):83–93.
involvement in neurologic disease. Neurology 72:1779–1785. Connor JR, Menzies SL, St Martin SM, Mufson EJ. 1992a. A histochem-
Benkovic SA, Connor JR. 1993. Ferritin, transferrin, and iron in selected ical study of iron, transferrin, and ferritin in Alzheimer’s diseased
regions of the adult and aged rat brain. J Comp Neurol 338:97–113. brains. J Neurosci Res 31(1):75–83.

598 • NEUROGLIA
Connor JR, Pavlick G, Karli D, Menzies SL, Palmer C. 1995a. A his- transferrin gene. Similar elements govern transcription in oligoden-
tochemical study of iron-positive cells in the developing rat brain. drocytes and in a neuronal cell line. J Biol Chem 269:24504–24510.
J Comp Neurol 355(1):111–123. Fang FC. 2011. Antimicrobial actions of reactive oxygen species. MBio
Connor JR, Roskams AJ, Menzies SL, Williams ME. 1993. Transferrin 2(5):1–6.
in the central nervous system of the shiverer mouse myelin mutant. Fisher M, Levine PH, Weiner BH, Vaudreuil CH, Natale A, Johnson
J Neurosci Res 36(5):501–507. MH, et al. 1988. Monocyte and polymorphonuclear leukocyte toxic
Corbin JG, Kelly D, Rath EM, Baerwald KD, Suzuki K, Popko B. 1996. oxygen metabolite production in multiple sclerosis. Inflammation
Targeted CNS expression of interferon-gamma in transgenic mice 12(2):123–131.
leads to hypomyelination, reactive gliosis, and abnormal cerebellar Forge JK, Pedchenko TV, LeVine SM. 1998. Iron deposits in the central
development. Mol Cell Neurosci 7(5):354–370. nervous system of SJL mice with experimental allergic encephalomy-
Cortese S, Konofal E, Bernardina BD, Mouren MC, Lecendreux M. elitis. Life Sci 63(25):2271–2284.
2009. Sleep disturbances and serum ferritin levels in children Francois C, Nguyen-Legros J, Percheron G. 1981. Topographical and
with attention-deficit/hyperactivity disorder. Eur Child Adolesc cytological localization of iron in rat and monkey brains. Brain Rex
Psychiatry 18(7):393–399. 215:317–322.
Craelius W, Migdal MW, Luessenhop CP, Sugar A, Mihalakis I. 1982. French HM, Reid M, Mamontov P, Simmons RA, Grinspan JB. 2009.
Iron deposits surrounding multiple sclerosis plaques. Arch Pathol Oxidative stress disrupts oligodendrocyte maturation. J Neurosci Res
Lab Med 106(8):397–399. 87(14):3076–3087.
Crichton RR, Wilmet S, Legssyer R, Ward RJ. 2002. Molecular and cel- Friede RL. 1962. The cytochemistry of normal and reactive astrocytes. J
lular mechanisms of iron homeostasis and toxicity in mammalian Neuropathol Exp Neurol 21:471–478.
cells. J Inorg Biochem 91(1):9–18. Gebril OH, Simpson JE, Kirby J, Brayne C, Ince PG. 2011. Brain iron
Dang TN, Bishop GM, Dringen R, Robinson SR. 2012. The metabolism dysregulation and the risk of ageing white matter lesions. Neuromol
and toxicity of hemin in astrocytes. Glia 59(10):1540–1550. Med 13:289–299.
de Arriba Zerpa GA, Saleh MC, Fernandez PM, Guillou F, Espinosa de Gelman BB, Rodriguez-Wolf MG, Wen J, Kumar S, Campbell GR, Herzog
los Monteros A, de Vellis J, et al. 2000. Alternative splicing prevents N. 1992. Siderotic cerebral macrophages in the acquired immunode-
transferrin secretion during differentiation of a human oligodendro- ficiency syndrome. Arch Pathol Lab Med 116(5):509–516.
cyte cell line. J Neurosci Res 61(4):388–395. Giometto B, Bozza F, Argentiero V, Gallo P, Pagni S, Piccinno MG.
de los Monteros AE, Korsak RA, Tran T, Vu D, de Vellis J, Edmond J. 1990. Transferrin receptors in rat central nervous system. An immu-
2000. Dietary iron and the integrity of the developing rat brain: a nocytochemical study. J Neurol Sci 98:81–90.
study with the artificially-reared rat pup. Cell Mol Biol (Noisy-le- Gorter JA, Mesquita AR, van Vliet EA, da Silva FH, Aronica E. 2005. Increased
grand) 46(3):501–515. expression of ferritin, an iron-storage protein, in specific regions of the
Dickinson TK, Connor JR. 1995. Cellular distribution of iron, trans- parahippocampal cortex of epileptic rats. Epilepsia 46(9):1371–1379.
ferrin, and ferritin in the hypotransferrinemic (Hp) mouse brain. Goto K, Mochizuki H, Imai H, Akiyama H, Mizuno Y. 1996. An
J Comp Neurol 355(1):67–80. immuno-histochemical study of ferritin in 1-methyl-4-phenyl-
Dickinson TK, Connor JR. 1998. Immunohistochemical analysis of trans- 1,2,3,6-tetrahydropyridine (MPTP)-induced hemiparkinsonian
ferrin receptor: regional and cellular distribution in the hypotransfer- monkeys. Brain Res 724(1):125–128.
rinemic (hpx) mouse brain. Brain Res 801(1–2):171–181. Guizzetti P. 1915. Principali risultati dell’applicazione grossolona a
Dringen R, Bishop GM, Koeppe M, Dang TN, Robinson SR. 2007. fresco delle reazioni istochimiche del ferro sulsistema nervosa cen-
The pivotal role of astrocytes in the metabolism of iron in the brain. trale dell’uomo e di aluni mammiferi domestici. Rio. Patol nerv men?
Neurochem Res 32(11):1884–1890. 20:102–l 17.
Du F, Qian C, Qian ZM, Wu XM, Xie H, Yung WH, et al. 2012. Hepcidin Haacke EM, Garbern J, Miao Y, Habib C, Liu M. 2010. Iron stores and
directly inhibits transferrin receptor 1 expression in astrocytes via a cerebral veins in MS studied by susceptibility weighted imaging. Int
cyclic AMP-protein kinase a pathway. Glia 59(6):936–945. Angiol 29(2):149–157.
Duce JA, Tsatsanis A, Cater MA, James SA, Robb E, Wikhe K, et al. 2010. Haacke EM, Makki M, Ge Y, Maheshwari M, Sehgal V, Hu J, et al. 2009.
Iron-export ferroxidase activity of beta-amyloid precursor protein is Characterizing iron deposition in multiple sclerosis lesions using sus-
inhibited by zinc in Alzheimer’s disease. Cell 142(6):857–867. ceptibility weighted imaging. J Magn Reson Imaging 29(3):537–544.
Dwork AJ, Schon EA, Herbert J. 1988. Nonidentical distribution of Haider L, Fischer MT, Frischer JM, Bauer J, Hoftberger R, Botond G,
transferrin and ferric iron in human brain. Neuroscience 27(1): et al. 2012. Oxidative damage in multiple sclerosis lesions. Brain
333–345. 134(Pt 7):1914–1924.
Dziembowska M, Tham TN, Lau P, Vitry S, Lazarini F, Dubois-Dalcq M. Hallgren B, Sourander P. 1958. The effect of age on the non-haemin iron
2005. A role for CXCR4 signaling in survival and migration of neu- in the human brain. J Neurochem 3(1):41–51.
ral and oligodendrocyte precursors. Glia 50:258–269. Han J, Day JR, Connor JR, Beard JL. 2003. Gene expression of transfer-
Erb GL, Osterbur DL, LeVine SM. 1996. The distribution of iron in the rin and transferrin receptor in brains of control vs. iron-deficient rats.
brain: a phylogenetic analysis using iron histochemistry. Brain Res Nutr Neurosci 6(1):1–10.
Dev Brain Res 93(1–2):120–128. Hartung HP, Schafer B, Heininger K, Toyka KV. 1988. Suppression of
Espinosa de los Monteros A, Chiapelli F, Fisher RS, de Vellis J. 1988. experimental autoimmune neuritis by the oxygen radical scavengers
Transferrin: an early marker of oligodendrocytes in culture. Int J Dev superoxide dismutase and catalase. Ann Neurol 23(5):453–460.
Neurosci 6(2):167–175. Hentze MW, Caughman SW, Rouault TA, Barriocanal JG, Dancis A,
Espinosa de los Monteros A, de Vellis J. 1988. Myelin basic protein and Harford JB et al. 1987. Identification of the iron-responsive element
transferrin characterize different subpopulations of oligodendrocytes for the translational regulation of human ferritin mRNA. Science
in rat primary glial cultures. J Neurosci Res 21(2–4):181–187. 238:1570–1573.
Espinosa de los Monteros A, Foucaud B. 1987. Effect of iron and Hill JM, Ruff MR, Weber RJ, Pert CB. 1985. Transferrin receptors in
transferrin on pure oligodendrocytes in culture; characterization rat brain: neuropeptide-like pattern and relationship to iron distribu-
of a high-affinity transferrin receptor at different ages. Brain Res tion. Proc Natl Acad Sci U S A 82(13):4553–4557.
432(1):123–130. Hulet SW, Hess EJ, Debinski W, Arosio P, Bruce K, Powers S, et al.
Espinosa de los Monteros A, Kumar S, Zhao P, Huang CJ, Nazarian R, 1999a. Characterization and distribution of ferritin binding sites in
Pan T, et al. 1999. Transferrin is an essential factor for myelination. the adult mouse brain. J Neurochem 72(2):868–874.
Neurochem Res 24(2):235–248. Hulet SW, Heyliger SO, Powers S, Connor JR. 2000. Oligodendrocyte
Espinosa de los Monteros A, Sawaya BE, Guillou F, Zakin MM, de progenitor cells internalize ferritin via clathrin-dependent receptor
Vellis J, Schaeffer E. 1994. Brain-specific expression of the human mediated endocytosis. J Neurosci Res 61(1):52–60.

IRON AND GLIA • 599


Hulet SW, Powers S, Connor JR. 1999b. Distribution of transferrin Li R, Luo C, Mines M, Zhang J, Fan GH. 2006. Chemokine CXCL12
and ferritin binding in normal and multiple sclerotic human brains. induces binding of ferritin heavy chain to the chemokine receptor
J Neurol Sci 165(1):48–55. CXCR4, alters CXCR4 signaling, and induces phosphorylation
Hyden HaP, A. 1960. A cytophysiological study of the functional rela- and nuclear translocation of ferritin heavy chain. J Biol Chem 281:
tionship between oligodendroglia and nerve cells of Deiter’s nucleus. 37616–37627.
J Neurochemistry 6 57–72. Lin HH, Connor JR. 1989. The development of the transferrin-transferrin
Jahanshad N, Kohannim O, Hibar DP, Stein JL, McMahon KL, de receptor system in relation to astrocytes, MBP and galactocerebroside
Zubicaray GI, et al. 2012. Brain structure in healthy adults is related in normal and myelin-deficient rat optic nerves. Brain Res Dev Brain
to serum transferrin and the H63D polymorphism in the HFE gene. Res 49(2):281–293.
Proc Natl Acad Sci U S A. E851-9, Epub 2012. Lozoff B, Beard J, Connor J, Barbara F, Georgieff M, Schallert T. 2006.
Jellinger KA, Kienzl E, Rumpelmaier G, Paulus W, Riederer P, Long-lasting neural and behavioral effects of iron deficiency in
Stachelberger H, et al. 1993. Iron and ferritin in substantia nigra in infancy. Nutr Rev 64(5 Pt 2):S34–43; discussion S72–91.
Parkinson’s disease. Adv Neurol 60:267–272. Luders E, Narr KL, Hamilton LS, Phillips OR, Thompson PM, Valle JS,
Jurewicz A, Matysiak M, Tybor K, Kilianek L, Raine CS, Selmaj K. et al. 2009. Decreased callosal thickness in attention-deficit/hyperac-
2005. Tumour necrosis factor-induced death of adult human oligo- tivity disorder. Biol Psychiatry 65(1):84–88.
dendrocytes is mediated by apoptosis inducing factor. Brain 128(Pt Magaki S, Raghavan R, Mueller C, Oberg KC, Vinters HV, Kirsch WM.
11):2675–2688. 2007. Iron, copper, and iron regulatory protein 2 in Alzheimer’s dis-
Kalman B. 2006. Role of mitochondria in multiple sclerosis. Curr Neurol ease and related dementias. Neurosci Lett 418:72–76.
Neurosci Rep 6(3):244–252. Mairuae N, Connor JR, Cheepsunthorn P. 2011. Increased cellular iron
Kaur C, Ling EA. 1995. Transient expression of transferrin receptors levels affect matrix metalloproteinase expression and phagocytosis in
and localisation of iron in amoeboid microglia in postnatal rats. activated microglia. Neurosci Lett 500(1):36–40.
J Anat 186 (Pt 1):165–173. Marta CB, Escobar Cabrera OE, Garcia CI, Villar MJ, Pasquini JM,
Kaur C, Ling EA. 1999. Increased expression of transferrin receptors and Soto EF. 2000. Oligodendroglial cell differentiation in rat brain is
iron in amoeboid microglial cells in postnatal rats following an expo- accelerated by the intracranial injection of apotransferrin. Cell Mol
sure to hypoxia. Neurosci Lett 262(3):183–186. Biol (Noisy-le-grand) 46(3):529–539.
Kienzl E, Puchinger L, Jellinger K, Linert W, Stachelberger H, Jameson Marta CB, Ortiz EH, Hallak M, Baron B, Guillou F, Zakin MM, et al.
RF. 1995. The role of transition metals in the pathogenesis of 2002. Changes in the expression of cytoskeletal proteins in the CNS
Parkinson’s disease. J Neurol Sci 134 Suppl:69–78. of transferrin transgenic mice. Dev Neurosci 24(2–3):242–251.
Kikutani H, Kumanogoh A. 2003. Semaphorins in interactions Mash DC, Pablo J, Flynn DD, Efange SM, Weiner WJ. 1990.
between T cells and antigen-presenting cells. Nat Rev Immunol Characterization and distribution of transferrin receptors in the rat
3(2):159–167. brain. J Neurochem 55(6):1972–1979.
Kim JH, Brown SL, Jenrow KA, Ryu S. 2008. Mechanisms of McTigue DM, Tripathi RB. 2008. The life, death and replacement of oli-
radiation-induced brain toxicity and implications for future clinical godendrocytes in the adult CNS. J Neurochem 107:1–19.
trials. J Neurooncol 87:279–286. Moos T. 1996. Immunohistochemical localization of intraneuronal
Kim Y, Kim SU. 1991. Oligodendroglial cell death induced by oxygen transferrin receptor immunoreactivity in the adult mouse central
radicals and its protection by catalase. J Neuroscience Research nervous system. J Comp Neurol 375: 675–692.
29:100–106. Moos T, Rosengren Nielsen T, Skjørringe T, Morgan EH. 2007. Iron
Kondo Y, Ogawa N, Asanuma M, Ota Z, Mori A. 1995. Regional dif- trafficking inside the brain. J Neurochem 103:1730–1740.
ferences in late-onset iron deposition, ferritin, transferrin, astrocyte Morath DJ, Mayer-Proschel M. 2001. Iron modulates the differentiation
proliferation, and microglial activation after transient forebrain isch- of a distinct population of glial precursor cells into oligodendrocytes.
emia in rat brain. J Cereb Blood Flow Metab 15(2):216–226. Dev Biol 237(1):232–243.
Kugler P. 1994. Glucose-6-phosphate dehydrogenase is enriched in oli- Møllgård K, Dziegielewska KM, Saunders NR, Zakut H. Soreq H.
godendrocytes of the rat spinal cord. Enzyme histochemical and 1988. Synthesis and localization of plasma proteins in the developing
immunocytochemical studies. Histochemistry 101(2):143–153. human brain. Integrity of the fetal blood-brain barrier to endogenous
Kumanogoh A, Kikutani H. 2003. Roles of the semaphorin family in proteins of hepatic origin. Dev Biol 128:207–221.
immune regulation. Adv Immunol 81:173–198. Nelissen K, Mulder M, Smets I, Timmermans S, Smeets K, Ameloot M,
Kumanogoh A, Marukawa S, Suzuki K, Takegahara N, Watanabe C, et al. 2012. Liver X receptors regulate cholesterol homeostasis in oli-
Ch’ng E, et al. 2002. Class IV semaphorin Sema4A enhances T-cell godendrocytes. J Neurosci Res 90(1):60–71.
activation and interacts with Tim-2. Nature 419(6907):629–633. Nemeth E. Ganz T. 2006. Regulation of iron metabolism by hepcidin.
Lane DJ, Robinson SR, Czerwinska H, Bishop GM, Lawen A. 2011. Two Annu Rev Nutr 26:323–42.
routes of iron accumulation in astrocytes: ascorbate-dependent fer- Nguyen-Legros J, Bizot J, Bolesse M, Pulicani JP. 1980. “Noir de diamin-
rous iron uptake via the divalent metal transporter (DMT1) plus an obenzidine”: une nouvelle methode histochimique de revelation
independent route for ferric iron. Biochem J 432(1):123–132. du fer exogene. [“Diaminobenzidine black” as a new histochemical
Lavi E, Strizki JM, Ulrich AM, Zhang W, Fu L, Wang Q et al. 1997. demonstration of exogenous iron (author’s transl)]. Histochemistry
CXCR-4 (Fusin), a co-receptor for the type 1 human immunode- 66:239–244.
ficiency virus (HIV-1), is expressed in the human brain in a variety Oide T, Yoshida K, Kaneko K, Ohta M, Arima K. 2006. Iron overload
of cell types, including microglia and neurons. Am J Pathol 151: and antioxidative role of perivascular astrocytes in aceruloplasmine-
1035–1042. mia. Neuropathol Appl Neurobiol 32(2):170–176.
LeVine SM. 1991. Oligodendrocytes and myelin sheaths in normal, Oloyede OB, Folayan AT, Odutuga AA. 1992 Effects of low iron status
quaking and shiverer brains are enriched in iron. J Neurosci Res and deficiency of essential fatty acids on some biochemical constitu-
29(3):413–419. ents of rat brain. Biochem Int 27 913–922.
LeVine SM. 1997. Iron deposits in multiple sclerosis and Alzheimer’s dis- Ortiz E, Pasquini JM, Thompson K, Felt B, Butkus G, Beard J, et al.
ease brains. Brain Res 760(1–2):298–303. 2004a. Effect of manipulation of iron storage, transport, or availabil-
Levison SW, Chuang C, Abramson BJ, Goldman JE. 1993. The migra- ity on myelin composition and brain iron content in three different
tional patterns and developmental fates of glial precursors in the animal models. J Neurosci Res 77(5):681–689.
rat subventricular zone are temporally regulated. Development Ortiz EH PL, Soto EF, Pasquini JM. 2005. Apotransferrin and
119(3):611–622. the cytoskeleton of oligodendroglial cells. J Neurosci Research
LeVine SM, Goldman JE. 1988. Embryonic divergence of oligodendro- 82:822–830.
cyte and astrocyte lineages in developing rat cerebrum. J Neurosci Paez PM, Marta CB, Moreno MB, Soto EF, Pasquini JM. 2002.
8(11):3992–4006. Apotransferrin decreases migration and enhances differentiation of

600 • NEUROGLIA
oligodendroglial progenitor cells in an in vitro system. Dev Neurosci Shimizu T, Wolfe LS. 1990. Arachidonic acid cascade and signal trans-
24(1):47–58. duction. J Neurochem 55:1–15.
Palmer C, Menzies SL, Roberts RL, Pavlick G, Connor JR. 1999a. Shoham S, Youdim MB. 2000. Iron involvement in neural damage and
Changes in iron histochemistry after hypoxic-ischemic brain injury microgliosis in models of neurodegenerative diseases. Cell Mol Biol
in the neonatal rat. J Neurosci Res 56(1):60–71. (Noisy-le-grand) 46(4):743–760.
Patel BN, David S. 1997. A novel glycosylphosphatidylinositol-anchored Shouman BO Mesbah A, Aly H 2008. Iron metabolism and lipid per-
form of ceruloplasmin is expressed by mammalian astrocytes. J Biol oxidation products in infants with hypoxic ischemic encephalopathy.
Chem 272(32):20185–20190. J Perinatol 28:487–491.
Patel BN, Dunn RJ, David S. 2000. Alternative RNA splicing generates Sindrilaru A, Peters T, Wieschalka S, Baican C, Baican A, Peter H, et al.
a glycosylphosphatidylinositol-anchored form of ceruloplasmin in 2011. An unrestrained proinflammatory M1 macrophage population
mammalian brain. J Biol Chem 275:4305–4310. induced by iron impairs wound healing in humans and mice. J Clin
Patel JR, McCandless EE, Dorsey D, Klein RS. 2010. CXCR4 promotes Invest 121(3):985–997.
differentiation of oligodendrocyte progenitors and remyelination. Skoff RP, Toland, D, and Nast E. 1980. Pattern of myelination and dis-
Proc Natl Acad Sci U S A 107:11062–11067. tribution of neuroglial cells along the developing optic system of the
Pedchenko TV, LeVine SM. 1998. Desferrioxamine suppresses experi- rat and rabbit. J Comp Neurol 191:237–252.
mental allergic encephalomyelitis induced by MBP in SJL mice. Song W, Su H, Song S, Paudel HK, Schipper HM. 2006. Over-expression
J Neuroimmunol 84(2):188–197. of heme oxygenase-1 promotes oxidative mitochondrial damage in rat
Pleasure D, Abramsky O, Silberberg D, Quinn B, Parris J, Sida T. 1977. astroglia. J Cell Physiol 206(3):655–663.
Lipid synthesis by an oligodendroglial fraction in suspension culture. Sorond FA, Ratan RR. 2000. Ironing-out mechanisms of neuronal
Brain Research 134:377–382. injury under hypoxic-ischemic conditions and potential role of iron
Probert L, Akassoglou K, Pasparakis M, Kontogeorgos G, Kollias chelators as neuroprotective agents. Antioxid Redox Signal 2(3):
G. 1995. Spontaneous inflammatory demyelinating disease in 421–436.
transgenic mice showing central nervous system-specific expres- Sow A, Lamant M, Bonny JM, Larvaron P, Piaud O, Lecureuil C, et al.
sion of tumor necrosis factor alpha. Proc Natl Acad Sci U S A 2006. Oligodendrocyte differentiation is increased in transferrin
92(24):11294–11298. transgenic mice. J Neurosci Res 83(3):403–414.
Qi Y, Jamindar TM, Dawson G. 1995. Hypoxia alters iron homeosta- Spatz H. 1922. Eisennachweis im Gehirn, besonders in Zentren des
sis and induces ferritin synthesis in oligodendrocytes. J Neurochem extrapyramidalen Systems. Z ges Neurol Psychiat 77:,261–390.
64(6):2458–2464. Taylor EM, Crowe A, Morgan EH. 1991. Transferrin and iron
Rajan KS, Colburn RW, Davis JM. 1976. Distribution of metal ions uptake by the brain: effects of altered iron status. J Neurochem
in the subcellular fractions of several rat brain areas. Life Sci 57(5):1584–1592.
18(4):423–431. Taylor EM, Morgan EH. 1990. Developmental changes in transferrin
Rathnasamy G, Ling EA, Kaur C. 2011. Iron and iron regulatory pro- and iron uptake by the brain in the rat. Brain Res Dev Brain Res
teins in amoeboid microglial cells are linked to oligodendrocyte 55(1):35–42.
death in hypoxic neonatal rat periventricular white matter through Texel SJ, Xu X, Harris ZL. 2008. Ceruloplasmin in neurodegenerative
production of proinflammatory cytokines and reactive oxygen/ diseases. Biochem Soc Trans 36(Pt 6):1277–1281.
nitrogen species. J Neurosci 31(49):17982–17995. Thorburne SK, Juurlink BH. 1996. Low glutathione and high iron gov-
Robb SJ, Connor JR. 1998. An in vitro model for analysis of oxidative ern the susceptibility of oligodendroglial precursors to oxidative
death in primary mouse astrocytes. Brain Res 788(1–2):125–132. stress. J Neurochem 67(3):1014–1022.
Robb SJ, Robb-Gaspers LD, Scaduto RC Jr., Thomas AP, Connor JR. Todorich B, Pasquini JM, Garcia CI, Paez PM, Connor JR. 2009.
1999. Influence of calcium and iron on cell death and mitochon- Oligodendrocytes and myelination: the role of iron. Glia 57(5):467–478.
drial function in oxidatively stressed astrocytes. J Neurosci Res Todorich B, Zhang X, Connor JR. 2011. H-ferritin is the major source of
55(6):674–686. iron for oligodendrocytes. Glia 59(6):927–935.
Roy CN, Custodio AO, de Graaf J, Schneider S, Akpan I, Montross LK, Todorich B, Zhang X, Slagle-Webb B, Seaman WE, Connor JR.
et al. 2004. An Hfe-dependent pathway mediates hyposideremia in 2008. Tim-2 is the receptor for H-ferritin on oligodendrocytes.
response to lipopolysaccharide-induced inflammation in mice. Nat J Neurochem 107(6):1495–1505.
Genet 36:481–485. Ueda F, Raja KB, Simpson RJ, Trowbridge IS, Bradbury MW. 1993. Rate
Saleh MC, Espinosa de los Monteros A, de Arriba Zerpa GA, Fontaine of 59Fe uptake into brain and cerebrospinal fluid and the influence
I, Piaud O, Djordjijevic D, et al. 2003. Myelination and motor coor- thereon of antibodies against the transferrin receptor. J Neurochem
dination are increased in transferrin transgenic mice. J Neurosci Res 60(1):106–113.
72(5):587–594. Vulpe CD, Kuo YM, Murphy TL, Cowley L, Askwith C, Libina N,
Sanchez-Abarca LI, Tabernero A, Medina JM. 2001. Oligodendrocytes et al. 1999. Hephaestin, a ceruloplasmin homologue implicated in
use lactate as a source of energy and as a precursor of lipids. Glia intestinal iron transport, is defective in the sla mouse. Nat Genet
36(3):321–329. 21:195–199.
Sanyal B, Polak PE, Szuchet S. 1996. Differential expression of the Warringa RA, Hoeben RC, Koper JW, Sykes JE, van Golde LM,
heavy-chain ferritin gene in non-adhered and adhered oligodendro- Lopes-Cardozo M. 1987. Hydrocortisone stimulates the develop-
cytes. J Neurosci Res 46(2):187–197. ment of oligodendrocytes in primary glial cultures and affects glu-
Schipper HM, Song W, Zukor H, Hascalovici JR, Zeligman D. 2009. cose metabolism and lipid synthesis in these cultures. Brain Res
Heme oxygenase-1 and neurodegeneration: expanding frontiers of 431(1):79–86.
engagement. J Neurochem 110(2):469–485. Watanabe Y, Takahashi A, Shimazu T. 1990. Neural control of bio-
Schonberg DL, McTigue DM. 2009. Iron is essential for oligodendro- synthesis and secretion of serum transferrin in perfused rat liver.
cyte genesis following intraspinal macrophage activation. Exp Neurol Biochem J 267(2):545–548.
218(1):64–74. Williams R, Buchheit CL, Berman NE, LeVine SM. 2012. Pathogenic
Schonberg DL, Popovich PG, McTigue DM. 2007. Oligodendrocyte gen- implications of iron accumulation in multiple sclerosis. J Neurochem
eration is differentially influenced by toll-like receptor (TLR) 2 and 120(1):7–25.
TLR4-mediated intraspinal macrophage activation. J Neuropathol Wrigglesworth J, Baum H. 1988. Iron dependent enzymes in the brain.
Exp Neurol 66(12):1124–1135. In Brain Iron: Neurochemical and Behavioural Aspects, ed. Y. MB,
Schulz KV, Vulpe CD, Harris LZ, David S. 2011. Iron efflux from oli- 25–66. London: Taylor and Francis.
godendrocytes is differentially regulated in gray and white matter. Wu LJ, Leenders AG, Cooperman S, Meyron-Holtz E, Smith S, Land
J Neurosci 31(37):13301–13311. W, et al. 2004. Expression of the iron transporter ferroportin in

IRON AND GLIA • 601


synaptic vesicles and the blood-brain barrier. Brain Res 1001: Zhang J, Stanton DM, Nguyen XV, Liu M, Zhang Z, Gash D, et al.
108–17. 2005a. Intrapallidal lipopolysaccharide injection increases iron and
Yang L, Fan M, Du F, Gong Q, Bi ZG, Zhu ZJ, et al. 2012. Hypoxic ferritin levels in glia of the rat substantia nigra and induces locomotor
preconditioning increases iron transport rate in astrocytes. Biochim deficits. Neuroscience 135(3):829–838.
Biophys Acta 1822(4):500–508. Zhang X, Haaf M, Todorich B, Grosstephan E, Schieremberg H,
Yu GS, Steinkirchner TM, Rao GA, Larkin EC. 1986. Effect of pre- Surguladze N, et al. 2005b. Cytokine toxicity to oligodendrocyte
natal iron deficiency on myelination in rat pups. Am J Pathol precursors is mediated by iron. Glia 52(3):199–208.
125(3):620–624. Zhang X, Surguladze N, Slagle-Webb B, Cozzi A, Connor JR. 2006a.
Zakin MM, Baron B, Guillou F. 2002. Regulation of the tissue-specific Cellular iron status influences the functional relationship between
expression of transferrin gene. Dev Neurosci 24(2–3):222–226. microglia and oligodendrocytes. Glia 54(8):795–804.
Zechel S, Huber-Wittmer K, von Bohlen O, Halbach. 2006. Distribution Zhao J, O’Connor T, Vassar R. 2011. The contribution of activated
of the iron-regulating protein hepcidin in the murine central nervous astrocytes to Abeta production: implications for Alzheimer’s disease
system. J Neurosci Res 84:790–800. pathogenesis. J Neuroinflammation 8:150.

602 • NEUROGLIA
MICROGLIA

603
This page intentionally left blank
47.
ROLE OF MICROGLIA IN THE NORMAL BRAIN
Frank Kirchhoff

A B B R E VI AT I O N S In this chapter the dynamic behavior of microglia and their


role for normal brain function is discussed and the following
2P-LSM two-photon laser-scanning microscopy points are addressed: the properties of microglial motility, its
5c-Nase 5c-nucleotidase mechanism and regulation as well as the role of microglia dur-
AMPA 2-amino-3-(5-methyl-3-oxo-1,2- oxazol- ing brain development in neuronal elimination and synaptic
4-yl)propanoic acid wiring (Fig. 47.1).
BDNF brain-derived neurotrophic factor
bFGF basic fibroblast growth factor
CNS central nervous system
EGFP enhanced green fluorescent protein 2 S U RVE I L L A N C E O F B R A I N F U N C T I O N
GFAP glial fibrillary acidic protein BY R A M I F I E D M I C R O G L I A
KCC K+-Cl– cotransporter
NBC1 Na+/bicarbonate cotransporter 1 Soon after entering the brain from the periphery, the amoe-
NDPase nucleoside diphosphatase boid precursor cells of microglia acquire a typical morphol-
NGF nerve growth factor ogy characterized by small rod-shaped somata with numerous
NO nitric oxide thin and highly ramified processes that extend radially. Their
NTPase nucleoside triphosphatase three-dimensional distribution in vivo appears rather homo-
PI3K phosphatidylinositol 3c-kinase geneous with a territorial organization and a typical cell-to-
PLOSL polycystic lipomembranous osteodysplasia cell distance of 50 to 60 μm. This kind of microglial cell has
with sclerosing leukoencephalopathy originally been described as resting. Indeed, if one monitors
PNPase purine nucleoside phosphorylase the position of microglial somata for several hours in vivo
SIRP signal regulatory protein using two-photon laser-scanning microscopy (2P-LSM)
TGF-E transforming growth factor-E (Helmchen and Denk 2005), the cell bodies generally remain
TNF-D tumor necrosis factor D stationary with only few signs of migration. Only 5% of
TSP thrombospondin microglial cells move their position minimally, by 1 to 2 μm/
trk tropomyosin-related kinase hour (Nimmerjahn et al. 2005). Since 2005 in vivo time-lapse
recordings of microglia became one of the most frequently
1 INTRODUCTION used experimental approaches to study their functional role.
In a transgenic mouse line in which the fractalkine receptor
Microglial glial cells represent a unique and abundant con- gene (cx3cr1) had been replaced by the open reading frame of
stituent of the central nervous system (CNS). Originating the enhanced green fluorescent protein (EGFP) all microglial
from primitive myeloid precursor cells of the embryonic yolk cells are fluorescently tagged and can be brightly and at high
sac, microglial cells populate the brain before developmental spatial resolution visualized by 2P-LSM ( Jung et al. 2000;
closure of the blood-brain barrier (Ginhoux et al. 2010). In Nimmerjahn et al. 2005).
the adult nervous system, they are almost evenly distributed In contrast to the somata microglial processes appear
throughout the various brain regions at a density of about highly motile with two different types of morphology
6,000 to 8,000 cells/mm3. Thereby, they roughly form 5% to changes: (1) growth and retraction of the cell processes them-
15% of the cellular elements with some variations in different selves, as well as (2) filopodia-like membrane protrusions that
brain regions and species (Lawson et al. 1990; Mittelbronn transiently appear at random locations along processes. These
et al. 2001). In general, they are described to comprise the structural changes of the processes as well as of the filopodia
main component of the innate immune system in the brain occur on a timescale of minutes (Fig. 47.2). Extensions and
by responding to any pathological insult. More recently, func- retractions display similar velocities of 1 to 3 μm/minute for
tions of microglia have been discovered that appear to be more processes and up to 4 μm/minute for filopodia. In general,
important for normal brain functions and that—in a strict thicker branches show slower extensions than thinner pro-
sense—are not related to immunological responses. However, cesses of higher order. The filopodia-like protrusions are of
our understanding of microglial function in the healthy ner- variable shape, but typically they form bulbous endings. These
vous system is only emerging and still limited. protrusions appear transiently for a few minutes, sometimes

605
~5mm
Motility Synaptic interactions Phagocytosis

Figure 47.1 Overview of Microglial Behavior in the Healthy Brain. Highly motile microglial processes continuously remodel their local environment
(left), structurally and functionally interact with synaptic elements (middle; dendritic branch and spines, green) through direct contacts and exchanges
of molecular signals, and contribute to restructuring of neuronal circuits by phagocytosing synaptic elements and newborn cells (right; cellular
inclusions, blue and green). Microglial morphology and behavior display variability across central nervous system regions and stages of the
lifespan. Figure from Tremblay and Majewska 2011.

microglia can continuously monitor the activities of neigh-


boring cells and effectively control the microenvironment.
In addition, with its phagocytic potential it can directly act
like a communal waste collection service and clear the paren-
chyma of accumulated metabolic products and deteriorated
tissue components. Indeed, membrane protrusions spontane-
ously engulf tissue components that can be further processed
in the microglial cell body.

3 M E C H A N I S M S A N D R E GU L AT I O N O F
Figure 47.2 Dynamic Motility of Microglial Processes in the Normal M I C R O G L I A L M OT I L I T Y, S W E L L I N G ,
Brain. Ten minutes of microglial movements in the mouse cortex (A)
and spinal cord (B). (Red) Process retractions at the start of recording.
A N D C Y TO S K E L ETO N R E A R R A N G E M E N T
(Green) Processes that extended and retracted during the recording.
(Blue) Process extensions. By addressing microglial motility, one has to distinguish mainly
three forms of cell movement and migration: (1) the baseline
even repeatedly, along main processes and at their terminal activity of microglial motility that is generated by everlast-
endings. Frequently, the protrusive activity is interrupted for ing extensions and retractions of microglial lamellipodia and
several minutes before further extensions or retractions occur. filopodia as well as being an indicator of their tissue survey-
Despite this dynamic change of motility, the number of tran- ing function, (2) directed growth of single processes along the
sient protrusions per cell (about 20 in the cortical gray matter) gradient of a locally released chemoattractant, and (3) cellular
and the total process length remains constant. Although the migration encompassing translocation of cell bodies. The lat-
morphological changes appear randomly, the overall motility ter two can be regarded as graded forms of cell activation.
is as fast and abundant that each point of the extracellular ter- Identifying the mechanisms of microglial motility and
ritory adjacent to a single microglia cell is encountered three to migration is still an intense area of research. Currently, swell-
four times during a day. Extended areas of extracellular space ing with the involvement of ion transporters and channels as
that appear as translucent pockets in electron micrographs fre- well as actin-mediated processes are considered. In particular,
quently surround microglial processes (Tremblay et al. 2010). the surveying motility of filopodia and lamellipodia seems
Probably, microglial cells themselves generate these regions by to be mediated by regulatory volume increases and decreases
secreting proteases such as matrix-metalloproteinases to facili- (Zierler et al. 2008). Process extension would then require the
tate the process movement (Crocker et al. 2008). controlled influx of osmolytes and water. And indeed, asym-
Although the resident microglia of the healthy, unper- metrical distributions of transporters and ion channels have
turbed brain have previously been called “resting,” these cells been observed. K+-Cl– cotransporter (KCC) mediate the
are structurally the most dynamic cells of the CNS. It is not osmolyte influx and induce process swelling. Cl– influx persists
too tempting to speculate that the dynamic but persistent because of activation of swelling-activated Cl– channels. In the
motility fulfills housekeeping functions such as the removal opposite direction, K+-efflux is evoked by intracellular Ca2+
of cellular debris or toxic metabolites. The processes seem to oscillations triggering Ca2+-activated K+ channels. Also, coop-
sample their three-dimensional territories. Interestingly, bor- eration of Na+/H+ exchangers and Na+-K+/2Cl– cotransport-
der zones between neighboring microglial cells are nonover- ers have been implicated in local swelling. For the NaHCO3
lapping. When microglial processes encounter one of another cotransporter NBC1 and the KCC transporters a polarized
cell, endings mutually repelled each other. Equipped with distribution has been suggested. The cytokine CCL21 that is
numerous transmitters, growth factor and cytokine receptor expressed after neuronal injury affects microglial motility. Its

606 • NEUROGLIA
action as a chemoattractant is accompanied by induction of a effectively activate the numerous purinoceptors of P1 and
distinct Cl– conductance (Rappert et al. 2002). P2-type in an autocrine and/or paracrine manner. Microglia
Part of the microglial motility is also mediated by the express all P1 receptors, that is, the adenosine receptors A1,
actin-based cytoskeleton because it can be blocked by cytoch- A2A, A2B, and A3. In addition, they express the ionotropic
alasin B. Interestingly, the rapid depolymerization and repo- P2 receptors P2X4 and P2X7 as well as the metabotropic P2
lymerization, as well as the cytoplasmic redistribution of actin receptors P2Y6 and P2Y12. Thereby, microglia are not only
filaments, is induced by microglial 2-amino-3-(5-methyl- the cell type expressing the most diverse set of purinergic sig-
3-oxo-1,2-oxazol-4-yl)propanoic acid (AMPA) type glutamate naling molecules, it is perfectly equipped to be the best sentinel
receptors (Noda et al. 2000). Thereby, microglial motility is at of the evolutionary oldest transmitter system. Although ATP
least partially modulated by excitatory neuronal activity. is regarded as an alert signal, adenosine has been attributed
Another important regulator of microglial motility is rep- a more neuroprotective function. The distinct spatially and
resented by components of the complement system. Short temporally defined composition of the extracellular milieu in
application of C5a induces a ruffling of microglial membranes purines such as ATP, ADP, and adenosine represents a sophis-
within seconds (Nolte et al. 1996). Subsequently, a con- ticated trigger signal for the motility and migratory behavior
comitant extension of lamellipodia and a change of the actin of microglia.
cytoskeleton are observed. Although the activation of C5a Equipped with this diversity of nucleotide sensing mol-
receptors induces intracellular rises of Ca2+, the microglial ecules, these receptors are used in distinct pathways. In the
motility appears to be independent from intracellular Ca2+. context of motility the ADP-preferring P2Y12 receptor is
However, the motility is effectively blocked by the G protein particularly important (Haynes et al. 2006). In the brain this
inhibitor pertussis toxin. Although C5a receptors are also receptor is predominantly expressed on microglia on the pro-
found to be coupled to K+ channels, and therefore might be cesses as well as on the somata. As soon as the receptors evenly
involved in swelling-mediated process extensions, these chan- distributed along the processes sense a gradient of ADP/ATP
nels are not linked to regulation of microglial motility. (e.g., elicited by a local brain injury or release of ATP from a
glass micropipette), the microglial processes extend and elon-
gate toward the source within minutes. In vivo this process out-
4 MICROGLIA AND PURINERGIC growth is not accompanied by cell migration. As soon as the
SIGNALING process reaches its target region, the P2Y12 receptor is down-
regulated to almost nondetectable levels. In P2Y12 receptor
Microglia not only recognize a wide range of signals, but also knock-out mice the outgrowth is significantly delayed, but not
display a rather broad plasticity in their response (Kettenmann completely abolished, suggesting that additional mechanisms
et al., 2011). Given the rather homogenous distribution of are involved (Haynes et al. 2006). In acutely isolated brain
microglia within various CNS regions, they contribute to slices, release of ATP associated with astroglial Ca2+ waves is
the structural and functional integrity of the CNS. Some sensed by microglia (Schipke et al. 2002). Subsequently, an
authors attribute a very low threshold of activation to micro- outwardly rectifying K+ conductance is induced. In platelets
glial cells. However, this is only partially true if one compares a functional and structural interaction of the adenosine recep-
cell responses evoked by triggers of normal brain function tor A2A and the P2Y1 receptor with P2Y12 has been found
with those induced during acute or chronic brain injuries. (Suzuki et al. 2011). The role of adenosine for microglial motil-
The primordial transmitter ATP, for example, activates neu- ity or migration has also been strengthened by the analysis of
rons, astrocytes or microglia at similar concentrations (see also mice deficient of the ectonucleotidase ENTPDase1 or CD39,
chapter 25). an enzyme that very rapidly converts ATP into AMP that can
Interestingly, microglia are probably the cell-type with the be readily hydrolyzed by 5c-nucleotidase, broadly expressed
broadest response pattern to purinergic signals. In the CNS, in the brain. In ENTPDase1 knock-out mice ATP is inactive
ATP is not only an energy metabolite itself and its derivatives to stimulate microglial motility and has to be substituted by
are released as important primary transmitter and cotransmit- exogenous soluble ectonucleotidase or adenosine to observe
ter by a range of release mechanisms, which include vesicular microglial outgrowth (Farber et al. 2008).
release, diffusion through gap-junction hemi-channels of the Analysis of the second messenger cascade revealed the
pannexin-type, or transporters. Highest concentrations of activation of phosphatidylinositol 3c-kinase (PI3K) and
ATP are released during programmed or necrotic cell death. Akt kinase (Irino et al. 2008). Subsequently, induction of
Therefore, ATP is regarded as a universal “danger” signal. integrin-E1-extracellular matrix interactions has been sug-
Indeed, the excess of ATP in the brain parenchyma is neu- gested to control the extension of microglial processes
rotoxic. On release, ATP is rapidly degraded by extracellular (Ohsawa et al. 2010). Required rises of intracellular Ca2+ could
ectonucleotidases to ADP, AMP, and adenosine, thereby gen- be provided by influx through the ionotropic P2X4 receptor
erating a complex “soup” of purinergic signaling molecules. (Ohsawa et al. 2007).
A complete set of various ectonucleotidases, namely nucleo- Although ATP/ADP represents a strong chemoattrac-
side triphosphatase (NTPase), nucleoside diphosphatase tant for microglial processes, the volatile transmitter nitric
(NDPase), 5c-nucleotidase (5c-Nase), and purine nucleoside oxide (NO) induces migratory behavior of whole cells (Dibaj
phosphorylase (PNPase) is directly expressed at the surface et al. 2010). When the NO-donor spermine-NONOate was
of microglia. After hydrolysis, the purine metabolites can injected into the mouse dorsal white matter of the spinal cord

ROLE OF MICROGLIA IN THE NORMAL BR AIN • 607


with a fine glass micropipette, microglial cells retracted their remained unclear whether the apparently random pattern
fine lamellipodia, converted into cells with a simpler, more of microglia motility was controlled by distinct mechanisms
elongated shape, and started to migrate toward the local NO other than the purinergic and NO signals released by local
source within minutes (Dibaj et al. 2010). A detailed phar- microinjuries.
macological analysis revealed that NO triggers the guanylate Although the first in vivo imaging experiments of micro-
cyclase pathway of microglia. For astrocytes it has already been glia revealed only a small influence of neuronal activity onto
shown that dibutyryl-cGMP and atrial natriuretic peptide (a the global microglial motility (Nimmerjahn et al. 2005), more
ligand for a particulate guanylate cyclase) induce process elon- recent studies demonstrate a very selective interaction of micro-
gation and redistribution of the intermediate filament pro- glial processes with synaptic terminals (Paolicelli et al. 2011;
tein glial fibrillary acidic protein (GFAP) and actin filaments Tremblay et al. 2010; Wake et al. 2009). In transgenic mice
(Boran and Garcia 2007). Expression of the small GTPase in which both cell types, neurons and microglia, were labeled
RhoA, which is involved in cellular migration and contrac- by expression of fluorescent proteins discrete morphological
tion, is regulated by protein kinase G, another component of interaction steps could be discerned by two-photon laser-scan-
the NO/cGMP signaling pathway (Sauzeau et al. 2003). ning microscopy (2P-LSM)(Wake et al. 2009; Tremblay et al.
The baseline motility is effectively modulated by ambient 2010). In the two studies different transgenic mice were used:
levels of purines such as ATP, uridine triphosphate, and their Thy1-EGFP (Feng et al. 2000) and Iba1-EGFP (Hirasawa et
diphosphate metabolites. So far, three different mechanisms al. 2005) by Wake and colleagues; Thy1-EYFP and CX3CR1-
have been reported that affect extracellular purine levels. EGFP by Tremblay and colleagues. During their tissue sur-
Although a block of GABAergic inhibition enhances motil- veying, microglial processes stop at different synapses with
ity by about 10% (Nimmerjahn et al. 2005), hydrolyzing ATP variable duration ranging from a few minutes to 1 hour to
to AMP by the hydrolyzing enzyme apyrase as well as block- establish direct contacts with neuronal synapses, which could
ing gap junction channels reduces the motility (Davalos et al. be recognized by the fluorescent protein–expression in the
2005). Because ATP is often coreleased during neurotrans- postsynaptic spine compartment. On serial electron micro-
mission, the block of inhibition results in increased ATP. graphs of imaged areas, it can be seen that microglial processes
Similarly, ATP can be released from astroglial pannexin-type touch also other synaptic elements, the presynaptic terminal,
gap junction hemichannels (Iglesias et al. 2009). perisynaptic process, and synaptic cleft (see Fig. 47.3). These
novel ultrastructural observations define microglia as a sec-
ond, additional glial element next to astrocytes (Perea et al.
5 M I C R O G L I A L C O N T R I B U T I O N TO 2009) with important influence on synaptogenesis and neu-
SY N A P S E F O R M AT I O N A N D N E U R O N A L rotransmission (Tremblay and Majewska 2011). Interestingly,
C O N N E C T I VI T Y these ultrastructural inspections revealed that the dendritic
shaft was not contacted by microglia, thereby strengthening
During development, and during synaptic plasticity as well, the specificity of microglia–synapse interaction. The contacts
microglia contribute to the formation of synapses as well in appeared rather regularly with a frequency of about once per
determining their synaptic efficacy. Thrombospondins (TSPs) hour. In contrast to the observation that microglial motility is
belong to extracellular matrix proteins, are mainly produced slightly enhanced when neuronal inhibition is blocked, reduc-
by astrocytes and induce synapse formation (Christopherson ing neuronal activity resulted in a reduced frequency of syn-
et al. 2005). In the early nervous system also microglia secrete aptic contacts. Permanent block of the visual input (either by
TSPs (Chamak et al. 1995; Moller et al. 1996). TSP interacts eye enucleation or tetrodotoxin injection into the retina) lead
with the integrin-associated protein CD47 that can form a to a reduction of neuronal activity in the observed layer two
link to the signal regulatory protein (SIRP) D SIRP D is a to three regions of the visual cortex and concomitant retrac-
transmembrane protein expressed by neurons and myeloid tion of microglial processes from the synapses (Wake et al.
lineage cells (Matozaki et al. 2009). The protein CD47 is a 2009). Under the condition of energy failure as observed in
widely expressed glycoprotein. In the CNS it is found on vas- the ischemic penumbra after transient occlusion of the middle
cular endothelial cells. The system is involved in the regulation cerebral artery, the duration of microglia–synapse contacts is
of migration and phagocytosis as well as immune homeostasis. prolonged to more than 1 hour. Because numerous presyn-
Via the release of TSPs by microglia SIRPD/CD47 also affects aptic terminals disappear during that period, it is likely that
neuronal networks by modulating synaptogenesis. microglia contribute to the increased structural reorganiza-
Discovering the dynamic extensions and retractions of tion in penumbra by sensing the functional status of individual
microglial processes in the normal brain led to a significant synapses. In an injury model (axotomy of vagal motoneurons)
change in our understanding of microglia. After the first in it was shown that synaptic activity decreased before synapse
vivo imaging studies, the term resting microglia characterizing elimination (Yamada et al. 2008).
the nonactivated state in the healthy brain was changed to sur- Microglia–neuron interaction also affects synapse forma-
veying microglia to indicate its constantly active role in moni- tion under less aggressive experimental conditions. Under
toring its environment (Hanisch and Kettenmann 2007). normal sensory stimulation dendritic spines with microglial
These early observations revealed repeated contacts with the contacts display a much larger size distribution (Tremblay
surfaces of all adjacent cell types, such as neurons, astrocytes, et al. 2010). A reversible change of synaptic activity results
or the endothelial cells of the brain capillaries. However, it in a corresponding morphological modification. Although

608 • NEUROGLIA
E Without microglia F
1000 With microglia 4000
Extracellular space area (nm2)
Exlracelluar space area (nm2)

800
3000
600
2000
400

1000
200

0 0
0 1000 2000 3000 4000 5000 6000 7000
Microglial process area (nm2)

Figure 47.3 Ultrastructural Interactions Between Microglia and Synapses During Normal Sensory Experience. A–C. electron microscopy images
showing IBA1-immunostained microglial (m+) cell bodies (A), as well as large (B) and small (C) processes, surrounded by extended extracellular space
(asterisks) and contacting axon terminals (blue), dendritic spines (pink), perisynaptic astrocytes (green), and synaptic clefts (arrowheads). d, dentrite; N,
nucleus; p, perikaryon. Scale bars = 250 nm. D. electron microscopy image showing extended microglia-associated extracellular spaces (asterisks) after
glutaraldehyde instead of acrolein fixation. The unlabeled microglial process (m) makes direct contacts with dendritic spines (s) and axon terminals (t),
and displays an inclusion (in), as well as a clathrin-coated pit (black arrow) at the site of contact with a spine. Scale bar = 250 nm. E. Extracellular space
areas with or without contact with IBA1-positive microglial process. F. Correlation between the areas of microglial processes and associated extracel-
lular space. From Tremblay et al. 2010.

ROLE OF MICROGLIA IN THE NORMAL BR AIN • 609


dark-adapted mice display more microglial processes and more (CD11b/CD18) on microglia (Linnartz et al. 2012). In vitro
contacts to spines and axon terminals, the extent of processes an additional mechanism of cell debris removal in the absence
and contact sites return to control levels after novel light expo- of inflammation has been shown that involves the triggering
sure. Interestingly, the visual experience also influences the receptor expressed on myeloid cells-2 (TREM2)–mediated
size of the extracellular perisynaptic space around the micro- signaling in microglia (Takahashi et al. 2005). Patients with
glial processes, thereby suggesting an important bidirectional loss-of-function mutations of TREM2 develop a chronic
signaling pathway from neurons to microglia that actively neurodegenerative disease, named Nasu-Hakola disease or
involves the modulation of the extracellular matrix. polycystic lipomembranous osteodysplasia with sclerosing leu-
Surprisingly, although the in vivo imaging and the ultra- koencephalopathy (PLOSL). PLOSL develops as a neurode-
structural studies convincingly demonstrated intense contacts generative disease with only latent inflammation and patients
and engulfments of synaptic structures by microglia, they die at the fourth or fifth decade of life. Because TREM2 is
have not yet provided clear evidences for microglial phago- required for tissue debris clearance by microglia, the disease
cytosis of synaptic components. It is, therefore, very likely highlights the essential function of microglia for CNS tissue
that membrane-bound ligand-receptor–mediated signaling homoeostasis (Neumann and Takahashi 2007).
between the microglial process and the synaptic elements Still intriguing is the possibility of direct sensing of the
exist. Several signaling pairs have already been identified. A synaptic status by recognizing the respective neurotransmit-
common feature is the activity of the ligand as a calming signal ter. Microglia are well equipped with a plethora of transmit-
for microglia. In the presence of the ligand microglia remain in ter and neurohormone receptors such as glutamate, GABA,
its surveying state, whereas its reduction or the corresponding or substance P (Hanisch and Kettenmann 2007; Pocock
inactivation of the receptor lead to strong microglial responses and Kettenmann 2007). However, the functional impact of
(Biber et al. 2007). these receptors for synaptic regulation has not yet been dem-
One of such signaling pairs is formed by fractalkine and onstrated conclusively. So far, in the healthy brain, the cor-
CX3CR1. Although CX3CR1 is a heptahelical, G protein– responding receptors and electrophysiological responses to
coupled receptor, fractalkine (CX3CL1) is a transmem- synaptic events could not be detected on microglia. Therefore
brane glycoprotein expressed by neurons that ectodomain more indirect ways of regulating neuronal activity, for exam-
can be proteolytically cleaved and released into the extracel- ple, via astrocytes, have to be considered (Fontainhas et al.
lular space. Fractalkine is the exclusive ligand for CX3CR1, 2011; Wu et al. 2008). Activation of microglia induces a rapid
in the CNS expressed only by microglia (Cook et al. 2001; production and release of ATP. Microglial ATP then triggers
Harrison et al. 1998). In CX3CR1-deficient mice the calm- astrocytes to amplify ATP production and release glutamate.
ing effect of fractalkine onto microglia is no longer exerted, Subsequently, astroglial glutamate increases EPSC frequency
resulting in increased neuronal cell death after stimulation of by modulation of the neuronal presynaptic metabotropic glu-
inflammatory responses by injection of lipopolysaccharide, tamate receptor mGluR5.
by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) Thus, microglia might act as a genuine regulator of neu-
intoxication leading to symptoms of Parkinson disease or in rotransmission and upstream partner of astrocytes.
genetically modified mice modeling amyotrophic lateral scle-
rosis (ALS) (Cardona et al. 2006). More information on the
pathology of microglia in neurodegeneration is provided in 6 FAC TO R S R E L E A S E D BY M I C R O G L I A
chapters 61, 63 and 65.
To prevent microglial activation fractalkine is tonically Numerous studies have shown that microglial cells can release
released throughout the CNS at high concentrations (>300 a multitude of growth factors such as interleukin-1E, trans-
pg of soluble fractalkine per mg of aqueous extract of an adult forming growth factor-E (TGF-E), basic fibroblast growth
mouse brain). However, the interaction of fractalkine and factor (bFGF), hepatocyte growth factor, brain-derived neu-
CX3CR1 is not only a static mechanism to silence microglia. rotrophic factor (BDNF), and nerve growth factor (NGF).
In the uninjured brain of CX3CR1-deficient mice significantly Therefore, the supernatant of microglial cell cultures display
less microglial cells were found during the first postnatal month a powerful neurotrophic action (Morgan et al. 2004) (see also
(Paolicelli et al. 2011). Although in these mice synapses were chapters 22 and 23). Although we can hypothesize that micro-
also engulfed by microglial processes, almost twice as many glia contribute to higher-order brain functions, we are still
dendritic spines as well as immature synapses were detected. missing not only a systematic approach to reveal the growth
This was accompanied by a decreased frequency of spontane- factor production by microglia of different brain regions, we
ous excitatory postsynaptic currents recorded from hippocam- also have to develop experimental paradigms for subsequent
pal pyramidal neurons in acutely isolated slices from 2- to functional evaluation in vivo.
3-week-old-mice. These two observations indicate insufficient Microglial-derived growth factors may also affect synap-
maturation of synapses and, hence of hippocampal connec- tic transmission directly, as it has been shown in the spinal
tivity. The fractalkine/CX3CR1 pair is a pivotal component cord by investigating the molecular mechanisms of neuro-
of synaptic pruning (Paolicelli et al. 2011). Subsequently, the pathic pain after peripheral nerve injury. The injury-mediated
synaptic elimination occurs via a classical complement cascade release of ATP stimulates microglia to release BDNF, which
involving synaptic or perisynaptic localization of C1q and C3 in turn changes the Cl–-equilibrium potential in lamina
(Stevens et al. 2007) as well as the complement receptor CR3 I neurons. Subsequently, GABA- and glycine-mediated

610 • NEUROGLIA
Cl– currents reverse and the corresponding postsynaptic mechanisms of neuron–microglia interactions responsible for
responses become excitatory, instead of inhibitory (Coull synapse detection, recognition, and engulfment present dur-
et al. 2005). BDNF, however, may act directly on micro- ing development might be similar with those required for
glia. It induced a sustained increase in [Ca2+]i through bind- removal of dying neurons by phagocytosis (Peri and Nusslein-
ing with the truncated tropomyosin-related kinase (trk) Volhard 2008).
B receptor, resulting in activation of the phospholipase C
pathway and store-operated Ca2+ entry in rodent microglial
cells (Mizoguchi et al. 2009). 8 S U M M A RY A N D P E R S P E C T I VE S
Recently, it has been shown in tissue culture and acutely
isolated brain slices that the proinflammatory cytokine tumor In the CNS microglia are pivotal cells that permanently con-
necrosis factor alpha (TNF-D)–regulated synaptic scaling trol the status of brain activity. For that purpose they show the
depending on neuronal activity (Stellwagen et al. 2006). following features: (1) They are evenly distributed throughout
Although it was not directly proven in that study, it is highly all brain regions. (2) They are equipped with a broad range
likely that the TNF-D was produced by microglia present in of transmitter, growth factor, and cytokine receptors to effec-
the culture. tively monitor the diversity of neural activity during devel-
These data highlight the important role of microglia in opment and in the adult individual. (3) Receptor activation
the effective regulation of distinct synaptic transmission path- occurs rapidly and results in drastic functional changes.
ways by releasing various peptides. However, more studies are From these points one can extrapolate the following puta-
required to reveal the molecular complexity and diversity of tive functions for the healthy brain:
microglial, astroglial, and neuronal interactions in common
1. During development, microglia can contribute to the
cell circuits.
structural reorganization, for example, by removal of
excess and dispensable neurons that die by apoptosis or by
fine tuning and shaping synaptic structures as well as the
7 M I C R O G L I A D U R I N G D E VE L O PM E N T
adjacent extracellular space.
Microglial cell do not only determine the correct number of 2. Microglia fulfill a housekeeping function by removing
synapses during development by a mechanism called synap- cellular waste products.
tic pruning, microglia may determine the correct number of
3. Microglia help to reshape brain connectivity during
neurons as well. However, during the development of the ner-
sensory input.
vous system, microglia display Janus-like properties. In some
instances they positively influence neurogenesis, whereas in 4. Microglia cure microinjuries, for example, by removal of
other they directly induce programmed cell death and elimi- cell debris after the death of single cells.
nate developing neurons. In tissue culture, for example, the
astroglial differentiation of neural precursors isolated from 5. Microglia are always in an alert state to sense acute or
the largest neurogenic niche of the brain, the supraventricu- chronic injuries.
lar zone, required the presence of microglia and their released Current research has not only identified the dynamic
interleukin-6 and leukemia-inhibitory factor (Nakanishi et al. activity of microglia in the brain, but also characterized sev-
2007; Walton et al. 2006). Differentiated astrocytes, in turn, eral molecular mechanisms of bidirectional neuron–microglia
seem to determine the final steps of microglia ramification interactions during development, learning, and experience.
that occurs later during development (Navascues et al. 2000). Present data also demonstrate that additional yet unknown
In other regions of the brain microglia regulate cell death. In mechanisms exist. In particular, we are still missing a com-
the chick retina microglia release NGF that activates the trkB prehensive description how microglia interact with other cell
receptor to eliminate developing neurons by inducing apopto- types at the circuit level, for example, the communication with
sis (Frade and Barde 1998). Similarly, in the developing mouse excitatory or inhibitory neurons via perisynaptic astrocytes,
cerebellum, microglia induced the cell death of Purkinje neu- the interactions with astrocytes and the brain vasculature and
rons produced in excess. In contrast with the retina, here the the influence on the oligodendrocyte lineage cells and NG2
cell death was mediated by microglial release of superoxide glia as well as axons and myelin. Furthermore, future work has
anion (O2–) generated during respiratory bursts (Marin-Teva not only to evaluate and quantify the relative importance of
et al. 2004). The translucent and developing zebrafish brain already known pathways, but also to address region-depen-
provides an excellent model system: With a width of 400 μm dent variations in microglia function.
the brain can be completely imaged. Because it contains only
about 30 microglial cells, all of them can be visualized almost
simultaneously (Peri and Nusslein-Volhard 2008). Time-lapse REFERENCES
in vivo imaging of microglia revealed that all apoptotic, but
still alive neurons were engulfed by microglial cells. Dead Biber K, Neumann H, Inoue K, Boddeke HW. 2007. Neuronal “on” and
“off ” signals control microglia. Trends Neurosci 30:596–602.
neurons, however, were found directly inside the microglia, Boran MS, Garcia A. 2007. The cyclic GMP-protein kinase G path-
outlining the phagocytic function of microglia during neu- way regulates cytoskeleton dynamics and motility in astrocytes.
ronal removal. This study also suggests that the molecular J Neurochem 102:216–230.

ROLE OF MICROGLIA IN THE NORMAL BR AIN • 611


Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra Linnartz B, Kopatz J, Tenner AJ, Neumann H. 2012. Sialic acid on
IM, et al. 2006. Control of microglial neurotoxicity by the fractalkine the neuronal glycocalyx prevents complement C1 binding and
receptor. Nat Neurosci 9:917–924. complement receptor-3-mediated removal by microglia. J Neurosci
Chamak B, Dobbertin A, Mallat M. 1995. Immunohistochemical detec- 32:946–952.
tion of thrombospondin in microglia in the developing rat brain. Marin-Teva JL, Dusart I, Colin C, Gervais A, van RN, Mallat M. 2004.
Neuroscience 69:177–187. Microglia promote the death of developing Purkinje cells. Neuron
Christopherson KS, Ullian EM, Stokes CC, Mullowney CE, Hell JW, 41:535–547.
Agah A, et al. 2005. Th rombospondins are astrocyte-secreted pro- Matozaki T, Murata Y, Okazawa H, Ohnishi H. 2009. Functions and
teins that promote CNS synaptogenesis. Cell 120:421–433. molecular mechanisms of the CD47-SIRPalpha signalling pathway.
Cook DN, Chen SC, Sullivan LM, Manfra DJ, Wiekowski MT, Trends Cell Biol 19:72–80.
Prosser DM, et al. 2001. Generation and analysis of mice lacking the Mittelbronn M, Dietz K, Schluesener HJ, Meyermann R. 2001. Local
chemokine fractalkine. Mol Cell Biol 21:3159–3165. distribution of microglia in the normal adult human central nervous
Coull JA, Beggs S, Boudreau D, Boivin D, Tsuda M, Inoue K, et al. 2005. system differs by up to one order of magnitude. Acta Neuropathol
BDNF from microglia causes the shift in neuronal anion gradient 101:249–255.
underlying neuropathic pain. Nature 438:1017–1021. Mizoguchi Y, Monji A, Kato T, Seki Y, Gotoh L, Horikawa H, et al.
Crocker SJ, Frausto RF, Whitton JL, Milner R. 2008. A novel method 2009. Brain-derived neurotrophic factor induces sustained elevation
to establish microglia-free astrocyte cultures: comparison of matrix of intracellular Ca2+ in rodent microglia. J Immunol 183:
metalloproteinase expression profi les in pure cultures of astrocytes 7778–7786.
and microglia. Glia 56:1187–1198. Moller JC, Klein MA, Haas S, Jones LL, Kreutzberg GW, Raivich G.
Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, et al. 2005. 1996. Regulation of thrombospondin in the regenerating mouse
ATP mediates rapid microglial response to local brain injury in vivo. facial motor nucleus. Glia 17:121–132.
Nat Neurosci 8:752–758. Morgan SC, Taylor DL, Pocock JM. 2004. Microglia release activators of
Dibaj P, Nadrigny F, Steffens H, Scheller A, Hirrlinger J, Schomburg neuronal proliferation mediated by activation of mitogen-activated
ED, et al. 2010. NO mediates microglial response to acute spinal cord protein kinase, phosphatidylinositol-3-kinase/Akt and delta-Notch
injury under ATP control in vivo. Glia 58:1133–1144. signalling cascades. J Neurochem 90:89–101.
Farber K, Markworth S, Pannasch U, Nolte C, Prinz V, Kronenberg Nakanishi M, Niidome T, Matsuda S, Akaike A, Kihara T, Sugimoto H.
G, et al. 2008. The ectonucleotidase cd39/ENTPDase1 modulates 2007. Microglia-derived interleukin-6 and leukaemia inhibitory fac-
purinergic-mediated microglial migration. Glia 56:331–341. tor promote astrocytic differentiation of neural stem/progenitor cells.
Feng G, Mellor RH, Bernstein M, Keller-Peck C, Nguyen QT, Eur J Neurosci 25:649–658.
Wallace M, et al. 2000. Imaging neuronal subsets in transgenic mice Navascues J, Calvente R, Marin-Teva JL, Cuadros MA. 2000. Entry,
expressing multiple spectral variants of GFP. Neuron 28:41–51. dispersion and differentiation of microglia in the developing central
Fontainhas AM, Wang M, Liang KJ, Chen S, Mettu P, Damani M, et al. nervous system. An Acad Bras Cienc 72:91–102.
2011. Microglial morphology and dynamic behavior is regulated by Neumann H, Takahashi K. 2007. Essential role of the microglial
ionotropic glutamatergic and GABAergic neurotransmission. PLoS triggering receptor expressed on myeloid cells-2. TREM2. for central
One 6:e15973. nervous tissue immune homeostasis. J Neuroimmunol 184:92–99.
Frade JM, Barde YA. 1998. Microglia-derived nerve growth factor causes Nimmerjahn A, Kirchhoff F, Helmchen F. 2005. Resting microglial cells
cell death in the developing retina. Neuron 20:35–41. are highly dynamic surveillants of brain parenchyma in vivo. Science
Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. 2010. 308:1314–1318.
Fate mapping analysis reveals that adult microglia derive from primi- Noda M, Nakanishi H, Nabekura J, Akaike N. 2000. AMPA-kainate
tive macrophages. Science 330:841–845. subtypes of glutamate receptor in rat cerebral microglia. J Neurosci
Hanisch UK, Kettenmann H. 2007. Microglia: active sensor and versa- 20:251–258.
tile effector cells in the normal and pathologic brain. Nat Neurosci Nolte C, Moller T, Walter T, Kettenmann H. 1996. Complement 5a
10:1387–1394. controls motility of murine microglial cells in vitro via activation of
Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, McNamara RK, an inhibitory G-protein and the rearrangement of the actin cytoskel-
et al. 1998. Role for neuronally derived fractalkine in mediating eton. Neuroscience 73:1091–1110.
interactions between neurons and CX3CR1-expressing microglia. Ohsawa K, Irino Y, Nakamura Y, Akazawa C, Inoue K, Kohsaka S. 2007.
Proc Natl Acad Sci U S A 95:10896–10901. Involvement of P2X4 and P2Y12 receptors in ATP-induced micro-
Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, glial chemotaxis. Glia 55:604–616.
et al. 2006. The P2Y12 receptor regulates microglial activation by Ohsawa K, Irino Y, Sanagi T, Nakamura Y, Suzuki E, Inoue K, et al.
extracellular nucleotides. Nat Neurosci 9:1512–1519. 2010. P2Y12 receptor-mediated integrin-beta1 activation regulates
Helmchen F, Denk W. 2005. Deep tissue two-photon microscopy. Nat microglial process extension induced by ATP. Glia 58:790–801.
Methods 2:932–940. Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P,
Hirasawa T, Ohsawa K, Imai Y, Ondo Y, Akazawa C, Uchino S, et al. et al. 2011. Synaptic pruning by microglia is necessary for normal
2005. Visualization of microglia in living tissues using Iba1-EGFP brain development. Science 333:1456–1458.
transgenic mice. J Neurosci Res 81:357–362. Perea G, Navarrete M, Araque A. 2009. Tripartite synapses: astro-
Iglesias R, Dahl G, Qiu F, Spray DC, Scemes E. 2009. Pannexin 1: cytes process and control synaptic information. Trends Neurosci
the molecular substrate of astrocyte “hemichannels.” J Neurosci 32:421–431.
29:7092–7097. Peri F, Nusslein-Volhard C. 2008. Live imaging of neuronal degradation
Irino Y, Nakamura Y, Inoue K, Kohsaka S, Ohsawa K. 2008. Akt activa- by microglia reveals a role for v0-ATPase a1 in phagosomal fusion in
tion is involved in P2Y12 receptor-mediated chemotaxis of microglia. vivo. Cell 133:916–927.
J Neurosci Res 86:1511–1519. Pocock JM, Kettenmann H. 2007. Neurotransmitter receptors on micro-
Jung S, Aliberti J, Graemmel P, Sunshine MJ, Kreutzberg GW, Sher A, glia. Trends Neurosci 30:527–535.
et al. 2000. Analysis of fractalkine receptor CX(3)CR1 function by Rappert A, Biber K, Nolte C, Lipp M, Schubel A, Lu B, et al. 2002.
targeted deletion and green fluorescent protein reporter gene inser- Secondary lymphoid tissue chemokine (CCL21) activates CXCR3
tion. Mol Cell Biol 20:4106–4114. to trigger a Cl—current and chemotaxis in murine microglia.
Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. 2011. Physiology J Immunol 168:3221–3226.
of microglia. Physiol Rev 91:461–553. Sauzeau V, Rolli-Derkinderen M, Marionneau C, Loirand G,
Lawson LJ, Perry VH, Dri P, Gordon S. 1990. Heterogeneity in the dis- Pacaud P. 2003. RhoA expression is controlled by nitric oxide
tribution and morphology of microglia in the normal adult mouse through cGMP-dependent protein kinase activation. J Biol Chem
brain. Neuroscience 39:151–170. 278:9472–9480.

612 • NEUROGLIA
Schipke CG, Boucsein C, Ohlemeyer C, Kirchhoff F and Kettenmann Tremblay M, Stevens B, Sierra A, Wake H, Bessis A, Nimmerjahn A. 2011.
H. 2002. Astrocyte Ca2+ waves trigger responses in microglial cells The role of microglia in the healthy brain. J Neurosci 31: 16064–16069.
in brain slices. FASEB J:10.1096/fj.01–0514fje. Wake H, Moorhouse AJ, Jinno S, Kohsaka S, Nabekura J. 2009. Resting
Stellwagen D, Malenka RC. 2000. Synaptic scaling mediated by glial microglia directly monitor the functional state of synapses in vivo and
TNF-a. Nature 440:1054–1059. determine the fate of ischemic terminals. J Neurosci 29: 3974–3980.
Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Walton NM, Sutter BM, Laywell ED, Levkoff LH, Kearns SM, Marshall
Nouri N, et al. 2007. The classical complement cascade mediates GP, et al. 2006. Microglia instruct subventricular zone neurogenesis.
CNS synapse elimination. Cell 131:1164–1178. Glia 54:815–825.
Suzuki T, Obara Y, Moriya T, Nakata H, Nakahata N. 2011. Functional Wu LJ, Steenland HW, Kim SS, Isiegas C, Abel T, Kaang BK, et al. 2008.
interaction between purinergic receptors: effect of ligands for Enhancement of presynaptic glutamate release and persistent inflam-
A2A and P2Y12 receptors on P2Y1 receptor function. FEBS Lett matory pain by increasing neuronal cAMP in the anterior cingulate
585:3978–3984. cortex. Mol Pain 4:40.
Takahashi K, Rochford CD, Neumann H. 2005. Clearance of apoptotic Yamada J, Hayashi Y, Jinno S, Wu Z, Inoue K, Kohsaka S, et al. 2008.
neurons without inflammation by microglial triggering receptor Reduced synaptic activity precedes synaptic stripping in vagal
expressed on myeloid cells-2. J Exp Med 201:647–657. motoneurons after axotomy. Glia 56:1448–1462.
Tremblay ME, Lowery RL, Majewska AK. 2010. Microglial interac- Zierler S, Frei E, Grissmer S, Kerschbaum HH. 2008. Chloride influx
tions with synapses are modulated by visual experience. PLoS Biol provokes lamellipodium formation in microglial cells. Cell Physiol
8:e1000527. Biochem 21:55–62.

ROLE OF MICROGLIA IN THE NORMAL BR AIN • 613


48.
FACTOR S CONTROLLING MICROGLIAL ACTIVATION
Uwe-Karsten Hanisch

A B B R E VI AT I O N S 8). Also later on, notions of their pathological and physiological


importance were inevitably linked to the process of activation.
AE amyloid-E However, the underlying mechanisms triggering a transforma-
AD Alzheimer disease tion and causing consequences for the central nervous system
APC antigen-presenting cell (CNS) tissue remained elusive, as was the nature of signals that
ALS amyotrophic lateral sclerosis would initiate the response. Now it is known that microglial
CNS central nervous system activation is complex regarding causes, courses, and outcomes.
COX cyclooxygenase Even the term activation may not adequately reflect shifts from
DAMP damage-associated molecular pattern the state in the healthy CNS to the behavior upon an insult
EAE experimental autoimmune encephalomyelitis because it implies nonactivity under normal conditions and
ECM extracellular matrix fails to acknowledge diversity of reactive phenotypes. Seminal
FcR Fc receptor discoveries over the recent years unraveled housekeeping func-
IFN interferon tions of the “resting” microglia (see chapter 47). Meanwhile, the
IGF insulin-like growth factor concept of reactive phenotype diversity is accepted and becom-
IL interleukin ing further refined. Common to all of these aspects, there are
iNOS inducible nitric oxide synthase molecular factors that govern a departure from the surveilling
LPS lipopolysaccharide mode, that shape executive performance to protect the CNS
M1 classically activated macrophages against threats, but that may occasionally lead to an inappro-
M2 alternatively activated macrophages priate reactivity by scale and properties. There is no shortage
mGluR metabotrobic glutamate receptor of literature reports on those factors, receptors, and signaling
MHC major histocompatibility complex cascades, conditions under which they may unfold an influence
MMP matrixmetalloproteinase and the proposed impact they leave. Here, a focus is given to
MS multiple sclerosis principles of actions along with a brief survey of the main classes
MyD88 myeloid differentiation primary response of factors controlling microglial activation and activities.
gene 88
NO nitric oxide
PAMP pathogen-associated molecular pattern 2 CONCEPTS OF MICROGLIAL
PD Parkinson disease AC T I VAT I O N A N D AC T I VAT I N G
PG prostaglandin FAC TO R S
PRR pattern recognition receptor
RNI reactive nitrogen intermediates Several roles of microglia in the CNS surveillance as well as
ROS reactive oxygen species preservation of its integrity and functionality have been unrav-
TGF transforming growth factor eled over the last decade (Davalos et al. 2005; Nimmerjahn
Th T helper (cell) et al. 2005; Paolicelli et al. 2011; Wake et al. 2009). Concepts
TLR Toll-like receptor and fundamental discoveries that boosted their formulation are
TNF tumor necrosis factor outlined in chapters 19 and 47. The process of activation—as
TRIF TIR-domain containing adaptor protein inducible shifts in activity states were traditionally termed—
inducing IFNE includes a dramatic rebuilding of the cytoskeleton and mem-
brane organization, induction of whole sets of genes and
release of direct and indirect gene products (also considering
1 INTRODUCTION synthesis by induced enzymes) as well as unfolding of func-
tional portfolios ranging from migration and phagocytosis
The conscious description of microglia by del Rio-Hortega (see chapter 49) to communication with the adaptive immu-
came already with the observation that they undergo morpho- nity, for example, contributions as antigen-presentating cells
logical changes in response to pathological events (see chapter (APC, see chapter 50). The present section stresses principles

614
of alert and activation as triggered and governed by molecu- species (ROS), of proteases and other mediators and expres-
larly identifiable signals. sion of an array of surface molecules for cellular communi-
cation, including major histocompatibility complex (MHC)
structures (Fig. 48.1B), are in support of defense and protective
2.1 L I P O P O LYS AC C H A R I D E A N D T H E FI R S T reactions, but some of these factors and their associated func-
E X P E R I M E N TA L D E M O N S T R AT I O N O F tions can also cause substantial damage (Block et al. 2007).
INDUCIBLE RESPONSES Such activated microglia can exhibit massive neurotoxicity.
Structurally, LPS varies by the type and number of acy-
Much of the knowledge about the molecular and cellular prin-
lations, by phosphorylation as well as by the size and the
ciples as well as consequences of microglial activation has been
composition of the carbohydrate moieties, giving rise to a
derived from experimental stimulations under controlled in
variety of chemotypes. Common to all is recognition by cell
vitro conditions (Kettenmann et al. 2011). Even though a loss
surface–expressed Toll-like receptor (TLR) 4, triggering ini-
of tissue context comes with unavoidable limitations (absence tial host defense responses of innate immune cells, including
of environmental cues with activity-controlling influence), the microglia (Regen et al. 2011). Yet even long before the dis-
steps and features of activation through many of the key recep- covery of TLRs, LPS was known and used for the profound
tor systems can be essentially recapitulated. Certain manipula- changes it triggers in macrophages and microglia (Schwartz
tion and monitoring options are still not equally accessible in et al. 2006).
vivo, giving value to work with cell preparations as a comple- Dramatic shape changes, including granular appearance,
mentary approach. If particular compounds should be nomi- already indicate a response. However, it is more the massive
nated for delivering basic insights into microglia activation, induction of release activities that would affect the CNS.
lipopolysaccharide (LPS) would lead the list. Morphological changes under LPS resembled the appearance
As a cell wall component of gram-negative strains, LPS is of microglia in the diseased CNS—and activation could not
responsible for many of the initial responses to such infections be tolerated by its vulnerable neur(on)al structures. In con-
(Beutler and Rietschel 2003). Production of cytokines, such clusion, microglia earned the reputation of a harmful cell,
as the pluripotent and proinflammatory tumor necrosis fac- almost like a risk factor (Schwartz et al. 2006). This notion
tor (TNF) alpha, interleukin 1 (IL-1), IL-6, or IL-12, and a dominated the view of microglia for years. Although it is
range of chemokines, like CCL2, CCL3, CCL5, CXCL1, and now known to be a poor reflection of the true physiological
CXCL2 with chemoattractive activity for monocytes, T-cell spectrum, the early experiments employing LPS (and other
populations, and neutrophils, organizes not only immune cell microbial agents) set the basis for experimental studies on fac-
infiltration and activities, but influences the functions and tor-initiated microglial responses. Responses can be versatile,
vitality of neurons and glial cells, including microglia them- depending on the driving stimulus and contextual signaling
selves (Hanisch, 2002; Kettenmann et al. 2011) (Fig. 48.1A). of additional factors. Even for TLR4, functional implications
Similarly, release of nitric oxide (NO) and reactive oxygen are diverse. Notably, microglia primarily serve supportive and

Figure 48.1 Microglial Responses to Stimulation of TLR4 with LPS. A. Mouse microglial cells were stimulated in vitro with S-LPS and Re-LPS at
various concentrations. The released cytokines and chemokines were determined in the culture supernatants after 18 hours. Absolute amounts were
then normalized to amounts obtained from stimulation at maximal agonist concentration. B. Microglia were challenged with Re-LPS (10–8 g/mL, 48
hours), stained with fluorescence-conjugated anti-MHCI and anti-CD11b antibodies and processed for flow cytometry. Analysis considered CD11b+
cells for MHCI expression. C. Microglia deficient in functional MyD88 or TRIF (myd88–/–, trif lps2) were stimulated with 10–7 g/mL of either LPS
chemotype for 18 hours. Cytokines and chemokines were analyzed as in (A), normalized to the release obtained with wild-type cells. Data are from
Regen et al. 2011.

FAC TO R S C O N T R O L L I N G M I C R O G L I A L AC T I VAT I O N • 615


protective functions under normal healthy conditions as well to chapter 49). Pioneering work exploiting facial nerve transec-
as in many (if not most) situations in which the CNS has to tion set the fundaments of targeting microglial reactions in vivo
cope with exogenous and endogenous threats. Yet most of the (Kreutzberg 1996). The process of microglial activation can
“daily” actions of microglia in solving minor and localized unfold the reactive repertoire of an innate immune cell. Stages
abnormalities probably remain unnoticed because they never were defined by morphological, molecular, and functional
surface with signs (Hanisch and Kettenmann 2007). characteristics, with full-blown reactive microglia presenting
like (other) macrophages (Davoust et al. 2008; Graeber 2010).
Yet microglia are embedded in a specialized tissue (brain paren-
2.2 G E N E R A L FE AT U R E S A N D P H A S E S O F
chyma), which has to protect itself from potentially damaging
T H E M I C RO G L I A L AC T I VAT I O N P RO C E S S
consequences of an immune reaction. This special condition
Until recently, microglia of the healthy adult CNS were con- has been described as “immune privilege” (Galea et al. 2007).
sidered quiescent, that is, functionally dormant, because of Microglia activation is thus a highly regulated process.
low (absent) expression of activation-associated molecules and Traditional concept oversimplified and generalized the
their ramified morphology. This changed with in vivo imaging adjustments to changes in their environment. The binary con-
studies demonstrating high motility of the cellular processes cept of activity states (“resting” versus “activated”) did not
for a scanning of the environment (Nimmerjahn et al. 2005; acknowledge default activities under normal conditions, nor
Paolicelli et al. 2011; Wake et al. 2009). the diversity of response options. Microglial activation is not an
Infection, trauma, ischemia, neurodegenerative diseases, “all-or-none” process, like an action potential. It does not follow
or altered neuronal activity, that is any disturbance or loss of a linear path to a uniform outcome. Instead, activated microg-
homeostasis, real or potential danger to the CNS can then lia acquire distinct functional profiles (David and Kroner 2011;
evoke rapid and profound changes in the microglial cell shape, Saijo and Glass 2011; Schwartz et al. 2006) (Fig. 48.2).
gene expression patterns, and functional behavior (Block et al. Features and consequences of activation are largely (yet
2007; Davoust et al. 2008; Kettenmann et al. 2011) (refer also not entirely) determined by the initial cause. This cause comes

Figure 48.2 States of Microglia Activity and Steps of the Activation Process. In the normal healthy (mature) CNS tissues, microglia present with a
ramified morphology previously considered a “resting” state, but recent investigations proved that they are active in terms of surveillance. This includes
scanning of their environment with motile processes and intimate contacts with synapses as well as permanent integration of signaling inputs through
an array of receptors. Appearance of molecular cues indicating disturbed homeostasis can instruct rapid changes in morphology, movement, expres-
sion, and functional profiles. Infections, ischemia, trauma, or cell impairment along with neurodegenerative and autoimmune processes deliver a
range of activating signals that are sensed by already expressed receptors. Transformations to alerted and finally fully activated states are, however, also
controlled by calming signals that inform microglia about the well-being of neurons. Loss of such inputs may set off an alert or lower thresholds for
activating signs. Similar to other macrophages, but in a CNS-adapted fashion, microglia will then commit to distinct reactive phenotypes. Programs
come with particular transcriptional profiles as well as nontranscriptional adjustments. Their choice is dictated by the initial stimuli and their context.
Initial programs may also undergo changes while the activation proceeds, governed by a disappearance of driving forces and influences from other resi-
dent CNS as well as invading immune cells. Activation may resolve thereafter. Deescalation may come with a redistribution of microglia, their decline
by cell death, or even a departure from the CNS. Microglia may reacquire a nonactivated state—or remain altered by the previous experience. Their
behavior on another confrontation may differ from that of completely naïve cells. Scheme adapted from Hanisch and Kettenmann 2007.

616 • NEUROGLIA
with prominent molecular signals that are indicative of the at all and which features are expressed. Sorting of activating
pathophysiological meaning for the CNS, such as an infec- signals could be based on exogenous or endogenous sources
tious threat or cell impairment by intrinsic complications. (infectious agents, self-derived molecules). Signals vary by
Microglia are prepared to understand many of these signals cellular origin and physiological implication (neurotransmit-
and call up a variety of programs to cope with the challenge. ters, inflammatory messengers). Delivery by autocrine or para-
Common to all activating scenarios is that microglia have to crine routes or “leakage” differentiates between professionally
give up the surveilling mode for a situation-adapted response secreted factors and material delivered by damage. Factors can
program, based on the balance of incoming signs (Hanisch be grouped by their biochemical or chemical nature or related
and Kettenmann 2007). receptors, action as a soluble ligand, or as anchored in a mem-
brane or the extracellular matrix (ECM), such as integrins.
Signals can also be sorted by principles setting off an alarm.
2.3 I N I T I AT I O N O F A R E S P O NS E
In one case, the signal has to appear in the extracellular space
Multiple signals converge on microglia under normal con- surrounding microglia, because it is usually not there at all
ditions as well as on impairment of the CNS. Each of these (bacteria), or close by, but not in physical contact (plasma
conditions comes with an ensemble of (molecular) factors proteins), or usually not present in signaling-relevant format
that will decide on whether microglia engage with a response (redox state, monomeric/aggregated, on necrotic cell death or

Table 48.1 LIGAND-RECEPTOR SYSTEMS FOR ACTIVATION AND MODULATION OF MICROGLIAL RESPONSES
FACTOR/COMPOUND CLASS LIGANDS AND RESPECTIVE RECEPTORS

Cell wall components, surface structures, Pam3CSK4, MALP-2, lipoteichoic acid, proteoglycans, various LPS chemotypes (S- and R-LPS),
DNA and RNA of viral, bacterial or zymosan, poly(I:C), poly(A:U), flagellin, ssRNA, CpG ODN as binding to PRR families, especially
fungal origin (PAMPs) TLR1/2, TLR3, TLR4, TLR5, TLR6/2, TLR7/8, and TLR9

Endogenous molecules (DAMPs) HMGB1, fibronectin, fibrinogen, tenascin, versican, hsp60, hsp72, S100A8/9, neutrophil elastase,
lactoferrin, serum amyloid A, AE, AE25–35, AE40, AE42, prion protein (PrP), oxidized low density
lipoprotein, fatty acids, hyaluronic acid, heparan sulfate (fragments), IgG/chromatin complexes,
endogenous RNA/DNA as binding to TLR2, TLR3, TLR4, TLR7/8, and TLR9 as well as other
(scavenger) receptors, RAGE

Cytokines IFNJ and IL-4/IL-13 with crucial instruction of phenotypes, IL-10 as an immunosuppressive factor,
TNFD, IL-1E, IL-18, IL-33 with activating contributions, IFNE with modulating influences, IL-2,
IL-15, TGFE, colony stimulating factors (M-CSF, GM-CSF), activities via their respective receptors
and receptor subtypes

Chemokines CCL2/CCR2 pair with key role in monocyte/microglia recruitment, CX3CL1/CX3CR1 with calming
control, both systems also with distinct roles in monocyte/microglia subset identification, ligands for
CCR1, CCR3, CCR5, CXCR1, CXCR3, CXCR4, IL-8R with diverse functional effects

Complement Complement factor C1q, C5a via complement receptors

Antibodies Immunoglobulins of diverse sub/classes (IgA, IgG, IgM), largely active as immune complexes

Neurotrophins/neurotrophic factors Brain-derived neurotrophic factor (BDNF), glial-derived neurotrophic factor (GDNF), nerve growth
(NF) factor (NGF), neurotrophin 3 (NT-3), NT-4

Coagulation factors Thrombin via proteinase-activated receptors (PAR) 1, 3, 4

Neurotransmitters/cotransmitters Adenosine, ADP, ATP via adenosine (A) and purinoreceptors (P2X, P2Y) of most diverse subtypes,
glutamate via AMPA and mGluR types, GABA via GABAB receptors, acetylcholine via nicotinic
receptors, nor/adrenaline via D and E subtypes of adrenergic receptors, dopamine via D1 and D2 recep-
tors, often with inhibitory and modulating influences on other receptor stimulations

Neurohormones/neuromodulators Bradykinin, neurokinin, substance P, VIP, endothelin, opioids, endorphins, angiotensin, somatostatin,
melatonin, platelet-activating factor (PAF), histamine, cannabinoids via their respective receptors and
receptor subtypes

Steroids, vitamins Gluco/corticoids via glucocorticoid as well as mineralocorticoid receptors, vitamin D3, importance for
attenuation of responses to other receptor activations (e.g., TLRs), own activities

Lipids and others Ceramides, gangliosides, prostaglandins, leukotrienes, lysophosphatidic acids


Systems with calming steady-state Ligand/receptor pairs with reciprocal expression by neurons and microglia, SIRP1D/CD47, CD200/
influences CD200R, CD22/CD45, and CX3CL1/CX3CR1

Examples are given as based on Chen and Nunez (2010), Hanisch and Kettenmann (2007), Hanisch et al. (2008), Kettenmann et al. (2011), and Piccinini and
Midwood (2010).

FAC TO R S C O N T R O L L I N G M I C R O G L I A L AC T I VAT I O N • 617


in neurodegenerative processes with protein deposition). All gene 88 (MyD88) or TIR-domain containing adaptor protein
of them alert microglia by (sudden, increased) presence as “on inducing IFNE (TRIF) (Lin et al. 2010; O’Neill and Bowie
signals.” Typical on signaling is mediated via TLRs or recep- 2007; Park et al. 2009). MyD88 and TRIF, in turn, organize
tors for TNFD or interferon (IFN) J (Table 48.1). In contrast, a downstream activation of kinase cascades and transcription
certain ligand–receptor pairs keep microglia in a steady state factors controlling interferons, cytokines, and chemokines
because their signaling imposes a calming influence, with an with importance for immune cell recruitment and activa-
“off signaling” effect on disruption (Biber et al. 2007; Hanisch tion (Kawai and Akira 2010). Although most TLRs depend
and Kettenmann 2007; Kettenmann et al. 2011; Saijo and Glass on MyD88, and only TLR3 solely relies on TRIF, TLR4 is
2011; van Rossum and Hanisch 2004). CD200/CD200R, in the unique situation to be linked to both adapters, which
CD47/SIRPD, CX3CL1/CX3CR1, or CD22/CD45 are organizes the various response elements (Fig. 48.1C). Toll-like
representative systems. Integration of both signal types deter- receptor expression in cells of the innate and adaptive immu-
mines whether surveilling microglia transform to other (exec- nity as well as in nonimmune cells has been associated with a
utive) activity states (see Fig. 48.2). A distinction by “on” and wide range of functions, including responses to PAMPs and
“off ” signals offers even more understanding of activation. On DAMPs (Glezer et al. 2006; Hanisch et al. 2008; Miller et al.
signals require the pre-existence of receptors—TLRs cover- 2005; Rivest 2009; Rubartelli and Lotze 2007).
ing multiple signs of infection. Blockage of constitutive off The unleashing and propagation of microglial activation
signaling can increase basal activities and enhance stimulated in (apparently noninfectious) CNS diseases has been attrib-
responses in situations of neuronal impairment of any kind, uted to some CNS-intrinsic signals that would be delivered
for example. Interruption of the current in wires of a shop- as a consequence of cell impairment. Now, more and more
ping window turns on an alarm. Microglia get alerted when of those factors are actually identified. DAMPs may not only
off signals fade. As a consequence, microglia can sense danger play roles in responses to trauma and cell injury, vascular dis-
by “known” and “unknown” causes. ruption, and ECM breakdown, and contribute to damaging
cascades following CNS complications, such as ischemia and
stroke. DAMP signaling likely participates also in neurodegen-
2.4 FAC TO R S C O N T RO L L I N G M I C RO G L I A L
erative processes, such as Alzheimer disease (AD), Parkinson
AC T I VAT I O N
disease (PD), or amyotrophic lateral sclerosis (ALS), and to
Pattern recognition receptors (PRRs) are germ line-encoded some extent in autoimmune diseases such as multiple sclerosis
sensors for microorganisms and enable the immune system (MS) (Chen and Nunez 2010).
to fight infectious threats. They bind an assortment of evolu- For microglia, especially (yet not only) TLR1, 2, 4, and 6
tionary conserved structural motifs within glycolipids, glyco- in their combinations or association with non-TLR receptors
peptides, lipoproteins, and proteoglycans as well as RNA and would play a role not only in the response to bacterial infec-
DNA of diverse microbial origins that are collectively termed tions but also for detection of endogenous factors, including
pathogen-associated molecular patterns (PAMPs) (Kawai several heat-shock proteins (hsp), fibronectin, lactoferrin,
and Akira 2010; Takeuchi and Akira 2010). Pattern recogni- tenascin, versican, heparan sulfate, hyaluronic acid, HMGB1,
tion receptors come in families as to structural and functional S100A8/S100A9, oxidized phopsholipids and lipoprotein
relations: NOD-like receptors (NLRs), RIG-1-like receptors A, fatty acids, eosinophil-derived neurotoxin, serum amy-
(RLRs), C-type lectin receptors (CLRs), absence in mela- loid, and amyloid-E (AE) (Hanisch and Kettenmann 2007;
noma 2-like receptors, and TLRs (Chen and Nunez 2010). Hanisch et al. 2008; Heneka et al. 2010; Lehnardt et al.
Significantly, PRRs also observe the inner world because some 2008; Piccinini and Midwood 2010; Stewart et al. 2010).
(notably TLRs) respond to molecules generated, released, or Thus, DAMPs may prepare and assist reactions to PAMPs,
modified in the context of tissue injury. Being of diverse origin such as in meningitis, or independently drive inflammatory
and physiological importance, as damage-associated molecular scenarios. Disturbed protein expression, altered turnover,
patterns (DAMPs) they gain a role as danger indicators (alarm- and processing as resulting in abnormal (poly)peptide struc-
ins) in situations that are not (yet) linked to infection and that ture, aggregation, and deposition are assumed to be key ele-
(ab)use (innate) immune cells in sterile inflammation (Chen ments of microglia activation and reaction in diseases such as
and Nunez 2010). Microglia express PRRs, including TLRs, AD. Factors such as AE in AD, which could misuse DAMP
and involve their obvious and cryptic signaling in probably all principles, are becoming increasingly better defined also as
scenarios of CNS diseases (Hanisch and Kettenmann 2007; to their receptors and signaling outcomes. This is illustrated
Hanisch et al. 2008; Kettenmann et al. 2011). Recognizing by a study reporting on an assembly of a TLR4/6 heterodi-
PAMPs and DAMPs, TLRs prototypically fulfill roles of mer with CD36 that can promote sterile inflammation to
omnipotent sensors of disturbed homeostasis (Hanisch and AE (Stewart et al. 2010). Of course, TLRs are not the only
Kettenmann 2007; Matzinger 2007). class of receptors sensing danger and damage, and micro-
Recognizing patterns rather than individual ligands, a glial responses are not only orchestrated by DAMPs (see
small number of TLRs (about 13 in mouse and humans) Table 48.1). Microglia are a source and target of a plethora
cover a wide range of relevant pathogens. Expressed on the of cytokines, which control diverse genes and functions by
cell surface or in endosomal compartments, TLRs signal via themselves but also exert remarkable modulatory influences
two major pathways that are characterized by the signaling on other receptor-signaling systems (Hanisch 2002; Häusler
adapter proteins myeloid differentiation primary response et al. 2002; Regen et al. 2011).

618 • NEUROGLIA
Among the cytokines/interferons, TNFD, IL-1, IL-4, may mainly exert modulating influences (Boucsein et al. 2003;
and IL-6 play essential roles as to their induction by diverse Kettenmann et al. 2011).
insults, multiple implications, profound effects, and key The list of microglia-activating factors comprises an
roles for governing microglial functions (Block et al. increasing number and diversity of additional molecules, con-
2007; Hanisch 2002; Hanisch and Kettenmann 2007; taining also lipid mediators or proteases with unusual receptor
Kettenmann et al. 2011; van Rossum et al. 2008). IL-1 and mechanisms, as surveyed in detail by Kettenmann et al. (2011).
its relatives are essential to CNS responses as (mainly but Examples are given in Table 48.1.
not only) mediated by microglia based on a deep involve-
ment in a regulation of (innate) immune function (Liew
2.5 C O M M IT M E N T TO R E AC T I VE P H E N OT Y P E S
et al. 2010; Prinz and Hanisch 1999; Smith 2011). Beneficial
outcomes of manipulating IL-1–like signaling, for example, A global impact on understanding microglial functions derived
via IL-1 receptor antagonist, point to an implementation from research on macrophage phenotypes, that is, their abil-
in CNS-harming processes, but IL-1 also has functions in ity to mount tailored responses (Gordon and Taylor 2005;
development or endocrine axis (Bilbo and Schwarz 2009; Martinez et al. 2006; Mosser and Edwards 2008; Murray and
Hanisch 2002). IL-1 can claim special relations to situations Wynn 2011).
with TLR-agonistic PAMP/DAMP involvement owing to Microbial agents such as LPS, or cytokines such as TNFD
common signaling pathways. or IFNJ, trigger the “classical” and “innate” (immune) activa-
For IL-6, ambiguous impacts on microglia result from its tion. Cells develop a phenotype with proinflammatory, APC,
wide array of proinflammatory as well as antiinflammatory and microbicidal activities in support of Th1-type adaptive
consequences. It is the induction by tissue damage that may immune responses, which can also result in tissue damage,
determine the diverse importance for the CNS. Similarly as however. Macrophages can “alternatively” commit to more
for IL-1, IL-6 functions are modulated at both the level of the antiinflammatory (inflammation-resolving) profiles under the
ligand as well as its receptor activity (Hanisch 2002). influence of the Th2 cytokines IL-4 and IL-13, with endocy-
IFNγ and IL-4 are key determinants for instruction of totic, parasite-killing, and tissue repair activities and fostering
general orientations seen with activated macrophages, includ- Th2 immune responses (Murray and Wynn 2011). Following
ing microglia (Hanisch and Kettenmann 2007; Kettenmann the Th1/Th2 paradigm, classically and alternatively activated
et al. 2011). Their roles are further discussed in the follow- macrophages were also termed M1/M2 types. The principle
ing. Importantly, production largely depends on other cells, appears to be evolutionarily conserved as it is seen in fishes.
namely Th1 and Th2 T-cell subsets, as well as natural killer Highlighted by the prominent expression of either the proin-
(NK) cells, indicating a prominent control of infiltrating flammatory IL-12 (IL-12hi) or the immunosuppressive IL-10
immune cells on microglia (Hanisch, 2002; Hanisch and (IL-10hi), M1 and M2 macrophages reveal characteristic (dis-
Kettenmann 2007; Häusler et al. 2002; Kettenmann et al. crete, partially reciprocal, and overlapping) patterns of induced
2011). cytokines/chemokines, enzymes, and substrates for ECM
Chemokines comprise a family of chemoattractive cytok- breakdown or rebuilding (Martinez et al. 2009) (Fig. 48.3).
ines exerting functions in cell migration under both physi- The M1 phenotype is accompanied by the release of
ological and pathophysiological conditions, but also beyond. TNFD, IL-1, IL-6, IL-23, CCL2, CCL3, CCL5, ROS,
Their structures, receptors, signaling, and (glial) physiology reactive nitrogen intermediates (RNI), and diverse matrix
are subjects of chapter 22. Receptors are expressed abun- metalloproteinases (e.g., MMP-1, -2, -7, -9, or -12) as well as
dantly in microglia. Combined with a promiscuous ligand (enhanced) expression of MHCII, CD16, CD32, CD64,
acceptance, induced chemokine cocktails exert a sophisti- CD80, CD86, and TLR family members—to name a few.
cated control. Special mention is made regarding members M2 macrophages rather induce IL-1 receptor antagonist
with exposed roles in the management of microglial func- (IL-1ra), CCL17, CCL22, coagulation factor XIII, decoy
tions (Kettenmann et al. 2011). CCL2 is of key importance IL-1RII, CD163, mannose/scavenger receptors, fibronectin,
as to its monocyte-attractive properties (Mildner et al. 2007). and ECM-cross-linking enzymes (David and Kroner 2011;
CX3CL1, also known as fractalkine, organizes a complex Mantovani et al. 2004; Mosser and Edwards 2008; Murray
control over microglia activities by calming effects and other and Wynn 2011; Ransohoff and Perry 2009). The IL-10lo/
involvement (Cardona et al. 2006; Prinz et al. 2011). CXCL10 IL-12hi versus IL-10hi/IL-12lo bifurcation is thus mirrored by
signaling via CXCR3 plays roles in diverse neuropathologies. preferential, selective, or even opposite induction of many
Involvement in NMDA-induced neurotoxicity unraveled additional genes. M1 macrophages express cyclooxygenase
cooperation with astrocytes for a complex function of micro- (COX) 2, whereas M2 orientation is associated with COX1
glia in diminished or enhanced cell death, depending on CNS (Martinez et al. 2006). Expression of inducible nitric oxide
regions (Kettenmann et al. 2011). synthase (iNOS) or arginase directs the use of arginine for
If a class of compounds is typical for the CNS, it is the range production of NO or synthesis of polyamines, the former
of neurotransmitters. As it is for astrocytes, some can be sensed being used for defense purposes, the latter being required for
by microglia. Interactions with neuronal signaling derive from tissue repair. Cells may switch between these pathways, M2
both their production and the expression of their receptors. being the homeostatic default setting (Martinez et al. 2006).
Although activating contributions are found largely for ATP This binary classification only temporarily served profile
and related purines, other transmitters and cotransmitters descriptions (Mantovani et al. 2004; Mosser and Edwards

FAC TO R S C O N T R O L L I N G M I C R O G L I A L AC T I VAT I O N • 619


Figure 48.3 Reactive Phenotypes of Macrophages/Microglia. Exposure to different stimuli leads to distinct functional consequences. Certain cytok-
ines drive macrophage reactions in support of inflammation and Th1 type of (adaptive) immune responses, highlighted by IL-12 release, but consisting
also of numerous other gene activations. An array of induced cytokines and chemokines, enzymes for ROS and NO synthesis, proteases (matrix metal-
loproteinases), as well as expression of surface molecules for cell and matrix interactions is thereby serving largely in defense reactions, partially with
the risk of tissue impairment. A macrophage of this orientation is commonly known as classically or M1 activated. In contrast, cytokines, namely IL-4
or IL-13, rather induce an alternative or M2 activation. This is characterized by prominent induction of IL-10 and a more or less complementary set of
genes, for cytokines, chemokines, enzymes, and substrates that together promote antiinflammatory and inflammation-resolving processes, Th2 types of
immune responses and tissue repair. As exemplified by mannose receptor and MHCII expression, there can be substantial overlaps in genes and their
functional aspects, suggesting that reactive phenotypes are not sharply separated entities. Indeed, phenotypes cannot easily be categorized by a M1/M2
bifurcation. Subtypes were defined by triggering stimuli and outcomes, such as M2a, M2b (type II) or M2c on infections, tissue damage, and exposure
to apoptotic cells and (myelin) debris, in association with tumors and metabolic disorders. The concept of phenotypes considers major orientations
relating to homeostasis, defense, and repair, but also that actual transcriptional and functional profiles are versatile and fine tuned, and probably even
flexible. Microglia are thought (and already shown) to commit to similar phenotypes within a M1/M2 spectrum, but respond in a fashion that is
adapted to CNS conditions. For details, see a recent survey (Kettenmann et al. 2011).

2008) (see Fig. 48.3). Exposure to immune complexes (IC) cannot match biological diversity. In addition to homeo-
can reverse LPS-induced toxicity and IL-12 production, static control and maintenance, cells act in immunoregula-
switching macrophages into an IL-10hi phenotype termed type tion and defense or healing and repair—with profiles of genes
II (M2b) (Anderson and Mosser 2002). FcJRI (CD64) seems and functions as to situations and location (Hanisch and
to be a critical FcR. M2b cells do not easily fit into a M1/M2 Kettenmann 2007; Mosser and Edwards 2008). Appropriate
sorting. They produce TNFD, and all three subtypes express phenotype initiation, propagation, maturation, and ter-
MHCII. Dampening of inflammatory activity by FcJR liga- mination are essential for success. Excessive acute, chronic,
tion was also observed with other proinflammatory stimuli, or maladapted responses can have detrimental outcomes
such as lipoteichoic acid, CD40 ligand, or hyaluronic acid ranging from hyperinflammation to immunosuppression in
(Gerber and Mosser 2001). The physiological impact of M2b infection, autoimmunity, cancer, and neurodegenerative and
macrophages is tremendous. Transfer of 106 cells/mouse res- metabolic diseases (Hanisch and Kettenmann 2007; Pollard
cued LPS-treated animals, whereas animals receiving control 2009).
macrophages succumbed to lethal endotoxemia (Anderson For microglia, a similar array of phenotypes can be
et al. 2002). In contrast to M1, M2b macrophages also induced assumed, and first studies report on the similarities and dif-
naïve CD4+ T cells to produce high IL-4 levels, even when ferences, although detailed experimental proof is pending
they were subsequently stimulated under nonbiasing condi- (David and Kroner 2011; Hanisch and Kettenmann 2007;
tions. M2b macrophage transfer to ovalbumin-immunized Kettenmann et al. 2011; Olah et al. 2012; Schwartz et al.
mice led to higher antibody titers and IgG1 preference, com- 2006). Phenotypic switches can occur without overt changes
pared with M1 delivery. Obviously, the macrophages instruct in morphology, rendering subtle functional reorganizations
T- and B-cell functions. likely to escape detection. Orientations for M1 and M2 (e.g.,
It turns out that macrophages, including CNS micro- under IFNJ, LPS, IL-4/IL-13, or IL-10) reveal essential over-
glia, take major orientations by the physiological context, lap with responses of macrophages (van Rossum et al. 2008).
although actual sets of genes and functions allow for rather Nevertheless, side-by-side comparisons also reveal differences,
flexible tuning. Accordingly, rigid phenotype classifications such as in the preferential induction of IL-10 versus TGFE, as

620 • NEUROGLIA
to the sensitivity to stimuli, organization of receptor/signal- it after injury, but eventually convert to profile for deescala-
ing, and functional consequences (Regen et al. 2011). tion and restoration.
The role of IFNJ and IL-4 for instruction of micro- Experimental instructions in vitro enforce orientations
glial phenotypes has been addressed by a number of studies which in vivo may follow (and build on) each other (Hanisch
(Butovsky et al. 2006a,b), also in relation to allowing, sup- and Kettenmann 2007). Defects in such transitions could be
porting, or impeding regenerative processes (Kempermann detrimental. Chronic activation may result from the persist-
and Neumann 2003). In one of them, microglia were forced ing presence of an activating stimulus that cannot be prop-
toxic by treatment with aggregated AE or LPS and trans- erly cleared or is constantly renewed. This might be the case
ferred to hippocampal slice preparations. Marked tissue for AE plaques in AD. Vicious cycles can (further) drive
damage was observed. On the other hand, microglia treated microglia owing to neuronal decline, fueling damage by self-
with IL-4 exerted neuroprotection. The effect was accom- propelling recruiting of microglia (Lehnardt et al. 2008).
panied by induction of MHCII, upregulated insulin-like This may apply to various degenerative diseases (Block and
growth factor (IGF) I, and altered TNFD production. IFNγ Hong 2007). Failure to progress through a sequence of
had some protective effect, but conferred at high levels a phenotypes could arrest microglia in a functional state that
phenotype that interfered with oligodendrogenesis, which can only transiently be tolerated by the CNS. Interestingly, cer-
could be overcome by IL-4. Injection of IL-4–educated tain elements of a microglial response can continue, whereas
microglia into the cerebrospinal fluid of rodents with experi- others run down with time. Repetitive challenges through
mental autoimmune encephalomyelitis (EAE) resulted TLRs can result in progressively declining responses, prob-
in increased oligodendrogenesis and ameliorated disease ably involving desensitization, as shown for LPS-triggered
symptoms. IL-4–induced oligodendrogenesis-promoting NO and TNFD production. In contrast, cells continue
orientation was further investigated, revealing that a releasing prostaglandin E2 (PGE2). Accordingly, iNOS expres-
CD11b+CD11c+MHCII+TNFD– phenotype capable of sion decreases, whereas COX2 and PGE synthase remain ele-
IGF-I delivery rescues cells, CNS tissue, and animals from vated. The enzymes thus demonstrate a dissociated regulation
the adverse effects of toxic compounds, which drive a with time. A “continuum” of microglial phenotypes (Town
CD11b+CD11c–MHCII–TNFD– microglia. Dendritic cell et al. 2005) or heterogeneity of reactive macrophages found
(DC) orientation with APC potential (CD11c, MHCII) in a tissue (Kigerl et al. 2009; Mantovani et al. 2004) may,
of “protective” microglia, their plasticity and neurogenesis therefore, not just reflect a variety of fixed options chosen a
support indicate that engagement with innate and adaptive priori. Cells may, indeed, switch phenotypes based on feed-
immunity (and even proinflammatory cytokines) may not backs. Still it has to be verified whether phenotypes prevail-
necessarily and always impair the CNS (Hanisch et al. 2008; ing at stages of a CNS lesion, as in MS or AD, are based on
Rivest 2009; Simard and Rivest 2006). distinct subpopulations with specialized duties or whether
Thus, harm might not only result from an overshoot- individual cells adapt performances. Especially little is known
ing (acute) reaction, but also from maladapted phenotypes. about human microglia and how reactive phenotypes are
Misinterpretation of DAMPs as signs of an exogenous threat installed and modulated—and how they compete in ambigu-
may trigger defense programs at a cost of neurons and glia ous situations.
(Popovich et al. 1999; Schwartz et al. 2006). On the other
hand, failure to fight off brain tumors or metastasis—or even
2.7 R E S P O NS E T E R M I NAT I O N A N D T H E FAT E
providing assistance—would be equally devastating (Pollard
O F P O S TAC T I VAT E D M I C RO G L I A
2009; Pukrop et al. 2010).
The simplest feedback on activation could be based on dis-
appearance or clearance of the stimulus. Desensitization of
2.6 C O N T RO L OVE R T H E AC T I VAT I O N C O U R S E
signaling cascades, as another mechanism, is frequently based
Microglial activation is likely not a monophasic event. For a on receptor internalization. In some cases, receptor endo-
population, or even a single cell, induced genes and functions cytosis is rather a principle for triggering intracellular path-
may change throughout the course (Hanisch and Kettenmann ways. TLR4 initiates MyD88 signaling from the surface and
2007) (see Fig. 48.2). In vivo evidence derives from injuries, continues with TRIF activation in endosomes (Zanoni et al.
ischemia, or autoimmune challenges (Kettenmann et al. 2011). 2011). In still other cases, signaling arrest is caused by block
Profile shifts were studied for macrophages in more detail, of receptor domains, as for G protein–coupled receptors.
but can also be shown for microglia. Microglial challenges by As to microglia, involved principles are as many-sided as
PAMPs trigger CCL2, CCL3, CCL5, CXCL1, and CXCL2. the multitude of expressed receptors. Failure of termi-
IFNγ, driving M1 polarization, was found to completely reor- nating the activation can fuel smoldering inflammation.
ganize the profile. It enhanced monocyte-attracting CCL2, However, the neuronal circuitry cannot tolerate such an envi-
suppressed signals for neutrophils and Th1 cells, but spared ronment for long (Schwartz et al. 2006). Not only a tight con-
those for Th2 attraction (Häusler et al. 2002). Using IFNJ, trol of the initiation of activation, but also of its termination,
Th1 cells could thus self-limit their own recruitment. In turn, is required.
Th2 cytokines, like IL-4, could then direct a M2-like pheno- Besides autonomous mechanisms, such as timed produc-
type in support of Th2 responses and repair. Microglia may tion of IL-10 or induction of intracellular regulators of sig-
start with an emergency response to fight an infection or limit naling, feedbacks from other cells may impose de-escalation.

FAC TO R S C O N T R O L L I N G M I C R O G L I A L AC T I VAT I O N • 621


Whether and how microglia redistribute after local prolif- see microbes, and if so, only in truly threatening situations.
eration and migratory congregation or whether coordinated However, the CNS is a heterogeneous tissue, probably more
decline takes place by cell death or departure from the CNS than any other organ accommodating macrophage-like cells.
has not yet been properly addressed. Nor are the factors known Microglia in the various CNS divisions cannot ignore their
that would govern such processes (Hanisch and Kettenmann distinctions.
2007) (see Fig. 48.2). Microglia distribute throughout the CNS, but the den-
Postactivated microglia—cells that had mounted a tran- sity varies (Lawson et al. 1990). Regions with a high micro-
sient reaction—may resume to the surveillance modus and glia index, such as the substantia nigra (twice as high as other
reacquire a naïve state. Alternatively, some microglia may also areas), could be especially affected by inflammatory factors. In
remain altered, by receptor expression, signaling efficacy or combination with properties of the intrinsic neuronal popu-
transcriptional regulation. Such effects were shown for extra- lation (pathways for neurotransmitter synthesis), oxidative
neural macrophages (DeSanta et al. 2007). Experience from stress can develop a strong impact (Block and Hong 2007).
a former activation, such as through TLR4 (DeSanta et al. Yet microglial density may not be the only determinant of
2009), could influence properties on re-challenge (e.g., by epi- regional differences (Hanisch and Kettenmann 2007).
genetic modification). Microglia may themselves present with diversity in recep-
Infectious complications especially in development do tor expression and signaling organization, underlying some
not only affect maturing CNS structures, such as white mat- heterogeneity in responses. Several studies revealed distinct
ter, and causing lasting impairment, but also affect microglia expression patterns for CD11c, CD34, CD40, CD45, CD86,
responses later in life (Bilbo and Schwarz 2009; Bilbo et al. CXCL14, FcJRII, IGF-I, IL-6, iNOS, integrins, MHCII,
2005). Signaling systems with importance for defense as well neurotrophins, Tim-3, TNFD, Trem2, or 5D4+ keratan sul-
as developmental processes may perturb critical maturation fate (de Haas et al. 2008; Kettenmann et al. 2011). Central
events when being induced by early infection. IL-1 serves as an nervous system regions vary by cell communities as well as
example because it shares MyD88 signaling with TLRs. The micro milieus defined by prevalent neurotransmitters, ECM
original identification of the name-giving Toll, the Drosophila components, and cues associated with myelinated or vascu-
homolog, was based on morphogenetic functions. Recent lar structures. Expression profiles may reflect adaptations in
studies addressed the TLR potential in mammalian develop- housekeeping duties and reactive options. This is indicated,
ment (Rolls et al. 2007). Immune activation may thus conflict for example, by the CXCL10/CXCR3 system and its role in
with ontogenetic IL-1R/TLR/MyD88 signaling. neuronal death and glial activation in the hippocampus (van
In combination with an assumed (life-)long tissue resi- Weering et al. 2010). These differences are disguised when
dence of the microglial cell pool, imprints of earlier activation studying bulk responses of entire populations without cellular
episodes should also be considered for CNS diseases that do or anatomical resolution.
not have overt infectious associations, including brain tumors, Response heterogeneity could yet also apply to micro-
AD, or neuropsychiatric disorders. Provocative resemblance glia within a local population, as it is indicated by discrete
to lymphocyte memory is “foisted” on purpose and has basal or regulated expression patterns for cells in the imme-
already been attributed to natural killer cells (Ugolini and diate vicinity (Fitzner et al. 2011; Schmid et al. 2002, 2009).
Vivier 2009). Subsets are distinguished by the clearance of oligodendrocytic
Practical implications of modulating (probably sustained) exosomes laden with myelin, a contribution to physiological
microglial activity on an insult also relate to tactile allodynia. myelin turnover (Fitzner et al. 2011). In turn, upregulation
Neuropathic pain can develop on nerve injury. A role for of MHCII appears to be assigned to a complementary sub-
microglia has been demonstrated, followed by an elegant dis- population. Compartmentalization of endogenous material
section of a chain of molecular events that involve CCL21 and clearance and antigen presentation potential could segregate
P2X4 (Biber et al. 2011). Not only does such work improve conflicting functions. In the case of myelin, failure in seques-
the understanding of how microglia engages with neuronal tration could cause (auto)immune responses. A split by per-
functions, but it also nominates candidates for therapeutic formance is also seen when microglia are exposed to myelin
interference. debris (Regen et al. 2011; van Rossum et al. 2008). Its removal
is a prerequisite for remyelination and may lower the risk for
autoimmune rechallenge. On the contrary, material taken up
3 C E N T R A L N E RVO U S SYS T E M–S P E C I F I C by microglia could be presented to T cells. In both cases, the
CONDITIONS IN CONTROL physiological disposal and removal of damaged myelin, micro-
OF MICROGLIAL FUNCTIONS glial subtypes behave differently, probably based on receptor
sets for recognition, binding, and regulation, like Fc, comple-
Tissue macrophages differ by their environment and func- ment, scavenger, phosphatidyl, and purine receptors or PRRs
tional requirements (Davoust et al. 2008; Murray and Wynn (Gitik et al. 2011; Makranz et al. 2006; Neumann et al. 2009;
2011; Prinz et al. 2011). Alveolar macrophages are exposed Rotshenker 2003, 2009; Town et al. 2005).
to airborne germs. Peritoneal macrophages may occasionally Evidence also points to subpopulations with distinct
detect trace amounts of agents spilling from the commensal electrophysiological properties (Kettenmann et al. 2011).
flora. The CNS harbors various types of macrophages, in addi- Privileged production of key cytokines, such as TNFD, by
tion to microglia proper. Parenchymal microglia may rarely subsets would grant them a master control among “the”

622 • NEUROGLIA
microglial response. Although the concept of monocyte/mac- Anderson CF, Mosser DM. 2002. A novel phenotype for an acti-
rophage diversity owing to origin and location has a long his- vated macrophage: the type 2 activated macrophage. J Leukoc Biol
72:101–106.
tory (Treves 1984), studies now trace precursors and routes Beutler B, Rietschel ET. 2003. Innate immune sensing and its roots: the
of settlement, turnover rates under normal conditions, and story of endotoxin. Nat Rev Immunol 3:169–176.
replenishment under disease conditions (Davoust et al. 2006; Biber K, Neumann H, Inoue K, Boddeke HW. 2007. Neuronal “On”
Mildner et al. 2007; Prinz et al. 2011) (see chapter 15). First and “Off ” signals control microglia. Trends Neurosci 30:596–602.
investigations deal with microglia/subset-specific defects, Biber K, Tsuda M, Tozaki-Saitoh H, Tsukamoto K, Toyomitsu E, Masuda
T, Boddeke H, Inoue K. 2011. Neuronal CCL21 up-regulates micro-
such as in Hoxb8, which correlate with dysfunctions even at glia P2X4 expression and initiates neuropathic pain development.
the behavioral level (Chen et al. 2010). A conscious consider- EMBO J 30:1864–1873.
ation of microglial heterogeneity could also open new avenues Bilbo SD, Biedenkapp JC, Der-Avakian A, Watkins LR, Rudy JW,
for selective manipulation. Maier SF. 2005. Neonatal infection-induced memory impairment
after lipopolysaccharide in adulthood is prevented via caspase-1 inhi-
bition. J Neurosci 25:8000–8009.
Bilbo SD, Schwarz JM. 2009. Early-life programming of later-life brain
4 S U M M A RY A N D P E R S P E C T I VE S and behavior: a critical role for the immune system. Front Behav
Neurosci 3:14.
Microglia are resident myeloid cells and serve as principal Block ML, Hong JS. 2007. Chronic microglial activation and progressive
innate immune cells of the CNS. Functions in the healthy dopaminergic neurotoxicity. Biochem Soc Trans 35:1127–1132.
Block ML, Zecca L, Hong JS. 2007. Microglia-mediated neurotoxicity:
mature CNS relate to a surveillance of tissue homeostasis and uncovering the molecular mechanisms. Nat Rev Neurosci 8:57–69.
appear to include previously unnoticed contributions to the Boucsein C, Zacharias R, Farber K, Pavlovic S, Hanisch UK, Kettenmann
maintenance of structural integrity, neurogenesis, neuronal H. 2003. Purinergic receptors on microglial cells: functional expres-
plasticity, and signaling. Developmental roles include but also sion in acute brain slices and modulation of microglial activation in
exceed phagocytosis. All these functions are tightly regulated vitro. Eur J Neurosci 17:2267–2276.
Butovsky O, Landa G, Kunis G, Ziv Y, Avidan H, Greenberg N, et al.
by signaling inputs as mediated through a broad array of con- 2006a. Induction and blockage of oligodendrogenesis by differently
stitutively expressed receptors, including those that prepare activated microglia in an animal model of multiple sclerosis. J Clin
microglia for responses to infections, tumors, cell impairment, Invest 116:905–915.
and tissue injury of the most diverse cause and nature. PAMPs Butovsky O, Ziv Y, Schwartz A, Landa G, Talpalar AE, Pluchino S, et al.
and DAMPs, cytokines and chemokines, immunoglobulins 2006b. Microglia activated by IL-4 or IFN-gamma differentially
induce neurogenesis and oligodendrogenesis from adult stem/pro-
and complements lipid mediators and neurotransmitters exert genitor cells. Mol Cell Neurosci 31:149–160.
profound influences on reactive phenotypes. Shifts from the Cardona AE, Pioro EP, Sasse ME, Kostenko V, Cardona SM, Dijkstra IM,
surveillance (resting) mode to executive states are controlled by et al. 2006. Control of microglial neurotoxicity by the fractalkine
the net influence of activating and calming factors. “Activation” receptor. Nat Neurosci 9:917–924.
may induce further upregulation of receptors, ion channels, and Chen GY, Nunez G. 2010. Sterile inflammation: sensing and reacting to
damage. Nat Rev Immunol 10:826–837.
other cognate molecules to install, enhance, or modify sensitiv- Chen SK, Tvrdik P, Peden E, Cho S, Wu S, Spangrude G, et al. 2010.
ity to additional signals or change expression to render cells Hematopoietic origin of pathological grooming in Hoxb8 mutant
unresponsive—as part of adapted reactions. Cell contact and mice. Cell 141:775–785.
messengers of resident CNS and invading immune cells may Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, et al. 2005.
subsequently further shape functional profiles and eventually ATP mediates rapid microglial response to local brain injury in vivo.
Nat Neurosci 8:752–758.
organize de-escalation and installation of a postactivation status. David S, Kroner A. 2011. Repertoire of microglial and macrophage
Although the list of reported factors in microglia control is ever responses after spinal cord injury. Nat Rev Neurosci 12:388–399.
growing, future challenges include the need for understand- Davoust N, Vuaillat C, Androdias G, Nataf S. 2008. From bone marrow
ing their integrative actions. Practical use shall be made based to microglia: barriers and avenues. Trends Immunol 29:227–234.
on factors that decide on protective versus harmful actions of Davoust N, Vuaillat C, Cavillon G, Domenget C, Hatterer E, Bernard A,
et al. 2006. Bone marrow CD34+/B220+ progenitors target the
microglia to develop strategies of therapeutic moderation. inflamed brain and display in vitro differentiation potential toward
microglia. FASEB J 20:2081–2092.
de Haas AH, Boddeke HW, Biber K. 2008. Region-specific expression
AC K N OW L E D G M E N T S of immunoregulatory proteins on microglia in the healthy CNS. Glia
56:888–894.
This work was supported by grants from the German Research DeSanta F, Narang V, Yap ZH, Tusi BK, Burgold T, Austenaa L,
et al. 2009. Jmjd3 contributes to the control of gene expression in
Council (DFG, SFB/TRR43 and FOR1336). Figures have LPS-activated macrophages. EMBO J 28:3341–3352.
been based on published work with respective permission DeSanta F, Totaro MG, Prosperini E, Notarbartolo S, Testa G, Natoli
(Hanisch and Kettenmann 2007; Kettenmann et al. 2011; G. 2007. The histone H3 lysine-27 demethylase Jmjd3 links inflam-
Regen et al. 2011). mation to inhibition of polycomb-mediated gene silencing. Cell
130:1083–1094.
Fitzner D, Schnaars M, van Rossum D, Krishnamoorthy G, Dibaj
P, Bakhti M, et al. 2011. Selective transfer of exosomes from
REFERENCES oligodendrocytes to microglia by macropinocytosis. J Cell Sci
124:447–458.
Anderson CF, Gerber JS, Mosser DM. 2002. Modulating macrophage Galea I, Bechmann I, Perry VH. 2007. What is immune privilege (not)?
function with IgG immune complexes. J Endotoxin Res 8:477–481. Trends Immunol 28:12–18.

FAC TO R S C O N T R O L L I N G M I C R O G L I A L AC T I VAT I O N • 623


Gerber JS, Mosser DM. 2001. Reversing lipopolysaccharide toxic- Matzinger P. 2007. Friendly and dangerous signals: is the tissue in con-
ity by ligating the macrophage Fc gamma receptors. J Immunol trol? Nat Immunol 8:11–13.
166:6861–6868. Mildner A, Schmidt H, Nitsche M, Merkler D, Hanisch UK, Mack M,
Gitik M, Liraz-Zaltsman S, Oldenborg PA, Reichert F, Rotshenker et al. 2007. Microglia in the adult brain arise from Ly-6ChiCCR2+
S. 2011. Myelin down-regulates myelin phagocytosis by microglia monocytes only under defined host conditions. Nat Neurosci
and macrophages through interactions between CD47 on myelin 10:1544–1553.
and SIRP-alpha (signal regulatory protein-alpha) on phagocytes. Miller SI, Ernst RK, Bader MW. 2005. LPS, TLR4 and infectious
J Neuroinflammation 8:24. disease diversity. Nat Rev Microbiol 3:36–46.
Glezer I, Lapointe A, Rivest S. 2006. Innate immunity triggers oligoden- Mosser DM, Edwards JP. 2008. Exploring the full spectrum of mac-
drocyte progenitor reactivity and confines damages to brain injuries. rophage activation. Nat Rev Immunol 8:958–969.
FASEB J 20:750–752. Murray PJ, Wynn TA. 2011. Protective and pathogenic functions of
Gordon S, Taylor PR. 2005. Monocyte and macrophage heterogeneity. macrophage subsets. Nat Rev Immunol 11:723–737.
Nat Rev Immunol 5:953–964. Neumann H, Kotter MR, Franklin RJ. 2009. Debris clearance by micro-
Graeber MB. 2010. Changing face of microglia. Science 330:783–788. glia: an essential link between degeneration and regeneration. Brain
Hanisch UK. 2002. Microglia as a source and target of cytokines. Glia 132:288–295.
40:140–155. Nimmerjahn A, Kirchhoff F, Helmchen F. 2005. Resting microglial cells
Hanisch UK, Johnson TV, Kipnis J. 2008. Toll-like receptors: roles in are highly dynamic surveillants of brain parenchyma in vivo. Science
neuroprotection? Trends Neurosci 31:176–182. 308:1314–1318.
Hanisch UK, Kettenmann H. 2007. Microglia: active sensor and versa- O’Neill LA, Bowie AG. 2007. The family of five: TIR-domain-containing
tile effector cells in the normal and pathologic brain. Nat Neurosci adaptors in Toll-like receptor signalling. Nat Rev Immunol 7:353–364.
10:1387–1394. Olah M, Amor S, Brouwer N, Vinet J, Eggen B, Biber K, et al. 2012.
Häusler KG, Prinz M, Nolte C, Weber JR, Schumann RR, Kettenmann Identification of a microglia phenotype supportive of remyelination.
H, et al. 2002. Interferon-gamma differentially modulates the release Glia 60:306–321.
of cytokines and chemokines in lipopolysaccharide- and pneumo- Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P,
coccal cell wall-stimulated mouse microglia and macrophages. Eur J et al. 2011. Synaptic pruning by microglia is necessary for normal
Neurosci 16:2113–2122. brain development. Science 333:1456–1458.
Heneka MT, Nadrigny F, Regen T, Martinez-Hernandez A, Park BS, Song DH, Kim HM, Choi BS, Lee H, Lee JO. 2009. The struc-
Dumitrescu-Ozimek L, Jardanhazi-Kurutz D, et al. 2010. Locus ceruleus tural basis of lipopolysaccharide recognition by the TLR4-MD-2
controls Alzheimer’s disease pathology by modulating microglial functions complex. Nature 458:1191–1195.
through norepinephrine. Proc Natl Acad Sci U S A 107:6058–6063. Piccinini AM, Midwood KS. 2010. DAMPening inflammation by mod-
Kawai T, Akira S. 2010. The role of pattern-recognition receptors in innate ulating TLR signalling. Mediators Inflamm 2010.
immunity: update on Toll-like receptors. Nat Immunol 11:373–384. Pollard JW. 2009. Trophic macrophages in development and disease.
Kempermann G, Neumann H. 2003. Neuroscience. Microglia: the Nat Rev Immunol 9:259–270.
enemy within? Science 302:1689–1690. Popovich PG, Guan Z, Wei P, Huitinga I, van RN, Stokes BT. 1999.
Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. 2011. Physiology Depletion of hematogenous macrophages promotes partial hindlimb
of microglia. Physiol Rev 91:461–553. recovery and neuroanatomical repair after experimental spinal cord
Kigerl KA, Gensel JC, Ankeny DP, Alexander JK, Donnelly DJ, Popovich injury. Exp Neurol 158:351–365.
PG. 2009. Identification of two distinct macrophage subsets with Prinz M, Hanisch UK. 1999. Murine microglial cells produce and
divergent effects causing either neurotoxicity or regeneration in the respond to interleukin-18. J Neurochem 72:2215–2218.
injured mouse spinal cord. J Neurosci 29:13435–13444. Prinz M, Priller J, Sisodia SS, Ransohoff RM. 2011. Heterogeneity
Kreutzberg GW. 1996. Microglia: a sensor for pathological events in the of CNS myeloid cells and their roles in neurodegeneration. Nat
CNS. Trends Neurosci 19:312–318. Neurosci 13:1227–1235.
Lawson LJ, Perry VH, Dri P, Gordon S. 1990. Heterogeneity in the dis- Pukrop T, Dehgani F, Han-Ning C, Lohaus R, Byanga K, Heermann S,
tribution and morphology of microglia in the normal adult mouse et al. 2010. Microglia promote colonization of brain tissue by breast
brain. Neuroscience 39:151–170. cancer cells in a Wnt-dependent way. Glia 58:1477–1489.
Lehnardt S, Schott E, Trimbuch T, Laubisch D, Krueger C, Wulczyn Ransohoff RM, Perry VH. 2009. Microglial physiology: unique stimuli,
G, et al. 2008. A vicious cycle involving release of heat shock protein specialized responses. Annu Rev Immunol 27:119–145.
60 from injured cells and activation of toll-like receptor 4 mediates Regen T, van RD, Scheffel J, Kastriti ME, Revelo NH, Prinz M, et al.
neurodegeneration in the CNS. J Neurosci 28:2320–2331. 2011. CD14 and TRIF govern distinct responsiveness and responses
Liew FY, Pitman NI, McInnes IB. 2010. Disease-associated functions of in mouse microglial TLR4 challenges by structural variants of LPS.
IL-33: the new kid in the IL-1 family. Nat Rev Immunol 10:103–110. Brain Behav Immun 25:957–970.
Lin SC, Lo YC, Wu H. 2010. Helical assembly in the Rivest S. 2009. Regulation of innate immune responses in the brain. Nat
MyD88-IRAK4-IRAK2 complex in TLR/IL-1R signalling. Nature Rev Immunol 9:429–439.
465:885–890. Rolls A, Shechter R, London A, Ziv Y, Ronen A, Levy R, et al. 2007.
Makranz C, Cohen G, Reichert F, Kodama T, Rotshenker S. 2006. cAMP Toll-like receptors modulate adult hippocampal neurogenesis. Nat
cascade (PKA, Epac, adenylyl cyclase, Gi, and phosphodiesterases) Cell Biol 9:1081–1088.
regulates myelin phagocytosis mediated by complement receptor-3 Rotshenker S. 2003. Microglia and macrophage activation and the regu-
and scavenger receptor-AI/II in microglia and macrophages. Glia lation of complement-receptor-3 (CR3/MAC-1)-mediated myelin
53:441–448. phagocytosis in injury and disease. J Mol Neurosci 21:65–72.
Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. 2004. Rotshenker S. 2009. The role of Galectin-3/MAC-2 in the activation of
The chemokine system in diverse forms of macrophage activation and the innate-immune function of phagocytosis in microglia in injury
polarization. Trends Immunol 25:677–686. and disease. J Mol Neurosci 39:99–103.
Martinez FO, Gordon S, Locati M, Mantovani A. 2006. Transcriptional Rubartelli A, Lotze MT. 2007. Inside,outside, upside down:
profi ling of the human monocyte-to-macrophage differentiation damage-associated molecular pattern molecules (DAMPs) and redox.
and polarization: new molecules and patterns of gene expression. Trends Immunol 28:429–436.
J Immunol 177:7303–7311. Saijo K, Glass CK. 2011. Microglial cell origin and phenotypes in health
Martinez FO, Helming L, Gordon S. 2009. Alternative activation of and disease. Nat Rev Immunol 11:775–787.
macrophages: an immunologic functional perspective. Annu Rev Schmid CD, Melchior B, Masek K, Puntambekar SS, Danielson PE,
Immunol 27:451–483. Lo DD, et al. 2009. Differential gene expression in LPS/IFNgamma

624 • NEUROGLIA
activated microglia and macrophages: in vitro versus in vivo. Treves AJ. 1984. The origin of monocyte-macrophage heterogeneity:
J Neurochem 109 Suppl 1:117–125. possible alternatives. Med Hypotheses 14:335–346.
Schmid CD, Sautkulis LN, Danielson PE, Cooper J, Hasel KW, Ugolini S, Vivier E. 2009. Immunology: natural killer cells remember.
Hilbush BS, et al. 2002. Heterogeneous expression of the trigger- Nature 457:544–545.
ing receptor expressed on myeloid cells-2 on adult murine microglia. van Rossum D, Hanisch UK. 2004. Microglia. Metab Brain Dis 19:
J Neurochem 83:1309–1320. 393–411.
Schwartz M, Butovsky O, Brück W, Hanisch UK. 2006. Microglial phe- van Rossum D, Hilbert S, Strassenburg S, Hanisch UK, Bruck W.
notype: is the commitment reversible? Trends Neurosci 29:68–74. 2008. Myelin-phagocytosing macrophages in isolated sciatic
Simard AR, Rivest S. 2006. Neuroprotective properties of the innate and optic nerves reveal a unique reactive phenotype. Glia 56:
immune system and bone marrow stem cells in Alzheimer’s disease. 271–283.
Mol Psychiatry 11:327–335. van Weering HR, Boddeke HW, Vinet J, Brouwer N, de Haas AH, van
Smith DE. 2011. The biological paths of IL-1 family members IL-18 and RN, et al. 2010. CXCL10/CXCR3 signaling in glia cells differen-
IL-33. J Leukoc Biol 89:383–392. tially affects NMDA-induced cell death in CA and DG neurons of
Stewart CR, Stuart LM, Wilkinson K, van Gils JM, Deng J, Halle A, the mouse hippocampus. Hippocampus 36:368–387.
et al. 2010. CD36 ligands promote sterile inflammation through Wake H, Moorhouse AJ, Jinno S, Kohsaka S, Nabekura J. 2009.
assembly of a Toll-like receptor 4 and 6 heterodimer. Nat Immunol Resting microglia directly monitor the functional state of synapses
11:155–161. in vivo and determine the fate of ischemic terminals. J Neurosci
Takeuchi O, Akira S. 2010. Pattern recognition receptors and inflamma- 29:3974–3980.
tion. Cell 140:805–820. Zanoni I, Ostuni R, Marek LR, Barresi S, Barbalat R, Barton GM,
Town T, Nikolic V, Tan J. 2005. The microglial “activation” continuum: et al. 2011. CD14 controls the LPS-induced endocytosis of Toll-like
from innate to adaptive responses. J Neuroinflammation 2:24. receptor 4. Cell 147:868–880.

FAC TO R S C O N T R O L L I N G M I C R O G L I A L AC T I VAT I O N • 625


49.
ROLES OF ACTIVATED MICROGLIA
Kelly R. Miller, Stefan Prokop, and Frank L. Heppner

A B B R E VI AT I O N S “resting” microglia in the healthy brain are highly dynamic


cells (Davalos et al. 2005; Nimmerjahn et al. 2005) (see
AA arachidonic acid chapter 47); therefore, microglia activation refers specifically
AD Alzheimer’s disease to a distinct repertoire of cellular behavior that is restricted to
ATP adenosine triphosphate situations in which there is a disturbance in CNS homeostasis.
BMEC brain microvascular endothelial cell Careful consideration of this definition highlights at least
[Ca2+]i intracellular calcium activity three critical aspects of microglia reactivity: (1) The range of
CNS central nervous system responses that microglia exhibit upon activation are widely
GM-CSF granulocyte-macrophage colony varying and highly context-dependent; (2) in the absence
stimulating factor of primary “microgliopathies” (Bianchin et al.), microglia
IL-1β interleukin-1 beta activation constitutes a reaction to an already present CNS
IL-3 interleukin-3 disturbance; and (3) microglia activation refers to an in vivo
IL-6 interleukin-6 cellular response. By nature, the role of microglia is to detect
JAK janus kinase and respond to homeostatic perturbations in their microen-
LPS lipopolysaccharide vironment; therefore, cells in vitro are likely to exhibit behav-
MAPK mitogen–activated protein kinase iors that do not translate to the in vivo situation. This chapter
M-CSF macrophage colony stimulating factor describes how microglia respond to CNS insult with a focus
MCP1 monocyte chemotactic protein-1 on the in vivo setting.
NADPH nicotinamide adenine dinucleotide phosphate The functional significance of microgliosis varies depend-
NgR Nogo receptor ing on the pathological circumstances. Reactive microglia are
NO nitric oxide capable of responding to brain insult with a wide repertoire of
NPY neuropeptide Y specialized activity (Fig. 49.1). One prominent role of activated
PMA phorbol myristate acetate microglia in instances of neuronal degeneration is phagocyto-
PNS peripheral nervous system sis of tissue debris. Microglia also serve as antigen-presenting
ROS reactive oxygen species cells (for details, see chapter 50). Moreover, upon activation,
STAT signal transducer and activator of microglia migrate to sites of damage and proliferate. Finally,
transcription activated microglia upregulate or produce de novo immu-
TNFα tumor necrosis factor alpha nomodulatory factors, including cytokines and chemokines,
that mediate both autocrine and paracrine signaling. With the
exception of antigen presentation (see chapter 50), each of the
1 INTRODUCTION aforementioned aspects of microglia reactivity is elaborated in
the sections to follow.
The term microglia activation is a somewhat ambiguous
and problematic phrase that is often dangerously overused.
Historically, this term has been introduced as a mainly descrip- 2 M I C R O G L I A P R O L I F E R AT I O N
tive idiom derived from the morphological appearance of
microglia in pathological conditions, which implies a uniform The mitotic potential of microglia in the adult brain was
cellular response to a variety of stimuli, while misleadingly also first demonstrated in the late 1960s using autoradiographic
assuming that microglial cells are quiescent bystanders in the studies employing [3H] thymidine incorporation in the con-
normal, nondiseased central nervous system (CNS) that only text of brain injury (Friede and Johnstone 1967; Kreutzberg
awaken when called on. Of course, the microglia cosmos is not 1966). Although microglial cells have been shown to divide
black and white, and this is why we need to adjust our view in the absence of injury, this phenomenon occurs to a much
on, and even the terms we use to describe, microglia activa- lesser degree in the healthy brain and is thought to reflect nor-
tion. So, what is microglia activation? To activate is “to make mal cell turnover (Dalton et al. 1968; Tonchev et al. 2003).
something active or more active” (Merriam-Webster Online Population expansion of microglia in response to CNS insult
Dictionary 2012). In regard to microglia biology, more active is allows a more robust cellular response than might otherwise
indeed the appropriate description. It is now well known that be possible by the existing pool of cells in the vicinity of brain

626
protein aggregates resting microglial cell injury/trauma

CNS Insult
infection ischemia

activated microglia cell

IL-1βra
NPY
cannabinoids
Nogo-A
anti-inflammatory drugs Apoptosis of excess
Stages of Microglia Activation

cells

1. Migration C5a
ATP
IL-1β 2. Proliferation
M-CSF
GM-CSF
IL-6
TNF-α
IL-1β
ATP
NADPH oxidase/H2O2

Ab
bacteria
phosphatidylsering
asialoglycoprotein
vitronectin
4. Phagocytosis 3. Cytokine production
IL-10
TGFb

IL-1β
IL-6
TNF-α
IFNγ
IL-2
M-CSF
Resolution of

resting microglial cell senescent microglial cell


Activation
Microglia

Figure 49.1 Functions of Activated Microglia. Initial CNS insult leading to microglia activation. Photomicrographs depict: (Upper left panel) Protein
aggregates (β-amyloid plaques [4G8 immunohistochemistry] left; and neurofibrillary tangles [AT-8 immunohistochemistry] right). (Lower left panel)
Infection (cerebral fungal abscess [PAS staining] left; and cerebral toxoplasmosis [H&E staining] right). (Upper right panel) Trauma (severe CNS
trauma with intracerebral hematoma). (Lower right panel) Ischemia (hypoxic neuronal damage [H&E staining] right; acute cerebral infarction [H&E
staining] left]. Microglia activation. Photomicrographs depict: (1) Migration—activated microglia [Iba-1 immunohistochemistry, brown] gathering
around cerebral β-amyloid plaques (4G8 immunohistochemistry, red) in a mouse model of AD. (2) Proliferation—proliferating microglia in the spinal
cord after peripheral nerve lesion (Iba-1 green; BrdU, blue). (3) Cytokine secretion—activated murine microglia after intracerebral LPS challenge
(Iba-1 immunohistochemistry, brown) signaling to astrocytes (GFAP immunohistochemistry, lower right) and neurons (H&E staining, lower left).
(4) Phagocytosis—in vitro phagocytosis assay using acute slice cultures (Iba-1 immunohistochemistry, red; Latex beads, green).

R O L E S O F AC T I VAT E D M I C R O G L I A • 627
damage; however, it is important to note that inhibition of (M-CSF) and granulocyte-macrophage colony stimulating
microglia proliferation does not necessarily prevent cellular factor (GM-CSF) (Kloss et al. 1997; Raivich et al. 1994);
reactivity and effector functions (Raivich et al. 1994). Despite cytokines, such as interleukin-1 beta (IL-1β) (Mander et al.
the numerous contexts in which microglial activation has 2006), tumor necrosis factor alpha (TNFα) (Ganter et al.
been observed, mitosis proves to be a prominent and consist- 1992; Merrill 1991), interleukin-6 (IL-6) (Kiefer et al. 1993;
ent component of the cellular response to injury. Notably, the Streit et al. 1998, 2000), and interleukin-3 (IL-3) (Kloss et al.
proliferative activity of microglia contrasts other CNS cells, 1997); chemokines, prominently including monocyte chemo-
including astrocytes, which undergo cell division in a much tactic protein-1 (MCP1/CCL2) (Hinojosa et al. 2011) and
more limited context (see chapter 51). purines, specifically ATP (Baricordi et al. 1999).
Much of our insight about microglia proliferation has been Binding of these aforementioned factors to their respec-
gained by studying regenerating nerve models (Cammermeyer tive receptors serves to activate many of the same signaling
1965; Kreutzberg 1966; Miller and Streit 2007; Streit and pathways that ultimately converge to induce gene transcrip-
Kreutzberg 1988). Data from these and other acute and tion of, among other transcripts, cell cycle regulators such
chronic neural injury models reveal that microglial prolifera- as the cell cycle–associated proteins cyclin D1, E, A, and
tion begins as early as 12 hours post-lesion, peaks between 3 cyclin-dependent kinase inhibitor p21. Specifically, the
and 15 days after insult depending on the nature of the injury, janus kinase ( JAK)-signal transducer and activator of tran-
and declines thereafter (Ladeby et al. 2005; Sjostrand 1971; scription (STAT) pathway ( JAK-STAT) plays a prominent
Streit and Kreutzberg 1988; Tonchev et al. 2003). Following role in signal transduction of a wide array of cytokines and
a proliferative burst, homeostasis of the microglia popula- growth factors leading to changes in cellular activity, includ-
tion is regulated through apoptotic death of excess cells, a ing proliferation (Aaronson and Horvath 2002). Among the
process that can be detected as early as 4 days postinjury and proliferation-inducing molecules described in the preced-
persists for up to 21 days (Gehrmann and Banati 1995; Jones ing, the CSFs and interleukins have been shown to initiate
et al. 1997). Although microglial mitotic activity typically microglia proliferation by signaling through the JAK-STAT
occurs during early stages of insult and subsequently resolves, pathway (Liva et al. 1999; Nosaka et al. 1999). Although
repeated stimulation in the form of lipopolysaccharide (LPS) common, the JAK-STAT pathway is certainly not exclusive
administration has been shown to induce continued prolifera- in the mediation of promitotic signaling cascades, and the
tion for up to 45 days (Shankaran et al. 2007). Nevertheless, Ras- mitogen–activated protein kinase (Ras-MAPK) path-
the exact proliferative capacity of microglia is not known, and way has also been described to act downstream of the men-
data from in vivo experiments wherein microglia were sub- tioned proliferation-inducing factors, including the CSFs
jected to repeated activation over the course of a year suggests and MCP-1, for example ( Jimenez-Sainz et al. 2003; Nicola
that the mitotic potential of microglia may be finite (Miller 1990). Similarly, ATP binding to the ionotropic P2X7 recep-
and Streit 2007), despite their telomerase enzyme expression tor on microglia has been shown to activate MAPK pathway
(Flanary and Streit 2004), which serves as a compensatory proteins, ERK 1/2 and MEK (Skaper et al. 2010) and leads
mechanism for telomere attrition. to transient Ca2+ currents in microglia (Eichhoff et al. 2011;
In contrast to the well-characterized proliferative response Oshimi et al. 1999).
of microglia, the mechanisms underlying its regulation prove An important regulator of microglia proliferation upon
more complex and challenging to delineate. stimulation by many of the discussed factors, specifically
Although microglia proliferation can be readily induced including IL-1β, TNFα, ATP, and GM-CSF as well as arachi-
by countless substances in vitro, the physiological relevance of donic acid (AA), phorbol myristate acetate (PMA) and fibrillar
such manipulations is not clear, and the highly reductionistic Aβ 1–40, is hydrogen peroxide (H2O2) generated from nico-
nature of the setting ensures that critical regulatory elements tinamide adenine dinucleotide phosphate (NADPH) oxidase
of microglial activity are lacking. Nonetheless, careful consid- (Nox) by microglia ( Jekabsone et al. 2006). H2O2 produced
eration of both in vivo and in vitro data together reveals some by the degeneration of superoxide can induce cell death at high
converging mechanisms governing microglial mitosis. concentrations, but at low concentrations can affect cell signal-
In principle, nearly any stimulus that induces microg- ing pathways, including those previously mentioned, through
lia activation will also serve to incite proliferation, whether the oxidation of target molecules (Groeger et al. 2009). In this
directly or indirectly. Therefore, it is beyond the scope of this way, H2O2 produced by the phagocyte-expressed Nox enzyme
chapter to detail every condition associated with microglia is capable of inhibiting enzymes, including phosphatases,
proliferation (see chapter 48 for more details), but discussion thereby influencing signal transduction pathways such as the
will rather focus on the most commonly identified signal- JAK-STAT and Ras-MAPK pathways ultimately resulting in
ing pathways mediating microglia mitosis. The best studied microglia mitosis.
models wherein microglia activation and subsequent cell divi-
sion has been described include nerve injury models, stroke,
infection models, including LPS administration and models 3 M I C R O G L I A M I G R AT I O N
of (degenerative) CNS disease, especially Alzheimer’s disease
(AD). These varying stimuli often elicit production of com- Although resting microglia engage in continual extension and
mon promitotic factors, including, but not limited to, growth retraction of their processes to survey the surrounding milieu,
factors, especially macrophage colony stimulating factor they are believed to exist as nonmigratory cells in the absence

628 • NEUROGLIA
of activation. However, in vitro and in vivo studies have dem- microglia migration varies depending on the inciting stimulus,
onstrated the migratory capacity of activated microglia in again highlighting the very specialized and heterogeneous
response to a multitude of chemoattractants and brain insult, responses of microglia to different types of CNS insult.
respectively. The ability of microglia to migrate to sites of Despite the multitude of activating stimuli so far shown
brain damage and disease is crucial for fulfillment of their role to induce microglia migration, some prevailing signaling
as CNS resident immunocompetent cells. Almost without molecules have been identified. The initial phase of chemot-
exception, microglia can be found perilesionally where they axis involves binding of a chemoattractant to its receptor.
secrete signaling molecules, including chemokines that serve Perhaps the most well-defined factor known to induce migra-
to alert other microglia and astrocytes and recruit more cells tory activity in microglia (and leukocytes) is complement
to the injury site, phagocytose tissue debris and pathogens, 5a (C5a). Complement proteins are known to be produced
and present antigen to peripherally derived immune cells. at sites of CNS insult, and thus serve as ideal candidates for
Other important activities of activated microglia that require mediation of microglia chemotaxis. The major C5a receptor,
migration to injured neurons involves synaptic stripping (see CD88, is expressed on microglia and has even been found to
section 5). This phenomenon has now been observed in be upregulated on microglia in the vicinity of amyloid plaques
numerous contexts, and in some instances is thought to in AD mouse models (Cudaback et al. 2011). Binding of C5a
protect neurons from excitotoxic inputs (Kreutzberg and to its receptor has been shown to induce almost immediate
Hager 1966; Streit and Kreutzberg 1988; Trapp et al. 2007). changes in microglia behavior, including ruffling of cellular
Additionally, in AD for example, microglia migrate to and membranes followed by lamellipodia extensions (Nolte et al.
cluster around amyloid plaques (Fig. 49.2), but their function 1996). This activity was accompanied by a rearrangement of
in this setting remains unresolved. Although microglia are the actin cytoskeleton and increased intracellular calcium
invariably present at sites of CNS pathology, the mechanisms activity [Ca2]i. Similarly, the neuronally derived signaling mol-
regulating migration, like all aspects of microglia reactivity, ecule fractalkine also elicits microglia chemotaxis in response
are complex and tightly regulated and also difficult to study to injury that is accompanied by increased [Ca2]i (Harrison
in living organisms. et al. 1998). As mentioned, microglia are sensitive to extracel-
Most knowledge gained about the specific molecules lular ATP levels, and data reveals that binding of ATP to puri-
that induce microglia mobilization and the signaling path- nergic receptors also serves to induce microglia migration. In
ways involved has been obtained from in vitro studies using fact, in contrast to the great deal of in vitro data gathered on
transwell migration assays involving the Boyden chamber and this topic, ATP-induced microglia motility has also been dem-
time-lapse imaging of cells in culture or ex vivo brain slices. onstrated in the living neocortex of mice using two-photon
One elegant study that provides great insight into the charac- imaging (Davalos et al. 2005). Several lines of evidence sug-
teristics of long-distance microglia migration was conducted gest that P2X4 and P2Y12 receptors may be the primary media-
using time lapse confocal imaging of in vivo–injured, acutely tors of ATP-induced microglia motility, but involvement of
isolated living brain slices (Carbonell et al. 2005). This study other purinergic receptors is not precluded (Honda et al.
revealed that extensive migration of microglia occurs as early 2001; Horvath and DeLeo 2009). Like C5a, ATP is present at
as 24 hours after injury and peaks at 3 days. Furthermore, micro- foci of brain damage or disease through release from distressed
glia reached a maximum speed of greater than 10 μm/minute. cells.
Interestingly, data from this study demonstrate that the speed of To balance pro-migratory signals, several factors have also
been identified that serve to inhibit microglia motility. In
general, antiinflammatory molecules that act to dampen the
overall microglia activation response can also inhibit migra-
tory activities. For example, exogenous agents ranging from the
tetracycline antibiotic minocycline to the antiinflammatory
spice curcumin inhibit numerous aspects of microglia reactiv-
ity, including migration (Karlstetter et al. 2011). Similarly, the
endogenously derived IL-1β receptor antagonist (IL-1βra)
and neuropeptide Y (NPY) inhibit IL-1β–induced microglia
migration (Ferreira et al. 2012). In addition, cannabinoids have
also been shown to hinder microglia chemotaxis toward the
microglia-derived virus-specified trans-activating protein, Tat
(Fraga et al. 2011). Alternatively, an example of a more spe-
cialized anti-migratory signaling axis involves Nogo-A and
Nogo receptor (NgR). In addition to neurons, microglia have
50 μm been found to express NgR, which when bound by its ligand,
Nogo-66 (a component of the myelin associated inhibitory
molecule, Nogo-A) mediates inhibition of microglia migration
Figure 49.2 Activated microglia gathering around a cerebral by affecting cell adhesion, polarization, and membrane protru-
β-amyloid plaque in a mouse model of Alzheimer’s disease (Iba-1 sion formation (Yan et al. 2011). Moreover, expression of NgR
immunohistochemistry, brown; 4G8 immunohistochemistry, red). and its coreceptor, Troy/Lingo-1, were found to be expressed

R O L E S O F AC T I VAT E D M I C R O G L I A • 629
by microglia in demyelinating lesions of multiple sclerosis, sug- resulting from cell dysfunction or chronic stimulation may be
gesting that this molecule may have a distinct role in regulation harmful to the surrounding brain tissue. Many factors secreted
of the microglial inflammatory response (Satoh et al. 2007). by microglia act in a paracrine fashion on neighboring cells,
Finally, in vitro findings point out that many CNS-resident such as neurons and astrocytes, but microglia also engage in
cells can and likely do participate in the complex and fine-tuned critical autocrine signaling that can serve to self-regulate other
orchestration of the brain’s intrinsic inflammatory response. aspects of cellular activation, such as morphology (see chapter 8
Specifically, evidence supports a role for soluble factors pro- for details), migration (discussed in the preceding) and fur-
duced by brain microvascular endothelial cells (BMECs) in ther cytokine elaboration. This section describes the modes of
inhibition of microglia migration (Wang et al. 2011). Although cytokine signaling that microglia engage in and highlights the
this was not an exhaustive enumeration of all factors that may varying and specific profiles of microglia cytokine elaboration
influence the motility of activated microglia, and there are sure that occur under different pathological circumstances.
to be many more interesting and important regulators identi-
fied in the future, the information known to date clearly indi-
4.1 AU TO C R I N E S I G NA L I N G
cates that like all aspects of microglia reactivity, perilesional
migration is a well-controlled process within the CNS. Autocrine signaling refers to the secretion of a substance by a
cell that acts on surface receptors of the same cell. Microglia
express myriad cytokine and chemokine receptors that bind
4 C Y TO K I N E A N D C H E M O K I N E molecules synthesized by the cells themselves (see chapters 19
PRODUCTION and 22 for more details).

The production of cytokines and chemokines following activa-


4.2 PA R AC R I N E S I G NA L I N G
tion is a prominent feature of reactive microgliosis and may per-
haps be the best-studied aspect of microglia function. Microglia Signaling molecules produced by activated microglia have a pro-
are capable of producing a great variety of cytokines and found impact on neighboring glial cells, neurons, and endothe-
chemokines, and new evidence for the impact of these signaling lial cells. The cytokine/chemokine signaling cascade initiated
molecules on the CNS, be it in disease or physiological settings, by activated microglia usually only affects tissue within the
is constantly added. Basic principles of cytokine and chemokine local microenvironment. This mechanism is thought to act as
production by activated microglia and important signaling mol- a safeguard to contain potentially harmful effects of overactive
ecules and interactions are discussed herein (Table 49.1). The cytokine production. However, long-range effects of signaling
interested reader is referred elsewhere for a detailed overview molecules secreted by activated microglia include cross-talk
of factors produced by activated microglia (Hanisch 2002) with the peripheral immune system, the recruitment of periph-
and to chapter 22 for a description of the receptors mediating eral cells to the brain, and influences on the endocrine system.
cytokine and chemokine signaling. Most evidence concerning Among the effects of microglia-mediated paracrine signaling,
cytokine/chemokine production by microglia has been derived it has been shown that cytokines derived from activated micro-
from in vitro studies using isolated cells, mainly from perinatal glia are important for astroglial responses to occur (Balasingam
tissue, mixed glial cultures, and organotypic slice culture sys- et al. 1996). An excellent example is the instance of neuro-
tems. Data extracted from these analyses have broadened our pathic pain, in which it is hypothesized that early microglia
understanding of microglia function, but must be interpreted activation serves to instigate astrocyte reactivity and promote
with care in light of the fact that primary cell cultures derived an inflammatory process that culminates in central sensitiza-
from perinatal tissue may not reflect the resting state of micro- tion (see chapter 68). This theory highlights the inhibition
glia, which is only achieved later in development (Draheim of microglia activation as a therapeutic target for this condi-
et al. 1999). Furthermore, a fact that should not be underes- tion. Among the numerous factors produced by microglia that
timated is that single microglia or mixed-glial culture systems act on astrocytes throughout the brain are IL-1β, IL-2, IL-6,
lack other cell types that serve to form important signaling net- TNFα, and M-CSF (for more detail, see chapters 47 and 53).
works in the in vivo setting. Finally, acute tissue damage occur- Furthermore, IL-1, IL-2, and IL-6 secreted by activated micro-
ring during preparation of organotypic slice cultures can lead to glia can impact growth of oligodendrocytes (Hanisch 2002),
inflammatory changes that may confound experimental results. and several microglia-derived cytokines have been implicated
These may be some of the very reasons why we still do not have in the pathogenesis of demyelinating diseases, such as multiple
a complete and accurate picture of “microglia activation” as it sclerosis (for more detail, see chapter 61). Lastly, microglia can
occurs in reality, that is, in the living mature brain upon distur- influence neuronal activity via the elaboration of cytokines,
bance of CNS homeostasis. including IL-1, IL-2, interferons, and TNFα, among others
Microglial elaboration of cytokines/chemokines is a tightly (Rothwell et al. 1996) (see chapters 53 and 68).
regulated and gradually escalating process. The production of
these factors by activated microglia occurs in a graded fashion
4.3 C O N T E X T-D E P E N D E N T C Y TO K I N E
in relation to the severity and nature of the insult. Although
P RO D U C T I O N
microglia cytokine/chemokine secretion is generally aimed at
restoring homeostasis after an insult and is therefore locally and As mentioned, microglia activation and the resulting cytokine/
temporally restricted, excess production of effector molecules chemokine response is a process that is tightly regulated and

630 • NEUROGLIA
Table 49.1 CYTOKINES AND CHEMOKINES PRODUCED BY ACTIVATED MICROGLIA
CYTOKINE/CHEMOKINE ABBREVIATION TYPE OF INJURY MAIN TARGETS REFS.

Growth regulated oncogene α GROα Inflammation Leucocytes; (Filipovic et al. 2003;


oligodendrocytes Janabi et al. 1999)

Interleukin-1α/β Il-1α/β Infection, ischemia, stroke, T and B cells, monocytes, (Hanisch 2002; Raivich
excitotoxicity, mechanic microglia; neurons; et al. 1999)
injury pituitary cells
Interleukin-1 receptor antagonist Il-1ra Infection, ischemia, stroke, T and B cells, monocytes, (Simi et al. 2007)
excitotoxicity, mechanic microglia; neurons;
injury pituitary cells
Interleukin-2 Il-2 Infection T and B cells, NK cells, (Hanisch 2002)
macrophages, microglia
Interleukin-3 Il-3 Infection, neurodegeneration hematopoietic progeni- (Lee et al. 1993)
tor cells, macrophages,
microglia
Interleukin-4 Il-4 Infection, neurodegeneration T and B cells, mac- (Hanisch 2002)
rophages, microglia
Interleukin-6 Il-6 Infection, trauma T and B cells, plasma cells, (Hanisch 2002; Raivich
liver cells, microglia et al. 1999)
Interleukin-10 Il-10 Infection, neurodegeneration T and B cells, mac- (Hanisch 2002;
rophages, microglia Kettenmann et al. 2011)
Interleukin-12 Il-12 Infection T and B cells, mac- (Hanisch et al. 2001;
rophages, microglia Taoufik et al. 2001)
Interleukin-13 Il-13 Infection T and B cells, mac- (Hanisch 2002;
rophages, microglia Kettenmann et al. 2011)
Interleukin-15 Il-15 Infection, mechanic injury T and B cells, NK cells, (Gomez-Nicola et al.
macrophages, microglia 2008; Hanisch 2002)
Interleukin-18 Il-18 Infection T and B cells, mac- (Alboni et al. 2010)
rophages, microglia
Gamma interferon inducible IP-10 (CXCL10) Infection, T cells, macrophages, ( Jack et al. 2005; Xia and
protein-10 neurodegeneration microglia, dendritic cells Hyman 1999)
Monocyte chemoattractant MCP-1 (CCL2) Neurodegeneration, isch- T cells, macrophages, (Xia and Hyman 1999)
protein-1 emia, epilepsy microglia
Macrophage colony stimulating M-CSF Ischemia, injury Hematopoietic stem cells, (Kettenmann et al. 2011)
factor Neurodegeneration macrophages, microglia,
neurons
Macrophage-derived chemokine MDC ?? Macrophages, microglia, (Godiska et al. 1997)
NK cells
Macrophage inflammatory MIP-1α Infection, ischemia, Macrophages, microglia (Diab et al. 1999)
protein-1α/-1β (CCL3)/1β(CCL4) neurodegeneration
Macrophage inflammatory MIP-2 (CXCL2) Infection Neutrophils, hematopoi- (Diab et al. 1999)
protein-2 etic stem cells
Macrophage inflammatory MIP-3β (CCL19) Neuroinflammation Dendritic cells, (Columba-Cabezas et al.
protein-3β lymphocytes 2003)
Transforming growth factor TGFβ Infection, tissue damage, Lymphocytes, monocytes, (Streit et al. 1998;
neurodegeneration microglia Wyss-Coray et al. 2001)
Tumor necrosis factor α TNFα Infection, injury, Neurons, astrocytes, (Hanisch 2002; Raivich
neurodegeneration microglia, lymphocytes et al. 1999)
Regulated on activation, normal RANTES (CCL5) Infection, neurodegeneration T cells, eosinophils, baso- (Hanisch 2002)
T cell expressed and secreted phils, leukocytes
Fractalkine CX3CL1 Neuronal injury Microglia (Hanisch 2002; Zujovic
et al. 2005)

R O L E S O F AC T I VAT E D M I C R O G L I A • 631
adapted to the respective pathological stimulus. Not only infection. On the other hand, an uncontrolled production
the specific factors themselves, but also the amounts that are of cytotoxic molecules can also harm the surrounding CNS
released are modulated to suit the severity and nature of the tissue. For example, high levels of IFN-γ can directly induce
tissue lesion. Low levels of factors, including M-CSF and apoptosis by causing upregulation of Fas and Fas ligand (Badie
TGFβ1, can be detected in unchallenged adult brains (Kiefer et al. 2000). Again, the simultaneous production of TGFβ1
et al. 1993; Streit et al. 1998). However, on activation, micro- can counteract some effects of IFN-γ and helps to control the
glia cells upregulate and secrete a panel of cytokines/chemok- inflammatory reaction.
ines that are common to many different pathological stimuli,
yet which are specifically adjusted to the particular injurious
4.3.3 Neurodegenerative Disease
setting at hand. Besides M-CSF and TGFβ1, the proinflam-
matory factors TNFα, IL-6, IL-1β, and nitric oxide (NO) Chronic activation of microglia and subsequent cytokine and
belong to the basic set of signaling molecules secreted by chemokine secretion has been described extensively in several
activated microglia (Raivich et al. 1999). neurodegenerative diseases (McGeer and McGeer 1997; Wyss-
Coray 2006) (see chapters 63, 64 and 65). This protracted
reactivity is characterized by upregulation of the mentioned
4.3.1 Brain Injury
cytokines in activated microglia, especially IL-1β and TNFα.
Traumatic spinal cord injury leads to a rapid upregulation of It has been proposed that microglia-mediated neuroinflam-
IL-1β, IL-6, and TNFα by activated microglia (Bartholdi mation serves to exacerbate neurodegenerative processes,
and Schwab 1997; Streit et al. 1998; Yang et al. 2005). thereby significantly contributing to disease progression. This
Alternatively, the regenerating facial nerve transection model, theory has been called the neuroinflammatory cascade hypothe-
one of the most widely used animal models for studying injury- sis (Streit 2004) in the context of AD and has fueled clini-
induced microglia activation, is associated with only low lev- cal trials on the use of antiinflammatory medications for AD
els of TNFα and IL-1β and a sustained secretion of IL-6. In treatment, with equivocal success. Notably, chronic microg-
contrast, IL-6 is rapidly downregulated after an initial spike lia activation in response to neurodegenerative conditions is
following traumatic spinal cord injury (Streit et al. 1998). associated with a “senescent” phenotype that does not support
Likewise, TNFα is strongly induced by severe neuronal dam- classical functions of activated microglia (Miller and Streit
age following traumatic brain injury or ischemia (Bruce et al. 2007). In this context, microglia-mediated bystander damage
1996; Seilhean et al. 1997), but is almost absent in the facial may occur as a result of a dysregulation of otherwise tightly
nerve axotomy paradigm (Streit et al. 1998). High levels of controlled microglia activity. Taken together, this suggests
TNFα have been associated with direct toxic effects on neu- that fine-tuned manipulation of specific microglial signaling
rons (Kraft et al. 2009) and myelin (Hartung 1993); however, pathways known to be altered in AD may be a successful ther-
animal studies demonstrate enhanced neuronal death and apeutic strategy. At present, the impact of microglia activation
poorer prognosis on deficiency of TNFα signaling (Yang et al. and subsequent cytokine/chemokine secretion on disease pro-
1998), indicating a neuroprotective effect of TNFα, especially gression is a matter of hot debate and the interested reader is
at low levels. Notably, this example serves to illustrate the dual referred to chapters 63, 64 and 65 and elsewhere (Wyss-Coray
effects that cytokines produced by activated microglia can 2006) for further reading.
have. Besides the mentioned factors that are primarily known
for their proinflammatory effects, microglia also produce the
4.3.4 Classical versus Alternative Activation
antiinflammatory molecule TGFβ1 on activation (Kiefer
et al. 1995), which serves to subdue the proinflammatory In analogy to peripheral macrophages, activated microglia can
response occurring after injury and prevent harmful effects on adopt two main functional phenotypes. Microglia may exhibit
CNS tissue. a classically activated phenotype that is induced by IFN-γ
or LPS and generally promotes Th1 responses and neuronal
injury. This “classical” response better mimics the activation
4.3.2 Infection
profile well-known from ex vivo studies. Under these circum-
Infection of the CNS or meninges also leads to prominent stances, microglia stereotypically secrete the proinflammatory
upregulation of the aforementioned cytokines in microglia. cytokines TNFα, IL-1β, and reactive oxygen species (ROS)
Besides robust increases in TNFα expression, secretion of and NO. Alternatively, IL-4 or IL-10 can induce an antiin-
IFN-γ is also observed in association with specific infectious flammatory or alternatively activated phenotype in microglia
conditions (Suzuki et al. 2005; Wang and Suzuki 2007). (Ebert et al. 2008; Ponomarev et al. 2007; Town et al. 2005).
Although microglia are not a primary source of IFN-γ under Microglia that exhibit the latter activation profile promote
most pathological circumstances, IFN-γ is a strong regulator resolution of inflammation and tissue repair, support Th2
of subsequent cytokine production by microglia and induces functions, and primarily secrete IL-4, IL-10, and IGF-1. This
the release of molecules important for phagocytosis, antigen concept highlights the functional plasticity of microglia and
presentation, and cytotoxic functions of microglia (Raivich is very helpful in explaining the differential impact of micro-
et al. 1999). This activation-linked signaling cascade is simi- glia cells in various disease contexts or during progression
lar to what can be observed with other tissue macrophages, of one and the same disease. Significantly, one must keep in
and which is known to aid in controlling and clearing the mind that these two distinct phenotypes describe the state of a

632 • NEUROGLIA
single, highly specialized and adaptive cell type under specific phagocytosis program that is not associated with a proinflam-
conditions rather than differing, that is, fixed subtypes within matory reaction, but rather with secretion of the antiinflam-
the microglia population. matory cytokines TGFβ and IL-10 (Ravichandran 2003). This
process highlights the fact that although microglial phago-
cytosis coincides with neuronal death, dying neurons signal
5 P H AG O C Y TO S I S BY AC T I VAT E D M I C RO G L I A
their demise and microglial cells act as responders, rather than
As the “cousins” of peripheral monocytes, microglia serve instigators of cell death. Notably, in contrast to peripheral ner-
as the brain’s intrinsic macrophages. In addition to the vous system (PNS) lesions, the assumption that synapses and
ever-more-recognized role of microglia-mediated phagocy- dendrites from injured CNS neurons are not automatically
tosis in the developing brain and homoeostatic maintenance retracted is substantiated by findings attained using the facial
of the adult CNS, phagocytic microglia play a pivotal part nerve axotomy model, in which it was demonstrated that
in CNS pathology and repair. Microglia engage in multiple microglia actively remove synapses from the perikaryon and
types of ingestion of extracellular substances, including classi- dendrites of injured neurons (Streit 2005). Migratory activity
cal phagocytosis, pinocytosis, and uptake of particles through positions microglia in close proximity to damaged cells, allow-
their processes. ing them to strip residual synapses and dendrites (Schiefer et al.
Phagocytosis describes a process in which a cell engulfs 1999). Similar results have been garnered from experimental
a solid object with its cytoplasm, forming a phagosome entorhinal cortex lesion models in which it was observed that
that is internalized via a specialized form of endocytosis. the signaling cascade initiated by injured neurons is important
The solid object is subsequently degraded in the lysosome for removal of denervated dendrites (Rappert et al. 2004). In
in either an oxygen-dependent or -independent fashion. response to neuronal necrosis, cell debris remains at the lesion
Oxygen-dependent degradation depends on the generation site, especially myelin. Also here, microglia take on the task
of ROS, wherein myeloperoxidase generates hypochlorite of clearing the remains. Finally, pinocytosis refers to uptake
that ultimately aids in the destruction of ingested bacteria. The of small molecules or fluid into endosomal vesicles that can
oxygen-independent system relies on the release of effector mol- fuse with the lysosome and lead to degradation of its contents.
ecules that are stored in cytosolic granules. Proteolytic enzymes, In contrast to phagocytosis, the process of pinocytosis is not
such as defensins and lysozymes as well as lactoferrin, which specific to the substance being transported.
sequesters iron and leads to unfavorable growth conditions, also Although microglia are the first line of defense in the
help to eliminate ingested bacteria (Flannagan et al. 2011). CNS, their phagocytic capacity relative to that of periph-
In contrast to instances of sublethal CNS injury, neuronal eral macrophages may be limited (Mosley and Cuzner 1996;
degeneration incites microglia to adopt a phagocytic pheno- Popovich et al. 1999), and newly recruited blood-borne mac-
type that represents an escalated form of activation not char- rophages within the injured or diseased brain may signifi-
acteristic of all reactive microglial cells. Phagocytic microglia cantly contribute to efficient clearance of cell debris (Stoll and
take on an ameboid morphology (which does not necessarily Jander 1999). Importantly, the removal of myelin debris by
mean that all ameboid microglia per definition are phagocytic) phagocytes is crucial for subsequent regeneration via axonal
characterized by shortened or absent processes, cytoplasmic outgrowth and remyelination, both of which have been shown
hypertrophy, and surface expression of macrophage mark- to be inhibited by the persistent presence of myelin molecules
ers such as CD68 (ED1) (Streit et al. 1999). The purpose of at the injury site (Kotter et al. 2005). This process was shown
microglia phagocytosis is to clear apoptotic neurons, necrotic to be more effective in young as compared with aged animals
tissue or microbes, and this process is finely attuned to suit the (Dubois-Dalcq et al. 2005; Shields et al. 1999), suggesting an
initiating trigger. age-related decline in macrophage/microglia function. Myelin
Infectious agents, in particular cell-wall components of degrading phagocytes have been extensively described in mul-
bacteria, activate microglia and stimulate phagocytosis via tiple sclerosis lesions (Bruck et al. 1995), but the functional
Toll-like receptor signaling. This promotes a proinflamma- impact of these cells on disease progression and pathogenesis
tory response characterized by secretion of IL-1, TNFα, and is still far from clear. The role of microglia in multiple sclerosis
NO production (Ravichandran 2003). This inflammatory is detailed in chapter 61.
reaction, in particular the production of ROS, is important Besides microbes, apoptotic cells and cellular debris, extra-
to kill and clear the infectious agent, but can also be harm- cellular aggregates of proteins are powerful activators of micro-
ful to the surrounding CNS tissue when exaggerated. Besides glia phagocytosis. In that respect, the relationship between the
direct degradation of invading microbes, antigens of phago- accumulation of Aβ in AD and associated phagocytic activity,
cytosed and degraded parasites are subsequently presented on or lack thereof, by microglia has been extensively studied and
the cell surface via MHC class II molecules to fuel activation is still a matter of vehement debate (see also chapter 64). There
of peripheral immune cells that further aid in clearance of the is abundant evidence that microglia are activated and recruited
CNS infection. to Aβ-plaques via a proinflammatory cascade. Moreover, is
Apoptotic neurons trigger phagocytosis by microglia on has been shown that microglia are capable of ingesting Aβ
exposure of internal cell wall components on the outer cell into their cytoplasm (Wisniewski et al. 1991). This finding
surface. Exposed entities of asialoglycoprotein, vitronectin has fueled numerous experimental studies indicating that
or phosphatidylserine on apoptotic cells bind to respective microglia-mediated clearance of Aβ is crucial for ameliora-
receptors on microglia (Witting et al. 2000) and initiate a tion of AD pathology (Tan et al. 2002; Town et al. 2008;

R O L E S O F AC T I VAT E D M I C R O G L I A • 633
Wyss-Coray et al. 2001). However, despite the fact that Aβ
can be demonstrated inside the cytoplasm of microglia, con-
clusive experimental proof of lysosomal degradation of Aβ by
microglia is lacking. In fact, in vitro studies show that although
Aβ is efficiently taken up by microglia, it is later found in the
medium again, indicating that microglia sample these extra-
cellular aggregates, but are apparently unable to degrade them,
resulting in re-release back into the extracellular space (Njie
et al. 2012). This activity may also contribute to the dynamic
nature of Aβ plaques. In accordance with these data, evidence
from analysis of post-mortem human AD tissue suggests that
microglia attempting to phagocytose Aβ may undergo a pro-
cess termed frustrated phagocytosis that could lead to produc-
tion and secretion of toxic molecules because the cell is not
capable of degrading the ingested agent (Streit 2004). Finally, 50 μm
chronic Aβ exposure has been postulated to induce premature
aging of microglia cells in a process called microglia senes- Figure 49.3 Alerted microglia at the border of microglia depleted cortical
cence, and it has been speculated that a subsequent partial loss area in ganciclovir treated CD11b-Herpes simplex virus thymidine kinase
of neuroprotective function may contribute to AD progres- mice (Iba-1 immunohistochemistry, brown).
sion. In support of this theory, data from experimental studies
reveal that conditional depletion of microglia from the brains
of AD transgenic mice does not affect the number of amy- are important for maintenance of the neuronal stem cell pool
loid plaques in the brain (Grathwohl et al. 2009). This find- in the hippocampus through apoptosis-mediated phagocyto-
ing does not necessarily mean that microglia are redundant in sis (Sierra et al. 2010). These physiological phagocytic activi-
AD pathogenesis, but rather suggests that microglia in the AD ties of microglia are primarily triggered by neuronal signaling
environment are, in fact, unable to efficiently clear Aβ. Thus, and are performed by nonactivated, ramified microglia and
removal of these cells from the system (at least for up to 1 associated with little or no inflammatory cytokine reaction.
month) does not result in the increase in plaques that might be
expected in the case of sudden removal of efficient mediators
of Aβ clearance, whereas functional parameters such as behav- 6 S U M M A RY A N D P E R S P E C T I VE S
ioral changes on microglia deficiency in the aforementioned
experimental AD setting have not been assessed. In addition Microglia are capable of mounting a rapid and robust response
to dying neurons, microglia have also been shown to engage to homeostatic disturbances in the CNS. Although basic prin-
in phagocytosis of other microglia cells (Falsig et al. 2008). ciples of microglia activation are stereotypic for all types of
A transgenic mouse model allowing for the conditional and insults, discriminating aspects of the reactive profile are highly
selective ablation of CD11b+ myeloid cells in the brain causes specialized and context dependent. Moreover, microgliosis
widespread death of microglia on pharmacological treatment, is a tightly regulated process balanced by incoming signals
reaching nearly 100%, depending on the treatment protocol from surrounding cells and through autocrine signaling. In
(Heppner et al. 2005). Interestingly, although some cell debris this regard, it is important to note that microglia activation
can be found throughout the microglia-free parenchyma, occurs as a response to signals from distressed CNS cells who
nearly all dead cells are cleared. Although this phenomenon themselves serve as instigators of reactivity. Therefore, it must
warrants further investigation, it is most likely that the cells do be assumed that in the absence of primary microglia dysfunc-
not all die at once, but are eliminated by a domino effect, leav- tion, reactive microgliosis detected in early disease stages, even
ing behind neighboring microglia to clear the remains of dead before signs of obvious pathology, likely occurs in response to
cells. In support of this hypothesis, highly “alerted” microglia signals from already afflicted neurons, and is not a primary
displaying an uncharacteristically large, bushy appearance can event in pathogenesis. In fact, the activation of microglia
be seen near the borders of microglia-depleted brain regions preceding other pathogenic signs may serve as a warning of
(Fig. 49.3). impending illness that could prove clinically valuable, and may
It is important to note that not all phagocytic activities also be useful for studying the region and cell-specific origin of
performed by microglia are carried out by activated cells under disease. On the other hand, new data are more and more sug-
conditions of duress. In the developing brain, microglia play a gestive of the fact that microglial function deteriorates with
critical role in removal of surplus neurons that have undergone advanced age, and this decline may be exacerbated by chronic
apoptosis via a caspase-3–mediated mechanism (Marin-Teva CNS pathology. More research is needed to understand the
et al. 2004). Furthermore, microglia are important for removal ways in which aging, repeated injury, such as head trauma, and
of synapses during development, a process also termed synap- chronic neurodegenerative disease affect microglia viability.
tic pruning, which involves the classical complement cascade In the event that microglia lose the ability to properly self-
(Stevens et al. 2007) and fractalkine signaling (Paolicelli et al. regulate their activational profile or respond to exogenous
2011). Last, it has recently been demonstrated that microglia regulatory signals, exaggerated and detrimental inflammatory

634 • NEUROGLIA
processes may ensue. In this way, dysfunctional microglia may Diab A, Abdalla H, Li HL, Shi FD, Zhu J, Hojberg B, Lindquist L,
also contribute to disease pathogenesis. An understanding of Wretlind B, Bakhiet M, Link H. 1999. Neutralization of macrophage
inflammatory protein 2 (MIP-2) and MIP-1alpha attenuates neutro-
the specific functional impediments exhibited by “senescent” phil recruitment in the central nervous system during experimental
microglia may provide clues regarding strategies to maintain bacterial meningitis. Infect Immun 67:2590–601.
or boost microglia health or substitute for lost CNS support. Draheim HJ, Prinz M, Weber JR, Weiser T, Kettenmann H, Hanisch
Technological advances, including live in vivo microscopy that UK. 1999. Induction of potassium channels in mouse brain microg-
allows for in situ visualization of behaving microglia, sophis- lia: cells acquire responsiveness to pneumococcal cell wall compo-
nents during late development. Neuroscience 89(4):1379–1390.
ticated transgenic mouse models that facilitate microglia- Dubois-Dalcq M, Ffrench-Constant C, Franklin RJ. 2005. Enhancing
specific manipulation, and ever-advancing high-throughput central nervous system remyelination in multiple sclerosis. Neuron
genetic and proteomic screening capabilities hold promise for 48(1):9–12.
an era of new discoveries in the field of microglia biology that Ebert S, Schoeberl T, Walczak Y, Stoecker K, Stempfl T, Moehle C, et al.
may facilitate therapeutic strategies aimed at regulating micro- 2008. Chondroitin sulfate disaccharide stimulates microglia to adopt
a novel regulatory phenotype. J Leukoc Biol 84(3):736–740.
glial responses to brain disease. Eichhoff G, Brawek B, Garaschuk O. 2011. Microglial calcium signal acts
as a rapid sensor of single neuron damage in vivo. Biochim Biophys
Acta 1813(5):1014–1024.
Falsig J, Julius C, Margalith I, Schwarz P, Heppner FL, Aguzzi A. 2008.
REFERENCES A versatile prion replication assay in organotypic brain slices. Nat
Neurosci 11(1):109–117.
Aaronson DS, Horvath CM. 2002. A road map for those who don’t Ferreira R, Santos T, Cortes L, Cochaud S, Agasse F, Silva AP, et al.
know JAK-STAT. Science 296(5573):1653–1655. 2012. Neuropeptide Y inhibits interleukin-1 beta-induced microglia
Alboni S, Cervia D, Sugama S, Conti B. 2010. Interleukin 18 in the motility. J Neurochem 120(1):93–105.
CNS. J Neuroinflammation 7:9. Filipovic R, Jakovcevski I, Zecevic N. 2003. GRO-alpha and CXCR2
Badie B, Schartner J, Vorpahl J, Preston K. 2000. Interferon-gamma in the human fetal brain and multiple sclerosis lesions. Dev Neurosci
induces apoptosis and augments the expression of Fas and Fas ligand 25:279–90.
by microglia in vitro. Exp Neurol 162(2):290–296. Flanary BE, Streit WJ. 2004. Progressive telomere shortening occurs in
Balasingam V, Dickson K, Brade A, Yong VW. 1996. Astrocyte reactiv- cultured rat microglia, but not astrocytes. Glia 45(1):75–88.
ity in neonatal mice: apparent dependence on the presence of reactive Flannagan RS, Jaumouille V, Grinstein S. 2012. The cell biology of phag-
microglia/macrophages. Glia 18(1):11–26. ocytosis. Annu Rev Pathol 7:61–98.
Baricordi OR, Melchiorri L, Adinolfi E, Falzoni S, Chiozzi P, Buell Fraga D, Raborn ES, Ferreira GA, Cabral GA. 2011. Cannabinoids
G, et al. 1999. Increased proliferation rate of lymphoid cells trans- inhibit migration of microglial-like cells to the HIV protein Tat.
fected with the P2X(7) ATP receptor. J Biol Chem 274(47): J Neuroimmune Pharmacol 6(4):566–577.
33206–33208. Friede RL, Johnstone MA. 1967. Responses of thymidine labeling of
Bartholdi D, Schwab ME. 1997. Expression of pro-inflammatory nuclei in gray matter and nerve following sciatic transection. Acta
cytokine and chemokine mRNA upon experimental spinal cord Neuropathol 7(3):218–231.
injury in mouse: an in situ hybridization study. Eur J Neurosci Ganter S, Northoff H, Mannel D, Gebicke-Harter PJ. 1992. Growth
9(7):1422–1438. control of cultured microglia. J Neurosci Res 33(2):218–230.
Bianchin MM, Martin KC, de Souza AC, de Oliveira MA, Rieder CR. Gehrmann J, Banati RB. 1995. Microglial turnover in the injured CNS:
2010. Nasu-Hakola disease and primary microglial dysfunction. Nat activated microglia undergo delayed DNA fragmentation following
Rev Neurol 6(9):2 p following 523. peripheral nerve injury. J Neuropathol Exp Neurol 54(5):680–688.
Bruce AJ, Boling W, Kindy MS, Peschon J, Kraemer PJ, Carpenter MK, Godiska R, Chantry D, Raport CJ, Sozzani S, Allavena P, Leviten D,
et al. 1996. Altered neuronal and microglial responses to excitotoxic Mantovani A, Gray PW. 1997. Human macrophage-derived che-
and ischemic brain injury in mice lacking TNF receptors. Nat Med mokine (MDC), a novel chemoattractant for monocytes, mono-
2(7):788–794. cyte-derived dendritic cells, and natural killer cells. J Exp Med
Bruck W, Porada P, Poser S, Rieckmann P, Hanefeld F, Kretzschmar 185:1595–604.
HA, et al. 1995. Monocyte/macrophage differentiation in early mul- Gomez-Nicola D, Valle-Argos B, Suardiaz M, Taylor JS, Nieto-Sampedro
tiple sclerosis lesions. Ann Neurol 38(5):788–796. M. 2008. Role of IL-15 in spinal cord and sciatic nerve after chronic
Cammermeyer J. 1965. Juxtavascular karyokinesis and microglia cell constriction injury: regulation of macrophage and T-cell infi ltration.
proliferation during retrograde reaction in the mouse facial nucleus. J Neurochem 107:1741–52.
Ergeb Anat Entwicklungsgesch 38:1–22. Grathwohl SA, Kalin RE, Bolmont T, Prokop S, Winkelmann G, Kaeser
Carbonell WS, Murase S, Horwitz AF, Mandell JW. 2005. Migration of SA, et al. 2009. Formation and maintenance of Alzheimer’s disease
perilesional microglia after focal brain injury and modulation by CC beta-amyloid plaques in the absence of microglia. Nat Neurosci
chemokine receptor 5: an in situ time-lapse confocal imaging study. J 12(11):1361–1363.
Neurosci 25(30):7040–7047. Groeger G, Quiney C, Cotter TG. 2009. Hydrogen peroxide as a
Columba-Cabezas S, Serafini B, Ambrosini E, Aloisi F. 2003. Lymphoid cell-survival signaling molecule. Antioxid Redox Signal 11(11):
chemokines CCL19 and CCL21 are expressed in the central nervous 2655–2671.
system during experimental autoimmune encephalomyelitis: impli- Hanisch UK. 2002. Microglia as a source and target of cytokines. Glia
cations for the maintenance of chronic neuroinflammation. Brain 40(2):140–155.
Pathol 13:38–51. Hanisch UK, Prinz M, Angstwurm K, Hausler KG, Kann O,
Cudaback E, Li X, Montine KS, Montine TJ, Keene CD. 2011. Kettenmann H, Weber JR. 2001. The protein tyrosine kinase inhib-
Apolipoprotein E isoform-dependent microglia migration. FASEB J itor AG126 prevents the massive microglial cytokine induction by
25(6):2082–2091. pneumococcal cell walls. Eur J Immunol 31:2104–15.
Dalton MM, Hommes OR, Leblond CP. 1968. Correlation of Harrison JK, Jiang Y, Chen S, Xia Y, Maciejewski D, McNamara RK,
glial proliferation with age in the mouse brain. J Comp Neurol et al. 1998. Role for neuronally derived fractalkine in mediating
134(4):397–400. interactions between neurons and CX3CR1-expressing microglia.
Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, et al. 2005. Proc Natl Acad Sci U S A 95(18):10896–10901.
ATP mediates rapid microglial response to local brain injury in vivo. Hartung HP. 1993. Immune-mediated demyelination. Ann Neurol
Nat Neurosci 8(6):752–758. 33(6):563–567.

R O L E S O F AC T I VAT E D M I C R O G L I A • 635
Heppner FL, Greter M, Marino D, Falsig J, Raivich G, Hovelmeyer N, Marin-Teva JL, Dusart I, Colin C, Gervais A, van Rooijen N, Mallat
et al. 2005. Experimental autoimmune encephalomyelitis repressed M. 2004. Microglia promote the death of developing Purkinje cells.
by microglial paralysis. Nat Med 11(2):146–152. Neuron 41(4):535–547.
Hinojosa AE, Garcia-Bueno B, Leza JC, Madrigal JL. 2011. CCL2/ McGeer EG, McGeer PL. 1997. The role of the immune system in neuro-
MCP-1 modulation of microglial activation and proliferation. degenerative disorders. Mov Disord 12(6):855–858.
J Neuroinflammation 8:77. Merriam-Webster Online Dictionary. 2012.
Honda S, Sasaki Y, Ohsawa K, Imai Y, Nakamura Y, Inoue K, et al. 2001. Merrill JE. 1991. Effects of interleukin-1 and tumor necrosis factor-alpha
Extracellular ATP or ADP induce chemotaxis of cultured microglia on astrocytes, microglia, oligodendrocytes, and glial precursors
through Gi/o-coupled P2Y receptors. J Neurosci 21(6):1975–1982. in vitro. Dev Neurosci 13(3):130–137.
Horvath RJ, DeLeo JA. 2009. Morphine enhances microglial migra- Miller KR, Streit WJ. 2007. The effects of aging, injury and disease on
tion through modulation of P2X4 receptor signaling. J Neurosci microglial function: a case for cellular senescence. Neuron Glia Biol
29(4):998–1005. 3(3):245–253.
Jack CS, Arbour N, Manusow J, Montgrain V, Blain M, McCrea E, Shapiro Mosley K, Cuzner ML. 1996. Receptor-mediated phagocytosis of myelin
A, Antel JP. 2005. TLR signaling tailors innate immune responses in by macrophages and microglia: effect of opsonization and receptor
human microglia and astrocytes. J Immunol 175:4320–30. blocking agents. Neurochem Res 21(4):481–487.
Janabi N, Hau I, Tardieu M. 1999. Negative feedback between prosta- Nicola NA. 1990. Granulocyte colony-stimulating factor. Immunol Ser
glandin and alpha- and beta-chemokine synthesis in human microg- 49:77–109.
lial cells and astrocytes. J Immunol 162:1701–6. Nimmerjahn A, Kirchhoff F, Helmchen F. 2005. Resting microglial cells
Jekabsone A, Mander PK, Tickler A, Sharpe M, Brown GC. 2006. are highly dynamic surveillants of brain parenchyma in vivo. Science
Fibrillar beta-amyloid peptide Abeta1–40 activates microglial prolif- 308(5726):1314–1318.
eration via stimulating TNF-alpha release and H2O2 derived from Njie EG, Boelen E, Stassen FR, Steinbusch HW, Borchelt DR, Streit WJ.
NADPH oxidase: a cell culture study. J Neuroinflammation 3:24. 2012. Ex vivo cultures of microglia from young and aged rodent brain
Jimenez-Sainz MC, Fast B, Mayor F Jr, Aragay AM. 2003. Signaling path- reveal age-related changes in microglial function. Neurobiol Aging
ways for monocyte chemoattractant protein 1-mediated extracellular 33(1):195 e1–12.
signal-regulated kinase activation. Mol Pharmacol 64(3):773–782. Nolte C, Moller T, Walter T, Kettenmann H. 1996. Complement 5a
Jones LL, Banati RB, Graeber MB, Bonfanti L, Raivich G, Kreutzberg controls motility of murine microglial cells in vitro via activation of
GW. 1997. Population control of microglia: does apoptosis play a an inhibitory G-protein and the rearrangement of the actin cytoskel-
role? J Neurocytol 26(11):755–770. eton. Neuroscience 73(4):1091–1107.
Karlstetter M, Lippe E, Walczak Y, Moehle C, Aslanidis A, Mirza M, Nosaka T, Kawashima T, Misawa K, Ikuta K, Mui AL, Kitamura T. 1999.
et al. 2011. Curcumin is a potent modulator of microglial gene expres- STAT5 as a molecular regulator of proliferation, differentiation and
sion and migration. J Neuroinflammation 8:125. apoptosis in hematopoietic cells. EMBO J 18(17):4754–4765.
Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. 2011. Physiology Oshimi Y, Miyazaki S, Oda S. 1999. ATP-induced Ca2+ response medi-
of microglia. Physiol Rev 91:461–553. ated by P2U and P2Y purinoceptors in human macrophages: signal-
Kiefer R, Lindholm D, Kreutzberg GW. 1993. Interleukin-6 and trans- ling from dying cells to macrophages. Immunology 98(2):220–227.
forming growth factor-beta 1 mRNAs are induced in rat facial nucleus Paolicelli RC, Bolasco G, Pagani F, Maggi L, Scianni M, Panzanelli P,
following motoneuron axotomy. Eur J Neurosci 5(7):775–781. et al. 2011. Synaptic pruning by microglia is necessary for normal
Kiefer R, Streit WJ, Toyka KV, Kreutzberg GW, Hartung HP. 1995. brain development. Science 333(6048):1456–1458.
Transforming growth factor-beta 1: a lesion-associated cytokine of Ponomarev ED, Shriver LP, Maresz K, Pedras-Vasconcelos J, Verthelyi D,
the nervous system. Int J Dev Neurosci 13(3–4):331–339. Dittel BN. 2007. GM-CSF production by autoreactive T cells is
Kloss CU, Kreutzberg GW, Raivich G. 1997. Proliferation of ramified required for the activation of microglial cells and the onset of experi-
microglia on an astrocyte monolayer: characterization of stimulatory mental autoimmune encephalomyelitis. J Immunol 178(1):39–48.
and inhibitory cytokines. J Neurosci Res 49(2):248–254. Popovich PG, Guan Z, Wei P, Huitinga I, van Rooijen N, Stokes BT.
Kotter MR, Zhao C, van Rooijen N, Franklin RJ. 2005. 1999. Depletion of hematogenous macrophages promotes partial
Macrophage-depletion induced impairment of experimental CNS hindlimb recovery and neuroanatomical repair after experimental
remyelination is associated with a reduced oligodendrocyte progeni- spinal cord injury. Exp Neurol 158(2):351–365.
tor cell response and altered growth factor expression. Neurobiol Dis Raivich G, Bohatschek M, Kloss CU, Werner A, Jones LL, Kreutzberg
18(1):166–175. GW. 1999. Neuroglial activation repertoire in the injured brain:
Kraft AD, McPherson CA, Harry GJ. 2009. Heterogeneity of microglia graded response, molecular mechanisms and cues to physiological
and TNF signaling as determinants for neuronal death or survival. function. Brain Res Brain Res Rev 30(1):77–105.
Neurotoxicology 30(5):785–793. Raivich G, Moreno-Flores MT, Moller JC, Kreutzberg GW. 1994.
Kreutzberg GW. 1966. Autoradiographische Untersuchung uber die Inhibition of posttraumatic microglial proliferation in a genetic
Beteiligung von Gliazellen an der axonalen Reaktion im Facialiskern model of macrophage colony-stimulating factor deficiency in the
der Ratte. Acta Neuropathol (Berl) 7:149–161. mouse. Eur J Neurosci 6(10):1615–1618.
Kreutzberg GW, Hager H. 1966. Electron microscopical demonstra- Rappert A, Bechmann I, Pivneva T, Mahlo J, Biber K, Nolte C, et al.
tion of acid phosphatase activity in the central nervous system. 2004. CXCR3-dependent microglial recruitment is essential for
Histochemie 6(3):254–259. dendrite loss after brain lesion. J Neurosci 24(39):8500–8509.
Ladeby R, Wirenfeldt M, Dalmau I, Gregersen R, Garcia-Ovejero D, Ravichandran KS. 2003. “Recruitment signals” from apoptotic cells:
Babcock A, et al. 2005. Proliferating resident microglia express the invitation to a quiet meal. Cell 113(7):817–820.
stem cell antigen CD34 in response to acute neural injury. Glia Rothwell NJ, Luheshi G, Toulmond S. 1996. Cytokines and their recep-
50(2):121–131. tors in the central nervous system: physiology, pharmacology, and
Lee TT, Martin FC, Merrill JE. 1993. Lymphokine induction of rat pathology. Pharmacol Ther 69(2):85–95.
microglia multinucleated giant cell formation. Glia 8:51–61. Satoh J, Tabunoki H, Yamamura T, Arima K, Konno H. 2007. TROY
Liva SM, Kahn MA, Dopp JM, de Vellis J. 1999. Signal transduction and LINGO-1 expression in astrocytes and macrophages/micro-
pathways induced by GM-CSF in microglia: significance in the con- glia in multiple sclerosis lesions. Neuropathol Appl Neurobiol
trol of proliferation. Glia 26(4):344–352. 33(1):99–107.
Mander PK, Jekabsone A, Brown GC. 2006. Microglia proliferation is Schiefer J, Kampe K, Dodt HU, Zieglgansberger W, Kreutzberg GW.
regulated by hydrogen peroxide from NADPH oxidase. J Immunol 1999. Microglial motility in the rat facial nucleus following periph-
176(2):1046–1052. eral axotomy. J Neurocytol 28(6):439–453.

636 • NEUROGLIA
Seilhean D, Kobayashi K, He Y, Uchihara T, Rosenblum O, Katlama C, cortex, and olfactory bulb of young adult macaque monkeys. Glia
et al. 1997. Tumor necrosis factor-alpha, microglia and astrocytes in 42(3):209–224.
AIDS dementia complex. Acta Neuropathol 93(5):508–517. Town T, Laouar Y, Pittenger C, Mori T, Szekely CA, Tan J, et al. 2008.
Shankaran M, Marino ME, Busch R, Keim C, King C, Lee J, et al. Blocking TGF-beta-Smad2/3 innate immune signaling mitigates
2007. Measurement of brain microglial proliferation rates in vivo in Alzheimer-like pathology. Nat Med 14(6):681–687.
response to neuroinflammatory stimuli: application to drug discov- Taoufi k Y, de Goer de Herve MG, Giron-Michel J, Durali D, Cazes E,
ery. J Neurosci Res 85(11):2374–2384. Tardieu M, Azzarone B, Delfraissy JF. 2001. Human microglial cells
Shields SA, Gilson JM, Blakemore WF, Franklin RJ. 1999. Remyelination express a functional IL-12 receptor and produce IL-12 following
occurs as extensively but more slowly in old rats compared to IL-12 stimulation. Eur J Immunol 31:3228–39.
young rats following gliotoxin-induced CNS demyelination. Glia Town T, Nikolic V, Tan J. 2005. The microglial “activation” continuum:
28(1):77–83. from innate to adaptive responses. J Neuroinflammation 2:24.
Sierra A, Encinas JM, Deudero JJ, Chancey JH, Enikolopov G, Trapp BD, Wujek JR, Criste GA, Jalabi W, Yin X, Kidd GJ, et al.
Overstreet-Wadiche LS, et al. 2010. Microglia shape adult hippocam- 2007. Evidence for synaptic stripping by cortical microglia. Glia
pal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem 55(4):360–368.
Cell 7(4):483–495. Wang J, Li PT, Du H, Hou JC, Li WH, Pan YS, et al. 2011. Impact of
Simi A, Tsakiri N, Wang P, Rothwell NJ. 2007. Interleukin-1 and inflam- paracrine signals from brain microvascular endothelial cells on micro-
matory neurodegeneration. Biochem Soc Trans 35:1122–6. glial proliferation and migration. Brain Res Bull 86(1–2):53–59.
Sjostrand J. 1971. Neuroglial proliferation in the hypoglossal nucleus Wang X, Suzuki Y. 2007. Microglia produce IFN-gamma independently
after nerve injury. Exp Neurol 30(1):178–189. from T cells during acute toxoplasmosis in the brain. J Interferon
Skaper SD, Debetto P, Giusti P. 2010. The P2X7 purinergic receptor: Cytokine Res 27(7):599–605.
from physiology to neurological disorders. FASEB J 24(2):337–345. Wisniewski HM, Barcikowska M, Kida E. 1991. Phagocytosis of beta/A4
Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, amyloid fibrils of the neuritic neocortical plaques. Acta Neuropathol
Nouri N, et al. 2007. The classical complement cascade mediates 81(5):588–590.
CNS synapse elimination. Cell 131(6):1164–1178. WittingA,MullerP,HerrmannA,KettenmannH,NolteC.2000.Phagocytic
Stoll G, Jander S. 1999. The role of microglia and macrophages in the clearance of apoptotic neurons by Microglia/Brain macrophages in
pathophysiology of the CNS. Prog Neurobiol 58(3):233–247. vitro: involvement of lectin-, integrin-, and phosphatidylserine-mediated
Streit WJ. 2004. Microglia and Alzheimer’s disease pathogenesis. recognition. J Neurochem 75(3):1060–1070.
J Neurosci Res 77(1):1–8. Wyss-Coray T. 2006. Inflammation in Alzheimer disease: driving force,
Streit WJ. 2005. Microglia and neuroprotection: implications for bystander or beneficial response? Nat Med 12(9):1005–1015.
Alzheimer’s disease. Brain Res Brain Res Rev 48(2):234–239. Wyss-Coray T, Lin C, Yan F, Yu GQ, Rohde M, McConlogue L, et al.
Streit WJ, Hurley SD, McGraw TS, Semple-Rowland SL. 2000. 2001. TGF-beta1 promotes microglial amyloid-beta clearance and
Comparative evaluation of cytokine profi les and reactive gliosis sup- reduces plaque burden in transgenic mice. Nat Med 7(5):612–618.
ports a critical role for interleukin-6 in neuron-glia signaling during Xia MQ, Hyman BT. 1999. Chemokines/chemokine receptors in the cen-
regeneration. J Neurosci Res 61(1):10–20. tral nervous system and Alzheimer’s disease. J Neurovirol 5:32–41.
Streit WJ, Kreutzberg GW. 1988. Response of endogenous glial cells to Yan J, Zhou X, Guo JJ, Mao L, Wang YJ, Sun J, Sun LX, Zhang LY,
motor neuron degeneration induced by toxic ricin. J Comp Neurol Zhou XF, Liao H. 2012. Nogo-66 inhibits adhesion and migra-
268(2):248–263. tion of microglia via GTPase Rho pathway in vitro. J Neurochem
Streit WJ, Semple-Rowland SL, Hurley SD, Miller RC, Popovich PG, 120:721–31.
Stokes BT. 1998. Cytokine mRNA profi les in contused spinal cord Yang GY, Gong C, Qin Z, Ye W, Mao Y, Bertz AL. 1998. Inhibition
and axotomized facial nucleus suggest a beneficial role for inflamma- of TNFalpha attenuates infarct volume and ICAM-1 expression in
tion and gliosis. Exp Neurol 152(1):74–87. ischemic mouse brain. Neuroreport 9(9):2131–2134.
Streit WJ, Walter SA, Pennell NA. 1999. Reactive microgliosis. Prog Yang L, Jones NR, Blumbergs PC, Van Den Heuvel C, Moore EJ,
Neurobiol 57(6):563–581. Manavis J, et al. 2005. Severity-dependent expression of
Suzuki Y, Claflin J, Wang X, Lengi A, Kikuchi T. 2005. Microglia pro-inflammatory cytokines in traumatic spinal cord injury in the
and macrophages as innate producers of interferon-gamma in the rat. J Clin Neurosci 12(3):276–284.
brain following infection with Toxoplasma gondii. Int J Parasitol Zujovic V, Benavides J, Vige X, Carter C, Taupin V. 2000. Fractalkine
35(1):83–90. modulates TNF-alpha secretion and neurotoxicity induced by micro-
Tan J, Town T, Crawford F, Mori T, DelleDonne A, Crescentini R, et al. glial activation. Glia 29(4):305–315.
2002. Role of CD40 ligand in amyloidosis in transgenic Alzheimer’s Zujovic V, Luo D, Baker HV, Lopez MC, Miller KR, Streit WJ, Harrison
mice. Nat Neurosci 5(12):1288–1293. JK. 2005. The facial motor nucleus transcriptional program in response
Tonchev AB, Yamashima T, Zhao L, Okano H. 2003. Differential to peripheral nerve injury identifies Hn1 as a regeneration-associated
proliferative response in the postischemic hippocampus, temporal gene. J Neurosci Res 82:581–91.

R O L E S O F AC T I VAT E D M I C R O G L I A • 637
50.
IMMUNE FUNCTIONS OF MICROGLIA
Trevor Owens

A B B R E VI AT I O N S responses are induced. However, subsequent effector response


must occur in tissues, wherever pathogens are found.
APC antigen-presenting cell
BBB blood-brain barrier
1.2 M A J O R H I S TO C O M PAT I B I L IT Y C O M P L E X I
CNS central nervous system
A N D C D8+ T C E L L S
DAMP danger/damage-associated molecule pattern
DC dendritic cell The antiviral major histocompatibility complex (MHC
EAE experimental autoimmune encephalomyelitis I)–restricted effector CD8+ T cell response is necessarily less
GFP green fluorescent protein dependent on professional APC, so that virus-infected cells
IFN interferon can be susceptible to immune attack, whatever their cell type
IGF1 insulin-like growth factor-1 or wherever located. Therefore, it is consistent that MHC I is
IL18bp IL18 binding protein widely expressed, essentially by all nucleated cells of the body,
LPS lipopolysaccharide and this includes all cells within the CNS. (The preceding
MS multiple sclerosis statements represent “general knowledge” and are difficult to
MyD88 myeloid differentiation primary response precisely reference. Entry points to an extensive literature can
gene-88 be found in Goverman, 2009).
NFkB nuclear factor kappa B
PAMP pathogen-associated molecular pattern
1.3 M A J O R H I S TO C O M PAT I B I L IT Y C O M P L E X I I
PD-1 programmed cell death-1
A N D C D4 + T C E L L S
PRR pattern recognition receptor
RIG-I retinoic acid inducing gene-1 Requirements for response of CD4+ T cells are more com-
RLH RIG-I–like helicase plex. These T cells are required for cellular immunity such
TGFE transforming growth factor-beta as directed against intracellular bacteria and the induction
TLR Toll-like receptor of inflammatory responses, as well as being required as help-
TNFD tumor necrosis factor-alpha ers for antibody responses. CD4+ T cells recognize antigenic
TRAF TNF receptor–associated factor; peptides in association with MHC II and this is not widely
TRIF Toll/IL1R-containing adapter inducing expressed. Hence, there is great interest in cells within tis-
IFNE sues that can express MHC II and whether they play a role in
directing immune responses in those tissues.

1 INTRODUCTION
1.4 C O S T I MU L AT I O N
Although essential for presentation of antigen to CD4+
1.1 A N T I G E N P R E S E N TAT I O N:
T cells, expression of MHC II is not the only requirement for a
GENERAL CONCEPTS
cell to function as an APC. Additional signals from costimula-
The concept of antigen presentation is a cornerstone of immu- tory ligands on APCs are required for optimal T-cell response
nology. T lymphocytes are, with B lymphocytes, the only two (Goverman 2009). It should be considered here whether
cell types that express clonally restricted receptors encoded the immune response being induced is primary or secondary.
by rearranged genes. Unlike the B cell receptor or cell surface The costimulatory requirements for induction of a primary
immunoglobulin, which can directly recognize conforma- T-cell response are stricter than for a secondary response. It
tional epitopes of antigenic moieties, the majority of T-cell makes sense that activated antiviral CD8+ T cells should be
receptors (i.e., alpha-beta receptors) are restricted to recogni- able to exert their effector function without too many con-
tion of peptides that are presented by association with MHC straints, for optimal protection against viruses. In the same
molecules. Immune dogma holds that antigen is primarily pre- way, effector CD4+ T-cell responses in tissues should be less
sented to T cells by so-called professional antigen presenting restrained than primary responses, because this enables pro-
cells (APC), within lymphoid tissue, which is where immune tection against pathogens and has obvious advantage for the

638
host. Naïve T-cell responses that may be induced in the con- therefore are candidate APCs for T-cell responses in the CNS
text of established inflammation, outside lymphoid tissue, (Hanisch and Kettenmann 2007). Consequently, we need to
would equally be expected to be costimulation-dependent. understand the role of microglia as antigen presenting cells in
Nevertheless, even for a secondary effector response, MHC the CNS. It is important to consider the circumstances under
II expression per se is not sufficient and costimulation is which they would mediate such a role and their status relative
required. to non-glial APCs.
Not only does recognition of MHC II+peptide per se
not necessarily lead to a productive T-cell response, but in
2.2 OT H E R C E N T R A L N E RVO US S Y S T E M–
the absence of any other signal, MHC II+peptide alone can
R E S I D E N T A N T I G E N-P R E S E N T I N G C E L L S
induce T-cell anergy, or antigen-specific nonresponsiveness.
Simultaneous engagement of costimulator receptor-ligand Are there other cells within the CNS with potential to pres-
pairs delivers signals that result in a proliferative and cytokine ent antigen to T cells? Astrocytes were shown to be capable
effector T-cell response. The paradigmatic costimulator mol- of presenting antigen for secondary T-cell activation in vitro,
ecules that are upregulated on microglia are B7.1/CD80 and especially after activation by inflammatory cytokines (Fontana
B7.2/CD86 (Goverman 2009). Costimulatory signals are et al. 1984), but subsequent studies show that they cannot pres-
delivered by these molecules via CD28 on the T cell. CD40 ent antigen for primary T-cell response (Williams et al. 1995),
is a cell surface receptor expressed by B cells and myeloid cells they do not normally express costimulator ligands, even in vitro
(including microglia) that is implicated in costimulation of (Williams et al. 1994), and their APC capability is strongly
T-cell responses, through engagement of CD40L/CD154 on influenced by co-cultured microglia, which must be taken into
activated CD4+ T cells. Opposing signals via CTLA4 or via consideration in any assay for astrocyte function (Holm et al.
the programmed cell death-1 (PD-1)/PD-L1 pathway keep 2012; Saura 2007; Williams et al. 1995). Astrocytes are not
T-cell responses in check (Goverman 2009). considered effective CNS APC for inflammatory responses,
although it has been reported that they might promote antiin-
flammatory Th2 cells (Aloisi et al. 1999).
2 A N T I G E N P R E S E N TAT I O N I N T H E
C E N T R A L N E RVO U S SYS T E M
2.3 D E N D R IT I C C E L L S
The central nervous system (CNS), like other organs of the The most potent antigen-presenting cell type in the body is
body, is protected from infection and tumor development by the dendritic cell (DC) (Goverman 2009). Myeloid DCs are
the immune system. Unlike most other organs, there is a lack implicated in induction of inflammatory immune responses
of lymphatic drainage from the CNS and transit of molecules such as in experimental autoimmune encephalomyelitis
from blood to the local circulation (cerebrospinal fluid) is (EAE), whereas plasmacytoid DC, that do not express myel-
tightly restricted by the blood-brain barrier (BBB) (Owens oid markers, are associated with Type I interferon (IFN) pro-
et al. 2008). These differences, although important, are often duction and immunoregulation (Goverman 2009). One of
overemphasized, because although immune cellular traffic the enduring paradigms in immunology is that of Langerhans
across the BBB is dependent on specific and well-regulated cells in skin, which after encounter with antigen migrate to
molecular interactions, it nevertheless occurs. This is evidenced lymphoid tissue, where they become DC and present anti-
by the deleterious consequences of its blockade as well as by gen to T cells. The debate whether a similar concept of APC
diseases such as multiple sclerosis, in which immune infiltra- migration applies to induction of immune responses against
tion is a pathological characteristic (Engelhardt and Ransohoff CNS antigens has been given some impetus by a study show-
2005). Therefore, there is a need for immune-competent cells ing that monocytes can migrate from CNS to deep cervical
in the CNS that can interact with, support, and regulate lymph nodes (Kaminski et al. 2012), which have been impli-
blood-derived immune responses. cated in CNS inflammation (van Zwam et al. 2009). It was
not demonstrated whether CNS-derived monocytes became
DC in lymph nodes.
2.1 M I C RO G L I A A S C E N T R A L N E RVO US SYS T E M
Dendritic cells are defined by their morphology and a pro-
A N T I G E N-P R E S E N T I N G C E L L S
file of surface markers, no one of which is sufficient for their
The requirements for a cell to function as an APC for a CD4+ identification. These include high levels of CD11c, MHC II,
T-cell response broadly include the ability to take up anti- and costimulator ligands, as well as absence of markers for other
gen, process it into peptides that can associate with MHC II, lineages. Expression of MHC II and CD103/integrin-alphaE
express MHC II on the cell surface, and express costimula- (OX62) was used to distinguish populations of rat DC in dura
tory ligands and mediators. These considerations direct atten- mater, leptomeninges, and choroid plexus from parenchymal
tion to the molecular requirements for antigen presentation microglia and tissue macrophages, and this and other studies
and whether microglia fulfill them. Microglia are implicated demonstrated that DC are not normally found in the CNS
in phagocytosis, inflammation, and cytotoxicity in the CNS parenchyma (Greter et al. 2005; McMenamin 1999; Prodinger
(Hanisch and Kettenmann 2007), and express all of the capa- et al. 2011). It has been proposed that DC in perivascular/sub-
bilities required for antigen presentation. Microglia are the arachnoid spaces act as gatekeepers for presentation of anti-
only glial cells that normally express MHC II at any level, gen to infiltrating T cells (Becher et al. 2006; Prodinger et al.

I M MU N E F U N C T I O N S O F M I C R O G L I A • 639
2011). Once the CNS becomes inflamed as in multiple sclero- Flow cytometric discrimination of microglia from
sis (MS) or EAE, then DC may infiltrate the parenchyma, as macrophages and DC in CNS
do activated macrophages. Macrophages, DC,
Activated macrophages are also potent APC, and it has T cells granulocytes
been reported that macrophages and DC are much more effec-
tive APC, for secondary T-cell response, than microglia (Ford Microglia
et al. 1995). However, comparison to splenic or thymic APC

CD45
as reference controls may set the bar too high for identification
of functional APC in the CNS. As discussed in the following, Non-bone marrow
there are reasons to suspect that CNS DC may be less potent derived cells
APC than splenic DC, so that microglia may be equally effec-
tive to the CNS-resident DC that are normally found within
the CNS. Nevertheless, the case for microglia as APC must
always be considered in light of the fact that other APC are
available either within the CNS or immediately adjacent. The
CDIIb
physiological role of microglial APC may reflect their loca-
tion rather than their potency. Figure 50.1 Flow Cytometric Discrimination of Microglia from Central
Nervous System–Infiltrating Myeloid Cells. The schematic illustrates
the principle whereby microglia and blood-derived myeloid cells, all of
3 I S O L AT I O N A N D A S S AY O F M I C R O G L I A which express CD11b/Mac1, can be distinguished by the relatively low
level of expression of CD45 by microglia, using flow cytometry. The red
population oval defines CD45dimCD11b+ microglia, which are drawn to
Apparently simple questions such as whether a given cell type not overlap with the CD45highCD11b+ macrophages DC and granulo-
can present antigen are complicated by the experimental dif- cytes. Microglia can increase their expression of CD45 when activated,
ficulty of isolating cells in an unaltered state from the tissue. but there is general agreement that this does not move them into the
Many studies have relied on in vitro culture for eventual purifi- CD45high category. Even if it did, the CD45dim population remains dis-
tinct from blood-derived cells, which are always CD45high. Non-myeloid
cation of glial cells, and this introduces possibilities for assay of CNS-infiltrating cells, which include T cells, do not express CD11b and
aberrant activation states, which in many cases may not reflect are identifiable using lineage-specific markers.
even activation states in vivo. Furthermore, the APC capabil-
ity of a given cell type may include contribution of cocultured 3.2 C O M P L E M E N T R EC E P TO R S
contaminant cells, as has been mentioned for astrocytes and
microglia (Holm et al. 2012; Saura 2007). Likewise, cocul- Additional receptors that are used in analysis and isolation of
tured DC may need to be considered in interpretation of in antigen-presenting microglia are the complement receptors.
vitro assays of microglial function. There are two prominent integrin beta2 family D/E heterodi-
meric receptors for complement that define myeloid cells, so
including microglia, as well as antigen-presenting function.
3.1 C D45 L EVE L S A S A M A R K E R F O R M I C RO G L I A
These are the complement receptor-3 or Mac-1/CD11b, and
Microglia, as cells of the myeloid lineage, are phenotypically complement receptor-4 or CD11c. They share expression of
similar to macrophages, and they are usually distinguished the beta2/CD18 chain, and are defined by their alpha chain
in situ by morphology and location, but this is not useful for expression (alphaM/ITGAM for CD11b, versus alphaX/
functional assays. Seminal studies by Sedgwick and Hickey ITGAX for CD11c).
led to understanding that parenchymal microglia can be dis-
tinguished from other myeloid lineage cells in CNS by their 3.3 C D11B
relatively low expression of CD45 (Ford et al. 1995; Sedgwick
et al. 1991). This allows them to be analyzed separately from CD11b is routinely used as a marker for myeloid cells in mice.
blood-derived cells, and to be isolated using flow cytometric CD11b-deficient mice showed delayed onset and attenuated
methods (see Fig. 50.1). This approach is most commonly EAE (Bullard et al. 2005). Transfer experiments reveal a role
used for rodent microglia given difficulties of accessing nor- for CD11b in T-cell priming, generating altered cytokine pro-
mal human tissue in sufficient quantities for isolation proce- files and attenuated ability to transfer EAE, and as expected
dures. Reduced CD45 expression (CD45low or CD45dim) is a role for host–resident myeloid cells in transfer recipients—
an experimentally useful characteristic that has been exploited these would include microglia.
in numerous studies (Babcock et al. 2006). Myeloid cells in
perivascular space, meninges, or subarachnoid space express
3.4 C D11C
CD45 at levels similar to those in blood (CD45high).
Notwithstanding persistent residual concerns that elevated Complement receptor-4 CD11c is upregulated by microglia
levels of CD45 on activated microglia may obscure distinc- under similar circumstances to B7 molecules and is implicated
tions, there is consensus that the CD45dim or CD45low cells in their antigen-presenting capability. CD11c is composed of a
in CNS isolates do not include any astrocytes, macrophages heterodimer of CD18 and CD11c. This molecule is implicated
or DC. in phagocytosis and is associated with antigen presentation.

640 • NEUROGLIA
CD11c is frequently used as a “shorthand” marker for DC, T cells after culture with cerebral endothelial cells (Bai et al.
although always with recognition that it is per se insufficient 2009). Central nervous system–resident dendritic cells (DC,
for their identification in absence of other DC markers. Mice CD45highCD11c+) may not be as effective as APC as splenic
that lack CD11c are resistant to EAE, although systemic defi- DC (Løbner and Owens, unpublished). The finding that a
ciency also affects the peripheral immune response (Bullard subset of CD11b+ CD11c+ CD45dim microglia were equivalent
et al. 2007). Adoptive transfer of wild-type encephalitogenic as APC to DC (Remington et al. 2007) in fact involved com-
T cells to CD11c-deficient mice resulted in markedly attenu- parison to splenic DC, so if CNS DC should be modulated by
ated EAE, indicating that a non–T-cell source of CD11c is the local milieu, then this would if anything be an understated
also critical. CD11c-deficient T cells were also compromised finding.
as inducers of EAE, showing altered cytokine profiles, reflect-
ing the impact of CD11c deficiency in priming; therefore,
inevitably, identification of the CD11c-expressing cell(s) that 5 A N T I G E N U P TA K E BY M I C R O G L I A
are critical for EAE is going to be complex (Bullard et al.
2007). For example, lack of CD11c+ DC has also been shown This occurs by phagocytosis or by pinocytocis. Both intro-
to attenuate EAE (Greter et al. 2005). duce cell-extrinsic proteins to the lysosomal compartment,
where subsequent fusion in proteolysomes generates peptides
that associate to MHC II (Goverman 2009). These pathways
4 D I R E C T D E M O N S T R AT I O N O F intersect with those involved in autophagy, although the pre-
M I C R O G L I A L A N T I G E N -P R E S E N T I N G cise role of cell-intrinsic autophagic mechanisms to presenta-
CELL FUNCTION tion of cell-extrinsic molecules remains unclear (Mizushima
and Komatsu 2011). Human and rodent microglia have
The first unequivocal assay of APC function by directly isolated been shown to be capable of phagocytosing cell debris, and
ex vivo microglia used flow cytometry to isolate them from rat infection- and degeneration-related material, in vitro and
CNS, using CD11b/c+ CD45dim as markers (Ford et al. 1995). in vivo (Bechmann and Nitsch 1997; Kielian et al. 2002;
Their ability to present antigen to myelin basic protein-specific Koenigsknecht and Landreth 2004; Ulvestad et al. 1994),
T cells for a secondary response was compared with that of albeit with some selectivity (Hughes et al. 2010). An alternate
CD11b/c+ CD45high macrophages/DC from CNS, using mechanism for antigen uptake involves incorporation of exo-
thymic APC as a reference control. Microglia induced 20-fold somes released by oligodendrocytes (Fitzner et al. 2011).
increases in thymidine incorporation by T cells, equivalent to What is clear is that microglia have the ability to ingest
the fold increases induced by CD45high cells isolated from the particulate matter from their milieu and generate from this
same CNS. However, the same microglia were relatively poor peptides that can associate with MHC II on the microglial cell
inducers of interleukin (IL) production (Ford et al. 1995). The surface. As direct demonstration that this normally occurs,
conclusion drawn by the authors was that microglia were infe- isolated microglia from CNS without any addition of antigen
rior as APC to macrophages/DC in the CNS, and it remains a could act as APCs in vitro for activation of myelin-specific T
commonly stated opinion today that despite the fact that micro- cells (Ford et al. 1995). The fact that the responses induced
glia are the only MHC II-expressing cells resident within the were suboptimal compared with those induced by APC that
CNS, their ability to present antigen to T cells is questionable had been antigen-loaded emphasizes the need for further
(Ford et al. 1995; Hanisch and Kettenmann 2007). Alternative development of microglial APC capability. Part of this may
interpretations of those data are possible. involve MHC II induction.
In a complementary study, Aloisi and colleagues (1999) iso-
lated microglia from neonatal mice and compared their ability
5.1 A N T I G E N P RO C E S S I N G/P ROT E A S O M E
to act as APC with that of lymph node DC, for the activation of
naïve and secondary Th1- or Th2-biased T-cell response. The key An important component of a cell’s capacity for antigen pre-
finding with relevance to this chapter was that IFNJ-activated sentation is the generation of peptides that can associate with
microglia were as effective as DC in induction of naïve or Th1 MHC. The 20S proteasome is a multicatalytic complex of
CD4+ T cells. Given that IFNJ is itself largely produced by Th1 protease subunits that achieves this goal in APCs. Microglia
T cells this of course represents a chicken-versus-egg argument have been shown to selectively substitute immunoproteasome
for any role for microglia in the initiation of a T cell response in for constitutive subunits in response to the bacterial Toll-like
the CNS, but nevertheless reinforces the core findings of Ford receptor-4 (TLR4)-ligand lipopolysaccharide (LPS) as well as
et al. (1995) that microglia are effective APC under appropriate in response to (IFNJ) (Stohwasser et al. 2000). Thus activated
circumstances (Aloisi et al. 1999). microglia adapt their proteasome structure toward an MHC I
antigen-processing capability. IFNJ has separately been shown
to induce both mRNA for MHC I and antigen processing com-
4.1 MO D U L AT I O N O F A N T I G E N-P R E S E N T I N G
ponents, with rapid kinetics, in mouse microglia. Importantly,
C E L L F U N C T I O N BY T H E C E N T R A L N E RVO US
these mRNAs were expressed at relatively high levels in naïve
SYSTEM ENVIRONMENT
unstimulated microglia, and then upregulated in response to
The CNS environment may also influence function of APC. coronoavirus infection, suggesting “innate immune prepared-
DC were shown to become microglia-like and inhibit CD4 ness” (Malone et al. 2008).

I M MU N E F U N C T I O N S O F M I C R O G L I A • 641
5.2 M A J O R H I S TO C O M PAT I B I L I T Y C O M P L E X I I are numerous other innate receptor families, the concept of
E X P R E S S I O N A N D P E P T I D E A S S O C I AT I O N innate signaling pathways that induce costimulation and anti-
gen presentation is exemplified by TLRs, and for the sake of
Although microglia, as members of the myeloid cell lineage,
this discussion it is assumed that similar principles apply to
can express MHC II, this is not always detectable in situ with
other receptors. The concept is important because it links
conventional staining techniques. It was described that only
antigen presentation to infection or loss of homeostasis and
5% of ex vivo isolated CD45low rat microglia expressed MHC
therefore satisfies a teleological consideration of Need driving
II, compared with 15% of CD45high cells (Ford et al. 1995).
Function. Such principles, originally devised for DC, resonate
It should be noted that neither population expressed high
particularly strongly for processes that might lead to immune
endogenous levels of MHC II. Once microglia are activated,
response within the CNS.
however, MHC II is upregulated and readily detectable. One
of the best known stimuli for induction of MHC II is the T-
and natural killer (NK)-cell cytokine IFNJ (Goverman 2009).
6.1 TO L L-L I K E A N D OT H E R I N NAT E R EC E P TO R S
As has already been discussed, this may not be so helpful for
consideration of innate antigen-presenting ability because pro- The TLRs are a family of Type I proteins almost all of which
duction of IFNJ implies that antigen presentation has already signal via IL1-receptor homologous domain interactions with
occurred, although of course it has relevance to induction of the cytoplasmic adaptor molecule myeloid differentiation pri-
secondary responses, an important aspect of MS. Stimuli that mary response gene-88 (MyD88) and downstream interme-
may be more meaningful in terms of induction of protective diates to induce transcriptional activation of genes encoding
immune responses would include pathogen-associated molec- costimulator receptors, cytokines, and chemokines (Fig. 50.3).
ular pattern (PAMP), or damage-associated molecular pattern Toll-like receptor 3 and under some circumstances TLR4 do
(DAMP) antigens, both of which signal via innate receptors not signal via MyD88, but use an alternative pathway to engage
(Mills 2011). A number of candidate CNS DAMPs have been with nuclear machinery (Kawai and Akira 2009). Microglia
identified (Amor et al. 2010). stand out from other CNS-resident cells in detectably express-
ing all members of the TLR family, and can upregulate their
expression in response to infection and inflammation (Kielian
6 I N N AT E R E C E P TO R S I G N A L I N G F O R 2006). Cooperation between TLRs in microglial signaling has
MICROGLIAL RESPONSE also been shown (Holley et al. 2012), which underscores that
the microglial response to local milieux is likely to be com-
Innate receptors recognize conserved epitopes whose pre- plex. Notably some of the TLRs are predominantly expressed
sentation in a tissue reflects either infection or damage. Such intracellularly, in phagosomes, in which case ligand access is
“molecular pattern” epitopes do not therefore signal non-self dependent on cellular uptake. Another class of intracellular
so much as loss of homeostasis. This is a very important con- innate receptors expressed by microglia, whose role in anti-
cept for the CNS where microglia (as described in chapters 47 gen presentation remains less well worked out, are the retinoic
to 49) are constantly surveying their surroundings. Encounter acid inducing gene-I (RIG-I)-like helicases, or RLH receptors
with innate ligands triggers microglia to upregulate MHC (Dann et al. 2011; Kawai and Akira 2009). These bind viral
and costimulatory molecules, that is to say, to become a “pro- RNA, similar to TLR3, 7, and 9 and so they act as primary
fessional APC” (Takeda and Akira 2001). This concept is signals of cellular infection. However, release of RNA and
illustrated in Figure 50.2. The innate or pattern recognition conformationally similar entities are also a consequence of cell
receptors (PRR) that play the most well understood role are damage and their uptake may also trigger microglial antigen
the TLRs ( Janeway and Medzhitov 2002). Although there presenting capability. Of particular interest is the ability of

PAMPs, DAMPs

IL12, IL23
phagocytosis
PRRs
Antigen Naive
T cell

Th1,
Costimulatory Th17
molecules

Immature APC Mature APC

Figure 50.2 Principles of Antigen Presentation. The figure shows that stimulation of immature APC by PAMPs or DAMPs, acting through PRRs,
induces expression of costimulatory ligands and maturation of the APC. Whether the PAMP/DAMP is a component of pathogen or tissue antigen, or
whether antigen is separately phagocytosed, the result is a mature APC expressing MHC+ peptide that is equipped to costimulate a T cell by virtue of
costimulator ligand and cytokine expression (IL12 and IL23 are shown). The result of this interaction is an effector T cell that secretes cytokines such
as IFNJ (Th1) and IL17 (Th17).

642 • NEUROGLIA
TLR7, TLR9 TLR3 RLH TLR4 TLR2, TLR4
7 C O S T I MU L AT I O N O F M I C R O G L I A L
RESPONSES

Although the molecular details remain elusive it is assumed


TIRAP TRAM TIRAP
MyD88
TRIF TRADD
TRIF MyD88
that expression of microglial components of both costimula-
tory and regulatory pathways is controlled by their milieu, via
innate receptors. Microglia have been shown to upregulate
TRAF6
B7.1/CD80 and B7.2/CD86 in EAE and in MS (Goverman
TRAF6
2009; Issazadeh et al. 1998; Windhagen et al. 1995). Functional
TRAF3
FADD relevance of B7 molecule expression was evidenced by resis-
tance to EAE in mice that lacked their expression (Chang et al.
Type I IFN NFκB
1999), although this did not directly implicate B7 in micro-
glial antigen presentation. Interestingly, in response to synap-
Figure 50.3 Signaling Pathways Through Pattern Recognition Receptors tic degeneration, an experimental system that does not have
for Type I Interferon Production. TLR3,4, 7, 9, and RLH receptors signal a destructive autoimmune component, microglia upregulated
for Type I IFN production. Type I IFN inducing pathways are shown B7.2/CD86 but not B7.1/CD80 (Bechmann et al. 2001). This
in red. The TLR7 and TLR9 (receptors for virus PAMPs) and TLR2 was interpreted to indicate that failure to upregulate B7.1/
and TLR4 (receptors for bacterial PAMPs) all engage the intracellular
signaling intermediate MyD88 via Toll/IL1 receptor domain containing CD80 might account for the lack of a destructive autoimmune
adapter protein TIRAP, and this leads to activation of TRAF6. In the T-cell response. However, a transgenic mouse in which micro-
case of TLR7 and TLR9, TRAF6 then engages TRAF3, which results glia expressed B7.2/CD86 developed spontaneous EAE-like
in type I IFN transcription and production (red arrows), whereas for symptoms with demyelination and a marked infiltration of
TLR2 and TLR4 (gray arrow), TRAF6 leads to nuclear factor kappa B CD8+ T cells to spinal cord (Zehntner et al. 2003). Although
(NFkB) activation and production of inflammatory cytokines. TLR4
also activates TRIF via TRIF-related adapter molecule TRAM, and this transgenic systems can sometimes have idiosyncratic mecha-
too can activate TRAF6 and NFkB (gray half arrow), but can also lead nistic aspects; this exemplifies the fundamental costimulatory
to TRAF3 activation and type I IFN (red half arrow). The viral PAMP paradigm. Induction of costimulation via for example infec-
receptor TLR3 and the RLH receptors also activate TRAF3, either via tion-related stimuli results in populations of primed or mature
TRIF (TLR3) or the TNF receptor-associated death domain TRADD APC that can most effectively present pathogenic epitopes to
(RLH), respectively, both inducing type I IFN. Signaling through all of
these innate receptors can also activate NFkB, shown by dotted arrows in host-protective T cells (Fig. 50.4). Unfortunately, this results
the background. The pathway to NFkB from RLH receptors involves the in their also being primed for presentation of myelin or other
Fas-associated death domain FADD. CNS-derived self-epitopes.

PRRs to trigger production of Type I IFN. This is signaled via


TNF receptor-associated factor-3 (TRAF3) accessed either via 1).
the MyD88 or Toll/IL1R-containing adapter inducing IFNE
(TRIF) pathways (see Fig. 50.3) (Kawai and Akira 2009). 2).
Microglia MHC/TCR
Innate receptors may also regulate cytokine responses by
microglia. Microglia are known to express TLR2 in models of CD28
CNS infection in which exogenous PAMPs are a likely trigger No costimulation-no response
(Nichols et al. 2009). In experimental brain abscesses induced
Infection or other stimuli
by Staphylococcus aureus, levels of IL17 and infiltrating Th17 as (PAMPs, DAMPs)
well as gamma-delta IL17 producing T cells were significantly 3).
enhanced in TLR2-knock-out mice (Nichols et al. 2009).
These findings potentially implicate TLR2 signals in microglial B7.2
regulation of the Th17 response in the CNS, as discussed in a
later section. In the brain abscess model, microglia were shown Demyelination
to express the IL17RA/CD217 receptor and so were also com-
petent to respond to IL17, although the predominant effect Figure 50.4 How Costimulation Regulates Antigen Presentation in
of IL17 in this system was to synergize with tumor necrosis the Central Nervous System. Microglia that have not been induced to
factor-alpha (TNFD) for induction of chemokine production express costimulator ligands do not activate T cells that may enter the
CNS (1), even though they may express antigenic peptides associ-
by macrophages (Nichols et al. 2009). Interestingly, microglia ated with MHC. Recognition of such peptide+MHC by T cells in
also upregulate TLR2 in response to noninfectious stimuli the absence of a costimulatory signal (CD28 is shown) may anergize
such as axonal lesion-induced synaptic degeneration (Babcock T cells (2), thus contributing to the noninflammatory CNS environ-
et al. 2006) and spinal cord injury (Kigerl et al. 2007), and in ment. Microglia that have encountered PAMP or DAMP signals as
mice that lack TLR2, the CNS response to axonal lesion was a consequence of CNS infection or damage upregulate costimulator
ligands (B7.2/CD86 is shown) (3), and can present antigen to T cells
marked by reduced production of TNFD, including by micro- for induction of effector function, which can result in demyelinat-
glia (Babcock et al. 2006). This speaks to the innate immune ing disease (4). The figure is modified from an original drawing by
preparedness of microglia, as mentioned. Zehntner et al. 2003.

I M MU N E F U N C T I O N S O F M I C R O G L I A • 643
Additional tuning influences on the T-cell response TGFβ (Millward et al. 2010). Such Th17 T cells are impli-
induced by antigen-presenting microglia are provided by cated in EAE and MS, even though IL17 itself may not be
cytokines, which in turn direct cytokine production by T cells. essential (Goverman 2009; Haak et al. 2009). Microglial
T cells that induce inflammation are characterized by cytokine functional and regional heterogeneity are likely to play a role
profiles, which include IFNJ as well as IL17, IL21, and gran- in this context. The fact that intrathecal delivery of adenovi-
ulocyte-macrophage colony-stimulating factor. Induction of ral IL18bp inhibited Th17 and EAE was interpreted to show
these proinflammatory cytokines is driven by synergistic or in that IL18bp production by cells at the BBB controls T cell
some cases opposing actions of IL12, IL18, and IL23, with par- inflammatory potential as they enter the CNS (Millward
ticipation of IL1, IL6, and transforming growth factor-beta et al. 2010). Whether the critical APC were DC or microglia
(TGFβ) (Gutcher and Becher 2007). All of these cytokines was not determined, but proximity to blood vessels and the
can be produced by microglia and as for costimulatory ligand/ BBB are a likely influence on microglial phenotype (Hanisch
receptors, it either has been shown or can be assumed that and Kettenmann 2007).
PAMP and DAMP signals control such glial response.

8.2 T Y P E I I N T E R FE RO N
8 C Y TO K I N E A N D C H E M O K I N E The type I IFNβ (used as a therapy for MS) has been shown
P R O D U C T I O N BY M I C R O G L I A to inhibit Th17 responses (Guo et al. 2008), and whether
IFNβ inhibits EAE or MS correlated inversely with IL17 lev-
Microglia are a source of as wide a range of cytokines and els (Axtell et al. 2010). Microglia express interferon regulatory
chemokines as other myeloid cells without distinguishing factor-7 in EAE (Salem et al. 2011) or in response to synaptic
characteristics from macrophages, for example. Under appro- degeneration (Khorooshi and Owens 2010), and they are con-
priate stimulus they can produce cytokines considered both sidered a likely source of type I IFN. It has been shown that
pro inflammatory and antiinflammatory, as well as chemok- the critical type I IFN-responding population for regulation
ines attractant for multiple T-cell subsets, macrophages, DC, of EAE are of the myeloid lineage and so might include micro-
and granulocytes (Hanisch and Kettenmann 2007). This glia (Prinz et al. 2008). This reinforces that microglia may act
broad capacity for cytokine and chemokine production sug- to dampen inflammation in the CNS.
gests that it is not only the cell but also its context that will
define the microglial contribution to any particular response.
9 C D 11 C + M I C R O G L I A
8.1 I M MU N O R E GU L ATO RY RO L E
CD11c is upregulated under inflammatory conditions by a sub-
F O R M I C RO G L I A L C Y TO K I N E S
population of microglia that have preferential ability to pres-
The possibility that microglia might subserve an immuno- ent antigen. It was reported that microglia become activated
regulatory function is of particular interest. They are impli- early in EAE and that these activated microglia upregulate
cated in inflammatory responses via “guilt by association,” MHC II, CD40, and CD86, as well as CD11c (Ponomarev
but their actual role remains unclear. Butovsky et al. showed et al. 2005). The use of radiation bone marrow chimeras in that
that microglia could be directed by the cytokine IL4 to a study, to distinguish microglia from peripheral myeloid cells,
CD11b+ CD11c+ phenotype associated with production of introduces a caution about the possibility of aberrant cell acti-
insulin-like growth factor-1 (IGF1) and neurogenic poten- vation (see Mildner et al. 2007). However, analogous findings
tial (Butovsky et al. 2006). Two recent genomewide array were made in mice undergoing cuprizone-induced demyelina-
screens of microglia isolated by cell sorting from mice have tion, wherein there is a pronounced activation of microglia in
shown that microglia display proregenerative gene expres- demyelinating corpus callosum. These were identified as being
sion profiles. In one case CD11b+ CD45dim microglia from greater than 90% CD45dim microglia and not macrophages by
the corpus callosum of mice undergoing cuprizone-induced flow cytometry (Remington et al. 2007). Among the CD45dim
demyelination and subsequent remyelination showed upreg- cells was a subpopulation that upregulated CD11c, which is
ulation of the CD11c-encoding gene itgax as well as chemok- not normally easily detectable on microglia (Remington et al.
ines and cytokines that would support oligodendrocyte 2007). Similar CD11c+ microglia were identified in the den-
precursor cell recruitment (Olah et al. 2012). Expression of tate gyrus of mice that had undergone experimental denerva-
IGF1 and other proneurogenic genes was shown for CD11b+ tion of the hippocampus (Babcock et al. 2008). Populations
CD45dim microglia isolated from the subventricular zone of of microglia from cuprizone-treated mice, which included
mice with EAE (Starossom et al. 2011). The authors called CD11c-positive microglia, were shown to be highly potent
attention to expression of IL18-binding protein (IL18bp), a antigen presenting cells for T-cell response induction in vitro
soluble factor that neutralizes an inhibitor of neuronal dif- (Remington et al. 2007). Correspondingly, one of the genes
ferentiation, by these microglia (Starossom et al. 2011). It strongly upregulated by CD11b+ CD45dim microglia sorted
has been reported that CD11b+ CD45dim microglia express from such tissues was the CD11c-encoding itgax gene (Olah
IL18bp in EAE, and shown that IL18bp can inhibit Th17 in et al. 2012). Unexpectedly, in that same study, neither CD40
the CNS when overexpressed using a viral vector. This was nor CD80 was detected as expressed and CD86 was not
likely caused by suppression of APC-derived IL6, IL23, and regulated.

644 • NEUROGLIA
9 L O C AT I O N O F A N T I G E N -P R E S E N T I N G so much their innate capability as APC as their location in the
C E L L S I N T H E C E N T R A L N E RVO U S tissue. A recent study tracked CD11c+ cells in mouse CNS via
SYSTEM itgax promoter-driven GFP expression. It is possible that some
of the cells that were visualized were microglia, because flow
By now the reader may be wondering whether microglia are cytometric analysis of relative CD45 levels was not employed
just immature CNS DC, given that they appear to present (Prodinger et al. 2011). It is of interest that CD11c promoter-
antigen equivalently and share at least some surface markers driven GFP+ cells in that study were localized juxtavascularly
once activated. Relative CD45 levels distinguish parenchymal at the glia limitans, which would place this subset of microglia
microglia from other myeloid cells in CNS, including DC, in communication with cells in perivascular space, a role that
but once they are sufficiently activated, overlap in phenotype, has been conventionally assigned to DC (Becher et al. 2006).
and function cannot be formally excluded (see Fig. 50.1). It Identification of CD45low CD11c+ microglia helps to rec-
is not easily resolved whether some of the CD45high CD11b+ oncile studies that reported parenchymal DC in mouse mod-
CD11c+ cells in inflamed CNS started out as CD45low micro- els of ischemia and brain infection (Fischer and Reichmann
glia. There are technical limitations to the extent to which 2001). Discrimination of microglia from blood-derived cells
individual cells originating within a tissue can be tracked to by measurement of relative CD45 expression was not done
other locations and phenotypes. Consensus at present is that in those studies, and it may be speculated that the CD11b+
DC are a distinct subset of myeloid cells, but one should keep CD11c+ cells that were described may have at least included
an open mind as to the extent to which this distinction is CD11c+ microglia. Curiously, neither itgax or the CD11b-
function- and context-dependent. encoding itgam genes, nor CD80 or CD86 were among those
It is of interest that CD11c serves as a marker for APCs upregulated in CD45dimCD11b+ microglia isolated from the
in the CNS, as elsewhere. It has been suggested that DC are subventricular zone of mice with EAE, although a plethora
primarily situated in perivascular locations appropriate to a of other genes associated with antigen presentation (includ-
“gatekeeper” function, whereas microglia are parenchymally ing CD40) were among the signature genes in that study
located and so may play more of a role in progression of EAE (Starossom et al. 2011).
and MS than in initiation (Becher et al. 2006; Greter et al.
2005) (Fig. 50.5). Such compartmentalization (both physi-
cal and intellectual) helps to resolve continuing debate over 10 R E S P O N S E TO C Y TO K I N E S
whether microglia can present antigen as effectively as DC, but
may also be an oversimplification. The CD11c+ DC is clearly Dialog between microglia and inflammatory cells is mediated
an effective APC, and it may be speculated that the primary at least in part via cytokines. This can result either in progres-
difference between them and microglia may turn out to be not sion or remission of the inflammatory response. For example,
microglia have been shown to express the IL-17RA/CD217
receptor for the inflammatory cytokine IL17A/F, which
Parenchymal
microglia Neuropil
defines Th17 (Das Sarma et al. 2009). In vitro studies show
that IL17 induces microglial as well as astroglial production
of chemokines such as MCP-1/CCL2, MCP-5/CCL12, and
Perivascular Glia limitans
space
KC/MIP-2/CXCL1 (Das Sarma et al. 2009). This would
predict amplification of the inflammatory response via glial-
DC
derived chemokines recruiting macrophages and granulo-
Blood Perivascular cytes. Because monocytes are also a potential source of IL17
vessel microglia (Moseley et al. 2003), the microglial response may not only
be directed by Th17, but also by infiltrating macrophages, and
Endothelium
microglia themselves are also reported to be a source of IL17
(Kawanokuchi et al. 2008; Lv et al. 2011). On the other hand
DC the IL17 response may itself be inhibited by mediators pro-
duced by microglia in the CNS. These include type I IFN and
Figure 50.5 Importance of Location for Antigen Presentation in the IL18bp (Millward et al. 2010; Salem et al. 2011). Microglia
Central Nervous System. T cells that enter the CNS in MS and EAE also respond to the immunomodulatory cytokine TGF-β
do so in postcapillary venules in which the vascular endothelial and glia which can derive from T-cell subsets as well as from other glial
limitans basement membranes are separated by a perivascular space. cells and neurons and is implicated in preserving the relatively
Dendritic cells in perivascular space are considered to play a role as
gatekeeper APC. Presentation of antigen by DC to T cells permits their
noninflammatory state of the CNS (Hanisch and Kettenmann
further activation and facilitates their transit across the glia limitans into 2007; Liu et al. 2006). Another well-known modulatory com-
the CNS parenchyma. Juxtavascular DC and (it is speculated) microglia ponent of the microglial environmental milieu is ATP acting
extend processes across the glia limitans, which can also present antigen via purinergic receptors, which has been shown to reduce the
to trafficking T cells. By contrast, parenchymal microglia, deeper in in vitro induction by LPS of IL12 as well as other cytokines and
the neuropil, only encounter T cells that make it that far. This imposes
restrictions on their capacity for T cell interaction, especially with naïve
chemokines (Boucsein et al. 2003). It should be kept in mind
T cells. The figure incorporates concepts described in Becher et al. that demonstration of microglial capability to respond to or
(2006), Owens et al. (2008), and Prodinger et al. (2011). produce mediators that modulate production of cytokines

I M MU N E F U N C T I O N S O F M I C R O G L I A • 645
in vitro is not always a predictor of microglial response in microglia subserve DC functions, including the ability to traf-
vivo, and there may also be issues of microglial heterogeneity fic out of the CNS for antigen presentation in lymph nodes A
(Hanisch and Kettenmann 2007). number of groups are engaged in addressing these issues, and
description of progress and advances can be found in chapters
15, 19, and 47 to 49.
11 M I C R O G L I A L R E GU L AT I O N O F
T CELLS
AC K N OW L E D G M E N T S
Regulation of inflammatory immune response also involves
direct cellular ligand–receptor interactions. The PD-1 recep- This work was supported by grants from the Danish Agency
tor on CD4+ T cells signals for reduced Th1 response when for Research and Innovation, the Danish Multiple Sclerosis
engaged by PD-L1/B7-H1/CD274 that is expressed by myel- Society, the Lundbeck Foundation, and the Novo Nordisk
oid cells, including microglia (Schreiner et al. 2008). This Foundation.
has been implicated in regulating relapses in EAE, as well as
in modulating inflammatory T cells in Theiler virus demy-
elination (Duncan and Miller 2011). Clearly, the stimuli for REFERENCES
induction of PD-L1/CD274 on microglia, as well as whether
Aloisi F, Ria F, Columba-Cabezas S, Hess H, Penna G, Adorini L. 1999.
there is heterogeneity of PD-L1 expression among microglia, Relative efficiency of microglia, astrocytes, dendritic cells and B cells
need to be defined, to more fully understand this microglial in naive CD4+ T cell priming and Th1/Th 2 cell restimulation. Eur J
regulation of Th1 response in the CNS. Of interest is that Immunol 29(9):2705–2714.
PD-L1 was found to be strongly upregulated by microglia Amor S, Puentes F, Baker D, van der Valk P. 2010. Inflammation in neu-
responding to cuprizone demyelination, an experimental sys- rodegenerative diseases. Immunology 129(2):154–169.
Arnett HA, Mason J, Marino M, Suzuki K, Matsushima GK, Ting JP.
tem that is T-cell–independent (Arnett et al. 2001; Olah et al. 2001. TNFalpha promotes proliferation of oligodendrocyte progeni-
2012). Again one sees evidence of innate programs of micro- tors and remyelination. Nat Neurosci 4(11):1116–1122.
glial response that predispose to interaction with cells of the Axtell RC, de Jong BA, Boniface K, van der Voort LF, Bhat R, De Sarno
immune system, in this case to modulate response. P, et al. 2010. T helper type 1 and 17 cells determine efficacy of
interferon-beta in multiple sclerosis and experimental encephalomy-
elitis. Nat Med 16(4):406–412.
12 S U M M A RY A N D P E R S P E C T I VE S Babcock AA, Toft-Hansen H, Owens T. 2008. Signaling through
MyD88 regulates leukocyte recruitment after brain injury. J Immunol
181(9):6481–6490.
Microglia secrete cytokines and chemokines and express sur- Babcock AA, Wirenfeldt M, Holm T, Nielsen HH, Dissing-Olesen L,
face molecules consistent with antigen-presenting function. Toft-Hansen H, et als. 2006. Toll-like receptor 2 signaling in response
Microglia have been directly demonstrated to present antigen to brain injury: an innate bridge to neuroinflammation. J Neurosci
to T cells and they are the best-equipped parenchymal CNS- 26(49):12826–12837.
resident cell to do this. Ongoing studies of the influence of the Bai B, Song W, Ji Y, Liu X, Tian L, Wang C, et al. 2009. Microglia and
microglia-like cell differentiated from DC inhibit CD4 T cell prolif-
CNS environment on the antigen-presenting ability of other eration. PLoS One 4(11):e7869.
cells such as DC suggest that microglia may be equally effec- Becher B, Bechmann I, Greter M. 2006. Antigen presentation in autoim-
tive APC as DC in situ. The challenge now is to devise ways munity and CNS inflammation: how T lymphocytes recognize the
to address this. Recent cell isolation and microarray studies brain. J Mol Med 84(7):532–543.
point the way toward more precise gene and proteomic analy- Bechmann I, Nitsch R. 1997. Astrocytes and microglial cells incorporate
degenerating fibers following entorhinal lesion: a light, confocal, and
ses of microglial subpopulations and the immune cells whose electron microscopical study using a phagocytosis-dependent label-
responses they control. Key questions for the future will be, ing technique. Glia 20(2):145–154.
where are the antigen-presenting microglia located in the Bechmann I, Peter S, Beyer M, Gimsa U, Nitsch R. 2001. Presence of
CNS? And how heterogeneous are microglia both with regard B7–2 (CD86) and lack of B7–1 (CD(80) on myelin phagocytosing
to tissue localization and in the context of response? It will be MHC-II-positive rat microglia is associated with nondestructive
immunity in vivo. Faseb J 15(6):1086–1088.
of interest to determine the extent to which the local environ- Boucsein C, Zacharias R, Farber K, Pavlovic S, Hanisch UK, Kettenmann
ment determines microglial phenotype, including antigen- H. 2003. Purinergic receptors on microglial cells: functional expres-
presenting ability, versus whether predetermined microglial sion in acute brain slices and modulation of microglial activation in
phenotypes define the local antigen-presenting environment. vitro. Eur J Neurosci 17(11):2267–2276.
Transplantation of microglial subpopulations is currently not Bullard DC, Hu X, Adams JE, Schoeb TR, Barnum SR. 2007.
p150/95 (CD11c/CD18) Expression is required for the develop-
technically feasible and would seem challenging, so a more ment of experimental autoimmune encephalomyelitis. Am J Pathol
plausible approach would be to control local production of 170(6):2001–2008.
selected candidate mediators. A further challenge will be to Bullard DC, Hu X, Schoeb TR, Axtell RC, Raman C, Barnum SR. 2005.
selectively activate or disable subpopulations of microglia in Critical requirement of CD11b (Mac-1) on T cells and accessory cells
otherwise physiologically normal circumstances, to assess their for development of experimental autoimmune encephalomyelitis.
J Immunol 175(10):6327–6333.
relative contribution to CNS immunity. The environmental Butovsky O, Koronyo-Hamaoui M, Kunis G, Ophir E, Landa G, Cohen
cues for microglial participation in immune responses need to H, et al. 2006. Glatiramer acetate fights against Alzheimer’s disease
be defined—unequivocal identification of CNS DAMPs and by inducing dendritic-like microglia expressing insulin-like growth
their receptors will be necessary. Finally, to what extent can factor 1. Proc Natl Acad Sci U S A 103(31):11784–11789.

646 • NEUROGLIA
Chang TT, Jabs C, Sobel RA, Kuchroo VK, Sharpe AH. 1999. Studies Kawanokuchi J, Shimizu K, Nitta A, Yamada K, Mizuno T, Takeuchi
in B7-deficient mice reveal a critical role for B7 costimulation in both H, et al. 2008. Production and functions of IL-17 in microglia.
induction and effector phases of experimental autoimmune encepha- J Neuroimmunol 194(1–2):54–61.
lomyelitis. J Exp Med 190(5):733–740. Khorooshi R, Owens T. 2010. Injury-induced type I IFN signaling
Dann A, Poeck H, Croxford AL, Gaupp S, Kierdorf K, Knust M, et al. regulates inflammatory responses in the central nervous system.
2011. Cytosolic RIG-I-like helicases act as negative regulators of ster- J Immunol 185(2):1258–1264.
ile inflammation in the CNS. Nat Neurosci 15(1):98–106. Kielian T. 2006. Toll-like receptors in central nervous system glial
Das Sarma J, Ciric B, Marek R, Sadhukhan S, Caruso ML, Shafagh J, inflammation and homeostasis. J Neurosci Res 83(5):711–730.
et al. 2009. Functional interleukin-17 receptor A is expressed in cen- Kielian T, Mayes P, Kielian M. 2002. Characterization of microglial
tral nervous system glia and upregulated in experimental autoim- responses to Staphylococcus aureus: effects on cytokine, costimula-
mune encephalomyelitis. J Neuroinflam 6:14. tory molecule, and Toll-like receptor expression. J Neuroimmunol
Duncan DS, Miller SD. 2011. CNS expression of B7-H1 regulates 130(1–2):86–99.
pro-inflammatory cytokine production and alters severity of Theiler’s Kigerl KA, Lai W, Rivest S, Hart RP, Satoskar AR, Popovich PG. 2007.
virus-induced demyelinating disease. PLoS One 6(4):e18548. Toll-like receptor (TLR)-2 and TLR-4 regulate inflammation,
Engelhardt B, Ransohoff RM. 2005. The ins and outs of T-lymphocyte gliosis, and myelin sparing after spinal cord injury. J Neurochem
trafficking to the CNS: anatomical sites and molecular mechanisms. 102(1):37–50.
Trends Immunol 26(9):485–495. Koenigsknecht J, Landreth G. 2004. Microglial phagocytosis of fibril-
Fischer HG, Reichmann G. 2001. Brain dendritic cells and macrophages/ lar {beta}-amyloid through a {beta}1 integrin-dependent mechanism.
microglia in central nervous system inflammation. J Immunol J Neurosci 24(44):9838–9846.
166(4):2717–2726. Liu Y, Teige I, Birnir B, Issazadeh-Navikas S. 2006. Neuron-mediated
Fitzner D, Schnaars M, van Rossum D, Krishnamoorthy G, Dibaj P, generation of regulatory T cells from encephalitogenic T cells sup-
Bakhti M, et al. 2011. Selective transfer of exosomes from oligo- presses EAE. Nat Med 12(5):518–525.
dendrocytes to microglia by macropinocytosis. J Cell Sci 124(Pt 3): Lv M, Liu Y, Zhang J, Sun L, Liu Z, Zhang S, et al. 2011. Roles of inflam-
447–458. mation response in microglia cell through Toll-like receptors 2/
Fontana A, Fierz W, Wekerle H. 1984. Astrocytes present myelin interleukin-23/interleukin-17 pathway in cerebral ischemia/reperfu-
basic protein to encephalitogenic T-cell lines. Nature 307(5948): sion injury. Neuroscience 176:162–172.
273–276. Malone KE, Stohlman SA, Ramakrishna C, Macklin W, Bergmann
Ford AL, Goodsall AL, Hickey WF, Sedgwick JD. 1995. Normal adult CC. 2008. Induction of class I antigen processing components in
ramified microglia separated from other central nervous system mac- oligodendroglia and microglia during viral encephalomyelitis. Glia
rophages by flow cytometric sorting. Phenotypic differences defined 56(4):426–435.
and direct ex vivo antigen presentation to myelin basic protein- reac- McMenamin PG. 1999. Distribution and phenotype of dendritic cells
tive CD4+ T cells compared. J Immunol 154(9):4309–4321. and resident tissue macrophages in the dura mater, leptomeninges,
Goverman J. 2009. Autoimmune T cell responses in the central nervous and choroid plexus of the rat brain as demonstrated in wholemount
system. Nat Rev Immunol 9(6):393–407. preparations. J Comp Neurol 405(4):553–562.
Greter M, Heppner FL, Lemos MP, Odermatt BM, Goebels N, Laufer T, Mildner A, Schmidt H, Nitsche M, Merkler D, Hanisch UK, Mack M,
et al. 2005. Dendritic cells permit immune invasion of the CNS in an et al. 2007. Microglia in the adult brain arise from Ly-6ChiCCR2+
animal model of multiple sclerosis. Nat Med 11(3):328–334. monocytes only under defined host conditions. Nat Neurosci
Guo B, Chang EY, Cheng G. 2008. The type I IFN induction pathway 10(12):1544–1553.
constrains Th17-mediated autoimmune inflammation in mice. J Clin Mills KH. 2011. TLR-dependent T cell activation in autoimmunity. Nat
Invest 118(5):1680–1690. Rev Immunol 11(12):807–822.
Gutcher I, Becher B. 2007. APC-derived cytokines and T cell polariza- Millward JM, Lobner M, Wheeler RD, Owens T. 2010. Inflammation
tion in autoimmune inflammation. J Clin Invest 117(5):1119–1127. in the central nervous system and Th17 responses are inhibited
Haak S, Croxford AL, Kreymborg K, Heppner FL, Pouly S, Becher B, by IFN-{gamma}-induced IL-18 binding protein. J Immunol
et al. 2009. IL-17A and IL-17F do not contribute vitally to autoim- 185(4):2458–2466.
mune neuro-inflammation in mice. J Clin Invest 119(1):61–69. Mizushima N, Komatsu M. 2011. Autophagy: renovation of cells and tis-
Hanisch UK, Kettenmann H. 2007. Microglia: active sensor and versa- sues. Cell 147(4):728–741.
tile effector cells in the normal and pathologic brain. Nat Neurosci Moseley TA, Haudenschild DR, Rose L, Reddi AH. 2003.
10(11):1387–1394. Interleukin-17 family and IL-17 receptors. Cytokine Growth Factor
Holley MM, Zhang Y, Lehrmann E, Wood WH, Becker KG, Kielian Rev 14(2):155–174.
T. 2012. Toll-like receptor 2 (TLR2)-TLR9 crosstalk dictates IL-12 Nichols JR, Aldrich AL, Mariani MM, Vidlak D, Esen N, Kielian T.
family cytokine production in microglia. Glia 60(1):29–42. 2009. TLR2 deficiency leads to increased Th17 infi ltrates in experi-
Holm TH, Draeby D, Owens T. 2012. Microglia are required for astro- mental brain abscesses. J Immunol 182(11):7119–7130.
glial toll-like receptor 4 response and for optimal TLR2 and TLR3 Olah M, Amor S, Brouwer N, Vinet J, Eggen B, Biber K, et al. 2012.
response. Glia 60(4):630–638. Identification of a microglia phenotype supportive of remyelination.
Hughes MM, Field RH, Perry VH, Murray CL, Cunningham C. 2010. Glia 60(2):306–321.
Microglia in the degenerating brain are capable of phagocytosis of Owens T, Bechmann I, Engelhardt B. 2008. Perivascular spaces and
beads and of apoptotic cells, but do not efficiently remove PrPSc, even the two steps to neuroinflammation. J Neuropathol Exp Neurol
upon LPS stimulation. Glia 58(16):2017–2030. 67(12):1113–1121.
Issazadeh S, Navikas V, Schaub M, Sayegh M, Khoury S. 1998. Kinetics Ponomarev ED, Shriver LP, Maresz K, Dittel BN. 2005. Microglial cell
of expression of costimulatory molecules and their ligands in murine activation and proliferation precedes the onset of CNS autoimmu-
relapsing experimental autoimmune encephalomyelitis in vivo. nity. J Neurosci Res 81(3):374–389.
J Immunol 161(3):1104–1112. Prinz M, Schmidt H, Mildner A, Knobeloch KP, Hanisch UK, Raasch J,
Janeway CA Jr, Medzhitov R. 2002. Innate immune recognition. Annu et al. 2008. Distinct and nonredundant in vivo functions of IFNAR
Rev Immunol 20:197–216. on myeloid cells limit autoimmunity in the central nervous system.
Kaminski M, Bechmann I, Pohland M, Kiwit J, Nitsch R, Glumm J. Immunity 28(5):675–686.
2012. Migration of monocytes after intracerebral injection at ento- Prodinger C, Bunse J, Kruger M, Schiefenhovel F, Brandt C, Laman JD,
rhinal cortex lesion site. J Leukoc Biol 6(3) [Epub ahead of print]. et al. 2011. CD11c-expressing cells reside in the juxtavascular paren-
Kawai T, Akira S. 2009. The roles of TLRs, RLRs and NLRs in pathogen chyma and extend processes into the glia limitans of the mouse ner-
recognition. Int Immunol 21(4):317–337. vous system. Acta Neuropathol 121(4):445–458.

I M MU N E F U N C T I O N S O F M I C R O G L I A • 647
Remington LT, Babcock AA, Zehntner SP, Owens T. 2007. Microglial Ulvestad E, Williams K, Bjerkvig R, Tiekotter K, Antel J, Matre R.
recruitment, activation, and proliferation in response to primary 1994. Human microglial cells have phenotypic and functional
demyelination. Am J Pathol 170(5):1713–1724. characteristics in common with both macrophages and dendritic
Salem M, Mony JT, Lobner M, Khorooshi R, Owens T. 2011. Interferon antigen-presenting cells. J Leukoc Biol 56(6):732–740.
regulatory factor-7 modulates experimental autoimmune encephalo- van Zwam M, Huizinga R, Heijmans N, van Meurs M, Wierenga-Wolf
myelitis in mice. J Neuroinflammation 8(1):181–189. AF, Melief MJ, et al. 2009. Surgical excision of CNS-draining lymph
Saura J. 2007. Microglial cells in astroglial cultures: a cautionary note. nodes reduces relapse severity in chronic-relapsing experimental auto-
J Neuroinflammation 4(1):26. immune encephalomyelitis. J Pathol 217(4):543–551.
Schreiner B, Bailey SL, Shin T, Chen L, Miller SD. 2008. PD-1 ligands Williams K, Ulvestad E, Antel JP. 1994. B7/BB-1 antigen expression on
expressed on myeloid-derived APC in the CNS regulate T-cell adult human microglia studied in vitro and in situ. Eur J Immunol
responses in EAE. Eur J Immunol 38(10):2706–2717. 24(12):3031–3037.
Sedgwick JD, Schwender S, Imrich H, Dorries R, Butcher GW, ter Williams KC, Dooley NP, Ulvestad E, Waage A, Blain M, Yong VW,
Meulen V. 1991. Isolation and direct characterization of resident et al. 1995. Antigen presentation by human fetal astrocytes with the
microglial cells from the normal and inflamed central nervous sys- cooperative effect of microglia or the microglial-derived cytokine
tem. Proc Natl Acad Sci U S A 88(16):7438–7442. IL-1. J Neurosci 15(3 Pt 1):1869–1878.
Starossom SC, Imitola J, Wang Y, Cao L, Khoury SJ. 2011. Subventricular Windhagen A, Newcombe J, Dangond F, Strand C, Woodroofe MN,
zone microglia transcriptional networks. Brain Behav Immun Cuzner ML, et al. 1995. Expression of costimulatory molecules B7–1
25(5):991–999. (CD80), B7–2 (CD86), and interleukin 12 cytokine in multiple scle-
Stohwasser R, Giesebrecht J, Kraft R, Muller EC, Hausler KG, rosis lesions. J Exp Med 182(6):1985–1996.
Kettenmann H, et al. 2000. Biochemical analysis of proteasomes Zehntner SP, Brisebois M, Tran E, Owens T, Fournier S. 2003.
from mouse microglia: induction of immunoproteasomes by Constitutive expression of a costimulatory ligand on antigen-presenting
interferon-gamma and lipopolysaccharide. Glia 29(4):355–365. cells in the nervous system drives demyelinating disease. FASEB J
Takeda K, Akira S. 2001. Roles of Toll-like receptors in innate immune 17(13):1910–1912.
responses. Genes Cells 6(9):733–742.

648 • NEUROGLIA
SECTION 3
ROLE OF GLIAL CELLS IN DISEASE
This page intentionally left blank
MECHANISMS OF GLIAL INJURY
This page intentionally left blank
51.
ASTROCY TE RESPONSES TO CENTRAL
NERVOUS SYSTEM INJURY AND DISEASE
Michael V. Sofroniew

A B B R E VI AT I O N S diseases. These findings have revealed that astrocyte reactivity is a


context-dependent, complex, and multivariate processes that has
ALS amyotrophic lateral sclerosis the potential to impact substantively on neural function in a vari-
APC adenomatous poliposis coli ety of ways. This chapter reviews current information about (1)
AQP4 aquaporin 4 cellular and molecular aspects of astrocyte changes in response to
ATP adenosine triphosphate injury and disease, (2) mechanisms that trigger and regulate these
BBB blood-brain barrier changes, and (3) functional consequences.
Ca2+ calcium
[Ca2+]i intracellular calcium activity
CNS central nervous system 2 T E R M I N O L O GY
CSPG chondroitin sulfate proteoglycans
EAAT2 excitatory amino acid transporter 2 The terminology used to describe astrocyte responses to injury
EAE experimental autoimmune encephalomyelitis can vary considerably among authors. There are no widely
EGF epidermal growth factor accepted categories that define different types of astrocyte
FGF fibroblast growth factor responses and the same terminology can be used to describe
GABA gamma-aminobutyric acid different things by different authors. In the interest of clarity it
GFAP glial fibrillary acid protein is useful to begin with a brief consideration of terminology and
Glt1 glutamate transporter 1 definitions of terms as used in this chapter (Table 51.1). Various
GP130 receptor for interleukin 6 terms that are in common use to describe astrocyte responses
GSNO S-nitrosoglutathione to injury or disease include: astrocyte reactivity, astrocyte acti-
INF-J interferon gamma vation, reactive astrogliosis, reactive astrocytosis, and glial scar
LPS lipopolysaccharide formation. As a general all-inclusive descriptor of astrocyte
NFκB nuclear factor kappa-light-chain-enhancer responses to stimuli associated with injury or disease, we pre-
of activated B cells fer the term reactive, as used in “reactive astrocytes” or “astro-
NGF nerve growth factor cyte reactivity” (see Table 51.1). Although the term astrocyte
NMO neuromyelitis optica activation is sometimes used to describe astrocyte responses to
NO nitric oxide injury or disease, we find this term confusing in light of the
SOC3 suppressor of cytokine signaling 3 now considerable evidence that astrocytes become activated
SOD superoxide dismutase in healthy tissue by exhibiting transient, ligand-evoked eleva-
STAT3 signal transducer and activator of tions in intracellular calcium activity ([Ca2+]i). This form of
transcription 3 healthy astrocyte activation is under intense investigation as
TGF-β transforming growth factor beta a potential means of mediating dynamic astrocyte functions,
VEGF-A vascular endothelial growth factor-A including interactions with synapses and regulation of blood
flow (Attwell et al. 2010; Barres 2008; Halassa and Haydon
2010) (see chapters 16, 17, 26, 37, 38, and 39). It seems appro-
1 INTRODUCTION priate at this time to differentiate this form of astrocyte activa-
tion in healthy physiological contexts from astrocyte reactivity
For well over 100 years, astrocytes have been known to undergo to stimuli associated with injury or disease.
anatomical changes in response to injury and disease in the mam- The terms reactive astrogliosis, reactive astrocytosis, and glial
malian central nervous system (CNS), and histopathological scar also deserve clarification (see Table 51.1). In this chapter we
changes in astrocytes are routinely used as hallmarks of disease or will use the term reactive astrogliosis to denote any astrocyte
damage in human CNS tissue. Until recently, however, the func- response, however mild or severe, undergone by astrocytes to any
tional implications of such astrocyte responses have been poorly form of CNS insult. Because the suffix “cytosis” is widely used
understood. Over the past 20 years, considerable information has in histopathology to denote an increase in cell number over that
accrued regarding molecular and functional changes associated normally observed, we will use the term reactive astrocytosis only
with astrocyte responses to many different types of injuries and when there is true evidence of astrocyte proliferation. Lastly, it

653
Table 51.1 TERMINOLOGY RELATED TO ASTROCYTE RESPONSES TO CNS INSULTS
TERM DEFINITION AS USED IN THIS ARTICLE

Reactive astrogliosis Umbrella term referring to all forms of astrocyte responses to all forms of CNS insults, ranging from very
mild reversible responses such as small changes in gene expression, to moderate responses involving large
changes in gene expression and structural changes such as hypertrophy, to severe responses that include
cell proliferation and permanent scar formation

Reactive astrocytosis Reactive proliferation of astrocytes that results from newly occurring cell division in direct response to a
CNS insult

Reactive astrocyte Umbrella term referring to an astrocyte that has responded in any way (mild to severe) to any form of
CNS insult

Scar forming astrocyte Reactive astrocyte that derives from newly proliferated cells and that lines the borders to lesions of
severely damaged tissue

Glial scar Multicellular structure that forms borders around severely damaged or inflamed CNS tissue, consisting of
newly proliferated, scar-forming astrocytes that abut and interact with fibromeningeal cells and fibro-
cytes (see Fig. 51.1D,E)

Mild to moderate astrogliosis Astrogliosis in which astrocytes hypertrophy but the individual domains of their processes are preserved
and tissue cytoarchitecture is preserved, and in which there is potential for resolution if the insult resolves
(see Fig. 51.1C)
Severe astrogliosis Astrogliosis in which there is astrocyte proliferation and substantial overlap of astrocyte processes with
loss of individual astrocyte domains; and in which there is permanent rearrangement of cytoarchitecture,
even if the insult resolves (see Fig. 51.1D,E)

deserves emphasis that the terms glial scar and reactive astrogliosis events that mediate different specific responses (Table 51.2).
are not synonymous (see Table 51.1), as discussed in detail in the The changes that can be associated with astrocyte reactivity
next section. range from reversible alterations in gene expression and cell
3 R E AC T I VE A S T R O G L I O S I S A S A hypertrophy with preservation of cellular domains and tissue
C O N T I N U U M O F C E L LU L A R C H A N G E S structure, to long lasting scar formation that involves cell
proliferation and rearrangement of tissue structure (see Fig. 51.1,
Contrary to commonly held beliefs, reactive astrogliosis is not Table 51.3) (Sofroniew 2009; Sofroniew and Vinters 2010).
a simple all-or-none stereotypic phenomenon. Instead, there Based on a large body of observations in experimental animals
is now ample experimental evidence that reactive astrogliosis and human pathological specimens, we have recently proposed
is a finely gradated continuum of changes that occur in a a definition of reactive astrogliosis that includes several grades
context-dependent manner and that can be independently of severity that may be commonly encountered in experimental
regulated by a multitude of different specific molecular signaling and clinical histopathological examinations (Sofroniew 2009;

Table 51.2 EXAMPLES OF MOLECULAR REGULATORS AND MODULATORS OF REACTIVE ASTROGLIOSIS


FUNCTIONAL CATEGORIES IMPLICATED FACTORS

A. Extracellular mediators

Cytokines and growth factors IL6, IL1β, TNFα, INFγ, CNTF, LIF, TGF β, IL10,
EGF, FGF2, endothelin

Neurotransmitters and modulators glutamate, noradrenalin

Small molecules released by cell injury ATP

Molecules of oxidative stress nitric oxide, reactive oxygen species

Ischemia-associated hypoxia and glucose deprivation

Infection-associated lipopolysaccharide (LPS)

Neurodegeneration associated β-amyloid


Systemic metabolic toxicity ammonium (NH4+)

B. Intracellular signaling molecules STAT3, NFκB, SOC3, MAP-kinase, SOX9, Nrf2, Olig2, cAMP, Nurr1, SMAD, mTOR

654 • NEUROGLIA
Sofroniew and Vinters 2010). It is important to emphasize scar formation can be seen adjacent to essentially all lesions of
that the different severities of reactive astrogliosis transition tissue damage formed by stroke, trauma, or infection (Fig. 51.2).
seamlessly along a continuum. Nevertheless, it is convenient Although differences in degree of reactive astrogliosis are readily
for purposes of description, classification, and discussion to apparent in different degrees of hypertrophy, upregulation of
recognize several broad categories as defined in the next three glial fibrillary acid protein (GFAP) and proliferation, much work
sections and as summarized in structural terms in Figure 51.1 remains to be done in characterizing differences in transcriptome
and Table 51.1. Representative examples of the continuous and proteome. Early studies in this direction show that stroke
gradations of reactive astrogliosis from mild to moderate to and lipopolysaccharide (LPS) mediate significantly different

A Healthy tissue
Cytoplasm &
cell membrane

Cytoskeleton

Figure 51.1 Changes Characteristic of Different Gradations of Reactive


B Mild to moderate reactive astrogliosis Astrogliosis. A. In healthy CNS tissue, the territories of astrocyte
Cytoplasm & processes have little or no overlap and many astrocytes do not express
cell membrane detectable levels of the prototypical astrocyte cytoskeletal protein, GFAP.
B. In mild to moderate reactive astrogliosis most (if not all) astrocytes
have upregulated expression of GFAP and exhibit hypertrophy of cyto-
Domain skeleton and stem processes, but with preservation of individual astrocyte
preservation domains and without pronounced overlap of astrocyte processes and
without cell proliferation. In addition there are variable changes in
Cytoskeletal
hypertrophy
molecular expression and associated functional activities. These changes
vary with insult severity and exhibit the potential for structural resolu-
tion if the triggering insult is removed or resolves. Such changes occur
C Severe diffuse reactive astrogliosis after mild metabolic or molecular insults, or mild infections or inflam-
matory activation, or after mild trauma, or at sites distant from a more
severe injury. C. Severe diffuse reactive astrogliosis is characterized by the
Process overlap appearance of newly proliferated astrocytes (with red nuclei in figure) in
with loss of addition to pronounced upregulation of GFAP expression with hypertro-
domains phy of cytoskeleton and stem processes. As a result there is considerable
intermingling of astrocyte processes and disruption of astrocyte domains,
Proliferated causing long lasting reorganization of tissue architecture. There are also
astrocytes substantial changes in molecular expression. Such changes are found in
areas surrounding severe focal lesions, infections or areas responding to
chronic neurodegenerative triggers. D. Severe reactive astrogliosis with
D Severe reactive astrogliosis with compact scar compact glial scar formation occurs along borders to areas of overt tissue
damage and inflammation after CNS trauma, stroke or infection. It is
Inflammatory characterized by newly proliferated astrocytes (with red nuclei in figure),
cell infiltrate particularly along the immediate borders to lesions, where scar forming
in tissue lesions astrocytes interact with other cell types such as fibroblasts and fibrocytes
to form a multicellular glial scar that also includes deposition of dense
collagenous extracellular matrix. In the immediate scar border, astrocytes
Fibroblasts
have densely overlapping processes. These astroglial scars function as
fibrocytes important barriers to inflammatory cells, infectious agents and non-CNS
etc cells in a manner that protects healthy tissue from nearby areas of intense
inflammation. In addition these scars act as barriers to axon regenera-
tion. Glial scar formation results in long lasting reorganization of tissue
Multicellular compact scar with barrier function architecture that appears to be long lasting or permanent. E. Perivascular
astrogliosis with scar formation is characterized by dense accumula-
E Perivascular astrogliosis with scar formation tions of the endfeet and processes of reactive astrocytes that align and
Perivascular scar intermingle around perivascular cuffs or clusters of inflammatory cell
with barrier function infiltrates. There are newly proliferated astrocytes (with red nuclei in
figure) as well as pronounced upregulation of GFAP expression with
Blood vessel hypertrophy of cytoskeleton and stem processes. These perivascular scars
form during certain CNS infections, in the vicinity of CNS traumatic
Perivascular cuff lesions and in autoimmune disorders. In a manner similar to astrocyte
of inflammatory scars that form around lesions after CNS trauma or stroke in (D), these
cell infiltrate perivascular astrocyte scars form important barriers that protect healthy
tissue by restricting the invasion of inflammatory cells from perivascular
clusters into CNS parenchyma.

A S T R O C Y T E R E S P O N S E S TO C E N T R A L N E RVO U S SYS T E M I N J U RY A N D D I S E A S E • 655


Table 51.3 EXAMPLES OF ASTROCYTE GENES POTENTIALLY INDUCED DURING REACTIVE ASTROGLIOSIS
FUNCTIONAL CATEGORIES EXAMPLES OF GENES

Cytokines IL6, IL1β, TNFα, INFγ, CNTF, LIF, TGF β, IL11, IL15,

Chemokines CXCL1, CXCL9, CXCL10, CCL2, CCL5, CCL7

Extracellular matrix MMP9, SEMA4A, TIMP1, chondroitin sulfate proteoglycans (various)

Growth factors and related BDNF, NGF, GDNF, Thsp1, GAP43, FGF2, PDGFb, BMP1, VEGFA

Astrocyte structural GFAP, VIM, NES

Small molecule producing enzymes NOS2, PTGES, SOD3


Intracellular signaling molecules STAT3, NFκB, SOC3, MAP-kinase, SOX9, Nrf2, Olig2, cAMP, Nurr1, SMAD, mTOR

For more details see references: Sofroniew 2009; Zamanian et al. 2012; Hamby et al. 2012.
Abbreviations as per nomenclature of Human Gene Compendium, GeneCards® www.genecards.org

changes in the astrocyte transcriptome in vivo (Zamanian et al. not be confused with the cell proliferation (astrocytosis) that
2012) and that different molecular stimulators of astrogliosis occurs in the more severe forms of reactive astrogliosis describe
mediate significantly different changes in the astrocyte below. Mild to moderate reactive astrogliosis is generally asso-
transcriptome in vitro (Hamby et al. 2012), as discussed in more ciated with mild nonpenetrating and noncontusive trauma, or
detail as folllows. with diffuse innate immune activation (viral infections, sys-
temic bacterial infections), or with areas that are some distance
to focal CNS lesions. It is noteworthy that in healthy tissue,
3.1 M I L D TO MO D E R AT E R E AC T I V E
the extensive network of finely branched processes of individ-
A S T RO G L I O S I S
ual astrocytes occupy contiguous, essentially nonoverlapping
Mild to moderate reactive astrogliosis consists of changes (up domains (see Fig. 51.1A) (Bushong et al. 2002) (see chapter 4),
or down) in gene expression that occur together with variable and that in mild to moderate reactive astrogliosis, there is
degrees of hypertrophy of cell body and stem processes, with- preservation of the individual nonoverlapping domains of
out loss of individual astrocyte domains and without astrocyte reactive astrocytes in spite of the hypertrophy of the cell body
proliferation (see Fig. 51.1B) (Sofroniew 2009; Sofroniew and and stem processes (see Fig. 51.1B) (Wilhelmsson et al. 2006).
Vinters 2010). There is increased expression of astrocyte struc- Experimental evidence suggests if the triggering mechanism
tural proteins such as the canonical astrocyte marker protein, is able to resolve, then mild or moderate reactive astrogliosis
GFAP. In healthy tissue, not all astrocytes express detectable exhibits the potential for resolution in which the astrocytes
levels of GFAP (see Fig. 51.1A), and so this increased GFAP return to a morphological appearance similar to that in healthy
expression can lead to increased detection of astrocytes in mild tissue, suggesting that there is little or no long-lasting reor-
to moderate reactive astrogliosis (see Fig. 51.1B) that should ganization of tissue architecture (Sofroniew 2009; Sofroniew
and Vinters 2010). The extent to which various molecular and
potential functional changes might resolve or persist is not well
known and this question warrants further investigation.

3.2 S EVE R E D I F F US E R E AC T I VE
A S T RO G L I O S I S
Severe diffuse reactive astrogliosis consists of changes (up or
down) in gene expression with pronounced up regulation of
GFAP expression, together with hypertrophy of cell body and
stem processes, as well as intermittent astrocyte proliferation
and loss of individual astrocyte domains such that there is sub-
stantive intermingling and overlapping of neighboring astro-
cyte processes (see Fig. 51.1C) (Oberheim et al. 2008; Sofroniew
Figure 51.2 Continuous Gradient of Reactive Astrogliosis from Mild to 2009; Sofroniew and Vinters 2010). These changes can result
Moderate to Scar Formation Adjacent to Tissue Lesions. Areas of severe in long lasting or permanent reorganization of tissue architec-
tissue damage after stroke, trauma, infection, or autoimmune inflamma- ture that can extend diffusely over substantive areas. This type
tory lesions become surrounded by compact glia scar. Spreading away
from the scar is a gradient of reactive astrogliosis that diminishes in
of response is generally found in areas that extend for some
severity with increasing distance from the lesion until there is a seamless distance away from severe focal lesions, or in areas responding
transition to healthy tissue. to chronic neurodegenerative triggers, or in response to certain

656 • NEUROGLIA
types of infection or seizures (Oberheim et al. 2008; Sofroniew striking differences in astrocyte changes along the continuum
and Vinters 2010). Because there can be considerable tissue of reactive astrogliosis, ranging from small modulations in
reorganization, the potential for resolution and return to nor- gene expression to compact scar formation, in response to
mal structure is reduced (see Fig. 51.1C) (Sofroniew 2009). insults of different kinds, are likely to be of consequence when
considering the functions and impact of reactive astrogliosis
on CNS functions, as discussed further.
3.3 S EVE R E R E AC T I V E A S T RO G L I O S I S WI T H
C O M PAC T G L I A L S C A R F O R M AT I O N
Severe reactive astrogliosis with compact glial scar forma- 4 S I G N A L I N G C A S C A D E S T H AT
tion consists of changes (up or down) in gene expression I N I T I AT E A N D R E GU L AT E R E AC T I VE
with pronounced upregulation of GFAP expression, together ASTROGLIOSIS
with hypertrophy of cell body and stem processes, as well as
pronounced astrocyte proliferation and the pronounced In agreement with the observations just described, which indi-
overlapping of astrocyte processes that interdigitate to form cate that reactive astrogliosis is not a single all-or-none phe-
compact borders that surround and demarcate areas of severe nomenon, available evidence also indicates that there is not a
tissue damage, necrosis, infection or autoimmune-triggered single genetic program that is simply turned on by different
inflammatory infiltration (see Fig. 51.1D) (Bush et al. 1999; stimuli and elicits all aspects of a stereotypic “reactive astroglio-
Drogemuller et al. 2008; Faulkner et al. 2004; Herrmann et al. sis” response. Instead, current evidence indicates that different
2008; Oberheim et al. 2008; Voskuhl et al. 2009). The borders aspects of reactive astrogliosis can be regulated separately and
formed by compact glial scars include other cell types, in partic- individually as called for in different situations by a wide vari-
ular meningeal and perivascular derived fibroblasts, fibrocytes, ety of different molecular mediators and signaling pathways
and other glial cells (Aldrich and Kielian 2011; Bundesen et al. (see Table 51.2) (Sofroniew 2009). Different molecular media-
2003; Herrmann et al. 2008; Reilkoff et al. 2011; Sofroniew tors that can influence astrocytes can be released in response
2009; Sofroniew and Vinters 2010). These multicellular glial to essentially all forms of CNS insults, ranging from subtle cel-
scars also deposit collagenous extracellular matrix that con- lular perturbations to intense tissue injury and cell death. Many
tains many molecular cues that inhibit axonal and cellular cell types can release molecular mediators of reactive astro-
migration (Silver and Miller 2004). It is important to note that gliosis including cells intrinsic to CNS tissue such as neurons,
although other cells are present in and around the multicellu- microglia, oligodendrocyte lineage cells, endothelia, pericytes,
lar glial scar, proliferating scar-forming astrocytes exert essen- fibromeningeal cells and other astrocytes, as well as nonneu-
tial scar organizing functions, such that scar formation does ral cells that gain entry into the CNS, such as bone marrow
not occur when astrocytes are dysfunctional (Bush et al. 1999; derived leukocytes, fibrocytes, and microbial infectious agents,
Faulkner et al. 2004; Herrmann et al. 2008). Triggering insults and in some cases cells that remain outside the CNS but release
include penetrating trauma, severe contusive trauma, invasive cytokines or toxins that can affect astrocytes. There is a broad
infections or abscess formation, neoplasm, chronic neurode- range of types of molecules that can trigger reactive astrogliosis
generation, systemically triggered inflammatory challenges. It including small molecules such as purines and transmitters, to
is noteworthy that glial scar formation is associated with sub- large polypeptide growth factors and cytokines (see Table 51.2)
stantive tissue reorganization and structural changes that are as reviewed recently (Sofroniew 2009; Sofroniew and Vinters
essentially permanent and persist even when triggering insults 2010). Some of these molecular mediators are released via spe-
may have resolved. An interesting and understudied variant cific signaling mechanisms, while others are released by simply
of astroglia scar formation is found surrounding perivascu- cell damage or cell death. Most of the extracellular molecu-
lar cuffs of leukocytes in CNS autoimmune disease and viral lar triggers of reactive astrogliosis have well defined recep-
infections, in which proliferated reactive astrocytes tightly tor targets that have the potential to initiate a wide variety of
surround infiltrating leukocytes that occupy the perivascular different intracellular signaling cascades that involve many dif-
space (see Fig. 51.1E) (Sofroniew and Vinters 2010; Voskuhl ferent second messenger systems as reviewed (Sofroniew 2009;
et al. 2009). Transgenic loss of function studies show that these Sofroniew and Vinters 2010). It is beyond the scope of this
reactive astrocytes exert barrier functions that limit the spread chapter to recapitulate the voluminous amount of information
of leukocytes and restrict them to the perivascular space, such available, but it is useful to consider certain basic examples that
that disruption of this barrier results in increased spread of point toward broad principles that appear to be emerging.
leukocytes and tissue damage in autoimmune conditions such Although we are in the early stages of dissecting the precise
as experimental autoimmune encephalitis (EAE) (Voskuhl et nature of molecular signaling routes between specific forms of
al. 2009). Thus, there appear to be interesting similarities in CNS damage or disease and specific aspects of reactive astroglio-
both structure and function between the glial scars that form sis, some evidence in this regard is available. A number of studies
around focal traumatic injuries and perivascular autoimmune from different laboratories have used transgenic mice with astro-
lesions. In both cases, disruption of the astrocyte scar leads to cyte specific gene deletions to examine roles of different compo-
widespread invasion of inflammatory cells into CNS paren- nents of signaling pathways involving STAT3, SOC3, or NFκB
chyma, indicative of essential barrier functions. in regulating reactive astrogliosis and its functions in differ-
Astrocyte contributions to glial scar formation can be ent models of CNS disorders. Briefly, deletion of STAT3 or its
regarded as the most extreme form of reactive astrogliosis. The associated membrane receptor, GP130, markedly attenuates

A S T R O C Y T E R E S P O N S E S TO C E N T R A L N E RVO U S SYS T E M I N J U RY A N D D I S E A S E • 657


certain aspects of reactive astrogliosis such as cell hypertrophy, et al. 2008). Another potential source might be the multipotent
upregulation of GFAP, and scar formation, and was associated progenitors in subependymal tissue that express GFAP (Garcia et
with increased spread of inflammation and infection, increased al. 2004) and generate progeny cells that migrate toward sites of
lesion size and demyelination, and impaired functional recov- injury after trauma or stroke (Ohab and Carmichael 2008), but
ery after spinal cord injury, autoimmune disease, or infection thus far no evidence has been presented that these particular pro-
(Drogemuller et al. 2008; Haroon et al. 2011; Herrmann et al. genitors give rise to newly generated scar-forming reactive astro-
2008; Okada et al. 2006). Deletion of SOC3 or NFκB also cytes. Additional studies will be required to establish the relative
attenuated certain aspects of reactive astrogliosis such as cell contributions of these potential sources.
hypertrophy and up regulation of GFAP, but was associated
with reduced inflammation and lesion size after spinal cord
injury or autoimmune disease (Brambilla et al. 2005, 2009; 5 E F F E C TO R M O L E C U L E S
Okada et al. 2006). Thus, different signaling pathways that can U P R E GU L AT E D A N D R E L E A S E D BY
trigger visible reactive astrogliosis in the form of cellular hyper- R E AC T I VE A S T R O C Y T E S
trophy and increased GFAP expression are involved in mediat-
ing either pro- or antiinflammatory effects that are associated Whole genome expression profiling is enabling the identifica-
with that reactive astrogliosis. tion of molecular changes associated with reactive astrogliosis
Findings such as those just described, combined with other in vivo (Zamanian et al. 2012) or in response to stimulation
available evidence, support a model in which there are many dif- with specific molecular mediators of reactive astrogliosis in vitro
ferent specific signaling mechanisms that trigger different specific (Hamby et al. 2012). Such studies are identifying a wide variety of
molecular, morphological, and functional changes in reactive potential effector molecules that may be produced and released
astrocytes. According to this model, not only will the responses by reactive astrocytes to influence other cell types, including
of reactive astrocytes vary in a context-dependent manner as many different cytokines, chemokines, growth factors, extra-
determined by different signals, but also the potential effects of cellular matrix molecules, and small signaling molecules, some
reactive astrogliosis on astrocyte functions will vary with different examples of which are given in Figure 51.3 and Table 51.3. It is
types of stimuli. In agreement with this model are recent findings also noteworthy that in vivo, different causes of reactive astro-
that different types of CNS insults, such as stroke or exposure to gliosis such as stroke and peripheral inoculation with LPS led
innate inflammatory mediators, have markedly different effects
on the genomic profiles of reactive astrocytes in vivo (Zamanian Microglia
et al. 2012), and that stimulation of astrocytes in vitro with dif- Synapse LIF
ferent inflammatory mediators also leads to broadly different NO LCN2 FGF2
changes in astrocyte genomic profiles (Hamby et al. 2012). This TNFα IL6
model also predicts that the impact of reactive astrogliosis on IL1β
its host neural tissue and associated neural functions will not be THBS1
Reactive Astrocyte
uniform or stereotypic, but will vary in a context-specific manner IL11
as determined by specific signaling mechanisms. BDNF CCL7
Specific molecular triggers of astrocyte proliferation have also
CXCL10
been identified. As discussed above, not all reactive astroglio- GDNF
sis induces cell proliferation, but in its most severe forms, reac- CCL2
tive astrogliosis can lead to the appearance of newly proliferated NGF CXCL1
astrocytes, particularly in scars that form borders around or along IGF1 VEGF-A
inflammatory cell infiltrates or tissue necrosis (see Fig. 51.1D,E). SEMA4A
GSNO wbc
Such astrocyte proliferation generally occurs in response to severe NO PGE
MMPs
tissue damage or inflammation after severe trauma, stroke, infec- Neuron CSPGs bv
tion, autoimmune responses, or degenerative disease (Drogemuller
et al. 2008; Faulkner et al. 2004; Voskuhl et al. 2009; White et
al.). The molecular triggers that lead to proliferation of reactive Figure 51.3 Examples of Effector Molecules Potentially Produced and
Released by Reactive Astrocytes in Response to CNS Insults. Reactive
astrocytes in vivo are incompletely characterized but include epi- astrocytes can be stimulated by different mediators to upregulate the
dermal growth factor (EGF), fibroblast growth factor (FGF), production of a wide variety of potential effector molecules that can
endothelin 1, and ATP (Gadea et al. 2008; Levison et al. 2000; influence different local cells in a paracrine fashion. Only a cross-section of
Neary and Zimmermann 2009). The source of newly divided molecular examples can be shown. The effector molecules include factors
scar-forming astrocytes is not well established, and there are sev- that can regulate blood flow or blood-brain barrier (NO, prostaglandin
E (PGE), VEGF-A); chemokines, cytokines, and growth factors that
eral potential possibilities. Perhaps the strongest evidence thus far can regulate white blood cells or microglia (CXCL1, CXCL10, CCL2,
is that mature astrocytes can reenter the cell cycle and proliferate CCL7, interleukin (IL)-1β, IL6, IL11, LIF, FGF2, tumor necrosis factor
during scar formation (Buffo et al. 2008; Bush et al. 1999; Gadea alpha (TNFα); growth factors and small molecules that can influence
et al. 2008). Evidence has also been presented that some prolifer- neurons and synapses, neurotrophic factor, glial cell-derived brain-derived
ating reactive astrocytes may derive from NG2 progenitor cells in neurotrophic factor (BDNF), nerve growth factor (NGF), insulin growth
factor (IGF)1, TNFα, thrombospondin (Thsp) 1, NO); as well as molecules
the local parenchyma (Magnus et al. 2008) or from ependymal that contribute to extracellular matrix (chondroitin sulfate proteoglycans
cell progenitors after injury or stroke (Carlen et al. 2009; Meletis (CSPG), matrix metallopeptidase 9 (MMP9), Semaphorin-4A (Sema4a)).

658 • NEUROGLIA
to significantly and markedly different changes in the astrocyte various kinds of essential beneficial effects after CNS insults. It is
transcriptome (Zamanian et al. 2012). In vitro, different media- also becoming clear that reactive astrocytes are not only hetero-
tors of reactive astrogliosis such as transforming growth factor geneous in their morphology as discussed above (see Fig. 51.1),
beta (TGF-β) or LPS plus interferon gamma (INF-γ) caused but also in the functions and effects that they exert in response
substantially and significantly different changes in the astrocyte to different molecular triggers released by different kinds of CNS
transcriptome, and combinatorial interactions among these dif- insults.
ferent mediators led to synergistic changes in gene expression
that could not be predicted simply by summing the effects of
7.1 R E AC T I VE A S T RO G L I O S I S ,
the individual mediators (Hamby et al. 2012). These findings
N EU RO P ROT EC T I O N A N D EFFEC TS O N
clearly demonstrate the heterogeneity and specificity of reactive
N EU RO NA L F U N C T I O N
astrogliosis under different conditions and as mediated by dif-
ferent molecular stimuli. Various different types of transgenic loss of function models
from multiple laboratories show that either ablation or attenua-
tion of reactive astrogliosis causes increased lesion size, increased
6 E F F E C T S O F R E AC T I VE A S T R O G L I O S I S neuronal loss, demyelination, and exacerbated loss of function
O N A S T R O C Y T E P H YS I O L O GY A N D O N after traumatic injury, stroke, autoimmune attack or infec-
F U N C T I O N S E X E RT E D BY A S T R O C Y T E S tion (Bush et al. 1999; Drogemuller et al. 2008; Faulkner et al.
I N T H E H E A LT H Y C E N T R A L N E RVO U S 2004; Herrmann et al. 2008; Li et al. 2008; Myer et al. 2006;
SYSTEM Okada et al. 2006; Voskuhl et al. 2009). A large number of
studies provide different kinds of evidence in vivo and in vitro
In comparison with the voluminous information accumulating that reactive astrocytes can protect CNS cells and tissue in
about morphological and genetic changes associated with reactive various ways, including by (1) uptake of potentially excitotoxic
astrogliosis, far less is known about the impact of reactive astro- glutamate (Bush et al. 1999; Rothstein et al. 1996; Swanson
gliosis on astrocyte physiology or on functions normally exerted et al. 2004), (2) protection from oxidative stress via glutathione
by astrocytes in the healthy CNS. As described elsewhere in this production (Chen et al. 2001; Shih et al. 2003; Swanson et al.
volume, astrocytes exert numerous essential functions in healthy 2004; Vargas et al. 2008), (3) protection via adenosine release
tissue including maintenance of extracellular ion homeostasis, (Lin et al. 2008), (4) protection from NH4+ toxicity (Rao
clearance of transmitters, provision of energy metabolites, regula- et al. 2005), (5) protection by degradation of amyloid-beta pep-
tion of blood flow, and interactions with synapses, and astrocytes tides (Koistinaho et al. 2004), and (6) stabilizing extracellular
exhibit ligand-evoked intracellular calcium ([Ca2+]i) increases that fluid and ion balance and reducing seizure threshold (Zador
are under intense investigation as potential means of mediating et al. 2009). In addition, recent findings indicate that reactive
dynamic astrocyte functions, including interactions with synapses astrogliosis can influence astrocyte functions in such a manner
and regulation of blood flow. The effects of reactive astrogliosis as to have effects on synaptic transmission. For example, down-
on these activities are not well defined and in some cases are only regulation of gamma-aminobutyric acid (GABA) transporter
beginning to be studied. One recent study reports that virally function by reactive astrocytes in periinfarct cortex was asso-
infected reactive astrocytes downregulated glutamine synthetase ciated with reduced cortical neuronal synaptic plasticity that
and that local neurons exhibited reduced inhibitory synaptic cur- could be reversed by inhibition of extrasynaptic GABAA recep-
rents (Ortinski et al. 2010). Other recent findings indicate that tor mediated currents (Clarkson et al. 2010). It can be speculated
exposure to inflammatory mediators alters astrocyte expression of that increasing GABA tone may initially be neuroprotective, but
a large number of G-protein coupled receptors and alters astro- later reduces capacity for plasticity and recovery. Together, these
cyte changes in [Ca2+]i evoked by specific ligands of these recep- findings indicate that reactive astrocytes can exert a range of
tors (Hamby et al. 2012). Together such findings point toward the functions that impact on neuroprotection, repair, and recovery
possibility of important effects of reactive astrogliosis on astrocyte in response to CNS insults of various kinds, including trauma,
physiology in ways that may impact on neural function, and sug- infection, stroke, and degenerative disease (Sofroniew 2005,
gest that this area warrants more extensive investigation. 2009; Sofroniew and Vinters 2010).

7.2 R E AC T I VE A S T RO C Y T E RO L E S I N B L O O D
7 F U N C T I O N S O F R E AC T I VE
B R A I N BA R R I E R B R E A K D OWN A N D R E PA I R ,
ASTROCY TES
A N D R EGU L AT I O N O F E D E M A
Glial scar formation is well known as an impediment to axon Many CNS insults result in leakiness of the blood-brain bar-
regeneration migration (see chapter 56). Perhaps for this reason, rier, including traumatic injury, stroke, inflammation and
reactive astrogliosis per se has sometimes been regarded as a purely certain forms of neurodegeneration, and in many cases, the
maladaptive obstacle to functional recovery after CNS damage blood-brain barrier (BBB) will repair spontaneously if the
and it has even been proposed that wholesale inhibition of reac- damaging insults resolve, such as in acute traumatic injury or
tive astrogliosis might be therapeutically beneficial. It is important stroke. Reactive astrocytes appear able to play critical roles
to emphasize that this is not the case. There are now many differ- in both breakdown and repair of BBB. Recent findings indi-
ent kinds of experimental evidence that reactive astrocytes exert cate that vascular endothelial growth factor-A (VEGF-A)

A S T R O C Y T E R E S P O N S E S TO C E N T R A L N E RVO U S SYS T E M I N J U RY A N D D I S E A S E • 659


derived from astrocytes mediates BBB breakdown and lympho- Although it may initially seem counterintuitive that astro-
cyte infiltration during autoimmune CNS inflammation (Argaw cytes could exert both pro- and antiinflammatory effects, we
et al. 2009, 2012). On the other hand, experimental loss of func- have recently proposed a functional model that reconciles this
tion studies demonstrate that astrocytes are essential for sponta- seeming paradox (Sofroniew 2009; Sofroniew and Vinters
neous posttraumatic repair of the blood-brain barrier (Bush et 2010). In this model, reactive scar-forming astrocytes function
al. 1999; Faulkner et al. 2004) even though astrocytes are not not only to attract and instruct inflammatory cells, in particular
essential for normal development of the blood- brain barrier at early times after insults, but also over time these astrocytes
(Daneman et al. 2010; Saunders et al. 2008) (see chapter 33). A form cell migration barriers that surround and demarcate areas
candidate molecule for astrocyte contribution to BBB repair is where intense inflammation is needed and that restrict the
S-nitrosoglutathione (GSNO), which is produced by astrocyte- spread of inflammatory cells and infectious agents in order to
like enteric glia and stimulates barrier properties in gut epithelia preserve nearby healthy tissue (Sofroniew 2005; Sofroniew
(Savidge et al. 2007). Astrocytes also play essential roles in reduc- 2009; Sofroniew and Vinters 2010) (see Fig 51.1D,E). This
ing vasogenic edema after trauma, stroke or obstructive hydro- model is congruent with the notion that evolutionary pressures
cephalus (Bush et al. 1999; Zador et al. 2009). shaping CNS injury responses are likely to have favored mecha-
nisms to keep small injuries small and uninfected, and that the
inhibition of the migration of inflammatory cells and infectious
agents and other cells might have led to the accidental byprod-
7.3 R E AC T I V E A S T RO G L I O S I S , S C A R
uct of inhibiting axon regeneration owing to the redundancy
F O R M AT I O N, A N D R E GU L AT I O N O F
between migration cues across cell types (Bush et al. 1999;
I N FL A M M AT I O N
Sofroniew 2005). In this context it is also interesting to consider
One of the essential functions exerted by reactive astrocytes the variant of astroglia scar found surrounding perivascular col-
appears to be that of formation of scars that create migration lections of leukocytes in CNS autoimmune disease and viral
barriers around collections of highly active inflammatory cell infections as discussed previously (see Fig. 51.1E) (Sofroniew
infiltrates that collect in areas of focal CNS lesions after trau- and Vinters 2010; Voskuhl et al. 2009). Recent findings show
matic injury, stroke, or suppurative infection, or in perivas- that the formation of such perivascular collections of leukocytes
cular clusters or cuffs during viral infections or autoimmune in the form of ectopic lymphoid follicles can be induced by the
disease (see Fig. 51.1D,E) (Sofroniew and Vinters 2010). As Th17 subtype of T cells (Peters et al. 2011). It is interesting to
described in this section, loss of function studies clearly show consider that interactions between T cells and astrocytes may
increased spread of inflammation and increased tissue damage be involved in the formation of such ectopic lymphoid follicles.
when astrocyte scar formation is experimentally attenuated. These follicles are surrounded by reactive scar-forming astro-
It has been recognized for some time that astrocytes in vitro cytes (see Fig. 51.1E) in both experimental animals and human
have the capacity to make many pro- or antiinflammatory mole- pathology (Sofroniew and Vinters 2010; Voskuhl et al. 2009)
cules in response to different kinds of stimulation (Eddleston and and fail to form when scar-forming astrocytes are experimentally
Mucke 1993; John et al. 2003; Sofroniew 2009), and can exert ablated in transgenic mice, resulting in the widespread spread
both pro- and antiinflammatory effects on microglia (Farina et al. of T cells in the neural parenchyma (Voskuhl et al. 2009). The
2007; Min et al. 2006). Recent studies from multiple laborato- nature and consequences of astrocyte–leukocyte interactions in
ries using transgenic loss of function models have extended such such follicles requires further study, but it is noteworthy that
observations and shown that reactive astrocytes in vivo can exert astrocytes produce high levels of diverse chemokines and cytok-
both pro- and antiinflammatory regulatory functions. For exam- ines in vivo and in vitro in response to innate inflammatory stim-
ple, ablation or attenuation of reactive scar-forming astrocytes can uli associated with injury and microbial infection (Hamby et al.
exacerbate the spread of inflammatory cells during (1) locally ini- 2012; John et al. 2003; Zamanian et al. 2012). It is interesting to
tiated innate inflammatory responses to traumatic injury (Bush speculate that such ectopic lymphoid follicles within the CNS
et al. 1999; Faulkner et al. 2004; Herrmann et al. 2008; Li may represent sites where astrocytes help to recruit and con-
et al. 2008; Okada et al. 2006), or (2) during peripherally ini- strain immune cells in sites (see Fig. 51.1E) where they may be
tiated adaptive immune responses such as EAE (Liedtke et al. instructed within the blood brain barrier by factors produced by
1998; Voskuhl et al. 2009). In this regard it is also noteworthy astrocytes and other cells, in such a manner that they may play
that attenuation of reactive astrocytes also leads to increased important roles in both adaptive situations such as viral infec-
spread of infection (Drogemuller et al. 2008). Nevertheless, tion and in maladaptive situations such as autoimmune disease.
reactive astrocytes also have proinflammatory potential, and
other studies provide evidence that deletion or knockdown of
certain molecules in astrocytes is associated with a reduction 8 R E AC T I VE A S T R O C Y T E S A N D
in inflammation after traumatic injury or experimental autoim- N O N - C E L L -AU TO N O M O U S
mune disease (Brambilla et al. 2005, 2009; Okada et al. 2006). N E U R O D E G E N E R AT I O N
Together these findings indicate that astrocyte involvement in
the regulation of CNS inflammation is complex and likely to The work summarized thus far in this chapter provides com-
be context dependent and regulated by multimodal extracellu- pelling evidence that reactive astrogliosis is a ubiquitous,
lar signaling events that may differ at different time points after complex, essential and beneficial part of the repair response
insults. to all CNS insults. Nevertheless, there is also evidence

660 • NEUROGLIA
that dysfunctions of astrocytes or reactive astrocytes can (7) contribution to chronic pain (Milligan and Watkins
contribute to, or to be a primary source of, CNS disease 2009). In these cases in which there is no obvious genetic
mechanisms (Sofroniew and Vinters 2010). In the healthy mutation, the gain of detrimental effects by reactive astrocytes
CNS, astrocytes exert many different essential functions, is incompletely understood but may result from specific sig-
as presented in other chapters of this volume. It is therefore naling cascades that result from complex interactions with
not surprising that there is a rapidly growing body of both other cell types such as microglia and other inflammatory cells
experimental and clinical evidence showing that genetic (Farina et al. 2007), or for example by simultaneous exposure
defects or molecular abnormalities within astrocytes or to microbial antigens such as lipopolysaccharide, which can
reactive astrocytes can precipitate non-cell-autonomous markedly increase the levels of potentially toxic nitric oxide
neuronal dysfunction or degeneration. These effects can produced by astrocytes in response to cytokines (Hamby
occur as a result of either gain of abnormal effects, or loss et al. 2006). Context-dependent acute or chronic combinato-
of normal functions, by astrocytes or reactive astrocytes. rial signaling events involving different cell types might thus
stimulate reactive astrocytes to produce potentially cytotoxic
levels of molecules or might lead to chronic inflammation or
8.1 G A I N O F D ET R I M EN TA L EFFEC TS BY
neuropathic pain. A greater understanding of such combina-
A S T RO C Y T E S O R R E AC T I V E A S T RO C Y T E S
torial signaling could facilitate targeted therapeutic strategies
There is evidence from both clinical and experimental stud- that preserve the beneficial and attenuate the potentially detri-
ies that under specific circumstances astrocytes or reactive mental aspects of reactive astrogliosis (Sofroniew 2009).
astrocytes have the potential to exert detrimental effects. In
some cases, there is a clear association with a genetic defect
8.2 L O S S O F E S S E N T I A L F U N C T I O NS BY
or molecular dysfunction. Clinical studies have identified at
A S T RO C Y T E S O R R E AC T I VE A S T RO C Y T E S
least two single gene mutations that cause gain of detrimental
effects by astrocytes and/or reactive astrocytes. In Alexander There are many potential ways in which loss of essential
disease, a dominant, gain-of-function mutation of the gene functions by astrocytes or reactive astrocytes might lead to
encoding GFAP is associated with changes in astrocytes that non-cell-autonomous neuronal dysfunction or degeneration.
resemble reactive astrogliosis, leukoencephalopathy, macroen- As summarized in many chapters in this volume, in healthy
cephalopathy, seizures, psychomotor disturbances, and prema- neural tissue, astrocytes play critical roles in many functions
ture death (Brenner et al. 2001) (see chapter 69). In a familial essential for neuronal health and function including homeo-
form of amyotrophic lateral sclerosis (ALS) or motor neuron stasis of extracellular fluid, ions and transmitters, regulation of
disease, a dominant gain-of-function mutation of the gene blood flow, energy provision and interactions with synapses.
encoding superoxide dismutase (SOD) leads to production by In addition, as summarized in this chapter, reactive astrocytes
reactive astrocytes of molecules that are toxic to motor neurons play critical roles in additional functions including neuropro-
(Di Giorgio et al. 2007; Nagai et al. 2007) (see chapter 63). In tection during various CNS insults, repair of the blood-brain
addition, gain of function transgenic mouse models indicate barrier, and regulation of inflammation. Although we are only
that selective targeting to astrocytes of a mutant form of the beginning to appreciate the specific mechanisms whereby
SOD associated with ALS leads to neuronal degeneration astrocyte dysfunction might impact on neural function, spe-
(Lobsiger and Cleveland 2007; Nagai et al. 2007; Yamanaka cific examples are becoming available from both experimental
et al. 2008). It is important to emphasize that in these examples studies and clinical observations.
of Alexander disease and familial ALS, the neuronal toxicity From an experimental perspective, there is now clear
and damage to neural tissue is caused by genetically mutated evidence that various single gene mutations that are tar-
and abnormal astrocytes rather than to the generic process of geted exclusively to astrocytes in transgenic mice can lead
reactive astrogliosis per se. In addition, there is also some evi- to loss of astrocyte functions that spontaneously precipitate
dence that the process of reactive astrogliosis might in some situ- non-cell-autonomous neuronal dysfunction and degeneration
ations exert detrimental effects, perhaps in an analogous fashion in the absence of additional CNS insults. For example, selec-
to the manner by which the process of inflammation can in tive deletion from astrocytes of the endoribonuclease, Dicer,
some situations exert detrimental effects. In this regard, there is leads to cell-non-autonomous neuronal degeneration of both
evidence from transgenic animals and other experimental mod- cerebellar granule neurons and Purkinje neurons (Tao et al.
els that reactive astrocytes may be stimulated by specific signal- 2011). Dicer-deficient mice exhibit normal motor develop-
ing cascades to gain detrimental effects such as (1) exacerbation ment and neurological morphology until mid-adolescence and
of inflammation via cytokine production (Brambilla et al. 2005, then invariably develop a fulminant neurological decline char-
2009), (2) production and release neurotoxic levels of reactive acterized by ataxia, severe progressive cerebellar degeneration,
oxygen species (Hamby et al. 2006; Swanson et al. 2004), (3) seizures, uncontrollable movements, and premature death.
release of potentially excitotoxic glutamate (Orellana et al. Concomitant with onset of symptoms, essential astrocytic
2009; Takano et al. 2005), (4) contribution to seizure genesis functions including glutamate uptake and antioxidant path-
(Jansen et al. 2005; Tian et al. 2005), (5) compromise of blood- ways are substantially impaired, leading to massive apoptosis
brain barrier function owing to VEGF-production (Argaw et al. of cerebellar granule cells and degeneration of Purkinje cells
2009), (6) cytotoxic edema during trauma and stroke through (Tao et al. 2011). Another example is that of the Wnt-signaling
aquaporin 4 (AQP4) overactivity (Zador et al. 2009), and pathway gene, adenomatous poliposis coli (APC), wherein

A S T R O C Y T E R E S P O N S E S TO C E N T R A L N E RVO U S SYS T E M I N J U RY A N D D I S E A S E • 661


deletion of APC selectively from astroglia leads to the grad- astrocytes. Differences in the molecular expression of dif-
ual but relentlessly progressive cell-non-autonomous neu- ferent forms of reactive astrocytes, and how such changes in
ronal degeneration of cerebellar Purkinje neurons (Wang et al. expression impact on essential functions normally exerted by
2011). APC-deficient mice develop morphologically normal astrocytes in healthy tissue, may have important ramifications
Bergmann astroglia during the juvenile period, but then their for outcome after different types of CNS insults. Additional
radial fibers become shortened, their cell bodies translocate to work is needed to address these interesting issues.
the molecular layer, and they lose their pial contact and trans- From a clinical perspective, the disorder neuromyelitis
form into stellate-shaped cells reminiscent of reactive astro- optica (NMO), a relapsing inflammatory demyelinating dis-
cytes. Purkinje neurons are normal in appearance throughout ease (see chapter 61), provides evidence that dysfunction or
the juvenile period and then begin gradually to degenerate loss of astrocytes can exacerbate CNS inflammation in a clini-
with a gradual diminution of the dendritic tree and eventual cally relevant context. Neuromyelitis optica is causally associ-
cell death such that there is significant loss of Purkinje neurons ated with autoantibodies that bind selectively to AQP4 on
and cerebellar atrophy by middle age. Granule neurons are not astrocytes (Lennon et al. 2005) and precipitate compliment-
visibly affected. It is also well established that genetic deletion mediated astrocyte lysis that is associated with pronounced
of the astrocyte glutamate uptake transporter, Glt1 (EAAT2) inflammation and tissue destruction (Lucchinetti et al. 2002;
will lead to seizures and excitotoxic neuronal death (Rothstein Roemer et al. 2007) and severe symptoms such as vision loss
et al. 1996) (see chapter 63). As regards reactive astrocytes, and paralysis (Wingerchuk et al. 1999). The neuropathologi-
there is evidence that single gene mutations targeted exclu- cal changes found in NMO appear in many ways to be simi-
sively to astrocytes can lead to loss of essential functions by lar to changes found in transgenic mouse models in which
reactive astrocytes that result in increased neuronal dysfunc- ablation of reactive scar-forming astrocytes results in greatly
tion and degeneration in the context of experimental models exacerbated inflammation and tissue destruction during CNS
of CNS injury or disease. For example, deletion of the signaling autoimmune inflammation (Voskuhl et al. 2009).
molecule STAT3 or the cytokine receptor GP130 can impair
function and increase tissue damage and neurodegeneration
in experimental models of CNS trauma, infection, and auto- 9 S U M M A RY A N D P E R S P E C T I VE S
immune disease (Drogemuller et al. 2008; Haroon et al. 2011;
Herrmann et al. 2008; Myer et al. 2006; Okada et al. 2006). In Astrocytes have been known to respond to CNS injury and
addition, as discussed above, the transgenically targeted selec- disease for well over 100 years. This response has often been
tive ablation of proliferating reactive astrocytes will cause the regarded as a stereotypic all-or-none phenomenon that uni-
failure of blood-brain barrier repair, increase inflammation, formly results in glial scar formation. Recent progress dem-
and exacerbate loss of function and neurodegeneration in var- onstrates that this is not the case and that instead, reactive
ious models of CNS trauma or autoimmune disease (Bush et astrogliosis is a finely gradated continuum of changes that
al. 1999; Faulkner et al. 2004; Myer et al. 2006; Voskuhl et al. range from subtle alterations in gene expression and cell
2009), indicating that the consequence of loss of astrocytes hypertrophy that are reversible, to scar formation with per-
because of injury or disease leads to devastating consequences manent tissue rearrangement. It has become clear that there
and the comprehensive degeneration of neural tissue. is not a single program for reactive astrogliosis with a simple
Another important and interesting question is the degree on/off switch, but that the different potential structural and
to which the process of reactive astrogliosis might in itself in functional changes associated with reactive astrogliosis that
some situations lead to a reduction or loss of essential func- can occur are regulated separately by many different poten-
tions exerted by astrocytes in healthy tissue. Some experimen- tial signaling mechanisms. For example, different signaling
tal evidence is already available in this regard. For example, events have been identified that separately regulate expression
recent findings indicate that reactive astrogliosis can influence of structural proteins, cell hypertrophy or cell division. Other
astrocyte functions that may impact on synaptic transmission. signaling mechanisms separately regulate expression of pro- or
Downregulation of GABA transporter function by reactive antiinflammatory molecules. In addition, although it is clear
astrocytes in periinfarct cortex was found to be associated that reactive scar forming astrocytes are associated with the
with reduced cortical neuronal synaptic plasticity and inhibi- inhibition of axon regrowth, a number of beneficial functions
tion of extrasynaptic GABAA currents was found to benefit have been identified that are mediated by reactive astrocytes,
recovery (Clarkson et al. 2010). Reactive astrogliosis associ- including neuroprotection, blood brain barrier repair, and
ated with viral infection reduced astrocyte glutamine synthe- the restriction of inflammation. Dysfunctions of astrocytes
sis and altered hippocampal neuronal excitability (Ortinski et and reactive astrocytes can also occur and contribute to CNS
al. 2010). In addition, after traumatic spinal cord injury, the disorders, either through gain of abnormal effects or loss of
majority of proliferating astrocytes were found to express low normal functions. Combinations of mouse models of trans-
or no detectable levels of a transgene reporting expression lev- genic astrocyte manipulations with experimental models of
els of the major astrocyte glutamate transporter, Glt1, which CNS injury or disease show that genetic modulations of reac-
may influence the ability of the injured tissue to cope with tive astrogliosis and scar formation can markedly alter tissue
potentially excitotoxic glutamate load (Lepore et al. 2011). As repair, disease progression and functional outcome. Ablation
discussed above, proliferating astrocytes are characteristic of of astrocytes and attenuation of certain astrocyte functions
scar formation and are intermingled with non-proliferating exacerbate disease progression and tissue degeneration and

662 • NEUROGLIA
worsen functional outcome, whereas deletion of certain astro- neurite outgrowth after ablation of scar-forming, reactive astrocytes
cyte genes appears to improve outcome in some situations. in adult transgenic mice. Neuron 23:297–308.
Bushong EA, Martone MA, Jones YZ, Ellisman MH. 2002. Protoplasmic
Collectively such findings point toward an enormous, yet astrocytes in CA1 atratum radiatum occupy separate anatomical
incompletely understood, potential for astrocytes to contrib- domains. J Neurosci 22:183–192.
ute to, or play primary roles in, disease processes, tissue repair, Carlen M, Meletis K, Goritz C, Darsalia V, Evergren E, Tanigaki K, et al.
and functional outcome in a wide variety of clinical condi- 2009. Forebrain ependymal cells are Notch-dependent and generate
tions including stroke, epilepsy, and neurodegenerative disease neuroblasts and astrocytes after stroke. Nat Neurosci 12(3):259–267.
Chen Y, Vartiainen NE, Ying W, Chan PH, Koistinaho J, Swanson
(Sofroniew and Vinters 2010). In this context it is noteworthy RA. 2001. Astrocytes protect neurons from nitric oxide toxicity by a
that genetic polymorphisms that impact selectively on astro- glutathione-dependent mechanism. J Neurochem 77(6):1601–1610.
cyte functions are likely to become of interest in clinical neu- Clarkson AN, Huang BS, Macisaac SE, Mody I, Carmichael ST. 2010.
roscience. Future studies are needed to identify specific factors Reducing excessive GABA-mediated tonic inhibition promotes func-
that might alter or perturb the normal process of reactive tional recovery after stroke. Nature 468(7321):305–309.
Daneman R, Zhou L, Kebede AA, Barres BA. 2010. Pericytes are
astrogliosis through different means such as (1) genetic muta- required for blood-brain barrier integrity during embryogenesis.
tions or polymorphisms, (2) autoimmune events, or (3) simul- Nature 468(7323):562–566.
taneous exposure to multiple stimulators that might interact Di Giorgio FP, Carrasco MA, Siao MC, Maniatis T, Eggan K. 2007.
synergistically. Non-cell autonomous effect of glia on motor neurons in an embry-
onic stem cell-based ALS model. Nat Neurosci 10(5):608–614.
Drogemuller K, Helmuth U, Brunn A, Sakowicz-Burkiewicz M, Gutmann
AC K N OW L E D G M E N T S DH, Mueller W, et al. 2008. Astrocyte gp130 expression is critical for the
control of Toxoplasma encephalitis. J Immunol 181(4):2683–2693.
Eddleston M, Mucke L. 1993. Molecular profi le of reactive astrocytes—
Work in the author’s laboratory is supported by grants from implications for their role in neurological disease. Neuroscience
the National Institutes of Health (RO1 NS057624), Roman 54:15–36.
Reed Spinal Cord Injury Research Fund, Adelson Medical Farina C, Aloisi F, Meinl E. 2007. Astrocytes are active players in cere-
Research Foundation, National Multiple Sclerosis Society, bral innate immunity. Trends Immunol 28:138–145.
Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew
and Wings for Life. MV. 2004. Reactive astrocytes protect tissue and preserve function
after spinal cord injury. J Neurosci 24:2143–2155.
Gadea A, Schinelli S, Gallo V. 2008. Endothelin-1 regulates astrocyte
REFERENCES proliferation and reactive gliosis via a JNK/c-Jun signaling pathway.
J Neurosci 28(10):2394–2408.
Aldrich A, Kielian T. 2011. Central nervous system fibrosis is associated Garcia ADR, Doan NB, Imura T, Bush TG, Sofroniew MV. 2004.
with fibrocyte-like infi ltrates. Am J Pathol 179(6):2952–2962. GFAP-expressing progenitors are the principle source of consti-
Argaw AT, Asp L, Zang J, Navrazhina K, Pham T, Mariani JN, et al. 2012. tutive neurogenesis in adult mouse forebrain. Nature Neurosci
Astrocyte-derived VEGF-A drives blood-brain barrier breakdown in 7:1233–1241.
CNS inflammatory disease. J Clin Invest 122:2454–2468. Halassa MM, Haydon PG. 2010. Integrated brain circuits: astrocytic
ArgawAT,GurfeinBT,ZhangY,ZameerA,JohnGR.2009.VEGF-mediated networks modulate neuronal activity and behavior. Annu Rev Physiol
disruption of endothelial CLN-5 promotes blood-brain barrier break- 72:335–355.
down. Proc Natl Acad Sci USA 106(6):1977–1982. Hamby ME, Coppola G, Geschwind DH, Khakh BS, Sofroniew MV.
Attwell D, Buchan AM, Charpak S, Lauritzen M, Macvicar BA, 2012. Inflammatory mediators alter astrocyte genomic profi les
Newman EA. 2010. Glial and neuronal control of brain blood flow. and G-protein receptor evoked calcium excitability. submitted for
Nature 468(7321):232–243. publication.
Barres BA. 2008. The mystery and magic of glia: a perspective on their Hamby ME, Hewett JA, Hewett SJ. 2006. TGF-beta1 potentiates astro-
roles in health and disease. Neuron 60(3):430–440. cytic nitric oxide production by expanding the population of astro-
Brambilla R, Bracchi-Ricard V, Hu WH, Frydel B, Bramwell A, cytes that express NOS-2. Glia 54(6):566–577.
Karmally S, et al. 2005. Inhibition of astroglial nuclear factor kappaB Haroon F, Drogemuller K, Handel U, Brunn A, Reinhold D, Nishanth
reduces inflammation and improves functional recovery after spinal G, et al. 2011. Gp130-dependent astrocytic survival is critical for
cord injury. J Exp Med 202:145–156. the control of autoimmune central nervous system inflammation.
Brambilla R, Persaud T, Hu X, Karmally S, Shestopalov VI, J Immunol 186(11):6521–6531.
Dvoriantchikova G, et al. 2009. Transgenic inhibition of astroglial Herrmann JE, Imura T, Song B, Qi J, Ao Y, Nguyen TK, et al. 2008.
NF-kappaB improves functional outcome in experimental autoim- STAT3 is a critical regulator of astrogliosis and scar formation after
mune encephalomyelitis by suppressing chronic central nervous system spinal cord injury. J Neurosci 28(28):7231–7243.
inflammation. J Immunol 182(5):2628–2640. Jansen LA, Uhlmann EJ, Crino PB, Gutmann DH, Wong M. 2005.
Brenner M, Johnson AB, Boespflug-Tanguy O, Rodriguez D, Goldman JE, Epileptogenesis and reduced inward rectifier potassium current in
Messing A. 2001. Mutations in GFAP, encoding glial fibrillary tuberous sclerosis complex-1-deficient astrocytes. Epilepsia 46(12):
acidic protein, are associated with Alexander disease. Nat Genet 1871–1880.
27(1):117–120. John GR, Lee SC, Brosnan CF. 2003. Cytokines: Powerful regulators of
Buffo A, Rite I, Tripathi P, Lepier A, Colak D, Horn AP, et al. 2008. glial cell activation. Neuroscientist 9:10–22.
Origin and progeny of reactive gliosis: a source of multipotent cells in Koistinaho M, Lin S, Wu X, Esterman M, Koger D, Hanson J, et al. 2004.
the injured brain. Proc Natl Acad Sci USA 105(9):3581–3586. Apolipoprotein E promotes astrocyte colocalization and degradation
Bundesen LQ, Scheel TA, Bregman BS, Kromer LF. 2003. Ephrin-B2 of deposited amyloid-beta peptides. Nat Med 10(7):719–726.
and EphB2 regulation of astrocyte-meningeal fibroblast interac- Lennon VA, Kryzer TJ, Pittock SJ, Verkman AS, Hinson SR. 2005. IgG
tions in response to spinal cord lesions in adult rats. J Neurosci marker of optic-spinal multiple sclerosis binds to the aquaporin-4
23(21):7789–7800. water channel. J Exp Med 202:473–477.
Bush TG, Puvanachandra N, Horner CH, Polito A, Ostenfeld T, Svendsen Lepore AC, O’Donnell J, Bonner JF, Paul C, Miller ME, Rauck B,
CN, et al. 1999. Leukocyte infi ltration, neuronal degeneration and et al. 2011. Spatial and temporal changes in promoter activity of the

A S T R O C Y T E R E S P O N S E S TO C E N T R A L N E RVO U S SYS T E M I N J U RY A N D D I S E A S E • 663


astrocyte glutamate transporter GLT1 following traumatic spinal Roemer SF, Parisi JE, Lennon VA, Benarroch EE, Lassmann H, Bruck W,
cord injury. J Neurosci Res 89(7):1001–1017. et al. 2007. Pattern-specific loss of aquaporin-4 immunoreactivity dis-
Levison SW, Jiang FJ, Stoltzfus OK, Ducceschi MH. 2000. IL-6-type tinguishes neuromyelitis optica from multiple sclerosis. Brain 130(Pt
cytokines enhance epidermal growth factor-stimulated astrocyte prolif- 5):1194–1205.
eration. Glia 32:328–337. Rothstein JD, Dykes-Hoberg M, Pardo CA, Bristol LA, Jin L, Kuncl RW,
Li L, Lundkvist A, Andersson D, Wilhelmsson U, Nagai N, Pardo AC, et al. 1996. Knockout of glutamate transporters reveals a major role for
et al. 2008. Protective role of reactive astrocytes in brain ischemia. astroglial transport in excitotoxicity and clearance of glutamate. Neuron
J Cereb Blood Flow Metab 28(3):468–481. 16(3):675–686.
Liedtke W, Edelmann W, Chiu FC, Kucherlapati R, Raine CS. 1998. Saunders NR, Ek CJ, Habgood MD, Dziegielewska KM. 2008. Barriers in
Experimental autoimmune encephalomyelitis in mice lacking glial the brain: a renaissance? Trends Neurosci 31(6):279–286.
fibrillary acidic protein is characterized by a more severe clinical Savidge TC, Newman P, Pothoulakis C, Ruhl A, Neunlist M, Bourreille A,
course and an infiltrative central nervous system lesion. Am J Pathol et al. 2007. Enteric glia regulate intestinal barrier function and
152(1):251–259. inflammation via release of S-nitrosoglutathione. Gastroenterology
Lin JH, Lou N, Kang N, Takano T, Hu F, Han X, et al. 2008. A cen- 132(4):1344–1358.
tral role of connexin 43 in hypoxic preconditioning. J Neurosci Shih AY, Johnson DA, Wong G, Kraft AD, Jiang L, Erb H, et al. 2003.
28(3):681–695. Coordinate regulation of glutathione biosynthesis and release by
Lobsiger CS, Cleveland DW. 2007. Glial cells as intrinsic components Nrf2-expressing glia potently protects neurons from oxidative stress.
of non-cell-autonomous neurodegenerative disease. Nat Neurosci J Neurosci 23(8):3394–3406.
10(11):1355–1360. Silver J, Miller JH. 2004. Regeneration beyond the glial scar. Nature Rev
Lucchinetti CF, Mandler RN, McGavern D, Bruck W, Gleich G, Ransohoff Neurosci 5:146–156.
RM, et al. 2002. A role for humoral mechanisms in the pathogenesis of Sofroniew MV. 2005. Reactive astrocytes in neural repair and protection.
Devic’s neuromyelitis optica. Brain 125(Pt 7):1450–1461. Neuroscientist 5:400–407.
Magnus T, Carmen J, Deleon J, Xue H, Pardo AC, Lepore AC, et al. 2008. Sofroniew MV. 2009. Molecular dissection of reactive astrogliosis and glial
Adult glial precursor proliferation in mutant SOD1G93A mice. Glia scar formation. Trends Neurosci 32(12):638–647.
56(2):200–208. Sofroniew MV, Vinters HV. 2010. Astrocytes: biology and pathology. Acta
Meletis K, Barnabe-Heider F, Carlen M, Evergren E, Tomilin N, Shupliakov Neuropathol 119(1):7–35.
O, et al. 2008. Spinal cord injury reveals multilineage differentiation of Swanson RA, Ying W, Kauppinen TM. 2004. Astrocyte influences on isch-
ependymal cells. PLoS Biol 6(7):e182. emic neuronal death. Curr Mol Med 4(2):193–205.
Milligan ED, Watkins LR. 2009. Pathological and protective roles of glia in Takano T, Kang J, Jaiswal JK, Simon SM, Lin JH, Yu Y, et al. 2005.
chronic pain. Nat Rev Neurosci 10(1):23–36. Receptor-mediated glutamate release from volume sensitive channels in
Min KJ, Yang MS, Kim SU, Jou I, Joe EH. 2006. Astrocytes induce astrocytes. Proc Natl Acad Sci U S A 102(45):16466–16471.
hemeoxygenase-1 expression in microglia: a feasible mechanism for Tao J, Wu H, Lin Q, Wei W, Lu XH, Cantle JP, et al. 2011. Deletion of
preventing excessive brain inflammation. J Neurosci 26:1880–1887. astroglial dicer causes non-cell-autonomous neuronal dysfunction and
Myer DJ, Gurkoff GG, Lee SM, Hovda DA, Sofroniew MV. 2006. Essential degeneration. J Neurosci 31(22):8306–8319.
protective roles of reactive astrocytes in traumatic brain injury. Brain Tian GF, Azmi H, Takano T, Xu Q, Peng W, Lin J, et al. 2005. An astro-
129(10):2761–2772. cytic basis of epilepsy. Nat Med 11(9):973–981.
Nagai M, Re DB, Nagata T, Chalazonitis A, Jessell TM, Wichterle H, Vargas MR, Johnson DA, Sirkis DW, Messing A, Johnson JA. 2008.
et al. 2007. Astrocytes expressing ALS-linked mutated SOD1 Nrf2 activation in astrocytes protects against neurodegeneration in
release factors selectively toxic to motor neurons. Nat Neurosci mouse models of familial amyotrophic lateral sclerosis. J Neurosci
10(5):615–622. 28(50):13574–13581.
Neary JT, Zimmermann H. 2009. Trophic functions of nucleotides in the Voskuhl RR, Peterson RS, Song B, Ao Y, Morales LB, Tiwari-Woodruff
central nervous system. Trends Neurosci 32(4):189–198. S, et al. 2009. Reactive astrocytes form scar-like perivascular barri-
Oberheim NA, Tian GF, Han X, Peng W, Takano T, Ransom B, et al. ers to leukocytes during adaptive immune inflammation of the CNS.
2008. Loss of astrocytic domain organization in the epileptic brain. J Neurosci 29(37):11511–11522.
J Neurosci 28(13):3264–3276. Wang X, Imura T, Sofroniew MV, Fushiki S. 2011. Loss of adenoma-
Ohab JJ, Carmichael ST. 2008. Poststroke neurogenesis: emerging princi- tous polyposis coli in Bergmann glia disrupts their unique archi-
ples of migration and localization of immature neurons. Neuroscientist tecture and leads to cell nonautonomous neurodegeneration of
14(4):369–380. cerebellar Purkinje neurons. Glia 59(6):857–868.
Okada S, Nakamura M, Katoh H, Miyao T, Shimazaki T, Ishii K, White RE, McTigue DM, Jakeman LB. 2010. Regional heterogeneity in
et al. 2006. Conditional ablation of Stat3 or Socs3 discloses a dual astrocyte responses following contusive spinal cord injury in mice.
role for reactive astrocytes after spinal cord injury. Nature Med J Comp Neurol 518(8):1370–1390.
12(7):829–834. Wilhelmsson U, Bushong EA, Price DL, Smarr BL, Phung V, Terada
Orellana JA, Saez PJ, Shoji KF, Schalper KA, Palacios-Prado N, Velarde V, M, et al. 2006. Redefi ning the concept of reactive astrocytes as
et al. 2009. Modulation of brain hemichannels and gap junction chan- cells that remain within their unique domains upon reaction to
nels by pro-inflammatory agents and their possible role in neurodegen- injury. Proc Natl Acad Sci U S A 103(46):17513–17518.
eration. Antioxid Redox Signal 11(2):369–399. Wingerchuk DM, Hogancamp WF, O’Brien PC, Weinshenker BG.
Ortinski PI, Dong J, Mungenast A, Yue C, Takano H, Watson DJ, 1999. The clinical course of neuromyelitis optica (Devic’s syn-
et al. 2010. Selective induction of astrocytic gliosis generates deficits in drome). Neurology 53(5):1107–1114.
neuronal inhibition. Nat Neurosci 13(5):584–591. Yamanaka K, Chun SJ, Boillee S, Fujimori-Tonou N, Yamashita H,
Peters A, Pitcher LA, Sullivan JM, Mitsdoerffer M, Acton SE, Franz B, et Gutmann DH, et al. 2008. Astrocytes as determinants of dis-
al. 2011. Th17 cells induce ectopic lymphoid follicles in central nervous ease progression in inherited amyotrophic lateral sclerosis. Nat
system tissue inflammation. Immunity 35(6):986–996. Neurosci 11(3):251–253.
Rao KV, Panickar KS, Jayakumar AR, Norenberg MD. 2005. Zador Z, Stiver S, Wang V, Manley GT. 2009. Role of aquaporin-4 in cere-
Astrocytes protect neurons from ammonia toxicity. Neurochem Res bral edema and stroke. Handb Exp Pharmacol 190:159–170.
30(10):1311–1318. Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, Giffard RG, et
Reilkoff RA, Bucala R, Herzog EL. 2011. Fibrocytes: emerging effector al. 2012. Genomic analysis of reactive astrogliosis. J Neurosci
cells in chronic inflammation. Nat Rev Immunol 11(6):427–435. 32:6391–6410.

664 • NEUROGLIA
52.
METAB OLIC INJURY OF
OLIGODENDROCY TES AND MYELIN
Peter K. Stys

A B B R E VI AT I O N S therefore possibly of limited effectiveness. This chapter will


summarize current knowledge of the molecular pathways,
AMPA α-amino-3-hydroxy-5-methyl-isoxazoIe-4- with an emphasis on Ca2+ dependent mechanisms, that may
propionate be involved in injury to the OL, its processes, and the vital
ADP adenosine diphosphate myelin sheath that plays such an important role in supporting
ASIC acid-sensing ion channel information transmission in the mammalian nervous system.
ATP adenosine triphosphate
CNS central nervous system
EAAT excitatory amino acid transporter 2 O L I G O D E N D R O C Y T E S T RU C T U R E ,
ER endoplasmic reticulum F U N C T I O N, A N D E N E R GY M ETA B O L I S M :
GluT glutamate transporter A B R I E F O VE RVI EW
GlyT glycine transporter
MS multiple sclerosis A detailed review of the development, structure, and function
NMDA N-methyl-d-aspartate of OLs, as well as metabolic pathways of energy production,
NO nitric oxide is presented elsewhere in this volume (see chapters 13, 20,
OL oligodendrocyte and 27). This section serves as a brief overview to put into
PNS peripheral nervous system context the demands placed on these glia, and as a result, their
TNFα tumor necrosis factor α predilection to injury. As OLs mature from precursors in the
developing CNS, their main role is to synthesize myelin, and
ensheath axons, thus tremendously improving conduction
1 INTRODUCTION velocity. This in turn allows axons to retain a much smaller
diameter, and thus greatly increases the packing density in
Oligodendrocytes (OLs) are one of the major glia of the cen- the CNS. This appears to be the main objective of myelina-
tral nervous system (CNS), subserving the important function tion, because the actual net energy saving may not be as great
of synthesizing myelin for ensheathment of axons in the brain as once assumed (Harris & Attwell 2012). Indeed, the syn-
and spinal cord. It is estimated that more than 95% of cal- thetic demands placed on myelinating OLs seem extreme:
losal axons in the human are myelinated (Aboitiz et al. 1992); each OL may synthesize myelin membrane whose total area
therefore, injury and death of oligodendrocytes have the pro- exceeds that of the parent soma by 10,000-fold, generating
pensity to cause major disruption both to the function and as much as 5,000 μm2 of membrane per day during the active
even long-term survival of vital axonal connections within the myelinating phase (Pfeiffer et al. 1993), placing an extreme
CNS. Numerous clinical disorders result in injury and death energy demand on these glia (Harris & Attwell 2012).
of OLs, and ultimately demyelination of axons. These include To fuel its energy needs, the OL possesses all the standard
trauma (brain and spinal cord injury), ischemia (acute stroke metabolic machinery and organelles (i.e., mitochondria) nec-
and more chronic vascular insufficiency leading to dementia), essary for glycolytic and oxidative adenosine triphosphate
infections, toxins, metabolic abnormalities, genetic diseases (ATP) synthesis. Oligodendrocytes express glucose transport-
resulting in leukodystrophy, and degenerative/inflammatory ers (Arluison et al. 2004), indicating that these glia can utilize
disorders whose causes are unknown such as Alzheimer’s glucose directly. However, glucose is not the only substrate that
disease and multiple sclerosis (for a more extensive list, see can support OL energy requirement: lactate can also be taken
Schmahmann et al. 2008). These conditions share, to a greater up by these cells, which they use for energy, lipid metabolism,
or lesser extent, pathology centered on the oligodendrocyte and myelin synthesis (Rinholm et al. 2011). Oligodendrocytes
and the myelin sheath, in addition to involving other elements are electrically polarized, with an estimated resting potential of
of the CNS. Yet we know comparatively little about the cellu- about −65 mV (Karadottir et al. 2005), and as with most elec-
lar and molecular pathophysiology of oligodendrocyte injury trically polarized cells, OLs must expend energy to maintain
and death, and as a result, any attempts at devising effective physiological transmembrane ionic gradients. They express
therapeutic interventions will be perforce incomplete, and all the major cation and anion transporters found in other

665
excitable and electrically polarized cells, including Na+/K+- chapter; the reader is referred elsewhere for details (Buntinx et al.
ATPase, Ca2+-ATPase, Na+ Ca2+ exchanger, a variety of anion 2002) (see chapter 50). Voltage- and store-operated Ca2+ chan-
transport proteins, and intracellular Ca2+ release channels nels have also been detected on OLs, and are likely involved in
(Ruiz et al. 2010; Mata & Fink 1989; reviewed in chapter 20). development and directional process growth (Butt 2006); their
All of these consume energy directly or indirectly, and many are role in OL injury is less established. Perhaps the best studied are
directly or indirectly involved in maintaining control over the neurotransmitter-gated channels, many of which are capable of
most important ion involved in cell signaling and death: cal- permeating damaging amounts of Ca2+ ions in a wide variety of
cium. Thus, an imbalance between Ca2+ entry into the cytosol pathological conditions.
(from extra- and/or intracellular sources) versus the cell’s abil- The effectors of cell destruction in the face of a pathologi-
ity to buffer this key cation, most often because of impairment cal intracellular Ca2+ rise are likely a collection of Ca2+ activated
of energy supply, leads to injury and death. This “final common enzyme systems normally tightly regulated and essential for
pathway” of cell injury (Schanne et al. 1979) applies equally to growth, repair and remodeling. The calpains are a family of Ca2+
OLs, and much of the remainder of this chapter will focus on activated proteases that have as substrates numerous OL and
mechanisms and conditions that lead to Ca2+ dyshomeostasis myelin proteins, ideally positioning these enzymes to wreak major
in OLs, and equally importantly, in the myelin sheath, that havoc if excessively activated (Vosler et al. 2008). Several phos-
responds uniquely to pathological stimuli independently of its pholipases, both Ca2+-dependent and independent, will in turn
parent cell body. degrade critical myelin lipids, which make up 70% of the content
of myelin. Peptidylarginine deiminase 2 is another Ca2+-activated
enzyme present in myelin that converts arginine residues in myelin
3 C A L C I U M O VE R L OA D I N J U R E S basic protein to citrulline. The alteration in net charge on this key
OLIGODENDROCY TES AND MYELIN structural protein may lead to destabilization of myelin, poten-
tially rendering it more susceptible to breakdown or immune
Perhaps the earliest compelling data on the importance of Ca2+ attack (Harauz & Musse 2007). Taken together, abnormal Ca2+
ions in degeneration of myelin and injury of OLs stemmed accumulation inside the OL soma, its processes, and myelin can
from experiments where Ca2+ ionophores were applied to in turn unleash a wide variety of nefarious biochemical pathways
myelinated fibers in the PNS and CNS, or to cultured OLs. that can severely damage or destroy the myelinating unit.
The latter suffered injury at ionophore concentrations that did
not affect other cell types (e.g., astrocytes), and myelin under-
went severe vesiculation (Scolding et al. 1992; Smith et al. 1985; 4 OLIGODENDROCYTES EXPRESS
Smith & Hall 1988). Also, OLs suffer injury upon release of C A L C I U M-P E R M E A B L E G LU TA M AT E
Ca2+ from internal stores (Benjamins & Nedelkoska 1996). R E C E P TO R S
Together these observations underscore the key role played by
an excess accumulation of intracellular Ca2+ levels, regardless Myelin and OLs are inexcitable, although they are electrically
of the source, as a fundamental mediator of demyelination and polarized (Karadottir et al. 2005) (this is speculative for myelin,
OL injury (Deng et al. 2003; Follett et al. 2004; Karadottir which is a rather unusual structure and difficult to study
et al. 2005; Matute 2010; Micu et al. 2006; Salter & Fern 2005). physiologically; for further discussion see Stys 2011). Therefore it
Clearly such experiments are deliberately contrived to directly was somewhat surprising to find voltage-gated ion channels and
raise intracellular Ca2+ levels in order to study the consequences, transmitter-gated receptors on these cells. In fact, OLs and their
and of course such chemical ionophore-induced Ca2+ overload progenitors express a surprisingly rich variety of voltage- and
would not occur in disease states. However, Ca2+ ionophores chemically-activated channels, including many Na+, K+ and Ca2+
mimic both complement and perforin attack, inducing lysis of channels, together with ionotropic and metabotropic glutamate
OLs (Jones et al. 1991). This suggests that even natural, albeit receptors, purinergic receptors, and GABA-sensitive channels.
pathological, conditions lead to direct OL/myelin Ca2+ excess The precise roles of these channels are unknown, but recent
and cell damage. In addition to nonspecific ionophore- (artificial data indicate they likely contribute importantly to development,
or natural) induced Ca2+ entry, OLs and perhaps surprisingly, proliferation, and migration (Domingues et al. 2010; see chapter
myelin itself, express channels and receptors that permeate Ca2+. 20). The common physiological effect of activation of most of
This likely represents a physiological signaling arrangement, but these channels is cell depolarization and cytosolic Ca2+ increase,
one that also promotes injury and demyelination if overdriven both through transmembrane flux and release from internal
under pathological conditions. Ca2+ stores. Taken together, short of being able to generate
In fact, there exist many factors and stimuli capable of induc- full-fledged action potentials, OLs appear to exhibit most of
ing Ca2+ fluctuations in OLs, likely playing a role in development the active signaling behavior of excitable neurons. It is therefore
and if excessive, in pathology. T-cell perforins trigger Ca2+ signals not surprising that like neurons, which are famous for their
as noted above (Jones et al. 1991), as well as a variety of inflamma- sensitivity to overactivation and suffering excitotoxicity in a wide
tory mediators including histamine and bradykinin (Bernstein variety of pathological conditions, OLs also suffer damage from
et al. 1996; He et al. 1996), arachidonic acid and its by-products overactivation of their surface channels and receptors and suffer
(Soliven et al. 1993), and lysophosphatidic acid (Moller et al. “excitotoxicity”; indeed, of all glia, those of the oligodendroglial
1999). The mechanisms involved in OL and myelin damage lineage are the most susceptible to this type of damage (Matute
by immune effectors are complex and beyond the scope of this et al. 2007a).

666 • NEUROGLIA
A series of reports from the 1990s unequivocally included mainly in gray matter OLs (Ziak et al. 1998). It wasn’t until
OLs into the family of cells that suffer damage upon excess almost a decade later that Attwell and colleagues reported
exposure to glutamate (Matute et al. 1997; Oka et al. 1993; functional NMDA receptors in OLs at all stages of matura-
Yoshioka et al. 1996). The receptors most responsible are the tion (Karadottir et al. 2005; Karadottir & Attwell 2007;
ionotropic AMPA and kainate receptors. Notably, OL AMPA Burzomato et al. 2010). These receptors are somewhat unique,
receptors lack the GluA2 (formerly GluR2) subunit, and the exhibiting weak Mg2+ block, which would imply that they
GluK2 (formerly GluR6) kainate subunit is minimally edited may generate a significant current even at the nominal rest-
(Matute et al. 2001), conferring substantial Ca2+ permeability ing potential of −60 to −65 mV. From knowledge of molecular
to both. Exposure of OLs to AMPA and kainate receptor ago- physiology of NMDA receptors, together with immunochem-
nists results in OL apoptosis, or after more chronic exposure, ical studies, it was suggested that the subunit composition may
OL necrosis, demyelination, and axonal damage (Matute 1998). also be unusual, composed of GluN1 (NR1), GluN2C/D
Given the documented vulnerability of OLs to Ca2+ overload (NR2C/D) and/or GluN3 (NR3) subunits. Probably not
and the highly Ca2+-permeable AMPA and kainate receptor coincidentally, mature myelin itself also expresses NMDA
varieties expressed by these cells, it is highly likely that OLs are receptors with similar subunit compositions (see next sec-
directly damaged by receptor overactivation and Ca2+ influx tion) (Burzomato et al. 2010; Micu et al. 2006; Pin ῀ a-Crespo
from outside the cell. Interestingly, AMPA receptor-induced et al. 2010). At variance with the above findings, more recent
Ca2+ entry can be further amplified by a Ca2+-induced Ca2+ electrophysiological experiments using acute brain slices from
release phenomenon in OLs (akin to cardiac Ca2+-induced transgenic mice revealed that OLs substantially downregulate
Ca2+ release), leading to further endoplasmic reticulum (ER) somatic AMPA and NMDA receptor densities and glutamate
stress, mitochondrial damage, and cell injury (Ruiz et al. receptor-coding mRNAs as they mature; although progenitor
2010). This phenomenon in OLs is curiously similar to what is NG2+ cells exhibit substantial NMDA receptor-dependent
described in white matter axons that are myelinated by these currents, in this study pre-myelinating and mature OLs did
same cells; here submyelinic axonal AMPA receptors, orga- not (De Biase et al. 2010). In line with these observations,
nized into discrete signaling “nanocomplexes” (Stirling & Stys Salter and Fern reported that OL somata preferentially express
2010), also trigger exaggerated Ca2+ release from the axoplas- AMPA/kainate receptor subunits, whereas the OL processes
mic reticulum, the axonal equivalent of the ER (Ouardouz et contain NMDA receptors (Salter & Fern 2005); because
al. 2009). Depending on the intensity and chronicity of recep- processes will be geographically and electrically distant from
tor activation, glutamate receptor-mediated OL Ca2+ overload the soma, they would tend not to be detected electrophysi-
may result not only in necrotic death, but may cause apopto- ologically during patch clamp recordings, consistent with the
sis. Interestingly, the apoptotic cascades triggered by AMPA absence of detectable NMDA currents noted above. Moreover,
and kainate receptors are distinct, indicating selective signal- ischemic injury of OL somata was prevented by AMPA/kain-
ing pathways. Kainate receptors activate caspases 9 and 3, ate receptor antagonists, but processes were only protected
whereas AMPA receptors induce caspase 8, truncation of the when NMDA receptors were blocked (Salter & Fern 2005).
Bid protein, followed by activation of caspase 3 and PARP-1 Similarly, Micu and coworkers found that ischemic Ca2+
polymerase (Matute et al. 2007a). The presence of AMPA and increase in the OL cell body was prevented by the AMPA/
kainate receptors on OLs, and their importance in meditating kainate antagonist NBQX, whereas myelin Ca2+ accumula-
injury to this critical cell, have been confirmed and extended tion was mainly dependent on NMDA receptors (Micu et al.
by many groups in a variety of models (reviewed in Karadottir 2006). Taken together, although the issue of NMDA recep-
& Attwell 2007). Glutamate receptor overactivation may also tors on OLs is still not completely settled, current evidence
result in indirect injury to OLs given that other glia such as suggests the following: (1) Immature OLs express all ionotro-
microglia, also express these receptors. The latter have been pic receptors on their soma, likely explaining their vulnerabil-
shown to release tumor necrosis factor α (TNFα) in response ity in pre- and early postnatal life (Fern & Moller 2000; Deng
to glutamate receptor activation, and the generation of reac- et al. 2003). (2) as OLs mature, somatic density of NMDA
tive oxygen and nitrogen species which could further exacer- receptors diminishes, perhaps not to zero, leaving AMPA/
bate damage to OLs (Matute et al. 2001). kainate receptors as the predominant forms on their cell bod-
The third major ionotropic glutamate receptor is the ies. (3) Instead, NMDA receptor expression seems to migrate
N-methyl-d-aspartate (NMDA) receptor, which is ubiqui- centrifugally, occupying processes and the myelin sheath.
tous in the mammalian CNS, subserving a key role in learning This framework implies that cytoprotection of OLs is com-
and memory. This receptor differs from the AMPA/kainate plicated, and will require a combination approach, because
classes being highly permeable to Ca2+, and thus implicated in both the soma, processes, and sheaths are essential for survival
many acute and chronic degenerative CNS disorders (Lau & and function of the entire myelinating unit, which will likely
Tymianski 2010). The presence of functional NMDA recep- suffer damage by overactivation of distinct receptor subtypes.
tors on OLs has been more controversial. A number of earlier Another important point pertaining to NMDA receptors is
studies failed to conclusively demonstrate these (Karadottir the fact that they require two obligatory agonists for activa-
& Attwell 2007). A report by Sykova and coworkers demon- tion: glutamate and glycine (or D-serine). The latter has not
strated NMDA-evoked currents in oligodendrocytes from been studied much in the context of OLs, and will be briefly
spinal gray matter, which diminished substantially during the discussed below. Although the previous discussion is focused
first 14 postnatal days, with only very weak currents detected on pathophysiology, the obvious question that arises is, what

M ETA B O L I C I N J U RY O F O L I G O D E N D R O C Y T E S A N D M Y E L I N • 667
is the physiological purpose of these receptors on OLs? glutamate–cystine exchange activity, driving uptake of cystine
Unmyelinated CNS axons have been shown to have vesicular in exchange for release of cytosolic glutamate, further increas-
release apparatus, forming synaptic specializations with OL ing toxic extracellular glutamate levels (Domercq et al. 2007).
precursor cells, and supporting AMPA receptor-mediated GluTs contribute to OL damage, and excitotoxicty in
activity-dependent glutamatergic transmission from axon to general, not only by reducing transmitter uptake, but also by
this glial precursor (Kukley et al. 2007; Ziskin et al, 2007). releasing glutamate from the cytosol. Because of the sensitivity
Moreover, demyelinated axons in the adult mouse corpus cal- of glutamate receptors (especially NMDA receptors) to this
losum form functional AMPA/kainate receptor-dependent transmitter, extracellular glutamate is maintained in the low
synapses onto adult-born OL precursor cells migrating from micromolar range. In contrast, cytoplasm of most cells con-
the subventricular zone after focal demyelination. This “syn- tains millimolar glutamate, and the intracellular space repre-
apse” is downregulated as the OL precursors mature and com- sents the major fraction of brain volume. Therefore, there is
plete their remyelination (Etxeberria et al. 2010). Therefore it potentially a large lethal source or glutamate lurking just on
is tempting to speculate that AMPA/kainate receptors play a the other side of every membrane of every cell in the CNS.
role in differentiation and migration of immature OLs during Under pathological conditions such as trauma or ischemia,
(re)myelination. As for NMDA receptors, their role in this cells accumulate Na+ and depolarize, two ideal stimuli that
process is less clear. Fields and colleagues showed that NMDA synergistically conspire to promote reverse operation of gluTs,
receptors, together with metabotropic glutamate receptors, which are electrogenic and driven by the prevailing Na+ (and
stimulated through vesicular release of glutamate induced K+) gradient (Danbolt 2001). Thus, reverse operation of glutTs
by electrical activity from axons undergoing myelination, are and release of glutamate is a major source of pathological glu-
involved in promoting local protein synthesis in myelinating tamate rise in the extracellular space, and causes injury to OLs,
processes (Wake et al. 2011). On the other hand, other reports neurons, and axons (Fern & Moller 2000). Indeed, any cell
indicate that these receptors are not involved in postnatal OL that is depolarized and loaded with Na+ under pathological
development or myelination (De Biase et al. 2011; Guo et al. conditions, and that expresses gluTs (which includes all major
2012). Given the predilection of “centrifugal” expression of cell types in white matter), will release abnormal amounts
OL NMDA receptors as alluded to above, perhaps their role of glutamate. This is particularly true of white matter axons,
has more to do with physiology of OL processes and axomy- which contain substantial quantities of glutamate in the axo-
elinic signaling (Stys 2011), possibly relevant to “metabolic” plasm that is readily released during injury (Li et al. 1999). As
support of the mature axomyelinic unit, but this is purely mentioned in the previous section, NMDA receptors require
speculative at this time. not only glutamate, but also glycine, for activation. As with
Although OLs appear to express all three classes of iono- glutamate, the CNS is equipped with Na+-dependent glycine
tropic glutamate receptors that are poised to cause damage, the transporters (glyT1 and 2) (Aragon and Lopez-Corcuera
other key part of the excitotoxic equation is clearance of this 2003). We have shown that in CNS white matter, glyTs con-
transmitter. This is effected by Na+-dependent glutamate trans- tribute importantly to Ca2+ overload in both OLs and myelin
porters (gluTs), of which 5 distinct types have been identified during chemical ischemia (Micu et al. 2007), indicating that
and are expressed in the mammalian CNS, denoted EAAT1 this transporter family may also play a major role in OL injury,
through 5 (excitatory amino acid transporter) (Danbolt and white matter damage in general, by virtue of its ability
2001). Glutamate transporters may contribute to OL excito- to source the other essential NMDA receptor coagonist. It is
toxicity passively, by reducing their capacity for extracellular interesting to note that inborn errors of metabolism leading
glutamate clearance, or actively, by releasing glutamate from to abnormally high or low levels of glycine and/or D-serine
the cytoplasm of cells during conditions of intracellular Na+ result in severe myelination defects (de Koning et al. 2000;
loading and depolarization, both of which will tend to drive Press et al. 1989), underscoring the potential importance of
gluTs in the glutamate release mode. The importance of con- this less-studied, but essential, NMDA receptor coagonist in
tinuous and competent glutamate uptake is underscored by myelination and white matter integrity.
reports that inhibition of these transporters, either pharma-
cologically or by antisense oligonucleotides, causes OL death,
demyelination, and axon damage in animal models (Domercq 5 OT H E R C A L C I U M-P E R M E A B L E
et al. 2005). Such experiments are very instructive because CHANNELS AND OLIGODENDROCYTE
they tell us that the basal cycling (i.e., release and reuptake) I N J U RY
of glutamate is substantial, even in white matter; impairment
of uptake, or excess release alone, is sufficient to cause severe While glutamate, its receptor family and its transporters, are
damage to white mater in general, and OLs in particular. the most thoroughly studied in the context of OLs, these
Impaired gluT expression and function has been implicated cells express additional receptors that are emerging as playing
in exacerbating excitotoxicty in human disorders such as mul- important roles in OL pathophysiology. As their name implies,
tiple sclerosis (Werner et al. 2001), where an inflammatory purinergic receptors are activated by purines and pyrimidines,
milieu with generation of proinflammatory cytokines such as and are divided into two main classes, P1 and P2, activated
TNFα and oxidative stress promotes downregulation of gluTs by adenosine and ATP/ADP, respectively. The P2 family is
(Domercq et al. 2007). To add additional insult to inflam- further subdivided into P2X and P2Y. The P1 and P2Y recep-
matory injury, infiltrating activated microglia increase their tors are G protein-coupled metabotropic, whereas P2X are

668 • NEUROGLIA
ionotropic receptors (Burnstock et al. 2011; Matute 2011). matter injury (Constantinou and Fern 2009; Nikolaeva et al.
Each of these main categories is itself further subdivided into 2009). The list is already large (for a review, see Domingues et
many subtypes. All four types of P1 receptor are expressed by al. 2010) and is likely to grow larger as time goes on, empha-
OLs, most of the Ca2+-permeable ionotropic P2X subtypes sizing the remarkable molecular complexity of nonsynaptic
(with the P2X7 receptor being particularly prominent) and white matter and its constituent cells.
several metabotropic P2Y subtypes (for review, see Matute
& Ransom 2012). Activation of these purinergic receptors by
adenosine and ATP results in elevation of cytosolic [Ca] in 6 MYELIN ALSO EXPRESSES CHANNELS
OLs, via release from internal Ca2+ stores or influx from the A N D R E C E P TO R S
extracellular space across the plasma membrane ( James & Butt
2002; Matute et al. 2007b). Under physiological conditions, Myelin is a very unusual structure, composed of multiple wraps
these receptors may function to transduce signals from axons of lipid-rich membranes emanating from OL processes in the
and astrocytes to control myelination (Butt 2011). Importantly, CNS (see chapter 44). Its main role is to increase the resistance
given their propensity to raise OL Ca2+ levels via stores release and reduce the capacitance of the axon membrane allowing
or permeation across the cell membrane, purinergic receptors rapid saltatory conduction along fibers that are one or two orders
are well positioned to effect injury to OLs under pathologi- of magnitude thinner than their unmyelinated equivalents,
cal conditions. The lifeblood of cellular energy supply, ATP, is an essential requirement in a high-density mammalian CNS.
paradoxically highly toxic when it accumulates outside the cell. Without an obvious intracellular compartment, electrical
Under pathological conditions, ATP can be released by several polarization or source of signaling molecules, it was assumed
mechanisms, including Ca2+-dependent exocytosis of synap- that myelin is an inert structure, playing its part only by virtue
tic vesicles, connexin- and pannexin-containing gap junction of its passive biophysical properties. A major stumbling block
hemichannels, and other ion channels, including P2X7 recep- was a lack of techniques capable of reporting physiological
tors themselves, setting the stage for a potentially dangerous responses from this structure, in contrast to neurons and glia
positive feedback loop. Although P2X7 receptors have a rela- where quantitative and sophisticated electrical and optical
tively low affinity for ATP, pathological conditions result in recordings have been applied for decades. Given biochemical,
substantial release of this nucleotide in sufficient amounts to immunohistochemical, proteomics-based, and cytochemical
activate these highly Ca2+-permeable receptors (Burnstock et evidence that myelin per se expresses functional ion channels
al. 2011). An important source of ATP release may be from and pumps, transporters and chemical receptors (Ishii et al.
astrocytes (Coco et al. 2003). This precipitates severe OL 2009; Jahn et al. 2009), we posited that it too can respond
injury (Matute et al. 2007b), and as will be discussed in the to stimuli, both physiological and pathological, much like its
next section, probably direct damage to myelin as well. parent OL and possibly even like neurons. We developed a
Acid-sensing ion channels (ASICs) are plasmalemmal technique based on 2-photon laser scanning microscopy that is
channels that are activated by low (<7) extracellular pH. These able to report Ca2+ fluctuations from the major dense line (the
cation channels are mainly permeable to Na+ but some isoforms thin cytosolic spiral) of mature CNS myelin (Micu et al. 2007).
exhibit substantial Ca2+ permeability (reviewed in Krishtal Using this method, we showed that mature CNS myelin suffers
2003). Cells of the OL lineage express ASICs, and in particular, a substantial increase in Ca2+ levels following chemical ischemia
the ASIC1a splice variant (Feldman et al. 2008), which is Ca2+ (Micu et al. 2006). Molecular dissection revealed a surprising
permeable. This would implicate these channels as potentially mechanism: while the ischemic Ca2+ rise in the OL cell body
important mediators of OL injury under acidotic conditions is mainly mediated by AMPA/kainate receptors, myelinic Ca2+
as occurs during ischemia or inflammation (Friese et al. 2007). overload does not passively follow the fate of the parent soma.
These channels can flux damaging amounts of Ca2+ directly, AMPA/kainate receptor blockers have only a modest effect on
and can contribute additional indirect damage by promoting myelinic Ca2+ increase, (nonetheless, the effect was significant,
Na+ entry and depolarization, in turn activating voltage-gated consistent with earlier immunohistochemical demonstration
Ca2+ channels, Na+ Ca2+ exchanger (Chen et al., 2007), and of GluA4 AMPA subunits in myelin [Li and Stys 2000], an
glutamate transporters (the latter two driven to import Ca2+ observation that was subsequently confirmed and extended to
and release glutamate respectively), all known to be expressed include the GluK5 [KA2] kainate receptor subunit [Brand-
by OLs. The importance of these channels is underscored by Schieber and Werner 2003]). Somewhat surprisingly, NMDA
reports of reduced tissue damage and improved clinical scores receptor inhibitors completely block the ischemic Ca2+ rise in
after experimental autoimmune encephalomyelitis in animals myelin (but not in OL soma) (Micu et al. 2006), emphasizing
genetically lacking ASIC1 or where this channel was pharma- the independent and molecularly unique response of the
cologically inhibited by amiloride (Friese et al. 2007). In con- sheath compared to its parent OL cell body. Using a variety
trast, activation of these channels promoted OL injury in an of specific pharmacological agents, it was determined that
ASIC-dependent manner (Vergo et al. 2011). myelinic receptors are likely composed of the obligatory
As more detailed experiments are focused on OLs and GluN1 (NR1) subunit and GluN2C-D (NR2C-D) subunits,
myelin, additional channels, receptors and transporters are with no evidence for GluN2A or B, which are prominent
being identified, any of which can participate in parallel to synaptic and extrasynaptic (respectively) receptors in neurons.
mediate OL injury and demyelination. For instance, recently Immunohistochemistry provided a direct demonstration of
nicotinic and adrenoreceptors have been implicated in white the presence of all the major NMDA receptor subunits on

M ETA B O L I C I N J U RY O F O L I G O D E N D R O C Y T E S A N D M Y E L I N • 669
CNS myelin, located in clusters, mainly along the inner and It may be no coincidence that myelin prominently expresses this
outer myelin loops (Micu et al. 2006) (Fig. 52.1). At the same unusual GluN1/GluN3 receptor, as it has been shown to be
time, Attwell and colleagues independently corroborated the weakly susceptible to voltage-dependent Mg block, and exhibits
presence of NMDA receptor subunits in myelin (Karadottir low Ca2+ permeability (reviewed in Henson et al. 2010). Unlike
et al. 2005), which was further confirmed by Fern and coworkers conventional synapses, where AMPA/kainate receptors are
(Alix and Fern 2009), lending credence to this unexpected present to depolarize the postsynaptic membrane and relieve
finding. A follow-up study revealed an even more unexpected NMDA receptors from their Mg block, perhaps myelin cannot
result. Using the same technique, but without chemical ischemia, undergo similar depolarizations, and therefore must contain
we subsequently showed that D-serine alone (a GluN1 glycine a receptor that is not strongly blocked at hyperpolarized
site agonist) elicits a substantial rise in myelinic Ca2+ levels potentials. Once activated however, the myelinic NMDA
(Pin῀ a-Crespo et al. 2010). Pharmacological dissection revealed receptor may need to show restraint with regards to how
that these responses are likely mediated by an unusual “glycine- much Ca2+ it permeates; the cytoplasmic volume of the inner
only” NMDA receptor, activated by glycine or D-serine alone mesaxon (the uncompacted inner tongue of myelin adjacent to
(Chatterton et al. 2002), without the need for NMDA or the axon) and the myelin spiral is tiny compared with a neurons
glutamate which bind to the GluN2 subunit. These “glycine- cytosolic volume; therefore, an NMDA receptor with limited
only” NMDA receptors are instead composed of glycine/D- Ca2+ permeability could suffice for whatever local signaling is
serine-binding subunits, GluN1 and GluN3, without GluN2. required, and in fact may be essential to avoid injuring the inner
loops. In summary, to date evidence indicates the inner myelin
loops contain GluA4 AMPA, and mainly GluN1/GluN2C-D
(or perhaps triheteromeric GluN1/GluN2C/GluN3A-
containing receptors [Burzomato et al. 2010]) and GluN1/
GluN3 “glycine-only” NMDA receptors, and that these
contribute importantly to myelin damage. The physiological
role of these receptors is unknown, but they may represent
the “postsynaptic” component of the proposed axomyelinic
synapse (Stys 2011). The potentially unique pharmacological
properties of the GluN1/GluN3 receptors raise intriguing
possibilities for rational design of selective therapeutic agents
for disorders where demyelination is prominent.
The rather unexpected finding of functional neurotrans-
mitter receptors on myelin was bolstered soon thereafter by
data from Matute and colleagues who reported the pres-
ence of Ca2+ permeable P2X7 receptors not only on OLs,
but also in myelin (Matute et al. 2007b). They found that
activation of P2X7 damaged OLs and promoted demyelina-
tion, similar to activation of glutamate receptors. Moreover,
P2X7 receptors are expressed on compact myelin, and like
NMDA receptors, appear to show a predilection for the
inner and outer myelin loops. Although these investigators
did not directly demonstrate that myelinic P2X7 receptors
cause Ca2+ overload in this structure (promotion or reduc-
tion of demyelination by modulating P2X7 may have been
due to injury or survival of the parent OLs in their study),
given the high Ca2+ permeability of P2X7 and their expres-
sion in myelin, it is highly likely that like NMDA receptors,
these purinergic receptors directly contribute to myelin
Ca2+ overload and demyelination. Although highly specu-
lative at this time, the emergence of various neurotrans-
mitter receptors, transporters and various ion exchangers/
cotransporters in myelin further raises the prospect that the
mature sheath is a far more active structure than previously
Figure 52.1 CNS Myelin Expresses Transmitter Receptors. A. Clusters
of NMDA receptor subunits (green) are seen surrounding axon cylinders thought, capable of specific signaling possibly originating
(neurofilament, red) of adult rat optic nerve axons that are fully myeli- from its ensheathed axon. Unfortunately for myelin, like
nated at this age. Scale bars = 2 μm. Immunogold labeling indicates that with glia and excitable neurons, when excessively activated
most receptors are located at the inner or outer lops of myelin. Scale bars: under pathological conditions, these same signaling path-
50 nm. B. Compact CNS myelin also expresses clusters of P2X7 puriner-
ways likely contribute to myelin damage and may represent
gic receptors with a similar distribution. Scale bars 300 nm. My: myelin,
Ax: axon. (A) Modified with permission from Micu et al. the most proximal events eventually leading to frank histo-
2006. (B) Modified with permission from Matute et al. 2007b). pathological demyelination.

670 • NEUROGLIA
GluT
(EAAT1) Ca AMPAR
K
Na KAR
glu mGluR
cystine/
glutamate glu
RyR
antiporter P2YR
Ca IP3R G
cystine ROS ER
-
...glutathione Na
Ca NKCC
K
Ca
NCX Cl

Na
- Ca
ATPase
Na Na-K NO
ATPase -
ONOO
Ca
ROS K
ASIC1
NO P2X7
ONOO
- NMDAR H+
Ca
Ca K Cl Ca Na, Ca
Ca K Na
ATPase
Na-K GluA4 GluK5 NMDAR P2X7 calpain
KCC
ATPase Ca
PAD2
MYELIN
NET (GluN1/2C,D/3A)
GluT (EAAT1)
Na NE

AXON

Figure 52.2 Schematic Illustrating Some Major Pathways That Normally Subserve Important Physiological Functions, but May be recruited to Operate
Inappropriately Contributing to Injury to Ols and Myelin. The OL soma expresses most major ion channels, transporters, and AMPA/kainate and
metabotropic glutamate receptors. Ca2+ homeostasis in maintained by the plasmalemmal Na+-Ca2+ exchanger (NCX) and Ca2+-ATPase, as well as the
ER Ca2+-ATPase (not shown). Pathological Ca2+ accumulation can be mediated by many systems, including reverse NCX, AMPA/kainate receptor
overactivation, acid-sensing ion channels (ASIC1a), and ionoptropic (P2X7) and metabotropic (P2Y) purinergic receptors. Additional Ca2+ can be
released from the ER via ryanodine (RyR) receptors in a Ca2+-induced Ca2+ release manner or via IP3 receptors. The Na+ K+-Cl– cotransporter (NKCC)
can load the OL soma with Na+ leading to further injury by reverse NCX or promoting glutamate release via reverse glutamate transport (gluT). OL
processes are preferentially damaged by NMDA receptors. Myelin itself expresses a variety of receptors (GluA4 AMPA, NMDA, KA2 kainate, and
P2X7 purinergic) all of which are Ca2+-permeable. Reactive oxygen and nitrogen species (ROS, NO, peroxynitrite [ONOO-]), generated locally or by
nearby cells, can diffuse and promote membrane and myelin damage through lipid peroxidation and can exacerbate mitochondrial dysfunction (see text
for more detail).

7 N O N –R E C E P TO R -M E D I AT E D I N J U RY mechanisms (Back et al. 1998). This powerful intracellular


antioxidant molecule depends on a steady supply of cystine,
Neuroinflammatory lesions are characterized by a very toxic supplied by uptake by the cystine/glutamate antiporter, which
milieu with elevated levels of nitric oxide (NO) (or reactive is then converted to cysteine, essential for glutathione synthe-
nitrogen species in general), cytokines, glutamate, and oxygen sis (Conrad and Sato 2012). Interfering with this system by
free radicals. Nitric oxide has both beneficial and deleterious cystine deprivation results in toxicity to OLs, especially imma-
effects (reviewed in Smith and Lassmann 2002); OLs appear ture OLs (Back et al. 1998). Upregulation of this antiporter,
particularly vulnerable to this free radical (Mitrovic et al. under conditions that require a response to increased oxida-
1994), with cell death proceeding either via necrosis or apop- tive stress, will release glutamate, potentially exacerbating
tosis depending on the length and intensity of NO exposure excitotoxicity (Conrad and Sato 2012) (see the preceding),
(Garthwaite et al. 2002). The particularly toxic metabolite of reflecting the complex interdependence of many pathways,
NO, peroxynitrite, damages cell membranes through lipid the net result of which is not always apparent.
peroxidation, permanently altering their chemical and physi- Regulation of cellular volume is a complex and essential
cal properties (van der Veen & Roberts 1999). This may be function of all cells, and depends on a complex collection
particularly important for demyelination, because the altered of channels and anion transporters coupled to various other
membrane composition promotes recognition and phagocy- ions (reviewed in Okada 2004). Given that the essential func-
tosis by macrophages (Graham et al. 1993). Reactive oxygen tion of myelin depends on its structural integrity, and on a
species are also toxic to OLs and likely myelin as well. Like tight apposition to its axon, swelling of this structure could
NO, these radicals are also generated in abundance by acti- adversely affect its function in a significant way, by altering its
vated macrophages and microglia, resulting in damage to electrical characteristics and promoting current leaks if vol-
white matter elements in neuroinflammatory conditions such ume changes disrupt the intimate axomyelinic relationship.
as multiple sclerosis (MS) (Haider et al. 2011; reviewed in The K+-Cl– cotransporter, one of the main anion cotransport-
van Horssen et al. 2011). Indeed, cells of the OL lineage have ers involved in volume regulation, is expressed in OLs and
developed powerful strategies to cope with free radical stress, also in myelin (Malek et al., 2003). Experiments on anoxic
with the glutathione system being one of the more important ex vivo white matter tracts revealed that while reducing Na+

M ETA B O L I C I N J U RY O F O L I G O D E N D R O C Y T E S A N D M Y E L I N • 671
influx through voltage-gated Na+ channels by TTX is highly (Karadottir et al. 2005). To date, reperfusion strategies are the
protective at restoring the total magnitude of evoked electrical only effective treatment for acute stroke. The burst of reactive
responses (Stys et al. 1992), the waveshapes of the compound oxygen species generated by mitochondria when tissue is reox-
action potentials, governed by velocities of constituents axons, ygenated (Piantadosi and Zhang 1996) causes additional dam-
remain significantly distorted (Malek et al. 2003). This suggests age, and OLs may be particularly vulnerable because of their
that Na+ channel blockade rescues axons from injury, but the high iron content (LeVine and Macklin 1990), which catalyzes
axomyelinic integrity continues to be disrupted, causing slow- the formation of free radicals and promotes lipid peroxidation.
ing and desynchronization of action potential propagation. This may be particularly important for lipid-rich myelin, and
Interestingly, addition of the K+ Cl– cotransporter inhibitor is consistent with the frequent demyelination seen after brain
furosemide significantly improves the shape, but not the mag- ischemia. Interestingly, detailed pathological studies indicate
nitude, of the postanoxic compound action potential, suggest- that the inner myelin loops (those adjacent to the axon) are
ing that myelin swelling mediated by aberrant Cl– movement preferentially damaged by ischemia as evidenced by loss of
(and in turn water) across the myelin lamellae, mediated by myelin-associated glycoprotein, together with apoptotic OLs,
the K+ Cl– co-transporter, was mainly responsible for disrupt- and often with still-preserved, though frequently spheroidized
ing the essential physical association between myelin sheath axons (Aboul-Enein et al. 2003). This attests to the high vul-
and axon (Malek et al. 2003). The Na+-K+-Cl- cotransporter nerability of OLs and myelin to ischemia. Moreover, the fact
has also been shown to promote pathological Na+ entry into that Ca2+-permeable myelinic NMDA receptors are preferen-
OLs, which then triggers reverse Na+ Ca2+ exchange resulting tially located on the inner loops, precisely where the earliest
in Ca2+ entry across the OL plasmalemma and additional Ca2+ myelin damage is observed, may not be a coincidence. In addi-
dependent injury (Chen et al. 2007). tion, expression of Ca2+-permeable P2X7 purinergic receptors
on OLs and inner myelin loops also contributes to ischemic
injury to these elements (Domercq et al. 2010).
8 I N J U RY TO O L I G O D E N D R O C Y T E S A N D Demyelinating neuroinflammatory conditions such as
MYELIN IN SPECIFIC DISORDER S multiple sclerosis are another prominent example of disorders
exhibiting prominent damage to OLs and myelin. Mechanisms
Many prevalent and devastating disorders of the brain and of damage by the immune system have been extensively studied
spine adversely affect the white matter, with OLs and myelin and are complex, the details being beyond the scope of this chap-
being prominent targets that are rarely if ever spared injury. ter. The reader is instead referred to chapter 61 and other reviews
Indeed, it is likely that OLs and myelin are probably affected, (Bruck 2005). In addition to the complex immunopathogenesis
directly or indirectly, in virtually every disorder of the CNS, of the MS plaque, inflammatory lesions represent a very toxic
therefore it is not possible to cover every one in this section. milieu with high levels of excitotoxins (glutamate, ATP), NO,
Instead, a few prominent and better understood conditions, at and a plethora of inflammatory effectors. The detailed pathways
least from the viewpoint of OL/myelin pathophysiology, will of glutamatergic and purinergic injury to OLs and myelin have
be mentioned. Moreover, there are many overlaps in pathways been described above. More relevant to MS and neuroinflam-
pathologically overdriven among various disorders (e.g., gluta- matory disease, however, are observations from animal and
mate excitotoxicity is prominent in ischemia, inflammation, human studies clearly indicating increased levels of glutamate
and trauma), therefore no clear lines should be drawn in the in the MS brain (Srinivasan et al. 2005), impaired homeostatic
discussion that follows. One of the commonest diseases is isch- mechanisms for glutamate (Werner et al., 2001), robust pro-
emia, either with an acute presentation as in ischemic stroke, or tective effects of AMPA/kainate (Pitt et al. 2000; Smith et al.
a more chronic scenario of longstanding (mainly subcortical) 2000), and NMDA receptor (Wallstrom et al. 1996; but see Guo
small vessel insufficiency and microinfarcts. Ischemia affects et al. 2012; Matute 2010) antagonists in rodent models of experi-
not only older subjects, but also the very young, in the form mental autoimmune encephalomyelitis (an experimental model
of perinatal ischemia resulting in the well-known pathology mirroring the inflammatory lesions of MS) and genome-wide
of periventricular leukomalacia (Back et al. 2007). Ischemia is association studies showing strong links with genes involved
particularly damaging because the general reduction of energy in glutamate homeostasis (Baranzini et al. 2010). Interestingly,
supply in cells promotes multiple points of failure, with an this latter study indicated the strongest correlation in patients
intracellular increase of [Ca2+]i being the final common path- with the greatest degree of progressive degeneration, suggest-
way leading to OL and myelin injury. Indeed, after neurons, ing a direct link to MS pathogenesis independent of immune
OLs are the most sensitive cells to hypoxia/ischemia (Fern & or inflammatory effects. Another very important mediator of
Moller 2000; reviewed in Matute et al. 2006), with immature cellular injury during inflammation is NO generation (Smith
OLs in the perinatal brain being even more sensitive probably & Lassmann 2002). Oligodendrocytes are far more sensitive
because of their collection of surface glutamate receptors. Influx to damage by NO (or more likely peroxynitrite, its highly toxic
of extracellular Ca2+ through AMPA/kainate receptors is prob- by-product (Jack et al. 2007)), than other glia (Mitrovic et al.
ably the most important route of Ca2+ overload in OLs (Matute 1994). Mechanisms include lipid peroxidation, with the large
2011). As noted in previous sections, OL processes and myelin area of myelin membrane conferring particular vulnerability.
are preferentially sensitive to NMDA receptor overactivation Peroxynitrite might indirectly damage OLs (and most other
(Micu et al. 2006; Salter & Fern 2005), though this distinction cells) by inhibiting mitochondrial metabolism, thus promoting
between soma and processes/myelin is certainly not absolute a state of “virtual hypoxia” (Stys 2004).

672 • NEUROGLIA
Trauma to the brain and spinal cord is an unfortunately is the most vulnerable in most disorders of the CNS. However,
common occurrence. The pathophysiology of secondary glia play critical roles as well, and in particular, the OL and
degenerative events triggered by the initial injury is diverse myelin, without which the neuron’s raison d’être is largely
(Farkas and Povlishoc 2007), but OL injury and demyelina- extinguished: if the neuron cannot transmit its output to its
tion are a nearly universal feature that may progress for weeks target, the neuron may as well not exist. Recent years have seen
or months following the initial trauma (Totoiu and Keirstead a growing interest in the fundamental mechanisms of OL and
2005). Extracellular glutamate has been shown to rise signifi- myelin injury, and this area is starting to catch up to the vast
cantly after both brain and spinal trauma (McAdoo and Wu knowledge base that has accumulated regarding the neuron
2008; Chamoun et al. 2010), triggering secondary excitotox- and the interneuronal synapse. The emerging complexity per-
icty as discussed above. Another major excitotoxin, ATP, also taining to the OL and myelin is at the same time fascinating,
increases substantially after traumatic brain and spinal cord and humbling, as even this relatively simple cell is turning out
injury, resulting in further injury to nearby OLs and neu- to be far more complex than once imagined. Perhaps the most
rons via P2X7 receptors (reviewed in Burnstock et al. 2011). interesting aspect of OL/myelin biology is the discovery of
Inflammation and relative ischemia are also prominent after an ever richer collection of neurotransmitter-based signaling
trauma, in turn entraining all the deleterious cascades associ- mechanisms on the OL and even the mature myelin sheath
ated with these conditions as detailed previously. The unfolded itself. Together with emerging data on transmitter release
protein response in response to ER stress has been shown to machinery in axons, these observations are painting a fas-
be directly involved in OL damage, and by extension in sec- cinating novel framework of an “axomyelinic” synapse (Stys
ondary demyelination, following spinal cord injury (Ohri 2011). This synapse may play a fundamentally important role
et al. 2011). Finally, neurodegenerative conditions, in particu- in myelination during development and repair, and may also
lar Alzheimer’s disease, also involve degeneration of white mat- control white matter plasticity in the mature CNS. Equally
ter, including injury to OLs and demyelination. Traditionally important, dysfunction of this synapse may participate in the
white matter pathology in Alzheimer’s has been ascribed to pathogenesis of a broad range of neurological disorders where
small vessel ischemia and Wallerian degeneration secondary to white matter is adversely affected. The challenge for the next
cortical atrophy. Recent evidence shows that Aβ alters NMDA decade will be to expand our knowledge of axoglial, and in
receptor kinetics leading to receptor overactivation, excessive particular, axomyelinic signaling, which in turn will pave the
Ca2+ influx, and neuronal injury (You et al. 2012). Given that road for developing potent and selective therapeutic agents
myelin also expresses NMDA receptors, it is conceivable that designed to mitigate damage to the white matter of the CNS.
a similar mechanism may promote direct injury to myelin.
Traumatic brain injury and chronic traumatic encephalopa-
thy also exhibit β-amyloid deposition ( Johnson et al. 2010), REFERENCES
suggesting a potential similar mechanism of myelin and white
matter damage after trauma. These proposed new mechanisms Aboitiz F, Scheibel AB, Fisher RS, Zaidel E. 1992. Fiber composition of
will require experimental confirmation however. the human corpus callosum. Brain Res 598:143–153.
Aboul-Enein F, Rauschka H, Kornek B, Stadelmann C, Stefferl A,
Bruck W, et al. 2003. Preferential loss of myelin-associated glycopro-
tein reflects hypoxia-like white matter damage in stroke and inflam-
9 S U M M A RY A N D P E R S P E C T I VE S matory brain diseases. J Neuropathol Exp Neurol 62:25–33.
Alix JJ, Fern R. 2009. Glutamate receptor-mediated ischemic injury of
Oligodendrocytes and the myelin they produce play a vital premyelinated central axons. Ann Neurol 66:682–693.
role for the proper operation of the CNS. Numerous disorders Aragon C, Lopez-Corcuera B. 2003. Structure, function and regulation
of glycine neurotransporters. Eur J Pharmacol 479:249–262.
result in damage to OLs and demyelination, causing major Arluison M, Quignon M, Thorens B, Leloup C, Penicaud L. 2004.
morbidity and mortality. As in many other cells, Ca2+ overload Immunocytochemical localization of the glucose transporter 2
plays a major role in the death of OLs and damage to myelin. (GLUT2) in the adult rat brain. II. Electron microscopic study.
Recent studies have revealed a remarkably complex collection J Chem Neuroanat 28:137–146.
of channels and receptors expressed on OL cell bodies, pro- Back SA, Gan X, Li Y, Rosenberg PA, Volpe JJ. 1998. Maturation-dependent
vulnerability of oligodendrocytes to oxidative stress-induced death
cesses, and myelin itself (Fig. 52.2), that contribute to Ca2+ caused by glutathione depletion. J Neurosci 18:6241–6253.
accumulation and cell injury in a variety of conditions ranging Back SA, Riddle A, McClure MM. 2007. Maturation-dependent vulnera-
from ischemia, trauma, neuroinflammation, and neurodegen- bility of perinatal white matter in premature birth. Stroke 38:724–730.
erative disorders. It is highly likely that there exist many more Baranzini SE, Srinivasan R, Khankhanian P, Okuda DT, Nelson SJ,
chemically-gated receptors on OL cell bodies, processes, and Matthews PM, et al. 2010. Genetic variation influences glutamate
concentrations in brains of patients with multiple sclerosis. Brain
myelin itself, that may provide unique therapeutic opportuni- 133:2603–2611.
ties and desired selectivity, both for protecting the myelinat- Benjamins JA, Nedelkoska L. 1996. Release of intracellular calcium
ing unit from injury, and conceivably for boosting repair and stores leads to retraction of membrane sheets and cell death in mature
remyelination after an insult. Experimental approaches aimed mouse oligodendrocytes. Neurochem Res 21:471–479.
at understanding mechanisms of neurological disease have Bernstein M, Lyons SA, Moller T, Kettenmann H. 1996.
Receptor-mediated calcium signalling in glial cells from mouse cor-
been heavily “neurocentric,” that is, biased toward elucidating pus callosum slices. J Neurosci Res 46:152–163.
cellular and molecular mechanisms of injury to neurons and Brand-Schieber E, Werner P. 2003. AMPA/kainate receptors in
synapses. This is understandable because the neuronal element mouse spinal cord: cell-specific display of receptor subunits by

M ETA B O L I C I N J U RY O F O L I G O D E N D R O C Y T E S A N D M Y E L I N • 673
oligodendrocytes and astrocytes and at the nodes of Ranvier. Glia Farkas O, Povlishock JT. 2007. Cellular and subcellular change evoked
42:12–24. by diff use traumatic brain injury: a complex web of change extending
Bruck W. 2005. The pathology of multiple sclerosis is the result of far beyond focal damage. Prog Brain Res 161:43–59.
focal inflammatory demyelination with axonal damage. J Neurol Feldman DH, Horiuchi M, Keachie K, Mccauley E, Bannerman P, Itoh
252(Suppl 5):v3–9. A, et al. 2008. Characterization of acid-sensing ion channel expres-
Buntinx M, Stinissen P, Steels P, Ameloot M, Raus J. 2002. sion in oligodendrocyte-lineage cells. Glia 56:1238–1249.
Immune-mediated oligodendrocyte injury in multiple sclerosis: Fern R, Moller T. 2000. Rapid ischemic cell death in immature oligo-
molecular mechanisms and therapeutic interventions. Crit Rev dendrocytes: a fatal glutamate release feedback loop. J Neurosci
Immunol 22:391–424. 20:34–42.
Burnstock G, Krugel U, Abbracchio MP, Illes P. 2011. Purinergic sig- Follett PL, Deng W, Dai W, Talos DM, Massillon LJ, Rosenberg PA,
nalling: from normal behaviour to pathological brain function. Prog et al. 2004. Glutamate receptor-mediated oligodendrocyte toxicity
Neurobiol 95:229–274. in periventricular leukomalacia: a protective role for topiramate. J
Burzomato V, Frugier G, Perez-Otano I, Kittler JT, Attwell D. 2010. The Neurosci 24:4412–4420.
receptor subunits generating NMDA receptor mediated currents in Friese MA, Craner MJ, Etzensperger R, Vergo S, Wemmie JA, Welsh MJ,
oligodendrocytes. J Physiol 588:3403–3414. et al. 2007. Acid-sensing ion channel-1 contributes to axonal degen-
Butt AM. 2006. Neurotransmitter-mediated calcium signalling in oligo- eration in autoimmune inflammation of the central nervous system.
dendrocyte physiology and pathology. Glia 54:666–675. Nat Med 13:1483–1489.
Butt AM. 2011. ATP: a ubiquitous gliotransmitter integrating Garthwaite G, Goodwin DA, Batchelor AM, Leeming K, Garthwaite J.
neuron-glial networks. Semin Cell Dev Biol 22:205–213. 2002. Nitric oxide toxicity in CNS white matter: an in vitro study
Chamoun R, Suki D, Gopinath SP, Goodman JC, Robertson C. 2010. using rat optic nerve. Neuroscience 109:145–155.
Role of extracellular glutamate measured by cerebral microdialysis in Graham A, Hogg N, Kalyanaraman B, O’Leary V, Darley-Usmar V,
severe traumatic brain injury. J Neurosurg 113:564–570. Moncada S. 1993. Peroxynitrite modification of low-density lipopro-
Chatterton JE, Awobuluyi M, Premkumar LS, Takahashi H, Talantova tein leads to recognition by the macrophage scavenger receptor. FEBS
M, Shin Y, et al. 2002. Excitatory glycine receptors containing the Lett 330:181–185.
NR3 family of NMDA receptor subunits. Nature 415:793–798. Guo F, Maeda Y, Ko EM, Delgado M, Horiuchi M, Soulika A, et al. 2012.
Chen H, Kintner DB, Jones M, Matsuda T, Baba A, Kiedrowski L, et Disruption of NMDA receptors in oligodendroglial lineage cells does
al. 2007. AMPA-mediated excitotoxicity in oligodendrocytes: role not alter their susceptibility to experimental autoimmune encephalo-
for Na(+)-K(+)-Cl(-) co-transport and reversal of Na(+)/Ca(2+) myelitis or their normal development. J Neurosci 32:639–645.
exchanger. J Neurochem 102:1783–1795. Haider L, Fischer MT, Frischer JM, Bauer J, Hoftberger R, Botond G,
Coco S, Calegari F, Pravettoni E, Pozzi D, Taverna E, Rosa P, et al. 2003. et al. 2011. Oxidative damage in multiple sclerosis lesions. Brain
Storage and release of ATP from astrocytes in culture. J Biol Chem 134:1914–1924.
278:1354–1362. Harauz G, Musse AA. 2007. A tale of two citrullines—structural and
Conrad M, Sato H. 2012. The oxidative stress-inducible cystine/gluta- functional aspects of myelin basic protein deimination in health and
mate antiporter, system x (c) (-): cystine supplier and beyond. Amino disease. Neurochem Res 32:137–158.
Acids 42:231–246. Harris JJ, Attwell D. 2012. The energetics of CNS white matter.
Constantinou S, Fern R. 2009. Conduction block and glial injury J Neurosci 32:356–371.
induced in developing central white matter by glycine, GABA, nora- He M, Howe DG, McCarthy KD. 1996. Oligodendroglial signal trans-
drenalin, or nicotine, studied in isolated neonatal rat optic nerve. duction systems are regulated by neuronal contact. J Neurochem
Glia 57:1168–1177. 67:1491–1499.
Danbolt NC. 2001. Glutamate uptake. Prog Neurobiol 65:1–105. Henson MA, Roberts AC, Perez-Otano I, Philpot BD. 2010. Influence
De Biase LM, Kang SH, Baxi EG, Fukaya M, Pucak ML, Mishina M, of the NR3A subunit on NMDA receptor functions. Prog Neurobiol
et al. 2011. NMDA receptor signaling in oligodendrocyte progenitors 91:23–37.
is not required for oligodendrogenesis and myelination. J Neurosci Ishii A, Dutta R, Wark GM, Hwang SI, Han DK, Trapp BD, et al. 2009.
31:12650–12662. Human myelin proteome and comparative analysis with mouse
De Biase LM, Nishiyama A, Bergles DE. 2010. Excitability and synap- myelin. Proc Natl Acad Sci U S A 106:14605–14610.
tic communication within the oligodendrocyte lineage. J Neurosci Jack C, Antel J, Bruck W, Kuhlmann T. 2007. Contrasting potential of
30:3600–3611. nitric oxide and peroxynitrite to mediate oligodendrocyte injury in
de Koning TJ, Jaeken J, Pineda M, Van Maldergem L, Poll-The BT, van multiple sclerosis. Glia 55:926–934.
der Knaap MS. 2000. Hypomyelination and reversible white mat- Jahn O, Tenzer S, Werner HB. 2009. Myelin proteomics: molecular
ter attenuation in 3-phosphoglycerate dehydrogenase deficiency. anatomy of an insulating sheath. Mol Neurobiol 40:55–72.
Neuropediatrics 31:287–292. James G, Butt AM. 2002. P2Y and P2X purinoceptor mediated Ca2+
Deng W, Rosenberg PA, Volpe JJ, Jensen FE. 2003. Calcium-permeable signalling in glial cell pathology in the central nervous system. Eur J
AMPA/kainate receptors mediate toxicity and preconditioning by Pharmacol 447:247–260.
oxygen-glucose deprivation in oligodendrocyte precursors. Proc Natl Johnson VE, Stewart W, Smith DH. 2010. Traumatic brain injury and
Acad Sci U S A 100:6801–6806. amyloid-beta pathology: a link to Alzheimer’s disease? Nat Rev
Domercq M, Etxebarria E, Perez-Samartin A, Matute C. 2005. Neurosci 11:361–370.
Excitotoxic oligodendrocyte death and axonal damage induced by Jones J, Frith S, Piddlesden S, Morgan BP, Compston DA, Campbell
glutamate transporter inhibition. Glia 52:36–46. AK, et al. 1991. Imaging Ca 2+ changes in individual oligodendro-
Domercq M, Perez-Samartin A, Aparicio D, Alberdi E, Pampliega O, cytes attacked by T-cell perforin. Immunology 74:572–577.
Matute C. 2010. P2X7 receptors mediate ischemic damage to oligo- Karadottir R, Attwell D. 2007. Neurotransmitter receptors in the life
dendrocytes. Glia 58:730–740. and death of oligodendrocytes. Neuroscience 145:1426–1438.
Domercq M, Sanchez-Gomez MV, Sherwin C, Etxebarria E, Fern Karadottir R, Cavelier P, Bergersen LH, Attwell D. 2005. NMDA recep-
R, Matute C. 2007. System xc- and glutamate transporter inhibi- tors are expressed in oligodendrocytes and activated in ischaemia.
tion mediates microglial toxicity to oligodendrocytes. J Immunol Nature 438:1162–1166.
178:6549–6556. Krishtal O. 2003. The ASICs: signaling molecules? Modulators? Trends
Domingues AM, Taylor M, Fern R. 2010. Glia as transmitter sources and Neurosci 26:477–483.
sensors in health and disease. Neurochem Int 57:359–366. Kukley M, Capetillo-Zarate E, Dietrich D. 2007. Vesicular glutamate
Etxeberria A, Mangin JM, Aguirre A, Gallo V. 2010. Adult-born SVZ release from axons in white matter. Nat Neurosci 10:311–320.
progenitors receive transient synapses during remyelination in corpus Lau A, Tymianski M. 2010. Glutamate receptors, neurotoxicity and neu-
callosum. Nat Neurosci 13:287–289. rodegeneration. Pflugers Arch 460:525–542.

674 • NEUROGLIA
LeVine SM, Macklin WB. 1990. Iron-enriched oligodendrocytes: a reex- Pin῀a-Crespo JC, Talantova M, Micu I, States B, Chen HS, Tu S, et al.
amination of their spatial distribution. J Neurosci Res 26:508–512. 2010. Excitatory glycine responses of CNS myelin mediated by NR1/
Li S, Mealing GA, Morley P, Stys PK. 1999. Novel injury mechanism in NR3 “NMDA” receptor subunits. J Neurosci 30:11501–11505.
anoxia and trauma of spinal cord white matter: glutamate release via Pitt D, Werner P, Raine CS. 2000. Glutamate excitotoxicity in a model
reverse Na+-dependent glutamate transport. J Neurosci 19:RC16. of multiple sclerosis. Nat Med 6:67–70.
Li S, Stys PK. 2000. Mechanisms of ionotropic glutamate receptor-mediated Press GA, Barshop BA, Haas RH, Nyhan WL, Glass RF, Hesselink JR.
excitotoxicity in isolated spinal cord white matter. J Neurosci 20: 1989. Abnormalities of the brain in nonketotic hyperglycinemia: MR
1190–1198. manifestations. AJNR Am J Neuroradiol 10:315–321.
Malek S, Coderre E, Stys PK. 2003. Aberrant chloride transport con- Rinholm JE, Hamilton NB, Kessaris N, Richardson WD, Bergersen
tributes to anoxic/ischemic white matter injury. J Neurosci 23: LH, Attwell D. 2011. Regulation of oligodendrocyte development
3826–3836. and myelination by glucose and lactate. J Neurosci 31:538–548.
Mata M, Fink DJ. 1989. Ca 2+-ATPase in the central nervous system: an Ruiz A, Matute C, Alberdi E. 2010. Intracellular Ca 2+ release through
EM cytochemical study. J Histochem Cytochem 37:971–980. ryanodine receptors contributes to AMPA receptor-mediated mito-
Matute C. 1998. Characteristics of acute and chronic kainate exci- chondrial dysfunction and ER stress in oligodendrocytes. Cell Death
totoxic damage to the optic nerve. Proc Natl Acad Sci U S A 95: Dis 1:e54.
10229–10234. Salter MG, Fern R. 2005. NMDA receptors are expressed in develop-
Matute C. 2010. Calcium dyshomeostasis in white matter pathology. ing oligodendrocyte processes and mediate injury. Nature 438:
Cell Calcium 47:150–157. 1167–1171.
Matute C. 2011. Glutamate and ATP signalling in white matter pathol- Schanne FA, Kane AB, Young EE, Farber JL. 1979. Calcium-dependence
ogy. J Anat 219:53–64. of toxic cell death: a final common pathway. Science 206:700–702.
Matute C, Alberdi E, Domercq M, Perez-Cerda F, Perez-Samartin A, Schmahmann JD, Smith EE, Eichler FS, Filley CM. 2008. Cerebral
Sanchez-Gomez MV. 2001. The link between excitotoxic oligo- white matter: neuroanatomy, clinical neurology, and neurobehav-
dendroglial death and demyelinating diseases. Trends Neurosci ioral correlates. Ann N Y Acad Sci 1142:266–309.
24:224–230. Scolding NJ, Morgan BP, Campbell AK, Compston DA. 1992. The role
Matute C, Alberdi E, Domercq M, Sanchez-Gomez MV, of calcium in rat oligodendrocyte injury and repair. Neurosci Lett
Perez-Samartin A, Rodriguez-Antiguedad A, et al. 2007a. Excitotoxic 135:95–98.
damage to white matter. J Anat 210:693–702. Smith KJ, Hall SM. 1988. Peripheral demyelination and remyelina-
Matute C, Domercq M, Sanchez-Gomez MV. 2006. Glutamate-mediated tion initiated by the calcium-selective ionophore ionomycin: in vivo
glial injury: mechanisms and clinical importance. Glia 53:212–224. observations. J Neurol Sci 83:37–53.
Matute C, Ransom BR. 2012. Roles of white matter in CNS Smith KJ, Hall SM, Schauf CL. 1985. Vesicular demyelinatin induced by
patho-physiologies. ASN Neuro 4:pii. raised intracellular calcium. J Neurol Sci 71:19–37.
Matute C, Sanchez-Gomez MV, Martinez-Millan L, Miledi R. 1997. Smith KJ, Lassmann H. 2002. The role of nitric oxide in multiple sclero-
Glutamate receptor-mediated toxicity in optic nerve oligodendro- sis. Lancet Neurol 1:232–241.
cytes. Proc Natl Acad Sci U S A 94:8830–8835. Smith T, Groom A, Zhu B, Turski L. 2000. Autoimmune encephalomy-
Matute C, Torre I, Perez-Cerda F, Perez-Samartin A, Alberdi E, elitis ameliorated by AMPA antagonists. Nat Med 6:62–66.
Etxebarria E, et al. 2007b. P2X(7) receptor blockade prevents ATP Soliven B, Takeda M, Shandy T, Nelson DJ. 1993. Arachidonic acid and
excitotoxicity in oligodendrocytes and ameliorates experimental its metabolites increase Cai in cultured rat oligodendrocytes. Am J
autoimmune encephalomyelitis. J Neurosci 27:9525–9533. Physiol 264:C632–640.
McAdoo DJ, Wu P. 2008. Microdialysis in central nervous system disor- Srinivasan R, Sailasuta N, Hurd R, Nelson S, Pelletier D. 2005. Evidence
ders and their treatment. Pharmacol Biochem Behav 90:282–296. of elevated glutamate in multiple sclerosis using magnetic resonance
Micu I, Ridsdale A, Zhang L, Woulfe J, McClintock J, Brantner CA, et spectroscopy at 3 T. Brain 128:1016–1025.
al. 2007. Real-time measurement of free Ca2+ changes in CNS myelin Stirling DP, Stys PK. 2010. Mechanisms of axonal injury: inter-
by two-photon microscopy. Nat Med 13:874–879. nodal nanocomplexes and calcium deregulation. Trends Mol Med
Micu I, Coderre E, Jiang Q, Ridsdale A, Zhang L, Woulfe J, et al. 2006. 16:160–170.
NMDA receptors mediate Ca accumulation in central nervous sys- Stys PK. 2004. Axonal degeneration in MS: is it time for neuroprotective
tem myelin during chemical ischemia. Nature 439:988–992. strategies? Ann Neurol 55:601–603.
Mitrovic B, Ignarro LJ, Montestruque S, Smoll A, Merrill JE. 1994. Nitric Stys PK. 2011. The axo-myelinic synapse. Trends Neurosci 34:
oxide as a potential pathological mechanism in demyelination: its differ- 393–400.
ential effects on primary glial cells in vitro. Neuroscience 61:575–585. Stys PK, Waxman SG, Ransom BR. 1992. Ionic mechanisms of anoxic
Moller T, Musante DB, Ransom BR. 1999. Lysophosphatidic injury in mammalian CNS white matter: role of Na+ channels and
acid-induced calcium signals in cultured rat oligodendrocytes. Na+-Ca 2+ exchanger. J Neurosci 12:430–439.
Neuroreport 10:2929–2932. Totoiu MO, Keirstead HS. 2005. Spinal cord injury is accompa-
Nikolaeva MA, Richard S, Mouihate A, Stys PK. 2009. Effects of the nied by chronic progressive demyelination. J Comp Neurol 486:
noradrenergic system in rat white matter exposed to oxygen-glucose 373–383.
deprivation in vitro. J Neurosci 29:1796–1804. van der Veen RC, Roberts LJ. 1999. Contrasting roles for nitric oxide and
Ohri SS, Maddie MA, Zhao Y, Qiu MS, Hetman M, Whittemore SR. peroxynitrite in the peroxidation of myelin lipids. J Neuroimmunol
2011. Attenuating the endoplasmic reticulum stress response improves 95:1–7.
functional recovery after spinal cord injury. Glia 59:1489–1502. van Horssen J, Witte ME, Schreibelt G, de Vries HE. 2011. Radical
Oka A, Belliveau MJ, Rosenberg PA, Volpe JJ. 1993. Vulnerability of oli- changes in multiple sclerosis pathogenesis. Biochim Biophys Acta
godendroglia to glutamate: pharmacology, mechanisms, and preven- 1812:141–150.
tion. J Neurosci 13:1441–1453. Vergo S, Craner MJ, Etzensperger R, Attfield K, Friese MA, Newcombe
Okada Y. 2004. Ion channels and transporters involved in cell volume reg- J, et al. 2011. Acid-sensing ion channel 1 is involved in both axonal
ulation and sensor mechanisms. Cell Biochem Biophys 41:233–258. injury and demyelination in multiple sclerosis and its animal model.
Ouardouz M, Coderre E, Zamponi GW, Hameed S, Yin X, Trapp BD, Brain 134:571–584.
et al. 2009. Glutamate receptors on myelinated spinal cord axons: II. Vosler PS, Brennan CS, Chen J. 2008. Calpain-mediated signaling mech-
AMPA and GluR5 receptors. Ann Neurol 65:160–166. anisms in neuronal injury and neurodegeneration. Mol Neurobiol
Pfeiffer SE, Warrington AE, Bansal R. 1993. The oligodendrocyte and 38:78–100.
its many cellular processes. Trends Cell Biol 3:191–197. Wake H, Lee PR, Fields RD. 2011. Control of local protein synthe-
Piantadosi CA, Zhang J. 1996. Mitochondrial generation of reactive oxy- sis and initial events in myelination by action potentials. Science
gen species after brain ischemia in the rat. Stroke 27:327–331. 333:1647–1651.

M ETA B O L I C I N J U RY O F O L I G O D E N D R O C Y T E S A N D M Y E L I N • 675
Wallstrom E, Diener P, Ljungdahl A, Khademi M, Nilsson CG, Olsson You H, Tsutsui S, Kannanayakal TJ, Chen L, Hameed S, Xia P, et al. 2012.
T. 1996. Memantine abrogates neurological deficits, but not CNS Aβ damages neurons by altering copper-dependent prion protein regu-
inflammation, in Lewis rat experimental autoimmune encephalomy- lation of NMDA receptors. Proc Natl Acad Sci USA 109:1737–1742.
elitis. J Neurol Sci 137:89–96. Ziak D, Chvatal A, Sykova E. 1998. Glutamate-, kainate- and
Werner P, Pitt D, Raine CS. 2001. Multiple sclerosis: altered gluta- NMDA-evoked membrane currents in identified glial cells in rat
mate homeostasis in lesions correlates with oligodendrocyte and spinal cord slice. Physiol Res 47:365–375.
axonal damage. Ann Neurol 50:169–180. Ziskin JL, Nishiyama A, Rubio M, Fukaya M, Bergles DE. 2007.
Yoshioka A, Bacskai B, Pleasure D. 1996. Pathophysiology of oligo- Vesicular release of glutamate from unmyelinated axons in white
dendroglial excitotoxicity. J Neurosci Res 46:427–437. matter. Nat Neurosci 10:321–330.

676 • NEUROGLIA
53.
INTERACTION OF MICROGLIA WITH NEURONS
AND ASTROCY TES UNDER LESIONED NEURONAL
CONDITIONS
Kazuyuki Nakajima and Shinichi Kohsaka

A B B R E VI AT I O N S and pathological conditions. However, it is not always entirely


clear how the glial cells and neurons interact, or for what rea-
BDNF brain-derived neurotrophic factor son. A good model for examining the cellular interactions
CD11b cluster of differentiation 11b between glial cells and neurons is the peripheral nerve injury
Cdk cyclin-dependent protein kinase model, which often involves injuries from the transection of
CdkI cyclin-dependent protein kinase inhibitor facial nerves (Kreutzberg 1996) or hypoglossal nerves (Morita
ChAT choline acetyltransferase et al. 1996). Since this injury system maintains the blood brain
CM conditioned medium barrier under normal conditions, the reactions between neu-
CNS central nervous system rons and glial cells can be observed in the parenchyma without
CNTF ciliary neurotrophic factor considering the effects of blood-derived cells and blood con-
CSF colony stimulating factor stituents. Initially, the facial nerve injury system results in an
ERK extracellular signal-regulated kinase alteration of motoneurons, followed by glial responses includ-
GM-CSF granulocyte, macrophage-CSF ing microglial activation/proliferation and astroglial activation
GDNF glial cell line-derived neurotrophic factor (Kreutzberg 1996; Streit et al. 1988). This chapter summarizes
GFAP glial fibrillary acidic protein current knowledge on the events in injured neurons, microglia,
GLAST glutamate aspartate transporter and astrocytes, as well as the interaction between microglia and
GLT-1 glutamate transporter-1 neurons/astrocytes, in the peripheral nervous injury system.
Iba1 ionized Ca2+ binding adapter molecule
IL interleukin
JNK c-jun N-terminal protein kinase 2 CHANGES IN INJURED NEURONS
MAPK mitogen-activated protein kinase
M-CSF macrophage-CSF Transection of the facial nerve in adult rats leads to a retro-
MPF mitosis promoting factor grade injury in the motoneuron cell bodies. In this case, the
NGF nerve growth factor injured motoneurons are viable and regenerate, unlike in
NO nitric oxide neonatal rats. The injured neurons begin to change their gene
NT neurotrophin expressions, morphology and functional state.
PA plasminogen activator In the early stages after transection, immediate early genes
PAI-1 plasminogen activator inhibitor-1 including c-jun, Jun-B, and TIS11 are induced in the injured
PCNA proliferating cell nuclear antigen motoneurons (Haas et al. 1993). Subsequently, these injured
PLGn plasminogen cells undergo changes in their expressions of molecules such
TGFβ transforming growth factor beta as cytoskeleton components, metabolic enzymes, neuropep-
TNFα tumor necrosis factor alpha tides, and cytokines (Moran and Graeber 2004). For example,
tPA tissue-type PA the transcriptions of actin and tubulin are downregulated in
uPA urokinase-type PA the injured motoneurons (McNamara et al. 2000). Stearoyl-
VAchT vesicular acetylcholine transporter CoA desaturase, which mainly plays a role in energy storage, is
strongly promoted (Schmitt et al. 2003). Neuron-specific eno-
lase levels are increased in the motoneurons following injury,
1 INTRODUCTION and may contribute to their survival (Angelov et al. 1994).
Damage-induced neuronal endopeptidase is enhanced in the
The nervous system is constructed of heterogeneous cell types motoneurons of the axotomized hypoglossal nucleus, possibly
(i.e., neurons and glial cells), and these cell types are thought to to participate in a self-protection mechanism (Kiryu-Seo et al.
interact closely with each other. These processes are mediated 2000). Calcitonin gene-related protein (Haas et al., 1990),
by many molecules acting as signaling molecules under normal GAP-43 (Schwaiger et al. 1998) (see chapters 48 and 51)

677
and interferon-gamma (Olsson et al. 1989) are elevated in A
response to neuronal injury. Accompanying these changes, the Iba1
functioning of the injured motoneurons declines along with
the downregulated levels of choline acetyltransferase (ChAT) GFAP
and the vesicular acetylcholine transporter (VAchT) (Tetzlaff
and Kreutzberg 1984). In fact the facial motoneurons reduce L R L R L R L R L R
the levels of ChAT and VAchT after the injury (Fig. 53.1). 1d 3d 5d 7d 14d

B CD11b
3 R E S P O N S E S O F M I C R O G L I A TO
M OTO N E U R O N I N S U LT

3.1 AC T I VAT I O N
The morphological change from a ramified form to a short
process-bearing form is a characteristic feature of microglia
observed after motoneuronal insult. Accompanying or after
the transformation/morphological change, microglia start
to increase the expression of various kinds of proteins. The
Figure 53.2 Activated Microglia in the Transected Facial Nucleus.
classically established proteins in activated microglia include A. Iba1 and GFAP levels in the time course. The same samples
immune-related proteins such as the cluster of differentiation used in Figure 53.1A were immunoblotted for Iba1 and GFAP.
11b (CD11b), CD4, and Fc receptor (Kreutzberg 1989). Later B. Immunohistochemistry for CD11b. The same tissue sections used in
studies showed that ionized Ca2+ binding adapter protein 1 Figure 53.1B were stained for CD11b. C, control side; T, transected side.
(Iba1) (Ito et al. 1998) and annexin III (Konishi et al. 2006)
are also induced in activated microglia. CD11b and Iba1 are “synaptic stripping.” Although the localization of the microglia
well-defined microglia-specific markers in the parenchyma of would seem to suggest that they are involved in the “stripping”
the nervous system (Fig. 53.2). For a list of other molecules of synaptic contact, later evidence indicates that synaptic with-
upregulated in microglia in the axotomized facial nucleus, see drawal from the motoneuron cell body following peripheral
the review by Moran and Graeber (2004). nerve injury is likely a neuron-autonomous event, and not one
In the axotomized facial nucleus (Blinzinger and induced by activated microglia (Perry and O’Connor 2010).
Kreutzberg 1968) and in the axotomized hypoglossal nucleus
(Gesase and Kiyama 2000), microglial processes are observed 3.2 P RO L I FE R AT I O N
to squeeze into the synaptic cleft, leading to the concept of
One of the responses of microglia to motoneuronal injury is
A proliferation. After the injury-induced morphological trans-
ChAT formation, microglia begin to increase their numbers in the
ipsilateral facial nucleus (Ito et al. 1998).
VAchT
3.2.1 Proliferation Factors and the Receptors
L R L R L R L R L R
1d 3d 5d 7d 14d
The factors controlling microglial proliferation have been
assumed as colony stimulating factors (CSFs) in in vitro
B ChAT VAchT studies, in which macrophage-CSF (M-CSF); granulocyte,
C T C T macrophage-CSF (GM-CSF); and multi-CSF (interleukin 3;
IL-3) exert almost equivalent proliferative effects on microglia
(Guilian and Ingleman 1988). However, among the growth fac-
tors potentially involved in microglial proliferation, M-CSF is
perhaps the most likely candidate based on the finding that
osteopetrotic mice (Wiktor-Jedrzejczak et al. 1990), which
cannot produce biologically active M-CSF, exhibit almost no
Figure 53.1 Injured Motoneurons in the Transected Facial Nucleus. increase in microglial number in the transected facial nucleus
A. ChAT and VAchT levels in the injured nucleus. The right (R) facial (Raivich et al. 1994). In fact, the amount of M-CSF increases
nerve of adult rats was cut, and after 1, 3, 5, 7 and 14 days the left (L) and in the ipsilateral facial nucleus at the time of microglial prolif-
right (R) facial nuclei of each rat were recovered. The tissue homogenate eration (Yamamoto et al. 2010). At the same time, the recep-
of each nucleus was immunoblotted for ChAT and VAchT. tor for M-CSF, cFms (Ross 2006), is also remarkably induced
B. Immunohistochemistry for ChAT and VAchT. Tissue sections of the
brainstem of an adult rat whose right facial nerve was transected 5 days in the ipsilateral facial nucleus (Fig. 53.3A). These results are
earlier was stained for ChAT (left side) and for VAchT (right side). essentially in good agreement with those from 125I-M-CSF-
C, control side; T, transected side. binding experiments (Raivich et al. 1991).

678 • NEUROGLIA
A suggesting that the induced CdkI brakes the progression of
M-CSF the microglial cell cycle advanced by MPF.
The participation of cyclin A and cyclin D in microglial
proliferation was reported in a GM-CSF-stimulated micro-
cFms
glial cell line (Koguchi et al. 2003). Zhang et al. (2009) dem-
onstrated that the microglial proliferation that occurs in focal
PCNA cerebral ischemia is inhibited by a reduction of cyclin A, B,
and E by applying a cell cycle inhibitor to the brain prior to
middle carotid artery occlusion.
Cyclin A

L R L R L R L R L R 3.2.3 Signaling Mechanism for Inducing


1d 3d 5d 7d 14d Microglial Proliferation
B CFms Iba1 Cyclin A Iba1
There is accumulating evidence that, after stimulation with a
mitogen, a specific signaling cascade for proliferation is acti-
vated in microglia. Liva et al. (1999) reported that, following
GM-CSF stimulation, extracellular signal regulated kinase
(ERK)1/2, Jak2, and STAT5A/5B were phosphorylated in
microglia in vitro. In human microglial cultures in which
microglia proliferate in a GM-CSF-dependent manner, Suh
et al. (2005) demonstrated that Hck tyrosine kinase and
the phophatidylinositol 3-kinase/AKt signaling cascade are
Figure 53.3 Proliferation of Microglia in the Transected Facial Nucleus. associated with microglial proliferation. The importance of
A. Time course of M-CSF, cFms, PCNA, and cyclin A levels. The same p38 mitogen-activated protein kinases (p38MAPKs) in the
samples used in Figure 53.1A were immunoblotted for M-CSF, cFms, microglial proliferation/activation induced by neuregulin-1
PCNA, and cyclin A. B. Immunohistochemistry for cFms and cyclin
A. The same tissue sections used in Figure 53.1B were dually stained for
has been reported in the rat dorsal horn (Calvo et al. 2010).
cFms/Iba1 (left side) and for cyclin A/Iba1 (right side). In the facial nerve transection system, cFms stimulated by
M-CSF is activated and transduces the signals to downstream
MAPKs (Yamamoto et al., 2012). The stimulation of micro-
glia with M-CSF resulted in the activation of ERK, c-Jun
Apart from the facial nerve transection model, other CSFs N-terminal kinase ( JNK), and p38MAPK, and JNK and
are also possible candidates for microglial mitogen. Human p38MAPK played a role in the induction of cyclins/PCNA
fetal and adult microglia in vitro respond to M-CSF and and cFms, respectively.
GM-CSF, and proliferate, but GM-CSF shows a much stron- Mander et al. (2006) reported that microglial proliferation
ger effect than M-CSF (Lee et al. 1994). Immunohistochemical in response to TNFα or IL-1β was mediated by hydrogen perox-
analyses have shown that GM-CSF is produced by reactive ide from nicotinamide adenine dinucleotide phosphate oxidase.
astrocytes in the lesion sites of degenerative diseases. In addi- This finding suggests that reactive oxygen species may be associ-
tion, in a study by Bartolini et al. (2011), granulocyte-CSF ated with a signaling cascade leading to microglial proliferation.
enhanced the proliferation of microglia in the normal cen-
tral nervous system (CNS). Thus, plural CSFs may serve as
3.2.4. Inhibition of Microglial Proliferation
microglial mitogens in normal and pathological states of the
CNS. Furthermore, particularly in inflammatory sites, cytok- Microglial proliferation occurring in the axotomized motor
ines such as tumor necrosis factor alpha (TNFα) and IL-1β nucleus can be blocked. In the transected facial nucleus, adminis-
(Ganter et al., 1992) may act as the mitogens. tration of adriamycin inhibits microglial proliferation (Graeber
et al. 1989). Similarly, in hypoglossal nuclei treated with cytosine
arabinoside, microglial proliferation is inhibited (Svensson and
3.2.2 Cell Cycle-Related Proteins
Aldskogius 1993). In the studies described above, these reagents
It is well known that cyclin pairs with a suitable cyclin-dependent were thought to act as blockers of DNA replication.
protein kinase (Cdk) and the resultant cyclin/Cdk complex
serves as a mitosis-promoting factor (MPF) that progresses
3.2.5 Features of Proliferating Microglia
the cell cycle and leads to cell proliferation (Sherr 1993). More
recently, it was revealed that cyclinA/Cdk2 and cyclinD/ It would be valuable to analyze the properties of proliferat-
Cdk4 serve as the MPFs in microglia of the transected facial ing microglia from the viewpoint of neurotrophic cells/cyto-
nucleus (Yamamoto et al. 2012). After axotomy of the facial toxic cells. From the observations reported so far, harmful
nerve, microglia begin to express the cell cycle-genes cyclin A molecules including TNFα, IL-1β, IL-6, nitric oxide (NO)
and proliferating cell nuclear antigen (PCNA) (Fig. 53.3B), the and superoxide anion are not significantly promoted in the
Cdk inhibitor (CdkI) p21, which serves in cell division arrest transected facial nucleus. On the other hand, in the injured
(Fischer et al., 2002), is also induced in the injured nucleus, facial nucleus, certain neurotrophic factors for motoneurons

I N T E R AC T I O N O F M I C R O G L I A W IT H N EU R O N S A N D A ST R O C Y T E S U N D E R L E S I O N E D N EU R O NA L C O N D IT I O N S • 679
are detected. One of these factors, transforming growth factor change their morphology and metabolic and functional state.
b1 (TGFβ1), is expressed in activated microglia (Kiefer et al. Simultaneously, such injured neurons are presumed to out-
1993). Enhanced levels of brain-derived neurotrophic fac- put various signaling molecules to trigger the activation of
tor (BDNF) are localized in motoneurons (Kobayashi et al. glial cells, including an electrical signal (Rishal and Fainzilber
1996). Apart from the neurotrophic ligands, receptors for 2010), CD200/CD200R interaction-derived signal (Lyons
glial cell line-derived neurotrophic factor (GDNF) (Burazin et al. 2007), chemokines (Nishiyori et al. 1998), cytokines
et al. 1998), BDNF (Kobayashi et al. 1996), leukemia inhibi- (Tsuda et al. 2009), nucleotides, and transmitters. In response
tory factor, and ciliary neurotrophic factor (CNTF) (Haas to the injury signals, microglia are thought to be activated
et al. 1999) are also enhanced in motoneurons. These results and/or proliferated. However, how the microglial properties
predict that these receptors will attempt to bind the ligands are modulated by the neuronal stimulus is poorly understood.
released from surrounding glial cells in the injured facial To clarify this matter, it is necessary to analyze the influence of
nucleus. Microglia and astrocytes in vitro exhibit the ability neuronal stimuli on microglial properties by using an in vitro
to produce a variety of neurotrophic factors. Accordingly, it is system.
possible that proliferating microglia play a role as neurotrophic
cells in the lesioned site.
4.1 N EU ROT RO P H I C A B I L IT Y
The observation that rubrospinal neurons degenerate in
the absence of microglial proliferation in the rubrospinal tract Microglia in vitro produce various kinds of neurotrophic
transection model indicates that proliferating microglia serve as factors (Nakajima and Kohsaka 2004). Whether or not
neurosupportive cells in vivo (Streit 2002). Lalancette-Hebert the microglial properties are altered by neuronal stimulus
et al. (2007) indicated that selective ablation of proliferating is an interesting question that remains to be answered. The
microglia leads to the exacerbation of ischemic injury, and stimulation of microglia with neuronal conditioned medium
conversely, stimulation of microglial proliferation by M-CSF (CM) enhanced the production of neurotrophic factors
results in an increase of insulin-like growth factor-1 levels. The including nerve growth factor (NGF), neurotrophin (NT)-
results suggest that proliferating microglia serve as neuropro- 4/5, TGFβ1, GDNF, fibroblast growth factor, and IL-3
tective cells in cerebral ischemia. (Nakajima et al. 2007). These results suggest that the neuronal
Another proof that proliferating microglia serve as neuro- stimulation changes microglia into more neurotrophic cells,
supportive cells in vivo was provided by Lopez-Redondo et al. which contribute to neuronal survival and regeneration in
(2000), who observed that these cells express high levels of glial vivo (Table 53.1).
type glutamate transporter-1 (GLT-1) in the area surrounding
the injured motoneuron cell body in the axotomized facial
4.2 E XC ITOTOX I N-S C AVE N G I N G A B I L I T Y
nucleus. The location of GLT-1–expressing microglia seems
to suggest that the microglia protect injured motoneurons Microglia cells in vitro express the glutamate transporter
from the abnormal glutamate-induced excitation that would GLT-1 (EAAT2) (Lopez-Redondo et al. 2000), and exhibit
be otherwise induced. a specific ability to uptake glutamate (Nakajima et al., 2001).
The glutamate uptake by microglia was enhanced by neu-
ronal CM, and its stimulation could be blocked by a GLT-1
4 R E S P O N S E O F M I C R O G L I A TO inhibitor (Nakajima et al. 2008). Neuronal CM triggers the
N E U R O N A L S T I MU L AT I O N S I N V I T RO upregulation of GLT-1, but not another glial-type glutamate
aspartate transporter (GLAST) (EAAT1). This implies that
As described above, an insult to the facial nerve may send a proliferating microglial cells at injured motoneurons reduce
signal to the motoneuron cell bodies that induces them to glutamate levels, and thereby reduce glutamate toxicity.

Table 53.1 RESPONSE OF MICROGLIA TO NEURONAL STIMULUS (NEURONAL CONDITIONED MEDIUM)


NONSTIMULATION NEURONAL CM-STIMULATION LPS-STIMULATION

mRNA PRODUCTION RELEASE mRNA PRODUCTION RELEASE mRNA PRODUCTION RELEASE

NGF + + − ++ ++ ++ + ++ ++

BDNF ++ ++ ++ ++ ++ ++ +++ +++ +++

NT-3 ++ − − + − − ++ +/− +/−

NT-4/5 ++ + + ++ +++ +++ ++ ++ ++

TGFb1 + + + +++ +++ +++ ++ ++ ++


GDNF + ++ ++ +++ +++ +++ ++ ++ −

The levels of mRNA expression and the protein production/secretion of NGF, BDNF, NT3, NT4/5, TGFb1, GDNF in nonstimulated microglia, lipopolysaccharide
(LPS)-stimulated microglia, and neuronal CM-stimulated microglia are summarized.

680 • NEUROGLIA
4.3 E X T R AC EL LU L A R P ROT EO LY T I C AC T I VIT Y The expression in the cells tells us that the cells are function-
ally in a phagocytic state.
In the axotomized facial nucleus, microglia change their mor-
phology, proliferate, and migrate to the motoneuron cell bod-
ies, and astrocytes also become hypertrophic and extend their 5.1 N EWB O R N R ATS
processes to form lamellar sheets (Kreutzberg 1996). These
events represent the occurrence of significant tissue remodel- The transection of newborn rat facial nerve leads to the
ing in the axotomized facial nucleus, and involve an associa- appearance of ED1-positive cells in the ipsilateral nucleus
tion of proteinases which stimulate tissue rearrangement by (Graeber et al. 1998). In the same study, the number of ED1-
degrading extracellular proteins. One of the proteases shown positive cells increased. Because the induction of the ED1
to play an important role in the tissue remodeling is plasmin antigen is associated with the formation of phagosomes, this
(Dano et al. 1985). Inactive proenzyme plasminogen (PLGn) finding demonstrated that most of the motoneurons had died.
is changed into an active proteinase plasmin by plasminogen The ED1-positive cells are microglia, which are identified by
activator (PA). PA is of two types: urokinase-type PA (uPA) Iba1 staining. In addition, some of the phagocytic microglia
and tissue-type PA (tPA). were proliferating and major histocompatibility complex
Tissue zymography of brainstem sections reveals that uPA class II-positive. Similar to the facial nerve transection model,
activity is induced in the axotomized facial nucleus (Nakajima motoneuronal cell death is observed in the transected newborn
et al. 1996). The uPA activity increases consistent with the rat hypoglossal nucleus (Snider, 1989) and in the newborn rat
time of microglial proliferation (Nakajima et al. 2004). This sciatic nerve transection paradigm (Iwasaki et al. 1995).
phenomenon suggests the presence of an interaction between
injured motoneurons and microglia. An in vitro study revealed 5.2 A D U LT R ATS
that the amounts of uPA released from microglia are enhanced
by coculture with neurons or by neuronal CM (Nakajima et al. Motoneurons in the adult rat facial nucleus undergo cell death,
2005). Thus, uPA induced in the axotomized facial nucleus and microglia transform to phagocytes when the facial nerve
is considered to be produced in microglia stimulated with is transected and ricin is administered to the transection site or
injured motoneurons. A substrate for uPA and tPA, PLGn, injected into the facial nerve (Streit and Kreutzberg 1988). In
was demonstrated to be produced in microglia (Nakajima addition to these models, cycloheximide induces phagocytes
et al. 1992). Thus, microglia are important cells for generating in the ipsilateral facial nucleus (Fig. 53.4). In the transected
plasmin in the CNS.
A plasmin-generating system (PLGn/PAs system) par-
A
ticipates in remodeling of the nervous system, including in
processes such as degradation of the extracellular matrix, and ED-1
transformation, activation, proliferation, and migration of
glial cells (Nakajima and Kohsaka 1996). At the same time,
plasmin activates inactive proforms such as procollagenase
(He et al. 1989), pro-uPA (Blasi et al. 1987), and pro-IL-8 Actin
(Nakagawa et al. 1991). Furthermore, plasmin is prerequisite
for activating latent TGFβ (Annes et al. 2003) and proNGF/ L R L R L R
proBDNF (Lu et al. 2005). Thus, through the plasmin-gen-
Rat 1 Rat 2 Rat 3
erating ability, activated and proliferating microglia are sug-
gested to contribute to tissue remodeling in the injured facial
B ED-1
nucleus.

5 R E S P O N S E S O F M I C R O G L I A TO
M OTO N E U R O N C E L L D E AT H

The response of microglia to motoneuron cell death is dif-


ferent from the response to injured but living motoneurons.
The motoneuron cell death can be seen in the transected facial
nucleus in newborn rats (Aldskogius and Thomander 1986;
Figure 53.4 Phagocytes Induced by Cycloheximide. A. Expression of
Graeber et al. 1998), in the ricin-administered facial nucleus the ED-1 protein. The first rat did not receive an operation (Rat-1). In
in adult rats (Streit and Kreutzberg 1988), and in the Cholera the second adult rat, the right facial nerve was transected (Rat-2). In
toxin-applied nucleus (Llewellyn-Smith et al. 2000). In these the third adult rat, the right facial nerve was cut, and a gelform soaked
situations, the microglia were transformed into phagocytes. in cycloheximide (CH) solution was administered at the injury site
This response is an essential process to remove needless struc- (Rat 3). After 5 days, each facial nucleus of the three rats was dissected,
and the tissue homogenate was immunoblotted for ED-1 and actin.
tures in advance of the reconstruction of cellular connections B. Immunohistochemistry for ED-1. Tissue sections of brainstem of
in the facial nucleus. An indicator of the phagocytes is CD68 Rat-3 in A were stained with ED-1 antibody. C, control side; T+CH,
(ED-1), which is a lysosomal protein (Damoiseaux 1994). transected and CH-treated side.

I N T E R AC T I O N O F M I C R O G L I A W IT H N EU R O N S A N D A ST R O C Y T E S U N D E R L E S I O N E D N EU R O NA L C O N D IT I O N S • 681
facial nucleus without treatment, ED1-positive cells are not receptor in astrocytes (Popratiloff et al. 1996). These upregu-
observed. However, the ED1-positive cells appeared in the lated molecules may be associated with the cellular and bio-
transected and cycloheximide-treated facial nucleus. These chemical events necessary for tissue remodeling in the injured
cells were PCNA-positive and cFms-positive, suggesting a facial nucleus.
state of proliferation. The influence of these activated astrocytes on neurons is
Dying neurons would be expected to express the “eat me” important for the survival and restoration of neuronal func-
signal (Elward and Gasque 2003) on the neuronal membrane. tion. Generally, the activated astrocytes have been viewed as
Receiving the signals by scavenger receptor and T cell immu- neuroprotective cells for supporting injured neurons by the
noglobin mucin 4, microglia internalize the dead neurons production of neurotrophic factors (Dong and Benveniste
by phagocytosis. The microglia are ED1-positive phagocytic 2001). The capacity has been ascertained in many in vitro stud-
cells (Kinchen and Ravichandran 2008). As a matter of fact, ies. Overexpression of GDNF by astrocytes prevents the pro-
the phagocytic activity is regulated by chemokines and neu- grammed cell death of motoneurons (Oppenheim et al. 2000)
rotransmitters, like ATP (Koizumi et al. 2007). and the axotomy-induced motoneuron cell death (Zhao et al.
2004). These results strongly suggest that the neurotrophic
factors produced in astrocytes are actually delivered to the
6 R E S P O N S E S O F A S T R O C Y T E S TO region surrounding the neurons in the injured peripheral ner-
M OTO N E U R O N I N S U LT vous system.
From the viewpoint of glutamate toxicity, astrocytes can be
regarded as a glutamate eliminator. Activated astrocytes at the
6.1 R E S P O N S I V E N E S S O F A S T RO C Y T E S
lesion site express highly active uptake system for glutamate,
Microglia are not the only cell type that responds to neuronal namely glutamate transporters (GLAST, GLT-1) (Mcnaugt
insult. In the facial nerve transection model, astrocytes also and Jenner 2000). The glutamate taken up into astrocytes is
undergo morphological changes and functional transforma- converted to glutamine by glutamine synthetase (Brusilow
tions. One of the responses is morphological change from a et al. 2010). Thus, astrocytes upon their activation are thought
protoplasmic form to a fibrous form (Graeber and Kreutzberg to act as glutamate eliminators. In addition to the glutamate-
1986). One of the cellular constituents upregulated in astro- uptake capacity, astrocytes have an ability to protect against
cytes is glial fibrillary acidic protein (GFAP), which is an the neuronal cell death induced by oxidative stress (Oshiro
astrocyte-specific intermediate protein and never expressed et al. 2008). Accordingly, activated astrocytes in the injured
in neurons and microglia. Although the level of the inter- facial nucleus are suggested to play a major role in neuropro-
mediate protein is low in astrocytes under a normal state, tection by releasing neurotrophic factors, and by eliminating
the level increases in astrocytes in the facial nucleus after the glutamate toxicity and oxidative stress.
transection, and the elevated levels continue for a long time
(Graeber and Kreutzberg 1988). Astrocytes in this state are
6.2 I N T E R AC T I O N B ET WE E N M I C RO G L I A
known as reactive astrocytes or activated astrocytes. The func-
A N D A S T RO C Y T E S
tional significance of GFAP in activated astrocytes is associ-
ated with their hypertrophy and lamella formation. Analysis As seen above, both astrocytes and microglia respond to neu-
of GFAP-knockout mice suggested that GFAP is neces- ronal injury, and they interact for the functional restoration of
sary to physically support the cellular construction in astro- neurons and tissue remodeling. The finding that the number
cytes (Liedtke et al. 1996). In addition, a role of GFAP as an of astrocytes in the transected facial nucleus of osteopetrotic
inhibitory barrier against axonal regeneration was reported in mice was less than that of control mice suggests the presence
GFAP/vimentin-deficient mice (Wilhelmsson et al. 2004). of an interaction between astrocytes and microglia (Raivich
In the CNS, astrocytes respond to neuronal damage et al. 1994). Other relevant phenomena include the associa-
including trauma, ischemia, and chemicals, and undergo pro- tion of the microglial cell number with astroglial prolifera-
liferation leading to astrogliosis (Zhang et al. 2010). However, tion in an animal model (Miller and McAllister 2007), and
astrocytes do not proliferate in the transected adult rat facial that the astrogliosis caused by microglia in vitro (Rohl et al.
nucleus. A similar phenomenon is observed in the adult hypo- 2007) leads to an intercellular interaction between astrocytes
glossal nucleus (Svensson et al. 1994). In contrast to the adult, and microglia. Therefore, astrocytes and microglia appear to
in the neonatal rats astrocytes proliferate in response to facial form an activation loop in which one cell type–derived sig-
nerve lesions (Graeber et al. 1998). nal further stimulates the other cell type. The mediators in
Some molecules are known to be promoted in astrocytes in the pathological state are predicted to be soluble molecules
response to neuronal injury. Plasminogen activator inhibitor-1 including proinflammatory cytokines, chemokines, proteases,
(PAI-1), which regulates PA activity, is induced in astrocytes nucleotides, and radicals, although many of the active materi-
in the transected facial nucleus (Reddington et al. 1994). als are not accurately identified yet.
Protein tyrosine phosphatase SHP1 is enhanced in astrocytes There are some reports that astrocytes activate or regu-
as well as microglia in the injured facial nucleus (Horvat et al. late microglial properties/functions. In a study by Rohl and
2001). In the transected hypoglossal nucleus, apolipoprotein J Sievers (2005), astrocytes stimulated microglia to enhance
is augmented in astrocytes (Svensson et al. 1995). Sciatic nerve NO production in a coculture system with trimethyltin, sug-
transection results in the elevation of N-methyl D-aspartate gesting that astrocytes potentiate the trimethyltin-triggered

682 • NEUROGLIA
microglial activation. A certain soluble molecule(s) is specu- At an early time point following injury, the injured motoneu-
lated to be the effective stimulator. Shih et al. (2006) reported rons downregulated function-related molecules such as ChAT
that astrocytes attenuate microglial activation by stimulating and VAchT. Subsequently, the microglia were activated and pro-
their Nrf2-dependent antioxidant gene expressions, includ- liferated. In the facial nerve transection model, enhanced levels
ing heme oxygenase-1, glutathione synthesis enzymes, and of M-CSF were suggested to trigger the induction of cFms and
superoxide dismutase expression, indicating that astrocytes cyclinA/D in microglia to advance the cell cycle. The putative
contribute to the regulation of microglia-triggered inflam- roles of the proliferating microglia were suggested to include
mation. Astrocytes in vitro suppressed the phagocytic activ- serving as neurotrophic/neurosupportive cells, as glutamate
ity of microglia in a coculture system containing senile plaque scavengers to prevent glutamate toxicity, and as a regulator of
cores. The responsible molecules are thought to be diffusible tissue remodeling. On the other hand, in response to motoneu-
factor(s) (DeWitt et al. 1998). ronal death, microglia are activated, proliferate, and become
On the other hand, in some cases microglia were shown phagocytes. These cells were not essentially inflammatory.
to be able to regulate astrocytic properties by releasing sol- In response to neuronal injury, astrocytes also become
uble molecules. Microglia-derived PLGn induced PAI-1 in activated and are suggested to act as neuroprotective cells.
astrocytes in vitro (Liu et al, 2000). Because PLGn and uPA Microglia and astrocytes are suggested to interact intimately
are produced in microglia and PAI-1 is produced in astro- with each other by using certain mediators, and through their
cytes (Reddington et al. 1994), a plasmin-generating system crosstalk, they participate in neurosupportive actions and in
could be regulated through the cellular interaction between the tissue remodeling.
microglia and astrocytes. Similarly, microglia-derived PLGn There are some points to be elucidated in the future. The
enhanced the production of TGFβ3, but not TGFβ1 or mechanism by which M-CSF is upregulated in microglia
TGFβ2, in astrocytes (Maeda et al. 2009). The TGFβs are stimulated with injured neurons is still uncertain. The signifi-
known to regulate the survival of neurons (Makwana et al. cance of proliferating microglia in the injured nervous system
2007). The neuroprotective function of astrocytes might be should be clarified in vivo. Furthermore, the mechanism by
regulated by microglial stimulus in vivo. which microglia change into phagocytes in vivo, and the prop-
erties of phagocytic microglia remain to be elucidated. And
finally, the astrocytic response to motoneuron insult, and the
7 S U M M A RY A N D P E R S P E C T I VE S interaction between astrocytes and microglia in injured ner-
vous system will need to be analyzed in detail. These studies
Peripheral nerve injury leads to motoneuron downregulation, will lead to a better understanding of the role of activated/
microglial activation/proliferation, and astroglial activation proliferating microglia, phagocytic microglia, and activated
in the ipsilateral nucleus. In this chapter, the events occurring astrocytes, and of the cellular interaction between astrocytes
in the injured motor nucleus were summarized (Fig. 53.5). and microglia in the lesioned nervous system.

Activation

Activation
Astrocytes

Proliferation Microglia

Cell death

Microglia

Astrocytes Injured motoneuron Phagocytes


tPA Microglia Ach↓
ChAT↓
PAI-1 uPA VAchT↓ Microglia

PLGn
proTGFβ3 Neurotrophic
NGF action
Plasmin Migration BNDF
NT-4/5
Extracellular Glutamate TGFβ1
proteolysis Microglia scavenger GDNF

Figure 53.5 Cellular Interaction Between Injured Motoneurons and Glial Cells. Activation, proliferation, enhanced neurotrophic action, association
with tissue remodeling of microglia, and astrocyte activation in the injured motor nucleus are depicted. The transformation of microglia into phago-
cytes in response to neuronal cell death is also shown.

I N T E R AC T I O N O F M I C R O G L I A W IT H N EU R O N S A N D A ST R O C Y T E S U N D E R L E S I O N E D N EU R O NA L C O N D IT I O N S • 683
REFERENCES axotomized facial motoneurons: evidence for a sequential postle-
sional function of the cytokines. J Neurobiol 41:559–571.
Aldskogius H, Thomander L. 1986. Selective reinnervation of somatoto- Haas CA, Streit WJ, Kreutzberg GW. 1990. Rat facial motoneurons
pically appropriate muscles after facial nerve transection and regen- express increased levels of calcitonin gene-related peptide mRNA in
eration in the neonatal rat. Brain Res 375:126–134. response to axotomy. J Neurosci Res 27:270–275.
Angelov DN, Neiss WF, Gunkel A, Guntinas-Lichius O, Stennert He C, Wilhelm SM, Pentland AP, Marmer BL, Grant GA, Eisen AZ, et al.
E. 1994. Axotomy induces intranuclear immunolocalization of 1989. Tissue cooperation in a proteolytic cascade activating human
neuron-specific enolase in facial and hypoglossal neurons of the rat. interstitial collagenase. Proc Natl Acad Sci U S A 86:2632–2636.
J Neurocytol 23:218–233. Horvat A, Schwaiger F, Hager G, Brocker F, Sreif R, Knyazev P, et al.
Annes JP, Munger JS, Rifk in D. 2003. Making sense of latent TGFb acti- 2001. A novel role for protein tyrosine phosphatase shp1 in con-
vation. J Cell Sci 116:217–224. trolling glial activation in the normal and injured nervous system.
Bartolini A, Vigliani MC, Magrassi L, Vercelli A, Rossi F. 2011. G-CSF J Neurosci 21:865–874.
administration to adult mice stimulates the proliferation of microglia Ito D, Imai Y, Ohsawa K, Nakajima K, Fukuuchi Y, Kohsaka S. 1998.
but does not modify the outcome of ischemic injury. Neurobiol Dis Microglia-specific localisation of calcium binding protein, Iba1. Mol
41:640–649. Brain Res 57:1–9.
Blasi F, Vassali JD, Dano K. 1987. Urokinase-type plasminogen activa- Iwasaki Y, Ikeda K, Shiojima T, Kinoshita M. 1995. CNQX prevents spi-
tor: Proenzyme, receptor, and inhibitors. J Cell Biol 104:801–804. nal motor neuron death following sciatic nerve transection in new-
Blinzinger K, Kreutzberg GW. 1968. Displacement of synaptic termi- born rats. J Neurol Sci 134:21–25.
nals from regenerating motoneurons by microglial cells. Zellforsch Kiefer R, Lindholm D, Kreutzberg GW. 1993. Interleukin-6 and trans-
Mikrosk Anat, 85:145–157. forming growth factor-b 1 mRNA are induced in rat facial nucleus
Brusilow SW, Koehler RC, Traystman RJ, Cooper AJ. 2010. Astrocyte following motoneuron axotomy. Eur J Neurosci 5:775–781.
glutamine synthetase: importance in hyperammonemic syndromes Kinchen JM, Ravichandran KS. 2008. Phagosome maturation: going
and potential target for therapy. Neurotherapeutics 7:452–470. through the acid test. Nat Rev Mol Cell Biol 9:781–795.
Burazin TC, Gundlach AL. 1998. Up-regulation of GDNFR-alpha and Kiryu-Seo S, Sasaki M, Yokohama H, Nakagomi S, Hirayama T, Aoki S.,
c-ret mRNA in facial motor neurons following facial nerve injury in et al. 2000. Damage-induced neuronal endopeptidase (DINE) is
the rat. Mol Brain Res 55:331–336. a unique metallopeptidase expressed in response to neuronal dam-
Calvo M, Zhu N, Tsantoulas C, Ma Z, Grist J, Loeb JA, et al. 2010. age and activates superoxide scavengers. Proc Natl Acad Sci U S A
Neuregulin-ErbB signaling promotes microglial proliferation and 97:4345–4350.
chemotaxis contributing to microgliosis and pain after peripheral Kobayashi NR, Bedard AM, Hincke MT, Tetzlaff W. 1996. Increased
nerve injury. J Neurosci 30:5437–5450. expression of BDNF and trkB mRNA in rat facial motoneurons after
Damoiseaux JG, Döpp EA, Calame W, Chao D, MacPherson GG, axotomy. Eur J Neurosci 8:1018–1029.
Dijkstra CD. 1994. Rat macrophage lysosomal membrane antigen Koguchi K, Nakatsuji Y, Okuno T, Sawada M, Sakoda S. 2003. Microglial
recognized by monoclonal antibody ED1. Immunology 83:140–147. cell cycle-associated proteins control microglial proliferation in vivo
Dano K, Adreasen PA, Grondahl-Hansen J, Kristensen P, Nielsen LS, and in vitro and are regulated by GM-CSF and density-dependent
Skriver L. 1985. Plasminogen activators, tissue degradation, and can- inhibition. J Neurosci Res 74:898–905.
cer. Cancer Res 44:139–226. Koizumi S, Shigemoto-Mogami Y, Nasu-Tada k, Shinozaki Y, Ohsawa
DeWitt DA, Perry G, Cohen M, Doller C, Silver J. 1998. Astrocytes K, Tsuda M, et al. 2007. UDP acting at P2Y6 receptors is a mediator
regulate microglial phagocytosis of senile plaque cores of Alzheimer’s of microglial phagocytosis. Nature 446;1091–1095.
disease. Exp Neurol 149:329–340. Konishi H, Namikawa K, Kiyama H. 2006. Annexin III implicated in
Dong Y, Benveniste EN. 2001. Immune function of astrocytes. Glia the microglial response to motor nerve injury. Glia 53:723–732.
36:180–190. Kreutzberg GW. 1996. Microglia: a sensor for pathological events in the
Elward K, Gasque P. 2003. “Eat me” and “don’t eat me” signals govern the CNS. Trend Neurosci 19:312–318.
innate immune response and tissue repair in the CNS: emphasis on Kreutzderg GW, Graeber MB, Streit W. 1989. Neuron-glial relation-
the critical role of the compliment system. Mol Immunol 40:85–94. ship during regeneration of motorneurons. Metabolic Brain Diseases
Fischer PM, Endicott J, Meijer L. 2002. Cyclin-dependent kinase inhibi- 4:81–85.
tors. Prog Cell Cycle Res 5:235–248. Lalancette-Hebert M, Gowing G, Simard A, Weng YC, Kriz J. 2007.
Ganter G, Northoff H, Männel D, Gebicke-Härter. 1992. Growth con- Selective ablation of proliferating microglial cells exacerbates isch-
trol of cultured microglia. J Nerurosci Res 33:218–230. emic injury in the brain. J Neurosci 27:2596–2605.
Gesase AP, Kiyama H. 2000. Morphological changes and expression of Lee SC, Liu W, Brosnan CF, Dickson DW. 1994. GM-CSF promotes
protein kinase CK2b subunit in the microglia after hypoglossal nerve proliferation of human fetal and adult microglia in primary cultures.
transection. J Neurocytol 29:61–66. Glia 12:309–318.
Giulian D, Ingleman JE. 1988. Colony-stimulating factors as promoters Liedtke W, Edelmann W, Bieri PL, Chiu FC, Cowan NJ, Kucherlapati
of ameboid microglia. J Neurosci 8:4707–4717. R, et al. 1996. GFAP is necessary for the integrity of CNS white mat-
Graeber MB, Kreutzberg GW. 1986. Astrocytes increase in glial fibril- ter architecture and long-term maintenance of myelination. Neuron
lary acidic protein during retrograde changes of facial motor neurons. 17:607–615.
J Neurocytol 15:363–373. Liu Y-M, Honda S, Kohsaka S, Nakajima K. 2000. Plasminogen enhances
Graeber MB, Kreutzberg GW. 1988. Delayed astrocyte reaction follow- the secretion of plasminogen activator-1 (PAI-1) from cultured rat
ing facial nerve axotomy. J Neurocytol 17:209–220. astrocytes. Neurosci Lett 282:137–140.
Graeber MB, Lopez-Redondo F, Ikoma E, Ishikawa M, Imai Y, Nakajima Liva SM, Kahn MA, Dopp JM, de Vellis J. 1999. Signal transduction
Ket al. 1998. The microglia/macrophage response in the neonatal rat pathways induced by GM-CSF in microglia: significance in the con-
facial nucleus following axotomy. Brain Res 813:241–253. trol of proliferation. Glia 26:344–352.
Graeber MB, Streit WJ, Kreutzberg GW. 1989. Formation of Llewellyn-Smith IJ, Martin CL, Arnolda LF, Minson JB. 2000.
microglia-derived brain macrophages is blocked by adriamycin. Acta Tracer-toxin: cholera toxin B-saporin as a model. J Neurosci Methods
Neuropathol 78:348–358. 103:83–90.
Haas CA, Donath C, Kreutzberg GW. 1993. Differential expression of Lopez-Redondo F, Nakajima K, Honda S, Kohsaka S. 2000. Glutamate
immediate early genes after transection of the facial nerve. Neurosci transporter GLT-1 is highly expressed in activated microglia follow-
53:91–99. ing facial nerve axotomy. Mol Brain Res 76:429–435.
Haas CA, Hofmann HD, Kirsch M. 1999. Expression of CNTF/ Lu B, Pang PT, Woo NH. 2005. The yin and yang of neurotrophin
LIF receptor components and activation of STAT3 signaling in action. Nature Rev Neurosci 6:603–614.

684 • NEUROGLIA
Lyons A, Downer EJ, CXrotty S, Nolan YM, Mills KH, Lynch MA. Nishiyori A, Minami M, Ohtani Y, Takami S, Yamamoto J, Kawaguchi
2007. CD200 ligand receptor interaction modulates microglial acti- N, et al. 1998. Localization of fractalkine and CX3CR1 mRNAs in
vation in vivo and in vitro: a role for IL-4. J Neurosci 27:8309–8313. rat brain: fractalkine play a role in signaling from neuron to micro-
Maeda S, Nakajima K, Tohyama Y, Kohsaka S. 2009. Characteristic glia? FEBS Lett 429:167–172.
response of astrocytes to plasminogen/plasmin to upregulate trans- Olsson T, Kristensson K, Ljungdahl A, Maehlen J, Holmdahl R,
forming growth factor beta 3 (TGFbeta3) production/secretion Klareskog L. 1989. Gamma-interferon-like immunoreactivity in axo-
through proteinase-activated receptor-1 (PAR-1) and the down- tomized rat motor neurons. J Neurosci 9:3870–3875.
stream phosphatidylinositol 3-kinase (PI3K)-Akt/PKB signaling Oppenheim RW, Houenou LJ, Parsadanian AS, Prevette D, Snider WD,
cascade. Brain Res 130:1–13. Shen L. 2000. Glial cell line-derived neurotrophic factor and develop-
Makwana M, Jones LL, Cuthill D, Heuer H, Bohatschek M, Hristova ing mammalian motoneurons: regulation of programmed cell death
M, et al. 2007. Endogenous transforming growth factor beta 1 sup- among motoneuron subtypes. J Neurosci 20:5001–5011.
presses inflammation and promotes survival in adult CNS. J Neurosci Oshiro S, Kawamura K, Zhang C, Sone T, Morioka SM, Kobayashi S,
27:11201–11213. et al. 2008. Microglia and astroglia prevent oxidative stress-induced
Mander PK, Jekabsone A, Brown GC. 2006. Microglia proliferation is neuronal cell death: Implications for aceruloplasminemia. Biochim
regulated by hydrogen peroxide from NADPH oxidase. J Immunol Biophys Acta 1782:109–117.
176: 1046–1052. Perry VH, O’Connor V. 2010. The role of microglia in synaptic strip-
McNamara RK, Jiang Y, Streit WJ, Lenox RH. 2000. Facial motor neu- ping and synaptic degeneration: a revised perspective. ASN Neuro
ron regeneration induces a unique apatial and temporal pattern of 2:281–291.
myristoylated alanine-rich C kinase substrate expression. Neurosci Popratiloff A, Kharazia VN, Weinberg RJ, Laonipon B, Rustioni A.
97:581–589. 1996. Glutamate receptors in spinal motoneurons after sciatic nerve
McNaugt KS, Jenner P. 2000. Extracellular accumulation of nitric transection. Neurosci 74:953–958.
oxide, hydrogen peroxide, and glutamate in astrocytic cultures Raivich G, Gehrmann J, Kreutzberg GW. 1991. Increase of mac-
following glutathione depletion, complex I inhibition, andor rophage colony-stimulating factor and granulocyte-macrophage
lipopolysaccharide-induced activation. Biochem Pharmacol colony-stimulating factor receptors in the regenerating rat facial
60:979–988. nucleus. J Neurosci Res 30:682–686.
Miller JM, McAllister JP. 2007. Reduction of astrogliosis and microglio- Raivich G, Moreno-Flores M, Moller JC, Kreutzberg GW. 1994.
sis by cerebrospinal fluid shunting in experimental hydrocephalus. Inhibition of posttraumatic microglial proliferation in a genetic
Cerebrospinal Fluid Res 4:5 model of macrophage colony-stimulating factor deficiency in the
Moran LB, Graeber MB. 2004. The facial nerve axotomy model. Brain mouse. Eur J Neurosci 6:1615–1618.
Res Rev 44:154–178. Reddington M, Haas CA, Kreutzberg GW. 1994. The plasminogen acti-
Morita N, Kiryu S, Kiyama H. 1996. p53-independent cyclin G expres- vator system in neurons and glia during motoneuron regeneration.
sion in a group of mature neurons and its enhanced expression during Neuropathol Appl Neurobiol 20:188–190.
nerve regeneration. J Neurosci 16:5961–5966. Rishal I, Fainzilber M. 2010. Retrograde signaling in axonal regenera-
Nakagawa H, Hatakeyama S, Ikesue A, Miyai H. 1991. Generation of tion. Exp Neurol 223:5–10.
interleukin-8 by plasmin from AVLPR-interleukin-8, the human Rohl C, Lucius R, Sievers J. 2007. The effect of activated microglia on
fibroblast-derived neutrophil chemotactic factor. FEBS Lett astrogliosis parameters in astrocyte cultures. Brain Res 1129:43–52.
282:412–414. Rohl C, Sievers J. 2005. Microglia is activated by astrocytes in trimethyl-
Nakajima K, Kohsaka S. 1996. Functional implication of microglia-derived tin intoxication. Toxicol Appl Pharmacol 204:36–45.
secretory proteases. In: Ling EA, Tan CK, Tan CBC (eds.), Topical Ross FP. 2006. M-CSF, c-Fms, and signaling in osteoclasts and their pre-
issues in microglial research. Singapore: Singapore Neuroscience cursors. Ann NY Acad Sci 1068:110–116.
Association, pp. 203–218. Schmitt AB, Breuer S, Liman J, Buss A, Schlangen C, Pech K, et al.
Nakajima K, Kohsaka S. 2004. Microglia: neuroprotective and neu- 2003. Identification of regeneration-associated genes after central
rotrophic cells in the central nervous system. Curr Drug Targets and peripheral nerve injury in the adult rat. BMC Neurosci 4:8.
Cardiovasc Haematol Disord 4:65–84. Schwaiger FW, Hager G, Raivich G, Kreutzberg GW. 1998. Cellular
Nakajima K, Reddington M, Kohsaka S, Kreutzberg GW. activation in neuroregeneration. Prog Brain Res 117:197–210.
1996. Induction of urokinase-type plasminogen activator in rat Sherr CJ. 1993. Mammalian G1 cyclins. Cell 73:1059–1065.
facial nucleus by axotomy of the facial nerve. J Neurochem 66: Shih AY, Fernandes HB, Choi FY, Kozoriz MG, Liu Y, Li P, et al. 2006.
2500–2505. Policing the police: Astrocytes modulate microglial activation.
Nakajima K, Tohyama Y, Kohsaka S, Kurihara,T. 2001. Ability of J Neurosci 26:3887–3888.
rat microglia to uptake extracellular glutamate. Neurosci Lett Snider WD, Thanedar S. 1989. Target dependence of hypoglossal
307:171–174. motor neurons during development in maturity. J Comp Neurol
Nakajima K, Tohyama Y, Kurihara T, Kohsaka S. 2004. 279:489–498.
Axotomy-dependent urokinase induction in the rat facial nucleus: Streit WJ. 2002. Microglia as neuroprotective, immunocompetent cells
possible stimulation of microglia by neurons. Neurochem Int of the CNS. Glia 40:133–139.
46:107–116. Streit WJ, Graeber MB, Kreutzberg GW. 1988. Functional plasticity of
Nakajima K, Tohyama Y, Kurihara T, Kohsaka S. 2005. Enhancement of microglia: A review. Glia 1:301–307.
urokinase-type plasminogen activator (uPA) secretion, but not that Streit WJ, Kreutzberg GW. 1988. Response of endogenous glial cells to
of substrate plasminogen (PGn), by rat microglia stimulated with motor neuron degeneration induced by toxic ricin. J Comp Neurol
neuronal conditioned medium. Neurosci Lett 378:13–17. 268:248–263.
Nakajima K, Tohyama Y, Maeda S, Kohsaka S, Kurihara T. 2007. Suh H-S, Kim M-O, Lee SC. 2005. Inhibition of granulocyte-macrophage
Neuronal regulation by which microglia enhance the production of colony-stimulating factor signaling and microglial proliferation by
neurotrophic factors for GABAergic, catecholaminergic, and cholin- anti-CD45RO: role of Hck tyrosine kinase and phophatidylinositol
ergic neurons. Neurochem Int 50:807–820. 3-kinase/Akt. J Immunol 174:2712–2719.
Nakajima K, Tsuzaki N, Nagata K, Takemoto N, Kohsaka S. 1992. Svensson M, Aldskogius H. 1993. Synaptic density of axotomized hypo-
Production and secretion of plasminogen in cultured rat brain micro- glossal motoneurons following pharmacological blockade of the
glia. FEBS Lett 308:179–182. microglial cell proloferation. Exp Neurol 120:123–131.
Nakajima K, Yamamoto S, Kohsaka S, Kurihara T. 2008. Neuronal stim- Svensson M, Liu L, Mattsson P, Morgan BP, Aldskogius H. 1995.
ulation leading to upregulation of glutamate transporter-1 (GLT-1) in Evidence for activation of the terminal pathway of complement and
rat microglia in vitro. Neurosci Lett 436:331–334. upregulation of sulfated glycoprotein (SGP)-2 in the hypoglossal

I N T E R AC T I O N O F M I C R O G L I A W IT H N EU R O N S A N D A ST R O C Y T E S U N D E R L E S I O N E D N EU R O NA L C O N D IT I O N S • 685
nucleus following peripheral nerve injury. Mol Chem Neuropathol Yamamoto S, Kohsaka S, Nakajima K. 2012. Role of cell cycle-associated
24:53–68. proteins in microglial proliferation in the axotomized rat facial
Svensson M, Mattsson P, Aldskogius H. 1994. A bromodeoxyuridine nucleus. Glia 60:570–581
labeling study of proliferating cells in the brainstem following hypo- Yamamoto S, Nakajima K, Kohsaka S. 2010. Macrophage-colony stimu-
glossal nerve transection. J Anat 185:537–542. lating factor as an inducer of microglial proliferation in axotomized
Tetzlaff W, Kreutzberg GW. 1984. Enzyme changes in the rat facial rat facial nucleus. J Neurochem 115:1057–1067.
nucleus following a conditioning lesion. Exp Neurol 85:547–564. Zhang Q, Chen C, Lü J, Xie M, Pan D, Luo X, et al. 2009. Cell cycle
Tsuda M, Masuda T, Kitano J, Shimoyama H, Tozaki-Saitoh H, Inoue K. inhibition attenuates microglial proliferation and production of
2009. IFN-g receptor signaling mediates spinal microglia activation IL-1beta, MIP-1alpha, and NO after focal cerebral ischemia in the
driving neuropathic pain. Proc Natl Acad Sci U S A 106:8032–8037. rat. Glia 57:908–920.
Wiktor-Jedrzejczak W, Bartocci A, Ferrante Jr AW, Ahmed-Ansari A, Zhang D, Hu X, Qian L, O’Callaghan JP, Hong J-S. 2010. Astrogliosis
Sell KW, Pollard JW, et al. 1990. Total absence of colony-stimulating in CNS pathologies: is there a role for microglia? Mol Neurobiol
factor 1 in the macrophage-deficient osteopetrotic (op/op) mouse. 41:232–241.
Proc Natl Acad Sci U S A 87:4828–4832. Zhao Z, Alam S, Oppenheim RW, Prevette DM, Evenson A, Parsadanian
Wilhelmsson U, Li L, Pekna M, Berthold CH, Blom S, Eliasson C, et al. A. 2004. Overexpression of glial cell line-derived neurotrophic factor
2004. Absence of glial fibrillary acidic protein and vimentin prevents in the CNS rescues motoneurons from programmed cell death and
hypertrophy of astrocytic processes and improves posttraumatic promotes their long-term survival following axotomy. Exp Neurol
regeneration. J Neurosci 24:5016–5021. 190:356–372.

686 • NEUROGLIA
54.
SCHWANN CELLS AND INJURY
Violetta Zujovic and Alexandros A. Lavdas

A B B R E VI AT I O N S of immune response, the support of axonal regeneration, and an


important actor in peripheral and central regeneration processes.
ADAM a disintegrin and metalloprotease Furthermore, we will also give an overview of all the pathologies
BACE β-site amyloid precursor protein cleaving that result from SC dysfunction.
enzyme
BDNF brain-derived neurotrophic factor
BM bone marrow 2 D E VE L O PM E N T
BMP bone morphogenic protein
Ccnd1 cyclinD1 Because a specific chapter (chapter 14) is dedicated to SC devel-
CMT Charcot-Marie-Tooth disease opment, we will give a general brief overview of all the features
CNS central nervous system of SC development that will be useful in understanding the pro-
DRG dorsal root ganglia cesses SCs go through during nerve degeneration-regeneration.
EAE experimental allergic encephalomyelitis
FGF fibroblast growth factor 2.1 T R A NS C R I P T I O N FAC TO R S
GFP green fluorescent protein
IFN interferon The embryonic phase of SC development involves three major
IL interleukin transitions. The transition from migrating neural crest cells to
LPS lipopolysaccharide SC precursor, the differentiation of SC precursors into imma-
MHC major histocompatibility complex ture SCs, and their final maturation to either myelinating or
miRNA micro RNA nonmyelinating SCs.
MS multiple sclerosis The time and space regulation of cell progression through SC
MOG myelin oligodendrocyte glycoprotein lineage is based on the balance of expression of transcription fac-
NGF nerve growth factor tors that can be divided in two groups, the negative and the posi-
NRG neuregulin tive regulators of myelination. Transcription factors such as Sox
NT neurotrophin (SRY-related HMG-box) 2, c-jun, Pax 3, Id2, Id 4, and Notch
OL oligodendrocytes signaling are designated as negative regulators, and Sox 10, Oct 6,
P0 peripheral myelin protein 0 and Krox 20 as positive regulators (see Fig. 54.1; chapter 14; and
PI-BMDC proinsulin-producing bone marrow derived Jessen and Mirsky 2008).
cells Recent data report that SC differentiation is also regulated
PLA2 phospholipase A2 at the epigenetic level. When micro RNA (miRNA), small non-
PNS peripheral nervous system coding RNA that bind to target RNA and inhibit their tran-
SC Schwann cell scription, are deleted in SCs by the specific ablation of DICER,
TACE TNFα-converting enzyme the SCs are “stuck” at the promyelinating stage (Yun et al. 2010).
TGF tumor growth factor Negative regulators of myelination, such as Sox 2, c-jun, and
TLR Toll-like receptors cyclin D1 are upregulated in SC-dicer mutants, whereas Krox 20
TNF tumor necrosis factor is dramatically downregulated (Yun et al. 2010). Thus, miRNA-
YFP yellow fluorescent protein 138 silencing results in an increase of c-jun and Sox 2. It appears
that miRNA are involved in promoting and stabilizing the dif-
ferentiated SC phenotype (Wienholds and Plasterk 2005).
1 INTRODUCTION
2.2 AXO NA L S I G NA L I N G
Schwann cells, the myelinating cells of the peripheral nervous
system (PNS), have attracted intense interest in the last decades, During development, the control of PNS myelination is
both because of their role in various pathological conditions and instructed by axonal signals, the most prominent being the neu-
because of their contribution to nervous system regeneration. regulins (NRGs), which are crucial at several stages of SC dif-
In this chapter, we document the numerous pleiotropic roles of ferentiation process (Jessen and Mirsky 2005). Neuregulins are
Schwann cells (SCs) particularly in repair processes such as being a family of growth factors with four different isoforms that act
a sensor of axonal integrity and injury, the activator and regulator through erbB tyrosine kinase receptors (Birchmeier and Nave
687
Figure 54.1 The balance between positive (red) and negative (green) regulators of SC program directs SC fate toward a myelinated or un/dedifferenti-
ated state. Sox 10 and Oct6 act in synergy to induce the expression of Krox 20 (purple arrow). High Krox 20 expression results in the triggering of the
myelination process along with the inhibition of negative regulators (yellow arrow). However after an injury, the reexpression of negative regulator
redirects SCs to an undifferentiated state. Micro RNA(miR)s stabilize the differentiated phenotype by inhibiting the expression of several negative
regulators and controlling the levels of Krox20.

2008). Axonally derived NRG1 signals via erbB2 and erbB3 phagocytose extracellular debris resulting from dying or dam-
receptor heterodimers present on cells of the SC lineage and is aged cells by activating the phospholipase A2 family of enzymes
required for SC precursor emergence, migration into the periph- (Martini et al. 2008). Another protease, the matrix metallopro-
eral nerve, and survival before differentiation into immature SCs tease 9, is also expressed by SCs after peripheral nerve injury and
(see chapter 14). is involved in both leukocyte migration and myelin breakdown
(Kobayashi et al. 2008).

3 S C H WA N N C E L L S ’ R O L E I N P N S I N J U RY
3.1.2 Dedifferentiation/Peripheral
3.1 T H E MU LT ITA S K I N G AC T I VIT Y O F Nervous System Stem Cells
S C H WA N N C E L L S I N P NS I N JU RY As in development, SC dedifferentiation is dependent on the
Schwann cells are the first sensors of axonal damage and balance between positive and negative regulators of myelina-
react quickly to peripheral nerve injury by orchestrating the tion (see Fig. 54.1). Upon nerve injury, negative regulators of
immune response, clearing myelin debris, supporting axonal myelination such as Notch1, Ccnd1, c-jun, Sox2, Id 4, and Id2
regeneration, and finally remyelinating regenerated axons (for are upregulated and drive SC dedifferentiation to a phenotype
overview see Fig. 54.2). similar to, but distinct from, that of immature SCs (Mirsky
et al. 2008; also chapter 14). One of the first negative regula-
tors of myelination to have been described is c-jun. The specific
3.1.1 Phagocytic Activity/Immune Mediation
deletion of c-jun in SCs leads to delay in SC dedifferentiation,
Schwann cell reaction to PNS injury is mediated through the phagocytic activity, and a loss of axon regenerative ability.
activation of Toll-like receptors (TLRs). The family of TLRs Sharing the same expression pattern as c-jun is sox2, which is
belongs to the group of pattern-recognition receptors that recog- also upregulated after injury. In a very elegant study, Woodhoo
nize specific conserved components of microorganisms (Takeda et al. (2009) successfully deciphered the role of Notch signal-
et al. 2003) and they play a critical role in various inflammatory ing in the SC dedifferentiation process. This study notably
disorders (Cook et al. 2004). Indeed, SCs express a variety of provided evidence that when Notch is ectopically activated in
TLRs such as TLR2, TLR3, TLR4, TLR7 constitutively, and noninjured nerve, it is sufficient to induce myelin breakdown
TLR1, which is upregulated after nerve axotomy (Goethals et and provoke SC dedifferentiation. It is the first study to dem-
al. 2010). Axonal injury results in the liberation of TLR ligand onstrate that overexpression of a negative regulator is sufficient
that will in turn activate the production by SCs of cytokines to overcome Krox 20 inhibition and control SC plasticity.
and chemokines that will amplify and fine tune the inflamma- The injury related changes that occur in dedifferentiated
tory response (Pineau et al. 2009). Indeed, the release of tumor SCs are also regulated at the epigenetic level. Indeed, immedi-
necrosis factor (TNF)α, interleukin (IL)α, ILβ, and CCL2 ately after injury most SC miRNAs are downregulated (Viader
and CCL3 (Perrin et al. 2005) by SCs will trigger macrophage et al. 2011). This allows the transcription of negative regula-
recruitment to the site of injury, macrophages that will support tors of myelination and the engagement of the dedifferentia-
SC action in clearing myelin debris. tion process. When Dicer expression is specifically deleted in
Denervated SCs are the major phagocytic cells for the first SCs, there is a delay in the transition from myelinating SCs
days after injury. They can degrade their own myelin but also to dedifferentiated SCs and from dedifferentiated SCs to

688 • ROLE OF GLIAL CELLS IN DISEASE


Figure 54.2 Overview of SC Participation in PNS Regeneration. After peripheral injury, SCs dedifferentiate, exert phagocytic activity, and regulate the
immune response. Once myelin and axonal debris are cleared, they form bands of Büngner, columns of cells that recreate a microenvironment permis-
sive for axonal regeneration. It is only when the axons have reinnervated their targets that SCs reacquire a remyelinating phenotype.

remyelinating SCs; indicating that SCs respond less efficiently and neuronal cytoskeleton through their interaction with their
to injury when miRNA are absent. Among the miRNA high- receptors (integrins, dystroglycan) present at the growth cone.
lighted in this study, miR-34a and miR-140 represent interest- Fibronectin is expressed at low levels in the SC basement mem-
ing targets. Note that miR-34 interacts and inhibits cyclinD1 brane, but is rapidly upregulated following PNS injury (Lefcort
(Ccnd1) transcription and interferes with Notch signaling, et al. 1992). Fibronectin acts as a substratum for migrating
whereas miR-140 maintains Krox 20 expression to a specific SCs (Baron-Van Evercooren et al. 1982b) and provides also
functional level. a direct substrate for adhesion and outgrowth of regenerat-
While the dedifferentiation process of myelinating SCs ing axons (Gardiner 2011). Schwann cells produce a specific
has been thoroughly studied, one should consider also the embryonic isoform of fibronectin, that is best fitted to interact
participation of PNS stem cells in the regenerative process. with α4β1 integrins present in the growth cone of regenerating
In recent years, two studies provided evidence for the per- axons (Baron-Van Evercooren et al. 1982b; Vogelezang et al.
sistence of neural crest–derived stem cells in adult dorsal 2001). Other ECM molecules that are upregulated after injury
root ganglia (DRG) (Li et al. 2007; Nagoshi et al. 2008). include the laminins. Two types of laminins are expressed in
Using lineage tracing with peripheral myelin protein 0 (P0) the intact nerve, laminin 2 composed of the α2β1γ1 subunits
and Wnt1-Cre/Floxed EGFP mice, Nagoshi and colleagues and laminin 8 composed of the α4β1 γ1 subunits (Patton et
demonstrated that the DRG-derived neural crest stem cells al. 1997); but after peripheral injury an increase of the α2, α4,
showed a great ability to form secondary spheres and a high β1, and γ1 chains is observed (Wallquist et al. 2002). Their
multipotency, giving rise to both neuronal and glial cells. essential role in nerve regeneration was shown by a series of
However their participation in the PNS repair process blocking experiments using function-blocking antibodies for
remains to be assessed. α2 laminin (Agius and Cochard 1998) and conditional knock
out for the γ1 laminin subunit (Chen and Strickland 2003),
both resulting in the inhibition of axonal growth (Gardiner
3.1.3 Axonal Growth Support: Extracellular
2011). But successful axonal regeneration is also regulated by
Matrix/Neurotrophins
the finely tuned timely expression of laminin receptors on
Next, the dedifferentiated SCs form Büngner bands and growth cone and SCs. Thus, following injury, α6β4 integrin
express surface molecules that guide regenerating fibers. The is down-regulated on SCs (Feltri et al. 1994) while α1β1 and
ability of SCs to support axonal regeneration is because of mul- α5β1 are induced in growth cone and SCs during the axonal
tiple factors. First, SCs produce extracellular matrix molecules regeneration phase (Lefcort et al. 1992; Siironen et al. 1992);
such as laminin and fibronectin that promote axonal growth conversely the reexpression of α6β4 integrin is correlated to
(Baron-Van Evercooren et al. 1982a). These molecules provide the remyelination process and full assembly of SC basal lamina
a molecular link between the extracellular matrix (ECM), SCs, (Feltri et al. 1994).

S C H WA N N C E L L S A N D I N J U RY • 689
Furthermore, SCs support axonal growth also by expressing regulatory element binding protein (SREBP). Leblanc and col-
multiple neurotrophins including nerve growth factor (NGF), leagues demonstrated that in SCs, Krox20 and SREBP trans-
neurotrophin-3 (NT3), brain-derived neurotrophic factor activators control several target gene promoters such as HMG
(BDNF), fibroblast growth factor (FGF), glial cell line-derived CoA reductase, a gene essential for cholesterol biosynthesis
neurotrophic factor and ciliary neurotrophic factor (Lavdas (Leblanc et al. 2005). Its role in myelination is further con-
et al. 2008), all known to be essential for axonal growth dur- firmed by SC-specific deletion of SREBP activation (Verheijen
ing development. After nerve injury, dedifferentiated SCs of the et al. 2009) and mutations perturbing cholesterol biosynthesis
distal nerve stump upregulate the synthesis of NGF, BDNF, and result in severe hypomyelination (Saher et al. 2009).
NT-4 but not NT-3 (Funakoshi et al. 1993). Neurotrophins are
able to promote axonal regrowth from proximal nerve stumps 3.2 S C H WA N N C E L L S I N P E R I P H E R A L N E RVO US
individually as well as in synergistic combinations (Cao and S YS T E M D E MY E L I NAT I N G D I S O R D E R S
Shoichet 2003). These molecules act through two different
types of receptors: high affinity Trk receptor tyrosine kinases, 3.2.1 Autoimmunity Related: Inflammation
expressed more prominently on axons, and low affinity p75 and Cytokines
neurotrophin receptors, which are also present on SCs (Chao Human inflammatory neuropathies are the results of an
2003). Although dedifferentiated SCs produce these growth immune attack against PNS autoantigens. According to the
factors, they also reexpress p75 after nerve injury (Funakoshi currently accepted model, even under normal conditions
et al. 1993), bind some of this protein, and in this way control there may be autoreactive T lymphocytes recognizing
their concentration and availability to the axons. To act prop- epitopes on the peripheral nerve, but their continuous
erly, the effect of neurotrophins has to be finely regulated both suppression ensures self-tolerance. During an infection,
in time and in quantity. For example, at low doses BDNF medi- these lymphocytes become activated upon encountering
ates its effects via both p75 and Trk receptor, but when high microbial epitopes. These epitopes can resemble endogenous
doses of BDNF are present the neurotrophin will preferentially peripheral nerve antigens, a situation termed molecular
signal through the p75 receptor, whose activation will inhibit mimicry (Yuki et al. 1993). For instance, in patients with
axonal growth (Gordon et al. 2011) (see chapter 55). Guillain–Barré syndrome, one of the most serious forms
of autoimmune neuropathies, epitopes shared between
Campylobacter jejuni, cytomegalovirus, or Haemophilus
3.1.4 Remyelination
influenzae and nerve fibers have been identified (Kieseier
Neuregulin-1 plays an essential role in the neuroregenerative et al. 2004). In this situation, autoreactive T lymphocytes
process. After peripheral nerve injury, there is dysregulation of stimulate B cells to produce autoantibodies, which in turn
the NRG1 isoform and erbB receptors (Nicolino et al. 2009) that block nerve conduction, activate complement, and trigger
might be one of the signals that instruct myelinating SCs to revert macrophage attack against peripheral nerve constituents
to a more immature phenotype (Guertin et al. 2005). An elegant (Sawant-Mane et al. 1996; Takigawa et al. 2000). Activated
recent study (Fricker et al. 2011) demonstrated that the targeted T cells cross the blood-nerve barrier and provoke local
deletion of axonal NRG1 results in slower axonal regeneration, inflammation by proinflammatory cytokines (Kieseier et al.
defects in remyelination, and incorrect reinnervation. The 2006). Attracted macrophages act as antigen-presenting cells,
amount of functional NRG1 present at the axonal surface release toxic mediators, and directly damage myelinating
regulates the myelination and remyelination process. Interestingly, SCs and axons (Kiefer et al. 2001). Functional consequences
two members of a disintegrin and metalloprotease (ADAM) are mainly determined by the degree of axonal degeneration.
secretase family have been implicated in the regulation of NRG1 The immune response may eventually be terminated by
type III activity notably by regulating its processing. The levels increased T-cell apoptosis, as shown in animal models of
of active NRG1 are regulated by proteolysis by β secretase β-site inflammatory neuropathies (Gold et al. 1997). Why has the
amyloid precursor protein cleaving enzyme (BACE) 1 and the problem of molecular mimicry not been resolved through
α-secretase TNFα-converting enzyme (TACE), which both evolutionary selective processes? Having to defend the
cleave NRG1type III. These two secretases have different cleavage body from as many invaders as possible, the immune system
sites and have an opposite effect on myelination because BACE1 works like a double-edged sword. It may be that the current
is a positive regulator of myelination and remyelination (Hu situation represents the optimum balance between response
et al. 2006; Shirakabe et al. 2001), whereas TACE is a negative against invading pathogens and autoimmunity.
regulator; these enzymes represent an interesting therapeutic Toll-like receptors are usually found on antigen-presenting
target because reducing TACE activity might increase the cells, such as dendritic cells, but have also been found on SCs
remyelination process (La Marca et al. 2011). (see 3.1.1). One of these molecules, TLR-2, expressed on pri-
To engage the remyelination process, the induction of mary human and rat SCs, is considered to be a target recep-
myelin genes has to be coordinated with high levels of lipid tor for Mycobacterium leprae (Harboe et al. 2005). Expression
synthesis required for the production of multi-layered choles- of various TLRs is inducible on rat SCs in vitro. For example,
terol rich membranes. Gene expression profiling revealed that stimulation of TLR-4 on SCs with lipopolysaccharides (LPS)
various lipid biosynthesis genes are upregulated after peripheral elicits the production of inflammatory mediators such as pro-
nerve injury (Verheijen et al. 2003). One of the key regulators tease inhibitors, chemokines, and growth factors (Qin et al.
of fatty acid and cholesterol biosynthesis in SCs is the sterol 2012; Shamash et al. 2002), suggesting that SCs can detect LPS

690 • ROLE OF GLIAL CELLS IN DISEASE


fragments and may act as a link between innate and acquired Fas on T cells with its ligand, FasL, a member of the TNF
immunity via TLR-4 activation in the inflamed PNS. family (Krammer 2000). This ligand has been shown to be
The presentation of antigens of cell-associated microorgan- expressed on SC surface after stimulation with proinflamma-
isms for recognition by T lymphocytes is performed by mol- tory cytokines in vitro, and functional analysis has suggested
ecules of the major histocompatibility complex (MHC). Two that the interaction between Fas on T cells and FasL on SCs
types of MHC gene products can be distinguished, class I and can promote apoptosis of T lymphocytes. Interestingly, FasL
class II MHC molecules, which differ both functionally and can also act as a signal-transducing molecule in SCs, leading to
in terms of cell expression, with all nucleated cells exhibiting the secretion of NGF, and may thus contribute to peripheral
MHC class I molecules and only macrophages, dendritic cells, nerve regeneration (Mimouni-Rongy et al. 2011).
and B lymphocytes expressing MHC class II molecules. The
levels of both class I and II molecules can be upregulated by
3.2.2 Myelin Malformation-Related: Genetic Causes
cytokines, particularly interferon (IFN)-γ and TNF-α. Other
cell types may acquire the ability to process and present anti- Charcot-Marie-Tooth (CMT) disease is a term used for
gens in inflammatory conditions, thus representing facultative inherited peripheral neuropathies, after the names of the three
antigen-presenting cells. Human and rat SCs in vitro have been investigators who described them in the late 19th century.
shown to constitutively express low levels of MHC class I; in In most cases, CMT is associated with autosomal dominant
the presence of activated T lymphocytes and upon stimulation inheritance, although a small proportion of X-linked domi-
with IFN-γ and TNF-α, significant amounts of MHC class II nant and autosomal recessive forms also exist. In addition,
molecules can also be detected (Gold et al. 1995). Also, SCs apparent sporadic cases occur, which may lead to new domi-
stain positive for MHC class II in human nerve biopsies from nantly inherited mutations (see chapter 62).
patients with Guillain–Barré syndrome, suggesting that these There is functional and anatomical evidence that axonal
cells may indeed act as facultative antigen-presenting cells degeneration is likely to be the result of abnormalities in
(Pollard et al. 1986). Schwann cells in vitro have been shown SC–axon interactions (Krajewski et al. 2000)). Trembler mice,
to present antigens to antigen-specific syngeneic T-cell lines which have a dysmyelinating peripheral neuropathy caused by
(Wekerle et al. 1986). Mycobacterium leprae readily invades and a point mutation in pmp22 similar to CMT1A, have signifi-
proliferates in nonmyelinating SCs (Rambukkana et al. 2002); cant changes in both axonal structure and function (Sahenk
myelinated SCs, on the other hand, are resistant to M. leprae et al. 1999).
invasion but undergo demyelination and axonal damage upon Mutations of P0 myelin protein (see chapter 7) cause
attachment to SC axon units, thus creating more fertile ground another form of CMT, CMT1B (Li et al. 2006). There exist
for the microbe expansion (Rambukkana 2004). Schwann cells many other, less common forms of CMT associated with dif-
have been shown to present M. leprae-derived antigens to lym- ferent types of mutations. Further analysis of these will be pre-
phocytes in an MHC II-dependent manner (Ford et al. 1993). sented in chapter 62.
A large body of evidence has accumulated over the last two Hereditary disorders of the PNS also trigger immune
decades implying that SCs can also produce and secrete a wide responses in the peripheral nerve. These responses are consid-
variety of cytokines, which could act as immunomodulators. ered to be important determinants of neuropathy. In CMT,
Cultured SCs have been shown to be able to produce IL-1, a infiltration of lymphocytes and macrophages has been demon-
cytokine involved in the initiation of an immune response. strated in peripheral nerves both in human patients (Vital et
Other proinflammatory cytokines, such as IL-6, TNF-α, and al. 1992) and animal models (Schmid et al. 2000). However,
TGF-β are generated and released by SCs under certain con- in addition to these secondary immune responses, a series
ditions, either in vitro or in vivo (Lisak et al. 1997). of studies have suggested an immunological involvement as
Proinflammatory cytokine production by SCs has also been a primary cause for disease in some genetically determined
shown to be regulated in an autocrine fashion (Qin et al. 2008). neuropathies. Mice with a heterozygous null mutation in P0
The specific receptors for some of the cytokines, such as TNF develop late-onset clinical paralysis associated with inflamma-
receptor, are constitutively expressed on SCs, whereas other tory pathology in the peripheral nerves. Young P0(+/-) mice,
proinflammatory and immunoregulatory mediators, such as without behavioral symptoms, have a defect in central toler-
prostaglandin E2, thromboxane A2, and leukotriene C4, are syn- ance to P0 and are more prone to induction of experimental
thesized in large amounts by SCs, and may regulate the immune autoimmune neuritis by sensitization against a P0 (180–199)
cascade within the inflamed PNS (Bonetti et al. 2000). peptide (Miyamoto et al. 2003). The P0 gene was found to
Schwann cells can also modulate immune reactivity by be transcribed in the thymus of wild-type and the P0 (+/-)
humoral mechanisms. One such pathway is through the produc- mice in an amount proportional to the gene dosage. Thus, a
tion and release of nitric oxide. Schwann cells possess inducible heterozygous mutation in an autoantigen could cause defective
nitrite oxide synthase, which is upregulated after stimulation central tolerance to this autoantigen. This finding may be rel-
with proinflammatory cytokines (Wohlleben et al. 1999). evant to CMT1B, in which P0 is affected. Through this mech-
Finally, SCs may be involved in the termination of auto- anism, autoimmune T cells may play some role in “genetic”
immune response in the PNS. The survival of lymphocytes diseases caused by a heterozygous gene defect. Increased
depends on a delicate balance between proapoptotic and immunogenicity owing to altered protein composition or
antiapoptotic factors. Lymphocyte apoptosis can be regu- mechanical instability of the myelin sheath are also possible
lated through the interaction of the cell surface receptor explanations for the immunoreactive nature of the defective

S C H WA N N C E L L S A N D I N J U RY • 691
myelin sheath. Molecules required for antigen presentation However, endogenous SCs that invade the CNS after
could communicate this information from SCs to immune injury are found mainly in areas lacking astrocytes, and
cells, and it has been proposed that a myelin protein mutant migration of SCs transplanted in rodent CNS white matter
Schwann cell would not demyelinate if it lacked the molecules is inhibited by astrocytes (Iwashita et al. 2000). When SCs
required for antigen processing and presentation (Meyer zu are grafted in the newborn CNS, in the presence of compet-
Horste et al. 2008)). ing endogenous oligodendrocytes, astrocytes are able to limit
their migration and prevent myelination by the transplanted
SCs. In addition, astrocytes are able to form basement
4 S C H WA N N C E L L S ’ R O L E I N C N S R E PA I R membranes around the area of the graft, effectively exclud-
ing the grafted cells from the host parenchyma altogether
In some pathological cases, SCs can participate in CNS (Baron-Van Evercooren et al. 1992). In vitro studies with
repair, in particular in the spinal cord (for an overview see cocultures of SCs and astrocytes have confirmed the mutual
Table 54.1). exclusivity of the two cell types and attempted to interpret it
based on the presence of extracellular matrix molecules, with
no consensus reached yet (Ghirnikar and Eng 1995; Wilby
4.1 E N D O G E N O US S C H WA N N C E L L R E PA I R I N
et al. 1999).
C N S M E C H A N I C A L I N JU RY
Schwann-cell–conditioned medium induces astrocyte
Numerous studies have demonstrated the capacity of SCs to proliferation and production of chondroitin sulfate proteo-
remyelinate CNS axons and promote axonal regeneration in glycan in a fibroblast growth factor receptor 1 (FGFR1)–
the injured CNS (Hagg and Oudega 2006) (see also chap- independent manner (Santos-Silva et al. 2007). In contrast,
ter 55). Schwann cells’ myelinating central axons have been stimulation of boundary formation, astrocytic hypertrophy
detected at the edge of the lesion cavity after photochemical and induction of GFAP expression in astrocytes by SCs is
thrombosis lesions in the rat (Bunge et al. 1994) and also after mediated in an FGF-dependent manner and is modulated
contusion spinal injuries in the monkey (Bresnahan 1978), by another extracellular matrix proteoglycan, heparin sulfate
whereas large numbers of SCs and axons have been reported proteoglycan. Identification of the factors secreted by SCs
in long-term spinal injury sites in human studies (Bunge that induce these astrocytic responses will give us a better
1994). In contusion spinal injuries of the rat, significant axonal understanding of the nature of astrocyte/SC interactions and
sprouting and regeneration occur and the amount of this will point us in the right direction in our quest to manipulate
regeneration is related, among other factors, to the amount of the inhibitory environment induced after injury to promote
SC invasion (Beattie et al. 1997). A recent study demonstrated regeneration (also see chapter 55).
the peripheral origin of invading SCs using genetic-labeling Interestingly, another inhibitor of axonal regeneration,
techniques for neural crest lineage cells (Nagoshi et al. 2011). semaphorin 3A, is repulsive for SCs in vitro and in vivo in a
Furthermore, the study showed that spinal cord injury induces model of spinal trauma. Treatment with a semaphorin 3A inhib-
the dedifferentiation of dorsal root SCs before their invasion itor was able to reverse this effect and increase SC-mediated
of the lesion. myelination and axonal regeneration (Kaneko et al. 2006).

Table 54.1 ENDOGENOUS SCHWANN CELL PARTICIPATION IN CENTRAL NERVOUS SYSTEM REPAIR

Humans Multiple sclerosis:


Feigin and Ogata 1971; Itoyama et al. 1983; Prineas and Connell 1979; Yamamoto et al. 1991
Neuromyelitis Optica:
Ikota et al. 2010
Spinal cord injury:
Buss et al. 2007
Animal models Development:
Focal irradiatiation of developing mouse spinal cord (Gilmore et al. 1983; Heard and Gilmore 1980)
Neuregulin 1 type III up-regulation in zebrafish CNS (Perlin et al. 2011)
Myelin Mutants:
Myelin deficient rat, taiep rat, canine shaking pup (Duncan and Hoffman 1997)
Demyelination:
Experimental allergic encephalomyelitis (Pender 1989)
Viral encephalomyelitis (Dal Canto and Lipton 1980; Miller et al. 1995)
Chemically induced demyelinating lesions in rodents (Blakemore et al. 1985) and macaques (Bachelin
et al. 2005; Lachapelle et al. 2005)
Spinal Cord Injury Models:
Photochemical thrombosis (Bunge et al. 1994)
Spinal cord injury (Bresnahan 1978; Nagoshi et al. 2011)

692 • ROLE OF GLIAL CELLS IN DISEASE


4 .2 E N D O G E N O US S C H WA N N C E L L arguing that the SCs derive from central stem/precursor cells,
R E MY E L I NAT I O N I N D E MY E L I NAT I N G and the other considering that SC remyelination is the conse-
DISORDERS quence of SC invasion from the PNS.
A few years ago, the possibility of the transdifferentiation
4.2.1 Schwann Cell Participation in
of central precursors to peripheral myelinating cells arose with
the Remyelination Process in MS and
a series of experiments. In vivo experiments using X-irradiated
Demyelinating Animal Models
ethidium bromide lesions, demonstrated that transplantation of
The dorsal root entry zone and motor axons exit represent FGF-2 expanded polysialic acid neural cell adhesion molecule
the transitional zone between the CNS and the PNS and (PSA-NCAM) glial restricted precursors (Keirstead et al. 1999),
is composed of the glia limitans, dense astrocytic processes myelin oligodendrocyte glycoprotein-expressing (MOG) oli-
covered by a basal lamina. godendrocyte precursors (Crang et al. 2004), or cloned human
This boundary can be transgressed in demyelinating con- neural precursors resulted in both SC and oligodendrocyte
ditions and the peripheral “invasion” of the CNS has been remyelination (Akiyama et al. 2001). Recent data identified a
observed in multiple sclerosis (MS) patients. Multiple scle- role for bone morphogenic protein (BMP) in instructing CNS
rosis is a demyelinating disease in which successive inflam- precursor into the SC fate. In vitro, BMP and FGF2 were found
matory attacks result in the specific destruction of central to cooperate to induce a neural crest-like fate including SC dif-
oligodendrocytes (OL) (see chapter 61). An interesting fea- ferentiation from fetal and adult neural stem cells (Sailer et al.
ture of MS is that during the development of the disease a 2005). In addition grafted OPCs were shown to differentiate
concomitant endogenous remyelination process is triggered. preferentially into SCs when in the presence of BMP (Talbott
In the CNS, this repair process is mainly conducted by OL et al. 2006). Depending on the environment they are trans-
that form new myelin sheaths around the axons, but the planted in, neural stem/progenitor cells derived from the spinal
presence of SC remyelination of CNS axons is also detected cord give rise to different cells (Mothe and Tator 2008). In the
in MS lesions (Itoyama et al. 1983; Yamamoto et al. 1991). adult shiverer mouse spinal cord, the neural stem/progenitor
Schwann cells invade the CNS mainly after the rupture of the cells generate oligodendrocytes, whereas in focal demyelinated
blood-brain barrier and their entry occurs in the optic nerve, lesion of the spinal cord they differentiate in both oligodendro-
spinal cord, brainstem, and cerebellum (reviewed in (Duncan cytes and SCs. A recent study explored the origin of remyelinat-
and Hoffman 1997). Intraspinal expansion of SC remyelina- ing SCs using transgenic Pdgfra-creERT2: Rosa26-YFP mice
tion was also detected in patients with Devic disease, a demy- that enabled the tracing of OL precursor cells (Zawadzka et al.
elinating disease affecting specifically the optic nerve and the 2010). They observed that after a demyelinating lesion of the
spinal cord (Ikota et al. 2010). spinal cord, 30% of new peripheral myelin colocalizes with the
New insights into the demyelination-remyelination mech- yellow fluorescent protein (YFP) reporter gene; providing evi-
anisms of MS have been made by the development of a vari- dence that some SCs might have a central origin.
ety of toxin-induced animal lesion models (see Baron-Van However the most favored hypothesis is that SCs come
Evercooren and Blakemore 2004) (see also chapter 57). All from peripheral nerve roots in the spinal cord and small nerves
these lesions are totally remyelinated and present the same that innervate blood vessels in the brain, because SC remyeli-
pattern, with SC remyelination at the center of the lesion in nation typically occurs in proximity to spinal/cranial nerves
an astrocytic free area and a oligodendrocytic remyelination at or around blood vessels (Duncan and Hoffman 1997). This is
the border where astrocytes are present. Schwann-cell–derived likely to occur because injection of lentiviral vectors encoding
myelin restores axonal conduction (Felts and Smith 1992) and green fluorescent protein (GFP) in the dorsal roots near a spi-
severe neurological deficits can be reversed when massive SC nal cord contusion injury lead to the presence of GFP labeled
entry occurs in response to spinal cord demyelination ( Jasmin SCs in the lesion (Oudega and Xu 2006). This invasion of the
et al. 2000). The recovery involves the establishment and main- CNS by SCs might be the result of a transgression of the fron-
tenance of nodes of Ranvier by endogenous SCs for more than tier between the CNS and PNS, with possible permissive con-
a 1-year period, with nodes displaying normal distribution of ditions that might be induced by the demyelinated lesion. So,
sodium and potassium channels (Black et al. 2006). the high SC remyelination observed in spinal cord of Devic
Although extremely useful, these models do not reflect the syndrome patients might be explained by the disturbance of
complex environment of MS lesions. A preferred MS model is the astrocytes present at the PNS/CNS frontier (Ikota et al.
experimental allergic encephalomyelitis (EAE), an inflamma- 2010).
tory demyelinating model induced by sensitizing the animals
with myelin-related proteins. As in MS lesions, SC-derived
myelin was detected in EAE models (Pender 1989) even after 5 M ETA B O L I S M-I N D U C E D S C H WA N N
longstanding disease duration. C E L L DA M AG E : F O C U S O N D I A B ET E S
MELLITUS
4.2.2 Origin of Remyelinating Schwann Cells
Neuropathy is the most common complication of diabetes,
Although the origin of remyelinating oligodendrocytes is well occurring in over 50% of all diabetic patients (Feldman et al.
described, the origin of remyelinating SCs remains a matter 1997). A multitude of biochemical changes are associated with
of debate. Two major trends emerge from the literature, one diabetic neuropathy: oxidative stress (Vincent et al. 2004),

S C H WA N N C E L L S A N D I N J U RY • 693
activation of the polyol pathway (Oates 2002), hypoxia and with both neurons and SCs are thus a major culprit in diabetic
ischemia (Low et al. 1989), activation of protein kinase C (Xia neuropathy (Terashima et al. 2012).
et al. 1995), mitogen-activated protein kinases (Tomlinson
1999), and inducible nitric oxide synthase (Vareniuk et al.
2008), increased advanced glycation end products and their 6 S U M M A RY A N D P E R S P E C T I VE S
receptors (Toth et al. 2008), and elevated cytokines such as
TNF (Yamakawa et al. 2011). In this chapter, we discussed the various properties of SCs
The participation of SCs in diabetic neuropathy is char- that implicate them in pathological states; we saw that these
acterized by degeneration of SCs and myelinated axons as cells participate in the causative processes of a variety of con-
well as loss of a population of dorsal root ganglia neurons ditions that affect the PNS, but also constitute a most impor-
(Schmeichel et al. 2003). The pathology of SCs in diabetes tant player in PNS regenerative processes. Last but not least,
is bad news for two reasons: it not only induces degenerative we discussed their participation in CNS repair. The informa-
phenomena, but it also makes regenerative processes more tion on SCs we have amassed over the last few decades allows
difficult because these cells are key players during nerve us not only to answer basic questions about their biology,
regeneration. but also to plan realistic strategies for using SCs for cell ther-
Biochemically, SCs are key players in hyperglycemia- apy. Their regeneration-supporting properties can be fully
induced damage, because they express the enzyme aldose exploited in the PNS in the construction of bioartificial nerve
reductase, which may be responsible for some of the harm- grafts to bridge peripheral nerve gaps resulting from injury,
ful effects of high glucose (Mizisin and Powell 1993). In and their ability to support the regrowth and myelination of
addition to the hyperglycemia-related mechanisms of nerve central axons makes them prime candidates for cell replace-
injury, experimental and clinical studies have suggested that ment therapy in the CNS, as will be discussed in chapter 57.
dyslipidemia related to obesity and type 2 diabetes can also
play a role in the development and progression of peripheral
neuropathy (Obrosova et al. 2007).
AC K N OW L E D G M E N T S
Growth-factor–related regulation of SC function is also
affected. An enhanced expression of p75 neurotrophin recep-
The authors would like to acknowledge all the former and
tor is found on diabetic SCs and, because this receptor is
present members of their labs who contributed directly
involved in an autocrine apoptotic loop, its increased expres-
or indirectly to the elaboration of the manuscript and in
sion may lead to increased cell loss (Eckersley et al. 2001).
particular Corinne Bachelin for her illustration skills. The
An interesting twist to the story of diabetic neuropathy
authors are also grateful to Anne Baron-Van Evercooren and
has been added recently (Chan et al. 2011). These investigators
Rhona Mirsky for their helpful suggestions and comments
describe the appearance of proinsulin-producing cells in multi-
that greatly improved the quality of the manuscript. Also,
ple organs and tissues of streptozotocin-induced diabetic mice.
the FP7 EU project grant Neurosign/Regpot for financial
Close examination of these cells revealed that they first appear
support.
in the bone marrow (BM) and eventually travel to different
peripheral organs and tissues through the circulation. The pro-
insulin-producing bone marrow-derived cells (PI-BMDCs)
appeared within one to three days of intermittent hyperglyce- REFERENCES
mia induced by repeated glucose injections. The initial conjec-
ture was that one could use such PI-BMDCs for diabetes cell Agius E, Cochard P. 1998. Comparison of neurite outgrowth induced by
intact and injured sciatic nerves: a confocal and functional analysis.
therapy, a thought reinforced by a report (Oh et al. 2004) that J Neurosci 18:328–338.
incubation in high-glucose medium induces insulin-gene tran- Akiyama Y, Honmou O, Kato T, Uede T, Hashi K, Kocsis JD. 2001.
scription in BM cells in vitro and that these cells could be trans- Transplantation of clonal neural precursor cells derived from adult
ferred to the kidney capsule of diabetic rodents and partially human brain establishes functional peripheral myelin in the rat spi-
ameliorate the hyperglycemia. However, it was found in vitro nal cord. Exp Neurol 167:27–39.
Bachelin C, Lachapelle F, Girard C, Moissonnier P, Serguera-Lagache C,
that the tiny amounts of PI produced by the PI-BMDCs were Mallet J, Fontaine D, Chojnowski A, Le Guern E, Nait-Oumesmar B,
not detectable in the culture medium and thus PI-BMDCs Baron-Van Evercooren A. 2005. Efficient myelin repair in the
induced by exposure to a high glucose medium had no thera- macaque spinal cord by autologous grafts of Schwann cells. Brain
peutic potential. Quite the contrary, these cells played a nega- Mar;128 (Pt3):540–549.
tive role. In fact, PI-BMDCs are a subpopulation of highly Baron-Van Evercooren A, Blakemore W. 2004. Remyelination through
engraftment. In: Lazzarini R, Griffin J, Lassmann H, Nave KA,
proinflammatory BMDCs specifically induced by diabetes Miller R, Trapp B (eds.), Myelin biology and disorders. San Diego:
that are capable of fusing with cells in the nerves. They seem Elsevier, pp. 143–172.
to play a central role in the pathogenesis of diabetic neuropa- Baron-Van Evercooren A, Clerin-Duhamel E, Lapie P, Gansmuller A,
thy and mediate much of the inflammation-related pathology Lachapelle F, Gumpel M. 1992. The fate of Schwann cells trans-
of diabetic complications. Importantly, these same cells have planted in the brain during development. Dev Neurosci 14:73–84.
Baron-Van Evercooren A, Kleinman HK, Ohno S, Marangos P, Schwartz
great fusogenic potential that can be demonstrated under in JP, Dubois-Dalcq ME. 1982a. Nerve growth factor, laminin, and
vitro conditions. They directly cause tissue damage by fusing fibronectin promote neurite growth in human fetal sensory ganglia
cultures. J Neurosci Res 8:179–193.

694 • ROLE OF GLIAL CELLS IN DISEASE


Baron-Van Evercooren A, Kleinman HK, Seppa HE, Rentier B, cells is polarized, developmentally regulated and axonally depen-
Dubois-Dalcq M. 1982b. Fibronectin promotes rat Schwann cell dent. Development 120:1287–1301.
growth and motility. J Cell Biol 93:211–216. Felts PA, Smith KJ. 1992. Conduction properties of central nerve
Beattie MS, Bresnahan JC, Komon J, Tovar CA, Van Meter M, Anderson fibers remyelinated by Schwann cells. Brain Res 574:178–192.
DK, et al. 1997. Endogenous repair after spinal cord contusion inju- Ford AL, Britton WJ, Armati PJ. 1993. Schwann cells are able to
ries in the rat. Exp Neurol 148:453–463. present exogenous mycobacterial hsp70 to antigen-specific T
Birchmeier C, Nave KA. 2008. Neuregulin-1, a key axonal signal that lymphocytes. J Neuroimmunol 43:151–159.
drives Schwann cell growth and differentiation. Glia 56:1491–1497. Fricker FR, Lago N, Balarajah S, Tsantoulas C, Tanna S, Zhu N, et
Black JA, Waxman SG, Smith KJ. 2006. Remyelination of dorsal col- al. 2011. Axonally derived neuregulin-1 is required for remyelina-
umn axons by endogenous Schwann cells restores the normal pattern tion and regeneration after nerve injury in adulthood. J Neurosci
of Nav1.6 and Kv1.2 at nodes of Ranvier. Brain 129:1319–1329. 31:3225–3233.
Blakemore WF, Crang AJ. 1985. The use of cultured autologous Funakoshi H, Frisen J, Barbany G, Timmusk T, Zachrisson O, Verge VM,
Schwann cells to remyelinate areas of persistent demyelination in et al. 1993. Differential expression of mRNAs for neurotrophins and
the central nervous system. J Neurol Sci 70(2):207–723. their receptors after axotomy of the sciatic nerve. J Cell Biol
Blakemore WF, Smith KJ. 1983. Node-like axonal specializations 123:455–465.
along demyelinated central nerve fibres: ultrastructural observa- Gardiner NJ. 2011. Integrins and the extracellular matrix: key media-
tions. Acta Neuropathol. 1983;60(3–4):291–296. tors of development and regeneration of the sensory nervous sys-
Bonetti B, Valdo P, Stegagno C, Tanel R, Zanusso GL, Ramarli D, tem. Dev Neurobiol 71:1054–1072.
et al. 2000. Tumor necrosis factor alpha and human Schwann Ghirnikar RS, Eng LF. 1995. Chondroitin sulfate proteoglycan stain-
cells: signalling and phenotype modulation without cell death. ing in astrocyte-Schwann cell co-cultures. Glia 14:145–152.
J Neuropathol Exp Neurol 59:74–84. Gilmore SA, Heard JK, Leiting JE. 1983. Patterns of x-radiation-
Bresnahan JC. 1978. An electron-microscopic analysis of axonal alter- induced Schwann cell development in spinal cords of immature
ations following blunt contusion of the spinal cord of the rhesus rats. Anat Rec Mar 205(3):313–319.
monkey (Macaca mulatta). J Neurol Sci 37:59–82. Gilmore SA, Sims TJ, Heard JK. 1982. Autoradiographic and ultra-
Bunge MB, Holets VR, Bates ML, Clarke TS, Watson BD. 1994. structural studies of areas of spinal cord occupied by Schwann cells
Characterization of photochemically induced spinal cord injury in and Schwann cell myelin. Brain Res 239(2):365–375.
the rat by light and electron microscopy. Exp Neurol 127:76–93. Goethals S, Ydens E, Timmerman V, Janssens S. 2010. Toll-like recep-
Bunge RP. 1994. The role of the Schwann cell in trophic support and tor expression in the peripheral nerve. Glia 58:1701–1709.
regeneration. J Neurol 242:S19–21. Gold R, Hartung HP, Lassmann H. 1997. T-cell apoptosis in autoim-
Buss A, Pech K, Kakulas BA, Martin D, Schoenen J, Noth J, Brook mune diseases: termination of inflammation in the nervous system
GA. 2007. Growth-modulating molecules are associated with and other sites with specialized immune-defense mechanisms.
invading Schwann cells and not astrocytes in human traumatic Trends Neurosci 20:399–404.
spinal cord injury. Brain 130(Pt 4):940–953. Gold R, Toyka KV, Hartung HP. 1995. Synergistic effect of IFN-gamma
Cao X, Shoichet MS. 2003. Investigating the synergistic effect of com- and TNF-alpha on expression of immune molecules and antigen
bined neurotrophic factor concentration gradients to guide axonal presentation by Schwann cells. Cell Immunol 165:65–70.
growth. Neuroscience 122:381–389. Gordon T, Tyreman N, Raji MA. 2011. The basis for diminished func-
Chan L, Terashima T, Urabe H, Lin F, Kojima H. 2011. Pathogenesis tional recovery after delayed peripheral nerve repair. J Neurosci
of diabetic neuropathy: bad to the bone. Ann N Y Acad Sci 31:5325–5334.
1240:70–76. Guertin AD, Zhang DP, Mak KS, Alberta JA, Kim HA. 2005.
Chao MV. 2003. Neurotrophins and their receptors: a conver- Microanatomy of axon/glial signaling during Wallerian degenera-
gence point for many signalling pathways. Nat Rev Neurosci tion. J Neurosci 25:3478–3487.
4:299–309. Hagg T, Oudega M. 2006. Degenerative and spontaneous regenerative
Chen ZL, Strickland S. 2003. Laminin gamma1 is critical for Schwann processes after spinal cord injury. J Neurotrauma 23:264–280.
cell differentiation, axon myelination, and regeneration in the Harboe M, Aseff a A, Leekassa R. 2005. Challenges presented by nerve
peripheral nerve. J Cell Biol 163:889–899. damage in leprosy. Lepr Rev 76:5–13.
Cook DN, Pisetsky DS, Schwartz DA. 2004. Toll-like receptors in the Heard JK, Gilmore SA. 1980. Intramedullary Schwann cell develop-
pathogenesis of human disease. Nat Immunol 5:975–979. ment following x-irradiation of mid-thoracic and lumbosacral spi-
Crang AJ, Gilson JM, Li WW, Blakemore WF. 2004. The remyeli- nal cord levels in immature rats. Anat Rec 197: 85–93.
nating potential and in vitro differentiation of MOG-expressing Hu X, Hicks CW, He W, Wong P, Macklin WB, Trapp BD, et al.
oligodendrocyte precursors isolated from the adult rat CNS. Eur J 2006. Bace1 modulates myelination in the central and peripheral
Neurosci 20:1445–1460. nervous system. Nat Neurosci 9:1520–1525.
Dal Canto MC, Lipton HL. 1980. Schwann cell remyelination and Ikota H, Iwasaki A, Kawarai M, Nakazato Y. 2010. Neuromyelitis
recurrent demyelination in the central nervous system of mice optica with intraspinal expansion of Schwann cell remyelination.
infected with attenuated Theiler’s virus. Am J Pathol 98(1):101–122 Neuropathology 30:427–433.
Dal Canto, MC, Rabinowitz, SG. 1982. Experimental models of Itoyama Y, Webster HD, Richardson EP, Jr., Trapp BD. 1983. Schwann
virus-induced demyelination of the central nervous system. Ann cell remyelination of demyelinated axons in spinal cord multiple
Neurol 11:109–127. doi: 10.1002/ana.410110202 sclerosis lesions. Ann Neurol 14:339–346.
Duncan ID, Hoff man RL. 1997. Schwann cell invasion of the central Iwashita Y, Fawcett JW, Crang AJ, Franklin RJ, Blakemore WF. 2000.
nervous system of the myelin mutants. J Anat 190 (Pt 1):35–49. Schwann cells transplanted into normal and X-irradiated adult
Eckersley L, Ansselin AD, Tomlinson DR. 2001. Effects of experimental white matter do not migrate extensively and show poor long-term
diabetes on axonal and Schwann cell changes in sciatic nerve isografts. survival. Exp Neurol 164:292–302.
Brain Res Mol Brain Res 92:128–137. Jasmin L, Janni G, Moallem TM, Lappi DA, Ohara PT. 2000. Schwann
Feigin I, Ogata J. 1971. Schwann cells and peripheral myelin within cells are removed from the spinal cord after effecting recovery from
human central nervous tissues: the mesenchymal character of paraplegia. J Neurosci 20:9215–9223.
Schwann cells. J Neuropathol Exp Neurol 30(4):603–612. Jessen KR, Mirsky R. 2005. The origin and development of glial cells
Feldman EL, Stevens MJ, Greene DA. 1997. Pathogenesis of diabetic in peripheral nerves. Nat Rev Neurosci 6:671–682.
neuropathy. Clin Neurosci 4:365–370. Jessen KR, Mirsky R. 2008. Negative regulation of myelination: rel-
Feltri ML, Scherer SS, Nemni R, Kamholz J, Vogelbacker H, Scott evance for development, injury, and demyelinating disease. Glia
MO, et al. 1994. Beta 4 integrin expression in myelinating Schwann 56:1552–1565.

S C H WA N N C E L L S A N D I N J U RY • 695
Kaneko S, Iwanami A, Nakamura M, Kishino A, Kikuchi K, Shibata development, myelination and dedifferentiation. J Peripher Nerv Syst
S, et al. 2006. A selective Sema3A inhibitor enhances regenerative 13:122–135.
responses and functional recovery of the injured spinal cord. Nat Miyamoto K, Miyake S, Schachner M, Yamamura T. 2003. Heterozygous
Med 12:1380–1389. null mutation of myelin P0 protein enhances susceptibility to
Keirstead HS, Ben-Hur T, Rogister B, O’Leary MT, Dubois-Dalcq autoimmune neuritis targeting P0 peptide. Eur J Immunol 33:
M, Blakemore WF. 1999. Polysialylated neural cell adhesion 656–665.
molecule-positive CNS precursors generate both oligodendrocytes Mizisin AP, Powell HC. 1993. Schwann cell injury is attenuated by
and Schwann cells to remyelinate the CNS after transplantation. aldose reductase inhibition in galactose intoxication. J Neuropathol
J Neurosci 19:7529–7536. Exp Neurol 52:78–86.
Kiefer R, Kieseier BC, Stoll G, Hartung HP. 2001. The role of mac- Mothe AJ, Tator CH. 2008. Transplanted neural stem/progenitor
rophages in immune-mediated damage to the peripheral nervous cells generate myelinating oligodendrocytes and Schwann cells
system. Prog Neurobiol 64:109–127. in spinal cord demyelination and dysmyelination. Exp Neurol
Kieseier BC, Hartung HP, Wiendl H. 2006. Immune circuitry in the 213:176–190.
peripheral nervous system. Curr Opin Neurol 19:437–445. Nagoshi N, Shibata S, Hamanoue M, Mabuchi Y, Matsuzaki Y,
Kieseier BC, Kiefer R, Gold R, Hemmer B, Willison HJ, Hartung Toyama Y, et al. 2011. Schwann cell plasticity aft er spinal cord
HP. 2004. Advances in understanding and treatment of immune- injury shown by neural crest lineage tracing. Glia 59:771–784.
mediated disorders of the peripheral nervous system. Muscle Nerve Nagoshi N, Shibata S, Kubota Y, Nakamura M, Nagai Y, Satoh E, et
30:131–156. al. 2008. Ontogeny and multipotency of neural crest-derived stem
Kobayashi H, Chattopadhyay S, Kato K, Dolkas J, Kikuchi S, Myers cells in mouse bone marrow, dorsal root ganglia, and whisker pad.
RR, et al. 2008. MMPs initiate Schwann cell-mediated MBP deg- Cell Stem Cell 2:392–403.
radation and mechanical nociception after nerve damage. Mol Cell Nicolino S, Panetto A, Raimondo S, Gambarotta G, Guzzini M,
Neurosci 39:619–627. Fornaro M, et al. 2009. Denervation and reinnervation of adult
Krajewski KM, Lewis RA, Fuerst DR, Turansky C, Hinderer SR, skeletal muscle modulate mRNA expression of neuregulin-1 and
Garbern J, et al. 2000. Neurological dysfunction and axonal ErbB receptors. Microsurgery 29:464–472.
degeneration in Charcot-Marie-Tooth disease type 1A. Brain 123 Oates PJ. 2002. Polyol pathway and diabetic peripheral neuropathy.
(Pt 7):1516–1527. Int Rev Neurobiol 50:325–392.
Krammer PH. 2000. CD95’s deadly mission in the immune system. Obrosova IG, Ilnytska O, Lyzogubov VV, Pavlov IA, Mashtalir N,
Nature 407:789–795. Nadler JL, et al “healthy” diet and aldose reductase inhibition.
Lachapelle F, Bachelin C, Moissonnier P, Nait-Oumesmar B, Diabetes 56:2598–2608.
Hidalgo A, Fontaine D, Baron-Van Evercooren A. 2005. Failure of Oh SH, Muzzonigro TM, Bae SH, LaPlante JM, Hatch HM, Petersen
remyelination in the nonhuman primate optic nerve. Brain Pathol. BE. 2004. Adult bone marrow-derived cells trans-differentiating
Jul 15(3):198–207. into insulin-producing cells for the treatment of type I diabetes.
La Marca R, Cerri F, Horiuchi K, Bachi A, Feltri ML, Wrabetz L, Lab Invest 84:607–617.
et al. 2011. TACE (ADAM17) inhibits Schwann cell myelination. Oudega M, Xu XM. 2006. Schwann cell transplantation for repair of
Nat Neurosci 14:857–865. the adult spinal cord. J Neurotrauma 23:453–467.
Lavdas AA, Papastefanaki F, Thomaidou D, Matsas R. 2008. Schwann Patton BL, Miner JH, Chiu AY, Sanes JR. 1997. Distribution and
cell transplantation for CNS repair. Curr Med Chem 15:151–160. function of laminins in the neuromuscular system of developing,
Leblanc SE, Srinivasan R, Ferri C, Mager GM, Gillian-Daniel AL, adult, and mutant mice. J Cell Biol 139:1507–1521.
Wrabetz L, et al. 2005. Regulation of cholesterol/lipid biosyn- Pender MP. 1989. Recovery from acute experimental allergic encepha-
thetic genes by Egr2/Krox20 during peripheral nerve myelination. lomyelitis in the Lewis rat. Early restoration of nerve conduction
J Neurochem 93:737–748. and repair by Schwann cells and oligodendrocytes. Brain 112 (Pt
Lefcort F, Venstrom K, McDonald JA, Reichardt LF. 1992. Regulation of 2):393–416.
expression of fibronectin and its receptor, alpha 5 beta 1, during develop- Perrin FE, Lacroix S, Aviles-Trigueros M, David S. 2005. Involvement
ment and regeneration of peripheral nerve. Development 116:767–782. of monocyte chemoattractant protein-1, macrophage inflamma-
Li HY, Say EH, Zhou XF. 2007. Isolation and characterization of neural crest tory protein-1alpha and interleukin-1beta in Wallerian degenera-
progenitors from adult dorsal root ganglia. Stem Cells 25:2053–2065. tion. Brain 128:854–866.
Li J, Bai Y, Ianakova E, Grandis M, Uchwat F, Trostinskaia A, et al. 2006. Perlin JR, Lush ME, Stephens WZ, Piotrowski T, Talbot WS. 2011.
Major myelin protein gene (P0) mutation causes a novel form of axonal Neuronal Neuregulin 1 type III directs Schwann cell migration.
degeneration. J Comp Neurol 498:252–265. Development 2011 Nov 138(21):4639–4648. Epub Sep 28.
Lisak RP, Skundric D, Bealmear B, Ragheb S. 1997. The role of cytokines Pollard JD, McCombe PA, Baverstock J, Gatenby PA, McLeod
in Schwann cell damage, protection, and repair. J Infect Dis 176 Suppl JG. 1986. Class II antigen expression and T lymphocyte sub-
2:S173–179. sets in chronic inflammatory demyelinating polyneuropathy.
Low PA, Lagerlund TD, McManis PG. 1989. Nerve blood flow and oxy- J Neuroimmunol 13:123–134.
gen delivery in normal, diabetic, and ischemic neuropathy. Int Rev Prineas JW, Connell F. 1979. Remyelination in multiple sclerosis.
Neurobiol 31:355–438. Ann Neurol 5(1):22–31.
Martini R, Fischer S, Lopez-Vales R, David S. 2008. Interactions between Qin Y, Cheng C, Wang H, Shao X, Gao Y, Shen A. 2008. TNF-alpha
Schwann cells and macrophages in injury and inherited demyelinating as an autocrine mediator and its role in the activation of Schwann
disease. Glia 56:1566–1577. cells. Neurochem Res 33:1077–1084.
Meyer zu Horste G, Hu W, Hartung HP, Lehmann HC, Kieseier BC. 2008. Qin Y, Hua M, Duan Y, Gao Y, Shao X, Wang H, et al. 2012.
The immunocompetence of Schwann cells. Muscle Nerve 37:3–13. TNF-alpha expression in schwann cells is induced by LPS and
Miller DJ, Asakura K, Rodriguez M. 1995. Experimental strategies to NF-kappaB-dependent pathways. Neurochem Res Apr 37(4):
promote central nervous system remyelination in multiple sclerosis: 722–731.
insights gained from the Theiler’s virus model system. J Neurosci Res Rambukkana A. 2004. Mycobacterium leprae-induced demyelina-
Jun 15;41(3):291–296. tion: a model for early nerve degeneration. Curr Opin Immunol
Mimouni-Rongy M, White JH, Weinstein DE, Desbarats J, Almazan 16:511–518.
G. 2011. Fas ligand acts as a counter-receptor in Schwann cells and Rambukkana A, Zanazzi G, Tapinos N, Salzer JL. 2002. Contact-
induces the secretion of bioactive nerve growth factor. J Neuroimmunol dependent demyelination by Mycobacterium leprae in the absence
230:17–25. of immune cells. Science 296:927–931.
Mirsky R, Woodhoo A, Parkinson DB, Arthur-Farraj P, Bhaskaran A, Sahenk Z, Chen L, Mendell JR. 1999. Effects of PMP22 duplication
Jessen KR. 2008. Novel signals controlling embryonic Schwann cell and deletions on the axonal cytoskeleton. Ann Neurol 45:16–24.

696 • ROLE OF GLIAL CELLS IN DISEASE


Saher G, Quintes S, Mobius W, Wehr MC, Kramer-Albers EM, Verheijen MH, Chrast R, Burrola P, Lemke G. 2003. Local regulation of fat
Brugger B, et al. 2009. Cholesterol regulates the endoplasmic retic- metabolism in peripheral nerves. Genes Dev 17:2450–2464.
ulum exit of the major membrane protein P0 required for periph- Viader A, Chang LW, Fahrner T, Nagarajan R, Milbrandt J. 2011.
eral myelin compaction. J Neurosci 29:6094–6104. MicroRNAs modulate Schwann cell response to nerve injury by rein-
Sailer MH, Hazel TG, Panchision DM, Hoeppner DJ, Schwab ME, McKay forcing transcriptional silencing of dedifferentiation-related genes.
RD. 2005. BMP2 and FGF2 cooperate to induce neural-crest-like J Neurosci 31:17358–17369.
fates from fetal and adult CNS stem cells. J Cell Sci 118:5849–5860. Vincent AM, Russell JW, Low P, Feldman EL. 2004. Oxidative stress in the
Santos-Silva A, Fairless R, Frame MC, Montague P, Smith GM, Toft pathogenesis of diabetic neuropathy. Endocr Rev 25:612–628.
A, et al. 2007. FGF/heparin differentially regulates Schwann cell Vital A, Vital C, Julien J, Fontan D. 1992. Occurrence of active demyelinat-
and olfactory ensheathing cell interactions with astrocytes: a role ing lesions in children with hereditary motor and sensory neuropathy
in astrocytosis. J Neurosci 27:7154–7167. (HMSN) type I. Acta Neuropathol 84:433–436.
Sawant-Mane S, Piddlesden SJ, Morgan BP, Holers VM, Koski CL. Vogelezang MG, Liu Z, Relvas JB, Raivich G, Scherer SS, ffrench-Constant
1996. CD59 homologue regulates complement-dependent cytoly- C. 2001. Alpha4 integrin is expressed during peripheral nerve
sis of rat Schwann cells. J Neuroimmunol 69:63–71. regeneration and enhances neurite outgrowth. J Neurosci 21:
Schmeichel AM, Schmelzer JD, Low PA. 2003. Oxidative injury and 6732–6744.
apoptosis of dorsal root ganglion neurons in chronic experimental Wallquist W, Patarroyo M, Thams S, Carlstedt T, Stark B, Cullheim
diabetic neuropathy. Diabetes 52:165–171. S, et al. 2002. Laminin chains in rat and human peripheral nerve:
Schmid CD, Stienekemeier M, Oehen S, Bootz F, Zielasek J, Gold distribution and regulation during development and after axonal
R, et al. 2000. Immune deficiency in mouse models for inherited injury. J Comp Neurol 454:284–293.
peripheral neuropathies leads to improved myelin maintenance. J Wekerle H, Schwab M, Linington C, Meyermann R. 1986. Antigen
Neurosci 20:729–735. presentation in the peripheral nervous system: Schwann cells
Shamash S, Reichert F, Rotshenker S. 2002. The cytokine network of present endogenous myelin autoantigens to lymphocytes. Eur J
Wallerian degeneration: tumor necrosis factor-alpha, interleukin-1alpha, Immunol 16:1551–1557.
and interleukin-1beta. J Neurosci 22:3052–3060. Wienholds E, Plasterk RH. 2005. MicroRNA function in animal
Shirakabe K, Wakatsuki S, Kurisaki T, Fujisawa-Sehara A. 2001. Roles of development. FEBS Lett 579:5911–5922.
Meltrin beta/ADAM19 in the processing of neuregulin. J Biol Chem Wilby MJ, Muir EM, Fok-Seang J, Gour BJ, Blaschuk OW, Fawcett
276:9352–9358. JW. 1999. N-Cadherin inhibits Schwann cell migration on astro-
Siironen J, Sandberg M, Vuorinen V, Roytta M. 1992. Laminin B1 and col- cytes. Mol Cell Neurosci 14:66–84.
lagen type IV gene expression in transected peripheral nerve: reinnerva- Wohlleben G, Hartung HP, Gold R. 1999. Humoral and cellular
tion compared to denervation. J Neurochem 59:2184–2192. immune functions of cytokine-treated Schwann cells. Adv Exp
Takeda K, Kaisho T, Akira S. 2003. Toll-like receptors. Annu Rev Immunol Med Biol 468:151–156.
21:335–376. Woodhoo A, Alonso MB, Droggiti A, Turmaine M, D’Antonio M,
Takigawa T, Yasuda H, Terada M, Haneda M, Kashiwagi A, Saito T, et al. Parkinson DB, et al. 2009. Notch controls embryonic Schwann
2000. The sera from GM1 ganglioside antibody positive patients with cell differentiation, postnatal myelination and adult plasticity. Nat
Guillain-Barre syndrome or chronic inflammatory demyelinating poly- Neurosci 12:839–847.
neuropathy blocks Na+ currents in rat single myelinated nerve fibers. Xia P, Kramer RM, King GL. 1995. Identification of the mechanism
Intern Med 39:123–127. for the inhibition of Na+,K(+)-adenosine triphosphatase by hyper-
Talbott JF, Cao Q, Enzmann GU, Benton RL, Achim V, Cheng XX, et al. glycemia involving activation of protein kinase C and cytosolic
2006. Schwann cell-like differentiation by adult oligodendrocyte pre- phospholipase A2. J Clin Invest 96:733–740.
cursor cells following engraftment into the demyelinated spinal cord is Yamakawa I, Kojima H, Terashima T, Katagi M, Oi J, Urabe H, et al.
BMP-dependent. Glia 54:147–159. 2011. Inactivation of TNF-alpha ameliorates diabetic neuropathy
Terashima T, Kojima H, Chan L. 2012. Bone marrow expression of in mice. Am J Physiol Endocrinol Metab 301:E844–852.
poly(ADP-ribose) polymerase underlies diabetic neuropathy via Yamamoto T, Kawamura J, Hashimoto S, Nakamura M. 1991.
hematopoietic-neuronal cell fusion. Faseb J 26:295–308. Extensive proliferation of peripheral type myelin in necrotic spinal
Tomlinson DR. 1999. Mitogen-activated protein kinases as glucose trans- cord lesions of multiple sclerosis. J Neurol Sci 102:163–169.
ducers for diabetic complications. Diabetologia 42:1271–1281. Yuki N, Taki T, Inagaki F, Kasama T, Takahashi M, Saito K, et al.
Toth C, Rong LL, Yang C, Martinez J, Song F, Ramji N, et al. 2008. 1993. A bacterium lipopolysaccharide that elicits Guillain-Barre
Receptor for advanced glycation end products (RAGEs) and experimen- syndrome has a GM1 ganglioside-like structure. J Exp Med
tal diabetic neuropathy. Diabetes 57:1002–1017. 178:1771–1775.
Vareniuk I, Pavlov IA, Obrosova IG. 2008. Inducible nitric oxide syn- Yun B, Anderegg A, Menichella D, Wrabetz L, Feltri ML, Awatramani
thase gene deficiency counteracts multiple manifestations of periph- R. 2010. MicroRNA-deficient Schwann cells display congenital
eral neuropathy in a streptozotocin-induced mouse model of diabetes. hypomyelination. J Neurosci 30:7722–7728.
Diabetologia 51:2126–2133. Zawadzka M, Rivers LE, Fancy SP, Zhao C, Tripathi R, Jamen F, et al.
Verheijen MH, Camargo N, Verdier V, Nadra K, de Preux Charles AS, 2010. CNS-resident glial progenitor/stem cells produce Schwann
Medard JJ, et al. 2009. SCAP is required for timely and proper myelin cells as well as oligodendrocytes during repair of CNS demyelina-
membrane synthesis. Proc Natl Acad Sci U S A 106:21383–21388. tion. Cell Stem Cell 6:578–590.

S C H WA N N C E L L S A N D I N J U RY • 697
This page intentionally left blank
RECOVERY OF NEURAL FUNCTION

699
This page intentionally left blank
55.
NERVE REGENERATION IN THE PERIPHERAL
NERVOUS SYSTEM
Tessa Gordon and Olawale AR Sulaiman

A B B R E VI AT I O N S despite the regenerative capacity of the peripheral nervous system


and provide the rationale for experimental approaches to improve
BDNF brain-derived neurotrophic factor axonal regeneration and in turn, functional recovery.
cAMP cyclic adenosine monophosphate
ES electrical stimulation
2 P E R I P H E R A L N E RVE I N J U RY
FGF fibroblast growth factor
GDNF glial-derived neurotrophic factor
IGF insulin-like growth factor 2.1 T H E C E L L B O DY R E S P O NS E A N D T H E
N-CAM neural cell adhesion molecule G ROW T H M O D E O F I N J U R E D N EU RO N S
NGF nerve growth factor
NT3 neurotrophin 3
2.1.1 Chromatolysis and Gene Expression:
NT4/5 neurotrophin 4/5
PMP22 peripheral myelin protein 22 kDa Neurons that incur axonal disruption or axotomy with loss of
RAG regeneration-associated gene contact with target connection undergo characteristic morpho-
SC Schwann cell logical or “chromatolytic” changes. The morphological changes
TGF-β transforming growth factor beta include swelling, movement of the nucleus and nucleolus to an
VEGF vascular endothelial growth factor eccentric position in the cell body, and dissolution of Nissl bod-
ies that comprise the rough endoplasmic reticulum. They reflect
a marked increase in mRNA synthesis and the change in gene
1 INTRODUCTION expression that converts the neurons from the normally “trans-
mitting” to the “growth” mode (Fu and Gordon 1997). They
The capacity for injured neurons to regenerate their axons and include the upregulation of growth associated genes such as
remake functional connections with target organs distinguishes actin, tubulin, and GAP43 and downregulation of genes such as
the peripheral from the central nervous system. This unique neurofilament that is normally responsible for sustaining axon
characteristic of the peripheral nervous system stems from the diameter (Fig. 55.1).
ability of the Schwann cells (SCs) as opposed to the inability of
the oligodendrocytes in the central nervous system, to support 2.1.2 Altered Gene Expression Is Transient
axonal regeneration (see chapters 54 and 56). In consequence,
The conversion of the neuron from a transmitting to a growth
return of function after peripheral nerve injuries contrasts with
mode involves the downregulation of transmitter-encoding
permanent deficits associated with central nerve injuries. Yet,
genes that include choline acetyltransferase in motoneurons
functional recovery after nerve injuries in humans is generally
and the upregulation of neurotrophic factors and their receptors
poor despite the capacity of injured peripheral nerves to regenerate
(Boyd and Gordon 2003b; Fu and Gordon 1997). The altered
their axons. Minimal or no recovery may be the outcome for
gene expression occurs within days to reach a maximum within
injuries that sever the peripheral nerve far from the target and
weeks, the time course varying according to the particular genes
that incur considerable delays before target reinnervation. This is
that are expressed. However, the expression is not maintained
regardless of considerable advances made in microsurgical repair
with mRNA levels returning to baseline levels (Sulaiman et al.
of the injured nerves (Sunderland 1978). The processes of axonal
2011) (see Fig. 55.1).
regeneration and target reinnervation involve many factors that
pertain to the injured neuron, the growth environment of the
distal nerve stumps, and denervated targets. The neurons must 2.2 C O N VE R S I O N O F T H E D I S TA L N E RVE
survive the injuries to mount a regenerative response, regenerate S T UM P TO A G ROW T H P E R M I S S I VE
their axons within the growth environment of the denervated S TAT E A F T E R I N JU RY
distal nerve stump, and reinnervate the appropriate targets to
restore function. This chapter reviews the response of SCs to 2.2.1 Wallerian Degeneration
peripheral nerve injury in animal models. This information will Axons that are separated from the cell body by nerve injury
provide insight into why functional recovery is often so poor undergo Wallerian degeneration with ultimate phagocytosis of
701
A GAP-43 ChAT

Tubulin AChE

Actin Neurofilament

Chromatolysis Axon Outgrowth Wallerian Degeneration

Neurotrophic Adhesion Myelination


Neurotrophic Neurotrophic factor & molecules, genes
factors factor receptors receptors mitogens/
receptors,
cytokines

B C
1.0 1.0
Gene expression

Gene expression
0.5 Neuron 0.5 Schwann cell
RAGs RAGs

0.0 0.0
07 100 200 07 100 200
Time after nerve injury (days) Time after nerve injury (days)
D E
1.2
Tubulin mRNA relative to 7d = 1

1600
*
% Change in GDNF mRNA

1.0 1400
1200 *
expression

0.8 1000
800 *
0.6 *
600
0.4 Intact 400
7 days 200
0.2 0
0 30 60 90 120 150 180 2d 7d 28d 90d 180d
Days after axotomy Days after chronic denervation

Figure 55.1 After nerve injury, regeneration-associated genes (RAGs) are upregulated transiently in the neurons (A,B) in concert with downregulation of
genes associated with normal synaptic transmission (A). Schwann cells in the denervated nerve stumps undergo proliferation during Wallerian degenera-
tion and express many RAGs in concert with downregulation of myelin-associated genes (A,C). The gene profiles support outgrowth of axons, but
their altered expression being transient, axonal growth declines with time as the growth response wanes and the growth support by Schwann cells
declines as the cells go into a senescent state. mRNA levels are plotted for tubulin in neurons (D) and GDNF in Schwann cells (E).

axonal and myelin debris by resident SCs and the macrophages and the ubiquitin-proteasome system (Zhai et al. 2003). The
that infiltrate the distal stump (Brushart 2011; Stoll and Muller axon debris releases mitogens that promote mitotic SC divi-
1999; Waller 1850) (see chapters 14 and 54) (Fig. 55.1). A sion and initiates a network of cytokines and transcription fac-
recent review breaks the events into three morphologically tors that stimulates myelin breakdown, macrophage invasion,
discernible stages: acute axonal degeneration, a latency period, and targets the myelin for phagocytosis by the SCs and mac-
and an abrupt granular degenerative stage (Wang et al. 2012). rophages (Brushart 2011; Fu and Gordon 1997). Schwann cell–
Within an hour of injury, “die back” in both the proximal derived neuregulins are not essential for this proliferative phase
and distal stumps is mediated by channel-mediated calcium (Atanasoski et al. 2006; Wang et al. 2012).
influx and activation of calpains that cleave neurofilament and The dividing SCs secrete many chemoattractive factors
microtubular-associated components such as spectrin and that include the cytokines, interleukin-1α, leukemia inhibi-
tubulin. In the peripheral nerve, this die back is to the first tory factor, and monocyte chemoattractant protein-1 (Fu and
node of Ranvier. The next stage of approximately 24 hours, Gordon 1997). They recruit macrophages into and through-
during which time action potentials continue to be conducted out the distal nerve stump, where they enhance SC prolifera-
(Miledi and Slater 1970) is followed by a longer period of dis- tion by releasing growth factors and assume the major role of
assembly of cytoskeletal proteins by calcium-activated calpains phagocytosis of axon and myelin debris (Avellino et al. 1995).

702 •
The growth factors include nerve growth factor (NGF), plate- appropriate distal pathways, although the process is not
let-derived growth factor, epithelial growth factor, and acidic precise (Al-Majed et al. 2000; Brushart 1993; Eberhardt
and basic fibroblast growth factors (FGF) (Fu and Gordon et al. 2006).
1997). Schwann cells and macrophage cytokines have been
implicated in both the enhancement (e.g., interleukin-1β and
tumor necrosis factor-α) and the curtailment (transforming 2.2.3 Transient Expression of Growth-Associated Genes
growth factor-β [TGF-β]) of phagocytosis and hence, the The growth supportive state of denervated SCs is transient
regulation of the month-long process of Wallerian degenera- like that of the growth state of axotomized neurons (Boyd
tion in the rat (Shamash et al. 2002; You et al. 1997). and Gordon 2003b; Fu and Gordon 1997) (see Fig. 55.1).
Neurotrophic factors such as GDNF that are upregulated
2.2.2 Conversion of Schwann Cells within weeks, are progressively downregulated to low lev-
to a Growth-Supportive State els within months after denervation (Hoke et al. 2002). The
low affinity p75 receptor expression and protein progres-
2.2.2.1 Expression of Growth-Associated Genes sively decline to undetectable levels within the same time
Schwann cells and macrophage-derived growth factors contrib- frame (Li et al. 1997; You et al. 1997). Interestingly, the
ute to the conversion of SCs from the myelinating phenotype declining expression of erb2/3 receptors for neuregulin is
associated with intact axons to the “growth-supportive” den- reversed when the chronically denervated nerve stump is
ervated phenotype in the absence of axon contact. The dener- placed into tissue culture (Li et al. 1997). The reduced expres-
vated SCs downregulate myelin-associated glycoproteins and sion of growth associated genes is accompanied by a transi-
proteins including P0, myelin basic protein, myelin-associated tion from the growth supportive state to a dormant state in
glycoprotein, peripheral myelin protein 22 kDa (PMP22), which the SCs atrophy and decline in number (Li et al. 1997;
connexin 32, periaxin, and transcription factors such as You et al. 1997).
Krox-20, Krox-24, and Oct-6. They upregulate genes for cell
adhesion molecules such as the immunoglobulins neural cell
adhesion molecule (N-CAM) and L1 and the cadherin super- 2.3 R EG E N E R AT I N G AXO NS A N D T H E I R
family. They also upregulate the genes for the large glycopro- I N T E R AC T I O NS WIT H S C H WA N N C E L L S
teins such as laminin that form the basal lamina substrate for 2.3.1 The “Bands of Büngner”
axonal regeneration, as well as many neurotrophins, growth
factors, and their receptors that play a role in regeneration of Schwann cells are normally anchored in intact nerve via
the axons from the proximal nerve stump (Boyd and Gordon attachments to the large glycoproteins of the basal lamina
2003b; Fu and Gordon 1997) (see Fig. 55.1) (see chapters 43 in the endoneurial tubes; these include laminin, fibronectin,
and 54). and collagen VI (Chernousov and Carey 2000). After injury,
The interleukins released in the distal nerve stumps after the denervated SCs upregulate genes for these glycoproteins
nerve injury have been implicated in the production of growth and other adhesion molecules, including NCAM and L1,
factors by the denervated SCs, including NGF in fibroblasts and N-cadherin that are members of the immunoglobulin
and SCs (Boyd and Gordon 2003b; Fu and Gordon 1997). and cadherin superfamilies, respectively (Brushart 2011; Ide
Transforming growth factor beta (TGF-β) has also been 1996; Martini and Schachner 1988). Thereby, they sustain the
shown to be essential for the neurotrophic effect of several basal lamina and they line the endoneurial tubes as the “bands
neurotrophic factors, including glial-derived neurotrophic fac- of Büngner” (Büngner 1891) to support the regeneration of
tor (GDNF), but the effects of TGF-β on SCs are phenotype- axons through the endoneurial tubes (Chernousov and Carey
dependent as they are in other cells (Einheber et al. 1995). 2000; Fu and Gordon 1997). Both the basal lamina and the
SCs are essential for axon regeneration, axon outgrowth, and
2.2.2.2 Motor and Sensory Specific Expression of elongation failing in their absence (Hall 1986).
Neurotrophic Factors Schwann cell–derived neuregulin likely plays a role in
The several growth factors expressed by denervated SCs the secondary proliferation of SCs as the regenerating axons
include the neurotrophins [NGF, brain-derived neurotrophic contact the SCs and in the remyelination of the axons once
factor (BDNF), neurotrophin-3 (NT-3) and neurotro- their processes are withdrawn (Fricker et al. 2011). There is
phin 4/5 (NT4/5)], members of the TGF-β superfamily pharmacological evidence that the neurotransmitters, acetyl-
including TGF-β and GDNF, vascular endothelial growth choline, and ATP that are released from growth cones act via
factor (VEGF), pleiotrophin, the insulin-like growth receptors on the SCs in the distal nerve stump to promote the
factors (IGF1 and 11), and FGF-1,2 (Brushart 2011). The withdrawal of their long processes and in turn, convert them
gene profile of neurotrophic factors differs in denervated SCs from the nonmyelinating growth permissive state to a myeli-
within endoneurial tubes that surround motor and sensory nating one (Vrbova et al. 2009). Neurotransmitter mediated
axons: BDNF is the predominant neurotrophin expressed by calcium influx has been demonstrated to initiate retraction of
motor SCs and the sensory SCs express a wider range of neu- long processes of denervated perisynaptic SCs (Georgiou et al.
rotrophins that include NGF and NT4/5 (Hoke et al. 2006). 1999). Nogo receptors on macrophages in the lesioned nerves
The profiles have been implicated in the preference shown for mediate their efflux out of the SC basal lamina during axon
motor and sensory neurons to regenerate their axons within extension (Fry et al. 2007).

N E RVE R E G E N E R AT I O N I N T H E P E R I P H E R A L N E RVO U S SYS T E M • 703


Diameters of the regenerating axons progressively increase 2.3.3 Sprouting, Axon Elongation, and
in proportion to the proximal axons with concomitant increase Reformation of Target Connections
in the myelin rings around the axons (Devor and Govrin-
Lippmann 1979). Full recovery of nerve fiber diameters, how- Parent axons in the proximal stump of injured peripheral
ever, depends on the reformation of functional contacts with nerve elaborate multiple growth cones and produce as many
denervated targets (Fu and Gordon 1997). as five daughter axons each that regenerate across the injury
site and into the distal nerve stumps (see Fig. 55.1). The fate of
these branches depends on the availability of a neural “guid-
2.3.2 Staggered Axonal Regeneration ing” structure distal to the injury site. In its absence, regenerat-
ing axons form a neuroma that is a mixture of immature nerve
The initial axon outgrowth within hours of nerve injury
fibers and connective tissue (Fu and Gordon 1997; Sunderland
is aborted before transport of elevated levels of actin and
1978). Axonal sprouts that advance distally from an axon
tubulin for axonal extension (Geraldo and Gordon-Weeks
within the proximal nerve stump comprise a “regenerating
2009). Outgrowth from the proximal nerve stump into the
unit” (Fu and Gordon 1997). The multiple axonal sprouts
distal stump is a protracted process especially after transec-
remain in the distal nerve stumps unless axons make target
tion injuries that disrupt the continuity of the basal lamina.
connections when all but one daughter axon are withdrawn
The accepted 1- to 3-mm/day regeneration rate in mammals
over months and even years (Aitken et al. 1947; Mackinnon
was calculated from rate at which regenerating axons covers
et al. 1991). The final diameter of the daughter axon depends
a distance. This was evaluated by the “pinch test” for sensory
on reformation of target contacts (Fu and Gordon 1997).
neurons and the rate of recovery of a given motor function,
At the neuromuscular junction, the specialized nonmyeli-
such as toe spreading for motoneuron regeneration (Gutmann
nating perisynaptic SCs normally respond to released trans-
et al. 1942; Sunderland 1947). When this plot was extended
mitters from the intact presynaptic terminals; they become
to the x-axis, the latent period of axon outgrowth was cal-
reactive, extend processes after denervation, and guide regen-
culated as a few days. However, the counts of backlabeled
erating axons back to the denervated endplates to reinnervate
motoneurons that regenerated their axons across a suture site
muscle (Son and Thompson 1995). These reactive perisynaptic
of a transected nerve demonstrated that only a small fraction
SCs resemble denervated SCs in the distal nerve stumps with
of motoneurons regenerate their axons into the distal stump
respect to their process formation. The perisynaptic Schwann
within 4 days, with the remainder progressively crossing the
cells and the basal lamina of the adult endplate are sufficient
suture site over a 30-day period (Al-Majed et al. 2002) (Fig.
to guide regenerating axons back to the endplate region,
55.2). This unexpected “staggering” of regenerating axons
even after ablation of the denervated muscle (Dunaevsky and
across the suture line accounted for our earlier finding of an
Connor 1998).
8- to 10-week period during which all motoneurons progres-
sively regenerate their axons over a 25-mm distance rather
than an expected shorter period of 2 to 3 weeks if regenera- 2.4 T E M P O R A L D EC L I N E I N R EG E N E R AT I V E
tion of axons was synchronous rather than staggered across C A PAC IT Y A N D S U P P O RT
the suture site (Al-Majed et al. 2000). Dorsal root ganglion
neurons demonstrate the same staggered outgrowth of axons 2.4.1 Poor Functional Recovery After Nerve
(Witzel et al. 2005). Cajal had described how, despite surgi- Injury in Humans
cal apposition of transected nerve stumps, the axons that grow Unfortunately, functional recovery in humans is frequently
out from the proximal nerve stump take tortuous routes to poor after peripheral nerve injuries, especially when nerve
navigate between surgically apposed proximal and distal nerve trunks close to the spinal cord and far from the target organs
stumps. In some cases the axons spiraled and some grew back are injured (Fig. 55.3). Under these conditions, the regenerat-
into the proximal nerve stump (Cajal 1928). In transgenic ing axons remain without target connections for months and
mice whose neurons express yellow fluorescent protein, the even years, a condition we refer to as chronic axotomy (Boyd
same tortuous pathways of axon growth were described after and Gordon 2002, 2003b; Fu and Gordon 1995a, 1997).
nerve transection and surgical repair (Witzel et al. 2005). Schwann cells in the distal nerve stumps and the target organs
Moreover, immunohistochemical visualization of events at are chronically denervated for long periods, conditions of
the suture site showed the same tortuous patterns of axon out- chronic SC denervation (Fu and Gordon 1995b; Gordon et al.
growth in the context of the slow migration of misaligned SCs 2003, 2011; Sulaiman and Gordon 2000, 2002; Sulaiman
into the disorganized laminin of the disrupted basal lamina et al. 2002) and chronic muscle denervation (Gordon et al.
(Witzel et al. 2005). 2011a), respectively. These periods may be months and years
Growth inhibitory molecules such as chondroitin for neurons to regenerate their axons and reinnervate targets
sulfate proteoglycans that are expressed in the endoneu- at 1 mm/day. The failure of functional return in the hand,
rium and the surrounding nerve sheaths, may serve to con- for example, is generally attributed to irreversible atrophy
fine regenerating axons to the growth supportive SC-basal of the denervated muscles and sense organs and fatty tissue
lamina conduits (Udina et al. 2006). Their breakdown by replacement (Sulaiman et al. 2011) (see Fig. 55.3). However,
chondroitinase ABC promotes axon outgrowth by a sepa- our experiments over the past approximately 15 years provide
rate signaling pathway to that of the neurotrophins (Zhou strong evidence that it is the progressive failure of chroni-
et al. 2006). cally axotomized neurons to regenerate their axons and the

704 • NEUROGLIA
A B

1.5 mm

25mm

Fluoro Fluoro
gold ruby

C 350 D 400
350
Labeled motoneurons

300 300
250
250 200
150
200 100
50
150 0
0 2 4 6 8 10 4d 1 wk 2 wk 3 wk 4 wk

E 350 F 400

350
300
Labeled motoneurons

All 300
250 250
200 200
150
150 Muscle
100 Stimulation
100 Sham
50
Cutaneous
50 0
0 2 4 6 8 10 4d 1 wk 2 wk 3 wk 4 wk

G H
350 180
160
300
Labeled motoneurons

All 140
250 120
100
200
80
Muscle
150 60
40
100
Cutaneous 20
50 0
0 2 4 6 8 10 0–4 4d– 1 wk– 2 wks– 3 wks–
Weeks after femoral ne
rve repair days 1 wk 2 wks 3 wks 4 wks
Weeks after femoral ne
rve repair

Figure 55.2 Numbers of retrogradely labelled femoral motoneurons that regenerated their axons 25 mm into muscle and cutaneous branches (A) and 1.5 mm (B) from
the site of transection and microsurgical repair are plotted as a function of time after repair with 1 hour sham or 20-Hz electrical stimulation. “Staggered motor axonal
regeneration” into the femoral nerve branches (C) and across the suture site (D) results in “preferential motor reinnervation” of the appropriate motor branch
(E). Electrical stimulation accelerates regeneration of axons across the suture site (F,H) and into the appropriate motor branch (G) with the arrows denoting a signifi-
cant increase (P < .01). The number of motoneurons in (H) represents new crossing events with each value being the mean motoneuron number labeled at the end of
the interval minus the mean labeled at the beginning of the interval. The rapid increase in crossing event that peaks between 1 and 2 weeks reflects the recruitment
of many motoneurons to regenerate across the suture line and penetrate the distal stump earlier with electrical stimulation than without stimulation. Adapted from
Al-Majed et al. 2000; Brushart et al. 2002.
progressive failure of chronically denervated SCs to support demonstrated in rat experiments in which the period of
the regenerating axons that play the predominant role in the motoneuronal axotomy was experimentally prolonged for
clinical failures of functional recovery after surgical repair up to a year before evaluating regenerative success (Boyd
(Boyd and Gordon 2002, 2003a,b; Fu and Gordon 1995a,b; and Gordon 2001, 2002, 2003a; Fu and Gordon 1995a). By
Gordon and Fu 1997; Gordon et al. 2011; Midha et al. 2005; counting backlabeled chronically axotomized motoneurons
Sulaiman and Gordon 2000, 2002, 2003, 2009; Sulaiman that regenerated their axons into a freshly denervated nerve
et al. 2002). The poor functional recovery is exacerbated by and counting reinnervated motor units to determine how
delayed nerve surgery. many motoneurons reinnervate denervated muscles, we
reported that these numbers progressively declined to
approximately 33% of those motoneurons that regenerated
2.4.2 Prolonged Axotomy of Neurons Reduces
after immediate axotomy (see Fig. 55.3). The exponential
Regenerative Capacity and Expression of
decline in regenerative capacity of the chronically
Regeneration Associated Genes
axotomized motoneurons was paralleled by a decline in the
The inherent ability of axotomized neurons to regenerate upregulated (RAGs Tetzlaff et al. 1996) (see Fig. 55.1).
axons is not sustained over long periods of time. This was

A 140
MN number
Percent of regenerating axons (%)

120 MU number

100

80

60

40

20
B 140
0 MN number
0 20 40 60 80 100 120 140 160 180 200
Percent of regenerating axons (%)

120 MU number
Days of chronic axotomy
100

C 80

60

40

20

Brachial plexus 0
injuries 0 20 40 60 80 100 120 140 160 180 200
Days of chronic denervation

800
mm
= 800
day
s

Median and
ulnar injuries
at the wrist
100 mm = Denervated
100 days hand muscle

Figure 55.3 Schematic illustration of proximal nerve injuries that require prolonged regeneration times for the regenerating axons to traverse lengthy
distances to reinnervate distant targets such as the muscles in the hand (C). Over these long periods, numbers of motoneurons that regenerate axons
(MN number) and reinnervate muscles (motor unit [MU] number) decline with time reflecting the reduced regenerative capacity of the chroni-
cally axotomized neurons (A) and the regenerative support by the chronically denervated Schwann cells (C). These combined account for the poor
functional outcomes of nerve surgeries that require regeneration over long distances. Chronic denervation of target organs does contribute a small
component to poor outcomes but irreversible atrophy of target organs and muscle in particular, is not the cause of these poor outcomes.

706 • NEUROGLIA
The dramatic negative effect of reduced regenerative capac- sluggish rate of muscle atrophy compared with a drastic loss of the
ity of the chronically axotomized motoneurons did not affect trophic environment of the distal nerve stumps after long-term
the ability of the neurons to reinnervate up to three times as denervation (Sulaiman and Gordon 2000). The declining num-
many muscle fibers as normal, the fewer motoneurons enlarg- ber of motoneurons that did regenerate their axons to make tar-
ing their motor units and the number of muscle fibers supplied get connections formed maximally enlarged motor units. These
by each reinnervating motoneuron (Fu and Gordon 1995b). enlarged units could not compensate for the approximately
Thus, chronic axotomy impairs the regenerative capacity of 95% reduction in numbers of functional motoneurons (Fu and
motoneurons, but does not impair the capacity of the smaller Gordon 1995b). The sustained capacity of denervated muscle
number of regenerated motor axons to make functional con- to accept reinnervation was also demonstrated by the parallel
nections with denervated muscle fibers. recovery of both muscle weight and isometric force. Nonetheless,
isolation of the effects of SC denervation from muscle denerva-
tion identified that chronically denervated muscle fibers did not
2.4.3 Prolonged Denervation Reduces the
fully recover their former size, arguing that there are limited
Capacity of Denervated Schwann Cells to Support
numbers of satellite cells that are incorporated into each atro-
Axonal Regeneration and Their Expression of
phic muscle fiber to recover muscle fiber cross-sectional area
Regeneration-Associated Genes
(Gordon et al. 2011a). Hence, progressive deterioration of the
The counts of axons regenerating into a chronically denervated growth-supportive capacity of SCs in the distal nerve stumps
nerve stump after 100 days in early experiments conducted during plays a primary role in poor functional recovery after nerve injury
World War II, indicated that the rate of outgrowth and the axon and the role of muscle atrophy is secondary.
numbers were not affected by delayed nerve repair (Gutmann It was striking that the long-term chronically denervated SCs
and Young 1944; Holmes and Young 1942). These findings with maintained their capacity to remyelinate the fewer axons that
evidence of extreme atrophy of the denervated muscles was the regenerated (see Fig. 55.3) (Sulaiman and Gordon 2000), partic-
basis for the enduring view that it is the irreversible atrophy of ularly in view of the progressive regression of the capacity of the
denervated targets and their replacement by connective tissue denervated SCs to sustain their growth permissive phenotype
and fat that is the prime basis for poor functional recovery after (see Fig. 55.1) and the progressive decline in numbers of growth
nerve repair (Gutmann and Young 1944; Holmes and Young supportive SCs in the chronically denervated distal nerve stumps
1942). However, counts of axons that regenerated into the distal (Li et al. 1997; You et al. 1997).
nerve stumps severely overestimate the number of neurons that
regenerate their axons, especially when regenerated axons are
2.5 M ISDIR E CT I ON OF R E G E N E R AT IN G AXONS
counted before target reinnervation. This is because each parent
axon gives rise to an average of five daughter sprouts (see section
2.3.3). Moreover, the axonal counts made just distal to the repair
2.5.1 Appropriate Reinnervation After
site are unlikely to represent the number of axons that regener-
Crush Injuries:
ate as far as the denervated muscle targets because of the progres-
sive loss of the capacity of chronically denervated SCs to support Functional outcomes after axonal injuries are best after nerve
axonal regeneration (Fig. 55.3B). We tested the latter possibility crush injuries in which the endoneurium remains anatomically
in experiments in which we enumerated both the number of intact all the way to the target and axonal sprouts of the regener-
freshly axotomized motoneurons that regenerated their axons ating unit are contained within the original endoneurium such
into chronically denervated nerve stumps and the number of rein- that the regenerating axons are “led” back to their original tar-
nervated motor units in the target muscle after cross-suture of a gets (Sunderland 1978). With time, reinnervated muscle fibers
freshly axotomized nerve to a chronically denervated distal nerve display the normal mosaic distribution of muscle fiber types,
stump (see Fig. 55.3A) (Fu and Gordon 1995b; Sulaiman and motor unit and muscle isometric forces recover fully, the original
Gordon 2000). Under conditions in which the effect of chronic number of functional motor units is restored, and the numbers
SC denervation was isolated from the effect of chronic muscle and diameters of the myelinated nerve fibers return to normal
denervation, we noted that the permissive environment provided (Gordon and Pattullo 1993).
by denervated SCs is short-lived with the numbers of motoneu-
rons regenerating their axons through chronically denervated SCs
declining rapidly (Gordon et al. 2011a). The numbers of freshly
2.5.2 Misdirection of Axons After Transection Injuries
axotomized motoneurons that regenerated their axons and rein- The number of axons that regenerate into distal nerve stumps
nervated denervated muscles via the chronically denervated nerve and successfully remake target connection is far more variable
stumps, declined in parallel to a value of approximately 5% (see for transection injuries that disrupt endoneurial tubes. Axonal
Fig. 55.3) (Fu and Gordon 1995b; Sulaiman and Gordon 2000). sprouts emanating from the proximal nerve stump may enter
These findings provide strong evidence that poor axonal several different endoneurial tubes with target destinations that
regeneration is owing to the progressive failure of axons to regen- they did not formerly supply as in the case of motor axons that
erate in the atrophic nerve stumps and not an inability of chroni- regenerate within pathways leading to the skin rather than mus-
cally denervated muscle fibers to accept reinnervation after long cle (Brushart 1993). Regenerating axons fail to find their origi-
delays. In fact, wet weights of reinnervated muscles, which are nal targets, especially when large nerve trunks are damaged and
directly related to their isometric forces, demonstrated a very require surgical repair.

N E RVE R E G E N E R AT I O N I N T H E P E R I P H E R A L N E RVO U S SYS T E M • 707


After microsurgical repair of severed nerves to the human neurons that regenerate in chronically denervated distal nerve
hand, voluntary recruitment of the reinnervated motor units stumps (see Fig. 55.1). The misdirection of regenerating axons to
revealed the random reinnervation of several muscles across inappropriate targets exacerbates the reduced numbers of neu-
the hand by the motoneurons that had previously supplied rons that regenerate their axons and reinnervate denervated tar-
only one muscle. The random distribution of regenerating gets. Strategies developed to accelerate axonal regeneration and
axons to different muscles essentially negated any fine con- counter the deleterious effects of long delays are presented here.
trol of hand movement (Thomas et al. 1987). Animal studies
of motor axonal regeneration also revealed random reinner-
3.1 AC C E L E R AT I N G AXO N O U TG ROW T H
vation of muscles after surgical repair of transected nerves.
Gillespie et al. (1987) demonstrated random distribution of
slow and fast motor units in reinnervated soleus and lateral
gastrocnemius rat muscles after surgical repair of the common
3.1.1 Conditioning Lesions Are Effective but
lateral gastrocnemius-soleus (LGS) nerve to the two muscles.
Not Feasible :
These studies demonstrate the inability of transected motor Several attempts have been made to accelerate the relatively slow
axons to “find” their original endoneurial tubes, leading to the rate of axonal regeneration to optimize the recovery of function
appropriate muscles. Experiments that applied retrograde dyes after nerve injuries. Of the many attempts made, only the condi-
to regenerated axons of transected sciatic and facial nerves in tioning crush lesion of the intact nerve before a more proximal
rats directly demonstrated misdirection of regenerating motor test crush lesion, significantly increases rate of axonal regenera-
axons from several different motoneuron pools to muscles tion (reviewed by Grafstein and McQuarrie 1978). The lesion ele-
that the motoneurons did not formerly supply (Brushart and vates mRNA expression of actin and tubulin and reduces levels of
Mesulam 1980). In cases in which the nerves supplied muscles neurofilament expression even further than normal in association
with antagonistic actions, axonal misdirection was associated with more rapid transport of these cytoskeletal proteins (Tetzlaff
with inappropriate movements, quite consistent with findings et al. 1996). The parallel increase in the rates of slow component b
that flexor and extensor motoneurons sustain their normal but of axonal transport of the cytoskeletal proteins and axonal regen-
inappropriate pattern of firing when they are directed to inap- eration in the injured neurons after the conditioning lesion pro-
propriately reinnervate extensor and flexor muscles, respec- vided further evidence for a direct causal link between these rates
tively (Gordon et al. 1986). (Hoffman and Lasek 1980; Jacob and McQuarrie 1991). The
more rapid transport of the cytoskeletal proteins into the growth
cones prevents their collapse (Jacob and McQuarrie 1991).
2.5.3 Preferential Motor Reinnervation
The conditioning lesion accelerates both outgrowth of
There are several peripheral nerve trunks whose branches regenerating axons that normally begins within hours of the
innervate skeletal muscles and sense organs in the skin and the injury and the later SC migration from the proximal stump after
joints. Examples include the femoral nerve with two branches, approximately 2 days (Torigoe et al. 1999). These findings argue
the muscle branch containing motor and sensory nerves to the for a role of both of the neurons and the SCs in the accelerated
quadriceps muscle, and the other pure sensory saphenous nerve axonal regeneration rate after a conditioning lesion. It has not yet
branch to the skin (Brushart 1993). Although reinnervation been ascertained as to whether the conditioning lesion, which so
of motor and sensory nerve branches is initially random, the effectively accelerates axonal outgrowth and regeneration rate, is
motoneurons that progressively regenerate their axons across the also effective in synchronizing the staggered axonal outgrowth
suture site send their axons into the appropriate motor branch across the suture site. This staggered outgrowth normally delays
(Al-Majed et al. 2000; Brushart 1993). This “preferential motor the process of axonal regeneration by many weeks (Al-Majed
reinnervation” emerged in parallel with the progressive or stag- et al. 2000; Brushart et al. 2002) (see Fig. 55.2). The condition-
gered regeneration of motor axons into the distal nerve stumps ing lesion is nevertheless the only technique that has been dem-
(see Fig. 55.2E,F) (Al-Majed et al. 2000). onstrated to accelerate the rate of regeneration. The important
An unusual acidic glycan associated with myelin profiles contraindication for the practical use of the conditioning lesion
of motor but not sensory mouse axons that is recognized by a to promote axon regeneration is that it must precede the test
monoclonal antibody L2/HNK-1 may be one mechanism of the injury to be effective (Jacob and McQuarrie 1993). This delay
preferential reinnervation of appropriate pathways (Eberhardt obviously negates the conditioning lesion as a clinically viable
et al. 2006; Martini et al. 1994). Another is the differential neu- technique to promote axonal regeneration in human patients.
rotrophic factor profile in SCs derived from sensory and motor
axons with the BDNF/NT4/5 profile likely to play an impor-
tant role in preferential reinnervation of motor pathways (Hoke
3.1.2 Brief Electrical Stimulation Accelerates
et al. 2006).
Axon Outgrowth
Early promising studies reported that continuous low fre-
quency electrical stimulation (ES) accelerates muscle rein-
3 S T R AT E G I E S TO P R O M OT E AXO N A L nervation (Nix and Hopf 1983; Pockett and Gavin 1985).
R E G E N E R AT I O N The data were not necessarily explained by more rapid nerve
regeneration. Using backlabeling to count motoneurons that
Long delays incurred by staggered and slow rate of axonal regen- regenerated their axons, we demonstrated that superthres-
eration frequently result in chronic axotomy of the injured hold low frequency ES immediately after the surgical repair
708 • NEUROGLIA
of transected femoral nerve accelerated axon regeneration of being ineffective, coincident with downregulation of neuronal
axons from all the axotomized motoneurons. The duration of BNDF (Geremia et al. 2007). Several laboratories have con-
the stimulation could be reduced to as short as 1 hour and the firmed the positive effect of ES in accelerating regeneration in
effect required that action potentials be conducted to the cell rat and mouse models of transected nerves (Asensio-Pinilla et al.
bodies (see Fig. 55.2G) (Al-Majed et al. 2000). Electrical stimu- 2009; Eberhardt et al. 2006; English et al. 2007; Franz et al.
lation accelerated axon outgrowth across the suture site (see Fig. 2008; Huang et al. 2010; Sharma et al. 2009; Yeh et al. 2010).
55.2F), but did not accelerate regeneration rate (see Fig. 55.4H)
(Brushart et al. 2002). The ES effect is to synchronize the
3.1.3 Electrical Stimulation Accelerates
growth of axons across the surgical suture line by accelerating
Target Reinnervation
the axon outgrowth from the proximal nerve stump and their
passage across the suture line (Brushart et al. 2002). Electrical stimulation accelerates functional return in rats
The accelerating effect of the 1-hour period of 20 Hz ES was (Eberhardt et al. 2006). In a proof of principle study of nerve
also demonstrated in sensory neurons where it promotes the regeneration and target reinnervation in humans, we counted
appropriate reinnervation of sensory nerve pathways (Brushart reinnervated motor units after carpal tunnel release surgery
et al. 2005). Only a 1-hour period stimulation was effective in with and without immediate 20-Hz 1-hour electrical stimula-
promoting sensory nerve regeneration, longer periods of ES tion (Gordon et al. 2010) (Fig. 55.4). Choosing only patients

Carpal Median nerve is released.


Incisition site tunnel

B 20Hz/
1hr ES

Median EMG
− nerve recording

Carpel
+ tunnel
release
surgery

C Control D Stimulation

400 * *
Number of innervated

300 Normal
motor units

300
Pre-op
x±SE
100

0
3m 6-8 m 12 m 3m 6-8 m 12 m

Figure 55.4 Immediately following open carpal tunnel release surgery (A) in patients with severe carpal tunnel syndrome (loss of >50% of functional motor
units in the thenar eminence muscle), the median nerve was stimulated at 20 Hz for 1 hour via implanted electrodes and action potentials generated were
monitored with electromyography. Mean (+SE) of the number of innervated motor units did not change from preoperative levels in the control unstim-
ulated group of patients. In contrast, there was already a significant increase by 6 months after the 1-hour stimulation with the number of functional
motor units at 1 year being equal to the normal levels.

N E RVE R E G E N E R AT I O N I N T H E P E R I P H E R A L N E RVO U S SYS T E M • 709


with severe carpal tunnel syndrome with electrophysiological 3.2 C O U N T E R AC T I N G T H E N EG AT I VE
evidence of loss of more than 50% of their functional motor EFFEC TS O F C H RO N I C AXOTO MY O F
units in the median eminence, the effects of the ES were strik- I N JU R E D N EU RO NS
ing. Especially as the number of reinnervated motor units did
3.2.1 By Neurotrophic Factors
not increase significantly even after 1 year without the stimu-
lation, the finding that the numbers had increased within 3 3.2.1.1 Exogenous Sources of Neurotrophic Factors
months in the stimulated group and reached normal levels 6 The expression of neurotrophic factors by axotomized neu-
months after the release and ES was striking (see Fig. 55.4). rons and denervated SCs peaks within weeks and declines
thereafter, the time course of the decline varying according
to the factor (Boyd and Gordon 2003b). In general, this time
3.1.4 Role of Neurotrophic Factors and cAMP course parallels the course of decline in capacity of chroni-
The effect of ES on accelerating axonal growth across a suture cally axotomized neurons to regenerate their axons (see sec-
site is associated with an accelerated and augmented upreg- tion 4.2). This suggested that exogenous application of these
ulation of endogenous neuronal BDNF and trkB followed factors might reverse the deleterious effects of chronic axo-
shortly after by elevated expression of RAGs (Al-Majed tomy on nerve regeneration (see Fig. 55.1). Indeed this proved
et al. 2000, 2004; Geremia et al. 2007). Knockout experi- to be the case. Exogenous supply of BDNF and/or GDNF
ments in transgenic mice demonstrated that positive ES significantly increased regenerating motoneuron numbers,
effect on regeneration is dependent on neuronal expression application of appropriate doses for a month during regen-
of NT4/5 (English et al. 2007). eration being effective in restoring regenerative competence
Strong links have also been made between activity-induced to that seen after immediate nerve repair (Boyd and Gordon,
elevations in BDNF, intracellular calcium and intracellular 2001, 2002, 2003a,b; Gordon et al. 2003). Not surprisingly,
cyclic adenosine monophosphate (cAMP) that in turn, these neurotrophic factors had no effect on regeneration
promote neurite outgrowth and axonal regeneration on both after immediate nerve suture. This is consistent with there
permissive and nonpermissive substrates in vitro and in vivo being sufficient endogenous supplies of neurotrophic fac-
(Qiu et al. 2002). The positive effects of ES in promoting axon tors by the axotomized neurons and SCs in the denervated
outgrowth is associated with elevation of cAMP, suggesting that distal nerve stumps after immediate nerve repair (Fig. 55.5).
the ES mediates its effect via its upregulation of neurotrophins GDNF acting on its receptors was effective at lower doses
and elevated cAMP, the effects likely converging on cAMP than BDNF and always facilitated axon regeneration (Boyd
response element-binding (CREB) to promote synthesis of and Gordon 2003a). BDNF had a bimodal dose-response
cytoskeletal proteins and/or rearrangement of these proteins effect with its trkB receptors mediating a positive effect on
to accelerate axon outgrowth (Aglah et al. 2008). Rolipram, a axon regeneration and p75 receptors a negative effect that
phosphodiesterase inhibitor that increases cAMP, mimicked acts to negate the positive effects of trkB activation (Boyd
ES in accelerating axon outgrowth (Udina et al. 2010). and Gordon 2001).

Distal stump
Neurotrophic factors

BDNF
and receptors

Distal
stump
MN GDNF
BDNF
Ret
TrkB
GFR-α

Motoneuron pool
Time (d) 0 10 20 30 40 50 60

10 mm

Site of
axotomy Schwann cell - growth supportive
Schwann cell - Atrophied

Figure 55.5 The time course of a month for all axons to regenerate across the suture site into the denervated distal nerve stump (Brushart et al. 2002)
corresponds with the time during which neurotrophic factors and their receptors in the distal nerve stump peak and, in most cases, decline. As a result
neurons regenerate their axons through the distal nerve stump as the Schwann cells progressively lose their growth support and become atro-
phic. Adapted from Furey et al. 2007.

710 • NEUROGLIA
3.2.1.2 Endogenous Sources of Neurotrophic Factors phenotype in addition to its mitogenic effects on the SCs
Low frequency ES upregulates BDNF and GDNF and its (Unsicker and Strelau 2000) (see section 2.2.2.1). TGF-β
receptors in axotomized motoneurons with NT4/5 appearing increased SC proliferation within 2 days in vitro and forsko-
to be a critical factor that mediates the neurotrophic effect of lin augmented the effects of TGF-β presumably by increas-
accelerated axon outgrowth (Al-Majed et al. 2000; English ing intracellular levels of cAMP. To assess the capacity of
et al. 2007). This is likely the reason for ES being effective in reactivated SCs to support axonal growth, we reactivated
promoting the regeneration of axons from chronically axo- chronically denervated SCs in isolated nerve stumps with
tomized neurons (Gordon et al. 2011b). The upregulation of TGF-β and forskolin for 2 days in vitro prior to placing
neurotrophic factors in freshly axotomized motoneurons was these reactivated SCs in a silastic cuff between proximal
effective in promoting axon regeneration even through chron- and distal nerve stumps in vivo. The effect was dramatic.
ically denervated nerves (Gordon et al. 2011b). The TGF-β- and forskolin-treated SCs increased the regen-
eration of axons through the silastic cuff fourfold as com-
3.2.2 FK506 Application pared with number of motoneurons that regenerated axons
into chronically denervated distal nerve stumps without
The immunophilin ligand FK506 that enhances neurite out- pretreatment. Moreover, the regenerated nerve fibers were
growth in vitro and accelerates regeneration rate after crush larger and better myelinated than those fibers that regener-
injury and after section and immediate resuture (Gold et al. ated through the untreated nerve explants (Sulaiman and
1995), was very effective in counteracting the negative effects Gordon 2002). These findings are consistent with previ-
of chronic axotomy to promote axonal regeneration. Twice as ous reports that chronically denervated SCs sustain their
many chronically axotomized motoneurons regenerated their capacity to undergo proliferation and remyelinate regen-
axons after 3-week daily subcutaneous injection in association erated axons (Sulaiman and Gordon 2000). They provide
with a corresponding increase in the number of regenerated promise that the reduced numbers of SCs that remain in
axons (Sulaiman et al. 2002). The effects are mediated via chronically denervated nerve stumps can be reactivated by
FKBP-52, a chaperone component of mature steroid recep- exposure to cytokines that are normally released during the
tor complexes, independent of the FK506-binding protein-12, process of Wallerian degeneration. The reinstated growth-
that mediates its immunosuppressive effects (Gold et al. 1999). supportive phenotype of the SCs is sufficient to support
Evidence of increased numbers of regenerated axons and the axonal regeneration.
more rapid progression of axon remyelination indicated that the
FK-506 also accelerated the rate of axon outgrowth, the effect
being observed in both motor and sensory neurons (Sulaiman 3.3.3 Side-to-Side Transfers to “Protect”
et al. 2002). Chronically Denervated Schwann Cells
Nerve autografts have been placed between a donor nerve and a
3.3 CO U N T E R AC T I N G T H E EFFEC TS O F recipient denervated distal nerve stump to promote axon crossing
P RO L O N G E D D E N E RVAT I O N O F T H E and growth within the denervated stump (Viterbo et al. 2009).
D I S TA L N E RV E S T U M P S Using this technique, we investigated whether these side-to-side
transfers could be used to “protect” a chronically denervated
3.3.1 Electrical Stimulation nerve stump before the ingrowth of axons after delayed nerve
Brief ES significantly increased the number of motor and sen- repair or from a more proximal nerve injury and surgical repair
sory neurons that regenerated their axons into a chronically (Ladak et al. 2011). Using a model of side-to-side bridging in rat
denervated nerve stump (Gordon et al. 2010). This finding hindlimb nerves, we reported that the small number of donor
indicated that ES may raise neurotrophic factor levels in the nerves that regenerate their axons into the denervated nerve stump
neurons sufficiently to “jump start” axon outgrowth into less do provide “protection” of the SCs, significantly increasing the
permissive growth environments provided by atrophic SCs number of neurons that regenerate their axons successfully into
in the chronically denervated nerve stumps. It is also possible the chronically denervated and protected distal nerve stumps.
that the ES may release sufficient neuregulins from the proxi- This technique is particularly attractive for the protection of
mal nerve stump to “reawaken” dormant SCs to support axon long denervated distal nerve stumps such as the ulnar nerve after
regeneration. Indeed, the effective myelination of those axons a brachial plexus injury because donor nerves that cross inserted
that do regenerate through chronically denervated nerve side-to-side bridges regenerate in both proximal and distal
stumps provides evidence that the reawakening is possible directions (Gordon and Borschel, unpublished findings). Hence
(Sulaiman and Gordon 2000). these donor nerves may protect the distal nerve sufficiently to
allow for more effective regeneration of axons over large distances
and in turn, to reinnervate denervated targets.
3.3.2 TGF-β to Promote Permissive Growth
Phenotype in Schwann Cells
A possible role of the cytokine TGF-β in counteracting the 4 S U M M A RY A N D P E R S P E C T I VE S
deleterious effects of chronic SC denervation was suggested
by the results of several in vitro studies that TGF-β plays a The contributions of the injured neurons, SCs and mac-
role in maintaining the growth-promoting denervated SC rophages to the regenerative process in the peripheral nervous

N E RVE R E G E N E R AT I O N I N T H E P E R I P H E R A L N E RVO U S SYS T E M • 711


system appear to be interlinked such that there is a window of Brushart TM. 1993. Motor axons preferentially reinnervate motor path-
opportunity during which the growth potential of the neurons ways. J Neurosci 13:2730–2738.
Brushart TM. 2011. Nerve Repair. New York: Oxford University Press.
translates into outgrowth and extension of regenerating axons. Brushart TM, Hoffman PN, Royall RM, Murinson BB, Witzel C,
This window is closely linked to the period during which there Gordon T. 2002. Electrical stimulation promotes motoneuron
is active Wallerian degeneration when macrophages interact regeneration without increasing its speed or conditioning the neuron.
with SCs to provide the permissive growth environment. The J Neurosci 22:6631–6638.
progressive but slow advancement of regenerating axons from Brushart TM, Jari R, Verge V, Rohde C, Gordon T. 2005. Electrical
stimulation restores the specificity of sensory axon regeneration. Exp
the proximal nerve across the site of nerve transection and Neurol 194:221–229.
repair severely slows the progress of axonal growth into the dis- Brushart TM, Mesulam MM. 1980. Transganglionic demonstration of
tal nerve stumps. The delay exacerbates the lengthy time peri- central sensory projections from skin and muscle with HRP-lectin
ods during which the neurons remain chronically axotomized conjugates. Neurosci Lett 17:1–6.
and the SCs remain chronically denervated. As we begin to Büngner OV. 1891. On the regeneration and degeneration of nerve-
processes to injury. Beitr Path Anat Allg Path 10:321–393.
understand the limitations to peripheral nerve regeneration, Cajal SR. 1928. Degeneration and regeneration of the nervous system.
we can begin to devise effective interventions to prolong the New York: Oxford University Press.
window of opportunity for axonal regeneration and promote Chernousov MA, Carey DJ. 2000. Schwann cell extracellular matrix
recovery of function after nerve injuries. Although exogenous molecules and their receptors. Histol Histopathol 15:593–601.
sources of neurotrophic factors for example, may be effective, Devor M, Govrin-Lippmann R. 1979. Maturation of axonal sprouts after
nerve crush. Exp Neurol 64:260–270.
we should continue to pursue methods that are not only read- Dunaevsky A, Connor EA. 1998. Stability of frog motor nerve terminals
ily applied clinically, as in electrical stimulation, but also that in the absence of target muscle fibers. Dev Biol 194:61–71.
effectively upregulate endogenous sources of molecules that Eberhardt KA, Irintchev A, Al-Majed AA, Simova O, Brushart TM,
are needed for effective nerve regeneration. Gordon T, et al. 2006. BDNF/TrkB signaling regulates HNK-1
carbohydrate expression in regenerating motor nerves and pro-
motes functional recovery after peripheral nerve repair. Exp Neurol
198:500–510.
REFERENCES Einheber S, Hannocks MJ, Metz CN, Rifk in DB, Salzer JL. 1995.
Transforming growth factor-beta 1 regulates axon/Schwann cell
Aglah C, Gordon T, Posse De Chaves EI. 2008. cAMP promotes neurite interactions. J Cell Biol 129:443–458.
outgrowth and extension through protein kinase A but independently English AW, Schwartz G, Meador W, Sabatier MJ, Mulligan A. 2007.
of Erk activation in cultured rat motoneurons. Neuropharmacology Electrical stimulation promotes peripheral axon regeneration
55:8–17. by enhanced neuronal neurotrophin signaling. Dev Neurobiol
Aitken JT, Sharman M, Young JZ. 1947. Maturation of peripheral nerve 67:158–172.
fibres with various peripheral connections. J Anat 81:1–22. Franz CK, Rutishauser U, Rafuse VF. 2008. Intrinsic neuronal proper-
Al-Majed AA, Brushart TM, Gordon T. 2000. Electrical stimulation accel- ties control selective targeting of regenerating motoneurons. Brain
erates and increases expression of BDNF and trkB mRNA in regener- 131:1492–1505.
ating rat femoral motoneurons. Eur J Neurosci 12:4381–4390. Fricker FR, Lago N, Balarajah S, Tsantoulas C, Tanna S, Zhu N, et al.
Al-Majed AA, Neumann CM, Brushart TM, Gordon T. 2000. Brief 2011. Axonally derived neuregulin-1 is required for remyelina-
electrical stimulation promotes the speed and accuracy of motor tion and regeneration after nerve injury in adulthood. J Neurosci
axonal regeneration. J Neurosci 20:2602–2608. 31:3225–3233.
Al-Majed AA, Tam SL, Gordon T. 2004. Electrical stimulation accel- Fry EJ, Ho C, David S. 2007. A role for Nogo receptor in macrophage
erates and enhances expression of regeneration-associated genes clearance from injured peripheral nerve. Neuron 53:649–662.
in regenerating rat femoral motoneurons. Cell Mol Neurobiol Fu SY, Gordon T. 1995a. Contributing factors to poor functional
24:379–402. recovery after delayed nerve repair: prolonged axotomy. J Neurosci
Asensio-Pinilla E, Udina E, Jaramillo J, Navarro X. 2009. Electrical 15:3876–3885.
stimulation combined with exercise increase axonal regeneration Fu SY, Gordon T. 1995b. Contributing factors to poor functional recov-
after peripheral nerve injury. Exp Neurol 219:258–265. ery after delayed nerve repair: prolonged denervation. J Neurosci
Atanasoski S, Scherer SS, Sirkowski E, Leone D, Garratt AN, Birchmeier 15:3886–3895.
C, et al. 2006. ErbB2 signaling in Schwann cells is mostly dispensable Fu SY, Gordon T. 1997. The cellular and molecular basis of peripheral
for maintenance of myelinated peripheral nerves and proliferation of nerve regeneration. Mol Neurobiol 14:67–116.
adult Schwann cells after injury. J Neurosci 26:2124–2131. Furey MJ, Midha R, Xu QG, Belkas J, Gordon T. 2007. Prolonged target
Avellino AM, Hart D, Dailey AT, MacKinnon M, Ellegala D, Kliot deprivation reduces the capacity of injured motoneurons to regener-
M. 1995. Differential macrophage responses in the peripheral and ate. Neurosurgery 60:723–732.
central nervous system during wallerian degeneration of axons. Exp Georgiou J, Robitaille R, Charlton MP. 1999. Muscarinic control of
Neurol 136:183–198. cytoskeleton in perisynaptic glia. J Neurosci 19:3836–3846.
Boyd JG, Gordon T. 2001. The neurotrophin receptors, trkB and p75, differ- Geraldo S, Gordon-Weeks PR. 2009. Cytoskeletal dynamics in
entially regulate motor axonal regeneration. J Neurobiol 49:314–325. growth-cone steering. J Cell Sci 122:3595–3604.
Boyd JG, Gordon T. 2002. A dose-dependent facilitation and inhibition Geremia NM, Gordon T, Brushart TM, Al-Majed AA, Verge VM. 2007.
of peripheral nerve regeneration by brain-derived neurotrophic fac- Electrical stimulation promotes sensory neuron regeneration and
tor. Eur J Neurosci 15:613–626. growth-associated gene expression. Exp Neurol 205:347–359.
Boyd JG, Gordon T. 2003a. Glial cell line-derived neurotrophic fac- Gillespie MJ, Gordon T, Murphy PR. 1986. Reinnervation of the lateral
tor and brain-derived neurotrophic factor sustain the axonal regen- gastrocnemius and soleus muscles in the rat by their common nerve.
eration of chronically axotomized motoneurons in vivo. Exp Neurol J Physiol 372:485–500.
183:610–619. Gillespie MJ, Gordon T, Murphy PR. 1987. Motor units and histochem-
Boyd JG, Gordon T. 2003b. Neurotrophic factors and their receptors in istry in rat lateral gastrocnemius and soleus muscles: evidence for
axonal regeneration and functional recovery after peripheral nerve dissociation of physiological and histochemical properties after rein-
injury. Mol Neurobiol 27:277–324. nervation. J Neurophysiol 57:921–937.

712 • NEUROGLIA
Gold BG, Densmore V, Shou W, Matzuk MM, Gordon HS. 1999. Mackinnon SE, Dellon AL, O’Brien JP. 1991. Changes in nerve fiber
Immunophilin FK506-binding protein 52 (not FK506-binding pro- numbers distal to a nerve repair in the rat sciatic nerve model. Muscle
tein 12) mediates the neurotrophic action of FK506. J Pharmacol Nerve 14:1116–1122.
Exp Ther 289:1202–1210. Martini R, Schachner M. 1988. Immunoelectron microscopic local-
Gold BG, Katoh K, Storm-Dickerson T. 1995. The immunosuppressant ization of neural cell adhesion molecule (L1, N-CAM, and
FK506 increases the rate of axonal regeneration in rat sciatic nerve. J Myelin-associated glycoprotein) in regenerating adult mouse sciatic
Neurosci 15:7509–7516. nerve. J Cell Biol 106:1735–1746.
Gordon T, Amirjani N, Edwards DC, Chan KM. 2010. Brief post-surgical Martini R, Schachner M, Brushart TM. 1994. The L2/HNK-1 carbohy-
electrical stimulation accelerates axon regeneration and muscle rein- drate is preferentially expressed by previously motor axon-associated
nervation without affecting the functional measures in carpal tunnel Schwann cells in reinnervated peripheral nerves. J Neurosci
syndrome patients. Exp Neurol 223:192–202. 14:7180–7191.
Gordon T, Falk V, Verge VMK, Tyreman N. 2010. Brief electrical stimu- Midha R, Munro CA, Chan S, Nitising A, Xu QG, Gordon T. 2005.
lation (ES) of transected peripheral nerve promotes axon regeneration Regeneration into protected and chronically denervated peripheral
through chronically denervated distal nerve stumps. Soc Neurosci nerve stumps. Neurosurgery 57:1289–1299.
35:541. Miledi R, Slater CR. 1970. On the degeneration of rat neuromuscular
Gordon T, Fu SY. 1997. Long-term response to nerve injury. Adv Neurol junctions after nerve section. J Physiol 207:507–528.
72:185–199. Nix WA, Hopf HC. 1983. Electrical stimulation of regenerating nerve
Gordon T, Pattullo MC. 1993. Plasticity of muscle fiber and motor unit and its effect on motor recovery. Brain Res 272:21–25.
types. Exerc Sport Sci Rev 21:331–362. Pockett S, Gavin RM. 1985. Acceleration of peripheral nerve regenera-
Gordon T, Stein RB, Thomas CK. 1986. Organization of motor units tion after crush injury in rat. Neurosci Lett 59:221–224.
following cross-reinnervation of antagonistic muscles in the cat hind Qiu J, Cai D, Dai H, McAtee M, Hoff man PN, Bregman BS, et al. 2002.
limb. J Physiol 374:443–456. Spinal axon regeneration induced by elevation of cyclic AMP. Neuron
Gordon T, Sulaiman OAR, Boyd JG. 2003. Experimental strategies to 34:895–903.
promote functional recovery after peripheral nerve injuries. J Periph Shamash S, Reichert F, Rotshenker S. 2002. The cytokine net-
Nerv Syst 8:236–250. work of Wallerian degeneration: tumor necrosis factor-alpha,
Gordon T, Tyreman N, Raji MA. 2011a. The basis for diminished func- interleukin-1alpha, and interleukin-1beta. J Neurosci 22:3052–3060.
tional recovery after delayed peripheral nerve repair. J Neurosci Sharma N, Coughlin L, Porter RG, Tanzer L, Wurster RD, Marzo SJ,
31:5325–5334. et al. 2009. Effects of electrical stimulation and gonadal steroids
Gordon T, Verge VMK, Tyreman N. 2011b. Brief one hour 20Hz elec- on rat facial nerve regenerative properties. Restor Neurol Neurosci
trical stimulation (ES) of chronically axotomized peripheral nerve 27:633–644.
promotes axon regeneration through freshly denervated distal nerve Son YJ, Thompson WJ. 1995. Schwann cell processes guide regeneration
stumps. Soc Neurosci 36:439. of peripheral axons. Neuron 14:125–132.
Grafstein B, McQuarrie IG. 1978. Role of the nerve cell body in axonal Stoll G, Muller HW. 1999. Nerve injury, axonal degeneration and neural
regeneration. In: Cotman CW (ed.), Neuronal plasticity. New York: regeneration: basic insights. Brain Pathol 9:313–325.
Raven, pp. 155–196. Sulaiman OA, Gordon T. 2009. Role of chronic Schwann cell denerva-
Gutmann E, Guttmann L, Medawar PB, Young JZ. 1942. The rate of tion in poor functional recovery after nerve injuries and experimental
regeneration of nerve. J Exp Biol 19:14–44. strategies to combat it. Neurosurgery 65:A105-A114.
Gutmann E, Young JZ. 1944. The re-innervation of muscle after various Sulaiman OAR, Gordon T. 2000. Effects of short- and long-term
periods of atrophy. J Anat 78:15–44. Schwann cell denervation on peripheral nerve regeneration, myelina-
Hall SM. 1986. Regeneration in cellular and acellular autografts in the tion, and size. Glia 32:234–246.
peripheral nervous system. Neuropathol Appl Neurobiol 12:27–46. Sulaiman OAR, Gordon T. 2002. Transforming growth factor-beta
Hoffman PN, Lasek RJ. 1980. Axonal transport of the cytoskeleton and forskolin attenuate the adverse effects of long-term Schwann
in regenerating motor neurons: constancy and change. Brain Res cell denervation on peripheral nerve regeneration in vivo. Glia 37:
202:317–333. 206–218.
Hoke A, Gordon T, Zochodne DW, Sulaiman OAR. 2002. A decline Sulaiman OAR, Gordon T. 2003. TGF-beta reverses the deleterious
in glial cell-line-derived neurotrophic factor expression is associated effect of long-term Schwann cell denervation on nerve regeneration
with impaired regeneration after long-term Schwann cell denerva- by inducing erbB3 receptor expression. Glia 24:A105–A114.
tion. Exp Neurol 173:77–85. Sulaiman OAR, Midha R, Gordon T. 2011. Pathophysiology of surgical
Hoke A, Redett R, Hameed H, Jari R, Zhou C, Li ZB, et al. 2006. nerve disorders. In: Winn HR (ed.), Yeomans neurological surgery.
Schwann cells express motor and sensory phenotypes that regulate Philadelphia: Saunders.
axon regeneration. J Neurosci 26:9646–9655. Sulaiman OAR, Midha R, Munro CA, Matsuyama T, Al-Majed AA,
Holmes W, Young JZ. 1942. Nerve regeneration after immediate and Gordon T. 2002. Chronic Schwann cell denervation and the pres-
delayed suture. J Anat 77:63–108. ence of a sensory nerve reduce motor axonal regeneration. Exp Neurol
Huang J, Lu L, Hu X, Ye Z, Peng Y, Yan X, et al. 2010. Electrical stimula- 176:342–354.
tion accelerates motor functional recovery in the rat model of 15-mm Sulaiman OAR, Voda J, Gold BG, Gordon T. 2002. FK506 increases
sciatic nerve gap bridged by scaffolds with longitudinally oriented peripheral nerve regeneration after chronic axotomy but not after
microchannels. Neurorehabil Neural Repair 24:736–745. chronic Schwann cell denervation. Exp Neurol 175:127–137.
Ide C. 1996. Peripheral nerve regeneration. Neurosci Res 25:101–121. Sunderland S. 1947. Rate of regeneration in human peripheral nerves:
Jacob JM, McQuarrie IG. 1991. Axotomy accelerates slow component b analysis of interval between injury and onset of recovery. Arch Neurol
of axonal transport. J Neurobiol 22:570–582. Psychiat 58(3):251–295.
Jacob JM, McQuarrie IG. 1993. Acceleration of axonal outgrowth in rat Sunderland S. 1978. Nerve and nerve injuries. Edinburgh: Livingstone.
sciatic-nerve at one week after axotomy. J Neurobiol 24:356–367. Tetzlaff W, Leonard C, Krekoski CA, Parhad IM, Bisby MA. 1996.
Ladak A, Schembri P, Olson J, Udina E, Tyreman N, Gordon T. 2011. Reductions in motoneuronal neurofi lament synthesis by successive
Side-to-side nerve grafts sustain chronically denervated peripheral axotomies: a possible explanation for the conditioning lesion effect
nerve pathways during axon regeneration and result in improved on axon regeneration. Exp Neurol 139:95–106.
functional reinnervation. Neurosurgery 68:1654–1666. Thomas CK, Stein RB, Gordon T, Lee RG, Elleker MG. 1987. Patterns
Li H, Terenghi G, Hall SM. 1997. Effects of delayed re-innervation of reinnervation and motor unit recruitment in human hand muscles
on the expression of c-erbB receptors by chronically denervated rat after complete ulnar and median nerve section and resuture. J Neurol
Schwann cells in vivo. Glia 20:333–347. Neurosurg Psychiatry 50:259–268.

N E RVE R E G E N E R AT I O N I N T H E P E R I P H E R A L N E RVO U S SYS T E M • 713


Torigoe K, Hashimoto K, Lundborg G. 1999. A role of migratory produced thereby in the structure of their primitive fibres. Philos
Schwann cells in a conditioning effect of peripheral nerve regenera- Trans R Soc London 140:423–429.
tion. Exp Neurol 160:99–108. Wang JT, Medress ZA, Barres BA. 2012. Axon degeneration: molecular
Udina E, Ladak A, Furey M, Brushart T, Tyreman N, Gordon T. 2010. mechanisms of a self-destruction pathway. J Cell Biol 196:7–18.
Rolipram-induced elevation of cAMP or chondroitinase ABC break- Witzel C, Rohde C, Brushart TM. 2005. Pathway sampling by regener-
down of inhibitory proteoglycans in the extracellular matrix pro- ating peripheral axons. J Comp Neurol 485:183–190.
motes peripheral nerve regeneration. Exp Neurol 223:143–152. Yeh CC, Lin YC, Tsai FJ, Huang CY, Yao CH, Chen YS. 2010. Timing
Unsicker K, Strelau J. 2000. Functions of transforming growth of applying electrical stimulation is an important factor decid-
factor-beta isoforms in the nervous system. Cues based on local- ing the success rate and maturity of regenerating rat sciatic nerves.
ization and experimental in vitro and evidence. Eur J Biochem Neurorehabil Neural Repair 24:730–735.
267:6972–6975. You S, Petrov T, Chung PH, Gordon T. 1997. The expression of the
Viterbo F, Amr AH, Stipp EJ, Reis FJ. 2009. End-to-side neurorrhaphy: low affinity nerve growth factor receptor in long-term denervated
past, present, and future. Plast Reconstr Surg 124:e351-e358. Schwann cells. Glia 20:87–100.
Vrbova G, Mehra N, Shanmuganathan H, Tyreman N, Schachner M, Zhai Q, Wang J, Kim A, Liu Q, Watts R, Hoopfer E, et al. 2003.
Gordon T. 2009. Chemical communication between regenerat- Involvement of the ubiquitin-proteasome system in the early stages of
ing motor axons and Schwann cells in the growth pathway. Eur J wallerian degeneration. Neuron 39:217–225.
Neurosci 30:366–375. Zhou FQ, Walzer M, Wu YH, Zhou J, Dedhar S, Snider WD. 2006.
Waller A. 1850. Experiments on the section of the glossopharyngeal and Neurotrophins support regenerative axon assembly over CSPGs by an
hypoglossal nerves of the frog, and observations of the alterations ECM-integrin-independent mechanism. J Cell Sci 119:2787–2796.

714 • NEUROGLIA
56.
NERVE FIBER REGENERATION IN THE CENTRAL
NERVOUS SYSTEM OF HIGHER VERTEBRATES
Anita D. Buchli and Martin E. Schwab

A B B R E VI AT I O N S and structural plasticity as well as functional recovery take


place.
ATP adenosine triphosphate In the past 25 years, molecules and factors responsible for
BBB blood-brain barrier the nonpermissive environment of the adult CNS and cellular
ChABC chondroitinase ABC mechanisms involved have been elucidated. In parallel, novel
CNS central nervous system therapeutic approaches are being developed to overcome the
CSPG chondroitin sulfate proteoglycan impediment of nerve fiber outgrowth leading to functional
CST corticospinal tract recovery.
DCC deleted in colorectal cancer
ECM extracellular matrix
IGF-1 insulin-like growth factor 1 2 T H E PAT H O P H YS I O L O GY O F S P I N A L
GAP-43 growth-associated protein 43 C O R D I N J U RY A N D S T R O K E
GDNF glial cell line–derived neurotrophic factor
GFAP glial fibrillary acidic protein Limited functionally relevant regeneration takes place sponta-
GPI glycosyl-phosphate-inositol neously following CNS injury. Animals that underwent experi-
HSPG heparin sulfate proteoglycan mental CNS injury or patients who suffered a stroke or spinal
HA hyaluronic acid cord injury (SCI) often show recovery of initial impairments
LAR leukocyte common antigen-related with time. The extent of recovery largely depends on the degree
LIF leukemia inhibitory factor of CNS damage; that is, after small lesions often near to full
MAG myelin-associated glycoprotein recovery can be achieved, whereas very large CNS lesions are
mTOR mammalian target of rapamycin accompanied by poor functional outcome. After large lesions
NgR Nogo-66 receptor the spontaneous recovery curve plateaus after approximately 8
OMpG oligodendrocyte-myelin glycoprotein to 12 weeks in rodents and several months in humans, leaving
PIP3 phosphatidylinositol (3,4,5)-triphosphate most functional improvements to the first months postinjury in
PI3K phosphoinositide-3-kinase humans.
PKA protein kinase A In animal models as well as in humans the final tissue dam-
PKC protein kinase C age after SCI is much larger than that of the first mechanical
PNS peripheral nervous system insult. Within the first few hours a variety of reactive processes
PTEN phosphatase and tensin homolog take place that are commonly termed secondary injury (Beattie
RGMA repulsive guidance molecule A 2004; Beattie et al. 2002a,b). In this second phase, vascular
SCI spinal cord injury changes, excitotoxic events, inflammation, and scarring further
unc-5c unc-5-homolog C contribute to the loss of tissue (Dumont et al. 2001; Schwab
wt wild-type and Bartholdi 1996). Starting hours to days after injury, plastic
changes take place at different CNS levels—from the cortex to
subcortical regions such as the brainstem and spinal cord. They
1 INTRODUCTION include biochemical, molecular, and synaptic changes, but also
gross anatomical adaptations resulting in the reorganization of
Although nerve fiber injury in the peripheral nervous sys- neuronal networks with a functionally relevant outcome.
tem (PNS) often leads to successful regeneration over long Although small lesions of the CNS can lead to strong and
distances, in the adult central nervous system (CNS) of often complete functional recovery, very large lesions have a
higher vertebrates spontaneous regeneration is almost absent. poor functional outcome. Currently available data mainly
Therefore, central nervous system trauma such as large spinal from animal experiments, but also from human patients indi-
cord or brain injuries or stroke frequently results in perma- cate that functional recovery probably depends mainly on
nent and severe deficits. During development, while circuits the amount of spared CNS tissue and plastic processes taking
are formed and fine tuned, the CNS is more adaptive to injury, place at different CNS levels.

715
Therapeutic interventions aim at increasing neuropro- 3.2 C E N T R A L N E RVO US SYS T E M MY E L I N
tection levels leading to a decrease of secondary damage. C O N TA I NS N E RVE C E L L G ROW T H
Rehabilitation and training vigorously applied in the first I N H I B ITO RY M O L EC U L E S
months after injury focus on enhancing spontaneous plastic
3.2.1 Nogo-A
adaptations which result in functional recovery after CNS
injury. Nogo-A, OMgp, and MAG are synthesized by oligodendro-
Neuroplasticity and axonal regeneration in the adult cytes and are enriched in myelin sheaths. Nogo-A was extracted
CNS are restricted by growth inhibitory molecules of myelin from a neurite growth inhibitory protein fraction of rat, bovine,
(Gonzenbach and Schwab 2008; Liu et al. 2006; Maier and and human spinal cord, respectively, and was shown to be a
Schwab 2006; Yiu and He 2006) and inhibitory factors pres- highly nonpermissive substrate in cell culture assays (Caroni
ent in the glial scar and extracellular matrix (Carulli et al. and Schwab 1988b; Spillmann et al. 1998). A monoclonal anti-
2005; Kwok et al. 2011; Liu et al. 2006; Yiu and He 2006). body raised against the rat neurite growth inhibitory fraction
Furthermore, complex processes of inflammation and second- enabled fibroblast spreading and neurite outgrowth of CNS
ary damage are being initiated after CNS injury (Barreto et al. neurons in vitro (Caroni and Schwab 1988a). In rat models of
2011; Norenberg et al. 2004). spinal cord injury/SCI and stroke application of these anti-
This chapter discusses the current state of knowledge on bodies promote regeneration of injured nerve fibers leading
the cellular and molecular processes taking place after CNS to improvements in functional recovery (Buchli et al. 2007;
injury and their influence on regeneration. Buchli and Schwab 2005; Markus et al. 2005; Papadopoulos
et al. 2002; Schnell and Schwab 1990; Seymour et al. 2005;
Tsai et al. 2007; Tsai et al. 2011; Zorner and Schwab 2010).
3 G L I A L A N D N E U R A L R E AC T I O N S TO Neutralization of Nogo-A or suppression of its downstream
C E N T R A L N E RVO U S SYS T E M DA M AG E effectors is accompanied by increased long-distance regen-
eration of lesioned axons as well as compensatory sprouting
of unlesioned fiber tracts in rodents and monkeys (Freund
3.1 T H E R EG E N E R AT I V E A B I L I T Y O F A D U LT
et al. 2006; Gonzenbach and Schwab 2008; Maier and Schwab
M A M M A L I A N N EU RO N S C A N B E A LT E R E D BY
2006; Schwab 2004; Yiu and He 2006).
T H E I R M I C RO E N V I RO N M E N T
Next to being expressed on myelinating oligodendrocytes,
First reported by Ramon y Cajal nearly a century ago (1928), Nogo-A is also found on neurons where it modulates growth
the intrinsic regenerative ability of the adult CNS and the cone motility and the dynamics of neurite formation (Montani
molecular components involved have only been unraveled et al. 2009; Petrinovic et al. 2010) both during development
in the last few decades (David and Aguayo 1981; Richardson and after injury (Cheatwood et al. 2008).
et al. 1984). Early studies by Ramon y Cajal were followed Nogo-A signaling is complex with different functions dur-
in the late 1970s by classic transplantation experiments by ing development and in the adult organism (Schwab 2010).
David and Aguayo (1981). After injury to spinal cord neurons In the mature CNS Nogo-A takes on the role of a negative
a peripheral nerve graft linking spinal cord areas rostral and regulator of neuronal growth—as described—and that of a
caudal to the injury site allowed long-distance regeneration modulator of LTP and synaptic stability (Aloy et al. 2006;
of damaged CNS axons into the peripheral nerve graft. This Delekate et al. 2011; Lee et al. 2008; Liu et al. 2003; Raiker
finding suggested that the glial environment of the periph- et al. 2010; Wang et al. 2002c), implying that it negatively
eral nerve was permissive to CNS axon regrowth and that the regulates both functional and structural plasticity in mature
regeneration failure in the CNS might result from a nonper- neuronal networks.
missive environment.
Myelin and the glial scar are currently the main inhibi- 3.2.2 MAG
tory structures described for nerve fiber regeneration in the
adult CNS. In vitro, oligodendrocytes, CNS white matter, MAG (Cao et al. 2010; Filbin 2008) is present in both
and myelin of adult rats were shown to be nonpermissive sub- CNS and PNS myelin and constitutes a member of sialic
strates for growing neurites and to induce growth cone col- acid binding proteins as well as of the immunoglobulin
lapse (Bandtlow et al. 1993; Schwab and Caroni 1988). superfamily. Early during development MAG functions
Several molecules, mainly expressed in myelin, have been as a neurite growth–promoting molecule ( Johnson et al.
described based on their ability to inhibit neurite outgrowth 1989), whereas in the mature CNS it takes on inhibitory
and induce growth cone collapse in primary neuron cultures: characteristics toward neurite outgrowth (McKerracher et al.
the myelin-associated inhibitors Nogo-A (Chen et al. 2000; 1994b; Mukhopadhyay et al. 1994). Cultures of dorsal root
GrandPre et al. 2000; Prinhja et al. 2000), myelin-associated ganglia growing on myelin of mice lacking MAG expression
glycoprotein (MAG) (McKerracher et al. 1994a), and oligo- made long extensions, whereas they barely grew on wild-
dendrocyte glycoprotein (OMgp) (Wang et al. 2002a), axon type myelin (Shen et al. 1998). However, MAG knock-out
guidance molecules ephrin B3 (Benson et al. 2005), Sema4D mice did not show major effects on axonal regeneration after
(Moreau-Fauvarque et al. 2003), Sema 5A (Goldberg et al. optic nerve or spinal cord lesion (Bartsch et al. 1995), indi-
2004; Kantor et al. 2004), netrin-1 (Low et al. 2008), and cating that MAG may not be involved in robust regeneration
tenascin-R ( Joester and Faissner 2001). after SCI.

716 • NEUROGLIA
3.2.3 OMgp Among these are growth factors, cytokines, trophic factors,
semaphorins, ephrins, Tenascin-R, Wnt, neugenin, neuronal
OMgp (Cao et al. 2010; Filbin 2008) is a GPI-linked, highly receptor protein tyrosine phosphatide sigma/Leukocyte
glycosylated membrane protein expressed in CNS myelin and common antigen-related (RPTPsigma/LAR) and chondroi-
different types of neurons. It is a potent inhibitor of neurite tin and heparan sulfate proteoglycans (CSPGs/HSPGs) on
outgrowth in vitro (Wang et al. 2002b) and inhibits axon astrocytes, which are described in the following. Through spe-
regeneration in vivo as shown in OMgp knock-out mouse cific membrane receptors some of these guidance cues eventu-
studies (Huang et al. 2005). The same OMgp-deficient mice ally converge onto the cytoskeleton, where they interact with
exhibited elevated collateral sprouting at the node of Ranvier, cytoskeletal and cytoskeletal-associated molecules within the
whereas OMgp mice with a mixed genetic background showed axon, whereas others influence protein synthesis in the soma
increased functional and anatomical regeneration after SCI (Fig. 56.1).
( Ji et al. 2008).
Nogo-A, MAG, and OMgp, although very different struc-
turally, interact with the same receptor, NgR1, leading to 3.4.1 Neurotrophin Signaling
growth cone collapse and inhibition of neurite outgrowth in
The mechanisms controlling the intrinsic axon growth abili-
vitro. However, the absence of NgR has no effect on inhibi-
ties during regeneration are still poorly understood. The
tion of neurite outgrowth in culture, and there is no improve-
tumor suppressor and tyrosine phosphatase PTEN regulates
ment in corticospinal tract regeneration in vivo, suggesting the
the activity of several molecular pathways, such as mTOR
existence of another, more specific Nogo-A receptor (Schwab
driven protein translation in the neuronal soma and cytoskel-
2010). On the other hand, axons become insensitive to MAG
etal assembly in axons, which are required for successful axon
or OMgp on cleavage of NgR and introduction of exogenous
regeneration. PTEN inactivation results in the accumulation
NgR to otherwise insensitive neurons renders them respon-
of PIP3 and the activation of AKT (Song et al. 2006) acting
sive to MAG or OMgp (Liu et al. 2002; Wang et al. 2002a).
on different downstream effectors in the soma and axon ter-
Although Nogo-A (Schwab 2010), MAG (McKerracher and
minal, which in turn control axon growth and injury-induced
Winton 2002), and OMgp (Dou et al. 2009) may be localized
axon regeneration. Neurotrophin signaling converges on the
on the surface of either oligodendrocytes or neurons, NgR and
PI3K/AKT pathway. The phenotypes of complete or condi-
a proposed unknown, Nogo-A specific receptor are expressed
tional knock-out of involved genes of this pathway indicate
on oligodendrocytes, where they assemble with different
that the pathway plays an important role for axon growth
membrane proteins to form a multimeric signaling complex.
(Eickholt et al. 2007; Zhong et al. 2007). PIP3/AKT signal-
Several signaling cascades that are not conclusively under-
ing leads to mTOR activation, the eventual outcome of which
stood seem to be involved in Nogo-A signaling (Gonzenbach
is an increase in protein synthesis, cell growth and nerve fiber
and Schwab 2008; Hannila and Filbin 2008; Joset et al. 2010;
regeneration (Guertin and Sabatini 2007; Liu et al 2010; Sun
Schwab 2010; Walmsley and Mir 2007).
et al. 2011).

3.3 O L I G O D E N D RO C Y T E I N JU RY C AUS E S C E L L 3.4.2 Insulin Growth Factor 1, Glial


D E AT H, MY E L I N DA M AG E , A N D MY E L I N L O S S Cell Line–Derived Neurotrophic Factor, and
Oligodendrocytes are particularly susceptible to toxic sub- Leukemia Inhibitory Factor
stances released to the close environment at the site of CNS In vivo growth factors do not exist in isolation and are modu-
injury. They acutely undergo both necrosis and apopto- lated by other factors, making it difficult to pin down certain
sis, with apoptosis continuing until chronic time points. biological effects to a single growth factor.
Oligodendrocyte loss leads to demyelination (see chapter 52), Insulin-like growth factor I (IGF-I) exhibits neuropro-
as well as impaired axon function and survival. In parallel, a tective effects and promotes neuronal differentiation and
rapid and prolonged oligodendrocyte progenitor cell (OPC) survival as well as myelin integrity and function (Carson
proliferative response occurs, especially at the lesion borders. et al. 1993; Russo et al. 2005). After ischemia, expression of
Proliferation of OPCs after injury is induced and controlled IGF-1 is induced in reactive microglia, oligodendrocytes,
by factors present at the site of injury, some of which are dis- astrocytes, and surviving neurons of the periinfarcted area
cussed in the following. Proliferating and migrating OPCs with other factors transporting IGF-1 from the site of synthe-
differentiate into myelinating oligodendrocytes, which remy- sis to the sites of action (Russo et al. 2005) (see also chapters
elinate demyelinated axons starting at 2 weeks post injury. 18 and 58).
Glial cell line–derived neurotrophic factor (GDNF) has
3.4 MO L ECU L A R C H A N G E S R E L EVA N T TO potent neuroprotective and neurotrophic effects on differ-
N EU R IT E G ROW T H A N D R EG E N E R AT I O N ent neuronal cell types of the CNS. GDNF neurotrophic
effects are mediated by a multisubunit receptor consisting
In addition to the neurite growth inhibitory factors present of the GPI linked GFRalpha1 and the transmembrane pro-
in myelin, proteins regulating axonal guidance during devel- tooncogene c-Ret. Neuroprotective effects of GDNF have
opment contribute to the failure of mature CNS neurons to been shown after treatment of SCI rats with recombinant
regenerate after injury (reviewed by Bashaw and Klein 2010). GDNF (Iannotti et al. 2004). GDNF also enhances axonal

N E RVE F I B E R R E G E N E R AT I O N I N T H E C E N T R A L N E RVO U S SYS T E M O F H I G H E R VE RT E B R AT E S • 717


Attractors/growth promotors:
Neurotrophic factors
ECM, cell adhesion molecules (CAM)
wnt4

Retrograde transport to the nucleus along microtubules

Glial or neuronal cell


nucleus
Local
protein
interactions

Neuronal growth machinery:


GAP-43, cytoskeletal proteins
Jak/Sat3 signalling (inhibited by SOCS3)
PI3K/AKT/mTOR signalling (inhib. by PTEN)
RhoA signalling
Repulsors/growth inhibitors:
Nogo-A, MAG, Omgp
Proteoglycans, ECM (HA, CSPGs)
Ephrins, semaphorins
Netrin
RGMA/neogenin
Wnt, wnt5a
Tenascin-R

Figure 56.1 Mechanisms Regulating Neurite Growth in the Adult Central Nervous System. Different growth inhibitory and promoting factors are present
in the ECM and expressed on neuronal and glial cells opposing the growth cone of neurons. The interaction of such guidance cues with specific receptor
components on the membrane of the growth cone eventually leads to rearrangements of the cytoskeleton or activation of gene transcription in
the nucleus.

regeneration and myelination after spinal cord injury (Zhang associated with CSPGs in the glial scar; in vitro blocking of
et al. 2009). the Sema3-CSPG interaction inhibits Sema3A repulsion, sug-
Axons of electrically active neurons release ATP, which in gesting a role in enhancing the inhibitory function of the glial
turn stimulates the production and release of leukemia inhibi- scar (Pasterkamp and Verhaagen 2006). Members of other
tory factor (LIF) from astrocytes enhancing myelination by classes of membrane-bound semaphorins have only recently
oligodendrocytes (Ishibashi et al. 2006) (see also chapters 20, been implicated in the failure of regeneration of injured CNS
25 and 45). axons. Sema4D is expressed on myelinating oligodendrocytes
after CNS injury where it inhibits axonal growth (Moreau-
Fauvarque et al. 2003) and seems to regulate the survival of
3.4.3 Semaphorins neuronal cells (Giraudon et al. 2004). Similarly, after injury
Semaphorins constitute a large family of secreted, membrane- Sema6B and Sema7A expression is upregulated in white
associated proteins the receptors, of which plexins and neuro- matter of the brain or spinal cord, respectively (Küry et al.
pilins, are found on myelinating oligodendrocytes (Bannerman 2004; Pasterkamp et al. 2007). The repulsive effect of sema-
et al. 2008; Spassky et al. 2002). Several members of class 3 phorins is mediated through interaction with plexin cell
semaphorins have been shown to act as axon guidance cues surface receptors on neurons, leading to collapse of the actin
during development. In the mature CNS the predominant cytoskeleton and retraction of outgrowing nerve fibers (Hung
neural response to semaphorins (and ephrins) is repulsive. and Terman 2011).
Their presence suggests a role in network stabilization by nar-
rowing down neuronal growth and survival. A proposed role 3.4.4 Ephrins and Eph Receptors
in remyelination builds on the findings that Sema3A expres-
sion is induced after injury in the mature rat CNS (De Winter Ephrins and their Eph receptors are membrane molecules
et al. 2002). Sema3s are also expressed by meningeal cells which—similar to semaphorins—play a role in axonal

718 • NEUROGLIA
pathfinding and target recognition during development important for neurite growth inhibition and growth cone
(Klein 2001) and some of which are constitutively expressed collapse mediated also by other inhibitory proteins such as
in the mature CNS of rodents and humans (Sobel 2005). Nogo-A, MAG, OMgp (via NgR signaling), Wnts, ephrin-A5,
EphrinAs are anchored to the membrane through a GPI tail, or Semaphorin-3A.
whereas ephrinBs are transmembrane proteins with an intra-
cellular cytosolic tail. The tyrosine kinase receptors EphAs and
3.4.7 Wnt Signaling
EphBs transmit ephrin-A and ephrin-B signaling, respectively
(Pitulescu and Adams 2010). Their role in the formation of the Wnts are axon guidance molecules particularly well described
glial scar, in neurite outgrowth inhibition and in apoptosis has for their role in developmental processes but which are also
been studied in vitro, after spinal cord, brain, or optic nerve involved in regeneration. Wnts bind to membrane bound
injury and in transgenic animals (Yaron and Sprinzak 2012). receptors that converge on downstream beta-catenin signal-
Of interest for regeneration after CNS injury is the ing, activating gene expression, and resulting in the regulation
enhanced expression of ephrinB3 in CNS myelin, the upregu- of a variety of processes.
lation of ephrinB2 in reactive astrocytes, and the increase in Three Wnts have been shown to be expressed after CNS
ephrinA5 expression around ischemic cortical lesions (Benson damage. Wnt1, Wnt5a (both repel descending CST axons
et al. 2005). Next to their role as growth inhibitory cues, Eph- during development), and the repulsive Wnt receptor Ryk are
ephrin signaling influences the formation of the glial scar expressed in spinal cord gray matter after SCI, whereas Wnt4
and apoptosis: After SCI in the rat EphA3 is upregulated in (attracts ascending sensory neurons during development) is
reactive astrocytes (Irizarry-Ramirez et al. 2005) and EphA7 induced close to the lesion site (Liu et al. 2008). After SCI,
regulates apoptosis of astrocytes (Figueroa et al. 2006). function blocking antibodies against Ryk promote axonal
Functional recovery has been described after blocking differ- sprouting beyond the lesion, implying that Wnt signaling
ent Ephrin-Eph interactions (Yaron and Sprinzak 2012). plays a role in inhibiting axon sprouting after injury.

3.4.5 Netrins 3.4.8 Hyaluronic Acid and Chondroitinsulfate


Proteoglycans
A third family of axonal guidance molecules involved in axon
regeneration are the secreted netrins. In the adult CNS, netrins Degradation of the ECM component hyaluronic acid (HA)
are expressed by neurons and myelinating oligodendrocytes induces activation and proliferation of astrocytes (Struve et al.
(Manitt et al. 2001). The response to netrin-1 is regulated by 2005). In vitro, HA decreases cell proliferation and reduces
the repertoire of netrin receptors expressed and their presenta- CSPG production (Khaing et al. 2011). The CSPGs neurocan,
tion on the cell surface as well as protein kinases PKA and PKC versican, aggrecan, and brevican can be anchored within the
within the neuron (Bouchard et al. 2004; Hong et al. 1999; ECM through binding to HA glycosaminoglycan, and treat-
Williams et al. 2003). In the visual system, netrin-1 and its ment with hyaluronidase leads to degradation of HA, result-
receptors DCC and Unc5c are expressed by nonlesioned adult ing in the release of these CSPGs from the ECM. Indeed,
retinal ganglion cells. After SCI, expression of the attraction- lesion of the rat nigrostriatal tract followed by treatment with
mediating netrin-1 receptor DCC decreases, whereas that of hyaluronidase led to partial degradation of HA and chondroi-
the repulsive receptor Unc5c returns to normal levels (Manitt tin sulfate as well as depletion of HA-binding CSPGs, which
et al. 2006). In areas surrounding the lesion site levels of in turn enhanced local sprouting at the lesion site (Moon et al.
netrin-1 remain unchanged in neurons and oligodendrocytes. 2003). This indicates that these three ECM components—HA,
Taken together, netrin-1 acts as an inhibitor of axon regenera- chondroitin sulfate, and HA-binding CSPGs—contribute to
tion in the mature CNS and may play a role in myelination. the lack of spontaneous axon regeneration after CNS injury.

3.4.6 RGMA/Neogenin 3.4.9 Tenascins


The repulsive guidance molecule A (RGMA) accumulates in Tenascins constitute a family of extracellular matrix (ECM)
the glial scar after SCI and induces growth cone collapse in glycoproteins known to regulate axonal guidance during
vitro. Under pathological conditions, RGMA accumulates development (Joester and Faissner 2001), but also involved in
at the lesion site (Schwab et al. 2005). Inhibition of RGMA myelination and cell adhesion to fibronectin. Tenascin-R is also
with a function blocking antibody after SCI in the rat model expressed in the mature CNS, where its expression is restricted
enhances growth of injured axons, promotes functional recov- mainly to oligodendrocytes and their precursors. It is secreted
ery (Hata et al. 2006), and induces plastic changes such as into the ECM and is upregulated at lesion sites after injury. In
synaptic rearrangements of spared axonal projections (Kyoto vitro it is inhibitory for retinal ganglion cell outgrowth and its
et al. 2007). This suggests that re-expression of embryonic expression in the optic nerve persists after injury (Becker et al.
repulsive cues in adult CNS contributes to the failure of axon 2000). Tenascin-R exerts its inhibitory function through bind-
regeneration in the CNS (Yamashita et al. 2007). ing to different membrane-bound molecules (Sandvig et al.
Repulsive guidance molecule A signaling converges on 2004). ChABC treatment of tenascin-R neutralizes binding to
the Rho/ROCK pathway in neurons through binding to the heparin-binding fibronectin fragment which in turn pro-
neogenin on the surface of neurons. Activation of RhoA is motes cell adhesion (Probstmeier et al. 2000) (Fig. 56.2).

N E RVE F I B E R R E G E N E R AT I O N I N T H E C E N T R A L N E RVO U S SYS T E M O F H I G H E R VE RT E B R AT E S • 719


Activated astroglia

Activated microglia
Oligodendrocytes
Differentiating OPC
Myelin debris Lesion cavity

Demyelinated axons Remyelination Sprouting axons

Figure 56.2 The Central Nervous System Environment Is Hostile for Axon Regeneration and Repair. After CNS injury nerve fibers are transected at the
site of injury and the surrounding tissue is damaged (red). During the early phase of injury, axon regneration is inhibited by myelin-associated inhibi-
tors (e.g., Nogo-A, MAG, EphA3, Sema3A) expressed on intact and damaged oligodendrocytes. Recruitment of inflammatory microglia and reactive
astrocytes to the site of lesion eventually leads to the formation of a glial scar, associated with an increased release of growth inhibitory CSPGs.
Spontaneously occurring processes supporting regeneration and repair (green) include the early neuroprotective effect of astrocytes, the astrocytic
production of antioxidants, the differentiation of OPCs into myelinating oligodendrocyes, and short-range regeneration and sprouting of axons.

3.5 A S T RO C Y T E R E S P O N S E TO I N JU RY well as the damaged axon. An attribute of the cellular response


Reactive astrocytes play a key role in neurotrauma, with to CNS injury is a rapid increase in damaging free radicals in
different roles during the course of the damage (see also both astrocytes and neurons, in response to which astrocytes
chapters 32, 51 and 57). produce beneficial antioxidants to enhance neuronal survival.
Studies with GFAP knockout mice suggest that modulat-
At an early stage they show neuroprotective functions,
ing the astrocytic response can be detrimental as well as bene-
whereas at later stages they facilitate the formation of a glial
ficial. These mice exhibit larger lesions than wt mice following
scar and inhibit CNS regeneration. Formation of a glial scar
brain injury, whereas mice lacking both GFAP and vimentin
results from the proliferation of astrocytes, oligodendrocyte
have impaired astrocyte activation and decreased glutamate
progenitor cells, meningeal fibroblasts, and pericytes migrat- uptake abilities but at the same time reveal posttraumatic
ing into the lesion (Chen et al. 2002; Göritz et al. 2011). regeneration (Wilhelmsson et al. 2004).
In response to CNS injury, reactive gliosis takes place,
which is characterized by hypertrophy of the cell bodies and
processes, altered gene expression, an increase in the expression 3.6 M I C RO G L I A A F T E R C E N T R A L
of the glial intermediate filament marker GFAP, of nestin and N E RVO US SYS T E M I N JU RY
vimentin, and proliferation bordering the wound that occurs The roles of microglia in the developing and adult CNS are
in a graded fashion in relation to the severity of the injury. covered by several chapters within this book (e.g., chapters
Glial scar formation is influenced by interleukins, trophic fac- 47–49). Here, we briefly summarize their roles in CNS trauma
tors, and cell adhesion molecules. The astrocyte-rich glial scar and regeneration.
has physical and chemical inhibitory properties: The processes Microglial cells react within minutes to traumatic tissue
of astrocytes become hypertrophied and interdigitated, form- destruction by increased motility, phagocytosis, and release of
ing a physical barrier for outgrowing neurites, and they express different factors such as pro-inflammatory and anti-inflamma-
inhibitory chondroitin sulfate proteoglycans (CSPGs). In tory cytokines, free radicals, antioxidants, and neurotrophic
addition, injury astrocytic gap junctions remain open for sub- and angiogenic factors. Depending on the microenvironment
stances such as proapoptotic factors, contributing to further and their state of activation, they can exacerbate neurodegen-
worsening of the cell injury (Giaume et al. 2010). eration or in contrast take on an important role in tissue repair
Next to enhancing glial scar formation in response to CNS and regeneration (Singh et al. 2011).
injury, astrocytes are important contributors to neuroprotec- Activated microglia can promote axon regeneration after
tion and repair (Ridet et al. 1997). The regulation of astro- CNS injury (Wake et al. 2009) and contribute to the plasticity
cytic production of growth factors and cytokines is complex of neuronal circuits. Resting microglia contact neuronal syn-
and involves the cross-talk between different types of glia as apses through their processes in a neuron activity–dependent

720 • NEUROGLIA
manner. After cerebral ischemia, these contacts are markedly translation for human patients. The main treatment strategies
prolonged and can lead to the disappearance of synapses, sug- include neurorehabilitative training—the only treatment that is
gesting that microglia monitor and react to the functional sta- routine in clinical settings today. Neurorehabilitation results in
tus of synapses (Wake et al. 2009). structural rearrangements, upregulation of growth factors, adhe-
Next to the glial–neuronal interactions the cross-talk sion molecules, and components of the synapse in spinal cord
between microglia and astrocytes seems to be pivotal for injured rats. Experimental and up-coming treatments include,
microglia activation. Intercellular astrocyte communication neuroprotective and antiinflammatory treatments, promotion
is typically mediated by gap junctions. Proinflammatory of fiber regeneration and compensatory sprouting, transplanta-
cytokines secreted by activated microglia inhibit astrocyte gap tion of bridges or stem cells to allow for growth over the lesion
junction communication influencing the role of astrocytes in and replacement of lost cell populations, and pharmacological or
providing neuronal support (Retamal et al. 2007). electrophysiological manipulation of residual networks. Several
of these treatment strategies disclose beneficial effects in animals
on the anatomical and/or functional level (Giger et al. 2010;
4 R E G E N E R AT I VE A N D C O M P E N S ATO RY Kwon et al. 2010, 2011; Noble et al. 2011; Tetzlaff et al. 2011;
F I B E R G R OW T H Zorner and Schwab 2010). Recent reviews on clinical trials in
spinal cord injury include Zorner and Schwab (2010) and Kwon
After damage or injury to the CNS, for example, after SCI or et al. (2010, 2011).
stroke, different types of recovery mechanisms can take place:
(1) regeneration of damaged axons involving elongation of the
4.1.1 Promotion of Fiber Regeneration
damaged axon end leading to reconnection with deafferentated
and Compensatory Sprouting
targets, and (2) compensatory sprouting of undamaged spared
axons to form new axonal connections with deafferentated tar- Initial molecular screens revealed the enhanced expression of
gets. The latter process is also termed compensatory plasticity. neurotrophic factors, axonal guidance molecules, and ECM
Interestingly, these two different recovery processes share similar proteins in denervated spinal cord tissue, along with enhanced
molecular mechanisms, leading to regrowth and regeneration. expression of growth-associated and cytoskeletal proteins in
Several neuronal intrinsic mechanisms are involved in neurons (Bareyre and Schwab 2003; Bareyre et al. 2002), mak-
axon growth and regeneration (Liu et al. 2011). MAPkinase, ing them key targets for regenerative treatment (Fig. 56.3).
Jak/Stat, and Akt/mTOR signaling play important roles for Inactivation of Nogo-A, NgR, CSPGs and their down-
the activation of the growth program in adult neurons (Gupta stream signaling molecules leads to long-distance regeneration
et al. 2011; Watanabe et al. 2011). Activation of these pathways of transected axons, and enhanced compensatory fiber growth
externally (e.g., by neurotrophic factors) or internally, by dele- (Carulli et al. 2005; Fournier et al. 2001; Schwab 2010). These
tion of crucial pathway inhibitors such as SOCS3 or PTEN processes are paralleled by functional recovery pointing to the
were recently shown to induce massive enhancement of fiber formation of new networks and functional reorganization
sprouting and regeneration after optic nerve or spinal cord taking place at different levels (Bradbury et al. 2002a; Li et al.
lesion (Liu et al. 2010; Sun et al. 2011). Cytoskeletal mRNAs 2004; Merkler et al. 2001). In a rat model of SCI sprouting of
and proteins, regulatory proteins such as GAP-43, tau, and axons into deafferentated areas of the spinal cord is paralleled by
MAPs, or the actin regulators cofilin/profilin, as well as cell improved functional outcome (Raineteau and Schwab 2001).
adhesion molecules are upregulated, allowing the formation Similar effects occur in monkeys subjected to high cervical
of growth cones, sprouts, and elongating axons. lesions. Anti-Nogo-A antibody treatment specifically enhances
A real gap of knowledge currently exists with regard to mech- recovery of manual dexterity, sprouting and regeneration of the
anisms of guidance and target finding for regenerating axons in corticospinal tract at the level of the spinal cord (Freund et al.
the adult CNS. The tissue composition, size, and architecture 2006, 2007). Based on these preclinical data a clinical trial apply-
are radically different from the developing CNS when these ing human anti-Nogo-A antibodies to paraplegic and tetraple-
fiber tracts originally grew. Information on detailed patterns of gic subjects is currently ongoing (http://clinicaltrials.gov/ct2/
guidance and growth factor expression in the adult, injured, or show/NCT00406016).
denervated CNS is rare. Still, in the spinal cord, regenerating Prevention of the scar formation is not yet possible but
corticospinal, red nucleus or serotonergic axons follow a rostral the inhibitory effects of CSPGs can be counteracted with
to caudal direction, and the reverse is true for ascending sensory experimental ChABC treatment. After SCI, treatment with
fibers, suggesting the presence of rostrocaudal cues. The posi- ChABC enhances axonal regeneration (Moon et al. 2001;
tion of the fibers is often abnormal, however. Detailed stud- Zuo et al. 1998), the restoration of postsynaptic activity and
ies on regulation of branch formation, target recognition, and functional recovery (Bradbury et al. 2002b).
specificity of synapse formation are lacking at present. Because of their ability to enhance the intrinsic cell
response of damaged neurons after injury, different trophic
factors have been applied after experimental SCI. Various
4.1 T R E AT M E N T S T R AT E G I E S
modes of delivery have been applied such as viral systems, and
Multiple attempts have been undertaken to enhance the lim- grafts of genetically modified cells that secrete these factors,
ited spontaneous recovery occurring after damage to the CNS pumps, or injections (Lacroix and Tuszynski 2000).
in experimental animal models and some are on their way to Application of neurotrophic factors results in neurite growth

N E RVE F I B E R R E G E N E R AT I O N I N T H E C E N T R A L N E RVO U S SYS T E M O F H I G H E R VE RT E B R AT E S • 721


A

Figure 56.3 Nerve Fiber Regeneration Can Be Enhanced After Central Nervous System Injury. After spinal cord injury in the rodent, axon regeneration
and sprouting can be encouraged around the lesion site by blocking neurite growth inhibiting ECM molecules (e.g., CSPGs), membrane-associated
proteins (e.g., Nogo-A), or downstream signaling molecules (e.g., PTEN, RhoA). A. In PTEN knock-out mice some axons grow to the distal spinal
cord regeneration e.g. 5mm caudal of the injury. B. In rats, after spinal cord lesion and ChABC treatment regenerating fibers transverse the lesioned
tissue. C. Spinal cord injured rats treated with anti-Nogo-A antibodies show regenerative sprouting (left) and fiber regeneration. D. Treatment with
C3 transferase to inactivate Rho stimulates axon regeneration after SCI. (A) From Liu et al. 2010. (B) From Bradbury et al. 2002. (C) From Liebscher
et al. 2005. (D) From Dergham et al. 2002. All are reprinted with permission.

722 • NEUROGLIA
promotion and long-distance regeneration as well as functional humans, myelin repair in the mouse was much more suc-
recovery in some models (Bregman et al. 2002; Lacroix and cessful (Lasiene et al. 2008). Oligodendrocyte replacement
Tuszynski 2000). by transplantation of precursor cells is a therapeutic option
that is currently considered for clinical trials. The use of oli-
godendrocyte-targeted mitogens or differentiation factors
4.1.2 Transplantation of Bridges or Stem Cells
to enhance myelin repair at sites of CNS trauma has not
The topic of cell-based therapies is discussed in chapter 57. been sufficiently tested experimentally so far.
A major obstacle for all cell replacement approaches—
including transplanted cells, tissues, or synthetic bridges—is
that after passing the bridge regenerating axons must re-enter 6 S U M M A RY A N D P E R S P E C T I VE S
the CNS tissue to find their targets. This process is severely
hampered by scar- and myelin-associated inhibitory factors. Much insight has been gained into the complex cross-talk
Different cell-based transplantation strategies attract and between neurons and different glial cells following CNS
support growing axons in the experimental animal model injury. Cellular, molecular, and secreted factors have been
(Rossi and Keirstead 2009). Some transplanted cells provide described, some of which influence nerve fiber sprouting,
cell replacement (e.g., for oligodendrocytes or neurons), which regeneration, and functional recovery in experimental animal
assume relay functions (Cummings et al. 2006; Hooshmand models. Treatments to counteract some of the cues that nega-
et al. 2009; Sharp et al. 2011), whereas others seem to confer tively control neurite outgrowth, to bridge the growth imped-
trophic or immunomodulatory effects. ing glial scar or boost plasticity are on their way to translation
for future therapies of spinal cord injury, brain injury, and
stroke.
4.1.3 Neuroprotective and Antiinflammatory
Treatments
Treatment with high doses of methylprednisolone, a cor- AC K N OW L E D G M E N T S
ticosteroid with antioxidant neuroprotective effects in
the animal, within the first hours after injury had modest The authors thanks the Journal of Neuroscience, Annals of
beneficial effects also in acute SCI patients (Bracken et al. Neurology, Nature, and Nature Neuroscience for their permis-
1998). Because of the small size of the effects and the sub- sion to reproduce published material in this chapter.
stantial side effects, the clinical use of methylprednisolone
therapy is being discontinued in many places at present.
Another experimental approaches for neuroprotection REFERENCES
in CNS-injured animals are the blockade of Rho GTPase
(Dubreuil et al. 2003). Rho blockade can also dampen the Aloy EM, Weinmann O, Pot C, Kasper H, Dodd DA, Rulicke T, et al.
regeneration inhibitory effects of Nogo-A and other growth 2006. Synaptic destabilization by neuronal Nogo-A. Brain Cell Biol
35:137–156.
inhibitors. A Rho-blocking drug, applied as a depot epidu- Arvanian VL, Schnell L, Lou L, Golshani R, Hunanyan A, Ghosh A,
rally on the acutely injured spinal cord, is currently under et al. 2009. Chronic spinal hemisection in rats induces a progres-
clinical investigation (http://clinicaltrials.gov/ct2/show/ sive decline in transmission in uninjured fibers to motoneurons.
NCT00610337). Exp Neurol 216:471–480.
Bandtlow CE, Schmidt MF, Hassinger TD, Schwab ME, Kater SB.
1993. Role of intracellular calcium in NI-35-evoked collapse of neu-
ronal growth cones. Science 259:80–83.
5 M Y E L I N R E PA I R A F T E R C E N T R A L Bannerman P, Ara J, Hahn A, Hong L, McCauley E, Friesen K, et al.
N E RVO U S SYS T E M I N J U RY 2008. Peripheral nerve regeneration is delayed in neuropilin
2–deficient mice. J Neurosci Res 86:3163–3169.
In the CNS, neurons and glia share a mutual dependence Bareyre FM, Haudenschild B, Schwab ME. 2002. Long-lasting sprout-
ing and gene expression changes induced by the monoclonal antibody
that becomes particularly evident in the process by which glia IN-1 in the adult spinal cord. J Neurosci 22:7097–7110.
forms myelin around axons. Signals regulating myelination are Bareyre FM, Schwab ME. 2003. Inflammation, degeneration and regen-
discussed in chapters 43–45. eration in the injured spinal cord: insights from DNA microarrays.
Oligodendrocytes are vulnerable by many influences that Trends Neurosci 26:555–563.
occur at sites of CNS injury and inflammation. Therefore, Barreto GE, Gonzalez J, Torres Y, Morales L. 2011. Astrocytic-neuronal
crosstalk: implications for neuroprotection from brain injury.
demyelination has been considered a major problem for Neurosci Res 71:107–113.
incomplete lesions in which spared axons can be nonfunc- Bartsch U, Bandtlow CE, Schnell L, Bartsch S, Spillmann AA, Rubin
tional because of local loss of myelin (Arvanian et al. 2009; BP, et al. 1995. Lack of evidence that myelin-associated glycopro-
Totoiu and Keirstead 2005). Oligodendrocyte proliferation tein is a major inhibitor of axonal regeneration in the CNS. Neuron
does happen, but the numbers of new oligodendrocytes 15:1375–1381.
Bashaw GJ, Klein R. 2010. Signaling from axon guidance receptors. Cold
around spinal cord lesion sites is modest and high num- Spring Harb Perspect Biol 2:a001941.
bers of demyelinated axons have been reported in human Beattie MS. 2004. Inflammation and apoptosis: linked therapeutic tar-
post-mortem material (Guest et al. 2005). In contrast with gets in spinal cord injury. Trends Mol Med 10:580–583.

N E RVE F I B E R R E G E N E R AT I O N I N T H E C E N T R A L N E RVO U S SYS T E M O F H I G H E R VE RT E B R AT E S • 723


Beattie MS, Harrington AW, Lee R, Kim JY, Boyce SL, Longo FM, et al. Delekate A, Zagrebelsky M, Kramer S, Schwab ME, Korte M. 2011.
2002a. ProNGF induces p74-mediated death of oligodendrocytes NogoA restricts synaptic plasticity in the adult hippocampus on a fast
following spinal cord injury. Neuron 36:375–386. time scale. Proc Natl Acad Sci U S A 108:2569–2574.
Beattie MS, Hermann GE, Rogers RC, Bresnahan JC. 2002b. Cell death Dou F, Huang L, Yu P, Zhu H, Wang X, Zou J, et al. 2009. Temporospatial
in models of spinal cord injury. Prog Brain Res 137:37–47. expression and cellular localization of oligodendrocyte myelin gly-
Becker T, Anliker B, Becker CG, Taylor J, Schachner M, Meyer RL, et al. coprotein (OMgp) after traumatic spinal cord injury in adult rats.
2000. Tenascin-R inhibits regrowth of optic fibers in vitro and per- J Neurotrauma 26:2299–2311.
sists in the optic nerve of mice after injury. Glia 29:330–346. Dubreuil CI, Winton MJ, McKerracher L. 2003. Rho activation patterns
Benson MD, Romero MI, Lush ME, Lu QR, Henkemeyer M, Parada LF. after spinal cord injury and the role of activated Rho in apoptosis in
2005. Ephrin-B3 is a myelin-based inhibitor of neurite outgrowth. the central nervous system. J Cell Biol 162:233–243.
Proc Natl Acad Sci U S A 102:10694–10699. Dumont RJ, Okonkwo DO, Verma S, Hurlbert RJ, Boulos PT, Ellegala
Bouchard J-F, Moore SW, Tritsch NX, Roux PP, Shekarabi M, Barker DB, et al. 2001. Acute spinal cord injury. Part I: Pathophysiologic
PA, et al. 2004. Protein kinase A activation promotes plasma mem- mechanisms. Clin Neuropharmacol 24:254–264.
brane insertion of dcc from an intracellular pool: a novel mechanism Eickholt BJ, Ahmed AI, Davies M, Papakonstanti EA, Pearce W, Starkey
regulating commissural axon extension. J Neurosci 24:3040–3050. ML, et al. 2007. Control of axonal growth and regeneration of
Bracken MS, Shepard MJ, Holford TR, Leo-Summers L, Aldrich EF, sensory neurons by the p110δ PI 3-kinase. PLoS ONE 2:e869.
Fazl M, et al. 1998. Methylprednisolone or tirilazad mesylate admin- Figueroa JD, Benton RL, Velazquez I, Torrado AI, Ortiz CM, Hernandez
istration after acute spinal cord injury: 1-year follow up. Results of CM, et al. 2006. Inhibition of EphA7 up-regulation after spinal cord
the third National Acute Spinal Cord Injured randomized controlled injury reduces apoptosis and promotes locomotor recovery. J Neurosci
trial. J Neurosurg 89:699–706. Res 84:1438–1451.
Bradbury EJ, Moon LD, Popat RJ, King VR, Bennett GS, Patel PN, et al. Filbin MT. 2008. PirB, a second receptor for the myelin inhibitors of
2002a. Chondroitinase ABC promotes functional recovery after spi- axonal regeneration Nogo66, MAG, and OMgp: implications for
nal cord injury. Nature 416:636–640. regeneration in vivo. Neuron 60:740–742.
Bregman BS, Coumans JV, Dai HN, Kuhn PL, Lynskey J, McAtee M, Fournier AE, GrandPre T, Strittmatter SM. 2001. Identification of
et al. 2002. Transplants and neurotrophic factors increase regenera- a receptor mediating Nogo-66 inhibition of axonal regeneration.
tion and recovery of function after spinal cord injury. Prog Brain Res Nature 409:341–346.
137:257–273. Freund P, Schmidlin E, Wannier T, Bloch J, Mir A, Schwab ME, et al.
Buchli AD, Rouiller E, Mueller R, Dietz V, Schwab ME. 2007. Repair of 2006. Nogo-A-specific antibody treatment enhances sprouting and
the injured spinal cord. A joint approach of basic and clinical research. functional recovery after cervical lesion in adult primates. Nat Med
Neurodegener Dis 4:51–56. 12:790–792.
Buchli AD, Schwab ME. 2005. Inhibition of Nogo: a key strategy Freund P, Wannier T, Schmidlin E, Bloch J, Mir A, Schwab ME, et al.
to increase regeneration, plasticity and functional recovery of the 2007. Anti-Nogo-A antibody treatment enhances sprouting of cor-
lesioned central nervous system. Ann Med 37:556–567. ticospinal axons rostral to a unilateral cervical spinal cord lesion in
Cao Z, Gao Y, Deng K, Williams G, Doherty P, Walsh FS. 2010. adult macaque monkey. J Comp Neurol 502:644–659.
Receptors for myelin inhibitors: Structures and therapeutic opportu- Giaume C, Koulakoff A, Roux L, Holcman D, Rouach N. 2010.
nities. Mol Cell Neurosci 43:1–14. Astroglial networks: a step further in neuroglial and gliovascular
Caroni P, Schwab ME. 1988a. Antibody against myelin-associated inhib- interactions. Nat Rev Neurosci 11:87–99.
itor of neurite growth neutralizes nonpermissive substrate properties Giger RJ, Hollis ER 2nd, Tuszynski MH. 2010. Guidance molecules in
of CNS white matter. Neuron 1:85–96. axon regeneration. Cold Spring Harb Perspect Biol 2:a001867.
Caroni P, Schwab ME. 1988b. Two membrane protein fractions from Giraudon P, Vincent P, Vuaillat C, Verlaeten O, Cartier L, Marie-Cardine
rat central myelin with inhibitory properties for neurite growth and A, et al. 2004. Semaphorin CD100 from activated t lymphocytes
fibroblast spreading. J Cell Biol 106:1281–1288. induces process extension collapse in oligodendrocytes and death of
Carson MJ, Behringer RR, Brinster RL, McMorris FA. 1993. Insulin-like immature neural cells. J Immunol 172:1246–1255.
growth factor I increases brain growth and central nervous system Goldberg JL, Vargas ME, Wang JT, Mandemakers W, Oster SF, Sretavan
myelination in transgenic mice. Neuron 10:729–740. DW, et al. 2004. An oligodendorcyte lineage-specific semaphorin,
Carulli D, Laabs T, Geller HM, Fawcett JW. 2005. Chondroitin Sema5A, inhibits axon growth by retinal ganglion cells. J Neurosci
sulfate proteoglycans in neural development and regeneration. 24:4989–4999.
CurrOpinNeurobiol 15 116–120. Gonzenbach RR, Schwab ME. 2008. Disinhibition of neurite growth to
Cheatwood JL, Emerick AJ, Schwab ME, Kartje GL. 2008. Nogo-A repair the injured adult CNS: focusing on Nogo. Cell Mol Life Sci
expression after focal ischemic stroke in the adult rat. Stroke 65:161–176.
39:2091–2098. Göritz C, Dias DO, Tomilin N, Barbacid M, Shupliakov O, Frisén J. 2011.
Chen MS, Huber AB, van der Haar ME, Frank M, Schnell L, Spillmann A pericyte origin of spinal cord scar tissue. Science 333:238–242.
AA, et al. 2000. Nogo-A is a myelin-associated neurite outgrowth GrandPre T, Nakamura F, Vartanian T, Strittmatter SM. 2000.
inhibitor and an antigen form monoclonal antibody IN-1. Nature Identification of the Nogo inhibitor of axon regeneration as a reticu-
403:434–439. lon protein. Nature 403:439–444.
Chen ZJ, Ughrin Y, Levine JM. 2002. Inhibition of axon growth by oli- Guertin DA, Sabatini DM. 2007. Defining the role of mTOR in cancer.
godendrocyte precursor cells. Molec Cell Neurosci 20:125–139. Cancer Cell 12:9–22.
Cummings BJ, Uchida N, Tamaki SJ, Anderson AJ. 2006. Human neu- Guest JD, Hiester ED, Bunge RP. 2005. Demyelination and Schwann
ral stem cell differentiation following transplantation into spinal cell responses adjacent to injury epicenter cavities following chronic
cord injured mice: association with recovery of locomotor function. human spinal cord injury. Exp Neurol 192:384–393.
Neurol Res 28:474–481. Gupta S, Mishra K, Surolia A, Banerjee K. 2011. Suppressor of cytokine
David S, Aguayo AJ. 1981. Axonal elongation into peripheral nervous signalling-6 promotes neurite outgrowth via JAK2/STAT5-mediated
system “bridges” after central nervous system injury in adult rats. signalling pathway, involving negative feedback inhibition. PLoS
Science 214:931–933. ONE 6:e26674.
De Winter F, Oudega M, Lankhorst AJ, Hamers FP, Blits B, Ruitenberg Hannila SS, Filbin MT. 2008. The role of cyclic AMP signaling in pro-
MJ, et al. 2002. Injury-induced class 3 semaphorin expression in the moting axonal regeneration after spinal cord injury. Exp Neurol
rat spinal cord. Exp Neurol 175:61–75. 209:321–332.

724 • NEUROGLIA
Hata K, Fujitani M, Yasuda Y, Doya H, Saito T, Yamagishi S, et al. 2006. Lasiene J, Shupe L, Perlmutter S, Horner P. 2008. No evidence for
RGMa inhibiton promotes axonal growth and recovery after spinal chronic demyelination in spared axons after spinal cord injury in a
cord injury. JCell Biol 173 47–58. mouse. J Neurosci 28:3887–3896.
Hong K, Hinck L, Nishiyama M, Poo MM, Tessier-Lavigne M, Stein E. Lee H, Raiker SJ, Venkatesh K, Geary R, Robak LA, Zhang Y, et al.
1999. A ligand-grated association between cytoplasmic domains of 2008. Synaptic function for the Nogo-66 receptor NgR1: regula-
UNC5 and DCC family receptors converts netrin-induced growth tion of dendritic spine morphology and activity-dependent synaptic
cone attraction to repulsion. Cell 97:927–941. strength. J Neurosci 28:2753–2765.
Hooshmand MJ, Sontag CJ, Uchida N, Tamaki S, Anderson AJ, Li W, Walus L, Rabacchi SA, Jirik A, Chang E, Schauer J, et al. 2004.
Cummings BJ. 2009. Analysis of host-mediated repair mechanisms A neutralizing anti-Nogo66 receptor monoclonal antibody reverses
after human CNS-stem cell transplantation for spinal cord injury: inhibition of neurite outgrowth by central nervous system myelin.
correlation of engraftment with recovery. PLoS ONE 4:e5871. J Biol Chem 279:43780–43788.
Huang JK, Phillips GR, Roth AD, Pedraza L, Shan W, Belkaid W, et al. Liu BP, Cafferty WB, Budel SO, Strittmatter SM. 2006. Extracellular
2005. Glial membranes at the node of Ranvier prevent neurite out- regulators of axonal growth in the adult central nervous system.
growth. Science 310 1813–1817. Philos Trans R Soc Lond B Biol Sci 361:1593–1610.
Hung R-J, Terman JR. 2011. Extracellular inhibitors, repellents, and Liu BP, Fournier A, Grand Pr T, Strittmatter SM. 2002. Myelin-associated
semaphorin/plexin/MICAL-mediated actin fi lament disassembly. glycoprotein as a functional ligand for the Nogo-66 receptor. Science
Cytoskeleton 68:415–433. 297:1190–1193.
Iannotti C, Ping Zhang Y, Shields CB, Han Y, Burke DA, Xu X-M. Liu K, Lu Y, Lee JK, Samara R, Willenberg R, Sears-Kraxberger I, et al.
2004. A neuroprotective role of glial cell line-derived neurotrophic 2010. PTEN deletion enhances the regenerative ability of adult corti-
factor following moderate spinal cord contusion injury. Exp Neurol cospinal neurons. Nat Neurosci 13:1075–1081.
189:317–332. Liu K, Tedeschi A, Park KK, He Z. 2011. Neuronal intrinsic mechanisms
Irizarry-Ramirez M, Villwon CA, Cruz-Orengo L, Figueroa J, Velasquez of axon regeneration. Annu Rev Neurosci 34:131–152.
I, Jones H, et al. 2005. Upregulation of EphA3 rerceptor after spinal Liu Y, Wang X, Lu CC, Kerman R, Steward O, Xu XM, et al. 2008.
cord injury. JNeurotrauma 22 929–935. Repulsive Wnt signaling inhibits axon regeneration after CNS injury.
Ishibashi T, Dakin KA, Stevens B, Lee RR, Kozlov SV, Stewart CL, J Neurosci 28:8376–8382.
et al. 2006. Astrocytes promote myelination in response to electrical Liu YY, Jin WL, Liu HL, Ju G. 2003. Electron microscopic localization
impulses. Neuron 49 823–832. of Nogo-A at the postsynaptic active zone of the rat. Neurosci Lett
Ji B, Case LC, Liu K, Shao Z, Lee X, Yang Z, et al. 2008. Assessment of 346:153–156.
functional recovery and axonal sprouting in oligodendrocyte-myelin Low K, Culbertson M, Bradke F, Tessier-Lavigne M, Tuszynski MH.
glycoprotein (OMgp) null mice after spinal cord injury. Mol Cell 2008. Netrin-1 is a novel myelin-associated inhibitor to axon growth.
Neurosci 39:258–267. J Neurosci 28:1099–1108.
Joester A, Faissner A. 2001. The structure and function of tenascins in Maier IC, Schwab ME. 2006. Sprouting, regeneration and circuit forma-
the nervous system. Matrix Biol 20:13–22. tion in the injured spinal cord: factors and activity. Philos Trans R
Johnson PW, Abramow-Newerly W, Seilheimer B, Sadoul R, Tropak MB, Soc Lond B Biol Sci 361:1611–1634.
Arquint M, et al. 1989. Recombinant myelin-associated glycoprotein Manitt C, Colicos MA, Thompson KM, Rousselle E, Peterson AC,
confers neural adhesion and neurite outgrowth function. Neuron Kennedy TE. 2001. Widespread expression of Netrin-1 by neurons
3:377–385. and oligodendrocytes in the adult mammalian spinal cord. JNeurosci
Joset A, Dodd DA, Halegoua S, Schwab ME. 2010. Pincher-generated 21:3911–3922.
Nogo-A endosomes mediate growth cone collapse and retrograde sig- Manitt C, Wang D, Kennedy TE, Howland DR. 2006. Positioned to
naling. J Cell Biol 188:271–285. inhibit: Netrin-1 and netrin receptor expression after spinal cord
Kantor DB, Chivatakarn O, Peer KL, Oster SF, Inatani M, Hansen injury. J Neurosci Res 84:1808–1820.
MJ, et al. 2004. Semaphorin 5A is a bifunctional axon guidance cue Markus TM, Tsai SY, Bollnow MR, Farrer RG, O’Brien TE,
regulated by heparan and chondroitin sulface proteoglycans. Neuron Kindler-Baumann DR, et al. 2005. Recovery and brain reorganiza-
44:961–975. tion after stroke in adult and aged rats. Ann Neurol 58:950–953.
Khaing ZZ, Milman BD, Vanscoy JE, Seidlits SK, Grill RJ, Schmidt CE. McKerracher L, David S, Jackson DL, Kottis V, Dunn RJ, Braun PE.
2011. High molecular weight hyaluronic acid limits astrocyte acti- 1994a. Identification of myelin-associated glycoprotein as a major
vation and scar formation after spinal cord injury. J Neural Eng myelin-derived inhibitor of neurite growth. Neuron 13:805–811.
8:046033. McKerracher L, Winton MJ. 2002. Nogo on the go. Neuron 36:345–348.
Klein R. 2001. Excitatory Eph receptors and adhesive ephrin ligands. Merkler D, Metz GAS, Raineteau O, Dietz V, Schwab ME, Fouad K.
Curr Opin Cell Biol 13:196–203. 2001. Locomotor recovery in spinal cord-injured rats treated with an
Küry P, Abankwa D, Kruse F, Greiner-Petter R, Müller HW. 2004. antibody neutralizing the myelin-associated neurite growth inhibitor
Gene expression profi ling reveals multiple novel intrinsic and extrin- Nogo-A. J Neurosci 21:3665–3673.
sic factors associated with axonal regeneration failure. Eur J Neurosci Michal S. 2004. Optic nerve crush: protection and regeneration. Brain
19:32–42. Res Bull 62:467–471.
Kwok JC, Dick G, Wang D, Fawcett JW. 2011. Extracellular matrix and Montani L, Gerrits B, Gehrig P, Kempf A, Dimou L, Wollscheid B,
perineuronal nets in CNS repair. Dev Neurobiol 71:1073–1089. et al. 2009. Neuronal Nogo-A modulates growth cone motility via
Kwon BK, Okon E, Hillyer J, Mann C, Baptiste D, Weaver LC, et al. Rho-GTP/LIMK1/cofi lin in the unlesioned adult nervous system.
2011. A systematic review of non-invasive pharmacologic neuro- J Biol Chem 284:10793–10807.
protective treatments for acute spinal cord injury. J Neurotrauma Moon LD, Asher RA, Rhodes KE, Fawcett JW. 2001. Regeneration of
28:1545–1588. CNS axons back to their target following treatment of adult rat brain
Kwon BK, Okon EB, Plunet W, Baptiste D, Fouad K, Hillyer J, et al. with chondroitinase ABC. Nature Neurosci 4:465–466.
2010. A systematic review of directly applied biologic therapies for Moon LDF, Asher RA, Fawcett JW. 2003. Limited growth of severed
acute spinal cord injury. Journal of Neurotrauma 28:1589–1610. CNS axons after treatment of adult rat brain with hyaluronidase.
Kyoto A, Hata K, Yamashita T. 2007. Synapse formation of the J Neurosci Res 71:23–37.
cortico-spinal axons is enhanced by RGMa inhibition after spinal Moreau-Fauvarque C, Kumanogoh A, Camand E, Jaillard C, Barbin
cord injury. Brain Res 1186:74–86. G, Boquet I, et al. 2003. The transmembrane semaphorin Sema4D/
Lacroix S, Tuszynski MH. 2000. Neurotrophic factors and gene therapy CD100, an inhibitor of axonal growth, is expressed on oligodendro-
in spinal cord injury. Neurorehabil Neural Repair 13:265–275. cytes and upregulated after CNS lesion. JNeurosci 23:9229–9239.

N E RVE F I B E R R E G E N E R AT I O N I N T H E C E N T R A L N E RVO U S SYS T E M O F H I G H E R VE RT E B R AT E S • 725


Mukhopadhyay G, Doherty P, Walsh FS, Crocker PR, Filbin MT. 1994. Schwab ME, Caroni P. 1988. Oligodendrocytes and CNS myelin are
A novel role for myelin-associated glycoprotein as an inhibitor of nonpermissive substrates for neurite growth and fibroblast spreading
axonal regeneration. Neuron 13:757–767. in vitro. J Neurosci 8:2381–2393.
Noble M, Mayer-Pröschel M, Davies JE, Davies SJA, Pröschel C. 2011. Seymour AB, Andrews EM, Tsai SY, Markus TM, Bollnow MR,
Cell therapies for the central nervous system: how do we identify Brenneman MM, et al. 2005. Delayed treatment with monoclonal
the best candidates? Current Opinion in Neurology 24:570–576 antibody IN-1 1 week after stroke results in recovery of function
10.1097/WCO.0b013e32834cd4c9. and corticorubral plasticity in adult rats. J Cereb Blood Flow Metab
Norenberg MD, Smith J, Marcillo A. 2004. The pathology of human 25:1366–1375.
spinal cord injury: defining the problems. JNeurotrauma 21: Sharp J, Hatch M, Nistor G, Keirstead H. 2011. Derivation of oligoden-
429–440. drocyte progenitor cells from human embryonic stem cells. Methods
Papadopoulos CM, Tsai SY, Alsbiei T, O’Brien TE, Schwab ME, Kartje Mol Biol 767:399–409.
GL. 2002. Functional recovery and neuroanatomical plasticity fol- Shen YJ, DeBellard ME, Salzer JL, Roder J, Filbin MT. 1998.
lowing middle cerebral artery occlusion and IN-1 antibody treatment Myelin-associated glycoprotein in myelin and expressed by Schwann
in the adult rat. Ann Neurol 51:433–441. cells inhibits axonal regeneration and branching. Mol Cell Neurosci
Pasterkamp RJ, Kolk S, Hellemons A, Kolodkin A. 2007. Expression 12:79–91.
patterns of semaphorin7A and plexinC1 during rat neural develop- Singh S, Swarnkar S, Goswami P, Nath C. 2011. Astrocytes and micro-
ment suggest roles in axon guidance and neuronal migration. BMC glia: responses to neuropathological conditions. Int J Neurosci
Dev Biol 7:98. 121:589–597.
Pasterkamp RJ, Verhaagen J. 2006. Semaphorins in axon regeneration: Sobel RA. 2005. Ephrin A receptors and ligands in lesions and
developmental guidance molecules gone wrong? Philos Trans R Soc normal-appearing white matter in multiple sclerosis. Brain Pathol
Lond B Biol Sci 361:1499–1511. 15:35–45.
Petrinovic MM, Duncan CS, Bourikas D, Weinman O, Montani L, Song K, Wang H, Krebs TL, Danielpour D. 2006. Novel roles of Akt
Schroeter A, et al. 2010. Neuronal Nogo-A regulates neurite fascicu- and mTOR in suppressing TGF-[beta]/ALK5-mediated Smad3 acti-
lation, branching and extension in the developing nervous system. vation. EMBO J 25:58–69.
Development 137:2539–2550. SpasskyN,deCastroF,LeBrasB,HeydonK,Quéraud-LeSauxF,Bloch-Gallego
Pitulescu ME, Adams RH. 2010. Eph/ephrin molecules—a hub for sig- E, et al. 2002. Directional guidance of oligodendroglial migration by
naling and endocytosis. Genes Dev 24:2480–2492. class 3 semaphorins and netrin-1. J Neurosci 22:5992–6004.
Prinhja R, Moore SE, Vinson M, Blake S, Morrow R, Christie G, Spillmann AA, Bandtlow CE, Lottspeich F, Keller F, Schwab ME. 1998.
et al. 2000. Inhibitor of neurite outgrowth in humans. Nature Identification and characterization of a bovine neurite growth inhibi-
403:383–384. tor (bNI-220). J Biol Chem 273:19283–19293.
Probstmeier R, Braunewell K-H, Pesheva P. 2000. Involvement of chon- Struve J, Maher PC, Li Y-q, Kinney S, Fehlings MG, Kuntz C, et al.
droitin sulfates on brain-derived tenascin-R in carbohydrate-dependent 2005. Disruption of the hyaluronan-based extracellular matrix in
interactions with fibronectin and tenascin-C. Brain Res 863:42–51. spinal cord promotes astrocyte proliferation. Glia 52:16–24.
Raiker SJ, Lee H, Baldwin KT, Duan Y, Shrager P, Giger RJ. 2010. Sun F, Park KK, Belin S, Wang D, Lu T, Chen G, et al. 2011. Sustained
Oligodendrocyte-myelin glycoprotein and Nogo negatively regulate axon regeneration induced by co-deletion of PTEN and SOCS3.
activity-dependent synaptic plasticity. J Neurosci 30:12432–12445. Nature 480:372–375.
Raineteau O, Schwab ME. 2001. Plasticity of motor systems after incom- Tetzlaff W, Okon EB, Karimi-Abdolrezaee S, Hill CE, Sparling JS,
plete spinal cord injury. Nat Rev Neurosci 2:263–274. Plemel JR, et al. 2011. A systematic review of cellular transplantation
Retamal MA, Froger N, Palacios-Prado N, Ezan P, Sáez PJ, Sáez JC, et al. therapies for spinal cord injury. J Neurotrauma 28:1611–1682.
2007. Cx43 Hemichannels and gap junction channels in astrocytes Totoiu MO, Keirstead HS. 2005. Spinal cord injury is accompanied by
are regulated oppositely by proinflammatory cytokines released from chronic progressive demyelination. J CompNeurol 486 373–383.
activated microglia. J Neurosci 27:13781–13792. Tsai SY, Markus TM, Andrews EM, Cheatwood JL, Emerick AJ, Mir
Richardson PM, Issa VMK, Aguayo AJ. 1984. Regeneration of long spi- AK, et al. 2007. Intrathecal treatment with anti-Nogo-A antibody
nal axons in the rat. J Neurocytol 13:165–182. improves functional recovery in adult rats after stroke. Exp Brain Res
Ridet JL, Malhotra SK, Privat A, Gage FH. 1997. Reactive astrocytes: 182:261–266.
cellular and molecular cues to biological function. Trends Neurosci Tsai SY, Papadopoulos CM, Schwab ME, Kartje GL. 2011. Delayed
20:570–577. anti-nogo-a therapy improves function after chronic stroke in adult
Rossi SL, Keirstead HS. 2009. Stem cells and spinal cord regeneration. rats. Stroke 42:186–190.
Curr Opin Biotechnol 20:552–562. Wake H, Moorhouse AJ, Jinno S, Kohsaka S, Nabekura J. 2009. Resting
Russo VC, Gluckman PD, Feldman EL, Werther GA. 2005. the microglia directly monitor the functional state of synapses in vivo and
insulin-like growth factor system and its pleiotropic functions in determine the fate of ischemic terminals. J Neurosci 29:3974–3980.
brain. Endocr Rev 26:916–943. Walmsley AR, Mir AK. 2007. Targeting the Nogo-A signalling pathway
Sandvig A, Berry M, Barrett LB, Butt A, Logan A. 2004. Myelin-, reac- to promote recovery following acute CNS injury. Curr Pharm Des
tive glia-, and scar-derived CNS axon growth inhibitors: expression, 13:2470–2484.
receptor signaling, and correlation with axon regeneration. Glia Wang KC, Koprivica V, Kim JA, Sivasankaran R, Guo Y, Neve RL, et al.
46:225–251. 2002a. Oligodendrocyte-myelin glycoprotein is a Nogo receptor
Schnell L, Schwab ME. 1990. Axonal regeneration in the rat spinal cord ligand that inhibits neurite outgrowth. Nature 16:1–4.
produced by an antibody against myelin-associated neurite growth Wang KC, Koprivica V, Kim JA, Sivasankaran R, Guo Y, Neve RL, et al.
inhibitors. Nature 343:269–272. 2002b. Oligodendrocyte-myelin glycoprotein is a Nogo receptor
Schwab JM, Conrad S, Monnier PP, Julien S, Mueller BK, Schluesener HJ. ligand that inhibits neurite outgrowth. Nature 417:941–944.
2005. Spinal cord injury-induced lesional expression of the repulsive Wang X, Chun SJ, Treloar H, Vartanian T, Greer CA, Strittmatter SM.
guidance molecule (RGM). EuropJNeurosci 21:1569–1576. 2002c. Localization of Nogo-A and Nogo-66 receptor proteins at
Schwab ME. 2004. Nogo and axon regeneration. Curr Opin Neurobiol sites of axon-myelin and synaptic contact. J Neurosci 22:5505–5515.
14:118–124. Watanabe H, Hongu T, Yamazaki M, Kanaho Y. 2011. Phospholipase
Schwab ME. 2010. Functions of Nogo proteins and their receptors in the D2 activation by p38 MAP kinase is involved in neurite outgrowth.
nervous system. Nat Rev Neurosci 11:799–811. Biochem Biophys Res Commun 413:288–293.
Schwab ME, Bartholdi D. 1996. Degeneration and regeneration of axons Wilhelmsson U, Li L, Pekna M, Verthold CH, Blom S, Eliasson C, et al.
in the lesioned spinal cord. Physiol Rev 76:319–370. 2004. Absence of glial fibrillary acidic protein and vimentin prevents

726 • NEUROGLIA
hypertrophy of astrocytic processes and improves post-traumatic Zhang H, Mei X, Zhang P, Ma C, White FA, Donnelly DF, et al. 2009.
regeneration. J Neurosci 24:5016–5021. Altered functional properties of satellite glial cells in compressed spi-
Williams EJ, Walsh FS, Doherty P. 2003. The FGF receptor uses the nal ganglia. Glia 57:1588–1599.
endocannabinoid signaling system to couple to an axonal growth Zhong J, Li X, McNamee C, Chen AP, Baccarini M, Snider WD. 2007.
response. J Cell Biol 160:481–486. Raf kinase signaling functions in sensory neuron differentiation and
Yamashita T, Mueller BK, Hata K. 2007. Neogenin and repulsive guid- axon growth in vivo. Nat Neurosci 10:598–607.
ance molecule signaling in the central nervous system. Curr Opin Zorner B, Schwab ME. 2010. Anti-Nogo on the go: from animal models
Neurobiol 17:29–34. to a clinical trial. Ann N Y Acad Sci 1198(Suppl 1):E22–34.
Yaron A, Sprinzak D. 2012. The cis side of juxtacrine signaling: a new role in Zuo J, Ferguson TA, Hernandez YJ, Stetler-Stevenson WG, Muir D.
the development of the nervous system. Trends Neurosci 35:230–239. 1998. Neuronal matrix metalloproteinase-2 degrades and inactivates
Yiu G, He Z. 2006. Glial inhibition of CNS axon regeneration. Nat Rev a neurite-inhibiting chondroitin sulfate proteoglycan. J Neurosci
Neurosci 7:617–627. 18:5203–5211.

N E RVE F I B E R R E G E N E R AT I O N I N T H E C E N T R A L N E RVO U S SYS T E M O F H I G H E R VE RT E B R AT E S • 727


57.
GLIAL CELL TRANSPLANTATION FOR CENTRAL
NERVOUS SYSTEM REPAIR
Anne Baron-Van Evercooren and Rebecca Matsas

A B B R E VI AT I O N S entered the segments of a peripheral nerve implanted into the


lesioned brain, and were myelinated by Schwann cells (SCs), the
BC boundary cap peripheral myelin-forming cells (Cajal 1928; Tello 1911). More
BMSC bone marrow mesenchymal cell than half a century later, William Blakemore demonstrated that
cAMP cyclic adenosine monophosphate SC remyelination occurred following placement of peripheral
CNS central nervous system nerve fragments over the areas of demyelination in the spinal
EAE experimental autoimmune encephalomyelitis cord (Blakemore 1977). Soon afterward Richardson et al. (1980)
ES embryonic stem cell published a landmark study that greatly influenced subsequent
FGF fibroblast growth factor research, showing that it was possible to bridge a complete
iPS induced pluripotent stem cell transection gap in the spinal cord with a peripheral nerve
LPC lysophosphatidylcholine implant. It was obvious that if the environment is permissive,
MBP myelin basic protein axons from central neurons are able to respond and regrow, and
MLD metachromatic leukodystrophy peripheral type myelination of CNS axons soon was described
MSC mesenchymal stem cell following engraftment of purified SCs (Duncan et al. 1981).
NCC Neural crest cell Madeleine Gumpel et al. then demonstrated for the first time
NC-SC Neural crest-derived stem cell that CNS fragment implantation could also lead to CNS-type
PS-NCAM poly-sialated-neural cell adhesion molecule myelination in myelin mutants (Lachapelle et al. 1983). Further
NPC neural stem/precursor cell experiments showed that graft-mediated oligodendrocyte
OEC olfactory ensheathing cell remyelination could be achieved in situations in which
OEp Olfactory ensheathing cell progenitor endogenous myelination/remyelination fails (Blakemore and
OPC oligodendrocyte progenitor cell Crang 1988). In the 1990s, Franklin et al. (1996) demonstrated
PDGF platelet-derived growth factor that transplantation of olfactory ensheathing cells (OECs)
PNS peripheral nervous system could also lead to robust remyelination of demyelinated CNS
PSA polysialic acid axons. Shortly afterward, stem cells derived from both neural
SC Schwann cell tissues (Hammang et al. 1997) and embryonic tissues (Brustle
SCI spinal cord injury et al. 1999) were identified as a promising source of myelin
SCP Schwann cell precursors following transplantation into myelin-deficient animals,
opening the field of cell therapy approaches based on ES- and
iPS-derived myelin forming cells.
1 INTRODUCTION
The complex changes occurring in spinal cord tissue
following injury have led to the idea of using combinatorial
This chapter provides an overview of the current knowledge
strategies. To repair the injured spinal cord it is essential to
relating to the use of transplanted glial cells and stem cells in the
limit the spread of secondary tissue damage, provide trophic
treatment of myelin diseases and spinal trauma. The chapter covers
support for neuroprotection, manipulate inflammation and
anatomical repair, including neuroregeneration and myelin repair,
scar formation, neutralize inhibitory molecules, promote
as well as bystander effects mediated by activation of endogenous
axonal growth and remyelination, and restore damaged
glia (oligodendrocyte progenitors, astrocytes) and modulation of
connectivity. Such combinatorial interventions include cellular
neuroinflammation.
transplants coupled with administration of neuroprotective
agents and growth factors, guidance channels to support
2 H I S TO RY O F C E L L T R A N S P L A N TAT I O N regrowing axons, addition of the enzyme chondroitinase to
F O R C E N T R A L N E RVO U S SYS T E M reduce inhibitory molecules or elevation of cyclic adenosine
M Y E L I N R E G E N E R AT I O N monophosphate (cAMP) to enhance axonal regeneration and
remyelination (Fouad et al. 2005; Pearse et al. 2004; Xu et al.
Santiago Ramon y Cajal and Francisco Tello were the first 1995). Although much of the work reported here is driven by
to indicate that central nervous system (CNS) axons can be exploration of the therapeutic potential of transplanted cells,
myelinated by exogenous cells. They observed that CNS axons these studies also stress the power of cell transplantation to
728
unravel the biology of glial cells during developmental and properties (Nashmi and Fehlings 2001). Because differentiation
pathological conditions. of endogenous oligodendrocyte progenitors (OPC) into oligo-
dendrocytes following SCI is not sufficient to achieve remyelina-
tion, myelinogenic cell engraftment has been proposed as a means
3 TA R G ET D I S E A S E S to promote axonal regeneration, restore myelination, and protect
denuded axons against subsequent degeneration. Engineering the
Transplantation of myelin-forming cells is considered as a transplanted cells to deliver growth factors associated with neural
potential treatment for leukodystrophies, multiple sclerosis, protection and regeneration to the lesion site is also attempted, so
and spinal cord injury. as to optimize the functional benefits.
The wide family of leukodystrophies are congenital myelin dis-
orders characterized by genetic abnormalities resulting in a failure
of myelin sheath formation and/or maintenance (see chapter 62). 4 ANIMAL MODELS
The mutations target genes encoding either proteins or enzymes
involved directly or indirectly in the development or maturation Animal models of myelin diseases in which either myelin is
of oligodendrocytes. They can lead to hypomyelination (lack the primary target or demyelination is secondary to axon dam-
of myelin), dysmyelination (abnormal myelin), or demyelina- age are distinguished here.
tion (myelin breakdown) and are characterized by severe motor,
sensory, and cognitive neurological deficits. One could there-
4.1 A N I M A L M O D E L S O F MY E L I N D I S E A S E S
fore speculate that in some of these diseases such as Pelizaeus
Merzbacher disease (PMD) introduction of genetically normal Two groups of models are used to study remyelination by grafted
myelinating cells could correct the genetic deficit in myelina- cells. These are models of hypomyelination/dysmyelination, and
tion. Transplanted cells may also serve as vehicles for the delivery models of induced demyelination of the adult CNS. Each of these
of normal enzymes to the endogenous cells, thus improving the models has advantages and disadvantages. The primary character-
functional deficit (Goldman and Windrem 2006). istic is that a substantial proportion of axons is available for the
Multiple sclerosis (MS) is an inflammatory CNS disorder transplanted cells to myelinate and therefore serves to assay the
characterized by disseminated demyelination and axonal loss biology and therapeutic potential of various cell preparations.
(see chapter 61). Even though remyelination occurs during
the early disease stages, it is progressively impaired, resulting
ultimately in lesions consisting of chronically demyelinated
4.1.1 Animal Models of Hypomyelination/
axons, axonal loss, and reactive gliosis. Given that axonal loss is
Dysmyelination
the leading cause of neurological disability characterizing MS This group includes animals characterized by (1) aberrant myelin
patients and that normal myelin is required for axonal integ- sheath production or maintenance owing to a defect in myelin
rity (Kornek et al. 2000), remyelination therapies are neces- protein–related genes (myelin mutants), or (2) progressive myelin
sary to ensure restoration of axonal function. In addition to loss resulting from abnormal accumulation of metabolites owing
myelin reconstruction, further benefits of cell therapy for MS to enzymatic deficiencies (models of Krabbe disease and metach-
have been suggested because transplanted cells may contribute romatic leukodystrophy). Because most myelin mutants are short
to axonal protection by suppressing ongoing inflammation lived, transplantation is usually performed in developing animals.
and activating endogenous repair. Neonate recipients have a highly permissive environment for
Spinal cord injury (SCI) is one of the most devastating condi- myelination because transplanted cells respond to developmen-
tions that affect the CNS and can lead to permanent disability. tal cues instead of reflecting chronic disease situation. In the case
It is characterized by lesions of CNS axons and a primary loss of mutants with an extended lifespan (e.g., canine shaking pup),
of cells at the site of injury. This results in posttraumatic inflam- transplantation has been performed into adult animals (Archer
matory responses, including activation of resident microglial et al. 1997). One of the advantages of these models is that myelin
cells and infiltration of circulating immune cells, which together produced by the transplanted cells can be readily recognized
with cytotoxic events, lead to secondary damages. These include because of its normal composition. For example, MBP posi-
formation of fluid-filled cysts, demyelination, and degeneration tive normal myelin is easily recognized in the shiverer mouse by
of axons. As a response to injury, a glial scar is formed primarily immunohistochemistry and electron microscopy (Lachapelle
by activated proliferating hypertrophic astrocytes or pericytes et al. 1983). Myelin mutants are classified into groups according to
(Goritz et al. 2011) generating a physical and chemical barrier the myelin protein–related gene affected (for review see Duncan
that seals off the injured tissue and restricts inflammation and cell et al. 2011. Major characteristics are listed in Table 57.1.
death. Although beneficial for tissue preservation during the early
stages after injury, eventually it creates a serious obstacle to regen-
eration. Inhibitory molecules, such as chondroitin sulfate proteo- 4.1.2 Induction of Demyelination in the Adult
glycans (Dou and Levine 1994), are upregulated within the glial Central Nervous System
scar, whereas myelin and myelin debris also constitute a major Models of induced demyelination include toxin-induced
source of factors preventing axonal growth and regeneration demyelination and induction of inflammatory pathologies of
(Caroni and Schwab 1988). Even after severe injuries some sur- the adult CNS. In toxin-induced models, cell grafting is gener-
viving axons persist in the subpial rim of white matter, but exhibit ally performed directly in the parenchyma, whereas in inflam-
demyelination and consequently dysfunctional conduction matory models cells are introduced in the venous or ventricular
G L I A L C E L L T R A N S P L A N TAT I O N F O R C E N T R A L N E RVO U S SYS T E M R E PA I R • 729
Table 57.1 MYELIN MUTANTS
SURVIVAL/CLINICAL
MUTANTS SPECIES DEFECT SIGNS PATHOLOGY DISEASE

Shiverer Mouse Deletion of 7 out of 9 2–3 Months Dysmyelination : lack of —


Mikoshiba et al. 1991 exons in MBP gene shivering MBP protein leading to
seizures uncompacted myelin

Long Evans Shaker (les) Rat Disruption of MBP 6–9 Months Dysmyelination/ —
Kwiecien et al. 1998 gene: insertion of demyelination lack of
retrotransposon in MBP protein leading to
noncoding region uncompacted myelin

Jimpy Mouse Base pair substitution 1-Month Oligodendrocyte death Severe form of
in 4 introns of seizures Hypomyelination PMD
PLP1 gene on X and mild inflammation
chromosomes Dysmyelination: remaining
myelin poorly compacted
Lack of PLP protein

Myelin deficient Rat X-linked mutants 23–25 Days Oligodendrocyte death Severe form of
Duncan and Milward 1995 with effected PLP1 Severe hypomyelination PMD
gene Remaining myelin lacks PLP
protein

PLP overexpressor Mouse PLP1 gene 23 Days (homozygous) Hypomyelination/ Severe form of
(PLOA) dysmyelination PMD
Griffiths et al. 1995 and major inflammation
2 Months Hypomyelination after 3–4
(heterozygous) months

Bradl et al. 1999 Rat PLP1 gene 23–24 Days Dysmyelination

Shaking pup Dog Point mutation in >2 Years Dysmyelination/ PMD


Archer et al. 1997 PLP1 gene hypomyelination

Mag-fyn KO Mouse Deficient for MAG Normal Hypomyelination


Biffiger et al. 2000 and fyn

Pex5 Mouse PEX5 12 Months Demyelination, ALD


Kassmann et al. 2007 axon loss, inflammation

Taiep Rat 17 Months


Duncan et al. 1992

Twitcher Mouse Galactocerebrosidase 34–45 Days Demyelination CNS Krabbe Globoïd


Duchen et al. 1980 and PNSecurreloboïd Leukodystrophy
macrophages

Primate

Canine

Arylsulfatase A (ASA) KO Mouse ASA 2 Years Sulfatide storage MLD


Hess et al. 1996 No obvious demyelination
Aspartoacylase KO Mouse Aspartoacylase 2–9 Months Myelin vacuolation, no Canavan disease
Matalon et al. 2000 demyelination

MBP, Myelin basic protein; MLD, metachromatic leukodystrophy; PLP, proteolipid protein; PMD, Pelizaeus-Merzbacher disease.

systems (reviewed in Tepavcevic and Baron-Van Evercooren by intraparenchymal zymogen injection and various virus-
2008). Focal models offer the possibility to graft cells either induced demyelinating diseases (see Table 57.3). These models
directly in the lesion to assay their repair potential or remotely offer an environment required to address more complex issues
form the lesion, to assay their ability to migrate. Their char- for cell therapy such as the ability of cells to gain access to
acteristics are summarized in Tables 57.2 and 57.3. Several lesions dispersed throughout the CNS, their survival within
models are characterized by more complex pathophysiologi- an inflammatory environment characterized by recurrent epi-
cal features than gliotoxin-induced models of demyelination, sodes of demyelination, as well as the ability of transplanted
including clinical deficits. These include experimental auto- preparations to sustain clinical recovery via neuroprotective
immune encephalomyelitis (EAE), demyelination induced and immunomodulatory properties.

730 • NEUROGLIA
Table 57.2 INDUCTION OF NONIMMUNE DEMYELINATION
AGENT/MODE SPECIES EXTENT CHRONICITY MODE PATHOLOGY

Lysolecithin Mouse, rat, rabbit, Focal Acute Membrane Focal loss of oligodendrocytes
intra-CNS injection cat, primate solubilizing agent and myelin, axon sparing, little
Hall et al. 1972 inflammation, followed by
remyelination.
Remyelination slower in
rabbits and nonhuman
primates than rodents
Ethidium bromide (EB) Rodents Focal but more DNA-intercalating Focal loss of oligodendrocyte
Intra-CNS injection extended agent and astrocytes leading to
than LPC demyelination, axon sparing,
lesions little inflammation

EB alone Acute Remyelination


Blakemore et al. 1982

EB combined with Chronic No remyelination


X-irradiation
Blakemore and Paterson,
1978
Cuprizone Rodents More diffuse Cuper chelator Widespread loss of
Diet oligodendrocytes, axon loss
Matsushima and
Morell 2001

6 wk Acute Remyelination
12 wk Chronic No remyelination

Table 57.3 INDUCTION OF IMMUNE-MEDIATED DEMYELINATION


INDUCTION MODE SPECIES EXTENT CHRONICITY MODE PATHOLOGICAL FEATURES DISEASE

Theilers’ virus (TMEV) Mouse Diffuse Persistent Viral- mediated Infection of neurons spreading MS
Lipton et al. 2005 to glial cells. including
oligodendrocytes, neuronal
degeneration, astrogliosis
Immune infiltrates
Mouse hepatitis virus Mouse Diffuse Acute Viral-mediated Transient replication in MS
(MHV) oligodendrocytes, leading to
Kristensson 1986 demyelination followed by slow
remyelination
EAE, peripheral Mouse, rat, Diffuse Monophasic, Immune-mediated Rupture of BBB, immune According to
injections of CNS guinea pig, relapse-remitting infiltrates, astrogliosis, axone loss, models :
homogenates, myelin, monkey (RR), oligodendrocyte ecurrentcurent RR, SP, or
myelin proteins, or primary (PP), events of demyelination followed PP MS
synthetic peptides and secondary by events of remyelination for
Gold et al. 2006 progressive (SP) RR, PP, and SP

Peripheral immunization Mouse, Rat Focal No prominent Immune-mediated Similar signs but localized in the Chronic MS
with myelin peptides clinical signs area of cytokine injections
followed by intra CNS
injections of cytokines
Kerschensteiner et al.
2004
Zymozan ip injection Rat Focal — Toll receptor-2 Focal inflammation, MS-like
Popovich et al. 2002 agonist demyelination, and axon loss

4.2 S P I NA L C O R D I N J U RY MO D E L S axons in the lesion, transplanted cells should ideally also sup-
port CNS axonal elongation and outgrowth. Available models
As mentioned, remyelination of demyelinated axons follow- reflect various degrees of traumatic injury and include partial
ing spinal cord injury appears fundamental to achieve func- or complete spinal cord transection, compression, or contu-
tional recovery. However, in addition to remyelinating the sion injury. Most cell transplantations are delivered directly at

G L I A L C E L L T R A N S P L A N TAT I O N F O R C E N T R A L N E RVO U S SYS T E M R E PA I R • 731


the site of injury or adjacent to it by injection of a cellular sus- The spinal cord becomes ischemic and the model mimics
pension via fine needles or capillaries. Transplantation is per- common clinical injuries and outcomes. An alternative to clip
formed either immediately after injury (acute phase) or after 1 compression is balloon compression in which a deflated
to 2 weeks following injury (subacute phase); unfortunately, balloon is inserted in the spinal cord at the desired level and is
only a few studies have dealt with chronic injuries (Tom et al. then inflated to cause tissue injury.
2009). The therapeutic potential of grafted cells is assessed by
demonstration of functional and histological recovery follow-
ing neuropathological analysis. 5 R O L E O F E XO G E N O U S G L I A L C E L L S I N
R E G E N E R AT I O N A N D M Y E L I N R E PA I R
4.2.1 Transection or Hemisection Models
The complete spinal cord transection injury model reflects symp- 5.1 A NATO M I C A L R E PA I R
toms of complete SCI in patients. Following laminectomy, spinal Glial cell types used for transplantation in CNS dysmyelina-
cord transection is performed with spring scissors. Occasionally, tion, demyelination, and SCI include Schwann cells, oligoden-
to attach the two ends for regeneration, a sterile gel or polymer droglial cells, and olfactory ensheathing cells (OECs). More
bridge is placed between the two severed spinal cord ends. The recently and rather unexpectedly, astrocytes have been used
advantage of this model is that the question of spared axons or in SCI, which when primed with certain factors appear to be
sprouting fibers is eliminated, allowing a clear identification of transformed from a growth-inhibitory to a growth-supporting
regenerated axons. The partial spinal transection injury model or phenotype (White et al. 2011). Recent data also indicate that
hemisection induces a less severe injury with motor dysfunction reactive astrocytes harvested from the lesion site after brain
appearing only in the ipsilateral injured side. injury acquire stem cell potential, suggesting that they may
be harnessed for repair strategies (Robel et al. 2011). Here, we
4.2.2 Contusion or Hemicontusion Models stress the role of these cells in regeneration and remyelination,
and refer to the repair potential of stem cells after transplanta-
Spinal contusion is the oldest and most widely used animal tion in models of dysmyelination, demyelination, and SCI.
model (Gruner 1992). In addition to motor dysfunction,
this injury elicits sensory dysfunction, including neuropathic
pain, tactile allodynia, and thermal hyperalgesia. Cervical
5.1.1 Committed Cells
contusion is rarely reported, because death can often occur
(Fehlings and Robins-Steele 2012). Therefore, cervical hemi- 5.1.1.1 Schwann Cells
contusion, following hemilaminectomy, is used instead. In Schwann cells (SCs) are the myelin-forming cells of the
cervical contusion models motor dysfunction appears in the peripheral nervous system. They are in intimate association
forelimbs (Gensel et al. 2006). The thoracic spinal cord con- and dynamic communication with neuronal axons regulat-
tusion model causing motor deficits in the hind limbs is more ing the development, maintenance, and function of peripheral
popular and is induced with impactors, such as the controlled nerves (see chapter 14). The well-established property of SCs
weight-drop impactor. to myelinate CNS demyelinated areas (Zujovic et al. 2007) has
fostered the wide use of these cells as an attractive cell source
to drive exogenous remyelination. Schwann cells are appealing
4.2.3 Compression Injury Models
candidates for autologous transplantation because they can be
Following laminectomy, compression injury is induced with grown and expanded from human biopsies (Fig. 57.1). Thus
clips calibrated to exert a force inducing moderate or severe the main advantage of using these peripheral nervous system
injury (Fehlings and Tator 1995). In either case the clip is (PNS) myelin-forming cells is that SCs can be obtained from
pressed dorsoventrally over the entire cord for 1 minute. neonate and adult sural nerve biopsies, cultured and expanded

Figure 57.1 Human Schwann Cells Isolated from Peripheral Nerve Biopsy. The culture was expanded in the presence of heregulin and Schwann cells
were purified from contaminating fibroblasts by immunopanning using anti-p75 neurotrophin receptor antibody. A. Purified Schwann cells were
transduced with a lentiviral vector for expression of the green fluorescent protein (green). B. Their purity (>95%) was assessed by immunofluorescence
staining for p75 (red). C. The merged picture is shown.

732 • NEUROGLIA
extensively in vitro, cryopreserved, and autotransplanted into
demyelinated CNS areas (Lavdas et al. 2008). Moreover, these
cells and their myelin are most likely not a target in MS. Rodent,
monkey, and human SCs have been successfully used to recon-
struct myelin sheaths and restore axonal conduction in dysmy-
elination, demyelination, and SCI. Moreover, remyelination by
exogenous SCs improves neurological functions in focally demy-
elinated areas of the CNS in rodents as well as in SCI (Honmou
et al. 1996). Finally, preliminary data indicate that SCs grafted
in a diffuse model of EAE, survive, migrate, and differentiate in
myelin-forming SCs (Zujovic et al. 2012). Importantly, autolo-
gous grafts of SCs in the primate demyelinated spinal cord have
been proved feasible (Bachelin et al. 2005).
Because of their inherent properties to sustain axonal
regeneration in the PNS, SCs were the first type of cells to
be transplanted into the injured spinal cord (Duncan et al.
1981). A number of studies have since demonstrated the abil-
ity of SCs or SC precursors to promote regeneration of the
lesioned spinal cord in rodent and primate models (for review
see Lavdas et al. 2008) (Fig. 57.2). Schwann cells have been
used for grafting either alone or in combination with other
cell types, such as olfactory ensheathing cells, with or without
growth factor administration (Fouad et al. 2005; Pearse et al.
2007). An alternative strategy has been to combine SC grafts
with sustained cAMP signaling to promote axonal regenera-
tion after injury (Pearse et al. 2004). Additionally, SCs have
been genetically engineered to secrete higher levels of growth
factors than they normally do, or other neuroprotective or
regeneration-promoting molecules (Girard et al. 2005; Lavdas
et al. 2010a; Papastefanaki et al. 2007).
One limitation in using SC grafts stems from their inabil-
ity to be efficiently integrated in an astrocyte-rich environ-
ment (Iwashita et al. 2000). To facilitate their incorporation
and at the same time introduce a cellular vector that would
render the CNS more permissive to regeneration, SCs were
engineered to express the polysialylated form of the neural cell
adhesion molecule NCAM (PSA-NCAM) on their surface
(Lavdas et al. 2006). Expression of PSA on NCAM results in
attenuation of cell-to-cell interactions and allows for structural
changes necessary for CNS repair (El Maarouf et al. 2006).
Transplantation of PSA-NCAM expressing rodent SCs in a
mouse model of spinal cord compression injury proved supe-
rior to using control SCs and resulted in a marked restoration
of hindlimb locomotor function (Papastefanaki et al. 2007).
Enhanced SC-astrocyte mixing, migration, and remyelina-
tion was also noted with transplantation of macaque SCs,
engineered to express PSA-NCAM, in a focal demyelination
model in nude mice (Bachelin et al. 2010).
Figure 57.2 Parasagittal Section of the Lesioned Mouse Spinal Cord
5.1.1.2 Olfactory Ensheathing Cells Transplanted with Schwann Cells, 4 Weeks After Compression Injury.
Olfactory ensheathing cells (OECs) are a distinct popula- A. Double labeling for fibronectin to illustrate the lesion site (blue).
tion of glial cells that wrap around—but do not myelinate— B. Glial fibrillary acid protein immunoreactivity (red). C. Transplanted
the axons of olfactory receptor neurons in their entire length, Schwann cells were isolated from transgenic mice expressing the green
as they extend from the olfactory epithelium in the PNS to fluorescence protein green fluorescent protein under the actin promoter
and appear in green. D. The merged picture. Orientation is shown in
the olfactory bulb in the CNS (Doucette 1990). Unlike SCs, (A). C, caudal; D, dorsal; R, rostral; V, ventral. Scale bar: 100 μm. From
OECs intermingle well with astrocytes (Lakatos et al. 2000) Lavdas et al. 2010b.
and have been shown to support the development and regen-
eration of the olfactory nervous system (Su and He 2010).

G L I A L C E L L T R A N S P L A N TAT I O N F O R C E N T R A L N E RVO U S SYS T E M R E PA I R • 733


Although OECs do not produce a myelin sheath around the boundary to form boundary cap (BC) cells (Fig. 57.3). More
axons of olfactory receptor neurons in situ, they can myelinate recently, embryonic Schwann cell precursors (SCp) or BC are
when presented with large-diameter axons in vitro (Babiarz the subject of growing interest for cell-based therapy. These
et al. 2011). Moreover, rodent, canine, human, or porcine immature cells appear to be less inhibited by CNS cues than
OECs isolated from the adult olfactory bulb are capable of neonatal and adult SCs and are therefore able to colonize
extensive functional remyelination following transplantation and remyelinate more efficiently the demyelinated or injured
into the demyelinated rat CNS (Deng et al. 2006). Because of CNS (Zujovic et al. 2010). Although embryonic BC and SCp
their biological properties and relative accessibility in humans differentiate in Schwann cells when transplanted in demyeli-
from the olfactory mucosa, OECs have also been considered nated lesions (Zujovic et al. 2010), they differentiate in oligo-
as candidate cells for autologous transplantation and have dendrocytes when confronted with CNS developmental cues
been tested in numerous animal models of SCI (Barnett and (Binder et al. 2011; Zujovic et al. 2011) (Fig. 57.4). Finally, SCp
Riddell 2007). Although promising, OEC transplantation promote extensive axon regeneration after transplantation in
has not yielded sufficient anatomical or functional recovery SCI (Agudo et al. 2008).
and, as with SCs, has led to implementation of combinatorial Neural crest–derived stem cells (NCSC) were also identified
approaches, such as cotransplantation with other cell types, in embryonic tissue, such as embryonic sciatic nerve (Morrison
genetic modification, or additional interventions, for example et al. 1999) and various adult tissues such as gut (Kruger et al.
(Cao et al. 2004). 2002), skin (McKenzie et al. 2006; Sieber-Blum et al. 2004),
heart (Tomita et al. 2005), DRG (Li et al. 2007; Nagoshi
et al. 2008), and bone marrow (Nagoshi et al. 2008; Wislet-
5.1.2 Stem Cells and Progenitor Cells
Gendebien et al. 2012). Although the regenerative potential of
5.1.2.1 Neural Crest–Derived Stem and bone marrow mesenchymal cells (BMSCs) after transplantation
Progenitor Cells in the injured spinal cord (Kocsis et al. 2002) or injured nerve
During development of the neural tube, neural crest cells (Keilhoff et al. 2006) suggested transdifferentiation of these cells
(NCC) colonize various embryonic tissues and differenti- into remyelinating SC, the identification of NC-SC in bone
ate into a wide range of cells, including neurons and glia in marrow suggest a possible NC origin of the remyelinating SC
the PNS, pigment cells in the skin, and endocrine cells in the in the mentioned studies. Stemlike cells can also be derived from
adrenal and thyroid gland. They also contribute to the mesen- adult peripheral nerves in response to injury (Takagi et al. 2011)
chymal lineage, giving rise to cartilage, bone, dermis, adipose or adult palatal ridges (Widera et al. 2011). However, so far, it
tissue from the craniofacial area, and vascular smooth muscle is not clear whether these cells arise from de-differentiation of
cells (pericytes). Within the PNS, SC go through different Schwann cells or a discrete population of unidentified stem cells.
transitional stages before becoming myelin competent cells Although the persistence of stem cell reservoirs in adult tissues
(Zujovic et al. 2007). Neural crest cells (NCC) differenti- opens new perspectives for cell therapy in demyelinating disor-
ate in SC precursors before becoming immature SC, and a ders and SCI, future work should further characterize the stem-
subpopulation of NCC arrest transiently at the PNS-CNS ness of these adult NC-SC compared to embryonic NC-SC.
BC

nmSC

NCC iSC
SCp

Pro-mSC mSC
Sox-9
Sox-2
Sox-10
Krox-20

Pluripotene Mullipotent Unipotent (SC)


Unipotent (SC)
(mesenchymal and (N,SC,SM, O) Poory migratory
Non migratory
neuroepithelial cells) Migratory
Migratory

Figure 57.3 Schematic Drawing Illustrating the Migration and Differentiation Potential of Different Stages (Defined by Stage-Specific Transcription
Factors) of the Schwann Cell Lineage After Engraftment in Different Situations in the Central Nervous System. NCCs migrate extensively and gener-
ate a wide variety of mesenchymal and neuroepithelial derivatives. Early stages of the SC lineage such as BC and SCP still migrate efficiently and can
generate multiple PNS and CNS derivatives (in developmental conditions). By contrast, more mature cells (immature SC and mature SC) do not
migrate efficiently and differentiate essentially in SC (myelinating or nonmyelinating) when grafted in the demyelinated CNS. BC, boundary cap cells;
iSC, immature Schwann cells; mSC, myelinating Schwann cells; N, neurons; NCC, neural crest cells; nmSC, nonmyelinating Schwann cells; SCP,
Schwann cell precursors; SM, smooth muscle cells.

734 • NEUROGLIA
Figure 57.4 Mouse Boundary Cap Cells Give Rise to Myelinating Oligodendrocytes After Transplantation into the Forebrain of the Newborn
Shiverer Brain. (A1–A3) YFP-positive cells (green) give rise to mature myelin basic protein (MBP) expressing oligodendrocytes with features of
myelin-forming cells extending processes to internodes (arrows). B1–3. Illustrated at higher magnification. C. Immunodetection for MBP indicated
widespread of BC-derived myelin patches in the subcortical white matter. D,E. Ultrastructural analysis confirms the presence of compacted donor
myelin (E, inset) compared with uncompacted shiverer myelin (D, inset).

Olfactory ensheathing cell progenitors OEp were found in and Blakemore 2002) (see chapter 13). In agreement, some
the human and rodent olfactory epithelium (Winstead et al. transplantation studies suggested that rodent or human oli-
2005). Transplantation into the demyelinated spinal cord lesion godendrocytes were capable of myelin formation (Baron-Van
indicated that exogenous rat OEp provide extensive remyelina- Evercooren and Blakemore 2004.). However, this possibility
tion (Markakis et al. 2009). Moreover, human OEp were trans- was not fully demonstrated mainly because of the difficulty
planted in the rodent traumatized spinal cord and shown to of obtaining cell preparations entirely devoid of OPC or the
rescue axotomized rubrospinal neurons and promote functional insufficient specificity of markers allowing the selection of the
recovery (Xiao et al. 2005). Yet their use in disease models of mature oligodendrocyte stage. Instead, OPC were the most
trauma or demyelination deserves further attention. extensively studied cells of the lineage (see Fig. 57.5). Rodent
Future studies should document the resistance to neuroin- and human OPC elicit vigorous remyelination when trans-
flammation and the neuroregenerative or neuroprotective planted in the developing brain of myelin mutants or focal
potential of these various adult NC-derived stem cells. lesions of demyelination (Groves et al. 1993; Lachapelle et al.
1983; Windrem et al. 2008). Interestingly, multiple injections
5.1.2.2 Oligodendrocyte Progenitors of these cells in the developing CNS parenchyma led to impres-
Until recently it was considered that exogenous cells that sive colonization of the entire neuraxis by donor-derived myelin
remyelinate axons in the adult CNS could be derived both (Archer et al. 1997). Moreover, remyelination by transplanted
from immature OPC and mature oligodendrocytes (Chari OPC is associated with functional recovery (Utzschneider

G L I A L C E L L T R A N S P L A N TAT I O N F O R C E N T R A L N E RVO U S SYS T E M R E PA I R • 735


Mature Olg
Pre-Olg Olg
NPC OPC

Olig1+
Olig2+/- NG2+ NG2+ O4+ Olig2+/–
Ollg 1, 2+ O4+ GalC+ Sox10+
Sox 10+ Ollg 1,2+ CNPase+ GalC+
PDGF-aR+ Sox 10+ Olig 1,2+ MOG+
A2B5+ PDGF-aR+ Sox10+ MBP+

Pluripotent Parenchymal OPC


Unipotent (O) Unipotent (O)
(N,O,A, SC) Multiotent (N,O,A, SC)
Migratory (+/–) Non mlgratory
Migratory Migratory (+)
SVZ OPC
Multiotent (N,O,A)
Migratory (++)

Figure 57.5 Schematic Drawing Illustrating the Migration and Differentiation Potential of Different Stages (Defined by Stage-Specific Markers) of
the Oligodendroglial Lineage After Engraftment in Different Situations in the Central Nervous System. Neural precursor cells migrate extensively and
generate neurons, astrocytes, oligodendrocytes and sometimes SC (demyelinating conditions). According to their origin, OPC migrate efficiently and
generate multiple CNS derivatives, including SC (demyelinating conditions). By contrast more mature cells (pre-Olg, Olg, and mature Olg) are non
migratory and generate solely and in limited numbers oligodendrocytes. A, astrocytes; N, neurons; NPC, neural precursor cells; Olg, oligodendro-
cytes; OPC, oligodendrocyte progenitor cells; Pre-Olg, pre-oligodendrocytes; SC, Schwann cells.

et al. 1994). Although OPCs were found to migrate poorly intra-CNS transplantation. Of concern were the regulation
in the adult normal CNS white matter, grafting CG4 cells in of their proliferation to avoid tumor formation, migration
a model of diffuse EAE (Tourbah et al. 1997) or at a distance capacity, differentiation in neural cell types other than oli-
of a focal inflammatory lesion (Wang et al. 2011b) indicated godendrocytes, and consequently their reduced ability to
that inflammation sustains OPC migration through normal reconstruct myelin in their host. Preclinical studies in a vari-
white matter and subsequent remyelination, a major issue for a ety of disease models established that tumor formation was
disease such as MS, which is characterized by randomly spread not an issue because these cells integrated efficiently within
lesions. the CNS parenchyma. Moreover, rodent and human NPCs
Most of the mentioned studies were performed isolat- migrate extensively (and thus more efficiently than OPC)
ing OPC from whole forebrain or spinal cord and did not leading to massive colonization of the neuraxis even in the
discriminate between parenchymal OPC and those that are adult CNS (Buchet et al. 2011).
localized in germinative niches such as the dentate gyrus Transplantation of NPCs in the developing forebrain
(DG) and subventricular zone (SVZ) (see chapters 5 and leads to the generation of multiple neural cell types, includ-
40). Transplantation of cells selected on the basis of the OPC ing astrocytes, neurons, and oligodendrocytes (Yandava
marker NG2 in the newborn brain indicated that postnatal et al. 1999). By contrast they generate astrocytes and oligo-
and adult SVZ NG2+ cells generate functional neurons in dendrocytes (Buchet et al. 2011) (Fig. 57.6), and sometimes
addition to CNS glia, and migrate more efficiently in the SCs (Keirstead et al. 1999), when grafted in the demyeli-
CNS compared with parenchymal NG2+ cells, which gen- nated spinal cord. Nevertheless, their capacity to generate
erate essentially glial cells and migrate poorly in the CNS oligodendrocytes seems to be region and species depen-
parenchyma (Aguirre and Gallo 2004). These studies clearly dent. However, it is limited when grafted in conditions of
underline region-specific differences among NG2+ cells focal inflammatory demyelination (Walczak et al. 2011).
(see chapters 5, 10 and 30). Adult SVZ fragments containing neural stem cells and
immature neural precursors and neurospheres form myelin
5.1.2.3 Central Nervous System Neural Stem/ when grafted into the neonate brain (Lachapelle et al. 2002;
Precursor Cells Zhang et al. 1999). This potential can be enhanced by prim-
Embryonic or adult neural stem/precursor cells (NPCs) ing SVZ donors with FGF (Lachapelle et al. 2002) (see
harvested from germinal areas of the CNS are typically cul- chapter 40). Most studies with NPC transplantation in SCI
tured and expanded as neurospheres in the presence of EGF have also reported that these cells integrate well and dif-
and/or FGF (Reynolds and Weiss 1996). They contain pre- ferentiate robustly in the host spinal cord, thus conferring
cursors for neurons, astrocytes, and oligodendrocytes and some degree of improvement (Karimi-Abdolrezaee et al.
also some self-renewing cells. In view of their self-renewability 2006). To increase the yield in neuronal or oligodendrocyte
and multipotency, NPCs are regarded as cells of high thera- progeny, rodent and human NPCs have been genetically
peutic interest for a variety of neurological diseases, includ- modified with the use of viral vectors to express neuronal
ing those affecting myelin, as well as CNS injuries. Numerous or glial fate determinants (Hwang et al. 2009; Maire et al.
studies have characterized the behavior of these cells after 2009; Makri et al. 2010).

736 • NEUROGLIA
Figure 57.6 Human Fetal Neural Progenitor Cells Give Rise to Myelinating Oligodendrocytes After Transplantation into the lysophosphatidylcholine
(LPC)-Induced Demyelination of the Shiverer Mouse Spinal Cord. A–D. Immunohistochemical labeling against myelin basic protein (MBP) (green)
illustrating human-derived myelin in the shiverer spinal cord at different rostro (A) caudal levels (D). E–H. Immunohistochemical detection of MBP
(red), human NOGO (green), and NF-200 (blue), illustrating multipolar human neural stem/precursor cell–derived oligodendrocytes extending
processes and wrapping axons in dorsal and ventral white matter. E,F. Cross-sections. F. An orthogonal view of (E). H. A higher magnification of (G)
illustrating MBP+ internodes and typical nodes of Ranvier. I–L. Ultrastructural analysis of myelin compaction in (I) shiverer mice without lesion, (J)
shiverer mice with LPC lesion, (K), shiverer mice with LPC lesion, and hNPC graft, and (L) higher magnification illustrating compaction of hNPC–
derived myelin. Scale bars (A–D), 200 μm; (E), 20 μm; (F,H) 5 μm; (G) 10 μm; (I–K) 250 nm; (L) 20 nm.

5.1.2.4 Embryonic Stem Cells and Induced be negligible in a completely paralyzed individual suffering
Pluripotent Stem Cells from SCI, the implications on other types of patients have to
OPCs and NPCs can also be derived from embryonic be appreciated. Notably, the use of iPS meets the needs for
stem cells (ES). A few studies reported the first evidence noninvasive isolation procedures. Rodent and human OPC
of myelination achieved by grafting ES-derived glial pro- were generated via differentiation of mouse iPS into NPCs,
genitors, for example (McDonald et al. 1999; Perez-Bouza and transplanted in demyelinating lesions, where they differ-
et al. 2005). Protocols were established to derive OPC from entiated into a few myelin-forming oligodendrocytes (Czepiel
human-ES cells (Hu et al. 2009). However, the number of et al. 2011; Pouya et al. 2011)). In addition, NPCs were derived
newly generated glial progenitors in vitro and in vivo is weak. from mouse embryonic iPS and human adult iPS and used in
Treatment of ES cells with all-trans retinoic acid alone or in transplantation paradigms for treatment of SCI (Nori et al.
combination with morphogens generated OPC-enriched 2011; Sakai et al. 2012; Tsuji et al. 2010)). Rodent and human
populations, which generated myelin after grafting into NC-PC can also be derived from ES cells and iPS (Lee et al.
the neonate shiverer brain (Izrael et al. 2007) and a model 2010; Menendez et al. 2011). Wang et al. (2011a) exploited the
of SCI (Keirstead et al. 2005). Treatment with interleukin repair potential of hiPS-NC by seeding them in nerve con-
6 (Zhang et al. 2006) or overexpression of Olig2 (Du et al. duits, and observed their differentiation in myelin-forming
2006) are other means to favor oligodendrogenesis from SCs, correlating with improved regeneration of the sciatic
mouse ES cells. nerve. However, the role of these PNS cells in CNS remyeli-
The future for regenerative medicine may reside in nation and regeneration has not been addressed.
induced pluripotent stem cells (iPS) as in theory they can be Although these pioneer studies remain to be confirmed and
derived from the patients’ own cells, expanded and used for their transplantation in various animal models of dysmyelination,
autologous transplantation (Fig. 57.7). Although endogenous demyelination, and SCI are required to demonstrate the
cells, such as Schwann cells, can also serve for autologous safety and efficacy of these novel cellular sources, these
transplantation, their use necessitates sacrificing a peripheral developments reported so far and the efforts made to improve
nerve. Although the significance of such an approach may the differentiation of iPS cells into myelin-forming candidates

G L I A L C E L L T R A N S P L A N TAT I O N F O R C E N T R A L N E RVO U S SYS T E M R E PA I R • 737


Figure 57.7 Schematic Representation of a Paradigm of Autologous Transplantation. The future for regenerative medicine may reside in cells harvested
from accessible human tissues, such as peripheral nerves (Schwann cells), olfactory mucosa (olfactory ensheathing cells), or the skin (fibroblasts for
derivation of induced pluripotent stem cells). Schwann cells and olfactory ensheathing cells may be cultured and expanded in vitro for autologous
transplantation, whereas induced pluripotent stem cells may be differentiated to neural precursors before being grafted into the injured central nervous
system.

open the perspective for autologous cell-based therapy for their intracerebroventricular delivery in a cuprizone-induced
demyelinating disorders and CNS trauma. model of demyelination (Einstein et al. 2009). Although
NPC remained at an immature state, enhanced remyelination
resulted from increased endogenous OPC proliferation, an
5.2 BYS TA N D E R E F F E C T S
effect mediated by platelet-derived growth factor (PDGF) A
A series of experimental studies suggest that both mesenchy- and fibroblast growth factor (FGF) 2, and their subsequent
mal stem cells (MSC) and NPCs on transplantation exert differentiation. In a recent study (Cusimano et al. 2012), it
other therapeutic effects than anatomical repair. Indeed, was shown that NPCs transplanted in a model of SCI also
NPCs or BMSC transplanted either by intrathecal or intrave- survived mostly undifferentiated within the host by finding
nous cell injection ameliorate the clinicopathological features their way toward a perilesional inflammatory-like vascular
of rodents with acute, chronic, and relapsing EAE (Martino niche, in close proximity to macrophages and microglial cells.
et al. 2010; Uccelli 2008). This phenomenon is caused by the The authors argue that from their homing site, undifferenti-
remarkable neuroprotective and immunomodulatory capaci- ated NPCs were able to control innate and adaptive immune
ties that NPC and BMSC exert within the CNS. These effects responses to induce tissue healing and functional recovery.
occur most likely via the release of soluble molecules (e.g., This and other studies highlight the remarkable potential of
cytokines, chemokines), which leads to downregulation of the transplanted NPCs, MSCs, and other cell types to influence
encephalitogenic inflammatory T cells, antigen-presenting bystander events, such as myelin repair and axonal rescue or
dendritic cells, and microglia/macrophages. Recent evidence regeneration after SCI.
shows that intravenously or subcutaneously transplanted The bystander effect of more committed cells is also the
NPCs might also exert immunomodulatory functions at the subject of recent studies. Olfactory ensheathing cells release
level of peripheral lymphoid organs (Pluchino et al. 2005). diffusible factors that regulate the proliferation and differ-
Although these could be features shared by immature cell entiation of NPCs and may be involved in neurogenesis,
types, similar bystander effects or immunomodulatory prop- which takes place throughout life in the olfactory system
erties of OPC or NC-SC were not reported. The bystander (Zhang et al. 2008). Additionally, SC transplants facilitate
effects of exogenous cells could also result from a trophic the emergence of host SCs in the lesioned spinal cord, which
effect on endogenous cells. Such trophic effect has been alongside with grafted SCs contribute to axon regenera-
reported for BMSC, but also more recently for NPCs after tion and protection. Yet their role in immunomodulation

738 • NEUROGLIA
has not been investigated in SCI or inflammatory CNS Barnett SC, Riddell JS. 2007. Olfactory ensheathing cell transplantation
demyelination. as a strategy for spinal cord repair—what can it achieve? Nat Clin
Pract Neurol 3(3):152–161.
Baron-Van Evercooren A, Blakemore WF. 2004. Remyelination through
engraftment. In: Lazzarini RA (ed.), Myelin biology and disorders.
6 S U M M A RY A N D P E R S P E C T I VE S San Diego: Elsevier, pp. 143–161.
Biffiger K, Bartsch S, Montag D, Aguzzi A, Schachner M, Bartsch U.
Over the years, the idea to treat demyelinating disorders or 2000. Severe hypomyelination of the murine CNS in the absence of
myelin-associated glycoprotein and fyn tyrosine kinase. J Neurosci.
SCI with cells capable of populating the lesion site and cre- Oct 1;20(19):7430–7.
ate a regeneration permissive environment within the CNS Binder E, Rukavina M, Hassani H, Weber M, Nakatani H, Reiff T, et al.
has attracted considerable attention. From the early days of 2011. Peripheral nervous system progenitors can be reprogrammed
peripheral nerve grafts to recent paradigms of cellular trans- to produce myelinating oligodendrocytes and repair brain lesions.
plants, the idea has emerged that stem cells and progenitor J Neurosci 31(17):6379–6391.
Blakemore WF. 1977. Remyelination of CNS axons by Schwann cells
cells are more promising, than committed cells. Moreover, transplanted from the sciatic nerve. Nature 266(5597):68–69.
combinatorial approaches hold promise as a means for- Blakemore WF. 1982. Ethidium bromide induced demyelination in the
ward. New knowledge has been acquired as to the fate of spinal cord of the cat. Neuropathol Appl Neurobiol 8(5):365–375.
transplanted cells in vivo and, most importantly, how donor Blakemore WF, Crang AJ. 1988. Extensive oligodendrocyte remyelina-
and host cells respectively interact. Thus, the notion of cell tion following injection of cultured central nervous system cells into
demyelinating lesions in adult central nervous system. Dev Neurosci
transplantation as a cell replacement strategy has shifted 10(1):1–11.
toward the concept that it represents a multifaceted thera- Blakemore WF, Patterson RC. 1978. Suppression of remyelination in the
peutic approach conferring neuroprotection, trophic support, CNS by X-irradiation. Acta Neuropathol 42(2):105–113.
immunomodulation, and manipulation of reactive gliosis, Bradl M, Bauer J, Inomata T, Zielasek J, Nave KA, Toyka K,
triggering endogenous regenerative processes in addition to Lassmann H, Wekerle H. 1999. Transgenic Lewis rats overexpressing
the proteolipid protein gene: myelin degeneration and its effect on
anatomical repair. T cell-mediated experimental autoimmune encephalomyelitis. Acta
Neuropathol. Jun;97(6):595–606.
Brustle O, Jones KN, Learish RD, Karram K, Choudhary K, Wiestler OD,
AC K N OW L E D G M E N T S et al. 1999. Embryonic stem cell-derived glial precursors: a source of
myelinating transplants. Science 285(5428):754–756.
Buchet D, Garcia C, Deboux C, Nait-Oumesmar B, Baron-Van
The authors thank Florentia Papastefanaki (RM’s lab) and Evercooren A. 2011. Human neural progenitors from different foe-
Delphine Buchet and Violetta Zujovic (ABVE’s lab) for pro- tal forebrain regions remyelinate the adult mouse spinal cord. Brain
viding original figures; Alexandros A. Lavdas and Violetta 134(Pt 4):1168–1183.
Zujovic for help with editing the manuscript, and all mem- Cajal SRy. 1928. Degeneration and regeneration of the nervous system.
bers of the Matsas and Baron-Van Evercooren laboratories for New York: Oxford University Press.
Cao L, Liu L, Chen ZY, Wang LM, Ye JL, Qiu HY, et al. 2004. Olfactory
their useful input. The authors acknowledge financial support ensheathing cells genetically modified to secrete GDNF to promote
to RM from FP7 REGPOT NEUROSIGN Project 264083 spinal cord repair. Brain 127(Pt 3):535–549.
and Fondation BNP Paribas; to ABVE from ELA, ARSEP Caroni P, Schwab ME. 1988. Antibody against myelin-associated inhibi-
Foundation and FP7 LEUKOTREAT Project. tor of neurite growth neutralizes nonpermissive substrate properties
of CNS white matter. Neuron 1(1):85–96.
Chari DM, Blakemore WF. 2002. New insights into remyelination fail-
ure in multiple sclerosis: implications for glial cell transplantation.
REFERENCES Mult Scler 8(4):271–277.
Cusimano M, Biziato D, Brambilla E, Donega M, Alfaro-Cervello
Agudo M, Woodhoo A, Webber D, Mirsky R, Jessen KR, McMahon SB. C, Snider S, et al. 2012. Transplanted neural stem/precursor cells
2008. Schwann cell precursors transplanted into the injured spinal instruct phagocytes and reduce secondary tissue damage in the
cord multiply, integrate and are permissive for axon growth. Glia injured spinal cord. Brain. 135:447–460.
56(12):1263–1270. Czepiel M, Balasubramaniyan V, Schaafsma W, Stancic M, Mikkers H,
Aguirre A, Gallo V. 2004. Postnatal neurogenesis and gliogenesis in the Huisman C, et al. 2011. Differentiation of induced pluripotent stem
olfactory bulb from NG2-expressing progenitors of the subventricu- cells into functional oligodendrocytes. Glia 59(6):882–892.
lar zone. J Neurosci 24(46):10530–10541. Deng C, Gorrie C, Hayward I, Elston B, Venn M, Mackay-Sim A,
Archer DR, Cuddon PA, Lipsitz D, Duncan ID. 1997. Myelination of et al. 2006. Survival and migration of human and rat olfactory
the canine central nervous system by glial cell transplantation: a ensheathing cells in intact and injured spinal cord. J Neurosci Res
model for repair of human myelin disease. Nat Med 3(1):54–59. 83(7):1201–1212.
Babiarz J, Kane-Goldsmith N, Basak S, Liu K, Young W, Grumet M. Dou CL, Levine JM. 1994. Inhibition of neurite growth by the NG2
2011. Juvenile and adult olfactory ensheathing cells bundle and chondroitin sulfate proteoglycan. J Neurosci 14(12):7616–7628.
myelinate dorsal root ganglion axons in culture. Exp Neurol 229(1): Doucette R. 1990. Glial influences on axonal growth in the primary
72–79. olfactory system. Glia 3(6):433–449.
Bachelin C, Lachapelle F, Girard C, Moissonnier P, Serguera-Lagache C, Du ZW, Li XJ, Nguyen GD, Zhang SC. 2006. Induced expression
Mallet J, et al. 2005. Efficient myelin repair in the macaque spinal cord of Olig2 is sufficient for oligodendrocyte specification but not
by autologous grafts of Schwann cells. Brain 128(Pt 3):540–549. for motoneuron specification and astrocyte repression. Mol Cell
Bachelin C, Zujovic V, Buchet D, Mallet J, Baron-Van Evercooren A. Neurosci 33(4):371–380.
2010. Ectopic expression of polysialylated neural cell adhesion mol- Duchen LW, Eicher EM, Jacobs JM, Scaravilli F, Teixeira F. 1980.
ecule in adult macaque Schwann cells promotes their migration and Hereditary leucodystrophy in the mouse: the new mutant twitcher.
remyelination potential in the central nervous system. Brain 133(Pt 2): Brain. Sep;103(3):695–710.
406–420.

G L I A L C E L L T R A N S P L A N TAT I O N F O R C E N T R A L N E RVO U S SYS T E M R E PA I R • 739


Duncan ID, Aguayo AJ, Bunge RP, Wood PM. 1981. Transplantation of Peters C, Gieselmann V. 1996. Phenotype of arylsulfatase A-deficient
rat Schwann cells grown in tissue culture into the mouse spinal cord. mice: relationship to human metachromatic leukodystrophy. Proc
J Neurol Sci 49(2):241–252. Natl Acad Sci USA. Dec 10;93(25):14821-6.
Duncan ID, Kondo Y, Zhang SC. 2011. The myelin mutants as models Honmou O, Felts PA, Waxman SG, Kocsis JD. 1996. Restoration of nor-
to study myelin repair in the leukodystrophies. Neurotherapeutics. mal conduction properties in demyelinated spinal cord axons in the
2011 Oct;8(4):607–24. adult rat by transplantation of exogenous Schwann cells. J Neurosci
Duncan ID, Lunn KF, Holmgren B, Urba-Holmgren R, Brignolo- 16(10):3199–3208.
Holmes L. 1992. The taiep rat: a myelin mutant with an associated Hu BY, Du ZW, Zhang SC. 2009. Differentiation of human oligoden-
oligodendrocyte microtubular defect. J Neurocytol. Dec;21(12): drocytes from pluripotent stem cells. Nat Protoc 4(11):1614–1622.
870–84. Hwang DH, Kim BG, Kim EJ, Lee SI, Joo IS, Suh-Kim H, et al. 2009.
Duncan ID, Milward EA. 1995. Glial cell transplants: experimental Transplantation of human neural stem cells transduced with Olig2
therapies of myelin diseases. Brain Pathol. Jul;5(3):301-10. transcription factor improves locomotor recovery and enhances
Einstein O, Friedman-Levi Y, Grigoriadis N, Ben-Hur T. 2009. myelination in the white matter of rat spinal cord following contusive
Transplanted neural precursors enhance host brain-derived myelin injury. BMC Neurosci 10:117.
regeneration. J Neurosci 29(50):15694–15702. Iwashita Y, Fawcett JW, Crang AJ, Franklin RJ, Blakemore WF. 2000.
El Maarouf A, Petridis AK, Rutishauser U. 2006. Use of polysialic acid Schwann cells transplanted into no

You might also like