Sei sulla pagina 1di 461

PROGRESS IN BRAIN RESEARCH

VOLUME 161

NEUROTRAUMA: NEW INSIGHTS INTO PATHOLOGY AND TREATMENT


Other volumes in PROGRESS IN BRAIN RESEARCH

Volume 124: Cerebellar Modules: Molecules, Morphology and Function, by N.M. Gerrits, T.J.H. Ruigrok and C.E. De Zeeuw (Eds.)
– 2000, ISBN 0-444-50108-8.
Volume 125: Transmission Revisited, by L.F. Agnati, K. Fuxe, C. Nicholson and E. Syková (Eds.) – 2000, ISBN 0-444-50314-5.
Volume 126: Cognition, Emotion and Autonomic Responses: the Integrative Role of the Prefrontal Cortex and Limbic Structures, by
H.B.M. Uylings, C.G. Van Eden, J.P.C. De Bruin, M.G.P. Feenstra and C.M.A. Pennartz (Eds.) – 2000, ISBN 0-444-
50332-3.
Volume 127: Neural Transplantation II. Novel Cell Therapies for CNS Disorders, by S.B. Dunnett and A. Björklund (Eds.) – 2000,
ISBN 0-444-50109-6.
Volume 128: Neural Plasticity and Regeneration, by F.J. Seil (Ed.) – 2000, ISBN 0-444-50209-2.
Volume 129: Nervous System Plasticity and Chronic Pain, by J. Sandkühler, B. Bromm and G.F. Gebhart (Eds.) – 2000, ISBN 0-444-
50509-1.
Volume 130: Advances in Neural Population Coding, by M.A.L. Nicolelis (Ed.) – 2001, ISBN 0-444-50110-X.
Volume 131: Concepts and Challenges in Retinal Biology, by H. Kolb, H. Ripps and S. Wu (Eds.) – 2001, ISBN 0-444-50677-2.
Volume 132: Glial Cell Function, by B. Castellano López and M. Nieto-Sampedro (Eds.) – 2001, ISBN 0-444-50508-3.
Volume 133: The Maternal Brain. Neurobiological and Neuroendocrine Adaptation and Disorders in Pregnancy and Post Partum, by
J.A. Russell, A.J. Douglas, R.J. Windle and C.D. Ingram (Eds.) – 2001, ISBN 0-444-50548-2.
Volume 134: Vision: From Neurons to Cognition, by C. Casanova and M. Ptito (Eds.) – 2001, ISBN 0-444-50586-5.
Volume 135: Do Seizures Damage the Brain, by A. Pitkänen and T. Sutula (Eds.) – 2002, ISBN 0-444-50814-7.
Volume 136: Changing Views of Cajal’s Neuron, by E.C. Azmitia, J. DeFelipe, E.G. Jones, P. Rakic and C.E. Ribak (Eds.) – 2002,
ISBN 0-444-50815-5.
Volume 137: Spinal Cord Trauma: Regeneration, Neural Repair and Functional Recovery, by L. McKerracher, G. Doucet and S.
Rossignol (Eds.) – 2002, ISBN 0-444-50817-1.
Volume 138: Plasticity in the Adult Brain: From Genes to Neurotherapy, by M.A. Hofman, G.J. Boer, A.J.G.D. Holtmaat, E.J.W.
Van Someren, J. Verhaagen and D.F. Swaab (Eds.) – 2002, ISBN 0-444-50981-X.
Volume 139: Vasopressin and Oxytocin: From Genes to Clinical Applications, by D. Poulain, S. Oliet and D. Theodosis (Eds.) – 2002,
ISBN 0-444-50982-8.
Volume 140: The Brain’s Eye, by J. Hyönä, D.P. Munoz, W. Heide and R. Radach (Eds.) – 2002, ISBN 0-444-51097-4.
Volume 141: Gonadotropin-Releasing Hormone: Molecules and Receptors, by I.S. Parhar (Ed.) – 2002, ISBN 0-444-50979-8.
Volume 142: Neural Control of Space Coding, and Action Production, by C. Prablanc, D. Pélisson and Y. Rossetti (Eds.) – 2003,
ISBN 0-444-509771.
Volume 143: Brain Mechanisms for the Integration of Posture and Movement, by S. Mori, D.G. Stuart and M. Wiesendanger (Eds.) –
2004, ISBN 0-444-513892.
Volume 144: The Roots of Visual Awareness, by C.A. Heywood, A.D. Milner and C. Blakemore (Eds.) – 2004, ISBN 0-444-50978-X.
Volume 145: Acetylcholine in the Cerebral Cortex, by L. Descarries, K. Krnjević and M. Steriade (Eds.) – 2004, ISBN 0-444-51125-3.
Volume 146: NGF and Related Molecules in Health and Disease, by L. Aloe and L. Calzà (Eds.) – 2004, ISBN 0-444-51472-4.
Volume 147: Development, Dynamics and Pathology of Neuronal Networks: From Molecules to Functional Circuits, by J. Van Pelt,
M. Kamermans, C.N. Levelt, A. Van Ooyen, G.J.A. Ramakers and P.R. Roelfsema (Eds.) – 2005, ISBN 0-444-51663-8.
Volume 148: Creating Coordination in the Cerebellum, by C.I. De Zeeuw and F. Cicirata (Eds.) – 2005, ISBN 0-444-51754-5.
Volume 149: Cortical Function: A View from the Thalamus, by V.A. Casagrande, R.W. Guillery and S.M. Sherman (Eds.) – 2005,
ISBN 0-444-51679-4.
Volume 150: The Boundaries of Consciousness: Neurobiology and Neuropathology, by Steven Laureys (Ed.) – 2005, ISBN 0-444-51851-7.
Volume 151: Neuroanatomy of the Oculomotor System, by J.A. Büttner-Ennever (Ed.) – 2006, ISBN 0-444-51696-4.
Volume 152: Autonomic Dysfunction after Spinal Cord Injury, by L.C. Weaver and C. Polosa (Eds.) – 2006, ISBN 0-444-51925-4.
Volume 153: Hypothalamic Integration of Energy Metabolism, by A. Kalsbeek, E. Fliers, M.A. Hofman, D.F. Swaab, E.J.W. Van
Someren and R. M. Buijs (Eds.) – 2006, ISBN 978-0-444-52261-0.
Volume 154: Visual Perception, Part 1, Fundamentals of Vision: Low and Mid-Level Processes in Perception, by S. Martinez-Conde,
S.L. Macknik, L.M. Martinez, J.M. Alonso and P.U. Tse (Eds.) – 2006, ISBN 978-0-444-52966-4.
Volume 155: Visual Perception, Part 2, Fundamentals of Awareness, Multi-Sensory Integration and High-Order Perception, by
S. Martinez-Conde, S.L. Macknik, L.M. Martinez, J.M. Alonso and P.U. Tse (Eds.) – 2006, ISBN 978-0-444-51927-6.
Volume 156: Understanding Emotions, by S. Anders, G. Ende, M. Junghofer, J. Kissler and D. Wildgruber (Eds.) – 2006, ISBN 978-0-
444-52182-8.
Volume 157: Reprogramming of the Brain, by A.R. Møller (Ed.) – 2006, ISBN 978-0-444-51602-2.
Volume 158: Functional Genomics and Proteomics in the Clinical Neurosciences, by S.E. Hemby and S. Bahn (Eds.) – 2006, ISBN 978-
0-444-51853-8.
Volume 159: Event-Related Dynamics of Brain Oscillations, by C. Neuper and W. Klimesch (Eds.) – 2006, ISBN 978-0-444-52183-5
Volume 160: GABA and the Basal Ganglia: From Molecules to Systems, by J.M. Tepper, E.D. Abercrombie and J.P. Bolam
(Eds.) – 2007, ISBN 978-0-444-52184-2
PROGRESS IN BRAIN RESEARCH

VOLUME 161

NEUROTRAUMA:
NEW INSIGHTS INTO PATHOLOGY AND
TREATMENT

EDITED BY

JOHN T. WEBER
Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands
and
School of Pharmacy, Memorial University of Newfoundland, St. John’s, NL, Canada

ANDREW I.R. MAAS


Department of Neurosurgery, Erasmus Medical Centre, Rotterdam, The Netherlands

AMSTERDAM – BOSTON – HEIDELBERG – LONDON – NEW YORK – OXFORD


PARIS – SAN DIEGO – SAN FRANCISCO – SINGAPORE – SYDNEY – TOKYO
2007
Elsevier
Radarweg 29, PO Box 211, 1000 AE Amsterdam, The Netherlands
Linacre House, Jordan Hill, Oxford OX2 8DP, UK

First edition 2007

Copyright r 2007 Elsevier B.V. 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 electronic, mechanical, photocopying,
recording or otherwise without the prior written permission of the publisher

Permissions may be sought directly from Elsevier’s Science & Technology Rights
Department in Oxford, UK: phone (+44) (0) 1865 843830; fax (+44) (0) 1865 853333;
e-mail: permissions@elsevier.com. Alternatively you can submit your request online by
visiting the Elsevier web site at http://www.elsevier.com/locate/permissions, and selecting
Obtaining permission to use Elsevier material

Notice
No responsibility is assumed by the publisher for any injury and/or damage to persons
or property as a matter of products liability, negligence or otherwise, or from any use
or operation of any methods, products, instructions or ideas contained in the material
herein. Because of rapid advances in the medical sciences, in particular, independent
verification of diagnoses and drug dosages should be made

Library of Congress Cataloging-in-Publication Data


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

British Library Cataloguing in Publication Data

Neurotrauma: new insights into pathology and treatment. –


(Progress in brain research; v. 161)
1. Nervous system – Wounds and injuries
I. Weber, John II. Maas, Andrew I.R.
617.40 8044

ISBN-13: 9780444530172

ISBN: 978-0-444-53017-2 (this volume)


ISSN: 0079-6123 (Series)

For information on all Elsevier publications


visit our website at books.elsevier.com

Printed and bound in The Netherlands

07 08 09 10 11 10 9 8 7 6 5 4 3 2 1
List of Contributors

J. Ai, St. Michael’s Hospital, Trauma Research, Toronto, ON M5B 1W8, Canada
G.J. Amelink, Rudolf Magnus Institute of Neuroscience, Department of Neurosurgery, University Medical
Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
A.J. Baker, St. Michael’s Hospital, Trauma Research, Toronto, ON M5B 1W8, Canada
D.C. Baptiste, Division of Cellular and Molecular Biology, Toronto Western Research Institute and
Krembil Neuroscience Centre, Toronto Western Hospital, 12th Floor Room 407, McLaughlin Pavilion,
399 Bathurst Street, Toronto, ON M5 T 2S8, Canada
M.D. Baumann, Department of Chemical Engineering and Applied Chemistry, University of Toronto,
Terrence Donnelly Centre for Cellular and Biomolecular Research, 164 College Street, Toronto, ON
M5S 3G9, Canada
S.J. Bernstein, Department of Bioengineering, University of Pennsylvania, 3320 Smith Walk, Philadelphia,
PA 19104-6392, USA
N. Biasca, Clinic of Orthopaedic, Sports Medicine and Traumatology, Department of Surgery, Spital
Oberengadin, CH-7503 Samedan/St. Moritz, Switzerland
O. Bloch, Department of Neurological Surgery, University of California, San Francisco, CA, USA
and Brain and Spinal Injury Center, University of California, 1001 Potrero Avenue, Room 101, San
Francisco, CA 94110, USA
H.M. Bramlett, The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of
Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL 33136, USA
M. Buchfelder, Department of Neurosurgery, Friedrich-Alexander-University, Erlangen-Nuremberg,
Schwabachanlage 6, D-91054 Erlangen, Germany
A. Buki, Department of Neurosurgery, Medical Faculty, Pécs University, Rét St. 2, Pécs, H-7624, Hungary
P. Bukovics, Department of Neurosurgery, University Medical School, Pécs University, Rét St. 2, Pécs,
H-7624, Hungary
M.R. Bullock, Department of Neurosurgery, Virginia Commonwealth University Medical Center,
Richmond, VA, USA
A.S. Cohen, Department of Pediatrics, University of Pennsylvania School of Medicine and Division of
Neurology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104-4318, USA
D.K. Cullen, Neural Injury Biomechanics and Repair Laboratory, Wallace H. Coulter Department of
Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive,
Atlanta, GA 30332-0535, USA
E. Czeiter, Department of Neurosurgery, University Medical School, Pécs University, Rét St. 2, Pécs,
H-7624, Hungary
W.D. Dietrich, The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of
Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL 33136, USA
T. Doczi, Department of Neurosurgery, University Medical School, Pécs University, Rét St. 2, Pécs,
H-7624, Hungary
J.J. Donkin, Discipline of Pathology, Level 3, Medical School North, The University of Adelaide,
Adelaide, SA 5005, Australia

v
vi

A.-C. Duhaime, Pediatric Neurosurgery, Children’s Hospital at Dartmouth, Dartmouth Hitchcock Medi-
cal Center, Lebanon, NH 03756, USA
S. Durham, Pediatric Neurosurgery, Children’s Hospital at Dartmouth, Dartmouth Hitchcock Medical
Center, Lebanon, NH 03756, USA
O. Farkas, Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia
Commonwealth University, 1101 E. Marshall St., 12-048 P.O. Box 980709, Richmond, VA 23298, USA
M.G. Fehlings, Division of Cellular and Molecular Biology, Toronto Western Research Institute and
Krembil Neuroscience Centre, Toronto Western Research Institute, 4th floor West Wing Room 449, 399
Bathurst Street, Toronto, ON M5 T 2S8, Canada
C.L. Floyd, Department of Physical Medicine and Rehabilitation, Center for Glial Biology in Medicine,
547 Spain Rehabilitation Center, University of Alabama at Birmingham, Birmingham, AL 35249, USA
B.F. Fuller, Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research at the
McKnight Brain Institute of the University of Florida, PO Box 100256, Gainesville, FL 32610, USA
D.M. Geddes-Klein, Department of Bioengineering, University of Pennsylvania, 3320 Smith Walk,
Philadelphia, PA 19104-6392, USA
P.B. Goforth, Department of Pharmacology and Toxicology, VCU School of Medicine, Virginia Common-
wealth University, Richmond, VA, USA
M.S. Grady, Department of Neurosurgery, University of Pennsylvania School of Medicine, Philadelphia,
PA, USA
B.C. Hains, Department of Neurology and Center for Neuroscience and Regeneration Research, LCI-707,
Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA and Rehabilitation
Research Center, VA Connecticut Healthcare System, West Haven, CT 06516, USA
I.K. Haitsma, Department of Neurosurgery, Erasmus Medical Center, ’s-Gravendijkwal 230, 3015 CE
Rotterdam, The Netherlands
I. Hassan, Discipline of Pathology, Level 3, Medical School North, The University of Adelaide, Adelaide,
SA 5005, Australia
R.L. Hayes, Department of Neuroscience, Center for Traumatic Brain Injury Studies at the McKnight
Brain Institute of the University of Florida, PO Box 100244, Gainesville, FL 32610, USA
F. Hesse, Department of Neurosurgery, Friedrich-Alexander-University, Erlangen-Nuremberg, Schwa-
bachanlage 6, D-91054 Erlangen, Germany
M. Horowitz, Laboratory of Environmental Physiology, Faculty of Dental Medicine, Hebrew University
of Jerusalem, Jerusalem 91120, Israel
P. Jendelova, Institute of Experimental Medicine ASCR, Videnska 1083, 14220 Prague 4, Czech Republic
C. Kang, Department of Chemical Engineering and Applied Chemistry, University of Toronto, Terrence
Donnelly Centre for Cellular and Biomolecular Research, 164 College Street, Toronto, ON M5S 3G9,
Canada
Y. Katayama, Department of Neurological Surgery, Nihon University School of Medicine, 30-1 Oyaguchi
Kamimachi, Itabashi-ku, Tokyo 173-8610, Japan
T. Kawamata, Department of Neurological Surgery, Nihon University School of Medicine, 30-1 Oyaguchi
Kamimachi, Itabashi-ku, Tokyo 173-8610, Japan
A. Kleindienst, Department of Neurosurgery, Friedrich-Alexander-University, Erlangen-Nuremberg,
Schwabachanlage 6, D-91054 Erlangen, Germany
F.H. Kobeissy, Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research at the
McKnight Brain Institute of the University of Florida, PO Box 100256, Gainesville, FL 32610, USA
J.D. Kocsis, Yale University School of Medicine, Neuroscience Research Center (127A), VA Connecticut
Health Care System, West Haven, CT 06516, USA
E.J.O. Kompanje, Department of Intensive Care, Erasmus MC University Medical Center Rotterdam,
’s-Gravendijkwal 230, P.O. Box 2040, 3015 CE Rotterdam, The Netherlands
vii

E. Kovesdi, Department of Neurosurgery, University Medical School, Pécs University, Rét St. 2, Pécs,
H-7624, Hungary
K.L. Lankford, Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West
Haven, CT 06516, USA
M.C. LaPlaca, Neural Injury Biomechanics and Repair Laboratory, Wallace H. Coulter Department of
Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive,
Atlanta, GA 30332-0535, USA
B. Li, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing,
P.R. of China
M.C. Liu, Department of Neuroscience, Center for Traumatic Brain Injury Studies at the McKnight Brain
Institute of the University of Florida, PO Box 100244, Gainesville, FL 32610, USA
N. Lukáčová, Institute of Neurobiology, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovak
Republic
B.G. Lyeth, Department of Neurological Surgery, University of California, 1515 Newton Court, One
Shields Avenue, Davis, CA 95816, USA
A.I.R. Maas, Department of Neurosurgery, Erasmus Medical Center, ’s-Gravendijkwal 230, 3015 CE
Rotterdam, The Netherlands
G.T. Manley, Department of Neurosurgery, University of California, 1001 Potrero Avenue, Room 101,
San Francisco, CA 94110, USA
J. Maršala, Institute of Neurobiology, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovak
Republic
W.L. Maxwell, Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
D.F. Meaney, Departments of Bioengineering and Neurosurgery, University of Pennsylvania, 3320 Smith
Walk, Philadelphia, PA 19104-6392, USA
M. Mesfin, Department of Bioengineering, University of Pennsylvania, 3320 Smith Walk, Philadelphia, PA
19104-6392, USA
W.J. Miller, Department of Bioengineering, University of Pennsylvania, 3320 Smith Walk, Philadelphia,
PA 19104-6392, USA
J.P. Muizelaar, Department of Neurosurgery, University of California at Davis Medical Center, 4860 Y
Street, Suite 3740, Sacramento, CA 95817, USA
M.W. Oli, Banyan Biomarkers Inc., 12085 Research Drive, Alachua, FL 32615, USA
J. Orendáčová, Institute of Neurobiology, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovak
Republic
A.K. Ottens, Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research at the
McKnight Brain Institute of the University of Florida, PO Box 100256, Gainesville, FL 32610, USA
J. Pal, Neurobiology Research Group of the Hungarian Academy of Sciences, University Medical School,
Pécs University, Rét St. 2, Pécs, H-7624, Hungary
E. Park, St. Michael’s Hospital, Trauma Research, Toronto, ON M5B 1W8, Canada
B.J. Pfister, Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ,
USA
N. Plesnila, Laboratory of Experimental Neurosurgery, Department of Neurosurgery and Institute
for Surgical Research, University of Munich Medical Center, Grosshadern, Marchioninistr 15, 81377
Munich, Germany
P. Poon, Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly
Center for Cellular and Biomolecular Research, Toronto, ON M5S 3E1, Canada
J.T. Povlishock, Department of Anatomy and Neurobiology, Medical College of Virginia Campus,
Virginia Commonwealth University, 1101 E. Marshall St., 12-048 Sanger Hall, P.O. Box 980709,
Richmond, VA 23298, USA
viii

G.R. Prado, Neural Injury Biomechanics and Repair Laboratory, Wallace H. Coulter Department
of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive,
Atlanta, GA 30332-0535, USA
C. Radtke, Department of Plastic, Hand and Reconstructive Surgery, Hannover Medical School, Hann-
over, Germany
D. Reglodi, Department of Anatomy, University Medical School, Pécs University, Szigeti u. 12, Pécs,
H-7624, Hungary
P. Reilly, Department of Neurosurgery, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
M. Sasaki, Department of Neurology and Center for Neuroscience and Regeneration Research, Yale
University School of Medicine, New Haven, CT 06510, USA
L.S. Satin, Department of Pharmacology and Toxicology, VCU School of Medicine, Virginia Common-
wealth University, Richmond, VA, USA
E. Schwarzbach, Department of Pharmacology, University of Pennsylvania School of Medicine,
Philadelphia, PA, USA
N.A. Shein, Department of Pharmacology, School of Pharmacy, Hebrew University of Jerusalem,
Jerusalem 91120, Israel
E. Shohami, Department of Pharmacology, School of Pharmacy, Hebrew University of Jerusalem,
Jerusalem, 91120, Israel
M.S. Shoichet, Department of Chemical Engineering and Applied Chemistry, University of Toronto,
Terrence Donnelly Centre for Cellular and Biomolecular Research, 160 College Street, Room 514,
Toronto, ON M5S 3E1, Canada
C.M. Simon, Neural Injury Biomechanics and Repair Laboratory, Wallace H. Coulter Department of
Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive,
Atlanta, GA 30332-0535, USA
P. Singh, Department of Bioengineering, University of Pennsylvania, 3320 Smith Walk, Philadelphia, PA
19104-6392, USA
F.J.A. Slieker, Department of Neurosurgery, Erasmus MC University Medical Center Rotterdam,
’s-Gravendijkwal 230, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
J.M. Spaethling, Department of Bioengineering, University of Pennsylvania, 3320 Smith Walk, Philadel-
phia, PA 19104-6392, USA
D.G. Stein, Department of Emergency Medicine, Emory University School of Medicine, Brain Research
Laboratory, Suite 5100, 1365B Clifton Road N.E., Atlanta, GA 30322, USA
N. Stocchetti, Ospedale Policlinico IRCCS, Milan University, Milan, Italy
E. Sykova, Institute of Experimental Medicine ASCR, EU centre of Excellence, Videnska 1083, 14220
Prague 4, Czech Republic
D. Szellar, Department of Neurosurgery, University Medical School, Pécs University, Rét St. 2, Pécs,
H-7624, Hungary
A. Tamas, Department of Anatomy, University Medical School, Pécs University, Szigeti u. 12, Pécs,
H-7624, Hungary
C.H. Tator, Krembil Neuroscience Centre, Toronto Western Research Institute and Department of
Surgery, University of Toronto, 399 Bathurst Street, Toronto, ON M5 T 2S8, Canada
E. Thornton, Discipline of Pathology, Level 3, Medical School North, The University of Adelaide,
Adelaide, SA 5005, Australia
R.J. Turner, Discipline of Pathology, Level 3, Medical School North, The University of Adelaide,
Adelaide, SA 5005, Australia
C. Van Den Heuvel, Discipline of Pathology, Level 3, Medical School North, The University of Adelaide,
Adelaide, SA 5005, Australia
ix

I. Vanický, Institute of Neurobiology, Slovak Academy of Sciences, Šoltésovej 4, 040 01 Košice, Slovak
Republic
B.H. Verweij, Rudolf Magnus Institute of Neuroscience, Department of Neurosurgery, University Medical
Center Utrech, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
R. Vink, Discipline of Pathology, Level 3, Medical School North, The University of Adelaide, Adelaide,
SA 5005, Australia
C.R. Von Reyn, Department of Bioengineering, University of Pennsylvania, 3320 Smith Walk, Philadel-
phia, PA 19104-6392, USA
K.K.W. Wang, Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research at the
McKnight Brain Institute of the University of Florida, PO Box 100256, Gainesville, FL 32610, USA
S.G. Waxman, Department of Neurology and Center for Neuroscience and Regeneration Research, LCI-
707, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA and Rehabilitation
Research Center, VA Connecticut Healthcare System, West Haven, CT 06516, USA
J.T. Weber, Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands and
School of Pharmacy, Memorial University of Newfoundland, Health Sciences Centre, 300 Prince Philip
Drive, St. John’s, NL A1B 3V6, Canada
X. Yao, Department of Neurological Surgery, University of California, San Francisco, CA, USA and Brain
and Spinal Injury Center, 1001 Potrero Avenue, Room 101, San Francisco, CA 94110, USA
Z. Zador, Department of Neurological Surgery, University of California, San Francisco, CA, USA and
Brain and Spinal Injury Center, 1001 Potrero Avenue, Room 101, San Francisco, CA 94110, USA
This page intentionally left blank
Preface

This current volume of Progress in Brain Research, entitled ‘‘Neurotrauma: New insights into pathology and
treatment’’ is a compilation of chapters on major topics which were discussed at the 8th International
Neurotrauma Symposium held in Rotterdam, The Netherlands in May of 2006. Neurotrauma, which
encompasses both traumatic brain injury (TBI) and spinal cord injury (SCI), is the leading cause of death
and disability in young adults, and the incidence in older patients is increasing. As such, neurotrauma is a
field in medicine with one of the highest unmet needs. Concentrated, focused and multidisciplinary efforts
are required to combat this important disease. Therefore, one of the major goals of the symposium was to
stimulate cross-talk between basic researchers and clinicians, as well as between individuals in the fields of
TBI and SCI. We feel that TBI and SCI researchers have much to learn from one another in their approach
to treating these disorders. In addition, researchers have an obligation to design and conduct clinically
relevant research, just as clinicians have an obligation to learn about the latest approaches to treating
neurotrauma. Exciting findings from basic research open opportunities for improving treatment results,
and this volume represents a unique and comprehensive overview of the latest findings and insights on
translational research in neurotrauma.
Experts who delivered presentations at the symposium have submitted the majority of chapters in this
volume. In general, these individuals were selected because the topics that they, and their research groups,
are currently pursuing are pertinent for understanding the pathology and treatment of neurotrauma. We
are grateful also to other experts, who were not in attendance in Rotterdam, for writing chapters in specific
areas of neurotrauma, which we felt were needed to make this a more comprehensive volume. The result,
we think, is a report on some of the most relevant and up-to-date topics in the neurotrauma field. Also, we
are proud that the author base is a true representation of the international neurotrauma society (INTS),
with contributors from Western and Eastern Europe, The United States, Canada, Asia and Australia. The
global problem of neurotrauma can best be served by a worldwide representation of neurotrauma
specialists combining and exchanging expertise.
We are extremely grateful to all of the participants, assistants and sponsors who made the 8th Inter-
national Neurotrauma Symposium a resounding success, and are especially gracious to all of the authors
who have taken the time to contribute to this timely volume. In addition, we would like to apologize to
some individuals or groups who may feel excluded, which was primarily due to space limitations. This
exclusion is by no means intentional, and we have no doubt that new and additional topics, which are not
covered in the current volume, will soon be represented elsewhere on the international stage.

John T. Weber
Andrew I.R. Maas

xi
This page intentionally left blank
Contents

List of Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v

Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi

Section I. Introduction

1. The impact of neurotrauma on society: an international perspective


P. Reilly (Adelaide, South Australia) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3

Section II. Biomechanics of Injury

2. CNS injury biomechanics and experimental models


M.C. LaPlaca, C.M. Simon, G.R. Prado and D.K. Cullen (Atlanta, GA, USA). . . . 13

3. Linking impact to cellular and molecular sequelae of CNS injury: modelling in vivo complexity
with in vitro simplicity
J.M. Spaethling, D.M. Geddes-Klein, W.J. Miller, C.R. von Reyn, P. Singh, M. Mesfin,
S.J. Bernstein and D.F. Meaney (Philadelphia, PA, USA) . . . . . . . . . . . . . . . . . . . 27

Section III. Pathological Mechanisms of Injury

4. Cellular and subcellular change evoked by diffuse traumatic brain injury: a complex web of
change extending far beyond focal damage
O. Farkas and J.T. Povlishock (Richmond, VA, USA) . . . . . . . . . . . . . . . . . . . . . 43

5. Astroglia: Important mediators of traumatic brain injury


C.L. Floyd and B.G. Lyeth (Davis, CA, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . 61

6. Rescuing neurons and glia: is inhibition of apoptosis useful?


E. Kovesdi, E. Czeiter, A. Tamas, D. Reglodi, D. Szellar, J. Pal, P. Bukovics,
T. Doczi and A. Buki (Pécs, Hungary) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 81

7. Substance P in traumatic brain injury


J.J. Donkin, R.J. Turner, I. Hassan and R. Vink (Adelaide, South Australia) . . . . . 97

xiii
xiv

8. Current concepts of cerebral oxygen transport and energy metabolism after severe traumatic
brain injury
B.H. Verweij, G.J. Amelink and J.P. Muizelaar (Utrecht, The Netherlands and
Sacramento, CA, USA). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 111

9. Progressive damage after brain and spinal cord injury: pathomechanisms and treatment strategies
H.M. Bramlett and W.D. Dietrich (Miami, FL, USA) . . . . . . . . . . . . . . . . . . . . . . 125

10. Injury-induced alterations in CNS electrophysiology


A.S. Cohen, B. J. Pfister, E. Schwarzbach, M.S. Grady, P.B. Goforth and
L.S. Satin (Philadelphia, PA, Newark, NJ and Richmond, VA, USA) . . . . . . . . . . 143

11. Traumatic injury of the spinal cord and nitric oxide


J. Mars̆ala, J. Orendáčová, N. Lukáčová and I. Vanický (Košice, Slovak Republic) 171

12. Aquaporins: role in cerebral edema and brain water balance


Z. Zador, O. Bloch, X. Yao and G.T. Manley (San Francisco, CA, USA) . . . . . . . 185

13. Sodium channel expression and the molecular pathophysiology of pain after SCI
B.C. Hains and S.G. Waxman (West Haven, CT, USA) . . . . . . . . . . . . . . . . . . . . 195

Section IV. Novel Aspects of Clinical Research in CNS Injury

14. Monitoring cerebral oxygenation in traumatic brain injury


I.K. Haitsma and A.I.R. Maas (Rotterdam, The Netherlands) . . . . . . . . . . . . . . . 207

15. Update on the treatment of spinal cord injury


D.C. Baptiste and M.G. Fehlings (Toronto, ON, Canada) . . . . . . . . . . . . . . . . . . 217

16. Cerebral contusion: a role for lesion progression


T. Kawamata and Y. Katayama (Tokyo, Japan) . . . . . . . . . . . . . . . . . . . . . . . . . 235

17. Ethical implications of time frames in a randomized controlled trial in acute severe traumatic
brain injury
E.J.O. Kompanje, A.I.R. Maas, F.J.A. Slieker and N. Stocchetti
(Rotterdam, The Netherlands and Milan, Italy). . . . . . . . . . . . . . . . . . . . . . . . . . 243

Section V. Emerging Topics in CNS Trauma

18. Experimental models of repetitive brain injuries


J.T. Weber (Rotterdam, The Netherlands) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 253

19. Minor traumatic brain injury in sports: a review in order to prevent neurological sequelae
N. Biasca and W.L. Maxwell (Samedan/St. Moritz, Switzerland
and Glasgow, UK) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263
xv

20. Traumatic brain injury in infants: the phenomenon of subdural hemorrhage with
hemispheric hypodensity (‘‘Big Black Brain’’)
A.-C. Duhaime and S. Durham (Lebanon, NH, USA) . . . . . . . . . . . . . . . . . . . . . 293

21. Traumatic brain injury and Alzheimer’s disease: a review


C. Van Den Heuvel, E. Thornton and R. Vink (Adelaide, Australia). . . . . . . . . . . 303

22. The neurotrophic protein S100B: value as a marker of brain damage and possible therapeutic
implications
A. Kleindienst, F. Hesse, M.R. Bullock and M. Buchfelder (Erlangen-Nuremberg,
Germany and Richmond, VA, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 317

23. Cerebellar injury: clinical relevance and potential in traumatic brain injury research
E. Park, J. Ai and A.J. Baker (Toronto, ON, Canada) . . . . . . . . . . . . . . . . . . . . . 327

24. Sex differences in brain damage and recovery of function: experimental and clinical findings
D.G. Stein (Atlanta, GA, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 339

25. Heat acclimation: a unique model of physiologically mediated global preconditioning


against traumatic brain injury
N.A. Shein, M. Horowitz and E. Shohami (Jerusalem, Israel). . . . . . . . . . . . . . . . 353

Section VI. The Future of Neurotrauma: Developing Novel Treatment Strategies

26. In vivo tracking of stem cells in brain and spinal cord injury
E. Sykova and P. Jendelova (Prague, Czech Republic) . . . . . . . . . . . . . . . . . . . . . 367

27. Intrathecal drug delivery strategy is safe and efficacious for localized delivery to the spinal cord
M.S. Shoichet, C.H. Tator, P. Poon, C. Kang and M.D. Baumann
(Toronto, ON, Canada). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 385

28. Decompression craniectomy after traumatic brain injury: recent experimental results
N. Plesnila (Munich, Germany) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 393

29. Novel neuroproteomic approaches to studying traumatic brain injury


A.K. Ottens, F.H. Kobeissy, B.F. Fuller, M.C. Liu, M.W. Oli,
R.L. Hayes and K.K.W. Wang (Gainesville and Alachua, FL, USA) . . . . . . . . . . 401

30. Remyelination of the injured spinal cord


M. Sasaki, B. Li, K.L. Lankford, C. Radtke and J.D. Kocsis (New Haven, CT, USA,
Chongqing, People’s Republic of China and Hannover, Germany) . . . . . . . . . . . . 419

Subject Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 435


This page intentionally left blank
SECTION I

Introduction
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 1

The impact of neurotrauma on society: an


international perspective

Peter Reilly

Department of Neurosurgery, Royal Adelaide Hospital, Adelaide, South Australia 5000

Abstract: Neurotrauma, in many countries and particularly in the younger age group kills more people
than AIDS or cancer but unlike these diseases the causes are known and it is preventable. The costs to
communities in terms of suffering and economics are enormous. The common causes are road traffic
accidents, falls and violence. Neurotrauma affects particularly the developing world where it consumes
already over stretched health resources. In the developed world steps to reduce the incidence of neuro-
trauma and to treat the victims have had some effect nevertheless it stills remains an endemic problem
which does not receive the public awareness or the political support it deserves. For the victims there is
general agreement on the principles of clinical management but often difficulties in applying early and
effective care in countries with the greatest need because of shortage of facilities and expertise. To reduce
the overall burden of neurotrauma demands actions which extend from the political to basic patient care.
There have been remarkable advances in the understanding of acute brain and spinal cord injury and
encouraging possibilities for effective neuroprotection, repair and regeneration but in the broader context
prevention of neurotrauma is the urgent imperative. In this endeavour the neuroscientist has knowledge
which informs and encourages policy makers to take the steps necessary to reduce injury. These steps
require political will and community support for hard decisions which impact on the way people conduct
their daily lives. The WHO predicts that unless there are changes in present policies and if there are no
additional road safety countermeasures put in place, there will be a major increase in road traffic fatalities
over the next 20 years and beyond (World Health Organisation. (2004). www.who.int).

Keywords: epidemiology; prevention; brain and spinal injury statistics

Introduction firearms that are easily available; for poor indus-


trial safety standards and for sports such as boxing
It is a sad paradox that one of the greatest health which aim to inflict neurotrauma or other sports
problems of this age is largely avoidable — neuro- which regularly do so.
trauma of the brain and spinal cord is caused by Some accidents are unavoidable and unforesee-
injury inflicted by misadventure, violence or care- able. Much of neurotrauma is all too predictable
lessness. The principle causes of neurotrauma and avoidable.
reflect choices by individuals and by society for In this introduction to the Proceedings of the
fast, freely available and personal transport; for 8th International Neurotrauma Symposium I wish
to consider some global aspects of neurotrauma
and the role of the International Neurotrauma
Corresponding author. Tel.: +61-8-8222-5232; Society (INTS) in the overall effort to reduce the
Fax: +61-8-8222-5668; E-mail: p.reilly@adelaide.edu.au impact of neurotrauma on societies.

DOI: 10.1016/S0079-6123(06)61001-7 3
4

Neurotrauma as a global problem These wide ranges no doubt reflect a combina-


tion of actual differences, varying definitions of
The global incidence of traumatic brain injury injury, different populations of inclusion and sam-
(TBI) is generally reported as 200 in 100,000 with pling errors (Bruns and Hauser, 2003).
a mortality of 20 per 100,000. Incidences reported A number of studies indicate important regional
from different countries range from 91 to 430 per variations. A higher mortality for patients injured
100,000 with mortalities of 9–89 per 100,000 in rural areas compared with urban areas has been
(Fearnside and Simpson, 2005). recorded in Australia and Taiwan (Chiu et al.,
Despite awareness of the causes and of the eco- 1995, 1997; Hillier et al., 1997).
nomic and human costs of neurotrauma the inci- Injury and mortality rates are bimodal affecting
dence remains depressingly high and in developing particularly the under 25 and over 65 years.
countries continues to rise. One of the difficulties in In the UK the TBI incidence of 10:100,000 rep-
describing accurately the global impact of neuro- resents 1% of all deaths but 20% of deaths in age
trauma is the variable way in which statistics are range 5–45 years. As in other developed countries,
recorded. Even in countries with well-developed TBI is the common cause of death in this age range.
data collection systems, information on the full
impact of head and spinal cord injury is difficult to
obtain. Mortality figures are often based on A&E The causes of neurotrauma
admissions or hospital separations and do not in-
clude pre hospital deaths which account for 50% The main causes of neurotrauma, transport acci-
of road trauma deaths. dents, falls and gunshot wounds, reflect societal
The incidence of spinal cord injury ranged from behaviour.
14.5 to 57.8 cases per million in a review of reported From the beginning motorised transport has in-
studies; the low figure of 14.5 from Australia did flicted injury to a degree which would undoubtedly
not include deaths at the scene of trauma (Ackery have shocked its inventors.
et al., 2004). By the 1930s the motor car ‘‘had emerged as the
Death and serious injury can be measured with most persistent killer in the western world’’ (Gilbert,
reasonable accuracy but the greater numbers of 1997). It is estimated that by 1997 25 million people
mild and moderate head injuries are less likely to had died on the roads (Odero et al., 1997). In 2002
be encompassed, yet they represent a major com- the global death rate from road traffic accidents was
ponent of the total burden of neurotrauma. Chang- 1.2 million and between 20 and 50 million people
ing practices in hospital admissions also affect are estimated to be injured or disabled each year
comparisons between studies. The greater use of (World Health Organisation, 2000, 2004). Of road
CT scanning has lead to fewer hospital admissions, trauma deaths it is estimated that 1/3–1/2 are due to
increasing the proportion of severe head injury ad- brain injury. Vehicular accidents are also the major
missions and reducing the mild and moderate head cause of spinal cord injury worldwide (Ackery et al.,
injury admissions (Thurman and Guerrero, 1999). 2004).
A true measure of incidence and prevalence of ne- The cost of road traffic injuries to society is
urotrauma would require that all head and spinal estimated as 2% of a country’s gross domestic
cord injuries be included. product (World Health Organisation, 2004).
In fact there is no fully comprehensive study of Global figures encompass important regional
the incidence of TBI in a defined population variations and trends (Soderlund and Zwi, 1995)
(Fearnside and Simpson, 2005). (Table 1). Ninety percent of road traffic accident
Similar problems affect spinal injury statistics deaths occur in low and middle income countries
(Ackery et al., 2004). and at nearly twice the population adjusted rate of
Despite these difficulties and limitations accurate high-income countries. Road traffic fatalities per
data are essential in order to develop prevention 100,000 population in the year 2000 ranged from
and management strategies. 28.3 in the African region to 5.9 in Great Britain
5

Table 1. Mechanisms of TBI

RTA Falls Violence Sport Occupation

Australia (Tate et al., 1998) 40 21 8 25


India (Gururaj, 2002) 45–60 20–30 10–20
China (Wang et al., 1986) 32 22 1 15 24
USA (CDC, 1999) 49 26 17

Note: Numbers are percentages of trauma victims in each of the respective categories. RTA, road traffic accidents.

Fig. 1. Road traffic fatality trends in three high-income countries (Australia, United Kingdom, United States of America). Sources:
Transport Safety Bureau, Australia; Department of Transport, United Kingdom; Fatality Analysis Reporting System, United States of
America (World Health Organisation, 2004).

(World Health Organisation, 2004). Furthermore (Chiu et al., 1995). This was considered to be
TBI from road traffic accidents has been falling in related to the rapidly increasing use of motor
high-income countries for some years. In Australia cycles, the most dangerous form of road transport
there has been an average yearly decline in TBI in current use anywhere in the world, and an
of 5%, most due to an 8% decline in vehicle increasing component of traffic in developing
occupancy injuries (O’Connor and Cripps, 1999). countries (World Health Organisation, 2004).
Between 1968 and 1983 road traffic accident Other causes of neurotrauma also show marked
mortality declined by more than 20% in Europe regional variation and changing trends. In the USA
(Fig. 1) In contrast it increased by more than firearm injuries exceeded road traffic accidents for
150% in Asian countries and more than 200% in the first time in 1990 and this trend contrasted with
African countries (Soderlund, 1995) (Fig. 2). a fall in road traffic accident deaths (Sosin et al.,
Traffic type has important influences on injury 1995). Other trends in injury causation have been
patterns. In Beijing one third of traffic deaths oc- documented in several countries. A review of head
cur among bicyclists. In India pedestrians account injury mortality from 1987 to 2000 in Sweden
for 15–35% of injuries, 2-wheeler occupants showed a constant rate of injury over this period,
20–40% and bicyclists 5–15% (Gururaj, 2002). however a fall in transport-related injury was bal-
Taiwan had a high rate of TBI from road traffic anced by an increase in falls below the age of 25 and
accidents reported in 1991 to be 89 per 100,000 over the age of 65 (Kleiven et al., 2003). In the UK
6

Fig. 2. Global and regional road fatality trends, 1987–1995. Data are displayed according to regional classification of TRL Ltd.,
United Kingdom. The World Health Organisation, 2004 document from which this comes states that reproduced with permission of
the authors namely, Jacobs G, Aeron-Thomas A, Astrop A. Estimating Global Road Fatalities. Crowthorne, Transport Research
Laboratory, 2000 (TRL Report 445).

there were regional differences between the inci- figures indicate a possible treatment rate of moder-
dence of road traffic accident versus falls and of ate head injury of 100–300 per 100,000 but the
alcohol-related accidents (Kay and Teasdale, 2001). actual rate may be greater than 600 per 100,000
Surveys from different countries also indicate (Cassidy et al., 2004). Thornhill et al. (2000)
variation in the causation of spinal cord injury reported that following mild head injury 51% had
between countries and over time. In most countries continuing symptoms and after moderate head in-
vehicular accidents are the leading cause of spinal jury 54%. Following mild head injury (GCS 13–15)
cord injury in the younger age group but, as with 79% show persistent headache, 59% memory prob-
head injury, this figure is falling while the incidence lems and 34% remained unemployed. Guerrero
of injury due to falls in the older age group is et al. (2000) found that at 1 year 15% still had
increasing (Cripps, 2006). Falls and violence are symptoms. This represents an enormous and often
more prevalent in developing countries and in the under treated group of potential disabilities.
lower economic strata of developed countries A pathophysiological basis has been identified
(Ackery et al., 2004). for continuing symptoms (Blumbergs et al., 1994;
Bigler, 2001). The ensuing neuropsychological
effects and the value of psychological therapy have
The community costs been documented (Ponsford et al., 2000; Carroll
et al., 2004; Cassidy et al., 2004; Ponsford, 2005).
Most studies of TBI focus on severe injury. Ninety
percent of patients presenting to A&E with head
injury in the UK had minor, 5% moderate and 5% Providing care
severe head injury. For every severe head injury
there were 17 mild or moderate injuries (Kay and Since the 1970s routine intracranial pressure
Teasdale, 2001). Most patients who suffer from mi- recording, the CT scan and Intensive Care Units
nor TBI make uneventful recoveries but many have focused treatment on severe head injury. The
suffer continuing, disabling symptoms. WHO general principles of management of brain and
7

spinal injury may be agreed upon but there are 9 months. There was a marked increase in crash
major difficulties in delivering care to patients in scene fatalities and in admissions of non helmeted
developing countries through shortage of neuro- crash survivors. It was concluded that helmet us-
surgeons, a lack of trained personal for primary age decreased fatalities by 20–40% (Bledsoe et al.,
care, particularly in rural areas, basic facilities and 2002). Other studies have supported the benefit of
transport systems for trauma victims. These pres- motor cycle helmets in reducing head and facial
sures are often compounded by the need to address injury (Wagle et al., 1993; Liu et al., 2003).
many other health problems with limited resources. Bicycle helmets have been mandatory in Australia
In such situations general surgeons may need to since 1990 and several studies have indicated their
be trained in acute neurosurgery. benefit (Attewell et al., 2001; Thompson et al.,
In Australia distance imposes special needs and 1999).
the general surgeons in Darwin, a city of 70,000 and There is great potential benefit for helmets in
2000 km from a neurosurgical centre, have man- developing countries where bicycle and increas-
aged acute neurosurgery for many years with sup- ingly motor bikes are a major cause of TBI.
port by neurosurgeons and teleradiology (Treacy Societal attitudes are clearly key to any injury
et al., 2005). To meet these needs protocols for on prevention campaign (Weinstein, 1987). Cars are
site management of neurotrauma by non neurosur- undoubtedly safer now than they were decades
geons have been developed (Neurosurgical Society ago. Standard safety ratings are accepted by most
of Australasia Guidelines, 2000). manufacturers, but they can be safer. It is often
remarked that safety does not sell compared with
appearance and performance.
What can be done? There appears to be an acceptance of the risk of
neurotrauma even though there must be few
Neurotrauma is a major public health challenge. families who do not have a victim of neuro-
Acknowledging that most TBI and spinal cord trauma of some degree. Those at highest risk have
injury is avoidable clearly emphasises the need for the least perception of risk (Gilk et al., 1999).
strategies to identify causes and prevent injury.
Motor vehicle accident rates have fallen in many
countries in response to public awareness cam- Community support for research
paigns and legislation (McDermott et al., 1996).
Each new legislated measure, for random alcohol Funding levels are a barometer of the value a so-
testing, speed cameras, seat belts or helmets, tends ciety puts on a particular field of research.
to be followed by a modest fall in accident rates The Brain Injury Association of America esti-
(Campbell, 1987; MacLennan et al., 2004). mates that TBI in the US costs the community
In response to the high number of motorcycle- $48.3 billion per year, $31.7 billion in hospitalisa-
related head injuries in Taiwan a concerted effort tion costs and $16.6 billion from fatal TBI. Spinal
was made to introduce motorcycle helmets from cord injury has been estimated to cost the US $9.7
1997. In the first year motorcycle-related head in- billion each year (Berkowitz, 1998). In 2006 the US
juries fell by 33%. There was a better than 90% National Institutes of Health, the major govern-
compliance rate with helmet wearing which was ment funding body, allocated $86 million for TBI
mandated by law (Chiu et al., 2000a, b). research and $87 million for spinal cord injury re-
Motorcycle helmets have now been introduced via search. Whether these are appropriate amounts
legislation in many countries including Australia. can be argued.
In the US the repeal of helmet legislation in In Australia the principle commonwealth funded
several states, provided a human experiment un- body, the National Health and Medical Research
likely to be approved by an ethics committee. Council provided $600,000 towards neurotrauma
In Arkansas repeal of the state helmet law in 1997 research in 2005–2006 from a total budget of $60
led to a fall in helmet usage from 97 to 52% over million, that is to say 1% of the research budget.
8

Researchers in neurotrauma in some states have providing an international forum for basic, preclini-
been more successful in gaining substantial grants cal and clinical research. It aims to encourage
from the state government third party insurers. young neuroscientists to pursue their interests and
One of the more innovative funding initiatives oc- in this way to foster research around the globe.
cured in Western Australia for 5 years fines raised Scientists have a role in neurotrauma prevention
by speed cameras amounting to $500,000 per year and treatment in every level. Scientific evidence
were allocated to trauma research. underpins effective political and social action. The
In the US and Australia the funds directed to- neuroscientist can play a vital role in informing
wards neurotrauma research appear small com- and stimulating policy makers towards action that
pared with the societal costs and in comparison will reduce the incidence of neurotrauma.
with the funds directed to other major diseases. The INTS plays a highly significant role in en-
Spinal research has profited greatly from patient couraging the worldwide search for better treat-
advocacy. A visit by Christopher Reeves to Sydney ments for victims of neurotrauma.
in 1999 lead to the NSW Premier’s Initiative re- It is important that the INTS liaises with other
search fund for spinal research. There are now neurotrauma organisations such as the WFNS,
moves to establish national platforms to coordinate National Neurotrauma Societies and regional
research and develop funds for spinal injury re- consortia so that the international neuroscience
search — important given the need to support high- community can use its wide experience and exper-
quality research in a relatively small population. tise to the best effect.

The challenge
References
Neurotrauma affects all communities, all age Ackery, A., Tator, C. and Krassioukov, A. (2004) A global
groups and walks of life. Despite understanding perspective on spinal cord injury epidemiology. J. Neuro-
of the causes and being able to treat victims better trauma, 21: 1355–1370.
it is an increasing global problem with important Attewell, R.G., Glase, K. and McFadden, M. (2001) Bicycle
helmet efficacy: a meta-analysis. Accid. Anal. Prev., 33:
regional variations. There is a lack of adequate
345–352.
comprehensive data of the incidence of neurotrau- Berkowitz, M., O’Leary, P., Kruse, D. and Harvey, C. (1998)
ma in any community, most of all in those com- Spinal cord injury: an analysis of medical and social costs.
munities which have the major burden of Demos Medical Publishing Inc., New York.
neurotrauma. Neurotrauma is a particular burden Bigler, E. (2001) The lesion(s) in traumatic brain injury:
implications for clinical neuropsychology. Arch. Clin.
on the developing countries which have the least
Neuropsychol., 16: 95–131.
capacity to manage it. Bledsoe, G.H., Schexnayder, S.M., Carey, M.J., Dobbins,
Reducing the incidence of neurotrauma can W.N., Gibson, W.D., Hindman, J.W., Collins, T., Wallace,
only occur by greater political action and the en- B.H., Cone, J.B. and Ferrer, T.J. (2002) The negative impact
gagement of societies in order to see neurotrauma of the repeal of the Arkansas motorcycle helmet law. J.
Trauma, 53: 1078–1087.
as avoidable.
Blumbergs, P.C., Scott, G. and Manavis, J. (1994) Staining of
amyloid precursor protein to study axonal injury in mild
head injury. Lancet, 344: 1055–1056.
The role of the INTS and neuroscience
Bruns Jr., J. and Hauser, W.A. (2003) The epidemiology of
traumatic brain injury: a review. Epilepsia, 44(Suppl 10):
The INTS comprises a worldwide body of experts 2–10.
in neurotrauma from clinical, non clinical and basic Campbell, B.J. (1987) Safety belt injury reduction related to
science backgrounds. Many are actively involved in crash severity and front seated position. J. Trauma, 27:
733–739.
acute neurotrauma care or working with victims of
Carroll, L.J., Cassidy, J.D. and Holm, L. (2004) Methodological
the delayed effects of traumatic brain or spinal cord issues and research recommendations for mild traumatic brain
injury or in injury prevention. The INTS aims to injury: the WHO Collaborating Centre Task Force on Mild
encourage research in all aspects of neurotrauma by Traumatic brain injury. J. Rehabil. Med., 43(Suppl): 113–135.
9

Cassidy, J.D., Carroll, L.J. and Peloso, P.M. (2004) Incidence, death in 137 road traffic fatalities in Victoria, Australia: an
risk factors and prevention of mild traumatic brain injury: overview. J. Trauma, 40: 520–535.
results of the WHO Collaborating Centre Task Force on Neurosurgical Society of Australasia Guidelines. (2000) The
Mild Traumatic brain injury. J. Rehabil. Med., 43(Suppl): Management of Acute Neurotrauma in Rural and Remote
26–28. Locations. A Set of Guidelines for the Care of Head and
Center for Disease Control and Prevention (CDC). (1999) Spinal Injuries. The Neurosurgical Society of Australasia,
Traumatic brain injury in the United States. A Report Royal Australasian College of Surgeons, Melbourne.
to Congress Current Data on Traumatic Brain Injury and O’Connor, P.J. and Cripps, R.A. (1999) Traumatic Brain Injury
Mortality. (TBI) Surveillance Issues. AIHW National Injury Surveil-
Chiu, W.T., Ho, Y.S. and Lee, Y.S. (2000a) Sharp decline of lance Unit, Flinders University Research Centre for Injury
injury mortality rate in a developing country. Am. J. Public Studies, Adelaide.
Health, 90(5): 793–796. Odero, W., Garner, P. and Zwi, A. (1997) Road traffic injuries
Chiu, W.T., Hung, C.C., Le, L.S., Lin, L.S., Shih, C.J. and in developing countries: a comprehensive review of epidemio-
LaPorte, R.E. (1997) Head injury in urban and rural logical studies. Trop. Med. Int. Health, 2: 445–460.
populations in a developing country. J. Clin. Neurosci., 4: Ponsford, J. (2005) Rehabilitation interventions after mild head
469–472. injury. Curr. Opin. Neurol., 18: 692–697.
Chiu, W.T., Hung, C.C. and Shih, C.J. (1995) Epidemiology Ponsford, J., Wilmott, C., Rothwell, A., Cameron, P., Kelly,
of head injury in rural Taiwan: a four year survey. J. Clin. A.M., Nelms, R., Curran, C. and Ng, K.T. (2000) Factors
Neurosci., 2: 210–215. influencing outcomes following mild traumatic brain injury
Chiu, W.T., Kuo, C.Y., Hung, C.C. and Chen, M. (2000b) The in adults. J. Int. Neuropsychol. Soc., 6: 568–597.
effect of the Taiwan motorcycle helmet use law on head Soderlund, N. and Zwi, A.B. (1995) Traffic related mortality in
injuries. Am. J. Public Health, 5(90): 793–796. industrialized and less developed countries. World Health
Cripps, R.A. (2006) Spinal Cord Injury, Australia 2003–04. Organisation Bull., 73: 175–182.
Australian Institute of Health and Welfare, Canberra. Sosin, D.M., Sniezek, J.E. and Waxweiler, R.J. (1995) Trends in
Fearnside, M.R. and Simpson, D.A. (2005) Epidemiology. death associated with traumatic brain injury, 1979 through
In: Reilly P.L. and Bullock R.B. (Eds.), Head Injury: 1992. JAMA, 273: 1778–1780.
Pathophysiology and Management (2nd ed). Hodder Tate, R.L., McDonald, S. and Lulham, J.M. (1998) Incidence
Arnold, London, pp. 3–25. of hospital-treated traumatic brain injury in an Australian
Gilbert, M. (1997) A History of the Twentieth Century, Vol. 1: community. Aust. N.Z. J. Public Health, 22: 419–423.
1900–1933. Harper Collins, London, 41p. Thompson, D.C., Rivara, F.P. and Thompson, R. (1999) Hel-
Gilk, D.C., Kronenfeld, J.J., Jackson, K. and Zhang, W. (1999) mets for preventing head and facial injuries in bicyclists. The
Comparison of traffic accident and chronic disease risk Cochrane Database of Systemic Reviews, Issue 4.
perceptions. Am. J. Health. Behav., 23: 198–209. Thornhill, S., Teasdale, G.M., Murray, G.D., McEwen, J.,
Guerrero, J.L., Thurman, D.J. and Sniezek, J.E. (2000) Roy, C.W. and Penny, K.I. (2000) Disability in young people
Emergency department visits associated with traumatic brain and adults one year after head injury: prospective cohort
injury: United States, 1995–1996. Brain Inj., 14: 181–186. study. BMJ, 320: 1631–1635.
Gururaj, G. (2002) Epidemiology of traumatic brain injuries: Thurman, D. and Guerrero, J. (1999) Trends in hospitalization
Indian scenario. Neurol. Res., 24: 24–28. associated with traumatic brain injury. JAMA, 282(10):
Hillier, S., Hillier, J. and Metzer, J. (1997) Epidemiology of 954–957.
traumatic brain injury in South Australia. Brain Inj., 11: Treacy, P.J., Reilly, P. and Brophy, B.P. (2005) Emergency
649–659. neurosurgery by general surgeons at a remote major hospital.
Kay, A. and Teasdale, G. (2001) Head injury in the United ANZ J. Surg., 75: 852–857.
Kingdom. World J. Surg., 25: 1210–1220. Wagle, V.G., Perkins, C. and Vallera, A. (1993) Is helmet use
Kleiven, S., Peloso, P.M. and von Holst, H. (2003) The epide- beneficial to motorcyclists? J. Trauma, 34: 120–122.
miology of head injuries in Sweden from 1987 to 2000. Inj. Wang, C.C., Schoenberg, B.S., Li, S., Yang, Y., Cheng, X. and
Control Saf. Promot., 10: 173–180. Bolis, L. (1986) Brain injury due to head trauma. Epidemio-
Liu, B., Norton, R., Blows, S. and Lo, S.K. (2003) Helmets for logy in urban areas of the People’s Republic of China. Arch.
preventing injury in motorcycle riders. The Cochrane Data- Neurol., 43: 570–572.
base of Systemic Reviews, Issue 4. Weinstein, N.D. (1987) Unrealistic optimism about susceptibi-
MacLennan, P.A., McGwin, G., Metzger, J., Moran, S.G. and lity to health problems: conclusions from a community-wide
Rue III, L.W. (2004) Risk of injury for occupants of motor sample. J. Behav. Med., 10: 481–500.
vehicle collisions from unbelted occupants. Inj. Prevent., 10: World Health Organisation. (2000) Injury: leading cause of the
363–367. global burden of disease. www.who.int
McDermott, F.T., Cordner, S.M. and Tremayne, A.B. (1996) World Health Organisation. (2004) World report on road
Evaluation of the medical management and preventability of traffic injury prevention. www.who.int
SECTION II

Biomechanics of Injury
This page intentionally left blank
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 2

CNS injury biomechanics and experimental models

M.C. LaPlaca, C.M. Simon, G.R. Prado and D.K. Cullen

Neural Injury Biomechanics and Repair Laboratory, Wallace H. Coulter Department of Biomedical Engineering, Georgia
Institute of Technology and Emory University, 313 Ferst Dr., Atlanta, GA 30332-0535, USA

Abstract: Traumatic brain injury (TBI) and traumatic spinal cord injury (SCI) are acquired when an
external physical insult causes damage to the central nervous system (CNS). Functional disabilities re-
sulting from CNS trauma are dependent upon the mode, severity, and anatomical location of the me-
chanical impact as well as the mechanical properties of the tissue. Although the biomechanical insult is the
initiating factor in the pathophysiology of CNS trauma, the anatomical loading distribution and the
resulting cellular responses are currently not well understood. For example, the primary response phase
includes events such as increased membrane permeability to ions and other molecules, which may initiate
complex signaling cascades that account for the prolonged damage and dysfunction. Correlation of insult
parameters with cellular changes and subsequent deficits may lead to refined tolerance criteria and facilitate
the development of improved protective gear. In addition, advancements in the understanding of injury
biomechanics are essential for the development and interpretation of experimental studies at both the in
vitro and in vivo levels and may lead to the development of new treatment approaches by determining
injury mechanisms across the temporal spectrum of the injury response. Here we discuss basic concepts
relevant to the biomechanics of CNS trauma, injury models used to experimentally simulate TBI and SCI,
and novel multilevel approaches for improving the current understanding of primary damage mechanisms.

Keywords: traumatic brain injury; traumatic spiral cord injury; neurotrauma; biomechanics; membrane
permeability; finite element analysis; injury tolerance criteria

Introduction mechanical input that has exceeded structural


limits of cells and tissue. Primary damage is
Traumatic brain injury (TBI) and spinal cord in- characterized by nonspecific cell loss as well as
jury (SCI) result in a range of deficits depending sublethal injury, which activates a cascade of seco-
on the insult severity and the anatomical region(s) ndary responses leading to prolonged cell death,
affected. In traumatic central nervous injury network dysfunction, and system level changes
(CNS) injury, a mechanical impact (caused by (Fig. 2). Although the mechanical impact is the
motor vehicle accidents, gunshot wounds, blows to initiating event in traumatic CNS injury, the rela-
the head or spine, etc.) induces a mechanical re- tionship between biomechanical inputs and the
sponse at the cell and tissue level that ultimately downstream pathological effects are not well un-
causes a pathophysiological injury response (as derstood. Investigation of relevant loading param-
shown in Fig. 1). In the acute phase of injury, pri- eters and the resulting cell and tissue responses in a
mary damage occurs as a direct result of a variety of model systems is imperative for deci-
phering injury-induced pathophysiological mecha-
Corresponding author. nisms and developing experimental models that
E-mail: michelle.laplaca@bme.gatech.edu hold fidelity to the human clinical situation.

DOI: 10.1016/S0079-6123(06)61002-9 13
14

Mechanical Mechanical Injury


Input Response Response

Fig. 1. Steps in CNS trauma. Traumatic brain and spinal cord injuries result from mechanical loading to the tissue. Pathophysiological
events are initiated by the mechanical tissue response to impact.
insult

secondary response

primary lifespan
response

Fig. 2. Temporal aspects of injury. Mechanical loading causes an acute primary phase followed by a prolonged secondary phase. The
primary response is characterized by nonspecific cell loss, which initiates a cascade of complex secondary events such as inflammation,
excitotoxicity, and neurodegeneration.

A notable application of the study of bio- to dysfunction are complex, yet can be simplified
mechanics in CNS trauma is the determination using controlled cellular injury models that ac-
of accurate tissue tolerances. Tissue tolerances are count for deformation magnitude and rate. Bio-
defined as the point at which structural and/or mechanically relevant in vitro TBI models, used in
physiological failure occurs. An improved under- combination with animal studies and computer
standing of injury biomechanics and the resulting simulations, may lead to improved cellular and
brain and spinal cord responses will ultimately fa- tissue injury tolerance criteria as well as a more
cilitate the development of improved protective complete understanding of the relationship bet-
gear (e.g., helmets and seat belts). Determination ween the biomechanical input and pathophysio-
of tolerance criteria requires information about the logical changes. This multilevel approach will be
forces and deformations that lead to failure, but discussed with respect to selection of experimental
the mechanical parameters (i.e., magnitude and models, development of mechanistically driven
rate of force and deformation) are only partially treatment strategies, and future research priorities.
understood. Tissue response and tolerance criteria
for humans are largely based on cadaveric studies,
but may not accurately represent the properties of Basic biomechanics
living tissue. Basic cell and animal studies, in
which a defined mechanical insult can be applied Biomechanics is the study of forces and physical
to live cells in culture or in an intact animal, have responses in stationary (static) and moving (dy-
an advantage for the determination of tissue tol- namic) biological systems. A system (in the case of
erance and may lead to the refinement of human traumatic CNS injury — the brain or spinal cord)
tolerance criteria. These tolerance criteria must be reacts in a specific way when a force, or load, is
model-independent and represent inherent system placed on it. These external loads may result in
properties. initial damage or lead to delayed damage. The
We will discuss basic biomechanical concepts as point at which loading causes tissue damage is the
they relate to traumatic brain and spinal cord in- threshold (or the tolerance) of the system and is
juries and present experimental models that have dependent on the type and duration of the load.
been developed and characterized in an attempt to The basic terms, or descriptors, that biomechani-
mimic the forces and deformations occurring in cians use to describe applied loads are force and
human CNS trauma. The mechanisms by which stress and the resulting responses are deformations
the mechanical response to a traumatic insult leads and strains.
15

Force is defined as the action of one body (a and strain are referred to as constitutive relation-
physical entity in the system, such as a windshield) ships and the resulting equations are used to de-
on another (as a result of an impact), which will fine behavior of the tissue (or the mechanical
cause acceleration of the second body (e.g., the response).
head) unless acted upon by an equal and opposite The basic mechanics terms defined above are
action counteracting the effect of the first body. valuable in describing the conditions that lead to
The unit is a Newton (N); 1 N is the force that will injuries, although several factors surround bio-
give 1 kg an acceleration of 1 m/s2 (English unit is mechanical analysis of damage prediction. Me-
pound-force, lbf). When forces are generated in chanical conditions can be referred to as the insult
tissue, deformation may ensue depending on the parameters and the result as the injury (Fig. 1).
material properties and the nature of the force itself. Two broad categories of insults can be defined as
Deformation is defined as the change in shape of a static and dynamic loading, with dynamic loading
body undergoing a force. A rigid body, for exam- being the most common. The mechanical response
ple, would experience extremely small deforma- to insult is the tissue deformation or strain and
tions, while biological tissue (usually referred to as will initiate the ensuing pathological events. The
deformable or nonrigid) can often undergo sub- insult parameters and the mechanical response
stantially large deformations. will dictate the types of injury (focal and/or
Stress is another term frequently used in bio- diffuse). We will consider the categories of insults,
mechanical analysis and refers to the distribution the mechanical response to traumatic insult,
of force relative to the area on which it acts. the types of injuries produced, as well as two
Normal stresses (designated by the Greek letter overlapping response phases (primary and second-
sigma (s)) act perpendicular to the surface, while ary) in light of the biomechanical fidelity of
shear stresses (designated by the Greek letter tau experimental models used to simulate these con-
(t)) act tangential to the surface. The unit is the ditions.
Pascal (Pa); 1 Pa ¼ 1 N/m2. A given force acting
on a small surface produces greater stress than the
same force acting over a larger surface. In other Traumatic mechanical insults
words, the amount of mechanical stress created by
a force is dependent on the size of the area over Loads are described as direct (e.g., physical con-
which the force is applied. The resulting strain tact between the head and another object) or in-
that occurs relates the deformed state of the body direct (e.g., as the result of motion of the head). In
to the undeformed state and is unitless. Exten- indirect loading, acceleration of the second body
sional strain is the change in length divided by the (e.g., the head) can act analogously to applied
original length (designated by the Greek letter forces. Loads can be translational (linear), rota-
epsilon) (e ¼ Dl/lo) and can be further classified as tional, or angular (a combination of translational
being in tension (positive strain) or compression and rotational). The type of force and the direc-
(negative strain). Extensional strain results from tion, or plane, of loading, will also affect the re-
stresses generated from linear (or translational) sulting mechanical response in the tissue. The
loads. Shear strain, often resulting from rotational extent and severity of deformation increases with
loads, is also the change in length divided by the increasing force, and this relationship is nonlinear.
original length (designated by the Greek letter In other words, the increase in tissue damage may
gamma) (g ¼ Dl/lo). Brain tissue is thought to be be greater than the proportional increase in force.
more sensitive to shear strain than extensional Static loading is a very slowly applied direct load.
strain (Holbourn, 1943). Therefore loading that Usually there are no deficits until there is sub-
involves rotation of the head has been thought to stantial tissue deformation. These loading condi-
result in more severe injuries, although this as- tions are relatively rare and often occur in human
sumption has recently been questioned (King entrapment situations (e.g., earthquakes). Dy-
et al., 2003). The relationships between stress namic loading, on the other hand, can occur quite
16

rapidly (under 1 s, often o50 ms) and is the most Mechanical response to traumatic insult
common cause of TBI and SCI. Dynamic loading
can further be broken down into impact loading A traumatic insult to brain or spinal cord will lead
(direct loading where an impact occurs with an to a mechanical response of the tissue that is de-
object hitting the head or the head hitting an pendent on the mode, severity, and anatomical lo-
object) or impulsive loading (indirect loading cation of the impact as well as the mechanical
where no contact occurs). Impact loading can be properties of the tissue. The mechanical properties
either focal or diffuse, depending on the magni- of a tissue vary from individual to individual, as
tude of the force and area of impact. Although well as with age and previous injuries or disease
pure impact would involve contact with no head (Prange and Margulies, 2002). In addition, cellular
movement, impact loading is usually a combina- orientation and tissue composition varies among
tion of contact forces — from the impact itself — anatomical regions of the brain and spinal cord,
and inertial forces — from the motion of the head creating nonuniform (or heterogeneous) mechani-
and the brain within the skull. It is important to cal properties that directly affect structural and
consider the size, mass, and hardness of the im- functional tolerances as well as the load distribution
pacting object as well as the surface area and ve- throughout the tissue upon mechanical loading.
locity at which contact occurs. For example, Because of the properties of soft tissues, like
impact with smaller objects (i.e., o2 in. in diam- brain and spinal cord, both the rate and the du-
eter) results in high local stress concentrations and ration of the insult will also influence the response.
therefore is associated with a greater risk for more Loads that are applied quickly may incur more
local and severe damage and is more likely to re- damage due to the material properties of CNS tis-
sult in tissue penetration. Impulsive loading is due sue. When loads are applied at a high rate, the
to inertial forces alone and leads to diffuse tissue cannot absorb (or reduce) the force fast
brain injuries. Models of impulsive loading in- enough and can fail both structurally and func-
clude angular acceleration of the head, yet many tionally. In contrast, slowly applied loads give the
of the models utilized for impact loading are de- tissue ‘‘time’’ to reduce the force and generally re-
signed to deliver a rapid bulk insult that has in- sult in less damage. For short durations of force,
ertial components. Ultimately, the response is much of the effects of the force are reduced. As the
dictated by the mechanical response of the tissue duration of force increases, less reduction occurs
or cells. and therefore less force is needed to produce tissue
Loads, in particular rotational inputs to the deformation. These behaviors are defined by a
brain, however, do not linearly scale between hu- mechanical property termed viscoelasticity.
mans and animal, as the mass of the brain is much
smaller. In fact, to produce an equivalent rota-
tional load in a rodent brain as in a human brain Types of traumatic CNS injury
the angular acceleration would need to be approx-
imately two orders of magnitude higher. This an- Focal injuries result from direct loading and can
atomical complexity introduces difficulty in often occur without widespread, or diffuse, dam-
directly linking pathological consequences to the age. Focal injuries are typically induced when an
biomechanical input. In addition to these con- object penetrates the skull or vertebral column as a
straints in animal modeling, the regional stresses result of a motor vehicle accident, gunshot wound,
and strains have yet to be well characterized. Fu- or a blow. As a result, macroscopically visible
ture investigations to determine the relationships damage is typically visible at the site of impact,
between biomechanical parameters and cellular and the clinical symptoms are often very specific to
responses will require a detailed spatial characteri- the area that is directly injured. Focal injuries to
zation of local cellular stresses and strains in ani- the brain include epidural hematomas and skull
mal models of CNS trauma. fracture (with or without brain damage). When
17

there is osteal or dural compromise, this is often injury. When the acceleration is translational, in-
termed open head injury in the clinical setting. juries tend to be localized to a smaller area. Ro-
Contact loading can also result in coup (at the site tational acceleration, on the other hand, can lead
of impact) and contra-coup (away from the site of to large strains deep within the brain, resulting in
impact) contusions to the brain, involving both diffuse axonal injury (DAI) (Gennarelli et al.,
cellular and vascular components. Focal injuries 1982). Most injuries seen clinically are a combina-
account for one-half of all severe head injuries, but tion of translational and rotational accelerations
two-third of all deaths in this group (Thurman and (referred to as angular acceleration). Diffuse inju-
Guerrero, 1999; Adekoya et al., 2002). ries are thought to occur as a result of not only the
SCI is most commonly caused by fracture and acceleration portion of loading, but also from the
dislocation of the spinal column, resulting in a fo- deceleration portion of the insult, creating very
cal injury. The mechanical impact causes displace- fast moving, uneven load distributions (Margulies
ment of bone fragments, intervertebral discs, or et al., 1990). Diffuse strains can lead to differential
ligaments, resulting in transient compression or movement of the skull relative to the brain, caus-
contusion of spinal cord tissue. Spinal cord is com- ing parasagittal bridging vein injury, as well as in-
pressed at the site of impact that causes the sur- tracerebral hemorrhage. Diffuse injury to the brain
rounding tissue to lengthen in the longitudinal tends to lead to widespread dysfunction, making
direction. Tissue near the center of the spinal cord these injuries the most prevalent cause of persistent
is most vulnerable, suggesting that the mechanical neurological disability. Clinically, diffuse injury is
loads are highest in this anatomical region. Large often seen in closed head injury and arises most
myelinated axons in the surrounding white matter often from motor vehicle accidents.
are also highly susceptible to mechanical damage,
due to stress concentrations at the nodes of
Ranvier (Maxwell, 1996). As in TBI, the rate, Experimental modeling of traumatic CNS injury
magnitude, and duration of the biomechanical in-
sult can dictate the injury response and may affect Experimental models of CNS injury have been in-
functional outcome. Slow stretching of the spinal valuable in the investigation of pathological mech-
cord results in very little tissue damage. In fact, anisms and treatment strategies. However, due to
increasing the length of the spinal cord up to twice the variable nature of clinical traumatic CNS in-
the original length results in very little damage if jury (e.g., inconsistencies in the anatomical loca-
the elongation is applied slowly (Shi and White- tion of impact and the magnitude and duration of
bone, 2006). However, biomechanical inputs ap- loading), experimental models must simplify the
plied rapidly or for an extended duration (longer human condition in order to create a reproducible
than 20–30 min) may surpass tissue thresholds and injury that can be utilized for controlled experi-
result in irreversible damage. mental testing. Although relevance to the clinic
Diffuse injuries are most often caused by inertial may be sacrificed, these simplifications allow the
loading, which describes the motion of objects. assessment of various outcome measures at the
The acceleration (velocity change divided by cellular, tissue, and organism level in response to
change in time) is an important parameter in de- defined bulk loading parameters.
termining tissue response. Higher accelerations In vivo animal models preserve much of the
correspond to higher forces (force equals mass complexity associated with human traumatic CNS
times acceleration, Newton’s second law). This injury while allowing the investigator to experi-
must be taken into account when establishing mentally manipulate certain parameters (e.g.,
thresholds for tissue damage. Because of the com- treatment variables, time of sacrifice) that are not
plex head-neck dynamics, the brain can undergo possible in humans. In the study of injury biome-
high acceleration when subjected to an external chanics, in vivo models provide a more complete
load and therefore TBI often manifests as a diffuse representation of the human brain and spinal cord
18

because they more closely mimic the material biomechanical parameters (e.g., deformation mode,
properties and anatomical architecture. Therefore, rate, and magnitude), allowing for systematic as-
the load distribution and structural failure in an- sessment of cellular responses to defined inputs.
imal models are expected to be similar to human The recent development of a three-dimensional (3-
injury when clinically relevant biomechanical load- D) model in which neural cells are cultured in a
ing parameters are applied in a scale-appropriate hydrogel offers an intermediate degree of complex-
manner. ity, as bulk deformation of the culture results
In vivo models commonly used in TBI and SCI in heterogeneous strain fields at the cellular
research have been used to experimentally repre- level depending on the orientation of the cell
sent aspects of the biomechanics of CNS trauma. within the matrix (LaPlaca et al., 2005; Cullen
Direct loading has been mimicked using contu- and LaPlaca, 2006).
sion, weight drop, fluid percussion, or compression
injuries. Contusion or weight drop involves brief,
rapid loading of CNS tissue using a piston or a Tolerance criteria for CNS injury
weight dropped from various heights (Dixon et al.,
1991; Anderson and Stokes, 1992; Marmarou To date, several cellular tolerance criteria have
et al., 1994; Young, 2002; Scheff et al., 2003). been established to describe the contribution of
These models are designed to deliver a rapid bulk both acceleration and pulse duration for a specific
insult that has both impact and inertial compo- head injury (e.g., skull fracture, concussion), in-
nents. Compression injury is also used to experi- cluding the Wayne State Tolerance Criteria
mentally replicate mechanical loads applied to (Lissner et al., 1960), the Gadd Severity Index
spinal cords over long durations (e.g., due to ab- (Gadd, 1966), and the Head Injury Criterion (Ver-
normal, prolonged twisting of the spine during an sace, 1971). The basic overlying principle is that
automobile accident) (Rivlin and Tator, 1978; short pulses of high acceleration can produce in-
Dolan and Tator, 1979). Inertial loading experi- jury, while lower accelerations require longer
enced during TBI is modeled with fluid percussion pulses to produce injury. These criteria have con-
injury (Dixon et al., 1987; McIntosh et al., 1989; tributed to the development of a fundamental
Thibault et al., 1992) (which has components of foundation; however, the tolerance stipulations
impact loads as well) and angular acceleration of have been based on cadaver or primate data in
the head (Gennarelli et al., 1982; Smith et al., which the measure of injury did not consider
1997), which results in characteristic pathophysio- damage at the cellular level. The efforts at the
logical changes such as DAI. National Highway Traffic & Safety Administra-
In vitro TBI models offer several advantages tion (NHTSA) have produced models of the head
over whole animal models, including control over in the SIMon project. The predictive capability of
cellular components and real-time measurement of SIMon and other computational models hinge on
acute responses. Neural cultures and tissue explants adoption of rational and experimentally verified
have been subjected to compression, tension, or thresholds for damage. Because different regions
shear to experimentally mimic aspects of CNS of CNS tissue have different cellular orientations
trauma (see Morrison et al., 1998b). Models in- and tissue composition, resulting in nonuniform
clude deformable membranes that are stretched (or heterogeneous) mechanical properties, struc-
biaxially (Ellis et al., 1995; Cargill and Thibault, tural and functional tolerances of the brain and
1996; Geddes and Cargill, 2001; Morrison et al., spinal cord differ depending on the region af-
1998a) or uniaxially (Pfister et al., 2003; Lusardi fected. More complex and realistic computer mod-
et al., 2004) to transfer strain to attached cells, els have been developed to provide more accurate
some with the capability of deforming neurites information relevant to the biomechanics of injury
aligned longitudinally to the strain field (Galbraith (e.g., Zhang et al., 2004). Iterative verification of
and Thibault, 1993; Smith et al., 1999). These in these models is imperative to their successful ap-
vitro models allow for isolation of specific plication. Measurement of brain tissue strain
19

during a dynamic mechanical event is exceedingly more susceptible to primary damage caused by the
difficult in the intact animal or postmortem human mechanical insult itself. Although identification of
subject (Hardy et al., 2003). Consequently, it has these regions would allow more accurate correla-
proven challenging to determine quantitative tol- tions between the mechanical input and patho-
erances to be used for the damage measures em- physiological responses, very little is currently
bedded in computer models. Current efforts have known about local cellular strains in animal mod-
utilized existing experimental data and scaling re- els of CNS trauma, mainly due to limitations in
lationships to empirically derive thresholds to pre- detection techniques.
dict physiological outcome in animal experiments One approach for addressing these technical
(Takhounts et al., 2003). Therefore, experimental limitations is the development of more sensitive
models that enable the correlation of strain and methods for the detection of mechanically induced
acute injury could potentially determine detailed damage. Although detection of structural failures
cellular tolerances. can be relatively obvious in some instances (such
as the presence of large focal lesions), more subtle
damage may also be present and can provide a
Response phases of traumatic CNS injury unique opportunity for assessment of local cellular
strains after trauma. Visualization of the anatom-
Acute cellular response ical localization of this mechanical damage can
provide a more sensitive measure of the load dis-
The initial damage that is a direct result of loading tribution throughout the tissue. We and others
to the brain is defined as the primary phase of in- have investigated nonspecific plasma membrane
jury. Biomechanicians study this phase in order to damage as an indicator of mechanical damage in
determine tissue tolerances to mechanical loading various models of TBI and SCI (Pettus et al., 1994;
because the effects of the mechanical insult can be LaPlaca et al., 1997; Shi and Borgens, 2000;
more easily isolated from biochemical events oc- Geddes et al., 2003; Farkas et al., 2006). This type
curring in the secondary or more chronic phase. of cellular damage occurs as a direct result of me-
Our understanding of tolerances at the cellular chanical loading, creating rips or tears in the
level is vital to developing better safety equipment plasma membrane at regions of high local strain.
and understanding mechanotransduction in the We have utilized Lucifer yellow as an indicator
pathological range. At the time of the insult there of acute biophysical membrane failure after TBI
may be a varying amount of primary damage that and SCI. Lucifer yellow is normally membrane-
results from the physical force itself. This includes impermeable; therefore, cellular presence of this
compromised skin, bony fractures, tissue tearing, molecule can be used to detect plasma membrane
cellular rupture, and reorientation of the tissue compromise. In these experiments, Lucifer yellow
components. If a deformation threshold is sur- was injected intrathecally 3 h prior to brain or spi-
passed, these structural failures result and can se- nal cord contusion, and animals were sacrificed
verely compromise brain function. 10 min after injury (a schematic of the injury de-
Due to the heterogeneity of CNS tissue, it is vices are illustrated in Fig. 3). Histological evi-
likely that loads and deformations experienced by dence demonstrated heterogeneous uptake of the
cells in various anatomical regions are not con- permeability marker in various anatomical loca-
sistent and cannot be accurately estimated by sim- tions (as shown in Fig. 4), indicating that the dis-
plistic models assuming homogeneity. Certain tribution of mechanical loading in CNS tissue is
anatomical regions may be subjected to more se- complex and not well understood. Although we
vere loading during impact because of differences have focused on acute membrane damage as an
in the material properties in that particular loca- indicator of the load distribution throughout the
tion (due to variations in cellular orientation, mye- brain and spinal cord, others have explored mem-
lination, etc.). Anatomical regions experiencing brane compromise as an initiator of downstream
larger strains would therefore be expected to be pathological events. Cell membrane damage can
20

A. Infinite Horizons spinal cord contusion device B. Cortical contusion impact device

Displacement
sensor and motor LVDT
controller

Impactor tip
Impactor tip Load cell
Air Stereotactic
Spinal clamps
tank frame
Control Box
500 µm

Fig. 3. In vivo contusion injury devices. Injury devices are used to experimentally deliver prescribed injury parameters to the exposed
brain or spinal cord. For example, the Infinite Horizons spinal cord contusion device (A) allows the user to select an impact force for
injury, while the controlled cortical impact device (B) utilizes a pneumatic system to injure the brain at a defined tissue displacement.

A. TBI B. SCI

100 µm

500 µm 500 µm

Fig. 4. Acute cellular permeability following TBI and SCI. In the acute phase of traumatic injury, the plasma membrane becomes
damaged due to local cellular strains that exceed structural thresholds. Lucifer yellow uptake in the injured brain (A) and spinal cord
(B) demonstrates a heterogeneous distribution of membrane failure, suggesting that loading is not evenly distributed throughout the
CNS parenchyma.

lead to abnormal ion movement across the mem- after focal injury in a contusion model (Fig. 4) as
brane, resulting in pathophysiological changes well as diffuse loading after impact acceleration
such as conduction block, neurofilament compact- injury (Farkas et al., 2006), with patterns of
ion, and impaired axonal transport (Pettus et al., marker uptake specific to the mode of impact. Be-
1994; Shi and Pryor, 2002). Thus, mechanical cause there is a correlation between injury severity
loading may directly result in pathophysiological and membrane compromise, permeability markers
changes. can therefore be used as an indicator of the extent
Experimental evidence has demonstrated that of local cellular loading parameters. For example,
the extent of membrane compromise is dependent experiments conducted in our laboratory have
on the magnitude and rate of strain (LaPlaca et al., demonstrated more extensive permeability marker
1997; Geddes et al., 2003; Shi and Whitebone, uptake in specific hippocampal regions after con-
2006). In addition, others have suggested that the tusion injury, suggesting that local cellular loading
mode of injury may play a critical role in dictating is more severe in certain anatomical locations.
the extent of mechanically induced cell membrane These data may explain the preferential cell death
damage (Geddes-Klein et al., 2006). After TBI, seen in these regions in the subacute and chronic
membrane disruption has been shown to occur phases, as mechanical damage during the initial
21

3-D Neural
Co-Cultures
piston

confocal microscope

3-D Cell Shearing Device 3-D Cell Compression Device

Trapezoidal Input

undeformed undeformed
Strain

0.50
0.25

10 20 30 40
Time (ms)
deformed deformed

Fig. 5. In vitro injury devices for shear and compression injuries. Neural cells cultured in a 3-D configuration were subjected to either
shear or compression injury with a prescribed strain magnitude and rate. This experimental model provides control over bulk material
deformation, while local strains may vary due to cellular orientation within the matrix.

impact in these regions may make the cells more permeability marker uptake per permeabilized cell,
susceptible to death and/or dysfunction during the potentially a gauge of local cellular strain/stress
secondary phase of injury. concentrations, was greater following shear defor-
Although in vivo models can provide a more mation (Fig. 7). Interestingly, the density of dead
anatomically accurate representation of the struc- cells was also significantly greater following shear
tural and functional damage associated with hu- deformation (5–7 fold increase) compared to com-
man CNS injury, in vitro models allow for more pression (2-fold increase), suggesting that there is a
thorough investigation of tissue tolerances because correlation between the degree of membrane per-
biomechanical insult parameters can be more pre- meability and the extent of cell death. This study
cisely controlled and manipulated. In a recent agrees with previous work demonstrating that
study, the effects of both shear and compression shear deformation is the primary mode of tissue
modes of impact were investigated (Fig. 5). This is failure (Holbourn, 1943; Sahay et al., 1992).
an example of how strain estimations derived from Although this study evaluated cellular responses
finite element analysis (FEA) can be applied to based on different modes of bulk deformation, lo-
simplified culture environments to isolate compo- cal cellular strains are heterogeneous, and may be
nents of the heterogeneous mechanical response a function of cell orientation with respect to the
(Fig. 6). Briefly, mixed cultures consisting of neu- bulk strain field (amongst other factors) (LaPlaca
rons and astrocytes were plated in a 3-D matrix et al., 2005; Cullen and LaPlaca, 2006). We have
and subjected to either shear or compressive load- demonstrated that neuronal response to loading
ing (0.50 strain at strain rates of 1, 10, or 30 s1). depends on cell orientation, and hence local cellu-
Both types of loading resulted in significant in- lar strain, where maximal neurite loss occurred at
creases in membrane permeability in a strain rate shear-dominated strain regimes (LaPlaca et al.,
dependent manner, with no differences in the den- 2005). Ongoing in vitro studies are aimed at de-
sity or percentage of permeabilized cells based on fining the biomechanical parameters (deformation
mode of deformation. However, the degree of mode, rate, and magnitude) that lead to structural
22

A
B

Finite element modeling of strain propagation following a focal insult


(controlled cortical impact) in a rat.
in vivo simulations in vitro
A.

Shear-dominated

shear

B.

Compression-dominated

compression

Unloaded region

static

Fig. 6. Finite element model (FEM) simulations and corresponding isolation of loading components in vitro. Traumatic loading to the
brain results in the generation of complex, heterogeneous strain patterns at tissue and cell levels. Heterogeneity in the cellular response
to traumatic loading may be due to several factors, including mode of deformation, cell population, and cell orientation. Neural cell
tolerances to traumatic loading may therefore be elucidated based on these parameters.
23

Static Control 1 s-1 rate 10 s-1 rate 30 s-1 rate

Compression

Shear

Fig. 7. Acute cellular permeability increases in vitro depend on mode of bulk loading. Representative confocal reconstructions of
calcein+ cells following static control conditions or mechanical loading (0.50 strain at 1, 10, or 30 s1 strain rate). Calcein, a normally
cell-impermeant molecule, was added to the extracellular space prior to loading but becomes intracellularly sequestered following
loading. Reconstructions from 50 mm thick z-stacks are shown here.

failure at the cellular level. Models of neural during a traumatic insult. The response (whether
trauma that represent the related biomechanics cellular or whole organism) can better represent
and pathophysiology are important for the eluci- the clinical setting and therefore potential treat-
dation of cellular tolerances and the development ments can be evaluated in a more relevant setting.
of mechanistically driven intervention strategies.
Future directions
Secondary response
Determination of tolerance criteria for traumatic
Primary damage initiates a cascade of secondary CNS injury will likely require a multilevel approach
responses, leading to cell death, network dysfunc- that incorporates both existing data and new
tion, and system-level changes (Fig. 8). While there knowledge from animal and cellular studies with
is no absolute time when primary damage evolves more refined computer modeling. Computer mode-
into delayed effects, the secondary phase of injury ling in the form of FEA can provide estimates of
can be defined as any consequence of the primary the mechanical response of tissue to a large range
insult. This may be in the acute (minutes to hours) of traumatic insult parameters, allowing parametric
period or in a more delayed fashion (days to analysis. These models need to contain anatomical
months) and is dependent on the severity of the detail (for both human and animals) and corre-
initial insult, as well as the health and age of the sponding mechanical property data to maintain the
individual. There is a role for biomechanics in de- highest possible fidelity. In addition, they should be
termining injury mechanisms in both the primary able to simulate large, high rate deformations for
and secondary phases of the injury response by both impact and inertial insult conditions. These
utilizing laboratory models that best mimic the estimated strain and stress patterns should be ver-
forces/stresses and deformations/strains that occur ified with in situ measurements when possible. This
24

MECHANICAL INSULT

MEMBRANE
MEMBRANE STRAIN PERMEABILITY

NON-SPECIFIC ION FLUX


DEPOLARIZATION
(e.g., Ca2+, Na+, K+)
abnormal cell
signaling energy
loss of
deficits
structural proteolytic
abnormal gene integrity degradation
expression

PERSISTENT DYSFUNCTION OR DEATH

Fig. 8. Simplified schematic of injury cascades initiated by mechanical loading. Mechanical injury may directly initiate downstream
pathophysiological events, but the cause-and-effect relationship has not been thoroughly explored. Plasma membrane damage is
hypothesized to trigger cell death or dysfunction through the inability to regulate ion flux.

represents an experimental challenge and is worthy highway and road safety by determining loading
of consideration with new advances in nano- and thresholds to the soft tissue of the brain and spinal
micro-fabrication techniques, which permit elec- cord. In addition to preventative strategies, bio-
tromechanical sensors to be instrumented. Animal mechanics plays an important role in experimental
models provide an opportunity to study the acute modeling which, in turn, is vital to the develop-
phase of injury and therefore can be correlated with ment and application of mechanistically inspired
estimated strain patterns in order to improve our pharmaceutical agents. By applying consistent and
current understanding of mechanotransduction. In clinically relevant mechanical parameters (e.g.,
addition, parallel long-term studies of delayed cell shear strain applied at high rates) to isolated
death and functional outcome can provide correl- neural cells or animal tissue, the response to me-
ative data to acute responses. Furthermore, the cell chanical disturbances can be assessed. The strain
response can be studied under very controlled con- response is dependent on the tissue heterogeneity,
ditions, and in vitro models of traumatic injury can namely the region-specific material properties and
be used to isolate elements of the mechanical re- tissue orientation, therefore making elucidation of
sponse and refine our understanding of cellular the cellular-level response to mechanical-trauma
tolerances. Altogether, these data (with known complex. The correlation of the injury response
temporal responses) can be applied to human mod- with strain enables detailed cellular tolerances that
els of traumatic injury (with unknown temporal can be used to predict human injury criteria using
responses) and tolerance criteria for humans ex- FEA. In addition to cellular-level investigations,
tracted and predicted for specific scenarios. biomechanical models can be utilized at the animal
level to achieve preclinical testing settings. Taken
together, multilevel investigations can be used to
Conclusion eventually decrease the incidence of traumatic
CNS injury and improve clinical outcomes.
Given the tremendous consequences that TBI and
SCI have on society, it is important to better un-
derstand the biomechanical circumstances as they Acknowledgements
relate to the physiological and clinical implica-
tions. Biomechanics can play a role in improving We acknowledge Liying Zhang and King Yang
preventative measures such as safety design in from Wayne State University for the FEA com-
automobiles and sports equipment, as well as puter simulations. Partial funding for the results
25

presented was provided by NSF (BES-0093830) magnitude-dependent increase in plasma membrane perme-
and by Cooperative Agreement No. DTNH22- ability. J. Neurotrauma, 20: 1039–1049.
Geddes-Klein, D.M., Schiffman, K.B. and Meaney, D.F. (2006)
01-H-07551 from the U.S. Department of Trans-
Mechanisms and consequences of neuronal stretch injury in
portation — National Highway Traffic Safety vitro differ with the model of trauma. J. Neurotrauma, 23:
Administration to the University of Alabama at 193–204.
Birmingham, Southern Consortium for Injury Gennarelli, T.A., Thibault, L.E., Adams, J.H., Graham, D.I.,
Biomechanics. Thompson, C.J. and Marcincin, R.P. (1982) Diffuse axonal
injury and traumatic coma in the primate. Ann. Neurol., 12:
564–574.
Hardy, W.N., Foster, C., Mason, M., Yang, K.H., King, A.I.
and Tashman, S. (2003) Investigation of head injury mech-
References anisms using neutral density technology and high-speed
biplanar X-ray. Stapp Car Crash J., 45: 337–368.
Adekoya, N., Thurman, D.J., White, D.D. and Webb, K.W. Holbourn, A.H. (1943) Mechanics of head injuries. Lancet, 2:
(2002) Surveillance for traumatic brain injury deaths — 438–441.
United States, 1989–1998. MMWR Surveill. Summ., 51: 1–14. King, A.I., Yang, K.H., Zhang, L., Hardy, W. and Viano, D.
Anderson, T.E. and Stokes, B.T. (1992) Experimental models (2003) Is head injury caused by linear or angular acceleration?
for spinal cord injury research: physical and physiological In: 2003 IRCOBI Conference. Lisbon, Portugal, pp. 1–12.
considerations. J. Neurotrauma, 9(Suppl 1): S135–S142. LaPlaca, M.C., Cullen, D.K., McLoughlin, J.J. and Cargill II.,
Cargill, R.S. and Thibault, L.E. (1996) Acute alterations in R.S. (2005) High rate shear strain of three-dimensional neu-
[Ca2+]i in NG108-15 cells subjected to high strain rate de- ral cell cultures: a new in vitro traumatic brain injury model.
formation and chemical hypoxia: an in vitro model for neural J. Biomech., 38: 1093–1105.
trauma. J. Neurotrauma, 13: 395–407. LaPlaca, M.C., Lee, V.M. and Thibault, L.E. (1997) An in vitro
Cullen, D.K. and LaPlaca, M.C. (2006) Neuronal response to model of traumatic neuronal injury: loading rate-dependent
high rate shear deformation depends on heterogeneity of the changes in acute cytosolic calcium and lactate dehydrogenase
local strain field. J. Neurotrauma, 23: 1304–1319. release. J. Neurotrauma, 14: 355–368.
Dixon, C.E., Clifton, G.L., Lighthall, J.W., Yaghmai, A.A. and Lissner, H.R., Lebow, M. and Evans, F.G. (1960) Experimental
Hayes, R.L. (1991) A controlled cortical impact model of studies on the relation between acceleration and intracranial
traumatic brain injury in the rat. J. Neurosci. Methods, 39: pressure changes in man. Surg. Gynecol. Obstet., 111:
253–262. 329–338.
Dixon, C.E., Lyeth, B.G., Povlishock, J.T., Findling, R.L., Lusardi, T.A., Rangan, J., Sun, D., Smith, D.H. and Meaney,
Hamm, R.J., Marmarou, A., Young, H.F. and Hayes, R.L. D.F. (2004) A device to study the initiation and propagation
(1987) A fluid percussion model of experimental brain injury of calcium transients in cultured neurons after mechanical
in the rat. J. Neurosurg., 67: 110–119. stretch. Ann. Biomed. Eng., 32: 1546–1558.
Dolan, E.J. and Tator, C.H. (1979) A new method for testing Margulies, S.S., Thibault, L.E. and Gennarelli, T.A. (1990)
the force of clips for aneurysms or experimental spinal cord Physical model simulations of brain injury in the primate.
compression. J. Neurosurg., 51: 229–233. J. Biomech., 23: 823–836.
Ellis, E.F., McKinney, J.S., Willoughby, K.A., Liang, S. and Marmarou, A., Foda, M.A., van den Brink, W., Campbell, J.,
Povlishock, J.T. (1995) A new model for rapid stretch-in- Kita, H. and Demetriadou, K. (1994) A new model of diffuse
duced injury of cells in culture: characterization of the model brain injury in rats. Part I: pathophysiology and biomechan-
using astrocytes. J. Neurotrauma, 12: 325–339. ics. J. Neurosurg., 80: 291–300.
Farkas, O., Lifshitz, J. and Povlishock, J.T. (2006) Mechano- Maxwell, W.L. (1996) Histopathological changes at central
poration induced by diffuse traumatic brain injury: an irre- nodes of Ranvier after stretch-injury. Microsc. Res. Tech.,
versible or reversible response to injury? J. Neurosci., 26: 34: 522–535.
3130–3140. McIntosh, T.K., Vink, R., Noble, L., Yamakami, I., Fernyak,
Gadd, C.W. (1966) Use of a weighted impulse criteria for es- S., Soares, H. and Faden, A.L. (1989) Traumatic brain injury
timating injury hazard. In: 10th Stapp Car Crash Conference in the rat: characterization of a lateral fluid percussion model.
1966, pp. 164–174. Neuroscience, 28: 233–244.
Galbraith, J. and Thibault, L.E. (1993) Mechanically induced Morrison III., B., Meaney, D.F. and McIntosh, T.K. (1998a)
depolarizations in the squid giant axon. J. Biomech. Eng., Mechanical characterization of an in vitro device designed to
115: 13–22. quantitatively injure living brain tissue. Ann. Biomed. Eng.,
Geddes, D.M. and Cargill II., R.S. (2001) An in vitro model of 26: 381–390.
neural trauma: device characterization and calcium response Morrison III., B., Saatman, K.E., Meaney, D.F. and McIntosh,
to mechanical stretch. J. Biomech. Eng., 123: 247–255. T.K. (1998b) In vitro central nervous system models of me-
Geddes, D.M., Cargill II., R.S. and LaPlaca, M.C. (2003) Me- chanically induced trauma: a review. J. Neurotrauma, 15:
chanical stretch to neurons results in a strain rate and 911–928.
26

Pettus, E.H., Christman, C.W., Giebel, M.L. and Povlishock, magnitude and rate of strain. J. Neurophysiol., 95:
J.T. (1994) Traumatically induced altered membrane perme- 3384–3390.
ability: its relationship to traumatically induced reactive ax- Smith, D.H., Chen, X.H., Xu, B.N., McIntosh, T.K., Gennar-
onal change. J. Neurotrauma, 11: 507–522. elli, T.A. and Meaney, D.F. (1997) Characterization of dif-
Pfister, B.J., Weihs, T.P., Betenbaugh, M. and Bao, G. (2003) fuse axonal pathology and selective hippocampal damage
An in vitro uniaxial stretch model for axonal injury. Ann. following inertial brain trauma in the pig. J. Neuropathol.
Biomed. Eng., 31: 589–598. Exp. Neurol., 56: 822–834.
Prange, M.T. and Margulies, S.S. (2002) Regional, directional, Smith, D.H., Wolf, J.A., Lusardi, T.A., Lee, V.M. and Meaney,
and age-depedent properties of the brain undergoing large D.F. (1999) High tolerance and delayed elastic response of
deformation. J. Biomed. Eng., 124: 244–252. cultured axons to dynamic stretch injury. J. Neurosci., 19:
Rivlin, A.S. and Tator, C.H. (1978) Effect of duration of acute 4263–4269.
spinal cord compression in a new acute cord injury model in Takhounts, E.G., Eppinger, R.H., Campbell, J.Q., Tannous,
the rat. Surg. Neurol., 10: 38–43. R.E. and Power, E.D. (2003) On the development of the SI-
Sahay, K.B., Mehrotra, R., Sachdeva, U. and Banerji, A.K. Mon finite element head model. Stapp Car Crash J., 47:
(1992) Elastomechanical characterization of brain tissues. J. 107–133.
Biomech., 25: 319–326. Thibault, L.E., Meaney, D.F., Anderson, B.J. and Marmarou,
Scheff, S.W., Rabchevsky, A.G., Fugaccia, I., Main, J.A. and A. (1992) Biomechanical aspects of a fluid percussion model
Lumpp Jr., J.E. (2003) Experimental modeling of spinal cord of brain injury. J. Neurotrauma, 9: 311–322.
injury: characterization of a force-defined injury device. J. Thurman, D. and Guerrero, J. (1999) Trends in hospitalization
Neurotrauma, 20: 179–193. associated with traumatic brain injury. JAMA, 282: 954–957.
Shi, R. and Borgens, R.B. (2000) Anatomical repair of nerve Versace, J. (1971) A review of the severity index. In: Proceed-
membranes in crushed mammalian spinal cord with polyeth- ings of the 15th Stapp Car Crash Conference, Society of
ylene glycol. J. Neurocytol., 29: 633–643. Automotive Engineers, New York, pp. 771–796.
Shi, R. and Pryor, J.D. (2002) Pathological changes of isolated Young, W. (2002) Spinal cord contusion models. Prog. Brain
spinal cord axons in response to mechanical stretch. Neuro- Res., 137: 231–255.
science, 110: 765–777. Zhang, L., Yang, K.H. and King, A.I. (2004) A proposed injury
Shi, R. and Whitebone, J. (2006) Conduction deficits and threshold for mild traumatic brain injury. J. Biomech. Eng.,
membrane disruption of spinal cord axons as a function of 126: 226–236.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 3

Linking impact to cellular and molecular sequelae of


CNS injury: Modeling in vivo complexity with in
vitro simplicity

Jennifer M. Spaethling, Donna M. Geddes-Klein, William J. Miller, Catherine R. von Reyn,


Pallab Singh, Mahlet Mesfin, Steven J. Bernstein and David F. Meaney

Departments of Bioengineering and Neurosurgery, University of Pennsylvania, 3320 Smith Walk, Philadelphia,
PA, 19104-6392, USA

Abstract: Traumatic brain injury (TBI) represents one of most common disorders to the central nervous
system (CNS). Despite significant efforts, though, an effective clinical treatment for TBI is not yet available.
The complexity of human TBI is modeled with a broad group of experimental models, with each model
matching some aspect of the human condition. In the past 15 years, these in vivo models were complemented
with a group of in vitro models, with these in vitro models allowing investigators to more precisely identify
the mechanism(s) of TBI, the different intracellular events that occur in acute period following injury, and
the possible treatment of this injury in vitro. In this paper, we review the available in vitro models to study
TBI, discuss their biomechanical basis for human TBI, and review the findings from these in vitro models.
Finally, we synthesize the current knowledge and point out possible future directions for this group of
models, especially in the effort toward developing new therapies for the traumatically brain injured patient.

Keywords: traumatic brain injury; biomechanics; in vitro models

Introduction Sattin, 2005). In the elderly, TBI is only eclipsed by


cardiovascular disease and cancer as a major cause
The enormous consequences of traumatic brain in- of death.
jury (TBI) continue in society, despite the rapid ad- Clinically, brain injuries are categorized broadly
vance of technology to reduce the severity of injuries as focal or diffuse (Gennarelli et al., 1982a). Focal
and new approaches in trauma patient care. Even injuries are readily observable lesions that appear
with these changes, traumatic brain injuries remain on standard CT or MRI scans, and include injuries
the leading cause of death in people less than 45 to the vasculature (epidural, subdural hematoma),
years old (Thurman et al., 1999). The incidence rate the microvasculature (cerebral contusions), and vis-
is equally startling — the number of people hospi- ible tears in the brain parenchyma (intracerebral
talized each year for traumatic brain injuries exceed hemorrhage). Diffuse injuries are diagnosed when
those diagnosed with multiple sclerosis, breast can- no visible lesions are present using conventional
cer, and spinal cord injury combined (Langlois and imaging, yet the patient has clear neurological
impairment. A major substrate of diffuse brain
Corresponding author. Tel.: +1 215 573 3155; Fax: +1 215 injuries is diffuse axonal injury (DAI), but this cat-
573 2071; E-mail: dmeaney@seas.upenn.edu egory of injury also includes diffuse brain swelling

DOI: 10.1016/S0079-6123(06)61003-0 27
28

and brain edema (Graham et al., 1995; Povlishock mechanisms for causing immediate impairment
and Katz, 2005). after brain injury, with the first modern studies
Generations of investigators directed their efforts dating back over six decades (Denny-Brown and
toward understanding the mechanisms of TBI. Russel, 1941; Denny-Brown, 1945). These studies
From these efforts, researchers and clinicians reco- were soon followed by a number of in vivo experi-
gnize that TBI for a given patient is not captured mental models that simulated these pressures to
well with only a ‘diffuse’ or ‘focal’ description. cause a concussive insult in animals (Stalhammar
Rather, many different injuries are grouped with and Olsson, 1975; Sullivan et al., 1976). The intra-
the broad clinical subtypes and leads to a tremen- cranial pressure patterns that are generated in the
dous diversity of injuries in the patient population. human brain during impact is known (Nahum et
Injury mechanisms share a similar diversity — the al., 1977) and is predicted accurately with compu-
mechanisms of injury for one specific traumatic in- tational models (e.g., Zhang et al., 2004). There-
jury may not apply universally to other injury fore, one can draw a clear relationship between the
types, and vice versa. As a field, we often use a pressures generated in the in vivo models and
reductionist approach to understand individual within the brain during TBI. The effect of transient
components of clinical TBI. pressure changes on tissue function, though, is less
Models to study the in vivo complexity of TBI clear. Of the possible mechanisms, there is only
exist in different species, along different length consensus that pressure cause damages is when the
scales that span from the single cell to the whole pressure drops below the cavitation threshold for
organism. The purpose of this review is to provide tissue, thereby causing immediate (primary) tissue
a current synthesis of the findings from models in- damage (Nusholtz et al., 1995). Much less agree-
tended to study TBI in vitro, with an emphasis on ment exists on whether high positive pressures will
discussing how these models relate to experimental cause impairment, or an additional physical injury
efforts using animal models of TBI. In turn, we will mechanism is needed.
summarize the findings from these models and Tissue deformation during impact is the
point out new areas of opportunity where these second major physical mechanism explaining
models may prove invaluable in developing new the primary patterns of injury in the brain.
treatments for TBI. Although the brain shape under pressure load-
ing largely does not change, the compliant
properties of the tissue make the brain susceptible
Biomechanical mechanisms that cause TBI in vivo to shearing deformations during impact. Until
recently, we knew little about the properties of
A discussion of the linkages between in vitro and in the brain during situations causing injury. Recent
vivo experimental TBI studies would be incomplete information suggests that the brain softens as it
without a brief review of the underlying biome- deforms (Prange and Margulies, 2002; Coats
chanical mechanisms that cause TBI. One major and Margulies, 2006), an intriguing material
aim of in vitro models is to replicate the underlying property that may lead to unexpected patterns
physical forces that the tissue experiences during of tissue damage. Although there is wide recog-
traumatic injury. However, the forces experienced nition that brain deformation/stress is a major
by the tissue during even a single head impact can mechanism leading to cellular damage, exactly
vary greatly with impact direction, force, and the how the stresses are transferred from the tissue
impacting surface. A person designing an in vitro to the cellular structures within the material are
model must ask — which of these forces to the not well known. Estimates of the tissue strain
tissue are important for causing injury? In addi- needed to cause injury are now available (Shreiber
tion, one asks — how does one control the inherent et al., 1997; Bain and Meaney, 2000; Zhu et al.,
variability of these forces? 2006), but there remains considerable discussion
Pressures developed within the brain during in- about how these strains should be modeled in
jury were considered one of the primary either in vivo or in vitro models, and if these
29

models can truly capture the complete simulation across species (Smith et al., 1995), and modifying
of injury. the model to injure different areas of the cortex.
Recent modifications of the model used a direct
skull impact, leading to a closed head impact
Reproducing injury mechanisms with in vivo TBI model that could be considered closer to the clin-
models ical condition. With the precise control of the
model, the cortical impact model is frequently the
Much like other diseases and disorders, an inves- choice when studying TBI in transgenic animals.
tigator faces several challenges when modeling hu- From an injury mechanism standpoint, the cortical
man TBI in an animal model. The clinical mix of model is qualitatively similar to the fluid percus-
focal and diffuse injuries conflicts with the need to sion technique — apply a mechanical input locally
develop a consistent, reliable, and repeatable to the cortex and study the progressive pattern of
model in the laboratory. Inevitably, the investiga- injury throughout the brain.
tor chooses which components of clinical TBI to A final method for TBI models is using acceler-
study in the laboratory. Here, we briefly categorize ation to injure the brain, reproducing a common
the different experimental models and describe mechanical loading that causes TBI in humans.
their uses. We use this description as background Studies show the necessary acceleration to cause
material for the in vitro models discussed in the injury increases quickly with decreasing brain mass
next section. (Ommaya et al., 1967); therefore, the acceleration
For nonpenetrating injuries, an early area of method is most readily used in large animal species
study was to model the effects of pressure loading that include the nonhuman primate and the min-
on the brain, as the concussive effects of pressure iature pig (Gennarelli et al., 1982b; Meaney et al.,
were reported in the early literature (Denny- 1995). Although models using acceleration input in
Brown, 1945). The effect of pressure led to the so small animals (e.g., rat, ferret) appear in the liter-
called ‘percussion concussion’ models (Gennarelli, ature (Marmarou et al., 1994; Xiao-Sheng et al.,
1994) that used a pulse of either air or fluid on the 2000; Gutierrez et al., 2001), it is difficult to sep-
exposed cortex to cause neurological impairment. arate the effects of the acceleration from the pos-
The most common model in current use is the fluid sible shape changes in the skull caused by the
percussion model, available in many species and relatively large force needed to create these accel-
used in different configurations (Stalhammar and erations. A combination of the acceleration input
Olsson, 1975; Sullivan et al., 1976; Dixon et al., and the higher proportion of white matter in the
1988; McIntosh et al., 1989a). Early work with gyrencephalic brains of the nonhuman primate and
fluid percussion showed how the pressure applied pig makes these models ideal for studying injury to
to the cortex was dissipated throughout the brain the white matter.
and the spinal canal (Stalhammar and Olsson,
1975). Later work showed that this pressure also
caused a movement of intracranial tissue, leading Broad categorization of in vivo models — what do
to pressure and strain throughout the brain they reproduce?
(Thibault et al., 1992). Possibly due to the com-
plex mechanics of this model, different variations The different in vivo models described above are
of the percussion model can lead to forms of injury generally designed to simulate the mechanisms of
that resemble components of human TBI. injury that occur in humans. Results from in vivo
A more recent technique to study brain injury in models are sometimes difficult to interpret, though,
vivo is to directly deform the brain with a solid as measurements of intracellular signaling, single
indentor, often referred to as the cortical impact cell function, and the role of different cell types are
model (Lighthall, 1988). The advantages of the difficult. Moreover, the effect of mechanical and
model include a highly quantified impact condi- hypoxic injury are not easily separable with in vivo
tion, an ability to easily scale the impact condition models, but are easily divided with in vitro
30

approaches. The use of in vitro models to simulate cuts can lead to gene expression changes over time
TBI began several decades ago and has signifi- (Raghupathi et al., 1998). This remains probably
cantly evolved in complexity (Morrison et al., the most precise model to study distal and proximal
1998b). In this section, we describe the in vitro effects of transection. The model best corresponds
models that are currently in use to study TBI, and to the physical disruption that can occur to some
provide an in vivo correlate for these in vitro neuronal processes following injury (Maxwell et al.,
methods. 1997), especially at the severe levels. The most im-
portant utility of the model, though, may be the
ability to distinguish the role of local and remote
Scratch model/laceration signaling on cell survival/death following injury
(Singleton et al., 2002).
The most direct method for mechanical injury is to
tear or lacerate cultures with a stylus or punch. The
Weight drop/compression
direct mechanical disruption of cultures is one of
the earliest methods to study the progression of in-
An additional and straightforward technique to
jury, beginning with tissue chunks (Epstein, 1971)
use on cultures is to mechanically compress the
but soon moving to mixed cultures of neurons and
cultures with a weight, akin to the weight drop
glia (Tecoma et al., 1989; Regan and Choi, 1994;
method developed initially by Allen (1911) to
Regan and Panter, 1995). The scratch method is
study spinal cord injury in vivo. Indeed, one of the
well suited for high throughput drug screening, and
first in vitro models for central nervous system
has been used by Faden and colleagues to examine
(CNS) injury used this technique of spinal cord
both inhibitors and antisense oligonucleotide treat-
cultures (Balentine et al., 1988). The technique is
ment (Faden et al., 1997; Mukhin et al., 1997,
well suited to organotypic cultures that have a de-
1998). This technology also scales easily to slice
fined thickness and more realistic 3D architecture,
culture tissue (Sieg et al., 1999), and has suggested
and can be used to study the effects of both me-
factors that influence the vulnerability or surviv-
chanical injury and a superimposed hypoxic injury
ability of neurons in different regions of the brain.
(Adamchik et al., 2000). The order of the injuries
The direct mechanical injury remains in use, now
can be changed, so that the mechanical injury can
with an emphasis that includes the release of mole-
be considered the secondary injury, or vice versa.
cules that could be considered biomarkers of the
The technique to compress the tissue construct can
injury (Yang et al., 2006) and the potential factors
also change; a dropped weight can be replaced by a
that can regulate the migratory activity of glial cells
rolling stainless steel bar, or a composite foam in-
to the injury site (Barral-Moran et al., 2003). The
dentor over a region of the culture (Adamchik
technique best correlates to the primary tissue tear-
et al., 2000). Recently, this type of model showed
ing that can occur in different brain regions fol-
the potential spreading depression that can occur
lowing severe head injury, a penetrating ballistic
from mechanical injury (Church and Andrew,
injury, or the local tissue damage that occurs with
2005). One primary disadvantage of this tech-
depressed skull fractures.
nique, though, is drawing the direct in vivo cor-
An interesting variation of the tearing model is
relate for this method. Crush injuries to the brain
using a laser to focally disrupt or transect the proc-
parenchyma are rare, and are complicated by
esses of neurons in culture (Gross et al., 1983). The
overlying skull fracture.
relative position of the laser cut can be controlled
and can be close to or distant from the neuronal
soma, yielding distinct differences in cell fate Cell/substrate stretch model
(Lucas, 1987). The distance from the lesion to the
soma also changes the subsequent ultrastructure Based on the number of completed studies, the
response, as well as the electrophysiological prop- most commonly used in vitro technique to study
erties of the cell (Lucas et al., 1985). Moreover, the the consequences of TBI in vitro is the cell stretch
31

(or substrate deformation) model. These models Identifying these early changes in CNS cells pro-
replicate the magnitude and rate of tissue defor- vides initial therapeutic targets. Knowing how the
mation that occurs in vivo during injury (Meaney mechanoactivated targets lead to subsequent intra-
et al., 1995), but often simplify the multiple oscil- cellular events and/or consequences will naturally
lations of tissue deformation into a single, transient generate new therapeutic targets, is perhaps as
stretch insult. One early feature of the model was important. In this section, we review the current
using a design where the cultured cells, plated to an knowledge on the early changes that occur follow-
elastic substrate, were simultaneously deformed in ing mechanical injury and their relative utility as a
two directions. Although first used in astrocytes, target for reversing the effects of the injury.
this technique was rapidly tested in many different
cell types of the CNS including neuron and neuron-
like cell lines, endothelial cells, and, more recently, NMDAR are the most commonly studied
microglia (Cargill and Thibault, 1996; Ellis et al., mechanoactivated target
1995; McKinney et al., 1996; Rzigalinski et al.,
1997). A variation of the initial stretch models is Alterations in ionic homeostasis have been ob-
now available, where the cultures are stretched served across nearly all in vitro and in vivo models
only in one direction (Lusardi et al., 2004). The of TBI and therefore provide a natural starting
stretch can be confined to just cultured axons to point for identifying mechanoactivated receptors.
model diffuse axonal injury (Smith et al., 1999). Due to the role of glutamate receptors in both
Methods to stretch dissociated cultures transfer physiologic (learning and memory) and pathologic
easily to studying the effects of stretch on tissue (stroke and epilepsy) conditions, N-methyl
slice cultures (Morrison et al., 1998a), where the D-aspartate receptors (NMDAR) are among the
transfer between the substrate stretch and the re- most widely studied receptors responsible for the
sulting tissue stretch are defined (Cater et al., 2006). cytosolic calcium overload (Gagliardi, 2000;
More recent work shows the cell stretch technique Arundine and Tymianski, 2004). Multiple models
can scale to study the effect of more complex, 3D of TBI have shown that the bulk of the loss of ionic
strain patterns on the morphology and viability of homeostasis can be attributed to activation of
cells in tissue constructs (LaPlaca et al., 2005). NMDARs. The connectivity of NMDARs to the
With the increasing number of controlled mechan- actin cytoskeleton may provide a potential mech-
ical manipulations available on cultures and tissue anism as to how these receptors are activated by
constructs, it is now possible to identify if CNS stretch (Geddes-Klein et al., 2006b). One unique
cells are uniquely vulnerable to a certain type of feature of mechanical injury to neurons is the
mechanical deformation (Geddes-Klein et al., mechanically induced reduction in the normal mag-
2006a), if the effects of injury are cumulative nesium block of the NMDAR (Zhang et al., 1996).
(Slemmer et al., 2002), or if the mechanism(s) of Altering the magnesium block of NMDARs
injury will change if the cells experience stretch, changes the relative influence of NMDAR even
compression, or fluid shear deformation (LaPlaca under normal neurotransmission signaling and
et al., 1997). provides a therapeutic target that has been found
to be somewhat effective in vivo (McIntosh et al.,
1989a, b). It is worth noting that mechanically in-
Mechanoactivation — the first therapeutic target duced changes in the NMDAR properties are not
observed across all models of TBI, including mixed
The increasing diversity of models used to study cultures (Faden et al., 2001), or when the cultures
TBI in vitro leads one to a common question — are subject to a sublethal stretch (Arundine et al.,
what processes do these forces activate rapidly fol- 2003, 2004). Additionally, the mechanically induced
lowing injury, and how do these ‘mechanoactivated’ activation or modulation of NMDARs leads to
signals lead to the pathological changes observed the propagation of intracellular calcium into adja-
hours to days following the initial injury? cent, uninjured cells (Lusardi et al., 2004). While
32

NMDAR antagonists have not been effective in the 2005), these mechanically initiated changes may
clinic, recent data suggests that targeting the loca- have long lasting effects on synaptic physiology
tion of NMDAR (synaptic vs. extrasynaptic) may following injury.
differentiate between the protective and excitotoxic
processes of NMDARs (DeRidder et al., 2006).
Voltage-gated sodium channels can contribute to
regional changes in neuronal axons
Are other glutamate receptors involved?
Voltage-gated sodium channels have been shown
to indirectly contribute to shifts in intracellular
The roles of other glutamate receptors are also be-
calcium following mechanical injury. The relative
ginning to generate serious attention in treating
role of the voltage-gated sodium channel only
traumatic brain injuries. a-Amino-3-hydroxy-5-
appears when the axonal segment of the neuron is
methyl-4-isoxazolepropionic acid receptors (AM-
deformed, potentially because the NMDAR and
PAR) undergo a unique transformation, losing their
AMPAR mediated changes dominate for dendritic
rapid desensitization property following mechanical
processes and the neuronal soma. Moreover, it is
injury (Goforth et al., 1999). The loss in desensiti-
worth noting, that little in vitro work exists for
zation is persistent for at least 24 h following injury,
myelinated axons due to the difficulties of creating
and can be avoided if the NMDARs are inhibited
myelinated cultures in vitro, even though it has
prior to mechanical injury (Goforth et al., 2004).
been shown that myelinated axons respond differ-
The impact of this loss in desensitization of the
ently to stretch than myelinated axon (Reeves
AMPARs on neuronal function is not yet well de-
et al., 2005). Studies show the sodium channel ac-
scribed, though. As AMPARs are centrally involved
tivation following mechanical injury leads to a
in fundamental processes such as synaptic plasticity
dramatic and sudden rise in axoplasmic calcium,
and metaplasticity, even a transient alteration in the
but not from calcium entering directly through the
normal desensitizing properties of the AMPAR can
sodium channel (Wolf et al., 2001). Rather, the
lead to immediate and long term changes in neu-
source of increased axoplasmic calcium is through
ronal network behavior.
voltage-gated calcium channels and through re-
Although there is evidence that inhibition of me-
versal of the sodium calcium exchanger. These
tabotropic glutamate receptors (mGluRs) will pro-
changes in the sodium channel occur simultane-
vide some protection against the effects of
ously with a larger, but reversible change in the
mechanical injury, it is not known if these changes
morphology of axons subjected to mechanical in-
are directly caused by mechanical modulation of
jury (Smith et al., 1999). Over time, these sodium
the mGluRs, or if these protective effects are from
channels are the targets of proteolysis, which can
simply inhibiting the activation of these receptors
lead to a sustained change in neuronal activity
from enhanced glutamate levels following injury
(Iwata et al., 2004).
(Faden et al., 2001; Movsesyan et al., 2001;
Movsesyan and Faden, 2006). One interesting
recent theory is the role of potential physical cou- Changes in mechanical permeability
pling between the group I mGluR receptors, IP3,
and phospholipase C (PLC) (Floyd et al., 2004). One final consequence of mechanical force is an
The role of different mGluR subtypes also reveals a immediate, but transient, change in the plasma
complex interplay between mGluRs after injury, membrane permeability termed ‘mechanoporation’.
where the inhibition of one subtype (type III) can In a series of studies, both the relative size and du-
eliminate the protection offered by an antagonist ration of these transient pores in the membrane were
directed against a second subtype (type II) estimated following neuronal stretch (Geddes and
(Movsesyan and Faden, 2006). Given the role of Cargill, 2001; Geddes et al., 2003). These effects are
mGluRs in either enhancing or modulating synap- also measured following traumatic injury in vivo,
tic efficacy (Gubellini et al., 2004; Simonyi et al., although these changes appear to occur over a
33

longer time period than in vitro preparations (Pettus injury, there is a delayed, but persistent depolari-
et al., 1994; Stone et al., 2004; Farkas et al., 2006). It zation that is linked to the alteration in the elect-
is not clear, though, how these changes in perme- rogenic sodium/potassium exchanger (Tavalin
ability can change for different neuronal regions et al., 1995, 1997). The depolarization is triggered
(axon, dendrite, soma), or if this process is limited to with a mild glutamate stimulation, and can be
one or more cell types. The effect of these transient attenuated by restoring intracellular ATP levels.
pores can be minimized by resealing the membrane Findings from several groups show that mechan-
with surfactants (Serbest et al., 2006). Even though ical injury leads to an enhanced response to gluta-
these changes may be brief, recent evidence suggests mate up to 24 h following injury (Weber et al.,
this mechanism may be capable of specifically stim- 1999; Arundine et al., 2004; Geddes-Klein et al.,
ulating different components of the mitogen- 2006a), in turn leading to an enhanced vulnerabil-
activated protein kinase (MAPK) cascade, and can ity to glutamate excitotoxicity (Arundine et al.,
therefore play a role in the ensuing cell death that 2003). Although most reports focus on the role of
occurs after injury (Serbest et al., 2006). However, the NMDAR as the underlying factor for this
the role of permeability increases in viability seems enhanced glutamate response, this is not the only
to be inconsistent across models, as some studies ligand-gated receptor that changes following in-
reveal no change in membrane resistance or perme- jury. The loss in desensitization of the AMPAR
ability to a fluorescent dye (Tavalin et al., 1995; appears in parallel with an enhancement of AMPA
Zhang et al., 1996; Smith et al., 1999). currents after injury, as does the enhancement of
g-aminobutyric acid (GABA) currents. Both the
AMPA and GABA current changes are linked to
Mechanoactivation cascade — the next target
the NMDAR, as inhibiting the NMDA prior to
injury eliminates the enhancement of both currents
Even though the early events of mechanical injury
(Goforth et al., 2004; Kao et al., 2004). These
in vitro are becoming clear, these efforts only begin
changes may also contribute to the changes in
to establish the complex cascade of events that will
neural network activity that appears following me-
influence neuronal or glial survival after TBI. The
chanical injury (Prado et al., 2005), although these
initial mechanoactivated receptors and ensuing loss
changes remain to be explored in detail. These
of ionic homeostasis activates a cascade of cellular
changes are likely among the initiating factors
processes that ultimately result in loss of function
that contribute to failure to induce LTP in in
or cell death. Understanding these cascades will be
vivo models and ultimately result in learning and
essential to developing novel therapeutic targets
memory deficits.
that are admissible in a clinically relevant thera-
Neuronal mitochondria are also affected, with a
peutic window. We term these next events as the
reduction in the mitochondria membrane potential
‘mechanoactivation cascade’. These cascades can
that can persist and which is dependent on the
occur rapidly following the initial mechanoactiva-
presence of surrounding glia (Ahmed et al., 2000,
tion step, or can progress more slowly over time. In
2002). A series of reports show that the normal
this section, we group these cascades according to
regulation of intracellular calcium stores are altered
the cell types studied to date in the CNS.
soon after mechanical injury, and this change in
capacitive calcium influx can alter the homeostatic
Neuronal cascades mechanisms for calcium induced calcium release
(Weber et al., 2001; Chen et al., 2004; Weber, 2004).
The changes in neurons following mechanical in- Cysteine protease activation is dependent on the
jury include alterations in the electrophysiological severity of the injury, and can be affected by
properties, organelle function, receptor profile, and NMDAR activation inhibition/activation (Pike
intracellular calcium buffering. Some of these et al., 2000; DeRidder et al., 2006). Inhibiting the
changes lead to neuronal vulnerability, while oth- activation of one cysteine protease, caspase-3, will
ers will transiently impair function. Early after have a short therapeutic window but also reveals
34

some crosstalk among the classic apoptotic and within astrocytes, but also mediate damage in
necrotic pathways (Knoblach et al., 2004). Gene adjacent neurons (Lamb et al., 1997). Changes in
expression changes correlate with the mechanical mitochondria membrane potential and ATP levels
input applied to the culture (Morrison et al., 2000); occur only transiently in astrocytes after injury, but
some of these changes reflect changes observed in the presence of the astrocytes in cultures change the
single neurons following TBI in vivo (O’Dell et al., time course of neuronal mitochondria changes
2000). The role of MAPKs in vitro are similar to (Ahmed et al., 2000). The presence of astrocytes
the role observed in vivo — extracellular signal also appears to influence the release of matrix met-
regulated kinase (ERK) inhibition may be a alloproteinases following injury in vitro, a factor
potential target (Mori et al., 2002), but the timing that may significantly affect the regeneration phase
of the therapy needs to be better defined (Dash after injury (Wang et al., 2002). Gene expression
et al., 2002). changes can be altered in the presence or absence of
glia (Katano et al., 1999), indicating that the cross-
over effect extends to the genomic level.
Astrocytic cascades

The role of astrocyte-based changes can influence What therapies have emerged?
not only glial reactivity in the injured brain, but
also neuronal activity through the coupling of as- A primary advantage of in vitro models is the
trocytes and neurons at the synapse. Intracellular ability to quickly test the efficacy of new or existing
calcium signaling in astrocytes after injury is compounds in reducing the effects of mechanical
coupled to the presence of extracellular calcium, injury. Most work to date, though, has focused on
and the absence of extracellular calcium will in- the mechanisms that contribute to either early
fluence astrocytic death (Rzigalinski et al., 1997). changes in intracellular signaling or cell function.
The complex intracellular regulation of calcium Surprisingly few studies have moved this work
also changes, as stimulation of mGluRs is less toward studying therapies that may be used for in
coupled to IP3 mediated calcium release and is vivo TBI treatment. Early work showed the effec-
regulated, in part, by PLC (Rzigalinski et al., 1998; tiveness of NMDA antagonists (Regan and Panter,
Floyd et al., 2001). Changes in intracellular 1995), free radical scavengers (Lamb et al., 1997;
sodium occur in parallel with these calcium Shah et al., 1997), and mGluR activation or inhi-
changes and are linked with glutamate uptake in bition (Allen et al., 1999; Movsesyan et al., 2001).
astrocytes, but may also be loosely coupled to More recent work includes the development of new
intracellular calcium changes through changes in peptides to reduce cell death after injury, even if the
the sodium calcium exchanger (Floyd et al., 2005). compound is delivered hours following injury
These changes in ion homeostasis lead to altera- (Faden et al., 2003, 2004, 2005). These peptides
tions in MAPK signaling (Neary et al., 2003), offer a strategic advantage over receptor antago-
some of which can be linked to the eventual glial nists by offering more selective inhibition of intra-
reactivity phase that forms an important part of cellular signaling triggered by a target receptor.
the neuropathology found in human TBI. Customized peptide designs can interfere with crit-
Perhaps more than studies using cultured neu- ical regulatory points in the mechanoactivation
rons, there are now several reports focusing on the cascade, such as the point where NMDAR activa-
‘crossover’ effect of mechanically injured astrocytes tion can lead to the formation of reactive oxygen
on other CNS cell types. For example, injured as- species. Using a peptide to inhibit the linkage
trocytes generate isoprostanes that can influence between the NMDAR and PSD-95, a prominent
vasoconstriction and reduce blood flow after postsynaptic density protein, leads to an interrup-
trauma (Hoffman et al., 2000). Generation of re- tion in the ROS cascade and will lead to neuronal
active oxygen species from mechanically injured protection (Arundine et al., 2004). Delayed efficacy
astrocytes can affect not only metabolic changes is also possible by using subunit specific
35

antagonists for the NMDAR (DeRidder et al., current work does not comprehensively address the
2006), perhaps due to the recent data showing the initial mechanoactivation process for all cell types
dual role for NMDAR in cell survival and in the brain; notable exceptions are the oligoden-
apoptosis (Hardingham. and Bading, 2003). drocytes, brain endothelial cells, and microglia.
In vitro models are also useful in determining Even within a given cell type such as neurons, there
more specific biomarkers for both TBI diagnosis are differences that appear among neurons from
and treatment. A group of released factors have different brain regions. Moreover, there is little
been found in the media following mechanical in- consensus information on how the culture platform
jury, with some contributing to the ensuing neu- — nearly pure or mixed cultures, tissue constructs,
ronal death. Injury using a stylus transection or organotypic slice cultures — contribute to the
method is inhibited with NMDAR antagonists measured response; information to date show the
and aminosteroids (Regan and Panter, 1995), and potential synergistic interactions that appear bet-
the surrounding media can contain free radicals ween different cell types. These areas, as well as
that, if applied to uninjured cultures, would cause many others, can provide more insight into how
apoptotic cell death within hours (Shah et al., these models will move toward representing the in
1997). A physical scratching injury also activates vivo environment following injury.
inflammatory mediated injury in neuronal cultures, In closing, it is worth noting that the natural
as well as astrocytic migration (Fitch et al., 1999), division between in vivo and in vitro models of
MAPK activation, and the release of matrix injury is slowly disappearing. The culturing tech-
metalloproteinases (MMPs) (Wang et al., 2002). niques are becoming more sophisticated, allowing
Mechanical stretch injury will cause the synaptic one to construct functional tissues with highly pre-
release of zinc (Cho et al., 2003), an increase in cise and more ‘in vivo like’ architectures. The
s100b in the media (Willoughby et al., 2004). It is methods to injure these cultures is now developed
interesting to note that at least some of these enough to mechanically load these constructs with
molecules are being investigated as potential any desired loading profile, mimicking the loading
biomarkers for in vivo injury (Pineda et al., 2004). profiles occurring in vivo following injury. In com-
parison, rapid advances with in vivo imaging tech-
nology now allows one to use fluorescent indicator
Pointing toward the future dyes in vivo to monitor shifts ion homeostasis, and
technology is already emerging to monitor the
Collectively, this work shows the extensive efforts electrical activity in living, awake animals over
completed in understanding how mechanical forces time. As a result, technologies and measurement
are transmitted to cells of the CNS, what processes normally restricted to in vitro systems are now
are activated by these mechanical forces, and how available for testing in the in vivo animal. The in-
this information reveals different intervention creasing convergence of these different approaches
points for treating the effects of mechanical injury will likely make comparative approaches in the fu-
in the CNS. Motivated by an understanding of the ture more common, and will lead to a more rapid
physical forces that occur within the tissue during translation of in vitro findings to the in vivo set-
injury, there are now models to study the effect of ting. Ultimately, this will translate into the more
both simple and more complex mechanical loading rapid testing of therapeutics in vivo that originated
to cultures. These models are well controlled, can from in vitro systems, and lead to more therapeutic
be easily extended to understand more realistic options for the traumatically brain injured patient.
loading conditions, and are even approaching the
very complex question of how these forces are
transferred to cells in 3D tissue. One needs to con- Acknowledgments
sider if additional in vitro models are needed, or if
the complex mechanical environment is suitably Funds for this work were provided by grants from
modeled with existing technology. Certainly, the the National Institutes of Health (RO1 HD-41699
36

and NS-35712) and the Commonwealth of Penn- dependent on tissue strain but not strain rate. J. Biomech.,
sylvania. 39(15): 2810–2818.
Chen, T., Willoughby, K.A. and Ellis, E.F. (2004) Group I
metabotropic receptor antagonism blocks depletion of cal-
cium stores and reduces potentiated capacitative calcium en-
References try in strain-injured neurons and astrocytes. J. Neurotrauma,
21(3): 271–281.
Adamchik, Y., Frantseva, M.V., Weisspapir, M., Carlen, P.L. Cho, I.H., Im, J.Y., Kim, D., Kim, K.S., Lee, J.K. and Han,
and Perez Velazquez, J.L. (2000) Methods to induce primary P.L. (2003) Protective effects of extracellular glutathione
and secondary traumatic damage in organotypic hippocam- against Zn2+-induced cell death in vitro and in vivo. J.
pal slice cultures. Brain Res. Brain Res. Protoc., 5(2): Neurosci. Res., 74(5): 736–743.
153–158. Church, A.J. and Andrew, R.D. (2005) Spreading depression
Ahmed, S.M., Rzigalinski, B.A., Willoughby, K.A., Sitterding, expands traumatic injury in neocortical brain slices. J.
H.A. and Ellis, E.F. (2000) Stretch-induced injury alters mi- Neurotrauma, 22(2): 277–290.
tochondrial membrane potential and cellular ATP in cultured Coats, B. and Margulies, S.S. (2006) Material properties of
astrocytes and neurons. J. Neurochem., 74(5): 1951–1960. porcine parietal cortex. J. Biomech., 39(13): 2521–2525.
Ahmed, S.M., Weber, J.T., Liang, S., Willoughby, K.A., Sit- Dash, P.K., Mach, S.A. and Moore, A.N. (2002) The role of
terding, H.A., Rzigalinski, B.A. and Ellis, E.F. (2002) extracellular signal-regulated kinase in cognitive and motor
NMDA receptor activation contributes to a portion of the deficits following experimental traumatic brain injury. Neuro-
decreased mitochondrial membrane potential and elevated science, 114(3): 755–767.
intracellular free calcium in strain-injured neurons. J. Neuro- Denny-Brown, D. (1945) Cerebral concussion. Physiol. Rev.,
trauma, 19(12): 1619–1629. 25: 296–325.
Allen, A. (1911) Surgery of experimental lesion of spinal cord Denny-Brown, D. and Russel, W.R. (1941) Experimental
equivalent to crush injury of fracture dislocation of spinal cerebral concussion. Brain, 64(2–3): 92–164.
column — a preliminary report. JAMA, 57(11): 878–890. DeRidder, M.N., Simon, M.J., Siman, R., Auberson, Y.P.,
Allen, J.W., Ivanova, S.A., Fan, L., Espey, M.G., Basile, A.S. Raghupathi, R. and Meaney, D.F. (2006) Traumatic mechan-
and Faden, A.I. (1999) Group II metabotropic glutamate ical injury to the hippocampus in vitro causes regional ca-
receptor activation attenuates traumatic neuronal injury and spase-3 and calpain activation that is influenced by NMDA
improves neurological recovery after traumatic brain injury. receptor subunit composition. Neurobiol. Dis., 22(1): 165–176.
J. Pharmacol. Exp. Ther., 290(1): 112–120. Dixon, C.E., Lighthall, J.W. and Anderson, T.E. (1988) Physio-
Arundine, M., Aarts, M., Lau, A. and Tymianski, M. (2004) logic, histopathologic, and cineradiographic characterization
Vulnerability of central neurons to secondary insults after in of a new fluid-percussion model of experimental brain injury
vitro mechanical stretch. J. Neurosci., 24(37): 8106–8123. in the rat. J. Neurotrauma, 5(2): 91–104.
Arundine, M., Chopra, G.K., Wrong, A., Lei, S., Aarts, M.M., Ellis, E.F., McKinney, J.S., Willoughby, K.A., Liang, S. and
MacDonald, J.F. and Tymianski, M. (2003) Enhanced vul- Povlishock, J.T. (1995) A new model for rapid stretch-
nerability to NMDA toxicity in sublethal traumatic neuronal induced injury of cells in culture: characterization of the
injury in vitro. J. Neurotrauma, 20(12): 1377–1395. model using astrocytes. J. Neurotrauma, 12(3): 325–339.
Arundine, M. and Tymianski, M. (2004) Molecular mecha- Epstein, M.H. (1971) Relative susceptibility of elements of the
nisms of glutamate-dependent neurodegeneration in ischemia cerebral cortex to mechanical trauma in the rat. J. Neu-
and traumatic brain injury. Cell Mol. Life Sci., 61(6): rosurg., 35(5): 517–522.
657–668. Faden, A.I., Fox, G.B., Di, X., Knoblach, S.M., Cernak, I.,
Bain, A.C. and Meaney, D.F. (2000) Tissue-level thresholds for Mullins, P., Nikolaeva, M. and Kozikowski, A.P. (2003)
axonal injury in an experimental model of CNS white matter Neuroprotective and nootropic actions of a novel cyclized
injury. J. Biomech. Eng., 122: 615–622. dipeptide after controlled cortical impact injury in mice. J.
Balentine, J.D., Greene, W.B. and Bornstein, M. (1988) In vitro Cereb. Blood Flow Metab., 23(3): 355–363.
spinal cord trauma. Lab. Invest., 58(1): 93–99. Faden, A.I., Ivanova, S.A., Yakovlev, A.G. and Mukhin, A.G.
Barral-Moran, M.J., Calaora, V., Vutskits, L., Wang, C., (1997) Neuroprotective effects of group III mGluR in trau-
Zhang, H., Durbec, P., Rougon, G. and Kiss, J.Z. (2003) matic neuronal injury. J. Neurotrauma, 14(12): 885–895.
Oligodendrocyte progenitor migration in response to injury Faden, A.I., Knoblach, S.M., Movsesyan, V.A. and Cernak, I.
of glial monolayers requires the polysialic neural cell- (2004) Novel small peptides with neuroprotective and noo-
adhesion molecule. J. Neurosci. Res., 72(6): 679–690. tropic properties. J. Alzheimers Dis., 6(Suppl 6): S93–S97.
Cargill II, R.S. and Thibault, L.E. (1996) Acute alterations in Faden, A.I., Movsesyan, V.A., Knoblach, S.M., Ahmed, F. and
[Ca2+]i in NG108-15 cells subjected to high strain rate de- Cernak, I. (2005) Neuroprotective effects of novel small pep-
formation and chemical hypoxia: an in vitro model for neural tides in vitro and after brain injury. Neuropharmacology,
trauma. J. Neurotrauma, 13(7): 395–407. 49(3): 410–424.
Cater, H.L., Sundstrom, L.E. and Morrison III, B. (2006) Faden, A.I., O’Leary, D.M., Fan, L., Bao, W., Mullins, P.G.
Temporal development of hippocampal cell death is and Movsesyan, V.A. (2001) Selective blockade of the
37

mGluR1 receptor reduces traumatic neuronal injury in vitro Graham, D.I., Adams, J.H., Nicoll, J.A., Maxwell, W.L. and
and improves outcome after brain trauma. Exp. Neurol., Gennarelli, T.A. (1995) The nature, distribution and causes
167(2): 435–444. of traumatic brain injury. Brain Pathol., 5(4): 397–406.
Farkas, O., Lifshitz, J. and Povlishock, J.T. (2006) Mechanop- Gross, G.W., Lucas, J.H. and Higgins, M.L. (1983) Laser mi-
oration induced by diffuse traumatic brain injury: an irre- crobeam surgery: ultrastructural changes associated with
versible or reversible response to injury? J. Neurosci., 26(12): neurite transection in culture. J. Neurosci., 3(10): 1979–1993.
3130–3140. Gubellini, P., Pisani, A., Centonze, D., Bernardi, G. and Ca-
Fitch, M.T., Doller, C., Combs, C.K., Landreth, G.E. and labresi, P. (2004) Metabotropic glutamate receptors and
Silver, J. (1999) Cellular and molecular mechanisms of glial striatal synaptic plasticity: implications for neurological dis-
scarring and progressive cavitation: in vivo and in vitro anal- eases. Prog. Neurobiol., 74(5): 271–300.
ysis of inflammation-induced secondary injury after CNS Gutierrez, E., Huang, Y., Haglid, K., Bao, F., Hansson, H.A.,
trauma. J. Neurosci., 19(19): 8182–8198. Hamberger, A. and Viano, D. (2001) A new model for diffuse
Floyd, C.L., Gorin, F.A. and Lyeth, B.G. (2005) Mechanical brain injury by rotational acceleration: I model, gross appear-
strain injury increases intracellular sodium and reverses Na+/ ance, and astrocytosis. J. Neurotrauma, 18(3): 247–257.
Ca2+ exchange in cortical astrocytes. Glia, 51(1): 35–46. Hardingham, G.E. and Bading, H. (2003) The Yin and Yang of
Floyd, C.L., Rzigalinski, B.A., Sitterding, H.A., Willoughby, NMDA receptor signalling. Trends Neurosci., 26(2): 81–89.
K.A. and Ellis, E.F. (2004) Antagonism of group I metabo- Hoffman, S.W., Rzigalinski, B.A., Willoughby, K.A. and Ellis,
tropic glutamate receptors and PLC attenuates increases in E.F. (2000) Astrocytes generate isoprostanes in response to
inositol trisphosphate and reduces reactive gliosis in strain- trauma or oxygen radicals. J. Neurotrauma, 17(5): 415–420.
injured astrocytes. J. Neurotrauma, 21(2): 205–216. Iwata, A., Stys, P.K., Wolf, J.A., Chen, X.H., Taylor, A.G.,
Floyd, C.L., Rzigalinski, B.A., Weber, J.T., Sitterding, H.A., Meaney, D.F. and Smith, D.H. (2004) Traumatic axonal in-
Willoughby, K.A. and Ellis, E.F. (2001) Traumatic injury jury induces proteolytic cleavage of the voltage-gated sodium
of cultured astrocytes alters inositol (1,4,5)-trisphosphate- channels modulated by tetrodotoxin and protease inhibitors.
mediated signaling. Glia, 33(1): 12–23. J. Neurosci., 24(19): 4605–4613.
Gagliardi, R.J. (2000) Neuroprotection, excitotoxicity and Kao, C.Q., Goforth, P.B., Ellis, E.F. and Satin, L.S. (2004)
NMDA antagonists. Arq. Neuropsiquiatr., 58(2B): 583–588. Potentiation of GABA(A) currents after mechanical injury of
Geddes, D.M. and Cargill II, R.S. (2001) An in vitro model of cortical neurons. J. Neurotrauma, 21(3): 259–270.
neural trauma: device characterization and calcium response Katano, H., Fujita, K., Kato, T., Asai, K., Kawamura, Y.,
to mechanical stretch. J. Biomech. Eng., 123(3): 247–255. Masago, A. and Yamada, K. (1999) Traumatic injury in vitro
Geddes, D.M., LaPlaca, M.C. and Cargill II, R.S. (2003) Sus- induces IEG mRNAs in cultured glial cells, suppressed by co-
ceptibility of hippocampal neurons to mechanically induced culture with neurons. Neuroreport, 10(12): 2439–2448.
injury. Exp. Neurol., 184(1): 420–427. Knoblach, S.M., Alroy, D.A., Nikolaeva, M., Cernak, I.,
Geddes-Klein, D.M., Schiffman, K.B. and Meaney, D.F. Stoica, B.A. and Faden, A.I. (2004) Caspase inhibitor
(2006a) Mechanisms and consequences of neuronal stretch z-DEVD-fmk attenuates calpain and necrotic cell death in
injury in vitro differ with the model of trauma. J. Neuro- vitro and after traumatic brain injury. J. Cereb. Blood Flow
trauma, 23(2): 193–204. Metab., 24(10): 1119–1132.
Geddes-Klein, D.M., Serbest, G., Mesfin, M.N., Cohen, A.S. Lamb, R.G., Harper, C.C., McKinney, J.S., Rzigalinski, B.A.
and Meaney, D.F. (2006b) Pharmacologically induced cal- and Ellis, E.F. (1997) Alterations in phosphatidylcholine me-
cium oscillations protect neurons from increases in cytosolic tabolism of stretch-injured cultured rat astrocytes. J. Neuro-
calcium after trauma. J. Neurochem., 97(2): 462–474. chem., 68(5): 1904–1910.
Gennarelli, T.A. (1994) Animate models of human head injury. Langlois, J.A. and Sattin, R.W. (2005) Traumatic brain injury
J. Neurotrauma, 11(4): 357–368. in the United States: research and programs of the Centers
Gennarelli, T.A., Spielman, G.M., Langfitt, T.W., Gildenberg, for Disease Control and Prevention (CDC). J. Head Trauma
P.L., Harrington, T., Jane, J.A., Marshall, L.F., Miller, J.D. Rehabil., 20(3): 187–188.
and Pitts, L.H. (1982a) Influence of the type of intracranial LaPlaca, M.C., Cullen, D.K., McLoughlin, J.J. and Cargill II,
lesion on outcome from severe head injury. J. Neurosurg., 56: R.S. (2005) High rate shear strain of three-dimensional neu-
26–32. ral cell cultures: a new in vitro traumatic brain injury model.
Gennarelli, T.A., Thibault, L.E., Adams, J.H., Graham, D.I., J. Biomech., 38(5): 1093–1105.
Thompson, C.J. and Marcincin, R.P. (1982b) Diffuse axonal LaPlaca, M.C., Lee, V.M. and Thibault, L.E. (1997) An in vitro
injury and traumatic coma in the primate. Ann. Neurol., 12: model of traumatic neuronal injury: loading rate-dependent
564–574. changes in acute cytosolic calcium and lactate dehydrogenase
Goforth, P.B., Ellis, E.F. and Satin, L.S. (1999) Enhancement release. J. Neurotrauma, 14(6): 355–368.
of AMPA-mediated current after traumatic injury in cortical Lighthall, J.W. (1988) Controlled cortical impact: a new experi-
neurons. J. Neurosci., 19(17): 7367–7374. mental brain injury model. J. Neurotrauma, 5: 1–15.
Goforth, P.B., Ellis, E.F. and Satin, L.S. (2004) Mechanical in- Lucas, J.H. (1987) Proximal segment retraction increases the
jury modulates AMPA receptor kinetics via an NMDA probability of nerve cell survival after dendrite transection.
receptor-dependent pathway. J. Neurotrauma, 21(6): 719–732. Brain Res., 425(2): 384–387.
38

Lucas, J.H., Gross, G.W., Emery, D.G. and Gardner, C.R. injury in vitro and in vivo by antagonizing N-methyl-D-aspart-
(1985) Neuronal survival or death after dendrite transection ate receptors. J. Pharmacol. Exp. Ther., 296(1): 41–47.
close to the perikaryon: correlation with electrophysiologic, Mukhin, A.G., Ivanova, S.A., Allen, J.W. and Faden, A.I.
morphologic, and ultrastructural changes. Cent. Nerv. Syst. (1998) Mechanical injury to neuronal/glial cultures in micro-
Trauma, 2(4): 231–255. plates: role of NMDA receptors and pH in secondary neu-
Lusardi, T.A., Rangan, J., Sun, D., Smith, D.H. and Meaney, ronal cell death. J. Neurosci. Res., 51(6): 748–758.
D.F. (2004) A device to study the initiation and propagation Mukhin, A.G., Ivanova, S.A., Knoblach, S.M. and Faden, A.I.
of calcium transients in cultured neurons after mechanical (1997) New in vitro model of traumatic neuronal injury:
stretch. Ann. Biomed. Eng., 32(11): 1546–1558. evaluation of secondary injury and glutamate receptor-me-
Marmarou, A., Foda, M.A., van den Brink, W., Campbell, J., diated neurotoxicity. J. Neurotrauma, 14(9): 651–663.
Kita, H. and Demetriadou, K. (1994) A new model of diffuse Nahum, A.M., Smith, R. and Ward, C.C. (1977) Intracranial
brain injury in rats. Part I: Pathophysiology and biomechan- pressure dynamics during head impact. In: 21st Annual Stapp
ics. J. Neurosurg., 80(2): 291–300. Car Crash Conference, Society of Automotive Engineers. San
Maxwell, W.L., Povlishock, J.T. and Graham, D.L. (1997) A Diego, CA.
mechanistic analysis of nondisruptive axonal injury: a review. Neary, J.T., Kang, Y., Willoughby, K.A. and Ellis, E.F. (2003)
J. Neurotrauma, 14(7): 419–440. Activation of extracellular signal-regulated kinase by stretch-
McIntosh, T.K., Vink, R., Noble, L., Yamakami, I., Fernyak, induced injury in astrocytes involves extracellular ATP and
S., Soares, H. and Faden, A.L. (1989a) Traumatic brain in- P2 purinergic receptors. J. Neurosci., 23(6): 2348–2356.
jury in the rat: characterization of a lateral fluid-percussion Nusholtz, G.S., Wylie, E.B. and Glascoe, L.G. (1995) Internal
model. Neuroscience, 28(1): 233–244. cavitation in simple head impact model. J. Neurotrauma,
McIntosh, T.K., Vink, R., Yamakami, I. and Faden, A.I. 12(4): 707–714.
(1989b) Magnesium protects against neurological deficit after O’Dell, D.M., Raghupathi, R., Crino, P.B., Eberwine, J.H.
brain injury. Brain Res., 482(2): 252–260. and McIntosh, T.K. (2000) Traumatic brain injury alters
McKinney, J.S., Willoughby, K.A., Liang, S. and Ellis, E.F. the molecular fingerprint of TUNEL-positive cortical neurons
(1996) Stretch-induced injury of cultured neuronal, glial, and in vivo: a single-cell analysis. J. Neurosci., 20(13): 4821–4828.
endothelial cells. Effect of polyethylene glycol-conjugated Ommaya, A.K., Yarnell, P., Hirsch, A.E. and Harris, E.H.
superoxide dismutase. Stroke, 27(5): 934–940. (1967) Scaling of experimental data on cerebral concussion in
Meaney, D.F., Smith, D.H., Shreiber, D.I., Bain, A.C., Miller, sub-human primates to concussion threshold for man. In:
R.T., Ross, D.T. and Gennarelli, T.A. (1995) Biomechanical 13th Stapp Car Crash Conference, Anaheim, CA, Society of
analysis of experimental diffuse axonal injury. J. Neurotrau- Automotive Engineers.
ma, 12(4): 689–694. Pettus, E.H., Christman, C.W., Giebel, M.L. and Povlishock,
Mori, T., Wang, X., Jung, J.C., Sumii, T., Singhal, A.B., Fini, J.T. (1994) Traumatically induced altered membrane perme-
M.E., Dixon, C.E., Alessandrini, A. and Lo, E.H. (2002) ability: its relationship to traumatically induced reactive axo-
Mitogen-activated protein kinase inhibition in traumatic nal change. J. Neurotrauma, 11(5): 507–522.
brain injury: in vitro and in vivo effects. J. Cereb. Blood Flow Pike, B.R., Zhao, X., Newcomb, J.K., Glenn, C.C., Anderson,
Metab., 22(4): 444–452. D.K. and Hayes, R.L. (2000) Stretch injury causes calpain
Morrison III, B., Eberwine, J.H., Meaney, D.F. and McIntosh, and caspase-3 activation and necrotic and apoptotic cell
T.K. (2000) Traumatic injury induces differential expression death in septo-hippocampal cell cultures. J. Neurotrauma,
of cell death genes in organotypic brain slice cultures deter- 17(4): 283–298.
mined by complementary DNA array hybridization. Neuro- Pineda, J.A., Wang, K.K. and Hayes, R.L. (2004) Biomarkers
science, 96(1): 131–139. of proteolytic damage following traumatic brain injury. Brain
Morrison III, B., Meaney, D.F. and McIntosh, T.K. (1998a) Pathol., 14(2): 202–209.
Mechanical characterization of an in vitro device designed to Povlishock, J.T. and Katz, D.I. (2005) Update of neuropathol-
quantitatively injure living brain tissue. Ann. Biomed. Eng., ogy and neurological recovery after traumatic brain injury.
26(3): 381–390. J. Head Trauma Rehabil., 20(1): 76–94.
Morrison III, B., Saatman, K.E., Meaney, D.F. and McIntosh, Prado, G.R., Ross, J.D., DeWeerth, S.P. and LaPlaca, M.C.
T.K. (1998b) In vitro central nervous system models of (2005) Mechanical trauma induces immediate changes in
mechanically induced trauma: a review. J. Neurotrauma, neuronal network activity. J. Neural Eng., 2(4): 148–158.
15(11): 911–928. Prange, M.T. and Margulies, S.S. (2002) Regional, directional,
Movsesyan, V.A. and Faden, A.I. (2006) Neuroprotective and age-dependent properties of the brain undergoing large
effects of selective group II mGluR activation in brain deformation. J. Biomech. Eng., 124(2): 244–252.
trauma and traumatic neuronal injury. J. Neurotrauma, Raghupathi, R., Grants, I., Rosenberg, L.J., McIntosh, T.K.
23(2): 117–127. and Lucas, J.H. (1998) Increased jun immunoreactivity in an
Movsesyan, V.A., O’Leary, D.M., Fan, L., Bao, W., Mullins, in vitro model of mammalian spinal neuron physical injury.
P.G., Knoblach, S.M. and Faden, A.I. (2001) mGluR5 J. Neurotrauma, 15(7): 555–561.
antagonists 2-methyl-6-(phenylethynyl)-pyridine and (E)-2- Reeves, T.M., Phillips, L.L. and Povlishock, J.T. (2005)
methyl-6-(2-phenylethenyl)-pyridine reduce traumatic neuronal Myelinated and unmyelinated axons of the corpus callosum
39

differ in vulnerability and functional recovery following Tavalin, S.J., Ellis, E.F. and Satin, L.S. (1995) Mechanical
traumatic brain injury. Exp. Neurol., 196(1): 126–137. perturbation of cultured cortical neurons reveals a stretch-
Regan, R.F. and Choi, D.W. (1994) The effect of NMDA, induced delayed depolarization. J. Neurophysiol., 74(6):
AMPA/kainate, and calcium channel antagonists on trau- 2767–2773.
matic cortical neuronal injury in culture. Brain Res., 633(1–2): Tavalin, S.J., Ellis, E.F. and Satin, L.S. (1997) Inhibition of
236–242. the electrogenic Na pump underlies delayed depolarization
Regan, R.F. and Panter, S.S. (1995) Traumatic neuronal injury of cortical neurons after mechanical injury or glutamate.
in cortical cell culture is attenuated by 21-aminosteroids. J. Neurophysiol., 77(2): 632–638.
Brain Res., 682(1–2): 144–150. Tecoma, E.S., Monyer, H., Goldberg, M.P. and Choi, D.W.
Rzigalinski, B.A., Liang, S., McKinney, J.S., Willoughby, K.A. (1989) Traumatic neuronal injury in vitro is attenuated by
and Ellis, E.F. (1997) Effect of Ca2+ on in vitro astrocyte NMDA antagonists. Neuron, 2(6): 1541–1545.
injury. J. Neurochem., 68(1): 289–296. Thibault, L.E., Meaney, D.F., Anderson, B.J. and Marmarou,
Rzigalinski, B.A., Weber, J.T., Willoughby, K.A. and Ellis, A. (1992) Biomechanical aspects of a fluid percussion model
E.F. (1998) Intracellular free calcium dynamics in stretch- of brain injuiry. J. Neurotrauma, 9(4): 311–322.
injured astrocytes. J. Neurochem., 70(6): 2377–2385. Thurman, D.J., Alverson, C., Dunn, K.A., Guerrero, J. and
Serbest, G., Horwitz, J., Jost, M. and Barbee, K. (2006) Mech- Sniezek, J.E. (1999) Traumatic brain injury in the United
anisms of cell death and neuroprotection by poloxamer 188 States: a public health perspective. J. Head Trauma Rehabil.,
after mechanical trauma. FASEB J., 20(2): 308–310. 14(6): 602–615.
Shah, P.T., Yoon, K.W., Xu, X.M. and Broder, L.D. (1997) Wang, X., Mori, T., Jung, J.C., Fini, M.E. and Lo, E.H. (2002)
Apoptosis mediates cell death following traumatic injury in Secretion of matrix metalloproteinase-2 and -9 after mechan-
rat hippocampal neurons. Neuroscience, 79(4): 999–1004. ical trauma injury in rat cortical cultures and involvement of
Shreiber, D., Bain, A.C. and Meaney, D.F. (1997) In vivo MAP kinase. J. Neurotrauma, 19(5): 615–625.
thresholds for mechanical injury to the blood brain barrier. J. Weber, J.T. (2004) Calcium homeostasis following traumatic
Passenger Cars, 106: 3792–3806. neuronal injury. Curr. Neurovasc. Res., 1(2): 151–171.
Sieg, F., Wahle, P. and Pape, H.C. (1999) Cellular reactivity to Weber, J.T., Rzigalinski, B.A. and Ellis, E.F. (2001) Traumatic
mechanical axonal injury in an organotypic in vitro model of injury of cortical neurons causes changes in intracellular
neurotrauma. J. Neurotrauma, 16(12): 1197–1213. calcium stores and capacitative calcium influx. J. Biol.
Simonyi, A., Schachtman, T.R. and Christoffersen, G.R. (2005) Chem., 276(3): 1800–1807.
The role of metabotropic glutamate receptor 5 in learning Weber, J.T., Rzigalinski, B.A., Willoughby, K.A., Moore, S.F.
and memory processes. Drug News Perspect., 18(6): 353–361. and Ellis, E.F. (1999) Alterations in calcium-mediated signal
Singleton, R.H., Zhu, J., Stone, J.R. and Povlishock, J.T. (2002) transduction after traumatic injury of cortical neurons. Cell
Traumatically induced axotomy adjacent to the soma does not Calcium, 26(6): 289–299.
result in acute neuronal death. J. Neurosci., 22(3): 791–802. Willoughby, K.A., Kleindienst, A., Muller, C., Chen, T., Muir,
Slemmer, J.E., Matser, E.J., De Zeeuw, C.I. and Weber, J.T. J.K. and Ellis, E.F. (2004) S100B protein is released by in
(2002) Repeated mild injury causes cumulative damage to vitro trauma and reduces delayed neuronal injury. J. Neuro-
hippocampal cells. Brain, 125(Pt 12): 2699–2709. chem., 91(6): 1284–1291.
Smith, D.H., Soares, H.D., Pierce, J.S., Pearlman, K., Saatman, Wolf, J.A., Stys, P.K., Lusardi, T., Meaney, D. and Smith, D.H.
K.E., Meaney, D.F., Dixon, C.E. and McIntosh, T.K. (1995) (2001) Traumatic axonal injury induces calcium influx mod-
A model of parasagittal controlled cortical impact in the ulated by tetrodotoxin-sensitive sodium channels. J. Neuro-
mouse: cognitive and histopathologic effects. J. Neurotrauma, sci., 21(6): 1923–1930.
12(2): 169–178. Xiao-Sheng, H., Sheng-Yu, Y., Xiang, Z., Zhou, F. and Jian-
Smith, D.H., Wolf, J.A., Lusardi, T.A., Lee, V.M. and Meaney, ning, Z. (2000) Diffuse axonal injury due to lateral head ro-
D.F. (1999) High tolerance and delayed elastic response of tation in a rat model. J. Neurosurg., 93(4): 626–633.
cultured axons to dynamic stretch injury. J. Neurosci., 19(11): Yang, X.F., Cao, F., Pan, D.S., Liu, W.G., Hu, W.W., Zheng,
4263–4269. X.J., Zhao, X.Q. and Lu, S.T. (2006) Establishment of a
Stalhammar, D. and Olsson, Y. (1975) Experimental brain mechanical injury model of rat hippocampal neurons in vitro.
damage from fluid pressures due to impact acceleration. Acta Chin. J. Traumatol., 9(1): 29–33.
Neurol. Scand., 52: 38–55. Zhang, L., Rzigalinski, B.A., Ellis, E.F. and Satin, L.S. (1996)
Stone, J.R., Okonkwo, D.O., Dialo, A.O., Rubin, D.G., Mutlu, Reduction of voltage-dependent Mg2+ blockade of NMDA
L.K., Povlishock, J.T. and Helm, G.A. (2004) Impaired ax- current in mechanically injured neurons. Science, 274(5294):
onal transport and altered axolemmal permeability occur in 1921–1923.
distinct populations of damaged axons following traumatic Zhang, L., Yang, K.H. and King, A.I. (2004) A proposed injury
brain injury. Exp. Neurol., 190(1): 59–69. threshold for mild traumatic brain injury. J. Biomech. Eng.,
Sullivan, H.G., Martinez, J., Becker, D.P., Miller, J.D., 126(2): 226–236.
Griffith, R. and Wist, A.O. (1976) Fluid-percussion model Zhu, Q., Prange, M. and Margulies, S. (2006) Predicting un-
of mechanical brain injury in the cat. J. Neurosurg., 45(5): consciousness from a pediatric brain injury threshold. Dev.
521–534. Neurosci., 28(4–5): 388–395.
SECTION III

Pathological Mechanisms of Injury


This page intentionally left blank
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 4

Cellular and subcellular change evoked by diffuse


traumatic brain injury: a complex web of change
extending far beyond focal damage

Orsolya Farkas and John T. Povlishock

Department of Anatomy and Neurobiology, Medical College of Virginia Campus, Virginia Commonwealth University,
P.O. Box 980709, Richmond, VA 23298, USA

Abstract: Until recently, our understanding of the cellular and subcellular changes evoked by diffuse
traumatic brain injury has been framed in the context of primary focal injury. In this regard, the ensuing
cell death cascades were linked to contusional-mediated changes associated with frank hemorrhage and
ischemia, and these were assumed to contribute to the observed apoptotic and necrotic neuronal death.
Little consideration was given to the potential that other non-contusional cell death cascades could
have been triggered by the diffuse mechanical forces of injury. While the importance of these classical,
contusion-related apoptotic and necrotic cell death cascades cannot be discounted with diffuse injury,
more recent information suggests that the mechanical force of injury itself can diffusely porate the
neuronal plasmalemma and its axolemmal membranes, evoking other forms of cellular response that can
contribute to cell injury or death. In this regard, the duration of the membrane alteration appears to be a
dependent factor, with enduring membrane change, potentially leading to irreversible damage, whereas
more transient membrane perturbation can be followed by cell membrane resealing associated with
recovery and/or adaptive change. With more enduring mechanical membrane perturbation, it appears
that some of the traditional death cascades involving the activation of cysteine proteases are at work.
Equally important, non-traditional pathways involving the lysosomal dependent release of hydrolytic
enzymes may also be players in the ensuing neuronal death. These mechanically related factors that
directly impact upon the neuronal somata may also be influenced by concomitant and/or secondary
axotomy-mediated responses. This axonal injury, although once thought to involve a singular intraaxo-
nal response to injury, is now known to be more complex, reflecting differential responses to injuries of
varying severity. Moreover, it now appears that fiber size and type may also influence the axon’s reaction
to injury. In sum, this review explicates the complexity of the cellular and subcellular responses evoked by
diffuse traumatic brain injury in both the neuronal somata and its axonal appendages. This review
further illustrates that our once simplistic views framed by evidence based upon contusional and/or
ischemic change do not fully explain the complex repertoire of change evoked by diffuse traumatic brain
injury.

Keywords: diffuse traumatic brain injury; neuronal injury; axonal injury; necrosis; apoptosis

Corresponding author. E-mail: jtpovlis@vcu.edu

DOI: 10.1016/S0079-6123(06)61004-2 43
44

Introduction appreciated phenomenon of diffuse traumatic


neuronal change.
Over the last 40 years, our understanding of the
complex pathobiology of traumatic brain injury
(TBI) has improved significantly. From both the Morphological characteristics of neuronal injury
clinical and basic science perspective, now most after DTBI
consider the pathobiology of TBI in the context of
focal as well as diffuse change (Povlishock and As noted above, unlike the wide body of literature
Katz, 2005). It is also recognized in the clinical on neuronal death after focal TBI, there are rela-
setting that focal injuries are typically embedded tively few descriptions of neuronal injury and
within concomitant diffuse pathologies, with the death occurring remote from contusional regions
diffuse pathologies being the major determinants (Dietrich et al., 1994a, b; Colicos et al., 1996;
of the adverse outcomes associated with TBI Hicks et al., 1996), or after DTBI (Smith et al.,
(Povlishock and Katz, 2005). Despite this recogni- 1997; Runnerstam et al., 2001; Cernak et al.,
tion of the importance of diffuse change, a dispro- 2002). These studies, similar to those focusing on
portionate number of basic science studies have focal TBI, describe two distinct forms of cell
focused on focal injuries, such as contusional change death, namely necrosis and apoptosis, within the
without a parallel consideration of any diffuse neocortex, hippocampus and diencephalon. With
changes that accompany the injury. In this review, diffuse injury, damaged/dying neurons have been
we attempt to address what is known as well as described not only in the neocortex in or near
what is controversial in our understanding of diffuse contusions, but also in regions remote from the
change within the traumatically injured brain paren- impact/contusional site, such as the C1, C2, C3
chyma, examining both the cellular and subcellular layers and the dentate hilus of hippocampus and
responses evoked by diffuse traumatic brain injury thalamus. Additional neuronal damage/death has
(DTBI) to the neurons and their axonal extensions. been described scattered in the caudate/putamen
In this review, we attempt also to consider new and the inferior and superior colliculi, consistent
experimental directions for this important area of with the diffuse nature of the injury. Like previ-
scientific inquiry. ous descriptions of necrosis, these damaged neu-
rons were dark and shrunken with Nissl staining
(Dietrich et al., 1994b; Hicks et al., 1996) or
eosinophilic with acid fuchsin staining (Cortez
Neuronal damage associated with DTBI et al., 1989; Dietrich et al., 1994a; Hicks et al.,
1996), revealing distorted profiles and vacuolizat-
Neuronal damage and death associated with several ion. At the ultrastructural level, such neurons
types of brain injury including TBI in humans and demonstrated increased cytoplasmic and nuclear
animals have been widely studied and, as such, have electron density, swollen mitochondria, vacuolated
been the focus of several major reviews (Adams cytoplasm, plasma membrane disruption, pyknotic
et al., 1980; Cervos-Navarro and Lafuente, 1991; nuclei and perisomatic glial swelling (Dietrich et al.,
Kotapka et al., 1992, 1993; Ross et al., 1993; 1994b). Importantly, neuronal necrosis has also
Kermer et al., 1999; Raghupathi, 2004; Yakovlev been observed after DTBI in the absence of any
and Faden, 2004). The majority of these studies and focal lesion, such as contusion or hematoma for-
reviews, however, have focused on focal injuries, mation. Specifically, neuronal necrosis bilaterally
addressing neuronal death in localized contusional scattered in the neocortex (Singleton et al., 2002;
or pericontusional regions. Because all these studies Singleton and Povlishock, 2004; Farkas et al., 2006)
provided excellent, detailed descriptions of the as well as in the cerebellum, and the C1 and C3
necrotic and apoptotic neuronal change associ- pyramidal layers and dentate hilus of the hippo-
ated with focal TBI, the present review follows a campus has been described (Kotapka et al., 1991;
different track, focusing primarily on the under Smith et al., 1997; Singleton et al., 2002; Singleton
45

and Povlishock, 2004). While this limited literature appreciated. It is believed that the balance between
supports the occurrence necrotic cell death with the anti- and pro-apoptotic signals determines if
DTBI, to date, the mechanisms underlying this the injured neuron dies or survives. At present,
necrosis are not well understood. The presence of apoptotic cell death is considered to play a role in
necrotic neurons remote from or in the absence of delayed neuronal death occurring several hours to
contusion suggests that the pathogenesis of this ne- weeks after DTBI, when the dominance of pro-
crosis does not follow from any overt destructive or apoptotic factors in the presence of a persistent
ischemic processes comparable to those occurring energy supply results in the activation of cysteine
within contusional foci. Unfortunately, our current proteases, such as caspases that are regulators and
beliefs regarding the pathogenesis of DTBI-induced effectors of apoptotic cell death.
necrotic change have been biased by the ischemic
literature, wherein disproportionate emphasis has
been placed on membrane pump failure and sub-
sequent ionic dysregulation and calpain-mediated Mechanisms eliciting neuronal injury and death
proteolysis. It is likely, however, that other poten- after DTBI
tially important factors and mechanisms associated
with direct mechanical perturbation and its sequel- Similar to their morphological differences, apop-
ae may be at work in the pathogenesis of this tosis and necrosis have also been distinguished by
diffuse necrotic neuronal death and, as such, merit the differences in their pathological mechanisms.
consideration (vide infra). As noted, apoptosis is an active process requir-
Like necrosis, scattered apoptosis has also ing energy while necrosis is a passive event that
been described following DTBI. Although under- results from energy failure and consequent loss of
appreciated and originally thought to take place ionic homeostasis with the ultimate recruitment of
only in ‘‘physiologic’’ programmed neuronal death an inflammatory response. Although both proc-
processes and/or slowly progressive neurodegene- esses were once considered distinct, recent evi-
rative diseases, its role in the pathobiology of dence demonstrates that necrosis and apoptosis
DTBI has become more apparent. Rink et al. can evolve in parallel in the same injured tissue.
(1995) first identified this process in TBI, positing Further, it has been recently suggested based upon
that it played a role in delayed neuronal damage the cellular microenvironment and energy supply
in contrast to necrosis that was linked to both that a switch between necrosis and apoptosis can
acute and delayed neuronal death. Neurons with occur within an individual cell (Kermer et al., 1999;
apoptotic morphology have been described in con- Kitanaka and Kuchino, 1999; Nicotera et al., 1999;
tusional foci, but more commonly have been iden- Raghupathi, 2004). This hybrid form of cell death,
tified either in the absence of contusions or in sometimes referred as aponecrosis, reflects the fact
foci remote from contusional change, such as the that these cells show the morphological character-
hippocampus and/or diencephalon (Colicos and istics of both types of cell death (Formigli et al.,
Dash, 1996; Colicos et al., 1996; Pravdenkova et al., 2000). To date, multiple reviews have considered
1996; Conti et al., 1998; Fox et al., 1998; Newcomb those cellular, subcellular and pathophysiological
et al., 1999; Lin et al., 2001; Runnerstam et al., cascades that may contribute to the above described
2001; Cernak et al., 2002, 2004; Raghupathi et al., necrotic and apoptotic neuronal cell death cascades
2002). At ultrastructural level, these apoptotic (Kermer et al., 1999; Raghupathi et al., 2000; Zipfel
neurons demonstrated cytoplasmic condensation, et al., 2000; Keane et al., 2001; Raghupathi, 2004;
nuclear pyknosis, chromatin condensation, cell Yakovlev and Faden, 2004). Accordingly, it is un-
rounding, membrane blebbing, cytoskeletal colla- necessaryto revisit these details. Rather, we provide
pse and, ultimately, disintegration of the cell into here only a summary of these well-characterized
small fragments forming apoptotic bodies. Similar processes, while providing, in subsequent passages,
to our understanding of necrosis, the pathogenesis more detail on other not well appreciated mecha-
of apoptosis occurring after DTBI is not well nisms related to the pathogenesis of DTBI.
46

Neuroexcitation and calcium dysregulation peroxidation, protein nitrosylation and DNA


degradation (Braughler and Hall, 1992).
Calcium dysregulation has been linked to both
necrotic and apoptotic processes, although differ-
ent intracellular cascades are involved. To date, Calpains and caspases
it is known that the traumatic episode can be
associated with generalized neuroexcitotoxicity, As noted above, increased intracellular Ca2+ can
involving excitatory amino acid (EAA) release activate proteases, such as calpain. Calpain, a
that can trigger several subcellular alterations via member of cysteine proteases, and its activation
Na+ and Ca2+ influx from the extracellular space. are well known to be associated with several
Using microdialysis, the EAA glutamate has been different types of brain injury, including DTBI
found to be elevated after TBI both in animals and (Kupina et al., 2001, 2002; Buki et al., 2003;
humans. While this glutamate elevation initially Farkas et al., 2006). It has several different iso-
was described after focal TBI (Faden et al., 1989; types, including m-calpain and m-calpain that are
Katayama et al., 1990; Palmer et al., 1993), recent ubiquitous in the central nervous system. Although
studies also showed the involvement of EAA calpains have several different substrates within the
release after DTBI (Runnerstam et al., 2001; Goda cell, such as cytoskeletal elements, neurofilaments,
et al., 2002; Fei et al., 2005). The subsequent mas- protein kinase C, calmodulin binding proteins and
sive Ca2+ influx via glutamate/NMDA channels transcription factors (for review, see Wang (2000);
triggers further Ca2+ release from intracellular Huang and Wang (2001)), one of the most widely
stores and thereby dramatically elevates free intra- investigated of the calpain substrates is spectrin.
cellular Ca2+. This increased cytosolic Ca2+ can Spectrin is the primary component of the neuronal
lead to activation of proteases, such as calpains, cytoskeleton and its cleavage by calpain results
phosphatases, protein kinases or nitric oxide synt- in specific and stable breakdown products, such as
hase, all which have been linked in various scenar- 150 and 145 kDa spectrin breakdown products
ios to either necrotic or apoptotic cell death (SBDPs). The presence of calpain-specific SBDPs
(Kermer et al., 1999). as a result of calpain-mediated spectrin proteolysis
(CMSP) has been routinely observed both after
experimental (Kampfl et al., 1996; Saatman et al.,
Free radicals 1996; Pike et al., 1998, 2001; Farkas et al., 2006)
and human TBI (McCracken et al., 1999; Pineda
Concurrent with and influenced by the glutamate et al., 2004; Farkas et al., 2005), and now is being
NMDA receptor activation and Ca2+ dysregula- used as one of the markers of Ca2+-induced cell
tion described above, the generation of oxygen injury and death. Excessive Ca2+ influx and the
free radicals can also occur after DTBI. Either concomitant calpain activation lead to the proteoly-
via the generation of nitric oxide, the accelerated tic degradation of intracellular proteins and mem-
metabolism of arachidonate acid via multiple branes. The ensuing cytoskeletal and membrane
pathways or the generation of superoxide anions damage can destroy cell integrity, cause increased
via an evoked inflammatory response, destruc- membrane permeability, compromise the transport
tive radical species can be generated with lethal of essential cell products, induce aberrant signaling
results for the brain (Kontos and Povlishock, cascades and finally lead to cell death (Kermer
1986; Povlishock and Kontos, 1992). With more et al., 1999). Although calpain activation can lead
localized radical generation and Ca2+ dysregula- to either necrotic or apoptotic change, most concur
tion, this creates the permissive environment for that in TBI it is primarily responsible for necrotic
mitochondrial damage, cytochrome C release and cell death (Wang, 2000; Raghupathi, 2004).
apoptotic cell death. With more generalized radi- Like calpains, caspases are also members of the
cal generation, the oxygen radicals participate cysteine protease family, with these proteases
in highly destructive processes including lipid thought to play a major role in apoptotic cell
47

death (Eldadah and Faden, 2000; Wang, 2000; Pro-apoptotic Bax overexpression has been
Raghupathi, 2004). Caspase-3, the common apop- demonstrated after DTBI (Cernak et al., 2002).
tosis effector, is activated via two major pathways, Concomitant decrease in the expression of the
either the mitochondrial intrinsic or the death anti-apoptotic Bcl-2 and BclXL has also been
receptor, extrinsic pathways. Briefly, the apoptotic reported (Felderhoff-Mueser et al., 2002). How-
stimuli can result in the permeabilization of the ever, some studies have demonstrated increased
mitochondrial membrane via different pathways, Bcl-2 expression after DTBI (Cernak et al., 2002).
subsequently triggering the release of cytocrome-c Overall, it has been suggested that the shift between
into the cytoplasm. Cytocrome-c binds to apopto- pro- and anti-apoptotic oncogene expression regu-
sis activating protein-1 (Apaf-1) and the complex lates the fate of the injured neuron, resulting in cell
activates caspase-9, which then activates caspase-3 death if the expression of pro-apoptotic proteins
via cleaving its proenzyme form. During the extrin- exceeds the expression of anti-apoptotic proteins,
sic pathways, apoptotic stimuli activate the death while promoting cell survival if the anti-apoptotic
receptors, such as TNFa1-receptor or Fas receptor. Bcl protein expression is upregulated over the pro-
The receptors recruit adapter proteins and form a apoptotic members of the Bcl-family (Merry and
complex, which then induces the autolytic activa- Korsmeyer, 1997).
tion of caspase-8, which activates caspase-3 (for
review, see Wang (2000); Yakovlev and Faden
(2004)). Caspase-3, similar to calpain, cleaves spec- Role of hydrolytic enzymes
trin in a specific manner resulting in a 120 kDa
SBDP that can be used for the specific detection In the previous passages, attention has been focused
of caspase-3 activity and apoptosis after TBI. The on those pathways that have received emphasis in
activation of caspase-3 has been observed in wide- terms of the apoptotic/necrotic pathways occurring
spread regions both after focal and DTBI (Pike with TBI. While it is clear that calcium dysregula-
et al., 1998, 2001; Beer et al., 2000; Clark et al., tion and cysteine protease activation are integral
2000; Keane et al., 2001; Yakovlev et al., 2001; to traumatically induced neuronal death, it is of
Cernak et al., 2002), as well as in human head injury note that other potentially important pathways and
(Clark et al., 1999; Pineda et al., 2004; Farkas et al., processes may be operant, although, they have been
2005). give little attention in the context of TBI. Foremost
among these are the calcium-mediated activation of
lysosomal hydrolytic pathways that have received
The Bcl-2 oncogene family considerable attention in non-TBI-related neuronal
death. Further, it is well known that lysosomes and
Integral to the apoptotic death cascade is the Bcl-2 their enzymes are involved in different cell death
proto-oncogene family, which is one of the key pathways in various non-CNS systems as reviewed
regulators of apoptosis. This family consists of by Kroemer and Jaattela (2005) and Yamashima
members with anti-apoptotic (Bcl-2 and BclXL) (2004). Multiple damaging insults are known to
and pro-apoptotic (Bax, Bad, Bid, BclXS) activity. result in the disruption of the lysosomal membrane
Typically, following apoptotic stimuli, Bax trans- leading to the release of several lysosomal enzymes,
locates from the cytoplasm to the mitochondrial such as cathepsins. It has been posited that low
membrane resulting in cytocrome-c release, which levels of stress result in limited lysosomal mem-
by binding with Apaf-1 activates caspases. Bax brane rupture and cathepsin release that in turn
is also suggested to directly activate caspase-3. elicit apoptosis. In contrast, high levels of stress
In contrast, anti-apoptotic Bcl-2 and BclXL can can cause generalized lysosomal membrane rupture
prevent the mitochondrial permeabilization by resulting in necrosis (Brunk et al., 1997). In the
inactivating Bax via heterodimerization. Bcl-2 later scenario, targeted Ca2+-mediated calpain
and BclXL can also inhibit caspase-3 activation activation and the likely concomitant production
(Merry and Korsmeyer, 1997; Yang et al., 1997). of free radicals are posited to destroy the lysosomal
48

membrane leading to the leakage of damaging et al., 2002). These TAI-linked neurons also re-
hydrolytic lysosomal enzymes into the cytoplasm to vealed a transient suppression of protein synthesis.
cause the digestion of the cell’s structural proteins Taken together, these transient reactive changes
(Yamashima, 2000; Yamashima et al., 2003). suggest a potential neuronal attempt at reorgan-
Despite the consistent involvement of lysosomes ization and repair, rather than the initiation of any
and cathepsin in cell death in multiple organs, prenecrotic/apoptotic change.
their contribution to neuronal death following
DTBI has not been rigorously evaluated and
obviously requires attention. Mechanoporation and the fate of neurons with
membrane disruption
Mechanically related factors
Using extracellular tracer infusion techniques,
multiple in vitro and in vivo studies have demon-
While the previous passages briefly reviewed some
strated that immediately following DTBI, other
of the neurochemical cascades triggered by the
non-axotomized neurons can take up either high
forces of injury, until recently there has not been
molecular weight tracers, such as horseradish per-
any parallel consideration that the mechanical
oxidase or different molecular weight dextrans
force of injury itself could directly perturb the
or other molecules normally excluded from the
neuronal cell membrane or its appendages lead-
neuronal cytoplasm by the intact cell membrane.
ing to mechanically induced neuronal death cas-
This immediate tracer uptake suggests that the me-
cades. In this regard, we comment here on recently
chanical force of the injury itself evoked neuronal
published data evaluating the potential damaging
cell membrane disruption (mechanoporation), that
neuronal somatic consequences of traumatically
then most likely allowed the influx of damaging
induced axotomy and/or the direct disruption of
ions through the compromised cell membrane
the neuronal plasmalemma.
(Geddes et al., 2003; Prado and LaPlaca, 2004;
Prado et al., 2004, 2005; Singleton and Povlishock,
Axotomy-related changes 2004).
Following these initial descriptions of neu-
Neuronal somatic changes occurring adjacent ronal membrane perturbation, subsequent studies
to sites of traumatic axonal injury (TAI) have revealed a rather heterogeneous neuronal response
been recognized previously (Van den Heuvel et al., to this disruption (Singleton and Povlishock, 2004;
1998, 1999; Cernak et al., 2004); yet, the rela- Farkas et al., 2006). Some neurons sustaining cell
tion of these events to any subsequent injury cas- membrane disruption revealed Fluoro Jade posit-
cades has not been appreciated. Recently, using ivity and ultrastructural evidence of overt neuron-
immunohistochemical techniques recognizing al necrosis. In contrast, other neurons sustaining
amyloid precursor protein (APP), a well accepted membrane disruption did not reveal comparable
marker of TAI, neuronal somata directly linked to signs of overt cell damage. Further, at the same
TAI were identified following DTBI in the cerebral time and in the same brain foci, different popula-
cortex, hippocampus and thalamus (Singleton tions of injured neurons did not reveal evidence
et al., 2002). These studies showed that contrary of membrane disruption, yet, they demonstrated
to contemporary thought, TAI, even in immediate either perisomatic axonal injury resulting in
proximity to the neuronal soma, did not result in increased neuronal somatic APP-positivity, or the
acute neuronal death. Rather, neurons sustaining non-axotomy-related induction of heat shock
TAI revealed signs of reactive change, including protein expression (Singleton and Povlishock,
the loss and degranulation of the rough endoplas- 2004), and/or CMSP (Farkas et al., 2006, vide
mic reticulum, disaggregation of polysomes and infra). In part, because of these divergent neuronal
dispersal of the Golgi apparatus without any cyto- responses, some of which were suggestive of cell
skeletal or mitochondrial alterations (Singleton death whereas others suggested recovery, the
49

potential for post-traumatic neuronal cell membrane reopening their membranes to now manifest endur-
resealing and recovery became a consideration. Via ing damage and/or additional neurons suffering
the administration of different extracellular tracers delayed membrane perturbation (Fig. 4) (Farkas
at varied times post-injury, in vitro studies have et al., 2006). Collectively, these in vivo studies clearly
revealed that the majority of neurons sustaining confirm the existence of DTBI-injured neuronal
mechanoporation could reseal their disrupted mem- membrane perturbation, while further illustrating
branes in the first minutes post-TBI (Geddes et al., the complex pathobiology associated with DTBI.
2003; Prado and LaPlaca, 2004; Prado et al., 2005). This complexity was further highlighted by re-
In contrast, in vivo studies did not confirm these cent in vitro studies that explored the potential re-
observations. Rather, in vivo studies revealed lationship of the above described altered membrane
that more than 50% of the tracer-containing corti- permeability to CMSP positing that CMSP co-ex-
cal neurons contained both pre- and post-injury isted with increased membrane permeability (Pike
administered tracers at several hours post-DTBI, et al., 2000; Liu and Schnellmann, 2003; Liu et al.,
suggesting enduring membrane permeability (Fig. 1) 2004). In contrast, with in vivo DTBI, the majority
(Farkas et al., 2006). Resealing was observed after of neurons sustaining either membrane disruption
DTBI, yet, to a lesser extent than suggested in vitro and/or demonstrating necrotic change did not re-
(Fig. 2) (Farkas et al., 2006). To further complicate veal CMSP, suggesting that membrane disruption
this issue, the recent finding of neurons demon- itself did not lead to calpain activation. Conversely,
strating enduring membrane permeability without with DTBI, neurons demonstrating CMSP did not
evidence of overt cell damage points to the occur- reveal membrane disruption, and, as the majority of
rence of potentially delayed membrane resealing, these CMSP-positive neurons revealed only limited
rather than the rapid resealing/repair observed in ultrastructural damage, this suggested that CMSP
vitro (Farkas et al., 2006). Further complexity was was not associated with neuronal death after DTBI
also associated with the finding that other neurons (Fig. 5) (Farkas et al., 2006). Taken together with
only revealed post-injury tracer uptake without other studies (Saatman et al., 1996; Brana et al.,
evidence of the pre-injury tracer, suggesting the 1999), these findings emphasize the need for caution
potential for delayed membrane disruption (Fig. 3) in interpreting the occurrence of CMSP and its
(Farkas et al., 2006). Although the underlying overall implications for the neuronal injury and
mechanism(s) of this delayed membrane disruption death associated with DTBI.
is (are) unknown, the possibility exists that the sus-
tained, elevated intracranial pressure occurring after
injury and persisting for several hours post-TBI Axonal damage associated with DTBI
(Farkas et al., 2006) could contribute to this pheno-
menon. Statistical analysis of the neuronal popu- In addition to diffuse neuronal perturbation and
lations demonstrating different tracer uptake over death described above, diffuse axonal injury is also
time post-DTBI has revealed significant tracer a distinguishing feature of DTBI, occurring across
distribution/redistribution consistent with the com- the spectrum of brain injury ranging from mild
plex neuronal membrane changes described above. through severe. Historically, the histological identi-
Specifically, between 4 and 8 h post-injury, the fication of diffuse axonal injury was based upon
proportion of neurons with delayed membrane per- the use of silver salts to detect, within the first days
turbation was significantly different from the pro- of injury, grossly swollen axonal bulbs that lacked
portion of resealed neurons as well as the proportion continuity with their downstream axonal partners
of those neurons with enduring membrane pertur- (Strich, 1956). This suggested their mechanical
bation. The change in the proportion of neurons transsection resulting in axonal retraction and
with resealed and enduring membrane perturbation axoplasmic pooling at the site of disconnection
over time was not significant. This redistribution of (Strich, 1956; Adams, 1982). More contempo-
membrane perturbation types between 4 and 8 h rary studies, however, have shown that in large
post-injury may be the result of resealed neurons part, this premise of transsection and retraction
50

Fig. 1. This figure illustrates the phenomenon of mechanically induced neuronal membrane disruption and its consequences for the
neuron. Via double labeled confocal images, panels A–C show neurons flooding with both dextrans with evidence of concomitant
cellular injury, reflected in their irregular, distorted profiles and vacuolization (arrows). In those cells showing the most severe damage,
note that the dextrans are also typically found within the nucleus (arrowhead). Note that other double-labeled neurons (D) dem-
onstrate little or no pathological damage and that despite homogenous tracer uptake no nuclear accumulation or vacuolization occurs
(arrows). Scale bars: 100 mm. Panel E illustrates three tracer flooded neurons, confirmed by routine fluorescent microscopy and
followed via EM. The most severely damaged neuron (asterisk) demonstrates increased electron density, organelle vacuolization
(arrows) and perisomatic glial ensheathment (arrowheads). The two other cells (double and triple asterisks) demonstrate little or no
pathological change. Note that the surrounding neuropil demonstrates little overt pathologic change consistent with the confocal
observations. Scale bar: 5 mm.

is not correct. Rather, it has been shown in mul- impairment of axonal transport, with progressive
tiple animal studies, as well as limited human in- local axonal swelling followed by detachment over
vestigations, that the forces of injury diffusely alter a post-traumatic course ranging from several hours
focal axonal segments. This results in a local up to a day (Povlishock and Jenkins, 1995). Given
51

Fig. 2. This figure illustrates the finding that although mechanoporation can occur, some neurons reseal their perturbated membranes
with increased post-traumatic survival. Here confocal images of pre-injury infused dextrans (A), post-injury-infused dextrans (B) and
their overlay (C) demonstrate some cortical neurons flooding with the pre-injury infused dextran alone without concomitant flooding
with the post-injury administrated tracer (arrows), all of which is suggestive of cell membrane closure/recovery. Note that one double-
flooded neuron demonstrating severe damage (arrowhead) and one double-labeled neuron demonstrating less severe pathology (double
arrowhead) are also shown. Scale bar: 50 mm. Panel D illustrates a routine fluorescent image that reveals a single labeled cell. In panel
E, the same neuron is visualized through the use of antibodies to the fluorophore and then carried to the EM level (F–G). Note that
neurons flooding with the pre-injury dextran alone do not show overt pathological damage. Immunoreactive products (anti-Alexa
Fluor IR) are labeled with arrows in panel G, which is an enlargement of the area, blocked out in panel F. Scale bar 2 mm. (Adapted
with permission from Farkas et al. (2006); Copyright 2006 by the Society for Neuroscience).

the fact that these reactive axonal changes were Cellular and subcellular factors related to the
found in scattered axons related to other intact ax- initiating pathogenesis of DTBI-associated axonal
ons and their vascular elements, this precluded the injury
potential for direct mechanical renting, suggesting
that more subtle intraaxonal changes were at work Appreciating that the above described cascades of
in the pathogenesis of this progressive axonal axonal perturbation leading to impaired transport
change leading to disconnection. and disconnection evolved over a relatively long
52

Fig. 3. In contrast to the phenomenon of resealing shown in Fig. 2, this figure provides evidence of delayed neuronal plasmalemmal
opening. Here confocal images of pre-injury infused dextran (A), post-injury-infused dextran (B) and their overlay (C) demonstrate
scattered neurons flooding with the post-injury infused dextran alone (arrows), among with double-flooded neurons (arrowheads).
Scale bar: 100 mm. (Adapted with permission from Farkas et al. (2006); Copyright 2006 by the Society for Neuroscience).

post-traumatic course, emphasis has been placed our laboratory probed these same segments with
upon identifying the initiating intraaxonal cellular antibodies targeting cytochrome C and caspase-
and subcellular factors both to understand better mediated spectrin proteolysis (Buki et al., 2000).
the pathobiology of this axonal injury and to Via this approach, we demonstrated that the dam-
develop therapies targeting these cellular and sub- aged mitochondria released cytochrome C that, in
cellular changes. While immediate physical trans- turn, activated caspase-mediated spectrin degrada-
section of the axon cylinder has been ruled out, tion (Buki et al., 2000; Buki and Povlishock, 2006).
the potential for focal disturbances in the axo- Collectively, both cysteine proteases, calpain and
lemma leading to local ionic dysregulation was caspase were recognized to participate in the deg-
evaluated in the experimental setting using extra- radation of the axonal cytoskeleton associated
cellular tracers normally excluded by the intact/un- with concomitant neurofilament side-arm cleavage,
altered axolemma. Through this approach, our neurofilament compaction and microtubular loss
laboratory showed that discreet axonal foci scat- (Buki and Povlishock, 2006). Because of these local
tered throughout the injured brain revealed evi- cytoskeletal abnormalities, these compacted axonal
dence of altered focal axolemmal permeability to segments could also be routinely identified by use
various normally excluded extracellular tracers of antibodies targeting altered neurofilament sub-
(Pettus and Povlishock, 1996; Povlishock and Pet- units (RMO14) that, in our hands, provided a sur-
tus, 1996). Moving on the premise that this alter- rogate marker for identifying such sites of injury
ation in axolemmal permeability would be (Povlishock et al., 1997). Based on these findings in
accompanied by local calcium dysregulation, we our lab, as well as others, it was assumed that these
probed the same axonal segments with antibodies intraaxonal changes universally led to an upstream
targeting calcium-activated, CMSP. In these stud- impairment of axonal transport leading to swelling
ies, CMSP was observed initially in the subaxo- and disconnection (Maxwell et al., 1997). However,
lemmal domain followed by its activation in the continued investigation failed to establish a routine
axon’s core, with a particular predilection for the correlation between these two axonal events, lead-
mitochondria that appeared swollen with disrupted ing us to question whether neurofilament compact-
cristae (Buki et al., 1999). Because such mi- ion and axonal swelling, consistently occurred
tochondrial damage appeared consistent with within the same axonal segment. More recent stud-
local calcium overloading and caspase activation, ies, using multiple strategies to qualitatively and
53

focally altered axolemmal permeability it appears,


as noted previously, that local calcium dysregula-
tion with the activation of the cysteine proteases,
causes local degradation of the axonal cytoskele-
ton, leading to axonal failure and disconnection
(Stone et al., 2004; Marmarou et al., 2005). Why
these sites of axonal injury do not reveal impaired
axonal transport and axonal swelling is unclear,
yet, it is conceivable that the suprathreshold
calcium uptake occurring at these sites most likely
converts anterograde to retrograde transport,
thereby precluding the development of reactive
axonal swelling (Sahenk and Lasek, 1988; Martz
Fig. 4. This figure provides a semiquantitative, graphic repre- et al., 1989). The validity of the above is supported
sentation of those different forms and prognosis of membrane by the use of various therapeutic strategies target-
perturbation following DTBI. Here the distribution of neurons
with resealed (gray column), enduring (white column) or delayed
ing calpain inhibition that, as such, significantly
membrane perturbation (black column) are shown after DTBI- reduce the numbers of axonal profiles showing the
induced membrane perturbation. As membranes remain closed, above described cysteine protease activation and
open or reseal in response to injury, tracer availability deter- cytoskeletal collapse (Buki et al., 2003; Buki and
mines the type of membrane perturbation assigned categorically Povlishock, 2006). In contrast, it appears that those
to each neuron. The pre-injury administration of Alexa Fluor
488 and post-injury administration of Texas Red conjugated
axons showing impaired axonal transport and local
dextrans permit the evaluation of neuronal membrane pertur- swelling do not sustain any alteration in local
bation distribution at 4 and 8 h after injury. Across time points, axolemmal permeability or any activation of the
the proportion of neurons with delayed membrane perturbation cysteine proteases (Povlishock and Stone, 2001).
is significantly different from the proportion of resealed neurons Rather, it is posited that other mechanisms are at
and the proportion of enduring membrane perturbation (w2,
po0.016). The change in the proportion of neurons with re-
work and that these are linked to more subtle
sealed and enduring membrane perturbation over time was not forms of calcium dysregulation. These potentially
significant (w2, p ¼ 0.19). The redistribution of the types of involve the activation of micromolar calpains to
membrane perturbation between 4 and 8 h post-injury may result trigger the activation of calcineurin that, in turn,
from resealed neurons reopening (gray arrow) to become neu- alters the microtubular network to disrupt local
rons with enduring damage and/or additional neurons (black
arrow) suffering delayed membrane perturbation.
axonal transport kinetics and thereby elicit the
swellings described above (Povlishock and Stone,
2001). Although limited direct evidence exists to
support this pathway, the use of calcineurin
quantitatively assess axonal numbers as well as the antagonists, such as FK506, directly attenuate the
spatial relationship of axons showing neurofila- numbers of axons showing impaired axonal
ment compaction and impaired transport have now transport and swelling while having no effect upon
convincingly shown that the above described sites those axons showing neurofilament compaction
of altered axonal permeability, cysteine protease and disconnection (Marmarou and Povlishock,
activation and cytoskeletal collapse do not rou- 2006). This supports the premise that calcineurin
tinely correlate with sites of impaired axonal trans- is integral to the pathogenesis of impaired axonal
port (Stone et al., 2004; Marmarou et al., 2005). transport and swelling. Collectively, these studies
This suggested that the neurofilament-compacted illustrate the complexity of the pathogenesis
axonal segments and swollen axonal segments dem- of diffuse axonal injury, suggesting at least two
onstrating impaired axonal transport most likely differing types of initiating mechanisms, with the
represent two different populations of injured ax- caveat that both populations of injured axons will
ons responding to the traumatic episode in different not likely be amenable to one form of therapeutic
fashions. For those axonal segments showing intervention.
54
55

Other potential cellular and subcellular factors axolemmal disruption was discerned. Rather,
related to diffuse axonal injury the depolarization associated with this injury
evoked sodium influx, the activation of voltage
In the above passages, we have focused upon the gated calcium channels and the concomitant
contemporary appreciation of the cellular and activation of sodium/calcium exchangers, all of
subcellular changes involved in diffuse axonal in- which contributed to local intraaxonal calcium
jury. Importantly, all these changes were believed overloading (Wolf et al., 2001). These calcium-me-
to be ongoing only in myelinated nerve fiber popu- diated changes, comparable with some of the
lations with no involvement of unmyelinated changes described in myelinated axons, were linked
axons that, in general, have received virtually no with the activation of proteases. These, in turn,
consideration either in the context of TBI (Jafari contributed to subsequent proteolysis of its NaCh
et al., 1998) or, for that matter, any other CNS subunit to promote a persistent elevation in
disorder. Recently, however, this perception was intracellular calcium, fueling additional pathologi-
changed by work conducted by Reeves et al. (2005) cal changes through many of the pathways ad-
who, through the use of electrophysiological meth- dressed above (Iwata et al., 2004). While these
ods, provided compelling evidence for unmyelin- changes obviously remain to be confirmed in vivo,
ated nerve fiber damage and dysfunction within they are intriguing and speak to yet another, differ-
the corpus callosum of traumatically brain-injured ent form of cellular and subcellular change that,
animals. Using an analysis of compound action in this case, involves a potential channelopathy as
potentials by examining two specific wave forms major player in the ensuing unmyelinated axonal
that can be independently related to either myelin- perturbation.
ated or unmyelinated axons populations, Reeves
and colleagues showed significant and sustained
depression of the compound action potentials Concluding comments
associated with the unmyelinated axon population.
These electrophysiological studies were accompa- In this brief review, we have attempted to critically
nied by routine morphological analysis using elec- evaluate current thought on the cellular and sub-
tron microscopy. Although these studies were cellular change evoked by DTBI at both the neu-
preliminary, they suggested that the morphological ronal somatic and axonal fronts. Recognizing that
progression of unmyelinated fiber change observed our appreciation of cellular and subcellular change
was quite dissimilar from that described above for in the context of TBI has been framed primarily by
myelinated axons. This potential difference is also our understanding of the pathobiology of contu-
partially supported by excellent in vitro studies that sional injury, this review cautions against moving
have examined non-myelinated neurites subjected on the assumption that these contusional cellular
to mechanical strains that ultimately led to local and subcellular cascades must transfer to those
axonal beading and disconnection (Wolf et al., neuronal cell bodies and their appendages that
2001; Iwata et al., 2004). In this pathology, how- sustain diffuse injury. Basic differences between
ever, no evidence of overt axolemmal change or focal and diffuse injury give rise to the possibility

Fig. 5. This figure illustrates the relation between CMSP and tracer uptake. Panel A shows triple labeled confocal image demon-
strating dextran flooded neurons as well as CMSP immunopositive neurons. Note that at both 4 and 8 h post-injury CMSP immuno-
reactive neurons were observed throughout the neocortex (arrows). Scale bar: 100 mm. In panels B–E confocal images of pre-injury
dextran flooding (B), post-injury dextran flooding (C), CMSP immunopositivity (D) and their overlay (E) demonstrate neurons
showing enduring membrane disruption reflected in their content of both tracers. Note that some neurons colocalize with CMSP-
immunopositivity (arrow), whereas other neurons demonstrate tracer flooding without CMSP (arrowheads). Also note that at these
same time points, several CMSP immunoreactive neurons also could be identified without concomitant tracer flooding (double
arrowhead). Lastly note that in the same region, a neuron demonstrating only the initial tracer flooding (big arrow), as well as a neuron
revealing only secondary tracer flooding (triple arrowhead) can also be seen. Scale bar: 50 mm. (Adapted with permission from Farkas
et al. (2006); Copyright 2006 by the Society for Neuroscience).
56

for the involvement of other cell perturbation/ slice cultures deprived of oxygen and glucose. Eur. J. Neurosci.,
death cascades that may not be operant with focal 11(7): 2375–2384.
Braughler, J.M. and Hall, E.D. (1992) Involvement of lipid per-
injury. Specifically, the pathogenesis of diffuse
oxidation in CNS injury. J. Neurotrauma, 9(Suppl 1): S1–S7.
injury highlights the potential for mechanically Brunk, U.T., Dalen, H., Roberg, K. and Hellquist, H.B. (1997)
induced membrane perturbation to serve as a more Photo-oxidative disruption of lysosomal membranes causes
significant player in the ensuing cellular and sub- apoptosis of cultured human fibroblasts. Free Radic. Biol.
cellular change. The potential for transient versus Med., 23(4): 616–626.
more enduring mechanically induced membrane Buki, A., Farkas, O., Doczi, T. and Povlishock, J.T. (2003) Pre-
injury administration of the calpain inhibitor MDL-28170 at-
perturbation may also influence subsequent neuron- tenuates traumatically induced axonal injury. J. Neurotrauma,
al somatic and axonal perturbation and/or death. 20(3): 261–268.
Additionally, the activation of other cell death Buki, A., Okonkwo, D.O., Wang, K.K. and Povlishock, J.T.
cascades including, but not limited to, activation (2000) Cytochrome c release and caspase activation in trau-
matic axonal injury. J. Neurosci., 20(8): 2825–2834.
of lysosomal-mediated change through the release
Buki, A. and Povlishock, J.T. (2006) All roads lead to discon-
of hydrolytic enzymes must now be considered. nection? Traumatic axonal injury revisited. Acta Neurochir.
Collectively, this review illustrates the complexity (Wien.), 148(2): 181–193.
of the pathobiology of DTBI and the danger Buki, A., Siman, R., Trojanowski, J.Q. and Povlishock, J.T.
in assuming that a singular mechanistic view of (1999) The role of calpain-mediated spectrin proteolysis in
cellular and subcellular change can translate to the traumatically induced axonal injury. J. Neuropathol. Exp.
Neurol., 58(4): 365–375.
diffusely injured brain. Cernak, I., Chapman, S., Hamlin, G. and Vink, R. (2002)
Temporal characterisation of pro- and anti-apoptotic mech-
anisms following diffuse traumatic brain injury in rats.
Abbreviations J. Clin. Neurosci., 9(5): 565–572.
Cernak, I., Vink, R., Zapple, D.N., Cruz, M.I., Ahmed, F.,
Chang, T., Fricke, S.T. and Faden, A.I. (2004) The patho-
CMSP calpain-mediated spectrin proteolysis
biology of moderate diffuse traumatic brain injury as iden-
DTBI diffuse traumatic brain injury tified using a new experimental model of injury in rats.
SBDP spectrin breakdown product Neurobiol. Dis., 17(1): 29–43.
TBI traumatic brain injury Cervos-Navarro, J. and Lafuente, J.V. (1991) Traumatic brain
injuries: structural changes. J. Neurol. Sci., 103(Suppl):
S3–S14.
Clark, R.S., Kochanek, P.M., Chen, M., Watkins, S.C.,
Acknowledgments Marion, D.W., Chen, J., Hamilton, R.L., Loeffert, J.E. and
Graham, S.H. (1999) Increases in Bcl-2 and cleavage of
This work was supported in part by NIH grants caspase-1 and caspase-3 in human brain after head injury.
FASEB J., 13(8): 813–821.
NS045824, NS047463 and HD055813. Clark, R.S., Kochanek, P.M., Watkins, S.C., Chen, M., Dixon,
C.E., Seidberg, N.A., Melick, J., Loeffert, J.E., Nathaniel,
P.D., Jin, K.L. and Graham, S.H. (2000) Caspase-3 medi-
References ated neuronal death after traumatic brain injury in rats.
J. Neurochem., 74(2): 740–753.
Adams, J.H. (1982) Diffuse axonal injury in non-missile head Colicos, M.A. and Dash, P.K. (1996) Apoptotic morphology of
injury. Injury, 13(5): 444–445. dentate gyrus granule cells following experimental cortical
Adams, J.H., Graham, D.I., Scott, G., Parker, L.S. and Doyle, impact injury in rats: possible role in spatial memory deficits.
D. (1980) Brain damage in fatal non-missile head injury. Brain Res., 739(1–2): 120–131.
J. Clin. Pathol., 33(12): 1132–1145. Colicos, M.A., Dixon, C.E. and Dash, P.K. (1996) Delayed,
Beer, R., Franz, G., Srinivasan, A., Hayes, R.L., Pike, B.R., selective neuronal death following experimental cortical im-
Newcomb, J.K., Zhao, X., Schmutzhard, E., Poewe, W. and pact injury in rats: possible role in memory deficits. Brain
Kampfl, A. (2000) Temporal profile and cell subtype distri- Res., 739(1–2): 111–119.
bution of activated caspase-3 following experimental trau- Conti, A.C., Raghupathi, R., Trojanowski, J.Q. and McIntosh,
matic brain injury. J. Neurochem., 75(3): 1264–1273. T.K. (1998) Experimental brain injury induces regionally
Brana, C., Benham, C.D. and Sundstrom, L.E. (1999) Calpain distinct apoptosis during the acute and delayed post-traumatic
activation and inhibition in organotypic rat hippocampal period. J. Neurosci., 18(15): 5663–5672.
57

Cortez, S.C., McIntosh, T.K. and Noble, L.J. (1989) Experi- Iwata, A., Stys, P.K., Wolf, J.A., Chen, X.H., Taylor, A.G.,
mental fluid percussion brain injury: vascular disruption and Meaney, D.F. and Smith, D.H. (2004) Traumatic axonal in-
neuronal and glial alterations. Brain Res., 482(2): 271–282. jury induces proteolytic cleavage of the voltage-gated sodium
Dietrich, W.D., Alonso, O., Busto, R., Globus, M.Y. and channels modulated by tetrodotoxin and protease inhibitors.
Ginsberg, M.D. (1994a) Post-traumatic brain hypothermia J. Neurosci., 24(19): 4605–4613.
reduces histopathological damage following concussive brain Jafari, S.S., Nielson, M., Graham, D.I. and Maxwell, W.L.
injury in the rat. Acta Neuropathol. (Berl.), 87(3): 250–258. (1998) Axonal cytoskeletal changes after nondisruptive
Dietrich, W.D., Alonso, O. and Halley, M. (1994b) Early axonal injury. II. Intermediate sized axons. J. Neurotrauma,
microvascular and neuronal consequences of traumatic 15(11): 955–966.
brain injury: a light and electron microscopic study in rats. Kampfl, A., Posmantur, R., Nixon, R., Grynspan, F., Zhao, X.,
J. Neurotrauma, 11(3): 289–301. Liu, S.J., Newcomb, J.K., Clifton, G.L. and Hayes, R.L.
Eldadah, B.A. and Faden, A.I. (2000) Caspase pathways, (1996) mu-Calpain activation and calpain-mediated cytoskele-
neuronal apoptosis, and CNS injury. J. Neurotrauma, 17(10): tal proteolysis following traumatic brain injury. J. Neurochem.,
811–829. 67(4): 1575–1583.
Faden, A.I., Demediuk, P., Panter, S.S. and Vink, R. (1989) Katayama, Y., Becker, D.P., Tamura, T. and Hovda, D.A.
The role of excitatory amino acids and NMDA receptors in (1990) Massive increases in extracellular potassium and the
traumatic brain injury. Science, 244(4906): 798–800. indiscriminate release of glutamate following concussive
Farkas, O., Lifshitz, J. and Povlishock, J.T. (2006) Mechanop- brain injury. J. Neurosurg., 73(6): 889–900.
oration induced by diffuse traumatic brain injury: an irre- Keane, R.W., Kraydieh, S., Lotocki, G., Alonso, O.F., Aldana,
versible or reversible response to injury? J. Neurosci., 26(12): P. and Dietrich, W.D. (2001) Apoptotic and antiapoptotic
3130–3140. mechanisms after traumatic brain injury. J. Cereb. Blood
Farkas, O., Polgar, B., Szekeres-Bartho, J., Doczi, T., Povlishock, Flow Metab., 21(10): 1189–1198.
J.T. and Buki, A. (2005) Spectrin breakdown products in the Kermer, P., Klocker, N. and Bahr, M. (1999) Neuronal death
cerebrospinal fluid in severe head injury: preliminary observa- after brain injury. Models, mechanisms, and therapeutic
tions. Acta Neurochir. (Wien.), 147(8): 855–861. strategies in vivo. Cell Tissue Res., 298(3): 383–395.
Fei, Z., Zhang, X., Jiang, X.F., Huang, W.D. and Bai, H.M. Kitanaka, C. and Kuchino, Y. (1999) Caspase-independent
(2005) Altered expression patterns of metabotropic glutamate programmed cell death with necrotic morphology. Cell
receptors in diffuse brain injury. Neurosci. Lett., 380(3): Death Differ., 6(6): 508–515.
280–283. Kontos, H.A. and Povlishock, J.T. (1986) Oxygen radicals in
Felderhoff-Mueser, U., Sifringer, M., Pesditschek, S., Kuckuck, brain injury. Cent. Nerv. Syst. Trauma, 3(4): 257–263.
H., Moysich, A., Bittigau, P. and Ikonomidou, C. (2002) Kotapka, M.J., Gennarelli, T.A., Graham, D.I., Adams, J.H.,
Pathways leading to apoptotic neurodegeneration following Thibault, L.E., Ross, D.T. and Ford, I. (1991) Selective
trauma to the developing rat brain. Neurobiol. Dis., 11(2): vulnerability of hippocampal neurons in acceleration-induced
231–245. experimental head injury. J. Neurotrauma, 8(4): 247–258.
Formigli, L., Papucci, L., Tani, A., Schiavone, N., Tempestini, Kotapka, M.J., Graham, D.I., Adams, J.H., Doyle, D. and
A., Orlandini, G.E., Capaccioli, S. and Orlandini, S.Z. (2000) Gennarelli, T.A. (1993) Hippocampal damage in fatal pae-
Aponecrosis: morphological and biochemical exploration of diatric head injury. Neuropathol. Appl. Neurobiol., 19(2):
a syncretic process of cell death sharing apoptosis and 128–133.
necrosis. J. Cell Physiol., 182(1): 41–49. Kotapka, M.J., Graham, D.I., Adams, J.H. and Gennarelli,
Fox, G.B., Fan, L., LeVasseur, R.A. and Faden, A.I. (1998) T.A. (1992) Hippocampal pathology in fatal non-missile human
Sustained sensory/motor and cognitive deficits with neuronal head injury. Acta Neuropathol. (Berl.), 83(5): 530–534.
apoptosis following controlled cortical impact brain injury in Kroemer, G. and Jaattela, M. (2005) Lysosomes and autophagy
the mouse. J. Neurotrauma, 15(8): 599–614. in cell death control. Nat. Rev. Cancer, 5(11): 886–897.
Geddes, D.M., Cargill, R.S. and LaPlaca, M.C. (2003) Kupina, N.C., Detloff, M.R., Dutta, S. and Hall, E.D. (2002)
Mechanical stretch to neurons results in a strain rate and Neuroimmunophilin ligand V-10,367 is neuroprotective after
magnitude-dependent increase in plasma membrane perme- 24-hour delayed administration in a mouse model of diffuse
ability. J. Neurotrauma, 20(10): 1039–1049. traumatic brain injury. J. Cereb. Blood Flow Metab., 22(10):
Goda, M., Isono, M., Fujiki, M. and Kobayashi, H. (2002) Both 1212–1221.
MK801 and NBQX reduce the neuronal damage after impact- Kupina, N.C., Nath, R., Bernath, E.E., Inoue, J., Mitsuyoshi,
acceleration brain injury. J. Neurotrauma, 19(11): 1445–1456. A., Yuen, P.W., Wang, K.K. and Hall, E.D. (2001) The novel
Hicks, R., Soares, H., Smith, D. and McIntosh, T. (1996) calpain inhibitor SJA6017 improves functional outcome after
Temporal and spatial characterization of neuronal injury delayed administration in a mouse model of diffuse brain
following lateral fluid-percussion brain injury in the rat. Acta injury. J. Neurotrauma, 18(11): 1229–1240.
Neuropathol. (Berl.), 91(3): 236–246. Lin, X., Zhi, D. and Zhang, S. (2001) Inhibiting effect of mod-
Huang, Y. and Wang, K.K. (2001) The calpain family and erate hypothermia on cell apoptosis after diffuse brain injury
human disease. Trends Mol. Med., 7(8): 355–362. in rats. Chin. J. Traumatol., 4(1): 14–19.
58

Liu, X. and Schnellmann, R.G. (2003) Calpain mediates caspase-3 proteolysis of alpha-spectrin after traumatic brain
progressive plasma membrane permeability and proteolysis injury. Neuroreport, 9(11): 2437–2442.
of cytoskeleton-associated paxillin, talin, and vinculin during Pineda, J.A., Wang, K.K. and Hayes, R.L. (2004) Biomarkers
renal cell death. J. Pharmacol. Exp. Ther., 304(1): 63–70. of proteolytic damage following traumatic brain injury. Brain
Liu, X., Van Vleet, T. and Schnellmann, R.G. (2004) The role Pathol., 14(2): 202–209.
of calpain in oncotic cell death. Annu. Rev. Pharmacol. Povlishock, J.T. and Jenkins, L.W. (1995) Are the pathobio-
Toxicol., 44: 349–370. logical changes evoked by traumatic brain injury immediate
Marmarou, C.R. and Povlishock, J.T. (2006) Administration and irreversible? Brain Pathol., 5(4): 415–426.
of the immunophilin ligand FK506 differentially attenuates Povlishock, J.T. and Katz, D.I. (2005) Update of neuropa-
neurofilament compaction and impaired axonal transport in thology and neurological recovery after traumatic brain
injured axons following diffuse traumatic brain injury. Exp. injury. J. Head Trauma Rehabil., 20(1): 76–94.
Neurol., 197(2): 353–362. Povlishock, J.T. and Kontos, H.A. (1992) The role of oxygen
Marmarou, C.R., Walker, S.A., Davis, C.L. and Povlishock, radicals in the pathobiology of traumatic brain injury. Hum.
J.T. (2005) Quantitative analysis of the relationship between Cell, 5(4): 345–353.
intra-axonal neurofilament compaction and impaired axonal Povlishock, J.T., Marmarou, A., McIntosh, T., Trojanowski,
transport following diffuse traumatic brain injury. J. Neuro- J.Q. and Moroi, J. (1997) Impact acceleration injury in the
trauma, 22(10): 1066–1080. rat: evidence for focal axolemmal change and related neuro-
Martz, D., Garner, J. and Lasek, R.J. (1989) Protein changes filament sidearm alteration. J. Neuropathol. Exp. Neurol.,
during anterograde-to-retrograde conversion of axonally 56(4): 347–359.
transported vesicles. Brain Res., 476(1): 199–203. Povlishock, J.T. and Pettus, E.H. (1996) Traumatically induced
Maxwell, W.L., Povlishock, J.T. and Graham, D.L. (1997) A axonal damage: evidence for enduring changes in axolemmal
mechanistic analysis of nondisruptive axonal injury: a review. permeability with associated cytoskeletal change. Acta
J. Neurotrauma, 14(7): 419–440. Neurochir. Suppl. (Wien.), 66: 81–86.
McCracken, E., Hunter, A.J., Patel, S., Graham, D.I. and Povlishock, J.T. and Stone, J.R. (2001) Traumatic axonal
Dewar, D. (1999) Calpain activation and cytoskeletal protein injury. In: Miller L.P., Hayes R.L. and Newcomb J.K. (Eds.),
breakdown in the corpus callosum of head-injured patients. Head Trauma: Basic, Preclinical and Clinical Directions.
J. Neurotrauma, 16(9): 749–761. Wiley, pp. 281–302.
Merry, D.E. and Korsmeyer, S.J. (1997) Bcl-2 gene family in Prado, G.R., Ross, J.D., DeWeerth, S.P. and LaPlaca,
the nervous system. Annu. Rev. Neurosci., 20: 245–267. M.C. (2005) Mechanical trauma induces immediate chan-
Newcomb, J.K., Zhao, X., Pike, B.R. and Hayes, R.L. (1999) ges in neuronal network activity. J. Neural Eng., 2(4):
Temporal profile of apoptotic-like changes in neurons and 148–158.
astrocytes following controlled cortical impact injury in the Prado, G., Caltagirone, C.L. and LaPlaca, M.C. (2004)
rat. Exp. Neurol., 158(1): 76–88. Neuronal plasma membrane disruption in the traumatically
Nicotera, P., Leist, M. and Manzo, L. (1999) Neuronal cell injured brain. J. Neurotrauma, 21: 1331.
death: a demise with different shapes. Trends Pharmacol. Prado, G. and LaPlaca, M.C. (2004) Neuronal plasma
Sci., 20(2): 46–51. membrane is transiently disrupted by mechanical trauma:
Palmer, A.M., Marion, D.W., Botscheller, M.L., Swedlow, an insight into the mechanisms involved. J. Neurotrauma, 21:
P.E., Styren, S.D. and DeKosky, S.T. (1993) Traumatic brain 1331.
injury-induced excitotoxicity assessed in a controlled cortical Pravdenkova, S.V., Basnakian, A.G., James, S.J. and Andersen,
impact model. J. Neurochem., 61(6): 2015–2024. B.J. (1996) DNA fragmentation and nuclear endonuclease
Pettus, E.H. and Povlishock, J.T. (1996) Characterization of a activity in rat brain after severe closed head injury. Brain Res.,
distinct set of intra-axonal ultrastructural changes associated 729(2): 151–155.
with traumatically induced alteration in axolemmal perme- Raghupathi, R. (2004) Cell death mechanisms following trau-
ability. Brain Res., 722(1–2): 1–11. matic brain injury. Brain Pathol., 14(2): 215–222.
Pike, B.R., Flint, J., Dutta, S., Johnson, E., Wang, K.K. Raghupathi, R., Conti, A.C., Graham, D.I., Krajewski, S.,
and Hayes, R.L. (2001) Accumulation of non-erythroid Reed, J.C., Grady, M.S., Trojanowski, J.Q. and McIntosh,
alpha II-spectrin and calpain-cleaved alpha II-spectrin break- T.K. (2002) Mild traumatic brain injury induces apoptotic
down products in cerebrospinal fluid after traumatic brain cell death in the cortex that is preceded by decreases in cellular
injury in rats. J. Neurochem., 78(6): 1297–1306. Bcl-2 immunoreactivity. Neuroscience, 110(4): 605–616.
Pike, B.R., Zhao, X., Newcomb, J.K., Glenn, C.C., Anderson, Raghupathi, R., Graham, D.I. and McIntosh, T.K. (2000)
D.K. and Hayes, R.L. (2000) Stretch injury causes calpain Apoptosis after traumatic brain injury. J. Neurotrauma,
and caspase-3 activation and necrotic and apoptotic cell 17(10): 927–938.
death in septo-hippocampal cell cultures. J. Neurotrauma, Reeves, T.M., Phillips, L.L. and Povlishock, J.T. (2005)
17(4): 283–298. Myelinated and unmyelinated axons of the corpus callosum
Pike, B.R., Zhao, X., Newcomb, J.K., Posmantur, R.M., differ in vulnerability and functional recovery following
Wang, K.K. and Hayes, R.L. (1998) Regional calpain and traumatic brain injury. Exp. Neurol., 196(1): 126–137.
59

Rink, A., Fung, K.M., Trojanowski, J.Q., Lee, V.M., Van den Heuvel, C., Blumbergs, P.C., Finnie, J.W., Manavis,
Neugebauer, E. and McIntosh, T.K. (1995) Evidence of J., Jones, N.R., Reilly, P.L. and Pereira, R.A. (1999) Upreg-
apoptotic cell death after experimental traumatic brain injury ulation of amyloid precursor protein messenger RNA in re-
in the rat. Am. J. Pathol., 147(6): 1575–1583. sponse to traumatic brain injury: an ovine head impact
Ross, D.T., Graham, D.I. and Adams, J.H. (1993) Selective model. Exp. Neurol., 159(2): 441–450.
loss of neurons from the thalamic reticular nucleus following Van den Heuvel, C., Lewis, S., Wong, M., Manavis, J., Finnie,
severe human head injury. J. Neurotrauma, 10(2): 151–165. J., Blumbergs, P.C., Jones, N. and Reilly, P. (1998) Diffuse
Runnerstam, M., Bao, F., Huang, Y., Shi, J., Gutierrez, E., neuronal perikaryon amyloid precursor protein immunore-
Hamberger, A., Hansson, H.A., Viano, D. and Haglid, K. activity in a focal head impact model. Acta Neurochir. Suppl.
(2001) A new model for diffuse brain injury by rotational (Wien.), 71: 209–211.
acceleration: II. Effects on extracellular glutamate, intracra- Wang, K.K. (2000) Calpain and caspase: can you tell the
nial pressure, and neuronal apoptosis. J. Neurotrauma, 18(3): difference? Trends Neurosci., 23(1): 20–26.
259–273. Wolf, J.A., Stys, P.K., Lusardi, T., Meaney, D. and Smith,
Saatman, K.E., Bozyczko-Coyne, D., Marcy, V., Siman, R. and D.H. (2001) Traumatic axonal injury induces calcium
McIntosh, T.K. (1996) Prolonged calpain-mediated spectrin influx modulated by tetrodotoxin-sensitive sodium channels.
breakdown occurs regionally following experimental brain J. Neurosci., 21(6): 1923–1930.
injury in the rat. J. Neuropathol. Exp. Neurol., 55(7): Yakovlev, A.G. and Faden, A.I. (2004) Mechanisms of neural
850–860. cell death: implications for development of neuroprotective
Sahenk, Z. and Lasek, R.J. (1988) Inhibition of proteolysis treatment strategies. NeuroRx, 1(1): 5–16.
blocks anterograde-retrograde conversion of axonally trans- Yakovlev, A.G., Ota, K., Wang, G., Movsesyan, V., Bao, W.L.,
ported vesicles. Brain Res., 460(1): 199–203. Yoshihara, K. and Faden, A.I. (2001) Differential expression
Singleton, R.H. and Povlishock, J.T. (2004) Identification and of apoptotic protease-activating factor-1 and caspase-3 genes
characterization of heterogeneous neuronal injury and death and susceptibility to apoptosis during brain development and
in regions of diffuse brain injury: evidence for multiple in- after traumatic brain injury. J. Neurosci., 21(19): 7439–7446.
dependent injury phenotypes. J. Neurosci., 24(14): Yamashima, T. (2000) Implication of cysteine proteases
3543–3553. calpain, cathepsin and caspase in ischemic neuronal death
Singleton, R.H., Zhu, J., Stone, J.R. and Povlishock, J.T. (2002) of primates. Prog. Neurobiol., 62(3): 273–295.
Traumatically induced axotomy adjacent to the soma does not Yamashima, T. (2004) Ca2+-dependent proteases in ischemic
result in acute neuronal death. J. Neurosci., 22(3): 791–802. neuronal death: a conserved ‘calpain-cathepsin cascade’ from
Smith, D.H., Chen, X.H., Xu, B.N., McIntosh, T.K., Gennarelli, nematodes to primates. Cell Calcium, 36(3–4): 285–293.
T.A. and Meaney, D.F. (1997) Characterization of diffuse ax- Yamashima, T., Tonchev, A.B., Tsukada, T., Saido, T.C.,
onal pathology and selective hippocampal damage following Imajoh-Ohmi, S., Momoi, T. and Kominami, E. (2003)
inertial brain trauma in the pig. J. Neuropathol. Exp. Neurol., Sustained calpain activation associated with lysosomal rup-
56(7): 822–834. ture executes necrosis of the postischemic CA1 neurons in
Stone, J.R., Okonkwo, D.O., Dialo, A.O., Rubin, D.G., Mutlu, primates. Hippocampus, 13(7): 791–800.
L.K., Povlishock, J.T. and Helm, G.A. (2004) Impaired Yang, J., Liu, X., Bhalla, K., Kim, C.N., Ibrado, A.M., Cai, J.,
axonal transport and altered axolemmal permeability occur Peng, T.I., Jones, D.P. and Wang, X. (1997) Prevention
in distinct populations of damaged axons following traumatic of apoptosis by Bcl-2: release of cytochrome c from
brain injury. Exp. Neurol., 190(1): 59–69. mitochondria blocked. Science, 275(5303): 1129–1132.
Strich, S.J. (1956) Diffuse degeneration of the cerebral white Zipfel, G.J., Babcock, D.J., Lee, J.M. and Choi, D.W. (2000)
matter in severe dementia following head injury. J. Neurol. Neuronal apoptosis after CNS injury: the roles of glutamate
Neurosurg. Psychiatry, 19: 163–185. and calcium. J. Neurotrauma, 17(10): 857–869.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 5

Astroglia: Important mediators of traumatic brain


injury

Candace L. Floyd1, and Bruce G. Lyeth2

1
Department of Physical Medicine and Rehabilitation, Center for Glial Biology in Medicine, 547 Spain Rehabilitation
Center, University of Alabama at Birmingham, Birmingham, AL 35249, USA
2
Department of Neurological Surgery, University of California, 1515 Newton Court, One Shields Avenue, Davis,
CA 95816, USA

Abstract: Traumatic brain injury (TBI) research to date has focused almost exclusively on the patho-
physiology of injured neurons with very little attention paid to non-neuronal cells. However in the past
decade, exciting discoveries have challenged this century-old view of passive glial cells and have led to a
reinterpretation of the role of glial cells in central nervous system (CNS) biology and pathology. In this
chapter we review several lines of evidence, indicating that glial cells, particularly astrocytes, are active
partners to neurons in the brain, and summarize recent findings that detail the significance of astrocyte
pathology in traumatic brain injury.

Keywords: astrocyte; glutamate; sodium; calcium; acidosis; mechanical strain injury

Introduction the significance of astrocyte pathology in traumatic


brain injury.
Traumatic brain injury (TBI) research to date has
focused almost exclusively on the pathophysiology
Astrocytes in normal brain function: recasting an
of injured neurons with very little attention paid to
old ‘‘star’’
non-neuronal cells. This near-exclusive focus on
neuroprotection likely reflects the predominant
One line of evidence that astrocytes are more than
paradigm of the neuroscience community as a
supportive connective tissue comes from recent de-
whole, which characterized glial cells as a special-
tailed analyses of the cellular morphology. A
ized type of connective tissue that merely provided
unique morphological feature of astrocytes is that
support for the neurons. However in the past dec-
nearly the entire cell surface is covered with proc-
ade, exciting discoveries have challenged this
esses that extend and become lamellae or filopodia
century-old view of passive glial cells and have
as indicated by three-dimensional reconstruction of
led to a reinterpretation of the role of glial cells in
electron micrographs. Most lamellae and filopodia
central nervous system (CNS) biology and patho-
originate from processes, which, like the cell
logy. In this chapter we review several lines of
body, contain organelles and cytoskeletal elements;
evidence, indicating that glial cells, particularly
however, the cell surface extensions do not contain
astrocytes, are active partners to neurons in the
organelles and intermediate filaments themselves
brain, and summarize recent findings which detail
and are therefore not visualized with GFAP
immunohistochemistry (Chao et al., 2002). Thus,
Corresponding author. E-mail: clfloyd@uab.edu GFAP immunohistochemistry provides only a

DOI: 10.1016/S0079-6123(06)61005-4 61
62

limited view of the total three-dimensional area and extracellular environment in ways that were tradi-
morphological complexity of a given astrocyte’s tionally the domain of neurons, the ‘‘excitable
domain. In proprotoplasmic astrocytes of the cor- cells’’ of the CNS.
tex, cell surface extensions account for 50–60% of Not only do astrocytes have the cyto-machinery
the cytoplasmic volume of the cell (the remainder is of excitable cells, but also they use it. Astrocyte
the cell body and processes) and cover 80% of excitability comprises activation by external or in-
the cell surface. Structurally, astrocytes are highly ternal signals followed by subsequent transfer of a
interdigitated such that neighboring processes often specific message to nearby cells and has been
correspond to different astrocytic cell bodies. Also termed ‘‘gliotransmission’’ (Bezzi and Volterra,
astrocyte processes frequently surround neuronal 2001). The foundation of astrocyte excitability is
synapses; for example in the rat neocortex, 56% of calcium signaling, both intracellular and intercel-
all synapses are contacted by astrocytes (Chao lular; and since astrocytes do not generate action
et al., 2002). Large variations in the structural potentials, imaging of calcium transients and os-
characteristics of glia-synaptic contacts exist such cillations has been more effective than traditional
that common morphologies run the gambit from electrophysiology techniques in studying astrocyte
partial to entire encasement of a single axo- excitation (Volterra and Meldolesi, 2005). Two
dendritic synapse or a triadic/serial synapse to well-documented forms of astrocyte excitation are
complete coverage of complex synapses including (a) neuron-dependent excitation and (b) neuron-
centro-dendritic, centro-axonic, and multicenteric independent excitation. Neuron-dependent excita-
glomeruli synapses (Chao et al., 2002). The exten- tion, mainly through elevations in transmitter
sive astrocyte–synapse connections have led to the outside of the synaptic cleft, was originally de-
hypothesis that many synapses are actually of a scribed for glutamate transmission in hippocam-
tripartite nature, comprising pre- and post-synaptic pus and cerebellum but now includes multiple
neuronal elements and a third glial component brain circuits as well as multiple receptors systems
(Haydon, 2001; Chao et al., 2002). including GABA, acetylcholine, dopamine, ATP,
A second line of evidence for the active role of BDNF, etc. (Haydon, 2001). Neuron-independent,
astrocytes in brain function is that astrocytes ex- or spontaneous, excitation of astrocytes occurs
press diverse neurotransmitter receptors that regu- mainly during development, but has also been
late the concentration of intercellular calcium demonstrated in adult preparations (Aguado et al.,
([Ca2+]i). Astrocytes express receptors for gluta- 2002) and may occur more frequently under path-
mate (Glaum et al., 1990; Holzwarth et al., 1994), ological conditions such as CNS injury as dis-
acetylcholine (Kondou et al., 1994; Shao and cussed later in this chapter. Neuron-independent
McCarthy, 1995), ATP (Neary and Zhu, 1994), calcium oscillations principally involve release of
and histamine (Peakman and Hill, 1995). Addi- calcium from intracellular stores, but calcium en-
tionally, astrocytes express voltage-gated Ca2+ try via voltage-gate channels may also play a role
channels similar to those in neurons (L- and (Parri et al., 2001; Nett et al., 2002; Parri and
N-type) and Ca2+ entry through these channels Crunelli, 2003). Spontaneous excitation of as-
alters [Ca2+]i (MacVicar et al., 1991; D’Ascenzo trocytes results in subsequent activation of neigh-
et al., 2004). Activation of voltage-gated Ca2+ boring cells, including astrocytes and neurons,
channels requires depolarization that has been which indicates that both cell types are sources of
shown in cultured astrocytes, organotypic cultures, excitation and communication networks may be
and acutely isolated astrocytes following far more complex than an astrocytic response to
local changes in extracellular K+ concentration neuronal excitation (Volterra et al., 2005).
(Verkhratsky et al., 1998). Importantly, astrocytes The cellular mechanisms of intercellular calcium
respond to neuronal transmitter release by gener- signaling in astrocytes remain an area of active in-
ation of calcium waves (Dani et al., 1992; Porter quiry. Comprehensive reviews detailing findings
and McCarthy, 1996). Thus astrocytes have intact leading to the current understanding of the mech-
cyto-machinery to sense and respond to the anisms of astrocyte intercellular calcium waves
63

have recently been published (Haydon, 2001; both shorter-range (gap-junction mediated) and
Volterra et al., 2005), but the most prevalent work- longer-range (ATP-mediated) calcium waves (for
ing hypotheses for these mechanisms are briefly de- review, see Haydon, 2001).
scribed here. Intercellular calcium signaling in as- One functional consequence of astrocyte cal-
trocyte comprises a calcium wave that is essentially cium signaling is glutamate release (Parpura et al.,
a sequence of increases in intracellular calcium that 1994; Bezzi et al., 1998). Using live-cell fluorescent
spreads, like a wave, throughout a group of as- imaging and cell culture, glutamate release from
trocytes. Astrocyte calcium waves were initially astrocyte was visualized by applying the cofactors
characterized in response to glutamate application NAD+ and the enzyme glutamate dehydrogenase
or mechanical stimulation to single astrocytes in in the bath so that any released glutamate is con-
cell culture (Cornell-Bell et al., 1990; Cornell-Bell verted to a-ketogluatarate and the NAD+ is re-
and Finkbeiner, 1991), and these findings have duced to the fluorescent NADH. Combination of
been corroborated in organotypic hippocampal this imaging technique with live-cell calcium im-
slice cultures (Harris-White et al., 1998), Müller aging demonstrated that a wave of extracellular
glia of acutely isolated retina (Newman and Zahs, glutamate accompanies an intercellular calcium
1998), and more recently in vivo (Nimmerjahn wave (Innocenti et al., 2000). Subsequent charac-
et al., 2004). In astrocytes, a ligand (such as gluta- terization of glutamate release from astrocytes has
mate) binds to a G-protein receptor coupled to shown that this release is not only related to in-
phosphoinositide-specific phospholipase C (PLC) tracellular calcium increases, but is actually cal-
which cleaves phosphatidylinositol 4,5 bisphos- cium dependent and is very likely regulated by
phate (PIP2) yielding the second messenger exocytosis (Parpura and Haydon, 2000; Anlauf
inositol-1,4,5 trisphosphate (IP3). IP3 then binds and Derouiche, 2005). In contrast to neurons
to the IP3 receptor (IP3R) located on intracellular where extracellular calcium is a main component
calcium stores, causing increased intracellular cal- of calcium-dependent exocytosis, both the IP3 and
cium. Elevated intracellular calcium in a single cell ryanodine-sensitive internal calcium stores seem to
can translate into an intercellular calcium wave by be key components in calcium-dependent gluta-
at least two known mechanisms, working inde- mate release from astrocytes (Araque et al., 1999).
pendently or in concert. First, an intercellular cal- In further support of the hypothesis that astrocytes
cium wave can spread between astrocytes via gap- exocytose glutamate, distinct vesicular compart-
junction mediated metabolic coupling. In this ments containing glutamate have been localized in
model, IP3 diffuses between gap junctions and astrocytes (Kreft et al., 2004); and astrocytes ex-
stimulates the release of intracellular calcium from press key elements of vesicular exocytosis,
adjacent astrocytes resulting in a wave of calcium. SNARE-family proteins (Parpura et al., 1995;
Additionally, an extracellular component may also Crippa et al., 2006). However, astrocyte exocytosis
be involved since studies show that calcium waves of glutamate is slower (2 orders of magnitude)
can travel between cells separated by a cell-free than neuronal counterparts (Kreft et al., 2004).
zone (Hassinger et al., 1996; Newman and Zahs, The primary implication of astrocyte release of
1997) and that waves follow the direction of extra- transmitters, such as glutamate, is modulation of
cellular perfusion (Hassinger et al., 1996). Since synaptic transmission in nearby neurons and this
waves of extracellular ATP can accompany inter- modulation has been shown in several experimen-
cellular calcium waves, the most likely candidate tal preparations. In cell culture, astrocytes were
for the extracellular message for calcium wave shown to either evoke or depress synaptic activa-
propagation is ATP (Cotrina et al., 1998b, 2000; tion and this activity was regulated by NMDA or
Guthrie et al., 1999). In this second model, as- metabotropic glutamate receptors (Araque et al.,
trocytes release ATP that then activates the 1999). In hippocampal slice cultures, glutamate
purinergic receptor P2Y1, thereby increasing IP3 release from astrocytes augmented interneuron
and producing a calcium signal in nearby cells. pyramidal synaptic connections (Kang et al.,
These two pathways may work together to produce 1998; Liu et al., 2004a) and activated kainate
64

receptors on interneurons (Liu et al., 2004b). compromise critical neuronal–glia interactions and
Functional consequences of astrocyte release of thus, may play a significant role in outcome after
D-serine have also been demonstrated in that as- injury. Perturbations in astrocyte function can also
trocyte-derived D-serine, through activation at the have indirect effects on normal brain function. For
allosteric glycine-binding site, facilitated NMDA example, alterations in glial glutamate transporter
receptor activity (Mothet et al., 2005; Panatier number or function could indirectly alter gluta-
et al., 2006). Also, as described above, astrocytes mate-mediated excitability. The recent findings
can release ATP. ATP can be converted to adeno- that D-serine, which is exclusively released from
sine, which then can hyperpolarize adjacent neu- astrocytes in the synapse, preferentially acts as
rons as has been shown in the retina (Newman, the NMDA receptor co-agonist (Boehning and
2003). Taken together, these studies demonstrate Snyder, 2003) would suggest that any injury-
that under physiological conditions astrocytes re- induced alteration in astrocyte function could ad-
lease transmitters that alter neuronal signaling. versely impact normal glutamatergic signaling.
While the effects of CNS injury on intra- and The majority of TBI laboratory studies that have
intercellular calcium signaling as well as on as- investigated astrocyte response to injury have fo-
trocyte neuronal signaling have yet to be fully elu- cused on the phenomena of reactive astrocytes char-
cidated, a strong body of evidence suggests that acterized by increased GFAP immunoreactivity,
astrocytes are not merely passive support cells but hypertrophy, and hyperplasia. The CNS responds
are active communication partners to neurons in within days to injury by producing reactive as-
the uninjured brain. Perhaps a better understand- trogliosis and glial scarring (McGraw et al., 2001).
ing of the effect of trauma on these elements of Although not completely understood, glial scarring
astrocyte communication will provide the infra- is thought to be an attempt by the CNS to restore
structure to develop therapeutic interventions that homeostasis by isolating the damaged region (Fitch
target multiple cell types in the injured brain. et al., 1999). However, the glial scar may interfere
with any subsequent neural repair or axonal regen-
eration (Ridet et al., 1997). Two days after midline
Revisiting and revising the ‘‘Classical View’’ of fluid percussion TBI in the rat, increased GFAP
astrocytes in CNS trauma staining and thickened glia consistent with reactive
astrocytes were observed in the CA3 hippocampus
As discussed in the introduction, TBI research to (D’Ambrosio et al., 1999) even though midline fluid
date has focused mainly on neuroprotection and percussion TBI is generally not associated with CA3
given little consideration to the role of glial cells in pyramidal cell loss (Lyeth et al., 1990). Reactive as-
injury. This has significant implications for the trogliosis and the formation of a glial scar have been
understanding of brain pathology after TBI viewed as both a promoter of and an impediment to
since the population of glial cells in brain is actu- CNS regeneration (Reier et al., 1983; Muller et al.,
ally much larger than neurons. Furthermore, the 1995; Ridet et al., 1997). This chapter focuses on
importance of the complex communication and events that involve TBI-induced astrocyte damage
interaction between astrocytes and neurons de- or death that likely occur prior to the reactive as-
scribed above are becoming more apparent in trocytic responses. Therapeutic interventions tar-
normal brain function and increasingly appear to geted at these early mechanisms of astrocyte injury
play a critical role in such perturbed brain func- may rescue both astrocytes and neurons and as a
tions as seizures and ischemia injury (Vernadakis, consequence may reduce glial scarring.
1996; Vesce et al., 1999). Increasingly, research
studies are indicating that astroglia cells are ad- Role of glutamate in astrocyte pathology in CNS
versely affected by trauma and ischemia and that trauma
damage to astrocytes also affects the fate of neu-
rons in the traumatically injured brain. Early im- During normal neuronal activity extracellular
pairment of astrocyte function after TBI may glutamate in the synaptic cleft must be rapidly
65

cleared to optimize the signal-to-noise ratio as well triggers activation of the Na+-K+ ATPase, which
as to prevent neuronal damage from excitotoxic- is fueled by the ATP produced by glycolysis (Silver
ity. Glutamate is removed from the synapse by and Erecinska, 1997). Further energy demand is
glutamate transporters located in the plasma mem- created by the conversion of glutamate to gluta-
brane of neurons and adjacent astrocytes (Kanner mine by glutamine synthetase. Energy demands of
and Schuldiner, 1987). The major glutamate re- the brain are met through several pathways includ-
moval mechanism is through the astrocyte-specific ing the oxidative phosphorylation, glycolytic, and
sodium-dependent glutamate transporters, GLT-1 glycogen pathways. Recent studies indicate that
and GLAST (Danbolt, 1994, 2001; Takahashi astrocytic glycolysis plays a major role in supplying
et al., 1997). GLT-1 is located primarily in the rat rapid energy demands of astrocytes (Schurr et al.,
forebrain while GLAST is found primarily in the 1999; Paemeleire and Leybaert, 2000). Glycolysis
cerebellum (Tanaka et al., 1997; Danbolt, 2001). also provides an important source for neuronal
Glutamate taken up by astrocytes is converted to energy substrates in the form of lactate for subse-
glutamine by glutamine synthetase, an energy-de- quent conversion to pyruvate for use in the TCA
pendent process requiring one molecule of ATP and oxidative phosphorylation pathways (Schurr
for each molecule of glutamate converted to gluta- et al., 1999; Paemeleire and Leybaert, 2000). The
mine (Daikhin and Yudkoff, 2000). Glutamine majority of excess lactate measured following TBI
then diffuses out of the astrocyte and is taken up is most likely of astrocytic origin and contributes to
by the presynaptic neuron for recycling back into lactic acidosis (Kawamata et al., 1992, 1995; Lyeth
glutamate via the enzyme glutaminase. et al., 1996). The lactate released as a by-product of
Glutamate excitotoxicity is a significant deter- glycolysis in astrocytes is taken up by neurons and
minant of TBI pathophysiology. Numerous labo- used as an aerobic energy substrate. In vitro slice
ratory studies (Faden et al., 1989; Katayama et al., experiments show that adjacent neurons use lactate
1990; Nilsson et al., 1990; Zhong et al., 2006) and generated in astrocytes. The elevated levels of lac-
several clinical studies (Zauner et al., 1996; Bullock tate in slices exposed to high glutamate and ample
et al., 1998; Koura et al., 1998) have documented glucose are significantly increased when lactate
an excessive glutamate release following TBI. The transport into neurons is inhibited (Schurr et al.,
elevated extracellular concentration of glutamate 1999). Furthermore, neuronal functional viability is
results in an excitotoxicity from excessive activa- also lost when lactate transport into neurons is in-
tion of ion-channel linked (Hayes et al., 1988; hibited (Schurr et al., 1999). Elevated tissue levels
McIntosh et al., 1989, 1998; Hicks et al., 1994) of lactate associated with hypoxia or ischemia have
and G-protein linked glutamate receptors (Hayes often been considered an indicator of anaerobic
et al., 1988; Hicks et al., 1994; Mukhin et al., 1996, metabolism. However, many studies now suggest
1997; Faden et al., 1997; Lyeth et al., 2001; that brain tissue also can produce lactate aerobi-
Zwienenberg et al., 2001). Experimental cerebral cally under certain physiological conditions when
ischemia is also associated with a large, rapid stimulated (Raichle, 1991; Wilsch et al., 1994;
increase in extracellular glutamate (Choi and Hyder et al., 1996; Hu and Wilson, 1997). Taken
Rothman, 1990). together, these results indicate that mechanical in-
jury to astrocytes rapidly elevates [Na+]i, followed
by a rapid energy demand, lactate production, and
Energy demands and acidosis in CNS trauma intracellular acidosis.
The role of lactacidosis and a fall in intracellular
The uptake of glutamate rapidly increases energy pH in brain injury and ischemia is well docu-
demands on astrocytes through several mecha- mented (Siesjo, 1988; Kraig and Chesler, 1990;
nisms. Mechanical damage to astrocytes elicits el- Staub et al., 1990; Nedergaard et al., 1991a, b).
evated [Na+]i in large part from cotransport with Electron microscopy studies show that acidosis
glutamate (Floyd et al., 2005). The sodium co- promotes cytotoxic edema that is localized pre-
transported into astrocytes along with glutamate dominantly in astrocytic endfeet (Garcia et al.,
66

1977; Jenkins et al., 1982). Acidosis-induced glial severe astrocytic edema, dissociation of the peri-
swelling is attributed to several membrane trans- vascular astrocyte endfeet from the capillary base-
porters including Na+/H+ antiporter (Grinstein ment membrane, and disruption of inter-astrocyte
and Rothstein, 1986), Cl/HCO3 exchanger gap junctions (Castejon, 1998). Electron micro-
(Kimelberg et al., 1990), and the Na+/HCO3 co- scopy of brain tissue from human cerebral contu-
transporter (Newman, 1991). Activation of these sion patients has revealed evidence of astrocytic
pH-stabilizing mechanisms during TBI-induced swelling as early as 3 h after injury, persisting for as
acidosis would elicit increases in glial intracellular long as 3 days (Bullock et al., 1991). It is important
Na+ and Cl along with osmotically obligated to consider that human tissue is essentially never
water leading to astrocyte swelling (Kempski et al., available for analysis during the first hours after
1988; Jakubovicz and Klip, 1989). Recent studies insult. Thus, there is a distinct possibility that hu-
have demonstrated that amiloride analogues that man astrocyte swelling may occur very rapidly after
inhibit the NHE-1 significantly reduce hippocam- injury as in animal models of ischemia and TBI. In
pal extracellular glutamate levels following tran- addition to cell swelling and raised intracranial
sient cerebral ischemia in rat (Phillis et al., 1998) pressure, the alterations in astrocyte ionic homeo-
and reduce CA1 hippocampal neuronal cell death stasis can likely initiate cell damage and death.
assessed at 6 days after transient cerebral ischemia There has been a dogma that astrocytes are
in the gerbil (Phillis et al., 1999). Phillis et al. highly resistant to hypoxic/ischemic insults com-
(1999) attributed their results to the actions of pared to neurons (Lukaszevicz et al., 2002). This
amiloride in reducing astrocyte swelling and dogma has been challenged by early suggestions
thereby increasing extracellular space. In a study and by later experiments demonstrating that as-
by Plesnila et al. (1999), astrocyte cultures exposed trocytes are indeed vulnerable to ischemic condi-
to lactic acid exhibited cell volume increases of tions. Over 20 years ago, Plum (Plum, 1983)
21% when pHi dropped to 6.8–6.2, levels observed suggested that the degree of infarction from focal
in TBI. Swelling was completely blocked with the ischemia might depend upon the viability of as-
NHE-1 inhibitor, amiloride. Ringel et al. (2000) trocytes. This proved to be an insightful prediction
found that inhibition of anion transports reduced and subsequent studies indicate that this sugges-
lactacidosis-induced swelling in astrocyte cultures. tion applies to TBI as well as cerebral ischemia. A
Activation of the pH-regulated membrane trans- number of in vitro studies have demonstrated that
porters during periods of intracellular acidosis astrocytes are indeed vulnerable to ischemia. As-
contribute to further elevations in [Na+]i and may trocytes exposed to hypoxia alone can require up
result in reversal of the sodium/calcium exchanger to 24 h exposure to produce widespread death
as discussed below. (Yu et al., 1989; Sochocka et al., 1994) even in the
absence of glucose (Kelleher et al., 1993). How-
ever, combining hypoxia with acidosis, a more re-
Acute astrocyte loss following CNS insult alistic model, accelerates astrocyte death to just a
few hours (Swanson et al., 1997). Recent in vitro
Astrocyte swelling is a prominent feature of both studies have examined this issue utilizing a novel
cerebral ischemia (Jenkins et al., 1982) and TBI experimental condition in which cultured as-
(Castejon, 1998) and is considered to be an ‘‘exag- trocytes were exposed to hypoxic, acidic, ion-
gerated extension of normal astrocyte function’’ shifted Ringers (HAIR) solution that mimics the
(Kimelberg, 1992). Astrocyte swelling is likely a environmental characteristics of ischemia and TBI
prominent component of raised ICP after both is- (Bondarenko and Chesler, 2001a). Their microen-
chemia and TBI and resulting in part from alter- vironment included glucose and thus mimicked
ations in ion-transport mechanisms that leads to incomplete ischemia as well as severe TBI. They
osmotically obligated water entry (Kimelberg found that as little as 15–20 min exposure to their
et al., 1990). Cortical biopsies of human TBI com- HAIR solution resulted in death of 60% of the
plicated with subdural hematoma have shown astrocytes (Bondarenko and Chesler, 2001a). The
67

same group subsequently reported that the NHE-1 and TBI (Rink et al., 1995; Newcomb et al., 1999).
inhibitor amiloride protected astrocytes exposed to Most of these studies have focused on apoptotic
the HAIR protocol. Furthermore, blocking the cell death of neurons. Astrocytes are also vulner-
Na+/Ca2+ exchanger with benzamil or KB- able to apoptotic cell death that may contribute to
R7943, which preferentially inhibits reverse Na+/ the pathogenesis of many acute and chronic ne-
Ca2+ exchange, protected astrocytes from the urodegenerative disorders (Takuma et al., 2004).
HAIR protocol (Bondarenko and Chesler, Hypoxia/ischemia induces mitochondrial depolari-
2001b; Bondarenko et al., 2005). These studies zation in astrocytes (Smith et al., 2003), that trig-
provide evidence that astrocytic intracellular aci- gers caspase-dependent cell death pathways and by
dosis activates NHE-1 leading to Na+ entry and poly(ADP-ribose) polymerase-1 cell death path-
the reversal of the Na+/Ca2+ exchanger, trigger- way (Giffard and Swanson, 2005). Astrocyte
ing astrocyte cell loss after ischemia. apoptosis can be detected by caspase activation
Recent in vivo studies provide further compelling by a variety of in vitro insults including staur-
evidence of early astrocyte loss following cere- osporin (Keane et al., 1997), cycloheximide (Tsuc-
bral ischemia. Liu et al. (1999) reported a 50% de- hida et al., 2002), and ischemic conditions (Yu
crease in GFAP mRNA and protein in the ischemic et al., 2001; Gabryel et al., 2002). Activation of
core within 5 h after permanent middle cerebral apoptotic cascades in astrocytes has also been re-
artery occlusion, and almost complete absence of ported after in vivo TBI. For example, accumula-
GFAP mRNA by 12 h. The loss of GFAP mRNA tion of active caspase-3 was observed for 5 days
preceded the loss of the mRNA for GLUT3, a after controlled cortical impact TBI in rats in
neuron-specific glucose transporter. Immunohisto- both neurons and astrocytes but, interestingly
chemical experiments using other astrocyte markers with a higher proportion of activated caspase-3
including S100beta, GSTYb1, and vimentin pro- in astrocytes than in neurons (Johnson et al.,
duced similar results. Interestingly, mRNA began 2005).
to increase in the peri-infarct area by 3 h and TBI and ischemia share a number of patho-
peaked at 48 h after ischemia, suggestive of reactive physiological similarities including excessive gluta-
astrocyte proliferation in the ischemic penumbra. mate release and selective neuronal cell loss. The
Using a rat subdural hematoma-induced ischemia role of astrocytes in uptake of glutamate may have
model, Jiang et al. (2000) reported complete loss of particular importance to the traumatically injured
GFAP immunostaining at 4 h after the insult in brain where large fluxes of extracellular glutamate
the ischemic core. Protoplasmic astrocytes appear (Faden et al., 1989; Katayama et al., 1990) likely
to be more vulnerable to ischemia than fibrous as- contribute to acute excitotoxic processes (Olney
trocytes. For example, following permanent middle and Ho, 1970; Hayes et al., 1988). Specifically,
cerebral artery occlusion in mice, Lukaszevicz et al. early impairment of astrocyte function after TBI
(2002) found that protoplasmic astrocytes (type 1, may compromise maintenance of homeostasis in
accounting for most of the astrocytes in gray the extracellular microenvironment and disrupt
matter) lost GFAP immunolabeling within minutes critical neuronal–glia interactions. Zhao et al.
in the ischemic core. In contrast, fibrous astrocytes (2003) demonstrated selective loss of GFAP and
(type 2, found predominantly in white matter) glutamine synthetase immunoreactivity in rat
displayed a transient hypertrophy with no conspic- brains as early as 1 h after lateral fluid percussion
uous cell death (Lukaszevicz et al., 2002). These TBI. The loss of immunoreactivity was limited to
studies provide compelling evidence that ischemia regions directly adjacent to areas of selective neu-
is associated with an early loss of astrocytes. ronal vulnerability, the hippocampus CA3. The
Glutamate excitotoxicity can lead to both ne- loss of immunoreactivity for astrocytes markers
crotic cell death and apoptotic programed cell preceded fluoro-jade detection of neuronal degen-
death following application of glutamate agonists eration by several hours suggesting that loss of
(Du et al., 1997; Nicotera et al., 1997) ischemia supporting astrocytes may contribute to subse-
(Namura et al., 1998; Graham and Chen, 2001) quent neuronal cell loss.
68

Unraveling the mechanisms of acute astrocyte Mechanical strain injury to astrocytes produces a
damage with in vitro mechanical injury rapid rise in [Ca2+]i that returns to near-basal lev-
els within 15 min. However, application of either
It is clear from the studies described above that in glutamate or the metabotropic glutamate group I
vivo experimental TBI or ischemia produces rapid (mGluRI) agonist trans-1-aminocyclopentyl-1-3-
and profound damage to astrocytes. Moreover, dicarboxylic acid (tACPD) after injury results in a
several in vitro models of ischemia also show acute significantly blunted rise in [Ca2+]i for up to 24 h
astrocyte pathology. Additionally, several of the post-injury, suggesting that the calcium signaling
astrocytic intracellular events occurring acutely machinery is disturbed by injury (Rzigalinski et al.,
after TBI have been characterized in a series of in 1998). Alterations in intracellular calcium dynam-
vitro studies using a mechanical injury model de- ics after mechanical strain injury may be related to
veloped by Ellis et al. (1995). In this model, cells alterations in inositol trisphosphate (IP3) signaling
are grown on a membrane that is subjected to a as strain injury to astrocytes causes a significant
rapid mechanical deformation that approximates increase in IP3 at 5, 15, and 30 min and at 24 and
the tensile strain or stretch that is a key component 48 h post-injury (Floyd et al., 2001). Interestingly,
in the acceleration/deceleration type of closed hu- the injury-induced increases in IP3 at 15 and
man TBI (Meaney et al., 1995) and in vivo brain 30 min post-injury correspond to time points when
injury (Margulies et al., 1990). This model has [Ca2+]i had returned to near normal levels; how-
been validated by demonstrating that in vitro ever, high IP3 should produce increased [Ca2+]i if
strain injury produces many of the post-traumatic Ca2+ signaling were functioning normally. Instead
responses observed with in vivo injury models in- the opposite was observed — elevated IP3 at time
cluding intracellular lesions to the mitochondria, points when intracellular calcium has returned to
Golgi, and cytoskeletal elements in astrocytes and basal levels which indicates that IP3 may be un-
neurons (Dietrich et al., 1994; Ellis et al., 1995; coupled from its target, the intracellular Ca2+
McKinney et al., 1996), increases in astrocyte and store. Additionally, antagonism of mGluRIs and
neuronal permeability (Ellis et al., 1995; Weber inhibition of PLC attenuated injury-induced un-
et al., 1999; Willoughby et al., 2004), activates coupling of IP3-mediated Ca2+ signaling, reduced
phospholipases (Lamb et al., 1997; Floyd et al., astrocyte damage (Floyd et al., 2001, 2004), and
2001), and induces free radical and isoprostane reduced injury-induced depletion of intracellular
formation (McKinney et al., 1996; Lamb et al., calcium stores (Chen et al., 2004). Thus, mechan-
1997; Hoffman et al., 2000) as well as release of the ical strain injury to astrocytes causes acute eleva-
injury markers S-100b and neuron-specific enolase tions in intracellular calcium and disruption of
(NSE; Slemmer et al., 2002, 2004). Strain-injury of IP3-mediated intracellular calcium signaling.
cultured neurons also alters the Mg2+ block of the Mechanical strain injury to astrocytes also sig-
NMDA receptor (Zhang et al., 1996) and produces nificantly increases intracellular sodium (Floyd
a novel stretch-induced delayed neuronal depo- et al., 2005). Mild and moderate strain injury pro-
larization, which may be related to transient neu- duced a rapid rise in intracellular sodium that
ronal dysfunction observed in vivo (Hamm et al., returned to near-basal levels by 20–25 min post-
1994; Tavalin et al., 1995). Strain-injury in neurons injury while severe injury produced increased in-
also increases activation of the AMPA receptor tracellular sodium for at least 50 min (the duration
by decreasing AMPA receptor desensitization of the experiment). These elevations in intracellu-
(Goforth et al., 1999). Thus, this well character- lar sodium were similar to those induced by exo-
ized mechanical injury model recapitulates many genous glutamate application and were reduced by
aspects of in vivo TBI and is a useful tool to ex- pre-injury application of the sodium-dependent
amine injury-induced changes in acute astrocyte glutamate uptake inhibitor TBOA, indicating that
pathology. sodium which accompanies glutamate uptake
A critical element in astrocytic calcium signa- contributes to injury-induced increases in intra-
ling is regulation of intracellular free Ca2+. cellular sodium. Comparison of the time course of
69

injury-induced elevations in intracellular sodium more prevalent in astrocytes than P2X (Illes and
and calcium indicated that there is significant Ribeiro, 2004). Nucleotides released from injured
overlap in the first 30 min post-injury, which raises cells can activate either the P2X or P2Y and cause
the possibility that sodium/calcium exchange may an increase in intracellular calcium (Illes and
be involved in the acute astrocyte pathology after Ribeiro, 2004) as well as activation of mitogen-
injury. The magnitude and direction of calcium activated protein kinase (MAPK) pathways
flux of the sodium/calcium exchanger is depend- (Gendron et al., 2003). Importantly, activation of
ent, in part, on the Na+ electrochemical gradient, P2X receptors on astrocytes is linked to neurode-
and under normal conditions the sodium/calcium generative events and astrogliosis (for review, see
exchanger operates in the ‘‘forward’’ or calcium- Neary et al., 1996); yet, activation of P2Y receptors
efflux mode to extrude calcium. Importantly, on astrocytes is protective. For example, activation
manipulations in uninjured astrocytes that raise of P2Y2 increases Bcl-2 family genes (Chorna et al.,
intracellular sodium have been shown to increase 2004), and activates extracellular signal-regulated
intracellular calcium by ‘‘reversal’’ of the sodium/ protein kinase (ERK) which has been linked to the
calcium exchanger or calcium-influx operation protective activation of CREB (Neary et al., 1996;
(Goldman et al., 1994). Thus, it was predicted Chorna et al., 2004). In an eloquent series of stud-
that the elevated intracellular sodium observed af- ies, Neary and colleagues have detailed the effects
ter mechanical strain injury may be sufficient to of mechanical strain injury to astrocytes on P2 re-
reverse the sodium/calcium exchanger and cause ceptor activation and found that injury induces
an influx of calcium. It was found that pre-injury a rapid release of ATP which, via P2 receptors,
blockade of the calcium-influx (reversed) mode activates ERK (Neary et al., 2003) and Akt
with the compound KB-R7943 significantly re- (Neary et al., 2005); increases expression and re-
duced intracellular calcium in the moderate and lease of a protein that induces synapse formation,
high levels of injury (but not at the low levels of thrombospondin-1 (Tran and Neary, 2006); and
injury), the magnitudes of injury with the largest also phosphorylates GSK3b at the Ser-9 location,
and more sustained elevations in intracellular so- thereby inhibiting activity (Neary and Kang, 2006).
dium (Floyd et al., 2005). Additionally, this sever- Thus, release of ATP from injured astrocytes can
ity of injury-dependent reversal of the sodium/ act not only as a stimulus for gliosis, but also as a
calcium exchanger was predicted by mathematical promoter of cell survival and synaptic plasticity.
modeling of the reversal potential of the sodium/ Further work is needed in this exciting area to bet-
calcium exchanger using experimental values of ter elucidate the complex relationship between P2
intercellular sodium and calcium concentrations activation, gliosis, and cell survival.
after mechanical strain injury. Taken together, Mechanical strain injury to astrocytes disrupts
these data suggest that increased intracellular so- many of the critical features of intra- and inter-
dium can cause a reversal of the sodium/calcium cellular calcium signaling in astrocytes. The known
exchanger to cause influx of calcium into the cells, changes in astrocyte signaling after mechanical in-
and that this only occurs with moderate or severe jury are summarized schematically in Fig. 1. Strain
injury. injury to astrocytes causes a rapid increase in in-
Mechanical strain injury to astrocytes causes tracellular calcium and sodium that is dependent
release of ATP (Ahmed et al., 2000; Neary et al., on the magnitude of injury (Floyd et al., 2005).
2005). ATP release following mechanical injury There are several mechanisms by which calcium is
not only has consequences for cellular energetics elevated following mechanical injury. Excessive
(Bambrick et al., 2004), but also may alter inter- glutamate following injury agonizes mGluRs to
cellular calcium signaling via P2 purinergic re- cause IP3-mediated calcium release (Floyd et al.,
ceptors. Cortical astrocytes express both the 2004). Theoretically, elevated glutamate following
ionotropic P2X receptors and the G-protein cou- injury could also activate the AMPA ionotropic
pled P2Y receptors (Lenz et al., 2000; Weisman glutamate channel as has been shown in neurons
et al., 2005); however, P2Y expression is much (Goforth et al., 2004). Although the effects of
70

ATP
P2YR Hemichannel?
Ca2+ mGluR ATP
Propidium
Iodide ?
AMPA MITOCHONDRIA
Na+ PLC

X
(reversed)

NC
IP3
Ca2+
1
T-
GL

Adjacent
Na+
Astrocyte TSP-2
Ca2+ Store Akt
ERK
Ca2+
Gap Junction

BCL-2
NUCLEUS

Fig. 1. Pathological events known to occur in astrocytes after mechanical strain injury. Mechanical strain injury to astrocytes results in
a rapid rise in intracellular calcium and sodium. Sources of injury-induced increases in intracellular calcium include activation of the
mGluRs, release of calcium from IP3-mediated intracellular calcium stores, reversal of the sodium/calcium exchanger, and possibly
AMPA receptor activation. Increases in intracellular sodium after mechanical injury are largely due to sodium-dependent uptake of
excessive glutamate. Injured astrocytes also release ATP. ATP could activate P2YRs and further increase intracellular calcium but also
could initiate cell survival pathways such as Bcl-2. Astrocytes are coupled by gap junctions, and small molecules such as IP3 or Ca2+
could travel between cells after injury. Propidium iodide, a marker for dead or damaged cells, could enter the astrocyte through
disruptions in the cell membrane and intercalate into the DNA. (See text for further details.) Abbreviations: protein kinase B/Akt
(Akt): (RS)-2-amino-3-(3-hydroxy-5-methylisoxazol-4-yl)propionate (AMPA), extracellular signal-regulated protein kinase (ERK),
glutamate transporter (GLT-1), inositol trisphosphate (IP3), metabotropic glutamate receptor (mGluR), sodium/calcium exchanger
(NCX), purinergic 2Y receptor (P2YR), phospholipase C (PLC).

mechanical injury on the subunit composition in astrocyte injury, as both increased intracellular
astrocytes has not been examined, yet, if the calcium and sodium are intimately involved. Ex-
GluR2 subunit is present, injury induced activa- cessive glutamate from mechanical injury also af-
tion would likely contribute to increase intracellu- fects the astrocyte by activation of sodium-
lar Na+. However, if the GluR2 subunit were dependent glutamate uptake that significantly
absent, the channel would also be permeable to elevates intracellular sodium. At moderate and se-
Ca2+ (Frandsen and Schousboe, 2003). Activation vere levels of injury, the elevation in intracellular
of either configuration of the AMPA receptor sodium is sufficient to cause reversal of the so-
would likely contribute to the pathophysiology of dium/calcium exchanger and produce calcium

Fig. 2. Effect of mechanical injury on functional coupling in astrocytes. Confluent primary astrocyte cultures we mechanically injured
and then functional coupling was analyzed using fluorescence recovery after photobleach (FRAP). Panel (A) shows representative
micrographs of uninjured (left column) or moderately injured (right column) astrocytes before, immediately after, and 15 s after
photobleach. Regions of interest (ROIs) were drawn around four cells per field. The ROIs outlined in red and green were bleached but
the ROIs in yellow and blue were not. A representative trace of a mildly injured astrocyte at 24 h post-injury is shown in panel (B).
Panel (C) shows quantification of the maximal recovery at various post-injury times. Mild injury increased and moderate injury
decreased functional coupling in astrocytes (n ¼ 3 separate experiments).
71
72

influx, thereby further contributing to elevated in- and production of oxygen-free radicals can inhibit
tracellular calcium (Floyd et al., 2005). Mechanical gap junction coupling (Bolanos and Medina, 1996;
strain injury causes release of ATP from damaged Martinez and Saez, 2000); yet, other evidence sug-
mitochondria (Ahmed et al., 2000; Neary et al., gests that gap junctions remain open during is-
2005), some of which is released into the extracel- chemia (Cotrina et al., 1998a). The role of gap
lular space. Although the mechanism of ATP re- junction communication in cell death following an
lease after injury is still unknown, candidates for ischemic insult is also unclear. Some groups report
release include hemichannels, ATP transporters, that inhibition of astrocyte gap junctions during
or vesicular exocytosis (Schwiebert, 2001). Extra- ischemic injury decreases the area of infarct and is
cellular ATP following injury can activate pur- neuroprotective (Lin et al., 1998; Frantseva et al.,
inergic receptors including the P2YRs. Activation 2002), suggesting that astrocyte gap junctions am-
of the P2YRs can increase intracellular calcium, plify damage and spread cell death signals to oth-
and also can have beneficial effects such as acti- erwise viable cells. An alternative hypothesis
vation of cytoprotective pathways and increased suggests that gap junctions enable astrocytes to
expression of Bcl-2 (Neary et al., 2003, 2005). use the syncytium to dissipate otherwise toxic con-
Propidium iodide, a marker of cell damage and centrations of glutamate or ions (Blanc et al., 1998;
death, could enter the cell via perturbations in the Siushansian et al., 2001). Part of the controversy
cell membrane. As astrocytes are extensively cou- over the role of gap junction communication in
pled via gap junctions, small molecules such as ischemic injury may be related to heterogeneity in
Ca2+ and IP3 could travel between cells to poten- gap junction coupling as well as regional or cel-
tially propagate an injury signal, as will be dis- lular differences in intracellular regulation of gap
cussed below. junctions (Zvalova et al., 2004). For example, gap
Work in uninjured astrocytes shows that gap junction hemichannels that open to the extracel-
junction communication is an important element lular space have been characterized (Contreras
in intercellular calcium signaling in astrocytes et al., 2002) and may be involved in calcium signa-
which facilitates intercellular transfer of ions (i.e., ling or ATP release (Stout et al., 2002; Bennett
Ca2+ or Na+) or second messengers (i.e., IP3). et al., 2003; Dermietzel et al., 2003).
Gap junctions are connections of intercellular The effect of traumatic brain injury on astrocyte
channels formed by hemichannels from adjoining gap junction communication has only recently
cell membranes, and the adult brain astrocytes are been investigated. Ohsumi et al. (2006) have shown
so abundantly interconnected that the network is that CX43 immunohistochemistry was increased in
regarded as a syncytium. Gap junctions are the hippocampus 24 and 72 h after lateral fluid
formed by the joining of 2 hemichannels, each of percussion injury. We evaluated the effect of me-
which comprises six protein subunits termed con- chanical strain injury on functional connectivity of
nexins. The connexin43 protein (Cx43) is highly gap junctions in astrocytes. In this experiment,
expressed in astrocyte gap junctions while other primary astrocyte cultures were grown on Flexs
connexin proteins comprise neuronal gap junc- plates and subjected to either a mild or moderate
tions (Yamamoto et al., 1990a, b; Giaume and rapid mechanical strain injury as previously de-
McCarthy, 1996). Gap junctions are not simply scribed (Floyd et al., 2005). Gap junction coupling
passive channels between cells, but functionally was evaluated at 30 min, 4 h, or 24 h after me-
respond to alterations in the cellular milieu that chanical strain injury using fluorescence recovery
alters gap junction assembly and permeability, after photobleach (FRAP). Astrocytes were
mainly via phosphorylation (Musil et al., 1990; bath loaded with 0.05 mg/ml 5-(and 6-) car-
Nagy and Li, 2000; Rouach et al., 2002; VanSlyke boxyfluorescein diacetate-AM (CFDA) which is
and Musil, 2005). Most studies of the effect of in- a cell membrane and gap junction permeable
jury on gap junction coupling have used models of dye that becomes membrane impermeable after
ischemic brain injury with divergent findings. In- de-esterification. Thus, when cells are visualized,
creases in intracellular calcium, decreases in pH, the CFDA is trapped within the cells but can
73

transfer between coupled cells via gap junctions. was decreased at these same time-points. Recovery
After the fluorescent moieties of the dye are of photobleach was assessed after severe strain in-
bleached by intense laser excitation in a cell, any jury, but many severely injured cells leaked dye
subsequent increase in fluorescent intensity, or re- into the extracellular compartment, rendering the
covery, is attributed to transfer of dye between gap severe injury data inconclusive (data not shown).
junctions and represents the extent of gap junction In summary, we found that mild injury increases
coupling. In this experiment, cells were visualized functional coupling and moderate injury decreases
on an upright Zeiss Axioscop 2 Laser scanning coupling. Additional experiments will be con-
confocal microscope (LSM510) using a Zeiss ducted to better understand the mechanisms of
Achroplan 20  dipping objective. The dye was these changes in gap junction coupling after me-
excited at 400 nm with emitted fluorescence above chanical strain injury and their role in the patho-
520 nm captured by a photo-multiplier tube. Two physiology of traumatic brain injury.
cells near the middle of the field were selected for
bleach and regions of interest (ROIs) were drawn.
Two additional ROIs were drawn for other cells in Conclusion
the field that were not subjected to photobleach.
Three field scans were obtained prior to bleach. With some notable exceptions such as the study of
Selected cells were bleached with 100 iterations at gliosis and astrocytic swelling, most TBI research
100% laser power. To assess recovery after bleach, has generally focused on the pathophysiology of
field scans were acquired every 2 s for the next neurons. However, many recent developments in
2.5 min (75 scans total). Digital images obtained the field of glial biology demonstrate the active
during experiments were saved on a PC for off-line nature of neuronal-astrocyte signaling indicating
analysis. Mean intensity values for each ROI were the vital importance of astrocytes in normal brain
divided by mean intensity values of reference (non- function. Furthermore, recent studies in Neuro-
bleached) cells to correct for photobleach of the trauma demonstrate the vulnerability of astrocytes
entire field. Figure 2(A) shows representative to CNS insults. Therefore, defining the time course
micrographs of uninjured astrocytes (left column) of astrocyte damage after TBI and uncovering the
and astrocyte subjected to a moderate mechanical mechanisms mediating that damage are critical to
strain injury (6.5 mm deformation) 30 min prior to a more complete understanding of TBI patho-
FRAP analysis. The ROIs outlined in red and physiology. Collectively, these findings are leading
green were bleached but the ROIs outlined in yel- to a novel departure from traditional approaches
low and blue were not. A representative trace of to TBI pathophysiology that have generally con-
the percentage of fluorescence recovery over time centrated on injury to neurons.
for an astrocyte mildly injured 24 h prior to FRAP
is shown in Fig. 2(B). Recovery of fluorescence is
rapid and begins to plateau by 400 s after bleach.
Quantification of the maximal recovery at various Acknowledgments
post-injury times is shown in Fig. 2(C). Uninjured
astrocytes were moderately coupled and recovery Support provided by UCD Health Systems Re-
after photobleach was between 30 and 40%. Neu- search Award (CLF), NIH NS29995 (BGL), NIH
rons are reportedly not extensively coupled and NS45136 (BGL).
were used as a procedural control with less than
5% fluorescence recovery detected in adjacent
neurons in co-cultures. In mildly injured as- References
trocytes, a significant increase in recovery after
Aguado, F., Espinosa-Parrilla, J.F., Carmona, M.A. and
photobleach was seen at 30 min, and 4 and 24 h Soriano, E. (2002) Neuronal activity regulates correlated
after mechanical strain injury. However, with network properties of spontaneous calcium transients in as-
moderate strain injury, recovery after photobleach trocytes in situ. J. Neurosci., 22: 9430–9444.
74

Ahmed, S.M., Rzigalinski, B.A., Willoughby, K.A., Sitterding, Synaptic Transmission. Oxford University Press, Oxford,
H.A. and Ellis, E.F. (2000) Stretch-induced injury alters pp. 3–23.
mitochondrial membrane potential and cellular ATP in cul- Chen, T., Willoughby, K.A. and Ellis, E.F. (2004) Group I
tured astrocytes and neurons. J. Neurochem., 74: 1951–1960. metabotropic receptor antagonism blocks depletion of cal-
Anlauf, E. and Derouiche, A. (2005) Astrocytic exocytosis ves- cium stores and reduces potentiated capacitative calcium en-
icles and glutamate: a high-resolution immunofluorescence try in strain-injured neurons and astrocytes. J. Neurotrauma,
study. Glia, 49: 96–106. 21: 271–281.
Araque, A., Sanzgiri, R.P., Parpura, V. and Haydon, P.G. Choi, D.W. and Rothman, S.M. (1990) The role of glutamate
(1999) Astrocyte-induced modulation of synaptic transmis- neurotoxicity in hypoxic-ischemic neuronal death. Annu.
sion. Can. J. Physiol. Pharmacol., 77: 699–706. Rev. Neurosci., 13: 171–182.
Bambrick, L., Kristian, T. and Fiskum, G. (2004) Astrocyte Chorna, N.E., Santiago-Perez, L.I., Erb, L., Seye, C.I., Neary,
mitochondrial mechanisms of ischemic brain injury and J.T., Sun, G.Y., Weisman, G.A. and Gonzalez, F.A. (2004)
neuroprotection. Neurochem. Res., 29: 601–608. P2Y receptors activate neuroprotective mechanisms in as-
Bennett, M.V., Contreras, J.E., Bukauskas, F.F. and Saez, J.C. trocytic cells. J. Neurochem., 91: 119–132.
(2003) New roles for astrocytes: gap junction hemichannels Contreras, J.E., Sanchez, H.A., Eugenin, E.A., Speidel, D.,
have something to communicate. Trends Neurosci., 26: Theis, M., Willecke, K., Bukauskas, F.F., Bennett, M.V. and
610–617. Saez, J.C. (2002) Metabolic inhibition induces opening of
Bezzi, P., Carmignoto, G., Pasti, L., Vesce, S., Rossi, D., unapposed connexin 43 gap junction hemichannels and re-
Rizzini, B.L., Pozzan, T. and Volterra, A. (1998) Prostaglan- duces gap junctional communication in cortical astrocytes in
dins stimulate calcium-dependent glutamate release in astro- culture. Proc. Natl. Acad. Sci. U.S.A., 99: 495–500.
cytes. Nature, 391: 281–285. Cornell-Bell, A.H. and Finkbeiner, S.M. (1991) Ca2+ waves in
Bezzi, P. and Volterra, A. (2001) A neuron-glia signalling net- astrocytes. Cell Calcium, 12: 185–204.
work in the active brain. Curr. Opin. Neurobiol., 11: Cornell-Bell, A.H., Finkbeiner, S.M., Cooper, M.S. and Smith,
387–394. S.J. (1990) Glutamate induces calcium waves in cultured as-
Blanc, E.M., Bruce-Keller, A.J. and Mattson, M.P. (1998) trocytes: long-range glial signaling. Science, 247: 470–473.
Astrocytic gap junctional communication decreases neuronal Cotrina, M.L., Kang, J., Lin, J.H., Bueno, E., Hansen, T.W.,
vulnerability to oxidative stress-induced disruption of Ca2+ He, L., Liu, Y. and Nedergaard, M. (1998a) Astrocytic gap
homeostasis and cell death. J. Neurochem., 70: 958–970. junctions remain open during ischemic conditions. J. Neuro-
Boehning, D. and Snyder, S.H. (2003) Novel neural modu- sci., 18: 2520–2537.
lators. Annu. Rev. Neurosci., 26: 105–131. Cotrina, M.L., Lin, J.H., Alves-Rodrigues, A., Liu, S., Li, J.,
Bolanos, J.P. and Medina, J.M. (1996) Induction of nitric oxide Azmi-Ghadimi, H., Kang, J., Naus, C.C. and Nedergaard,
synthase inhibits gap junction permeability in cultured rat M. (1998b) Connexins regulate calcium signaling by control-
astrocytes. J. Neurochem., 66: 2091–2099. ling ATP release. Proc. Natl. Acad. Sci. U.S.A., 95:
Bondarenko, A. and Chesler, M. (2001a) Calcium dependence 15735–15740.
of rapid astrocyte death induced by transient hypoxia, aci- Cotrina, M.L., Lin, J.H., Lopez-Garcia, J.C., Naus, C.C. and
dosis, and extracellular ion shifts. Glia, 34: 143–149. Nedergaard, M. (2000) ATP-mediated glia signaling. J.
Bondarenko, A. and Chesler, M. (2001b) Rapid astrocyte death Neurosci., 20: 2835–2844.
induced by transient hypoxia, acidosis, and extracellular ion Crippa, D., Schenk, U., Francolini, M., Rosa, P., Verderio, C.,
shifts. Glia, 34: 134–142. Zonta, M., Pozzan, T., Matteoli, M. and Carmignoto, G.
Bondarenko, A., Svichar, N. and Chesler, M. (2005) Role of (2006) Synaptobrevin2-expressing vesicles in rat astrocytes:
Na+-H+ and Na+-Ca2+ exchange in hypoxia-related acute insights into molecular characterization, dynamics and
astrocyte death. Glia, 49: 143–152. exocytosis. J. Physiol., 570: 567–582.
Bullock, R., Maxwell, W.L., Graham, D.I., Teasdale, G.M. and Daikhin, Y. and Yudkoff, M. (2000) Compartmentation of
Adams, J.H. (1991) Glial swelling following human cerebral brain glutamate metabolism in neurons and glia. J. Nutr.,
contusion: an ultrastructural study. J. Neurol. Neurosurg. 130: 1026S–1031S.
Psychiatry, 54: 427–434. D’Ambrosio, R., Maris, D.O., Grady, M.S., Winn, H.R. and
Bullock, R., Zauner, A., Woodward, J.J., Myseros, J., Choi, Janigro, D. (1999) Impaired K(+) homeostasis and altered
S.C., Ward, J.D., Marmarou, A. and Young, H.F. (1998) electrophysiological properties of post-traumatic hippo-
Factors affecting excitatory amino acid release following se- campal glia. J. Neurosci., 19: 8152–8162.
vere human head injury. J. Neurosurg., 89: 507–518. Danbolt, N.C. (1994) The high affinity uptake system for ex-
Castejon, O.J. (1998) Morphological astrocytic changes in citatory amino acids in the brain. Prog. Neurobiol., 44:
complicated human brain trauma: a light and electron micro- 377–396.
scopic study. Brain Inj., 12: 409–427. Danbolt, N.C. (2001) Glutamate uptake. Prog. Neurobiol., 65:
Chao, T.I., Rickmann, M. and Wolff, J.R. (2002) The synapse- 1–105.
astrocyte boundry: an anatomical basis for an inegrative role Dani, J.W., Chernjavsky, A. and Smith, S.J. (1992) Neuronal
of glia in synaptic transmission. In: Volterra A., Magistretti P. activity triggers calcium waves in hippocampal astrocyte net-
and Haydon P.G. (Eds.), The Tripartite Synapse: Glia in works. Neuron, 8: 429–440.
75

D’Ascenzo, M., Vairano, M., Andreassi, C., Navarra, P., Azzena, microscopy of feline cerebral cortex after middle-cerebral-
G.B. and Grassi, C. (2004) Electrophysiological and molecular artery occlusion. Virchows Arch. B. Cell Pathol., 25: 191–206.
evidence of L-(Cav1), N-(Cav2.2), and R-(Cav2.3) type Ca2+ Giaume, C. and McCarthy, K.D. (1996) Control of gap-junc-
channels in rat cortical astrocytes. Glia, 45: 354–363. tional communication in astrocytic networks. Trends Neuro-
Dermietzel, R., Meier, C., Bukauskas, F. and Spray, D.C. sci., 19: 319–325.
(2003) Following tracks of hemichannels. Cell Commun. Giffard, R.G. and Swanson, R.A. (2005) Ischemia-induced
Adhes., 10: 335–340. programmed cell death in astrocytes. Glia, 50: 299–306.
Dietrich, W.D., Alonso, O., Busto, R., Globus, M.Y. and Glaum, S.R., Holzwarth, J.A. and Miller, R.J. (1990) Gluta-
Ginsberg, M.D. (1994) Post-traumatic brain hypothermia mate receptors activate Ca2+ mobilization and Ca2+ influx
reduces histopathological damage following concussive brain into astrocytes. Proc. Natl. Acad. Sci. U.S.A., 87: 3454–3458.
injury in the rat. Acta Neuropathol., 87: 250–258. Goforth, P.B., Ellis, E.F. and Satin, L.S. (1999) Enhancement
Du, Y., Bales, K.R., Dodel, R.C., Hamilton-Byrd, E., Horn, of AMPA-mediated current after traumatic injury in cortical
J.W., Czilli, D.L., Simmons, L.K., Ni, B. and Paul, S.M. neurons. J. Neurosci., 19: 7367–7374.
(1997) Activation of a caspase 3-related cysteine protease is Goforth, P.B., Ellis, E.F. and Satin, L.S. (2004) Mechanical
required for glutamate-mediated apoptosis of cultured cere- injury modulates AMPA receptor kinetics via an NMDA
bellar granule neurons. Proc. Natl. Acad. Sci. U.S.A., 94: receptor-dependent pathway. J. Neurotrauma, 21: 719–732.
11657–11662. Goldman, W.F., Yarowsky, P.J., Juhaszova, M., Krueger, B.K.
Ellis, E.F., McKinney, J.S., Willoughby, K.A., Liang, S. and and Blaustein, M.P. (1994) Sodium/calcium exchange in rat
Povlishock, J.T. (1995) A new model for rapid stretch- cortical astrocytes. J. Neurosci., 14: 5834–5843.
induced injury of cells in culture: characterization of the Graham, S.H. and Chen, J. (2001) Programmed cell death in
model using astrocytes. J. Neurotrauma, 12: 325–339. cerebral ischemia. J. Cereb. Blood Flow Metab., 21: 99–109.
Faden, A.I., Demediuk, P., Panter, S.S. and Vink, R. (1989) Grinstein, S. and Rothstein, A. (1986) Mechanisms of regula-
The role of excitatory amino acids and NMDA receptors in tion of the Na+/H+ exchanger. J. Membr. Biol., 90: 1–12.
traumatic brain injury. Science, 244: 798–800. Guthrie, P.B., Knappenberger, J., Segal, M., Bennett, M.V.,
Faden, A.I., Ivanova, S.A., Yakovlev, A.G. and Mukhin, A.G. Charles, A.C. and Kater, S.B. (1999) ATP released from as-
(1997) Neuroprotective effects of group III mGluR in trau- trocytes mediates glial calcium waves. J. Neurosci., 19:
matic neuronal injury. J. Neurotrauma, 14: 885–895. 520–528.
Fitch, M.T., Doller, C., Combs, C.K., Landreth, G.E. and Hamm, R.J., Pike, B.R., O’Dell, D.M. and Lyeth, B.G. (1994)
Silver, J. (1999) Cellular and molecular mechanisms of glial Traumatic brain injury enhances the amnesic effect of an
scarring and progressive cavitation: in vivo and in vitro anal- NMDA antagonist in rats. J. Neurosurg., 81(2): 267–271.
ysis of inflammation-induced secondary injury after CNS Harris-White, M.E., Zanotti, S.A., Frautschy, S.A. and
trauma. J. Neurosci., 19: 8182–8198. Charles, A.C. (1998) Spiral intercellular calcium waves in
Floyd, C.L., Gorin, F.A. and Lyeth, B.G. (2005) Mechanical hippocampal slice cultures. J. Neurophysiol., 79: 1045–1052.
strain injury increases intracellular sodium and reverses Na(+)/ Hassinger, T.D., Guthrie, P.B., Atkinson, P.B., Bennett, M.V.
Ca(2+) exchange in cortical astrocytes. Glia, 51: 35–46. and Kater, S.B. (1996) An extracellular signaling component
Floyd, C.L., Rzigalinski, B.A., Sitterding, H.A., Willoughby, in propagation of astrocytic calcium waves. Proc. Natl. Acad.
K.A. and Ellis, E.F. (2004) Antagonism of group I meta- Sci. U.S.A., 93: 13268–13273.
botropic glutamate receptors and PLC attenuates increases in Haydon, P.G. (2001) Glia: listening and talking to the synapse.
inositol trisphosphate and reduces reactive gliosis in strain- Nat. Rev. Neurosci., 2: 185–193.
injured astrocytes. J. Neurotrauma, 21: 205–216. Hayes, R.L., Jenkins, L.W., Lyeth, B.G., Balster, R.L.,
Floyd, C.L., Rzigalinski, B.A., Weber, J.T., Sitterding, H.A., Robinson, S.E., Clifton, G.L., Stubbins, J.F. and Young,
Willoughby, K.A. and Ellis, E.F. (2001) Traumatic injury of H.F. (1988) Pretreatment with phencyclidine, an N-methyl-D-
cultured astrocytes alters inositol (1,4,5)- trisphosphate-me- aspartate antagonist, attenuates long-term behavioral deficits
diated signaling. Glia, 33: 12–23. in the rat produced by traumatic brain injury. J. Neurotrau-
Frandsen, A. and Schousboe, A. (2003) AMPA receptor- ma, 5: 259–574.
mediated neurotoxicity: role of Ca2+ and desensitization. Hicks, R.R., Smith, D.H., Gennarelli, T.A. and McIntosh, T.K.
Neurochem. Res., 28: 1495–1499. (1994) Kynurenate is neuroprotective following experimental
Frantseva, M.V., Kokarovtseva, L. and Perez Velazquez, J.L. brain injury in the rat. Brain Res., 655: 91–96.
(2002) Ischemia-induced brain damage depends on specific Hoffman, S.W., Rzigalinski, B.A., Willoughby, K.A. and Ellis,
gap-junctional coupling. J. Cereb. Blood Flow Metab., 22: E.F. (2000) Astrocytes generate isoprostanes in response to
453–462. trauma or oxygen radicals. J. Neurotrauma, 17(5): 415–420.
Gabryel, B., Adamczyk, J., Huzarska, M., Pudelko, A. and Holzwarth, J.A., Gibbons, S.J., Brorson, J.R., Philipson, L.H.
Trzeciak, H.I. (2002) Aniracetam attenuates apoptosis of as- and Miller, R.J. (1994) Glutamate receptor agonists stimulate
trocytes subjected to simulated ischemia in vitro. Neurotoxi- diverse calcium responses in different types of cultured rat
cology, 23: 385–395. cortical glial cells. J. Neurosci., 14: 1879–1891.
Garcia, J.H., Kalimo, H., Kamijyo, Y. and Trump, B.F. (1977) Hu, Y. and Wilson, G.S. (1997) A temporary local energy pool
Cellular events during partial cerebral ischemia. I. Electron coupled to neuronal activity: fluctuations of extracellular
76

lactate levels in rat brain monitored with rapid-response Kempski, O., Staub, F., Jansen, M., Schodel, F. and Baeth-
enzyme-based sensor. J. Neurochem., 69: 1484–1490. mann, A. (1988) Glial swelling during extracellular acidosis
Hyder, F., Chase, J.R., Behar, K.L., Mason, G.F., Siddeek, M., in vitro. Stroke, 19: 385–392.
Rothman, D.L. and Shulman, R.G. (1996) Increased Kimelberg, H.K. (1992) Astrocytic edema in CNS trauma. J.
tricarboxylic acid cycle flux in rat brain during forepaw stim- Neurotrama, 9(Suppl. 1): S71–S81.
ulation detected with 1H[13C]NMR. Proc. Natl. Acad. Sci. Kimelberg, H.K., Goderie, S.K., Higman, S., Pang, S. and
U.S.A., 93: 7612–7617. Waniewski, R.A. (1990) Swelling-induced release of gluta-
Illes, P. and Ribeiro, J.A. (2004) Molecular physiology of P2 mate, aspartate, and taurine from astrocyte cultures. J.
receptors in the central nervous system. Eur. J. Pharmacol., Neurosci., 10: 1583–1591.
483: 5–17. Kondou, H., Inagaki, N. and Fukui, H. (1994) Formation of
Innocenti, B., Parpura, V. and Haydon, P.G. (2000) Imaging inositol phosphates mediated by M3 muscarinic receptors in
extracellular waves of glutamate during calcium signaling in type-1 and type-2 astrocytes from neonatal rat cerebral cor-
cultured astrocytes. J. Neurosci., 20: 1800–1808. tex. Neurosci. Lett., 180: 131–134.
Jakubovicz, D.E. and Klip, A. (1989) Lactic acid-induced Koura, S.S., Doppenberg, E.M., Marmarou, A., Choi, S.,
swelling in C6 glial cells via Na+/H+ exchange. Brain Res., Young, H.F. and Bullock, R. (1998) Relationship between
485: 215–224. excitatory amino acid release and outcome after severe hu-
Jenkins, L.W., Becker, D.P. and Coburn, T.H. (1982) A quan- man head injury. Acta Neurochir. Suppl., 71: 244–246.
titative analysis of glial swelling and ischemic neuronal injury Kraig, R.P. and Chesler, M. (1990) Astrocytic acidosis in
following complete cerebral ischemia. In: Go K.G. and Baeth- hyperglycemic and complete ischemia. J. Cereb. Blood Flow
mann A. (Eds.), Recent Progress in the Study and Therapy of Metab., 10: 104–114.
Brain Edema. Plenum Press, New York, pp. 523–537. Kreft, M., Stenovec, M., Rupnik, M., Grilc, S., Krzan, M.,
Jiang, X.B., Ohno, K., Qian, L., Tominaga, B., Kuroiwa, T., Potokar, M., Pangrsic, T., Haydon, P.G. and Zorec, R.
Nariai, T. and Hirakawa, K. (2000) Changes in local cerebral (2004) Properties of Ca(2+)-dependent exocytosis in cul-
blood flow, glucose utilization, and mitochondrial function tured astrocytes. Glia, 46: 437–445.
following traumatic brain injury in rats. Neurol. Med. Chir., Lamb, R.G., Harper, C.C., McKinney, J.S., Rzigalinski, B.A.
40: 16–28. and Ellis, E.F. (1997) Alterations in phosphatidylcholine
Johnson, E.A., Svetlov, S.I., Wang, K.K., Hayes, R.L. and metabolism of stretch-injured cultured rat astrocytes. J.
Pineda, J.A. (2005) Cell-specific DNA fragmentation may be Neurochem., 68(5): 1904–1910.
attenuated by a survivin-dependent mechanism after trau- Lenz, G., Gottfried, C., Luo, Z., Avruch, J., Rodnight, R., Nie,
matic brain injury in rats. Exp. Brain Res., 167: 17–26. W.J., Kang, Y. and Neary, J.T. (2000) P(2Y) purinoceptor
Kang, J., Jiang, L., Goldman, S.A. and Nedergaard, M. (1998) subtypes recruit different mek activators in astrocytes. Br. J.
Astrocyte-mediated potentiation of inhibitory synaptic trans- Pharmacol., 129: 927–936.
mission. Nat. Neurosci., 1: 683–692. Lin, J.H., Weigel, H., Cotrina, M.L., Liu, S., Bueno, E.,
Kanner, B.I. and Schuldiner, S. (1987) Mechanism of transport Hansen, A.J., Hansen, T.W., Goldman, S. and Nedergaard,
and storage of neurotransmitters. CRC Crit. Rev. Biochem., M. (1998) Gap-junction-mediated propagation and amplifi-
22: 1–38. cation of cell injury. Nat. Neurosci., 1: 494–500.
Katayama, Y., Becker, D.P., Tamura, T. and Hovda, D.A. Liu, D., Smith, C.L., Barone, F.C., Ellison, J.A., Lysko, P.G.,
(1990) Massive increases in extracellular potassium and the Li, K. and Simpson, I.A. (1999) Astrocytic demise precedes
indiscriminate release of glutamate following concussive delayed neuronal death in focal ischemic rat brain. Brain Res.
brain injury. J. Neurosurg., 73: 889–900. Mol. Brain Res., 68: 29–41.
Kawamata, T., Katayama, Y., Hovda, D.A., Yoshino, A. and Liu, Q.S., Xu, Q., Arcuino, G., Kang, J. and Nedergaard, M.
Becker, D.P. (1992) Administration of excitatory amino acid (2004a) Astrocyte-mediated activation of neuronal kainate
antagonists via microdialysis attenuates the increase in glu- receptors. Proc. Natl. Acad. Sci. U.S.A., 101: 3172–3177.
cose utilization seen following concussive brain injury. J. Liu, Q.S., Xu, Q., Kang, J. and Nedergaard, M. (2004b) As-
Cereb. Blood Flow Metab., 12: 12–24. trocyte activation of presynaptic metabotropic glutamate re-
Kawamata, T., Katayama, Y., Hovda, D.A., Yoshino, A. and ceptors modulates hippocampal inhibitory synaptic
Becker, D.P. (1995) Lactate accumulation following concus- transmission. Neuron Glia Biol., 1: 307–316.
sive brain injury: the role of ion fluxes induced by excitatory Lukaszevicz, A.C., Sampaio, N., Guegan, C., Benchoua, A.,
amino acids. Brain Res., 674: 196–204. Couriaud, C., Chevalier, E., Sola, B., Lacombe, P. and On-
Keane, R.W., Srinivasan, A., Foster, L.M., Testa, M.P., Ord, teniente, B. (2002) High sensitivity of protoplasmic cortical
T., Nonner, D., Wang, H.G., Reed, J.C., Bredesen, D.E. and astroglia to focal ischemia. J. Cereb. Blood Flow Metab., 22:
Kayalar, C. (1997) Activation of CPP32 during apoptosis of 289–298.
neurons and astrocytes. J. Neurosci. Res., 48: 168–180. Lyeth, B.G., Gong, Q.Z., Dhillon, H.S. and Prasad, M.R.
Kelleher, J.A., Chan, P.H., Chan, T.Y. and Gregory, G.A. (1996) Effects of muscarinic receptor antagonism on the
(1993) Modification of hypoxia-induced injury in cultured rat phosphatidylinositol bisphosphate signal transduction path-
astrocytes by high levels of glucose. Stroke, 24: 855–863. way after experimental brain injury. Brain Res., 742: 63–70.
77

Lyeth, B.G., Gong, Q.Z., Shields, S., Muizelaar, J.P. and competent and -deficient cell lines. J. Cell Biol., 111:
Berman, R.F. (2001) Group I metabotropic glutamate 2077–2088.
antagonist reduces acute neuronal degeneration and behavi- Nagy, J.I. and Li, W.E. (2000) A brain slice model for in vitro
oral deficits after traumatic brain injury in rats. Exp. Neurol., analyses of astrocytic gap junction and connexin43 regula-
169: 191–199. tion: actions of ischemia, glutamate and elevated potassium.
Lyeth, B.G., Jenkins, L.W., Hamm, R.J., Dixon, C.E., Phillips, Eur. J. Neurosci., 12: 4567–4572.
L.L., Clifton, G.L., Young, H.F. and Hayes, R.L. (1990) Namura, S., Zhu, J., Fink, K., Endres, M., Srinivasan, A.,
Prolonged memory impairment in the absence of hippocam- Tomaselli, K.J., Yuan, J. and Moskowitz, M.A. (1998) Ac-
pal cell death following traumatic brain injury in the rat. tivation and cleavage of caspase-3 in apoptosis induced by
Brain Res., 526: 249–258. experimental cerebral ischemia. J. Neurosci., 18: 3659–3668.
MacVicar, B.A., Hochman, D., Delay, M.J. and Weiss, S. Neary, J.T. and Kang, Y. (2006) P2 purinergic receptors signal
(1991) Modulation of intracellular Ca++ in cultured as- to glycogen synthase kinase-3beta in astrocytes. J. Neurosci.
trocytes by influx through voltage-activated Ca++ channels. Res., 84: 515–524.
Glia, 4: 448–455. Neary, J.T., Kang, Y., Tran, M. and Feld, J. (2005) Traumatic
Margulies, S.S., Thibault, L.E. and Gennarelli, T.A. (1990) injury activates protein kinase B/Akt in cultured astrocytes:
Physical model simulations of brain injury in the primate. J. role of extracellular ATP and P2 purinergic receptors. J. Ne-
Biomech., 23: 823–836. urotrauma, 22: 491–500.
Martinez, A.D. and Saez, J.C. (2000) Regulation of astrocyte Neary, J.T., Kang, Y., Willoughby, K.A. and Ellis, E.F. (2003)
gap junctions by hypoxia-reoxygenation. Brain Res. Brain Activation of extracellular signal-regulated kinase by stretch-
Res. Rev., 32: 250–258. induced injury in astrocytes involves extracellular ATP and
McGraw, J., Hiebert, G.W. and Steeves, J.D. (2001) Modulat- P2 purinergic receptors. J. Neurosci., 23: 2348–2356.
ing astrogliosis after neurotrauma. J. Neurosci. Res., 63: Neary, J.T., Rathbone, M.P., Cattabeni, F., Abbracchio, M.P.
109–115. and Burnstock, G. (1996) Trophic actions of extracellular
McIntosh, T.K., Juhler, M. and Wieloch, T. (1998) Novel nucleotides and nucleosides on glial and neuronal cells.
pharmacologic strategies in the treatment of experimental Trends Neurosci., 19: 13–18.
traumatic brain injury: 1998. J. Neurotrauma, 15: 731–769. Neary, J.T. and Zhu, Q. (1994) Signaling by ATP receptors in
McIntosh, T.K., Vink, R., Noble, L., Yamakami, I., Fernyak, astrocytes. Neuroreport, 5: 1617–1620.
S., Soares, H. and Faden, A.I. (1989) Traumatic brain injury Nedergaard, M., Goldman, S.A., Desai, S. and Pulsinelli, W.A.
in the rat: characterization of a lateral fluid-percussion (1991a) Acid-induced death in neurons and glia. J. Neurosci.,
model. Neuroscience, 28: 233–244. 11: 2489–2497.
McKinney, J.S., Willoughby, K.A., Liang, S. and Ellis, E.F. Nedergaard, M., Kraig, R.P., Tanabe, J. and Pulsinelli,
(1996) Stretch-induced injury of cultured neuronal, glial, and W.A. (1991b) Dynamics of interstitial and intracellular
endothelial cells: effect of polyethylene glycol-conjugated pH in evolving brain infarct. Am. J. Physiol., 260:
superoxide dismutase. Stroke, 27: 934–940. R581–R588.
Meaney, D.F., Smith, D.H., Shreiber, D.I., Bain, A.C., Miller, Nett, W.J., Oloff, S.H. and McCarthy, K.D. (2002) Hippo-
R.T., Ross, D.T. and Gennarelli, T.A. (1995) Biomechanical campal astrocytes in situ exhibit calcium oscillations that
analysis of experimental diffuse axonal injury. J. Neurotrau- occur independent of neuronal activity. J. Neurophysiol., 87:
ma, 12: 689–694. 528–537.
Mothet, J.P., Pollegioni, L., Ouanounou, G., Martineau, M., Newcomb, J.K., Zhao, X., Pike, B.R. and Hayes, R.L. (1999)
Fossier, P. and Baux, G. (2005) Glutamate receptor activa- Temporal profile of apoptotic-like changes in neurons and
tion triggers a calcium-dependent and SNARE protein-de- astrocytes following controlled cortical impact injury in the
pendent release of the gliotransmitter D-serine. Proc. Natl. rat. Exp. Neurol., 158: 76–88.
Acad. Sci. U.S.A., 102: 5606–5611. Newman, E.A. (1991) Sodium-bicarbonate cotransport in reti-
Mukhin, A., Fan, L. and Faden, A.I. (1996) Activation of me- nal Muller (glial) cells of the salamander. J. Neurosci., 11:
tabotropic glutamate receptor subtype mGluR1 contributes 3972–3983.
to post-traumatic neuronal injury. J. Neurosci., 16: Newman, E.A. (2003) Glial cell inhibition of neurons by release
6012–6020. of ATP. J. Neurosci., 23: 1659–1666.
Mukhin, A.G., Ivanova, S.A. and Faden, A.I. (1997) mGluR Newman, E.A. and Zahs, K.R. (1997) Calcium waves in retinal
modulation of post-traumatic neuronal death: role of glial cells. Science, 275: 844–847.
NMDA receptors. Neuroreport, 8: 2561–2566. Newman, E.A. and Zahs, K.R. (1998) Modulation of neuronal
Muller, H.W., Junghans, U. and Kappler, J. (1995) Astroglial activity by glial cells in the retina. J. Neurosci., 18:
neurotrophic and neurite-promoting factors. Pharmacol. 4022–4028.
Ther., 65: 1–18. Nicotera, P., Ankarcrona, M., Bonfoco, E., Orrenius, S. and
Musil, L.S., Cunningham, B.A., Edelman, G.M. and Good- Lipton, S.A. (1997) Neuronal necrosis and apoptosis: two
enough, D.A. (1990) Differential phosphorylation of the gap distinct events induced by exposure to glutamate or oxidative
junction protein connexin43 in junctional communication- stress. Adv. Neurol., 72: 95–101.
78

Nilsson, P., Hillered, L., Ponten, U. and Ungerstedt, U. (1990) Porter, J.T. and McCarthy, K.D. (1996) Hippocampal as-
Changes in cortical extracellular levels of energy-related trocytes in situ respond to glutamate released from synaptic
metabolites and amino acids following concussive brain in- terminals. J. Neurosci., 16: 5073–5081.
jury in rats. J. Cereb. Blood Flow Metab., 10: 631–637. Raichle, M.E. (1991) The metabolic requirements of functional
Nimmerjahn, A., Kirchhoff, F., Kerr, J.N. and Helmchen, F. activity in the human brain: a positron emission tomography
(2004) Sulforhodamine 101 as a specific marker of astroglia study. Adv. Exp. Med. Biol., 291: 1–4.
in the neocortex in vivo. Nat. Methods, 1: 31–37. Reier, P.J., Stensaas, L.J. and Guth, L. (1983) The astrocytic
Ohsumi, A., Nawashiro, H., Otani, N., Ooigawa, H., Toyooka, scar as an impediment to regeneration in the central nervous
T., Yano, A., Nomura, N. and Shima, K. (2006) Alteration system. In: Kao C.C., Bunge R.P. and Reier P.J. (Eds.),
of gap junction proteins (connexins) following lateral fluid Spinal Cord Reconstruction. Raven Press, New York,
percussion injury in rats. Acta Neurochir. Suppl., 96: pp. 163–198.
148–150. Ridet, J.L., Malhotra, S.K., Privat, A. and Gage, F.H. (1997)
Olney, J.W. and Ho, O.L. (1970) Brain damage in infant mice Reactive astrocytes: cellular and molecular cues to biological
following oral intake of glutamate, aspartate or cysteine. function. Trends Neurosci., 20: 570–577.
Nature, 227: 609–611. Ringel, F., Chang, R.C., Staub, F., Baethmann, A. and Ples-
Paemeleire, K. and Leybaert, L. (2000) Ionic changes accom- nila, N. (2000) Contribution of anion transporters to the ac-
panying astrocytic intercellular calcium waves triggered by idosis-induced swelling and intracellular acidification of glial
mechanical cell damaging stimulation. Brain Res., 857: cells. J. Neurochem., 75: 125–132.
235–245. Rink, A., Fung, K.M., Trojanowski, J.Q., Lee, V.M., Ne-
Panatier, A., Theodosis, D.T., Mothet, J.P., Touquet, B., Pol- ugebauer, E. and McIntosh, T.K. (1995) Evidence of apo-
legioni, L., Poulain, D.A. and Oliet, S.H. (2006) Glia-derived ptotic cell death after experimental traumatic brain injury in
D-serine controls NMDA receptor activity and synaptic the rat. Am. J. Pathol., 147: 1575–1583.
memory. Cell, 125: 775–784. Rouach, N., Avignone, E., Meme, W., Koulakoff, A., Venance,
Parpura, V., Basarsky, T.A., Liu, F., Jeftinija, K., Jeftinija, S. L., Blomstrand, F. and Giaume, C. (2002) Gap junctions and
and Haydon, P.G. (1994) Glutamate-mediated astrocyte- connexin expression in the normal and pathological central
neuron signalling. Nature, 369: 744–747. nervous system. Biol. Cell, 94: 457–475.
Parpura, V. and Haydon, P.G. (2000) Physiological astrocytic Rzigalinski, B.A., Weber, J.T., Willoughby, K.A. and Ellis,
calcium levels stimulate glutamate release to modulate adja- E.F. (1998) Intracellular free calcium dynamics in stretch-
cent neurons. Proc. Natl. Acad. Sci. U.S.A., 97: 8629–8634. injured astrocytes. J. Neurochem., 70: 2377–2385.
Parpura, V., Liu, F., Brethorst, S., Jeftinija, K., Jeftinija, S. and Schurr, A., Miller, J.J., Payne, R.S. and Rigor, B.M. (1999) An
Haydon, P.G. (1995) Alpha-latrotoxin stimulates glutamate increase in lactate output by brain tissue serves to meet the
release from cortical astrocytes in cell culture. FEBS Lett., energy needs of glutamate-activated neurons. J. Neurosci.,
360: 266–270. 19: 34–39.
Parri, H.R. and Crunelli, V. (2003) The role of Ca2+ in the Schwiebert, E.M. (2001) ATP release mechanisms, ATP recep-
generation of spontaneous astrocytic Ca2+ oscillations. Ne- tors and purinergic signalling along the nephron. Clin. Exp.
uroscience, 120: 979–992. Pharmacol. Physiol., 28: 340–350.
Parri, H.R., Gould, T.M. and Crunelli, V. (2001) Spontaneous Shao, Y. and McCarthy, K.D. (1995) Receptor-mediated cal-
astrocytic Ca2+ oscillations in situ drive NMDAR-mediated cium signals in astroglia: multiple receptors, common stores
neuronal excitation. Nat. Neurosci., 4: 803–812. and all-or-nothing responses. Cell Calcium, 17: 187–196.
Peakman, M.C. and Hill, S.J. (1995) Adenosine A1 receptor- Siesjo, B.K. (1988) Acidosis and ischemic brain damage. Ne-
mediated changes in basal and histamine-stimulated levels of urochem. Pathol., 9: 31–88.
intracellular calcium in primary rat astrocytes. Br. J. Pharma- Silver, I.A. and Erecinska, M. (1997) Energetic demands of the
col., 115: 801–810. Na+/K+ ATPase in mammalian astrocytes. Glia, 21: 35–45.
Phillis, J.W., Estevez, A.Y., Guyot, L.L. and O’Regan, M.H. Siushansian, R., Bechberger, J.F., Cechetto, D.F., Hachinski,
(1999) 5-(N-Ethyl-N-isopropyl)-amiloride, an Na(+)-H(+) V.C. and Naus, C.C. (2001) Connexin43 null mutation in-
exchange inhibitor, protects gerbil hippocampal neurons creases infarct size after stroke. J. Comp. Neurol., 440:
from ischemic injury. Brain Res., 839: 199–202. 387–394.
Phillis, J.W., O’Regan, M.H. and Song, D. (1998) 5-(N-Ethyl- Slemmer, J.E., Matser, E.J., De Zeeuw, C.I. and Weber, J.T.
N-isopropyl)-amiloride inhibits amino acid release from the (2002) Repeated mild injury causes cumulative damage to
ischemic rat cerebral cortex: role of Na+-H+ exchange. hippocampal cells. Brain, 125: 2699–2709.
Brain Res., 812: 297–300. Slemmer, J.E., Weber, J.T. and De Zeeuw, C.I. (2004) Cell
Plesnila, N., Haberstok, J., Peters, J., Kolbl, I., Baethmann, A. death, glial protein alterations and elevated S-100 beta release
and Staub, F. (1999) Effect of lactacidosis on cell volume and in cerebellar cell cultures following mechanically induced
intracellular pH of astrocytes. J. Neurotrauma, 16: 831–841. trauma. Neurobiol. Dis., 15: 563–572.
Plum, F. (1983) What causes infarction in ischemic brain?: the Smith, I.F., Plant, L.D., Boyle, J.P., Skinner, R.A., Pearson,
Robert Wartenberg lecture. Neurology, 33: 222–233. H.A. and Peers, C. (2003) Chronic hypoxia potentiates
79

capacitative Ca2+ entry in type-I cortical astrocytes. J. transduction after traumatic injury of cortical neurons. Cell
Neurochem., 85: 1109–1116. Calcium, 26: 289–299.
Sochocka, E., Juurlink, B.H., Code, W.E., Hertz, V., Peng, L. Weisman, G.A., Wang, M., Kong, Q., Chorna, N.E., Neary,
and Hertz, L. (1994) Cell death in primary cultures of mouse J.T., Sun, G.Y., Gonzalez, F.A., Seye, C.I. and Erb, L. (2005)
neurons and astrocytes during exposure to and ‘recovery’ Molecular determinants of P2Y2 nucleotide receptor func-
from hypoxia, substrate deprivation and simulated ischemia. tion: implications for proliferative and inflammatory path-
Brain Res., 638: 21–28. ways in astrocytes. Mol. Neurobiol., 31: 169–183.
Staub, F., Baethmann, A., Peters, J., Weigt, H. and Kempski, Willoughby, K.A., Kleindienst, A., Muller, C., Chen, T., Muir,
O. (1990) Effects of lactacidosis on glial cell volume and J.K. and Ellis, E.F. (2004) S100B protein is released by in
viability. J. Cereb. Blood Flow Metab., 10: 866–876. vitro trauma and reduces delayed neuronal injury. J. Ne-
Stout, C.E., Costantin, J.L., Naus, C.C. and Charles, A.C. urochem., 91: 1284–1291.
(2002) Intercellular calcium signaling in astrocytes via ATP Wilsch, V.W., Pidoplichko, V.I., Opitz, T., Shinozaki, H. and
release through connexin hemichannels. J. Biol. Chem., 277: Reymann, K.G. (1994) Metabotropic glutamate recep-
10482–10488. tor agonist DCG-IV as NMDA receptor agonist in imma-
Swanson, R.A., Farrell, K. and Stein, B.A. (1997) Astrocyte ture rat hippocampal neurons. Eur. J. Pharmacol., 262:
energetics, function, and death under conditions of incom- 287–291.
plete ischemia: a mechanism of glial death in the penumbra. Yamamoto, T., Hertzberg, E.L. and Nagy, J.I. (1990a)
Glia, 21: 142–153. Epitopes of gap junctional proteins localized to neuronal
Takahashi, M., Billups, B., Rossi, D., Sarantis, M., Hamann, M. subsurface cisterns. Brain Res., 527: 135–139.
and Attwell, D. (1997) The role of glutamate transporters in Yamamoto, T., Ochalski, A., Hertzberg, E.L. and Nagy, J.I.
glutamate homeostasis in the brain. J. Exp. Biol., 200: 401–409. (1990b) On the organization of astrocytic gap junctions in rat
Takuma, K., Baba, A. and Matsuda, T. (2004) Astrocyte brain as suggested by LM and EM immunohistochemistry of
apoptosis: implications for neuroprotection. Prog. Ne- connexin43 expression. J. Comp. Neurol., 302: 853–883.
urobiol., 72: 111–127. Yu, A.C., Gregory, G.A. and Chan, P.H. (1989) Hypoxia-in-
Tanaka, J., Ichikawa, R., Watanabe, M., Tanaka, K. and In- duced dysfunctions and injury of astrocytes in primary cell
oue, Y. (1997) Extra-junctional localization of gluta- cultures. J. Cereb. Blood Flow Metab., 9: 20–28.
mate transporter EAAT4 at excitatory Purkinje cell synapses. Yu, A.C., Wong, H.K., Yung, H.W. and Lau, L.T. (2001)
Neuroreport, 8: 2461–2464. Ischemia-induced apoptosis in primary cultures of astrocytes.
Tavalin, S.J., Ellis, E.F. and Satin, L.S. (1995) Mechanical Glia, 35: 121–130.
perturbation of cultured cortical neurons reveals a stretch- Zauner, A., Bullock, R., Kuta, A.J., Woodward, J. and Young,
induced delayed depolarization. J. Neurophysiol., 74(6): H.F. (1996) Glutamate release and cerebral blood flow
2767–2773. after severe human head injury. Acta Neurochir. Suppl., 67:
Tran, M.D. and Neary, J.T. (2006) Purinergic signaling induces 40–44.
thrombospondin-1 expression in astrocytes. Proc. Natl. Zhang, L., Rzigalinski, B.A., Ellis, E.F. and Satin, L.S. (1996)
Acad. Sci. U.S.A., 103: 9321–9326. Reduction of voltage-dependent Mg2+ blockade of NMDA
Tsuchida, T., Kato, T., Yamada, A. and Kawamoto, K. (2002) current in mechanically injured neurons. Science, 274(5294):
Cycloheximide induces apoptosis of astrocytes. Pathol. Int., 1921–1923.
52: 181–185. Zhao, X., Ahram, A., Berman, R.F., Muizelaar, J.P. and Lyeth,
VanSlyke, J.K. and Musil, L.S. (2005) Cytosolic stress reduces B.G. (2003) Early loss of astrocytes after experimental trau-
degradation of connexin43 internalized from the cell surface matic brain injury. Glia, 44: 140–152.
and enhances gap junction formation and function. Mol. Zhong, C., Zhao, X., Van, K.C., Bzdega, T., Smyth, A., Zhou,
Biol. Cell, 16: 5247–5257. J., Kozikowski, A.P., Jiang, J., O’Connor, W.T., Berman,
Verkhratsky, A., Orkand, R.K. and Kettenmann, H. (1998) R.F., Neale, J.H. and Lyeth, B.G. (2006) NAAG peptidase
Glial calcium: homeostasis and signaling function. Physiol. inhibitor increases dialysate NAAG and reduces glutamate,
Rev., 78: 99–141. aspartate and GABA levels in the dorsal hippocampus fol-
Vernadakis, A. (1996) Glia-neuron intercommunications and lowing fluid percussion injury in the rat. J. Neurochem., 97:
synaptic plasticity. Prog. Neurobiol., 49: 185–214. 1015–1025.
Vesce, S., Bezzi, P. and Volterra, A. (1999) The active role of Zvalova, D., Cordier, J., Mesnil, M., Junier, M.P. and Chneiw-
astrocytes in synaptic transmission. Cell Mol. Life Sci., 56: eiss, H. (2004) p38/SAPK2 controls gap junction closure in
991–1000. astrocytes. Glia, 46: 323–333.
Volterra, A. and Meldolesi, J. (2005) Astrocytes, from brain Zwienenberg, M., Gong, Q.Z., Berman, R.F., Muizelaar, J.P.
glue to communication elements: the revolution continues. and Lyeth, B.G. (2001) The effect of groups II and III
Nat. Rev. Neurosci., 6: 626–640. metabotropic glutamate receptor activation on neuronal in-
Weber, J.T., Rzigalinski, B.A., Willoughby, K.A., Moore, S.F. jury in a rodent model of traumatic brain injury. Neurosur-
and Ellis, E.F. (1999) Alterations in calcium-mediated signal gery, 48: 1119–1127.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 6

Rescuing neurons and glia: is inhibition of apoptosis


useful?

E. Kovesdi1, E. Czeiter1, A. Tamas2, D. Reglodi2, D. Szellar1, J. Pal3, P. Bukovics1,3,


T. Doczi1 and A. Buki1,

1
Department of Neurosurgery, University Medical School, Pécs University, Pécs, Hungary, Rét u. 2. H-7624, Hungary
2
Department of Anatomy, University Medical School, Pécs University, Pécs, Hungary, Szigeti u. 12. H-7624, Hungary
3
Neurobiology Research Group of the Hungarian Academy of Sciences, University Medical School, Pécs University, Pécs,
Hungary, Rét u. 2. H-7624, Hungary

Abstract: Traumatic brain injury (TBI) represents a leading cause of death in western countries. Despite all
research efforts we still lack any pharmacological agent that could effectively be utilized in the clinical
treatment of TBI. Detailed unraveling of the pathobiological processes initiated by/operant in TBI is a
prerequisite to the development of rational therapeutic interventions. In this review we provide a summary
of those therapeutic interventions purported to inhibit the cell death (CD) cascades ignited in TBI. On
noxious stimuli three major forms of CD, apoptosis, autophagia and necrosis may occur. Apoptosis can be
induced either via the mitochondrial (intrinsic) or the receptor mediated (extrinsic) pathway; endoplasmic
reticular stress is the third trigger of caspase-mediated apoptotic processes. Although, theoretically pan-
caspase inhibition could be an efficient tool to limit apoptosis and thereby the extent of TBI, potential
cross-talk between various avenues of CD suggests that more upstream events, particularly the preservation
of the cellular energy homeostasis (cyclosporine-A, poly ADP ribose polymerase (PARP) inhibition, hy-
pothermia treatment) may represent more efficient therapeutic targets hopefully also translated to the
clinical care of the severely head injured.

Keywords: cell death; caspases; calpain; traumatic brain injury; traumatically induced axonal injury;
necrosis; apoptosis

Introduction per year for TBI is $60 billion. The TBI-related


death rate of the population under 35 years of age
Traumatic brain injury (TBI) puts an extreme is 3.5 times that of cancer and heart disease to-
burden on societies worldwide. More than five gether (Lewin, 1991; Thurman et al., 1999). De-
million Americans have a TBI-related long-term or spite of the social and economical significance of
lifelong need for help for daily activities. Long- TBI we still lack any efficient specific pharmaco-
term consequences include functional changes logical approach to interfere with the damaging
such as cognitive deficit, epilepsy, hypopytuitar- consequence of injury (Narayan et al., 2002). The
ism and increased risk for Alzheimer’s disease and therapeutic approaches utilized to date are basi-
Parkinson’s disease. The cumulative societal cost cally not different from those of the late seventies
to early eighties; a noteworthy reduction in mor-
Corresponding author. Tel.: +36-30-411-3349; Fax: +36-72- tality was only achieved via the implementation of
535931; E-mail: andras.buki@aok.pte.hu scientific evidence based therapeutic guidelines.

DOI: 10.1016/S0079-6123(06)61006-6 81
82

While such reorganization of the system and eve- processes are also associated with energy deple-
ryday practice of TBI care led to marked reduction tion/negative energetic state of the cell (Cai et al.,
in mortality and morbidity, the clinical care for the 1998; Arden and Betenbaugh, 2004). Recent ob-
head injured has reached its limits (Vukic et al., servations proved that both in focal and diffuse
1999; Ghajar, 2000). Thereby, now basic scientists forms of experimental TBI in injured soma as well
and/or pharmaceutical research should provide as in axons, various proteolytic processes are trig-
further ammunition for clinicians to fight the silent gered and are responsible for necrotic demise
epidemic represented by TBI. The importance of (Bartus, 1997; Yamashima, 2004; Farkas et al.,
translational studies in TBI is further highlighted 2006). These studies demonstrated that mechanic
by data from the field of health care management, or enzymatic alterations of membrane integrity in
indicating that effective treatment of TBI is con- conjunction with altered function of ionic pumps
sidered one of the most cost-efficient medical inter- and channels should lead to intracellular calcium
ventions (Saltman and Findling, 1997). accumulation which in turn results in the activa-
Although detailed analysis of the pathobiolog- tion of neutral proteases, primarily the cysteine-
ical processes activated in TBI are beyond the aspartate protease calpain (Bartus, 1997; Stys and
scope of this work, the authors will provide a short Jiang, 2002). Besides direct proteolysis associated
theoretical background to each section addressing with calpain activation, indirect proteolytic conse-
various approaches to rescue neurons and glia in quences such as calpain-mediated lysosomal rup-
TBI, while also referring to other chapters of this ture and cathepsin activation (Yamashima, 2004)
work for further relevant information. also contribute to cellular demise.
Although the association and feedback connec-
tion between permeability changes, activation of
Cell death routes proteolytic processes, cellular demise and func-
tional alterations following TBI is not entirely de-
On noxious influences or agents three major forms veloped, preliminary data suggest that inhibition
of cell death (CD) may occur: autophagic, necrotic of necrotic enzymes may facilitate functional re-
and apoptotic. covery (Saatman et al., 1996).
Autophagy is characterized by lysosomal se- Regardless of the initiating factors and the route
questration and degradation of cellular compo- leading to altered intracellular ionic homeostasis,
nents. This mode of cell death has its potential role increased level of intracellular Ca++ represents a
in physiological processes as well as in neurode- permissive environment for the activation of an-
generative disorders (for review, see Assuncao and other form of CD that is apoptosis (Cai et al., 1998).
Linden, 2004). Apoptosis, frequently referred to as programmed
While primary brain injury leads to focal and cell death plays a pivotal role in embryogenesis
diffuse alterations in brain parenchyma, which are and is frequently initiated by a non-lethal stim-
predominantly necrotic by nature, secondary brain ulus leading to cell death via the activation of
damage practically triggered and initiated at the proteolytic cascades. Characteristic features of
moment of TBI is also associated with apopto- apoptosis include DNA laddering (detectable by
tic damage. In fact, as time elapses post-injury, electrophoresis), DNA fragmentation (TUNEL/
apoptotic processes become more dominant and in terminal deoxynucleotidyl-transferase-mediated de-
late phases post-injury autophagy may also partici- oxyuridine triphosphate-biotin nick-end labeling),
pate particularly in axonal demise (Lockshin and sub-2N DNA (FACS), rounding/blebbing of the
Zakeri, 2004; Ringger et al., 2004; Gallyas et al., cell, fragmentation of nuclei with marginalization
2006). of chromatin, exteriorization of phosphatidylserine
Necrosis is characterized with swelling and rup- (annexin V binding) (for review, see Arden and
ture of the cell (or its axon); membrane damage Betenbaugh, 2004; Lockshin and Zakeri, 2004).
and activation of inflammatory reactions is a con- As apoptosis requires a relatively intact energetic
sistent feature of necrotic cell death. Such necrotic state of the cell, most probably it will not be
83

localized to the core of tissue laceration (focal dam- Knoblach et al. (2002), in the model of lateral fluid
age) rather it will involve remote/diffuse (‘‘penum- percussion TBI in accordance with other, previous
bra’’) area of the central nervous system (CNS). In reports (Yakovlev et al., 1997; Beer et al., 2000;
the last decade multiple lines of evidences pointed to Clark et al., 2000), demonstrated caspase-3 and -9
the activation of apoptotic processes in various activation in the cerebral cortex and the hippo-
forms of TBI (Rink et al., 1995; Conti et al., 1998; campus 1–12 h post-injury. Although these workers
Buki et al., 2000; Ginis et al., 2000). were not able to demonstrate considerable activa-
Studies in more severe, focal forms of TBI have tion of caspase-8, in other models including cortical
proven cytochrome c (cyto c) release and caspase-3 impact TBI caspase-8 (representing the induction
activation in the cerebral cortex 6–24 h post-injury of the extrinsic route of apoptosis) was also found
(Sullivan et al., 2002). Caspase-12 activation was activated in both neurons and glia (Beer et al.,
also demonstrated in cortex, hippocampus and 2001).
cortical astrocytes in this model of TBI implicating
endoplasmic reticulum-associated apoptotic proc-
esses (vide infra) in the pathogenesis of cell loss Apoptotic pathways
(Larner et al., 2004) These authors also demon-
strated capsase-7 activation in the same model Apoptosis could be initiated via three major path-
peaking at day five post-injury (Larner et al., 2005). ways (Fig. 1), and in most of these pathways

Fig. 1. The three major routes of apoptosis. Note that different caspases participate at initiation and the execution of the cell death
processes. Also note the crucial role of calcium and the mitochondrion (for further details, see text).
84

intracellular Ca++, mitochondria and the caspase benzodiazepine receptor, hexokinase, creatine kin-
enzyme family play a crucial role. The extrinsic ase and Bcl-2 proteins (Susin et al., 1999) (some of
receptor-mediated route utilizes specific cell sur- these Bcl-2 proteins harbor anti-apoptotic poten-
face receptors belonging to the tumor necrosis tial, while others possess pro-apoptotic activity
factor (TNF) super family where binding of the (Reed, 1998; Scorrano and Korsmeyer, 2003)).
‘‘death signal’’ (such as Fas by Fas ligand) triggers Opening of the MPTP leads to the release of pro-
the activation of the caspase enzyme cascade apoptotic substances — cyto c, second mito-
(Chinnaiyan et al., 1995). Caspases are cysteine- chondrial activator of caspases (SMAC)/direct
aspartate proteases which are present in the cell in inhibitor of apoptosis protein (IAP) binding pro-
the inactive proenzyme (zymogen) form. Specifi- tein with low pI (DIABLO) and apoptosis induc-
cally, ligand–receptor interaction in the extrinsic ing factor (AIF). When released, cyto c together
route will activate pro-caspase-8, which, in turn with the apoptosis activating factor 1 (Apaf-1),
activates pro-caspase-3. dATP and cytosolic pro-caspase-9 forms the ‘‘apo-
Similarly to this pathway, the final executor of ptosome’’ resulting in the generation of active ca-
the second, intrinsic pathway is also caspase-3, spase-9 which, in turn triggers the transition of
however, this route is mediated by mitochondrial pro-caspase-3 to active caspase-3 (Cai et al., 1998;
alterations (Cai et al., 1998; Kruman and Mattson, Adams and Cory, 2002).
1999; Adams and Cory, 2002; Kroemer, 2003). The third, most recently described route of
Mitochondria play a key role in apoptosis. Seve- apoptosis is initiated by endoplasmatic reticulum
ral studies demonstrated both in vitro and in vivo (ER)-stress leading to caspase-12 activation. This
that TBI leads to a perturbation of the energy latter enzyme can act either as an executor caspase
homeostasis (Giza and Hovda, 2001). Partially due or via direct or indirect activation of caspase-3
to excessive firing of neurons and/or ionic imbal- (Oyadomari et al., 2002; Zong et al., 2003).
ance due to altered pump/channel function as well
as mechanoporation in at least a subpopulation of
cell bodies and axons, a clear metabolic dysfunc- Crossroads of cell death
tion occurs, leading to a shift toward anaerobic
glycolysis, lactate-production and the accumula- To develop rationally targeted therapeutic strate-
tion of reactive oxygen species (ROS) (Pettus et al., gies beyond understanding these basic mechanisms
1994; Stys and Jiang, 2002; Singleton and Povli- of cell death, it is also mandatory to unravel those
shock, 2004; Farkas et al., 2006). N-methyl-D- potential routes via the pathways initiated by noxi-
aspartate (NMDA) receptor activation in neurons, ous stimuli may communicate or interact.
failure of the Na-K-ATPase in axons and me- Such connections or ‘‘detours’’ exist within the
chanoporation in some soma and axons in con- apoptotic machinery itself as well as between var-
junction with altered energy homeostasis leads to ious major pathways of cell death that is necrosis,
the accumulation of intracellular (intraaxonal) apoptosis and autophagy (Denecker et al., 2001;
Ca++ (Siesjo et al., 1999; Stys and Jiang, 2002). Lockshin and Zakeri, 2004). In addition to their
Mitochondria accumulate the excess amount role in the maintenance of the ionic and energy
of Ca++ leading to swelling and dissipation of homeostasis and triggering the intrinsic pathway
their membrane potential, initiating the perm- of apoptosis, mitochondria also serve as a cross-
ealization of their outer membrane for solutes road or common pathway for all routes of apo-
under 1500 Da, widely explained by the opening ptotic cell death (Cai and Jones, 1998; Saikumar
of the mitochondrial permeability transition et al., 1998) (Fig. 1).
pore (MPTP) (Zoratti and Szabo, 1995; Susin To this end, besides pro-apoptotic members of
et al., 1998; Kruman and Mattson, 1999). This the Bcl-2 family the mitochondrial membrane also
purported pore is consisting of the adenine nuc- contains other pro-apoptotic proteins such as Bax
leotide translocase (ANT), voltage dependent an- and Bak. These are responsible for the cross-talk
ion channel (VDAC), cyclophilin D, peripheral between mitochondria (that is the intrinsic
85

pathway of apoptosis) and the other two avenues When we consider, how to inhibit apoptosis, we
of apoptotic cell death; the extrinsic route is con- also have to answer, whether inhibition of
verging to these pro-apoptotic proteins via the ca- apoptosis is useful. Theoretically, in a philosophi-
spase-8 Bid–Bax/Bak interaction while the cal approach, we may conclude that a lethal or
endoplasmatic reticulum stress also interacts with even non-lethal stimulus may ignite any of the
these proteins as well as via sending direct Ca++ above-detailed pathways. Due to the cross-talk
signals to the mitochondria (Jurgensmeier et al., between these avenues and the internal diversity of
1998; Schendel et al., 1998; Ferri and Kroemer, the individual pathways it is quite possible that
2001). pharmacological intervention to halt a death proc-
Not only the major intracellular ‘‘death-ave- ess may just shift the cell death mechanism to an-
nues’’ but also the redundancy of the effector ca- other pathway of cellular demise. Inhibition of
spases provide considerable versatility for cell apoptotic cell death can lead to autophagy instead
death; studies with knock-out mice as well as ca- of resulting in cell survival (Lang-Rollin et al.,
spase-inhibitor studies have proven that an injured 2003). Similarly, although the application of ca-
cell has the potential to select between various ef- spase inhibitors is a popular way to interfere with
fector caspases and such decision is most probably apoptosis, more upstream targets should be advo-
influenced by various, physiologically active in- cated for pharmacological intervention. Specifi-
hibitors of apoptosis (Assuncao and Linden, cally, as caspase-9 and -3 represent the post-
2004). mitochondrial phase of cell death their activation
Alternative, protease-mediated pathways may requires the release of cyto c and other pro-apo-
also contribute to cell death. Ubiquitination of ptotic substances. Such excessive mitochondrial
some caspases and their subsequent destruction by damage also points to a considerable disruption of
the proteosome may participate in the regulation the electron transport chain leading to mass gene-
of apoptosis (Lockshin and Zakeri, 2002; Ditzel ration of free radicals as well as to altered energy
et al., 2003; Varshavsky, 2003). Recent observa- homeostasis both facilitating the activation of ne-
tions have proven that disconnection of the cell crotic processes in the cell. In traumatic axonal
from its neighboring structures specifically from injury (TAI) the activation of caspases leads to
the extracellular matrix should contribute to the irreversible digestion of the membrane skeleton
initiation of cell death mechanisms. This process, and other cytoskeletal structures indicative of im-
primarily mediated by matrix metalloproteases is minent, irreversible axonal demise (Wang et al.,
frequently described with the Greek word ‘‘an- 1998; Buki et al., 2000) (Fig. 2). The relevance of
oikis’’ (homelessness) (Lockshin and Zakeri, 2002; such co-activation of the ‘‘necrotic’’ and ‘‘apopto-
Abe et al., 2004). tic’’ machinery was also demonstrated in an ele-
The above-detailed complexity of the cell death gant study by Liu et al. (2006) in hippocampal
machinery provides multiple choices for a cell to lysates from rats which have undergone controlled
react to a noxious stimulus. Apoptotic cell death cortical impact injury, where degradome-studies
requires energy and phagocytes that scavenge the identified various common targets of calpain-2 and
dying cells. In cell cultures, in vitro in the absence caspase-3 including beta-II-spectrin, synaptotag-
of phagocytes the final mechanisms of cell death min-1 and striatin. Similar ‘‘collaboration’’ bet-
are always necrotic, despite of the initiating agent ween calpain and caspase-3 was also demonstrated
and the route the cell entered to die (Arden and by Warren et al. (2005) in metamphetamine
Betenbaugh, 2004; Assuncao and Linden, 2004). induced experimental neurotoxicity.
Limited sources of ATP can easily halt the apo-
ptotic processes and the cell may fail to display
apoptotic morphology, however, this does not Inhibition of apoptosis
mean an arrest of cell death rather the initiation of
autophagic or necrotic processes (Cai et al., 1998; Several studies with caspase-inhibitors corroborate
Reed, 1998; Saikumar et al., 1998). — at least from some aspects — the above-described
86

(2004) were able to prove significant improvement


of motor and neurological function using the
pan-caspase inhibitor z-VAD-fmk, they also stress
that this beneficial effect was purportedly also
attributable to the inhibition of calpain that is
co-inhibition of necrosis in the dose they applied.
In their review Lockshin and Zakeri (2004) also
stress that co-inhibition of lysosomal proteases
may lead to overestimation of the efficacy of
caspase-inhibition.
Keeping these warnings in mind one should
conclude that inhibition of apoptosis alone might
be ‘‘too little, too late’’. Thus, the most promising
way to interfere with the progression of the patho-
biological processes evoked by/operant in TBI
must either be the application of therapeutic
agents with multiple targets like the above men-
tioned pan-caspase inhibitors or targeting pre-
mitochondrial/mitochondrial events in the course
of cellular demise.
As far as multitargetic therapeutic approaches
are considered, sex steroids should be included to
our list; several studies suggested that steroids
could have beneficial effects both on neurons and
glia. In ischemic and TBI sex steroids have been
Fig. 2. Images of damaged axonal profiles from the medial le-
mniscus of rat exposed to impact acceleration brain injury proven to inhibit necrotic as well as apoptotic cell
(60 min post-injury). Cytochrome c release from damaged mito- death (Roof and Hall, 2000). Although the exact
chondria (immunofluorescent label on A) and caspase-3 acti- mechanism of their action still requires further in-
vation (immunofluorescent label on B) are co-localised in the vestigation, sex steroids are thought to inhibit lipid
same damaged axons (digital overlay on C) pointing to the
peroxydation and the formation of ROS while also
contribution of calcium-induced mitochondrial damage and
caspase activation to the pathogenesis of traumatically induced exerting anti-apoptotic properties facilitating the
diffuse brain injury (axonal injury). expression of anti-apoptotic proteins including
bcl-2 and Bax (Soustiel et al., 2005). Similarly, est-
rogen treatment has a positive effect on oligoden-
critical attitude to the usefulness of inhibiting the droglia function (Curry and Heim, 1966) and
effector-phase of apoptosis alone. Abrahamson astrocytic scar formation (Garcia-Estrada et al.,
et al. (2006) demonstrated that a pan-caspase in- 1999). Testosterone can influence the expression of
hibitor (BAF) significantly reduced the formation the astrocytic water channel protein aquaporin-4
of caspase-cleaved amyloid precursor protein frag- indicating that sex steroids may also exert their
ments also leading to a reduced volume of hippo- action on the formation of brain edema — a major
campal cell loss, however, in their experiment player in secondary brain injury (Gu et al., 2003).
performed in mice applying the cortical impact In the last two decades several studies suggested
model of injury significant necrotic areas where that poly(ADP-ribose)polymerase-1 (PARP-1, EC
still noted in the cortex and the hippocam- 2.4.2.30) inhibition might be beneficial in the in-
pus. In another work, the pan-caspase inhibitor hibition of CNS injury of various origin (Burkle,
FK-011 was not able to achieve reduction in 2001; Besson et al., 2003, 2005). This enzyme was
cortical lesion size measured 7 days post-injury originally associated with DNA-repair function:
(Sullivan et al., 2002). Although Knoblach et al. stress induced strand DNA breaks activate PARP
87

that will transfer ADP-ribose units to nuclear pro- 1998). While classic thought appreciated calpain as
teins from NAD. This energy consuming process an agent contributing to necrotic processes several
was recognized to lead to NAD depletion and loss lines of evidences demonstrated that calpain can
of ATP leading to a collapse in cellular energy participate in the induction of the mitochondrial
homeostasis and cell death (Burkle, 2001). While (intrinsic) pathway of apoptosis. This communi-
long-term inhibition of PARP could theoretically cation between the classic, Ca++-induced necrotic
lead to mutagenesis and carcinogenesis, in the processes and the apoptotic enzyme system is par-
acute phase such intervention would be beneficial tially executed that way that calcium accumulation
in terms of restoring/preserving the cellular energy leads to axolemmal permeability alteration
pool. As it has recently been stressed by Komjati et through proteolytic modification of the subaxo-
al. (2005) PARP inhibition is not only interfering lemmal network (cortical cytoskeleton) which in
with ROS-generated necrosis. Beyond representing turn at least in some neurons and in axons con-
a potential pathway in initiating and/or modulat- tributes to further membrane-permeability
ing necrosis via AIF, PARP has a substantial role changes (Buki et al., 1999b). As it has been de-
in the induction of apoptosis. Via the NF-kappaB- scribed in the above passages, excessive influx of
pathway PARP is also capable of influencing the Ca++ induces mitochondrial damage via MPTP-
expression of inflammatory cytokines and media- opening. In addition to this pathway, calpain itself
tors (Komjati et al., 2005). is capable — as it has been demonstrated in liver
In the last few years several PARP inhibitors — to open the MPTP via its direct proteolytic
have been proven to inhibit TBI. In the lateral modification (Aguilar et al., 1996). Once MPTP is
fluid percussion head injury model LaPlaca et al. open and the intrinsic route, thus caspase-3 is
(2001) have demonstrated that the PARP inhibitor activated, it can cleave calpastatin, the major
GPI 1650 significantly reduced the lesion volume inhibitor of calpain, contributing to further un-
while not influencing the number of TUNEL posi- controlled activation of calpain. Taking into con-
tive cells. sideration such interactions and the temporal/
Other experiments proved that PARP inhibitors causative relationship between calpain and caspase
were able to improve the neurological score of in- activation a logical way to influence the release
jured animals without reducing the extent of the of apoptotic enzymes should be the inhibition of
cellular lesion (Besson et al., 2005). These data in calcium-mediated proteolytic alterations that is
conjunction with those results indicating that the inhibition of calpain.
PARP inhibitors reduce the extent of TAI as- Recent observations indicated that systemic ad-
sessed by beta amyloid precursor protein immuno- ministration of calpain inhibitors reduced the
reactivity implicate the inhibition of TAI in the overall degree of the cerebral ischemia (Bartus,
neuroprotective effects of PARP inhibitors. 1997). Further, Saatman et al. (1996) demon-
Fluid percussion TBI is not the only model strated that the use of calpain inhibitors resulted in
where PARP inhibitors proved their neuroprotec- improved behavioral outcome in a contusional
tive effect: 3-aminobenzanide significantly reduced model of TBI. Intriguingly enough, in an extension
the lesion size in cold injury (Hortobagyi et al., of this work Saatman et al. failed to detect any
2003) and proved beneficial functional effects significant change in the size of contusion, con-
(PJ34) as well as improved effectiveness of neural cluding that the beneficial therapeutic effects of
stem cell transportation (Lacza et al., 2003). AK-295 could be explained by inhibiting TAI in
In the last decades, several studies implicated the remote areas (Saatman et al., 2000). The correct-
cytosolic neutral cysteine protease calpain (EC ness of this assumption was further supported by
3.4.22.17) in the pathogenesis of both diffuse and the finding that in a rodent model of TAI the cell
focal TBI (Bartus, 1997; Kampfl et al., 1997; Buki permeable peptidyl-aldehyde calpain inhibitor
et al., 1999b). This Ca++-induced enzyme is ca- MDL-28170 (carbobenzylzoxy-Val-Phe-H) proved
pable of reversibly cleaving spectrin and other to prevent axonal injury in the brainstem fiber
constituents of the cytoskeleton (Wang et al., tracts (Buki et al., 2003). Most recent observations
88

indicate that this agent is also potent in terms of some aspects of functional outcome following TBI
reducing the length of axonal segments displaying (Riess et al., 2001). Preliminary data regarding the
traumatically induced axolemmal permeability use of CsA in head injury in man indicate that this
changes frequently associated with the activation compound is safe and potentially beneficial (Alves
of the Ca++-induced protease calpain coupled in et al., 2003).
some axons to the initial, strain-induced axolem- Another immunophilin ligand FK506, a sub-
mal mechanoporation (Büki et al., unpublished) stance that has no known effect on MPT and ex-
(vide supra). Although physical and chemical erts its action via inhibition of calcineurin activity
properties of MDL-28170 do not favor its appli- (Liu et al., 1991; Kuroda et al., 1999) has also
cation in the clinical care, the family of such cell- displayed beneficial effects on TAI in a pre-injury
permeable calpain-inhibitors that readily cross administration paradigm (Singleton et al., 2001)
the blood brain barrier and exert their beneficial and reduced the complications associated with
effects in a relatively wide therapeutic window rapid re-warming following hypothermic interven-
(Bartus, 1997; Markgraf et al., 1998) should be tion for TAI (Suehiro et al., 2001).
considered candidates for further pharmacological Inhibition of the Ca++-induced phosphatase
modifications and studies. calcineurin is supposed to be beneficial in axonal
Calpain inhibitors are of particular interest as injury by preventing dephosphorylization of the
recent reports demonstrated that besides decreas- neurofilament side arms that should decrease the
ing the formation of cleavage products of this repelling forces between the neurofilaments
cysteine protease primarily participating in necro- thereby permitting NFC leading to impairment
tic processes, they are capable of inhibiting the of axoplasmatic transport (Povlishock et al., 1997;
formation of caspase-cleaved byproducts, thereby Okonkwo et al., 1998). Such dephosphorylated
apoptosis too (Kawamura et al., 2005). neurofilaments would be more susceptible for pro-
In the line of therapeutic approaches aiming to teolytic degradation by calpain and caspase (Pant,
inhibit the mitochondrial/premitochondrial phase 1988). It is of note that calcineurine might also
of apoptosis one of the most promising interven- participate in the signaling pathways regulating
tions is the use of the inhibitors of mPTP opening. proliferative responses of astrocytes and its inhi-
Among these interventions the immunophilin lig- bition by CsA as well as FK-506 diminishes as-
and cyclosporine A (CsA) has been used in a set of trocytic reactions (Pyrzynska et al., 2001).
experiments targeting TAI in the rodent impact Calcineurin also contributes to the control of
acceleration model of TBI. First, immuno-electron synaptic activity by inactivating dynamin I and
microscopic studies by Okonkwo and Povlishock synapsin I, proteins participating in neurotrans-
(1999) demonstrated that CsA significantly atten- mitter release (Cousin and Robinson, 2001) and by
uated mitochondrial swelling and rupture, trans- dephosporylation-mediated induction of nitric ox-
lating into reduced numbers of damaged, ide synthase leading to increased nitric oxide level
disconnected axonal segments displaying immonu- and neurotransmitter release (Dawson et al.,
reactivity for the transport protein beta amyloid 1993).
precursor protein. Subsequent studies also proved Other important aspects of TBI might also be
that both pre- and post-injury administration influenced by immunophyllin ligands. Specifically,
of CsA significantly reduced the extent of TAI CsA inhibited ischemia-induced edema in vitro,
detected by immunohistochemical markers of suggesting that such a beneficial effect should also
calpain-mediated spectrin proteolysis, axonal be worth investigating in secondary brain injury
disconnection and cytoskeletal alterations — ne- where hypoperfusion-hypoxya-induced edema and
urofilament compaction (NFC). Dose response tissue damage is an issue, too (MacGregor et al.,
curves for the beneficial effects of CsA in the 2003).
experimental model of TBI have also been estab- CsA also proved neuroprotective in the hands of
lished (Okonkwo et al., 2003). More recent stud- Gabbita et al. (2005) in the controlled cortical im-
ies also demonstrated that CsA limits/improves pact model where it inhibited the TBI-induced
89

increase of cleaved tau (cytoskeletal protein) level studies indicate that PACAP is a promising ther-
in the ipsilateral hippocampus and its accumula- apeutic agent in traumatic brain and spinal cord
tion in the CSF of injured rodents. Van Den et al. injuries, which is further supported by our obser-
(2004) have demonstrated another beneficial effect vations.
of CsA in TBI proving that 30 min post-injury Unfortunately, the exact mechanism underlying
administration of CsA reduced APP mRNA at 2 the in vivo neuroprotective effect of PACAP has
and 6 h post-injury and likewise reduced APP ac- not been clarified so far. In vitro studies assign
cumulation in injured cell bodies. Nevertheless, both anti-apoptotic and anti-inflammatory actions
they did not prove axonal preservation in terms of to PACAP. In cerebellar granule cells, PACAP
APP-immunoreactivity in their experiments. While significantly inhibited the activation of caspase-3
these results might reflect the complexity in the (Vaudry et al., 2004). PACAP was proven to be a
pathogenesis of TBI, it is of note that several potent inactivator of induced microglial release of
studies have proved that APP immunoreactivity pro-inflammatory cytokines and nitric oxide
alone roughly underestimates the extent of TBI (Kong et al., 1999; Kim et al., 2000; Delgado et
and, in some axon-populations other mechanisms al., 2003). It is also believed to influence mi-
than impaired axoplasmatic transport and axonal tochondrial integrity as PACAP inhibited the mi-
swelling should be held accountable for axonal tochondrial Ca++ uptake induced inactivation of
demise (Singleton and Povlishock, 2004; Farkas aconitase, a key mitochondrial enzyme influencing
et al., 2006). the viability of neurons (Tabuchi et al., 2003).
While, without doubt, a continuous develop- In the last few years our laboratories extensively
ment of various anti-apoptotic or, even more investigated the purported neuroprotective role of
promising, anti-apoptotic, anti-necrotic drugs PACAP in a diffuse model of TBI. Intriguingly
should be a major goal of pharmaceutical re- enough, we were not able to reproduce neuropro-
search, neuroprotective substances physiologically tective effects in terms of preserving axonal integ-
produced in the brain itself should also be taken rity assessed by APP-immunoreactivity (vide
into account. To this end, a bulk of recently col- supra) with the administration paradigm (125 mg
lected evidences point to the neuroprotective ef- pre-injury, iv) that had worked in a middle cere-
fects of pituitary adenylate cyclase activating bral artery occlusion model of stroke (Reglodi et
polypeptide (PACAP). al., 2002). Nevertheless, we were able to establish
This member of the vasoactive intestinal peptide the dose-response curve for intracerebroventricu-
(VIP)/secretin/glucagon peptide family (Miyata et lar (icv) administration of PACAP, proving that
al., 1989) was discovered as a hypothalamic pep- 100 mg PACAP significantly reduced the density of
tide on its potential of increasing adenylate cyclase damaged, immunoreactive axons in the cortico-
activity in the pituitary gland and proved to exert spinal tract (Farkas et al., 2004). Our most recent
various effects in the central and peripheral nerv- observations proved that a considerable therapeu-
ous systems including neurotrophic and neuropro- tic window exists for post-injury PACAP treat-
tective ones (Arimura, 1998; Vaudry et al., 2000). ment demonstrating significant axonal protection
In the extradural static weight compression model in an icv administration paradigm 2 h following
of spinal cord injury in rat, post-injury PACAP impact acceleration TBI (Tamas et al., 2006).
treatment significantly reduced the number of A new avenue of pharmacotherapy targeting
apoptotic cells assessed by TUNEL in the spinal apoptosis in TBI is represented by the inhibition
cord (Katahira et al., 2003). of cell cycle proteins. Di Giovanni et al. (2005)
Further, PACAP has been shown to eliminate demonstrated that the cell cycle inhibitor flavopir-
the increase of TNF after spinal cord transection idol was not only capable of inhibiting etoposide-
(Kim et al., 2000) and in experimental TBI post- induced, capase-mediated cell death in cultured
injury induction of PACAP-mRNA and its upreg- primary cortical neurons but also inhibited the
ulation paralleled the decrease in the number of proliferation of astrocytes and microglia and de-
apoptotic cells (Skoglosa et al., 1999). These creased the extension of TBI-induced tissue lesion
90

and improved functional recovery in the lateral experimental models applied should be a prereq-
fluid percussion rodent model. uisite for successful development of neuroprotec-
As proteolytic processes are key players in TBI- tive agents. As far as study design is considered we
evoked apoptosis, the list of potential therapeutic should appreciate that the rather complex and
measures would not be complete without men- versatile nature of TBI occurring in human is hard
tioning the neuroprotective capacity of therapeutic to copy in any experimental model existing cur-
hypothermia. This intervention is purported to act rently in our hands. In the overwhelming majority
via general inhibition or slowing of the proteolytic of cases human TBI is a mixture of focal and
processes which could provide an extended ther- diffuse injuries complicated with secondary brain
apeutic window for further, specific therapeutic injury primarily caused by hypoxia and hypoper-
approaches while also enabling the neurons to fusion. The most frequently used models like con-
keep their mitochondria up and running and their trolled cortical impact or moderate/severe forms of
local energy homeostasis intact (Buki et al., fluid percussion brain injury produce mass tissue
1999a). Although the use of controlled hypother- destruction where strain and shearing-force distri-
mia in the treatment of human TBI remains con- bution leads to excessive influx and release of
troversial, on the basis of multiple lines of evidence Ca++, and excitatory amino acids, activation and
this therapeutic modality might be worth further generation of ROS. Although such models may
investigations and probably further clinical studies mimic the situation in the most severe forms of
planned on the basis of past experience could re- human TBI, they ignite such a bulk of various
solve the controversy concerning the role of hypo- pathways that could hardly be controlled with a
thermia in the treatment of the severely head drug exerting its effect on a single target.
injured (Clifton, 2004). Specifically, on the basis of While diffuse brain injury models seem to pro-
our current knowledge on its action, hypothermia vide a better environment to study more subtle
should be introduced relatively earlier in the care forms of TBI and theoretically provide a more
of TBI paying special attention to gradual re- convenient field to investigate the effect of pharma-
warming in conjunction with introduction of other ceutical agents, recent observations shed light on
treatment strategies such as FK-506 that has al- the extremely complex and heterogenous nature of
ready proved its potential to inhibit diffuse brain both diffuse axonal and neuronal injury. Specifi-
injury associated with post-hypothermic rewarm- cally, in different fiber populations considerably
ing (Suehiro et al., 2001). different markers of axonal damage may appear
representing heterogeneity in the pathobiology as
well as in therapeutic efficacy (Stone et al., 2004).
Concluding remarks Similarly, acceleration–deceleration injury evokes
various forms of neuronal-cellular damage indi-
There are several controversies in the development cating a likewise complex field to target therapeu-
of novel pharmacotherpeutic agents aimed at the tically (Singleton and Povlishock, 2004; Farkas
inhibition of pathobiological processes evoked by et al., 2006).
TBI. While everybody in the field considers the In light of the above passages and the diversity
treatment of TBI an extremely cost efficient medi- and interactivity of cell death pathways, a fatalistic
cal procedure, we still lack any novel, efficient approach would be to conclude that when a cell is
therapeutic agent. Despite of the promising results committed to die due to an extrinsic or intrinsic
achieved with various pre-clinical studies all can- death signal it will do so regardless of whatever
didates failed at the clinical phase and so far none therapeutic measures have been taken; inhibition
of them proved useful for the everyday practice. of apoptosis will ignite autophagic or necrotic
While detailed analysis of such failure is well be- cell death, ultimately leading to cellular demise.
yond the scope of this review, the authors feel ap- Nevertheless, the authors still feel appropriate to
propriate to note that more detailed understanding address this very issue from a different point of
and appreciation of cell death mechanisms and view with the believe that combined therapeutic
91

approaches or ‘‘polyhparmacia’’ might success- References


fully target intermingled death pathways with re-
sulting improved morphological as well as Abe, Y., Yamamoto, T., Sugiyama, Y., Watanabe, T., Saito,
functional outcome. To this end one can envision N., Kayama, H. and Kumagai, T. (2004) ‘‘Anoikis’’ of
oligodendrocytes induced by Wallerian degeneration: ultra-
a scenario where ultra-early (pre-hospital) appli-
structural observations. J. Neurotrauma, 21: 119–124.
cation of therapeutic measures aimed to slow Abrahamson, E.E., Ikonomovic, M.D., Ciallella, J.R., Hope,
down proteolytic processes (such as hypothermia C.E., Paljug, W.R., Isanski, B.A., Flood, D.G., Clark, R.S.
treatment) and preserving energy homeostasis and DeKosky, S.T. (2006) Caspase inhibition therapy abol-
(PARP-inhibition, CsA-treatment) is followed or ishes brain trauma-induced increases in Abeta peptide: im-
plications for clinical outcome. Exp. Neurol., 197: 437–450.
joined by caspase and calpain inhibitors, or
Adams, J.M. and Cory, S. (2002) Apoptosomes: engines for
PACAP treatment. caspase activation. Curr. Opin. Cell Biol., 14: 715–720.
As far as the title of the present article is con- Aguilar, H.I., Botla, R., Arora, A.S., Bronk, S.F. and Gores,
sidered, to date it is hard to predict, whether in- G.J. (1996) Induction of the mitochondrial permeability
hibition of apoptosis is useful; one can only transition by protease activity in rats: a mechanism of he-
patocyte necrosis. Gastroenterology, 110: 558–566.
conclude that it is most probably beneficial in
Alves, O., Tolias, C.M., Lewis, C., Hayes, R.L., Choi, S.C.,
conjunction with other measures taken to inhibit Gilman, C., Enriquez, P., Povlishock, J.T. and Bullock, M.R.
various different pathways of traumatically in- (2003) Brain neurochemical alterations in patients with severe
duced neural demise. brain injury treated with cyclosporin-a. A placebo-controlled
study. Preliminary report. J. Neurotrauma, 20: 1125.
Arden, N. and Betenbaugh, M.J. (2004) Life and death in
Abbreviations mammalian cell culture: strategies for apoptosis inhibition.
Trends Biotechnol., 22: 174–180.
Arimura, A. (1998) Perspectives on pituitary adenylate cyclase
AIF apoptosis inducing factor activating polypeptide (PACAP) in the neuroendocrine, en-
CD cell death docrine, and nervous systems. Jpn. J. Physiol., 48: 301–331.
CsA cyclosporine A Assuncao, G.C. and Linden, R. (2004) Programmed cell deaths.
ER endoplasmatic reticulum Apoptosis and alternative deathstyles. Eur. J. Biochem., 271:
1638–1650.
IAP inhibitor of apoptosis protein
Bartus, R. (1997) The calpain hypothesis of neurodegeneration:
Icv intracerebroventricular (icv) evidence for a common cytotoxic pathway. Neuroscientist, 3:
MPTP mitochondrial permeability tran- 314–327.
sition pore Beer, R., Franz, G., Krajewski, S., Pike, B.R., Hayes, R.L.,
NMDA N-methyl-D-aspartate Reed, J.C., Wang, K.K., Klimmer, C., Schmutzhard, E., Po-
ewe, W. and Kampfl, A. (2001) Temporal and spatial profile
PARP-1 poly(ADP-ribose)polymerase 1
of caspase 8 expression and proteolysis after experimental
TAI traumatic axonal injury traumatic brain injury. J. Neurochem., 78: 862–873.
TBI traumatic brain injury Beer, R., Franz, G., Srinivasan, A., Hayes, R.L., Pike, B.R.,
TNF tumor necrosis factor Newcomb, J.K., Zhao, X., Schmutzhard, E., Poewe, W. and
Kampfl, A. (2000) Temporal profile and cell subtype distri-
bution of activated caspase-3 following experimental trau-
matic brain injury. J. Neurochem., 75: 1264–1273.
Besson, V.C., Croci, N., Boulu, R.G., Plotkine, M. and Marc-
Aknowledgments hand-Verrecchia, C. (2003) Deleterious poly(ADP-ribose)po-
lymerase-1 pathway activation in traumatic brain injury in
rat. Brain Res., 989: 58–66.
Supported by the Hungarian Science Found Besson, V.C., Zsengeller, Z., Plotkine, M., Szabo, C. and
(OTKA T048724) and the Bekesi Scholarship of Marchand-Verrecchia, C. (2005) Beneficial effects of PJ34
the Hungarian Academy of Sciences. The authors and INO-1001, two novel water-soluble poly(ADP-ribose)
thank for the invaluable advices and help of Pro- polymerase inhibitors, on the consequences of traumatic
brain injury in rat. Brain Res., 1041: 149–156.
fessor John T. Povlishock, Department of Anat-
Buki, A., Farkas, O., Doczi, T. and Povlishock, J.T. (2003)
omy and Neurobiology, Medical College of Preinjury administration of the calpain inhibitor MDL-28170
Virginia, Virginia Commonwealth University, attenuates traumatically induced axonal injury. J. Neuro-
Richmond, VA 23298-0709. trauma, 20: 261–268.
92

Buki, A., Koizumi, H. and Povlishock, J.T. (1999a) Moderate Ditzel, M., Wilson, R., Tenev, T., Zachariou, A., Paul, A.,
posttraumatic hypothermia decreases early calpain-mediated Deas, E. and Meier, P. (2003) Degradation of DIAP1 by the
proteolysis and concomitant cytoskeletal compromise in N-end rule pathway is essential for regulating apoptosis. Nat.
traumatic axonal injury. Exp. Neurol., 159: 319–328. Cell Biol., 5: 467–473.
Buki, A., Okonkwo, D.O., Wang, K.K. and Povlishock, J.T. Farkas, O., Lifshitz, J. and Povlishock, J.T. (2006) Mechano-
(2000) Cytochrome c release and caspase activation in trau- poration induced by diffuse traumatic brain injury: an irre-
matic axonal injury. J. Neurosci., 20: 2825–2834. versible or reversible response to injury? J. Neurosci., 26:
Buki, A., Siman, R., Trojanowski, J.Q. and Povlishock, J.T. 3130–3140.
(1999b) The role of calpain-mediated spectrin proteolysis in Farkas, O., Tamas, A., Zsombok, A., Reglodi, D., Pal, J., Buki,
traumatically induced axonal injury. J. Neuropathol. Exp. A., Lengvari, I., Povlishock, J.T. and Doczi, T. (2004) Effects
Neurol., 58: 365–375. of pituitary adenylate cyclase activating polypeptide in a rat
Burkle, A. (2001) Physiology and pathophysiology of poly model of traumatic brain injury. Regul. Pept., 123: 69–75.
(ADP-ribosyl)ation. Bioessays, 23: 795–806. Ferri, K.F. and Kroemer, G. (2001) Organelle-specific initiation
Cai, J. and Jones, D.P. (1998) Superoxide in apoptosis. Mito- of cell death pathways. Nat. Cell Biol., 3: E255–E263.
chondrial generation triggered by cytochrome c loss. J. Biol. Gabbita, S.P., Scheff, S.W., Menard, R.M., Roberts, K., Fug-
Chem., 273: 11401–11404. accia, I. and Zemlan, F.P. (2005) Cleaved-tau: a biomarker of
Cai, J., Yang, J. and Jones, D.P. (1998) Mitochondrial control neuronal damage after traumatic brain injury. J. Neurotrau-
of apoptosis: the role of cytochrome c. Biochim. Biophys. ma, 22: 83–94.
Acta, 1366: 139–149. Gallyas, F., Pal, J., Farkas, O. and Doczi, T. (2006) The fate of
Chinnaiyan, A.M., O’Rourke, K., Tewari, M. and Dixit, V.M. axons subjected to traumatic ultrastructural (neurofilament)
(1995) FADD, a novel death domain-containing protein, in- compaction: an electron-microscopic study. Acta Neuro-
teracts with the death domain of Fas and initiates apoptosis. pathol. (Berl.), 111: 229–237.
Cell, 81: 505–512. Garcia-Estrada, J., Luquin, S., Fernandez, A.M. and Garcia-
Clark, R.S., Kochanek, P.M., Watkins, S.C., Chen, M., Dixon, Segura, L.M. (1999) Dehydroepiandrosterone, pregnenolone
C.E., Seidberg, N.A., Melick, J., Loeffert, J.E., Nathaniel, and sex steroids down-regulate reactive astroglia in the male
P.D., Jin, K.L. and Graham, S.H. (2000) Caspase-3 mediated rat brain after a penetrating brain injury. Int. J. Dev. Neuro-
neuronal death after traumatic brain injury in rats. J. Neuro- sci., 17: 145–151.
chem., 74: 740–753. Ghajar, J. (2000) Traumatic brain injury. Lancet, 356: 923–929.
Clifton, G.L. (2004) Is keeping cool still hot? An update on Ginis, I., Hallenbeck, J.M., Liu, J., Spatz, M., Jaiswal, R. and
hypothermia in brain injury. Curr. Opin. Crit. Care, 10: Shohami, E. (2000) Tumor necrosis factor and reactive ox-
116–119. ygen species cooperative cytotoxicity is mediated via inhibi-
Conti, A.C., Raghupathi, R., Trojanowski, J.Q. and McIntosh, tion of NF-kappaB. Mol. Med., 6: 1028–1041.
T.K. (1998) Experimental brain injury induces regionally Giza, C.C. and Hovda, D.A. (2001) The neurometabolic cas-
distinct apoptosis during the acute and delayed post-trau- cade of concussion. J. Athl. Train, 36: 228–235.
matic period. J. Neurosci., 18: 5663–5672. Gu, F., Hata, R., Toku, K., Yang, L., Ma, Y.J., Maeda, N.,
Cousin, M.A. and Robinson, P.J. (2001) The dephosphins: Sakanaka, M. and Tanaka, J. (2003) Testosterone up-
dephosphorylation by calcineurin triggers synaptic vesicle regulates aquaporin-4 expression in cultured astrocytes.
endocytosis. Trends Neurosci., 24: 659–665. J. Neurosci. Res., 72: 709–715.
Curry III, J.J. and Heim, L.M. (1966) Brain myelination after Hortobagyi, T., Gorlach, C., Benyo, Z., Lacza, Z., Hortobagyi,
neonatal administration of oestradiol. Nature, 209: 915–916. S., Wahl, M. and Harkany, T. (2003) Inhibition of neuronal
Dawson, T.M., Steiner, J.P., Dawson, V.L., Dinerman, J.L., nitric oxide synthase-mediated activation of poly(ADP-rib-
Uhl, G.R. and Snyder, S.H. (1993) Immunosuppressant ose) polymerase in traumatic brain injury: neuroprotection
FK506 enhances phosphorylation of nitric oxide synthase by 3-aminobenzamide. Neuroscience, 121: 983–990.
and protects against glutamate neurotoxicity. Proc. Natl. Jurgensmeier, J.M., Xie, Z., Deveraux, Q., Ellerby, L., Brede-
Acad. Sci. U.S.A., 90: 9808–9812. sen, D. and Reed, J.C. (1998) Bax directly induces release of
Delgado, M., Leceta, J. and Ganea, D. (2003) Vasoactive in- cytochrome c from isolated mitochondria. Proc. Natl. Acad.
testinal peptide and pituitary adenylate cyclase-activating Sci. U.S.A., 95: 4997–5002.
polypeptide inhibit the production of inflammatory media- Kampfl, A., Posmantur, R.M., Zhao, X., Schmutzhard, E.,
tors by activated microglia. J. Leukoc. Biol., 73: 155–164. Clifton, G.L. and Hayes, R.L. (1997) Mechanisms of calpain
Denecker, G., Vercammen, D., Declercq, W. and Vandena- proteolysis following traumatic brain injury: implications for
beele, P. (2001) Apoptotic and necrotic cell death induced by pathology and therapy: implications for pathology and ther-
death domain receptors. Cell Mol. Life Sci., 58: 356–370. apy: a review and update. J. Neurotrauma, 14: 121–134.
Di Giovanni, S., Movsesyan, V., Ahmed, F., Cernak, I., Schin- Katahira, M., Yone, K. and Arishima, Y. (2003) The neuro-
elli, S., Stoica, B. and Faden, A.I. (2005) Cell cycle inhibition protective effects of PACAP on spinal cor injury (SCI) in
provides neuroprotection and reduces glial proliferation and rats. Regul. Pept., 15: 49.
scar formation after traumatic brain injury. Proc. Natl. Acad. Kawamura, M., Nakajima, W., Ishida, A., Ohmura, A., Miura,
Sci. U.S.A., 102: 8333–8338. S. and Takada, G. (2005) Calpain inhibitor MDL 28170
93

protects hypoxic-ischemic brain injury in neonatal rats by Lewin, I.C.F. (1991) Update of Max, W., Mackenzie, E.J.,
inhibition of both apoptosis and necrosis. Brain Res., 1037: Rice, D.P. Head injuries: costs and consequences. J. Head
59–69. Trauma Rehabil., 67: 76–91.
Kim, W.K., Kan, Y., Ganea, D., Hart, R.P., Gozes, I. and Liu, J., Farmer Jr., J.D., Lane, W.S., Friedman, J., Weissman,
Jonakait, G.M. (2000) Vasoactive intestinal peptide and pi- I. and Schreiber, S.L. (1991) Calcineurin is a common target
tuitary adenylyl cyclase-activating polypeptide inhibit tumor of cyclophilin-cyclosporin A and FKBP-FK506 complexes.
necrosis factor-alpha production in injured spinal cord and Cell, 66: 807–815.
in activated microglia via a cAMP-dependent pathway. Liu, M.C., Akle, V., Zheng, W., Dave, J.R., Tortella, F.C.,
J. Neurosci., 20: 3622–3630. Hayes, R.L. and Wang, K.K. (2006) Comparing calpain- and
Knoblach, S.M., Alroy, D.A., Nikolaeva, M., Cernak, I., Sto- caspase-3-mediated degradation patterns in traumatic brain in-
ica, B.A. and Faden, A.I. (2004) Caspase inhibitor z-DEVD- jury by differential proteome analysis. Biochem. J., 394:
fmk attenuates calpain and necrotic cell death in vitro and 715–725.
after traumatic brain injury. J. Cereb. Blood Flow Metab., Lockshin, R.A. and Zakeri, Z. (2002) Caspase-independent cell
24: 1119–1132. deaths. Curr. Opin. Cell Biol., 14: 727–733.
Knoblach, S.M., Nikolaeva, M., Huang, X., Fan, L., Krajew- Lockshin, R.A. and Zakeri, Z. (2004) Apoptosis, autophagy,
ski, S., Reed, J.C. and Faden, A.I. (2002) Multiple caspases and more. Int. J. Biochem. Cell Biol., 36: 2405–2419.
are activated after traumatic brain injury: evidence for in- MacGregor, D.G., Avshalumov, M.V. and Rice, M.E. (2003)
volvement in functional outcome. J. Neurotrauma, 19: Brain edema induced by in vitro ischemia: causal factors and
1155–1170. neuroprotection. J. Neurochem., 85: 1402–1411.
Komjati, K., Besson, V.C. and Szabo, C. (2005) Poly (ADP- Markgraf, C.G., Velayo, N.L., Johnson, M.P., McCarty, D.R.,
ribose) polymerase inhibitors as potential therapeutic agents Medhi, S., Koehl, J.R., Chmielewski, P.A. and Linnik, M.D.
in stroke and neurotrauma. Curr. Drug Targets CNS Neurol. (1998) Six-hour window of opportunity for calpain in-
Disord., 4: 179–194. hibition in focal cerebral ischemia in rats. Stroke, 29:
Kong, L.Y., Maderdrut, J.L., Jeohn, G.H. and Hong, J.S. 152–158.
(1999) Reduction of lipopolysaccharide-induced neurotoxic- Miyata, A., Arimura, A., Dahl, R.R., Minamino, N., Uehara,
ity in mixed cortical neuron/glia cultures by femtomolar A., Jiang, L., Culler, M.D. and Coy, D.H. (1989) Isolation of
concentrations of pituitary adenylate cyclase-activating poly- a novel 38 residue-hypothalamic polypeptide which stimu-
peptide. Neuroscience, 91: 493–500. lates adenylate cyclase in pituitary cells. Biochem. Biophys.
Kroemer, G. (2003) Mitochondrial control of apoptosis: an in- Res. Commun., 164: 567–574.
troduction. Biochem. Biophys. Res. Commun., 304: 433–435. Narayan, R.K., Michel, M.E., Ansell, B., Baethmann, A.,
Kruman, I.I. and Mattson, M.P. (1999) Pivotal role of mito- Biegon, A., Bracken, M.B., Bullock, M.R., Choi, S.C., Clif-
chondrial Ca++ uptake in neural cell apoptosis and necrosis. ton, G.L., Contant, C.F., Coplin, W.M., Dietrich, W.D.,
J. Neurochem., 72: 529–540. Ghajar, J., Grady, S.M., Grossman, R.G., Hall, E.D.,
Kuroda, S., Janelidze, S. and Siesjo, B.K. (1999) The immuno- Heetderks, W., Hovda, D.A., Jallo, J., Katz, R.L., Knoller,
suppressants cyclosporin A and FK506 equally ameliorate N., Kochanek, P.M., Maas, A.I., Majde, J., Marion, D.W.,
brain damage due to 30-min middle cerebral artery occlusion Marmarou, A., Marshall, L.F., McIntosh, T.K., Miller, E.,
in hyperglycemic rats. Brain Res., 835: 148–153. Mohberg, N., Muizelaar, J.P., Pitts, L.H., Quinn, P.,
Lacza, Z., Horvath, E.M., Komjati, K., Hortobagyi, T., Szabo, Riesenfeld, G., Robertson, C.S., Strauss, K.I., Teasdale, G.,
C. and Busija, D.W. (2003) PARP inhibition improves the Temkin, N., Tuma, R., Wade, C., Walker, M.D., Weinrich,
effectiveness of neural stem cell transplantation in experi- M., Whyte, J., Wilberger, J., Young, A.B. and Yurkewicz, L.
mental brain trauma. Int. J. Mol. Med., 12: 153–159. (2002) Clinical trials in head injury. J. Neurotrauma, 19:
Lang-Rollin, I.C., Rideout, H.J., Noticewala, M. and Stefanis, 503–557.
L. (2003) Mechanisms of caspase-independent neuronal Okonkwo, D.O., Melon, D.E., Pellicane, A.J., Mutlu, L.K.,
death: energy depletion and free radical generation. J. Neuro- Rubin, D.G., Stone, J.R. and Helm, G.A. (2003) Dose-
sci., 23: 11015–11025. response of cyclosporin A in attenuating traumatic axonal
LaPlaca, M.C., Zhang, J., Raghupathi, R., Li, J.H., Smith, F., injury in rat. Neuroreport, 14: 463–466.
Bareyre, F.M., Snyder, S.H., Graham, D.I. and McIntosh, Okonkwo, D.O., Pettus, E.H., Moroi, J. and Povlishock, J.T.
T.K. (2001) Pharmacologic inhibition of poly(ADP-ribose) (1998) Alteration of the neurofilament sidearm and its rela-
polymerase is neuroprotective following traumatic brain in- tion to neurofilament compaction occurring with traumatic
jury in rats. J. Neurotrauma, 18: 369–376. axonal injury. Brain Res., 784: 1–6.
Larner, S.F., Hayes, R.L., McKinsey, D.M., Pike, B.R. and Okonkwo, D.O. and Povlishock, J.T. (1999) An intrathecal
Wang, K.K. (2004) Increased expression and processing of bolus of cyclosporin A before injury preserves mitochondrial
caspase-12 after traumatic brain injury in rats. J. Neuro- integrity and attenuates axonal disruption in traumatic brain
chem., 88: 78–90. injury. J. Cereb. Blood Flow Metab., 19: 443–451.
Larner, S.F., McKinsey, D.M., Hayes, R.L. and Wang, K.K. Oyadomari, S., Araki, E. and Mori, M. (2002) Endoplasmic
(2005) Caspase 7: increased expression and activation after reticulum stress-mediated apoptosis in pancreatic beta-cells.
traumatic brain injury in rats. J. Neurochem., 94: 97–108. Apoptosis, 7: 335–345.
94

Pant, H.C. (1988) Dephosphorylation of neurofilament proteins Schendel, S.L., Montal, M. and Reed, J.C. (1998) Bcl-2 family
enhances their susceptibility to degradation by calpain. Bio- proteins as ion-channels. Cell Death Differ., 5: 372–380.
chem. J., 256: 665–668. Scorrano, L. and Korsmeyer, S.J. (2003) Mechanisms of cyto-
Pettus, E.H., Christman, C.W., Giebel, M.L. and Povlishock, chrome c release by proapoptotic BCL-2 family members.
J.T. (1994) Traumatically induced altered membrane perme- Biochem. Biophys. Res. Commun., 304: 437–444.
ability: its relationship to traumatically induced reactive axo- Siesjo, B.K., Hu, B. and Kristian, T. (1999) Is the cell death
nal change. J. Neurotrauma, 11: 507–522. pathway triggered by the mitochondrion or the endoplasmic
Povlishock, J.T., Marmarou, A., McIntosh, T., Trojanowski, reticulum? J. Cereb. Blood Flow Metab., 19: 19–26.
J.Q. and Moroi, J. (1997) Impact acceleration injury in the Singleton, R.H. and Povlishock, J.T. (2004) Identification and
rat: evidence for focal axolemmal change and related neuro- characterization of heterogeneous neuronal injury and death
filament sidearm alteration. J. Neuropathol. Exp. Neurol., in regions of diffuse brain injury: evidence for multiple in-
56: 347–359. dependent injury phenotypes. J. Neurosci., 24: 3543–3553.
Pyrzynska, B., Lis, A., Mosieniak, G. and Kaminska, B. (2001) Singleton, R.H., Stone, J.R., Okonkwo, D.O., Pellicane, A.J.
Cyclosporin A-sensitive signaling pathway involving calcine- and Povlishock, J.T. (2001) The immunophilin ligand FK506
urin regulates survival of reactive astrocytes. Neurochem. attenuates axonal injury in an impact acceleration model of
Int., 38: 409–415. traumatic brain injury. J. Neurotrauma, 18: 607–614.
Reed, J.C. (1998) Bcl-2 family proteins. Oncogene, 17: Skoglosa, Y., Lewen, A., Takei, N., Hillered, L. and Lindholm,
3225–3236. D. (1999) Regulation of pituitary adenylate cyclase activating
Reglodi, D., Tamas, A., Somogyvari-Vigh, A., Szanto, Z., polypeptide and its receptor type 1 after traumatic brain in-
Kertes, E., Lenard, L., Arimura, A. and Lengvari, I. (2002) jury: comparison with brain-derived neurotrophic factor and
Effects of pretreatment with PACAP on the infarct size and the induction of neuronal cell death. Neuroscience, 90:
functional outcome in rat permanent focal cerebral ischemia. 235–247.
Peptides, 23: 2227–2234. Soustiel, J.F., Palzur, E., Nevo, O., Thaler, I. and Vlodavsky,
Riess, P., Bareyre, F.M., Saatman, K.E., Cheney, J.A., Lifshitz, E. (2005) Neuroprotective anti-apoptosis effect of estrogens
J., Raghupathi, R., Grady, M.S., Neugebauer, E. and McIn- in traumatic brain injury. J. Neurotrauma, 22: 345–352.
tosh, T.K. (2001) Effects of chronic, post-injury Cyclosporin Stone, J.R., Okonkwo, D.O., Dialo, A.O., Rubin, D.G., Mutlu,
A administration on motor and sensorimotor function fol- L.K., Povlishock, J.T. and Helm, G.A. (2004) Impaired ax-
lowing severe, experimental traumatic brain injury. Restor. onal transport and altered axolemmal permeability occur in
Neurol. Neurosci., 18: 1–8. distinct populations of damaged axons following traumatic
Ringger, N.C., O’Steen, B.E., Brabham, J.G., Silver, X., brain injury. Exp. Neurol., 190: 59–69.
Pineda, J., Wang, K.K., Hayes, R.L. and Papa, L. (2004) Stys, P.K. and Jiang, Q. (2002) Calpain-dependent neurofila-
A novel marker for traumatic brain injury: CSF alphaII- ment breakdown in anoxic and ischemic rat central axons.
spectrin breakdown product levels. J. Neurotrauma, 21: Neurosci. Lett., 328: 150–154.
1443–1456. Suehiro, E., Singleton, R.H., Stone, J.R. and Povlishock, J.T.
Rink, A., Fung, K.M., Trojanowski, J.Q., Lee, V.M., Neuge- (2001) The immunophilin ligand FK506 attenuates the ax-
bauer, E. and McIntosh, T.K. (1995) Evidence of apoptotic onal damage associated with rapid rewarming following
cell death after experimental traumatic brain injury in the rat. posttraumatic hypothermia. Exp. Neurol., 172: 199–210.
Am. J. Pathol., 147: 1575–1583. Sullivan, P.G., Keller, J.N., Bussen, W.L. and Scheff, S.W.
Roof, R.L. and Hall, E.D. (2000) Gender differences in acute (2002) Cytochrome c release and caspase activation after
CNS trauma and stroke: neuroprotective effects of estrogen traumatic brain injury. Brain Res., 949: 88–96.
and progesterone. J. Neurotrauma, 17: 367–388. Susin, S.A., Lorenzo, H.K., Zamzami, N., Marzo, I., Snow,
Saatman, K.E., Murai, H., Bartus, R.T., Smith, D.H., Hay- B.E., Brothers, G.M., Mangion, J., Jacotot, E., Costantini,
ward, N.J., Perri, B.R. and McIntosh, T.K. (1996) Calpain P., Loeffler, M., Larochette, N., Goodlett, D.R., Aebersold,
inhibitor AK295 attenuates motor and cognitive deficits fol- R., Siderovski, D.P., Penninger, J.M. and Kroemer, G.
lowing experimental brain injury in the rat. Proc. Natl. Acad. (1999) Molecular characterization of mitochondrial apopto-
Sci. U.S.A., 93: 3428–3433. sis-inducing factor. Nature, 397: 441–446.
Saatman, K.E., Zhang, C., Bartus, R.T. and McIntosh, T.K. Susin, S.A., Zamzami, N. and Kroemer, G. (1998) Mito-
(2000) Behavioral efficacy of posttraumatic calpain inhibi- chondria as regulators of apoptosis: doubt no more. Biochim.
tion is not accompanied by reduced spectrin proteolysis, Biophys. Acta, 1366: 151–165.
cortical lesion, or apoptosis. J. Cereb. Blood Flow Metab., Tabuchi, A., Funaji, K., Nakatsubo, J., Fukuchi, M., Tsuchiya,
20: 66–73. T. and Tsuda, M. (2003) Inactivation of aconitase during the
Saikumar, P., Dong, Z., Weinberg, J.M. and Venkatachalam, apoptosis of mouse cerebellar granule neurons induced by a
M.A. (1998) Mechanisms of cell death in hypoxia/reoxygen- deprivation of membrane depolarization. J. Neurosci. Res.,
ation injury. Oncogene, 17: 3341–3349. 71: 504–515.
Saltman, R.B. and Findling, R.L. (1997) European Health Care Tamas, A., Zsombok, A., Farkas, O., Reglodi, D., Pal, J., Buki,
Reform. Analysis of current strategies. In: WHO Regional A., Lengvari, I., Povlishock, J.T. and Doczi, T. (2006) Post-
Publications European Series 72 Copenhagen: WHO. injury administration of pituitary adenylate cyclase activating
95

polypeptide (PACAP) attenuates traumatically induced ax- treatment and outcome. Acta Neurochir. (Wien.), 141:
onal injury in rats. J. Neurotrauma, 23: 686–695. 1203–1208.
Thurman, D.J., Alverson, C., Dunn, K.A., Guerrero, J. and Wang, K.K., Posmantur, R., Nath, R., McGinnis, K., Whitton,
Sniezek, J.E. (1999) Traumatic brain injury in the United M., Talanian, R.V., Glantz, S.B. and Morrow, J.S. (1998)
States: A public health perspective. J. Head Trauma Re- Simultaneous degradation of alphaII- and betaII-spectrin by
habil., 14: 602–615. caspase 3 (CPP32) in apoptotic cells. J. Biol. Chem., 273:
Van Den, H.C., Donkin, J.J., Finnie, J.W., Blumbergs, P.C., 22490–22497.
Kuchel, T., Koszyca, B., Manavis, J., Jones, N.R., Reilly, P.L. Warren, M.W., Kobeissy, F.H., Liu, M.C., Hayes, R.L., Gold,
and Vink, R. (2004) Downregulation of amyloid precursor M.S. and Wang, K.K. (2005) Concurrent calpain and ca-
protein (APP) expression following post-traumatic cyclo- spase-3 mediated proteolysis of alpha II-spectrin and tau in
sporin-A administration. J. Neurotrauma, 21: 1562–1572. rat brain after methamphetamine exposure: a similar profile
Varshavsky, A. (2003) The N-end rule and regulation of to traumatic brain injury. Life Sci., 78: 301–309.
apoptosis. Nat. Cell Biol., 5: 373–376. Yakovlev, A.G., Knoblach, S.M., Fan, L., Fox, G.B., Good-
Vaudry, D., Cottet-Rousselle, C., Basille, M., Falluel-Morel, night, R. and Faden, A.I. (1997) Activation of CPP32-like
A., Fournier, A., Vaudry, H. and Gonzalez, B.J. (2004) Pi- caspases contributes to neuronal apoptosis and neurological
tuitary adenylate cyclase-activating polypeptide inhibits ca- dysfunction after traumatic brain injury. J. Neurosci., 17:
spase-3 activity but does not protect cerebellar granule 7415–7424.
neurons against beta-amyloid (25–35)-induced apoptosis. Yamashima, T. (2004) Ca2+-dependent proteases in ischemic
Regul. Pept., 123: 43–49. neuronal death: a conserved ‘calpain-cathepsin cascade’ from
Vaudry, D., Gonzalez, B.J., Basille, M., Yon, L., Fournier, A. nematodes to primates. Cell Calcium, 36: 285–293.
and Vaudry, H. (2000) Pituitary adenylate cyclase-activating Zong, W.X., Li, C., Hatzivassiliou, G., Lindsten, T., Yu, Q.C.,
polypeptide and its receptors: from structure to functions. Yuan, J. and Thompson, C.B. (2003) Bax and Bak can lo-
Pharmacol. Rev., 52: 269–324. calize to the endoplasmic reticulum to initiate apoptosis. J.
Vukic, M., Negovetic, L., Kovac, D., Ghajar, J., Glavic, Z. and Cell Biol., 162: 59–69.
Gopcevic, A. (1999) The effect of implementation of guide- Zoratti, M. and Szabo, I. (1995) The mitochondrial permeabi-
lines for the management of severe head injury on patient lity transition. Biochim. Biophys. Acta, 1241: 139–176.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 7

Substance P in traumatic brain injury

James J. Donkin1, Renee J. Turner1, Islam Hassan1 and Robert Vink1,2,

1
Discipline of Pathology, University of Adelaide, Adelaide, South Australia
2
Centre for Neurological Diseases, The Hanson Institute, Adelaide, South Australia

Abstract: Recent evidence has suggested that neuropeptides, and in particular substance P (SP), may play a
critical role in the development of morphological injury and functional deficits following acute insults to the
brain. Few studies, however, have examined the role of SP, and more generally, neurogenic inflammation,
in the pathophysiology of traumatic brain injury and stroke. Those studies that have been reported suggest
that SP is released following injury to the CNS and facilitates the increased permeability of the blood brain
barrier, the development of vasogenic edema and the subsequent cell death and functional deficits that are
associated with these events. Inhibition of the SP activity, either through inhibition of the neuropeptide
release or the use of SP receptor antagonists, have consistently resulted in profound decreases in edema
formation and marked improvements in functional outcome. The current review summarizes the role of SP
in acute brain injury, focussing on its properties as a neurotransmitter and the potential for SP to adversely
affect outcome.

Keywords: neurotrauma; traumatic brain injury; edema; neuropeptides

Introduction such as helmets, airbags and seatbelts, little can be


done to prevent primary injury, and such injury
Traumatic brain injury is the leading cause of may be regarded as irreversible. In contrast, seco-
death and disability in people under the age of 40 ndary injury is made up of the delayed biochemical
years (Fleminger and Ponsford, 2005) with inci- and physiological factors that are initiated by the
dence rates estimated at 150–250 cases per 100,000 primary event, and these secondary injury factors
populations per year (Leon-Carrion et al., 2005). are thought to account for much of the morbidity
The cost for rehabilitation and care of such indi- following brain injury (McIntosh et al., 1996). This
viduals to the community runs into billion of dol- secondary injury cascade evolves over minutes to
lars annually. Despite the enormity of this public days and even months after the initial event, and
health problem, no effective treatment currently as such, there are opportunities for interventional
exists. It is now accepted that brain injury results pharmacology to prevent further injury and im-
in the development of neurologic deficits through prove outcome. As a result, research has focused
two main mechanisms. Firstly, the primary event on the identification of secondary injury factors
includes the mechanical processes such as shear- and the development of novel therapies that
ing, laceration and stretching of nerve fibres that attenuate, or even prevent, their action.
occurs at the time of the injury (Graham et al., A number of secondary injury factors have been
1992, 1996). Besides the use of preventive measures identified to date including blood brain barrier
(BBB) opening, edema formation, release of neuro-
Corresponding author. Tel.: +61-8–8222-3092; Fax: +61-8- transmitters such as excitatory amino acids, ion
8222-3093; E-mail: Robert.Vink@adelaide.edu.au changes, oxidative stress and bioenergetic failure,

DOI: 10.1016/S0079-6123(06)61007-8 97
98

amongst others. At the cellular level, the initial resident microglia. Changes in inflammatory cells
effect of mechanical impact is to increase the se- are paralleled by proliferation of astrocytes
lective permeability of the cell membrane and this (Hausmann and Betz, 2000), proliferation of cap-
occurs to varying degrees depending on the severity illaries, swelling of their endothelium and by the
of injury. This effect, known as mechanoporation formation of perivascular edema (Bullock et al.,
(Gennarelli and Graham, 1998), allows for the in- 1991; Vaz et al., 1997). The changes often culmi-
creased movement of ions into and out of cells nate in a gliotic scar studded with hemosiderin-
along their natural concentration gradients. Thus, laden macrophages.
calcium (Ca2+), sodium (Na+) and chloride (Cl) The osmotic subroutine occurs because the net
ions enter cells whilst potassium (K+) and magne- influx of ions is much greater than the net efflux of
sium (Mg2+) ions are lost from the cells. From this ions. Consequently, water is osmotically obligated
point, the pathological changes might be con- to follow the passage of ions into cells. This leads
sidered to differentiate into two subroutines ac- to cellular swelling, referred to as cytotoxic edema.
cording to whether these alterations in ion Glia also swell due to the fact that they function in
concentration cause effects due to their chemical the uptake of the K+ accumulating in the extra-
properties (the enzymatic subroutine) or due to cellular fluid (Reilly, 2001). This glial swelling may
their physical properties (the osmotic subroutine). further compromise cerebral perfusion by com-
The enzymatic subroutine revolves around the pressing the small blood vessels running amidst the
influx of calcium ions, which activates several cel- glial cells. Alternatively, water may be obligated to
lular enzyme cascades. These enzyme cascades me- follow an osmotic gradient generated by the pas-
diate cellular dysfunction, including activation of sage of proteins and ions from the vasculature to
calpains, axonal injury, accumulation of free rad- the brain interstitium. This edema is known as
ical species, increased production of nitric oxide vasogenic edema and is associated with an in-
and induction of proinflammatory gene expres- creased permeability of the BBB, best observed in
sion, which can potentially culminate in cell death the first 5 h after the TBI (O’Connor et al., 2003).
(Obrenovitch and Urenjak, 1997; Xiong et al., The microvasculature in the injury zone is affected
1997; Vespa et al., 1998). Among these different such that capillaries exhibit increased permeability
mechanisms of delayed cell damage in TBI, in- and arterioles lose their capacity to regulate blood
flammation is the predominant mechanism in the flow (Dietrich et al., 1994). Although the exact
case of contusions (Graham et al., 2002). The in- mechanisms of BBB disruption are unknown, it is
flammatory reaction consists of various compo- hypothesised that inflammatory mediators play a
nents that evolve at their own specific rate and role, possibly through receptor-mediated actions.
according to their own specific pattern as the age Among these inflammatory mediators, neuropep-
of the lesion increases (Oehmichen and Raff, 1980; tides such as substance P (SP), released from peri-
Oehmichen et al., 1986; Cervos-Navarro and vascular axons, are prime candidates.
Lafuente, 1991). For example, in terms of inflam- It is clear that the development of edema is
matory cell infiltration, several microscopic studies common to both the enzymatic and osmotic sub-
of human injury have demonstrated a distinct time routines of injury following TBI, and its adverse
course (Holmin et al., 1998; Hausmann et al., consequences on outcome through effects on in-
1999; Engel et al., 2000). In lesions aged up to 24 h, tracranial pressure (ICP) have been well described
the cellular component of inflammation was (Marmarou et al., 2000). Current protocols for the
represented by margination of neutrophils (also management of raised ICP include pharmacologi-
referred to polymorphonuclear leukocytes or cal regimens such as administration of hyperos-
PMNLs) in the vessels, whereas at 3–5 days of motic agents and barbiturates, or induction
survival, the inflammatory cell reaction consisted of hyperventilation and hypothermia, as well as
of tissue infiltration of not only neutrophils, but surgical procedures such as drainage of cerebro-
also monocyte/macrophages and CD4- and CD8- spinal fluid (CSF) and decompressive craniotomy
positive T-lymphocytes, as well as an activation of (Graham et al., 2002). Unfortunately, in terms of
99

improving patient survival rates and functional widely accepted (Otsuka and Takahashi, 1977;
outcome, these interventions have essentially been Nicoll et al., 1980; Pernow, 1983; Otsuka and
inadequate, largely because they do not address Yoshioka, 1993). In terms of structure, the fact that
the fundamental issue of what specific mechanisms certain neuropeptides share specific amino acid
are associated with edema development after TBI. sequences allows this vast collection of molecules
Recent studies have suggested that neuropeptides, to be sorted into families, such as the tachykinin
and in particular SP, may play a critical role in family which includes SP, calcitonin gene-related
edema formation, not only in terms of vasogenic peptide (CGRP) and neurokinin A (NKA). The
edema associated with increased BBB permeabil- tachykinin peptide family certainly represents one
ity, but also in the later cytotoxic phase of edema of the largest peptide families described in animals
development (Nimmo et al., 2004). Its involvement (Severini et al., 2002).
in the pathophysiology of TBI therefore seems to
straddle both the enzymatic and osmotic subrou-
Synthesis
tines of injury.
The formation of the peptide bonds of neuropep-
tides necessitates that they be synthesised on ribo-
Substance P
somes, structures present exclusively in the cell
body. It is common for the mRNA encoding
SP was first identified in the early part of 1930
neuropeptides to initially be translated into a
(Von Euler and Gaddum, 1931), initially as a crude
larger protein precursor. Two genes exist, the
extract isolated from equine brain and gut. The
preprotachykinin A (PPTA) gene and PPTB gene.
letter P derives from the ‘powder’ they extracted
The PPTA gene can express four different forms of
that contained the active substance. It was found to
mRNA through alternative splicing, two of which
have potent hypotensive and smooth muscle
(the b and g forms) encode synthesis of both SP
contractile properties (Von Euler and Gaddum,
and NKA. Expression of SP and its mRNA is
1931), and was identified in high concentrations in
widely abundant in both CNS and PNS (Harrison
the dorsal root of the spinal cord, leading to the
and Geppetti, 2001). aPPTA expression is more
proposal that it was a neuronal sensory transmitter
abundant in the brain, whilst bPPT-A and gPPTA
associated with pain transmission (Lembeck,
mRNAs predominate in peripheral tissues (Kotani
1953). Today, it is accepted that SP is released
et al., 1986). The b and g forms of PPTA mRNA
from both central and peripheral endings of pri-
also encode synthesis of neuropeptide K (NPK)
mary afferent neurons and functions as a neuro-
and neuropeptide g (NPg), which are elongated
transmitter (Otsuka and Yoshioka, 1993).
forms of NKA. However their function has not
Susan Leeman and colleagues (Chang et al.,
been fully elucidated. The PPTB gene gives rise to
1971) identified SP as an undecapeptide in the early
neurokinin B (NKB) (Hokfelt et al., 2001).
1970s, and were the first to synthesise the com-
pound (Tregear et al., 1971) and set up radio-
immunoassays (Powell et al., 1973). Such advances Localisation
allowed the effects of SP to be tested in physiolog-
ical models (Takahashi et al., 1974; Henry, 1976), SP immunoreactivity has been demonstrated in
using antibodies to monitor SP by radioimmuno- the rhinencephalon, telencephalon, basal ganglia,
assay and immunohistochemistry (Hokfelt et al., hippocampus, amygdala, septal areas, diencepha-
1975; Nilsson et al., 1975; Takahashi and Otsuka, lon, hypothalamus, mesencephalon, metencepha-
1975; Cuello and Kanazawa, 1978; Ljungdahl et al., lon, pons, myelencephalon and spinal cord
1978; Costa et al., 1980; Schultzberg et al., 1980) (Shults et al., 1984). Nerve fibres containing SP-
and demonstrating controlled neuronal release like immunoreactivity are common in most
(Otsuka and Konishi, 1976; Olgart et al., 1977). autonomic ganglia (Helke et al., 1982; Helke and
It’s role as a neurotransmitter was subsequently Phillips, 1988; Bergner et al., 2000), and SP
100

immunoreactivity has also been described in trige- cleavage of SP in vivo (Nadel, 1991). NEP has
minal and dorsal root ganglia (Lee et al., 1985; been demonstrated to metabolise SP in the brain
Gibbins et al., 1987) and intrinsic neurons of the (Hooper and Turner, 1987), spinal cord (Sakurada
gut (Sternini et al., 1995). In the autonomic ganglia et al., 1990) and peripheral tissues (Di Maria et al.,
SP is thought to play a modulatory role, the best 1998), while ACE has been reported to degrade SP
characterised response being observed in guinea pig in plasma (Wang et al., 1991), CSF and substantia
inferior mesenteric ganglion. In these neurons SP nigra. ACE has also been shown to contribute to
mimics a slow depolarisation, which can be evoked the degradation of fragments released from NEP.
by repetitive afferent nerve stimulation (Dun and Both NEP and ACE catalyse the hydrolysis bonds
Minota, 1981). Peripheral inflammation also leads of SP, leaving the peptide lacking the carboxyl
to an increase in SP immunoreactivity within the terminal regions required to bind to the tachykinin
superficial spinal cord (Marlier et al., 1991) and receptors (Skidgel and Erdos, 1987).
increased SP release (Schaible et al., 1990). Acti-
vation or damage to neurons leads to changes
Receptors
in neuropeptide biosynthesis that results from
induction of neuropeptide gene expression
The biological actions of SP are mediated by
(Hokfelt et al., 1994). Specifically, the expression
tachykinin (neurokinin: NK) receptors (Harrison
of PPT mRNA (Noguchi et al., 1988) and
and Geppetti, 2001), rhodopsin-like membrane
SP (NK1) receptor mRNA (McCarson, 1999) are
structures consisting of seven hydrophobic trans-
upregulated in the periphery during noxious stim-
membrane domains, connected by extracellular
ulation or neurogenic inflammation (Harrison and
and intracellular loops and coupled to G-proteins
Geppetti, 2001).
(Nakanishi, 1991; Gerard et al., 1993; Maggi and
SP is released from its precursor by the actions
Schwartz, 1997). There are three types of mam-
of proteases called convertases. Cleavage points
malian tachykinin receptors that have been cloned:
for the convertases on the PPT gene are doublets
NK1, NK2 and NK3 exhibiting preferences for SP,
of cationic residues (Harrison and Geppetti, 2001).
NKA and NKB, respectively (Regoli et al., 1994).
After release, the only mechanisms to terminate
Endogenous tachykinins are not highly selective
the action of neuropeptides are diffusion away
for any given receptor, and may act on all three
from the receptor site or degradation by extracel-
receptors with varying affinities under certain
lular peptidases. The slow nature of these proc-
conditions such as receptor availability or high
esses accounts for the prolonged effects of
peptide concentrations. For this reason SP acti-
neuropeptides (Kandel and Squire, 2000).
vates not only NK1 receptors, but also NK2
and NK3 receptors in a number of tissues (Regoli
et al., 1994).
Metabolism

Enzymes involved in metabolising SP include neu- Functions


tral endopeptidase (NEP) (Matsas et al., 1984),
SP-degrading enzyme (Probert and Hanley, 1987), SP has been implicated in memory and reinforce-
angiotensin-converting enzyme (ACE) (Skidgel ment processes. The amino terminus (NH2) of SP
and Erdos, 1987), dipeptidyl aminopeptide IV has been found to be involved in the memory pro-
(Heymann and Mentlein, 1978), post-proline end- moting effects of SP while the carboxy terminus
opeptidase (Blumberg et al., 1980), cathepsin-D (COOH) is involved in reinforcing properties. An
(Azaryan and Galoyan, 1988) and cathepsin-E NK1 antagonist (WIN 51708) was found to block
(Kageyama, 1993). While all of these enzymes these actions, indicating that the behavioural effects
are known to cleave SP in vitro, their individual of SP are mediated by NK1 receptor (Hasenohrl
cellular localisation suggests that NEP and/or et al., 2000). SP is also expressed widely in areas of
ACE are most likely to be involved in the the brain involved in fear producing pathways,
101

including the amygdala, septum, hippocampus, dorsal root ganglia. The concept of neurogenic
hypothalamus and periaqueductal gray (Rupniak inflammation has since evolved to encompass
and Kramer, 1999), and accordingly is released in vasodilatation, plasma extravasation and neuron-
response to aversive stimuli. As expected, injection al hypersensitivity caused by the release of neuro-
of SP into regions such as the periaqueductal gray peptides, including SP and CGRP, from sensory
modulates defensive reactions, while administration neurons (Black, 2002). The effects of sensory ne-
of an NK1 antagonist inhibits long-lasting audible uropeptides are particularly prominent at the level
vocalisation, which is a sign of anxiety and fear of the vasculature where they cause vasodilation
(Severini et al., 2002). Intracerebroventricular of arterioles, plasma protein extravasation in
injection of SP results in many diverse effects in post-capillary venules and leukocyte adhesion to
rodents including increased blood pressure and endothelial cells of venules (Geppetti et al., 1995).
heart rate, increased hindlimb rearing behaviour, Additional tissue-specific responses produced by
scratching, skin biting and grooming. neurogenic inflammation include smooth muscle
In bronchial smooth muscle, activation of sen- relaxation/contraction in the urinary bladder, ure-
sory neurons leads to vasodilation and increased ter and iris, inotropic and chronotropic effect on
vascular permeability, effects that are abolished by the heart and bronchoconstriction in the airways
pretreatment with capsaicin, an agent that causes amongst others (Geppetti et al., 1995).
neuropeptide depletion, or by treatment with a SP Peptide-containing primary sensory neurons are
antagonist. Histamine antagonists also blocked characterised by their unique sensitivity to caps-
nearly all of the stimulatory effects of SP. There- aicin, the pungent ingredient found in capsicum
fore, SP containing neurons play a major role in (Szallasi and Blumberg, 1999). The recent cloning
the local regulation of airway resistance and in- of the channel operated by capsaicin, the transient
terstitial fluid transfer in various pathological con- receptor potential vanilloid receptor-1 (TRPVR-1)
ditions (Lundberg et al., 1983). (Caterina et al., 1997), has clarified the molecular
basis of the selective action of capsaicin on sensory
neurons. This seven transmembrane domain pro-
Trigeminovascular system
tein is a non-selective cation channel, whose en-
dogenous stimulants include heat (>431C) and
Cerebral blood vessels are innervated by a combi-
protons. Subsets of primary sensory neurons are
nation of sympathetic, parasympathetic and trige-
stimulated selectively by capsaicin, causing the
minal somatic nerve fibres all of which are
release of sensory neuropeptides and promoting
important in cerebrovascular regulation (Atalay
neurogenic inflammation. At higher concentra-
et al., 2002). The trigeminal component of this in-
tions capsaicin kills neurons, blocking the genesis
nervation is commonly referred to as the trige-
of subsequent neurogenic inflammatory responses
minovascular system. This system has been shown
(Szallasi and Blumberg, 1999). These neurons are
to transmit pain sensation from the dura mater
defined as ‘‘capsacin-sensitive’’ due to the specific
and cranial vessels (Huber, 1899; Penfield, 1932,
excitatory/desensitisation effect of capsaicin
1934, 1940; Feindel et al., 1960). The perivascular
(Szolcsanyi and Mozsik, 1984).
endings of trigeminovascular fibres contain several
neurotransmitters including SP (Edvinsson et al.,
1989), CGRP (Uddman et al., 1985; McCulloch
Edema
et al., 1986), NKA (Edvinsson et al., 1988), nitric
oxide and amylin (Edvinsson et al., 2001).
Of all the secondary injury factors involved in
the development of neuronal dysfunction, edema
Neurogenic inflammation formation is thought to be central to outcome fol-
lowing injury (Lobato et al., 1988; Sarabia et al.,
Bayliss (1901) initially described vasodilatation of 1988). This is particularly the case in younger vic-
lower limb vessels following stimulation of the tims of TBI where formation of edema within the
102

brain has been found to be responsible for 50% of


all death and disability (Feickert et al., 1999). The
mechanisms associated with cerebral edema for-
mation remain largely unknown, however, inves-
tigation of peripheral tissues (Woie et al., 1993)
has demonstrated an association between neuro-
peptides, development of increased vascular per-
meability and subsequent edema formation.
Termed neurogenic inflammation, the process in-
volves the stimulation of the slow velocity C-fibres
(nociceptors), initiating the release of neuropep-
tides (Woie et al., 1993) including SP, CGRP and
NKA, which produce vasodilation, edema and Fig. 1. Alterations in brain water content at 5 h following
tissue swelling. While SP has been recognised as diffuse traumatic brain injury in rats. Vehicle treated controls
being primarily associated with increased vascular showed significantly more (po0.001) edema than either sham
permeability, SP is stored and co-released from or capsaicin pre-treated animals.
sensory nerve endings with CGRP, which is a
most potent endogenous vasodilator, and displays Yonehara et al., 1987; De and Ghosh, 1990; Laird
potent edema producing activity in the presence of et al., 2000). It is also well known that SP is the
SP (Severini et al., 2002). neuropeptide responsible for increased vascular
When left unchecked cerebral edema results in permeability, whereas CGRP is primarily associ-
an increase in ICP that may lead to a decrease in ated with vasodilation. Finally, Kramer et al.
tissue perfusion, localised hypoxia and ischemia, (2003) have demonstrated in studies of cardiac is-
and in severe cases brain herniation and death. It is chaemia that SP release is increased with magne-
therefore of paramount importance that edema sium depletion; declines in magnesium
genesis is inhibited following TBI. Whilst a concentration have been widely described follow-
number of studies have investigated the role of ing TBI (Vink et al., 1987, 1988). Therefore it is
classical inflammation in edema formation follow- feasible to propose that drugs acting on SP recep-
ing TBI (Stahel et al., 1998; Lenzlinger et al., 2001; tors, and in particular NK1 receptor (Hokfelt
Stein and Hoffman, 2003; Besson et al., 2005), to et al., 2001), may be beneficial in disease treat-
date only one study has examined the role of ne- ment. Studies of neurogenic inflammation follow-
urogenic inflammation post-trauma. Nimmo et al. ing acute brain injury have provided evidence
(2004) demonstrated that capsaicin administered supporting such a possibility.
prior to TBI significantly attenuated BBB opening,
edema formation and the development of both
motor and cognitive deficits. The authors con- Acute CNS injury
cluded that a capsaicin-induced depletion of ne-
uropeptides clearly prevented the development of Virtually all blood vessels of the body are sur-
neurogenic inflammation, and inhibited develop- rounded by sensory nerve fibres that contain both
ment of vasogenic edema (Fig. 1). Although, this CGRP and SP. Cerebral arteries, in particular,
study demonstrated a role for neurogenic inflam- appear to receive a dense supply of these neurones,
mation in TBI, the identification of which neuro- and it is therefore consistent that these neurones
peptide is primarily involved in the formation of have a role as mediators of the inflammatory
increased BBB permeability and edema formation process following injury. Studies of migraine
was not established. (Ferrari, 1998) have indeed demonstrated that
Previous studies of peripheral edema have neuropeptides are a therapeutic target to reduce
shown that SP is the neuropeptide most closely vascular permeability. However, only a limited
associated with capsaicin sensitivity (Saria, 1984; number of studies have demonstrated that
103

neurogenic inflammation may play a role in acute example, in a rodent cerebral stroke model, the
injury. Significant amounts of SP release has been NK1 receptor antagonist (SR140333) reduced inf-
detected in the nervous system following both pe- arct volume after focal ischaemia, implying that
ripheral nerve injury (Malcangio et al., 2000), that SP might play a role in exacerbating ischaemic
traumatic spinal cord injury (Sharma et al., 1990) damage (Yu et al., 1997). However, despite these
and more recently, in vitro studies of endothelium positive findings there has been no further work
stimulation (Annunziata et al., 2002). In our own published in this area. Interestingly, serum levels
TBI studies, perivascular SP immunoreactivity has of SP have been measured in humans with both
been shown to increase after TBI (Fig. 2), irre- transient ischaemic attack and complete stroke,
spective of the injury model (focal versus diffuse) and these have been found to be significantly el-
or severity of injury. In ischaemia, SP immunore- evated compared with controls (Bruno et al.,
activity has been shown to increase in GABAergic 2003). In other organs, post-ischaemic blockade
interneurons around regions of infarction, and of tachykinin receptors have been shown to inhibit
transiently expressed in cerebrovenular endotheli- vascular permeability, neutrophil recruitment,
um (Stumm et al., 2001). Our own studies have intestinal haemorrhage and neutropaenia follow-
shown increased SP immunoreactivity following ing ischaemia and reperfusion of the superior
ischaemic brain injury in the rat, which was asso- mesenteric artery in the rat (Souza et al., 2002).
ciated with a significant increase in edema forma- Similarly, SP antagonists reduced post-ischaemic
tion (Turner et al., 2006). With respect to the glial myocardial injury in rats with dietary Mg
response after acute injury, receptor-binding sites deficiency (Kramer et al., 1997), and the authors
for SP have been shown to increase on glia after suggest that SP may play an early critical role
neuronal injury (Mantyh et al., 1989). Because SP in inflammatory/pro-oxidant responses following
is known to regulate inflammatory and immune ischaemia. This finding is of particular interest to
responses in peripheral tissue, it therefore may TBI as traumatic injury produces sustained decline
regulate the glial response to injury. Subsequent in intracellular magnesium, implying that SP may
studies have confirmed that SP receptors are significantly contribute to the detrimental effects
expressed on astrocytes after injury and may there- of magnesium deficiency (Heath and Vink, 1996).
fore be linked to their transformation to reactive
astrocytes (Lin, 1995). This increase was not ob-
served in undamaged areas. NK1 receptor antagonists
Several studies using NK1 receptor antagonists
also support a role for SP in neurogenic inflam- A number of groups have hypothesised that tachy-
mation following ischaemic injury, although few kinin receptor antagonists may have several ther-
have been applied to studies of the CNS. For apeutic applications (Watling, 1992; Lowe

Fig. 2. SP immunoreactivity at 5 h following diffuse traumatic brain injury in the rat in (A) sham, uninjured rat cortex and (B) injured
rat cortex. Note the qualitative increase in perivascular SP immunoreactivity (bar ¼ 100 mm).
104

et al., 1994; Rupniak et al., 2000). The notion of neurogenic inflammation, as well as improve motor
antagonising SP was first raised by Leban et al. and cognitive function after TBI. In guinea pig
(1979) when examining the effects of SP agonists skin, administration of the NK1 receptor antago-
in the guinea pig ileum. Subsequently, Folkers nist RP 67580 was able to inhibit SP-induced ed-
et al. (1981) discussed the chemical design of SP ema formation and white blood cell accumulation
antagonists, before Engberg et al. (1981) develop- (Campos and Calixto, 2000). In vitro studies of
ed the first synthetic peptide antagonist (D-Pro, endothelial injury have supported a potential role
D-Trp)-SP for use in the CNS, specifically the for SP antagonists in the treatment of BBB dys-
block of locus coeruleus (LC) neurones. However, function by demonstrating that such antagonists
the affinity and metabolic instability of peptides neutralised increased BBB permeability, upregula-
limited their usefulness for in vivo studies. It was tion of MHC-II molecules, reduced expression of
the development of the first non-peptide SP anta- ICAM-1 and prevented associated cell morpholog-
gonist by Snider et al. (1991), who showed that ical changes (Annunziata et al., 2002).
CP-96345 was a potent, competitive and highly
selective antagonist of the NK1 receptor, that ini-
tiated a new wave of interest in these compounds. Conclusion
This antagonist was further refined to create the
highly potent n-acetyl-L-tryptophan benzyl esters While a role for neurogenic inflammation in vas-
(MacLeod et al., 1993, 1995), and it was the 3,5-bis cular permeability and edema formation has been
(trifluoromethyl) benzyl ester (L-732138) that led described in peripheral tissues for a number of
to the inhibition of SP-induced inositol phosphate years, few studies have examined the potential for
accumulation in Chinese hamster ovarian cells neurogenic inflammation to influence BBB perme-
expressing the human NK1 receptor. L-732138 ability and edema formation after traumatic brain
was then used as a starting point to identify high- injury. Those studies that have investigated a role
affinity SP receptor antagonists with improved in for neuropeptides in acute brain injury have dem-
vivo activity. Rupniak and Kramer (1999) first onstrated that inhibition of release attenuates BBB
described the efficacy of SP antagonists in the permeability and edema formation after injury,
treatment of experimental depression and emesis, and results in an associated improvement in func-
with the NK1 receptor antagonists being able to tional outcome. Immunohistochemistry studies
decrease anxiety (Santarelli et al., 2001) and de- have demonstrated that increased SP levels are
pression with fewer side effects than other drugs observed perivascularly, confirming a potential
of choice for the treatment of depression (Severini role in vasogenic edema formation. Given the
et al., 2002). Ranga and Krishnan (2002) subse- apparent lack of side effects from this class of
quently published the first use of the SP antagonist compound, and the potential to improve post-
MK-0869 in the treatment of clinical depression traumatic anxiety and depression, inhibition of the
and anxiety. NK1 receptor antagonists have also SP pathway using NK1 receptor antagonists is ex-
pected to provide a novel approach to the man-
been tested in dental pain, osteoarthritis, ne-
uropathic pain and migraine however no analge- agement of edema formation and improvement of
functional outcome after TBI.
sic effects have been reported in studies to date
(Hokfelt et al., 2001).
An alternative to inhibiting SP binding is to References
decrease neuropeptide release and depleting neuro-
peptide stores using the vanilloid agonist capsaicin Annunziata, P., Cioni, C., Santonini, R. and Paccagnini, E.
(Cadieux et al., 1986; Kashiba et al., 1997). Nimmo (2002) Substance P antagonist blocks leakage and reduces
activation of cytokine-stimulated rat brain endothelium.
et al. (2004) were able to demonstrate that neuro-
J. Neuroimmunol., 131: 41–49.
peptide-depletion prior to induction of TBI Atalay, B., Bolay, H., Dalkara, T., Soylemezoglu, F., Oge, K.
attenuated the development of BBB dysfunction and Ozcan, O.E. (2002) Transcorneal stimulation of trigemi-
and vasogenic edema formation associated with nal nerve afferents to increase cerebral blood flow in rats with
105

cerebral vasospasm: a noninvasive method to activate the Di Maria, G.U., Bellofiore, S. and Geppetti, P. (1998) Regu-
trigeminovascular reflex. J. Neurosurg., 97: 1179–1183. lation of airway neurogenic inflammation by neutral endo-
Azaryan, A.V. and Galoyan, A.A. (1988) Substrate specificity peptidase. Eur. Respir. J., 12: 1454–1462.
of cerebral cathepsin D and high-Mr aspartic endopeptidase. Dietrich, W.D., Alonso, O. and Halley, M. (1994) Early
J. Neurosci. Res., 19: 268–271. microvascular and neuronal consequences of traumatic
Bayliss, W.M. (1901) On the origin from the spinal cord of the brain injury: a light and electron microscopic study in rats.
vaso-dilator of the hindlimb, and on the nature of these fib- J. Neurotrauma, 11: 289–301.
ers. J. Physiol. (Lond.), 32: 1025–1043. Dun, N.J. and Minota, S. (1981) Effects of substance P on
Bergner, A.J., Murphy, S.M. and Anderson, C.R. (2000) After neurones of the inferior mesenteric ganglia of the guinea-pig.
axotomy, substance P and vasoactive intestinal peptide ex- J. Physiol., 321: 259–271.
pression occurs in pilomotor neurons in the rat superior cer- Edvinsson, L., Brodin, E., Jansen, I. and Uddman, R. (1988)
vical ganglion. Neuroscience, 96: 611–618. Neurokinin A in cerebral vessels: characterization, localiza-
Besson, V.C., Chen, X.R., Plotkine, M. and Marchand- tion and effects in vitro. Regul. Pept., 20: 181–197.
Verrecchia, C. (2005) Fenofibrate, a peroxisome prolifera- Edvinsson, L., Ekman, R. and Thulin, T. (1989) Reduced levels
tor-activated receptor alpha agonist, exerts neuroprotective of calcitonin gene-related peptide (CGRP) but not substance
effects in traumatic brain injury. Neurosci. Lett., 388: 7–12. P during and after treatment of severe hypertension in man.
Black, P.H. (2002) Stress and the inflammatory response: a re- J. Hum. Hypertens, 3: 267–270.
view of neurogenic inflammation. Brain Behav. Immun., 16: Edvinsson, L., Elsas, T., Suzuki, N., Shimizu, T. and Lee, T.J.
622–653. (2001) Origin and co-localization of nitric oxide synthase,
Blumberg, S., Teichberg, V.I., Charli, J.L., Hersh, L.B. CGRP, PACAP, and VIP in the cerebral circulation of the
and Mckelvy, J.F. (1980) Cleavage of substance P to an rat. Microsc. Res. Tech., 53: 221–228.
N-terminal tetrapeptide and a C-terminal heptapeptide by a Engberg, G., Svensson, T.H., Rosell, S. and Folkders, K. (1981)
post-proline cleaving enzyme from bovine brain. Brain Res., A synthetic peptide as an antagonist of substance P. Nature,
192: 477–486. 293: 222–223.
Bruno, G., Tega, F., Bruno, A., Graf, U., Corelli, F., Molfetta, Engel, S., Schluesener, H., Mittelbronn, M., Seid, K., Adjodah,
R. and Barucco, M. (2003) The role of substance P in cere- D., Wehner, H.D. and Meyermann, R. (2000) Dynamics of
bral ischemia. Int. J. Immunopathol. Pharmacol., 16: 67–72. microglial activation after human traumatic brain injury are
Bullock, R., Maxwell, W.L., Graham, D.I., Teasdale, G.M. and revealed by delayed expression of macrophage-related proteins
Adams, J.H. (1991) Glial swelling following human cerebral MRP8 and MRP14. Acta Neuropathol. (Berl.), 100: 313–322.
contusion: an ultrastructural study. J. Neurol. Neurosurg. Feickert, H.J., Drommer, S. and Heyer, R. (1999) Severe head
Psychiatry, 54: 427–434. injury in children: impact of risk factors on outcome.
Cadieux, A., Springall, D.R., Mulderry, P.K., Rodrigo, J., J. Trauma, 47: 33–38.
Ghatei, M.A., Terenghi, G., Bloom, S.R. and Polak, J.M. Feindel, W., Penfield, W. and Mc, N.F. (1960) The tentorial
(1986) Occurrence, distribution and ontogeny of CGRP nerves and localization of intracranial pain in man. Neuro-
immunoreactivity in the rat lower respiratory tract: effect of logy, 10: 555–563.
capsaicin treatment and surgical denervations. Neuroscience, Ferrari, M.D. (1998) Migraine. Lancet, 351: 1043–1051.
19: 605–627. Fleminger, S. and Ponsford, J. (2005) Long term outcome after
Campos, M.M. and Calixto, J.B. (2000) Neurokinin mediation traumatic brain injury. BMJ, 331: 1419–1420.
of edema and inflammation. Neuropeptides, 34: 314–322. Folkers, K., Horig, J., Rosell, S. and Bjorkroth, U. (1981)
Caterina, M.J., Schumacher, M.A., Tominaga, M., Rosen, Chemical design of antagonists of substance P. Acta Physiol.
T.A., Levine, J.D. and Julius, D. (1997) The capsaicin re- Scand., 111: 505–506.
ceptor: a heat-activated ion channel in the pain pathway. Gennarelli, T.A. and Graham, D.I. (1998) Neuropathology of
Nature, 389: 816–824. the head injuries. Semin. Clin. Neuropsychiatry, 3: 160–175.
Cervos-Navarro, J. and Lafuente, J.V. (1991) Traumatic brain Geppetti, P., Bertrand, C., Ricciardolo, F.L. and Nadel, J.A.
injuries: structural changes. J. Neurol. Sci., 103(Suppl): S3–S14. (1995) New aspects on the role of kinins in neurogenic in-
Chang, M.M., Leeman, S.E. and Niall, H.D. (1971) Amino- flammation. Can. J. Physiol. Pharmacol., 73: 843–847.
acid sequence of substance P. Nat. New Biol., 232: 86–87. Gerard, N.P., Bao, L., Xiao-Ping, H. and Gerard, C. (1993)
Costa, M., Cuello, A.C., Furness, J.B. and Franco, R. (1980) Molecular aspects of the tachykinin receptors. Regul. Pept.,
Distribution of enteric neurons showing immunoreactivity 43: 21–35.
for substance P in the guinea-pig ileum. Neuroscience, 5: Gibbins, I.L., Furness, J.B. and Costa, M. (1987) Pathway-
323–331. specific patterns of the co-existence of substance P, calcitonin
Cuello, A.C. and Kanazawa, I. (1978) The distribution of sub- gene-related peptide, cholecystokinin and dynorphin in neu-
stance P immunoreactive fibers in the rat central nervous rons of the dorsal root ganglia of the guinea-pig. Cell Tissue
system. J. Comp. Neurol., 178: 129–156. Res., 248: 417–437.
De, A.K. and Ghosh, J.J. (1990) Inflammatory responses in- Graham, D.I., Clark, J.C., Adams, J.H. and Gennarelli,
duced by substance P in rat paw. Indian J. Exp. Biol., 28: T.A. (1992) Diffuse axonal injury caused by assault. J. Clin.
946–948. Pathol., 45: 840–841.
106

Graham, D.I., Gennarelli, T.A. and McIntosh, T.K. (2002) Kageyama, T. (1993) Rabbit procathepsin E and cathepsin E.
Greenfield’s neuropathology. Arnold, New York. Nucleotide sequence of cDNA, hydrolytic specificity for bio-
Graham, D.I., Gentleman, S.M., Nicoll, J.A., Royston, M.C., logically active peptides and gene expression during devel-
Mckenzie, J.E., Roberts, G.W. and Griffin, W.S. (1996) opment. Eur. J. Biochem., 216: 717–728.
Altered beta-APP metabolism after head injury and its Kandel, E.R. and Squire, L.R. (2000) Neuroscience: breaking
relationship to the aetiology of Alzheimer’s disease. Acta down scientific barriers to the study of brain and mind.
Neurochir. Suppl., 66: 96–102. Science, 290: 1113–1120.
Harrison, S. and Geppetti, P. (2001) Substance P. Int. J. Bio- Kashiba, H., Ueda, Y. and Senba, E. (1997) Systemic capsaicin
chem. Cell Biol., 33: 555–576. in the adult rat differentially affects gene expression for ne-
Hasenohrl, R.U., Souza-Silva, M.A., Nikolaus, S., Tomaz, C., uropeptides and neurotrophin receptors in primary sensory
Brandao, M.L., Schwarting, R.K. and Huston, J.P. (2000) neurons. Neuroscience, 76: 299–312.
Substance P and its role in neural mechanisms governing Kotani, Y., Hirota, Y., Sugiyama, K., Joh, S., Shibutani, T.,
learning, anxiety and functional recovery. Neuropeptides, 34: Matsuura, H. and Inoki, R. (1986) Effects of noxious stimuli
272–280. and anesthetic agents on substance P content in rat central
Hausmann, R. and Betz, P. (2000) The time course of the vas- nervous system. Jpn. J. Pharmacol., 40: 143–147.
cular response to human brain injury: an immunohisto- Kramer, J.H., Mak, I.T., Phillips, T.M. and Weglicki, W.B.
chemical study. Int. J. Legal Med., 113: 288–292. (2003) Dietary magnesium intake influences circulating pro-
Hausmann, R., Kaiser, A., Lang, C., Bohnert, M. and Betz, P. inflammatory neuropeptide levels and loss of myocardial tol-
(1999) A quantitative immunohistochemical study on the erance to postischemic stress. Exp. Biol. Med. (Maywood),
time-dependent course of acute inflammatory cellular re- 228: 665–673.
sponse to human brain injury. Int. J. Legal Med., 112: Kramer, S.D., Jakits-Deiser, C. and Wunderli-Allenspach, H.
227–232. (1997) Free fatty acids cause pH-dependent changes in
Heath, D.L. and Vink, R. (1996) Traumatic brain axonal injury drug–lipid membrane interactions around physiological pH.
produces sustained decline in intracellular free magnesium Pharm. Res., 14: 827–832.
concentration. Brain Res., 738: 150–153. Laird, J.M., Olivar, T., Roza, C., De Felipe, C., Hunt, S.P. and
Helke, C.J., Neil, J.J., Massari, V.J. and Loewy, A.D. (1982) Cervero, F. (2000) Deficits in visceral pain and hyperalgesia
Substance P neurons project from the ventral medulla to the of mice with a disruption of the tachykinin NK1 receptor
intermediolateral cell column and ventral horn in the rat. gene. Neuroscience, 98: 345–352.
Brain Res., 243: 147–152. Leban, J., Rackur, G., Yamaguchi, I., Folkers, K., Bjorkroth,
Helke, C.J. and Phillips, E.T. (1988) Substance P antagonist- U., Rosell, S., Yanaihara, N. and Yanaihara, C. (1979) Syn-
induced spinal cord vasoconstriction: effects of thyrotropin- thesis of substance P analogs and agonistic and antagonistic
releasing hormone and substance P agonists. Peptides, 9: activities. Acta Chem. Scand. B, 33: 664–668.
1307–1315. Lee, Y., Kawai, Y., Shiosaka, S., Takami, K., Kiyama, H.,
Henry, J.L. (1976) Effects of substance P on functionally iden- Hillyard, C.J., Girgis, S., Macintyre, I., Emson, P.C. and
tified units in cat spinal cord. Brain Res., 114: 439–451. Tohyama, M. (1985) Coexistence of calcitonin gene-related
Heymann, E. and Mentlein, R. (1978) Liver dipeptidyl amino- peptide and substance P-like peptide in single cells of the
peptidase IV hydrolyzes substance P. FEBS Lett., 91: 360–364. trigeminal ganglion of the rat: immunohistochemical analy-
Hokfelt, T., Kellerth, J.O., Nilsson, G. and Pernow, B. (1975) sis. Brain Res., 330: 194–196.
Experimental immunohistochemical studies on the localiza- Lembeck, F. (1953) Central transmission of afferent impulses.
tion and distribution of substance P in cat primary sensory III. Incidence and significance of the substance P in the dorsal
neurons. Brain Res., 100: 235–252. roots of the spinal cord. Naunyn Schmiedebergs Arch. Exp.
Hokfelt, T., Pernow, B. and Wahren, J. (2001) Substance P: a Pathol. Pharmakol., 219: 197–213.
pioneer amongst neuropeptides. J. Intern. Med., 249: 27–40. Lenzlinger, P.M., Morganti-Kossmann, M.C., Laurer, H.L.
Hokfelt, T., Zhang, X. and Wiesenfeld-Hallin, Z. (1994) and McIntosh, T.K. (2001) The duality of the inflammatory
Messenger plasticity in primary sensory neurons following response to traumatic brain injury. Mol. Neurobiol., 24:
axotomy and its functional implications. Trends Neurosci., 169–181.
17: 22–30. Leon-Carrion, J., Dominguez-Morales Mdel, R., Barroso,
Holmin, S., Soderlund, J., Biberfeld, P. and Mathiesen, T. Y.M.J.M. and Murillo-Cabezas, F. (2005) Epidemiology
(1998) Intracerebral inflammation after human brain contu- of traumatic brain injury and subarachnoid hemorrhage.
sion. Neurosurgery, 42: 291–298; discussion 298–299. Pituitary, 8: 197–202.
Hooper, N.M. and Turner, A.J. (1987) Isolation of two differ- Lin, R.C. (1995) Reactive astrocytes express substance-P
entially glycosylated forms of peptidyl-dipeptidase A (angio- immunoreactivity in the adult forebrain after injury. Neuro-
tensin converting enzyme) from pig brain: a re-evaluation of report, 7: 310–312.
their role in neuropeptide metabolism. Biochem. J., 241: Ljungdahl, A., Hokfelt, T. and Nilsson, G. (1978) Distribution
625–633. of substance P-like immunoreactivity in the central nervous
Huber, G.C. (1899) Observations on the innervation of intra- system of the rat-I. Cell bodies and nerve terminals. Neuro-
cranial vessels. J. Comp. Neurol., 9: 1–34. science, 3: 861–943.
107

Lobato, R.D., Sarabia, R., Cordobes, F., Rivas, J.J., Adrados, cerebrovascular regulation. Proc. Natl. Acad. Sci. U.S.A., 83:
A., Cabrera, A., Gomez, P., Madera, A. and Lamas, E. 5731–5735.
(1988) Posttraumatic cerebral hemispheric swelling. Analysis McIntosh, T.K., Smith, D.H., Meaney, D.F., Kotapka, M.J.,
of 55 cases studied with computerized tomography. J. Neuro- Gennarelli, T.A. and Graham, D.I. (1996) Neuropathological
surg., 68: 417–423. sequelae of traumatic brain injury: relationship to neuro-
Lowe III, J.A., Drozda, S.E., Mclean, S., Bryce, D.K., Craw- chemical and biomechanical mechanisms. Lab. Invest., 74:
ford, R.T., Snider, R.M., Longo, K.P., Nagahisa, A. and 315–342.
Tsuchiya, M. (1994) Aza-tricyclic substance P antagonists. Nadel, J.A. (1991) Neutral endopeptidase modulates neuro-
J. Med. Chem., 37: 2831–2840. genic inflammation. Eur. Respir. J., 4: 745–754.
Lundberg, J.M., Martling, C.R., Saria, A., Folkers, K. and Nakanishi, S. (1991) Mammalian tachykinin receptors. Annu.
Rosell, S. (1983) Cigarette smoke-induced airway oedema Rev. Neurosci., 14: 123–136.
due to activation of capsaicin-sensitive vagal afferents and Nicoll, R.A., Schenker, C. and Leeman, S.E. (1980) Substance
substance P release. Neuroscience, 10: 1361–1368. P as a transmitter candidate. Annu. Rev. Neurosci., 3:
Macleod, A.M., Cascieri, M.A., Merchant, K.J., Sadowski, S., 227–268.
Hardwicke, S., Lewis, R.T., Macintyre, D.E., Metzger, J.M., Nilsson, G., Larsson, L.I., Hakanson, R., Brodin, E., Pernow,
Fong, T.M., Shepheard, S., et al. (1995) Synthesis and B. and Sundler, F. (1975) Localization of substance P-like
biological evaluation of NK1 antagonists derived from immunoreactivity in mouse gut. Histochemistry, 43: 97–99.
L-tryptophan. J. Med. Chem., 38: 934–941. Nimmo, A.J., Cernak, I., Heath, D.L., Hu, X., Bennett, C.J.
Macleod, A.M., Merchant, K.J., Cascieri, M.A., Sadowski, S., and Vink, R. (2004) Neurogenic inflammation is associated
Ber, E., Swain, C.J. and Baker, R. (1993) N-acyl-L-try- with development of edema and functional deficits following
ptophan benzyl esters: potent substance P receptor antago- traumatic brain injury in rats. Neuropeptides, 38: 40–47.
nists. J. Med. Chem., 36: 2044–2045. Noguchi, K., Morita, Y., Kiyama, H., Ono, K. and Tohyama,
Maggi, C.A. and Schwartz, T.W. (1997) The dual nature of the M. (1988) A noxious stimulus induces the preprotachykinin-
tachykinin NK1 receptor. Trends Pharmacol. Sci., 18: A gene expression in the rat dorsal root ganglion: a quanti-
351–355. tative study using in situ hybridization histochemistry. Brain
Malcangio, M., Ramer, M.S., Jones, M.G. and Mcmahon, S.B. Res., 464: 31–35.
(2000) Abnormal substance P release from the spinal cord O’Connor, C.A., Cernak, I. and Vink, R. (2003) Interaction
following injury to primary sensory neurons. Eur. J. Neuro- between anesthesia, gender, and functional outcome task
sci., 12: 397–399. following diffuse traumatic brain injury in rats. J. Neuro-
Mantyh, P.W., Johnson, D.J., Boehmer, C.G., Catton, M.D., trauma, 20: 533–541.
Vinters, V.H., Maggio, J.E., Too, H.P. and Vigna, S.R. Obrenovitch, T.P. and Urenjak, J. (1997) Is high extracellular
(1989) Substance P receptor binding sites are expressed by glutamate the key to excitotoxicity in traumatic brain injury?
glia in vivo after neuronal injury. Proc. Natl. Acad. Sci. J. Neurotrauma, 14: 677–698.
U.S.A., 86: 5193–5197. Oehmichen, M., Eisenmenger, W., Raff, G. and Berghaus, G.
Marlier, L., Poulat, P., Rajaofetra, N. and Privat, A. (1991) (1986) Brain macrophages in human cortical contusions as
Modifications of serotonin-, substance P- and calcitonin indicator of survival period. Forensic Sci. Int., 30: 281–301.
gene-related peptide-like immunoreactivities in the dorsal Oehmichen, M. and Raff, G. (1980) Timing of cortical contu-
horn of the spinal cord of arthritic rats: a quantitative sion. Correlation between histomorphologic alterations and
immunocytochemical study. Exp. Brain Res., 85: 482–490. post-traumatic interval. Z. Rechtsmed., 84: 79–94.
Marmarou, A., Fatouros, P.P., Barzo, P., Portella, G., Yoshi- Olgart, L., Gazelius, B., Brodin, E. and Nilsson, G. (1977) Re-
hara, M., Tsuji, O., Yamamoto, T., Laine, F., Signoretti, S., lease of substance P-like immunoreactivity from the dental
Ward, J.D., Bullock, M.R. and Young, H.F. (2000) Contri- pulp. Acta Physiol. Scand., 101: 510–512.
bution of edema and cerebral blood volume to traumatic Otsuka, M. and Konishi, S. (1976) Release of substance P-like
brain swelling in head-injured patients. J. Neurosurg., 93: immunoreactivity from isolated spinal cord of newborn rat.
183–193. Nature, 264: 83–84.
Matsas, R., Kenny, A.J. and Turner, A.J. (1984) The meta- Otsuka, M. and Takahashi, T. (1977) Putative peptide neuro-
bolism of neuropeptides. The hydrolysis of peptides, includ- transmitters. Annu. Rev. Pharmacol. Toxicol., 17: 425–439.
ing enkephalins, tachykinins and their analogues, by Otsuka, M. and Yoshioka, K. (1993) Neurotransmitter func-
endopeptidase-24.11. Biochem. J., 223: 433–440. tions of mammalian tachykinins. Physiol. Rev., 73: 229–308.
McCarson, K.E. (1999) Central and peripheral expression Penfield, W. (1932) Intracerebral vascular nerves. Arch. Neurol.
of neurokinin-1 and neurokinin-3 receptor and substance Psychiatry, 27: 30–44.
P-encoding messenger RNAs: peripheral regulation during Penfield, W. (1934) A contribution to the mechanism of intra-
formalin-induced inflammation and lack of neurokinin cranial pain. Proc. Assoc. Res. Nerv. Mental Dis., 15:
receptor expression in primary afferent sensory neurons. 399–415.
Neuroscience, 93: 361–370. Penfield, W. (1940) Dural headache and the innervation of the
McCulloch, J., Uddman, R., Kingman, T.A. and Edvinsson, L. dura mater. Arch. Neurol. Psychiatry, 44: 43–75.
(1986) Calcitonin gene-related peptide: functional role in Pernow, B. (1983) Substance P. Pharmacol. Rev., 35: 85–141.
108

Powell, D., Leeman, S., Tregear, G.W., Niall, H.D. and Potts distribution of substance P and substance P receptors in the
Jr., J.T. (1973) Radioimmunoassay for substance P. Nat. rat central nervous system. Peptides, 5: 1097–1128.
New Biol., 241: 252–254. Skidgel, R.A. and Erdos, E.G. (1987) Cleavage of peptide
Probert, L. and Hanley, M.R. (1987) The immunocytochemical bonds by angiotensin I converting enzyme. Agents Actions
localisation of ‘substance-P-degrading enzyme’ within the rat Suppl., 22: 289–296.
spinal cord. Neurosci. Lett., 78: 132–137. Snider, R.M., Longo, K.P., Drozda, S.E., Lowe III, J.A. and
Ranga, K. and Krishnan, R. (2002) Clinical experience with Leeman, S.E. (1991) Effect of CP-96,345, a nonpeptide sub-
substance P receptor (NK1) antagonists in depression. J. stance P receptor antagonist, on salivation in rats. Proc. Natl.
Clin. Psychiatry, 63(Suppl 11): 25–29. Acad. Sci. U.S.A., 88: 10042–10044.
Regoli, D., Boudon, A. and Fauchere, J.L. (1994) Receptors Souza, D.G., Mendonca, V.A., De, A.C.M.S., Poole, S. and
and antagonists for substance P and related peptides. Teixeira, M.M. (2002) Role of tachykinin NK receptors on
Pharmacol. Rev., 46: 551–599. the local and remote injuries following ischaemia and reper-
Reilly, P.L. (2001) Brain injury: the pathophysiology of the first fusion of the superior mesenteric artery in the rat. Br. J.
hours. ‘Talk and die revisited’. J. Clin. Neurosci., 8: 398–403. Pharmacol., 135: 303–312.
Rupniak, N.M., Carlson, E.C., Harrison, T., Oates, B., Seward, Stahel, P.F., Morganti-Kossmann, M.C. and Kossmann, T.
E., Owen, S., De Felipe, C., Hunt, S. and Wheeldon, A. (1998) The role of the complement system in traumatic brain
(2000) Pharmacological blockade or genetic deletion of sub- injury. Brain Res. Brain Res. Rev., 27: 243–256.
stance P (NK(1)) receptors attenuates neonatal vocalisation Stein, D.G. and Hoffman, S.W. (2003) Concepts of CNS plas-
in guinea-pigs and mice. Neuropharmacology, 39: ticity in the context of brain damage and repair. J. Head
1413–1421. Trauma Rehabil., 18: 317–341.
Rupniak, N.M. and Kramer, M.S. (1999) Discovery of the Sternini, C., Su, D., Gamp, P.D. and Bunnett, N.W. (1995)
antidepressant and anti-emetic efficacy of substance P recep- Cellular sites of expression of the neurokinin-1 receptor in the
tor (NK1) antagonists. Trends Pharmacol. Sci., 20: 485–490. rat gastrointestinal tract. J. Comp. Neurol., 358: 531–540.
Sakurada, T., Hara, A., Matsumura, H., Yamada, H., Stumm, R., Culmsee, C., Schafer, M.K., Krieglstein, J. and
Sakurada, S. and Kisara, K. (1990) A substance P analogue Weihe, E. (2001) Adaptive plasticity in tachykinin and tachy-
reduces amino acid contents in the rat spinal cord. Pharma- kinin receptor expression after focal cerebral ischemia is
col. Toxicol., 66: 75–76. differentially linked to gabaergic and glutamatergic cerebro-
Santarelli, L., Gobbi, G., Debs, P.C., Sibille, E.T., Blier, P., cortical circuits and cerebrovenular endothelium. J. Neuro-
Hen, R. and Heath, M.J. (2001) Genetic and pharmacologi- sci., 21: 798–811.
cal disruption of neurokinin 1 receptor function decreases Szallasi, A. and Blumberg, P.M. (1999) Vanilloid (capsaicin)
anxiety-related behaviors and increases serotonergic func- receptors and mechanisms. Pharmacol. Rev., 51: 159–212.
tion. Proc. Natl. Acad. Sci. U.S.A., 98: 1912–1917. Szolcsanyi, J. and Mozsik, G. (1984) Effects of capsaicin on the
Sarabia, R., Lobato, R.D., Rivas, J.J., Cordobes, F., Rubio, J., development of gastric mucosal damage by different necro-
Cabrera, A., Gomez, P., Munoz, M.J. and Madera, A. (1988) tizing agents and of gastric cytoprotection by PGI2
Cerebral hemisphere swelling in severe head injury patients. atropine and cimetidine on rats. Acta Physiol. Hung., 64:
Acta Neurochir. Suppl. (Wien.), 42: 40–46. 287–291.
Saria, A. (1984) Substance P in sensory nerve fibres contributes Takahashi, T., Konishi, S., Powell, D., Leeman, S.E. and
to the development of oedema in the rat hind paw after Otsuka, M. (1974) Identification of the motoneuron-depo-
thermal injury. Br. J. Pharmacol., 82: 217–222. larizing peptide in bovine dorsal root as hypothalamic sub-
Schaible, H.G., Jarrott, B., Hope, P.J. and Duggan, A.W. stance P. Brain Res., 73: 59–69.
(1990) Release of immunoreactive substance P in the spinal Takahashi, T. and Otsuka, M. (1975) Regional distribution of
cord during development of acute arthritis in the knee joint substance P in the spinal cord and nerve roots of the cat and
of the cat: a study with antibody microprobes. Brain Res., the effect of dorsal root section. Brain Res., 87: 1–11.
529: 214–223. Tregear, G.W., Niall, H.D., Potts Jr, J.T., Leeman, S.E. and
Schultzberg, M., Hokfelt, T., Olson, L., Alund, M., Nilsson, Chang, M.M. (1971) Synthesis of substance P. Nat. New
G., Terenius, L., Elde, R., Goldstein, M. and Said, S. (1980) Biol., 232: 87–89.
Substance P, VIP, enkephalin and somatostatin immunore- Turner, R.J., Blumbergs, P.C., Sims, N.R., Helps, S.C., Rod-
active neurons in intestinal tissue transplanted to the anterior gers, K.M. and Vink, R. (2006) Increased substance P
eye chamber. J. Auton. Nerv. Syst., 1: 291–303. immunoreactivity and edema formation following reversible
Severini, C., Improta, G., Falconieri-Erspamer, G., Salvadori, ischemic stroke. Acta Neurochir. Suppl., 96: 263–266.
S. and Erspamer, V. (2002) The tachykinin peptide family. Uddman, R., Edvinsson, L., Ekman, R., Kingman, T. and
Pharmacol. Rev., 54: 285–322. McCulloch, J. (1985) Innervation of the feline cerebral
Sharma, H.S., Nyberg, F., Olsson, Y. and Dey, P.K. (1990) vasculature by nerve fibers containing calcitonin gene-related
Alteration of substance P after trauma to the spinal cord: an peptide: trigeminal origin and co-existence with substance P.
experimental study in the rat. Neuroscience, 38: 205–212. Neurosci. Lett., 62: 131–136.
Shults, C.W., Quirion, R., Chronwall, B., Chase, T.N. and Vaz, R., Sarmento, A., Borges, N., Cruz, C. and Azevedo, I.
O’donohue, T.L. (1984) A comparison of the anatomical (1997) Ultrastructural study of brain microvessels in patients
109

with traumatic cerebral contusions. Acta Neurochir. (Wien.), and neurokinin A by plasma dipeptidyl(amino)peptidase IV,
139: 215–220. aminopeptidase M and angiotensin converting enzyme. Pep-
Vespa, P., Prins, M., Ronne-Engstrom, E., Caron, M., Shal- tides, 12: 1357–1364.
mon, E., Hovda, D.A., Martin, N.A. and Becker, D.P. (1998) Watling, K.J. (1992) Nonpeptide antagonists herald new era in
Increase in extracellular glutamate caused by reduced cerebral tachykinin research. Trends Pharmacol. Sci., 13: 266–269.
perfusion pressure and seizures after human traumatic brain Woie, K., Koller, M.E., Heyeraas, K.J. and Reed, R.K. (1993)
injury: a microdialysis study. J. Neurosurg., 89: 971–982. Neurogenic inflammation in rat trachea is accompanied by
Vink, R., McIntosh, T.K., Demediuk, P. and Faden, A.I. increased negativity of interstitial fluid pressure. Circ. Res.,
(1987) Decrease in total and free magnesium concentration 73: 839–845.
following traumatic brain injury in rats. Biochem. Biophys. Xiong, Y., Peterson, P.L., Muizelaar, J.P. and Lee, C.P. (1997)
Res. Commun., 149: 594–599. Amelioration of mitochondrial function by a novel antioxi-
Vink, R., McIntosh, T.K., Demediuk, P., Weiner, M.W. and dant U-101033E following traumatic brain injury in rats.
Faden, A.I. (1988) Decline in intracellular free Mg2+ is as- J. Neurotrauma, 14: 907–917.
sociated with irreversible tissue injury after brain trauma. Yonehara, N., Shibutani, T. and Inoki, R. (1987) Contribution
J. Biol. Chem., 263: 757–761. of substance P to heat-induced edema in rat paw. J. Pharma-
Von Euler, U.S. and Gaddum, J.H. (1931) An unidentified de- col. Exp. Ther., 242: 1071–1076.
pressor substance in certain tissue extracts. J. Physiol. Yu, Z., Cheng, G., Huang, X., Li, K. and Cao, X. (1997)
(Lond.), 72: 74–87. Neurokinin-1 receptor antagonist SR140333: a novel type
Wang, L.H., Ahmad, S., Benter, I.F., Chow, A., Mizutani, S. of drug to treat cerebral ischemia. Neuroreport, 8:
and Ward, P.E. (1991) Differential processing of substance P 2117–2119.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 8

Current concepts of cerebral oxygen transport and


energy metabolism after severe traumatic brain injury

B.H. Verweij1,, G.J. Amelink1 and J.P. Muizelaar2

1
Rudolf Magnus Institute of Neuroscience, Department of Neurosurgery, University Medical Center Utrecht,
Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
2
Department of Neurosurgery, University of California at Davis Medical Center, 4860 Y Street, Suite 3740, Sacramento,
CA 95817, USA

Abstract: Before energy metabolism can take place, brain cells must be supplied with oxygen and glucose.
Only then, in combination with normal mitochondrial function, sufficient energy (adenosine tri-phosphate
(ATP)) can be produced. Glucose is virtually the sole fuel for the human brain. The brain lacks fuel stores
and requires a continuous supply of glucose and oxygen. Therefore, continuous cerebral blood flow (CBF),
cerebral oxygen tension and delivery, and normal mitochondrial function are of vital importance for the
maintenance of brain function and tissue viability. This review focuses on three main issues: (1) Cerebral
oxygen transport (CBF, and oxygen partial pressure (PO2) and delivery to the brain); (2) Energy metabo-
lism (glycolysis, mitochondrial function: citric acid cycle and oxidative phosphorylation); and (3) The role
of the above in the pathophysiology of severe head injury. Basic understanding of these issues in the normal
as well as in the traumatized brain is essential in developing new treatment strategies. These issues also play
a key role in interpreting data collected from monitoring techniques such as cerebral tissue PO2, jugular
bulb oxygen saturation (SjvO2), near infra red spectroscopy (NIRS), microdialysis, intracranial pressure
monitoring (ICP), laser Doppler flowmetry, and transcranial Doppler flowmetry — both in the experi-
mental and in the clinical setting.

Keywords: traumatic brain injury; metabolism; mitochondrial function; mitochondria; lactate; cerebral
blood flow; ischemia

Cerebral oxygen transport electro-encephalographic slowing and at 20 ml/


100 g/min loss of consciousness occurs, but may be
Cerebral blood flow tolerated without long term functional conse-
quences. Below a CBF of 18 ml/100 g/min, ionic
Under normal conditions, the brain has critical homeostasis becomes jeopardized and neurons
thresholds for cerebral blood flow (CBF) as well as convert to anaerobic metabolism (Jones et al.,
oxygen tension (PO2). If CBF is reduced, neuronal 1981; Siesjo, 1992; Schroder et al., 1996). At a
events will result (Astrup, 1982). Normal CBF CBF of 10 ml/100 g/min, membrane integrity is
is 50 ml/100 g brain tissue/min (Sokoloff, 1960). lost and irreversible brain damage is inevitable.
If CBF is reduced to 25 ml/100 g/min there is Tissue infarction is related to CBF but is also time
dependent as shown in Fig. 1 (Jones et al., 1981).
Corresponding author. Tel.: +31-30-2507059; Arterial blood contains 13 vol% of O2, while
Fax: +31-30-2542100; E-mail: bverweij@umcutrecht.nl jugular venous blood contains 6.7 vol%, for an

DOI: 10.1016/S0079-6123(06)61008-X 111


112

Fig. 1. Schematic showing the relation between CBF and ischemic duration/tissue infarction. (Adapted with permission from Jones
et al., 1981.)

arterio venous difference of oxygen (AVDO2) of Another important factor to be considered is


(136.7) ¼ 6.3 vol% (ml of O2/100 ml of blood). carbon dioxide (CO2) reactivity: with hyperventi-
Knowing how much blood is flowing to the brain lation (resulting in low blood CO2 levels) cerebral
(50 ml/100 g brain tissue/min) and how much vasoconstriction occurs with an ensuing lower CBF
oxygen the brain extracts from this blood and higher AVDO2, whereas with high blood CO2
(AVDO2), one can calculate cerebral metabolic levels the reverse occurs (Muizelaar et al., 1991).
rate of oxygen (CMRO2): Autoregulation is fundamentally different from
CO2 reactivity: while in both metabolic and
CMRO2 ¼ CBF  AVDO2
pressure autoregulation vessel diameter changes
which is generally 3.2 ml (of O2/100 g of brain are compensatory responses to maintain a constant
tissue/min) (Gibbs et al., 1942). AVDO2, in CO2 reactivity the diameter changes
are primary and CBF and AVDO2 follow
passively. Thus, CO2 reactivity differs from any
Cerebrovascular reactivity before-mentioned type of autoregulation in that
AVDO2 changes. It appears not to be an adaptive
There are two circumstances in which the brain response of the brain to changing circumstances.
governs its own flow. In one, CBF changes
proportionally with changes in CMRO2 — meta-
bolic autoregulation. In the other, CBF remains Physiology of cerebral blood flow (CBF) and
constant despite changes in blood pressure or ICP cerebral blood volume (CBV)
(together CPP ¼ MABP  ICP, where CPP is
cerebral perfusion pressure and MABP is mean The factors governing CBF are expressed in the
arterial blood pressure) — pressure autoregulation Hagen–Poiseuille equation:
— and blood viscosity — viscosity autoregulation
(McHenry et al., 1974; Muizelaar et al., 1986; CPP  d 4
CBF ¼ k 
Muizelaar, 1989). 8lv
Metabolic-, pressure-, and viscosity-autoregula- where k is a constant, CPP cerebral perfusion
tion have in common that AVDO2 remains essen- pressure in turn defined by mean arterial blood
tially constant (under physiologic conditions), pressure (MABP) minus intracranial pressure
considering CMRO2 ¼ CBF  AVDO2. (ICP), d diameter of the blood vessel, l the length
113

of the blood vessel (which is practically constant), the tissue, cerebral tissue PO2 is best described as a
and v blood viscosity. The most powerful factor in continuum that can vary from 90 mm Hg very
this equation is vessel diameter. For instance, the close to capillaries to much less than 34 mm Hg in
maximum constriction that can be obtained by more distal regions (Zauner et al., 2002).
hyperventilation is 20% from normal baseline Reductions of partial pressure of arterial oxygen
(Kontos et al., 1977). This however, leads to a (PaO2) with normal rates of CBF also lead to func-
decrease in CBF of 60% from a normal value of tional deficits. A reduction of PaO2 to 65 mm Hg
50 ml/100 g/min to 20 ml/100 g/min. Practically all induces in humans an impaired ability to perform
of this diameter regulation takes place in the complex tasks. Short-term memory is impaired at
microcirculation, especially in the arterioles with a 55 mm Hg. A PaO2 of 30 mm Hg causes loss of
diameter of 300–15 m (Kontos et al., 1977, 1987). consciousness (Siesjö, 1978). In animal models,
The most intense changes in diameter and there- PaO2 reduction to 36 mm Hg cause intracellular
fore, cerebral blood volume occur in the microcir- acidosis, reductions in phosphocreatine (PCr) and
culation. Although it is unclear how much blood is ATP, and increases in intracellular lactate levels
to be found in this part of the cerebral circulation, it (Xiong et al., 1997). Normal human brain has a
is estimated to be one-third of 60 ml of total blood critical tissue PO2 between 15 and 20 mm Hg, below
volume in the brain, i.e., 20 ml under normal con- which infarction (depending on the duration) of
ditions (Muizelaar, 1989). With diameter ranging tissue may occur (Fleckenstein et al., 1990; Maas
between 80 and 160% of baseline, this translates et al., 1993; Meixenberger et al., 1993; Kiening
into volume ranging between 64 and 256% of the et al., 1996; van Santbrink et al., 1996; Wu and
baseline 20 ml; 13 ml with maximum vasoconstric- Saggau, 1997; Zauner et al., 1997; Van den Brink
tion, and 51 ml with maximal vasodilatation. et al., 2000).
Although many different factors are essential in Neuronal mitochondria require an intracellular
maintaining adequate CBF, it is suggested that PO2 of at least 1.5 mm Hg to maintain aerobic
local metabolic factors are of primary importance metabolism (Chance et al., 1973; Siesjö and Siesjö,
in the local tissue regulation. Under normal 1996). If cellular PO2 is low, the driving force to
circumstances, in areas of increased brain activity, deliver oxygen to the mitochondria is dramatically
vasoactive substances are released which alter reduced. The minimum tissue PO2 required to
vascular tone and local perfusion. Increased per- provide sufficient intracellular oxygen is unknown.
fusion then creates a washout effect, which leads to In addition, it has been proposed that the diffusion
reduction of perfusion. This feedback system distance for oxygen from the microvasculature
allows for the modification of CBF for short may increase after TBI due to astrocytic swelling,
periods during times of increased metabolic generalized tissue swelling, and tissue damage. In
requirements. Several metabolic factors play a role these situations the brain might require higher
in the autoregulation of CBF under normal con- tissue oxygen tensions to maintain sufficient tissue
ditions, such as CO2/H+ (pH), K+, adenosine, oxygenation (Zauner et al., 2002).
prostaglandins, and nitric oxide (NO), as well as
serotonin, histamine, neuropeptide Y, vasoactive
intestinal peptide, calcitonin generated peptide, Oxygen and hemoglobin
and others.
Flow of oxygen from the alveoli to the mitochon-
dria in the brain is dependent on hemoglobin
Brain tissue oxygen tension and PO2.
Once oxygen has diffused from the alveoli into
Under physiological conditions, a linear relation- pulmonary blood, it is transported by hemoglobin
ship exists between arterial PO2 and brain PO2 with to the cerebral tissue capillaries where it is released
arterial levels being 90 mm Hg and cerebrovenous for use, by mitochondria. The presence of hemo-
levels 35 mm Hg. Because oxygen is consumed in globin in the erythrocytes of the blood allows
114

Fig. 2. The oxygen–hemoglobin saturation curve, including the effect of hypothermia and hyperventilation. (Adapted with permission
from Guyton, 1986.)

transporting 30–100 times as much oxygen as could Cerebral energy metabolism


be transported simply in the form of dissolved
oxygen in blood without hemoglobin. The affinity Under normal circumstances, the brain requires
of oxygen for hemoglobin is best expressed by the large amounts of energy. Although the brain com-
oxygen–hemoglobin saturation curve as seen in prises only 2–3% of whole body weight, up to 20%
Fig. 2 (Guyton, 1986). This physiological system is of energy generated in the whole body is used by
unique in that it favors the binding of oxygen to the brain.
hemoglobin in the lungs and the release of oxygen Fifty percent of the energy produced by the brain
in the periphery. In the lungs, the curve favors is needed for synaptic activity; 25% is used for
100% hemoglobin saturation at or around restoring ionic gradients across the cell membrane.
normal alveolar oxygen tensions, whereas in the The remaining energy is spent on biosynthesis
periphery the rate of oxygen delivery is propor- such as maintaining membrane integrity and other
tional to the difference in oxygen partial pressure processes. If the synthesis of ATP is insufficient,
(PO2) between capillary blood and the tissue cells. homeostatic mechanisms deteriorate, intracellular
Oxygen use, by the mitochondria, is responsible for concentration of calcium increases, and cell death is
creating the driving force for oxygen delivery. inevitable.
It is interesting to note the effect of temperature Most of the energy is consumed by the neurons.
and hyperventilation on tissue oxygenation. A Although glial cells account for almost half of the
decrease in temperature as well as hyperventilation brain volume, they have a much lower metabolic
(alkalosis), shifts P50 (i.e., the oxygen tension at rate and account for less than 10% of total cerebral
which hemoglobin is 50% saturated) to the left thus energy consumption (Siesjo, 1984).
reducing tissue oxygenation due to increased Hb-O2 Under normal conditions, almost all energy in
affinity and thus, decreased O2 unloading to tissues. our body is produced by aerobic metabolism
115

(Stryer, 1988). Krebs described three stages in the Glycolysis


generation of energy:
Figure 3b illustrates the steps in the glycolysis that
are carried out in the cytoplasm (Stryer, 1988;
– Large molecules in food are broken down into
Zauner et al., 2002). GLUT-1 transports glucose
smaller units. Proteins are hydrolyzed to
across the blood-brain barrier. GLUT-1 also
amino acids, polysaccharides are hydrolyzed
mediates uptake into the astrocyte while GLUT-3
to simple sugars such as glucose, and fats are
does the same for neurons. The expression of these
hydrolyzed to glycerol and fatty acids. No
GLUT transporters is up-regulated in experimental
useful energy is generated in this phase.
models of hypoxia. The latter results in increased
– These numerous small molecules are degraded
import of glucose for energy production.
to a few simple units that play a central role in
Glycolysis is regulated by the enzyme phospho-
metabolism. Most of them are converted in
fructokinase-1. Increased ATP demand will activate
the acetyl unit of acetyl co-enzyme A (acetyl
this enzyme by increasing cellular cAMP levels
CoA). A small amount of ATP is generated at
and thereby increasing the rate of glycolytic ATP
this stage.
generation.
– Acetyl-CoA brings acetyl units into the citric
If mitochondria become dysfunctional, even
acid cycle, where they are completely oxidized
after restoring blood flow, a small amount of
to CO2. Four pairs of electrons are transferred
ATP can still be formed by glycolysis because this
to NAD+ and FAD for each acetyl group
process does not require oxygen. In this process
that is oxidized. Then ATP is generated as
only a few percent of the total energy in the
electrons flow from the reduced forms of these
glucose molecule is released.
carriers to O2 during oxidative phosphorylat-
The law of mass action states that as the end
ion. Thus, most of the energy is generated in
products of a chemical reaction build up in the re-
the third stage.
acting medium the rate of the reaction approaches
zero, thus preventing further production of ATP.
The metabolic patterns of the brain are strik- The two end products in the glycolytic reactions
ingly different from other organs in their use of (pyruvate and hydrogen ions) are combined with
fuel to meet their energy needs (Guyton, 1986). NAD+ to form NADH and H+. The quantities of
Glucose is virtually the sole fuel for the human these end products increase and react with each
brain, except during prolonged starvation. The other to form lactic acid. This lactic acid can diffuse
brain lacks fuel stores and hence requires a con- readily into the extracellular fluids and even into
tinuous supply of glucose, which enters freely at all the intracellular fluids of other less active cells.
times. It consumes 120 g daily, which corres- Therefore, lactic acid represents an ‘‘escape’’ into
ponds to an energy input of 420 kcal. The brain which the glycolytic end products can be directed,
accounts for some 60% of utilization of glucose by thus allowing glycolysis to proceed far longer than
the whole body in resting state. During starvation, would be possible if the pyruvate and hydrogen
ketone bodies (acetoacetate and 3-hydroxybuty- were not removed from the reacting medium.
rate) partly replace glucose as fuel for the brain. Glycolysis could proceed for only seconds without
Acetoacetate is activated by the transfer of CoA this conversion. Instead, it can proceed for several
from succinyl CoA to give acetoacetyl CoA. minutes, supplying the body with ‘‘considerable’’
Cleavage by thiolase then yields two molecules quantities of ATP. In the human at rest, 5–10%
of acetyl CoA, which enter the citric acid cycle. of the glucose consumed by the body manifests as
Fatty acids do not serve as fuel for the brain a net output of lactate into blood (Siesjö, 1978;
because they are bound to albumin in plasma and Guyton, 1986; Sokoloff, 1989; Andersen and
so they do not traverse the blood-brain barrier. Marmarou, 1992).
In essence, ketone bodies are transportable Once pyruvate has been synthesized it can either
equivalents of fatty acids. reversibly be converted to lactate and accumulated,
116

Fig. 3. (a) Schematic showing glycolysis and Krebs cycle. (Adapted with permission from Magistretti et al., 1999; Zauner et al., 2002.)
(b) Schematic showing mitochondrial electron transport. (Adapted with permission from Magistretti et al., 1999; Zauner et al., 2002.)
(c) Schematic showing the Magistretti model of coupled metabolism. (Adapted with permission from Magistretti et al., 1999; Zauner
et al., 2002.)
117

converted to amino acid alanine, or enter the – Respiratory assemblies contain numerous
citric acid cycle to produce energy via oxidative electron carriers, such as the cytochromes.
phosphorylation. The step-by-step transfer of electrons from
NADH or FADH2 to O2 through these car-
riers leads to pumping of protons out of the
Citric acid cycle mitochondrial matrix. A proton-motive force
is generated consisting of a pH gradient and a
The citric acid cycle that takes place in the mi- transmembrane electric potential. ATP is
tochondria is shown in Fig. 3a (Stryer, 1988; synthesized when protons flow back to the
Zauner et al., 2002). Under aerobic conditions, the mitochondrial matrix through an F0F1 ATP
next step in the aerobic generation of energy from synthase complex. Thus oxidation and phos-
glucose is the oxidative decarboxylation of pyruv- phorylation are coupled by a proton gradient
ate to form acetyl CoA. This activated acetyl unit across the inner mitochondrial membrane.
is then completely oxidized to CO2 by the citric
acid cycle, a series of reactions that is also known Traditionally cerebral energy production has
as the tricarboxylic acid cycle or the Krebs cycle. been considered to consist mainly of aerobic
The citric acid cycle is the final common pathway metabolism of glucose. Although it has long been
for the oxidation of fuel molecules; it also serves as assumed that glia and neurons use glucose as their
a source of building blocks for biosynthesis. sole energy source, recent information has sug-
gested otherwise; astrocytes may have the ability
to transport glucose across the blood-brain barrier
Oxidative phosphorylation via GLUT-1 and anaerobically metabolize it to
lactate. Lactate is then released into the extracel-
Figure 3b shows the electron transport chain lular space, where it is taken up by neurons and
(Stryer, 1988; Zauner et al., 2002). The NADH consumed aerobically to generate energy as seen in
and FADH2 formed in glycolysis, fatty acid oxi- Fig. 3c (Vibulsreth et al., 1987; Walz and Mukerji,
dation, and the citric acid cycle are energy-rich 1988; Magistretti et al., 1999).
molecules because each contains a pair of electrons With increasing neuronal activity, potassium and
with a high transfer potential. These electrons are glutamate are released into the extracellular space
subsequently donated to molecular oxygen, result- and are taken up by the astrocytes in an energy-
ing in a large amount of free energy, which can be dependent fashion causing increased astrocytic
used to generate ATP. Oxidative phosphorylation glycolysis. In traumatic brain injury conditions,
is the process in which ATP is formed as electrons aerobic metabolism is diminished due to reductions
are transferred from NADH or FADH2 to O2 by a in cellular oxygen, or due to mitochondrial dys-
series of electron carriers. This process acts as a function, causing increased lactate accumulation.
major source of ATP in aerobic organisms. Some
salient features of this process are (Stryer, 1988):
Mitochondria
– Respiratory assemblies that are located in the
inner membrane of mitochondria carry out Mitochondria are oval-shaped organelles, typically
oxidative phosphorylation. The citric acid 2 mm in length and 0.5 mm in diameter. Techniques
cycle and the pathway of the fatty acid for isolating mitochondria were devised in the late
oxidation, which supply most of the NADH 1940s. Eugene Kennedy and Albert Lehninger sub-
and FADH2, are in the adjacent mitochon- sequently discovered that mitochondria contain the
drial matrix. respiratory assembly, the enzymes of both the citric
– The oxidation of NADH yields 3 ATP, acid cycle and fatty acid oxidation. Electron micro-
whereas the oxidation of FADH2 yields 2 scopic studies by George Palade and Fritjof
ATP. Oxidation and phosphorylation are Sjöstrand revealed that mitochondria have two
coupled processes. membrane systems: an outer membrane and a
118

cerebral oxygen transport and energy metabolism


will be mentioned: Traumatic intracranial hema-
tomas (intraparenchymal, subdural, and epidural)
are also common after head injury. Epidural hem-
atomas (in up to 5% of all patients admitted to
hospitals for head injury, and 9% of those with
severe head injury) are often the result of skull
fractures causing rupture of underlying arteries or
veins. If arterial in origin they can enlarge very
quickly and cause rapid neurological deterioration.
If surgical intervention is prompt, and no other
brain injury is present, outcome can be favorable.
Fig. 4. Schematic of mitochondrion.
Acute subdural hematoma (ASDH) occurs in up
to 25% of all patients with severe head injury
highly folded inner membrane. The inner mem- (Richards and Hoff, 1974; Hubschmann and
brane is folded into a series of internal ridges called Nathanson, 1985). In comatose patients with TBI,
cristae. Hence, there are two compartments in ASDH carries the highest mortality rate: 60–90
mitochondria: the intermembrane space between (Jamieson and Yelland, 1972; Gennarelli et al.,
the outer and inner membranes, and the matrix, 1989; Wilberger et al., 1991). In this group outcome
which is bounded by the inner membrane (Fig. 4). is strikingly unfavorable due to decreased energy
Oxidative phosphorylation takes place in the metabolism. On one hand, decreased energy
inner mitochondrial membrane, in contrast with metabolism is due to ischemia caused by increased
most of the reactions of the citric acid cycle and intracranial pressure and therefore decreased per-
fatty acid oxidation, which occur in the matrix. fusion pressure; on the other hand due to the un-
The outer membrane is quite permeable to most derlying damaged brain being unable to use oxygen
small molecules and ions because it contains many because of damaged mitochondria (Verweij et al.,
copies of porin, a transmembrane protein with a 2000b, 2001).
large pore. In contrast, the inner membrane is
intrinsically impermeable to nearly all ions and
polar molecules. Specific protein carriers transport Hypotension and hypoxia
molecules such as adenosine di-phosphate (ADP)
and long chain fatty acids across the inner mito- The majority of the potential clinical events after
chondrial membrane. neurotrauma have been investigated with respect to
their frequency of occurrence and impact on out-
come, both for the prehospital and intensive care
Pathophysiology unit (ICU) periods (Chesnut et al., 1993; Jones
et al., 1994). These studies have uniformly identi-
Traumatic brain damage injury may be divided fied hypotension (SABPo90 mm Hg) and hypoxia
into primary and secondary types of injury (PaO2o60 torr) as the most influential. These
(Verweij and Muizelaar, 1996). Mechanical forces parameters, amenable to therapeutic manipulation,
acting at the moment of injury damage the blood seem to be the most significant predictors of poor
vessels, axons, neurons, and glia of the brain outcome, independent of their etiologies and
initiating an evolving sequence of secondary pre-resuscitation of secondary insults.
changes that result in complex cellular, inflamma- Admission to the ICU does not eliminate
tory, neurochemical, and metabolic alterations. secondary brain injury. Jones et al. (1994) have
It is not within the scope of this review to reported the results of computerized online
describe all the changes occurring in the brain after evaluation of 14 variables in 124 head-injured
severe head injury; only those directly related to patients of a variety of grades admitted to the
119

neurosurgical ICU. More than one episode of vessel diameter is to decrease blood viscosity.
hypotension occurred in 73% of all patients, Again, this decrease itself leads to vasoconstriction
with median durations of 29 min (SABPo90 mm if viscosity autoregulation is intact, and it has been
Hg), 22 min (SABPo80 mm Hg), and 32 min argued that the viscosity lowering effect mediates a
(SABPo70 mm Hg). In 40% of all cases, more good deal of mannitol’s effect on ICP (besides the
than one episode of hypoxia occurred with an av- osmotic effect) (Muizelaar et al., 1983, 1984, 1986).
erage duration of 12 min (PaO2o60 torr), 19 min When viscosity autoregulation is not intact, lower-
(PaO2o52 torr), and 20 min (PaO2o45 torr). It ing viscosity with mannitol can maintain CBF de-
has also been demonstrated that these secondary spite cerebral vasoconstriction associated with
insults do not only occur in the ICU, but also hyperventilation (Cruz et al., 1990). As stated pre-
during patient transport in X-ray and in OR suites viously, CBV can vary between 13 and 51 ml in the
(Andrews et al., 1990). microcirculation. To comprehend what these differ-
ences in volume mean to ICP, one must consider the
pressure volume index (PVI) (Marmarou et al.,
ICP and cerebral circulation 1978). Pressure volume index is defined as the
amount of fluid (in ml) needed to add to the intra-
According to the Monro–Kellie doctrine, ICP is cranial space to make ICP rise tenfold (or withdraw
governed by the interplay between the volumes of to decrease ICP tenfold):
brain (including cytotoxic edema), cerebrospinal DV
fluid (including vasogenic or extracellular edema) PVI ¼
Log½ICPbefore =ICPafter 
and the blood within the cerebral blood vessels, all
of which is within the confines of the rigid skull Normal PVI is 20–25 ml (Shapiro et al., 1980).
(Monro, 1783; Kellie, 1824). An increase in vol- However PVI has been observed as low as 6 ml
ume in one of these compartments (or epidural, after severe head injury, which indicates that in
subdural, or intracerebral hematoma) leads to a going from normoventilation (PaCO2 ¼ 30 mm
rise in ICP, unless this increase is compensated by Hg) to strong hyperventilation (PaCO2 ¼ 18 mm
an equal decrease in one of the two remaining Hg) resulting in vasoconstriction, ICP could
compartments. The natural defense against rising theoretically be decreased tenfold (Bouma et al.,
ICP with brain swelling is the displacement of CSF 1992a). (That this is not always desirable may be
from the skull: hence small compressed ventricles clear from the following example: PaCO2 36 mm
and absence of basal cisterns on computer tomo- Hg, ICP 40 mm Hg, MABP 100 mm Hg, CBF
graphy (CT) scans after severe trauma. Sometimes 30 ml/100 g/min; AVDO2 6 vol% - CMRO2
this process can be palliated by drainage through a 1.8 ml/100 g/min; now with hyperventilation to
ventricular catheter. get ICP below the desired 20 mm Hg: PaCO2 ¼ 22,
When one considers the Hagen–Poiseuille equa- ICP 20 mm Hg, MABP 100 mg Hg - CPP from 60
tion, there are two practical methods of maintaining to 80 - CBF to 40 ml/100 g/min.) However, be-
CBF during vasoconstriction. The first is to increase cause of 20% vasoconstriction and diameter being
CPP, which can be done by raising the blood pres- to the fourth power in the Hagen–Poiseuille equa-
sure. When pressure autoregulation is intact, this tion, CBF drops to 16 ml — well below the thresh-
maneuver in and of itself will cause vasoconstric- old for infarction (Jones et al., 1981), and especially
tion, and this has occasionally been used to decrease so after severe head injury. Moreover, as AVDO2
ICP (Muizelaar, 1989). More important, however, cannot rise above 10, CMRO2 will drop to 1.6 ml/
is the need to avoid low blood pressure, and hence, 100 g/min.
the effect of ‘‘perfusion pressure therapy’’ may be If ICP is uncontrollable barbiturates are some-
due in part to the simple avoidance of arterial hy- times administered, lowering the cerebral meta-
potension (Rosner and Daughton, 1990; Bouma bolic demands. If metabolic autoregulation is still
et al., 1992a; Rosner et al., 1995). The second intact this will result in decreased blood volume
method to maintain CBF in the face of decreasing and therefore reduced ICP.
120

All of these examples are common in clinical Lactate/hyperglycolysis


practice, and thus it may be obvious that a good
monitor is required to guide the management of In animals and humans it has been repeatedly
ICP, blood pressure, ventilatory parameters, and shown that TBI induces increased brain lactate
blood viscosity. Although monitoring of CBF and/ production, which normalizes gradually after the
or AVDO2 is ideal, there are no practical ways to first few days in those who survive, but remains at
do this continuously; therefore the authors revert levels five to ten times normal in those who suc-
to a derivate of AVDO2, i.e., SjvO2 (Cruz, 1988; cumb. Microdialysis studies have demonstrated
Robertson et al., 1989; Gopinath et al., 1994). that extracellular fluid glucose declines to extremely
low levels when lactate is increasing (Goodman
et al., 1999; Zauner et al., 2002). If aerobic metab-
olism fails, anaerobic metabolism remains, resulting
Ischemia and CMRO2 in hyperglycolysis and lactate production. A shift to
anaerobic metabolism will occur if ischemia is
Secondary cerebral ischemia is very common after present. If brain oxygen tension levels fall below
severe head injury and is associated with an un- 20 mm Hg aerobic metabolism ceases to occur
favorable outcome (Graham and Adams, 1971; (Zauner et al., 1997). However, if mitochondrial
Bouma et al., 1991; Siesjö and Siesjö, 1996). As dysfunction occurs, aerobic metabolism will also
discussed earlier, CBF is closely regulated by a shift toward anaerobic metabolism inducing the
number of mechanisms. However, after trauma same phenomena (Verweij et al., 2000b).
these mechanisms can fail resulting in ischemia. In human positron emission tomography (PET)
This is especially important as the brain seems more studies, increases in glucose metabolism are demo-
vulnerable to ischemia after trauma and this vul- nstrated especially in the zone around contusions
nerability persists for at least 24 h (Jenkins, 1989). It and in the hemisphere underlying hematomas
has also been demonstrated that CBF is low in the (Bergsneider et al., 1997). Such tissues are often
hyperacute post-traumatic period (Bouma et al., adjacent to ‘‘low-density’’ cytotoxic edema areas
1991, 1992b). Increased intracranial pressure (as in on CT scan. Human PET studies at later time
cerebral edema and subdural hematoma) and there- points (1–4 weeks post-ictus) and mitochondrial
fore decreased cerebral perfusion pressure appears analyses in the acute stage have shown uniformly
also to be an important cause of ischemia as well as decreased metabolism, both for glucose and oxy-
too vigorous hyperventilation (Muizelaar et al., gen in humans (Verweij et al., 2000b, 2001).
1991; Verweij et al., 2001). Marmarou as well as others have shown through
Salvant and Muizelaar suggested that a parallel animal experiments that anaerobic cerebral metab-
reduction in CBF and CMRO2 without an increase olism with generation of lactate appears to occur
in AVDO2 is consistent with diminished metabo- even in the absence of blood flow limitation (De
lism and may be due to mitochondrial dysfunction. Salles et al., 1987). It has also been found by meas-
Verweij et al. (2000b) demonstrated mitochondrial uring brain oxygenation, CO2 generation, pH and
dysfunction in isolated mitochondria from human temperature in parallel with extracellular fluid lac-
tissue. In a normal coupled relationship between tate, and glucose levels measured by microdialysis
AVDO2 and CBF (AVDO2 ¼ CMRO2/CBF), that lactate generation is increased in 65% of
AVDO2 remains unchanged when the CMRO2 measurements, even in the presence of adequate
changes. If however CMRO2 remains constant, CBF and brain oxygen levels (Zauner et al., 1997).
changes in AVDO2 reflect uncoupled changes in Mitochondrial dysfunction will explain lactate
CBF (Robertson et al., 1989). If CBF decreases production in these circumstances (Verweij et al.,
following head injury, AVDO2 will increase as the 2000b).
brain compensates by extracting a greater amount High levels of ECF lactate might be harmful
of oxygen. A further uncompensated decline in to the injured brain. Marked cerebral acidosis
CBF leads to ischemia and a fall in CMRO2. may exacerbate calcium-mediated damage to
121

intracellular enzyme systems and may also inter- oxygen delivery to the brain and the extraction of
fere with ion-channel function (Siesjo, 1992). High oxygen by the brain (Cruz, 1988; Cruz et al., 1990;
tissue lactate levels could foster a decline in brain Gopinath et al., 1994); (2) local brain tissue oxime-
pH, as has been shown in numerous post- try by a single Clark-type electrode (or multi-
traumatic animal and human studies. electrode also measuring PCO2, pH, and tempera-
The restoration of circulation after ischemia is ture), which gives information of local cerebral
accompanied by normalization of the tissue lactate tissue PO2. The relationship between SjvO2, cerebral
concentration and the lactate-to-pyruvate ratio. tissue PO2, and CBF has been well documented.
Initially there is an increase in pyruvate concen- An increase in the concentration of tissue lactate
tration as lactate is converted back to pyruvate. in the brain indicates a shift from aerobic to an-
Animal studies have demonstrated that if the aerobic metabolism in an attempt to maintain ATP
resuscitation interval is prolonged, tissue lactate production. This shift occurs in case of low cere-
remains elevated during reperfusion, suggesting bral tissue PO2 that has been shown to occur in
that residual tissue lactic acidosis is a sign of 25–39% of patients with severe TBI in the first
mitochondrial dysfunction. Since mitochondrial 12 h post- injury. Low brain tissue PO2 also closely
dysfunction is reversible, therapeutic intervention correlates with low regional CBF. It has also been
might be possible (Verweij et al., 1997; Xiong shown that low brain tissue PO2 is strongly corre-
et al., 1998; Berman et al., 2000). lated with high levels of dialysate lactate in the
Rapid reversal of acidosis may be unfavorable, brain (Zauner et al., 2002). Increasing the concen-
as mild acidosis might be beneficial (pH paradox) tration of inspired oxygen to 100% has shown to
during recovery from hypoxia. There is little direct increase brain tissue PO2. In a study by Menzel
evidence to demonstrate that lactate alone, or et al. (1999), statistically significant correlation
substantial intracellular acidosis alone, is toxic to existed between brain tissue PO2 and CPP. Brain
normal cerebral tissues. This may be attributable lactate measured by microdialysis remained high
to preserved high-energy phosphate concentra- during the entire period rising later on. No corre-
tions, which allow potential intracellular buffering lations were found between ICP, CPP, brain tissue
and transport of hydrogen ions from the cell. PO2, or lactate (brain tissue PO2 increases over the
During ischemia, however, acidosis may injure first 30 h with an overshoot at 36–48 h). The latter
neurons by denaturation of proteins, lead to dam- could be explained by mitochondrial dysfunction
age of astrocytes owing to failure of membrane (Verweij et al., 2000b).
transport systems, and cause promotion of iron-
dependent free radical formation. These events can Mitochondrial function after severe head injury
cause the inhibition of glycolysis by the complete
inhibition of the glycolytic phosphofructokinase at Mitochondria play a vital role in cell survival and
a pH of 6.5 or below. tissue development by virtue of their role in energy
Mild acidosis might be protective in vitro and in metabolism and apoptosis (Nicholls and Budd,
vivo in models of ischemia/hypoxia, by slowing 2000; Friberg and Wieloch, 2002). Since neuronal
enzymatic processes and reducing energy consump- tissue stores and anaerobic glycolysis provides
tion and free radical production. Nonetheless, ATP sufficient to maintain cellular function for
tissue acidosis is consistently associated with only 1–2 min, mitochondrial generation of ATP is
worsened ischemic outcome in vivo, which may of vital importance (Siesjo, 1992). Neuronal mito-
be augmented by hyperglycemia. chondria have a high capacity to store calcium
ions, thereby protecting neurons against transient
Cerebral tissue PO2/SjvO2 elevations in intracellular calcium concentrations
during neuronal hyperactivity. This calcium se-
Cerebral oxygenation is currently monitored in two questration requires negative mitochondrial matrix
ways: (1) measurement of jugular bulb oxygen sat- potential and therefore full functional mito-
uration, which reflects the relationship between chondria with access to oxygen and pyruvate.
122

In the past, research efforts in patients with Abbreviations


severe head injury have focused on optimizing the
delivery of oxygen and glucose to the injured brain ADP adenosine diphosphate
in an attempt to maintain the ATP supply and to AMP adenosine monophosphate
avoid neuronal damage. However, limiting factors ASDH traumatic acute subdural hematoma
in synthesizing ATP are not only inadequate ATP adenosine triphosphate
delivery of oxygen and glucose but also impair- AVDO2 arteriovenous oxygen content
ment of mitochondrial function (Verweij et al., difference
1997, 2000b). Severe TBI with or without hypoxia CBF cerebral blood flow
and or ischemia results in a number of biochemical CBV cerebral blood volume
processes such as amino acid efflux and oxygen free CMRO2 cerebral metabolic rate of oxygen
radical production. This ultimately leads to mas- CoA co-enzyme A
sive ion shifts with increased calcium in the intra- CPP cerebral perfusion pressure
cellular compartment (Fineman et al., 1993). It has CSF cerebrospinal fluid
previously been demonstrated that experimental CT computerized tomography
TBI perturbs calcium homeostasis with an over- EEG electro-encephalogram
load of cytosolic calcium and excessive calcium FAD flavin adenine dinucleotide
adsorption by the mitochondrial membranes GCS Glasgow coma scale
(Sciamanna et al., 1992; Verweij et al., 1997; Xiong GLUT glucose transporter
et al., 1997). This inhibits mitochondrial function, ICP intracranial pressure
even when there are sufficient oxygen and substrate MABP mean arterial blood pressure
present. In rats, mitochondrial dysfunction begins NAD nicotinamide adenine dinucleotide
1 h after TBI and persists for at least 14 days, with NIRS near infra red spectroscopy
the maximum level of dysfunction occurring at NO nitric oxide
12–72 h (Verweij et al., 1997). The same phenom- PaCO2 arterial PCO2 tension
ena occur in patients (Verweij et al., 2000b). PaO2 arterial oxygen partial pressure
A number of critical mechanisms have been PET positron emission tomography
elucidated by which mitochondria are involved PO2 oxygen partial pressure
in cell death. Elevated cytosolic Ca2+ and oxida- PVI pressure volume index
tive stress both contribute to the opening of the SABP systolic arterial blood pressure
mitochondrial permeability transition pore (PTP), SjvO2 jugular venous oxygen saturation
which depolarizes the mitochondrion and leads to TBI traumatic brain injury
mitochondrial swelling and subsequent release of
cytochrome ‘‘c’’ from the intermembrane space.
Cytochrome ‘‘c’’ normally functions as part of the References
respiratory chain, but when released into the
cytosol (as a result of PTP opening) it becomes a Andersen, B.J. and Marmarou, A. (1992) Functional compart-
critical component of the apoptosis execution mentalization of energy production in neural tissue. Brain
Res., 585: 190–195.
machinery, where it activates caspases (cysteine
Andrews, P.J., Piper, I.R., Dearden, N.M., et al. (1990) Seco-
aspartate proteases) and (if ATP is available) ndary insults during intrahospital transport of head injured
causes apoptotic cell death (Young, 1992). Thus, a patients. Lancet, 335(8685): 327–330.
number of new targets for therapeutic intervention Astrup, J. (1982) Energy requiring cell functions in the ischemic
have emerged (Verweij et al., 1997, 2000a; Xiong brain: their critical supply and possible inhibition in protec-
tive therapy. J. Neurosurg., 56: 482–497.
et al., 1998; Berman et al., 2000). These pos-
Bergsneider, M., Hovda, D.A., Shalmon, E., et al. (1997) Cere-
sible interventions have recently been described bral hyperglycolysis following severe traumatic brain injury
in a well-written review by Merenda and in humans: a positron emission tomographic study. J.
Bullock (2006). Neurosurg., 86: 241–251.
123

Berman, R.F., Verweij, B.H. and Muizelaar, J.P. (2000) Gopinath, S.P., Robertson, C.S., Contant, C.F., et al. (1994)
Neurobehavioral protection by the neuronal calcium chan- Jugular venous desaturation and outcome after head injury.
nel blocker ziconotide in a model of traumatic diffuse brain J. Neurol. Neurosurg. Psychiatry, 57(6): 717–723.
injury in rats. J. Neurosurg., 93(5): 821–828. Graham, D.I. and Adams, J.H. (1971) Ischemic brain damage
Bouma, G.J., Muizelaar, J.P., Bandoh, K., et al. (1992a) Blood in fatal head injuries. Lancet, 1: 265–266.
pressure and intracranial pressure-volume dynamics in severe Guyton, A.C. (1986) Textbook of Medical Physiology (Seventh
head injury: relationship with cerebral blood flow. J. Neuro- edition). W.B. Saunders Company, Philadelphia, pp. 481–525.
surg., 77(1): 15–19. Hubschmann, O.R. and Nathanson, D.C. (1985) The role of
Bouma, G.J., Muizelaar, J.P., Choi, S.C., et al. (1991) Cerebral calcium and cellular membrane dysfunction in experimental
bloodflow and metabolism after severe traumatic brain injury: trauma and subarachnoid hemorrhage. J. Neurosurg., 6:
the elusive role of ischemia. J. Neurosurg., 75(5): 685–693. 698–703.
Bouma, G.J., Muizelaar, J.P., Stringer, W.A., et al. (1992b) Jamieson, K.G. and Yelland, J.D.N. (1972) Surgically treated
Ultra-early evaluation of regional cerebral blood flow in traumatic subdural hematomas. J. Neurosurg., 37: 137–149.
severely head-injured patients using xenon-enhanced Jones, P.A., Andrews, P.J., Midgley, S., et al. (1994) Measuring
computirized tomography. J. Neurosurg., 77: 360–368. the burden of secondary insults in head-injured patients dur-
van den Brink, W.A., van Santbrink, H., Steyerberg, E.W., ing intensive care. J. Neurosurg. Anesthesiol., 6(1): 4–14.
et al. (2000) Brain oxygen tension in severe head injury. Jones, T., Morawetz, R., Crowell, R.M., et al. (1981) Threshold
Neurosurgery, 46(4): 868–876. of focal cerebral ischemia in awake monkeys. J. Neurosurg.,
Chance, B., Oshimo, N., Sugamo, T. and Mayersky, A. (1973) 54: 773–782.
Basic principles of tissue oxygen determination from mito- Kellie, G. (1824) On death from cold, and on congestions of the
chondrial signals. In: Bruley D.F. (Ed.), Oxygen Transport to brain: an account of the appearances observed in the dissec-
Tissue. Plenum Publishing, New York, pp. 277–292. tion of two of three individuals presumed to have perished in
Chesnut, R.M., Marshall, S.B., Piek, J., et al. (1993) Early and the storm of 3rd November, 1821; with some reflections on
late systemic hypotension as a frequent source of cerebral the pathology of the brain. TRANS Med. Chir. Soc., Edin-
ischemia following severe brain injury in the traumatic coma burgh, 1: 84–169.
data bank. Acta Neurochir. Suppl. (Wein.), 59: 121–125. Kiening, K.L., Unterberg, A.W., Bardt, T.F., Schneider, G.H.
Cruz, J. (1988) Continuous versus serial global hemometabolic and Lanksch, W.R. (1996) Monitoring of cerebral oxygenation
monitoring: application in acute brain trauma. Acta in patients with severe head injuries. J. Neurosurg., 85: 751–757.
Neurochir. (Wien.), 42: 35–39. Kontos, H.A., Raper, A.J. and Patterson, J.L. (1977) Analysis
Cruz, J., Miner, M.E., Allen, S.J., et al. (1990) Continuous of vasoactivity of local pH, PCO2, and bicarbonate on pial
monitoring of cerebral oxygenation in acute brain injury: vessels. Stroke, 8(3): 358–360.
injection of mannitol during hyperventilation. J. Neurosurg., Kontos, H.A., Wei, E.P., Navari, R.M., et al. (1987) Responses
73(5): 725–730. of cerebral arteries and arterioles to acute hypotension and
De Salles, A.A.F., Muizelaar, J.P. and Young, H.F. (1987) hypertension. Am. J. Physiol., 234(4): H371–H383.
Hyperglycemia, cerebrospinal fluid lactic acidosis, and cere- Maas, A.I., Fleckenstein, W., de Jong, D.A. and van Santbrink,
bral blood flow in severely head injured patients. Neurosur- H. (1993) Monitoring cerebral oxygenation. Acta Neurochir.
gery, 21: 45–50. Suppl. (Wien.), 59: 50–57.
Fineman, I., Hovda, D.A. and Smith, M. (1993) Concussive brain Magistretti, P.J., Pellerin, L., Rothman, D.L. and Shulman,
injury is associated with a prolonged accumulation of calcium: R.G. (1999) Energy on demand. Science, 283: 496–497.
a 45Ca autoradiographic study. Brain Res., 624: 94–102. Marmarou, A., Shulman, K. and Rosende, R. (1978) A non-
Fleckenstein, W., Maas, A.I., Nollert, G. and de Long, D.A. linear analysis of the cerebrospinal fluid system and intra-
(1990) Oxygen pressure in cerebrospinal fluid. In: Flecken- cranial pressure dynamics. J. Neurosurg., 48: 332–344.
stein W. and Maas A.I. (Eds.), Oxygen Transport. Blackwell, McHenry Jr., L.C., West, J.W., Coopers, E.S., et al. (1974)
Berlin, pp. 368–393. Cerebral autoregulation in man. Stroke, 5(6): 695–706.
Friberg, H. and Wieloch, T. (2002) Mitochondrial permeability Meixenberger, J., Dings, J., Kuhnigk, H. and Roosen, K. (1993)
transition in neurodegeneration. Biochimie, 84: 241–250. Studies of tissue PO2 in normal and pathophysiological hu-
Gennarelli, T.A., Champion, H.R. and Sacco, W.J. (1989) man brain cortex. Acta Neurochir. Suppl. (Wien.), 59: 58–63.
Mortality of patients with head injury and extracranial injury Menzel, M., Doppenberg, E.M.R., Zauner, A., et al. (1999)
treated in trauma centers. J. Trauma, 29: 1193–1202. Increased inspired oxygen concentration as a factor in im-
Gibbs, E.L., Lennox, W.G., Nims, L.F., et al. (1942) Arterial proved brain tissue oxygenation and tissue lactate levels after
and cerebral venous blood: arterial venous differences in severe human head injury. J. Neurosurg., 91: 1–10.
man. J. Biol. Chem., 144: 325–332. Merenda, A. and Bullock, R. (2006) Clinical treatments for
Goodman, J.C., Valadka, A.B., Gopinath, S.P., Uzura, M. and mitochondrial dysfunction after brain injury. Curr. Opin.
Robertson, C.S. (1999) Extracellular lactate and glucose al- Crit. Care, 12: 90–96.
terations in the brain after head injury measured by micro- Monro, A. (1783) Observations on the structure and functions
dialysis. Crit. Care Med., 27: 1965–1973. of the nervous system. Creech and Johnson, Edinburgh, p. 5.
124

Muizelaar, J.P. (1989) Cerebral blood flow, cerebral blood Handbook of Physiology. Section 11: Neurophysiology,
volume, and cerebral metabolism after severe head injury. Vol. 3. American Physiological Society, Washington DC,
In: Becker D.P. and Gudeman S.K. (Eds.), Textbook of pp. 1843–1864.
Head Injury. W.B. Saunders Company, Philadelphia, PA, Sokoloff, L. (1989) Circulation and Energy Metabolism of the
pp. 221–240. Brain. Raven Press, New York, pp. 541–564.
Muizelaar, J.P., Lutz, H.A. and Becker, D.P. (1984) Effect of Stryer, L. (1988) Biochemistry (Third edition). W.H. Freeman
mannitol on ICP and CBF and correlation with pressure and Company, New York, pp. 313–544.
autoregulation in severely head-injured patients. J. Neuro- Verweij, B.H. and Muizelaar, J.P. (1996) Avoiding secondary
surg., 61(4): 700–706. brain injury after severe head trauma: monitoring and man-
Muizelaar, J.P., Marmarou, A., Ward, J.D., et al. (1991) agement. J. Craniomaxillofac. Trauma, 2(3): 8–17.
Adverse effects of prolonged hyperventilation in patients Verweij, B.H., Muizelaar, J.P. and Vinas, F.C. (2001) Hyper-
with severe head injury: a randomized clinical trial. J. Neuro- acute measurement of intracranial pressure, cerebral perfu-
surg., 75: 731–739. sion pressure, jugular venous oxygen saturation, and laser
Muizelaar, J.P., Wei, E.P., Kontos, H.A., et al. (1983) Mannitol Doppler flowmetry, before and during removal of traumatic
causes compensatory cerebral vasoconstriction and vasodil- acute subdural hematoma. J. Neurosurg., 95(4): 569–572.
atation in response to blood viscosity changes. J. Neurosurg., Verweij, B.H., Muizelaar, J.P., Vinas, F.C., Peterson, P.L.,
59(5): 822–828. Xiong, Y. and Lee, C.P. (1997) Mitochondrial dysfunction
Muizelaar, J.P., Wei, E.P., Kontos, H.A., et al. (1986) Cerebral after experimental and human brain injury and its possible
blood flow is regulated by changes in blood pressure and in reversal with a selective N-type calcium channel antagonist
blood viscosity alike. Stroke, 17(1): 44–48. (SNX-111). Neurol. Res., 19(3): 334–339.
Nicholls, D.G. and Budd, S.L. (2000) Mitochondrial and neu- Verweij, B.H., Muizelaar, J.P., Vinas, F.C., Peterson, P.L.,
ronal survival. Physiol. Rev., 80: 315–360. Xiong, Y. and Lee, C.P. (2000a) Improvement in mito-
Richards, T. and Hoff, J. (1974) Factors affecting survival from chondrial dysfunction as a new surrogate efficiency measure
acute subdural hematoma. Surgery, 75: 253–258. for preclinical trials: dose-response and time-window profiles
Robertson, C.S., Narayan, R.K., Gokaslan, Z.L., et al. (1989) for administration of the calcium channel blocker Ziconotide in
Cerebral arteriovenous oxygen difference as an estimate of experimental brain injury. J. Neurosurg., 93(5): 829–834.
cerebral blood flow in comatose patients. J. Neurosurg., Verweij, B.H., Muizelaar, J.P., Vinas, F.C., Peterson, P.L.,
70(2): 222–230. Xiong, Y. and Lee, C.P. (2000b) Impaired cerebral mito-
Rosner, M.J. and Daughton, S. (1990) Cerebral perfusion pres- chondrial function after traumatic brain injury in humans. J.
sure management in head injury. J. Trauma, 30(8): 933–940. Neurosurg., 93(5): 815–820.
Rosner, M.J., Rosner, S.D. and Johnson, A.H. (1995) Cerebral Vibulsreth, S., Hefti, F., Ginsberg, M.D., Dietrich, W.D.
perfusion pressure: management protocol and clinical results. and Busto, R. (1987) Astrocytes protect cultured neurons
J. Neurosurg., 83(6): 949–962. from degeneration induced by anoxia. Brain Res., 422:
Schroder, M., Muizelaar, J.P., Kuta, A., et al. (1996) Thresh- 303–311.
olds for cerebral ischemia after severe head injury: relation- Walz, W. and Mukerji, S. (1988) Lactate production and release
ship with late CT findings and outcome. J. Neurotrauma, 13: in cultured astrocytes. Neurosci. Lett., 86: 296–300.
17–23. Wilberger Jr., J.E., Harris, M. and Diamond, D.L. (1991) Acute
van Santbrink, H., Maas, A.I. and Avezaat, C.J. (1996) subdural hematoma: morbidity, mortality, and operative
Continuous monitoring of partial pressure of brain tissue timing. J. Neurosurg., 74: 212–218.
oxygen in patients with severe head injury. Neurosurgery, Wu, L.G. and Saggau, P. (1997) Presynaptic inhibition of elic-
38(1): 21–31. ited neurotransmitter release. Trends Neurosci., 20: 204–212.
Sciamanna, M.A., Zinkel, J., Fabi, A.Y., et al. (1992) Ischemic Xiong, Y., Gu, Q. and Peterson, P.L. (1997) Mitochondrial
injury to rat forebrain mitochondria and cellular calcium dysfunction and calcium perturbation induced by traumatic
homeostasis. Biochim. Biophys. Acta, 1134: 223–232. brain injury. J. Neurotrauma, 14: 23–34.
Shapiro, K., Marmarou, A. and Shulman, K. (1980) Charac- Xiong, Y., Peterson, P.L., Verweij, B.H., et al. (1998) Mi-
terization of clinical CSF dynamics and neural axis compli- tochondrial dysfunction after experimental traumatic brain
ance using the pressure-volume index: I. The normal pressure injury: combined efficacy of SNX-111 and U-101033E.
volume index. Ann. Neurol., 7(6): 508–514. J. Neurotrauma, 15(7): 531–544.
Siesjö, B.K. (1978) Brain Energy Metabolism. Wiley, Chichester. Young, W. (1992) Role of calcium in central nervous systems
Siesjö, B.K. (1984) Cerebral circulation and metabolism. J. injuries. J. Neurotrauma, 9(Suppl 1): S9–S25.
Neurosurg., 60(5): 883–908. Zauner, A., Daugherty, W.P., Bullock, M.R., et al. (2002) Brain
Siesjö, B.K. (1992) Pathofysiology and treatment of focal cerebral oxygenation and energy metabolism: Part 1-Biological func-
ischemia. Part I: pathofysiology. J. Neurosurg., 77: 169–184. tion and pathophysiology. Neurosurgery, 51: 289–302.
Siesjö, B.K. and Siesjö, P. (1996) Mechanisms of secondary Zauner, A., Doppenberg, E.M., Woodward, J.J., Choi, S.C.,
brain injury. Eur. J. Anaesthesiol., 13: 247–268. Young, H.F. and Bullock, R. (1997) Continuous monitoring
Sokoloff, L. (1960) Metabolism of the central nervous system in of cerebral substrate delivery and clearance. Neurosurgery,
vivo. In: Field J., Magoun H.W. and Hall V.E. (Eds.), 41: 1082–1091.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 9

Progressive damage after brain and spinal cord


injury: pathomechanisms and treatment strategies

Helen M. Bramlett and W. Dalton Dietrich

Department of Neurological Surgery, Neurotrauma Research Center, The Miami Project to Cure Paralysis, University of
Miami Miller School of Medicine, 1095 NW 14th Terrace, Miami, FL 33136, USA

Abstract: The pathophysiology of brain and spinal cord injury (SCI) is complex and involves multiple
injury mechanisms that are spatially and temporally specific. It is now appreciated that many of these injury
mechanisms remain active days to weeks after a primary insult. Long-term survival studies in clinically
relevant experimental studies have documented the structural changes that continue at the level of the insult
as well as in remote brain structures. After traumatic brain injury (TBI), progressive atrophy of both gray
and white matter structures continues up to 1 year post-trauma. Progressive changes may therefore underlie
some of the long-term functional deficits observed in this patient population. After SCI, similar features of
progressive injury are observed including delayed cell death of neurons and oligodendrocytes, axonal
demyelination of intact fiber tracts and retrograde tract degeneration. SCI also leads to supraspinal changes
in cell survival and remote brain circuitry. The progressive changes in multiple structures after brain and
SCI are important because of their potential consequences on chronic or developing neurological deficits
associated with these insults. In addition, the better understanding of these injury cascades may one day
allow new treatments to be developed that can inhibit these responses to injury and hopefully promote
recovery. This chapter summarizes some of the recent data regarding progressive damage after CNS trauma
and mechanisms underlying these changes.

Keywords: traumatic brain injury; spinal cord injury; progressive damage; pathophysiology; treatment

Introduction important from the prospective of clarifying mech-


anisms underlying cell death, but more impor-
Recent studies from both experimental and clinical tantly provide new targets for therapeutic
investigations have emphasized the progressive intervention. Indeed, the observation that proc-
nature of central nervous system (CNS) injury. esses which potentially affect long-term outcome
In contrast to the initial concept that the majority may be active days or even months after injury
of damage occurs at the time of the primary is- provides new targets to improve outcome after
chemic or traumatic insult, new evidence empha- CNS injury. This new way of assessing and treat-
sizes that acute injury can initiate a variety of ing acute injury is also important as we think
pathophysiological cascades that lead to second- about how acute insults such as ischemic or trau-
ary injury mechanisms associated with subacute as matic injuries may enhance the vulnerability of the
well as progressive injury. These findings are aging brain to later occurring neurodegenerative
diseases. The main objective of this chapter is to
Corresponding author. Tel.: +1-305-243-8926; Fax: +1-305- summarize recent date emphasizing the progres-
243-3914; E-mail: hbramlett@miami.edu sive nature of lesion pathology after brain and

DOI: 10.1016/S0079-6123(06)61009-1 125


126

spinal cord injury (SCI) and highlight potential TBI (Povlishock and Christman, 1995; Graham
therapeutic strategies that may be relevant to these et al., 2000b; Leclereq et al., 2001). Diffuse axonal
devastating insults. injury underlies a major mechanism for the mor-
bidity and mortality associated with TBI (Adams
et al., 1989, 1991).
Traumatic brain injury Various animal models have been developed
that mimic some of the structural and behavioral
Traumatic brain injury (TBI) is a leading cause of consequences of human TBI (Gennarelli et al.,
morbidity and mortality in the United States 1982; Cernak, 2005). Although no one model of
(Langlois et al., 2000). In 2000, half of a million TBI exactly represents the human condition of
new cases of moderate and severe TBI were re- brain trauma, these models are important in the
ported. Traumatic insults commonly occur in understanding of critical pathomechanisms re-
young adults as a consequence of traffic and sponsible for traumatic events and the eventual
sporting accidents. More recently, there has been testing of novel therapeutic interventions. One
an increase in the number of traumatic brain in- model, fluid-percussion (F-P) brain injury has been
sults in the elderly due to falls and other traumatic used commonly in these preclinical TBI studies
insults. In developing countries, a significant rise (Dixon et al., 1987). For example, after moderate
in vehicular accidents has also been observed. parasagital F-P brain injury, the extravasation of
Thus, the magnitude of the problem internation- the protein tracer horseradish peroxidase (HRP) is
ally merits increased concern regarding the pre- observed in specific brain regions associated
vention as well as treatment of TBI. with acute vascular damage due to shearing forces
The pathophysiology of TBI is complicated produced by the insult (Dietrich et al., 1994). In
and involves both primary and secondary insults other studies, enduring changes in axolemmal per-
(Graham et al., 2000a; Bramlett and Dietrich, meability has been reported in white matter tracts,
2004). Primary insults due to impact injury result not necessarily associated with overt damage
in the rupture of membranes that lead to acute (Povlishock and Pettus, 1996). Thus, abnormal
damage of neuronal, glial and vascular compo- permeability of a variety of cellular membranes are
nents. These membrane disturbances also cause rendered leaky to tracers after TBI (Pettus and
metabolic stress that initiates a cascade of cellular Povlishock, 1996).
and molecular mechanisms that can initiate both More recently, evidence for apoptotic cell death
reparative as well as destructive processes. Acute has been demonstrated in models of TBI as well as
consequences of TBI include alterations in the SCI (Crowe et al., 1997; Beattie et al., 2002; Lu
blood-brain barrier (BBB) as well as complex et al., 2003). McIntosh and colleagues (Rink et al.,
changes in local cerebral blood flow (LCBF) and 1995) first provided ultrastructural evidence indi-
metabolism (Cortez et al., 1989; Hovda et al., cating that neurons may die by apoptotic mecha-
1991; Dietrich et al., 1994). Trauma-induced glial nisms after F-P brain injury. In that study,
swelling as a consequence of both vasogenic and neurons demonstrated the classical appearance of
cytotoxic edema is also a common early conse- apoptotic bodies within the cell nucleus. More re-
quence to TBI (Bullock et al., 1991; Kimbelberg cently, molecular and biochemical approaches
and Norenberg, 1994). Early metabolic and hemo- have been used to shed light on the various recep-
dynamic events on their own can lead to necrotic tor and intracellular cell signaling mechanisms re-
cell death in specific vulnerable populations of sponsible for apoptotic cell death (Keane et al.,
cells. Severe reductions in LCBF are observed in 2001a, b). The importance of this line of research
some TBI patients and may indicate ischemic to the present discussion is that emerging evidence
events (Bramlett and Dietrich, 2004). In human supports the concept that apoptotic pathways may
TBI, specific gray and white matter structures in- be activated for long periods after CNS injury
cluding the corpus callosum and hippocampus are and that mechanisms similar to programmed cell
frequently damaged after moderate and severe death may be responsible for some of the
127

progressive changes seen in the brain late after occurred without any evidence of a systemic seco-
trauma (Williams et al., 2001). ndary insult and therefore had no obvious under-
In addition to apoptosis, inflammatory and im- lying mechanism.
mune processes also appear to play an important In experimental studies of TBI, the majority of
role in pathophysiology of TBI (Shohami et al., investigations have evaluated traumatic outcome
1994; Schwab et al., 2001; Morganti-Kosmann in the acute or subacute post-traumatic period.
et al., 2002). Following brain injury, indicators of Thus, until recently, little information was avail-
the activation of pro-inflammatory processes in- able to determine the more chronic consequences
cluding increased expression of pro-inflammatory of experimental TBI. However, in 1997, the
cytokines such as IL-1b, TNFa and IL-6 have chronic histopathological consequences of moder-
been described (Holmin et al., 1997; Morganti- ate F-P brain injury were first assessed at 2 months
Kossman et al., 1997; Kinoshita et al., 2002). Also, after trauma. Bramlett et al. (1997a) reported a
specific receptors that interact with these cytokines significant enlargement of the lateral ventricle and
are expressed after injury and lead to the stimu- with associated atrophy of cerebral cortical areas
lation of inflammatory signaling cascades respon- within the traumatized hemisphere. This study was
sible for the expression of inflammatory genes the first to provide histopathological evidence for
(Lotocki et al., 2004). Thus, experimental studies structural changes continuing to occur weeks to
that have targeted inflammatory processes by ap- months after TBI. In subsequent studies, other in-
plying antibodies or blockers to inflammation vestigators using similar or different injury models
have in some cases lead to improved outcome in have complemented and extended these initial
the subacute post-traumatic setting. For example, findings by assessing even longer periods of post-
in several studies, treatment with the interleukin-1 traumatic injury (Smith et al., 1997; Pierce et al.,
receptor antagonist has been shown to lead to 1998; Dixon et al., 1999; Bramlett and Dietrich,
improved histopathological and behavioral out- 2002). For example, Smith et al. (1997) reported
come after TBI and cerebral ischemia (Toulmond progressive injury in various forebrain areas 1 year
and Rothwell, 1995; Rothwell and Luheshi, 2000). after moderate lateral F-P injury. In a model of
In reference to the present discussion, recent controlled cortical impact (CCI) injury, Dixon
evidence for inflammatory processes remaining et al. (1999) also reported progressive damage and
activated up to 1 year after experimental TBI long lasting behavioral deficits in that trauma
(Nonaka et al., 1999) and even longer after clinical model of focal damage. Thus, in various labora-
TBI (Gentleman et al., 2004) emphasizes the po- tories using different injury models, evidence of
tential role of inflammation in progressive damage progressive damage has been demonstrated. Most
after trauma. recently, Bramlett and Dietrich (2002) assessed
alterations in white matter tracts 1 year after mod-
erate F-P brain injury (Fig. 1). In that study,
Clinical evidence for progressive injury significant atrophy of various white matter struc-
tures within the traumatized hemisphere provided
In patients following TBI, magnetic resonance im- direct evidence of progressive white matter pa-
aging (MRI) approaches have identified evidence thology in widespread forebrain circuits. These
for progressive atrophy of specific brain regions at newer data emphasize the important fact that
chronic periods after trauma (Cullum and Bigler, white matter as well as gray matter structures may
1986; Anderson and Bigler, 1995; van der Naalt be highly vulnerable to progressive damage after
et al., 1999). In these clinical studies, evidence for relatively moderate degrees of trauma. Thus, ther-
the enlargement of ventricle structures and atro- apies may have to be developed that target both
phy of specific gray and white matter structures gray and white matter pathology after TBI
has been demonstrated. In one study, enlargement (Medana and Esiri, 2003). The importance of
of the lateral ventricle was identified as a relatively white matter vulnerability will again be empha-
late occurrence after TBI. This delayed response sized in the SCI discussion.
128

Fig. 1. Double-stained H&E and Luxol-fast blue sections 1 year after TBI or sham procedure. (A) TBI animal showing gross atrophy
with marked expansion of the ipsilateral lateral ventricle. (B) Sham operated animal appearing unremarkable. (C) Higher magni-
fication of external capsule thinning (arrows) after TBI. Reprinted from Bramlett and Dietrich (2002) with kind permission of Springer
Science and Business Media.

Pathomechanisms underlying progressive injury structure and function of an axon and/or circuit.
Oligodendrocyte death would lead to demyelina-
As previously discussed, both apoptotic and in- tion of projecting axons that would be expected
flammatory cascades are felt to underlie some of to result in axonal dysfunction and possible
the acute and subacute pathophysiological mech- progressive damage (Waxman, 1989). Indeed,
anisms that are responsible for cellular dysfunction electrophysiological studies have demonstrated
and death. Recent data also indicate that these the adverse effects of demyelination on action
mechanisms may be active weeks and months after potentials and axonal survival (Waxman et al.,
trauma and also participate in the progressive na- 1994). Thus, progressive apoptotic cell death can
ture of TBI. For example, prolonged apoptotic have a variety of adverse consequences on the
cell death has been demonstrated in several CNS structure and function of both gray and white
injury models (Emery et al., 1998; Beattie et al., matter structures.
2002). In these studies neuronal, microglial and In the area of inflammation, recent data has
oligodendrocyte apoptosis has been reported days also emphasized the progressive nature of that re-
to weeks after injury. Delayed neuronal apoptosis sponse to TBI. Several studies have provided evi-
may lead to the continued removal of axon pro- dence for long-term inflammatory responses to
jections that could ultimately lead to patterns of TBI. In brain sections stained for macrophage
deafferentation syndromes in brain regions remote activation for example, evidence for inflammatory
from the primary site of damage. A good example events is sometimes also seen many weeks after a
of this type of progressive and remote damage is traumatic brain insult. Gentleman et al. (2004)
the delayed thalamic pathology observed after reported microglial activation, weeks after TBI.
parasagittal or lateral F-P brain injury (Bramlett Similarly, inflammatory cells have been observed
et al., 1997a, b). Apoptotic death of oligodendro- chronically in an animal model of TBI
cytes could also have devastating effects on both (Rodriguez-Paez et al., 2005). In this study,
129

Fig. 2. Light level micrographs of toluidine blue stained thick plastic sections of control (A) and traumatized tissue, specifically the
lateral posterior thalamic nuclei, (100  ) at 3 days (B), 15 days (C), 3 months (D), 9 months (E) and 12 months (F) after TBI. (A)
Control tissue shows myelinated figures oriented perpendicular and parallel to the plain of the section. Normal appearing neuronal cell
bodies (N), astrocytes (black arrow) and blood vessels (V) are also apparent. (B) At 3 days, axonal abnormalities including changes in
axoplasmic density, unraveling of the myelin sheath (black pointer) and irregular swollen myelinated profiles (arrowhead) were
observed. There is also a proliferation of microglial cells (open arrow) associated with neuronal (N) and astrocytic (black arrow)
swelling. Normal appearing oligodendrocytes (double arrows) are observed. (C) At 15 days, there is an apparent increase in overall
tissue vacuolation, which appears to be associated with axonal (arrowhead) and non-axonal (black arrow) profiles. An increase in
microglial cells (open arrows) as well as unraveled myelin figures (black pointer) are observed. (D) At 3 months, parenchymal
vacuolation is still apparent with increased numbers of microglial cells (open arrows), swollen axonal figures (arrowhead) and irregular
myelin profiles (black pointer). (E) At 9 months, a dramatic number of vacuolated profiles are observed related to inflammatory cells
scattered among tissue debris. An amorphous crystallized material (black star) was observed inside the vacuolated profiles. There
appears to be a progressive increase in the size of the vacuolated profiles as the post-injury time increases. (F) At 12 months, the
vacuolization (large black arrows) continues to progress in association with a further decrease in numbers of myelinated axons without
the presence of the amorphous material described above. Reprinted from Rodriguez-Paez et al. (2005) with kind permission of Springer
Science and Business Media.
130

macrophage/microglia infiltration and swollen ax- after injury (Rancan et al., 2004). In the future,
ons were observed as late as 6 months using both surrogate biochemical markers of tissue damage
light and electron microscopic analysis within may be developed to predict and treat progressive
several vulnerable structures (Fig. 2). Addition- injury mechanisms.
ally, a temporal decline in the number of myelin- Abnormal protein aggregation has been impli-
ated axons within the cerebral cortex (Fig. 3) and cated in the pathogenesis of a number of neuro-
thalamus were reported which may be due to the logical diseases including Alzheimer’s and
prolonged inflammatory response observed in this Parkinson’s disease (Chaudhuri and Paul, 2006).
study. Nonaka et al. (1999) also provided evidence Recent evidence indicates that abnormal protein
for inflammatory processes being active up to 1 aggregation also occurs in models of cerebral is-
year after TBI. In that study, NF-kappaB was chemia and TBI (Blumbergs et al., 1994; Graham
seen in both cortical and sub-cortical brain regions et al., 1995; Lewen et al., 1995; Bramlett et al.,
undergoing progressive atrophy. Thus, the poten- 1997b; Hamberger et al., 2003). In models of TBI,
tial for macrophage/microglia released toxic immunocytochemical localization of beta APP
substances leading to tissue damage is a real pos- and other proteins has been identified in gray as
sibility even weeks after TBI. To support this as- well as white matter tracts (Sherriff et al., 1994;
sumption, recent biomarker studies that have Bramlett et al., 1997b). Whether evidence for pro-
measured levels of pro-inflammatory mediators longed periods of protein aggregation can be cor-
in the cerebral spinal fluid (CSF) and plasma of related with progressive tissue damage merits
TBI patients have reported elevated levels, days continued study.

Fig. 3. Estimation of myelinated axons in the cerebral cortex of animals following TBI or Sham surgery at 3 days, 15 days, 1 month,
3 months, 6 months, 9 months and 12 months. A significant difference was found for both group (po0.001) and time (po0.001)
between TBI and Sham animals. TBI results in a decrease in the number of myelinated axons compared to Sham animals at all time
points analyzed. In addition, there was a temporal decrease in the number of myelinated axons in the Sham animals as well. The
decrease in the axonal numbers of the Sham animals may be due to a normal aging process. Reprinted from Rodriguez-Paez et al.
(2005) with kind permission of Springer Science and Business Media.
131

Spinal cord injury addition, calpain activation has also been empha-
sized as an acute injury mechanism (Ray et al.,
Each year in the United States, approximately 2003). Many of these injury cascades have been
11,000 new spinal cord injuries are recorded. Cur- targets for therapeutic interventions (Blight and
rently there are over 250,000 individuals living Zimber, 2001).
with chronic SCI and its devastating consequences More recently, evidence for apoptotic cell death
in the United States. Similar to what has been de- has been reported in a number of SCI models
scribed with TBI, evidence for lesion progression (Springer et al., 1999; Ozawa et al., 2002; Keane
has also been demonstrated in both experimental et al., 2006). A complex integrated apoptotic path-
and clinical conditions of SCI (Wallace et al., way involving both extrinsic and intrinsic apopto-
1987; Bunge et al., 1993; Crowe et al., 1997; Bruce tic cascades has been reported. Also, the study of
et al., 2000; Hill et al., 2001; Guest et al., 2005; pro- and anti-apoptotic molecules, which can con-
Totoiu and Keirstead, 2005). Because SCI fre- trol apoptotic cell death, is an exciting area of
quently occurs in young people, it is equally im- current investigation. Recent work has concen-
portant that we understand the mechanisms trated on mechanisms that allow for the commu-
underlying chronic progressive damage and de- nication between the external environment with
velop therapeutic interventions to retard cell intracellular processes involved in cell survival and
death, axonal degeneration and demyelination in death. After trauma for example, specific death
SCI patients. The pathophysiology of acute SCI is receptors have been shown to accumulate within
multifactorial and like TBI, includes both primary specific areas of the plasma membrane called lipid
and secondary injury mechanisms (Nashmi and rafts that contain high concentrations of choles-
Fehlings, 2001; Keane et al., 2006). Primary injury terol and sphingolipid (Lotocki et al., 2004). Thus,
mechanisms include acute SC compression, im- after SCI and brain trauma, TNF1 receptors ac-
paction, laceration, shear damage and missile in- cumulate in lipid rafts and assist in the formation
jury (Norenberg et al., 2004). These acute injury of cytoplasmic platforms that allow scaffolding
mechanisms initiate a complex cascade of second- proteins to accumulate; leading to the increased
ary injury mechanisms that may remain activated interactions of proteins that lead to the activation
for months or years after injury. These initial of intracellular cascades associated with cell sur-
traumatic events lead to vascular damage and vival and death (Keane et al., 2006). Thus, re-
hemorrhage, alterations in spinal cord blood flow, search is currently being undertaken to understand
vascular thrombosis, vasospasm and loss of auto- this relatively acute injury response to neuronal
regulation. As a consequence to cellular membrane vulnerability and survival. Because a large amount
damage, metabolic abnormalities and electrolytic of cell death occurs in the acute post-traumatic
shifts in ions occur (LoPachin and Lehning, 1997; period, evidence for these pathomechanisms has
Li et al., 2000). In addition, neurotransmitters are been commonly reported during these periods.
released into the extracellular space leading to the However, since cell death during delayed
abnormal activation of various receptors and in- post-traumatic periods is spread out and more
tracellular signaling processes (Keane et al., 2006). difficult to identify, other approaches including
Other classical injury cascades that are activated immunocytochemistry must be used to investigate
after SCI include free radical formation, lipid per- later occurring injury mechanisms.
oxidation and edema formation. All of these acute In addition, experimental and clinical investiga-
processes lead to the acute destruction of gray and tions have also demonstrated that apoptotic cell
white matter structures (Blight, 1985; Schwab and death, demyelination, remyelination and axonal
Bartholdi, 1996; Rosenberg and Wrathall, 1997). degeneration may occur weeks to months after
As in brain injury, inflammatory cascades are experimental and clinical SCI (Bunge et al., 1961;
also activated after SCI (Blight, 1992; Crowe et al., Blight, 1993; Quencer and Bunge, 1996; Schwab
1997; Popovich et al., 2002; Keane et al., 2006). In and Bartholdi, 1996; Crowe et al., 1997; Emery
132

et al., 1998; Bruce et al., 2000; Hill et al., 2001; demyelination in an animal model of SCI. Data
Guest et al., 2005; Totoiu and Keirstead, 2005). from that study indicated that chronic progressive
For example, in a study by Crowe et al. (1997), demyelination after thoracic injury in rats does
apoptotic cells were identified from 6 h to 3 weeks occur for days after injury (Fig. 5). Interesting, a
after experimental SCI. Apoptotic cell death was process of secondary demyelination was reported
specifically shown to be present in the white matter around 120–450 days post injury. These studies
tracts where apoptotic cells were shown to be pos- underscore the importance of targeting demyeli-
itive for cellular markers of oligodendrocytes. This nation in the development of therapeutic interven-
observation is important because it indicates tions and again emphasize the progressive nature
oligodendrocyte cell death with resulting demyeli- of neurodegeneration after SCI.
nation could be an active mechanism in the pro- In human tissue, evidence for chronic demyeli-
gressive injury cascades associated with human nation has also been reported (Bunge et al., 1993;
SCI. Following this particular observation, vari- Norenberg et al., 2004; Guest et al., 2005). In the
ous laboratories have also reported apoptotic cell study by Guest et al. (2005), evidence for axonal
death in the spinal cord including neurons, demyelination, even a decade after human trau-
oligodendrocytes and inflammatory cells (Emery matic SCI was presented. Although the response
et al., 1998; Beattie et al., 2002). In addition to was very heterogeneous among the SCI specimens
clarifying mechanisms of injury, these results are evaluated, the potential for demyelination in spe-
also important because they provide new potential cific spinal cord circuits in which axon profiles re-
targets for therapeutic interventions directed to- mained intact was observed. In this regard,
ward the acute as well as chronic post-traumatic evidence for spontaneous remyelination of intact
period (Springer et al., 1999; Ozawa et al., 2002; CNS axons by invading Schwann cells has also
Demjen et al., 2004). been observed (Guest et al., 2005; Totoiu and
Evidence for apoptotic cell death and Schwa- Keirstead, 2005). This observation is important
nnosis has also been reported in specimens ob- because other studies have reported similar cellular
tained from patients that survived long periods responses to injury that indicate some endogenous
after SCI. In a study by Emery et al. (1998), apo- reparative mechanisms including remyelination
ptotic cells were identified using the apoptotic are activated after SCI (Hagg and Oudega, 2006).
marker caspase-3. In that study, oligodendrocytes In a clinical study by Bruce et al. (2000), evi-
stained positively for this indicator of apoptotic dence for Schwannosis was seen in 32 out of 65
injury in specific white matter tracts. Again, this cases that survived after 24 years. The incidence of
clinical observation is important because of the Schwannosis rose to 82% in SCI patients who
role of the oligodendrocyte in axonal myelination. survived more than 4 months. Associated with
We know from various electrophysiological stud- Schwannosis was intense chondroitin sulfate pro-
ies that if an axon is demyelinated by a toxic or teoglycan (CSPG) staining. This observation is
traumatic insult, that axon’s ability to propagate important because the CSPGs are thought to be
axon potentials is severely affected (Blight and inhibitory molecules that reduce axonal regenera-
Young, 1989; Waxman, 1989). Thus, strategies tion after CNS injury (Snow et al., 1990). Thus the
that would target degenerative mechanisms or ax- continued clarification of both the positive and
onal conduction blockage could potentially im- negative consequences of Schwann cell responses
prove outcome in SCI subjects (Blight and Young, to acute and chronic SCI remains an important
1989). Hill et al. (2001) have reported on extensive area of investigation.
dieback of the corticospinal tract chronically after The use of human spinal cords donated for re-
SCI (Fig. 4). However, there was a regenerative search is also allowing other injury mechanisms to
response of this tract along with the reticulospinal be evaluated in human tissues (Norenberg et al.,
tract evidenced by an extension of collaterals into 2004). At The Miami Project to Cure Paralysis,
the lesion matrix. Recently, Totoiu and Keirstead approximately 115 human spinal cords have been
(2005) have assessed chronic progressive collected from individuals surviving from 24 h to
133

Fig. 4. Appearance of impact site after a 12.5 mm contusion injury 1 dpi (A–C), 3 dpi (A, D, E), 8 dpi (A, F, G), 21 dpi (A, H, I) and 14
weeks (A, J, K, L) after injury. (A) The progression of cavitation. At 1 day, the impact site is hemorrhagic but no cavitation is present,
and RBCs extend up to an additional 5 mm rostral (arrow); between 3, 8 and 21 days the injury site becomes progressively more defined,
and it appears as a dark region in low magnification at 8 dpi and is clearly defined at 21 days. At 21 days and 14 weeks a cellular mass is
present within the cavity attached to the spared rim of white matter at several points via trabeculae (arrow). One dpi (B) RBCs and (C)
damaged axons at the impact site. Three dpi (D) damaged axons (small arrow) intermixed with blood cell infiltrate (large arrow) at the
impact site and (E) some macrophages (large arrowhead) have infiltrated the rim of spared white matter (small arrows). Eight dpi (F)
macrophages are densely packed at the center of the cavity while (G) open spaces (small arrow) are present between groups of
macrophages and other nonfluorescently labeled cells (large arrowhead) that appear in scattered bands within the cavity. Twenty-one
dpi (H) trabeculae, thin tissue bridges with regions of no tissue beside them, are beginning to form (small arrow), and macrophages
(large arrow head) are still present within the cavity in association with the trabeculae or as (I) macrophage rafts. Fourteen weeks after
injury (J–L) autofluorescing macrophages (large arrowhead) are present within (J) fibrous trabeculae and (K) cellular trabeculae as well
as (L) the cellular infiltrate rostral to the cavity. Bar in (A), 1 mml; all other bars, 100 mm. (C, E, F), same magnification. (B, D, G–L),
same magnification. dpi ¼ days post injury. Reprinted from Hill et al. (2001) with permission from Elsevier.
134

Fig. 5. Quantification of demyleinated and remyelinated axons at different time points post injury. The numbers of axons at the six
cranial and six caudal levels examined for all animals within a group were averaged to generate each point on the graph. The number of
demyleinated axons was highest at 1 day post injury and decreased substantially by 7 days post injury. Thereafter, demyelination was a
chronic and progressive phenomenon. Both oligodendrocyte and Schwann cell remyelination was present at all subsequent time points
post injury. Reprinted from Totoiu and Keirstead (2005) with permission from John Wiley & Sons, Inc.

24 years after injury. Corresponding MRI and which is an important secondary injury mechanism
immunocytochemical techniques are used to anal- after SCI (Gris et al., 2004).
yze the injured tissue (Bunge et al., 1993; Becerra Evidence for retrograde degeneration of cortical
et al., 1995; Emery et al., 1998; Guest et al., 2005). spinal tracts (CST) has also been shown in human
With this approach, investigators have assessed specimens. Following damage to thoracic cortical
the acute as well as more chronic immunohisto- spinal tracts for example, evidence for retrograde
pathological consequences of traumatic human degeneration can be seen in remote spinal tracts
SCI. For example, in early specimens taken 3 days (Bunge et al., 1993; Norenberg et al., 2004). In
after injury H&E staining shows damage to gray addition to retrograde degeneration, evidence for
and white matter areas with a well identified cen- Wallerian degeneration can also be obtained using
tral hemorrhagic lesion (Bunge et al., 1993). If repetitive MR imaging procedures in SCI patients
specimens are stained with various immunocyto- (Quencer and Bunge, 1996). These studies empha-
chemical approaches, evidence of gliosis in the size the widespread changes that occur as a con-
form of increased glial fibrillary acidic protein sequence to local SCI. With the development and
(GFAP) staining is seen surrounding the contusive improvement of new imaging techniques, acute
site. Also, the accumulation of invading inflam- damage as well as the progressive nature of human
matory cells such as polymorphonuclear leuko- SCI will be more easily investigated. In acute in-
cytes (PMNL) and macrophages can be visualized jury settings, better evidence for local cord com-
(Fleming et al., 2006). These types of studies con- pression will provide important information to
ducted on human tissues are important because help guide surgical approaches to target ischemic
they verify some of the acute and more chronic events. In chronic postoperative SCI cases, both
histopathological changes that are reported in cord compression and the formation and progres-
preclinical animal studies. This information is sion of cavities can be visualized in patients expe-
currently of importance to the development of riencing neurological symptoms including chronic
new treatments targeting inflammatory processes, pain. With time, some lesions expand to form cysts
135

that can have a dramatic effect on progression of McDonald et al., 2002; Sabbah et al., 2002; Lotze
neurological symptoms. These imaging techniques et al., 2006). As specific inputs to the cerebral cor-
will continue to be improved and allow specialized tex are removed after SCI, other cortical areas may
treatments to be developed for an individual pa- take over function. Thus, electrophysiological and
tient undergoing progressive injuries. metabolic studies have shown evidence for cortical
plasticity after SCI (Hoffman and Field-Fote,
2007; Kim et al., 2006). These observations not
Supraspinal alterations after SCI only emphasize the potential for plasticity occur-
ring in remote brain regions after SCI, but also
In addition to changes occurring at the level of the may be important as we think about repairing the
injured spinal cord, it is also clear that SCI leads to nervous system in the chronically injured spinal
alterations in supraspinal areas of the neuroaxis cord. If brain circuits and/or cortical maps were
(Jain et al., 1997; Raineteau et al., 2001; Hains significantly altered due to chronic SCI, would
et al., 2003; Hubscher and Johnson, 2006; Kim such a consequence affect the ability to repair the
et al., 2006). In this regard, there is a rich literature nervous system and return function? Only after
on experimental SCI lesioning studies in rodents successful regenerative approaches are developed
and non-human primates showing evidence for in the chronically injured state can these questions
degeneration of neurons in the cerebral cortex af- be answered.
ter injury. Some data indicate that cell death oc-
curs through apoptotic mechanisms whereas more
recently, additional evidence indicates that cortical Therapeutic interventions targeting progressive
cell bodies as a consequence of SCI may only un- injury
dergo severe atrophy but not actually die. In these
cases, the local addition of neurotrophic factors to Based on the complexity of brain and SCI, it is
a particular brain region reverses some of these clear that the injury mechanisms are multifactorial
morphological changes. and may require a combinational therapeutic ap-
In addition to cellular changes, evidence for cir- proach. In the area of brain and SCI, various
cuit reorganization and plasticity resulting in de- neuroprotective agents have been evaluated
activation and reactivation of certain brain areas (Narayan et al., 2002). Neuroprotective agents
has been demonstrated after clinical and experi- such as methylprednisolone, GM1 gangliosides,
mental SCI (Jain et al., 1997; Raineteau et al., lazeroids, calcium and sodium channel blockers,
2001). Chronic SCI has also been shown to induce growth factors as well as blockers of excitototoxic
changes in the response of thalamic neurons to process have been reported to be effective in some
physiological activation (Hubscher and Johnson, animal models. More recently, the use of anti-in-
2006). Thus, SCI leads to alterations in brain cir- flammatory strategies, calpain antagonists, anti-
cuits that are responsible for assessing normal and apoptotic strategies as well as agents targeting
abnormal sensation. In this regard, some research- cAMP have also been investigated with various
ers believe that this circuit plasticity in addition to results (Blight and Zimber, 2001).
other events including chronic inflammation may Mild hypothermia that targets multiple injury
account for the late development of chronic ne- cascades has been tested with various degrees of
urogenic pain in patients with SCI. A clearer un- success after both SCI and TBI (Hayashi et al.,
derstanding of local circuit changes in response to 2004; Guest and Dietrich, 2005). Although hypo-
SCI may provide important information regarding thermia was used in the 60 s to target both brain
how to treat these patients with neurogenic pain. and SCI, profound hypothermia (271C) commonly
As a consequence of chronic SCI, the cerebral produced severe effects on cardiac function and
cortex may also undergo plastic changes in terms increased infection rates in patients. However, in
of shifts in cortical map architecture (Topka et al., the mid-80 s, the importance of mild to moderate
1991; Jain et al., 1997; Bruehlmeier et al., 1998; hypothermic treatment was demonstrated in
136

ischemic and traumatic animal models. Busto et al. oxide (NO) synthesis. The inducible form of nitric
(1987) first reported that a reduction of just 2 or 3 oxide synthase (iNOS) produces free radicals that
degrees in the temperature of the brain provided can be destructive to tissue survival. Post-traumatic
dramatic protection of CA1 neurons within the hypothermia following TBI also reduces iNOS ac-
post-ischemic hippocampus. Subsequent studies tivity and may improve outcome by affecting free
demonstrated that mild hypothermia was also radical generation as well (Chatzipanteli et al.,
protective when initiated at various times after 1999).
cerebral ischemia or TBI. Recently, mild hypo- Mild hypothermia has been shown to be neuro-
thermia has been shown to have a dramatic effect protective and promote functional outcome after
on SCI. In a study by Yu et al. (2000), the bene- TBI (Hayashi et al., 2004). Bramlett et al. (1995)
ficial effects of systemic hypothermia on locomo- first reported that post-traumatic hypothermia im-
tor outcome and histopathological damage were proved cognitive function in rats. These studies are
reported after contusion SCI in rats. In that study, important because cognitive deficits are some of
mild hypothermia (331C) initiated 30 min after SCI the most severe consequences of mild, moderate
significantly improved open locomotor function and severe TBI. In regards to potential treatments
and significantly reduced contusion volume weeks for progressive injury, post-traumatic hypothermia
after injury. Because of these exciting preclinical has also been shown to reduce the progressive na-
findings, multiple clinical trials have been initiated ture of F-P brain injury in rats (Bramlett et al.,
in patients with various neurological insults in- 1997a). In that study, a period of 3 h of moderate
cluding cardiac arrest, stroke, TBI and SCI. Re- hypothermia significantly protected against pro-
cent multicenter trials in cardiac arrest patients gressive damage and enlargement of the lateral
have shown a dramatic benefit with mild hypo- ventricle at 2 months after TBI. Thus, in addition
thermia in that patient population (Bernard et al., to the acute benefits of therapeutic hypothermia,
2002; Hozler, 2002). Also, individual and multi- this therapy may also limit the amount of pro-
center trials have reported that mild hypothermia gressive damage after trauma. Additional studies
shows promise in improving outcome in severe will be required to determine what duration of the
TBI patients (Jiang et al., 2006). Clinical investi- hypothermic therapy is most beneficial in provid-
gations are currently determining the effects of ing long-term benefits in terms of functional
mild hypothermia involving SCI (Guest and outcome.
Dietrich, 2005). In this regard, mild hypothermia Finally, recent data indicate that different types
may be used during elective surgeries or in the of cellular transplantation strategies can promote
early post-injury periods. repair and improve recovery in animal models of
The beneficial effects of mild hypothermia in- brain and SCI (Schouten et al., 2004; Schwab
volve multiple injury pathomechanisms. Various et al., 2006; Thuret et al., 2006). In some cases, cell
studies have shown that modifying brain or SC transplantation strategies have been initiated to
temperatures significantly affects injury-induced replace dysfunctional or dead neurons injured by
excitatory neurotransmitter levels, free fatty acid the insult. Thus, an exciting field of research is
formation, BBB breakdown and the formation of currently directed toward stem cell biology and the
edema. Mild hypothermia after SCI has been potential for neural stem cells to repopulate the
shown to significantly reduce the acute inflamma- injured nervous system and improve function. Cell
tory response to injury (Chatzipanteli et al., 2000). survival, migration, control of cellular differenti-
Because the early inflammatory response to injury ation and the integration of new cells into existing
is thought to represent an important secondary in- circuits are some of the challenges currently being
jury mechanism, the ability of mild hypothermia to addressed with these approaches. Alternatively,
limit the accumulation of inflammatory cells after the transplantation of specific populations of
SCI represents an important therapeutic target for restricted progenitor cells that differentiate into
treatment. Free radical formation can come from myelin-forming cells is another important research
multiple sources including prostaglandin and nitric direction (Cao et al., 2005; Keirstead et al., 2005;
137

Karimi-Abdolrezaee et al., 2006). In recent studies, Blight, A.R. (1985) Delayed demyelination and macrophage
grafted cells have been shown to form morpho- invasion: a candidate for secondary cell damage in spinal
cord injury. Cent. Nerv. Syst. Trauma, 2: 299–315.
logically normal-appearing myelin sheaths around
Blight, A.R. (1992) Macrophages and inflammatory damage in
damaged axons. Importantly, experimental treat- spinal cord injury. J. Neurotrauma, 9(Suppl 1): S83–S91.
ments targeting neuronal or glia damage after Blight, A.R. (1993) Remyelination, revascularization, and re-
CNS injury leads to improvements in functional covery of function in experimental spinal cord injury. Adv.
recovery. Because progressive damage after brain Neurol., 59: 91–104.
and SCI can affect a variety of different cell types, Blight, A.R. and Young, W. (1989) Central axons in injured cat
spinal cord recover electrophysiological function following
these cellular therapies may be helpful in both ne- remyelination by Schwann cells. J. Neurol. Sci., 91: 15–34.
uroprotective as well as reparative strategies tar- Blight, A.R. and Zimber, M.P. (2001) Acute spinal cord injury:
geting the long-term consequences of CNS injury. pharmacotherapy and drug development perspectives. Curr.
Continued research into the progressive nature of Opin. Investig. Drugs, 2: 801–808.
Blumbergs, P.C., Scott, G., Manavis, J., Wainwright, H., Simp-
brain and SCI should provide new targets for
son, D.A. and McLean, A.J. (1994) Staining of amyloid pre-
treatment and better outcome in patients that sus- cursor protein to study axonal damage in mild head injury.
tain these devastating injuries. Lancet, 344: 1055–1056.
Bramlett, H.M. and Dietrich, W.D. (2002) Quantitative struc-
tural changes in white and gray matter 1 year following
Acknowledgments traumatic brain injury in rats. Acta Neuropathol. (Berl.), 103:
607–614.
Bramlett, H.M. and Dietrich, W.D. (2004) Pathophysiology of
This work was supported in part by NIH grants cerebral ischemia and brain trauma: similarities and differ-
NS30291, NS38665 and DAMD17-02-1-0190. The ences. J. Cereb. Blood Flow Metab., 24: 133–150.
authors also thank the members of the Bramlett/ Bramlett, H.M., Dietrich, W.D., Green, E.J. and Busto, R.
Dietrich laboratory for their important contribu- (1997a) Chronic histopathological consequences of fluid-
percussion brain injury in rats: effects of post-traumatic
tions to this research field.
hypothermia. Acta Neuropathol. (Berl.), 93: 190–199.
Bramlett, H.M., Green, E.J., Dietrich, W.D., Busto, R., Globus,
M.Y-T. and Ginsberg, M.D. (1995) Post-traumatic brain
References hypothermia provides protection from sensorimotor and
cognitive behavioral deficits. J. Neurotrauma, 12: 289–298.
Adams, J.H., Doyle, D., Ford, I., Gennarelli, T.A., Graham, Bramlett, H.M., Kraydieh, S., Green, E.J. and Dietrich, W.D.
D.I. and McLellan, D.R. (1989) Diffuse axonal injury in head (1997b) Temporal and regional patterns of axonal damage
injury: definition, diagnosis and grading. Histopathology, 15: following traumatic brain injury: a beta-amyloid precursor
49–59. protein immunocytochemical study in rats. J. Neuropathol.
Adams, J.H., Graham, D.I., Gennarelli, T.A. and Maxwell, Exp. Neurol., 56: 1132–1141.
W.L. (1991) Diffuse axonal injury in non-missile head injury. Bruce, J.H., Norenberg, M.D., Kraydieh, S., Puckett, W.,
J. Neurol. Neurosurg. Psychiatry, 54: 481–483. Marcillo, A. and Dietrich, W.D. (2000) Schwannosis: role of
Anderson, C.V. and Bigler, E.D. (1995) Ventricular dilation, gliosis and proteoglycan in human spinal cord injury. J.
cortical atrophy, and neuropsychological outcome following Neurotrauma, 17: 781–788.
traumatic brain injury. J. Neuropsychiatry Clin. Neurosci., 7: Bruehlmeier, M., Dietz, V., Leenders, K.L., Roelcke, U., Miss-
42–48. imer, J. and Curt, A. (1998) How does the human brain deal
Beattie, M.S., Hermann, G.E., Rogers, R.C. and Bresnahan, with a spinal cord injury? Eur. J. Neurosci., 10: 3918–3922.
J.C. (2002) Cell death in models of spinal cord injury. In: Bullock, R., Maxwell, W.L., Graham, D.I., Teasdale, G.M. and
McKerracher L., Doucet G. and Rossignol S. (Eds.), Adams, J.H. (1991) Glial swelling following human cerebral
Progress in Brain Research, Vol. 137. Elsevier, Amsterdam, contusion: an ultrastructural study. J. Neurol. Neurosurg.
pp. 37–47. Psychiatry, 54: 427–434.
Becerra, J.L., Puckett, W.R., Hiester, E.D., Quencer, R.M., Bunge, M.B., Bunge, R.P. and Ris, H. (1961) Ultrastructural
Marcillo, A.E., Post, M.J. and Bunge, R.P. (1995) MR- study of remyelination in an experimental lesion in adult cat
pathologic comparisons of wallerian degeneration in spinal spinal cord. J. Biophys. Biochem. Cytol., 10: 67–94.
cord injury. AJNR Am. J. Neuroradiol., 16: 125–133. Bunge, R.P., Puckett, W.R., Becerra, J.L., Marcillo, A. and
Bernard, S.A., Gray, T.W., Buist, M.D., Jones, B.M., Silvester, Quencer, R.M. (1993) Observations on the pathology of hu-
W., Gutteridge, G. and Smith, K. (2002) Treatment of com- man spinal cord injury. A review and classification of 22 new
atose survivors of out-of-hospital cardiac arrest with induced cases with details from a case of chronic cord compression
hypothermia. N. Engl. J. Med., 346: 557–563. with extensive focal demyelination. Adv. Neurol., 59: 75–89.
138

Busto, R., Dietrich, W.D., Globus, M.Y.-T., Valdes, I., Fleming, J.C., Norenberg, M.D., Ramsay, D.A., Dekaban,
Scheinberg, P. and Ginsberg, M.D. (1987) Small differences G.A., Marcillo, A.E., Saenz, A.D., Pasquale-Styles, M.,
in intraischemic brain temperature critically determine the Dietrich, W.D. and Weaver, L.C. (2006) The cellular inflam-
extent of ischemic neuronal injury. J. Cereb. Blood Flow matory response in human spinal cords after injury. Brain,
Metab., 7: 729–738. 129: 3259–3269.
Cao, Q., Xu, X.M., DeVries, W.H., Enzmann, G.U., Ping, P., Gennarelli, T.A., Thibault, L.E., Adams, J.H., Graham, D.I.,
Tsoulfas, P., Wood, P.M., Bunge, M.B. and Whittemore, Thompson, C.J. and Marcincin, R.P. (1982) Diffuse axonal
S.R. (2005) Functional recovery in traumatic spinal cord in- injury and traumatic coma in the primate. Ann. Neurol., 12:
jury after transplantation of multineurotrophin-expressing 564–574.
glial-restricted precursor cells. J. Neurosci., 25: 6947–6957. Gentleman, S.M., Leclercq, P.D., Moyes, L., Graham, D.I.,
Cernak, I. (2005) Animal models of head trauma. NeuroRx., 2: Smith, C., Griffin, W.S. and Nicoll, J.A. (2004) Long-term
410–422. intracerebral inflammatory response after traumatic brain
Chatzipanteli, K., Wada, K., Busto, R. and Dietrich, W.D. injury. Forensic Sci. Int., 146: 97–104.
(1999) Effects of moderate hypothermia on constitutive and Graham, D.I., Gentleman, S.M., Lynch, A. and Roberts, G.W.
inducible nitric oxide synthase activities after traumatic brain (1995) Distribution of beta-amyloid protein in the brain fol-
injury in the rat. J. Neurochem., 72: 2047–2052. lowing severe head injury. Neuropathol. Appl. Neurobiol.,
Chatzipanteli, K., Yanagawa, Y., Marcillo, A.E., Kraydieh, S., 21: 27–34.
Yezierski, R.P. and Dietrich, W.D. (2000) Posttraumatic Graham, D.I., McIntosh, T.K., Maxwell, W.L. and Nicoll, J.A.
hypothermia reduces polymorphonuclear leukocyte accumu- (2000a) Recent advances in neurotrauma. J. Neuropathol.
lation following spinal cord injury in rats. J. Neurotrauma, Exp. Neurol., 59: 641–651.
17: 321–332. Graham, D.I., Raghupathi, R., Saatman, K.E., Meaney, D.
Chaudhuri, T.K. and Paul, S. (2006) Protein-misfolding dis- and McIntosh, T.K. (2000b) Tissue tears in the white matter
eases and chaperone-based therapeutic approaches. FEBS J., after lateral fluid percussion brain injury in the rat: relevance
273: 1331–1349. to human brain injury. Acta Neuropathol. (Berl.), 99:
Cortez, S.C., McIntosh, T.K. and Noble, L.J. (1989) Experi- 117–124.
mental fluid percussion brain injury: vascular disruption and Gris, D., March, D.R., Oatway, M.A., Chen, Y., Hamilton,
neuronal and glial alternations. Brain Res., 482: 271–282. E.F., Dekaban, G.A. and Weaver, L.C. (2004) Transient
Crowe, M.J., Bresnahan, J.C., Shuman, S.L., Masters, J.N. and blockade of the CD11d/CD18 integrin reduces secondary
Beattie, M.S. (1997) Apoptosis and delayed degeneration af- damage after spinal cord injury, improving sensory, auto-
ter spinal cord injury in rats and monkeys. Nat. Med., 3: nomic, and motor function. J. Neurosci., 24: 4043–4051.
73–76. Guest, J.D. and Dietrich, W.D. (2005) Spinal cord ischemia and
Cullum, C.M. and Bigler, E.D. (1986) Ventricle size, cortical trauma. In: Tisherman S.A. and Sterz F. (Eds.), Therapeutic
atrophy and the relationship with neuropsychological status Hypothermia. Springer, New York, pp. 101–118.
in closed head injury: a quantitative analysis. J. Clin. Exp. Guest, J.D., Hiester, E.D. and Bunge, R.P. (2005) Demyelina-
Neuropsychol., 8: 437–452. tion and Schwann cell responses adjacent to injury epicenter
Demjen, D., Klussmann, S., Kleber, S., Zulani, C., Stieltjes, B., cavities following chronic human spinal cord injury. Exp.
Metzger, C., Hirt, U.A., Walczak, H., Falk, W., Essig, M., Neurol., 192: 384–393.
Edler, L., Krammer, P.H. and Martin-Villalba, A. (2004) Neu- Hagg, T. and Oudega, M. (2006) Degenerative and spontane-
tralization of CD95 ligand promotes regeneration and func- ous regenerative processes after spinal cord injury. J. Neuro-
tional recovery after spinal cord injury. Nat. Med., 10: 389–395. trauma, 23: 264–280.
Dietrich, W.D., Alonso, O. and Halley, M. (1994) Early mi- Hains, B.C., Willis, W.D. and Hulsebosch, C.E. (2003) Tem-
crovascular and neuronal consequences of traumatic brain poral plasticity of dorsal horn somatosensory neurons after
injury: a light and electron microscopic study in rats. J. acute and chronic spinal cord hemisection in rat. Brain Res.,
Neurotrauma, 11: 289–301. 970: 238–241.
Dixon, C.E., Kochanek, P.M., Yan, H.Q., Schiding, J.K., Hamberger, A., Huang, Y.L., Zhu, H., Bao, F., Ding, M.,
Griffith, R.G., Baum, E., Marion, D.W. and DeKosky, S.T. Blennow, K., Olsson, A., Hansson, H.A., Viano, D. and
(1999) One-year study of spatial memory performance, brain Haglid, K.G. (2003) Redistribution of neurofilaments and
morphology, and cholinergic markers after moderate con- accumulation of beta-amyloid protein after brain injury by
trolled cortical impact in rats. J. Neurotrauma, 16: 109–122. rotational acceleration of the head. J. Neurotrauma, 20:
Dixon, C.E., Lyeth, B.G., Povlioshock, J.T., Findling, R.L., 169–178.
Hamm, R.J., Marmarou, A., Young, H.F. and Hayes, R.L. Hayashi, N., Bullock, R., Dietrich, W.D. and Maekawa, T.
(1987) A fluid percussion injury model of experimental brain (2004) Hypothermia for Acute Brain Damage. Springer, New
injury in the rat. J. Neurosurg., 67: 110–119. York.
Emery, E., Aldana, P., Bunge, M.B., Puckett, W., Srinivasan, Hill, C.E., Beattie, M.S. and Bresnahan, J.C. (2001) Degener-
A., Keane, R.W., Bethea, J. and Levi, A.D. (1998) Apoptosis ation and sprouting of identified descending supraspinal ax-
after traumatic human spinal cord injury. J. Neurosurg., 89: ons after contusive spinal cord injury in the rat. Exp. Neurol.,
911–920. 171: 153–169.
139

Hoffman, L.R. and Field-Fote, E.C. (2007) Cortical reorgan- Kinoshita, K., Chatzipanteli, K., Vitarbo, E., Truettner, J.S.,
ization following bimanual training and somatosensory Alonso, O.F. and Dietrich, W.D. (2002) Interleukin-1beta
stimulation in cervical spinal cord injury: a case report. messenger ribonucleic acid and protein levels after fluid-
Phys. Ther., 87: 208–223. percussion brain injury in rats: importance of injury severity
Holmin, S., Schalling, M., Hojeberg, B., Nordqvist, A.C., and brain temperature. Neurosurgery, 51: 195–203.
Skeftruna, A.K. and Mathiesen, T. (1997) Delayed cytokine Langlois, J.A., Rutland-Brown, W. and Thomas, K.E. (2000)
expression in rat brain following experimental contusion. J. Traumatic Brain Injury in the United States: Emergency De-
Neurosurg., 86: 493–504. partment Visits, Hospitalizations, and Deaths. Centers for
Hovda, D.A., Yoshino, A., Kawamata, T., Katayama, Y. and Disease Control and Prevention, National Center for Injury
Becker, D.P. (1991) Diffuse prolonged depression of cerebral Prevention and Control, Atlanta, GA.
oxidative metabolism following concussive brain injury in the Leclercq, P.D., McKenzie, J.E., Graham, D.I. and Gentleman,
rat: a cytochrome oxidase histochemistry study. Brain Res., S.M. (2001) Axonal injury is accentuated in the caudal corpus
567: 1–10. callosum of head-injured patients. J. Neurotrauma, 18: 1–9.
Hozler M., for the Hypothermia after Cardiac Arrest Study Lewen, A., Li, G.L., Nilsson, P., Olsson, Y. and Hillered, L.
Group. (2002) Mild therapeutic hypothermia to improve the (1995) Traumatic brain injury in rat produces changes of
neurological outcome after cardiac arrest. N. Engl. J. Med., beta-amyloid precursor protein immunoreactivity. Neurore-
346: 549–556. port, 6: 357–360.
Hubscher, C.H. and Johnson, R.D. (2006) Chronic spinal cord Li, S., Jiang, Q. and Stys, P.K. (2000) Important role of reverse
injury induced changes in the responses of thalamic neurons. Na(+)-Ca(2+) exchange in spinal cord white matter injury
Exp. Neurol., 197: 177–188. at physiological temperature. J. Neurophysiol., 84:
Jain, N., Catania, K.C. and Kaas, J.H. (1997) Deactivation and 1116–1119.
reactivation of somatosensory cortex after dorsal spinal cord LoPachin, R.M. and Lehning, E.J. (1997) Mechanism of cal-
injury. Nature, 386: 495–498. cium entry during axon injury and degeneration. Toxicol.
Jiang, J.Y., Xu, W., Li, W.P., Gao, G.Y., Bao, Y.H., Liang, Appl. Pharmacol., 143: 233–244.
Y.M. and Luo, Q.Z. (2006) Effect of long-term mild hypo- Lotocki, G., Alonso, O.F., Dietrich, W.D. and Keane, R.W.
thermia or short-term mild hypothermia on outcome of pa- (2004) TNF receptor 1 and its signaling intermediates are
tients with severe traumatic brain injury. J. Cerebral Blood recruited to lipid rafts in the traumatized brain. J. Neurosci.,
Flow Metab., 26: 771–776. 24: 11010–11016.
Karimi-Abdolrezaee, S., Eftekharpour, E., Wang, J., Mors- Lotze, M., Laubis-Herrmann, U. and Topka, H. (2006) Com-
head, C.M. and Fehlings, M.G. (2006) Delayed transplanta- bination of TMS and fMRI reveals a specific pattern of re-
tion of adult neural precursor cells promotes remyelination organization in M1 in patients after complete spinal cord
and functional neurological recovery after spinal cord injury. injury. Restor. Neurol. Neurosci., 24: 97–107.
J. Neurosci., 26: 3377–3389. Lu, J., Moochhala, S., Shirhan, M., Ng, K.C., Tan, M.H., Teo,
Keane, R.W., Davis, A.R. and Dietrich, W.D. (2006) Inflam- A.L. and Ling, E.A. (2003) Nitric oxide induces macrophage
matory and apoptotic signaling after spinal cord injury. apoptosis following traumatic brain injury in rats. Neurosci.
J. Neurotrauma, 23: 335–344. Lett., 339: 147–150.
Keane, R.W., Kraydieh, S., Lotocki, G., Alonso, O.F., Aldana, McDonald, J.W., Becker, D., Sadowsky, C.L., Jane, Sr., J.A.,
P. and Dietrich, W.D. (2001a) Apoptotic and antiapoptotic Conturo, T.E., Schultz, L.M. (2002) Late recovery following
mechanisms after traumatic brain injury. J. Cereb. Blood spinal cord injury. Case report and review of the literature.
Flow Metab., 21: 1189–1198. J. Neurosurg., 97(Suppl 2): 252–265
Keane, R.W., Kraydieh, S., Lotocki, G., Bethea, J.R., Krajew- Medana, I.M. and Esiri, M.M. (2003) Axonal damage: a key
ski, S., Reed, J.C. and Dietrich, W.D. (2001b) Apoptotic and predictor of outcome in human CNS diseases. Brain, 126:
anti-apoptotic mechanisms following spinal cord injury. 515–530.
J. Neuropathol. Exp. Neurol., 60: 422–429. Morganti-Kossman, M.C., Lenzlinger, P.M., Hans, V., Stahel,
Keirstead, H.S., Nistor, G., Bernal, G., Totoiu, M.O., Cloutier, P., Csuka, E., Ammann, E., Stocker, R., Trentz, O. and
F., Sharp, K. and Steward, O. (2005) Human embryonic stem Kossmann, T. (1997) Production of cytokines following brain
cell-derived oligodendrocyte progenitor cell transplants injury: beneficial and deleterious for the damaged tissue.
remyelinate and restore locomotion after spinal cord injury. Mol. Psychiatry, 2: 133–136.
J. Neurosci., 25: 4694–4705. Morganti-Kossmann, M.C., Rancan, M., Stahel, P.F. and
Kim, B.G., Dai, H.N., McAtee, M., Vicini, S. and Bregman, Kossmann, T. (2002) Inflammatory response in acute trau-
B.S. (2006) Remodeling of synaptic structures in the motor matic brain injury: a double-edged sword. Curr. Opin. Crit.
cortex following spinal cord injury. Exp Neurol., 198: Care, 8: 101–105.
401–415. van der Naalt, J., Hew, J.M., van Zomeren, A.H., Sluiter, W.J.
Kimbelberg, H.K. and Norenberg, M.D. (1994) Astroglial re- and Minderhoud, J.M. (1999) Computed tomography and
sponse to CNS trauma. In: Salzman S.K. and Faden A.I. magnetic resonance imaging in mild to moderate head injury:
(Eds.), The Neurobiology of Central Nervous System early and late imaging related to outcome. Ann. Neurol., 46:
Trauma. Oxford University Press, New York, pp. 193–208. 70–78.
140

Narayan, R.K., Michel, M.E., Ansell, B., Baethmann, A., Raineteau, O., Fouad, K., Noth, P., Thallmair, M. and
Biegon, A., Bracken, M.B., Bullock, M.R., Choi, S.C., Schwab, M.E. (2001) Functional switch between motor
Clifton, G.L., Contant, C.F., Coplin, W.M., Dietrich, W.D., tracts in the presence of the mAb IN-1 in the adult rat.
Ghajar, J., Grady, S.M., Grossman, R.G., Hall, E.D., Proc. Natl. Acad. Sci. USA, 98: 6929–6934.
Heetderks, W., Hovda, D.A., Jallo, J., Katz, R.L., Knoller, Rancan, M., Bye, N., Otto, V.I., Trentz, O., Kossmann, T.,
N., Kochanek, P.M., Maas, A.I., Majde, J., Marion, D.W., Frentzel, S. and Morganti-Kossmann, M.C. (2004) The
Marmarou, A., Marshall, L.F., McIntosh, T.K., Miller, E., chemokine fractalkine in patients with severe traumatic brain
Mohberg, N., Muizelaar, J.P., Pitts, L.H., Quinn, P., injury and a mouse model of closed head injury. J. Cereb.
Riesenfeld, G., Robertson, C.S., Strauss, K.I., Teasdale, G., Blood Flow Metab., 24: 1110–1118.
Temkin, N., Tuma, R., Wade, C., Walker, M.D., Weinrich, Ray, S.K., Hogan, E.L. and Banik, N.L. (2003) Calpain in the
M., Whyte, J., Wilberger, J., Young, A.B. and Yurkewics, L. pathophysiology of spinal cord injury: neuroprotection with
(2002) Clinical trials in head injury. J. Neurotrauma, 19: calpain inhibitors. Brain Res. Rev., 42: 169–185.
503–557. Rink, A., Fung, K.M., Trojanowski, J.Q., Lee, V.M.,
Nashmi, R. and Fehlings, M.G. (2001) Mechanisms of axonal Neugebauer, E. and McIntosh, T.K. (1995) Evidence of
dysfunction after spinal cord injury: with an emphasis on the apoptotis cell death after experimental traumatic brain
role of voltage-gated potassium channels. Brain Res. Brain injury. Am. J. Pathol., 147: 1575–1583.
Res. Rev., 38: 65–191. Rodriguez-Paez, A.C., Brunschwig, J.P. and Bramlett, H.M.
Nonaka, M., Chen, X.H., Pierce, J.E., Leoni, M.J., McIntosh, (2005) Light and electron microscopic assessment of pro-
T.K., Wolf, J.A. and Smith, D.H. (1999) Prolonged activa- gressive atrophy following moderate traumatic brain injury
tion of NF-kappaB following traumatic brain injury in rats. in the rat. Acta Neuropathol., 109: 603–616.
J. Neurotrauma, 16: 1023–1034. Rosenberg, L.J. and Wrathall, J.R. (1997) Quantitative analysis
Norenberg, M.D., Smith, J. and Marcillo, A. (2004) The pa- of acute axonal pathology in experimental spinal cord con-
thology of human spinal cord injury: defining the problems. tusion. J. Neurotrauma, 14: 823–838.
J. Neurotrauma, 21: 429–440. Rothwell, N.J. and Luheshi, G.N. (2000) Interleukin 1 in the
Ozawa, H., Keane, R.W., Marcillo, A.E., Diaz, P.H. and brain: biology, pathology and therapeutic target. TINS, 23:
Dietrich, W.D. (2002) Therapeutic strategies targeting 618–625.
caspase inhibition following spinal cord injury in rats. Exp. Sabbah, P., De, S.S., Leveque, C., Gay, S., Pfefer, F., Nioche,
Neurol., 177: 306–313. C., Sarrazin, J.L., Barouti, H., Tadie, M. and Cordoliani,
Pettus, E.H. and Povlishock, J.T. (1996) Characterization of a Y.S. (2002) Sensorimotor cortical activity in patients with
distinct set of intra-axonal ultrastructural changes associated complete spinal cord injury: a functional magnetic resonance
with traumatically induced alteration in axolemmal perme- imaging study. J. Neurotrauma, 19: 53–60.
ability. Brain Res., 722: 1–11. Schouten, J.W., Fulp, C.T., Royo, N.C., Saatman, K.E.,
Pierce, J.E., Smith, D.H., Trojanowski, J.Q. and McIntosh, Watson, D.J., Snyder, E.Y., Trojanowski, J.Q., Prockop,
T.K. (1998) Enduring cognitive, neurobehavioral and histo- D.J., Maas, A.I. and McIntosh, T.K. (2004) A review and
pathological changes persist for up to one year following rationale for the use of cellular transplantation as a thera-
severe experimental brain injury in rats. Neuroscience, 87: peutic strategy for traumatic brain injury. J. Neurotrauma,
359–369. 21: 1501–1538.
Popovich, P.G., Guan, Z., McGaughy, V., Fisher, L., Hickey, Schwab, M.E. and Bartholdi, D. (1996) Degeneration and
W.F. and Basso, D.M. (2002) The neuropathological and regeneration of axons in the lesioned spinal cord. Physiol.
behavioral consequences of intraspinal microglial/macroph- Rev., 76: 319–370.
age activation. J. Neuropathol. Exp. Neurol., 61: 623–633. Schwab, J.M., Brechtel, K., Mueller, C.A., Failli, V., Kaps,
Povlishock, J.T. and Christman, C.W. (1995) The pathobiology H.P., Tuli, S.K. and Schluesener, H.J. (2006) Experimental
of traumatically induced axonal injury in animals and hu- strategies to promote spinal cord regeneration — an integra-
mans: a review of current thoughts. J. Neurotrauma, 12: tive perspective. Prog. Neurobiol., 78: 91–116.
555–564. Schwab, J.M., Seid, K. and Schluesener, H.J. (2001) Traumatic
Povlishock, J.T. and Pettus, E.H. (1996) Traumatically induced brain injury induces prolonged accumulation of cyclooxy-
axonal damage: evidence for enduring changes in axolemmal genase-1 expressing microglia/brain macrophages in rats. J.
permeability with associated cytoskeletal change. Acta Neurotrauma, 18: 881–890.
Neurochir. Suppl., 66: 81–86. Sherriff, F.E., Bridges, L.R. and Sivaloganathan, S. (1994)
Quencer, R.M. and Bunge, R.P. (1996) The injured spinal cord: Early detection of axonal injury after human head trauma
imaging, histopathologic clinical correlates, and basic science using immunocytochemistry for beta-amyloid precursor pro-
approaches to enhancing neural function after spinal cord tein. Acta Neuropathol. (Berl.), 87: 55–62.
injury. Spine, 21: 2064–2066. Shohami, E., Novikov, M., Bass, R., Yamin, A. and Gallily, R.
Raineteau, O. and Schwab, M.E. (2001) Plasticity of motor (1994) Closed head injury triggers early production of TNF
systems after incomplete spinal cord injury. Nat. Rev. alpha and IL-6 by brain tissue. J. Cereb. Blood Flow Metab.,
Neurosci., 2: 263–273. 14: 615–619.
141

Smith, D.H., Chen, X.H., Pierce, J.E., Wolf, J.A., Trojanowski, Wallace, M.C., Tator, C.H. and Lewis, A.J. (1987)
J.Q., Graham, D.I. and McIntosh, T.K. (1997) Progressive Chronic regenerative changes in the spinal cord after
atrophy and neuron death for one year following brain cord compression injury in rats. Surg. Neurol., 27:
trauma in the rat. J. Neurotrauma, 14: 715–727. 209–219.
Snow, D.M., Lemmon, V., Carrino, D.A., Caplan, A.I. and Waxman, S.G. (1989) Demyelination in spinal cord injury. J.
Silver, J. (1990) Sulfated proteoglycans in astroglial barriers Neurol. Sci., 91: 1–14.
inhibit neurite outgrowth in vitro. Exp. Neurol., 109: 111–130. Waxman, S.G., Utzschneider, D.A. and Kocsis, J.D. (1994)
Springer, J.E., Azbill, R.D. and Knapp, P.E. (1999) Activation Enhancement of action potential conduction following de-
of the caspase-3 apoptotic cascade in traumatic spinal cord myelination: experimental approaches to restoration of func-
injury. Nat. Med., 5: 943–946. tion in multiple sclerosis and spinal cord injury. Prog. Brain
Thuret, S., Moon, L.D. and Gage, F.H. (2006) Therapeutic Res., 100: 233–243.
interventions after spinal cord injury. Nat. Rev. Neurosci., 7: Williams, S., Raghupathi, R., MacKinnon, M.A., McIntosh,
628–643. T.K., Saatman, K.E. and Graham, D.I. (2001) In situ
Topka, H., Cohen, L.G., Cole, R.A. and Hallet, M. (1991) DNA fragmentation occurs in white matter up to 12 months
Reorganization of corticospinal pathways following spinal after head injury in man. Acta Neuropathol. (Berl.), 102:
cord injury. Neurology, 41: 1276–1283. 581–590.
Totoiu, M.O. and Keirstead, H.S. (2005) Spinal cord injury is Yu, C.G., Jimenez, O., Marcillo, A.E., Weider, B., Bangerter,
accompanied by chronic progressive demyelination. J. Comp. K., Dietrich, W.D., Castro, S. and Yezierski, R.P. (2000)
Neurol., 486: 373–383. Beneficial effects of modest systemic hypothermia on loco-
Toulmond, S. and Rothwell, N.J. (1995) Interleukin-1 receptor motor function and histopathological damage following con-
antagonist inhibits neuronal damage caused by fluid percus- tusion-induced spinal cord injury in rats. J. Neurosurg.,
sion injury in the rat. Brain Res., 671: 261–266. 93(Suppl 1): 85–93.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 10

Injury-induced alterations in CNS


electrophysiology $

Akiva S. Cohen1,, Bryan J. Pfister2, Elizabeth Schwarzbach3, M. Sean Grady4,


Paulette B. Goforth5 and Leslie S. Satin5

1
Department of Pediatrics, University of Pennsylvania, School of Medicine and Division of Neurology, Children’s Hospital
of Philadelphia, Philadelphia, PA, USA
2
Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
3
Department of Pharmacology, University of Pennsylvania, School of Medicine, Philadelphia, PA, USA
4
Department of Neurosurgery, University of Pennsylvania, School of Medicine, Philadelphia, PA, USA
5
Department of Pharmacology and Toxicology, VCU School of Medicine, Virginia Commonwealth University, Richmond,
VA, USA

Abstract: Mild to moderate cases of traumatic brain injury (TBI) are very common, but are not always
associated with the overt pathophysiogical changes seen following severe trauma. While neuronal death has
been considered to be a major factor, the pervasive memory, cognitive and motor function deficits suffered
by many mild TBI patients do not always correlate with cell loss. Therefore, we assert that functional
impairment may result from alterations in surviving neurons. Current research has begun to explore CNS
synaptic circuits after traumatic injury. Here we review significant findings made using in vivo and in vitro
models of TBI that provide mechanistic insight into injury-induced alterations in synaptic electrophysio-
logy. In the hippocampus, research now suggests that TBI regionally alters the delicate balance between
excitatory and inhibitory neurotransmission in surviving neurons, disrupting the normal functioning of
synaptic circuits. In another approach, a simplified model of neuronal stretch injury in vitro, has been used
to directly explore how injury impacts the physiology and cell biology of neurons in the absence of
alterations in blood flow, blood brain barrier integrity, or oxygenation associated with in vivo models of
brain injury. This chapter discusses how these two models alter excitatory and inhibitory synaptic trans-
mission at the receptor, cellular and circuit levels and how these alterations contribute to cognitive
impairment and a reduction in seizure threshold associated with human concussive brain injury.

Keywords: TBI; electrophysiology; hippocampus; cortical neurons; excitation; inhibition; stretch injury;
synapse

Introduction upon healthcare systems worldwide. Many who


survive the initial traumatic incident endure conti-
Traumatic brain injury (TBI) is a major public nuing problems with memory, cognition and
health problem, which has had a significant impact motor function that are debilitating to their daily
$ lives. While TBI has been extensively studied from
Supported by NIH-NINDS RO1 NS 45975 (ASC), Common-
wealth Neurotrauma Initiative Fund CNI 04-094 (LSS). both a clinical and a basic science standpoint,
Corresponding author. Tel.: +1-215-590-1472; many of these studies are descriptive rather than
E-mail: Cohena@email.chop.edu mechanistic, often focusing on the behavioral

DOI: 10.1016/S0079-6123(06)61010-8 143


144

consequences and neuronal cell loss associated understanding how injury impacts the physiology
with severe TBI in human patients or animal and cell biology of neurons and neural circuits. We
models. However, the extensive and widespread believe that comparing and contrasting results
primary neuronal destruction seen in severe cases obtained using these two very different models is
of TBI is generally irreversible and thus, in all productive and leads to a deeper understanding of
likelihood, therapeutically intractable. the mechanisms evoked by physical trauma. In the
Mild and moderate TBI, on the other hand, is last section, we summarize what has been learned
much more commonly encountered than severe to date, and suggest some directions for future
cases. From the authors’ standpoint, the pervasive research.
deficits associated with mild injury do not neces-
sarily require the frank loss of neuronal or glial cells
in the CNS, but result from the disruption of the In vivo models of TBI
synaptic function of surviving neurons, negatively
impacting the brain function of TBI patients. It is Fluid percussion injury
our contention that understanding how the delicate
balance of excitation and inhibition in CNS synap- LFPI is a reliable and reproducible experimental
tic circuits is disturbed after TBI will help explain model of concussive TBI implemented in rodents.
and ultimately treat disturbances of cognition and LFPI is widely used since it reproduces many
memory in mildly and moderately injured patients. pathophysiological features of mild to moderate
In contrast to the large number of histological, be- human TBI including blood brain barrier break-
havioral or biochemical studies that have appeared down, neuronal cell loss, gliosis, perturbation of
in the literature to date, there have been relatively ionic homeostasis and long-term alterations in
few studies of CNS electrophysiology after trauma. cognitive and motor function (Dixon et al., 1987;
The aim of the present review, is to emphasize and McIntosh et al., 1989; Thompson et al., 2005). In
discuss the significant progress made recently using this model, a pendulum strikes a piston sending a
in vivo and in vitro models of TBI. Furthermore, to brief (15 ms) percussion pulse of saline into the
understand the mechanisms of brain injury at the extradural space of the closed cranial cavity caus-
level of synaptic electrophysiology, we highlight the ing a brief displacement and deformation of the
need for more mechanistically designed studies of brain mimicking concussive injury. The severity of
synaptic electrophysiology in the future. injury is set by adjusting the height of the pendu-
The chapter is divided into two major sections. lum. LFPI results in both focal as well as diffuse
The first part is concerned with how TBI results brain injury consisting of a cortical contusion at
in specific changes in neuronal function that dis- the impact site and cell death and axonal damage
rupt the function of synaptic circuits in the brain, in the hippocampus, thalamus and cortex (Dixon
affecting the balance between excitatory and in- et al., 1987; McIntosh et al., 1989; Carbonell et al.,
hibitory neurotransmission. Here we focus on 1998; Thompson et al., 2005). LFPI has been ex-
studies of hippocampal slices from rodents sub- tensively characterized and widely used in rats as a
jected to lateral fluid percussion injury (LFPI) model, but has recently been adapted for use in
in vivo, a widely used experimental model of mice (Carbonell et al., 1998; Witgen et al., 2005).
TBI (Dixon et al., 1987; McIntosh et al., 1989; Importantly, the use of mice to study TBI will al-
Thompson et al., 2005). In the second part, we low investigators to exploit transgenic and knock-
discuss studies obtained using a simplified model out mouse technology to directly test the role of
of TBI, where cortical pyramidal neurons, co- specific genes or gene mutations on TBI, as well as
cultured with glia, are subjected to a defined lowering the costs associated with the use of rats.
degree of tensile stretch in vitro. Use of this model In this review chapter, we will include data
has allowed perturbations to be delivered in the obtained using fluid percussion to injure mice to
absence of changes in blood flow, blood brain demonstrate that these results are consistent with
barrier integrity or oxygenation, and facilitate the rat LFPI model.
145

Experimental LFPI and the hippocampus including impaired cognitive function and the
development of seizures (Cave and Squire, 1991;
In humans, TBI results in heterogeneous changes in Annegers et al., 1996; Asikainen et al., 1999).
brain function, including cognitive impairments in Within the hippocampus there are three intercon-
learning and memory. Damage to the hippocampus nected subregions referred to collectively as the
associated with significant impairment of learning ‘Trisynaptic Circuit’. This circuit consists of the
and memory was first recognized in studies of bi- dentate gyrus (DG), area CA3, and area CA1, each
lateral damage to the hippocampus in humans having distinct physiological roles (see Fig. 1). The
(Rempel-Clower et al., 1996; Deweer et al., 2001). DG ‘‘filters’’ out aberrant or excessive input to
Subsequently, a large body of knowledge has ac- the hippocampus from propagating further along
cumulated indicating that, while not exclusively re- the hippocampal circuit. Area CA3 ‘‘amplifies’’ the
sponsible for memory function, the hippocampus activity that the DG has allowed to enter, while
plays a central role in memory consolidation and area CA1 ‘‘transduces’’ processed hippocampal in-
retrieval (Cave and Squire, 1991; Miller et al., 1998; formation outflow to the cerebral cortex.
Bohbot et al., 2000). Moreover, the hippocampus is It is believed that the DG acts as a filter because
frequently damaged in TBI patients, thus likely it is itself extraordinarily resistant to the genera-
contributing to observed cognitive deficits includ- tion of synchronized bursting that is characteristic
ing impaired learning and memory (Kotapka et al., of epileptic seizures (Heinemann et al., 1992;
1992, 1993, 1994; Bigler et al., 1997). Lothman et al., 1992). This dampening of synap-
The dependence of memory acquisition on tic input may thus be critical in protecting the
the hippocampus is similar in humans and rats, fragile neurons within the hippocampus from
which both show selective loss of recently ac- excessive activation and seizure formation. The
quired memory and the failure to imprint new filtering or gatekeeper behavior of the DG is due
memories after hippocampal damage (He et al., to three main mechanisms: (1) the intrinsic mem-
2001; Liu and Bilkey, 2001). In rodents, LFPI brane properties of dentate granule cells (DGCs)
produces a consistent injury to the hippocampus tend to resist excessive excitation (Fricke and
including neuronal loss within all hippocampal Prince, 1984), (2) at the circuit level, the dearth of
subregions as well as physiologic, ionic and neuro- interconnections between DGCs impedes synchro-
chemical changes in the dentate hilus and nization since neurons firing inappropriately
areas CA1 and CA3. This damage has been cor- cannot easily transmit this overexcitation to their
related to visuo-spatial memory deficits observed neighbors (Claiborne et al., 1986) and (3) the effi-
in the Morris water maze assay (Smith et al., cacy of DG-mediated filtering is further enhanced
1991; Gorman et al., 1993) and to hippocampal- by robust surround inhibition of the DG (Sloviter,
dependent cognitive impairment demonstrated by 1994).
changes in the conditioned fear response (Hogg In area CA3, pyramidal neurons are highly in-
et al., 1998a, b; Witgen et al., 2005). Accordingly, terconnected to each other via recurrent axon col-
LFPI in the rat and mouse is currently the most laterals (Dudek et al., 1986; Wong et al., 1986),
widely accepted experimental model to mimic hip- providing the anatomical substrate for efficacious
pocampal injury. synchronization and signal amplification. Thus,
area CA3 is highly susceptible to recurrent excita-
tion and is consequently prone to seizure genera-
Functional electrophysiological changes in the tion and injury-related pathology (i.e., if the DG
hippocampus after TBI does not filter excessive input). This activity is in
turn transmitted via CA3 axons (Schaffer collat-
Normal hippocampal function is directly deter- erals) to area CA1, which excite both pyramidal
mined by a delicate balance between neuronal ex- neurons as well as inhibitory interneurons, thereby
citation and inhibition. Perturbation of this state of regulating both inhibition and excitation in area
equilibrium can have catastrophic consequences, CA1.
146

Fig. 1. Illustration of the hippocampal ‘Trisynaptic circuit’. This circuit consists of three interconnected subregions: the dentate gyrus
(DG), area CA3 and area CA1. The DG is thought to filter aberrant or excessive input from the entorhinal cortex via the perforant
pathway. Filtered activity is relayed to area CA3 via the mossy fibers where it is amplified and sent onward to area CA1 via the Schaffer
collaterals where the information is then transduced out to the cerebral cortex.

The role of GABAergic inhibition in the initiating, synchronizing and terminating both
hippocampus normal and pathological neuronal network acti-
vity in the central nervous system. A majority of
Pathological alterations in the effectiveness of inhibitory synapses are localized on neuronal
GABAA-mediated inhibition are implicated in somata and axon initial segments, since synaptic
many neurological disease processes, including input to these regions are highly efficacious in
TBI (Reeves et al., 1995; Toth et al., 1997; regulating neuronal output. Synaptic localization,
Santhakumar et al., 2001). GABA is the principal coupled with the heterogeneous patterns of in-
inhibitory neurotransmitter in the mammalian terneuronal innervation of excitatory (principal)
brain and not only controls the degree of neuronal neurons (Freund and Buzsaki, 1996), endows
activation but, importantly, is also critical in individual GABAAergic interneurons with the
147

capability of synchronizing the activity of hun- Injury-induced functional alterations in area CA1
dreds to thousands of principal neurons within
the hippocampus (Cobb et al., 1995). The syn- Area CA1: injury-induced alterations in
chronization of neocortical structures is known to hippocampal area CA1 excitability
subserve important functions, such as the process-
ing of sensory information (Lytton and Sejnowski, To evaluate hippocampal function after injury,
1991; Singer and Gray, 1995) and memory con- several groups have specifically investigated
solidation and formation (Joliot et al., 1994). changes in area CA1 excitability. Electrophysio-
Therefore, oscillations which are synchronized by logical studies on hippocampal slices from injured
GABAAergic inhibition can be viewed as being rodents using LFPI generally show a decrease in
critical for normal hippocampal function. excitability in hippocampal area CA1. D’Ambrosio
GABAA receptors are pentameric structures et al. (1998) initially found, 2 days post-LFPI, that
composed of several related subunit families. At increased simulation intensity is required to over-
present, eight different GABAA receptor subunit come the threshold of postsynaptic population
families have been cloned (with many families spikes (PS) as well as field extracellular postsynap-
having multiple members), including a (1–6), b tic potentials (fEPSP). In addition, the potentials
(1–4), g (1–3), d, r(1–3), e, p and y (Barnard et al., exhibit smaller amplitudes and decreased linear
1998). The subunit stoichiometry is considered to slopes after injury. These results suggest a decrease
be a primary determinant of the pharmacology of in area CA1 excitability, which was further corro-
the receptor. Although subunit composition is borated and expanded upon using mice 1 week
modulable under different developmental and after LFPI (Witgen et al., 2005). Input/output
pathological conditions, to date no injury-induced (I/O) curves, relating the stimulus intensity as a
alterations in GABAA receptor subunit composi- function of the linear slope of the fEPSP are shifted
tion have been reported. downward after injury compared with curves
Surprisingly, there have been few studies ex- obtained from naı̈ve/sham mice, Fig. 2 (Witgen
ploring functional changes in hippocampal cir- et al., 2005). In addition, the threshold for evoking
cuitry following TBI. Instead, most studies to date PS in slices from injured animals is correspondingly
have attempted to correlate regional hippocampal increased. The observed shifts in the I/O curve and
cell loss with behavioral changes induced by TBI, the PS threshold denote a decrease in excitability,
despite the gap between these two divergent end- and are indicative of diminished synaptic commu-
points. While hippocampal cell loss is often hypo- nication between pre and postsynaptic neurons.
thesized to alter the balance between neuronal While these studies in hippocampal slices estab-
excitation and inhibition which is necessary for lish reduced excitability in area CA1 after LFPI,
proper hippocampal function, iterations in func- earlier in vivo studies reported increased excitabil-
tion do not always correlate with the degree of ity during the acute period of 2 h–2 days following
observed cell death (Lyeth et al., 1990). Recent injury. Stimulation thresholds for the generation
studies have supported the alternative hypothesis of PS in vivo decreases within 2–3 h after LFPI
that alterations in the behavior of surviving neu- (Miyazaki et al., 1992), and the decrease persists
rons may be a key. Specifically, a few experimental for 2 days, returning to control levels by 7 days
studies using LFPI indicate that TBI affects exci- after LFPI (Reeves et al., 1995, 1997b). Although
tatory and inhibitory synaptic transmission, giving lowered thresholds suggest increased excitability,
rise to dysfunctional hippocampal circuits in ro- the amplitudes of the PS and fEPSPs are reduced
dents despite the absence of significant cell death compared with controls. The discrepant nature of
(Reeves et al., 1995; Toth et al., 1997; D’Ambrosio these results could be explained by differences in
et al., 1998; Witgen et al., 2005). However, the injury severity in the respective studies, the use of
precise cellular mechanism(s) responsible for the in vivo vs. in vitro slice preparations, differences in
TBI-induced pathological alterations in hippocam- the recording techniques used, or the difference in
pal function currently remain unknown. post-injury time points chosen. An additional
148

Fig. 2. Extracellular input/output curves for area CA1 evoked by afferent Schaffer collateral stimulation. LFPI in mice results in
smaller fEPSP slopes compared with those from sham animals. Reprinted from Neuroscience, Witgen et al. (2005), with permission
from Elsevier.

report recently suggested increased excitability of NMDA-mediated depolarizing potentials (AMPA-


area CA1 at 1 week after LFPI (Akasu et al., 2002). and GABAergic activity were pharmacologically
blocked) (Cohen and Abraham, 1996) in brain slices
from sham vs. LFPI mice, Fig. 3 (Schwarzbach
Area CA1: Glutamate evoked currents et al., 2006). These data show that in mild to mod-
erately injured mice, the amplitude of NMDA
Alterations in excitatory synaptic transmission are potentials (Figs. 3B, C) is significantly smaller than
one potential mechanism underlying the injury- in slices from sham animals (Figs. 3A, C). To
induced shifts in area CA1 excitability seen evaluate NMDA receptor alterations in individual
following TBI. Supporting the hypothesis that hippocampal neurons, whole-cell patch voltage-
excitatory neurotransmission is decreased after clamp recordings were performed in visually identi-
injury, a variety of immunocytochemical studies fied pyramidal neurons in hippocampal slices
of the hippocampus indicate that the number of (Schwarzbach et al., 2006). Focal application of
N-methyl-D-aspartate (NMDA) receptors decreases glutamate results in significantly smaller NMDA
within 12 –24 h post-injury, with no associated isolated excitatory current (sodium, AMPA and
changes in non-NMDA receptors such as AMPA GABA currents blocked) in slices from LFPI
or kainate receptors (Miller et al., 1990; Sihver (Fig. 3E) vs. sham (Fig. 3D) mice. Furthermore,
et al., 2001; Osteen et al., 2004). by subtracting the NMDA-mediated currents from
The NMDA receptor is important for the induc- the total glutamate-evoked currents, the AMPA-
tion of excitatory synaptic plasticity (see below). ergic component could be determined. This
Therefore, to examine whether injury causes demonstrates there is a significant reduction in
NMDA receptor dysfunction, extracellular record- AMPA as well as NMDA currents in LFPI slices
ing techniques were used to measure evoked compared with sham controls (Fig. 3G).
149

FPI SHAM

D E

BMI BMI
TTX APV TTX APV
CNQX CNQX
glycine glycine

40 80
F G

20 40
* *

0 0
FPI SHAM FPI SHAM

Fig. 3. Injury diminishes isolated NMDA receptor potentials and EPSCs following injury. (A) Sham; (B) FPI, trace of fEPSP, isolated
NMDA potential, recording after addition of APV to ensure the waveform is NMDA dependent. Scale bar: 0.5 mV, 10 ms.
(C) Histogram showing the quantification of the reduction in the peak amplitude of the NMDA potential recorded in slices from FPI
mice compared with sham animals (n ¼ 11 and 7 for FPI and sham, respectively, po0.05, denoted by *). (D) Sham; (E) FPI,
glutamatergic currents were recorded by focal application of glutamate (100 mM, 50 ms, 40 psi), the NMDA component was isolated
with the addition of CNQX to the bath, and at the end of each experiment, APV was perfused to ensure the current is NMDA-
dependent. Scale bar: 15 pA, 100 ms. (F) The histogram shows FPI causes a significant reduction in the peak amplitude of the NMDA
current. (G) The histogram is created by subtracting the isolated NMDA-mediated current from the entire glutamate evoked current to
determine the AMPA-mediated current. This shows FPI also causes a significant reduction in the peak amplitude of the AMPA
currents (* denotes significance, po0.05).

Area CA1: changes in inhibitory synaptic termination of excitatory neuronal activity. There-
transmission fore, augmentation of inhibitory activity could
also contribute to decreased area CA1 excitability
While we have presented evidence showing (Fig. 2). Furthermore, because synaptic plasticity,
decreases in excitatory synaptic transmission after especially long-term potentiation (LTP), requires a
injury, alterations in overall synaptic excitation may precise functional balance of inhibition and excita-
also reflect changes in inhibitory circuits as these tion, alterations in either excitation or inhibition
effects may be additive postsynaptically. Specifi- may interfere with functional information process-
cally, interneurons innervate CA1 pyramidal ing. A role for GABAA alteration as a cause for
neurons to provide GABAA-mediated inhibition memory impairments is supported by the findings
control over the initiation, synchronization and that GABAA receptor immunolocalization in area
150

CA1 increases after LFPI (Reeves et al., 1997a, b) hundreds of synapses (Mody et al., 1994). Specifi-
and that the administration of GABAA receptor cally, changes in the number of postsynaptic recep-
antagonists reduces cognitive deficits in rats after tors are generally accepted to cause changes in
FPI (O’Dell and Hamm, 1995). mIPSC amplitude, while shifts in receptor subunit
To test whether LFPI leads to augmented composition and/or GABA release kinetics alter
GABAA-mediated inhibition, miniature inhibitory mIPSC kinetics. In addition, a change in the
postsynaptic currents (mIPSCs) were recorded in total number of active synapses (i.e., presynaptic
slices from sham vs. injured mice 1 week post- release sites) is reflected by variations in mIPSC
injury. Miniature IPSCs are spontaneously occur- frequency.
ring events, reflecting the spontaneous release of The median mIPSC amplitude in CA1 neurons
single packets of GABA from individual presy- recorded from FPI animals is significantly greater
naptic terminals of GABAergic interneurons. than in sham animals (Figs. 4A, B) (Witgen et al.,
Analysis of mIPSCs provides detailed information 2005). The significant increase in mIPSC ampli-
about the properties of postsynaptic receptors tude is further demonstrated by calculating total
because mIPSCs result from the activation of mIPSC charge transfer (i.e., the amount of current
individual synapses, in contrast to stimulation flowing during an individual mIPSC and calcu-
studies which involve the activation of tens to lated by integrating the total area associated with

Fig. 4. Whole-cell voltage clamp recordings from area CA1 pyramidal neurons from FPI animals demonstrate increases in spon-
taneous miniature (mIPSC) activity 1 week after LFPI. (A) Cumulative frequency amplitude histogram for neurons from sham and
LFPI animals. (B) The mIPSCs are larger in CA1 pyramidal neurons from LFPI mice. Histograms of mean median (C) mIPSC
amplitude and (D) net charge transfer demonstrate significant increases in inhibitory activity of neurons from LFPI mice compared
with control animals. (E) Histogram demonstrating that mIPSC frequency is not significantly different in sham compared with LFPI
slices. Reprinted from Neuroscience, Witgen et al. (2005), with permission from Elsevier.
151

an mIPSC) from the two populations of neurons. memory deficits observed following TBI (Lyeth
Miniature IPSC charge transfer in neurons in et al., 1990; Smith et al., 1991; Gorman et al.,
slices from FPI animals is 155% of that calculated 1993).
for the sham population (Fig. 4D). Interestingly, The complement of LTP is long-term depression
neither the 50% decay time (T50), weighted (LTD), which is a decrease in net synaptic efficacy
decay or the frequency of occurrence of mIPSCs seen after low frequency stimulation (Christie et
recorded in slices from injured animals are signifi- al., 1994; Bear and Abraham, 1996). Unlike LTP,
cantly different from values obtained in slices from this form of synaptic plasticity can be generated in
sham animals (Fig. 4E) suggesting that concussive slices from LFPI mice and rats (D’Ambrosio et al.,
brain injury does not alter the number of active 1998; Schwarzbach et al., 2006), demonstrating
presynaptic inhibitory synapses or GABAA recep- the ability of neurons to express some forms of
tor subunit composition. The enhanced action synaptic plasticity even after LFPI.
potential-independent spontaneous inhibitory ac-
tivity in area CA1 following FPI would be expec-
ted to contribute to the reduced excitability in this Mechanisms of LTP
region. Qualitatively similar results were seen in
stretch injured cortical neurons where the amplitude The progression of LTP under physiological con-
of GABAA-mediated currents are directly increased ditions consists of pre to postsynaptic induction,
following injury (see below; Kao et al., 2004). postsynaptic expression and subsequent long-term
expression and maintenance. Results reported in
the early literature were mixed and often discrep-
Area CA1: excitatory synaptic plasticity ant as to the mechanisms involved in injury-
induced loss of LTP. Sanders et al. (2000) suggests
Long-term potentiation that while there are major deficits in the main-
tenance phase, measurements at 5 min show no
Persistent and life-long cognitive impairment is a impairment in potentiation and that TBI may not
hallmark of brain injury pathology (Barth et al., affect the mechanisms essential to LTP induction
1983; Bennett-Levy, 1984; Lyeth et al., 1990; and expression. This is the only study, however,
McAllister, 1992; McAllister et al., 1999) and even that does not show a depression of the fEPSP
mild TBI can damage the fine structure of the immediately after injury.
hippocampus (Kotapka et al., 1991; Lowenstein Post-tetanic potentiation (PTP), an initial com-
et al., 1992; Grady et al., 2003), contributing to a ponent of LTP also cannot be observed after LFPI
reduction in information processing (Mathias et al., (Sick et al., 1998; Schwarzbach et al., 2006). Since,
2004) and learning and memory impairments. LTP, PTP is believed to be due to residual presynaptic
a form of activity-dependent plasticity, is currently calcium buildup resulting in increased neurotrans-
our best physiological correlate of learning and mitter release (Del Castillo and Katz, 1954; Martin
memory. LTP can be experimentally induced in and Pilar, 1964; Barrett and Stevens, 1972; Hirst
area CA1 and is characterized by a long-lasting et al., 1981), this led to the hypothesis that injury is
potentiation of fEPSP slope above baseline in re- affecting LTP via action on presynaptic mecha-
sponse to tetanic stimulation of the Schaffer col- nisms. To determine if there are alterations in pre-
laterals. Several rodent LFPI models report a synaptic transmitter release following TBI, paired
consistent inability to induce and maintain LTP pulse studies (two identical stimulus pulse sepa-
in area CA1 in the ipsilateral hippocampus in rated by less than 100 ms) were compared in sham
vivo (Miyazaki et al., 1992; Reeves et al., 1995, vs. injured slices. Paired pulse facilitation (PPF) in
1997b) and in vitro beginning at 2 h and persisting area CA1 results in a modest augmentation of
to 8 weeks post-injury (D’Ambrosio et al., 1998; the second fEPSP from the excess calcium in
Sick et al., 1998; Sanders et al., 2000; Schwarzbach the presynaptic terminal, whereas impaired PPF
et al., 2006) and may contribute to learning and would denote alterations in release probability
152

have occurred. Reeves et al. (2000) demonstrated and excitation (Liu et al., 2004). As previously
that the second response is enhanced within 1 h reviewed, injury leads to a downward ampli-
after LFPI, returning to control levels by 7 days tude shift in the area CA1 I/O curve (see Fig. 2),
after injury. Schwarzbach et al. (2006), however, demonstrating decreased excitability due to aug-
using the same interpulse interval, did not observe mented inhibition (Witgen et al., 2005). Thus, an
differences in PPF between sham vs. injured 7 days initial hypothesis postulates that augmented inhi-
after injury. Methodological or different recording bition is the reason why LTP cannot be induced
configurations may underlie this discrepancy. after TBI in area CA1. However, fully blocking
Synaptic plasticity, like proper synaptic function inhibition with the GABAA antagonist bicuculline
in general, requires a balance between inhibition methiodide does not rescue LTP (Fig. 5C) refuting

Fig. 5. Inability to induce area CA1 LTP but not LTD following lateral LFPI in injured mice (open circles) compared with sham (filled
circles). (A) High frequency tetanic stimulation (arrows) results in facilitation of the fEPSP in hippocampal slices from sham but not
from injured mice. (B) Low frequency stimulation resulted in LTD of the fEPSP in slices derived from both injured and sham animals.
(C) Histogram demonstrating that LTP can be induced in slices from sham and naı̈ve animals using either the standard HFS or theta
burst stimulation (TBS); however, with both protocols, LTP cannot be induced in slices from LFPI animals. However, LTP can be
induced and maintained in area CA1 in the hippocampus contralateral to injury. Area CA1 LTP cannot be rescued by varying the
stimulation protocol, altering extracellular Ca2+ and Mg2+ concentrations or bath application of BMI. (D) LTD induced by a LFS
(line) in slices from FPI animals does not make subsequent LTP induction by HFS (arrows) possible. Responses were followed for an
additional 30 min post-tetanic stimulation, no potentiation (even a return to baseline) was observed. Reprinted from Hippocampus,
Schwarzbach et al. (2006), with permission from Wiley-Liss, Inc.
153

the hypothesis that overwhelming inhibition alone shown in another study to increase after FPI in rat
underlies dysfunctional LTP (Schwarzbach et al., brain (Atkins et al., 2006). Thus, Western blot
2006). analysis of subcellular fractions from the hippo-
Data suggesting that LTD can be induced in campus or parietal cortex regions reveal a redis-
slices from injured animals show that plasticity tribution of phosphorylated a-CaMKII from the
is possible in mice that have undergone FPI. cytosolic to membrane fractions 30 min after mode-
D’Ambrosio et al. (1998) suggested that it is not rate lateral FPI (Atkins et al., 2006). Presently
alterations in the induction mechanisms which more studies are therefore needed to determine
induce LTP, but rather that injury itself potenti- whether reduced CaMKII function contributes to
ates transmission leading to a saturation of LTP. injury-induced deficits in LTP expression.
Therefore, if injury prepotentiates synapses, teta- The data presented above describe a consistent
nic presynaptic stimulation will be unable to fur- inability to induce area CA1 LTP following LFPI
ther potentiate the fEPSP. To test this hypothesis, and explore many possible contributing mecha-
slices from injured animals were first ‘‘depotenti- nisms. The work has presented many valuable
ated’’ (using the same low frequency stimulation injury-induced alterations in the cascade leading to
protocol used to classically induce LTD) and increased synaptic efficacy, however the inability
slices were then subsequently tetanized with a high to induce and maintain LTP does not seem to be
frequency, presynaptic stimulus in an attempt to due to one finite cause.
induce LTP. However, using this protocol did
not result in either LTP or, a return to baseline in
slices from FPI animals (Fig. 5D). This suggests Injury-induced functional alterations in the DG
that the mechanism(s) specific for the induction of
LTP, and not for synaptic plasticity in general, DG: injury-induced alterations in DG excitability
are in fact altered by injury (Schwarzbach et al.,
2006). The DG is thought to act as a filter to dampen
The anatomical substrate for LTP induction and synaptic input from the perforant pathway (PP)
expression is the dendritic spine (Leuner et al., before it is amplified by area CA3. The lack of
2003; Segal, 2005). Anatomical studies using interconnections between DGCs (Claiborne et al.,
lucifer yellow illustrate that following FPI, the 1986) and robust feed-forward and feed-back
mean apical and basal dendritic spine size (mm2) inhibition (Sloviter, 1994) function to impede syn-
significantly increases (Fig. 6; Schwarzbach et al., chronization and endow the DG with its filtering
2006). This increased spine size could effect the ability. Studies on the electrophysiological prop-
threshold level of calcium required for LTP erties of the DG have consistently shown that
induction, which may be kept to a minimum due LFPI leads to increased DG excitability in rodents,
to small spine size (Malenka and Nicoll, 1999; effectively reducing its filtering capacity. While
Gazzaley et al., 2002). significant cell death has been observed in the hilar
It is known from many studies that the cal- region, the preferential survival of excitatory neu-
cium and calmodulin-dependent protein kinase rons is not found (Lowenstein et al., 1992; Toth
a-CaMKII, which is abundant postsynaptically, is et al., 1997; Santhakumar et al., 2000).
critical for the induction of LTP (Malenka et al., In injured animals, stimulation of the PP evokes
1989). Hence, a functional decrease in a-CaMKII repetitive PS firing (Lowenstein et al., 1992; Santha-
or a decrease in its phosphorylating activity after kumar et al., 2000; Golarai et al., 2001), reduced
injury could contribute to an inability to induce PS threshold and increased spike amplitude (Toth
LTP. Western blot analysis demonstrates a signifi- et al., 1997; Santhakumar et al., 2000, 2001; Witgen
cant decrease in the alpha form of CaMKII pro- et al., 2005) compared with non-injured animals.
tein expression in regionally dissected area CA1 Studies have also shown that the amplitude and
tissue from FPI injured animals (Schwarzbach slope of fEPSPs substantially increases in injured
et al., 2006). However, CaMKII activity has been animals (Coulter et al., 1996; Golarai et al., 2001;
154

Fig. 6. Increased dendritic spine size in slices from LFPI mice. (A) CA1 pyramidal neuron and its spiny dendrites from an injured
mouse loaded with Lucifer yellow. (pcl, pyramidal cell layer; sl, stratum lucidum; so, stratum oriens.) Scale bar: 10 mm. (B) Higher
magnification of area highlighted by box in section A showing fourth order basal dendrite segment spines. Scale bar: 1 mm. (C) Higher
magnification of area highlighted by box in section A showing second order apical dendrite segment spines. Scale bar: 1 mm.
(D) Neurolucida drawing of the spiny segment shown in B. Scale bar: 1 mm. (E) Comparison of the distribution of dendritic spine size
of sham (black) and FPI (gray) apical (E) and basal (F) CA1 pyramidal cells. Reprinted from Hippocampus, Schwarzbach et al. (2006),
with permission from Wiley-Liss, Inc.
155

Fig. 7. Extracellular input/output curves for the dentate gyrus evoked by afferent perforant path stimulation. LFPI in mice results in
increased fEPSP slopes compared with those from sham animals. Reprinted from Neuroscience, Witgen et al. (2005), with permission
from Elsevier.

Witgen et al., 2005). Consistent with rat models, pathway and could be blocked with AMPA re-
1 week after LFPI in the mouse, the I/O curve ceptor antagonist(s) (Santhakumar et al., 2000).
(measuring the normalized slope of the fEPSP vs. The overall conclusion of this study is that exci-
the stimulation intensity), is significantly shifted tatory mossy cells become ‘‘irritable’’ and lead
upward with respect to amplitude as compared with to an increased excitatory drive onto both DG
sham animals (Fig. 7). This injury-induced shift in excitatory and inhibitory neurons (Santhakumar
the I/O curve suggests that net synaptic efficacy et al., 2000).
increases in the mouse DG after LFPI. More recently studies have supported this
Since the discharge of DGCs is under tight in- hypothesis showing mossy fiber sprouting at 2–3
hibitory control by GABAergic interneurons, it is and 14–15 weeks after a weight drop model of
constructive to completely antagonize inhibition, injury (Golarai et al., 2001). In another study,
in order to test whether injury-induced alterations mossy fiber sprouting led to increased output
in DG excitability are due to changes in gluta- from the excitatory principal cells of the DG to
matergic synaptic activity. One study presents data other granule cells and interneurons augmenting
supporting injury-induced alterations in the exci- excitation (Santhakumar et al., 2001). This was
tatory limb of the DG (Santhakumar et al., 2000). based on findings that injury caused an increase
Using slices from injured rats 1 week post-LFPI, in the evoked PS lasting 1 month post-LFPI and
afferent PP stimulation causes DG neurons to re- sIPSC from granule cells exhibited a higher
spond with protracted depolarizations and aug- frequency through 6 months after LFPI. The
ments action potential discharges in the absence of cause of this increased sIPSC was demonstrated
GABAAergic inhibition. The late depolarizations to be due to the glutamatergic excitatory drive to
exhibited in DGCs are mediated by a polysynaptic interneurons.
156

DG: changes in inhibitory synaptic transmission (1–4 h) and lasting up to 4 days post-LFPI (Ross
and Soltesz, 2000). This depolarization is mediated
While it has been shown that surviving DGCs by an injury-induced reduction in the activity of
sustain TBI-induced increases in excitability, alter- the Na+/K+ pump (ATPase), which contributes
ations in inhibitory function cannot be ruled out. to maintaining the resting membrane potential.
Many studies demonstrate neuronal loss in the This 10 mV depolarization transiently raises the
hilar region, however, no preferential survival of excitability of the GABAAergic network in the
inhibitory neurons is observed, including soma- early post-traumatic DG.
tostatin, parvalbumin and cholecystokinin positive
interneurons (Lowenstein et al., 1992; Toth et al.,
TBI effects seizure threshold
1997; Santhakumar et al., 2000). While inhibitory
neuronal loss could result in a decrease of GABA-
As previously discussed, the DG’s filtering efficacy
ergic inhibitory efficacy, functional changes in
is crucial to preventing seizure activity. Thus, a
GABA receptors is also a possibility.
dysfunction in this region is a plausible expla-
Indeed, studies show injury-induced alterations
nation for the post-traumatic epilepsy observed
in the inhibitory network of the DG. By 1 week
(Annegers et al., 1996). Santhakumar et al. (2001)
post-injury in the rat, there is a decrease in mIPSCs
show that there is a decrease in the threshold to
frequency, limiting GABAergic inhibition without
generate sustained (30+ min) of seizure like acti-
altering mIPSC kinetics or amplitude (Toth et al.,
vity to tetanic stimulation, however no spontane-
1997). Similar results are found in the mouse but
ous seizure like activity is present in area CA1 at 3
the amplitudes of mIPSCs recorded in slices from
months (after recovery period).
injured animals 1 week post-LFPI are smaller than
Furthermore, slices generated from LFPI
sham animals (Figs. 8A–D), while the kinetics of
animals have a lower threshold for developing
mIPSCs are not altered. The consequence of the
self-sustained seizure activity 1 week post-injury
decrease in mIPSC amplitude is demonstrated by a
(Coulter et al., 1996) and persisting up to 3 months
significant reduction in total mIPSC net charge
(Santhakumar et al., 2001). Coulter et al. (1996)
transfer in neurons from FPI animals, which was
found greater disinhibition in the DG of slices
76% of that calculated for neurons from sham
from FPI animals 1 week after injury when com-
slices (Fig. 8E). Furthermore, the mean frequency
pared with both sham-operated and epileptic ani-
of occurrence of mIPSCs is significantly lower
mals, which could be a possible explanation for the
in neurons from FPI animals than that present
observed decreased seizure threshold. In addition,
in DG neurons in sham slices (Fig. 8F). These
using the weight drop model of TBI, Golarai et al.
alterations in GABAAergic function demonstrate
(2001) demonstrate that injured animals have a
that spontaneous inhibitory activity (known to be
lower seizure threshold to pentylenetetrazole-
mediated by basket and axo-axonic interneurons)
induced convulsions 15 weeks post-TBI (Golarai
regulating feed-forward inhibition on dentate gran-
et al., 2001).
ule neurons is significantly compromised 1 week
post-LFPI.
Interestingly, the frequency of spontaneous ac- Cellular mechanisms of abnormal electrophysiology
tion potential-mediated inhibitory postsynaptic after trauma: the effect of tensile strain on
currents is enhanced up to 5 months after injury. ionotropic (excitatory and inhibitory) receptors of
This increase in spontaneous inhibitory post- cortical neurons in an in vitro model of TBI
synaptic current (sIPSC) frequency is due to en-
hanced excitatory drive (from irritable mossy cells) As illustrated above, TBI induces regional altera-
onto inhibitory interneurons (Santhakumar et al., tions in excitatory and inhibitory synaptic function
2000). Additionally, LFPI also causes a transient in hippocampal slices from rats and mice subjected
(approximately 10 mV) depolarized shift in DG to LFPI. Elucidation of the molecular sites and
interneurons exhibited immediately after injury biochemical pathways that mediate synaptic
157

Fig. 8. Whole-cell voltage clamp recordings from dentate granule neurons from injured mice demonstrate reductions in spontaneous
miniature (mIPSC) activity 1 week after LFPI. The mIPSCs are smaller and less frequent in dentate granule neurons from LFPI
animals. (A) Continuous sweeps of mIPSCs from sham (left panel) and LFPI (right panel) dentate granule cells at 60 mV. (B)
Cumulative frequency amplitude histogram for neurons in (A) with representative mIPSCs from sham and injured dentate granule
neurons. Histograms of mean median (D) mIPSC amplitude, (E) net charge transfer, and (F) mIPSC frequency demonstrate significant
reductions in inhibitory activity of neurons from LFPI mice compared with sham animals. Reprinted from Neuroscience, Witgen et al.
(2005), with permission from Elsevier.

dysfunction in the CNS would provide favorable mechanical forces directly impact upon neurons
targets for putative pharmacological therapeutic during traumatic impact to trigger secondary con-
treatments directed toward improving cognitive sequences of injury or affect neuronal function in-
function and memory after TBI. Yet, presently, directly by the release of excitatory and other CNS
little is known in detail regarding the cellular neurotransmitters during the so-called ‘depolari-
mechanisms underlying dysfunctional neurotrans- zation impact’ phase of trauma (Katayama et al.,
mission. For example, it is still unresolved whether 1990). Investigators have thus begun to examine
158

the cellular processes that result from mechanical 1996; Goforth et al., 1999, 2004; Kao et al., 2004).
trauma in further detail and more directly by using Ellis et al. (1995) developed an in vitro TBI model
simplified in vitro cell injury models. in which neonatal rat cortical neurons are co-
In the earliest in vitro models of mechanical cultured with astrocytes in 6-well plates having
trauma, pyramidal neurons were injured by deformable SILASTIC (Dow Corning) bottoms
scratching a cultured neuronal cell layer with a (FlexCell Plates; Ellis et al., 1995). An injury con-
glass sylet (Tecoma et al., 1989), transecting cells in troller used in this model applies a controlled pulse
a manner similar to tissue damage incurred during of compressed nitrogen to individual wells via an
penetrating TBI. Alternative in vitro models have airtight gasket (Fig. 9; Ellis et al., 1995). Displace-
been developed to simulate the forces (tensile, ment of the SILASTIC membrane with its adher-
compressional or torsional) acting on brain tissue ent cells thereby rapidly stretches the cells within
during blunt trauma or acceleration/deceleration 100 ms, mimicking the deformation forces that
injury, in order to more fully characterize the contribute to rapid acceleration/deceleration in-
mechanisms by which mechanical deformation (or jury (Pike, 2001). It is important to emphasize
tensile strain) leads to changes in neurons and glia that the forces produced by this model are likely
(Pike, 2001). Models of tensile strain (stretch) typi- to be highly relevant and akin to those encoun-
cally use pneumatic or mechanical translational tered by the brain in vivo following acceleration/
devices to deform a stretchable substrate upon deceleration induced TBI, as evidenced by bio-
which various types of neurons or brain slices are mechanical studies and simulations carried out
cultured and are thus adherent (Ellis et al., 1995; using computer modeling (Margulies et al., 1990;
Morrison et al., 1998, 2006; Arundine et al., 2003). Meaney et al., 1995).
Shear strain or torque, which approximates the in- The Ellis model produces a range of injury levels,
ertial loading caused by the rapid acceleration of ranging from mild to moderate to severe. Signifi-
brain tissue during TBI, has been modeled by forc- cantly, the model generates an injury in a very re-
ing fluid over stationary cells using a cell shearing producible manner. Access to the multiple wells
injury device (CSID) (LaPlaca and Thibault, 1998), provides good experimental control, and facilitates
or a newly developed electro-mechanical cell shear- studies of pharmacological agents to probe the
ing model, which can be used to examine the effects mechanisms of trauma or to evaluate potential
of torsional stress applied to three dimensional therapeutic agents. The model also facilitates
neuronal cultures (LaPlaca et al., 2005). studies of cell viability by using propidium iodide
As simplified systems, in vitro models allow in- staining (McKinney et al., 1996), mitochondrial
jury to be studied in the absence of ischemia, membrane potential measurement using rhodamine
blood/brain barrier considerations, or other 123 (Ahmed et al., 2000, 2002), cell calcium levels
changes, and the models provide increased exper- measured with fura-2 (Rzigalinski et al., 1997,
imental control of tissue oxygenation, the cell type 1998a, b; Weber et al., 2001, 2002), electrophysio-
to be studied, availability of nutrients, temperature logy (Tavalin et al., 1995, 1997; Zhang et al., 1996;
and ionic and drug concentrations. For electro- Goforth et al., 1999, 2004; Lea et al., 2002; Kao
physiological studies, in vitro models enable rapid et al., 2004) and biochemical assays of either the
drug or agonist application, patch clamping and attached cells or the bathing media (Lamb et al.,
intracellular calcium measurements to be carried 1997; Rzigalinski et al., 1999; Hoffman et al.,
out with relative ease. In vitro models are thus well 2000).
suited for examining the direct effects of a me- Mild injury due to a 5.7 mm displacement of the
chanical insult on ion channel or receptor function. Silastic membrane (corresponding to 31% stretch of
Using the in vitro model of stretch injury devel- brain cells) releases glutamate from astrocytes, with
oped by Ellis et al. (1995) several studies reveal peak glutamate occurring within 5 min after injury,
alterations in the function of neurotransmitter re- and then declining within an hour (Rzigalinski et al.,
ceptors, including AMPA, NMDA and GABAA 1998a, b). The model also produces a transient rise
receptors following mechanical strain (Zhang et al., in neuronal [Ca2+]i which persists for 410 min and
159

Fig. 9. In vitro TBI model of mechanical injury. An injury controller applies a timed pulse of nitrogen to cultured cells grown on a
deformable SILASTIC membrane to mimic stretch deformation in TBI. Reprinted from J. Neurotrauma, Ellis et al. (1995), with
permission from Mary Ann Liebert, Inc., Publishers.

is partially attenuated by the NMDA antagonist Stretch injury alters cortical glutamate receptor
MK-801 (Ahmed et al., 2002). In addition [Ca2+]i function
responses of mildly injured neurons to exogenous
glutamate are potentiated 15 min–24 h post-injury Excitatory synaptic transmission is mediated by
(Weber et al., 1999). The Ellis model reproduces AMPA and NMDA glutamate receptor subtypes.
many features of whole animal TBI including Both NMDA and AMPA receptors are heteromul-
changes in cell viability (McKinney et al., 1996), timers, composed of combinations of NR1 and
neuronal and glial [Ca2+]i homeostasis (McKinney NR2A-D or GluR1-4 subunits, respectively. The
et al., 1996; Zhang et al., 1996; Rzigalinski et al., NMDA current of control, uninjured neurons is
1998a, b; Weber et al., 1999, 2002), the status of well known to be characterized by its ‘‘J-shaped’’
endoplasmic reticulum (ER) Ca stores (Weber et al., voltage dependence, which is due to external
1999; Floyd et al., 2001), free radicals (McKinney voltage-dependent Mg2+ block of NMDA recep-
et al., 1996), phosphoinositide metabolism (Floyd tors (NMDARs) (Mayer et al., 1984; McBain and
et al., 2001), mitochondrial function (Ahmed et al., Mayer, 1994; Dingledine et al., 1999). However, in
2000, 2002) and electrophysiology (Tavalin et al., mildly injured neurons, voltage-dependent Mg2+
1995, 1997; Ahmed et al., 2000). block of NMDARs is greatly reduced (Fig. 10;
160

2003). Furthermore, the effect of mGluR agonists


and antagonists on injury-induced NMDA potent-
iation is dependent on the presence or absence of
glial cells (Lea et al., 2002, 2003). In pure neuronal
cultures, inhibition of either mGluR1 or mGluR5
prevents stretch-induced NMDA receptor potent-
iation, suggesting a contributing role of Group I
mGluRs in NMDA alteration (Lea et al., 2002,
2003). However, while mGluR5 inhibition also at-
tenuates NMDA alterations in neurons grown in
mixed cultures with glia, inhibition of mGluR 1
augments injury-induced NMDA potentiation (Lea
et al., 2002, 2003). Activation of mGluR1 also
differentially modulates the effect of injury on
NMDA receptors, depending on the presence or
Fig. 10. Stretch injury reduces Mg2+ block of NMDARs, as absence of glia (Lea et al., 2003). The above data
evidenced by linearization of the J-shaped NMDA current highlight the complexity of the biochemical and
(I)–voltage (V) curve. I–V currents generated by the application
cellular pathways leading to alterations in NMDA
of 200 mM NMDA and 10 mM glycine to an uninjured control
and a mildly stretch-injured cortical pyramidal neuron. Re- receptors after mechanical injury and suggest the
printed from Science, Zhang et al. (1996), with permission from possible involvement of not only neuronal but also
AAAS. glial mGluRs in this process.
Reduced Mg2+ block of NMDARs has been
Zhang et al., 1996). Thus, injury shifts the IC50 of observed in other pathological conditions, includ-
Mg2+ inhibition of NMDA current by 100-fold, ing crush injury of motoneuron axons (Furukawa,
and reduces its Bmax by 50% (Zhang et al., 1996). 2000), axotomization of spinal cord motoneurons
Moreover, raising [Mg2+]o restores some of the (Abdrachmanova et al., 2002), kainate lesioning of
block to the channels (Zhang et al., 1996). The loss hippocampal neurons (Chen et al., 1999) and peri-
of Mg2+ sensitivity of NMDARs results in turn in pheral inflammation of dorsal horn neurons (Guo
potentiated [Ca2+]i responses to exogenously ap- and Huang, 2001). The neurohormone somatosta-
plied NMDA (Zhang et al., 1996). This enhanced tin also reduces the Mg2+ block of hippocampal
NMDA-elicited [Ca2+]i response was recently du- NMDARs by increasing PKC activity (Pittaluga
plicated in a different in vitro model of stretch in- et al., 2000). Thus, loss of Mg2+ block appears to
jury (Geddes-Klein et al., 2006), and in other be a general mechanism of traumatic injury and
studies by Lea et al. (2002, 2003, see below). Im- neuromodulation.
portantly, pretreating neurons with the PKC in- Stretch injury via the Ellis in vitro model also
hibitor calphostin C, partially prevents the loss of modulates AMPARs in cortical neurons. Goforth
Mg2+ block (Zhang et al., 1996), consistent with et al. (1999) found mild injury similar to that used
an earlier report that PKC modulates the Mg2+ to modify NMDA current also increases whole-cell
sensitivity of NMDA receptors in nodose neurons steady-state AMPAR current density (Goforth
(Chen and Huang, 1992). et al., 1999; Fig. 11A). Stretch does not appear to
Using the stretch-injury model, Faden and co- modify the sensitivity of AMPARs to AMPA,
workers confirmed that in vitro injury indeed de- AMPAR current–voltage properties or the appar-
creases external Mg2+ block of NMDARs and ent number of AMPA receptors on pyramidal neu-
increases NMDA current in cultured neurons (Lea rons. However, pharmacological reduction of
et al., 2002). The effect of stretch injury on NMDA AMPAR desensitization using cyclothiazide or ac-
receptors was shown to be altered by the activation tivation of AMPARs with the weakly AMPAR
of the Group I metabotropic glutamate receptors desensitizing agonist kainate, offsets the observed
(mGluRs), mGluR1 and mGluR5 (Lea et al., 2002, differences between control and injured AMPA
161

Fig. 11. Stretch injury alters AMPAR kinetics and currents. (A) Whole-cell AMPA-elicited currents in control and injured cultured
cortical pyramidal neurons. Stretch injury reduces fast desensitization of AMPARs, resulting in larger steady-state currents.
(B) AMPAR currents recorded from outside/out patches in response to a 100 ms pulse of 1 mM glutamate+20 mM APV. (C) Mean
20–80% activation time (top) and desensitization rate (bottom) of AMPAR currents recorded from outside/out patches (*po0.05).
Reprinted from J. Neurosci., copyright 1999 by the Society for Neuroscience, and J. Neurotrauma, Goforth et al. (2004), with
permission from Mary Ann Liebert, Inc., Publishers.

currents, suggesting that injury suppresses AMPAR kinetics observed following mild mechanical stretch
desensitization (Goforth et al., 1999). Changes in (Goforth et al., 2004). CaMKII is [Ca2+]i -activated,
AMPAR kinetics surprisingly even persist in cell is known to phosphorylate the GluR1 subunit of the
free membrane patches excised from injured neu- AMPA receptor and is thought to participate in the
rons vs. controls and include slower AMPAR induction of LTP in area CA1 of the hippocampus
activation as well as desensitization (Fig. 11B). (Lisman et al., 1997; Malenka and Nicoll, 1997,
AMPA current alterations persist for at least 24 h 1999; Malinow et al., 2000; Pickard et al., 2001;
post-injury and stretching neurons with the NMDA Fink and Meyer, 2002; Malinow and Malenka,
antagonist APV present prevents the alterations 2002; Song and Huganir, 2002). In support of the
in AMPA currents (Goforth et al., 2004). This involvement of CAMKII in injury, measurements
suggests that NMDAR activation is again a pre- of CaMKII activity in cell lysates from control
requisite for AMPAR modulation by injury. and stretch-injured neurons show an increase in
CaMKII is well known to be an important the percent of autophosphorylated autonomous
mediator of synaptic modulation in the CNS (see CaMKII activity at 10 min post-injury (R. Tombes,
above in CA1 plasticity section). CaMKII also P. Goforth and L. Satin, unpublished data). This is
appears to play an important role in mediating consistent with a demonstrated increase in CamKII
changes in ionotropic receptors associated with after FPI in vivo, which is accompanied by
TBI in cortical neurons. Thus, pretreatment of increased expression of phosphorylated GluR1, an
cultured cortical neurons with the CaMKII inhib- AMPA receptor subunit and CaMKII substrate
itor KN-93 (N-[2-[[[3-(4-chlorophenyl)-2-propenyl] (Atkins et al., 2006).
methylamino]methyl]phenyl]-N-(2-hydroxyethyl)-4 Although studies indicate that the effect of
methoxybenzenesulphonamide) in the Ellis TBI stretch injury on NMDA and AMPA receptors
model also prevents the alterations in AMPAR appear to involve the activation of the protein
162

kinases PKC and CaMKII, respectively (Zhang whether similar alterations in AMPA and NMDA
et al., 1996; Goforth et al., 2004), it is not known receptors occur in different brain regions (i.e., hippo-
whether activation of these kinases after injury campal CA1, CA3 and DG vs. cortical neurons)
leads to direct phosphorylation of ionotropic gluta- and determine the time course of these alterations.
mate receptors or associated proteins. Modulation
of AMPARs by phosphorylation reflects a com-
plex balance of kinase and phosphatase activity Stretch injury alters cortical GABAA receptor
that can affect not only receptor subunits but an- function
choring or modulatory proteins that interact with
AMPARS. Thus, it is important to determine the As discussed above, many studies have demon-
phosphorylation state of specific AMPA and strated injury-induced dysfunctional inhibitory
NMDA subunits after injury in a highly quanti- synaptic transmission in the hippocampus (Toth
tative manner. It is also possible that injury alters et al., 1997; Witgen et al., 2005). As with glutamate
the expression and/or localization of ionotropic receptors, a study of cortical GABAA receptor
glutamate receptors and these changes may or may function suggests that direct alterations in GABAA
not be dependent on phosphorylation. receptors likely contributes to inhibitory dysfunc-
Changes in glutamate receptor function such as tion after trauma (Kao et al., 2004). Using the Ellis
those described above following stretch injury in vitro model, Kao et al. (2004) demonstrate that
would be expected to contribute to dysfunctional mild mechanical injury augments GABA-activated
synaptic transmission in CNS circuits impacted currents, which were blocked by bicuculline,
by mechanical trauma. In fact preliminary data confirming their mediation by GABAA receptors
demonstrate decreased mEPSC amplitude, and (Fig. 12). Stretch injury does not shift the EC50 or
slowed mEPSC rise time and duration in cortical current–voltage relationship of these GABAA cur-
pyramidal neurons observed immediately after rents. However, as reported for AMPA receptors,
mild stretch injury (Satin, unpublished data). GABA current potentiation is prevented by injur-
Injury-induced reductions in AMPA rise time ing neurons in the presence of the NMDA re-
may contribute to decreased mEPSC amplitudes, ceptor antagonist APV or the CaMKII inhibitor
while the slowing of the mEPSC time course may KN-93 (Kao et al., 2004). CaMKII has been
be effected by reduced AMPA receptor desensiti- shown to phosphorylate GABAA channels (Swope
zation as well increased contribution of NMDA et al., 1999), and inhibiting the expression of
receptor activation after mechanical injury (Zhang the CaMKII alpha subunit is known to decrease
et al., 1996; Goforth et al., 1999, 2004). Sponta- GABAA receptor function (Churn et al., 2000).
neous glutamatergic EPSCs of cortical pyramidal More studies are needed to determine the mech-
neurons also exhibit reduced amplitude following anism mediating enhanced GABA current, which,
mild stretch injury (Satin, unpublished data). in theory, could result from either an increase in
These data are in general agreement with studies the number of functional GABAA channels in the
that show attenuation of glutamate-evoked cur- neuronal plasma membrane, an increase in the
rents recorded from pyramidal CA1 neurons from unitary current through open GABAA channels or
mice subjected to FPI (Schwarzbach et al., 2006). an increase in the probability of opening of a fixed
The discrepant findings regarding the relative number of GABAA channels.
contribution of NMDA receptors to EPSCs after As with glutamate receptors, direct alterations
injury may be due, in part, to the examination of in GABAA channels would likely contribute to
different brain regions (cortex vs. hippocampus), dysfunctional inhibitory synaptic transmission
or the post-injury time point at which studies were after TBI. In fact, miniature as well as spontane-
conducted (immediately vs. 7 days post-injury). To ous IPSCs recorded from stretch-injured cortical
further elucidate the role of direct glutamate re- neurons exhibit increased amplitude (Goforth and
ceptor alterations in dysfunctional neurotransmis- Satin, unpublished data), but no change in mIPSC
sion after TBI, it will be important to determine frequency, such as observed in hippocampal
163

Fig. 12. Stretch injury potentiates GABA-elicited whole-cell current. (A) Patch-clamp recordings of whole-cell currents elicited by the
application of 50 mM GABA (filled bar) for a control neuron (left) and a stretched-injured neuron (right). Neurons were voltage-
clamped to –60 mV, and the rapid application of GABA-induced inward current. GABA-elicited currents of mildly injured neurons
were increased in amplitude compared with uninjured control neurons. (B) Amplitude histograms of GABA current density shown for
control neurons (left) and stretch-injured neurons (right). Currents were elicited by 50 mM GABA, and were normalized by neuron
capacitance to yield GABA current density (pA/pF). Mean current densities obtained were 20.271.7 pA/pF (mean7SE, n ¼ 69) for
control neurons and 41.272.6 pA/pF (n ¼ 82) for injured neurons (po0.001; Student’s t test). Reprinted from J. Neurotrauma, Kao et
al. (2004), with permission from Mary Ann Liebert, Inc., Publishers.

CA1 neurons from mice subjected to FPI in vivo in cortical neurons. Such changes occurring in
(Witgen et al., 2005). postsynaptic neurotransmitter receptor function
The data discussed clearly demonstrate that would thus be expected to contribute to dysfunc-
there are prominent injury-induced alterations in tional synaptic transmission in vivo or in brain
the function of glutamate and GABA receptors slices from TBI rodents. To underscore an
164

enduring theme of this chapter, we stress that the blocker TTX (tetrodotoxin) or a bathing solution
changes observed in synaptic function in vitro lacking calcium. However, SIDD is unaffected by
following injury were seen using levels of injury the AMPA receptor (AMPAR) antagonist CNQX
unable to cause significant derangement of cell (6-cyano-7-nitroquinoxaline-2,3-dione) indicating
attachment, cell morphology or outright cell that the requirement for glutamate receptor acti-
death. This emphasizes the importance of study- vation is subtype specific (Tavalin et al., 1997).
ing changes in synaptic function after mild and Soltesz and colleagues (Ross and Soltesz, 2000)
moderate TBI, which as a topic needs far more found that the membrane potentials of DGCs in
emphasis by the neurotrauma field. brain slices from TBI rats are also depolarized due
Thus, the detailed study of CNS synaptic mech- to the same ionic mechanism identified using the
anisms and their exquisite sensitivity to mechanical Ellis model by Tavalin et al. (Ross and Soltesz,
deformation remains highly significant for the field 2000). Functionally, SIDD would be expected to
and will likely result in new insights of importance contribute to the disruptions in ionic homeostasis,
to the development of new approaches for treating and changes in neuronal excitability known to
TBI patients. occur after TBI.

Stretch injury alters Na+/K+ ATPase activity Conclusions

In addition to abnormal neurotransmitter receptor Normal brain function is critically dependent on


function, neurotransmission and neuronal excita- the activation and tight regulatory control of
bility in the CNS may also be modulated by highly organized and precise neural circuits, the
trauma-induced alterations in ion pump and trans- fundamental unit of which is the synapse. It is well
porter activity. Under normal conditions, the documented that TBI disrupts normal brain func-
resting membrane potentials of many pyramidal tion, resulting in behavioral, motor and cognitive
neurons includes an electrogenic component medi- deficits such as impaired memory, attention and
ated by the basal activity of plasma membrane executive function, as well as slowed processing
Na+/K+ ATPases or ‘Na pumps’ (Tavalin et al., speed (Salmond and Sahakian, 2005). We postu-
1995, 1997). This occurs because of unequal (i.e., late that such deficits most likely reflect abnormal
electrogenic) fluxes of Na+ out of the neuron synaptic transmission after TBI.
and K+ into the neuron driven by the Na+/K+ Synapses are either excitatory or inhibitory,
ATPase, which is mediated energetically by ATP defined by activation of the specific neurotransmit-
hydrolysis. In vitro studies of hippocampal neurons ter released. Overall neuronal activity is shaped by
in brain slices from TBI rats and cortical neurons the integration of inhibitory and excitatory synap-
subjected to mild stretch result in clear injury- tic potentials, and slight changes in the balance of
induced decrease in Na+/K+ ATPase activity, excitation and inhibition, or their speed of occur-
which in turn leads to depolarized neuronal resting rence can produce profound changes in brain func-
membrane potentials recorded in both the in vitro tion by affecting synaptic information processing.
stretch and LFPI injury models (Tavalin et al., Synaptic inputs moreover are not static, but rather
1997; Ross and Soltesz, 2000). In the mildly quite plastic and are capable of being modulated in
stretch-injured neurons, resting membrane poten- response to physiological stimulation, resulting in
tial depolarization (magnitude 10–12 mV), deve- changes in the number or pattern of synaptic con-
loped 15–60 min after injury and was termed nections, presynaptic release of neurotransmitter or
‘‘stretch-induced delayed depolarization’’ (or the functional properties of neuronal postsynaptic
‘‘SIDD’’) (Tavalin et al., 1995). SIDD results from receptors (Malenka and Bear, 2004). Pathological
a Ca2+-influx dependent loss of electrogenic Na/K alterations in the strength, timing or number of
ATPase activity and is prevented by pretreatment synaptic inputs would be expected to affect overall
with the NMDA antagonist APV, the Na+ channel integration of synaptic potentials in individual
165

neurons, leading to abnormal cell firing and dys- Asikainen, I., Kaste, M. and Sarna, S. (1999) Early and late
functional information processing and transmis- posttraumatic seizures in traumatic brain injury rehabilita-
sion of signals within the important neural circuits tion patients: brain injury factors causing late seizures and
of the cerebral cortex and hippocampus, as well as influence of seizures on long-term outcome. Epilepsia, 40(5):
584–589.
other brain regions.
Atkins, C.M., Chen, S., Alonso, O.F., Dietrich, W.D. and Hu,
In order to understand the mechanisms underly- B.R. (2006) Activation of calcium/calmodulin-dependent
ing the cognitive changes that occur after TBI, it is protein kinases after traumatic brain injury. J. Cereb. Blood
thus critically important to elucidate the effects of Flow Metab., 26(12): 1507–1518.
injury on each key component of brain function: Barnard, E.A., Skolnick, P., Olsen, R.W., Mohler, H., Sieghart,
neuronal circuitry, cell to cell synaptic transmission W., Biggio, G., Braestrup, C., Bateson, A.N. and Langer,
S.Z. (1998) International Union of Pharmacology. XV. Sub-
and intrinsic cellular electrical activity. Despite im- types of gamma-aminobutyric acid a receptors: classification
portant advances made in this area of investigation, on the basis of subunit structure and receptor function.
most of the mechanisms involved in mediating the Pharmacol. Rev., 50(2): 291–313.
effects of TBI on the electrophysiology of neurons Barrett, E.F. and Stevens, C.F. (1972) The kinetics of trans-
and synapses in the CNS still remain unknown. mitter release at the frog neuromuscular junction. J. Physiol.,
227(3): 691–708.
These mechanisms and their molecular and genetic Barth, J.T., Macciocchi, S.N., Giordani, B., Rimel, R., Jane,
underpinnings are therefore a major gap in our J.A. and Boll, T.J. (1983) Neuropsychological sequelae of
knowledge of the effects of TBI on the brain and minor head injury. Neurosurgery, 13(5): 529–533.
should therefore be considered the next frontier of Bear, M.F. and Abraham, W.C. (1996) Long-term depression
in hippocampus. Annu. Rev. Neurosci., 19: 437–462.
brain injury research. We thus believe that they de-
Bennett-Levy, J.M. (1984) Long-term effects of severe closed
serve much more attention by investigators work- head injury on memory: evidence from a consecutive series of
ing in this field and hope that the present review will young adults. Acta Neurol. Scand., 70(4): 285–298.
stimulate new investigations of synaptic function Bigler, E.D., Blatter, D.D., Anderson, C.V., Johnson, S.C.,
and neuronal electrophysiology following TBI. Gale, S.D., Hopkins, R.O. and Burnett, B. (1997) Hippo-
campal volume in normal aging and traumatic brain injury.
AJNR Am. J. Neuroradiol., 18(1): 11–23.
Bohbot, V.D., Allen, J.J. and Nadel, L. (2000) Memory deficits
References characterized by patterns of lesions to the hippocampus and
parahippocampal cortex. Ann. N.Y. Acad. Sci., 911: 355–368.
Abdrachmanova, G., Teisinger, J. and Vyklicky Jr., L. (2002) Carbonell, W.S., Maris, D.O., McCall, T. and Grady, M.S.
Axotomy-induced changes in the properties of NMDA re- (1998) Adaptation of the fluid percussion injury model to the
ceptor channels in rat spinal cord motoneurons. J. Physiol., mouse. J. Neurotrauma, 15(3): 217–229.
538(Pt 1): 53–63. Cave, C.B. and Squire, L.R. (1991) Equivalent impairment of
Ahmed, S.M., Rzigalinski, B.A., Willoughby, K.A., Sitterding, spatial and nonspatial memory following damage to the hu-
H.A. and Ellis, E.F. (2000) Stretch-induced injury alters mi- man hippocampus. Hippocampus, 1(3): 329–340.
tochondrial membrane potential and cellular ATP in cultured Chen, L. and Huang, L.Y. (1992) Protein kinase C reduces
astrocytes and neurons. J. Neurochem., 74(5): 1951–1960. Mg2+ block of NMDA-receptor channels as a mechanism of
Ahmed, S.M., Weber, J.T., Liang, S., Willoughby, K.A., Sit- modulation. Nature, 356(6369): 521–523.
terding, H.A., Rzigalinski, B.A. and Ellis, E.F. (2002) Chen, Y., Chad, J.E., Cannon, R.C. and Wheal, H.V. (1999)
NMDA receptor activation contributes to a portion of the Reduced Mg2+ blockade of synaptically activated N-methyl-
decreased mitochondrial membrane potential and elevated D-aspartate receptor-channels in CA1 pyramidal neurons in
intracellular free calcium in strain-injured neurons. J. Neuro- kainic acid-lesioned rat hippocampus. Neuroscience, 88(3):
trauma, 19(12): 1619–1629. 727–739.
Akasu, T., Muraoka, N. and Hasuo, H. (2002) Hyperexcitability Christie, B.R., Kerr, D.S. and Abraham, W.C. (1994) Flip side
of hippocampal CA1 neurons after fluid percussion injury of of synaptic plasticity: long-term depression mechanisms in
the rat cerebral cortex. Neurosci. Lett., 329(3): 305–308. the hippocampus. Hippocampus, 4(2): 127–135.
Annegers, J.F., Rocca, W.A. and Hauser, W.A. (1996) Causes Churn, S.B., Sombati, S. and Delorenzo, R.J. (2000) Inhibition
of epilepsy: contributions of the Rochester epidemiology of CaMKII alpha subunit expression results in decreased
project. Mayo Clin. Proc., 71(6): 570–575. GABAA receptor function. 2000 Abstract Viewer and Itinerary
Arundine, M., Chopra, G.K., Wrong, A., Lei, S., Aarts, M.M., Planner. Washington D.C. Society for Neuroscience. Online.
MacDonald, J.F. and Tymianski, M. (2003) Enhanced vul- Claiborne, B.J., Amaral, D.G. and Cowan, W.M. (1986) A light
nerability to NMDA toxicity in sublethal traumatic neuronal and electron microscopic analysis of the mossy fibers of the
injury in vitro. J. Neurotrauma, 20(12): 1377–1395. rat dentate gyrus. J. Comp. Neurol., 246(4): 435–458.
166

Cobb, S.R., Buhl, E.H., Halasy, K., Paulsen, O. and Somogyi, P. Goforth, P.B., Ellis, E.F. and Satin, L.S. (1999) Enhancement
(1995) Synchronization of neuronal activity in hippocampus by of AMPA-mediated current after traumatic injury in cortical
individual GABAergic interneurons. Nature, 378(6552): 75–78. neurons. J. Neurosci., 19(17): 7367–7374.
Cohen, A.S. and Abraham, W.C. (1996) Facilitation of long- Goforth, P.B., Ellis, E.F. and Satin, L.S. (2004) Mechanical
term potentiation by prior activation of metabotropic gluta- injury modulates AMPA receptor kinetics via an NMDA
mate receptors. J. Neurophysiol., 76(2): 953–962. receptor-dependent pathway. J. Neurotrauma, 21(6):
Coulter, D.A., Rafiq, A., Shumate, M., Gong, Q.Z., De- 719–732.
Lorenzo, R.J. and Lyeth, B.G. (1996) Brain injury-induced Golarai, G., Greenwood, A.C., Feeney, D.M. and Connor, J.A.
enhanced limbic epileptogenesis: anatomical and physiolog- (2001) Physiological and structural evidence for hippocampal
ical parallels to an animal model of temporal lobe epilepsy. involvement in persistent seizure susceptibility after trau-
Epilepsy Res., 26(1): 81–91. matic brain injury. J. Neurosci., 21(21): 8523–8537.
D’Ambrosio, R., Maris, D.O., Grady, M.S., Winn, H.R. and Gorman, L.K., Shook, B.L. and Becker, D.P. (1993) Traumatic
Janigro, D. (1998) Selective loss of hippocampal long-term brain injury produces impairments in long-term and recent
potentiation, but not depression, following fluid percussion memory. Brain Res., 614(1–2): 29–36.
injury. Brain Res., 786(1–2): 64–79. Grady, M.S., Charleston, J.S., Maris, D., Witgen, B.M. and
Del Castillo, J. and Katz, B. (1954) Statistical factors involved Lifshitz, J. (2003) Neuronal and glial cell number in the hippo-
in neuromuscular facilitation and depression. J. Physiol., campus after experimental traumatic brain injury: analysis
124(3): 574–585. by stereological estimation. J. Neurotrauma, 20(10): 929–941.
Deweer, B., Pillon, B., Pochon, J.B. and Dubois, B. (2001) Is Guo, H. and Huang, L.Y. (2001) Alteration in the voltage de-
the HM story only a ‘‘remote memory’’? Some facts about pendence of NMDA receptor channels in rat dorsal horn
hippocampus and memory in humans. Behav. Brain Res., neurones following peripheral inflammation. J. Physiol.,
127(1–2): 209–224. 537(Pt 1): 115–123.
Dingledine, R., Borges, K., Bowie, D. and Traynelis, S.F. He, J., Yamada, K., Zou, L.B. and Nabeshima, T. (2001) Spa-
(1999) The glutamate receptor ion channels. Pharmacol. tial memory deficit and neurodegeneration induced by the
Rev., 51(1): 7–61. direct injection of okadaic acid into the hippocampus in rats.
Dixon, C.E., Lyeth, B.G., Povlishock, J.T., Findling, R.L., J. Neural. Transm., 108(12): 1435–1443.
Hamm, R.J., Marmarou, A., Young, H.F. and Hayes, R.L. Heinemann, U., Beck, H., Dreier, J.P., Ficker, E., Stabel, J. and
(1987) A fluid percussion model of experimental brain injury Zhang, C.L. (1992) The dentate gyrus as a regulated gate for
in the rat. J. Neurosurg., 67(1): 110–119. the propagation of epileptiform activity. Epilepsy Res. Sup-
Dudek, F.E., Snow, R.W. and Taylor, C.P. (1986) Role of pl., 7: 273–280.
electrical interactions in synchronization of epileptiform Hoffman, S.W., Rzigalinski, B.A., Willoughby, K.A. and Ellis,
bursts. Adv. Neurol., 44: 593–617. E.F. (2000) Astrocytes generate isoprostanes in response to
Ellis, E.F., McKinney, J.S., Willoughby, K.A., Liang, S. and trauma or oxygen radicals. J. Neurotrauma, 17(5): 415–420.
Povlishock, J.T. (1995) A new model for rapid stretch- Hogg, S., Perron, C., Barneoud, P., Sanger, D.J. and Moser,
induced injury of cells in culture: characterization of the P.C. (1998a) Neuroprotective effect of eliprodil: attenuation
model using astrocytes. J. Neurotrauma, 12(3): 325–339. of a conditioned freezing deficit induced by traumatic injury
Fink, C.C. and Meyer, T. (2002) Molecular mechanisms of of the right parietal cortex in the rat. J. Neurotrauma, 15(7):
CaMKII activation in neuronal plasticity. Curr. Opin. Ne- 545–553.
urobiol., 12(3): 293–299. Hogg, S., Sanger, D.J. and Moser, P.C. (1998b) Mild traumatic
Floyd, C.L., Rzigalinski, B.A., Weber, J.T., Sitterding, H.A., lesion of the right parietal cortex in the rat: characterisation
Willoughby, K.A. and Ellis, E.F. (2001) Traumatic injury of a conditioned freezing deficit and its reversal by dizocil-
of cultured astrocytes alters inositol (1,4,5)-trisphosphate- pine. Behav. Brain Res., 93(1–2): 157–165.
mediated signaling. Glia, 33(1): 12–23. Joliot, M., Ribary, U. and Llinas, R. (1994) Human oscillatory
Freund, T.F. and Buzsaki, G. (1996) Interneurons of the hippo- brain activity near 40 Hz coexists with cognitive temporal
campus. Hippocampus, 6(4): 347–470. binding. Proc. Natl. Acad. Sci. U.S.A., 91(24): 11748–11751.
Fricke, R.A. and Prince, D.A. (1984) Electrophysiology of Kao, C.Q., Goforth, P.B., Ellis, E.F. and Satin, L.S. (2004)
dentate gyrus granule cells. J. Neurophysiol., 51(2): 195–209. Potentiation of GABA(A) currents after mechanical injury of
Furukawa, Y. (2000) Reduction of voltage-dependent magne- cortical neurons. J. Neurotrauma, 21(3): 259–270.
sium block of N-methyl-D-aspartate receptor-mediated cur- Katayama, Y., Becker, D.P., Tamura, T. and Hovda, D.A.
rent by in vivo axonal injury. Neuroscience, 96(2): 385–392. (1990) Massive increases in extracellular potassium and the
Gazzaley, A., Kay, S. and Benson, D.L. (2002) Dendritic spine indiscriminate release of glutamate following concussive
plasticity in hippocampus. Neuroscience, 111(4): 853–862. brain injury. J. Neurosurg., 73(6): 889–900.
Geddes-Klein, D.M., Schiffman, K.B. and Meaney, D.F. (2006) Kotapka, M.J., Gennarelli, T.A., Graham, D.I., Adams, J.H.,
Mechanisms and consequences of neuronal stretch injury in Thibault, L.E., Ross, D.T. and Ford, I. (1991) Selective vul-
vitro differ with the model of trauma. J. Neurotrauma, 23(2): nerability of hippocampal neurons in acceleration-induced
193–204. experimental head injury. J. Neurotrauma, 8(4): 247–258.
167

Kotapka, M.J., Graham, D.I., Adams, J.H. and Gennarelli, cell death following traumatic brain injury in the rat. Brain
T.A. (1992) Hippocampal pathology in fatal non-missile hu- Res., 526(2): 249–258.
man head injury. Acta Neuropathol. (Berl.), 83(5): 530–534. Lytton, W.W. and Sejnowski, T.J. (1991) Simulations of
Kotapka, M.J., Graham, D.I., Adams, J.H., Doyle, D. and cortical pyramidal neurons synchronized by inhibitory inter-
Gennarelli, T.A. (1993) Hippocampal damage in fatal pae- neurons. J. Neurophysiol., 66(3): 1059–1079.
diatric head injury. Neuropathol. Appl. Neurobiol., 19(2): Malenka, R.C. and Bear, M.F. (2004) LTP and LTD: an
128–133. embarrassment of riches. Neuron, 44(1): 5–21.
Kotapka, M.J., Graham, D.I., Adams, J.H. and Gennarelli, Malenka, R.C., Kauer, J.A., Perkel, D.J. and Nicoll, R.A.
T.A. (1994) Hippocampal pathology in fatal human head (1989) The impact of postsynaptic calcium on synaptic
injury without high intracranial pressure. J. Neurotrauma, transmission — its role in long-term potentiation. Trends
11(3): 317–324. Neurosci., 12(11): 444–450.
Lamb, R.G., Harper, C.C., McKinney, J.S., Rzigalinski, B.A. Malenka, R.C. and Nicoll, R.A. (1997) Silent synapses speak
and Ellis, E.F. (1997) Alterations in phosphatidylcholine up. Neuron, 19(3): 473–476.
metabolism of stretch-injured cultured rat astrocytes. J. Malenka, R.C. and Nicoll, R.A. (1999) Long-term potentiation
Neurochem., 68(5): 1904–1910. — a decade of progress? Science, 285(5435): 1870–1874.
LaPlaca, M.C., Cullen, D.K., McLoughlin, J.J. and Cargill II, Malinow, R., Mainen, Z.F. and Hayashi, Y. (2000) LTP mecha-
R.S. (2005) High rate shear strain of three-dimensional neu- nisms: from silence to four-lane traffic. Curr. Opin.
ral cell cultures: a new in vitro traumatic brain injury model. Neurobiol., 10(3): 352–357.
J. Biomech., 38(5): 1093–1105. Malinow, R. and Malenka, R.C. (2002) AMPA receptor
LaPlaca, M.C. and Thibault, L.E. (1998) Dynamic mechanical trafficking and synaptic plasticity. Annu. Rev. Neurosci.,
deformation of neurons triggers an acute calcium response 25: 103–126.
and cell injury involving the N-methyl-D-aspartate glutamate Margulies, S.S., Thibault, L.E. and Gennarelli, T.A. (1990)
receptor. J. Neurosci. Res., 52(2): 220–229. Physical model simulations of brain injury in the primate.
Lea, P.M., Custer, S.J., Vicini, S. and Faden, A.I. (2002) J. Biomech., 23(8): 823–836.
Neuronal and glial mGluR5 modulation prevents stretch- Martin, A.R. and Pilar, G. (1964) Presynaptic and post-synap-
induced enhancement of NMDA receptor current. Pharmacol. tic events during post-tetanic potentiation and facilitation in
Biochem. Behav., 73(2): 287–298. the avian ciliary ganglion. J. Physiol., 175: 17–30.
Lea, P.M.T., Custer, S.J., Stoica, B.A. and Faden, A.I. (2003) Mathias, J.L., Bigler, E.D., Jones, N.R., Bowden, S.C., Barrett-
Modulation of stretch-induced enhancement of neuronal Woodbridge, M., Brown, G.C. and Taylor, D.J. (2004)
NMDA receptor current by mGluR1 depends upon presence Neuropsychological and information processing performance
of glia. J. Neurotrauma, 20(11): 1233–1249. and its relationship to white matter changes following
Leuner, B., Falduto, J. and Shors, T.J. (2003) Associative moderate and severe traumatic brain injury: a preliminary
memory formation increases the observation of dendritic study. Appl. Neuropsychol., 11(3): 134–152.
spines in the hippocampus. J Neurosci, 23(2): 659–665. Mayer, M.L., Westbrook, G.L. and Guthrie, P.B. (1984)
Lisman, J., Malenka, R.C., Nicoll, R.A. and Malinow, R. Voltage-dependent block by Mg2+ of NMDA responses in
(1997) Learning mechanisms: the case for CaM-KII. Science, spinal cord neurones. Nature, 309(5965): 261–263.
276(5321): 2001–2002. McAllister, T.W. (1992) Neuropsychiatric sequelae of head in-
Liu, L., Wong, T.P., Pozza, M.F., Lingenhoehl, K., Wang, Y., juries. Psychiatr. Clin. North Am., 15(2): 395–413.
Sheng, M., Auberson, Y.P. and Wang, Y.T. (2004) Role of McAllister, T.W., Saykin, A.J., Flashman, L.A., Sparling,
NMDA receptor subtypes in governing the direction of hippo- M.B., Johnson, S.C., Guerin, S.J., Mamourian, A.C.,
campal synaptic plasticity. Science, 304(5673): 1021–1024. Weaver, J.B. and Yanofsky, N. (1999) Brain activation dur-
Liu, P. and Bilkey, D.K. (2001) The effect of excitotoxic lesions ing working memory 1 month after mild traumatic brain in-
centered on the hippocampus or perirhinal cortex in object jury: a functional MRI study. Neurology, 53(6): 1300–1308.
recognition and spatial memory tasks. Behav. Neurosci., McBain, C.J. and Mayer, M.L. (1994) N-methyl-D-aspartic acid
115(1): 94–111. receptor structure and function. Physiol. Rev., 74(3):
Lothman, E.W., Stringer, J.L. and Bertram, E.H. (1992) The 723–760.
dentate gyrus as a control point for seizures in the hippo- McIntosh, T.K., Vink, R., Noble, L., Yamakami, I., Fernyak,
campus and beyond. Epilepsy Res. Suppl., 7: 301–313. S., Soares, H. and Faden, A.L. (1989) Traumatic brain injury
Lowenstein, D.H., Thomas, M.J., Smith, D.H. and McIntosh, in the rat: characterization of a lateral fluid-percussion
T.K. (1992) Selective vulnerability of dentate hilar neurons model. Neuroscience, 28(1): 233–244.
following traumatic brain injury: a potential mechanistic link McKinney, J.S., Willoughby, K.A., Liang, S. and Ellis, E.F.
between head trauma and disorders of the hippocampus. (1996) Stretch-induced injury of cultured neuronal, glial, and
J. Neurosci., 12(12): 4846–4853. endothelial cells. Effect of polyethylene glycol-conjugated
Lyeth, B.G., Jenkins, L.W., Hamm, R.J., Dixon, C.E., Phillips, superoxide dismutase. Stroke, 27(5): 934–940.
L.L., Clifton, G.L., Young, H.F. and Hayes, R.L. (1990) Meaney, D.F., Smith, D.H., Shreiber, D.I., Bain, A.C., Miller,
Prolonged memory impairment in the absence of hippocampal R.T., Ross, D.T. and Gennarelli, T.A. (1995) Biomechanical
168

analysis of experimental diffuse axonal injury. J. Neuro- Reeves, T.M., Lyeth, B.G., Phillips, L.L., Hamm, R.J. and
trauma, 12(4): 689–694. Povlishock, J.T. (1997a) The effects of traumatic brain injury
Miller, L.A., Lai, R. and Munoz, D.G. (1998) Contributions of on inhibition in the hippocampus and dentate gyrus. Brain
the entorhinal cortex, amygdala and hippocampus to human Res., 757(1): 119–132.
memory. Neuropsychologia, 36(11): 1247–1256. Reeves, T.M., Zhu, J., Povlishock, J.T. and Phillips, L.L.
Miller, L.P., Lyeth, B.G., Jenkins, L.W., Oleniak, L., Panchision, (1997b) The effect of combined fluid percussion and ento-
D., Hamm, R.J., Phillips, L.L., Dixon, C.E., Clifton, G.L. rhinal cortical lesions on long-term potentiation. Neurosci-
and Hayes, R.L. (1990) Excitatory amino acid receptor sub- ence, 77(2): 431–444.
type binding following traumatic brain injury. Brain Res., Rempel-Clower, N.L., Zola, S.M., Squire, L.R. and Amaral,
526(1): 103–107. D.G. (1996) Three cases of enduring memory impairment
Miyazaki, S., Katayama, Y., Lyeth, B.G., Jenkins, L.W., after bilateral damage limited to the hippocampal formation.
DeWitt, D.S., Goldberg, S.J., Newlon, P.G. and Hayes, R.L. J. Neurosci., 16(16): 5233–5255.
(1992) Enduring suppression of hippocampal long-term Ross, S.T. and Soltesz, I. (2000) Selective depolarization of in-
potentiation following traumatic brain injury in rat. Brain terneurons in the early posttraumatic dentate gyrus: involve-
Res., 585(1–2): 335–339. ment of the Na(+)/K(+)-ATPase. J. Neurophysiol., 83(5):
Mody, I., De Koninck, Y., Otis, T.S. and Soltesz, I. (1994) 2916–2930.
Bridging the cleft at GABA synapses in the brain. Trends Rzigalinski, B.A., Liang, S., McKinney, J.S., Willoughby, K.A.
Neurosci., 17(12): 517–525. and Ellis, E.F. (1997) Effect of Ca2+ on in vitro astrocyte
Morrison, III, B., Cater, H.L., Benham, C.D. and Sundstrom, injury. J. Neurochem., 68(1): 289–296.
L.E. (2006) An in vitro model of traumatic brain injury uti- Rzigalinski, B.A., Weber, J.T., Willoughby, K.A. and Ellis,
lising two-dimensional stretch of organotypic hippocampal E.F. (1998a) Intracellular free calcium dynamics in stretch-
slice cultures. J. Neurosci. Methods, 150(2): 192–201. injured astrocytes. J. Neurochem., 70(6): 2377–2385.
Morrison, III, B., Meaney, D.F. and McIntosh, T.K. (1998) Rzigalinski, B.A., Weber, J.T., Willoughby, K.A., L.S., Wood-
Mechanical characterization of an in vitro device designed to ward, J.J. and Ellis, E.F. (1998b). Glutamate release and en-
quantitatively injure living brain tissue. Ann. Biomed. Eng., hanced receptor sensitivity in stretch-injured neurons. J
26(3): 381–390. Neurotrauma, 15: 894.
O’Dell, D.M. and Hamm, R.J. (1995) Chronic postinjury Rzigalinski, B.A., Willoughby, K.A., Hoffman, S.W., Falck,
administration of MDL 26,479 (Suritozole), a negative J.R. and Ellis, E.F. (1999) Calcium influx factor, further ev-
modulator at the GABAA receptor, and cognitive impair- idence it is 5,6-epoxyeicosatrienoic acid. J. Biol. Chem.,
ment in rats following traumatic brain injury. J. Neurosurg., 274(1): 175–182.
83(5): 878–883. Salmond, C.H. and Sahakian, B.J. (2005) Cognitive outcome in
Osteen, C.L., Giza, C.C. and Hovda, D.A. (2004) Injury- traumatic brain injury survivors. Curr. Opin. Crit. Care, 11:
induced alterations in N-methyl-D-aspartate receptor subunit 111–116.
composition contribute to prolonged 45 calcium accumula- Sanders, M.J., Sick, T.J., Perez-Pinzon, M.A., Dietrich, W.D.
tion following lateral fluid percussion. Neuroscience, 128(2): and Green, E.J. (2000) Chronic failure in the maintenance of
305–322. long-term potentiation following fluid percussion injury in
Pickard, L., Noel, J., Duckworth, J.K., Fitzjohn, S.M., Henley, the rat. Brain Res., 861(1): 69–76.
J.M., Collingridge, G.L. and Molnar, E. (2001) Transient Santhakumar, V., Bender, R., Frotscher, M., Ross, S.T.,
synaptic activation of NMDA receptors leads to the insertion Hollrigel, G.S., Toth, Z. and Soltesz, I. (2000) Granule cell
of native AMPA receptors at hippocampal neuronal plasma hyperexcitability in the early post-traumatic rat dentate
membranes. Neuropharmacology, 41(6): 700–713. gyrus: the ‘irritable mossy cell’ hypothesis. J. Physiol.,
Pike, B.R. (2001) In vitro models of traumatic brain injury. 524(Pt 1): 117–134.
In: Miller L.P. and Hayes R.L. (Eds.), Head Trauma: Ba- Santhakumar, V., Ratzliff, A.D., Jeng, J., Toth, Z. and Soltesz,
sic, Preclinical and Clinical Directions. Wiley, New York, I. (2001) Long-term hyperexcitability in the hippocampus af-
pp. 37–64. ter experimental head trauma. Ann. Neurol., 50(6): 708–717.
Pittaluga, A., Bonfanti, A. and Raiteri, M. (2000) Somatostatin Schwarzbach, E., Bonislawski, D.P., Xiong, G. and Cohen,
potentiates NMDA receptor function via activation of A.S. (2006) Mechanisms underlying the inability to induce
InsP(3) receptors and PKC leading to removal of the area CA1 LTP in the mouse after traumatic brain injury.
Mg(2+) block without depolarization. Br. J. Pharmacol., Hippocampus, 16(6): 541–550.
130(3): 557–566. Segal, M. (2005) Dendritic spines and long-term plasticity. Nat.
Reeves, T.M., Kao, C.Q., Phillips, L.L., Bullock, M.R. and Rev. Neurosci., 6(4): 277–284.
Povlishock, J.T. (2000) Presynaptic excitability changes fol- Sick, T.J., Perez-Pinzon, M.A. and Feng, Z.Z. (1998) Impaired
lowing traumatic brain injury in the rat. J. Neurosci. Res., expression of long-term potentiation in hippocampal slices 4
60(3): 370–379. and 48 h following mild fluid-percussion brain injury in vivo.
Reeves, T.M., Lyeth, B.G. and Povlishock, J.T. (1995) Long- Brain Res., 785(2): 287–292.
term potentiation deficits and excitability changes following Sihver, S., Marklund, N., Hillered, L., Langstrom, B., Wata-
traumatic brain injury. Exp. Brain Res., 106(2): 248–256. nabe, Y. and Bergstrom, M. (2001) Changes in mACh,
169

NMDA and GABA(A) receptor binding after lateral fluid- Thompson, H.J., Lifshitz, J., Marklund, N., Grady, M.S.,
percussion injury: in vitro autoradiography of rat brain Graham, D.I., Hovda, D.A. and McIntosh, T.K. (2005) Lat-
frozen sections. J. Neurochem., 78(3): 417–423. eral fluid percussion brain injury: a 15-year review and eva-
Singer, W. and Gray, C.M. (1995) Visual feature integration luation. J. Neurotrauma, 22(1): 42–75.
and the temporal correlation hypothesis. Annu. Rev. Neuro- Toth, Z., Hollrigel, G.S., Gorcs, T. and Soltesz, I. (1997)
sci., 18: 555–586. Instantaneous perturbation of dentate interneuronal net-
Sloviter, R.S. (1994) The functional organization of the hippo- works by a pressure wave-transient delivered to the neocor-
campal dentate gyrus and its relevance to the pathogenesis of tex. J. Neurosci., 17(21): 8106–8117.
temporal lobe epilepsy. Ann. Neurol., 35(6): 640–654. Weber, J.T., Rzigalinski, B.A., Willoughby, K.A., Moore, S.F.
Smith, D.H., Okiyama, K., Thomas, M.J., Claussen, B. and and Ellis, E.F. (1999) Alterations in calcium-mediated signal
McIntosh, T.K. (1991) Evaluation of memory dysfunction transduction after traumatic injury of cortical neurons. Cell
following experimental brain injury using the Morris water Calcium, 26(6): 289–299.
maze. J. Neurotrauma, 8(4): 259–269. Weber, J.T., Rzigalinski, B.A. and Ellis, E.F. (2001) Traumatic
Song, I. and Huganir, R.L. (2002) Regulation of AMPA injury of cortical neurons causes changes in intracellular
receptors during synaptic plasticity. Trends Neurosci., calcium stores and capacitative calcium influx. J. Biol.
25(11): 578–588. Chem., 276(3): 1800–1807.
Swope, S.L., Moss, S.J., Raymond, L.A. and Huganir, R.L. Weber, J.T., Rzigalinski, B.A. and Ellis, E.F. (2002) Calcium
(1999) Regulation of ligand-gated ion channels by protein responses to caffeine and muscarinic receptor agonists are
phosphorylation. Adv. Second Messenger Phosphoprotein altered in traumatically injured neurons. J. Neurotrauma,
Res., 33: 49–78. 19(11): 1433–1443.
Tavalin, S.J., Ellis, E.F. and Satin, L.S. (1995) Mechanical Witgen, B.M., Lifshitz, J., Smith, M.L., Schwarzbach, E.,
perturbation of cultured cortical neurons reveals a stretch- Liang, S.L., Grady, M.S. and Cohen, A.S. (2005) Regional
induced delayed depolarization. J. Neurophysiol., 74(6): hippocampal alteration associated with cognitive deficit
2767–2773. following experimental brain injury: a systems, network
Tavalin, S.J., Ellis, E.F. and Satin, L.S. (1997) Inhibition of and cellular evaluation. Neuroscience, 133(1): 1–15.
the electrogenic Na pump underlies delayed depolarization Wong, R.K., Traub, R.D. and Miles, R. (1986) Cellular basis of
of cortical neurons after mechanical injury or glutamate. neuronal synchrony in epilepsy. Adv. Neurol., 44: 583–592.
J. Neurophysiol., 77(2): 632–638. Zhang, L., Rzigalinski, B.A., Ellis, E.F. and Satin, L.S. (1996)
Tecoma, E.S., Monyer, H., Goldberg, M.P. and Choi, D.W. Reduction of voltage-dependent Mg2+ blockade of NMDA
(1989) Traumatic neuronal injury in vitro is attenuated by current in mechanically injured neurons. Science, 274(5294):
NMDA antagonists. Neuron, 2(6): 1541–1545. 1921–1923.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 11

Traumatic injury of the spinal cord and nitric oxide

Jozef Maršala, Judita Orendáčová, Nadežda Lukáčová and Ivo Vanický

Institute of Neurobiology, Slovak Academy of Sciences, Košice, Slovak Republic

Abstract: In the current report, we summarize our findings related to the involvement of nitric oxide (NO)
in the pathology of spinal cord trauma. We initially studied the distribution of nitric oxide synthase (NOS)-
immunolabeled and/or nicotinamide adenine dinucleotide phosphate diaphorase (NADPHd; which is
highly colocalized with NOS)-stained somata and fibers in the spinal cord of the rabbit. Segmental and
laminar distribution of NADPHd-stained neurons in the rabbit revealed a large number of NADPHd-
stained neurons in the spinal cord falling into six categories, N1–N6, while others could not be classified.
Large numbers of NADPHd-stained neurons were identified in the superficial dorsal horn and around the
central canal. Four morphologically distinct kinds of NADPHd-stained axons 2.5–3.5 mm in diameter were
identified throughout the white matter in the spinal cord. Moreover, a massive occurrence of axonal
NADPHd-staining was detected in the juxtagriseal layer of the ventral funiculus along the rostrocaudal
axis. The prominent NADPHd-stained fiber bundles were identified in the mediobasal and central portion
of the ventral funiculus. The sulcomarginal fasciculus was found in the basal and medial portion of the
ventral funiculus in all cervical and thoracic segments. Since the discovery that NO may act as a neuronal
transmitter, an increasing interest has focused on its ability to modulate synaptic function. NO passes
through cell membranes without specific release or uptake mechanisms inducing changes in signal-related
functions by several means. In particular, the activation of the soluble guanylyl cyclases (sGC), the for-
mation of cyclic guanosine 30 ,50 -monophosphate (cGMP) and the action of cGMP-dependent protein
kinases has been identified as the main signal transduction pathways of NO in the nervous system including
spinal cord. It is known that the intracellular level of cGMP is strictly controlled by its rate of synthesis via
guanylyl cyclases (GC) and/or by the rate of its degradation via 30 ,50 -cyclic nucleotide phosphodiesterases
(PDE). GC can be divided into two main groups, i.e., the membrane-bound or particular guanylyl cyclase
(pGC) and the cytosolic or sGC. In the spinal cord, the activation of pGC has only been demonstrated for
natriuretic peptides, which stimulate cGMP accumulation in GABA-ergic structures in laminae I–III of the
rat cervical spinal cord. These neurons are involved in controlling the action of the locomotor circuit. In
view of the abundance of NO-responsive structures in the brain, it is proposed that NO–cGMP signaling
will be part of neuronal information processing at many levels. In relation to this, we found that surgically
induced Th7 constriction of 24 h duration stimulated both the constitutive NOS activity and cGMP level by
120 and 131%, respectively, in non-compartmentalized white matter of Th8–Th9 segments, located just
caudally to the site of injury. NO-mediated cGMP formation was only slightly increased in the dorsal
funiculus of Th5–Th9 segments. There are some other sources that may influence the NO-mediated cGMP
formation in spinal cord. A high level of glutamate produced at the site of the lesion and an excessive
accumulation of intracellular Ca2+ may stimulate NOS activity and create suitable conditions for NO
synthesis and its adverse effect on white matter. An increased interest has focused on the role of NO at the

Corresponding author. Tel.: +421 55 6785 062; Fax: +421 55


6785 074; E-mail: marsala@saske.sk

DOI: 10.1016/S0079-6123(06)61011-X 171


172

site of injury and in areas located close to the epicenter of the impact site and, in these connections an
upregulation of NOS was noted in neurons and interneurons. However, the upregulation of NOS expres-
sion was also seen in interneurons located just rostrally and caudally to the lesion. A quantitative analysis
of laminar distribution of multiple cauda equina constriction (MCEC) induced NADPHd-stained neurons
revealed a considerable increase in these neurons in laminae VIII–IX 8 h postconstriction, and a highly
statistically significant increase of such neurons in laminae VII–X 5 days postconstriction in the
lumbosacral segments. Concurrently, the number of NADPHd-stained neurons on laminae I–II in LS
segments was greatly reduced. It is concluded that a greater understanding of NO changes after spinal cord
trauma is essential for the possibility of targeting this pathway therapeutically.

Keywords: spinal cord; trauma; cauda equina constriction; nitric oxide

The occurrence of NADPH diaphorase-stained and/ containing widely branching NADPHd-stained


or bNOS immunoreactive neurons in the gray neurons (Anderson, 1992; Valtschanoff et al.,
matter of the spinal cord 1992; Dun et al., 1993; Lee et al., 1993; Saito
et al., 1994; Vizzard et al., 1994b; Wetts and
Small, morphologically heterogeneous popula- Vaughn, 1994; Burnett et al., 1995; Maršala et al.,
tions of neurons containing nicotinamide adenine 1997, 1998).
dinucleotide phosphate diaphorase (NADPHd), A study from our laboratory aimed at segmental
and/or brain nitric oxide synthase (bNOS), an en- and laminar distribution of NADPHd-stained
zyme known to generate nitric oxide (NO), have neurons in the rabbit revealed a large number of
been identified at various sites in the central ner- NADPHd-positive neurons in the spinal cord fall-
vous system (CNS) in a number of mammalian ing into six categories, N1–N6, while others could
species including the human brain (Kowall et al., not be classified (Maršala et al., 1999). Large
1985, 1987; Kowall and Mueller, 1988; Mizukawa numbers of NADPHd-stained neurons were iden-
et al., 1989; Bredt et al., 1990; Vincent and tified in the superficial dorsal horn and around the
Kimura, 1992; Egberongbe et al., 1994; Vincent, central canal at all spinal levels and in the inter-
1994). Immunohistochemistry of bNOS revealed mediolateral cell column at thoracic and upper
that the occurrence of this enzyme is almost com- lumbar levels. NADPHd-stained somata of the
pletely homotopic with the localization of neurons pericentral region were divided into a thin sub-
stained for NADPHd (Lukáčová et al., 1999). ependymal cell column containing longitudinally
It has long been known that NADPHd-stained arranged, small bipolar neurons with processes
neurons are present in the spinal cord, especially in penetrating deeply into the intermediolateral cell
the substantia gelatinosa, lamina X, and the inter- column and/or running rostrocaudally in the sub-
mediolateral cell column (Thomas and Pearse, ependymal layer. The second pericentral cell col-
1964; Mizukawa et al., 1989; Blottner and umn located more laterally in lamina X contains
Baumgarten, 1992; Bredt and Snyder, 1992). How- NADPHd-stained neurons with long dendrites ra-
ever, the analysis of NADPHd-stained and/or diating in the transverse plane. In the pericentral
bNOS-immunoreactive neurons in the spinal cord region (lamina X), close association of NADPHd-
of different animal species (Anderson, 1992; stained somata and fibers, and mostly longitudi-
Valtschanoff et al., 1992; Dun et al., 1993; Vizzard nally oriented blood vessels was detected. Somata
et al., 1994a, 1995) revealed a morphologically of the sacral parasympathetic nucleus seen in seg-
heterogeneous pattern of NADPHd-stained neu- ments S1–S3, exhibited prominent NADPHd
ronal pools ranging from bipolar, poorly branched staining accompanied by heavily stained fibers ex-
NADPHd-stained neurons in the superficial dor- tending from Lissauer’s tract through lamina I
sal horn to highly differentiated neurons in the along the lateral edge of the dorsal horn to lamina
pericentral region (lamina X), deep dorsal horn V. A massive dorsal gray commissure, highly posi-
(laminae IV–V), and dorsal gray commissure tive in NADPHd staining, was found in segments
173

Sl–S3. Scattered positive cells were also found in NADPHd-staining could not be detected in the
the deeper dorsal horn, ventral horn, and white lateral funiculus consistent with the location of the
matter. Fiber-like NADPHd staining was found in dorsal spinocerebellar tract. Numerous, mostly
the superficial dorsal horn and pericentral region thin NADPHd-stained axonal profiles could be
in all cervical, thoracic, and lumbosacral segments. detected in the dorsolateral funiculus in all seg-
Dense, punctate, non-somatic NADPHd staining ments studied and in juxtagriseal portion of the
was detected in the superficial dorsal horn, in the lateral funiculus in the extent of the cervical and
pericentral region all along the rostrocaudal axis lumbar enlargement. A massive occurrence of axo-
and in the phrenic nucleus (segments C4–C5), nal NADPHd-staining was detected in the
dorsal nucleus (segments Th2–L2), Onuf’s nucleus juxtagriseal layer of the ventral funiculus along
(segments S1–S3), and the dorsal part of the dorsal the rostrocaudal axis of the spinal cord. The
gray commissure (segments S1–S3; Maršala et al., prominent NADPHd-stained bundles containing
1999). smooth, thick, non-varicose axons were identified
in the mediobasal and central portion of the ven-
tral funiculus. First, the sulcomarginal fasciculus
The distribution of NADPH diaphorase-positive was found in the basal and medial portion of the
and/or NOS immunolabeled fibers in spinal cord ventral funiculus in all cervical and upper thoracic
white matter segments. Second, more caudally, a long proprio-
spinal bundle displaying prominent NADPHd-
The funicular distribution of NADPHd-stained staining was localized in the central portion of the
axons was examined in the white matter of the ventral funiculus throughout the Th3–L3 segments
rabbit spinal cord by using horizontal, para- (Maršala et al., 2003).
sagittal, and transverse sections. Four morphologi-
cally distinct kinds of NADPHd-stained axons
2.5–3.5 mm in diameter were found in the sulco-
marginal fasciculus as a part of the ventral funi- NO/cGMP signaling in the spinal cord
culus in the cervical and upper thoracic segments
and in the long propriospinal bundle of the ventral For a long time it was believed that interneuronal
funiculus in Th3–L3 segments. Varicose NADPHd- communication was realized exclusively via synap-
stained axons of the sympathetic preganglionic tic contacts. However, since the discovery of NO
neurons, characterized by widely spaced varicosi- as a neuronal transmitter (Snyder and Bredt, 1991;
ties, were found in the ventral funiculus of Th2–L3 Dawson et al., 1992), an increasing interest has
segments. A third kind of NADPHd-stained ultra- focused on its ability to modulate synaptic func-
fine axons, 0.3–0.5 mm in diameter with numerous tions. NO passes through cell membranes without
varicosities mostly spherical in shape, was identified specific release or uptake mechanisms and acts di-
in large number within Lissauer’s tract. The last rectly on surrounding neural tissue in extended
group of NADPHd-stained axons, 1.0–1.5 mm in spatial limits of approximately 300–400 mm (Wood
diameter, occurred in the Lissauer tract. Most of and Garthwaite, 1994; Vincent, 1994). After its
these axons were traceable for considerable dis- synthesis in the soma of neurons, NO induces
tances and generating spherical and elliptical forms changes in signal-related functions in several ways.
of varicosities. In particular, the activation of soluble guanylyl
The majority of NADPHd-stained axons iden- cyclases (sGC), the formation of cyclic guanosine
tified in the cuneate and gracile fascicles were 30 ,50 -monophosphate (cGMP), and the action of
concentrated in the deep portion of the cGMP-dependent protein kinases have been iden-
dorsal funiculus. An extremely reduced number tified as the main signal transduction pathway of
of NADPHd-stained axons, confirmed by a com- NO in the nervous system (Bredt and Snyder,
puter-assisted image-processing system were found 1992; Wang and Robinson, 1997; Smolenski et al.,
in the dorsal half of the gracile fascicle. Axonal 1998).
174

It is known that the intracellular level of cGMP of different subunits of sGC is associated with the
is strictly controlled by its rate of synthesis via GCs binding of NO to a heme group, leading to for-
and/or by the rate of its degradation via 30 ,50 -cyclic mation of a nitrosyl–heme complex and conse-
nucleotide phosphodiesterases (PDEs; Beavo, quent conformational change (Humbert et al.,
1995; Lucas et al., 2000; Francis et al., 2001). GC 1990). The activation of sGC and the formation of
can be divided into two main groups, i.e., the cGMP have been identified as the main NO/cGMP
membrane-bound or pGC and the cytosolic or signal transduction pathway in the CNS (Bredt
sGC (Schmidt, 1992; Murad, 1994; Wedel and and Snyder, 1992; Wang and Robinson, 1997;
Garbers, 1997). PDEs comprise a large group of Smolenski et al., 1998). In view of the abundance
enzymes that hydrolyze cGMP to its inactive 50 - of NO-responsive structures in the brain, it is pro-
derivate. PDE families (PDE1–11) and a large posed that NO-cGMP signaling will be part of
number of their isoforms are characterized with neuronal information processing at many levels.
distinct localization pattern in the brain. However, To date, little is known about the role of NO-
only three PDE families, i.e., PDE5, PDE9, and a cGMP signaling in the spinal cord. Morris et al.
photoreceptor-specific PDE6, identified in retina, (1994) showed that the NO-cGMP pathway is
specifically use cGMP as a substrate (Beavo, 1995; located primarily in the superficial layers of the
van Staveren et al., 2003). cGMP can be degraded dorsal horn in the neonatal rat spinal cord. The
by dual-substrate PDEs (PDE1, PDE2, PDE10, NO-mediated cGMP response in immature spinal
and PDE11) as well. cord was also confirmed by Vles et al. (2000). The
pGC is a large transmembrane molecule that has increased level of cGMP in the dorsal horn of the
a receptor domain at the outside and the cata- spinal cord seems to be associated with hyper-
lytic site at the inside of the cell (Vles et al., 2000). algesia through activation of cGMP dependent
This enzyme is activated through interaction of protein kinase Ia (Aley et al., 1998; Tao et al.,
peptide hormones with the receptor domain. In the 2000). NO-mediated cGMP was also identified in
spinal cord, the activation of pGC has only been parvalbumin-immunoreactive (GABA-ergic) neu-
demonstrated for atrial natriuretic peptide (ANP), rons and in the axon terminals of the ventral horn
which stimulates cGMP accumulation in GABA- probably apposing the motor neurons (Vles et al.,
ergic structures of laminae I–III of the rat cervical 2000).
spinal cord (Vles et al., 2000); these neurons are
involved in controlling the action of the spinal loco-
motor circuit. The GABA-ergic agonist, Baclofen, NO-mediated cGMP synthesis in the spinal cord
a well-known drug in the treatment of spasticity, after traumatic injury
has been shown to express an effect on ANP-
mediated cGMP synthesis in the superficial dorsal Recent data from our laboratory have shown that
horn layers of the cervical spinal cord (de Louw surgically induced Th7 constriction of 24 h dura-
et al., 2002). These data propose that the clinical tion stimulated both the constitutive NOS activity
effect of Baclofen on the reduction of spasticity and cGMP level by 120 and 131%, respectively, in
may be involved in the triggering of a phosphory- non-compartmentalized white matter of Th8–Th9
lation/dephosphorylation cycle of the intracellular segments, located just caudally to the site of injury
part of the ANP receptor and, subsequently, a (Lukáčová et al., 2002). NO-mediated cGMP for-
downregulation of the pGC activity (Potter and mation was only slightly increased in the dorsal
Hunter, 2001). funiculus of Th5–Th9 segments, composed entirely
sGC is a heterodimeric enzyme composed of of long ascending axons. However, a strong in-
four different subunits (a1, a2, b1, b2; Russwurm crease of cGMP formation was noted in the lateral
et al., 1998); however, all catalytically active GC funiculus of segments located below the site of Th7
isoforms are built of the b1 subunit and either the constriction. These results suggest a decrease of
a1 or the a2 subunit (Gupta et al., 1997; Koesling the axoplasmic transport in ascending NOS-
and Friebe, 2000; Koglin et al., 2001). Activation immunoreactive (NOS-IR) axons, which clearly
175

prevail in the lateral funiculus in comparison with of NOS accompanied by an increase in cellular
descending axons. The NOS-IR neurons of the in- cGMP (Strosznajder and Chalimoniuk, 1996;
termediolateral cell column sending many NOS-IR Toborek et al., 2000). Morton and Bredt (1998)
dendrites into the lateral funiculus (Maršala have suggested that in primary cell cultures
et al., 1998, 1999), together with the NOS-IR ax- taken from the cerebellum of nNOS knockout
ons of the lateral spinothalamic tract may, upon mice, norepinephrine is able to stimulate cGMP
midthoracic constriction, provide the substrate for formation independently from sGC. In addition,
NO/cGMP formation. A significant decrease in the a selective inhibitor of sGC points to a possibility
level of cGMP found in the ventral funiculus of that extracellular cGMP released under basal
Th5–Th6 segments correlates with a significant de- conditions in the brain might come from both
crease of both the constitutive NOS activity, at- NO-sensitive and NO-insensitive GCs (Vallebuona
tributable to a slight constriction of the superficial and Raiteri, 1993; Luo et al., 1994; Fedele et al.,
portion of the ventral funiculus at Th7 level, and 1996). The elevation of cGMP levels could be
the axoplasmic transport of the NOS affecting elicited by atrial natriuretic factor, possibly
thick, highly NOS-IR axons (Lukáčová et al., localized in astroglial cells (de Vente et al., 1990).
2000). These axons form an ascending and, to a These non-neuronal elements form dense perinodal
lesser extent, a descending propriospinal bundle, processes approaching the node of Ranvier
connecting NOS-IR neurons in lumbosacral en- (Ransom et al., 1993) and, as such, are located
largement with the ventral motor nucleus at at sites known for a high rate of transmembrane
cervicothoracic level (Sherrington and Laslett, ion traffic between the axolemma and the extra-
1903; Barilari and Kuypers, 1969; Yezierski cellular space (Stys et al., 1991; Stys and Steffensen,
et al., 1980; Maršala et al., 2004; Lukáčová 1996). Based on biochemical and immunocyto-
et al., 2006). The detection of a considerably higher chemical experiments we can conclude that the
radioassay-assessed NOS activity in the ventral NO/cGMP signaling pathway is implicated in the
funiculus of the lumbosacral segments, in compar- pathology of SCI.
ison to cervical segments, supports the conclusions
based on immunocytochemical and histochemical
studies (Lukáčová et al., 1999; Lukáčová and NOS in the spinal cord: effect of traumatic injury in
Pavel, 2000). There are some other sources that the epicenter of injury
may influence the NO-mediated cGMP formation
in spinal cord injury (SCI). A high level of gluta- Spinal cord neurons are susceptible to toxic effects
mate produced at the site of the lesion in the of excitatory amino acids because of the excitatory
spinal cord (Mc Adoo et al., 1997) and an excessive amino acid glutamate, which is markedly released
accumulation of intracellular Ca2+ (Lehotský from glutamatergic neurons to the extracellular
et al., 1999) may stimulate NOS activity and space, stimulates N-methyl-D-aspartate (NMDA)
create suitable conditions for NO synthesis and receptors located on nitrergic neurons, and con-
its adverse effect on white matter. An extreme secutively enhances the release of NO (Prast and
production of NO and superoxide, appearing Philippu, 2001). Both NO and glutamate, when
early after SCI in its close vicinity, may stimulate overstimulated, can initiate a neurotoxic cascade.
the formation of peroxynitrite (ONOO) that An increasing interest has focused on the role of
in turn may break up, thus giving nitrogen NO at the site of SCI and in areas located close to
dioxide and a compound having a hydroxyl-like the epicenter of the impact site.
reactivity leading to oxidation of protein, DNA, Acute changes in NO production after SCI have
and membrane-damaging lipid peroxidation been studied by Liu et al. (2000), demonstrating a
(Lukáčová et al., 1994). Furthermore, the release rapid increase of NO immediately after injury fol-
of free fatty acids, particularly arachidonic acid, in lowed by a rapid decline during approximately the
SCI (Horrocks et al., 1984; Halát et al., 1987; first 60 min after injury. A microdialysis study by
Nakano et al., 1990) may lead to an overexpression Nakahara et al. (2002) has also demonstrated a
176

second wave of NO production after SCI in the delayed enhancement in cNOS activity observed
rat — the second wave was observed 24 h and 3 at day 7 in the hemisected region may result from
days after injury. Long-term changes in the pro- the alterations in the neuronal circuitry (Xu et al.,
duction of parenchymal NO after SCI have been 2000), subsequent proliferation of fibers contain-
studied by detecting activities of NOS at various ing NOS, and the expression of NOS observed in
posttraumatic periods. Recent data obtained by axonal swelling after SCI (Guizar-Sahagun et al.,
Trudrung et al. (2000) have shown the upregula- 1998). A strong increase of iNOS activity was
tion of NOS expression in neurons adjacent to the noted after thoracic spinal cord hemisection in
lesion on both sides of a complete transverse lesion both postoperative times studied. These data sug-
of the spinal cord. Yick et al. (1998) have described gest that an increase of iNOS expression within
the increase of nNOS expression in Clarke’s col- polymorphonuclear leucocyte and macrophages
umn 3 days after hemisection, reaching its maxi- (Xu et al., 2001; Chatzipanteli et al., 2002), ap-
mum 20 days after injury. The upregulation of pearing obvious from 3 h up to 14 days, is con-
NOS expression was also seen in interneurons lo- sistent with the inflammatory response. In CNS
cated rostrally and caudally to the lesion (Wu inflammation, infiltrated monocytes and macroph-
et al., 1994; Sharma et al., 2000; Lukáčová et al., ages are the primary source of iNOS, but the
2005). Surgically induced Th7 constriction of 24 h presence of endotoxins and proinflammatory
duration caused an increase of calcium-dependent cytokines induce similar response in astrocytes
nitric oxide synthase (cNOS) activity in lateral and microglia, the resident CNS immune cells
funiculus just below the site of injury and a strong (Murphy et al., 1993). After SCI, iNOS positivity
decrease in the ventral funiculus of two segments, has been observed and colocalized with macroph-
located above the injured site (Lukáčová et al., ages, neurons, astrocytes, and oligodendrocytes
2000, 2002). Diaz-Ruiz et al. (2002) studied the (Kwak et al., 2005). Obviously, it is difficult to
changes in constitutive and inducible NOS (iNOS) specify unambiguously all the cell types that ex-
activities after severe SCI in rats that survived press iNOS in the damaged and hemorrhaged tis-
2–72 h. They observed a significant increase in sue invaded by leucocytes. Chatzipanteli et al.
cNOS after 4 and 8 h, followed by a decline to (2002) have identified polymorphonuclear leuco-
normal levels. In contrast, iNOS levels gradually cytes as a significant cellular source of iNOS pro-
increased after injury, and the difference reached tein (Chatzipanteli et al., 2002). Satake et al.
statistical significance at 72 h posttrauma. Simi- (2000) has identified macrophages and perivascu-
larly, time-dependent decrease in cNOS activity lar cells as predominant iNOS positive cells after
and the expression of iNOS were noted in trau- SCI. A rapid formation and release of NO by
matized Th10 segment and in segments located in cNOS activity early after SCI (Hamada et al.,
close vicinity of spinal cord trauma (Chatzipanteli 1996) and later, increased formation of NO via
et al., 2002). This study noted a significant de- iNOS in the affected spinal cord region (Chatzi-
crease in cNOS activities at 3 h postinjury, and a panteli et al., 2002) may accelerate neurodestruc-
gradual return to control values within 24 h. tive events and lead to a secondary inactivation of
Again, iNOS activities displayed contrasting cNOS enzyme activity, possibly by NO binding to
changes. Activity was increased in all spinal cord the heme iron of the enzyme (Rogers and Ignarro,
segments and time points studied (3 h-3 days). The 1992) and/or by the phosphorylation of cNOS by
most robust increase was detected at 24 h at rostral proteinkinases (Dalkara and Moskowitz, 1997).
and caudal segments relative to injury site. Data Similarly, the reduction in blood flow and decrease
obtained recently from our laboratory have shown in oxygen tension caused by the structural damage
a remarkable decrease of cNOS activity in the in- of cell bodies might be responsible for decreased
jured site analyzed 1 day after Th9–Th10 spinal cNOS activity at the site of injury (Tator and
cord hemisection (Lukáčová et al., 2006). At 7th Fehlings, 1991; Vanderkooi et al., 1991; Kim et al.,
day of survival the enzyme activity returned to 1993; Rengasamy and Johns, 1993). These data
approximately half of the control value. The provide strong evidence that intrinsic properties of
177

neurons contribute to the different cellular re- et al., 2000, 2001). NADPHd histochemistry was
sponses. The changes in the constitutive and iNOS used as a marker of NOS-containing neurons. The
activities observed after spinal cord hemisection appearance and the time course of Fos-like
within the site of injury support the participation of immunoreactive, NADPHd, and double-labeled
NO in the pathology of SCI and result in a de- neurons was studied at 2 and 8 h postconstriction,
creased neuronal viability. In principle, the primary characterized as the incipient phase of cauda
injury of the spinal cord evokes an inflammatory equina syndrome. An increase in Fos-like
response marked by generation of tumor necrosis immunoreactivity in superficial laminae (I–II)
factor-a, the accumulation and adhesion of ne- and enhanced NADPHd staining of lamina VIII
utrophils, and the activation of endothelial cells neurons were found. A statistically significant in-
and macrophages at the site of injury (Taoka et al., crease in Fos-like immunoreactive neurons was
1997). These events, in turn, may contribute to found in laminae I–II and VIII–X 8 h postcon-
neuronal death or injury by inducing an excessive striction, and in contrast, a prominent decrease in
formation of NO and oxygen free radicals (Merril Fos-like immunoreactive neurons was found in
et al., 1993). Another very important mediator of laminae I–II, accompanied by a statistically sig-
secondary CNS injury is oxidative-nitrosative stress nificant increase in Fos-like immunoreactive neu-
(Hall and Braughler, 1993), induced by antioxida- rons in more ventrally located laminae VII–X at 5
tive depletion and/or an excess production of ox- days postconstriction. A quantitative analysis of
ygen free radicals and NO at the site of injury. NO laminar distribution of constriction-induced
is able to promote oxidative damage by reacting NADPHd-stained neurons revealed a considera-
with superoxide anion to form a strong oxidant, ble increase in these neurons in laminae VIII–IX
ONOO, and by perturbing ion metabolism (Beck- 8 h postconstriction and highly statistically signif-
man et al., 1990; Reif and Simmons, 1990). The icant increase in NADPHd-stained neurons in
rate of ONOO formation is three times faster than laminae VII–X 5 days postconstriction. Concur-
the dismutation of the superoxide anion to hydro- rently, the number of NADPHd-stained neurons
gen peroxide, the reaction catalyzed by superoxide in laminae I–II was greatly reduced. While a low
dismutase (SOD) (Dawson and Dawson, 1996). number of double-labeled neurons was found
Thus, at appropriate concentrations NO can effec- throughout the gray matter of lower lumbar and
tively compete with SOD for superoxide anion. In sacral segments at 2 h postconstriction, a statisti-
addition, NOS may generate ONOO directly by cally significant number of double-labeled neurons
producing both NO and superoxide (Iadecola, was found in lamina X 8 h and in laminae VII–X 5
1997). NO can oxidize lipids, proteins, and nucleic days postconstriction (Orendáčová et al., 2000). A
acid (Dawson and Dawson, 1994) causing spinal prominent involvement of the spinal cord neurons
cord cytotoxicity either directly through NO or its appearing in the lumbosacral segments at the be-
derived species. Taken together, such processes ginning, and seen early (8 h postconstriction) and
point to a possibility that NO represents a critical in fully developed cauda equina syndrome (5 days
factor in determining the extent of the functional postconstriction), results in a Fos-like immunore-
impairment of the spinal cord. activity and strongly enhanced NADPHd staining
of some neuronal pools.

NADPHd-exhibiting and Fos-like immunolabeling


of intrinsic spinal cord neurons in a model of MCEC Immunolabeling and cNOS activity in the incipient
cauda equina syndrome of the dog
We found considerable differences in the segmen-
tal and laminar distribution of Fos-like immuno- Remarkable changes appeared in the distribution
reactive and NADPHd-stained neurons in the and appearance of NOS-IR neurons in the lower
lower lumbar and sacral segments of the dog spi- lumbar and sacral segments analyzed 5 days post-
nal cord using the model of MCEC (Orendáčová constriction in a dog model of cauda equina
178

constriction (Maršala et al., 2003). While NOS- cNOS activity in the lateral funiculus of the upper
immunolabeled neurons detected in the upper lumbar segments, no changes of cNOS activity
lumbar segments in the intermediate zone and could be detected in the white matter funiculus of
ventral horns were seen dispersed across both re- the upper and lower lumbar segments (Maršala
gions in a quite small number, passing more cau- et al., 2003). An increase of cNOS activity was
dally, the number of NOS-immunolabeled neurons detected in the non-compartmentalized white mat-
was steadily growing, reaching in the sacral seg- ter of S1–S3 segments, a finding strongly contrast-
ments not only to the lateral portion of the inter- ing with a statistically significant decrease of
mediate zone, but also encroaching to the lateral cNOS activity assessed in the cauda equina.
and ventralmost portion of the ventral horn
(Orendáčová et al., 2001). Along with this increas- Therapeutic interventions and NO reduction after
ing density of NOS-IR neurons, several more or spinal trauma
less loosely dispersed dotted NOS-IR foci ap-
peared in the ventral horn and in the dorsal gray Therapies aimed at limiting the evolution of seco-
commissure of S1–S3 segments. Highly positive, ndary damage have a long history and are still
usually small and middle-sized NOS-IR neurons extensively studied both in experimental and clin-
could be detected around the periphery of the ical use. Intravenous steroids (methylpredniso-
Onuf’s nucleus in S1–S3 segments. Under normal lone) have been registered for clinical use in
circumstances, such NOS-IR positivity could be acute SCI, although there is still considerable de-
seen only scarcely. It should be noted that in- bate whether this therapy was really proved to be
creased dotted non-somatic NOS-IR positivity safe and efficacious. Importantly, there is strong
was often detected around interneurons in the evidence that secondary damage is associated with
core region of the ventral horn and in the inter- invading inflammatory cells. Experimental results
mediate zone. In some NOS-IR positive foci, the have shown that depletion of hematogeneous ma-
dark small profiles truly matched the outlines of crophages (Popovich et al., 1999) and inhibiting
these small neurons having, of course, no somatic diapedesis of blood borne leucocytes by antibodies
NOS-IR positivity. These findings clearly docu- directed against leucocyte adhesion molecules
ment a high activation of NOS containing neurons (Mabon et al., 2000; Bao et al., 2004; Gris et al.,
in this compression-induced cauda equina injury. 2004) reduces secondary damage after SCI, and
Considerable segmental and regional differences improves motor, sensitive, and autonomic func-
of cNOS activity were found in the gray and white tion recovery in experimental animals.
matter regions of the lumbosacral segments of the However, as posttraumatic inflammation also
dog 5 days after MCEC surgery (Lukáčová et al., has a potentially beneficial role, better understand-
2004). The enzyme activity was assessed by a ra- ing of the molecular mechanisms and their timing
dioassay (Bredt and Snyder, 1990) in the gray in this process would allow more specific treat-
matter, divided into dorsal horn, intermediate ments. These might diminish destructive processes,
zone, ventral horn, and in the white matter, di- while preserving the protective and regenerative
vided into dorsal, lateral, and ventral columns, re- functions of inflammatory cells. Evolving under-
spectively, comprising the upper (L1–L3) and standing of the role of posttraumatic NO changes
lower (L4–L7) lumbar segments, then in the non- represents one example of such therapeutic strat-
compartmentalized gray and white matter of egy refinement.
S1–S3 segments and cauda equina. While a statis- Experiments with genetically manipulated ani-
tically significant decrease of cNOS activity was mals demonstrated improved recovery with both
found in all gray matter regions of the upper lum- neuronal NOS-deficient (Farooque et al., 2001)
bar segments, a statistically significant increase of and iNOS-deficient mice (Isaksson et al., 2005).
cNOS activity could be detected in the ventral Pharmacological inhibition of NO production by
horn of the lower lumbar segments. With the ex- aminoguanidine has been shown to improve func-
ception of a statistically significant increase of tional recovery in various independent studies
179

(Chatzipanteli et al., 2002; Zhang et al., 2003; Soy References


et al., 2004). The effect of a more specific iNOS
inhibitor, 1400W has been tested in a traumatic Aley, K.O., McCarter, G. and Levine, J.D. (1998) Nitric oxide
signaling in pain and nociceptor sensitization in the rat. J.
brain injury model, where it dramatically reduced
Neurosci., 18: 7008–7014.
lesion volume, even if administered with 18 h delay Anderson, C.R. (1992) NADPH diaphorase-positive neurons in
(Jafarian-Tehrani et al., 2005). the rat spinal cord include a subpopulation of autonomic
In conclusion, SCI is a very complex pathologi- preganglionic neurons. Neurosci. Lett., 139: 280–284.
cal situation. It is believed that effective therapy Bao, F., Chen, Y., Dekaban, G.A. and Weaver, L.C. (2004)
will require combined approaches for minimizing Early anti-inflammatory treatment reduces lipid peroxidation
and protein nitration after spinal cord injury in rats. J. Neuro-
adverse reactions in the tissue and maximizing the chem., 88: 1335–1344.
regenerative capacity of the spinal cord (Ramer Barilari, M.G. and Kuypers, J.M. (1969) Propriospinal fibers
et al., 2000; Thuret et al., 2006). Precise under- interconnecting the spinal enlargements in the cat. Brain
standing of the timing of iNOS expression in spe- Res., 14: 321–330.
Beavo, J.A. (1995) Cyclic nucleotide phosphodiesterases: func-
cific models of SCI, and development of new more
tional implications of multiple isoforms. Physiol. Rev., 75:
specific inhibitors might represent one component 725–748.
of such effective therapy. Beckman, J.S., Beckman, T.W., Chen, J., Marshall, P.A. and
Freeman, B.A. (1990) Apparent hydroxyl radical production
by peroxynitrite: implications for endothelial injury from
Abbreviations nitric oxide and superoxide. Proc. Natl. Acad. Sci. U.S.A.,
87: 1620–1624.
Blottner, D. and Baumgarten, H. (1992) Nitric oxide synthase
ANP atrial natriuretic peptide (NOS) containing sympathoadrenal cholinergic neurons of
cGMP cyclic guanosine 30 ,50 -monophos- the rat IML-cell column: evidence from histochemistry,
phate immunohistochemistry and retrograde labeling. J. Comp.
CNS central nervous system Neurol., 316: 45–56.
Bredt, D.S., Hwang, P.M. and Snyder, S.H. (1990) Localization
GC guanylyl cyclase
of nitric oxide synthase indicating a neuronal role for nitric
iNOS inducible nitric oxide synthase oxide. Nature, 347: 768–770.
MCEC multiple cauda equina constric- Bredt, D.S. and Snyder, S.H. (1990) Isolation of nitric oxide
tion synthase, a calmodulin-requiring enzyme. Proc. Natl. Acad.
NADPHd nicotinamide adenine dinucleo- Sci. U.S.A., 87: 682–685.
Bredt, D.S. and Snyder, S.H. (1992) Nitric oxide: a novel neu-
tide phosphate diaphorase
ronal messenger. Neuron, 8: 3–11.
NMDA N-methyl-D-aspartate Burnett, A.L., Saito, S., Maguire, M.P., Yamaguchi, H.,
NO nitric oxide Chang, T.S.K. and Hanley, D.F. (1995) Localization of ni-
NOS nitric oxide synthase tric oxide synthase in spinal nuclei innervating pelvic ganglia.
NOS-IR NOS-immunoreactive J. Urol., 153: 212–217.
Chatzipanteli, K., Garcia, R., Marcillo, A.E., Loor, K.E.,
ONOO peroxynitrite
Kraydieh, S. and Dietrich, W.D. (2002) Temporal and seg-
PDE phosphodiesterase mental distribution of constitutive and inducible nitric oxide
pGC particular guanylyl cyclase synthases after traumatic spinal cord injury: effect of amino-
SCI spinal cord injury guanidine treatment. J. Neurotrauma, 5: 639–651.
sGC soluble guanylyl cycle Dalkara, T. and Moskowitz, M.A. (1997) Neurotoxic and ne-
uroprotective roles of nitric oxide in cerebral ischaemia. Int.
SOD superoxide dismutase
Rev. Neurobiol., 40: 319–336.
Dawson, T.M. and Dawson, V.L. (1994) gp120 neurotoxicity in
primary cortical cultures. Adv. Neuroimmunol., 4: 167–173.
Dawson, T.M., Dawson, V.L. and Snyder, S.H. (1992) A novel
neuronal messenger molecule in brain: the free radical, nitric
Acknowledgments
oxide. Ann. Neurol., 32: 297–311.
Dawson, V.L. and Dawson, T.M. (1996) Nitric oxide actions in
This work was supported by the APVT grant: 51- neurochemistry. Neurochem. Int., 29: 97–110.
013002 and VEGA grants: 2/5134/25, 2/5135/25, Diaz-Ruiz, A., Ibarra, A., Perez-Severiano, F., Guizar-Sahagun,
and 2/6212/26. G., Grijalva, I. and Rios, C. (2002) Constitutive and inducible
180

nitric oxide synthase activities after spinal cord contusion in Iadecola, C. (1997) Bright and dark sides of nitric oxide in
rats. Neurosci. Lett., 319: 129–132. ischemic brain injury. Trends Neurosci., 20: 132–139.
Dun, N.J., Dun, S.L., Wu, S.Y., Förstermann, U., Schmidt, Isaksson, J., Farooque, M. and Olsson, Y. (2005) Improved
H.H.H.W. and Tseng, L.F. (1993) Nitric oxide synthase functional outcome after spinal cord injury in iNOS-deficient
immunoreactivity in the rat, mouse, cat and squirrel monkey mice. Spinal Cord, 43: 167–170.
spinal cord. Neuroscience, 54: 845–857. Jafarian-Tehrani, M., Louin, G., Royo, N.C., Besson, V.C.,
Egberongbe, Y.I., Gentleman, S.M., Falkai, P., Bogerts, B., Bohme, G.A., Plotkine, M. and Marchand-Verrecchia, C.
Polak, J.M. and Roberts, G.W. (1994) The distribution of (2005) 1400W, a potent selective inducible NOS inhibitor:
nitric oxide synthase immunoreactivity in the human brain. improves histopathological outcome following traumatic
Neuroscience, 59: 561–578. brain injury in rats. Nitric Oxide, 12: 61–69.
Farooque, M., Isaksson, J. and Olsson, Y. (2001) Improved Kim, N., Vardi, Y., Padma-Nathan, H., Daley, J., Goldstein, I.
recovery after spinal cord injury in neuronal nitric oxide and Saenz de Tejada, I. (1993) Oxygen tension regulates the
synthase-deficient mice but not in TNF-alpha-deficient mice. nitric oxide pathway: physiological role in penile erection.
J. Neurotrauma, 18: 105–114. J. Clin. Invest., 91: 437–442.
Fedele, E., Jin, Y., Varnier, G. and Raiteri, M. (1996) In vivo Koesling, D. and Friebe, A. (2000) Enzymology of soluble
microdialysis study of a specific inhibitor of soluble guanylyl guanylyl cyclase. In: Mayer B. (Ed.), Nitric Oxide. Springer,
cyclase on the glutamate receptor/nitric oxide/cyclic GMP Berlin, pp. 93–109.
pathway. Br. J. Pharmacol., 119: 590–594. Koglin, M., Vehse, K., Budaeus, L., Scholz, H. and Behrends,
Francis, S.H., Turko, I.V. and Corbin, J.D. (2001) Cyclic nuc- S. (2001) Nitric oxide activates the beta 2 subunit of soluble
leotide phosphodiesterases: relating structure and function. guanylyl cyclase in the absence of a second subunit. J. Biol.
Prog. Nucleic Acid Res. Mol. Biol., 65: 1–52. Chem., 276: 30737–30743.
Gris, D., Marsh, D.R., Oatway, M.A., Chen, Y., Hamilton, Kowall, N.W., Beal, M.F., Ferrante, R.J. and Martin, J.B.
E.F., Dekaban, G.A. and Weaver, L.C. (2004) Transient (1985) Topography of nicotinamide adenine dinucleotide
blockade of the CD11d/CD18 integrin reduces secondary phosphate-diaphorase staining neurons in rat striatum. Neuro-
damage after spinal cord injury, improving sensory, auto- sci. Lett., 59: 61–66.
nomic, and motor function. J. Neurosci., 24: 4043–4051. Kowall, N.W., Ferrante, R.J., Beal, M.F., Richardson, Jr.,
Guizar-Sahagun, G., Garcia-Lopez, P., Espitia, A.L., Grijalva, E.P., Sofroniew, M.V., Cuello, A.C. and Martin, J.B. (1987)
I., Franco-Bourland, R.E. and Madrazo, I. (1998) Transitory Neuropeptide Y, somatostatin and reduced nicotinamide ad-
expression of NADPH diaphorase (NOS) in axonal swellings enine dinucleotide phosphate diaphorase in the human stria-
after spinal cord injury. NeuroReport, 9: 2899–2902. tum: a combined immunocytochemical and enzyme
Gupta, G., Azam, M., Yang, L. and Danziger, R.S. (1997) The histochemical study. Neuroscience, 20: 817–828.
beta2 subunit inhibits stimulation of the alpha1/beta1 form Kowall, N.W. and Mueller, M.O. (1988) Morphology and dis-
of soluble guanylyl cyclase by nitric oxide: potential relevance tribution of nicotinamide adenine dinucleotide phosphate
to regulation of blood pressure. J. Clin. Invest., 100: (reduced form) diaphorase reactive neurons in human brain-
1488–1492. stem. Neuroscience, 26: 645–654.
Halát, G., Lukáčová, N., Chavko, M. and Maršala, J. (1987) Kwak, E.K., Kim, J.W., Kang, K.S., Lee, Y.H., Hua, Q.H.,
Effects of incomplete ischemia and subsequent recirculation Park, T.I., Park, J.Y. and Sohn, Y.K. (2005) The role of
on free palmitate, stearate, oleate and arachidonate levels in inducible nitric oxide synthase following spinal cord injury in
lumbar and cervical spinal cord of rabbit. Gen. Physiol. rat. J. Korean Med. Sci., 20: 663–669.
Biophys., 6: 387–399. Lee, J.H., Price, R.H., Williams, F.G., Mayer, B. and Beitz,
Hall, E.D. and Braughler, J.M. (1993) Free radicals in CNS A.J. (1993) Nitric oxide synthase is found in some spin-
injury. Res. Publ. Assoc. Res. Nerv. Ment. Dis., 71: 81–105. othalamic neurons and in neuronal processes that appose
Hamada, Y., Ikata, T., Katoh, S., Tsuchiya, K., Niwa, M., spinal neurons that express Fos induced by noxious stimu-
Tsutsumishita, Y. and Fukuzawa, K. (1996) Roles of nitric lation. Brain Res., 608: 324–333.
oxide in compression injury of rat spinal cord. Free Radic. Lehotský, J., Kaplán, P., Račay, P., Mézešová, V. and Raey-
Biol. Med., 20: 1–9. maekers, L. (1999) Distribution of plasma membrane Ca2+
Horrocks, L.A., Dorman, R.V. and Porcellati, G. (1984) Fatty pump (PMCA) isoforms in the gerbil brain: effect of
acids and phospholipids in brain during ischemia. In: Bes P., ischemia-reperfusion injury. Neurochem. Int., 35: 221–227.
Braquet R., Paoletti R. and Siesjö B.K. (Eds.), Cerebral Is- Liu, D., Ling, X., Wen, J. and Liu, J. (2000) The role of reactive
chemia Excerpta Medica. Elsevier, Amsterdam, Congress nitrogen species in secondary spinal cord injury: formation of
Series 654A, pp. 211–226. nitric oxide, peroxynitrite, and nitrated protein. J. Neuro-
Humbert, P., Niroomand, F., Fischer, G., Mayer, B., Koesling, chem., 75: 2144–2154.
D., Hinsch, K.D., Gausepohl, H., Frank, R., Schultz, G. and de Louw, A.J., de Vente, J., Steinbusch, H.P., Steinbusch,
Bohme, E. (1990) Purification of soluble guanylyl cyclase H.W., Troost, J. and Vles, J.S. (2002) Baclofen inhibits ANP-
from bovine lung by a new immunoaffinity chromatographic mediated cyclic GMP synthesis in the rat cervical spinal cord.
method. Eur. J. Biochem., 190: 273–278. Neurosci. Lett., 321: 120–122.
181

Lucas, K.A., Pitari, G.M., Kazerounian, S., Ruiz-Stewart, I., Maršala, J., Lukáčová, N., Čı́žková, D., Lukáč, I., Kuchárová,
Park, J., Schulz, S., Chepenik, K.P. and Waldman, S.A. K. and Maršala, M. (2004) Premotor nitric oxide synthase
(2000) Guanylyl cyclases and signaling by cyclic GMP. immunoreactive pathway connecting lumbar segments with
Pharmacol. Rev., 52: 375–414. the ventral motor nucleus of the cervical enlargement in the
Lukáčová, N., Chavko, M. and Maršala, J. (1994) Ischemia/ dog. J. Chem. Neuroanat., 27: 43–54.
reperfusion-induced lipid peroxidation in the central nervous Maršala, J., Maršala, M., Vanický, I. and Taira, Y. (1999)
system. Biologia, 49: 871–879. Localization of NADPHd exhibiting neurons in the spinal
Lukáčová, N., Čı́žková, D., Maršala, M., Jalč, P. and Maršala, cord of the rabbit. J. Comp. Neurol., 406: 263–284.
J. (1999) Segmental and laminar distributions of nicotin- Maršala, J., Vanický, I., Maršala, M., Jalč, P., Orendáčová, J.
amide adenine dinucleotide phosphate–diaphorase expressing and Taira, Y. (1998) NADPH diaphorase in the spinal cord
and neuronal nitric oxide synthase–immunoreactive neurons of dogs. Neuroscience, 85: 847–862.
versus radioassay detection of catalytic nitric oxide synthase Mc Adoo, J., Hughes, M.G., Xu, G.Y., Robak, G. and DeC-
activity in the rabbit spinal cord. Neuroscience, 94: 229–237. astro, Jr., R., (1997) Microdialysis studies of the role of
Lukáčová, N., Čı́žková, D., Maršala, M., Pavel, J., Jalč, P., chemical agents in secondary damage upon spinal cord in-
Šulla, I., Kafka, J. and Maršala, J. (2000) Effect of midtho- jury. J. Neurotrauma, 14: 507–515.
racic spinal cord constriction on catalytic nitric oxide synt- Merril, J.E., Ignarro, L., Sherman, M.P., Melinek, J. and Lane,
hase activity in the white matter columns of rabbit. T.E. (1993) Microglial cell cytotoxicity of oligodendrocytes is
Neurochem. Res., 25: 1139–1148. mediated through nitric oxide. J. Immunol., 151: 2132–2141.
Lukáčová, N., Kafka, J., Čı́žková, D., Maršala, M. and Mizukawa, K., McGeer, P.L., Vincent, S.R. and McGeer, E.G.
Maršala, J. (2004) The effect of cauda equina constriction (1989) Distribution of reduced-nicotinamide-adenine-dinuc-
on nitric oxide synthase activity. Neurochem. Res., 29: leotide-phosphate diaphorase-positive cells and fibers in the
429–439. cat central nervous system. J. Comp. Neurol., 279: 281–311.
Lukáčová, N., Kolesárová, M., Kuchárová, K., Pavel, J., Morris, R., Southam, E., Gittins, S.R., de Vente, J. and Garth-
Kolesár, D., Radoňák, J., Maršala, M., Chalimoniuk, M., waite, J. (1994) The NO-cGMP pathway in neonatal rat
Langfort, J. and Maršala, J. (2006) The effect of a spinal cord dorsal horn. Eur. J. Neurosci., 6: 876–879.
hemisection on changes in nitric oxide synthase pools in the Morton, D.B. and Bredt, D.S. (1998) Norepinephrine increases
site of injury and in regions located far away from the injured cyclic GMP levels in cerebellar cells from neuronal nitric
site. Cell. Mol. Neurobiol., 26: 1365–1383. oxide synthase knockout mice. J. Neurochem., 71: 440–443.
Lukáčová, N. and Pavel, J. (2000) Catalytic nitric oxide synt- Murad, F. (1994) Regulation of cytosolic guanylyl cyclase by
hase activity in the white and gray matter regions of the nitric oxide: the NO-cyclic GMP signal transduction system.
spinal cord of rabbits. Physiol. Res., 49: 167–173. Adv. Pharmacol., 26: 19–33.
Lukáčová, N., Pavel, J., Jalč, P., Čı́žková, D., Maršala, M., Murphy, S., Simmons, M.L., Agullo, L., Garcia, A., Feinstein,
Silvašiová, M., Lukáč, I. and Maršala, J. (2002) The effect of D.L., Galea, E., Reis, D.J., Minc-Golomb, D. and Schwartz,
spinal cord constriction on NOS-cGMP signaling pathway in J.P. (1993) Synthesis of nitric oxide in CNS glial cells. Trends
the white matter of rabbit. Biologia, 57: 789–795. Neurosci., 16: 323–328.
Lukáčová, N., Pavel, J., Kuchárová, K. and Maršala, J. (2005) Nakahara, S., Yone, K., Setoguchi, T., Yamaura, I., Arishima,
Plasticity of the spinal neural circuitry in nitric oxide synthase Y., Yoshino, S. and Komiya, S. (2002) Changes in nitric
immunoreactive pathways after spinal cord hemisection. In: oxide and expression of nitric oxide synthase in spinal cord
Coleman R.M. (Ed.), Trends in Neurochemistry Research. after acute traumatic injury in rats. J. Neurotrauma, 19:
Nova Publishers, New York, pp. 189–222. 1467–1474.
Luo, D., Leung, E. and Vincent, S.R. (1994) Nitric oxide- Nakano, S., Kogure, K., Abe, K. and Yae, T. (1990) Ischemia-
dependent efflux of cGMP in rat cerebellar cortex: an in vivo induced alterations in lipid metabolism of the gerbil cerebral
microdialysis study. J. Neurosci., 14: 263–271. cortex: I. Changes in free fatty acid liberation. J. Neuro-
Mabon, P.J., Weaver, L.C. and Dekaban, G.A. (2000) Inhibi- chem., 54: 1911–1916.
tion of monocyte/macrophage migration to a spinal cord in- Orendáčová, J., Čı́žková, D., Kafka, J., Lukáčová, N.,
jury site by an antibody to the integrin alphaD: a potential Maršala, M., Šulla, I., Maršala, J. and Katsube, N. (2001)
new anti-inflammatory treatment. Exp. Neurol., 166: 52–64. Cauda equina syndrome. Prog. Neurobiol., 64: 613–637.
Maršala, J., Kafka, J., Lukáčová, N., Čı́žková, D., Maršala, M. Orendáčová, J., Maršala, M., Šulla, I., Kafka, J., Jalč, P.,
and Katsube, N. (2003) Cauda equina syndrome and nitric Čı́žková, D., Taira, Y. and Maršala, J. (2000) Incipient cauda
oxide synthase immunoreactivity in the spinal cord of the equina syndrome as a model of somatovisceral pain in dogs:
dog. Physiol. Res., 52: 481–496. spinal cord structures involved as revealed by the expression
Maršala, J., Kluchová, D. and Maršala, M. (1997) Spinal cord of c-fos and NADPH diaphorase activity. Neuroscience, 95:
gray matter layers rich in NADPH diaphorase positive neu- 543–557.
rons are refractory to ischemia-reperfusion-induced injury: a Popovich, P.G., Guan, Z., Wei, P., Huitinga, I., van Rooijen,
histochemical and silver impregnation study in rabbit. Exp. N. and Stokes, B.T. (1999) Depletion of hematogenous ma-
Neurol., 145: 165–179. crophages promotes partial hindlimb recovery and
182

neuroanatomical repair after experimental spinal cord injury. Beavo, J.A. and de Vente, J. (2003) mRNA expression pat-
Exp. Neurol., 158: 351–365. terns of the cGMP-hydrolyzing phosphodiesterases types 2,
Potter, L.R. and Hunter, T. (2001) Guanylyl cyclase-linked 5, and 9 during development of the rat brain. J. Comp. Ne-
natriuretic peptide receptors: structure and regulation. J. urol., 467: 566–580.
Biol. Chem., 276: 6057–6060. Strosznajder, J. and Chalimoniuk, M. (1996) Biphasic enhance-
Prast, H. and Philippu, A. (2001) Nitric oxide as modulator of ment of nitric oxide synthase activity and cGMP level fol-
neuronal function. Prog. Neurobiol., 64: 51–68. lowing brain ischemia in gerbils. Acta Neurobiol. Exp., 56:
Ramer, M.S., Harper, G.P. and Bradbury, E.J. (2000) Progress 71–81.
in spinal cord research: a refined strategy for the Interna- Stys, P.K. and Steffensen, I. (1996) Na(+)-Ca2+ exchange in
tional Spinal Research Trust. Spinal Cord, 38: 449–472. anoxic/ischemic injury of CNS myelinated axons. Ann. N.Y.
Ransom, B.R., Waxman, S.G. and Stys, P.K. (1993) Anoxic Acad. Sci., 779: 366–378.
injury of central myelinated axons: ionic mechanisms and Stys, P.K., Waxman, S.G. and Ransom, B.R. (1991) Na+-Ca2+
pharmacology. Res. Publ. Assoc. Res. Nerv. Ment. Dis., 71: exchanger mediates Ca2+ influx during anoxia in mammalian
121–151. central nervous system white matter. Ann. Neurol., 30:
Reif, D.W. and Simmons, R.D. (1990) Nitric oxide mediates 375–380.
iron release from ferritin. Arch. Biochem. Biophys., 283: Tao, Y.X., Hassan, A., Haddad, E. and Johns, R.A. (2000)
537–541. Expression and action of cyclic GMP-dependent protein kin-
Rengasamy, A. and Johns, R.A. (1993) Regulation of nitric ase I alpha in inflammatory hyperalgesia in rat spinal cord.
oxide synthase by nitric oxide. Mol. Pharmacol., 44: 124–128. Neuroscience, 95: 525–533.
Rogers, N.E. and Ignarro, L.J. (1992) Constitutive nitric oxide Taoka, Y., Okajima, K., Uchiba, M., Murakami, K., Kushi-
synthase from cerebellum is reversibly inhibited by nitric ox- moto, S., Johno, M., Naruo, M., Okabe, H. and Takatsuki,
ide formed from L-arginine. Biochem. Biophys. Res. Co- K. (1997) Role of neutrophils in spinal cord injury in the rat.
mmun., 189: 242–249. Neuroscience, 79: 1177–1182.
Russwurm, M., Behrends, S., Harteneck, C. and Koesling, D. Tator, C.H. and Fehlings, M.G. (1991) Review of the secondary
(1998) Functional properties of a naturally occurring isoform injury theory of acute spinal cord trauma with emphasis on
of soluble guanylyl cyclase. Biochem. J., 335: 125–130. vascular mechanisms. J. Neurosurg., 75: 15–26.
Saito, S., Kidd, G.J., Trapp, B.D., Dawson, T.M., Bredt, D.S., Thomas, E. and Pearse, A.G.E. (1964) The solitary active cells:
Wilson, D.A., Traystmann, R.J., Snyder, S.H. and Hanley, histochemical demonstration of damage-resistant nerve cells
D.F. (1994) Rat spinal cord neurons contain nitric oxide with a TPN-diaphorase reaction. Acta Neuropathol., 3:
synthase. Neuroscience, 59: 447–452. 238–249.
Satake, K., Matsuyama, Y., Kamiya, M., Kawakami, H., Thuret, S., Moon, L.D. and Gage, F.H. (2006) Therapeutic
Iwata, H., Adachi, K. and Kiuchi, K. (2000) Nitric oxide via interventions after spinal cord injury. Nat. Rev. Neurosci., 7:
macrophage iNOS induces apoptosis following traumatic 628–643.
spinal cord injury. Brain Res. Mol. Brain Res., 85: 114–122. Toborek, M., Garrido, R., Malecki, A., Kaiser, S., Mattson,
Schmidt, H.H. (1992) NO, CO and OH: endogenous soluble M.P., Hennig, B. and Young, B. (2000) Nicotine attenuates
guanylyl cyclase-activating factors. FEBS Lett., 307: 102–107. arachidonic acid-induced overexpression of nitric oxide synt-
Sharma, H.S., Westman, J., Gordh, T. and Nyberg, F. (2000) hase in cultured spinal cord neurons. Exp. Neurol., 161:
Spinal cord injury induced c-fos expression is reduced by 609–620.
p-CPA, a serotonin synthesis inhibitor: an experimental Trudrung, P., Wirth, U. and Mense, S. (2000) Changes in the
study using immunohistochemistry in the rat. Acta Neuro- number of nitric oxide-synthesizing neurons on both sides of
chir. Suppl., 76: 297–301. a chronic transection of the rat spinal cord. Neurosci. Lett.,
Sherrington, C.S. and Laslett, E.E. (1903) Observations on 287: 125–128.
some spinal reflexes and the inter connection of spinal seg- Vallebuona, F. and Raiteri, M. (1993) Monitoring of cyclic
ments. J. Physiol. (Lond.), 29: 58–96. GMP during cerebellar microdialysis in freely-moving rats as
Smolenski, A., Burkhardt, A.M., Eigenthaler, M., Butt, E., an index of nitric oxide synthase activity. Neuroscience, 57:
Gambaryan, S., Lohmann, S.M. and Walter, U. (1998) 577–585.
Functional analysis of cGMP-dependent protein kinases I Valtschanoff, J.G., Weinberg, R.J. and Rustioni, A. (1992)
and II as mediators of NO/cGMP effects. Naunyn NADPH diaphorase in the spinal cord of rats. J. Comp.
Schmiedebergs Arch. Pharmacol., 358: 134–139. Neurol., 321: 209–222.
Snyder, S.H. and Bredt, D. (1991) Nitric oxide as a neuronal Vanderkooi, J.M., Erecinska, M. and Silver, I.A. (1991) Oxygen
messenger. Trends Pharmacol. Sci., 12: 125–128. in mammalian tissue: methods of measurement and affinities
Soy, O., Aslan, O., Uzun, H., Barut, S., Igdem, A.A., Belce, A. of various reactions. Am. J. Physiol., 260: C1131–C1150.
and Colak, A. (2004) Time-level relationship for nitric oxide de Vente, J., Manshanden, C.G., Sikking, R.A., Ramaekers,
and the protective effects of aminoguanidine in experimental F.C. and Steinbusch, H.W. (1990) A functional parameter to
spinal cord injury. Acta Neurochir. (Wien.), 146: 1329–1336. study heterogeneity of glial cells in rat brain slices: cyclic
van Staveren, W.C., Steinbusch, H.W., Markerink-van Itter- guanosine monophosphate production in atrial natriuretic
sum, M., Repaske, D.R., Goy, M.F., Kotera, J., Omori, K., factor (ANF)-responsive cells. Glia, 3: 43–54.
183

Vincent, S.R. (1994) Nitric oxide: a radical neurotransmitter in Wetts, R. and Vaughn, J.E. (1994) Choline acetyltransferase
the central nervous system. Prog. Neurobiol., 42: 129–160. and NADPH diaphorase are co-expressed in rat spinal cord
Vincent, S.R. and Kimura, H. (1992) Histochemical mapping of neurons. Neuroscience, 63: 1117–1124.
nitric oxide synthase in the rat brain. Neuroscience, 46: Wood, P.L. and Garthwaite, J. (1994) Models for the diffu-
755–784. sional spread of nitric oxide: implications for neuronal nitric
Vizzard, M.A., Erdman, S.L. and de Groat, W.C. (1995) In- oxide signalling and its pharmacological properties. Neuro-
creased expression of neuronal nitric oxide synthase (NOS) in pharmacology, 33: 1235–1244.
visceral neurons after nerve injury. J. Neurosci., 15: Wu, W., Liuzzi, F.J., Schinco, F.P., Depto, A.S., Li, Y., Mong,
4033–4045. J.A., Dawson, T.M. and Snyder, S.H. (1994) Neuronal nitric
Vizzard, M.A., Erdman, S.L., Erickson, V.L., Stewart, oxide synthase is induced in spinal neurons by traumatic in-
R.J., Roppolo, J.R. and de Groat, W.C. (1994a) Localiza- jury. Neuroscience, 61: 719–726.
tion of NADPH diaphorase in the lumbosacral spinal cord Xu, J., Kim, G.M., Chen, S., Yan, P., Ahmed, S.H., Ku, G.,
and dorsal root ganglia of the cat. J. Comp. Neurol., 339: Beckman, J.S., Xu, X.M. and Hsu, C.Y. (2001) iNOS and
62–75. nitrotyrosine expression after spinal cord injury. J. Neuro-
Vizzard, M.A., Erdman, S.L., Roppolo, J.R., Förstermann, U. trauma, 18: 523–532.
and de Groat, W.C. (1994b) Differential localization of neu- Xu, W., Liu, L., Smith, G.C. and Charles, G. (2000) Nitric
ronal nitric oxide synthase immunoreactivity and NADPH- oxide upregulates expression of DNA-PKCs to protect cells
diaphorase activity in the cat spinal cord. Cell. Tissue Res., from DNA-damaging anti-tumour agents. Nat. Cell. Biol., 2:
278: 299–309. 339–345.
Vles, J.S., de Louw, A.J., Steinbusch, H., Markerink-van Itter- Yezierski, R.P., Culberson, J.L. and Brown, P.B. (1980) Cells of
sum, M., Steinbusch, H.W., Blanco, C.E., Axer, H., Troost, origin of propriospinal connections to cat lumbosacral gray
J. and de Vente, J. (2000) Localization and age-related as determined with horseradish peroxidase. Exp. Neurol., 69:
changes of nitric oxide- and ANP-mediated cyclic-GMP syn- 493–512.
thesis in rat cervical spinal cord: an immunocytochemical Yick, L.W., Wu, W., So, K.F. and Wong, S.Y. (1998) Time
study. Brain Res., 857: 219–234. course of NOS expression and neuronal death in Clarke’s
Wang, X. and Robinson, P.J. (1997) Cyclic GMP-dependent nucleus following traumatic injury in adult rat spinal cord.
protein kinase and cellular signaling in the nervous system. Neurosci. Lett., 241: 155–158.
J. Neurochem., 68: 443–456. Zhang, X.Y., Zhou, C.S., Jin, A.M., Tian, J., Zhang, H., Yao,
Wedel, B.J. and Garbers, D.L. (1997) New insights on the W.T. and Zheng, G. (2003) Effect of aminoguanidine on the
functions of the guanylyl cyclase receptors. FEBS Lett., 410: recovery of rat hindlimb motor function after spinal cord
29–33. injury. Di Yi Jun Yi Da Xue Xue Bao, 23: 687–689.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 12

Aquaporins: role in cerebral edema and brain water


balance$

Zsolt Zador1,2, Orin Bloch1,2, Xiaoming Yao1,2 and Geoffrey T. Manley1,2,

1
Department of Neurological Surgery, University of California, San Francisco, CA, USA
2
Brain and Spinal Injury Center, University of California, San Francisco, CA 94110, USA

Abstract: The regulation of water balance in the brain is crucial. A disruption in this equilibrium causes an
increase in brain water content that significantly contributes to the pathophysiology of traumatic brain
injury, hydrocephalus, and a variety of neurological disorders. The discovery of the aquaporin (AQP)
family of membrane water channels has provided important new insights into the physiology and pathology
of brain water homeostasis. A number of recent studies are described in the review that demonstrated the
important role of AQP1 and AQP4 in brain water balance and cerebral edema. Phenotypic analyses of
AQP deficient mice have allowed us to explore the role of these membrane water channels in the mech-
anisms of cytotoxic edema, vasogenic edema, and CSF production. These studies indicate that AQP4 plays
significant role in the development of cytotoxic edema and the absorption of excess brain water resulting
from vasogenic edema. They also have demonstrated the role of AQP1 in CSF production and maintenance
of steady-state ICP. The ability to modulate water flux through AQP deletion has provided new insights
into brain water homeostasis and suggested a number of new research directions. However, these efforts
have not yet translated to the treatment human clinical diseases. These advances will require the deve-
lopment of AQP inhibitors and activators to establish the benefit modulating the function of these water
channels.

Keywords: water transport; aquaporin; edema; brain injury; stroke; epilepsy; cytotoxic; vasogenic

Introduction water transport (Verkman, 2002). To date, over 12


different members of the AQP family have been
Water is regarded as the matrix of life: it is an identified, and a number of them have been shown
essential solvent for all living organisms. Although to contribute to rapid water flux in various tissues
it diffuses through tissues relatively freely, the including the kidney, lung, gastrointestinal tract,
transport of water across cell membranes is facil- and central nervous system (CNS) (Agre et al.,
itated by highly efficient transmembrane channels 2002; Verkman, 2002).
called aquaporins (AQPs). The selective water flow The regulation of water balance in the brain is
through AQPs is passive, moving along osmotic crucial. Normally, water transport is tightly regu-
gradients, which provide the driving force for lated to maintain a strict homeostatic balance bet-
ween the cerebral vascular, brain tissue, and
$
Supported by NIH NS050173 and the UCSF Brain and Spi- cerebrospinal fluid (CSF) compartments. A dis-
nal Injury Center. ruption in this equilibrium causes an increase in
Corresponding author. Tel.: +415-206-4536; Fax: +415-206- brain water content that significantly contributes
3948; E-mail: manleyg@neurosurg.ucsf.edu to the pathophysiology of traumatic brain injury,

DOI: 10.1016/S0079-6123(06)61012-1 185


186

hydrocephalus, and a variety of neurological dis- expression pattern suggests they may contribute to
orders (Fishman, 1975). The rigid nature of the the underlying mechanism of cerebral edema.
skull provides little capacity to buffer intracranial
volume changes, and beyond a limited threshold
the intracranial pressure (ICP) rises rapidly. The Aquaporins and brain water balance
increased ICP can ultimately lead to the impair-
ment of cerebral blood flow resulting in further The brain is composed of nearly 78% water
brain injury and death. In spite of the clinical im- (McIlwain and Bachelard, 1985). While the ma-
portance of brain water balance, the molecular jority of the brain water is intracellular, it is also
mechanisms remain poorly understood and the distributed in the extracellular space and the CSF
therapeutic interventions have improved little over compartments (cerebral ventricles and sub-
past 80 years (Weed and McKibben, 1919). A arachnoid space). Water enters the brain through
number of recent studies have demonstrated that the blood-brain barrier or across the choroids
the AQP water channels play a significant role in plexus (Fig. 1). Under normal physiological con-
brain water balance and cerebral edema. Thus, ditions, the extracellular fluid in the brain is be-
they may provide a novel molecular target for the lieved to flow into the cerebral ventricles or
treatment of a number of neurological disorders. subarachnoid space and exit the brain through
The aim of this review is to summarize the current the arachnoid granulations into to the venous sys-
understanding of AQPs in cerebral edema and tem (Abbott, 2004). Recent studies of transgenic
brain water balance. mice lacking AQP1 and AQP4 have helped to de-
fine the role of these channels in CSF production
and absorbtion.
Aquaporin expression in the CNS The choroid plexus epithelium expresses a va-
riety of ion transporters and channels that provide
A number of AQPs are expressed in the brain and the osmotic driving force for para- and transcel-
spinal cord. Two of these water channels, AQP1 lular water flux into the cerebral ventricles to form
and AQP4, are expressed in the brain at tissue– CSF (Wright et al., 1977). The rate of CSF
fluid interfaces important for brain water balance. secretion by the choroid plexus is substantial,
AQP1 is expressed in the apical membrane of the comparable to the rate of fluid reabsorption in the
choroid plexus epithelium (Hasegawa et al., 1994; proximal tubules (Welch, 1963). Earlier studies
Speake et al., 2003), where it has been shown to had demonstrated a high osmotic–diffusion ratio
facilitate CSF secretion into the cerebral ventricles of the choroids plexus, suggesting the presence of
(Oshio et al., 2005). AQP4 is expressed in the a water pore-like pathway (Wright et al., 1977).
basolateral membrane of the ependymal cells lin- To explore this hypothesis, our group developed
ing the cerebral ventricles, where it may play a novel methods to measure choroids plexus water
role in water transport between the brain paren- permeability and CSF production in wildtype
chyma and CSF compartments. In addition, and AQP1-null mice. We found that deletion of
AQP4 is expressed in the in the astocytic foot AQP1 reduced osmotically induced water trans-
processes in direct contact with the cerebral port in the choroid plexus by fivefold. Interest-
blood vessels that comprise the blood-brain ingly, CSF production was significantly reduced in
barrier. Highly polarized AQP4 expression is also AQP1-null mice, but only by 20–25%, indicating a
found in the dense astrocyte cell processes that substantial contribution of extrachoroidal fluid
form the glia limitans, a structure that is adjacent production by the brain parenchyma. This result
to the CSF-filled subarachnoid space. The expres- supported previous studies in which removal of
sion of AQP1 and AQP4 at these tissue–fluid in- the choroid plexus in rhesus monkeys (Milhorat
terfaces indicate a potential role in maintaining et al., 1971) and dogs (Bering and Sato, 1963)
brain water homeostasis by facilitating water flux resulted in only a 30–50% decrease in the CSF
between these compartments. Furthermore, their production.
187

BRAIN SURFACE
Dura
Arachnoid Sinus Arachnoid granulation
2
Pia 1
AQP1
AQP4 Ependyma

Endothelial Astrocyte Choroid


cell foot processes plexus

EXTRACELLULAR
VENTRICLE
SPACE

PARENCHYMA P
V

Fig. 1. Aquaporins and brain water balance. The production and circulation of cerebrospinal fluid (CSF) in the brain is facilitated by
the highly polarized expression of aquaporin-1 (AQP1) and aquaporin-4 (AQP4). (1) Water transport from the vasculature into the
ventricle is facilitated by AQP1 expressed in the apical membrane of the choroid plexus, and via AQP4 in the ependymal lining.
(2) Another potential pathway for water flux into the subarachnoid space via the astrocytic processes lining the pial membrane that
heavily express AQP4. Fluid from the subarachnoid space is drained through the arachnoid granulations into the low-pressure venous
sinus that exits the cranium. (3) Extracellular fluid may alternatively pass through the perivascular processes of astrocytes as another
pathway to drain fluid into the vascular compartment.

The rates of CSF production and absorption These phenotypic studies of mice lacking AQP1
must be equal in the steady-state. Hydrocephalus and AQP4 provide evidence for the role of these
and increased ICP can occur when the balance water channels in the physiology of brain water
between CSF production and absorption is dis- homeostasis under normal and abnormal condi-
turbed. A reversal of CSF flow can be seen in ex- tions (Oshio et al., 2003; Manley et al., 2004). A
perimental models of obstructive hydrocephalus, key feature in a number of neurological disorders
where outflow-obstruction drives CSF into the is an imbalance in brain water homeostasis that
brain parenchyma causing extracellular edema leads to cerebral edema (Fishman, 1975). The re-
(Hiratsuka et al., 1982; Braun et al., 1997). Given mainder of the review focuses on the compensa-
the widespread expression of AQP4 at the tory and maladaptive mechanisms linked to these
brain–CSF interface, we examined whether this water channels and the discussion of their poten-
water channel might facilitate the transparenchy- tial as targets for therapeutic interventions in brain
mal absorption of CSF when the normal ventricu- edema.
lar outflow pathways are blocked (Bloch et al.,
2006). Using a mouse model of obstructive
hyprocephalus, we found that AQP4-null mice Cerebral edema
had an accelerated course of ventricular enlarge-
ment and ICP elevation compared with wildtype Cerebral edema is defined as an abnormal increase
mice, indicating a role for AQP4 in CSF absorp- in brain water content. Klatzo (1994) described
tion. We hypothesized that AQP4 most likely par- two distinct forms of cerebral edema based their
ticipates in clearance of excess parenchymal fluid pathogenesis (Fig. 2): vasogenic (extracellular) and
seen in this model. cytotoxic (intracellular) brain edema. In most
188

A Cytotoxic B Vasogenic

Swollen foot processes Astrocyte


Capillary foot
lumen process

TJ

Endothelial
cell
Capillary Endothelial cell
lumen

Fig. 2. Principles of cytotoxic and vasogenic edema. (A) The pathologic swelling of astrocytic foot processes is the main contributor to
cytotoxic edema. The tight junctions (TJ) help to establish the blood-brain barrier (BBB). (B) The disruption of the BBB allows
extravasation of the fluid from the brain capillary into the extracelullar space resulting in vasogenic edema.

human disease, both forms of edema are found, Aquaporins and cytotoxic edema
however one type typically dominates depending
on the particular disorder. However, these distinct Although water intoxication produces a pure cel-
edema types can be purely created in experimental lular edema, in the brain the swelling is predom-
models allowing independent study of the molec- inantly localized to the glial processes around the
ular mechanisms involved in their evolution. capillaries with sparing of the neurons (Wasterlain
Vasogenic (extracellular) edema results from ex- and Torack, 1968; Kimelberg, 1995). Characteris-
travasation of fluid secondary to defects in the tic swelling of astrocytic foot processes is also
blood-brain barrier, and is seen in experimental found in brain tissue from head injured patients
models of CNS inflammation (infection), tumors (Bullock et al., 1991). The selective swelling of
(Davies, 2002), and cold injury (Chan et al., 1991; astrocytes following a systemic perturbation in
Oury et al., 1993). Another mechanism of extra- osmolality raised questions about the mechanism
cellular edema is the pressure driven accumulation of this phenomenon (Kimelberg, 1995). Localiza-
of CSF in the brain interstitium of the periven- tion studies of AQP4 suggested that it might play a
tricular parenchyma as previously described for role in glial-specific swelling. Using the water in-
obstructive hydrocephalus. In contrast, cytotoxic toxication model, our group demonstrated a sig-
edema is characterized by acute cellular swelling in nificant reduction in astrocytic foot process
the presence of an intact blood-brain barrier. A swelling in AQP4-null mice (Manley et al., 2000).
classic example of this edema type is modeled with This was associated with a decrease in brain water
water intoxication. In this model, rapid intraperi- content and a profound improvement in survival
toneal water infusion causes serum hyponatremia (Fig. 3). Using a clinically relevant model of is-
that creates an osmotic gradient driving water chemic stroke, with its predominantly cytotoxic
entry into the brain, resulting in pure cytotoxic edema, the AQP4-null mice also had decreased
edema. Given the highly polarized expression of cerebral edema and improved outcome.
AQP4 at the blood-brain barrier, and the bi- Similar studies were conducted with the dystro-
directional nature of water flux through this chan- phin null mdx-bgeo transgenic mouse and the
nel, it is not surprising that AQP4 participates in a-syntrophin null mouse (Frigeri et al., 2001; Vajda
the molecular mechanisms of both types of edema. et al., 2002; Amiry-Moghaddam et al., 2003).
189

A B
100
AQP4+/+
0 min
80 AQP4−/− AQP4−/−
Survival (%)

60
15 min
40

20 AQP4+/+
30 min
0
0 20 40 60 80 100 1440
1.045 1.047 1.049 1.051 1.053
Time (min)
Specific gravity (g/cm3)

C D E
ICP cmH2O at 60min % Infused Non-Infused Water content
water (µL)
80
82 30

60

81
20

40

80
10
20

79
0 0
+/+ −/− +/+ −/−

Fig. 3. AQP4 deletion in models of cytotoxic (A, B) and vasogenic edema (C, D, E). AQP4 deletion improves short-term survival
following water intoxication produced by intraperitoneal fluid injection (A). Specific gravity of brain tissue display substantially higher
values for AQP4/ mice 30 min after water intoxication (B), indicating reduced brain water content compared with AQP4+/+
littermates. The clearance of edema fluid is impaired in AQP4 null mice following intraparenchimal fluid injection. Higher ICP (C) and
brain water content (D) is seen in AQP4-null mice compared with AQP4 controls. Brain water content of AQP4-null mice was also
higher than the wildtype controls following brain abscess implantation, a clinically relevant model of vasogenic edema.

These mice have normal levels of AQP4 protein, To further clarify the role of AQP4 in astrocyte
but because membrane localization of AQP4 re- swelling, osmotically induced volume changes
quires dystrophin-associated protein complex and were assessed in primary astrocyte cultures de-
a-syntrophin (Frigeri et al., 2001; Neely et al., rived from AQP4-null mice (Solenov et al., 2004).
2001), AQP4 expression in the astrocytic foot The measurement of rapid changes in cell volume
processes is reduced (Vajda et al., 2002; Amiry- was possible by adapting and validating a method
Moghaddam et al., 2003). As a result, these mice of calcein quenching (Hamann et al., 2002) to
are regarded as alternative models for the AQP4- measure water permeability in astrocytes. We
null genotype. In general, these studies have con- found that water permeability (Pf) was decreased
firmed and extended our observations regarding by sevenfold in the AQP4-null astrocytes com-
the role of AQP4 in cytotoxic edema (Vajda et al., pared with wildtype astrocytes. Using small inter-
2002; Amiry-Moghaddam et al., 2003). fering RNA (siRNA) targeted to reduce the level
190

of AQP4 expression, a 50% reduction in apparent diffuse passively from the vascular bed into the
water permeability of wildtype astrocytes was brain parenchyma, while all remaining molecules
found (Nicchia et al., 2003), which also caused a are trafficked via active transport across the blood-
68% inhibition of cell growth. In contrast, there brain barrier (Rowland et al., 1992).
was no alteration in cell morphology or growth Surrounding the endothelia are the astrocytic
characteristics in the primary cultures of the foot processes, or ‘‘end-feet’’ (Janzer and Raff,
AQP4-null astrocytes. 1987). These highly specialized processes have
Astrocytes constitute the largest pool of cells in multiple ion transporters and channels that are
the human brain, outnumbering neurons by at specifically targeted to the foot processes indicat-
least 10 to 1. Their numbers and ability to rapidly ing that they also play a key role in blood-brain
swell in response to an osmotic gradient highlights barrier function. Membranes of astrocyte foot
their importance in the pathogenesis of cellular processes also contain numerous arrays of intra-
edema. Studies of astrocytes in which AQP4 has membrane particles in freeze-fracture electron
been deleted or knocked down, confirm that AQP4 micrographs (FFEM). These particles, which are
is the principal water channel in these cells. From a regular square arrays with a characteristic cobble-
clinical perspective, the phenotypic studies of stone pattern, are referred to as square arrays or
AQP4-null mice have clearly demonstrated that orthogonal arrays of particles (OAPs) (Landis and
deletion or reduction of AQP4 reduces edema and Reese, 1974). Based on the finding that AQP4 was
improves neurological outcome in well-established present in the same cell types in which OAPs were
model where cytotoxic edema is in primary patho- identified, it was proposed that AQP4 is an OAP
physiological mechanism. Together these studies protein (Frigeri et al., 1995). Support for this
indicate that AQP4 inhibition may be a promising hypothesis came from FFEM studies of brain
target for drug discovery for the treatment of from AQP4-null mice that were found not to have
cytotoxic cerebral edema. OAPs (Verbavatz et al., 1997). Subsequent anti-
AQP4 antibody labeling of OAPs in brain tissue
confirmed that OAPs contain AQP4 (Rash et al.,
Aquaporins and vasogenic edema 1998). The polarized expression and co-localiza-
tion of ion and water channels in the astrocyte foot
The key feature of vasogenic edema is the disrup- processes suggest that these structures may facil-
tion of the blood-brain barrier and subsequent itate ion and water transport across the blood-
leakage of the intravascular fluid into the extra- brain barrier.
cellular space of the brain parenchyma. The blood- Given that the formation of vasogenic edema is
brain barrier is essential for the normal function of primarily due to the disruption of the blood-brain
the brain. It provides a highly selective barrier be- barrier and subsequent leakage of the intravascu-
tween the vascular space and the brain tissue that lar fluid into the extracellular space, a role for as-
allows for maintenance of the microenvironment trocyte water channels in the formation of this
that is crucial for overall homeostasis of the brain. type of edema was not anticipated. However, re-
The anatomical substrates of the blood-brain bar- sults from multiple independent experiments sug-
rier are the capillary endothelium and the astocytic gest a definite role for AQP4 in the clearance of
foot processes that surround the cerebral capillar- extracellular fluid, and the resolution of vasogenic
ies (Reese and Karnovsky, 1967). edema.
Unlike the microvasculature in the rest of the As with the CSF, the amount of interstitial fluid
body, the cerebral capillaries are not fenestrated present in extracellular space is determined by the
and contain few endocytic vesicles (Sage et al., balance of fluid production and clearance from the
1998) suggesting limited diffusion and transcellu- brain parenchyma. Similarly, when the clearance
lar transport. Molecular tracking studies show or absorption is impaired, fluid accumulates and
that only small lipophilic molecules are allowed to ICP increases. To directly examine the role of
191

AQP4 in clearance of extracellular fluid, artificial been shown to be upregulated (Vizuete et al.,
CSF was continuously infused into the brain 1999); perhaps as an adaptive response to edema.
parenchyma of wildtype and AQP4-null mice From a clinical perspective, the phenotypic studies
(Fig. 3). In comparison with the wildtype mice, of AQP4-null mice indicate that deletion of AQP4
AQP4-null mice had significantly more brain water reduces the clearance of extracellular fluid and
and elevated ICP (Papadopoulos et al., 2004). Al- improves outcome in models where vasogenic
though the exact mechanism is yet to be defined, edema is the primary pathophysiological mecha-
these results demonstrate that AQP4 is involved in nism. Thus, AQP4 inhibition would not be indi-
the clearance of extracellular fluid from the brain cated for the treatment of vasogenic edema.
parenchyma. Whether or not strategies aimed at increasing
Our group has used a number of models where AQP4 expression or enhancing water permeability
vasogenic edema is the predominant form of ed- will improve outcome from vasogenic edema, re-
ema, including cortical freeze injury, tumor im- main to be investigated.
plantation, and brain abscess. Cortical freeze
injury lead to a marked disruption of the blood-
brain barrier in both wildtype and AQP4-null Aquaporins, ICP, and traumatic brain injury
mice, however AQP4-null mice had a significantly
greater increase in brain water content and ICP One of the more intriguing and clinically relevant
(Papadopoulos et al., 2004). Peritumoral vaso- findings has been the observation that ICP is sig-
genic edema was generated by implanting me- nificantly reduced in AQP1-null mice (Oshio et al.,
lanoma cells into the striatum of AQP4-null and 2005). ICP is the dynamic result of the interaction
wildtype mice. In accordance with the results from of the CSF, cerebral blood, and brain tissue com-
previous models, there was higher ICP and accel- partments. The individual effect of each compart-
erated neurological deterioration in the AQP4 de- ment on ICP is difficult to assess due to the
ficient mice (Papadopoulos et al., 2004). A similar complexity of the physiologic relationship between
result was obtained with implantation of Staphylo- these compartments. However, if the intracranial
coccus aureus to create brain abscesses in the stria- compliance is assumed to be constant, the
tum of mice. In addition to increased peri-abscess steady-state ICP can be expressed using the fol-
edema and increased ICP, AQP4 deficient mice lowing equation introduced by Marmarou:
demonstrated a greater mortality despite equiva- ICP ¼ If  Rout+Pss, where If is CSF formation
lent abscess size and cytokine levels (Bloch et al., rate, Rout is outflow resistance, and Pss is sagittal
2005). In all of the mentioned experiments, the sinus pressure (Marmarou et al., 1978). As previ-
primary lesion size and degree of BBB permeabil- ously described, AQP1-null mice have a reduced
ity were similar, indicating that the forces driving CSF formation rate that accounts for some of the
the development of vasogenic edema were equiv- 50% reduction in ICP seen in these mice. The de-
alent in both groups and that differences were due letion of AQP1 also leads to polyuria and hypo-
to impaired fluid clearance from the extracellular volemia due to its role in fluid absorption in the
space in AQP4-null mice. kidney (Schnermann et al., 1998). We suspected
Fluid reabsorption from the brain extracellular that this urinary concentrating defect might also
space is thought to occur by bulk flow clearance decreases the central venous pressure, which in
into the CSF and by transport back into cerebral turn, is in continuity with the sagittal sinus venous
capillaries (Reulen et al., 1978; Marmarou et al., pressure. This was indeed the case, indicating that
1994). Although a clear understanding of brain the effects of AQP1 deletion on venous pressure
water homeostasis remains to be fully elucidated, accounted for the balance of the reduction in ICP.
several lines of evidence point towards an impor- These results demonstrate that two different
tant role for AQP4. In pathological states that in- AQP1-dependent mechanisms act synergistically
crease extracellular fluid, AQP4 expression has to significantly decrease in ICP.
192

Fig. 4. The beneficial effect of AQP1 deletion in brain injury. (A) The absence of AQP1 improves survival in focal brain injury. (B) The
ICP peak is reduced in AQP1-null mice following cold injury compared with wildtype controls.

ICP management has been the cornerstone of conditions and suggest a novel application of
traumatic brain injury care for many years. It is AQP1 inhibitors for pharmacological treatment of
well known that when ICP is elevated and cannot elevated ICP.
be reduced, nearly all patients with traumatic
brain injury will die (Miller et al., 1981). All cur-
rent surgical and non-surgical treatments of ele- Conclusion
vated ICP are directed towards decreasing the
volume in one of the three major compartments — Phenotypic analyses of AQP deficient mice have
brain, blood, or CSF (Brain Trauma Foundation, allowed us to explore the role of these membrane
American Association of Neurological Surgeons water channels in the mechanisms of cytotoxic ed-
and Joint Section on Neurotrauma and Critical ema, vasogenic edema, and CSF production.
Care, 1996). The ability of AQP1 deletion to re- These studies indicate that AQP4 plays significant
duce two of the three principal determinants of role in the development of cytotoxic edema and
steady-state ICP, CSF production, and venous the absorption of excess brain water resulting from
pressure, suggested that AQP1deletion might be vasogenic edema. They also have demonstrated
protective in a model of brain trauma (Fig. 4). In a the role of AQP1 in CSF production and mainte-
focal model of brain injury, the peak ICP was 60% nance of steady-state ICP. The ability to modulate
lower in AQP1-null mice compared with wildtype water flux through AQP deletion has provided new
littermates. In addition, a dramatic improvement insights into brain water homeostasis and sug-
in survival rate was seen in the AQP1 deficient gested a number of new research directions. How-
mice: 84% compared with the 25% seen in wild ever, these efforts have not yet translated to the
type controls. Currently, there is no consensus on treatment human clinical diseases. These advances
the treatment of elevated ICP. These results pro- will require the development of AQP inhibitors
vide evidence for the importance of CVP and CSF and activators to establish the benefit modulating
production on ICP under normal and pathological the function of these water channels.
193

References Janzer, R.C. and Raff, M.C. (1987) Astrocytes induce blood-
brain barrier properties in endothelial cells. Nature,
Abbott, N.J. (2004) Evidence for bulk flow of brain interstitial 325(6101): 253–257.
fluid: significance for physiology and pathology. Neurochem. Kimelberg, H.K. (1995) Current concepts of brain edema. Re-
Int., 45(4): 545–552. view of laboratory investigations. J. Neurosurg., 83(6):
Agre, P., King, L.S., et al. (2002) Aquaporin water channels: 1051–1059.
from atomic structure to clinical medicine. J. Physiol., 542 Klatzo, I. (1994) Evolution of brain edema concepts. Acta
(Pt 1): 3–16. Neurochir. Suppl. (Wien.), 60: 3–6.
Amiry-Moghaddam, M., Otsuka, T., et al. (2003) An alpha- Landis, D.M. and Reese, T.S. (1974) Arrays of particles in
syntrophin-dependent pool of AQP4 in astroglial end-feet freeze-fractured astrocytic membranes. J. Cell Biol., 60(1):
confers bidirectional water flow between blood and brain. 316–320.
Proc. Natl. Acad. Sci. U.S.A., 100(4): 2106–2111. Manley, G.T., Binder, D.K., et al. (2004) New insights into
Bering, E. and Sato, O. (1963) Hydrocephalus: changes in for- water transport and edema in the central nervous system
mation and absorption of cerebrospinal fluid within the cere- from phenotype analysis of aquaporin-4 null mice. Neuro-
bral ventricles. J. Neurosurg., 20: 1050. science, 129(4): 983–991.
Bloch, O., Auguste, K.I., Manley, G.T. and Verkman, A.S. Manley, G.T., Fujimura, M., et al. (2000) Aquaporin-4 deletion
(2006) Accelerated progression of kaolin-induced hydroceph- in mice reduces brain edema after acute water intoxication
alus in aquaporin-4-deficient mice. J. Cereb. Blood Flow and ischemic stroke. Nat. Med., 6(2): 159–163.
Metab., 26(12): 1527–1537. Marmarou, A., Hochwald, G., et al. (1994) Brain edema reso-
Bloch, O., Papadopoulos, M.C., et al. (2005) Aquaporin-4 gene lution by CSF pathways and brain vasculature in cats. Am. J.
deletion in mice increases focal edema associated with Physiol., 267(2 Pt 2): H514–H520.
staphylococcal brain abscess. J. Neurochem., 95(1): 254–262. Marmarou, A., Shulman, K., et al. (1978) A nonlinear analysis
Brain Trauma Foundation, American Association of Neuro- of the cerebrospinal fluid system and intracranial pressure
logical Surgeons, Joint Section on Neurotrauma and Critical dynamics. J. Neurosurg., 48(3): 332–344.
Care. (1996) Guidelines for the management of severe head McIlwain, H. and Bachelard, H.S. (1985) Biochemistry and the
injury. J. Neurotrauma, 13(11): 641–734. Central Nervous System. Churchill Livingstone, Edinburgh.
Braun, K.P., Dijkhuizen, R.M., et al. (1997) Cerebral ischemia Milhorat, T., Hammock, M., et al. (1971) Cerebrospinal fluid
and white matter edema in experimental hydrocephalus: a production by the choroid plexus and brain. Science,
combined in vivo MRI and MRS study. Brain Res., 757(2): 173(994): 330–332.
295–298. Miller, J.D., Butterworth, J.F., et al. (1981) Further experience
Bullock, R., Maxwell, W.L., et al. (1991) Glial swelling follow- in the management of severe head injury. J. Neurosurg.,
ing human cerebral contusion: an ultrastructural study. J. 54(3): 289–299.
Neurol. Neurosurg. Psychiatry, 54(5): 427–434. Neely, J.D., Amiry-Moghaddam, M., et al. (2001) Syntrophin-
Chan, P.H., Yang, G.Y., et al. (1991) Cold-induced brain ed- dependent expression and localization of Aquaporin-4 water
ema and infarction are reduced in transgenic mice overex- channel protein. Proc. Natl. Acad. Sci. U.S.A., 98(24):
pressing CuZn-superoxide dismutase. Ann. Neurol., 29(5): 14108–14113.
482–486. Nicchia, G.P., Frigeri, A., et al. (2003) Inhibition of aquaporin-
Davies, D.C. (2002) Blood-brain barrier breakdown in septic 4 expression in astrocytes by RNAi determines alteration in
encephalopathy and brain tumours. J. Anat., 200(6): 639–646. cell morphology, growth, and water transport and induces
Fishman, R.A. (1975) Brain edema. N. Engl. J. Med., 293(14): changes in ischemia-related genes. FASEB J., 17(11):
706–711. 1508–1510.
Frigeri, A., Gropper, M.A., et al. (1995) Localization of MIWC Oshio, K., Song, Y., et al. (2003) Aquaporin-1 deletion reduces
and GLIP water channel homologs in neuromuscular, epi- osmotic water permeability and cerebrospinal fluid produc-
thelial and glandular tissues. J. Cell Sci., 108(Pt 9): tion. Acta Neurochir. Suppl., 86: 525–528.
2993–3002. Oshio, K., Watanabe, H., et al. (2005) Reduced cerebrospinal
Frigeri, A., Nicchia, G.P., et al. (2001) Aquaporin-4 deficiency fluid production and intracranial pressure in mice lacking
in skeletal muscle and brain of dystrophic mdx mice. FASEB choroid plexus water channel Aquaporin-1. FASEB J., 19(1):
J., 15(1): 90–98. 76–78.
Hamann, S., Kiilgaard, J.F., et al. (2002) Measurement of cell Oury, T.D., Piantadosi, C.A., et al. (1993) Cold-induced brain
volume changes by fluorescence self-quenching. J. Fluoresc., edema in mice. Involvement of extracellular superoxide dis-
12: 139–145. mutase and nitric oxide. J. Biol. Chem., 268(21):
Hasegawa, H., Ma, T., et al. (1994) Molecular cloning of a 15394–15398.
mercurial-insensitive water channel expressed in selected Papadopoulos, M.C., Manley, G.T., et al. (2004) Aquaporin-4
water-transporting tissues. J. Biol. Chem., 269(8): 5497–5500. facilitates reabsorption of excess fluid in vasogenic brain ed-
Hiratsuka, H., Tabata, H., et al. (1982) Evaluation of periven- ema. FASEB J., 18(11): 1291–1293.
tricular hypodensity in experimental hydrocephalus by metriz- Rash, J.E., Yasumura, T., et al. (1998) Direct immunogold
amide CT ventriculography. J. Neurosurg., 56(2): 235–240. labeling of aquaporin-4 in square arrays of astrocyte and
194

ependymocyte plasma membranes in rat brain and spinal Vajda, Z., Pedersen, M., et al. (2002) Delayed onset of brain
cord. Proc. Natl. Acad. Sci. U.S.A., 95(20): 11981–11986. edema and mislocalization of aquaporin-4 in dystrophin-null
Reese, T.S. and Karnovsky, M.J. (1967) Fine structural local- transgenic mice. Proc. Natl. Acad. Sci. U.S.A., 99(20):
ization of a blood-brain barrier to exogenous peroxidase. J. 13131–13136.
Cell Biol., 34(1): 207–217. Verbavatz, J.M., Ma, T., et al. (1997) Absence of orthogonal
Reulen, H.J., Tsuyumu, M., et al. (1978) Clearance of edema arrays in kidney, brain and muscle from transgenic knockout
fluid into cerebrospinal fluid. A mechanism for resolution of mice lacking water channel aquaporin-4. J. Cell Sci., 110(Pt
vasogenic brain edema. J. Neurosurg., 48(5): 754–764. 22): 2855–2860.
Rowland, L.P., Fink, M.E., et al. (1992) Cerebrospinal fluid: Verkman, A.S. (2002) Physiological importance of aquaporin
blood-brain barrier, brain oedema and hydrocephalus. In: water channels. Ann. Med., 34(3): 192–200.
Kandel E.R., Schwartz J.H. and Jessel T.M. (Eds.), Princi- Vizuete, M.L., Venero, J.L., et al. (1999) Differential upregu-
ples of Neuroscience. Elsevier, New York, pp. 1050–1060. lation of aquaporin-4 mRNA expression in reactive as-
Sage, M.R., Wilson, A.J., et al. (1998) Contrast media and trocytes after brain injury: potential role in brain edema.
the brain. The basis of CT and MR imaging enhancement. Neurobiol. Dis., 6(4): 245–258.
Neuroimaging Clin. N. Am., 8(3): 695–707. Wasterlain, C.G. and Torack, R.M. (1968) Cerebral edema
Schnermann, J., Chou, C.-L., et al. (1998) Defective proximal in water intoxication. II. An ultrastructural study. Arch.
tubular fluid reabsorption in transgenic aquaporin-1 null Neurol., 19(1): 79–87.
mice. PNAS, 95(16): 9660–9664. Weed, L.H. and McKibben, P.S. (1919) Experimental alteration
Solenov, E., Watanabe, H., et al. (2004) Sevenfold-reduced os- of brain bulk. Am. J. Physiol., 48: 531–558.
motic water permeability in primary astrocyte cultures from Welch, K. (1963) Secretion of cerebrospinal fluid by choroid
AQP-4-deficient mice, measured by a fluorescence quenching plexus of the rabbit. Am. J. Physiol., 205(3): 617–624.
method. Am. J. Physiol. Cell Physiol., 286(2): C426–C432. Wright, E.M., Wiedner, G., et al. (1977) Fluid secretion by the
Speake, T., Freeman, L.J., et al. (2003) Expression of aquaporin frog choroid plexus. Exp. Eye Res., 25(Suppl): 149–155.
1 and aquaporin 4 water channels in rat choroid plexus.
Biochim. Biophys. Acta, 1609(1): 80–86.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 13

Sodium channel expression and the molecular


pathophysiology of pain after SCI

Bryan C. Hains and Stephen G. Waxman

Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine,
New Haven, CT 06510, USA; Rehabilitation Research Center, VA Connecticut Healthcare System, West Haven,
CT 06516, USA

Abstract: The chronic pain that develops as a result of spinal cord injury (SCI) is extremely debilitating and
remains largely unmanageable by current therapeutic strategies. Voltage-gated sodium channels regulate
the biophysical properties, and thus firing characteristics, of neurons. After SCI the repertoire of sodium
channels produced by dorsal horn nociceptive neurons is altered, enabling neurons to fire at higher than
normal rates in response to unchanged peripheral stimuli as well as to generate spontaneous discharges in
the absence of stimuli, resulting in the genesis of neuropathic pain. Our results have shown increased
expression of the Nav1.3 sodium channel in the spinal cord and thalamus. Nav1.3 upregulation allows
dorsal horn neurons to generate ramp currents, enhanced persistent currents, and shifts in steady-state
activation and inactivation. Further downstream, Nav1.3 causes increased spontaneous and evoked firing
of neurons in the ventroposterior lateral (VPL) nucleus of the thalamus. Nav1.3 also underlies changes in
burst firing properties of VPL neurons. The combination of spinal and thalamic generation and ampli-
fication of pain by Nav1.3 dysregulation contributes to post-SCI chronic pain. If proven to be similar in
humans, targeting of this system after SCI may offer hope for treatment of clinical pain.

Keywords: spinal cord injury; pain; sodium channels; thalamus; dorsal horn

Introduction Turner et al., 2001), even to a greater extent than


the motor impairment (Mariano, 1992; Stormer
Although less apparent than paralysis, the preva- et al., 1997). Of patients with low thoracic or
lence of chronic pain disability after spinal cord lumbrosacral lesions, 37% are willing to trade
injury (SCI) is widespread. Sixty to eighty percent the possibility of recovery for pain relief
of persons who have sustained SCI, regardless of (Nepomuceno et al., 1979).
the level or completeness of lesion or of the type of Nociceptive signals are transmitted through the
injury, experience clinically significant pain after dorsal horn of the spinal cord. The spinal cord
injury (Finnerup et al., 2001; Siddall et al., 2003). dorsal horn contains the primary synapses through
The pain, which can have a pricking, burning, or which afferent somatosensory information, related
aching quality (Cairns et al., 1996), can be so se- to touch, pressure, brush, temperature, and noxi-
vere as to produce a drastic impairment in daily ous stimuli, is received from the periphery. Dorsal
routines and quality of life (Rintala et al., 1998; horn sensory neurons receive this information pri-
marily from the skin, perform a degree of process-
Corresponding author. Tel.: +203-785-6351; Fax: +203-785- ing, and transmit signals through distinct tracts
7826; E-mail: stephen.waxman@yale.edu
within the spinal cord to supraspinal structures

DOI: 10.1016/S0079-6123(06)61013-3 195


196

where pain is interpreted and perceived. Within Ten genes encode molecularly distinct voltage-
each level of the nervous system including the spi- gated sodium channels, at least seven of which are
nal cord, nociceptive signals are subject to a degree expressed in the rat nervous system. In the adult
of modulation by circuitry that it passes through. spinal cord dorsal horn, Nav1.1, Nav1.2, and
Experimental SCI induces electrophysiological Nav1.6 are strongly detectable, whereas Nav1.3
changes in dorsal horn and thalamic sensory expression, while present at early ontogenetic
neurons that contribute to pain-like behaviors states, decreases with development and is nearly
in animals. These include shifts in proportions of undetectable in adults (Felts et al., 1997).
cells responding to evoked noxious stimulation,
increases and irregularity in spontaneous back-
ground activity, increased evoked activity to (for- SCI and dorsal horn ion channel dysregulation
merly) innocuous and noxious stimuli, increases
in after discharge activity following stimulation, We have studied sodium channel expression at
and emergence of abnormal burst firing. several levels along the pain-signaling pathway in a
Since the nature of the applied stimuli does not model of contusive SCI in which, as in humans
change, other mechanisms must account for the with non-penetrating SCI, there is central necrosis,
alterations in stimulus processing that lead to cen- surrounded by a rim of surviving ascending and
tral pain after SCI. Central changes in expression descending axons at the level of the injury (Hains
of molecules such as ion channels, neurotransmit- et al., 2004). In this rodent model in situ hybrid-
ters, and their receptors, contribute to altered sen- ization and immunocytochemistry with an iso-
sory processing by changing the electrophysiologic form-specific Nav1.3 antibody show that
excitability, and therefore output, of spinal sen- expression of Nav1.3 is upregulated within dorsal
sory neurons. Specific molecular changes in the horn neurons caudal to the level of the injury
expression of voltage-gated sodium channels have (Hains et al., 2003). These studies show that, 4
recently been shown by our laboratory to contrib- weeks after SCI, expression of Nav1.3 is increased,
ute to the development and maintenance of central within neurons contained within laminae I–VI in
pain following SCI. the lumbar dorsal horn (L3–L5) (Fig. 1B). Nav1.3
does not co-localize with OX-42, a marker for ac-
tivated microglia (which proliferate after SCI and
are present in all laminae within the dorsal horn
Spinal cord sodium channels (Hains and Waxman, 2006)), or with GFAP, a
marker for astrocytes (which also proliferate in all
Action potential generation and propagation by spinal laminae after SCI). CGRP, restricted to the
sensory neurons relies on multiple isoforms of terminals of primary afferent fibers within laminae
voltage-gated sodium channels (termed Nav). The I–II, does not co-localize with Nav1.3. The sub-
selective expression of ensembles of sodium chan- stance-P receptor NK1R, found in second-order
nels, unique in different types of neurons, tunes the nociceptive neurons within laminae I–IV, does
biophysical properties of each cell. Within the however co-localize with Nav1.3, indicating that
normal nervous system, properties fundamental to Nav1.3 is upregulated within nociceptive dorsal
neuronal function such as activation threshold, horn neurons.
inactivation, refractory period, rates of repriming, In this model, 88% of sampled lumbar dorsal
and the ability to generate and conduct high- horn units show increased evoked activity to nat-
frequency trains of action potentials all depend on ural peripheral stimuli (brush, press, pinch, graded
the type(s) of sodium channels expressed within a von Frey filaments, and 471C thermal stimuli). In
given neuron (Waxman, 2000). As might be ex- comparison to intact animals in which dorsal horn
pected, dysregulation of channel expression can neurons fire at rates of 5–21 Hz, SCI animals
abnormally reconfigure neuronal function in dis- demonstrate increased evoked rates of up to
ease states. 55–60 Hz (Fig. 1E).
197

Fig. 1. Nav1.3 mRNA is expressed at low levels in naı̈ve animals (A), but is upregulated in lumbar dorsal horn neurons after spinal cord
injury (SCI) (B). Treatment with antisense (AS) oligodeoxynucleotides generated against Nav1.3 reduces Nav1.3 transcripts after SCI (C).
Corresponding unit recordings show evoked activity to peripheral stimulation (BR, brush; PR, press; PI, pinch, increasing strength von
Frey filament stimulation, and noxious thermal heating (471C)), after SCI (E) compared with controls (D). After Nav1.3 AS delivery,
evoked activity of dorsal horn neurons resembles that found in naı̈ve levels (F). Adapted with permission from Hains et al. (2003).

The Nav1.3 sodium channel plays a role in decreases the evoked hyperresponsiveness of
maintaining neuronal hyperresponsiveness to dorsal horn multi-receptive neurons (Fig. 1F),
peripheral stimulation, as well as pain-related whereas administration of Nav1.3 mismatch
behaviors after SCI, as evidenced through selec- (MM) sequence, used as a control, has no effect.
tive knock-down of Nav1.3 expression via anti- Electrophysiological recordings show that 4 days
sense (AS) oligodeoxynucleotide administration after initiation of Nav1.3 AS, high levels of
(Hains et al., 2003). Lumbar intrathecal admin- evoked activity of dorsal horn neurons in SCI
istration of Nav1.3 AS very effectively reduces animals are markedly decreased in response to all
the expression of Nav1.3 within dorsal horn peripheral stimuli, and only 20% of sampled units
neurons after SCI (Fig. 1C), and significantly are hyperexcitable.
198

Nav1.3 and neuronal hyperresponsiveness We recently examined the biophysical properties


of sodium currents in acutely dissociated dorsal
Expression of the Nav1.3 sodium channel after horn neurons after chronic SCI, and found a
SCI uniquely configures neurons to fire in an ab- number of changes in sodium current characteris-
normal, heightened manner. Nav1.3 produces a tics via whole-cell patch-clamp recordings (Fig. 2A)
rapidly repriming tetrodotoxin-sensitive sodium (Lampert et al., 2006). First, we found shifts in the
current that promotes neuronal firing at higher- activation and inactivation properties of the volt-
than-normal frequencies (Cummins et al., 2001). age-gated sodium currents to more positive poten-
Since stimulus intensity is encoded in the dorsal tials after SCI (Fig. 2B). Second, persistent sodium
horn by the rate of firing, this change in neuronal currents in dorsal horn neurons from SCI animals
processing of afferent sensory information serves were increased when compared with cells from in-
to amplify incoming signals, such that perceived tact animals (Fig. 2C). Third, the ramp current
pain thresholds are lowered after SCI. elicited in response to slow depolarizing potentials

Fig. 2. Whole-cell patch-clamp recordings of acutely dissociated lumbar dorsal horn neurons 28 days after T9 SCI reveal changes in
sodium current properties. Representative current traces from intact and SCI animals, elicited by stepwise depolarizations from a
holding potential of 120 mV to voltages ranging form 110 to +40 mV (A). Steady-state inactivation (open squares: intact, filled
squares: SCI) measured as relative available current after a 500 ms prepulse to the indicated potential. Steady-state activation shown as
relative conductance (B). Note that SCI shifts steady-state inactivation in dorsal horn neurons toward the curve for axotomized DRG
neurons, in which Nav1.3 is upregulated. Representative current traces showing enhancement of persistent current after SCI (C).
Vertical lines delimit the region in which the mean persistent current was assessed. Inset: Representative record from a human
embryonic kidney cell heterologously transfected with Nav1.3, showing persistent current. Representative current traces in response to
a 200 ms voltage ramp from 120 to +30 mV showing an increase in ramp currents in SCI but not intact animals (D). Adapted with
permission from Lampert et al. (2006).
199

was increased after SCI (Fig. 2D). The shift in ventroposterior lateral (VPL) nucleus of the
steady-state inactivation and the increase in the thalamus (Jones, 1998). VPL neurons are involved
persistent and ramp current all would be expected in sensory-discriminative aspects of pain process-
to contribute to dorsal horn neuron hyperrespon- ing. Thalamic changes have been associated with
siveness, which is observed after SCI. All of these pain following SCI in humans (Lenz et al., 1989;
properties, which are consistent with upregulation Pattany et al., 2002), primates (Weng et al., 2000),
of Nav1.3, can contribute to hyperresponsiveness and rodent models (Koyama et al., 1993; Gerke
of dorsal horn neurons, after SCI. et al., 2003), but the underlying molecular mech-
anisms are still not fully understood.
After SCI, expression of the Nav1.3 sodium
Thalamic Nav1.3 dysregulation channel is upregulated in VPL neurons (Fig. 3B),
and an increase in the spontaneous activity and
Dorsal horn neurons project rostrally within the responses to natural stimuli is observed (Hains
spinothalamic tract to third-order neurons of the et al., 2005). Lumbar administration of Nav1.3 AS

Fig. 3. Expression of Nav1.3 protein within the ventral posterolateral (VPL) nucleus of the thalamus is absent in intact animals (A),
but 28 days after SCI Nav1.3 expression is upregulated within VPL and to some degree the ventral posteromedial (VPM) nuclei.
Intrathecal Nav1.3 antisense (AS) administration (C) significantly reduced the expression of Nav1.3 within the VPL (D). Long-term
recording of a single VPL unit after SCI with a hindpaw receptive field revealed high spontaneous discharge activity and evoked
responsiveness to brush (BR) stimulation of the receptive field (E). Following topical application of lidocaine (lido) and cord tran-
section (tx), evoked responses were abolished however high spontaneous activity persisted. Adapted with permission from Hains et al.
(2005).
200

reduced the number of neurons displaying Nav1.3 (Hains et al., 2006). Furthermore, we observed
upregulation within the VPL (Fig. 3C), and re- that Nav1.3 AS returned the number of spikes/
versed thalamic electrophysiologic abnormalities burst, burst duration, and interburst interval to-
caused by SCI. Importantly, we also observed an ward control levels following SCI. Our data are
increased level of spontaneous background activ- similar to those reported by Lenz et al. (1989,
ity in VPL neurons associated with SCI which 1994) in humans with post-SCI pain showing that
persists after spinal cord transection (Fig. 3E) that thalamic neurons exhibit oscillatory burst firing
disconnects the site of SCI and ascending projec- characterized by high discharge rates, and decel-
tions from the lumbar enlargement from the eration of firing rate throughout the burst period,
thalamus, indicating a degree of autonomous and suggest that altered sodium channel expres-
hyperexcitability of the thalamus. We cannot ex- sion within thalamic neurons contributes to these
clude the possibility that factors other than Nav1.3 functional abnormalities.
also contribute to the reconfiguration of the firing
properties of thalamic units following SCI since
the magnitudes of change of Nav1.3 expression Multi-tiered alterations in nociceptive processing
levels did not perfectly match the magnitude of the
electrophysiologic changes. However, our results Our findings demonstrate changes in excitability
demonstrate changes in sodium channel expression and expression of Nav1.3 within dorsal horn and
within the thalamus that are associated with VPL neurons following SCI. We have also ob-
abnormal sensory processing and chronic neuro- served that hyperexcitability of thalamic neurons
pathic pain after SCI. following SCI is to at least some degree autono-
The thalamus also undergoes a change in burst mous, since it persists following spinal cord tran-
firing properties after SCI. Bursts are involved in section which abolishes ascending barrages from
normal and pathological perceptual processing; spinal cord neurons near or below the injury site.
however, rhythmic oscillation of burst firing is ob- Together with our earlier results on dorsal root
served in pathophysiological conditions, and it has ganglion neurons (Cummins and Waxman, 1997;
been suggested that abnormal thalamic activity Cummins et al., 2001), these studies provide evi-
may contribute to the perception of chronic pain dence for a link between pain after SCI and mo-
(Lenz et al., 1989, 1994; Jeanmonod et al., 1993; lecular changes in pain-signaling neurons,
McCormick, 1999). Furthermore, the information suggesting that dysregulation of sodium channel
content of bursts is higher than for single-spikes in Nav1.3 expression at both spinal and supraspinal
the visual system (Reinagel et al., 1999), and the levels contribute to altered processing of somato-
overall probability of generating at least one sensory information and chronic neuropathic pain
postsynaptic spike (in the pain-processing sensory after SCI. Specifically, our results indicate that
cortex) is higher for bursts than for single-spikes there is upregulated expression of Nav1.3 in first-,
(Csicsvari et al., 1998). In neurons of the ventro- second-, and third-order neurons of the pain path-
basal thalamus, increased gain or transfer ratio way after contusive SCI, and suggest that this
induced by burst firing results in increased tha- leads to enhanced neuronal excitability at each of
lamocortical efficacy, enhancing the postsynaptic these multiple levels.
response (Swadlow and Gusev, 2001). Thus, it is As originally proposed by Yezierski (2001), SCI
possible that pathological burst firing after SCI can trigger the activity of pain generators and
may more potently activate cortical circuits in- amplifiers within the CNS. On the basis of our
volved in pain perception. observations, we propose an integrated model
We recently showed that after SCI, burst firing (Fig. 4) in which increased Nav1.3 expression
intervals become more regular, spike events are within pain-signaling neurons at multiple levels of
reduced within each burst, acceleration in burst the neuraxis causes exaggerated nociceptive signa-
duration occurs in bursts containing higher spike ling by way of spontaneous firing, a lowered
counts, and shifts occur among spike firing modes threshold for firing in response to synaptic drive,
201

Fig. 4. Model whereby after SCI, multi-tiered dysregulation of sodium channel expression contributes to the pathological ampli-
fication and generation of aberrant nociceptive information, and ultimately chronic pain. In comparison to intact animals (A), second-
order neurons within the dorsal horn increase the gain with which they respond to innocuous and noxious inputs (10 ) after SCI (B). The
exaggerated responses of these dorsal horn neurons in response to peripheral stimulation poise them to act as an amplifier of normal as
well as inappropriate pathological nociceptive signals (20 ). After SCI, the thalamus also acts as a pain amplifier by responding in an
increased manner to nociceptive information relayed from the dorsal horn, and in addition, thalamic circuitry can act as an au-
tonomous pain signal generator (30 ). Treatment strategies that target one or more components of this pathway may be of value in
combating chronic pain following SCI.

and increased synaptic drive due to enhanced ex- (Waxman and Hains, 2006). Abnormal processing
citability of presynaptic elements. According to at thalamic levels would then be expected to fur-
this model, second-order neurons within the dorsal ther exaggerate abnormal firing patterns from the
horn increase the gain with which they respond to spinal cord after SCI. Our observations of in-
innocuous and noxious inputs after SCI. The ex- creased primary burst firing activity and reduced
aggerated responses of these dorsal horn neurons silence in VPL neurons after SCI leads us to sug-
configures them to act as an amplifier of normal as gest that, following SCI, an increased level of
well as inappropriate pathological nociceptive sig- abnormal afferent firing is being forwarded to
nals. cortical structures involved in interpreting pain.
Because the thalamus is involved not only in
relaying, but also processing incoming information
from the spinal cord en route to the cortex, injury- Conclusion
induced changes in spinal pain generator circuitry
may feed aberrant signals into the injured thala- SCI induces nervous-system-wide changes in neu-
mus which further processes and amplifies the sig- ronal firing properties which contribute to chronic
nals before relaying messages that are interpreted pain, some of which are governed by the patho-
as signaling pain to suprathalamic structures logical expression of the Nav1.3 sodium channel.
202

By prevention of the upregulation of Nav1.3, Finnerup, N.B., Johannesen, I.L., Sindrup, S.H., Bach, F.W.
knock-down of Nav1.3 after it is upregulated, or and Jensen, T.S. (2001) Pain and dysesthesia in patients
with spinal cord injury: a postal survey. Spinal Cord, 39:
selective pharmacological targeting of Nav1.3
256–262.
channels deployed after injury, it is possible that Gerke, M.B., Duggan, A.W., Xu, L. and Siddall, P.J. (2003)
the molecular amplifiers and generators of chronic Thalamic neuronal activity in rats with mechanical allodynia
pain associated with SCI may be effectively tar- following contusive spinal cord injury. Neuroscience, 117:
geted and muted so that chronic pain can be 715–722.
ameliorated. Hains, B.C., Klein, J.P., Saab, C.Y., Craner, M.J., Black, J.A.
and Waxman, S.G. (2003) Upregulation of sodium channel
Nav1.3 and functional involvement in neuronal hyperexcit-
ability associated with central neuropathic pain after spinal
Acknowledgments cord injury. J. Neurosci., 23: 8881–8892.
Hains, B.C., Saab, C.Y., Lo, A.C. and Waxman, S.G. (2004)
The authors thank Dr. Joel Black for valuable Sodium channel blockade with phenytoin protects spinal
cord axons, enhances axonal conduction, and improves func-
experimental advice. This work was supported in
tional motor recovery after contusion SCI. Exp. Neurol., 188:
part by grants from the Medical Research Service 365–377.
and Rehabilitation Research Service, Department Hains, B.C., Saab, C.Y. and Waxman, S.G. (2005) Changes in
of Veterans Affairs, and the National Multiple electrophysiological properties and sodium channel Nav1.3
Sclerosis Society. The Center for Neuroscience and expression in thalamic neurons after spinal cord injury.
Brain, 128: 2359–2371.
Regeneration Research is a Collaboration of the
Hains, B.C., Saab, C.Y. and Waxman, S.G. (2006) Alterations
Paralyzed Veterans of America and the United in burst firing of thalamic VPL neurons and reversal by
Spinal Association. BCH was funded by The Na(v)1.3 antisense after spinal cord injury. J. Neurophysiol.,
Christopher Reeve Paralysis Foundation (HB1- 95: 3343–3352.
0304-2), the NIH/NINDS (1 F32 NS046919-01), Hains, B.C. and Waxman, S.G. (2006) Activated microglia
contribute to the maintenance of chronic pain after spinal
and Pfizer (Scholar’s Grant in Pain Medicine).
cord injury. J. Neurosci., 26: 4308–4317.
Jeanmonod, D., Magnin, M. and Morel, A. (1993) Thalamus
and neurogenic pain: physiological, anatomical, and clinical
References data. Neuroreport, 4: 475–478.
Jones, E.G. (1998) Viewpoint: the core and matrix of thalamic
Cairns, D.M., Adkins, R.H. and Scott, M.D. (1996) Pain and organization. Neuroscience, 85: 331–345.
depression in acute traumatic spinal cord injury: origins of Koyama, S., Katayama, Y., Maejima, S., Hirayama, T., Fujii,
chronic problematic pain? Arch. Phys. Med. Rehab., 77: M. and Tsubokawa, T. (1993) Thalamic neuronal hyperac-
329–335. tivity following transection of the spinothalamic tract in the
Cummins, T.R., Aglieco, F., Renganathan, M., Herzog, R.I., cat: involvement of N-methyl-D-aspartate receptor. Brain
Dib-Hajj, S.D. and Waxman, S.G. (2001) Nav1.3 sodium Res., 612: 345–350.
channels: rapid repriming and slow closed-state inactivation Lampert, A., Hains, B.C. and Waxman, S.G. (2006) Upregu-
display quantitative differences after expression in a mam- lation of persistent and ramp sodium current in dorsal horn
malian cell line and in spinal sensory neurons. J. Neurosci., neurons after spinal cord injury. Exp. Brain Res., 174:
21: 5952–5961. 660–666.
Cummins, T.R. and Waxman, S.G. (1997) Downregulation of Lenz, F.A., Kwan, H.C., Dostrovsky, J.O. and Tasker, R.R.
tetrodotoxin-resistant sodium currents and upregulation of a (1989) Characteristics of the bursting pattern of action po-
rapidly repriming tetrodotoxin-sensitive sodium current in tentials that occurs in the thalamus of patients with central
small spinal sensory neurons after nerve injury. J. Neurosci., pain. Brain Res., 496: 357–360.
17: 3503–3514. Lenz, F.A., Kwan, H.C., Martin, R., Tasker, R., Richardson,
Csicsvari, J., Hirase, H., Czurko, A. and Buzsaki, G. (1998) R.T. and Dostrovsky, J.O. (1994) Characteristics of somato-
Reliability and state dependence of pyramidal cell-interneu- topic organization and spontaneous neuronal activity in the
ron synapses in the hippocampus: an ensemble approach in region of the thalamic principal sensory nucleus in patients
the behaving rat. Neuron, 21: 179–189. with spinal cord transection. J. Neurophysiol., 72: 1570–1587.
Felts, P.A., Yokoyama, S., Dib-Hajj, S., Black, J.A. and Wax- Mariano, A.J. (1992) Chronic pain and spinal cord injury. Clin.
man, S.G. (1997) Sodium channel alpha-subunit mRNAs I, J. Pain, 8: 87–92.
II, III, NaG, Na6 and hNE (PN1): different expression pat- McCormick, D.A. (1999) Are thalamocortical rhythms the
terns in developing rat nervous system. Mol. Brain Res., 45: Rosetta Stone of a subset of neurological disorders? Nat.
71–82. Med., 5: 1349–1351.
203

Nepomuceno, C., Fine, P.R., Richards, J.S., Gowens, H., dysaesthesiae in spinal cord injury patients: results of a multi-
Stover, S.L., Rantanuabol, U. and Houston, R. (1979) Pain centre study. Spinal Cord, 35: 446–455.
in patients with spinal cord injury. Arch. Phys. Med. Re- Swadlow, H.A. and Gusev, A.G. (2001) The impact of ‘burst-
habil., 60: 605–609. ing’ thalamic impulses at a neocortical synapse. Nat. Neuro-
Pattany, P.M., Yezierski, R.P., Widerstrom-Noga, E.G., Bowen, sci., 4: 402–408.
B.C., Martinez-Arizala, A., Garcia, B.R. and Quencer, R.M. Turner, J.A., Cardenas, D.D., Warms, C.A. and McClellan,
(2002) Proton magnetic resonance spectroscopy of the C.B. (2001) Chronic pain associated with spinal cord injuries:
thalamus in patients with chronic neuropathic pain after spi- a community survey. Arch. Phys. Med. Rehabil., 82:
nal cord injury. Am. J. Neuroradiol., 23: 901–905. 501–509.
Reinagel, P., Godwin, D., Sherman, S.M. and Koch, C. (1999) Waxman, S.G. (2000) The neuron as a dynamic electrogenic
Encoding of visual information by LGN bursts. J. Neuro- machine: modulation of sodium-channel expression as a basis
physiol., 81: 2558–2569. for functional plasticity in neurons. Philos. Trans. R. Soc.
Rintala, D.H., Loubser, P.G., Castro, J., Hart, K.A. and Lond. B. Biol. Sci., 355: 199–213.
Fuhrer, M.J. (1998) Chronic pain in a community-based Waxman, S.G. and Hains, B.C. (2006) Fire and phantoms after
sample of men with spinal cord injury: prevalence, severity, spinal cord injury: Na+ channels and central pain. Trends
and relationship with impairment, disability, handicap, and Neurosci., 29: 207–215.
subjective well-being. Arch. Phys. Med. Rehabil., 79: Weng, H.R., Lee, J.I., Lenz, F.A., Schwartz, A., Vierck, C.,
604–614. Rowland, L. and Dougherty, P.M. (2000) Functional plas-
Siddall, P.J., McClelland, J.M., Rutkowski, S.B. and Cousins, ticity in primate somatosensory thalamus following chronic
M.J. (2003) A longitudinal study of the prevalence and char- lesion of the ventral lateral spinal cord. Neuroscience, 101:
acteristics of pain in the first 5 years following spinal cord 393–401.
injury. Pain, 103: 249–257. Yezierski, R.P. (2001) Pain following spinal cord injury: patho-
Stormer, S., Gerner, H.J., Gruninger, W., Metzmacher, K., physiology and central mechanisms. In: Sandkühler J., et al.
Follinger, S., Wienke, C., Aldinger, W., Walker, N., Zimmer- (Eds.), Progress in Brain Research. Elsevier, Amsterdam,
mann, M. and Paeslack, V. (1997) Chronic pain/ pp. 429–449.
This page intentionally left blank
SECTION IV

Novel Aspects of Clinical Research in CNS Injury


This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 14

Monitoring cerebral oxygenation in traumatic


brain injury

Iain K. Haitsma and Andrew I.R. Maas

Department of Neurosurgery, Erasmus Medical Center, ’s-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands

Abstract: Ischemia is a common problem after traumatic brain injury (TBI) that eludes detection with
standard monitoring. In this review we will discuss four available techniques (SjVO2, PET, NIRS and
PbrO2) to monitor cerebral oxygenation. We present technical data including strengths and weaknesses of
these systems, information from clinical studies and formulate a vision for the future.

Keywords: TBI; PbrO2; brain tissue oxygen tension; SjVO2; PET; NIRS; tissue oxygen reactivity; advanced
neuromonitoring

Introduction (dis)advantages, the available clinical data with


results of interventions with CPP and respiratory
Treatment of patients with severe traumatic brain variations and will conclude summarizing our view
injury (TBI) is largely focused on the prevention of of the future with these new technologies.
secondary insults. Standard monitoring consists
of measuring intra cranial pressure (ICP),
(mean) arterial blood pressure (MAP) and thus Monitoring technologies
calculating the cerebral perfusion pressure
(CPP ¼ MAPICP). This has led to ICP and Global vs. focal monitoring
CPP driven treatment protocols (Rosner et al.,
1995; Nordstrom, 2005). However these measure- The different measurement modalities have differ-
ments do not give further information on the ent working areas. Some work very focally (e.g.
existence of ischemia which is common in post brain tissue oxygen tension measurement) others
mortem studies after TBI (Graham et al., 1978, globally (e.g. jugular bulb oxygenation). This leads
1989). Much effort has gone into the development to differences in their use and interpretation of
of extra monitoring of the injured brain, so called their values (Sarrafzadeh et al., 1998; Gopinath
multi modal monitoring (Unterberg et al., 1997; et al., 1999; Bellander et al., 2004; Engstrom et al.,
Meixensberger et al., 1998; Mulvey et al., 2004; 2005). The advantage of a global measurement is
De Georgia and Deogaonkar, 2005; Vespa, 2005). the information on a large part of the injured
This review will focus on monitoring oxygenation brain, more so because most treatments in TBI are
in the injured brain. We will discuss the available systemic.
technologies with special emphasis on brain The more focal measurements have been typi-
tissue oxygen tension measurement, their cally used in two ways. Either they have been ap-
plied to non-injured areas in essence using them
Corresponding author. Tel.: +31-10-463-4077; Fax: +31-10- as global monitors or to the penumbra of focal
463-4075; E-mail: airmaas@erasmusmc.nl lesions, for instance contusions. The second

DOI: 10.1016/S0079-6123(06)61014-5 207


208

Table 1. Information regarding measurement technologies

Technique Catheter location Global vs. focal Continuous or intermittent

Jugular bulb oximetry Intravenous Global Continuous or intermittent


PET Extracranial Global and focal Intermittent
Near infra red spectroscopy Extracranial Focal Continuous or intermittent
Brain tissue oxygen tension Intraparenchymal Focal Continuous

approach will provide earlier information on the 1993b; Fortune et al., 1994; Kiening et al., 1996;
state of the tissue most threatened at that time. Meixensberger et al., 1998; Gopinath et al., 1999).
However there are practical problems in defining One article is devoted to possible technical prob-
the penumbra zone, in accurately positioning the lems with jugular oximetry (Dearden and Midgley,
catheter and finally in interpreting the absolute 1993). Others claim to have little difficulties with
values measured. jugular venous oxygen saturation (SjVO2) meas-
The data regarding the different systems com- urements (Cruz, 1997). Apart from measuring
bined with their ability to measure continuously or jugular oxygen saturation further information can
intermittently are summarized in Table 1. be gained by calculating the arteriovenous differ-
ence in oxygen content (AVDO2) or even cerebral
extraction of oxygen (Le Roux et al., 1997; Cruz,
Jugular bulb oximetry 1998).
In conclusion, it can be stated that jugular
The jugular bulb, the cranial tip of the internal oximetry is a relatively invasive mode of measure-
jugular vein, contains pure cerebrovenous blood. ment that can be subject to technical difficulties,
Back in the early 1900s the jugular vein was the best side of measurement is the topic of debate.
pierced to gain information on cerebrovenous It can give valuable information on oxygen con-
oxygenation (White and Baker, 2002). Later in sumption either continuously or intermittent.
the 1940s it was used for calculating CBF by Kety
and Schmidt (1945). Since the early 1990s litera-
ture has accumulated on the value of measuring
the oxygen saturation of jugular blood (Robertson PET scan
et al., 1992; Cruz, 1993a; De Deyne, 1999; White
and Baker, 2002). Using 15O2 as an inhalant information can be
The measurement can be done with intermittent gained on oxygen consumption using positron
blood gas samples or continuously by inserting emission tomography (PET) scanning. This
a fiberoptic catheter. The side of cannulation is method is non-invasive, it does however expose
debated. Some propose to use the right side, as the the patient to radiation and more importantly de-
right jugular bulb is usually dominant, others test pends on the patient being out of his relatively safe
for dominancy of a side, but alternatively the ICU environment for a longer period. Other
choice can be made to measure on the side that is downsides include the intermittent character and
most injured. Variability exists if both sides are the limited availability of this technique. Informa-
measured continuously (Metz et al., 1998). This of tion can be gained on cerebral blood flow (CBF),
course raises questions on whether the values can oxygen extraction fraction (OEF) and cerebral
really be interpreted as indicative of the whole metabolic rate of O2 (CMRO2). The calculations
brain. Technical problems have been reported are regional, but the whole brain can be measured.
with continuous measurements with time of Using voxel-based analysis the ischemic burden
good quality ranging from 43 to 91.5% (Cruz, can be quantified (Coles et al., 2004b).
209

Near infra red spectroscopy (NIRS) thick probes, usually via a bolt system. The Licox
system uses a Clark type electrode, which gener-
NIRS is a non-invasive measurement of Hb oxy- ates a current that is dependent on the amount of
genation. As the name implies it uses near infrared oxygen near the catheter tip. The Paratrend also
light that can cross the skull. Hb is a strong uses a Clark electrode and later changes to an
absorber of near infrared light and the amount of optode system in the Neurotrend. The Para/
absorption depends on the degree of Hb oxygen- Neurotrend supplies information on local temper-
ation. Information can be supplied in the percent- ature, pCO2 and pH as well. The Licox now also
age of HbO2 saturation, or absolute changes in incorporates a temperature sensor. Complication
HbO2, deoxyhemoglobin and total hemoglobin rates are equally low as in the use of intraparen-
in micromolar concentrations, referenced to an chymal ICP monitors and data are reliable with
arbitrary baseline (Cho et al., 2000). Technical very low catheter drift rates (Kiening et al., 1996;
problems have precluded wide scale use of this non- Dings et al., 1998; van den Brink et al., 2000;
invasive measurement. If the source and detector Hoelper et al., 2005). Time of good data quality
are not far enough apart, measurements are influ- of up to 95% has been reported (Meixensberger
enced by subcutaneous blood flow. With complete et al., 1998). Catheters are usually inserted in the
ischemia near normal values have been found, pos- frontal white matter and give continuous informa-
sibly related to venous blood sequestered in the tion on the local balance between oxygen supply
infracted tissue (Gomersall et al., 1997). and uptake. After insertion, a run in time of 1–2 h
Because of these technical problems, especially is advised before reliable data are produced (Dings
in TBI patients, NIRS is currently rarely used in et al., 1998; van den Brink et al., 2000). We reco-
adults. With recent modifications its use might mmend performing an oxygen challenge after
increase again (Al-Rawi, 2005). Technical limita- catheter insertion to verify its proper placement,
tions are less relevant in neonates. as even small hematomas around the catheter tip
can lower the oxygen readings, which would result
in no or very little increase in PbrO2 with a large
Brain tissue oxygen tension (PbrO2) increase in inspiratory oxygen fraction (FiO2) or
more appropriate arterial oxygen tension (PaO2)
Following extensive pre-clinical work, the intro- (van den Brink et al., 1998).
duction of clinical PbrO2 measurements was pio- In conclusion, PbrO2 measurement is an inva-
neered by the Rotterdam group (Maas et al., 1993) sive, but safe and reliable measurement that gives
and the technique formally introduced during the continuous information on local oxygenation.
first congress on cerebral oxygenation organized
by the Rotterdam, Berlin and Wurzburg groups
in 1995. Initial publications followed rapidly
(Kiening et al., 1996; van Santbrink et al., 1996; Clinical data
Meixensberger et al., 1998). Many articles includ-
ing review articles are available on this subject Jugular oximetry
(Andrews, 2001; Haitsma and Maas, 2002; Bader
et al., 2003; Littlejohns et al., 2003; De Georgia Low SjVO2 values (below 50–55%) are related to
and Deogaonkar, 2005; Vespa, 2005; Rose et al., poorer outcome (Gopinath et al., 1994; Fandino
2006). Most reports use one of the two technolo- et al., 2000; Perez et al., 2003). The same holds
gies — the Licox system (Integra lifesciences, for values higher than 75% (Cormio et al., 1999;
Plainsboro, NJ) or the Para/Neurotrend system Macmillan et al., 2001). Others have challenged
(Charbel et al., 1997) (Diametrics Medical Ltd., these classic cut-off values after measurements in
UK). Unfortunately the Para/Neurotrend system patients with Cushing syndrome (44.7–69.5%)
is no longer available. Both techniques require (Chieregato et al., 2003). A limited improvement
intraparenchymal insertion of the 0.5–0.8 mm as opposed to clear improvement in increased
210

AVDO2 after treatment is also related to poorer hyperventilation (Diringer et al., 2000). In a study
prognosis (Le Roux et al., 1997). with 13 TBI patients receiving hyperventilation,
In two important publications the Houston CBF was reduced to even below 20 ml/100 g/min,
group compared ICP (CPP50 mmHg, PaCO2 but no changes in oxygen extraction fraction were
25–30 mmHg) and CBF (CPP70 mmHg, PaCO2 noted. It was concluded that no energy failure
35 mmHg) regimens to each other (Robertson took place during hyperventilation (Diringer et al.,
et al., 1999; Contant et al., 2001). In the CBF 2002). Another study confirmed that hyperventi-
group jugular desaturations were less frequent lation lowered CBF but not to the level of ischemia
(30% vs. 50.6%). No difference in outcome was (Coles et al., 2002).
observed, possibly because benefits of treatment
of jugular desaturations were offset by a fivefold
NIRS
increase in ARDS in the CBF group. These pub-
lications led to lowering of the recommended CPP
Few clinical studies with regard to oxygenation
level in the guidelines from 70 to 60 mmHg.
measurements have been published on NIRS.
Hyperventilation leads to a decrease in SjVO2
Interesting is its possible use as a detector of
and increase in AVDO2, these effects however can
intracranial hematomas. In the presence of a sub-
be offset by hyperoxygenation (Matta et al., 1994;
dural hematoma or bloody contusion, or even in
Thiagarajan et al., 1998). A treatment protocol
the event of massive blood in the subarachnoid
using the cerebral extraction of oxygen as guide for
space, absorption is high, thus preventing suffi-
treatment including targeted hyperventilation has
cient light to return to the detectors. This phe-
shown promising results in adults with TBI and
nomenon may be used to detect delayed traumatic
diffuse brain swelling and separately in children
hematomas, leading to better timed follow-up CTs
with severe TBI and increased ICP (Cruz, 1998;
and operations (Gopinath et al., 1993, 1995).
Cruz et al., 2002).
In TBI patients with decreased CBF hyperbaric
O2 therapy can increase CBF and CMRO2 at 1 PbrO2
and 6 h after treatment, at constant AVDO2
(Rockswold et al., 2001). Increased ICP and CSF Low PbrO2 values are observed in up to 50% of
lactate levels were also lowered. patients during the first 24 h with depth and du-
ration of tissue hypoxia being related to outcome
and being an independent predictor of unfavorable
PET measurements outcome and death (Bardt et al., 1998; Valadka
et al., 1998; van den Brink et al., 2000; Stiefel et al.,
Even with PET measurements debate exists on the 2006). No absolute value can be given as a cut-off
frequency of regional ischemia (Coles et al., 2004a; point for tissue hypoxia but values ranging from
Vespa et al., 2005). Ischemia was defined as the 10 to 20 mmHg in uninjured frontal white matter
oxygen extraction fraction at a cerebral venous are mentioned based on prognostic analysis or
oxygen (CVO2) content below 3.5 ml/100 ml. One related to CBF values (Zauner et al., 1997; Bardt
study reports ischemic volume ranging from 1 to et al., 1998; Doppenberg et al., 1998b; Valadka
16% (Coles et al., 2004a), the other maximally 1% et al., 1998; van den Brink et al., 2000).
of brain volume in TBI patients (Vespa et al., A sudden fall in PbrO2 to zero, without the
2005). development of a hematoma around the catheter,
Increasing CPP from 70 to 90 mmHg produces is considered an early sign of brain death (van
a significant fall in ischemic brain volume (IBV) Santbrink et al., 1996; Palmer and Bader, 2005).
which is most pronounced in patients with a larger Recently interest has increased on testing auto-
IBV (Coles et al., 2004c). regulation (Mascia et al., 2000; Czosnyka et al.,
A study published in 2000 found a decrease 2001; Steiner et al., 2002; Cremer et al., 2004).
in CBF but no change in CMRO2 with Patients with mean CPP levels close to their
211

optimal CPP are more likely to have a favorable and Stocchetti, 1999; Bressack and Schiffman,
outcome (Steiner et al., 2002). The response of 2003). A report by the Milan group showed re-
PbrO2 to changes in CPP is also dependent on the duced lactate levels, but not the lactate–pyruvate
state of autoregulation (Lang et al., 2003; Cremer ratio with hyperoxia which was interpreted as ab-
et al., 2004). With increased ICP a CPP reduction sence of improvement in cerebral metabolism
below 77 mmHg can lead to a further ICP increase (Magnoni et al., 2003). A more recent report by
and decrease in PbrO2 (Cremer et al., 2004). the Richmond and Bern groups also demonstrated
Higher CPP levels might be useful in normalizing a fall in lactate–pyruvate ratio after hyperoxia
tissue oxygenation in ischemic areas (Stocchetti compared to a historical control group (Tolias
et al., 1998). A historical case control study by the et al., 2004). Nevertheless, even oxygen can be
Leipzig group from 2003 with increased CPP for considered a drug and is not without its side effects
low PbrO2 values showed less frequent cerebral (Lodato, 1990). Further reports on outcome meas-
hypoxia, but no difference in 6-month outcome ures with hyperoxia treatment are eagerly awaited.
(Meixensberger et al., 2003). PbrO2 decreases with hypothermia with an extra
One publication by the Pennsylvania group reduction below 351C (Gupta et al., 2002) in one
claimed reduced patient death using both ICP study, others report an increase in PbrO2 with
and PbrO2 monitoring (Stiefel et al., 2005). This hypothermia (Zhi et al., 1999; Jia et al., 2005).
was based on a historical case control study, in Decompressive craniectomies for intractable
which patients with ICP and PbrO2 monitoring ICP leads to increased PbrO2 (Jaeger et al., 2003;
first were treated according to an ICP/CPP proto- Stiefel et al., 2004).
col, later patients were entered in a stepwise pro-
tocol that also treated PbrO2 values below
25 mmHg. Mortality rate dropped from 44 to Technology comparisons
25%. The major critique on this study concerns its
retrospective nature. SjVO2 values correlate with IBV, defined as the
Hyperventilation usually lowers PbrO2 (Schneider area with CVO2 content p3.5 ml/100 ml as meas-
et al., 1998), and reactivity is higher in patients ured by PET scan, with SjVO2 o50% occurring at
with poorer prognosis, statistically significant on IBV at 1375% (Coles et al., 2004a).
day 5 after injury (Carmona Suazo et al., 2000). In a study in 14 patients NIRS detected twice
Increases in PbrO2 with hyperventilation in TBI as many events as SjVO2 monitoring although
have however also been reported (Dings et al., the clinical significance of this is not known
1996; Imberti et al., 2000). (Kirkpatrick et al., 1995). Another study found
PbrO2 can be increased by increasing FiO2/ poor correlation between NIRS and SjVO2 mon-
PaO2. itoring even with SjVO2 desaturations below 55%
Tissue oxygen response (TOR) can be calculated (Lewis et al., 1996). In a study in 60 children
as: without TBI, SjVO2 and NIRS correlated signifi-
cantly but NIRS had lower sensitivity (Nagdyman
PbrO2 1
TOR ¼  et al., 2005).
PaO2 PbrO2 SjVO2 and PbrO2 correlate closely especially
Higher values of TOR are related to poorer when CPP falls below 60 mmHg (Kiening et al.,
outcome and may reflect a loss of oxygen auto- 1996). This correlation is less in areas with focal
regulation (van Santbrink et al., 1996, 2003). pathology (Gupta et al., 1999). Correlation be-
Several groups have investigated the effects of tween SjVO2 and PbrO2 during CO2 reactivity
increasing FiO2/PaO2 to supranormal levels. In testing is low (Fandino et al., 1999).
1999 the Richmond group reported a fall in brain In a study in 36 TBI patients moderate hyper-
lactate levels measured by microdialysis after ventilation reduced SjVO2, but not below 50%
administering 100% O2 (Menzel et al., 1999a, b). with PbrO2 values below 10 mmHg (Imberti et al.,
These results were not without controversy (Rossi 2002), in some tests SjVO2 and PbrO2 changed in
212

different directions, supporting the concept that Acknowledgments


these technologies are complementary.
In a 2002 study PbrO2 did not correlate with The work of Dr. Haitsma is supported by NWO
cerebral end capillary O2 tension as measured with grant: 920-03-130. The Department of Neurosur-
PET. The change in both values during hyperven- gery, EMC, has received in natural support of
tilation was however significantly correlated. research in neurocritical care from GMS mbh.
PbrO2 is strongly correlated with local CBF (Kiel, Germany) and Codman/Johnson & Johnson
measurement (Doppenberg et al., 1998a; Jaeger (Raynham, MA, USA).
et al., 2005).

References
Summary and conclusions
Al-Rawi, P.G. (2005) Near infrared spectroscopy in brain
ICP and CPP measurements alone are inadequate injury: today’s perspective. Acta Neurochir. Suppl., 95:
for detecting ischemia. The above-mentioned tech- 453–457.
Andrews, R.J. (2001) Monitoring for neuroprotection. New
niques can provide valuable information on oxy- technologies for the new millennium. Ann. N.Y. Acad. Sci.,
genation. An ideal technique (non-invasive, 939: 101–113.
continuous, reliable and easy to use on the ICU) Bader, M.K., Littlejohns, L.R. and March, K. (2003) Brain
does not exist. To gain maximal insight the com- tissue oxygen monitoring in severe brain injury, II. Implica-
tions for critical care teams and case study. Crit. Care Nurse,
bination of a local and global monitor should
23: 29–38, 40–42, 44.
suffice. However the advantages in outcome with Bardt, T.F., Unterberg, A.W., Hartl, R., Kiening, K.L.,
extra measurements or targeted treatment have Schneider, G.H. and Lanksch, W.R. (1998) Monitoring of
not yet been unequivocally proven. Any effort to brain tissue PO2 in traumatic brain injury: effect of cerebral
do so should be stimulated and probably be part hypoxia on outcome. Acta Neurochir. Suppl., 71: 153–156.
Bellander, B.M., Cantais, E., Enblad, P., Hutchinson, P.,
of a multi-center initiative, for instance Brain-IT.
Nordstrom, C.H., Robertson, C., Sahuquillo, J., Smith, M.,
Recent reports of improved survival after PbrO2 Stocchetti, N., Ungerstedt, U., Unterberg, A. and Olsen,
measurement and treatment (Stiefel et al., 2005) N.V. (2004) Consensus meeting on microdialysis in neuroin-
and reports of improved metabolism with hyper- tensive care. Intensive Care Med., 30: 2166–2169.
oxia (Tolias et al., 2004) seem very promising. Bressack, M.A. and Schiffman, J. (2003) Hyperoxia: good or
The same holds true for the reports on targeted bad? J. Neurosurg., 98: 223–224; author reply 224–225.
Carmona Suazo, J.A., Maas, A.I., van den Brink, W.A.,
hyperventilation with jugular bulb measurements van Santbrink, H., Steyerberg, E.W. and Avezaat, C.J. (2000)
(Cruz, 1998; Cruz et al., 2002). These studies de- CO2 reactivity and brain oxygen pressure monitoring in se-
serve verification in other centers. Because of their vere head injury. Crit. Care Med., 28: 3268–3274.
broad availability SjVO2 and PbrO2 measurements Charbel, F.T., Hoffman, W.E., Misra, M., Hannigan, K. and
Ausman, J.I. (1997) Cerebral interstitial tissue oxygen ten-
seem to be the most logical for multi center studies
sion, pH, HCO3, CO2. Surg. Neurol., 48: 414–417.
with a slight preference for the Licox system be- Chieregato, A., Calzolari, F., Trasforini, G., Targa, L. and
cause of its ease of use. After stepwise implemen- Latronico, N. (2003) Normal jugular bulb oxygen saturation.
tations of new monitoring techniques a software J. Neurol. Neurosurg. Psychiatry, 74: 784–786.
system capable of integrating all values and sup- Cho, H., Nemoto, E.M., Sanders, M., Fernandez, K. and
plying recommendations for further diagnostic Yonas, H. (2000) Comparison of two commercially available
near-infrared spectroscopy instruments for cerebral oximetry.
steps or treatment will be crucial. Until then we Technical note. J. Neurosurg., 93: 351–354.
will eagerly await and participate in further studies Coles, J.P., Fryer, T.D., Smielewski, P., Chatfield, D.A.,
on autoregulation guided CPP treatment, effect Steiner, L.A., Johnston, A.J., Downey, S.P., Williams,
of hyperoxygenation treatment, targeted hyper- G.B., Aigbirhio, F., Hutchinson, P.J., Rice, K., Carpenter,
T.A., Clark, J.C., Pickard, J.D. and Menon, D.K. (2004a)
ventilation and using multimodality monitoring
Incidence and mechanisms of cerebral ischemia in early clini-
as secondary endpoints in the use of treatments cal head injury. J. Cereb. Blood Flow Metab., 24: 202–211.
such as barbiturate coma, hypothermia and de- Coles, J.P., Fryer, T.D., Smielewski, P., Rice, K., Clark, J.C.,
compressive craniectomy. Pickard, J.D. and Menon, D.K. (2004b) Defining ischemic
213

burden after traumatic brain injury using 15O PET imaging Dings, J., Meixensberger, J., Amschler, J., Hamelbeck, B. and
of cerebral physiology. J. Cereb. Blood Flow Metab., 24: Roosen, K. (1996) Brain tissue PO2 in relation to cerebral
191–201. perfusion pressure, TCD findings and TCD-CO2-reactivity
Coles, J.P., Minhas, P.S., Fryer, T.D., Smielewski, P., after severe head injury. Acta Neurochir., 138: 425–434.
Aigbirihio, F., Donovan, T., Downey, S.P., Williams, G., Dings, J., Meixensberger, J., Jager, A. and Roosen, K. (1998)
Chatfield, D., Matthews, J.C., Gupta, A.K., Carpenter, T.A., Clinical experience with 118 brain tissue oxygen partial pres-
Clark, J.C., Pickard, J.D. and Menon, D.K. (2002) Effect of sure catheter probes. Neurosurgery, 43: 1082–1095.
hyperventilation on cerebral blood flow in traumatic head Diringer, M.N., Videen, T.O., Yundt, K., Zazulia, A.R., Aiyagari,
injury: clinical relevance and monitoring correlates. Crit. V., Dacey Jr., R.G., Grubb, R.L. and Powers, W.J. (2002) Re-
Care Med., 30: 1950–1959. gional cerebrovascular and metabolic effects of hyperventilation
Coles, J.P., Steiner, L.A., Johnston, A.J., Fryer, T.D., after severe traumatic brain injury. J. Neurosurg., 96: 103–108.
Coleman, M.R., Smieleweski, P., Chatfield, D.A., Aigbirhio, F., Diringer, M.N., Yundt, K., Videen, T.O., Adams, R.E.,
Williams, G.B., Boniface, S., Rice, K., Clark, J.C., Pickard, J.D. Zazulia, A.R., Deibert, E., Aiyagari, V., Dacey Jr., R.G.,
and Menon, D.K. (2004c) Does induced hypertension reduce Grubb Jr., R.L. and Powers, W.J. (2000) No reduction
cerebral ischaemia within the traumatized human brain? in cerebral metabolism as a result of early moderate hyper-
Brain, 127: 2479–2490. ventilation following severe traumatic brain injury. J. Neuro-
Contant, C.F., Valadka, A.B., Gopinath, S.P., Hannay, H.J. surg., 92: 7–13.
and Robertson, C.S. (2001) Adult respiratory distress syn- Doppenberg, E.M., Zauner, A., Bullock, R., Ward, J.D.,
drome: a complication of induced hypertension after severe Fatouros, P.P. and Young, H.F. (1998a) Correlations bet-
head injury. J. Neurosurg., 95: 560–568. ween brain tissue oxygen tension, carbon dioxide tension,
Cormio, M., Valadka, A.B. and Robertson, C.S. (1999) Ele- pH, and cerebral blood flow–a better way of monitoring the
vated jugular venous oxygen saturation after severe head in- severely injured brain? Surg. Neurol., 49: 650–654.
jury. J. Neurosurg., 90: 9–15. Doppenberg, E.M., Zauner, A., Watson, J.C. and Bullock, R.
Cremer, O.L., van Dijk, G.W., Amelink, G.J., de Smet, A.M., (1998b) Determination of the ischemic threshold for brain
Moons, K.G. and Kalkman, C.J. (2004) Cerebral hemody- oxygen tension. Acta Neurochir. Suppl., 71: 166–169.
namic responses to blood pressure manipulation in severely Engstrom, M., Polito, A., Reinstrup, P., Romner, B.,
head-injured patients in the presence or absence of intracra- Ryding, E., Ungerstedt, U. and Nordstrom, C.H. (2005)
nial hypertension. Anesth. Analg., 99: 1211–1217, table of Intracerebral microdialysis in severe brain trauma: the im-
contents. portance of catheter location. J. Neurosurg., 102: 460–469.
Cruz, J. (1993a) Cerebral oxygenation. Monitoring and man- Fandino, J., Stocker, R., Prokop, S. and Imhof, H.G. (1999)
agement. Acta Neurochir. Suppl., 59: 86–90. Correlation between jugular bulb oxygen saturation and
Cruz, J. (1993b) On-line monitoring of global cerebral hypoxia partial pressure of brain tissue oxygen during CO2 and O2
in acute brain injury. Relationship to intracranial hyperten- reactivity tests in severely head-injured patients. Acta
sion. J. Neurosurg., 79: 228–233. Neurochir., 141: 825–834.
Cruz, J. (1997) Continuous monitoring of partial pressure Fandino, J., Stocker, R., Prokop, S., Trentz, O. and Imhof,
of brain tissue oxygen in patients with severe head injury. H.G. (2000) Cerebral oxygenation and systemic trauma
Neurosurgery, 40: 1104. related factors determining neurological outcome after brain
Cruz, J. (1998) The first decade of continuous monitoring of injury. J. Clin. Neurosci., 7: 226–233.
jugular bulb oxyhemoglobinsaturation: management strate- Fortune, J.B., Feustel, P.J., Weigle, C.G. and Popp, A.J. (1994)
gies and clinical outcome. Crit. Care Med., 26: 344–351. Continuous measurement of jugular venous oxygen satura-
Cruz, J., Nakayama, P., Imamura, J.H., Rosenfeld, K.G., tion in response to transient elevations of blood pressure in
de Souza, H.S. and Giorgetti, G.V. (2002) Cerebral extrac- head-injured patients. J. Neurosurg., 80: 461–468.
tion of oxygen and intracranial hypertension in severe, acute, Gomersall, C.D., Joynt, G.M., Gin, T., Freebairn, R.C. and
pediatric brain trauma: preliminary novel management strat- Stewart, I.E. (1997) Failure of the INVOS 3100 cerebral oxi-
egies. Neurosurgery, 50: 774–779; discussion 779–780. meter to detect complete absence of cerebral blood flow. Crit.
Czosnyka, M., Smielewski, P., Piechnik, S., Steiner, L.A. and Care Med., 25: 1252–1254.
Pickard, J.D. (2001) Cerebral autoregulation following head Gopinath, S.P., Robertson, C.S., Contant, C.F., Hayes, C.,
injury. J. Neurosurg., 95: 756–763. Feldman, Z., Narayan, R.K. and Grossman, R.G. (1994)
Dearden, N.M. and Midgley, S. (1993) Technical considera- Jugular venous desaturation and outcome after head injury.
tions in continuous jugular venous oxygen saturation meas- J. Neurol. Neurosurg. Psychiatry, 57: 717–723.
urement. Acta Neurochir. Suppl., 59: 91–97. Gopinath, S.P., Robertson, C.S., Contant, C.F., Narayan, R.K.,
De Deyne, C. (1999) Jugular bulb oximetry: the link between Grossman, R.G. and Chance, B. (1995) Early detection of de-
cerebral and systemic management of severe head injury. layed traumatic intracranial hematomas using near-infrared
Intensive Care Med., 25: 430–431. spectroscopy. J. Neurosurg., 83: 438–444.
De Georgia, M.A. and Deogaonkar, A. (2005) Multimodal Gopinath, S.P., Robertson, C.S., Grossman, R.G. and Chance, B.
monitoring in the neurological intensive care unit. Neuro (1993) Near-infrared spectroscopic localization of intracranial
logist, 11: 45–54. hematomas. J. Neurosurg., 79: 43–47.
214

Gopinath, S.P., Valadka, A.B., Uzura, M. and Robertson, C.S. Kirkpatrick, P.J., Smielewski, P., Czosnyka, M., Menon, D.K.
(1999) Comparison of jugular venous oxygen saturation and and Pickard, J.D. (1995) Near-infrared spectroscopy use in
brain tissue PO2 as monitors of cerebral ischemia after head patients with head injury. J. Neurosurg., 83: 963–970.
injury. Crit. Care Med., 27: 2337–2345. Lang, E.W., Czosnyka, M. and Mehdorn, H.M. (2003)
Graham, D.I., Adams, J.H. and Doyle, D. (1978) Ischaemic Tissue oxygen reactivity and cerebral autoregulation
brain damage in fatal non-missile head injuries. J. Neurol. after severe traumatic brain injury. Crit. Care Med., 31:
Sci., 39: 213–234. 267–271.
Graham, D.I., Ford, I., Adams, J.H., Doyle, D., Teasdale, G.M., Le Roux, P.D., Newell, D.W., Lam, A.M., Grady, M.S. and
Lawrence, A.E. and McLellan, D.R. (1989) Ischaemic brain Winn, H.R. (1997) Cerebral arteriovenous oxygen difference:
damage is still common in fatal non-missile head injury. a predictor of cerebral infarction and outcome in patients
J. Neurol. Neurosurg. Psychiatry, 52: 346–350. with severe head injury. J. Neurosurg., 87: 1–8.
Gupta, A.K., Al-Rawi, P.G., Hutchinson, P.J. and Lewis, S.B., Myburgh, J.A., Thornton, E.L. and Reilly, P.L.
Kirkpatrick, P.J. (2002) Effect of hypothermia on brain tis- (1996) Cerebral oxygenation monitoring by near-infrared
sue oxygenation in patients with severe head injury. Br. J. spectroscopy is not clinically useful in patients with severe
Anaesth., 88: 188–192. closed-head injury: a comparison with jugular venous bulb
Gupta, A.K., Hutchinson, P.J., Al-Rawi, P., Gupta, S., Swart, M., oximetry. Crit. Care Med., 24: 1334–1338.
Kirkpatrick, P.J., Menon, D.K. and Datta, A.K. (1999) Meas- Littlejohns, L.R., Bader, M.K. and March, K. (2003) Brain
uring brain tissue oxygenation compared with jugular venous tissue oxygen monitoring in severe brain injury, I. Research
oxygen saturation for monitoring cerebral oxygenation after and usefulness in critical care. Crit. Care Nurse, 23: 17–25;
traumatic brain injury. Anesth. Analg., 88: 549–553. quiz 26–27.
Haitsma, I.K. and Maas, A.I. (2002) Advanced monitoring in Lodato, R.F. (1990) Oxygen toxicity. Crit. Care Clin., 6:
the intensive care unit: brain tissue oxygen tension. Curr. 749–765.
Opin. Crit. Care, 8: 115–120. Maas, A.I., Fleckenstein, W., de Jong, D.A. and van Santbrink, H.
Hoelper, B.M., Alessandri, B., Heimann, A., Behr, R. and (1993) Monitoring cerebral oxygenation: experimental studies
Kempski, O. (2005) Brain oxygen monitoring: in-vitro accu- and preliminary clinical results of continuous monitoring of
racy, long-term drift and response-time of Licox- and Neuro- cerebrospinal fluid and brain tissue oxygen tension. Acta Neuro-
trend sensors. Acta Neurochir. (Wien.), 147: 767–774. chir. Suppl., 59: 50–57.
Imberti, R., Bellinzona, G. and Langer, M. (2002) Cerebral Macmillan, C.S., Andrews, P.J. and Easton, V.J. (2001) In-
tissue PO2 and SjvO2 changes during moderate hyper creased jugular bulb saturation is associated with poor out-
ventilation in patients with severe traumatic brain injury. come in traumatic brain injury. J. Neurol. Neurosurg.
J. Neurosurg., 96: 97–102. Psychiatry, 70: 101–104.
Imberti, R., Ciceri, M., Bellinzona, G. and Pugliese, R. (2000) Magnoni, S., Ghisoni, L., Locatelli, M., Caimi, M., Colombo, A.,
The use of hyperventilation in the treatment of plateau Valeriani, V. and Stocchetti, N. (2003) Lack of improvement in
waves in two patients with severe traumatic brain injury: cerebral metabolism after hyperoxia in severe head injury: a
contrasting effects on cerebral oxygenation. J. Neurosurg. microdialysis study. J. Neurosurg., 98: 952–958.
Anesthesiol, 12: 124–127. Mascia, L., Andrews, P.J., McKeating, E.G., Souter, M.J.,
Jaeger, M., Soehle, M. and Meixensberger, J. (2003) Effects of Merrick, M.V. and Piper, I.R. (2000) Cerebral blood flow
decompressive craniectomy on brain tissue oxygen in patients and metabolism in severe brain injury: the role of pressure
with intracranial hypertension. J. Neurol. Neurosurg. Psy- autoregulation during cerebral perfusion pressure manage-
chiatry, 74: 513–515. ment. Intensive Care Med., 26: 202–205.
Jaeger, M., Soehle, M., Schuhmann, M.U., Winkler, D. and Matta, B.F., Lam, A.M. and Mayberg, T.S. (1994) The influ-
Meixensberger, J. (2005) Correlation of continuously moni- ence of arterial oxygenation on cerebral venous oxygen
tored regional cerebral blood flow and brain tissue oxygen. saturation during hyperventilation. Can. J. Anaesth., 41:
Acta Neurochir. (Wien.), 147: 51–56; discussion 56. 1041–1046.
Jia, J., Lin, Y.Q., Liu, W.F., Zhong, T.A., Zhang, J., Ye, Y. Meixensberger, J., Jaeger, M., Vath, A., Dings, J., Kunze, E.
and Xu, Y.Q. (2005) Study of the effects of mild hypothermia and Roosen, K. (2003) Brain tissue oxygen guided treatment
on cerebral PO2, PCO2 and pH and body temperature in supplementing ICP/CPP therapy after traumatic brain injury.
patients with acute severe head injury. Chin. J. Traumatol., 8: J. Neurol. Neurosurg. Psychiatry, 74: 760–764.
138–141. Meixensberger, J., Jager, A., Dings, J., Baunach, S. and
Kety, S.S. and Schmidt, C.F. (1945) The determination of cere- Roosen, K. (1998) Multimodal hemodynamic neuromoni-
bral blood flow in man by the use of nitrous oxide in low toring–quality and consequences for therapy of severely head
concentrations. Am. J. Physiol., 143: 53–66. injured patients. Acta Neurochir. Suppl., 71: 260–262.
Kiening, K.L., Unterberg, A.W., Bardt, T.F., Schneider, G.H. Menzel, M., Doppenberg, E.M., Zauner, A., Soukup, J.,
and Lanksch, W.R. (1996) Monitoring of cerebral oxygen- Reinert, M.M. and Bullock, R. (1999a) Increased inspired
ation in patients with severe head injuries: brain tissue PO2 oxygen concentration as a factor in improved brain tissue
versus jugular vein oxygen saturation. J. Neurosurg., 85: oxygenation and tissue lactate levels after severe human head
751–757. injury. J. Neurosurg., 91: 1–10.
215

Menzel, M., Doppenberg, E.M., Zauner, A., Soukup, J., Schneider, G.H., Sarrafzadeh, A.S., Kiening, K.L., Bardt, T.F.,
Reinert, M.M., Clausen, T., Brockenbrough, P.B. and Unterberg, A.W. and Lanksch, W.R. (1998) Influence of hy-
Bullock, R. (1999b) Cerebral oxygenation in patients after perventilation on brain tissue-PO2, PCO2, and pH in patients
severe head injury: monitoring and effects of arterial hype- with intracranial hypertension. Acta Neurochir. Suppl., 71:
roxia on cerebral blood flow, metabolism and intracranial 62–65.
pressure. J. Neurosurg. Anesthesiol., 11: 240–251. Steiner, L.A., Czosnyka, M., Piechnik, S.K., Smielewski, P.,
Metz, C., Holzschuh, M., Bein, T., Woertgen, C., Rothoerl, R., Chatfield, D., Menon, D.K. and Pickard, J.D. (2002) Con-
Kallenbach, B., Taeger, K. and Brawanski, A. (1998) Moni- tinuous monitoring of cerebrovascular pressure reactivity
toring of cerebral oxygen metabolism in the jugular bulb: allows determination of optimal cerebral perfusion pressure
reliability of unilateral measurements in severe head injury. in patients with traumatic brain injury. Crit. Care Med., 30:
J. Cereb. Blood Flow Metab., 18: 332–343. 733–738.
Mulvey, J.M., Dorsch, N.W., Mudaliar, Y. and Lang, E.W. Stiefel, M.F., Heuer, G.G., Smith, M.J., Bloom, S., Maloney-
(2004) Multimodality monitoring in severe traumatic brain Wilensky, E., Gracias, V.H., Grady, M.S. and LeRoux, P.D.
injury; The role of brain tissue oxygenation monitoring. (2004) Cerebral oxygenation following decompressive hemi-
Neurocrit. Care, 391–402. craniectomy for the treatment of refractory intracranial
Nagdyman, N., Fleck, T., Schubert, S., Ewert, P., Peters, B., hypertension. J. Neurosurg., 101: 241–247.
Lange, P.E. and Abdul-Khaliq, H. (2005) Comparison bet- Stiefel, M.F., Spiotta, A., Gracias, V.H., Garuffe, A.M.,
ween cerebral tissue oxygenation index measured by near- Guillamondegui, O., Maloney-Wilensky, E., Bloom, S.,
infrared spectroscopy and venous jugular bulb saturation in Grady, M.S. and LeRoux, P.D. (2005) Reduced mortality
children. Intensive Care Med., 31: 846–850. rate in patients with severe traumatic brain injury treated
Nordstrom, C.H. (2005) Physiological and biomechanical prin- with brain tissue oxygen monitoring. J. Neurosurg., 103:
ciples underlying volume-targeted therapy – the "Lund Con- 805–811.
cept". Neurocrit. Care, 83–96. Stiefel, M.F., Udoetuk, J.D., Spiotta, A.M., Gracias, V.H.,
Palmer, S. and Bader, M.K. (2005) Brain tissue oxygenation in Goldberg, A., Maloney-Wilensky, E., Bloom, S. and
brain death. Neurocrit. Care, 17–22. LeRoux, P.D. (2006) Conventional neurocritical care and
Perez, A., Minces, P.G., Schnitzler, E.J., Agosta, G.E., Medina, S.A. cerebral oxygenation after traumatic brain injury. J. Neuro-
and Ciraolo, C.A. (2003) Jugular venous oxygen saturation or surg., 105: 568–575.
arteriovenous difference of lactate content and outcome in chil- Stocchetti, N., Chieregato, A., De Marchi, M., Croci, M.,
dren with severe traumatic brain injury. Pediatr. Crit. Care Med., Benti, R. and Grimoldi, N. (1998) High cerebral perfusion
4: 33–38. pressure improves low values of local brain tissue O2 tension
Robertson, C.S., Contant, C.F., Gokaslan, Z.L., Narayan, R.K. (PtiO2) in focal lesions. Acta Neurochir. Suppl., 71: 162–165.
and Grossman, R.G. (1992) Cerebral blood flow, arteriove- Thiagarajan, A., Goverdhan, P.D., Chari, P. and Somasunde-
nous oxygen difference, and outcome in head injured patients. ram, K. (1998) The effect of hyperventilation and hyperoxia
J. Neurol. Neurosurg. Psychiatry, 55: 594–603. on cerebral venous oxygen saturation in patients with trau-
Robertson, C.S., Valadka, A.B., Hannay, H.J., Contant, C.F., matic brain injury. Anesth. Analg., 87: 850–853.
Gopinath, S.P., Cormio, M., Uzura, M. and Grossman, R.G. Tolias, C.M., Reinert, M., Seiler, R., Gilman, C., Scharf, A.
(1999) Prevention of secondary ischemic insults after severe and Bullock, M.R. (2004) Normobaric hyperoxia–induced
head injury. Crit. Care Med., 27: 2086–2095. improvement in cerebral metabolism and reduction in intra-
Rockswold, S.B., Rockswold, G.L., Vargo, J.M., Erickson, C.A., cranial pressure in patients with severe head injury: a pro-
Sutton, R.L., Bergman, T.A. and Biros, M.H. (2001) Effects spective historical cohort-matched study. J. Neurosurg., 101:
of hyperbaric oxygenation therapy on cerebral metabolism 435–444.
and intracranial pressure in severely brain injured patients. Unterberg, A.W., Kiening, K.L., Hartl, R., Bardt, T.,
J. Neurosurg., 94: 403–411. Sarrafzadeh, A.S. and Lanksch, W.R. (1997) Multimodal
Rose, J.C., Neill, T.A. and Hemphill, III, J.C. (2006) Contin- monitoring in patients with head injury: evaluation of the
uous monitoring of the microcirculation in neurocritical care: effects of treatment on cerebral oxygenation. J. Trauma, 42:
an update on brain tissue oxygenation. Curr. Opin. Crit. S32–S37.
Care, 12: 97–102. Valadka, A.B., Gopinath, S.P., Contant, C.F., Uzura, M. and
Rosner, M.J., Rosner, S.D. and Johnson, A.H. (1995) Cerebral Robertson, C.S. (1998) Relationship of brain tissue PO2 to
perfusion pressure: management protocol and clinical results. outcome after severe head injury. Crit. Care Med., 26:
J. Neurosurg., 83: 949–962. 1576–1581.
Rossi, S. and Stocchetti, N. (1999) Brain tissue oxygenation. J. van den Brink, W.A., Haitsma, I.K., Avezaat, C.J.,
Neurosurg., 91: 1065–1067. Houtsmuller, A.B., Kros, J.M. and Maas, A.I. (1998) Brain
Sarrafzadeh, A.S., Kiening, K.L., Bardt, T.F., Schneider, G.H., parenchyma/PO2 catheter interface: a histopathological
Unterberg, A.W. and Lanksch, W.R. (1998) Cerebral oxy- study in the rat. J. Neurotrauma, 15: 813–824.
genation in contusioned vs. nonlesioned brain tissue: moni- van den Brink, W.A., van Santbrink, H., Steyerberg, E.W.,
toring of PtiO2 with Licox and Paratrend. Acta Neurochir. Avezaat, C.J., Suazo, J.A., Hogesteeger, C., Jansen, W.J.,
Suppl., 71: 186–189. Kloos, L.M., Vermeulen, J. and Maas, A.I. (2000) Brain
216

oxygen tension in severe head injury. Neurosurgery, 46: Vespa, P.M. (2005) Multimodality monitoring and telemoni-
868–876; discussion 876–878. toring in neurocritical care: from microdialysis to robotic
van Santbrink, H., Maas, A.I. and Avezaat, C.J. (1996) telepresence. Curr. Opin. Crit. Care, 11: 133–138.
Continuous monitoring of partial pressure of brain tissue White, H. and Baker, A. (2002) Continuous jugular venous
oxygen in patients with severe head injury. Neurosurgery, 38: oximetry in the neurointensive care unit – a brief review:
21–31. [L’oxymetrie continue de la veine jugulaire a l’unite des soins
van Santbrink, H., vd Brink, W.A., Steyerberg, E.W., intensifs neurologiques – une breve revue]. Can. J. Anaesth.,
Carmona Suazo, J.A., Avezaat, C.J. and Maas, A.I. (2003) 49: 623–629.
Brain tissue oxygen response in severe traumatic brain Zauner, A., Doppenberg, E.M., Woodward, J.J., Choi, S.C.,
injury. Acta Neurochir. (Wien.), 145: 429–438; discussion Young, H.F. and Bullock, R. (1997) Continuous monitoring
438. of cerebral substrate delivery and clearance: initial experience
Vespa, P., Bergsneider, M., Hattori, N., Wu, H.M., Huang, S.C., in 24 patients with severe acute brain injuries. Neurosurgery,
Martin, N.A., Glenn, T.C., McArthur, D.L. and Hovda, D.A. 41: 1082–1091; discussion 1091–1093.
(2005) Metabolic crisis without brain ischemia is common after Zhi, D.S., Zhang, S. and Zhou, L.G. (1999) Continuous mon-
traumatic brain injury: a combined microdialysis and positron itoring of brain tissue oxygen pressure in patients with severe
emission tomography study. J. Cereb. Blood Flow Metab., head injury during moderate hypothermia. Surg. Neurol., 52:
25(6): 763–774. 393–396.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 15

Update on the treatment of spinal cord injury

Darryl C. Baptiste1 and Michael G. Fehlings1,2,

1
Division of Cell and Molecular Biology, Toronto Western Research Institute and Krembil Neuroscience Centre, Toronto
Western Hospital, Toronto, ON, Canada
2
Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University of Toronto, Toronto,
ON, Canada

Abstract: Acute spinal cord injury (SCI) is a devastating neurological disorder that can affect any indi-
vidual at a given instance. Current treatment options for SCI include the use of high dose methyl-
prednisolone sodium succinate, a corticosteroid, surgical interventions to stabilize and decompress the
spinal cord, intensive multisystem medical management, and rehabilitative care. While utility of these
therapeutic options provides modest benefits, there is a critical need to identify novel approaches to treat or
repair the injured spinal cord in hope to, at the very least, improve upon the patient’s quality of life.
Thankfully, several discoveries at the preclinical level are now transitioning into the clinical arena. These
include the Surgical Treatment for Acute Spinal Cord Injury Study (STASCIS) Trial to evaluate the role
and timing of surgical decompression for acute SCI, neuroprotection with the semisynthetic second gene-
ration tetracycline derivative, minocycline; aiding axonal conduction with the potassium channel blockers,
neuroregenerative/neuroprotective approaches with the Rho antagonist, Cethrins; the use of anti-NOGO
monoclonal antibodies to augment plasticity and regeneration; as well as cell-mediated repair with stem
cells, bone marrow stromal cells, and olfactory ensheathing cells. This review overviews the pathobiology of
SCI and current treatment choices before focusing the rest of the discussion on the variety of promising
neuroprotective and cell-based approaches that have recently moved, or are very close, to clinical testing.

Keywords: acute spinal cord injury; pathobiology; pharmacologic therapy; neuroprotection; rehabilitation;
cell-mediated repair

Introduction
modest financial incentive for the private sector to
make and market new medications, SCI can po-
Acute or traumatic spinal cord injury (SCI) occurs
tentially be considered an ‘‘orphan disease.’’ This is
with an annual incidence of 11.5–53.4 cases per
troubling since the epidemiological predominance
million within developed nations, with the causes of
for this debilitating neurological disorder tends to
these injuries ranging from motor vehicle accidents
affect individuals aged in their 3rd decade of life; a
and community violence to recreational activities
time typically associated with one’s prime earning
and workplace-related injuries (Sekhon and
potential (Sekhon and Fehlings, 2001). Coupled
Fehlings, 2001). SCI affects fewer than 200,000
with the necessity to provide lifelong health care
people in the United States and due to the perceived
support in the form of therapy and rehabilitation, a
significant economic strain is therefore placed on
Corresponding author. Tel.: +1 416 603 5229 (lab); +1 416 society by SCI. Moreover, therapies for SCI have
603 5800 ext. 2973 (office); Fax: +1 416 603 5745; a high likelihood of being translatable for other
E-mail: michael.fehlings@uhn.on.ca neurological indications such as stroke and

DOI: 10.1016/S0079-6123(06)61015-7 217


218

traumatic brain injury. Thus, we would argue that cord blood flow dynamics and local ischemia-
SCI presents excellent opportunities for commer- reperfusion. This initial trauma is then followed by
cialization of therapies, which target central nerv- ‘‘secondary’’ biochemical events that set the stage
ous system disorders characterized by cell death or for membrane dissolution and cellular death.
loss of key neural circuits or pathways. Some notable ‘‘secondary’’ etiological events
Since the published results from the landmark include disturbances to ionic homeostasis (i.e.,
National Acute Spinal Cord Injury Study (NASCIS) irregular regulation of cellular calcium, sodium,
II trial during the early 90 s, steroid use with and potassium trafficking), glutamate-mediated
methylprednisolone has become widely used in the excitotoxicity, mitochondrial instability, reactive
treatment of SCI (Bracken et al., 1990, 1992). How- oxygen species generation, elevated activation of
ever, with methylprednisolone only yielding modest proteolytic enzymes such as caspases and calpains,
improvements in neurological recovery coupled with and inflammation.
the potential risks of impaired wound healing and Cell death following SCI likely occurs as a con-
sepsis the utility of methylprednisolone has been tinuum of necrosis and apoptosis. While necrosis
seriously questioned (Coleman et al., 2000; Hurlbert, predominates immediately following the primary
2000). Moreover, findings from the Maryland traumatic episode, researchers have observed de-
monosialotetrahexosylganglioside sodium salt layed neuronal and oligodendroglial apoptosis, al-
(GM-1) clinical trial suggested that GM-1 had the beit the former occurs to a lesser degree, relative to
ability to enhance recovery of lower extremities, the latter (Casha et al., 2001). Applied neuropro-
which had provided strong rationale to further in- tective approaches have been tested in SCI models
vestigate the efficacy of low and high dose GM-1 with the intention that drug-mediated protective
following standard treatment with methylpredniso- effects will lead to preservation of neuronal and
lone in the Sygen multicenter study (Geisler et al., glial cell populations via targeting one or more of
1991). However, despite GM-1 demonstrating the aforementioned secondary injury events. How-
improved rates of recovery, along with improved ever, to date, few pharmacological approaches
bladder/bowel function, sacral sensation, and anal have been successful in translating therapeutic
contraction over the first 3 months post-injury the value in patients.
prospectively planned analysis at 6 months was neg-
ative (Geisler et al., 2001). Hence, with no univer-
sally accepted standard therapy in place, novel Current non-pharmacological treatment options for
therapeutic approaches for SCI are urgently re- SCI
quired. Surely, such a finding can only be made
through further understanding of the etiological Surgery
events following traumatic SCI. As such, we owe
much of our current state of knowledge regarding In SCI, primary mechanical forces together with
the etiology of SCI to those involved in basic sci- secondary injurious mechanisms collectively con-
entific research utilizing clinically relevant animal tribute to nervous tissue damage. Experimental
models of SCI, often involving extradural spinal evidence exists to support surgical decompression
cord contusion or compression in rodents (Rivlin following persistent compression of the spinal cord
and Tator, 1978; Gruner, 1992; Stokes et al., 1992; following SCI (Delamarter et al., 1995). However,
Joshi and Fehlings, 2002a, b). despite its widespread use in patients with SCI, the
role of surgery in improving neurological recovery
remains controversial because of the absence of
Pathobiological mechanisms of SCI randomized, controlled clinical trials. Further-
more, the presence and duration of a therapeutic
The pathobiology of SCI follows a biphasic proc- window, during which surgical decompression
ess, involving a ‘‘primary’’ traumatic insult to the could mitigate the secondary mechanisms of SCI
spinal cord that initiates a disruption in spinal have yet to be clearly defined.
219

The timing of surgical intervention in the treat- surgical management following SCI will only be
ment of experimental and clinical SCI was recently demonstrated clearly with a carefully designed
reviewed (Fehlings and Perrin, 2006). The authors prospective controlled trial to assess the optimum
conducted a MEDLINE search of the literature timing of decompressive surgery in SCI. These re-
from 1966 to 2005, dealing with the role of de- sults spurred the STASCIS to embark on a pro-
compression in SCI using the medical subject spective, multicenter study in North America to
heading of ‘‘decompression’’ and ‘‘spinal cord in- determine the effectiveness of early surgical de-
jury.’’ The overall conclusions from this extensive compression and stabilization procedures for re-
search revealed that decompressive surgery per- ducing the possibility of further damage to the
formed within the first 24 h of injury could be spinal cord following acute compression of the
conducted safely. It is the senior authors’ recom- cervical spinal cord. This study is currently under-
mendation that urgent decompression be per- way and is being converted into a collaborative
formed within the first 24 h of injury following trial under the auspice of the Spine Trauma Study
an isolated cervical SCI only if hemodynamic sta- Group (STSG).
bility can be maintained.
The Spinal Cord Injury Committee of the Joint
Section on Neurotrauma and Critical Care of the Rehabilitation
American Association of Neurological Surgeons
(AANS) and the Congress of Neurological Sur- Gait rehabilitation is a specific component of
geons (CNS) formed the Surgical Treatment for physical rehabilitation for persons with subacute
Acute Spinal Cord Injury Study (STASCIS) group or chronic motor-incomplete SCI. One novel
in 1992. By 1996, STASCIS was joined by the method of gait rehabilitation involves the use of
Joint Section on Spinal Disorders and Peripheral an overhead support point and a harness (i.e.,
Nerves of the AANS/CNS. The principal aim of body weight supported or BWS). The BWS strat-
the STASCIS group was to conduct a randomized egy has been combined with treadmill-based gait
prospective controlled trial to examine the role and training in recent studies with dramatic results
timing of decompressive surgery performed on the (Hicks et al., 2005). This combination of BWS and
spinal cord and cauda equina of patients after treadmill rationally evolved from the idea that
acute spinal injury. man possesses a central pattern generator, a cir-
A multicenter, retrospective study was per- cuitry located within the spinal cord capable of
formed in 36 North American centers to examine driving involuntary locomotor movements,
the use and timing of surgery in patients who have similar to lower mammals (Duysens and Van de
sustained SCI (Tator et al., 1999). The study was Crommert, 1998; Van de Crommert et al., 1998).
performed to obtain information required for the Indeed, an electrical train of stimuli applied over
planning of a randomized controlled trial in which the second lumbar segment with a frequency of
early and late decompressive surgery are com- 25–60 Hz and an amplitude of 5–9 V was shown to
pared. A total of 585 patients aged 16–75 years be effective in inducing rhythmic, alternating
with acute SCI or cauda equina injury were ad- stance and swing phases of the lower limbs in
mitted to participating centers, although approxi- paraplegic subjects, providing proof of a central
mately half of the patients were excluded due to pattern generator in humans (Dimitrijevic et al.,
late admission, age, gunshot wound, or absence of 1998). Thus, it is believed that the combination of
signs of compression on imaging. The timing of BWS and treadmill training will enhance the
surgery varied widely, where 23.5, 15.8, 19, and output by the intrinsic central pattern generator.
41.7% of the patients received surgery within 24, The effects of long-term BWS treadmill training
25–48, 48–96 h, and 5 days post-injury, respec- on functional walking ability and perceived quality
tively. The findings from this study have not led to of life were recently assessed in 14 persons with
unequivocal support for varied timing of surgical chronic incomplete SCIs (Hicks et al., 2005). Study
management in SCI patients. Thus, the benefit of subjects, classified on entrance as being ASIA
220

(American Spinal Injury Association) grade B or demyelinated axon cannot transmit motor or
C, participated in three weekly sessions of BWS sensory impulses. When an axon is demyelinated
treadmill training over the course of 1 year and after injury, large numbers of potassium channels
were assessed for improvements in functional abil- are exposed to the extracellular space and permit
ity and quality of life throughout the year and up potassium ion channels to open and release
to 8 months thereafter. The effect of this regimen potassium ions. Thus, the result of demyelinated
showed that all subjects required less body weight axons and potassium channel exposure is a
support, and were able to increase both walking reduced safety factor of action potential propaga-
speed and distance per session, as well as improved tion across the demyelinated region of the axon
over-ground walking ability. Heightened physical (Nashmi and Fehlings, 2001). As such, pharma-
ability was correlated with an enhanced perspec- cological antagonism of exposed potassium
tive of life. By the 8-month follow-up subjects channels on demyelinated axons with Fampri-
demonstrated diminished performance in treadmill dines (4-aminopyridine or 4-AP) had been
sessions with stable over-ground performance. hypothesized to restore the ability of axons to
Their quality of life reflected slight disappoint- transmit electrical impulses.
ment in their decreased physical function. Thus, In a randomized, double-blind, crossover de-
overall the BWS treadmill training appeared to be signed trial, where each patient received 4-AP and
a useful form of rehabilitation. a vehicle placebo on different occasions by infu-
However, the efficacy of BWS treadmill training sion over 2 h, separated by 2 weeks, 4-AP treat-
may not confer additional benefit over other forms ment demonstrated a significant temporary
of intensive rehabilitation therapies. Recently, the neurologic improvement in five of six patients
efficacy of step training with BWS treadmill train- with incomplete SCI. No effect was detected in
ing was compared with over-ground practice to the two cases of complete paraplegia and one of two
efficacy of a defined over-ground mobility therapy severe incomplete cases (Hansebout et al., 1993).
in 146 patients with incomplete SCI admitted for More recently, in a prospective, randomized,
inpatient rehabilitation in six regional centers double-blind, placebo-controlled, crossover trial
(Dobkin et al., 2006a). Following receipt of 12 15 chronic ambulatory SCI patients were ran-
weeks of BWS treadmill training or controlled domized to receive either an initial 2 weeks of
over-ground mobility training, primary outcome 40 mg/day, oral 4-AP medication of placebo or
variables of functional independence measures for immediate-release, 4-AP before being crossed over
locomotion or walking speed and distance for to an alternate medication for the next 2 weeks of
ASIA grade A and B SCI patients, or ASIA grade the study (DeForge et al., 2004). Evaluations for
C and D SCI patients, respectively, obtained at the the study were conducted at baseline before
6 month endpoint, did not support the expectation initiating treatment, 2 weeks and 4 weeks post-
that BWS treadmill training would be more effica- treatment through measurement of dynamometer
cious than intensive over-ground mobility therapy. lower limb isometric muscle force and biomechan-
ical gait assessments. Patients were also required to
give their subjective impressions of their received
Improving axonal conduction in the injured spinal therapy through a survey on exit of the study. The
cord results of the study demonstrated that despite
positive feelings towards the treatment no statis-
Fampridines tical differences were noted between placebo and
4-AP therapies.
Physical trauma to the spinal cord results in de- Information regarding the current status of Fa-
myelination of preserved spinal tracts. Without mpridines clinical testing for SCI is not yet pub-
insulating sheaths of myelin, these surviving axons lished, but can be found at the Acorda Therapeutics
become less efficient in their ability to transmit web site. In 2004, Acorda Therapeutics completed
electrical impulses to be conducted. As a result, the two Phase III clinical trials with their proprietary
221

Fampridines (Sustained Release) formulation. Nei- methylprednisolone sodium succinate (MPSS)


ther of these studies demonstrated statistical sig- consisting of 100 or 1000 mg bolus followed by
nificance in their primary endpoints, reduction of 25 or 250 mg MPSS daily thereafter for 10 days,
spasticity, as measured by the Ashworth score, and respectively (Bracken et al., 1984). However the
improvement of participant’s Subject Global Im- 6-month follow-up data for NASCIS I revealed no
pression rating. However, in one of the studies, the difference in neurological recovery of motor
data showed a positive trend toward improvement function or pinprick and light touch sensation
on the Ashworth score (http://www.acorda.com/ between the two treatment arms of the study.
pipeline_fampridine_sci1.asp). Moreover, increased fatality, wound infections
of both trauma and operative sites were associ-
ated to a greater extent with high-dose MPSS
Potassium and sodium channel blockade
(Bracken et al., 1984).
Further animal testing later generated data to
While the preclinical findings strongly support the
suggest that the NASCIS I MPSS dose used was
use of potassium channel blockade with 4-amino-
not sufficient to elicit notable therapeutic efficacy
pyridine to improve axonal conduction following
related to improved protection from ischemic
trauma-induced demyelination, the clinical utility
insults and calcium-dependent degradation of
of 4-aminopyridine is limited due to its associated
neurofilament cytoskeletal proteins (Braughler
adverse effects, which include restlessness, confu-
and Hall, 1984). Thus, the second NASCIS trial
sion, and infrequent generalized tonic-clonic sei-
was designed to evaluate the efficacy of high-dose
zure (Bever, 1994).
MPSS administered as a 30 mg/kg bolus over the
In an effort to minimize these unwanted side
first hour followed by an infusion of 5.4 mg/kg/h
effects Sanofi-Aventis has designed HP184 (N-[N-
over the next 23 h to placebo, and to a group of
propyl]-N-[3-fluoro-4-pyridinyl]-1H-3-methylindole-
patients receiving the opioid receptor antagonist
1-amine hydrochloride), a pharmacological blocker
naloxone (Bracken et al., 1990). The trial was fur-
capable of antagonizing both potassium and
ther designed to investigate the effects of differing
sodium channels. Sanofi-Aventis is currently in
times for drug administration following SCI. An
the process of conducting a Phase II, double-blind,
analysis of all patients who entered into the trial
placebo-controlled, multicenter study to assess the
failed to demonstrate a significant difference
efficacy and safety of HP184 at 100, 200, and
among the treatments. However, upon stratifica-
400 mg doses administered orally once daily for 24
tion of the data on the basis of time to loading
weeks in 18–65 year old patients with chronic SCIs
dose (less than 8 h vs. greater than 8 h from SCI)
classed ASIA grade C or D. Sanofi-Aventis will
and adjustment for the severity of SCI (complete
also assess the effect of HP184 treatment on
vs. incomplete), analysis of the results from this
recovery of walking function and spasticity.
second study at the 6-week and 6-month follow-up
revealed that patients treated with MPSS within
Neuroprotective/neuroregenerative approaches to the first 8 h of injury had significantly improved
treating the injured spinal cord motor and sensory function in comparison to pa-
tients receiving placebo, naloxone, or MPSS at
Methylprednisolone sodium succinate later times. Furthermore, these differences re-
mained significant 1 year following SCI (Bracken
Preclinical research originally identified methyl- et al., 1992). None of the differences in patients
prednisolone as a potent inhibitor of lipid peroxi- taking naloxone or in patients treated with
dation following traumatic injury (Hall and MPSS more than 8 h after SCI were statistically
Braughler, 1981, 1982). These findings led to the significant.
initiation of NASCIS I, which compared the effec- Despite the beneficial therapeutic effects demon-
tiveness of treating SCI patients with an adminis- strated with MPSS treatment, the results from the
tered intravenous (i.v.) standard or high dose NASCIS II trial have not been universally
222

accepted (Casha et al., 2001; Hurlbert, 2001; despite these concerns the NASCIS II and III trials
AANS/CNS, 2002). Concerns surrounding the effectively demonstrated the validity for targeting
small sample population size for the groups show- secondary injurious pathological events in SCI,
ing beneficial effects, performance of medical and while also emphasizing the importance of timing
surgical protocols by participating centers in a with applied interventions.
nonstandardized manner, and the fact that there
were no functional outcome measures defined to
assess whether the improvements noted with Monosialotetrahexosylganglioside
MPSS treatments were correlated with clinical sig-
nificance all reduced enthusiasm for the indication Monosialotetrahexosylganglioside GM-1 sodium
of MPSS for acute SCI. salt is a naturally occurring compound, which re-
In an attempt to resolve some of the issues sides within cell membranes of the mammalian
raised from the second NASCIS trial, a third CNS (Geisler et al., 2001). Several preclinical stud-
NASCIS trial was organized to include a func- ies reporting neuroprotective and neuroregenera-
tional independence measure (Bracken et al., tive actions by GM-1 in experimental models of
1997). A total of 499 patients suffering a SCI ischemia and injury have been reported (Agnati
within 8 h were enrolled into the study and re- et al., 1983; Fass and Ramirez, 1984; Bose et al.,
ceived 30 mg/kg bolus of MPSS before random- 1986). These findings have led to the initiation of
ization. Patients in the 24 h regimen group received the Maryland GM-1 Ganglioside Study, a small
a MPSS infusion of 5.4 mg/kg/h over 24 h, those in randomized, placebo-controlled, double-blind trial
the 48 h regimen group received a MPSS infusion designed to investigate the efficacy of GM-1 in
of 5.4 mg/kg/h for 48 h and those in the tirilazad patients with cervical and thoracic SCIs (Geisler
group received a 2.5 mg/kg bolus infusion of et al., 1991). In this trial, 34 patients completed the
tirilazad mesylate (TM; a ‘‘lazaroid’’ with inhibi- test-drug protocol that consisted of either 100 mg
tory effects on lipid peroxidation without gluco- GM-1 or placebo i.v. per day for 18–32 doses, with
corticoid side effects) every 6 h for 48 h. The results the first dose beginning within 72 h of the onset of
from this 3rd study concluded that no benefit was SCI. The results from this study demonstrated the
associated with extending MPSS treatment beyond effectiveness for GM-1 to improve the recovery of
24 h if MPSS had been administered within the neurologic function after 1 year and provided the
first 3 h of SCI, while those that started MPSS necessary rationale to support the recruitment of
therapy between 3 and 8 h demonstrated improve- SCI patients for a larger GM-1 study.
ments in motor capabilities if drug infusion was The randomized double-blind Sygens Multi-
continued for 48 h in comparison to 24 h infusion. center Acute Spinal Cord Injury Study was com-
It should be noted that the guidelines committee pleted in the late 1990s and reported in 2001. In
of the AANS/CNS Joint Section on Disorders of this trial, all 797 patients were randomized to re-
the Spine and Peripheral Nerves on reviewing the ceive a standard 30 mg/kg bolus of methyl-
evidence regarding the use of MPSS in the treat- prednisolone followed by 5.4 mg/kg/h infusion of
ment of acute SCI in adults concluded that its use methylprednisolone for 23 h within the first 8 h of
could only be supported at the level of a treatment injury. Placebo, low-dose GM-1 (300 mg loading
option (AANS/CNS, 2002). However, it is the au- dose followed by 100 mg/day for 56 days) or high-
thors’ view that the intense criticism directed at the dose GM-1 (600 mg loading dose followed by
NASCIS II and III trials must be balanced by the 200 mg/day for 56 days) was then randomly as-
current lack of alternative neuroprotective strate- signed following the completion of corticosteroid
gies for acute SCI. Moreover, the modest thera- administration to avoid unwanted drug interac-
peutic benefit demonstrated by MPSS may tions between methylprednisolone and GM-1.
potentially prove to impart a major benefit on The findings from this larger trial demonstrated
cervical SCI patient’s functional independence and that patients receiving either low- or high-dose
quality of life (Sekhon and Fehlings, 2001). Still, GM-1 had enhanced recovery of neurologic
223

function by 3 months post-injury regardless of biochemical monitoring in the acute setting of SCI;
their baseline severity, and showed a trend for im- to monitor the safety and tolerance of i.v. mino-
proved bowel/bladder function, sacral sensation, cycline administration; to assess systemic and cer-
and anal contraction (Geisler et al., 2001). How- ebral spinal fluid (CSF) absorption characteristics
ever, the primary outcome variable, the percentage of i.v. minocycline administration; and to assess
of marked recovery, at the designated 6 month serial CSF interleukin 1 beta (IL-1b) and nitric
time-point was negative. While no future clinical oxide concentration, which correspond to the ac-
trials with Sygen are currently planned, the spon- tivities of caspase-1 and -3, respectively, along with
soring company Fidia is evaluating this option tumor necrosis factor alpha (TNF-a) levels to
(Corporate Communications Fidia Farmaceutici reflect the levels of a major pro-inflammatory
SpA, Personal communications). cytokine.

Minocycline Cethrins

The broad-spectrum antibiotic, minocycline, is a CNS myelin inhibits axon growth due to the ex-
lipophilic derivative of tetracycline that has demon- pression of several extracellular growth-inhibitory
strated ability to provide neuroprotection via intra- proteins, including myelin-associated glycoprotein
peritoneal (Wells et al., 2003; Teng et al., 2004) or (MAG), myelin oligodendrocyte glycoprotein
i.v. (Xu et al., 2004) routes of administration. (MOG), and neurite outgrowth (Nogo). It is now
Mechanisms attributed to the protective actions evident that the growth-inhibitory effects of these
elicited by minocycline include ability to overcome proteins are exerted through the regulatory actions
glutamate-mediated excitotoxicity (Tikka and of small intracellular GTPase-associated signaling
Koistinaho, 2001; Baptiste et al., 2004), anti- proteins, Rho and Rac (Niederost et al., 2002;
inflammatory effects through blocking the activa- Winton et al., 2002). Rho and Rac can exist in one
tion of microglial cells (Dommergues et al., 2003; of two possible forms — an active GTP-bound
Baptiste et al., 2005), inhibiting both cytochrome c state, and an inactive GTP-bound state. In the ac-
release and caspase-dependent apoptotic neuronal tive conformation, these small GTPase-associated
death (Zhu et al., 2002), as well as a capacity to proteins work to inhibit neurite outgrowth. For
antagonize the activities of matrix metal- example, purified cerebellar granule cells grown on
loproteinases-2 (Cho et al., 2006) and -9 (Brundula Nogo-A, Nogo-66, or MAG containing substrates
et al., 2002). Additionally, minocycline was shown have all been shown to result in inhibited neurite
to reduce oligodendrocyte apoptosis, corticospinal outgrowth (Niederost et al., 2002). Furthermore,
tract dieback, and lesion size, while improving increased levels of Rho-GTP were detected in
functional outcome following a C7–C8 dorsal col- pheochromocytoma cell line cultures that had been
umn transection SCI in rats (Stirling et al., 2004). plated on myelin, MAG, or poly-L-lysine contain-
Considering, these findings together with the rel- ing substrates by measuring the precipitate from
atively safe clinical track record of minocycline has cell homogenates mixed with Rho binding domain
made this pharmacological approach a very prom- isolated from rhotekin, a GTP-interacting protein
ising candidate for the SCI. (Dubreuil et al., 2003). Moreover, MAG, MOG,
A Phase I/II pilot study to investigate the effi- or Nogo-mediated neurite outgrowth inhibition
cacy of intravenously administered minocycline in can be relieved with the addition of Rho-A-
patients with acute SCI is currently underway in associated kinase ROCK inhibitor (Niederost
Calgary, Alberta. The study has aimed to enroll et al., 2002).
patients above 16 years of age possessing a Preclinical studies now support the hypothesis
SCI between C1 and T11. The major objectives that SCI is associated with an upregulation of
of this trial will be to assess the feasibility of GTP-bound Rho. Dubreuil et al. (2003) demon-
both minocycline administration and clinical and strated that Rho-GTP levels are robustly elevated
224

following transection or contusion cord injuries in can be encouraged further upon manipulation of
comparison to uninjured control animals. Rho ac- the local tissue microenvironment.
tivation following transection-induced injury be- Years later, Caroni et al. (1988) reported that
gan as early as 1.5 h post-SCI and was sustained nonpermissive substrates within the CNS were
for 7 days (Dubreuil et al., 2003). Addition of the actually associated with oligodendrocytes and my-
Rho-selective antagonist, C3–05 transferase, in- elin. Moreover, monoclonal antibody or pharma-
jected in a fibrin matrix to the lesion site following cological treatment directed against white matter
cord transection, or C3–05 alone into the contused nonpermissive substrates allowed neuron out-
cord demonstrated ability to reduce RhoA activa- growth (Caroni and Schwab, 1988; Savio and
tion levels back to physiological conditions Schwab, 1989). By 2000, the gene nogo, which can
(Dubreuil et al., 2003). Also, treatment with be transcribed to yield Nogo-A, -B, and -C mRNA
C3–05 reduced neuronal and glial cell apoptosis products, was cloned (Chen et al., 2000). Nogo-A
that would pathologically occur in secondary in- is a high molecular weight transmembrane protein
jury processes of SCI (Dubreuil et al., 2003). Thus, and potent inhibitor of neurite outgrowth pro-
this novel approach has demonstrated multifac- duced by oligodendrocytes. Inhibition of Nogo-A
eted therapeutic actions, namely neuroregenerative with the selective monoclonal antibody IN-1 or
and neuroprotective properties. with the peptide antagonist NEP1-40, in spinally
BioAxone Inc., a biopharmaceutical company injured rats demonstrates neutralization of myelin-
based in Montreal, Quebec, recently sponsored a dependent neurite outgrowth blockade, improved
Phase I/IIa multinational clinical trial (lead site in axonal regeneration, and enhanced rates of func-
Toronto, Ontario) designed to evaluate the safety tional locomotor recovery (Chen et al., 2000; Li
and pharmacokinetics of rising dosages of Cethrin and Strittmatter, 2003).
(BA-210), a recombinant fusion protein composed Novartis Institutes for BioMedical Research has
of C3 transferase and a transport sequence that since taken a focused interest in developing anta-
aids the protein’s ability to breach cellular mem- gonists for Nogo-A and its corresponding recep-
branes together with a fibrin sealant, following a tor, Nogo-66, to promote neuroregeneration
single extradural administration during surgery for (Wiessner et al., 2003; Walmsley et al., 2004).
acute thoracic and cervical SCI. The trial was Currently, Novartis is sponsoring a Phase I clinical
conducted in nine centers located in Canada and trial to evaluate the safety of anti-Nogo-A mono-
the USA. In December 2005, the U.S. Food & clonal antibody therapy.
Drug Administration granted Orphan Drug Status
to Cethrins, which will likely hasten clinical eval-
uation of BioAxone’s lead product through reduc- Autologous macrophages
ing clinical development costs and facilitating
regulatory filings in other countries. It is antici- The CNS was once considered as an ‘‘immune-
pated that the results of this Phase I/IIa trial will privileged’’ environment that was neither suscepti-
become available in early 2007. ble to nor could evoke an inflammatory response
(Lucas et al., 2006). Consequently, the hypothesis
that inflammation may be harnessed to heal the
Nogo injured spinal cord has not been received without
skepticism. Although evidence exists to support a
It has long been thought that the capacity for the pathological role for inflammation in the etiology
CNS to repair lesions through regeneration was of many CNS disorders (Stoy et al., 2005; Man
limited. However, hope in this context has been et al., 2007; Miklossy et al., 2006; Noorbakhsh
invigorated since the landmark discoveries by et al., 2006), it is very likely that inflammatory me-
Richardson et al. (1980), which have provided diators possess dual roles, with detrimental acute
the first indication that CNS neurons possess an effects together with healing effects in longer-term
intrinsic ability to regenerate following injury that repair and recovery (Lucas et al., 2006).
225

Preclinical investigations involving rats that had cord lesion. The results from this study at 1-year
received a single complete spinal cord transection follow-up demonstrated that the therapy was safe
together with local implantation of ex vivo- and warranted further investigation in a randomi-
activated homologous macrophages incubated zed Phase II, multicenter clinical trial within the
with homologous peripheral nerves demonstrated USA and Israel. However, despite the FDA grant-
that stimulation of the host immune response can ing an orphan drug status to ProCord on Septem-
promote partial recovery of motor function, as ber 13, 2004, recruitment into this trial is currently
demonstrated through hind limb movement, plan- suspended due to presumed financial challenges
tar paw placement, and weight-bearing steps, related to the conduct of such a large complex
along with electrophysiological assessments via trial. Clinical follow-up is currently being con-
cortically evoked hind limb muscle response ducted for those patients that have already been
(Rapalino et al., 1998). Additionally, macrophag- admitted.
es incubated with autologous skin were found to
be just as efficacious (Rapalino et al., 1998). The
mechanism of action associated with autologous Cell-mediated repair of the injured spinal cord
ex vivo-activated macrophages is believed to be
both neuroprotective and neuroregenerative. Ne- Neural stem and progenitor cells
uroprotective properties likely arise due to the
ability of activated macrophages to synthesize and A variety of cell types have been investigated for
secrete concurrently protective cytokines IL-1b their potential use in cell-mediated tissue repair
and IL-6, together with the trophic factor, brain following SCI. Theses include neural stem cells
derived neurotrophic factor, and the chemokine, (NSCs), neural progenitor cells (NPCs), Schwann
IL-8, while reducing levels of the neurotoxic cyto- cells, oligodendroglial precursor cells (OPCs),
kine, TNF-a. In addition, regenerative properties bone marrow stem cells (BMSCs), and olfactory
may result due to macrophage-mediated phagocy- ensheathing cells (OECs).
tosis of myelin debris, an event that would effec- NSCs and NPCs have functional differences,
tively make the environment of the injured spinal where the former represents an uncommitted cell
cord less cytotoxic and more responsive to axonal type that can divide repeatedly while maintaining
regeneration (Bomstein et al., 2003). potency to generate differentiated cell types (neu-
From 2000 to 2003, Proneuron Biotechnologies rons and glia), whereas the latter represents a kind
conducted a Phase I, open-label nonrandomized of cell that exhibits a limited number of cell divi-
safety study in patients diagnosed with a complete sions before it differentiates into a specific cell type
SCI (ASIA grade A) within C5–T11 within 14 days (Pollard et al., 2006). Despite the fact that stem/
of injury at centers located in Israel and Belgium precursor cells may be derived from a variety of
(Knoller et al., 2005). ASIA grade A patients sources, research with these cells raises both po-
were the only SCI population admitted into the litical and ethical concerns. As a result, stem cell
ProCords study, in an effort to limit the con- technology is trapped within an ethical dilemma of
founding interpretation of spontaneous recovery balancing heightened emotions regarding embryo/
that would result from incomplete injuries (ASIA fetal research with the potentially therapeutic
grades B, C, or D). benefits to patients with desperate hopes.
Proneuron Biotechnologies’ proprietary proce- NSCs have been isolated from the adult human
dure for obtaining activated autologous macroph- hippocampus and lateral ventricle wall (Johansson
ages consists of isolating white lymphocytes from et al., 1999b). Additionally, postmortem analysis
the patient’s blood and incubating them ex vivo of tissue derived from the hippocampus and sub-
with autologous dermis. A single dose of 4 million ventricular zone of the caudate nucleus revealed
activated autologous macrophages was adminis- that NPCs reside in the adult human brain
tered by four microinjections into the spinal cord (Eriksson et al., 1998). Thus, these findings show
parenchyma at the caudal border of the spinal that neurogenesis occurs throughout life and
226

supports the hypothesis that the mature CNS pos- may act as a physical bridge, allowing the normal
sesses an innate ability to repair damaged tissue. In functioning of undamaged or regenerated axons.
light of these findings researchers have attempted Moreover, these cells are capable of secreting nu-
to demonstrate the utility harnessing the capabil- merous trophic factors including brain derived ne-
ities of endogenous stem/progenitor cells following urotrophic factor, glial derived neurotrophic
traumatic injuries (Johansson et al., 1999a; Kernie factor, nerve growth factor, and ciliary neurotro-
et al., 2001). However, the results of these studies phic factor (Pellitteri et al., 2006), while also being
demonstrated that the endogenous stem/progeni- able to remyelinate (Kohama et al., 2001) and
tor supply tend to move toward an astrocytic lin- guide regenerating CNS axons to their intended
eage (Johansson et al., 1999a; Kernie et al., 2001). tracts (Zheng and Kuffler, 2000). The transplant
Studies involving transplantation of NSCs/ procedure would involve removing a small amount
NPCs have shown greater promise for traumatic of the patient’s own peripheral nerve tissue, iso-
injuries than attempts to stimulate or control en- lating Schwann cells from the tissue, growing them
dogenous neurogenesis. For instance Cummings in plastic culture dishes in an incubator, and then
et al. (2005) demonstrated that human CNS stem implanting the cultured Schwann cells into the site
cells grown as neurospheres survive, migrate, and of SCI.
express differentiation markers for neurons and A recent report has demonstrated the efficacy of
oligodendrocytes after long-term engraftment in subarachnoid space transplantation of Schwann
the injured spinal cord. Furthermore, the stem cell cells into the contused epicenter in adult rats 7
engraftment was associated with improved loco- days post-SCI resulting in significantly enhanced
motor recovery with diminished astrocytic differ- locomotor recovery and significantly greater ax-
entiation, remyelination in both the spinally onal density at the lesion epicenter (Firouzi et al.,
injured and myelin-deficient mice, along with elec- 2006). Furthermore, this route for transplantation
tron microscope evidence consistent with synapse was without notable reduction in tissue integrity or
formation between stem cells and host neurons augmented levels of pro-inflammatory and pro-
(Cummings et al., 2005). However, despite signifi- apoptotic mediators (Firouzi et al., 2006). How-
cant advances in this, adult NSCs remain an elu- ever, in light of these advances with Schwann cells,
sive cell for study, and researchers are facing many researchers have still not found a solution to per-
challenges to the development of therapeutic ap- mit regenerating axons to extend beyond the
plications from adult NSC research. Among these transplant due to the inhibitory nature of the sur-
challenges are the identification and characteriza- rounding glial scar (Oudega and Xu, 2006). This
tion of NSCs, the understanding of the physiology problem likely calls for a combinatorial approach,
of newly generated neuronal cells in the adult which makes the local microenvironment more re-
brain, and the isolation and culture of homoge- ceptive for regeneration.
nous populations of NSCs/NPCs from the adult
CNS for cell-based therapy. Furthermore, meas-
ures must be taken to improve on the survival of Bone marrow stromal cells
transplanted stem/progenitor cells in the hostile
environment of the injured spinal cord (Karimi- Bone marrow stromal cells (BMSCs) provide yet
Abdolrezaee et al., 2006). another source for obtaining pluripotent stem
cells. Bone marrow contains a population of pluri-
potent cells that have the capacity to migrate to-
Schwann cells wards a lesion and release various growth factors
to facilitate regeneration and lesion repair. Under
Schwann cells are a type of glial cell responsible physiological settings BMSCs function to regulate
for forming myelin sheaths around peripheral hematopoiesis; however, if these cells are grown
nerves. Researchers are excited about the use of away from their natural environment these cells
Schwann cells in neural repair because these cells can be readily and effectively propagated and
227

manipulated genetically to yield a variety of cell administration and a group of acute SCI patients
types including phenotypes that closely resemble (10–30 days post-SCI) against patients sustaining
neurons (Clark and Keating, 1995). The advantage chronic SCIs. Motor evoked potential, somato-
of using bone marrow as a source for stem cells is sensory evoked potential, magnetic resonance im-
that they are relatively easy to isolate, the cells aging, and ASIA scores were used in patient
grow well in tissue culture, BMSCs may be used in follow-up. The results of this study demonstrated
autologous transplantation protocols, and these that the BMSC-mediated repair is associated with
BMSCs have already received approval for modest improvements in acute patients and that
hematopoietic diseases (Syková et al., 2006). BMSC transplantation is a relatively safe proce-
Hofstetter et al. (2002) differentiated BMSCs in dure. Thus, it is anticipated that a Phase II clinical
vitro into neuronal-like cells and investigated their trial designed to test the efficacy will be initiated in
fate following immediate or delayed injection into the near future.
the contused spinal cord of adult rats. Their find-
ings demonstrated that BMSCs administered 1-
week post-SCI had better rates of survival than Oligodendrocyte precursor cells
BMSCs administered at the time of injury. Fur-
thermore, the delayed treatment was associated Since the neurological disability in SCI is associ-
with significantly improved locomotor function. ated with oligodendrocyte death, resulting in de-
Despite their success in improving neurological myelination and axonal degeneration, one would
recovery rates, electrophysiological and anticipate that OPC-mediated repair would pro-
immunohistochemical analysis of these differenti- duce significant improvements in neural recovery.
ated BMSCs demonstrated that the transplanted In preclinical research OPC transplant alone or in
cells were likely rather immature, which lacked combination with the glycoprotein molecule Sonic
functional sodium and potassium channels. Still hedgehog (Shh), to further drive OPCs toward an
the effectiveness of this approach administered oligodendroglial lineage, was administered to
within a clinically relevant therapeutic window of adult rats 5 days following a moderate thoracic
opportunity makes BMSCs a promising strategy SCI (Bambakidis and Miller, 2004). The results of
for SCI. To date, two clinical trials have been this treatment regimen demonstrated that trans-
preformed in patients with SCI. plantation of OPCs improves axonal conduction
In South Korea, Park et al. (2005) evaluated the and spinal cord function in the injured spinal cord.
therapeutic efficacy of combining autologous Furthermore, the therapeutic benefits were more
BMSC transplantation, administered directly into pronounced when OPC treatment was given in
the spinal cord lesion site, with granulocyte ma- combination with Shh, while treatment of injured
crophage-colony stimulating factor (GM-CSF), rats with Shh on its own resulted in the prolifer-
given subcutaneously, in six patients with com- ation of an endogenous population of neural pre-
plete SCIs. At the 6- and 18-month follow-up pe- cursor cells (Bambakidis and Miller, 2004).
riods, four of the six patients showed neurological The most frequently investigated OPC is O-2A,
improvements by two ASIA grade (from ASIA A which can differentiate into oligodendroglial cells
to ASIA C), while another improved from ASIA A and type-2 astrocytes in vitro, but only into
to ASIA B. Moreover, BMSC transplantation to- oligodendrocytes following in vivo transplantation
gether with GM-CSF was not associated with in- into brain or spinal cord (Cao et al., 2002). O-2A
creased morbidity or mortality. cell transplant demonstrates ample capacity to
The second clinical trial, which was conducted migrate to regions adjacent to the site of in-
in Prague, Czech Republic, is a Phase I study that jury, differentiate into oligodendrocytes, remyelin-
has enrolled 20 patients with transversal SCIs to ate axons, and improve on functional, behavioral,
test the safety of autologous bone marrow cell electrophysiological, and morphological meas-
implantation (Syková et al., 2006). The investiga- urable outcome variables (Lee et al., 2005).
tors compared intra-arterial versus i.v. routes of Still, a major shortcoming associated with
228

O-2A-mediated repair is their relatively slow pro- (T4-T10) spinal cord, 6 months to 3 years post-
liferation rates, which effectively limits their ability injury (Feron et al., 2005). Only eight patients were
to self-renewal. admitted into the trial (four patients to receive OEC
transplantation and four sham-operated controls).
All participants were closely monitored before the
Olfactory ensheathing cells trial was started and for 3 years following the trial
admittance. All patients underwent regular MRI
OECs are specialized glial cells that surround the examination, along with physical, psychiatric,
olfactory sensory axons of the first cranial nerve neurological, and neurophysiological assessments
within the nose, which possess neuroregenerative by medical staff and physiotherapy and occupa-
properties akin to Schwann cells of the peripheral tional therapy assessments by respective therapists.
nervous system (Doucette, 1995). OECs are suit- By far the largest of clinical trials testing the
able candidates for cell-mediated repair following efficacy of OEC transplants to treat SCI patients
SCI because this cell type can support axonal comes from Beijing, China, where 171 subjects
outgrowth and regrowth through the adult CNS; have participated (Huang et al., 2003). Patients
they are able to remyelinate the axons that have aged 2–64 years, had traumatic or acute compress-
undergone primary demyelination; and they can ive injuries due to epidural hematoma. Outcome
migrate within the CNS and coexisting in an variables included motor score, light touch, and
astrocyte-rich environment and therefore fully pin prick scores according to the International
integrating within a lesion (Barnett, 2004). This Standards for Neurological and Functional Clas-
latter characteristic of OECs to integrate with sification of SCI by ASIA and the International
astrocytes sets them apart from Schwann cells. Society of Paraplegia before and 2 and 8 weeks
There are now several reports documenting the post-intervention. The conclusions made from this
robust therapeutic efficacy of OEC transplantation study were that OEC transplantation can partially
in animal models of SCI. For instance, Li et al. preserve spinal functions regardless of patient age.
(1998) have demonstrated that cultured OECs in- However, the recent publication by Dobkin et al.
jected into focal lesions within the adult rat cor- (2006b) warns the SCI community of the proce-
tical spinal tract begin to regenerate within the first dure of OEC transplantation as performed by Dr.
week following transplantation. Moreover, the cut Huang as not meeting international standards of a
corticospinal axons extended caudally and en- Phase I safety or Phase II efficacy trial. Consid-
sheathed by P0-positive peripheral myelin had ac- ering the rather poorly defined inclusion and ex-
cumulated into parallel bundles, which clusion criteria, coupled with the inconsistencies in
appropriately extended across the full length of OEC injection sites, a lack of observed functional
the lesioned area and reentered the caudal part of benefit, and perioperative morbidity caution must
the host corticospinal tract (Li et al., 1998). be taken when assessing these anecdotal claims.
Ramon-Cueto et al. (2000) showed that OEC
transplantation can provide improved rates of
functional and structural recovery following a Conclusion
complete spinal cord transection from 3 to 7
months post-transplant and corticospinal, raphe- In addition to methylprednisolone, GM-1 ganglio-
spinal, and coeruleospinal tracts all regenerated side, and Fampridine a number of novel pharma-
for long distances within caudal cord stumps. cological and cell-based approaches appear to be
With these and other findings promoting the on the horizon for the therapeutic management of
utility for OECs to be used in neural repair follow- SCI (Table 1). Considering the complexity and
ing SCI clinical trials have arisen around the world. multifactorial nature of pathobiological mecha-
A Phase I clinical trial was initiated in 2001, in nisms that contribute to SCI, it is very likely that
Brisbane, Australia, to investigate the safety of combinatorial approaches will have the greatest
OEC transplantation into the injured thoracic opportunity to improve on the quality of life of
229

Table 1. Select pharmacological and cell-based therapies for spinal cord injury

Approach Proposed mechanism of action

Methylprednisolone A glucocorticosteroid that has demonstrated ability to inhibit posttraumatic lipid peroxidation and
inflammation in animal models of spinal cord injury (SCI), and has shown ability to improve
neurological recovery in spinal cord injured humans

GM-1 ganglioside A naturally occurring compound, residing within cell membranes of the mammalian CNS that has been
shown to promote neuritic sprouting, and has demonstrated ability to oppose excitotoxicity and
apoptosis
Minocycline A second generation tetracycline derivative with demonstrated ability to inhibit excitotoxicity, oxidative
stress, microglial cell activation, and caspase-dependent and caspase-independent-mediated pathways of
neuronal death
Fampridines A specific voltage-dependent potassium channel blocker shown to restore action potential conduction,
and enhance synaptic transmission in damaged demyelinated nerve fibers via blockade of exposed
potassium channels within the internodal membrane of injured axons

HP 184 A use- and frequency-dependent antagonist of sodium channels, and voltage-dependent blocker of
potassium channels designed to have less associated adverse effects than Fampridines
Cethrins A Rho antagonist, administered directly into the injured spinal cord in combination with a fibrin sealant
for improved drug delivery, to allow reversal of abnormal Rho activation in neurons and glial cells, and
promotion of neuroprotection and neuroregeneration following acute SCI
Anti-Nogo-A A monoclonal antibody targeted at Nogo-A to neutralize neurite outgrowth inhibition, and allow
monoclonal antibodies enhanced regeneration, compensatory sprouting, structural reorganization, plasticity, and functional
recovery following SCI
Procords Autologous macrophages, activated through co-incubation with regenerative autologous skin tissue, are
implanted into the injured spinal cord to elevate the secretion of protective cytokines, interleukin-1 beta
(IL-1b) and IL-6, the trophic factor, brain derived neurotrophic factor (BDNF), and the chemokine IL-
8, while reducing the secretion of the proinflammatory cytokine, tumor necrosis factor alpha (TNF-a);
all of which helps to make the injured environment of the lesioned spinal cord more feasible for axonal
regeneration, reduce cyst formation, and improve motor recovery

spinally injured individuals. Moreover, preclinical Agnati, L.F., Fuxe, K., Calza, L., Benfenati, F., Cavicchioli, L.,
analysis view SCI in a larger view with a thorough Toffano, G. and Goldstein, M. (1983) Gangliosides increase
examination of potentially beneficial effects in the survival of lesioned nigral dopamine neurons and favour
the recovery of dopaminergic synaptic function in striatum of
addition to ambulation. For example, neuropro- rats by collateral sprouting. Acta Physiol. Scand., 119:
tective/regenerative approaches that yield modest 347–363.
effects in enhancing rates of locomotor recovery Bambakidis, N.C. and Miller, R.H. (2004) Transplantation of
would be of bladder/bowel function and relieve oligodendrocyte precursors and sonic hedgehog results in
chronic pain. With regard to cell-based ap- improved function and white matter sparing in the spinal
cords of adult rats after contusion. Spine J., 4: 16–26.
proaches, in addition to political and religious Baptiste, D.C., Hartwick, A.T., Jollimore, C.A., Baldridge,
conflicts, it is clear that careful planning and W.H., Seigel, G.M. and Kelly, M.E. (2004) An investigation
design is required to obtain meaningful data. of the neuroprotective effects of tetracycline derivatives in
experimental models of retinal cell death. Mol. Pharmacol.,
66: 1113–1122.
Baptiste, D.C., Powell, K.J., Jollimore, C.A., Hamilton, C.,
References LeVatte, T.L., Archibald, M.L., Chauhan, B.C., Robertson,
G.S. and Kelly, M.E. (2005) Effects of minocycline and
AANS/CNS. (2002) Pharmacological therapy after acute cer- tetracycline on retinal ganglion cell survival after axotomy.
vical spinal cord injury. Neurosurgery, 50: S63–S72. Neuroscience, 134: 575–582.
230

Barnett, S.C. (2004) Olfactory ensheathing cells: unique glial nonpermissive substrate properties of CNS white matter.
cell types? J. Neurotrauma, 21: 375–382. Neuron, 1: 85–96.
Bever Jr., C.T. (1994) The current status of studies of amino- Casha, S., Yu, W.R. and Fehlings, M.G. (2001) Oligoden-
pyridines in patients with multiple sclerosis. Ann. Neurol., droglial apoptosis occurs along degenerating axons and is
36(Suppl): S118–S121. associated with FAS and p75 expression following spinal
Bomstein, Y., Marder, J.B., Vitner, K., Smirnov, I., Lisaey, G., cord injury in the rat. Neuroscience, 103: 203–218.
Butovsky, O., Fulga, V. and Yoles, E. (2003) Features of Chen, M.S., Huber, A.B., van der Haar, M.E., Frank, M.,
skin-coincubated macrophages that promote recovery from Schnell, L., Spillmann, A.A., Christ, F. and Schwab, M.E.
spinal cord injury. J. Neuroimmunol., 142: 10–16. (2000) Nogo-A is a myelin-associated neurite outgrowth in-
Bose, B., Osterholm, J.L. and Kalia, M. (1986) Ganglioside- hibitor and an antigen for monoclonal antibody IN-1. Na-
induced regeneration and reestablishment of axonal continu- ture, 403: 434–439.
ity in spinal cord-transected rats. Neurosci. Lett., 63: 165–169. Cho, K.O., La, H.O., Cho, Y.J., Sung, K.W. and Kim, S.Y.
Bracken, M.B., Collins, W.F., Freeman, D.F., Shepard, M.J., (2006) Minocycline attenuates white matter damage in a rat
Wagner, F.W., Silten, R.M., Hellenbrand, K.G., Ransohoff, model of chronic cerebral hypoperfusion. J. Neurosci. Res.,
J., Hunt, W.E., Perot Jr., P.L., et al. (1984) Efficacy of me- 83: 285–291.
thylprednisolone in acute spinal cord injury. JAMA, 251: Clark, B.R. and Keating, A. (1995) Biology of bone marrow
45–52. stroma. Ann. N.Y. Acad. Sci., 770: 70–78.
Bracken, M.B., Shepard, M.J., Collins Jr., W.F., Holford, T.R., Coleman, W.P., Benzel, D., Cahill, D.W., Ducker, T., Geisler,
Baskin, D.S., Eisenberg, H.M., Flamm, E., Leo-Summers, F., Green, B., Gropper, M.R., Goffin, J., Madsen III, P.W.,
L., Maroon, J.C., Marshall, L.F., et al. (1992) Methyl- Maiman, D.J., Ondra, S.L., Rosner, M., Sasso, R.C., Trost,
prednisolone or naloxone treatment after acute spinal cord G.R. and Zeidman, S. (2000) A critical appraisal of the re-
injury: 1-year follow-up data. Results of the second national porting of the national acute spinal cord injury studies (II
acute spinal cord injury study. J. Neurosurg., 76: 23–31. and III) of methylprednisolone in acute spinal cord injury. J.
Bracken, M.B., Shepard, M.J., Collins, W.F., Holford, T.R., Spinal Disord., 13: 185–199.
Young, W., Baskin, D.S., Eisenberg, H.M., Flamm, E., Leo- Cummings, B.J., Uchida, N., Tamaki, S.J., Salazar, D.L.,
Summers, L., Maroon, J., et al. (1990) A randomized, con- Hooshmand, M., Summers, R., Gage, F.H. and Anderson,
trolled trial of methylprednisolone or naloxone in the treat- A.J. (2005) Human neural stem cells differentiate and pro-
ment of acute spinal-cord injury. Results of the second mote locomotor recovery in spinal cord-injured mice. Proc.
national acute spinal cord injury study. N. Engl. J. Med., 322: Natl. Acad. Sci., U.S.A., 102: 14069–14074.
1405–1411. DeForge, D., Nymark, J., Lemaire, E., Gardner, S., Hunt, M.,
Bracken, M.B., Shepard, M.J., Holford, T.R., Leo-Summers, Martel, L., Curran, D. and Barbeau, H. (2004) Effect of 4-
L., Aldrich, E.F., Fazl, M., Fehlings, M., Herr, D.L., Hitc- aminopyridine on gait in ambulatory spinal cord injuries: a
hon, P.W., Marshall, L.F., Nockels, R.P., Pascale, V., Perot double-blind, placebo-controlled, crossover trial. Spinal
Jr., P.L., Piepmeier, J., Sonntag, V.K., Wagner, F., Cord, 42: 674–685.
Wilberger, J.E., Winn, H.R. and Young, W. (1997) Admini- Delamarter, R.B., Sherman, J. and Carr, J.B. (1995) Patho-
stration of methylprednisolone for 24 or 48 hours or tirilazad physiology of spinal cord injury. Recovery after immediate
mesylate for 48 hours in the treatment of acute spinal cord and delayed decompression. J. Bone Joint Surg. Am., 77:
injury. Results of the third national acute spinal cord injury 1042–1049.
randomized controlled trial. National acute spinal cord in- Dimitrijevic, M.R., Gerasimenko, Y. and Pinter, M.M. (1998)
jury study. JAMA, 277: 1597–1604. Evidence for a spinal central pattern generator in humans.
Braughler, J.M. and Hall, E.D. (1984) Effects of multi-dose Ann. N.Y. Acad. Sci., 860: 360–376.
methylprednisolone sodium succinate administration on in- Dobkin, B., Apple, D., Barbeau, H., Basso, M., Behrman, A.,
jured cat spinal cord neurofilament degradation and energy DeForge, D., Ditunno, J., Dudley, G., Elashoff, R., Fugate,
metabolism. J. Neurosurg., 61: 290–295. L., Harkema, S., Saulino, M. and Scott, M. (2006a) Weight-
Brundula, V., Rewcastle, N.B., Metz, L.M., Bernard, C.C. and supported treadmill vs. over-ground training for walking
Yong, V.W. (2002) Targeting leukocyte MMPs and transmi- after acute incomplete SCI. Neurology, 66: 484–493.
gration: minocycline as a potential therapy for multiple scle- Dobkin, B.H., Curt, A. and Guest, J. (2006b) Cellular trans-
rosis. Brain, 125: 1297–1308. plants in China: observational study from the largest human
Cao, Q., Benton, R.L. and Whittemore, S.R. (2002) Stem cell experiment in chronic spinal cord injury. Neurorehabil.
repair of central nervous system injury. J. Neurosci. Res., 68: Neural Repair, 20: 5–13.
501–510. Dommergues, M.A., Plaisant, F., Verney, C. and Gressens, P.
Caroni, P., Savio, T. and Schwab, M.E. (1988) Central nervous (2003) Early microglial activation following neonatal excito-
system regeneration: oligodendrocytes and myelin as non- toxic brain damage in mice: a potential target for neuropro-
permissive substrates for neurite growth. Prog. Brain Res., tection. Neuroscience, 121: 619–628.
78: 363–370. Doucette, R. (1995) Olfactory ensheathing cells: potential for
Caroni, P. and Schwab, M.E. (1988) Antibody against glial cell transplantation into areas of CNS injury. Histol.
myelin-associated inhibitor of neurite growth neutralizes Histopathol., 10: 503–507.
231

Dubreuil, C.I., Winton, M.J. and McKerracher, L. (2003) Rho Huang, H., Chen, L., Wang, H., Xiu, B., Li, B., Wang, R.,
activation patterns after spinal cord injury and the role of Zhang, J., Zhang, F., Gu, Z., Li, Y., Song, Y., Hao, W.,
activated Rho in apoptosis in the central nervous system. J. Pang, S. and Sun, J. (2003) Influence of patients’ age on
Cell Biol., 162: 233–243. functional recovery after transplantation of olfactory en-
Duysens, J. and Van de Crommert, H.W. (1998) Neural control sheathing cells into injured spinal cord injury. Chin. Med. J.
of locomotion: the central pattern generator from cats to (Engl.), 116: 1488–1491.
humans. Gait Posture, 7: 131–141. Hurlbert, R.J. (2000) Methylprednisolone for acute spinal cord
Eriksson, P.S., Perfilieva, E., Bjork-Eriksson, T., Alborn, A.M., injury: an inappropriate standard of care. J. Neurosurg., 93:
Nordborg, C., Peterson, D.A. and Gage, F.H. (1998) Ne- 1–7.
urogenesis in the adult human hippocampus. Nat. Med., 4: Hurlbert, R.J. (2001) The role of steroids in acute spinal cord
1313–1317. injury: an evidence-based analysis. Spine, 26: S39–S46.
Fass, B. and Ramirez, J.J. (1984) Effects of ganglioside treat- Johansson, C.B., Momma, S., Clarke, D.L., Risling, M., Len-
ments on lesion-induced behavioral impairments and sprout- dahl, U. and Frisen, J. (1999a) Identification of a neural stem
ing in the CNS. J. Neurosci. Res., 12: 445–458. cell in the adult mammalian central nervous system. Cell, 96:
Fehlings, M.G. and Perrin, R.G. (2006) The timing of surgical 25–34.
intervention in the treatment of spinal cord injury: a system- Johansson, C.B., Svensson, M., Wallstedt, L., Janson, A.M.
atic review of recent clinical evidence. Spine, 31: S28–S36. and Frisen, J. (1999b) Neural stem cells in the adult human
Feron, F., Perry, C., Cochrane, J., Licina, P., Nowitzke, A., brain. Exp. Cell Res., 253: 733–736.
Urquhart, S., Geraghty, T. and Mackay-Sim, A. (2005) Au- Joshi, M. and Fehlings, M.G. (2002a) Development and char-
tologous olfactory ensheathing cell transplantation in human acterization of a novel, graded model of clip compressive spi-
spinal cord injury. Brain, 128: 2951–2960. nal cord injury in the mouse: Part 1. Clip design, behavioral
Firouzi, M., Moshayedi, P., Saberi, H., Mobasheri, H., Abol- outcomes, and histopathology. J. Neurotrauma, 19: 175–190.
hassani, F., Jahanzad, I. and Raza, M. (2006) Transplanta- Joshi, M. and Fehlings, M.G. (2002b) Development and char-
tion of Schwann cells to subarachnoid space induces repair in acterization of a novel, graded model of clip compressive
contused rat spinal cord. Neurosci. Lett., 402: 66–70. spinal cord injury in the mouse: Part 2. Quantitative neuro-
Geisler, F.H., Coleman, W.P., Grieco, G. and Poonian, D. anatomical assessment and analysis of the relationships be-
(2001) The Sygen multicenter acute spinal cord injury study. tween axonal tracts, residual tissue, and locomotor recovery.
Spine, 26: S87–S98. J. Neurotrauma, 19: 191–203.
Geisler, F.H., Dorsey, F.C. and Coleman, W.P. (1991) Recov- Karimi-Abdolrezaee, S., Eftekharpour, E., Wang, J., Mors-
ery of motor function after spinal-cord injury — a randomi- head, C.M. and Fehlings, M.G. (2006) Delayed transplanta-
zed, placebo-controlled trial with GM-1 ganglioside. N. Engl. tion of adult neural precursor cells promotes remyelination
J. Med., 324: 1829–1838. and functional neurological recovery after spinal cord injury.
Gruner, J.A. (1992) A monitored contusion model of spinal J. Neurosci., 26: 3377–3389.
cord injury in the rat. J. Neurotrauma, 9: 123–128. Kernie, S.G., Erwin, T.M. and Parada, L.F. (2001) Brain re-
Hall, E.D. and Braughler, J.M. (1981) Acute effects of intra- modeling due to neuronal and astrocytic proliferation after
venous glucocorticoid pretreatment on the in vitro peroxi- controlled cortical injury in mice. J. Neurosci. Res., 66:
dation of cat spinal cord tissue. Exp. Neurol., 73: 321–324. 317–326.
Hall, E.D. and Braughler, J.M. (1982) Effects of intravenous Knoller, N., Auerbach, G., Fulga, V., Zelig, G., Attias, J.,
methylprednisolone on spinal cord lipid peroxidation and Bakimer, R., Marder, J.B., Yoles, E., Belkin, M., Schwartz, M.
Na++K+-ATPase activity. Dose-response analysis during and Hadani, M. (2005) Clinical experience using incubated au-
1st hour after contusion injury in the cat. J. Neurosurg., 57: tologous macrophages as a treatment for complete spinal cord
247–253. injury: phase I study results. J. Neurosurg. Spine, 3: 173–181.
Hansebout, R.R., Blight, A.R., Fawcett, S. and Reddy, K. Kohama, I., Lankford, K.L., Preiningerova, J., White, F.A.,
(1993) 4-Aminopyridine in chronic spinal cord injury: a con- Vollmer, T.L. and Kocsis, J.D. (2001) Transplantation of
trolled, double-blind, crossover study in eight patients. J. cryopreserved adult human Schwann cells enhances axonal
Neurotrauma, 10: 1–18. conduction in demyelinated spinal cord. J. Neurosci., 21:
Hicks, A.L., Adams, M.M., Martin Ginis, K., Giangregorio, 944–950.
L., Latimer, A., Phillips, S.M. and McCartney, N. (2005) Lee, K.H., Yoon do, H., Park, Y.G. and Lee, B.H. (2005)
Long-term body-weight-supported treadmill training and Effects of glial transplantation on functional recovery fol-
subsequent follow-up in persons with chronic SCI: effects lowing acute spinal cord injury. J. Neurotrauma, 22:
on functional walking ability and measures of subjective well- 575–589.
being. Spinal Cord, 43: 291–298. Li, S. and Strittmatter, S.M. (2003) Delayed systemic Nogo-66
Hofstetter, C.P., Schwarz, E.J., Hess, D., Widenfalk, J., El receptor antagonist promotes recovery from spinal cord in-
Manira, A., Prockop, D.J. and Olson, L. (2002) Marrow jury. J. Neurosci., 23: 4219–4227.
stromal cells form guiding strands in the injured spinal cord Li, Y., Field, P.M. and Raisman, G. (1998) Regeneration of
and promote recovery. Proc. Natl. Acad. Sci. U.S.A., 99: adult rat corticospinal axons induced by transplanted olfac-
2199–2204. tory ensheathing cells. J. Neurosci., 18: 10514–10524.
232

Lucas, S.M., Rothwell, N.J. and Gibson, R.M. (2006) The role Savio, T. and Schwab, M.E. (1989) Rat CNS white matter, but
of inflammation in CNS injury and disease. Br. J. Pharma- not gray matter, is nonpermissive for neuronal cell adhesion
col., 147(Suppl 1): S232–S240. and fiber outgrowth. J. Neurosci., 9: 1126–1133.
Man, S.M., Ma, Y.R., Shang, D.S., Zhao, W.D., Li, B., Guo, Sekhon, L.H. and Fehlings, M.G. (2001) Epidemiology, demo-
D.W., Fang, W.G., Zhu, L. and Chen, Y.H. (2007) Periph- graphics, and pathophysiology of acute spinal cord injury.
eral T cells overexpress MIP-1 alpha to enhance its Spine, 26: S2–S12.
transendothelial migration in Alzheimer’s disease. Neurobiol. Stirling, D.P., Khodarahmi, K., Liu, J., McPhail, L.T.,
Aging, 28: 485–496. McBride, C.B., Steeves, J.D., Ramer, M.S. and Tetzlaff, W.
Miklossy, J., Doudet, D.D., Schwab, C., Yu, S., McGeer, E.G. (2004) Minocycline treatment reduces delayed oligodendro-
and McGeer, P.L. (2006) Role of ICAM-1 in persisting in- cyte death, attenuates axonal dieback, and improves func-
flammation in Parkinson disease and MPTP monkeys. Exp. tional outcome after spinal cord injury. J. Neurosci., 24:
Neurol., 197: 275–283. 2182–2190.
Nashmi, R. and Fehlings, M.G. (2001) Mechanisms of axonal Stokes, B.T., Noyes, D.H. and Behrmann, D.L. (1992) An
dysfunction after spinal cord injury: with an emphasis on the electromechanical spinal injury technique with dynamic
role of voltage-gated potassium channels. Brain Res. Brain sensitivity. J. Neurotrauma, 9: 187–195.
Res. Rev., 38: 165–191. Stoy, N., Mackay, G.M., Forrest, C.M., Christofides, J., Eger-
Niederost, B., Oertle, T., Fritsche, J., McKinney, R.A. and ton, M., Stone, T.W. and Darlington, L.G. (2005) Try-
Bandtlow, C.E. (2002) Nogo-A and myelin-associated glyco- ptophan metabolism and oxidative stress in patients with
protein mediate neurite growth inhibition by antagonistic Huntington’s disease. J. Neurochem., 93: 611–623.
regulation of RhoA and Rac1. J. Neurosci., 22: 10368–10376. Syková, E., Jendelova, P., Urdzikova, L., Lesny, P. and Hejcl,
Noorbakhsh, F., Tsutsui, S., Vergnolle, N., Boven, L.A., A. (2006) Bone marrow stem cells and polymer hydrogels:
Shariat, N., Vodjgani, M., Warren, K.G., Andrade-Gordon, two strategies for spinal cord injury repair. Cell. Mol.
P., Hollenberg, M.D. and Power, C. (2006) Proteinase-acti- Neurobiol., 26: 1111–1127.
vated receptor 2 modulates neuroinflammation in experi- Tator, C.H., Fehlings, M.G., Thorpe, K. and Taylor, W. (1999)
mental autoimmune encephalomyelitis and multiple sclerosis. Current use and timing of spinal surgery for management of
J. Exp. Med., 203: 425–435. acute spinal cord injury in North America: results of a
Oudega, M. and Xu, X.M. (2006) Schwann cell transplanta- retrospective multicenter study. J. Neurosurg., 91: 12–18.
tion for repair of the adult spinal cord. J. Neurotrauma, 23: Teng, Y.D., Choi, H., Onario, R.C., Zhu, S., Desilets, F.C.,
453–467. Lan, S., Woodard, E.J., Snyder, E.Y., Eichler, M.E. and
Park, H.C., Shim, Y.S., Ha, Y., Yoon, S.H., Park, S.R., Choi, Friedlander, R.M. (2004) Minocycline inhibits contusion-
B.H. and Park, H.S. (2005) Treatment of complete spinal triggered mitochondrial cytochrome c release and mitigates
cord injury patients by autologous bone marrow cell trans- functional deficits after spinal cord injury. Proc. Natl. Acad.
plantation and administration of granulocyte-macrophage Sci. U.S.A., 101: 3071–3076.
colony stimulating factor. Tissue Eng., 11: 913–922. Tikka, T.M. and Koistinaho, J.E. (2001) Minocycline provides
Pellitteri, R., Russo, A. and Stanzani, S. (2006) Schwann cell: a neuroprotection against N-methyl-D-aspartate neurotoxicity
source of neurotrophic activity on cortical glutamatergic by inhibiting microglia. J. Immunol., 166: 7527–7533.
neurons in culture. Brain Res., 1069: 139–144. Van de Crommert, H.W., Mulder, T. and Duysens, J. (1998)
Pollard, S.M., Conti, L., Sun, Y., Goffredo, D. and Smith, A. Neural control of locomotion: sensory control of the central
(2006) Adherent neural stem (NS) cells from fetal and adult pattern generator and its relation to treadmill training. Gait
forebrain. Cereb. Cortex, 16: i112–i120. Posture, 7: 251–263.
Ramon-Cueto, A., Cordero, M.I., Santos-Benito, F.F. and Walmsley, A.R., McCombie, G., Neumann, U., Marcellin, D.,
Avila, J. (2000) Functional recovery of paraplegic rats and Hillenbrand, R., Mir, A.K. and Frentzel, S. (2004) Zinc
motor axon regeneration in their spinal cords by olfactory metalloproteinase-mediated cleavage of the human Nogo-66
ensheathing glia. Neuron, 25: 425–435. receptor. J. Cell Sci., 117: 4591–4602.
Rapalino, O., Lazarov-Spiegler, O., Agranov, E., Velan, G.J., Wells, J.E., Hurlbert, R.J., Fehlings, M.G. and Yong, V.W.
Yoles, E., Fraidakis, M., Solomon, A., Gepstein, R., Katz, (2003) Neuroprotection by minocycline facilitates significant
A., Belkin, M., Hadani, M. and Schwartz, M. (1998) recovery from spinal cord injury in mice. Brain, 126:
Implantation of stimulated homologous macrophages results 1628–1637.
in partial recovery of paraplegic rats. Nat. Med., 4: 814–821. Wiessner, C., Bareyre, F.M., Allegrini, P.R., Mir, A.K.,
Richardson, P.M., McGuinness, U.M. and Aguayo, A.J. (1980) Frentzel, S., Zurini, M., Schnell, L., Oertle, T. and Schwab,
Axons from CNS neurons regenerate into PNS grafts. Na- M.E. (2003) Anti-Nogo-A antibody infusion 24 hours
ture, 284: 264–265. after experimental stroke improved behavioral outcome and
Rivlin, A.S. and Tator, C.H. (1978) Effect of duration of acute corticospinal plasticity in normotensive and spontane-
spinal cord compression in a new acute cord injury model in ously hypertensive rats. J. Cereb. Blood Flow Metab., 23:
the rat. Surg. Neurol., 10: 38–43. 154–165.
233

Winton, M.J., Dubreuil, C.I., Lasko, D., Leclerc, N. and Zheng, M. and Kuffler, D.P. (2000) Guidance of regen-
McKerracher, L. (2002) Characterization of new cell perme- erating motor axons in vivo by gradients of diffusible
able C3-like proteins that inactivate Rho and stimulate neu- peripheral nerve-derived factors. J. Neurobiol., 42:
rite outgrowth on inhibitory substrates. J. Biol. Chem., 277: 212–219.
32820–32829. Zhu, S., Stavrovskaya, I.G., Drozda, M., Kim, B.Y., Ona, V.,
Xu, L., Fagan, S.C., Waller, J.L., Edwards, D., Borlongan, Li, M., Sarang, S., Liu, A.S., Hartley, D.M., Wu du, C.,
C.V., Zheng, J., Hill, W.D., Feuerstein, G. and Hess, D.C. Gullans, S., Ferrante, R.J., Przedborski, S., Kristal, B.S. and
(2004) Low dose intravenous minocycline is neuroprotective Friedlander, R.M. (2002) Minocycline inhibits cytochrome c
after middle cerebral artery occlusion-reperfusion in rats. release and delays progression of amyotrophic lateral sclero-
BMC Neurol., 4: 7. sis in mice. Nature, 417: 74–78.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 16

Cerebral contusion: a role model for lesion


progression

Tatsuro Kawamata1,2, and Yoichi Katayama1,2

1
Department of Neurological Surgery, Nihon University School of Medicine, Tokyo, Japan
2
The Japan Neurotrauma Data Bank Committee (The Japan Society of Neurotraumatology, The Japanese Council of
Traffic Science)

Abstract: The early massive edema caused by severe cerebral contusion results in progressive intracranial
pressure (ICP) elevation and clinical deterioration within 24–72 h post-trauma. Surgical excision of the
necrotic brain tissue represents the only therapy, which can provide satisfactory control of the elevated ICP
and clinical deterioration. In this chapter, we review the results of our clinical studies regarding the
pathophysiology of contusion edema and evaluate the effects of surgical treatment, i.e. contusion necro-
tomy, by analyzing the data from the Japan Neurotrauma Data Bank.

Keywords: cerebral contusion; brain edema; diffusion-weighted image; vascular permeability; tissue
osmolality; contusion necrotomy

Introduction therapy, which can provide satisfactory control of


the elevated ICP. The precise mechanism under-
In patients with cerebral contusions, two types of lying such an early massive edema is not yet clearly
edema can be clinically recognized. One is the early understood. We review the results of our clinical
massive edema that occurs within the period of studies (Katayama et al., 1990, 1992, 1998; Kushi
24–72 h post-trauma. This type of edema creates et al., 1994; Kawamata et al., 2000; Katayama and
strong mass effect resulting in progressive elevation Kawamata, 2003), which have provided several
of the intracranial pressure (ICP) and clinical dete- lines of evidence to suggest that a large amount of
rioration known as talk-and-deteriorate (Katayama edema fluid is accumulated in the necrotic brain
et al., 1990). The other is the delayed peri-contusion tissue within the central area of contusion, and this
edema, which is typically seen in the white matter contributes to the early massive edema. The clini-
adjacent to the cerebral contusion, at several days cal results of surgical treatment, i.e. contusion
post-trauma by T2-weighted magnetic resonance necrotomy, are also discussed.
(MR) imaging. This type of edema rarely causes
ICP elevation leading to fatal deterioration.
Despite intensive medical therapy, the elevated Histopathology of cerebral contusion
ICP in patients with early massive edema is often
uncontrollable and fatal. In such instances, surgi- The classical histopathological study of Lindenberg
cal excision of the necrotic brain tissue is the only and Freytag (1957) demonstrated the presence of
two components of cerebral contusion; one is the
Corresponding author. Tel.: +81-3-3972-8111 ext. 2481; central (core) area in which cells undergo necrosis
Fax: +81-3-3554-0425; E-mail: kawamata@med.nihon-u.ac.jp as the primary consequence of mechanical injury

DOI: 10.1016/S0079-6123(06)61016-9 235


236

(contusion necrosis proper), and the other is the


peripheral (rim) area in which cellular swelling oc-
curs as a consequence of ischemia. A clear demar-
cation line separates these two components. The
area of contusion necrosis is histopathologically
evident as early as at 3 h post-trauma, as a primary
brain damage (Eriskat et al., 1994). The cellular
elements in the central area, both neuronal as well
as glial cells, uniformly undergo shrinkage, and
then disintegration, homogenation and cyst forma-
tion eventually. In contrast, cell swelling is pre-
dominant in the peripheral area (Lindenberg and
Freytag, 1957). The ischemia in the peripheral area
is largely attributable to microthrombosis, which is
a main cause of secondary brain damages. Numer-
ous clinical studies have demonstrated a decrease in
cerebral blood flow in the central as well as the
peripheral areas of contusion (e.g., Alexander
et al., 1994).

MR diffusion study (ADC mapping)

We have investigated the evolution of cerebral


contusion by diffusion-weighted MR imaging and
apparent diffusion co-efficient (ADC) mapping in Fig. 1. A representative case of cerebral contusion in the acute
phase post-trauma. Upper: A CT scan revealed low-density
head trauma patients (Kawamata et al., 2000). area in bilateral frontal lobe. Lower: Diffusion-weighted image
Following conventional T1- and T2-weighted MR demonstrated low intensity core with surrounding high inten-
imaging, diffusion-weighted MR images are ob- sity rim, which was typical pattern of diffusion-weighted image
tained, and ADC mapping is computed from var- in the acute phase (o48 h post-trauma) of cerebral contusion
ious b factor diffusion images. without massive hemorrhage.
The diffusion-weighted MR images demonstrate
a low intensity core in the central area, beginning
at approximately 24 h post-trauma. The ADC during this period, which can be termed a halo ap-
value within this central area clearly increases pearance (Fig. 2). The ADC value decreases in the
during the period of 24–72 h post-trauma (Fig. 1; peripheral area during the period of 24–72 h post-
Kawamata et al., 2000). This elevated ADC value trauma (Kawamata et al., 2000). This decreased
appears to represent the contusion necrosis proper, ADC value appears to represent the cellular swell-
since cellular disintegration and homogenization ing, which would result in shrinkage of the extra-
in this area would result in an expansion of the cellular space. The ADC values between the central
extracellular space. and peripheral areas are maximally dissociated
The diffusion-weighted MR images demonstrate during the period of 24–72 h post-trauma.
a high intensity rim in the peripheral area of contu- The ADC value in the peripheral area shifts
sion beginning at approximately 24 h post-trauma, from a decrease to an increase after 72 h post-
which corresponds with the timing of the elevated trauma. At the same time, an increase in ADC
ICP and neurological deterioration. The combina- value becomes evident in the adjacent white mat-
tion of a low intensity core and a high intensity rim ter. This increase in ADC value in the adjacent
is a consistent finding in the cerebral contusion white matter appears to represent the delayed
237

Fig. 2. Bilateral contusion demonstrating a halo appearance with a combination of a low intensity core and a high intensity rim on
diffusion-weighted image (left). On ADC mapping (right), the peripheral rim showed low ADC value representing the cellular swelling
resulting from shrinkage of the extracellular space.

peri-contusion edema, which can commonly be seen appear to contradict the widely held view that an
on T2-weighted MR images as vasogenic edema. increased cerebrovascular permeability is respon-
sible for the development of contusion edema
(Marmarou, 2003).
Increased cerebrovascular permeability We examined the changes in cerebrovascular per-
meability employing intravenous slow infusion of
Since gadolinium (Gd)-DTPA, like plasma pro- Gd-DTPA and delayed MR imaging (Kushi et al.,
tein, does not normally cross the blood-brain bar- 1994). In general, increases in cerebrovascular per-
rier, enhancement with Gd-DTPA, if observed in meability can be detected more clearly by intrave-
association with cerebral contusion, implies an in- nous slow infusion of high dose contrast medium
creased cerebrovascular permeability. In a previ- and delayed neuroimaging (Ito et al., 1988). This
ous study (Lang et al., 1991), Gd-DTPA was technique revealed that cerebral contusions can be
administered intravenously by bolus injection and enhanced at as early as 24–48 h post-trauma. In MR
MR imaging was undertaken soon after the ad- imaging undertaken at 2 h after Gd-DTPA admini-
ministration. Such a procedure failed to detect any stration, enhancement on T1-weighted images was
increase in cerebrovascular permeability associ- observed in either the central area or the peripheral
ated with cerebral contusions during the initial few area, or both. It is evident that water supply from
days post-trauma. Enhancement with Gd-DTPA the blood vessels is not completely interrupted even
has been reported to become detectable at 6–9 in the central area of contusion.
days post-trauma. The delayed peri-contusion edema has commonly
Furthermore, an immunohistochemical study of been attributed to an increased cerebrovascular
the post-mortem brain in such patients failed to permeability. As mentioned above, enhancement
reveal any plasma protein leakage around the con- with Gd-DTPA has been reported to become de-
tused brain areas during this early period (Todd tectable at 6–9 days post-trauma. It is also possible,
and Graham, 1990). Evaluations of the vascular however, that resolution of the cellular swelling
permeability by 99mTc pertechnetate single pho- in the peripheral area of contusion might permit
ton emission tomography failed to reveal any evi- propagation of the edema fluid accumulated within
dence of an increased vascular permeability within the central area to the adjacent white matter. This
the area of contusion during the initial few days would lead to edema appearance on CT and MR
post-trauma (Bullock et al., 1990). Such findings images which resembles that induced by an
238

increased cerebrovascular permeability, i.e. vaso- crescent-shaped zone still existed at 2 weeks post-
genic edema. trauma in some cases (Fig. 4). The very high ADC
value appears to represent edema fluid accumula-
tion within the central area of contusion.
Edema fluid accumulation in the central area Edema fluid accumulation within the necrotic
brain tissue is also suggested by the formation of a
In approximately 50% of patients with cerebral fluid–blood interface within cerebral contusions
contusions, a crescent-shaped zone of very high (Fig. 5). The fluid–blood interface can be formed
ADC value develops at the border between the without a fluid cavity. When we carry out surgery,
central and peripheral areas beginning at approxi- we find no fluid cavity but softened and water-rich
mately 24 h post-trauma (Fig. 3; Kawamata et al., necrotic brain tissue is present (Katayama et al.,
2000). This crescent-shaped zone was always lo- 1992). Within the contusion necrosis, hemorrh-
cated within the central area of contusion. The age undergoes enlargement within the initial 6 h

Fig. 3. Cerebral contusion in the temporal tip at 45 h post-trauma. Left: CT scan showed small low-density area in the temporal tip.
Middle: T2-weighted MRI revealed mixed intensity area in the core of contusion. Right: ADC mapping demonstrated a crescent-
shaped zone (arrows) of very high ADC value, as high as that of CSF, representing edema fluid accumulation within the central area of
contusion.

Fig. 4. Cerebral contusion in the subacute phase, 2 weeks post-trauma. Left: T2-weighted image. Middle: Diffusion-weighted image.
Right: ADC mapping. A crescent shaped zone still existed (arrows). ADC value in the peripheral area of contusion increased,
indicating that vasogenic edema turned to be predominant.
239

Fig. 5. CT scan and its scheme of cerebral contusion in the temporal tip (10 h post-trauma), showing fluid–blood interface in the area
of contusion necrosis. (A) edema fluid; (B) hematoma; (C) fluid–blood interface; (D) layering of red blood cells.

post-trauma (Katayama et al., 1990), and red edema fluid accumulation in the central area and
blood cells diffusely permeate the softened necrotic contribute to the early massive edema of cerebral
brain tissue (Lindenberg and Freytag, 1957). The contusion.
fluid–blood interfaces observed within the central
area of contusion may represent layering of red
Surgical treatment
blood cells in the softened necrotic brain tissue,
which has accumulated voluminous edema fluid.
We investigated the effects of surgical excision of
the necrotic brain tissue in patients with severe cere-
bral contusion. The data from the Japan Neuro-
Osmotic potential of the contusion necrosis
trauma Data Bank (JNTDB) in which a total of
1002 patients suffering severe traumatic brain
We have reported that necrotic brain tissue sam-
injury registered during the period between 1998
pled from the central area of contusion during
and 2001 were analyzed (Nakamura et al., 2002).
surgery shows a very high osmolality, reaching
Among these patients, 182 (18%) demonstrated
350–400 mOsm (Katayama et al., 1998). It is un-
severe cerebral contusions as the major lesions
certain whether or not such a marked increase in
contributing to their clinical status.
osmolality is osmotically active and causes edema
fluid accumulation. It appears that expansion of
the extracellular space in the central area increases Results
the capacitance for edema fluid accumulation. In
contrast, shrinkage of the extracellular space in the Among the 182 patients with severe cerebral con-
peripheral area increases the resistance for edema tusion, 121 (66%; Group I) were treated conserv-
fluid propagation or resolution. We hypothesize atively and the remaining 61 (34%; Group II)
that the barrier formed by swollen cells in the peri- underwent surgery. The ratio of selecting surgical
pheral area may prevent edema fluid propagation management was far lower in patients with cere-
and also help to generate osmotic potentials across bral contusion (34719%), as compared with those
the central and peripheral areas. Since blood flow with acute epidural hematoma (88711%) or acute
is greatly reduced but is not completely interrupted subdural hematoma (68718%). There was a huge
in the contused brain tissue, water is supplied from variation in the ratio of selecting surgical mana-
the blood vessels into the central area. We suggest gement (9–77%) among the contributing centers
that a combination of these events may facilitate (n ¼ 10) of this data bank. While older patients,
240

especially those between 40 and 60 years old, mortality between the two groups was observed
tended to undergo surgery more frequently, even when the analysis was restricted to patients
younger patients tended to be treated conserva- who definitely demonstrated ‘‘talk-and-deteriorate’’
tively. There was, however, no significant differ- (64% vs. 22%; p ¼ 0.026; n ¼ 29; Table 2).
ence in age between Groups I and II (47.8723.8 The indications for surgical intervention in case
vs. 54.4719.5 years). The surgical management of severe cerebral contusion remain controversial
involved internal decompression (complete exci- (Bullock et al., 1989). The huge variation in ratio
sion of the necrotic brain tissue and evacuation of of selecting surgical management among the
clots) with or without external decompression in present centers reflects a diversity in management
most patients (90%) of Group II. The remaining policy and an absence of consensus regarding the
patients underwent external decompression alone. indications for surgery.
Surgery was performed at 1.886.1 (19.5724.2) h, The higher mortality in Group I, as compared
mostly (73%) within 24 h post-trauma. with Group II, suggests that the surgery performed
Group I demonstrated a clearly poorer outcome in the Group II patients helped to prevent their
on the Glasgow outcome scale (GOS) at 6 months clinical deterioration and death. Such an effect of
post-trauma (Table 1). The mortality was clearly surgery was most striking in those patients who
higher in Group I, as compared with Group II were scored at 9 or better at the time of admission.
(48% vs. 23%; p ¼ 0.0001; n ¼ 182). A difference It is by no means certain whether patients in
in mortality between the two groups was noted in Group II who were scored at 9 or better before
patients who were scored at 8 or less on the GCS at surgery would have deteriorated or not if surgery
the time of admission, but did not reach a statis- had not been carried out. An effect of surgery on
tically significant level (Table 1). The most striking mortality is evident, however, since a difference in
difference was observed in patients who were scored mortality was clearly observed even when the
at 9 or better on the GCS at the time of admission analysis was restricted to patients who definitely
(Table 1). The mortality was clearly higher in demonstrated ‘‘talk-and-deteriorate’’. This finding
Group I, as compared with Group II (56% vs. strongly suggests that the surgery itself was the
17%; p ¼ 0.017; n ¼ 45). A clear difference in major reason for the improved mortality in Group
II. In other words, death was probably prevented
by the surgery in many patients of Group II.
Table 1. Outcome (6 months post-trauma)
The present findings support our hypothesis that
GOS (%) early massive edema is caused by cerebral contu-
sion through the presence of necrotic brain tissue,
n GR MD SD VS D and indicate that surgical excision of the necrotic
GCS on admission: 3–5 brain tissue is the only therapy which can provide
Conservative 47 7 2 11 11 70 satisfactory control of the progressive elevation of
Surgical 11 9 9 27 0 55 the ICP and clinical deterioration in many cases.
GCS on admission: 6–8 Surgical intervention should be considered in
Conservative 58 29 21 10 10 29
Surgical 21 24 29 24 10 14
Table 2. Outcome (6 months post-trauma) in patients demon-
GCS on admission: 9–15 strating ‘‘talk-and-deteriorate’’
Conservative 16 19 13 13 0 56
Surgical 29 28 28 17 10 17 GOS (%)

Total n GR MD SD VS D
Conservative 121 19 12 12 9 48
Surgical 61 23 25 21 8 23 Conservative 11 18 9 9 0 64
Surgical 18 11 22 39 6 22
Note: GCS, Glasgow coma scale; GOS, Glasgow outcome scale.
 p ¼ 0.017. Note: GCS, Glasgow coma scale; GOS, Glasgow outcome scale.
 p ¼ 0.0001. p ¼ 0.026.
241

patients with severe cerebral contusion who demon- Eriskat, J., Schurer, L., Kempski, O. and Baethmann, A. (1994)
strate ‘‘talk-and-deteriorate’’. Growth kinetics of a primary brain tissue necrosis from focal
lesion. Acta Neurochir., s60: 425–427.
Ito, U., Reulen, H.-J., Tomita, H., Ikeda, J., Saito, J. and
Conclusion Maehara, T. (1988) Formation and propagation of brain
oedema fluid around human brain metastasis: a CT study.
Acta Neurochir., 90: 35–41.
There is at present no established medical treat- Katayama, Y. and Kawamata, T. (2003) Edema fluid accumu-
ment which can effectively inhibit edema fluid ac- lation within necrotic brain tissue as a cause of the mass
cumulation within cerebral contusions. The most effect of cerebral contusion in head trauma patient. Acta
Neurochir., s86: 323–327.
effective therapy for ameliorating the potentially
Katayama, Y., Mori, T., Maeda, T. and Kawamata, T. (1998)
fatal edema is surgical excision of the necrotic Pathogenesis of the mass effect of cerebral contusions: a
brain tissue. The effects of surgical excision of ne- rapid increase in osmolality within the contusion necrosis.
crotic brain tissue have commonly been accounted Acta Neurochir., s71: 289–292.
for on the basis of an increased space compensa- Katayama, Y., Tsubokawa, T., Kinoshita, K. and Himi, K.
tion for mass lesions. It is possible, however, that (1992) Intra-parenchymal fluid-blood levels in traumatic in-
tracerebral hematomas. Neuroradiology, 34: 381–383.
excision of the necrotic brain tissue does mean Katayama, Y., Tsubokawa, T., Miyazaki, S., Kawamata, T.
elimination of the cause of edema fluid accumu- and Yoshino, A. (1990) Oedema fluid formation within con-
lation. If the ICP is elevated by early massive ed- tused brain tissue as a cause of medically uncontrollable el-
ema due to cerebral contusion and the elevated evation of intracranial pressure in head trauma patients. Acta
ICP is medically uncontrollable, surgical excision Neurochir., s51: 308–310.
Kawamata, T., Katayama, Y., Aoyama, N. and Mori, T. (2000)
of the necrotic brain tissue would appear to rep- Heterogeneous mechanisms of early edema formation in cere-
resent the therapy of choice, regardless of the size bral contusion: diffusion MRI and ADC mapping study.
of the associated hemorrhages. Surgery should be Acta Neurochir., s76: 9–12.
considered in patients who are scored at 9 or better Kushi, H., Katayama, Y., Shibuya, T., Tsubokawa, T. and
Kuroha, T. (1994) Gd-DTPA enhanced magnetic resonance
at the time of admission, as soon as they show
imaging of cerebral contusions. Acta Neurochir., s60:
clinical deterioration. 472–474.
Lang, D.A., Hadley, D.M., Teasdale, G.T., Macpherson, P.
and Teasdale, E. (1991) Gadolinium DTPA enhanced
References magnetic resonance imaging in acute head injury. Acta
Neurochir., 109: 5–11.
Alexander, M.J., Martin, N.A., Khanna, M., Caron, M. and Lindenberg, R. and Freytag, E. (1957) Morphology of cortical
Becker, D.P. (1994) Regional cerebral blood flow trends in contusions. AMA Arch. Pathol., 63: 23–42.
head injured patients with focal contusions and cerebral Marmarou, A. (2003) Pathophysiology of traumatic brain
edema. Acta Neurochir., s60: 479–481. edema: current concepts. Acta Neurochir., s86: 7–10.
Bullock, R., Golek, J. and Blake, G. (1989) Traumatic intra- Nakamura, N., Yamaura, A., Shigemori, M., Ono, J., Kawamata,
cerebral hematoma Which patients should undergo surgical T., Sakamoto, T. and Japanese Data Bank Committee for
evacuation? CT scan features and ICP monitoring as a basis Traumatic Brain Injury. (2002) Epidemiology, prevention and
for decision making. Surg. Neurol., 32: 181–187. countermeasures against severe traumatic brain injury in Japan
Bullock, R., Statham, J., Patterson, D., Wyper, D., Hadley, D. and abroad. Neurol. Res., 24: 45–53.
and Teasdale, E. (1990) The time course of vasogenic oedema Todd, N.V. and Graham, D.I. (1990) Blood-brain barrier
after focal human head injury: evidence from SPECT mapping damage in traumatic brain contusion. Acta Neurochir., s51:
of blood brain barrier defects. Acta Neurochir., s51: 286–288. 296–299.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 17

Ethical implications of time frames in a randomized


controlled trial in acute severe traumatic brain injury

Erwin J.O. Kompanje1,2,, Andrew I.R. Maas2, Franc- ois J.A. Slieker2 and
Nino Stocchetti3

1
Department of Intensive Care, Erasmus MC University Medical Center Rotterdam, P.O. Box 2040, 3000 CA Rotterdam,
The Netherlands
2
Department of Neurosurgery, Erasmus MC University Medical Center Rotterdam, P.O. Box 2040, 3000 CA Rotterdam,
The Netherlands
3
Ospedale Policlinico IRCCS, Milan University, Milan, Italy

Abstract: Objectives: To analyze factors determining the time between injury and study drug administra-
tion (SDA) in a randomized controlled trial (RCT) of acute severe traumatic brain injury (TBI) and to
discuss the ethical implications. Methods: Time frames prior to SDA, differentiated per country, were
analyzed in a recently conducted RCT in severe TBI. Per protocol, the time window for SDA was 6 h after
injury. We selected patients for whom written proxy consent (PC) was obtained prior to SDA (n ¼ 631).
Results: The time between injury and admission to the neurotrauma center (NTC) varied per country from
1.16 to 2.35 h, but CT scan was obtained on average within 1 h of admission. The median time between
injury and CT scan was within 3 h in all but one country. The broadest time window was observed between
CT scan and obtaining required PC (1.71–2.74 h). The median time between injury and PC varied between
countries from 3.75 to 5.00 h. After consent had been obtained, almost all patients subsequently received
study drug within 1 h. In 85.3% of all cases time between injury and SDA exceeded 4 h, in 60% 5 h.
Conclusions: The requirement of written PC causes a significant delay in SDA in TBI. With deferred
consent, the first dose of an investigational drug could potentially be administered directly after completion
of the admission CT scan, which reduce the time to SDA by 50%. We argue that randomization under
deferred consent is ethically defendable for emergency research in severe TBI. Recommendations for
patient protection are proposed.

Keywords: traumatic brain injury; informed consent; emergency trial; deferred consent

Introduction TBI is about 30% and a significant disability per-


sists in a further 35–40%. These data signify an
Severe traumatic brain injury (TBI) remains a ethical imperative to develop and test neuropro-
major cause of death and disability afflicting tective agents and other new therapeutic strategies.
mostly young adult males and elderly people, Various neuroprotective agents, mainly targeting
and results in high economic costs to society specific pathophysiologic mechanisms, have been
(McGarry et al. 2002). The fatality rate for severe tested in TBI, but convincing benefit has not been
shown (Maas et al., 1999). Randomized controlled
Corresponding author. Tel.: +31-6-53837655; Fax: +31-10- trials (RCTs) in emergency and intensive care
4634075; E-mail: e.j.o.kompanje@erasmusmc.nl medicine pose complex ethical and methodological

DOI: 10.1016/S0079-6123(06)61017-0 243


244

challenges (Maas et al., 2004; Kompanje et al., proxy consent (PC), and between PC and study
2005). drug administration (SDA)) in a recently
Specific ethical issues pertaining to clinical completed RCT in TBI.
testing of neuroprotective agents in TBI include
the emergency nature of the research, the incapac-
ity of the patients to informed consent before in- Materials and methods
clusion, short therapeutic time windows, and a
risk–benefit ratio based on concept that in relation We analyzed critical time frames in a multi-center
to the severity of the trauma, significant adverse placebo-controlled phase III trial, investigating the
side effects may be acceptable for treatments efficacy and safety of a single dose of dexanabinol
with proven benefit. The relevance of these issues in severe TBI. This RCT was conducted from
and implications for trial design are, however, January 2001 to March 2004 in Europe, Israel,
not fully recognized in- and outside the expert Australia, and the United States. Dexanabinol is a
field. Legislation in countries in the European synthetic cannabinoid analogue with strong
Union is being amended to comply with the neuroprotective potential and devoid of psycho-
European Union Directive 2001/20/EC (European tropic activity. The protocol stipulated SDA
Union, 2001). In this new European legislation, within 6 h after injury. The study recruited 861
emergency research under deferred or waiver of patients with severe TBI. No beneficial effect of
consent is not permitted. This will impede or dexanabinol was found. Full details and results of
even obviate emergency research phase III trials in the study have been published (Maas et al., 2006).
TBI in the European countries (Kompanje and In total, 7164 patients were screened for partic-
Maas, 2004; Silverman et al., 2004; Liddell et al., ipation in the trial. Enrollment criteria were not met
2006a, b). in 6303 patients. Of these, the sole reason for
We aimed to analyze the implications of critical exclusion was the time window in 671 patients and
time frames (time between injury and admission to in a further 944 patients inability to give study
a neurotrauma center (NTC), between admission medication within 6 h was one of the reasons for
and the first cranial CT scan, between CT scan and exclusion (Fig. 1).

Time window as main reason for exclusion

4688

5000

4000

3000

944
2000 671

1000

0
time window as time window +
other reason for
only reason for other reason for
exclusion
exclusion exclusion

patients 671 944 4688

Fig. 1. Reasons for exclusion (n ¼ 6303).


245

In view of the severity of the brain injury, in- incomplete screening logs due to HIPAA regula-
formed consent could not be obtained from the tions (Maas et al., 2005; Kompanje and Maas,
patients themselves. PC was accepted in all coun- 2006), and patients from Australia seeing the
tries. Deferred consent was allowed in Australia, different infrastructure of this country in compar-
Austria, Finland, and in some centers in France ison to European countries.
and Germany. Consent by an independent physi-
cian was allowed in Israel, Italy, Spain, and the
United Kingdom. In all cases of deferred consent, Results
subsequent written assent by patient or proxy was
obtained. The median age of our study population was 32
For the analysis of time frames we selected pa- years (IQR 23–44); 520 (82.4%) were male, 111
tients enrolled in Europe and Israel, in whom PC (17.6%) female. The time between injury and ad-
was obtained before SDA (n ¼ 631). We excluded mission to the NTC varied between countries from
patients from the United States for reason of 1.16 to 2.35 h (Table 1, Fig. 2). A total of 501

Table 1. Time windows per country (median+IQR)

Country (N) Hours between injury Hours between injury Hours between injury Hours between injury
and admission NTC and CT scan median and obtained consent and SDA median
median (IQR) (IQR) median (IQR) (IQR)

Belgium (23) 0.93 (0.65–1.27) 1.80 (1.28–2.27) 3.75 (2.75–4.75) 4.60 (3.98–5.42)
Netherlands (73) 1.00 (0.75–1.33) 1.65 (1.32–2.00) 4.53 (3.95–5.05) 5.53 (5.07–5.75)
Israel (116) 0.93 (0.72–1.40) 1.91 (1.58–2.47) 4.01 (3.20–4.83) 4.67 (4.00–5.33)
Spain (75) 1.33 (0.97–1.67) 2.07 (1.65–2.53) 4.17 (3.33–5.00) 5.17 (4.30–5.58)
Germany (109) 1.20 (0.88–2.00) 1.65 (1.30–2.13) 4.08 (3.42–4.98) 5.25 (4.25–5.67)
Italy (146) 1.25 (0.83–2.60) 1.77 (1.40–2.35) 4.92 (4.08–5.28) 5.50 (4.98–5.75)
France (34) 2.17 (1.42–3.00) 3.08 (1.97–3.53) 5.00 (4.50–5.38) 5.75 (5.17–5.83)
Other countriesa (55) 1.47 (1.00–2.67) 1.82 (1.33–2.50) 4.00 (3.08–4.75) 5.25 (4.33–5.75)
a
Countries with small patient populations (United Kingdom, Denmark, Austria, Poland, and Turkey) were combined.

other

france

italy

germany

spain

israel

netherlands

belgium

injury 1h 2h 3h 4h 5h 6h
time between injury and admission NTC
time between admission and ct-scan
time between ct-scan and obtained consent
time between consent and SDA

Fig. 2. Time between injury and admission neurotrauma center, time between admission and first CT scan, time between first CT scan
and informed consent for inclusion in trial and time between consent and start study drug admission.
246

379 informed consent before inclusion, and a


400
risk–benefit ratio based on the concept that in
350 relation to the severity of the trauma, significant
adverse side effects may be acceptable for treat-
300
ments with proven benefit (Kompanje et al., 2005).
number of patients

250

200 168
The emergency nature of research and short
150 therapeutic time windows in TBI
100 70
Severe TBI is an emergent and life-threatening
50 13 condition and existing therapy is unsatisfactory
1
0 given the high morbidity and mortality. Neuro-
1-2hours 2-3hours 3-4hours 4-5hours >5hours logical damage does not only occur at the moment
time between injury and study drug admission of impact, but can evolve over the following hours
and days. Deleterious effects of this progressive
Fig. 3. Distribution of number of patients. Time between injury
and study drug administration (n ¼ 631). damage are determined at clinical and biochemical
levels. This has led to the development of new
(79.4%) patients were directly admitted to the pharmaceuticals with promising potential to limit
NTC, and 130 (20.6%) concerned secondary refer- secondary damage and to improve outcome. One
rals. In 71 secondarily referred patients the CT of these new pharmaceuticals was dexanabinol,
scan was made before admission to the NTC. The but efficacy in the treatment of severe TBI was not
time between admission and the first diagnostic demonstrated (Maas et al., 2006). Recruitment in
CT scan was within 1 h in all patients. With the this study was relatively slow and the majority of
exception of France, in all countries the median patients enrolled toward the end of the 6 h time
time between injury and CT scan was within 3 h window stipulated in the protocol. We hypothe-
(Table 1). The broadest time window was found sized that the relatively slow recruitment and lack
between the admission CT scan and obtaining the of demonstrable efficacy in the clinical situation
required PC (between 1.71 and 2.74 h). The median may have been influenced by informed consent
time between injury and obtained PC varied be- procedures and resulting delays in SDA. Early in-
tween 3.75 and 5.00 h (IQR 2.75–5.38 h) (Table 1). tervention would appear crucial to the effect of
After consent had been given, almost all patients neuroprotective agents. Experimental data have
subsequently received the study drug within 1 h consistently shown better protection the sooner an
(Table 1, Fig. 2). In 85.3% of all cases the time agent is administered after TBI (Hoff, 1986). For
between injury and SDA exceeded 4 h, in 60% of dexanabinol, experimental studies have shown
the cases it even exceeded 5 h (Fig. 3). In total, 139 beneficial effects with administration within 3 h
patients were randomized with deferred consent. after injury, demonstrating protection against
The median time between injury and SDA in this breakdown of the blood-brain barrier, reduction
group was 4.75 h (IQR 3.90–5.67). These 139 pa- of edema formation, and improved outcome
tients are not included in our analysis. (Shohami, 1995). No significant reduction of cer-
ebral edema was noted if the drug was administered
between 4 and 6 h after injury, but some improve-
Discussion ment in neurological symptoms was found. Based
on these findings, it may be concluded that in the
Specific ethical issues pertaining to clinical evalu- experimental model the pathophysiologic time win-
ation of neuroprotective agents in TBI include the dow can be determined at 3 h. Whether this exper-
emergency nature of the research, short therapeu- imental time window may be extrapolated to the
tic windows, the incapacity of the patients to clinical situation in patients with TBI remains
247

uncertain. Prespecified subgroup (patients who (CRASH trial management group, 2004). The ob-
received the SD within 4 h and patients who re- servation in the cohort of patients enrolled in the
ceived the SD after 4 h) analysis showed no signifi- dexanabinol trial with deferred or waiver of con-
cant differential treatment effect (Maas et al., sent, that (proxy) assent was obtained later in all
2006). Nonetheless, it may be concluded in general cases supports the concept of accepting deferred or
that chances of efficacy are greater if treatment is waiver of consent for emergency research in TBI
provided earlier. We found that in almost all of the trials.
studied cases the time between injury and comple-
tion of the primary diagnostic CT scan remained
within 3 h post injury, which corresponds to the The acute incapacity of the patients and informed
therapeutic time window in the animal model. In consent
60% of the cases, however, the time between injury
and SDA was more than 5 h, and in 85.3% of all All patients with severe TBI are unconscious, and
cases more than 4 h (Fig. 3). consequently informed consent can never be ob-
We have shown that the main determinant of tained from the patients themselves. As substitute
the time to SDA is formed by the time required to for informed consent, a legal representative must
obtain informed (proxy) consent, and these results give consent for inclusion in research, or in the
indicate that delays in SDA could be reduced by absence thereof, by proxies; alternatively consent
50% with the adoption of deferred consent or may be deferred or waived. We have argued that
waiver of consent. We could, however, not deferred consent is preferable. However, the new
demonstrate shorter inclusion times in the cohort European Union Directive (2001/20/EC) (Euro-
of patients randomized with deferred consent. The pean Union, 2001; Liddell et al., 2006a, b) stipu-
reason for the delay in this cohort was that most lates a requirement for informed (proxy) consent.
investigators waited for PC before randomization, This is motivated by respect for the autonomy of
and only used deferred consent when at the end of patients, and to ensure that that patient’s wishes
the inclusion boundary. These observations favor are guaranteed as far as possible. The requirement
adopting deferred consent procedures in trials in for PC assumes that relatives are available in emer-
acute TBI as primary approach, rather than as gency situations, and that these relatives can be fully
‘‘ultimum refugium’’, only to be undertaken if PC informed and given sufficient time to make a bal-
cannot be obtained. Other studies have demon- anced, ethically valid decision in a relatively short
strated advantages in using the deferred consent time period under emotional distress. Even when
and waiver of consent in emergency research. In proxies are available, many are not aware of the
the National Acute Brain Injury Study: Hypother- patient’s wishes (Luce, 2003). Surrogate decision
mia (NABIS-H) the adoption of waiver of consent makers for critical care research resulted in false-
resulted in higher enrollment and reduced the time positive consent rates of 16–20.3% (Coppolino
between injury and treatment by approximately and Ackerson, 2001). The emotional nature of an
45 min (Clifton et al., 2002). In this study, relatives emergency situation limits the reliability of PC
of only 11 out of 113 patients arrived within 6 h for clinical research (Mason and Allmark, 2000;
after the injury. In a septic shock trial the inves- Coppolino and Ackerson, 2001; Hsieh et al.,
tigators could not contact the proxies within the 2001). Under emergency circumstances, PC does
inclusion time in 74% of the cases, and these were not seem to secure proper patient/subject protec-
included under waiver of consent (Annane et al., tion. In our experience, the validity of informed
2004). In the CRASH trial, mean time to random- consent and PC given in an emergency situation is
ization was significantly longer in those hospitals at least troubling. When consent for clinical re-
where consent was required compared with those search is sought during an emergency situation,
it was not (4.4 h [SE ¼ 0.21] versus 3.2 h comprehension is generally less than optimal (Cut-
[SE ¼ 0.16]), the difference in the mean time to tini, 2000; Sugarman, 2000; Williams et al., 2003).
randomization was 1.2 h [95% CI 0.7–1.8 h] A small minority realizes that pharmacological
248

trials are designed to assess not only efficacy but in treatment initiation, as we have shown by our
safety as well (Harth and Thong, 1995). Are pa- analysis. The possible therapeutic benefit, with
tients willing to be represented by their close rel- short therapeutic time windows in experimental
atives? Roupie et al. (2000) found that only 40.6% models, forms the moral justification for random-
of 1089 patients wanted their spouse/partner to be izing patients under deferred consent or waiver of
their surrogate, 28% wanted to be represented by consent within a sufficient period of time. The
the physician in charge of their care. risks should however be acceptable in relation to
One study searching for public views on emer- the severity of the disease or injury.
gency exception to informed consent found that For an effective agent in life-threatening condi-
most of 530 people (88%) believed that research tion adverse effects may be expected and accepted
subjects should be informed prior to being en- in some patients. Careful monitoring and follow-
rolled, while 49% believed enrolling patients with- up of such adverse events is mandatory.
out prior consent would be acceptable in an For trials under deferred consent or waiver of
emergency situation and 70% (369) would not ob- consent in acute emergency situations we propose
ject to be entered into such a study without pro- to institute an independent safety committee, un-
viding prospective informed consent (McClure der the auspices of regulatory authorities. Such an
et al., 2003). In another study 11 of 12 stroke independent safety committee, without (financial)
patients stated that, if the patient or family was ties to industry or investigators, offers the best
not able to consent, the treating physician should safeguard for patient protection. The obligation
make the decision for inclusion in an emergency to such a committee is based on the experience of
trial (Blixen and Agich, 2005). a dramatically harmful outcome in some trials
Furthermore, the requirement for all patients to under waiver of consent in other fields of medicine
give written informed consent before enrollment (Freeman, 2001; Lewis et al., 2001).
can result in major selection biases, such that en- Clinical research in emergency situations with-
rolled patients may not be representative of the out prospective informed or PC is ethically chal-
typical patient (Tu et al., 2004). lenging. Severe TBI is without doubt an emergent
Although respect for a patient’s autonomy is a and life-threatening condition and existing therapy
guiding principle in human rights, we doubt very is unsatisfactory. This should qualify severe TBI as
much whether this can be guaranteed by manda- emergency exception to informed consent or de-
tory PC in acute TBI research. A balance should ferred consent for randomized clinical controlled
be sought between optimal patient protection and trials involving pharmacological agents with
research to advance the standards of clinical care promising therapeutic benefit facing short thera-
in emergency situations to the benefit of society. peutic time windows. Randomized placebo-con-
trolled investigations are necessary to determine
the safety and efficacy of new developed agents
Risk– benefit ratio and patient protection under these circumstances. The requirement for
prior written PC causes significant delays in SDA.
In our opinion the balance between risk and ben- With deferred consent or waiver of consent the
efit should be the guiding principle in emergency first dose of the experimental drug can be admin-
research in severe TBI. This also applies to the istered directly after completion of the first diag-
nature and the type of consent procedures. The nostic CT scan, which is very close to the
ethical principle of respect for the autonomy of the experimental therapeutic time window. Randomi-
patient underpinning the informed consent proce- zed controlled phase III trials investigating the
dures is not valid for acutely incapacitated patients safety and efficacy of agents with promising ben-
as TBI victims (Kompanje et al., 2005). Significant efit, conducted in acute emergency situations with
concerns have been raised on the validity and eth- short therapeutic time windows, should allow ran-
ics of PC in acute emergency situations, and the domization under deferred consent or waiver of
required written consent causes a significant delay consent. This is ethically defendable, off course
249

with proper safeguard from an independent safety Kompanje, E.J.O. and Maas, A.I.R. (2004) ‘Treat first, ask
committee. Making progress in knowledge of later?’ Emergency research in acute neurology and neurotrau-
treatment in acute neurological and other inten- matology in the European Union. Intensive Care Med., 30:
sive care conditions is only possible if national 168–169.
Kompanje, E.J.O. and Maas, A.I.R. (2006) Is the Glasgow
regulations and legislations allow waiver of con-
Coma Scale score protected health information? The effect of
sent or deferred consent for clinical trials (Lemaire, new United States regulations (HIPAA) on completion of
2005). As two of us have said before: ‘treat first, screening logs in emergency research trials. Intensive Care
ask later’ is ethically defendable in emergency and Med., 32: 313–314.
intensive care medicine research (Kompanje and Lemaire, F. (2005) Waiving consent for emergency research.
Maas, 2004). Eur. J. Clin. Invest., 35: 287–289.
Lewis, R.J., Berry, D.A., Cryer, H., Fost, N., Krome, R.,
Washington, G.R., Houghton, J., Blue, J.W., Bechhofer, R.,
Cook, T. and Fisher, M. (2001) Monitoring a clinical trial
conducted under the Food and Drug Administration regu-
References lations allowing a waiver of prospective informed consent:
the diaspirin cross-linked hemoglobin traumatic hemorrhagic
Annane, D., Outlin, H., Fisch, C. and Bellissant, E. (2004) The shock efficacy trial. Ann. Emerg. Med., 38: 397–404.
effect of waiving consent on enrolment in a sepsis trial. Inten- Liddell, K., Chamberlain, D., Menon, D.K., Bion, J., Kom-
sive Care Med., 30: 632–637. panje, E.J.O., Lemaire, F., Druml, C., Vrhovac, B.,
Blixen, C.E. and Agich, G.J. (2005) Stroke patients preferences Wiedermann, C.J. and Sterz, F. (2006a) The European Clin-
and values about emergency research. J. Med. Ethics, 31: ical Trial Directive revisited: the VISEAR recommendations.
608–611. Resuscitation, 69: 9–14.
Clifton, G.L., Knudson, P. and McDonald, M. (2002) Waiver Liddell, K., Kompanje, E.J.O., Lemaire, F., Vrhovac, B., Menon,
of consent in studies of acute brain injury. J. Neurotrauma, D.K., Bion, J., Chamberlain, D., Wiedermann, C.J. and
19: 1121–1126. Druml, C. (2006b) Recommendations in relation to the EU
Coppolino, M. and Ackerson, L. (2001) Do surrogate decision Clinical Trials Directive and medical research involving inca-
makers provide accurate consent for intensive care research? pacitated adults. Wien. Klin. Wochenschr., 118: 183–191.
Chest, 119: 603–612. Luce, J.M. (2003) Is the concept of informed consent applicable
CRASH trial management group. (2004) Research in emer- to clinical research involving critically ill patients? Crit. Care
gency situations: with or without relatives consent. Emerg. Med., 31(Suppl): S153–S160.
Med. J., 21: 703. Maas, A.I.R., Kompanje, E.J.O., Slieker, F.J.A. and Stocchetti,
Cuttini, M. (2000) Proxy informed consent in pediatric re- N. (2005) Differences in completion of screening logs bet-
search: a review. Early Hum. Dev., 60: 89–100. ween Europe and the United States in an Emergency phase
European Union. (2001) Directive 2001/20/EC of the European III trial resulting from HIPAA requirements. Ann. Surg.,
Parliament and the Council of 4 April 2001 on the approxi- 241(2): 382–383.
mation of the laws, regulations and administrative provi- Maas, A.I.R., Marmarou, A., Murray, G.D. and Steyerberg,
sions of the Member States relating to the implementation E.W. (2004) Clinical trials in traumatic brain injury: current
of good clinical practice in the conduct of clinical trials on problems and future solutions. Acta Neurochir., 89: 113–118.
medicinal products for human use. Off. J. Eur. Comm., Maas, A.I.R., Murray, G., Henney, H., Kassem, N., Legrand,
L121/34. V., Mangelus, M., Muizelaar, J.P., Stocchetti, N., Knoller,
Freeman, D.B. (2001) Safeguarding patients in clinical trials N. and on behalf of the Pharmos TBI Investigators. (2006)
with high mortality rates. Am. J. Crit. Care Med., 164: Efficacy and safety of dexanabinol in sever traumatic brain
190–192. injury: results of a phase III randomized placebo controlled
Harth, S.C. and Thong, Y.H. (1995) Parental perceptions and clinical trial. Lancet Neurol., 5: 38–45.
attitudes about informed consent in clinical research involv- Maas, A.I.R., Steyerberg, E.W. and Murray, G.D. (1999) Why
ing children. Soc. Sci. Med., 40: 1573–1577. have recent trials of neuroprotective agents in head injury
Hoff, J.T. (1986) Cerebral protection. J. Neurosurg., 65: failed to show convincing efficacy? A pragmatic analysis and
579–591. theoretical considerations. Neurosurgery, 44: 1286–1298.
Hsieh, M., Dailey, M.W. and Callaway, C.W. (2001) Surrogate Mason, S.A. and Allmark, P.J. (2000) Obtaining informed
consent by family members for out-of-hospital cardiac arrest consent to neonatal randomised controlled trials: interviews
research. Acad. Emerg. Med., 8: 851–853. with parents and clinicians in the Euricon study. Lancet, 356:
Kompanje, E.J.O., Maas, A.I.R., Hilhorst, M.T., Slieker, 2045–2051.
F.J.A. and Teasdale, G.M. (2005) Ethical considerations on McClure, K.B., Delorio, N.M., Gunnels, M.D., Ochsner, M.J.,
consent procedures for emergency research in severe and Biros, M.H. and Schmidt, T.A. (2003) Attitudes of emer-
moderate traumatic brain injury. Acta Neurochir. (Wien.), gency department patients and visitors regarding emergency
147: 633–640. exception from informed consent in resuscitation research,
250

community consultation and public notification. Acad. Em- Silverman, H.J., Druml, C., Lemaire, F. and Nelson, R. (2004)
erg. Med., 10: 352–359. The European Union Directive and the protection of inca-
McGarry, L.J., Thompson, D., Millham, F.H., Cowell, L., pacitated subjects in research: an ethical analysis. Intensive
Snyder, P.J., Lenderking, W.R. and Weinstein, M.C. (2002) Care Med., 30: 1723–1729.
Outcomes and costs of acute treatment of traumatic brain Sugarman, J. (2000) Is the emperor really wearing new clothes?
injury. J. Trauma, 53: 1152–1159. Informed consent for acute coronary syndromes. Am. Heart
Roupie, E., Santin, A., Boulme, R., Wartel, J.S., Lepage, E., J., 140: 2–3.
Lemaire, F., Lejonc, J.L. and Montagne, O. (2000) Patients Tu, J.V., Willison, D.J., Silver, F.L., Fang, J., Richards, J.A.,
preferences concerning medical information and surrogacy: Laupacis, A. and Kapral, M.K. (2004) Impracticability of
results of a prospective study in a French emergency depart- informed consent in the registry of the Canadian stroke net-
ment. Intensive Care Med., 26: 52–56. work. NEJM, 350: 1414–1421.
Shohami, E. (1995) Long-term effect of HU-211, a novel Williams, B.F., French, J.K. and White, H.D. (2003) Informed
competitive NMDA antagonist, on motor and memory consent during the clinical emergency of acute myocardial
functions after closed head injury in the rat. Brain Res., infarction (HERO-2 consent substudy): a prospective obser-
674: 55–62. vational study. Lancet, 361: 918–922.
SECTION V

Emerging Topics in CNS Trauma


This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 18

Experimental models of repetitive brain injuries

John T. Weber

Department of Neuroscience, Erasmus Medical Centre, Rotterdam, The Netherlands

Abstract: Repetitive traumatic brain injury (TBI) occurs in a significant portion of trauma patients, es-
pecially in specific populations, such as child abuse victims or athletes involved in contact sports (e.g.
boxing, football, hockey, and soccer). A continually emerging hypothesis is that repeated mild injuries may
cause cumulative damage to the brain, resulting in long-term cognitive dysfunction. The growing attention
to this hypothesis is reflected in several recent experimental studies of repeated mild TBI in vivo. These
reports generally demonstrate cellular and cognitive dysfunction after repetitive injury using rodent TBI
models. In some cases, data suggests that the effects of a second mild TBI may be synergistic, rather than
additive. In addition, some studies have found increases in cellular markers associated with Alzheimer’s
disease after repeated mild injuries, which demonstrates a direct experimental link between repetitive TBI
and neurodegenerative disease. To complement the findings from humans and in vivo experimentation, my
laboratory group has investigated the effects of repeated trauma in cultured brain cells using a model of
stretch-induced mechanical injury in vitro. In these studies, hippocampal cells exhibited cumulative damage
when mild stretch injuries were repeated at either 1-h or 24-h intervals. Interestingly, the extent of damage
to the cells was dependent on the time between repeated injuries. Also, a very low level of stretch, which
produced no cell damage on its own, induced cell damage when it was repeated several times at a short
interval (every 2 min). Although direct comparisons to the clinical situation are difficult, these types of
repetitive, low-level, mechanical stresses may be similar to the insults received by certain athletes, such as
boxers, or hockey and soccer players. This type of in vitro model could provide a reliable system in which to
study the mechanisms underlying cellular dysfunction following repeated injuries. As this area of TBI
research continues to evolve, it will be imperative that models of repetitive injury replicate injuries in
humans as closely as possible. For example, it will be important to model appropriately concussive episodes
versus even lower level injuries (such as those that might occur during boxing matches). Suitable inter-
injury intervals will also be important parameters to incorporate into models. Additionally, it will be crucial
to design and utilize proper controls, which can be more challenging than experimental approaches to single
mild TBI. It will also be essential to combine, and compare, data derived from in vitro experiments with
those conducted with animals in vivo. These issues, as well as a summary of findings from repeated TBI
research, are discussed in this review.

Keywords: hippocampus; in vivo models; in vitro; mild injury; repeated injury; TBI; trauma

Corresponding author. Present address: School of Pharmacy, Introduction


Memorial University of Newfoundland, Health Sciences
Centre, St. John’s, NL, Canada. Tel.: +1-709-777-7022; Repetitive injuries occur in a considerable portion
Fax: +1-709-777-7044; E-mail: jweber@mun.ca of individuals who experience a traumatic brain

DOI: 10.1016/S0079-6123(06)61018-2 253


254

injury (TBI). Child abuse victims, and in particu- Experimental studies of repeated mild TBI in vivo
lar, victims of shaken baby syndrome, are often
subjected to multiple injuries to the head (Shannon It should be made clear that when discussing ex-
et al., 1998). Spousal abuse also contributes to the perimental studies of repetitive TBI in vivo, this
number of repeated brain injuries (Roberts et al., does not include studies of secondary insults, such
1990). However, many injuries of these types go as a mechanical insult to the head followed by a
unreported, and it is difficult to assess how many duration of ischemia or glutamate exposure. Re-
insults a patient may have suffered. Arguably, peated TBI experimentation can be defined as an
athletes represent the largest group of patients that initial mechanical injury to the head followed by
are at risk for experiencing repeated brain injuries another mechanical insult to the head of the same
(Kelly and Rosenberg, 1997; Kelly, 1999; Powell or different degree. Based on these criteria, little
and Barber-Foss, 1999). In fact, sports and recre- attention was given to these types of experiments
ational activities contribute to approximately 10% before the year 2000, with only a few repetitive
of all mild injuries, and these individuals are at injury studies being published (Olsson et al., 1971;
high risk of experiencing a second TBI (Woo and Weitbrecht and Noetzel, 1976; Kanayama et al.,
Thoidis, 2000). Also, in comparison to child or 1996). Several additional in vivo studies of repeated
spousal abuse victims, there is generally better injuries in rodents have now been conducted in the
documentation of how many brain injuries an in- past decade (Allen et al., 2000; Laurer et al., 2001;
dividual has sustained due to recreational or sports DeFord et al., 2002; Uryu et al., 2002; Conte et al.,
related activities, making this population easier to 2004; Creeley et al., 2004; Raghupathi et al., 2004;
study. Dobrowolska and Gibson, 2005; Gibson and
The idea that multiple head injuries in athletes Schalles, 2005; Longhi et al., 2005; Yoshiyama et
could lead to clinical problems has long been sug- al., 2005). All of these repeated mild injury studies
gested. For example, the development of dementia were conducted using rodent models of TBI with
pugilistica in professional boxers is believed to be the exception of the study by Raghupathi et al.
caused by the multiple hits to the head that boxers (2004), which reported a pediatric model of re-
endure over the course of their career (Jordan, peated injury conducted in pigs.
2000). Also, recent evidence has shown that the Repetitive TBI generally occurs at a mild level,
number of concussions is inversely related to per- therefore experimental models have been used
formance on several neuropsychological tests in which are minimally invasive and do not require a
soccer players (Matser et al., 1999, 2001) and craniotomy, such as weight drop models. The
jockeys who have experienced multiple concuss- models must also be administered at a level that
ions generally display more cognitive dysfunctions produces minimal, or preferably, no fatality. Indi-
than those who have had a single injury (Wall viduals who have suffered from a mild TBI often
et al., 2006). complain of cognitive difficulties post-injury.
When studying repetitive brain trauma in ath- Therefore, repeated injury studies usually evalu-
letes, we can gain much information about the ate cognitive function, for example using the
pathology and progress of such injuries from the Morris water maze (MWM) test, as well as the
injured athletes themselves, for example by meas- extent of cell abnormalities in the cortex and hip-
uring changes in cognitive and motor perform- pocampus. The hippocampus in particular has re-
ance. However, these injuries are generally at a ceived significant attention in the study of repeated
mild level, and therefore, except in rare cases when mild TBI, because it plays a critical role in certain
athletes die as a result of the insult, we cannot types of learning and aspects of memory storage.
assess the changes that have occurred in the brain Experimental and clinical data have demonstrated
at the cellular and sub-cellular levels. In order to not only the importance of this brain region in
compile this type of information, we must turn to learning and memory, but also that the hippo-
experimental models of TBI. campus is uniquely vulnerable to injury, even after
255

mild brain trauma (Lyeth et al., 1990; Lowenstein Repetitive injury and neurodegenerative disease
et al., 1992). In a study by DeFord et al. (2002),
repeated mild injuries were administered to mice It has been noted for some time that there is a
(four times every 24 h), followed by MWM testing correlation between the occurrence of TBI and
and histological analysis. Significant learning defi- the further development of neurodegenerative dis-
cits were found after repeated injuries, which were ease later in life. In fact, TBI is considered to be
not evident after a single injury. These deficits oc- one of the most robust risk factors for developing
curred even in absence of cell death within the Alzheimer’s disease (AD) (Szczygielski et al.,
cortex and hippocampus. These studies were ex- 2005). There is also evidence that genetic predis-
tended by Gibson and Schalles (2005) using the position may increase one’s risk of developing
same injury model; they demonstrated impaired AD, such as possession of the apolipoprotein E e4
function in the Barnes circular maze after multiple, allele. A phenomenon known as chronic TBI oc-
but not single injuries. Cognitive deficits after curs in a significant amount of professional boxers
multiple mild TBIs (using MWM analysis) have (Jordan, 2000), with the most serious form,
also been demonstrated in a similar study using a dementia pugilistica, resulting in severe motor
weight drop model (Creeley et al., 2004). and cognitive dysfunctions. It is now known that
In order to most closely mimic the type of insult the pathology of AD and dementia pugilistica are
that athletes may receive, Laurer et al. (2001) de- quite similar (Geddes et al., 1999; Schmidt et al.,
veloped an injury regimen that they described as 2001). Although the epidemiological data are
‘‘concussive’’; this model was used for subsequent quite strong, very little has been done to make
studies as well (Uryu et al., 2002; Conte et al., 2004; the mechanistic link between repeated mild TBI
Longhi et al., 2005). In an assessment of cognitive and the development of either AD or dementia
and motor function after repeated injury in mice, pugilistica.
Laurer et al. (2001) found that the brain was more A few studies have found increases in cellular
vulnerable to a second insult if the second injury markers associated with AD after repeated mild
occurred 24 h after the first. Even though no cog- injuries (Kanayama et al., 1996; Uryu et al., 2002;
nitive deficits were demonstrated in mice receiving Conte et al., 2004; Dobrowolska and Gibson,
repeated injuries, there was a decrease in motor 2005). For example, Kanayama et al. (1996)
function and some neuronal loss. The authors also showed an increase in tau immunoreactivity in
stated that the effects of a second mild TBI may be neurons and Dobrowolska and Gibson (2005) re-
synergistic, rather than additive. To analyze further ported increased amyloid precursor protein fol-
the effects of lengthening the inter-injury interval, lowing multiple injuries. Uryu et al. (2002) and
Longhi et al. (2005) investigated repetitive injuries Conte et al. (2004) used a transgenic mouse model
three, five, and seven days apart. Animals that re- of AD-like amyloidosis, and demonstrated that
ceived repeated injuries three or five days apart ex- amyloid levels and deposits increased in the brain
hibited cognitive dysfunction not evident in sham after repeated injuries, but not after a single insult.
animals or those injured only once. However, no Yoshiyama et al. (2005) utilized the same trans-
deficits were observed when the injury interval was genic mouse model and analyzed the effects of four
extended to seven days. This experimental evidence injuries given per day, once a week, for a period of
— that the brain can recover from a first injury, four weeks, in order to simulate dementia pugi-
given a sufficient amount of time — is appealing, listica. After nine months, only one mouse showed
especially in relation to establishing ‘‘return-to- pathology consistent with this syndrome. Al-
play’’ guidelines for athletes. Overall, the evidence though the findings were not robust, this is per-
from in vivo experimental models suggests that re- haps the only study to attempt to model dementia
petitive mild TBI causes more cognitive and cellular pugilistica, and more studies are warranted.
dysfunction than a single injury, if the brain is not The overall findings of these studies are quite
given a sufficient amount of time to recover. important, because they can demonstrate a direct
256

experimental link between repeated mild TBI and protein (a biomarker of injury commonly em-
the development of AD-like pathology. Generally, ployed in the clinic) than cultures that received a
it takes years before the onset of symptoms of ne- second injury at 24 h. Cultures injured 24 h apart
urodegenerative disorders after an individual has also exhibited less staining with the intravital dye,
experienced a TBI. Therefore, it requires an ex- propidium iodide, than those injured 1 h apart. As
tremely long amount of time to gather this type of shown in some in vivo studies, these findings sug-
epidemiological data from the human population. gest that a level of injury, which produces meas-
This area of research, in particular, is where ex- urable damage or dysfunction on its own, may
perimental models could truly help decipher the cause cumulative damage if repeated within
mechanisms by which neurodegenerative disease a certain time frame (Laurer et al., 2001; Longhi
may be triggered by repetitive brain injury. et al., 2005).
We also investigated the effects of a very low
level of stretch, which produced no overt cell dam-
Studying repeated injuries in vitro age (Slemmer and Weber, 2005). This ‘‘subthresh-
old’’ level of stretch did not cause significant
Several in vitro approaches have now been devel- damage or death, even when it was repeated at a
oped to study traumatic injury, which utilize cells 1-h interval. However, this low level of stretch did
grown in culture (Morrison et al., 1998; Weber, induce cell damage when it was repeated several
2004; LaPlaca et al., 2005; also see Chapter 3 by times at a short interval (every 2 min), indicated by
Spaethling et al., in this volume). My laboratory increased propidium iodide staining, neuronal
group utilizes a model of stretch-induced mechan- loss, and an increase in NSE release. Although
ical injury in vitro originally developed by Ellis direct comparisons to the clinical situation are
et al. (1995). We have characterized this stretch difficult, these types of repetitive, low-level, me-
injury model in cell cultures composed of neurons chanical stresses may be similar to the insults re-
and glia from murine hippocampus (Slemmer ceived by certain athletes, such as boxers and
et al., 2002; Slemmer and Weber, 2005), cortex hockey and soccer players (Matser et al., 1998,
(Engel et al., 2005), and cerebellum (Slemmer 1999; Jordan, 2000; Webbe and Ochs, 2003;
et al., 2004). Wennberg and Tator, 2003). This type of in vitro
To complement the findings from humans and model could comprise a reliable system in which to
from in vivo experimentation, we have recently study the mechanisms underlying cellular dysfunc-
conducted a series of studies investigating the tion following repeated injuries. In addition, this
effects of repeated trauma on hippocampal cells approach could provide a means for relatively
(Slemmer et al., 2002; Slemmer and Weber, 2005). rapid screening of potential therapeutic strategies
In these studies, we utilized a mild level of stretch for both single and repeated mild TBI.
injury that produces some measurable damage to
cells when administered a single time. When mild
stretch injuries were repeated at either 1-h or 24-h The phenomenon of preconditioning
intervals, cells exhibited cumulative damage. For
example, cultures that received a second insult Several studies have indicated that an initial, very
displayed a significant loss of neurons not evident mild insult to either cultured cells or to the brain
in cultures that received only one injury. Addi- itself, may provide some protection from a second,
tionally, cultures injured twice released a signifi- more severe insult, a finding that has been termed
cant level of neuron specific enolase (NSE), which ‘‘preconditioning’’. Ischemic preconditioning, in
was not observed in cultures injured a single time. which a brief exposure to ischemia renders the
Interestingly, the extent of damage to the cells was brain more resistant to subsequent longer periods
dependent on the time between repeated injuries. of ischemia, has been well described (for review,
For example, cultures that received a second insult see Schaller and Graf, 2002). There is also evidence
1 h after the first injury released more S-100B of preconditioning cross-tolerance. For example,
257

brief ischemia lessens damage following TBI in Future directions


vivo (Pérez-Pinzón et al., 1999). Another interest-
ing phenomenon is that heat acclimation (chronic Experimental evidence suggests that animals can
exposure to moderate heat) can also provide re- demonstrate cognitive deficits and cellular dys-
sistance to TBI (see Chapter 25 by Shein et al., in function after repetitive mild TBI, even though
this volume). the injury may not necessarily lead to cell death
In our in vitro studies, we observed a novel form (Kanayama et al., 1996; DeFord et al., 2002).
of mechanical preconditioning. When hippocam- Therefore, rather than trying to prevent cells from
pal cultures were administered a subthreshold level dying after repeated injuries, it is probably more
of stretch 24 h prior to a mild stretch, there was a important to learn how to restore normal cellular
significant decrease in released S-100B protein physiology after a traumatic episode. Combining
compared to cultures that were injured at a mild studies at the cellular and behavioral levels is es-
level alone (Slemmer and Weber, 2005). This ob- sential, and one particular area of interest is the
servation suggests some form of protection initi- evaluation of the effects of repeated TBI on synap-
ated by this low level of stretch. A similar finding tic plasticity in the hippocampus. This ability of
in vivo was reported by Allen et al. (2000). In their neurons to undergo changes in synaptic strength,
study, rats received a series of mild injuries spaced such as long-term potentiation (LTP), is postu-
three days apart using a weight drop model. Some lated to be a cellular correlate of learning and
of these animals received a severe injury after the memory (Bliss and Collingridge, 1993; Malenka
repetitive mild injuries. Motor function deficits and Nicoll, 1999). Several studies have reported
were evident in severely injured animals, but not in impaired hippocampal LTP after TBI in vivo
animals that received repeated mild injuries or re- (Albensi, 2001; Weber, 2004). One area of future
peated mild injuries followed by a severe injury. research could focus on restoring mechanisms of
This last observation suggests a preconditioning synaptic plasticity after injury (such as LTP), as
effect. well as correlated hippocampal-mediated behavi-
The most important question now is how do we oral tasks.
utilize this information? One can imagine the ethi- The hippocampus shares neuronal projections
cal implications of suggesting to people that a mild with areas of the cerebral cortex, which undoubt-
insult to their brains may in fact protect them from edly also contributes to memory formation and
worse insults in the future. We still have much to storage. Indeed, alterations in synaptic plasticity
learn about preconditioning. For example, what is may also occur directly in the cortex after repeated
the threshold for mechanical insults between ini- mild TBI. Therefore, although the hippocampus
tiating protective versus damaging mechanisms in may play a central role in the cognitive dysfunc-
the brain? A clearer understanding of the mecha- tion observed after mild TBI, it is important not to
nisms by which this protection is elicited holds overlook contributions from other brain areas as
potential for the management of mild TBI. The well. Along these lines, since some repeated injury
fact that different stressors can protect the brain studies demonstrate motor dysfunction, it may
from TBI (i.e. cross-tolerance) suggests that the also be appropriate to investigate cellular physiol-
same or similar mechanisms are responsible for the ogy and synaptic plasticity in the cerebellum
endogenous protection. Increasing the expression (Hansel et al., 2001; Weber et al., 2003; Slemmer
of these protective systems could not only be a et al., 2005) after repetitive TBI.
reliable way for managing mild TBI, but also The utilization of proper parameters for re-
could provide resistance in individuals who may be peated injury studies may be even more crucial
at risk of sustaining an additional head injury, than deciding on appropriate research directions.
such as athletes. Both in vivo and in vitro models For example, what are the best inter-injury inter-
could provide reliable systems in which to study val, or intervals, to use? Although 24 h between
the mechanisms underlying the preconditioning injuries is the most common (and perhaps practi-
phenomenon. cal) interval in the laboratory (Weitbrecht and
258

Noetzel, 1976; Laurer et al., 2001; DeFord et al., (Yoshiyama et al., 2005), the number of injuries
2002; Uryu et al., 2002; Conte et al., 2004; Creeley should certainly be increased.
et al., 2004; Dobrowolska and Gibson, 2005; What are the proper controls and endpoints to
Gibson and Schalles, 2005; Yoshiyama et al., use for repeated injury studies? For in vivo studies
2005), is it the most appropriate in mimicking analyzing the effects of a single TBI, the issue of
what occurs in humans? Also, how many injuries controls is fairly straightforward. Sham animals
should a researcher administer? If one is attempt- are treated at an equivalent time as injured ani-
ing to model concussive episodes, then two, three, mals, and the analysis, cellular or behavioral, is
or four may be enough, as this may closely mimic a also performed at the same timepoint. However,
true situation, especially with athletes. However, when comparing uninjured animals to animals
when attempting to recreate dementia pugilistica that have received more than one injury, what is

Fig. 1. Issues for consideration when designing repeated TBI experiments (i.e. choosing proper timepoints for controls and behavioral/
tissue analysis). (A) Hypothetical timeline for two injuries 24 h apart. (B) Timeline for greater than two injuries. T ¼ time.
259

the proper comparison? For example, if an animal Abbreviations


receives an injury on day one, an additional injury
on day two, and analysis takes place on day three, AD Alzheimer’s disease
does one compare the data with sham animals LTP long-term potentiation
from day one, or from day two (or both, see MWM Morris water maze
Fig. 1A). The issue is further complicated when NSE neuron specific enolase
comparing repeatedly injured animals to animals TBI traumatic brain injury
that have received a single TBI. If the comparison
concerns animals that undergo four injuries or a
single injury, are the single insult animals injured
at the same time as injury one in the repeated
group, or at the same time as the fourth injury (see Acknowledgments
Fig. 1B)? This decision will also affect the endpoint
as well. For example, if animals or tissue are The in vitro work discussed in this manuscript was
analyzed one day after the fourth injury, then four supported by grants from the Research Council
days will have passed for the single injury group if for Earth and Life Sciences (ALW) and Research
those animals were injured on day one. This Council for Medical Sciences (MW) with financial
difference in time could affect the observations. aid from the Netherlands Organization for Scien-
We have struggled with these issues when design- tific Research (NWO), a Breedtestrategie subsidie
ing our in vitro experiments. One could argue that from Erasmus Medical Centre, and funding from
if a long enough period of time passes after the Hersenstichting Nederland. I would also like to
injuries, such as weeks or months, then the effect thank Jennifer E. Slemmer for editorial assistance.
of when the single insult animals were injured will
be negligible. Admittedly, this would be more
References
proper for comparison to the human situation in
which the effects of mild TBI can be manifested for Albensi, B.C. (2001) Models of brain injury and alterations in
weeks, months, or even years. However, this is of- synaptic plasticity. J. Neurosci. Res., 65: 279–283.
ten not practical for many laboratories, as the Allen, G.V., Gerami, D. and Esser, M.J. (2000) Conditioning
costs of housing animals for months can at times effects of repetitive mild neurotrauma on motor function in
an animal model of focal brain injury. Neuroscience, 99:
be prohibitive. Also, conducting long-term experi-
93–105.
ments in vitro is limited, since the cells generally Bliss, T.V. and Collingridge, G.L. (1993) A synaptic model of
remain viable for only a few weeks. This raises a memory: long-term potentiation in the hippocampus. Nature,
critical point as to the relevance of repeated injury 361: 31–39.
studies in vitro. I strongly believe that in vitro ex- Conte, V., Uryu, K., Fujimoto, S., Yao, Y., Rokach, J., Long-
hi, L., Trojanowski, J.Q., Lee, V.M.-Y., McIntosh, T.K. and
periments can deliver information about the cel-
Pratico, D. (2004) Vitamin E reduces amyloidosis and im-
lular mechanisms of repeated injury that are proves cognitive function in Tg2576 mice following repetitive
difficult to obtain in vivo, and that it is essential concussive brain injury. J. Neurochem., 90: 758–764.
to combine data derived from in vitro experiments Creeley, C.E., Wozniak, D.F., Bayly, P.V., Olney, J.W. and
with those conducted with animals in vivo. How- Lewis, L.M. (2004) Multiple episodes of mild traumatic brain
injury result in impaired cognitive performance in mice.
ever, I am unsure how to directly compare the
Acad. Emerg. Med., 11: 809–819.
data. For example, is a 24 h injury interval in vitro DeFord, S.M., Wilson, M.S., Rice, A.C., Clausen, T., Rice,
equivalent to 24 h in vivo? The more consensus L.K., Barabnova, A., Bullock, R. and Hamm, R.J. (2002)
that exists on these issues with individuals who Repeated mild brain injuries result in cognitive impairment in
conduct repeated injury studies, the easier it will be B6C3F1 mice. J. Neurotrauma, 19: 427–438.
Dobrowolska, J.A. and Gibson, C.J. (2005) Immunohisto-
to compare the data, and the stronger a case can
chemical analysis of amyloid precursor protein in the hippo-
be made for showing unequivocally, that repeated campus and cortex of mice following repeated mild traumatic
mild TBI could lead to long-term dysfunction in brain injuries. Soc. Neurosci. Abst., 31: Program Number
humans. 334.7.
260

Ellis, E.F., McKinney, J.S., Willoughby, K.A., Liang, S. and Matser, E.J.T., Kessels, A.G.H., Lezak, M.D., Jordan, B.D.
Povlishock, J.T. (1995) A new model for rapid stretch- and Troost, J. (1999) Neuropsychological impairment in am-
induced injury of cells in culture: characterization of the ateur soccer players. JAMA, 282: 971–973.
model using astrocytes. J. Neurotrauma, 12: 325–339. Matser, J.T., Kessels, A.G.H., Jordan, B.D., Lezak, M.D. and
Engel, D.C., Slemmer, J.E., Vlug, A.S., Maas, A.I. and Weber, Troost, J. (1998) Chronic traumatic brain injury in profes-
J.T. (2005) Combined effects of mechanical and ischemic in- sional soccer players. Neurology, 51: 791–796.
jury to cortical cells: secondary ischemia increases damage Matser, J.T., Kessels, A.G.H., Lezak, M.D. and Troost, J.
and decreases effects of neuroprotective agents. Neurophar- (2001) A dose-response relation of headers and concussions
macology, 49: 985–995. with cognitive impairment in professional soccer players.
Geddes, J.F., Vowles, G.H., Nicoll, J.A. and Revesz, T. (1999) J. Clin. Exp. Neuropsychol., 23: 770–774.
Neuronal cytoskeletal changes are an early consequence of Morrison III, B., Saatman, K.E., Meaney, D.F. and McIntosh,
repetitive head injury. Acta Neuropathol. (Berl.), 98: T.K. (1998) In vitro central nervous system models of
171–178. mechanically induced trauma: a review. J. Neurotrauma, 15:
Gibson, C.J. and Schalles, M.D. (2005) Barnes circular maze 911–928.
deficits in repeated mildly brain injured mice. Soc. Neurosci. Olsson, Y., Rinder, L., Lindgren, S. and Stalhammar, D. (1971)
Abst., 31: Program Number 334.6. Studies on vascular permeability changes in experimental
Hansel, C., Linden, D.J. and D’Angelo, E. (2001) Beyond par- brain concussion. 3. A comparison between the effects of
allel fiber LTD: the diversity of synaptic and non-synaptic single and repeated sudden mechanical loading of the brain.
plasticity in the cerebellum. Nat. Neurosci., 4: 467–475. Acta Neuropathol. (Berl.), 19: 225–233.
Jordan, B.D. (2000) Chronic traumatic brain injury associated Pérez-Pinzón, M.A., Alonso, O., Kraydieh, S. and Dietrich,
with boxing. Semin. Neurol., 20: 179–185. W.D. (1999) Induction of tolerance against traumatic
Kanayama, G., Takeda, M., Niigawa, H., Ikura, Y., Tamii, H., brain injury by ischemic preconditioning. Neuroreport, 10:
Taniguchi, N., Kudo, T., Miyamae, Y., Morihara, T. and 2951–2954.
Nishimura, T. (1996) The effects of repetitive mild brain in- Powell, J.W. and Barber-Foss, K.D. (1999) Traumatic brain
jury on cytoskeletal protein and behavior. Methods Find. injury in high school athletes. JAMA, 282: 958–963.
Exp. Clin. Pharmacol., 18: 105–115. Raghupathi, R., Mehr, M.F., Helfaer, M.A. and Margulies,
Kelly, J.P. (1999) Traumatic brain injury and concussion in S.S. (2004) Traumatic axonal injury is exacerbated following
sports. JAMA, 282: 989–991. repetitive closed head injury in the neonatal pig. J. Neuro-
Kelly, J.P. and Rosenberg, J.H. (1997) Diagnosis and manage- trauma, 21: 307–316.
ment of concussion in sports. Neurology, 48: 575–580. Roberts, G.W., Whitwell, H.L., Acland, P.R. and Bruton, C.J.
LaPlaca, M.C., Cullen, D.K., McLoughlin, J.J. and Cargill II, (1990) Dementia in a punch-drunk wife. Lancet, 335:
R.S. (2005) High rate shear strain of three-dimensional neu- 918–919.
ral cell cultures: a new in vitro traumatic brain injury model. Schaller, B. and Graf, R. (2002) Cerebral ischemic precondi-
J. Biomech., 38: 1093–1105. tioning. An experimental phenomenon or a clinical impor-
Laurer, H.L., Bareyre, F.M., Lee, V.M., Trojanowski, J.Q., tant entity of stroke prevention? J. Neurol., 249: 1503–1511.
Longhi, L., Hoover, R., Saatman, K.E., Raghupathi, R., Schmidt, M.L., Zhukareva, V., Newell, K.L., Lee, V.M.-Y. and
Hoshino, S., Grady, S.M. and McIntosh, T.K. (2001) Mild Trojanowski, J.Q. (2001) Tau isoform profile and phosphoryl-
head injury increasing the brain’s vulnerability to a second ation state in dementia pugilistica recapitulate Alzheimer’s
concussive impact. J. Neurosurg., 95: 859–870. disease. Acta Neuropathol. (Berl.), 101: 518–524.
Longhi, L., Saatman, K.E., Fujimoto, S., Raghupathi, R., Shannon, P., Smith, C.R., Deck, J., Ang, L.C., Ho, M. and
Meaney, D.F., Davis, J., McMillan, B.S.A., Conte, V., Becker, L. (1998) Axonal injury and the neuropathology of
Laurer, H.L., Stein, S., Stocchetti, N. and McIntosh, T.K. shaken baby syndrome. Acta Neuropathol. (Berl.), 95:
(2005) Temporal window of vulnerability to repetitive 625–631.
experimental concussive brain injury. Neurosurgery, 56: Slemmer, J.E., De Zeeuw, C.I. and Weber, J.T. (2005) Don’t get
364–374. too excited: mechanisms of glutamate-mediated Purkinje cell
Lowenstein, D.H., Thomas, M.J., Smith, D.H. and McIntosh, death. Prog. Brain Res., 148: 367–390.
T.K. (1992) Selective vulnerability of dentate hilar neurons Slemmer, J.E., Matser, E.J.T., De Zeeuw, C.I. and Weber, J.T.
following traumatic brain injury: a potential mechanistic link (2002) Repeated mild injury causes cumulative damage to
between head trauma and disorders of the hippocampus. hippocampal cells. Brain, 125: 2699–2709.
J. Neurosci., 12: 4846–4853. Slemmer, J.E. and Weber, J.T. (2005) The extent of damage
Lyeth, B.G., Jenkins, L.W., Hamm, R.J., Dixon, C.E., Phillips, following repeated injury to hippocampal cells is dependent
L.L., Clifton, G.L., Young, H.F. and Hayes, R.L. (1990) on the severity of insult and inter-injury interval. Neurobiol.
Prolonged memory impairment in the absence of hippocam- Dis., 18: 421–431.
pal cell death following traumatic brain injury in the rat. Slemmer, J.E., Weber, J.T. and De Zeeuw, C.I. (2004) Cell
Brain Res., 526: 249–258. death, glial protein alterations and elevated S-100b protein in
Malenka, R.C. and Nicoll, R.A. (1999) Long-term potentiation cerebellar cell cultures following mechanically induced
— a decade of progress? Science, 285: 1870–1874. trauma. Neurobiol. Dis., 15: 563–572.
261

Szczygielski, J., Mautes, A., Steudel, W.I., Falkai, P., Bayer, calcium transients in Purkinje cell dendrites. Proc. Natl.
T.A. and Wirths, O. (2005) Traumatic brain injury: cause or Acad. Sci. U.S.A., 100: 2878–2883.
risk of Alzheimer’s disease? A review of experimental studies. Weitbrecht, W.U. and Noetzel, H. (1976) Autoradiographic
J. Neural Transm., 112: 1547–1564. investigations in repeated experimental brain concussion
Uryu, K., Laurer, H., McIntosh, T., Pratico, D., Martinez, D., (author’s transl.) [Article in German]. Arch. Psychiatr.
Leight, S., Lee, V.M.-Y. and Trojanowski, J.Q. (2002) Re- Nervenkr., 223: 59–68.
petitive mild brain trauma accelerates Ab deposition, lipid Wennberg, R.A. and Tator, C.H. (2003) National Hockey
peroxidation, and cognitive impairment in a transgenic mouse League reported concussions, 1986–87 and 2001–02. Can. J.
model of Alzheimer amyloidosis. J. Neurosci., 22: 446–454. Neurol. Sci., 30: 206–209.
Wall, S.E., Williams, W.H., Cartwright-Hatton, S., Kelly, T.P., Woo, B.H. and Thoidis, G. (2000) Epidemiology of traumatic
Murray, J., Murray, M., Owen, A. and Turner, M. (2006) brain injury. In: Woo B.H. and Nesathurai S. (Eds.), The
Neuropsychological dysfunction following repeat concussions Rehabilitation of People with Traumatic Brain Injury. Black-
in jockeys. J. Neurol. Neurosurg. Psychiatry, 77: 518–520. well Science, Malden, pp. 13–17.
Webbe, F.M. and Ochs, S.R. (2003) Recency and frequency of Yoshiyama, Y., Uryu, K., Higuchi, M., Longhi, L., Hoover, R.,
soccer heading interact to decrease neurocognitive perform- Fujimoto, S., McIntosh, T., Lee, V.M.-Y. and Trojanowski,
ance. App. Neurol., 10: 31–41. J.Q. (2005) Enhanced neurofibrillary tangle formation, cer-
Weber, J.T. (2004) Calcium homeostasis following traumatic ebral atrophy, and cognitive deficits induced by repetitive
neuronal injury. Curr. Neurovasc. Res., 1: 151–171. mild brain injury in a transgenic tauopathy mouse model.
Weber, J.T., De Zeeuw, C.I., Linden, D.J. and Hansel, C. J. Neurotrauma, 22: 1134–1141.
(2003) Long-term depression of climbing fiber-evoked
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 19

Minor traumatic brain injury in sports: a review in


order to prevent neurological sequelae

Nicola Biasca1, and William L. Maxwell2

1
Clinic of Orthopaedic, Sports Medicine and Traumatology, Department of Surgery, Spital Oberengadin, CH-7503
Samedan/St. Moritz, Switzerland
2
Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK

Abstract: Minor traumatic brain injury (mTBI) is caused by inertial effects, which induce sudden rotation
and acceleration forces to and within the brain. At less severe levels of injury, for example in mTBI, there is
probably only transient disturbance of ionic homeostasis with short-term, temporary disturbance of brain
function. With increased levels of severity, however, studies in animal models of TBI and in humans have
demonstrated focal intra-axonal alterations within the subaxolemmal, neurofilament and microtubular
cytoskeletal network together with impairment of axoplasmic transport. These changes have, until very
recently, been thought to lead to progressive axonal swelling, axonal detachment or even cell death over a
period of hours or days, the so-called process of ‘‘secondary axotomy’’. However, recent evidence has
suggested that there may be two discrete pathologies that may develop in injured nerve fibers. In the TBI
scenario, disturbances of ionic homeostasis, acute metabolic changes and alterations in cerebral blood flow
compromise the ability of neurons to function and render cells of the brain increasingly vulnerable to the
development of pathology. In ice hockey, current return-to-play guidelines do not take into account these
new findings appropriately, for example allow returning to play in the same game. It has recently been
hypothesized that the processes summarized above may predispose brain cells to assume a vulnerable state
for an unknown period after mild injury (mTBI). Therefore, we recommend that any confused player with
or without amnesia should be taken off the ice and not be permitted to play again for at least 72 h.

Keywords: minor traumatic brain injury, mTBI; neuromechanics; neuropathology; neurobiology and
associated neurometabolic changes of mTBI; process of delayed axotomy; vulnerability of cells in the brain;
return to play guidelines; ice hockey

Introduction nervous system. This is especially true in ice


hockey, which has many inherent features that
Ice hockey is one of the speediest, competitive may predispose players to injury, including high
contact team sports involving both rapid changes acceleration–deceleration, rapid changes of direc-
of direction of the whole body and controlled ag- tion, shooting, body checking and a low friction
gression. Athletes are therefore exposed to an in- ice surface. Before the introduction of full-face
creased risk of a high-velocity collision that may masks and helmets, head injuries, including inju-
result in a sports-related injury to the central ries to the face, scalp and brain, accounted for at
least 50% of all serious injuries in ice hockey
Corresponding author. Tel.: +41-81-851-85-15; Fax: +41-81- (Pashby, 1993). Indeed, before the use of any
851-85-16; E-mail: Biasca@medicmotion.com type of helmet and before the mandatory use of

DOI: 10.1016/S0079-6123(06)61019-4 263


264

standardized helmets, in the early 1960s, fatalities the brain is exposed to a linear acceleration, but
secondary to head injuries have been reported in not so in angular acceleration. This set the stage
Sweden, Canada and the United States (Pashby, for further inquiry into the effects of acceleration
1993). Since 1963, however, there have been no versus impact trauma to the head (Denny and
fatalities from head injury reported in Sweden Russel, 1941; Holbourn, 1943; Gennarelli, 1991;
(Odelgard, 1989). Elson and Ward, 1994). It is now generally ac-
However, an alarming and persistently high rate cepted that in patients who experience blunt head
of minor traumatic brain injuries or mTBI, often injury, nerve fibers/axons are particularly suscep-
termed cerebral concussion worldwide over the tible to damage. It is also now well recognized that
last decade and a half has been reported (Clayton, diffuse axonal injury (DAI) is a consistent feature
1997; Dick, 1997; Biasca, 2000; Tegner, 2000; of severe human TBI, particularly those injuries
www.hockeyinjuries). Although the majority of involving rapid acceleration/deceleration of the
cases of traumatic brain injury (TBI) do occur af- brain (Denny and Russel, 1941; Gennarelli, 1991;
ter Motor Vehicle Accidents and falls, it is notable Gennarelli et al., 1998; Kelly, 1999; Graham et al.,
that sport activities are responsible for approxi- 2000). Recently, there have been considerable ad-
mately 15–25% of all cases (Gennarelli, 1991; vances in our understanding of the nature and time
McGehee, 1996). Recent evidence, obtained over course of axonal injury (AI) after TBI (Graham
the last 15 years, has suggested that mTBI may be et al., 2000). There is also increasing evidence
more common and more serious than previously that an mTBI may represent the mildest form
thought worldwide (Denny and Russel, 1941; of a diffuse brain injury. The descriptor ‘‘minor
Gennarelli, 1991; Castaldi, 1993; Tegner and Lore- Traumatic Brain Injury’’ (mTBI) has been used to
ntzon, 1996; Clayton, 1997; Dick, 1997; Gennarelli refer to such an injury (Gennarelli et al., 1998;
et al., 1998; Biasca et al., 1999; Roberts et al., Kelly, 1999; Graham et al., 2000).
1999; Biasca, 2000; Ranalli, 2000; Tegner, 2000, The purpose of this manuscript is to review re-
2001; www.hockeyinjuries; Burke, 2001; Laprade, cent studies in the epidemiology, neuromechanics,
2001; Pashby et al., 2001). According to the Center neuropathology, neurobiology and associated ne-
for Disease Control in the United States, there are urometabolic changes in mTBI in order to under-
more than 300,000 sports-related mTBI each year stand why, following a ‘‘mild’’ insult to the head,
and certainly this number underestimates the true brain cells are more susceptible and are more
level of incidence, because many of these injuries ‘‘vulnerable’’ to a second and perhaps more dam-
are not reported (MMWR, 1997). aging accident.
There has long been controversy concerning the
structural basis of mTBIs, like concussion. How-
ever, the importance of acceleration forces was al- Epidemiology of mTBI
ready established by the early 1940s (Denny and
Russel, 1941; Holbourn, 1943; Gennarelli, 1991; Injuries to the head constitute a significant pro-
Elson and Ward, 1994). Historically, many inves- portion of the total number of injuries in many,
tigators of the early 1990s were of the belief that different contact sports (Kelly et al., 1991; Cantu,
mTBI of the brain could occur without evidence of 1992; Biasca et al., 1995; Tegner and Lorentzon,
cellular or vascular lesions of the cerebrum, for 1996; Clayton, 1997; Dick, 1997; Roberts et al.,
example a number of studies have been cited by 1999; Biasca, 2000; Tegner, 2000; Tegner et al.,
Denny and Russel (1941), Gennarelli (1991) and 2000; Laprade, 2001; Pashby et al., 2001; Sports-
Elson and Ward (1994). Further, areas of pet- Related Recurrent Brain Injuries–United States,
echiae, in the absence of other lesions, have been 1997). National and international analyses report
reported after fatal brain injury (Elson and Ward, that the proportion of mTBI to the overall number
1994). of injuries fluctuates in American football between
Holbourn (1943) stated that the relative motion 3 and 24%, and in soccer between 4 and 22%
between brain constituents is insignificant when (Castaldi, 1993; Tegner and Lorentzon, 1996;
265

Clayton, 1997; Dick, 1997; Gennarelli et al., 1998; League (NHL), John Powell (2001) reported an
Biasca et al., 1999; Roberts et al., 1999; Biasca, increased incidence of mTBI from 2% in the sea-
2000; Ranalli, 2000; Tegner, 2000, 2001; Tegner son 1989–1990, to 4.9% in 1995–1996, to 8% in
et al., 2000; Burke, 2001; Laprade, 2001; Pashby each of the last two Seasons 1999–2001 for which
et al., 2001). data is available (www.hockeyinjuries). Lastly,
In ice hockey, more specifically, the proportion mTBI represented 20% of all head injuries in the
of mTBI to the overall number of injuries fluctu- Swiss National Ice Hockey Leagues A and B dur-
ates between 2 and 20%. More detailed analysis ing the seasons 1996–1998 (Fig. 1) (Biasca, 2000).
has also shown that the frequency of mTBI has However, the diagnosis of an mTBI is subject to
risen markedly over the last 15 years. In the Ca- widespread controversy both in definition and
nadian Hockey League (CHL) Clayton (1997) re- the degree of severity of injury. This controversy
ported that the proportion of mTBI rose from 4% has contributed to the current paucity of reliable
of all injuries in the period 1991–1996, to 8% in epidemiological data. It has recently been sug-
1997, 14% in 1998 and 1999 and 17% in the period gested that it would be helpful to have a world-
1999–2000. The National Collegiate Athletic wide-standardized assessment for mTBI and a
Association (NCAA) Injury Surveillance System centralized data pool using the International
recorded a similar increase in North America Jun- Sports Injury System ‘‘ISIS’’. Such would greatly
ior Hockey where the incidence of mTBI almost facilitate international comparison through use of
doubled between 1986 (0.7 injuries per 1000 AE in an Internet-based database system (Biasca and
1986) and 1996 (1.5 injuries per 1000 AE in 1996) Tegner, 2001).
(Dick, 1997). Similarly, in a study in Scandinavia,
one Swedish ice hockey team reported a rising in-
cidence of mTBI over the last 15 years from 2% of Neuromechanics of mTBI
all injuries in the season 1986–1987 to 18% in the
season 2000–2001 (Tegner, 2000; Tegner et al., The most common mechanical input to the head is
2000). In parallel, in the professional Ice Hockey dynamic loading of either impact or impulsive type

Fig. 1. Percent of mTBI, i.e. cerebral concussion, to all injuries in Sweden, Canada, North America and Switzerland from 1986 to 2001
(Clayton, 1997; Biasca et al., 2000; Tegner et al., 2000; Tegner, 2000; www.hockeyinjuries).
266

(Elson and Ward, 1994; Gennarelli, 1991; Bailes both more frequent and more damaging to the
and Cantu, 2001; Graham et al., 2000). substance of the brain because the substance of the
Impact loading requires a direct blow to the head brain is exposed to mechanical loading over a very
occurring in a period of less than 200 ms (Elson short time span — 2–20 ms — and has a very low
and Ward, 1994; Gennarelli, 1991) and, in most tolerance of tensile and shear strain. Thus, the
cases, in less than 20 ms (Graham and Gennarelli, highest levels of damage to the brain result from
1997). It occurs when a blunt object strikes the exposure to shearing stresses where individual ax-
head and usually initiates a combination of con- ons are stretched.
tact and inertial forces that result in a series of Inertial forces are responsible for the two most
events. The forces vary with the size of the im- important types of damage encountered in blunt
pacting object and the magnitude of the force deli- head injury:
vered to the contact point, the so-called contact First, acute subdural hematoma (SDH) resulting
phenomena (Elson and Ward, 1994; Graham et al., from tearing of subdural bridging veins. Such in-
2000). These contact phenomena are a complex jury occurs when the rise time and duration of
group of mechanical events that occur both locally inertial loading to the head is relatively short over
and distant from the point of the impact. They 2–12 ms (Gennarelli and Thibault, 1982). SDH
may cause laceration of the scalp, skull deforma- occurs most frequently in patients that have
tions, fractures, cerebral contusions and propaga- experienced a fall or been exposed to an assault
tion of shock waves that travel through the skull (Graham and Gennarelli, 1997).
and brain. Protective headgear is designed to Second, widespread damage in the white matter
reduce the severity of these injuries (i.e., skull de- resulting in diffuse axonal injury DAI (Elson and
formation, fracture and hematoma). Ward, 1994; Gennarelli, 1991; Graham et al.,
Impulsive loading, in contrast, occurs when the 2000) where inertial injury to the head occurs over
head is put into motion and accelerated as a result a longer time span — 11–22 ms (Gennarelli et al.,
of either an impact to another part of the body, or 1982). Adams et al. (1989) defined DAI and dis-
as a result of a secondary response to a direct im- tinguished three levels or grades of severity of in-
pact. Impulsive loading occurs also when the mov- jury to white matter — grade I in which there is
ing head is stopped without it striking anything or widespread damage to axons within the cerebral
is arrested by impact (Elson and Ward, 1994; hemispheres; grade II in which there is, in addition
Gennarelli, 1991; Graham et al., 2000). Under to the above, the occurrence of focal lesions in the
these conditions the resulting head injuries are corpus callosum often related to small hemorrh-
solely caused by inertia resulting from the manner ages (petechia) or tissue tears and grade III in
(translation, rotation, angulation) in which the which there is also damage to axons within the
head is moved. In biomechanical terms this results rostral brainstem.
in shearing stresses within the brain substance, re- The brain, which is housed in the protective
sulting in the incidence of foci of strain. Strain is bony cranium and bathed by a layer of cerebro-
the amount of deformation of brain or other tissue spinal fluid (CSF), has freedom of movement be-
as a result of the application of mechanical force. fore it abuts the cranium (i.e., smooth intracranial
There are three types of strain: compression, ten- surfaces, CSF interfaces). The CSF provides a nat-
sion and shear. The brain is highly incompressible ural shock-absorbing system, converting focally
but will deform in shear most easily when sub- applied external stress to compressive stress, fol-
jected to rotational loads (reviewed by Margulies lowing the contours of the sulci and gyri, and dis-
and Thibault, 1989). Individual axons can sustain tributing the force in a uniform fashion. The CSF,
only tensile strain, which is the amount of elon- however, does not provide complete protection
gation that occurs when a material is stretched. against shearing forces being induced in the brain,
Biological materials withstand strain if they are especially when rotational forces are applied to the
deformed slowly. But in head-injury impulsive and head and shearing forces occur at those sites in the
inertial loading, giving rise to dynamic strain, is brain where rotational gliding is hindered — for
267

example by the relatively rigid membranes forming head travels in an arc then we have a rotational
the falx cerebri and the tentorium cerebelli. The acceleration (Bishop, 1997, 2000; Bishop and
incidence of DAI is greatest when the brain is ex- Arnold, 1993; Bailes and Cantu, 2001). Focal
posed to rotational forces arising during a hori- brain injuries are more common after a linear
zontal/coronal displacement of the head. Inertial acceleration and diffuse brain injuries after a
effects result in the skull moving laterally before rotational acceleration (Bishop, 1997, 2000; Bishop
the brain does and the brain may continue to move and Arnold, 1993). However, in humans, almost
after the head has stopped moving. The falx and any injury event involves a combination of these
tentorium are firmly adherent to the internal sur- two and separation of brain responses due to
face of the skull. The brain, to the contrary, floats rotation and linear acceleration is almost impossi-
in CSF. The mid-line falx will therefore move ble (Bishop, 1997, 2000; Bishop and Arnold, 1993).
within a different time frame to the brain. The Often the mechanism of a head injury is complex
medial side of one cerebral hemisphere may contact involving a combination of focal and diffuse com-
the surface of the falx while the other hemisphere ponents. Direct head contact may induce both a
continues moving. Structures that cross the mid- rotational acceleration to cause both diffuse and
line, for example nerve fibers in the corpus callo- focal injury (Bishop, 1997, 2000; Bishop and
sum will therefore be exposed to tensile strain as Arnold, 1993; Gennarelli et al., 1998; Kelly, 1999;
one hemisphere moves away from the other. Thus Graham et al., 2000). Further, the direction in
nerve fibers passing through the corpus callosum or which the head moves plays an important role in
through the cerebral peduncles have a greater risk determining the amount and distribution of axonal
of being exposed to tensile strain and thus to in- damage in a given situation (Gennarelli et al.,
jury. In the last several years a widely held con- 1998).
sensus has developed that there is a spectrum of
levels of injury to nerve fibers when they are ex-
posed to tensile strain. When an athlete or patient mTBI in ice hockey and correlation with the type of
experiences either mild concussion — (disturbance blow to the head
of neurological function without loss of conscious-
ness), classical cerebral concussion — (a tempo- Specifically in relation to ice hockey, following
rary, reversible loss of neurological function with examination of 11 cases of mTBI in the profes-
temporary loss of consciousness for up to 6 h) or sional NHL, in the Canadian Amateur Hockey
more severe head injury when the patient is un- League and in the Swiss Ice Hockey League, it was
conscious for more than 6 h — is now thought that proposed that three different possible mechanisms
nerve fibers are injured in all of the above clinical may be responsible for causing mTBI in ice hockey
scenarios. The distinction between the above levels (Bishop, 2001):
of injury is thought to reflect the number and dis- 1. a direct eccentric blow to the head,
tribution of injured fibers within the white matter 2. a direct blow to the face and
of the brain. The less severe forms of injury in 3. a blow directed to the chin.
which only a small number of fibers are injured are
now thought to be the principal causes of an mTBI A direct eccentric blow to the head, not passing
(Elson and Ward, 1994; Gennarelli et al., 1998; through the center of mass of the head, may ex-
Kelly, 1999; Graham et al., 2000). The situation is pose brain tissue to many forces. These may be
further complicated in that an inertial loading of simplified as a force Ft with translatory compo-
the brain and head, whether caused by an impact nents (with transversal and axial forces) and a
or by an impulsive loading is influenced by the di- force Fr with rotatory components in the coronal,
rection of movement of the head during the injury sagittal and transverse (horizontal) planes (Fig. 2).
episode. If the acceleration causes a straight move- The rotatory component in the coronal plane, af-
ment of the center of the head, then we have a ter a blow to the side of the head, causes the most
linear acceleration; if the center of gravity of the severe injuries within the brain while a blow in the
268

Fig. 3. Example of a direct blow to the face or jaw: There is no


protective equipment, such as helmets and mouth guards, which
may be effective in reducing any rotatory component. (Mt:
Fig. 2. Example of a direct eccentric blow to the head: a pro- Moving force in the transversal plane; Mf: moving force in the
tective headgear, with its padding, helps to reduce the effect of frontal plane; Ft: force in the transversal plane.) Reprinted with
the final force F and therefore may reduce the risk of a focal permission and courtesy of the player.
head injury. (Mt: Moving force in the transversal plane; Ms:
moving force in the sagittal plane; Mf: moving force in the
frontal plane; Ft1: force 1 in the transversal plane; Ft2: force 2 injury, but although development of padding ma-
in the transversal plane; Fa: force in the axial plane.) Reprinted terials has resulted in improvement of the energy
with permission and courtesy of the player. absorption characteristics of helmets, the crucial
factor causing mTBI is widely acknowledged to be
sagittal plane (flexion/extension of the cervical the effect of rotational acceleration of the brain.
vertebral joints) is tolerated best (Gennarelli et al., A direct blow to the face may cause both a force
1998). Protective headgear including padding, Ft with translatory and force Fr with rotatory
helps to reduce the overall effect of the final force components. In ice hockey contacts to the jaw and
F, and also reduces the contributory effects of face, a similar mechanism to the knockout punch
contact and rotational phenomenon. Hockey hel- in boxing, are numerous. As a result there is often
mets were originally developed to reduce the risk a significant rotational acceleration of the head
of serious head injuries, precipitated by direct and brain (Fig. 3).
blunt traumas and provided protection against Gennarelli et al. (1982) found that the severity
brain injury death and/or intracranial hematoma of experimental TBI in a non-human primate
(Odelgard, 1989; Bishop, 1997, 2000; Bishop and appeared to be related to the plane of rotational
Arnold, 1993; Dixon and Brodie, 1993; Pashby, acceleration. For equivalent levels of angular ac-
1993; Stoner and Kreating, 1993; Pashby et al., celeration, nerve fibers in the white matter of the
2001; Tegner, 2001). The two most critical func- brain were most vulnerable to injury if the head
tions of helmets are to reduce impact energy and was rotated laterally (vide supra). When the brain
load distribution to the head. These properties re- moved laterally tensile strain occurred in a coronal
duce the magnitude of the forces applied to the plane parallel to the long axis of nerve fibers
head, reduce the stress and strain to which the crossing the sagittal plane. Lesser degrees of injury
skull and brain are exposed and thus reduce the were obtained when the brain moved either in
severity of mTBI and head injury. Thus, protective the sagittal or horizontal planes (Gennarelli
headgear first of all reduces the risk of a focal head et al., 1982). Recently Smith et al. (2000) have
269

suggested that the severity of experimental TBI


may be more of a reflection of the severity of ax-
onal damage in specific regions of the brain, most
notably the brainstem, rather than the total sum of
AI distributed throughout the organ (Smith et al.,
2000). Following rotation of the head in the axial
plane, substantially more axonal damage was
found in the brainstem, compared with that in-
curred when the head was rotated in the coronal
plane (Smith et al., 2000). But care needs to be
taken in the interpretation of these results and
their comparability with injury in the human. In
the pig the brain stem lies in the same approximate
longitudinal axial plane as the cerebrum. In the
human, however, the brain stem lies at an angle of
some 1201 to the cerebrum. Thus, in the pig an
axial injury which occurs in the horizontal plane of
the head (Fig. 1 in Smith et al., 2000) will provide Fig. 4. Example of a direct blow to the chin: mouth guards may
have the ability to absorb the impact loading from the chin and
the greatest mechanical strain to nerve fibers at the to distribute the remaining energy, throughout the resilience of
lateral limits of the brain stem. Indeed, Smith et al. its material combined with its design, over a much larger sur-
(2000) illustrate such data in Fig. 4 of their paper. face area, thereby reducing the final force on the brain. (Ft:
In humans, however, because of the different Force in the transversal plane.) Reprinted with permission and
planes of orientation of the longitudinal axes of courtesy of the player.
the cerebrum and brain stem mechanical strain will
be placed largely on those nerve fibers lying par-
allel to the direction of displacement of the head. mandatory, worldwide since June 2002. This
That is in coronal and parasagittal fibers within means that every player making contact to the
the cerebral hemispheres. Nonetheless, Smith et al. head and neck with the body, elbow, shoulder,
(2000) are the first to suggest (1) a relationship knee or stick of an opposing player will be penal-
between the plane of head rotational acceleration ized, at the discretion of the referee, with either a
and the distribution of AI and (2) that injury to Minor (2 min) or Major (5 min) Penalty plus au-
axons in the brainstem plays a major role in the tomatic Game Misconduct or even Match Penalty.
induction of an immediate post-traumatic coma. Prevention strategies, such as the introduction of
With specific regard to sports-related injuries, no ‘‘Checking from behind’’ Rules in 1994, have be-
protective equipment, which may reduce the ef- come effective during the last years in decreasing
fects of this rotatory force (Fr), is presently avail- the number of severe spinal injuries worldwide.
able. For this reason it is necessary to eliminate the Therefore, it is hoped and expected that these
practice by players of checking against the head new ‘‘Head-Checking’’ rules should reduce the in-
and neck of other players and which may thereby cidence and severity of mTBI.
expose the brain to rotatory acceleration/deceler- A blow directed to the chin may cause a trans-
ation. The Rules Committee of the International latory force Ft, which is transmitted from the chin
Ice Hockey Federation IIHF, under the chairman- through the lower jaw, through the temporoman-
ship of Mr. Philippe Lacarrière, has therefore in- dibular joint at the base of the skull and then to
troduced in 2002 new ‘‘Head-Checking Rules’’ the brain. These forces may also propagate shock
(IIHF Rule No. 540: http://www.iihf.com// waves through the brain, which may induce mTBIs
hockey/rules/rules.htm) in order to eliminate (Fig. 4).
every intentional or unintentional check or blow Mouth guards alone or in conjunction with ad-
to the head and neck. This rule has been made ditional face protection, have been demonstrated
270

to be effective in preventing and/or reducing the Neuropathology of mTBI


incidence of dental and orofacial injuries (Piccin-
inni, 2001). Numerous anecdotal reports have in- The pathology of human TBI is complex and het-
dicated a correlation between the use of mouth erogeneous, comprising either single or a combi-
guards and prevention of mTBI. Piccininni (2001) nation of focal and diffuse lesions (Gennarelli,
focused on two primary mechanisms to explain 1991; Elson and Ward, 1994; Graham et al., 2000;
this correlation. The first mechanism was dissipa- Tegner, 2001). Trauma to the skull and brain
tion of forces delivered to the maxilla, skull and manifests in a variety of pathological hallmarks
temporomandibular joint complex when the man- reflecting the distribution of the impact energy.
dible received a blow from the front whereby the Sharp objects at high velocity tend to cause per-
force applied to the mandible is transmitted to forating focal cortical contusions (FCC). Colli-
both temporomandibular joints, absorbed in part sions against sharp edges lead to non-penetrating,
by the articular disc and fibrous capsule of those mostly focal head injuries (FHI). Blunt trauma —
joints before being transmitted to the temporal where the head does not necessarily hit anything
bone and the base of the skull. The second mech- but is exposed to rapid acceleration and deceler-
anism was the stabilization of the skull through the ation — often leads to diffuse trauma, that is DAI.
increased contraction of the muscles of mastica- FCC, FHI and DAI often occur together at any
tion, principally the massetur and temporalis mus- combination of lesion severity (Elson and Ward,
cles, when clenching the lower jaw. This may be 1994; Graham et al., 2000).
enhanced with the presence of a mouth guard (Pi- FHI are caused by forces of contact and head
ccininni, 2001). For this reason Piccininni (2001) acceleration from direct blunt trauma, such as be-
stated that mouth guards can and should be pro- ing struck with a hockey stick or a puck, or falling
moted as effective devices for the prevention of on the ice surface and striking the head. These
dental and orofacial injuries and that properly de- types of injuries may produce a focal cortical con-
signed mouth guards may also play a role in the tusion with well-demarcated damage to the paren-
reduction of incidence or severity of mTBI. How- chyma of the brain and associated disruption of
ever, evidence of any injury protection using arterial and venous vessels in the region of the
mouth guards is derived from case series and ret- contusion. Such gross lesions may readily be ob-
rospective injury surveys. Further epidemiological served in CT scans, MRI or macroscopically at
research using the ISIS will surely contribute to an autopsy. FHI often result in fracture of the skull,
increased understanding of the incidence and with or without an associated epidural hematoma,
mechanism of any injury and allow a reduction in cortical contusions and in intracranial hem-
of the risk of injury. The use of properly fitted orrhage (Gennarelli, 1991; Bishop, 1997; Bishop
mouth guards would no doubt reap numerous and Arnold, 1993; Elson and Ward, 1994; Kelly,
benefits in term of reducing medical, financial, 1999; Graham et al., 2000; Tegner, 2001). Tissue
cognitive, psychological and social consequences damage occurs either by direct mechanical de-
of any dental and cerebral injuries, like mTBI, at a struction of brain parenchyma, compression
minimal cost. As a result, the Rules Committee of through interstitial bleeding or by secondary inf-
the IIHF has made in 2002 some rule changes. arction following disrupted circulation. Wearing
This rule will be mandatory, as a first step, for all an internationally approved, standardized helmet
junior ice hockey players younger than 20 years will clearly reduce the number and severity of these
and not wearing full-face masks. As a next step, focal injuries (Bishop, 1997; Bishop and Arnold,
this rule should be mandatory for all players born 1993).
after December 31, 1975 and for all players wear- Diffuse head injuries, on the other hand, are
ing full-face masks as well, because a full-face caused by the inertial effect of either a mechanical
mask alone can not reduce the forces transmitted blow to the head or rapid rotation acceleration/
to the brain from any blow to the jaw or chin deceleration phenomena as a result of a blow to
(Biasca et al., 2002). another part of the body or a fall (Gennarelli,
271

1991; Elson and Ward, 1994; Graham et al., 2000; provided considerable advances in the understand-
Tegner, 2001). In ice hockey, such injuries may ing of the nature and time course of TAI in and
involve axonal damage due to excessive angular following TBI (Povlishock et al., 1983; Grady et
acceleration (e.g., after an abrupt body check) al., 1993; Blumbergs et al., 1994, 1995; Christman
caused by either direct or indirect inertial loading et al., 1994; Elson and Ward, 1994; Pettus et al.,
of the head and are characterized by a global dis- 1994; Sherriff et al., 1994a, b; Blumbergs, 1997;
ruption of neurological function (Gennarelli, 1991; Maxwell et al., 1997; Gennarelli et al., 1998; Gra-
Elson and Ward, 1994; Graham et al., 2000; ham et al., 2000).
Tegner, 2001). It is now well recognized that (1) Graham et al. (2000) reported that over the last
traumatic axonal injury (TAI) is a consistent several years a consensus has arisen that the prime
feature of human TBI, particularly those injuries site of injury in an injured axon in TBI is the axo-
involving rapid acceleration/deceleration of the lemma. There is now a consensus that pathology in
brain (Elson and Ward, 1994; Graham et al., 2000) the injured axon then occurs because of loss of
and (2) that these injuries represent a continuous homeostatic mechanisms that maintain differential
spectrum of the same pathology: a progressive ionic gradients necessary for the normal electrical
widespread but heterogeneous damage to axons activity of the axon.
(Gennarelli, 1991; Elson and Ward, 1994; Graham TAI has been shown in a variety (fluid percus-
et al., 2000; Tegner, 2001). sion, cortical impact, rotational acceleration and
The majority of studies of TAI have long been stretch-injury) of experimental animal models of
focused upon human DAI and animal models human head injury (Povlishock et al., 1983; Pettus
thereof. Here TAI has been linked to the morbid- et al., 1994; Graham et al., 2000). Povlishock et al.
ity associated with that condition (Strich, 1961, (1983) reported TBI after fluid percussion injury
1970; Nevin, 1967; Peerless and Rewcastle, 1967; and controlled cortical impact in rats, cats and
Oppenheimer, 1968; Clark, 1974; Adams et al., swine. Gennarelli et al. (1982) reported TBI after
1982; Gennarelli et al., 1982; Povlishock et al., rotational head acceleration in a non-human pri-
1983; Pettus et al., 1994; Maxwell et al., 1997). mate, and Ross et al. (1995) in miniature swine.
Historically, it was suggested that forces acting Lastly, Gennarelli et al. (1989) reported TAI after
during TBI and TAI tear or shear axons, causing controlled stretch-injury to the guinea pig optic
them to retract from the site of injury and expel a nerve.
ball of axoplasm. This forms a reactive swelling or A consensus concerning the sequence of path-
retraction ball, the histological structural feature ologies within injured axons has developed from
then used to diagnose DAI (Adams et al., 1982). the above studies of moderate-to-severe TAI gen-
More recent studies, however, have not provided erated in numerous, different laboratories. This
support for the concept that large numbers of sequence may be summarized very briefly as fol-
axons are sheared at the time of injury. Rather lows. First, injury to the axolemma; second, swell-
experimental, animal models have demonstrated ing of axonal mitochondria as a result of specific
that traumatic injuries elicit a delayed or second- damage to the internal mitochondrial membrane;
ary axotomy wherein the trauma initially injures third, loss of axonal microtubules and alterations
an axon, which, however, remains in continuity, of the intra-axonal relationship of neurofilaments;
causing it, over a period of at least several hours, fourth, the occurrence of so-called ‘‘axonal swell-
to fail at one or more foci, and disconnect. A re- ings’’ (foci of increased axonal diameter at which
active swelling of classic description then forms there is still continuity of the axon on both prox-
(Povlishock et al., 1983; Povlishock, 1986, 1992, imal and distal sides of the region of increased
1993; Gennarelli, 1991; Pettus et al., 1994; Povli- axonal diameter) and finally axonal disconnection
shock and Christman, 1995; Povlishock and Jenk- to form so-called degeneration bulbs or ‘‘axonal
ins, 1995; Povlishock and Pettus, 1996; Maxwell et bulbs’’ (Povlishock et al., 1983, 1997; Pettus et al.,
al., 1997; Gennarelli et al., 1998; Graham et al., 1994; Povlishock and Pettus, 1996; Maxwell et al.,
2000). Further, studies within the last decade have 1997, 1999; Okonkwo and Povlishock, 1999).
272

Povlishock and colleagues initiated such study


in traumatically induced axons using the antero-
grade tracer horseradish peroxidase (HRP) (Po-
vlishock et al., 1983; Erb and Povlishock, 1988).
Their approach was based on the premise that ax-
ons tear at the moment of injury. At injury, axons
laden with anterogradely transported protein
would rupture and expel a mass of protein (per-
oxidase)-containing axoplasm that could be easily
visualized using histochemical methods at by both
light and electron microscopes (Povlishock et al.,
1983). But evidence for tearing or shearing — that
is loss of the integrity of the axolemma and the
presence of label free within the periaxonal space
— was not obtained (Erb and Povlishock, 1988).
Rather at 1 h and later after injury a local, lobular
pooling of intra-axonal peroxidase still in conti-
nuity with both proximal and distal parts of the
peroxidase containing axon occurred. Moreover,
the diameter of the focal pools of HRP increased
in diameter — from 10 mm at 1 h to 18 mm at 6 h
after injury (Erb and Povlishock, 1988). In addi-
tion, multilobulated profiles interconnected by Fig. 5. Light micrograph (A) illustrates the changes in intra-
thin HRP labeled bands were observed. Evidence axonal anterograde horseradish peroxidise passage following
for loss of continuity of labeled axons was only fluid percussion injury. Note that within several hours of the
traumatic insult the axon shows a lobulated swelling with a site
described at 2 h and later after injury. The pooling
of axonal narrowing (red arrow). Reprinted with permission
of peroxidase was suggested to occur because of a and courtesy from Maxwell et al. (1997). Light micrograph (B)
focal impairment of axonal transport. Continued illustrates a lobulated terminal swelling after axonal disconnec-
anterograde tracer delivery to the site, resulted in tion to form a conspicuous proximal bulb. Human material
focal expansion of diameter of the axon, swelling labelled with antibodies for beta amyloid precursor protein.
and ultimately detachment from its distal coun-
terpart (Figs. 5A, B) (Povlishock et al., 1983; loss of axonal microtubules and a reduced spacing
Povlishock, 1992; Pettus et al., 1994; Maxwell between, or compaction, of neurofilaments. How-
et al., 1997). ever, such alterations were not representative of all
A second approach using HRP, as a marker of axons demonstrating pathology. In some axons,
AI (Pettus et al., 1994; Pettus and Povlishock, despite the fact that peroxidase had not entered the
1996), was to infuse peroxidase into the CSF of axoplasm, the components of the cytoskeleton no
anaesthetized animals prior to TBI (Pettus and longer ran parallel to the long axis of the axon but
Povlishock, 1996). The intact axolemma normally adopted a spiral course through the axoplasm
excludes molecules as large as HRP from the axo- (Pettus and Povlishock, 1996). Thus in mild ex-
plasm. But, within 5 min after TBI, axons were perimental TBI neurofilament misalignment and
obtained that contained peroxidase and such labe- axonal swelling were not associated with the pas-
led axons were obtained up to 6 h, the end of the sage of peroxidase from the extracellular to the
experimental period examined, after injury. Thus intra-axonal compartment (Pettus et al., 1994,
TBI resulted in damage to the axolemma of some 1996). The latter findings provided support for the
axons such that peroxidase was able to enter the concept that there is a spectrum of AI in TAI.
axoplasm (Pettus et al., 1994; Pettus and Povli- More recently, this concept has been strengthened
shock, 1996). In the aforesaid axons there was also by the findings of other investigators who have
273

shown that TAI does not affect only large axons, b-APP in relatively normal-looking axons (Gen-
as had been suggested in the mid 1980s, but also tleman et al., 1995) and may simply represent
injures small, myelinated fibers that occur, for ex- changes in axoplasmic transport of a temporary
ample in the optic nerve (Jafari et al., 1997, 1998). nature in axons which, hypothetically, have a po-
Indeed, these latter studies showed that alterations tential for recovery (Gennarelli, 1991; Gentleman
for numbers of and spacing between microtubules et al., 1995; Gennarelli et al., 1998). The time
and neurofilaments differed between nerve fibers course of recovery is unclear, but it is interesting to
of different axonal diameter. Most recently, a fur- note that Blumbergs et al. (1994) reported that b-
ther complication of the study of developing pa- APP accumulation can be shown in humans as
thology in TAI has been indicated in that long as 99 days after the initial injury (Blumbergs
immunocytochemical labeling of injured axons et al., 1994, 1995; Blumbergs, 1997).
for either b-amyloid precursor protein, a normal Overall, these immunocytochemical alterations
component of axoplasm, or compacted neurofila- have provided further confidence that, after TAI,
ments (Stone et al., 2001) has suggested that one there is a focal impairment of axoplasmic trans-
subtype of injured axons undergo a progressive port, leading to progressive axonal swelling and
focal swelling resulting in disconnection or sec- detachment/secondary axotomy over a period of
ondary axotomy between 2 and 6 h while others several hours or days after injury (Grady et al.,
show compaction of neurofilaments within min- 1993; Christman et al., 1994; Pettus et al., 1994;
utes of injury but do not undergo any further Povlishock and Christman, 1995; Maxwell et al.,
change over the following several hours. 1997; Graham et al., 2000).
Over the last decade use of antibodies against However, patients with mTBI rarely come to
amyloid precursor protein (APP) (Sherriff et al., neuropathological examination. Nonetheless,
1994a, b; Gentleman et al., 1995; McKenzie et al., there is increasing evidence that some permanent
1996) has been adopted as the marker of choice for damage may be present (Christman et al., 1994;
detecting TAI in the diagnostic laboratory. APP is Blumbergs, 1997; Gennarelli et al., 1998; Graham
a membrane-spanning glycoprotein originating et al., 2000). Initially it was thought that mTBI
from a gene on chromosome 21 and of unknown might produce only temporary disturbances of
function (Hartmann, 2001). It is transported by brain function without gross structural changes.
fast axoplasmatic transport, is located at both the Now there is increasing evidence that mTBI, in
pre- and post-synaptic sites and accumulates both animals and humans, can lead to an impair-
within axonal swellings in TBI very quickly in hu- ment of axoplasmic transport, which results in a
mans (Blumbergs et al., 1994; Blumbergs, 1997) as progressive pathology comparable with that de-
a result of loss of fast axonal transport. However, scribed earlier in moderate or severe TBI (vide
the recent findings by Stone et al. (2001) now supra). Thus axonal swellings form in mTBI, fol-
question the sensitivity of a-APP immunohisto- lowed by axonal detachment in ‘‘delayed axo-
chemistry alone as a tool for demonstrating TAI. tomy’’ (Blumbergs et al., 1995; Povlishock and
Rather, a range of immonocytochermical markers Christman, 1995; Maxwell et al., 1997; Gennarelli
should be used because use of APP labeling prob- et al., 1998; Graham et al., 2000). This process of
ably underestimates the true number of axons un- delayed axotomy is a complex network of inter-
dergoing pathology. Indeed, use of the antibody acting functional, structural, cellular and molecu-
Ab38 that labels sites of calpain-mediated spectrin lar changes (Gennarelli et al., 1998). Fairly recent
proteolysis (Buki et al., 1999), shows increased evidence (Maxwell et al., 1999) has provided fur-
density of axons/mm2 labeled for Ab38 (calpain- ther confidence for the concept that tensile strain
mediated spectrin proteolysis) in the pyramidal to an axon leads to injury to the axolemma. Cyto-
tracts and medial lemniscus of rats after impact chemical techniques which label for activity of
acceleration injury at 60–120 compared with Ca2+-ATPase and Na+/K+-ATPase (pNNP-ase)
15–30 min after injury. In addition, a-APP labe- show loss of activity of these two membrane
ling techniques have also shown accumulation of pumps after injury. Loss of such membrane pump
274

activity would result in a loss of transmembrane manifested as either long-term compaction of ne-
homeostasis for ions, and in particular for Ca2+. urofilaments or loss of fast axonal transport lead-
The resulting influx of calcium down the normal ing to secondary axotomy.
diffusion gradient, which maintains Ca2+ levels APP as a marker for TAI, is of particular im-
10,000 times higher in the extra-axonal compart- portance with regard to mTBI following the very
ment in the normal axon, will result in elevation of important observation of multifocal AI within the
intra-axonal concentrations of Ca2+ and allow fornices in human beings with a recorded loss of
activation of calpain-mediated spectrin proteloysis consciousness of as little as 60 s (Blumbergs et al.,
(CMSP). Spectrin is an integral component of the 1994). Thus TAI has been documented after rel-
subaxolemma cytoskeleton. Buki et al. (1999) atively mild head injury and in milder grades of
showed that CMSP breakdown products appear DAI in humans (Christman et al., 1994; Blumb-
first within the subaxolemma compartments of in- ergs et al., 1995; Povlishock and Christman, 1995;
jured axons at 15 min and only extend throughout Povlishock and Jenkins, 1995). There is also a
the whole diameter of the axoplasm at 1 h after growing consensus that mTBI may represent the
injury. Unfortunately CMSP immunocytochemis- mildest form of TAI (Povlishock et al., 1983; Elson
try was not examined beyond 2 h after injury. But, and Ward, 1994; Pettus et al., 1994; Gennarelli
quantitative freeze-fracture studies provided mor- et al., 1998). At mild levels of AI, a primary mis-
phological evidence of damage to cell membranes alignment of the components of the axonal cyto-
(Maxwell et al., 1999), both the axolemma and skeleton occurs in which neurofilaments and
myelin sheath, up to 24 h after TAI. In parallel, microtubules assume a spiral orientation within
excess axoplasmic calcium leads to a dysfunction the axoplasm independent of damage to or an in-
and a breakdown of neurofilaments and microtu- crease in permeability of the axolemma as may be
bules. Further, release of acetylcholine contributes indicated through use of HRP tracers within the
not only to the depolarization of neurons but also CSF (Sherriff et al., 1994b; Gentleman et al., 1995;
to calcium-associated excitoxic tissue damage. Pettus and Povlishock, 1996; Maxwell et al., 1997).
Thus there is increasing evidence that the initial In damaged axons regions of increased axonal di-
site of damage is the axolemma. Such damage may ameter are termed axonal swellings. Here conti-
then allow abnormal ionic or molecular influx into nuity of the axon is maintained on both the
the injured nerve fibers. Over time, these changes proximal and distal sides of the swelling (Fig. 6)
lead to foci of disruption of axonal structure and (Povlishock et al., 1983; Pettus et al., 1994; Sherriff
impaired axonal transport culminates in secondary et al., 1994b; Gentleman et al., 1995; Maxwell
or delayed axotomy. et al., 1997).
Thus, experimental studies of TAI, over the last
decade, have lead to the now widely held consen-
sus that there is a spectrum of pathological axonal
changes reflecting both the severity of injury (Po-
vlishock et al., 1983; Pettus and Povlishock, 1996)
and size of axons (Maxwell et al., 1997, 2003). The
situation is complicated because there is increasing
evidence that the spectrum of axonal response to
TAI may reflect either or a combination of (1) the
level of tensile strain applied to individual axons at
the time of injury, (2) the degree of damage to the
axolemma leading to the injured axon entering the
‘‘pathological cascade’’ which results in secondary
Fig. 6. This light micrograph of human material labelled for
axotomy, (3) the size of the axon before injury in beta amyloid precursor protein, illustrates, early in the post-
that large and small axons respond differently to traumatic course, the occurrence of two regions of increased
TAI and (4) whether the axonal response is axonal caliber where the two swellings are still linked.
275

In parallel with moderate or severe TAI, in saturation of the mechanism of mitochondrial se-
mTBI in sports-related injuries the magnitude, du- questration of excess cytoplasmic calcium. This
ration and rate of onset of angular acceleration as causes damage to and swelling of mitochondria
well as the direction of head motion during the (Okonkwo and Povlishock, 1999), together with
injury episode will determine the amount of axonal enlargement of the injured axon. Hypothetically,
damage, the severity and the reversibility of the this mild impairment of axoplasmic transport is
clinical syndromes and the resulting neurological reflected, ultrastructurally, as a non-linear align-
deficits (Gennarelli et al., 1998; Graham et al., ment of the components of the axonal cytoskele-
2000). Although, hypothetically, both the number ton. It is currently hypothesized that most of these
and overall anatomical distribution of injured ax- axons recover and thus restitution of structure and
ons is lower and less widespread compared with function may be expected.
the severe traumatic state, the very presence of But, if the level of stretch is between 10 and 20%
immunocytochemically labeled, damaged axons is (Stages III and IV respectively) (Fig. 7), then the
indicative of AI and thought to reflect the lower axon cannot fully restore ionic and osmotic home-
end of the spectrum of TAI (Povlishock, 1992, ostasis and, as a consequence, numerous patho-
1993). It is now becoming accepted that there are a logical changes are initiated (Gennarelli et al.,
range of categories of diffuse brain injury, which 1998). Recent evidence (Stone et al., 2001) suggests
have the same pathophysiology within the indi- that there may be two separate pathological path-
vidual, injured axons; but a different clinical out- ways followed by different axons (Fig. 7).
come. The latter, hypothetically, relates to the As the level of clinical injury progresses from
severity of axonal elongation resulting from me- less to more severe, greater amounts of damage to
chanical strain placed upon axons at the time of individual axons and greater numbers of axons are
injury (Elson and Ward, 1994; Graham et al., thought to be involved. In injuries from which the
2000). patient apparently recovers, for example mTBI,
Current thinking with regard to the spectrum of there is presently no evidence for Stages III and IV
axonal responses between momentary periods of damage. This is due to the fact that the patient
unconsciousness on the playing field, mild concus- resumes normal activities with no detectable
sion and concussion may be summarized as fol- change in their performance and that Stages III
lows in terms of the degree of mechanical strain and IV may only be determined post-mortem.
placed upon axons at the time of injury and their Therefore, it is suggested that Stages I/II represent
physiological/pathological responses. the great majority of axonal injuries experienced,
for example by ice hockey and other contact sports
(1) At the lowest level of strain (5% or less
players.
elongation) (Stage I) (Fig. 7), ionic influx of
various species occurs and causes temporary
(2) In the context of mTBI, the influence of the
failure of the generation and propagation of
degree of damage to the axolemma upon
action potentials (Gennarelli et al., 1998).
subsequent axonal responses has not been
Influx of Ca2+, Na+ and Cl and the efflux
investigated in any systematic or controlled,
of K+ at this level of injury are fully restored
experimental manner. But there is now good
in a matter of minutes and the damaged
evidence that the integrity of and the func-
axon recovers its function completely.
tional state of the axolemma are related to
With increasing strain applied to the axon, a axonal responses after TAI. The structural
series of increasingly severe mechanisms and path- and physiological integrity of the axolemma
ological changes occur. If the level of axonal is essential for the generation and propaga-
stretch is in the 5–10% range (Stage II) (Fig. 7) tion of action potentials. It is suggested that
then additional ionic perturbations are associated perhaps electrophysiological assessment of
with either fluid fluxes in an attempt to maintain membrane function would possibly provide
osmotic balance (Gennarelli et al., 1998) or the best means of analysis of the functional
276

WALLERIAN DEGERATION

(1 to 12 weeks)
(24 hours - 7 days)
PROXIMAL REMNANT OF AXON Proteolysis of axonal components
reduced calibre of axon but myelin figures remain
neurofilament compaction over DISTAL REMNANT OF AXON
30-50 µm of axonal length

SECONDARY AXOTOMY
(minimum of 4 hours - 7 days)

(up to 12 weeks)
15 - 20% strain (4 hours - 99 days)
axonal disconnection Compaction of neurofilaments
reduced axonal cross-sectional area
PRIMARY AXOTOMY disruption of the myelin sheath increased g ratio
(less than 1 hour) enlarged periaxonal space
focal compaction of neurofilaments
disruption of the axolemma
proteolysis/destruction of neurofilaments

% strain currently unknown (30 min-7 days)


more than 20% strain multiple foci of neurofilament compaction
membrane renting/fragmentation swelting of/damage to mitochondria
proteolysis of axonal cytoskeleton neurofilament compaction
in zone of axotomy marked infolding of the exolemma
10-15% strain (1.5 - 6 hours)
axonal swelling
loss of axonal transport
loss of microtubules
µM calpain/phosphatase/kinase activation
collapse/loss of neurofilament sidearms
compaction of neurofilaments

5 - 10% strain (15 min - 24 hours)


functional and structural damage to the axolemma
depolarisation and loss of ionic homeostasis
loss of and/or de-activation of ion channels and ATP
dependent membrane pumps, mitochondrial damage
KEY
5% or less strain (seconds) Effects of injury
translent depolarisation Mechanical strain
(Time scale)
Recovery
mechanism and ultrastructural changes
INJURY DESCRIPTOR

Fig. 7. Schematic overview of current thinking with regards to AI in human DAI and animal diffuse traumatic brain injury. (Modified
from Maxwell et al., 1977.)

integrity of an injured axon. The authors are time required for repolarization increases in
aware of only two experimental, electro- a non-linear manner. At loadings resulting
physiological studies concerning axonal re- in more than 20% elongation the axolemma
sponses to the application of tensile strain is irreparably damaged (Galbraith et al.,
(Tomei et al., 1990; Galbraith et al., 1993) 1993). There is thus an apparent, critical
and only the former involved experiments on threshold at or around 20% elongation at
mammalian axons. With increasing loading which the axolemma is irreparably damaged.
from 0 to 20% elongation there is rapid In mammalian myelinated axons, analysis of
depolarization following by a slow repolar- visual evoked potentials in the visual cortex
ization. But with an increased loading, the after TAI to optic nerve showed (1) a
277

reduction in signal amplitude to 33% of which are not damaged to the degree that
control values immediately after injury, to large molecules such as HRP may enter the
66% at 24 h and 77% at 1 week. (2) An in- axoplasm.
creased signal latency to 126% of control (3) Evidence obtained over the last several years
values after injury, 119% at 24 h and 97% at has shown that neurofilaments and micro-
1 week in an animal model (guinea pig optic tubules demonstrate a spectrum or range of
nerve stretch-injury) in which the current responses after TAI. Current results suggest
literature documents that only 20% of fibers that at lower or mild levels of injury, there is
provide morphological evidence for injury focal disarray of neurofilament and micro-
(Gennarelli et al., 1989). Thus, at the func- tubule alignment within the axoplasm, but
tional level, axons are rapidly depolarized at no loss of microtubules (Povlishock et al.,
TAI (Stage I, Fig. 7). However, in this ex- 1983; Pettus and Povlishock, 1996; Maxwell
perimental paradigm, recovery of activity et al., 1997, 2003). But, in moderate or se-
occurs relatively slowly, will vary with the vere TAI there is (1) rapid loss of microtu-
initial loading, and total recovery is not ob- bules (Maxwell and Graham, 1997) and (2) a
tained even at 1 week after injury (Tomei et reduction in the spacing between adjacent
al., 1990). Two research groups have pro- neurofilaments, resulting in both an in-
vided morphological evidence for damage to creased density of neurofilaments per unit
the axolemma. First, after fluid percussion area of the axoplasm (Pettus and Povli-
injury, influx of HRP from the CSF into shock, 1996; Povlishock et al., 1997) and
damaged axons was shown (Pettus and Po- reduced interneurofilament spacing (Jafari
vlishock, 1996). But, in molecular terms et al., 1997, 1998; Maxwell et al., 2003).
HRP, is a large molecule and thus influx The term applied to this latter change in the
into the axoplasm of an injured axon would organization of the axonal cytoskeleton is
necessitate the appearance of relatively large that the neurofilaments are said to be ‘‘com-
‘‘holes’’ in the membrane. Hypothetically, pacted’’.
such damage would probably result from
elongation of axons by more that 20% of More recently, quantitative analysis has shown
their pre-injury length. It is noteworthy that that alterations in the organization of the axonal
in the same study the authors noted axons in cytoskeleton after TAI differ (1) in the time course
which there was axonal pathology but that of the response by microtubules and neurofila-
HRP had not entered the axoplasm. It is ments and (2) in different sized axons. After TAI
currently hypothesized that such damage in the guinea pig optic nerve compaction of ne-
represents a mild level of injury to those ax- urofilaments is most marked in smaller axons. In-
ons. Second, using the same model as Tomei deed, the reduced spacing between neurofilaments
et al. (1990), Maxwell et al. (1999) provided has been correlated with a reduction in the calibre
both cytochemical evidence for loss of mem- of axons (Jafari et al., 1997; Maxwell et al., 2003).
brane pump activity — vide supra — and But in larger axons of the optic nerve there is an
quantitative data for an alteration in the increased spacing between components of the
number and distribution of intramembra- cytoskeleton throughout the length of the nerve.
nous particles (IMPs) visualized through In these larger fibers, nonetheless, there are focal
freeze-fracture techniques. It is currently be- sites of compacted neurofilaments. But these sites
lieved that the altered distribution of IMPs are associated with loss of the axolemma and pos-
reflects changes in the distribution and sibly are sites at which the axon is undergoing the
number of transmembrane proteins some final stages of secondary axotomy (Jafari et al.,
of which are membrane pumps and ion 1998; Maxwell et al., 2003). In other animal
channels. Hypothetically, such changes may models, however, for example cortical impact
also reflect damage to the axolemma in fibers and fluid percussion injury, there is compaction
278

of neurofilaments in relatively large fibers (Pettus cerebral blood flow (CBF) and cerebral glucose
and Povlishock, 1996; Povlishock et al., 1997; metabolism have all been documented in brain-
Stone et al., 2001). injured patients (Giza and Hovda, 2000). Utilizing
animal models, investigators have been able to
(4) Recent, novel work has suggested that com- better characterize trauma-induced ionic flux and
paction of neurofilaments may occur in ax- provide quantitative data in support of the corre-
ons other than those which progress to the sponding metabolic dysfunction that has become
formation of axonal swellings and secondary the clinically, accepted diagnostic descriptor of
axotomy (Stone et al., 2001). However, only mTBI. Microdialysis probes have detected ionic
one research group has so far described such disturbances (Katayama et al., 1990; Kawamata et
a finding and thus there is presently no other al., 1992; Hovda et al., 1999; Cantu, 2000; Reinert
material that would allow discussion. But, it et al., 2000; Klatky et al., 2002) and nuclear mag-
is included here as a pointer to the fact that netic resonance spectroscopy (MRS) has recently
attention should be paid to this phenomenon been used to demonstrate neurochemical altera-
in later investigations of TAI. tions associated with pathological conditions after
Clearly, much further work needs to be done to TBI (McIntosh et al., 1987, 1988; Pettus and Po-
fully understand the significance of differing cyto- vlishock, 1996; Hovda, 1996; Cecil et al., 1998;
skeletal responses to (1) axonal loading, (2) dif- Hovda et al., 1999; Bigler, 1999; Garnett et al.,
ferent responses by axons within different CNS 2000; Giza and Hovda, 2000; Brooks et al., 2001;
tracts and (3) between axons that are undergoing Sinson et al., 2001). Positron emission tomography
secondary axotomy and those that are not. How- (PET) has provided quantitative data for altera-
ever, in summary, three different types of evidence tions in cerebral glucose metabolism (Nedd et al.,
support the hypothesis that damage to the axo- 1993; Ruff et al., 1994; Gross et al., 1996; Abdel-
lemma is key to the development of pathology in Dayem et al., 1998; Bigler, 1999; Ricker et al.,
injured axons after TAI. Overall, there is variation 2001; Levine et al., 2002; Umile et al., 2002) while
in the sites of axonal damage within the brain of both radioisotope studies (Hovda et al., 1999;
each and any patient and axonal cytoskeletal re- Ricker et al., 2001) and transcranial Doppler im-
sponses may differ between axons in different CNS aging (Ng et al., 2000; Muller et al., 2001) have
tracts. It is intriguing as to whether these detailed been used to follow changes in CBF. Fairly re-
differences in axonal response to TAI are impor- cently, application of the above investigative tech-
tant in consideration of the question as to why niques have been applied to human head-injured
some fibers within a particular field are injured patients. These techniques have confirmed and ex-
while other, spatially closely related fibers are not tended the experimental data derived from in vivo
in cases of TAI. It is suggested that the above animal models (Pettus and Povlishock, 1996; Cecil
should be rigorously considered in the formulation et al., 1998; Hovda et al., 1999) by providing good
of any future experimental studies of TAI. evidence that exactly comparable pathophysiolog-
ical responses occur in head-injured patients.
Thus, there is increasingly convincing evidence
Neurobiology and associated neurometabolic that mTBI triggers a complex and interwoven se-
changes of mTBI quence of ionic and metabolic events from which
damaged cells may eventually either recover or de-
In recent years extensive knowledge concerning generate and die. The metabolic cascade is a mul-
pathophysiological processes of mTBI has been tidimensional process (Hovda, 1996; Giza and
gleaned from in vivo and in vitro experiments Hovda, 2000). To briefly summarize, after mTBI,
(Yaghmai and Povlishock, 1992; Hovda, 1996; there is a significant K+ efflux from injured cells,
Pettus and Povlishock, 1996; Cecil et al., 1998; owing to mechanical deformation of the axolemma
Hovda et al., 1994, 1999; Giza and Hovda, 2000; and opening of voltage-dependent K+ channels.
Maroon et al., 2000). Alterations in ionic fluxes, These lead to neuronal firing, depolarization
279

and release of excitatory neurotransmitters. In the et al., 2000). Traumatic, ischemic or anoxic injury
normal brain, excess extracellular K+ is subject to to axons is associated with widespread neuronal
reuptake by surrounding glial cells. This compen- depolarization and release of EAA neurotransmit-
satory mechanism can maintain physiologic extra- ters such as glutamate, leading to the opening of
cellular K+ levels even after mild TBI or ongoing NMDA receptor-associated ion channels and influx
seizure activity (Hovda et al., 1999; Matsushita et of Ca2+ (Graham et al., 2000). The resulting post-
al., 2000; Reinert et al., 2000). But more severe TBI, traumatic Ca2+ storm has been documented using
45
where greater numbers of axons and neurons are Ca-autoradiography after a lateral fluid percus-
injured however, will overcome this glial safety sion injury (Fineman et al., 1993), and indirectly,
valve. Initially there is a slow rise in extracellular through use of cytochemical techniques, as a result
K+. But as the physiological ceiling for K+ bal- of redistribution of membrane pump calcium-AT-
ance is breached, an abrupt increase occurs, leading Pase and Ca2+ influx into myelinated nerve fibers
to depolarization, release of excitatory amino acids of the guinea pig optic nerve after stretch-injury
(EAAs) and a further massive K+ flux though (Maxwell et al., 1995).
EAA/ligand-gated ion channels (Hovda et al., Finally, the injured axon may lose the capacity
1999; Yamamoto et al., 1999). Following this wave to buffer, extrude or to bind the intracellular cal-
of excitation neurons are hyperpolarized and there cium. When the calcium load exceeds the capacity
is consequent suppression of neuronal activity, re- of axoplamic buffers, there is a consensus that ac-
ferred to as ‘‘spreading depression’’, which is tivation of calpains and phospholipases initiate
thought responsible for the neurologic dysfunction processes of cytoskeletal and membrane degener-
across the cortical surface noted in the clinical set- ation. Several recent studies have documented
ting of seizure propagation and migraine aura. both acute calpain activation and regional calpain-
When the extracellular potassium concentration in- induced cytoskeletal proteolysis (Buki et al., 1999;
creases beyond the normal, physiological upper Graham et al., 2000; Saatman et al., 2003). This
limit (4–5 mmol/L) to levels of 20–50 mmol/L and initiates a cascade of structural changes in injured
greater, then inhibition of the action potential and axons beginning with breakdown of the subaxo-
loss of consciousness may occur (Maroon et al., plasmic cytoskeleton (Buki et al., 1999), parts of
2000). The EAAs, especially glutamate, activated the cytoskeleton related to swollen, damaged mi-
kainate and a-amino-3-hydroxy-5-methylisoxazole- tochondria, and later (between 30 and 120 min af-
4-propionic acid (AMPA) receptors located on the ter injury) compaction of cytoskeletal elements
cell body which open channels permeable to both throughout the remnants of the axoplasm. This
Na+ and K+. EAAs also activate N-methyl-D-as- pathology results in focal impairment of axoplas-
partate (NDMA) receptors permeable to Ca2+ as mic transport leading to focal accumulation of
well as Na+ and K+. The release of glutamate transported materials and loss of delivery of mol-
permits a rapid and sustained influx of Ca2+, which ecules required for repair of the damaged axolem-
leads to dysfunction of the oxidative metabolism as ma. This leads to more widespread damage to the
a result of damage to mitochondria (Fineman et al., axolemma (Buki et al., 1999; Maxwell et al., 1999),
1993; Maxwell et al., 1999) and which is amelio- generation of numerous injurious compounds, in-
rated by therapy using cyclosporin A (Okonkwo et cluding free radicals and various inflammatory
al., 1999). The cell therefore becomes increasingly mediators, for example cytokines, prostaglandins
dependent on ATP generated through glycolysis. and leukotrienes. At this stage, since the cell body
Alterations in brain Ca2+ homeostasis through is now also damaged, the potential for axonal re-
loss/inactivation of membrane pump Ca2+ ATPase covery is lost and pathological changes in the in-
and receptors/channels associated with Ca2+ entry jured axon continue until the axon undergoes
(voltage sensitive channels or ionophore-associated secondary axotomy (Maxwell et al., 1997; Gen-
glutamate receptors, such as NMDA receptors) narelli et al., 1998).
have been also associated with regional cerebral Calcium influx may also activate breakdown/
edema, vasospasm and delayed cell death (Graham loss of microtubules (Maxwell and Graham, 1997;
280

Maxwell et al., 1999) since calcium both prevents physiological stress to which cells in the brain are
assembly of microtubules and causes rapid disas- thereby exposed may make those cells much more
sembly of pre-existing polymers (Matsumura and susceptible to secondary injury. Indeed, CBF may
Hayashi, 1976; Langford, 1978). In addition to remain depressed for several days after TBI,
calcium activating calpains (Buki et al., 1999), ac- thereby limiting the ability of the brain to respond
cumulation above normal axoplasmic levels may adequately to subsequent perturbations in energy
activate, cystein proteases, spectrin proteases, cal- demand (Cantu, 2000).
cineurin and phospholipase A2 proteases. Further, Bergsneider et al. (2000) suggested that there are
post-traumatic depolarization will activate three distinct periods of altered tissue/brain me-
phospholipase C. Collectively these act to degrade tabolism following almost all types of animal ex-
a wide range of cytoskeletal and membrane com- perimental TBI — fluid percussion, cortical impact
ponents and eventually lead to cell damage or and SDH — and head-injury in humans. In sum-
death. More specifically, in axons after TAI, post- mary, the initial response is a total-body increase
traumatic elevated concentrations of Ca2+ within in glucose utilization governed by pathophysio-
the axoplasm leads to compaction (i.e., a reduction logic processes, which lead to the endogenous re-
in the spacing between neighboring) of neurofila- lease of EAAs and adrenergic neurotransmitters
ments. This has recently become a major patho- (Bergsneider et al., 2000), which are independent
logical marker of TAI (Pettus and Povlishock, of the functional state of neuronal activation and,
1996; Jafari et al., 1997, 1998; Povlishock et al., we suggest, may reflect responses to release of the
1997; Buki et al., 1999; Stone et al., 2001; Maxwell above by traumatized neurons. This total-body
et al., 2003). Experimental studies following a lat- hypermetabolic state, reflects the activation of en-
eral fluid percussion brain injury using 45Ca auto- ergy-consuming, ATP-dependent, ion pumps
radiography have shown that Ca2+ accumulation which attempt to redress abnormal ion fluxes (Ho-
continues for several days, only returning to con- vda, 1996; Hovda et al., 1999; Bergsneider et al.,
trol levels by the 4th day after the injury (Hovda et 2000; Giza and Hovda, 2000). Secondly, related
al., 1999; Samii et al., 1999). In certain situations, regions of the brain undergo a period of reduced
it may require several seconds or longer for ion glucose utilization, which, over the course of 4
concentrations in injured axons to rise above the days, return to normal values (Fineman et al.,
threshold required for initiation of the develop- 1993). Little is known of the processes affecting
ment of pathology. In mTBI, the presence of such glucose utilization during this second phase of
a time course may explain why some athletes can metabolic depression although it has been sug-
walk from the field and then collapse unconscious gested that Ca2+ sequestration by mitochondria
on the sideline (Maroon et al., 2000). An injured leads to opening of the permeability transition
axon will, probably, attempt to restore membrane pore of the inner mitochondrial membrane (Ok-
potential and recover homeostasis. Membrane inkwo and Povlishock, 1999) and inhibition of
pumps and transporters (e.g., NPP-ase, Na+/ oxidative phosphorylation and energy transduct-
K+-ATPase) and membrane pump Ca2+ATPase ion (Fineman et al., 1993; Bergsneider et al., 2000).
may work overtime, consuming increased amounts This leads to cells utilizing the anaerobic, glycoly-
of ATP. To meet these elevated ATP requirements, tic pathway to synthesize high energy compounds
there is a marked up-regulation of cellular glycol- needed to fuel membrane pumps. Indeed, one ex-
ysis, which occurs within minutes after TBI. Cor- perimental model utilizing cytochemical labeling
related with this period of hyperglycolysis, there is for activity of two ATP-dependent ionic pumps
an increased synthesis of lactate and other toxic has shown that there is a time course of at least 1 h
products (i.e., free fatty acids) in cells of the brain after injury for the loss of pump activity of both
and CSF which leads to further damage to neu- p-NPPase and membrane pump Ca2+ -ATPase
rons. In parallel with this, CBF is reduced to and that this loss of activity extends for at least
oligaemic levels. The resultant mismatch between 24 h after injury (Maxwell et al., 1999). The cor-
oxygen, glucose delivery and consumption and the related influx of Ca2+ into injured axons has been
281

suggested to stimulate both synthesis of glycogen Giza and Hovda, 2000), does not normalize until
and reduce axonal transport of phosphofructoki- between 5 and 10 days after injury (Kawamata
nase leading to increased content of glycogen in et al., 1992) and may result in reduction of both
injured axons with 15 min of injury. Moreover, protein synthesis and oxidative phosphorylation.
there is loss of glycogen deposits in injured axons The term ‘‘energy crisis’’ has been coined to refer
between 7 and 14 days after injury. This loss, to this state of metabolic disequilibrium. The mag-
it is suggested, may correlate with the reported nitude of this ‘‘energy crisis’’ has important impli-
return to normal glucose utilization by 4 days cations for cellular recovery and for increased
(Fineman et al., 1993) and recovery of metabolic vulnerability to secondary insults. As a result cells
function (Pappius, 1981; Fineman et al., 1993; in the brain are more susceptible and are more
Hovda, 1996) which showed that neurological ‘‘vulnerable’’ to damage or even death should the
deficits persist both as long as there is metabolic patient experience a second insult of, even, lesser
depression, and that the rate of recovery of be- intensity (Kawamata et al., 1992; Hovda, 1996;
havioral function parallels the recovery of meta- Buki et al., 1999; Hovda et al., 1999; Giza and
bolic function in the relevant regions of the brain. Hovda, 2000).
However, at present, the mechanisms leading to Overall, the above findings suggest that loss of
the third phase (recovery) are not well understood consciousness after head trauma, the development
although they do appear to be spontaneous of secondary brain damage and the enhanced vul-
(Bergsneider et al., 2000). nerability of cells in the brain to a second insult
Overall, these studies have demonstrated that can be explained largely on the basis of abnormal
the brain is not metabolically quiescent or stag- ionic fluxes, acute metabolic changes and loss of
nant after TBI, but instead exhibits a metabolic autoregulation of CBF immediately after mTBI
response that is both dynamic and stereotypic (Maroon et al., 2000). However, following a trau-
(Fineman et al., 1993). Recent studies in head-in- matic insult to the head, other cells, perhaps the
jured humans (Bergsneider et al., 2000) further most important of which are smooth muscle and
raise the possibility that the extent of the cerebral endothelial cells in the walls of the micro-
metabolic rate of glucose (CMRglc) and oxygen vasculature of the brain, are also damaged. This
(CMRO2) depression after TBI is governed by the results in changes in the extracellular milieu and
severity of injury rather than the level of con- compromise of the ability of neurons to function
sciousness. Hypothetically, it is possible that the and initiate a number of processes that might re-
relative demand placed upon the glycolytic path- establish the normal pericellular environment (Ho-
way will vary with the severity of the injury, as a vda et al., 1999). It is now evident that after mTBI
function of CMRO2. At this time there is too little the stoichiometric relationship of metabolism and
evidence to be able to suggest that in mTBI pa- the coupling to CBF is fundamentally compro-
tients the CMRglc may be a better indicator of mised. There is also evidence that such breakdown
neurological status. For example, Bergsneider et is thought to be responsible for the degree and
al. (2000) cite two head-injured patients with com- extent of vulnerability following mTBI (Hovda et
parable CMRglc where one patient was ambula- al., 1999). However, the concept of injury-induced
tory with a Glasgow Coma Score (GCS) of 15 vulnerability to even minor changes in CBF, in-
while the other patient was comatose and had to creased intracranial pressure and apnea (Hovda,
be mechanically ventilated. Nonetheless, it is sug- 1996; Hovda et al., 1999; Giza and Hovda, 2000)
gested that, with data obtained from a larger al- has risen from studies of patients severely enough
iquot of patients, the CMRglc may prove to be a injured to have been admitted to hospital and,
more sensitive indicator of neurological status in most probably, in intensive care. That is a patient
the mTBI patient. that has most likely experienced moderate or se-
Furthermore, evidence that alteration of glucose vere head-injury. A basic tenet of this review is
metabolism is not restricted to the first hours fol- that the concept of injury-induced vulnerability
lowing mTBI (Hovda, 1996; Hovda et al., 1999; should also be a major concern in the management
282

of head-injured athletes who have experienced sensitivity of commercially available detection kits
mTBI but are not hospitalized. However, there is (Sangtec 100s, AB Sangtec Medical, S-161 02
now an increasing acceptance that injured axons Bromma, Sweden) to analyze NSE and S-100b
(1) undergo or demonstrate a wide spectrum of concentrations in serum samples, have recently at-
physiopathological responses with a time course tracted growing attention in clinical research and
extending to at least several hours, (2) may be diagnostic practice. Both proteins are now ac-
physiologically and functionally stressed after in- cepted as specific neurobiochemical markers of
jury and (3) as a result, may be more susceptible to brain damage in clinical and experimental ischemic
any second, even a milder, insult for at least sev- brain infarction and TBI (Moore, 1965; Skogseid
eral tens of hours. Therefore, after any type of et al., 1992; Ingebrigsten, 1995; Ingebrigsten
head-injury, which may potentiate the develop- et al., 1995, 1997, 1999, 2000; Yamazaki et al.,
ment of pathology within axons and their cell 1995; Ross et al., 1996; Woertgen et al., 1997;
bodies of the injured sportsman, should be advised McKeating et al., 1998; Raabe et al., 1998;
not to resume play in the same game. But rather, Hermann et al., 1999, 2000, 2001; Romner et al.,
those sportsmen should be encouraged to rest. 2000; Biberthaler et al., 2001; De Kruijk et al.,
This rest could, we suggest, provide mildly injured 2001; Pleines et al., 2001).
neurons and other cells the opportunity to respond The serum S-100b protein (S-100b) is a member
to post-traumatic increased demands for energy of a large family of Ca2+-binding proteins, the
and would allow re-establishment of normal chem- cellular synthesis of which has been localized pre-
ical and ionic cellular peri- and intracellular envi- dominantly in astroglia and Schwann cells. When
ronments. Moore discovered the S-100 protein, he suggested
that it was located only in brain tissue (Moore,
1965). Later studies, however, demonstrated its
Biochemical markers of mTBI presence in the body in different forms, and that
the b form predominates in the brain (Moore,
During the past decade neurobiochemical markers 1965; Ingebrigsten et al., 1995, 1997, 1999, 2000;
of brain damage have gained increasing interest in Woertgen et al., 1997; Ingebrigsten, 1998; McK-
experimental and clinical neurotraumatology. Pro- eating et al., 1998; Raabe et al., 1998; Hermann et
teins synthesized in astrogial cells or neurons have al., 1999, 2001; Romner et al., 2000; Biberthaler et
been proposed as markers of cell damage in the al., 2001; Pleines et al., 2001). Increased serum S-
central nervous system. The most common include 100b protein concentrations in peripheral blood
fibrillary acid protein, myelin basic protein, crea- are considered as a marker for dysfunction of the
tine kinase isoenzyme BB, neuron specific enolase blood-brain barrier and therefore, protein S-100b
(NSE) and protein serum-100b. Increased levels of release into peripheral blood may indicate func-
these proteins in the CSF and serum have been tional brain dysfunction without the necessary
observed in patients with various neurological dis- presence of a pathology visible through use of CT
eases (Skogseid et al., 1992; Ingebrigsten, 1995; imaging (Moore, 1965; Ingebrigsten et al., 1995,
Ingebrigsten et al., 1995, 1997; Yamazaki et al., 1997, 1999, 2000; Woertgen et al., 1997; Ingebrigs-
1995; Ross et al., 1996; Woertgen et al., 1997; ten, 1998; McKeating et al., 1998; Raabe et al.,
McKeating et al., 1998; Raabe et al., 1998; Her- 1998; Hermann et al., 1999, 2001; Romner et al.,
mann et al., 1999, 2000, 2001; De Kruijk et al., 2000; Biberthaler et al., 2001; Pleines et al., 2001).
2001; Ringger et al., 2004). Structural proteins of Little is known regarding the mechanism by which
astroglial (glial fibrillary acidic protein) or neu- S-100b passes through the blood-brain barrier and
ronal (neurofilament protein) brain tissue have enters the blood. However, it may be posited that
mostly been used in experimental settings. Alter- where there is loss of autoregulation and stripping
natively, and with a much greater applicability in of endothelium, as has, for example been noted in
the clinical environment, protein serum S-100b and ischemia or severe TBI (Maxwell, 1999; Maxwell
NSE are two markers which, due to present high et al., 1992) that damage to the microvasculature
283

of the brain that has been described in DAI magnetization transfer imaging, allow for the de-
may provide a pathway for passage of S-100b from tection of diffuse white matter brain damage or
injured glial cells into blood. The detection of axonal brain injury with a greater sensitivity than
protein S-100b in peripheral blood may therefore conventional CT procedures. Correspondingly
reflect either an ongoing pathophysiological these probably result in a better prediction of out-
cascade that leads to secondary brain damage or come to the patient (Ingebrigsten et al., 1999).
result from necrosis (apoptosis) of previously NSE is an isoenzyme of enolase and is predom-
damaged cells (Hermann et al., 2000). inantly found in the cytoplasm of neurons, in ne-
However, a recent study showed that the sensi- uroendocrine cells, smooth muscle fibers and
tivity of S-100b measurement for the identification adipose tissue (Skogseid et al., 1992; Yamazaki et
of patients with a pathological CT scan was high al., 1995; Ross et al., 1996; McKeating et al., 1998;
(90%), but the specificity was only 65% with a Hermann et al., 1999, 2000, 2001; De Kruijk et al.,
cutoff level of 0.5 ng/ml (Ingebrigtsen et al., 2000). 2001; Pleines et al., 2001). The cytoplasmic enzyme
Biberthaler et al. (2001) found an even higher sen- is liberated by cell destruction. It has been sug-
sitivity of S-100b measurement (100%) than that gested, therefore, that increased levels of NSE in
of Ingebrigtsen et al., but that the specificity was CSF and peripheral blood indicate neuronal dam-
reduced (40.5%). Therefore, measurement of this age. Herrmann et al. (1999, 2000) demonstrated an
parameter may be helpful as an additional screen- association between the release of both NSE and
ing tool to identify risk groups in a cohort of S-100b protein and the severity of a TBI. Notably,
mTBI patients. Biberthaler et al. (2001) suggested patients with mTBI showed a different temporal
using a cutoff point at a concentration of 0.1 ng/ml profile of NSE in their serum to moderate or se-
plasma since this concentration was the lowest vere TBI patients (Hermann et al., 2000). A sig-
obtained in the serum of patients with mTBI but nificant difference for the concentration of NSE in
lacking signs of intracerebral injury on cranial serum from mild and moderate to severe head in-
computed tomography. jury patients was only obtained up to 24 h after
Further, it has recently been demonstrated that injury. Thereafter serum values for either mTBI
the early post-traumatic elevated serum levels of S- or moderate to severe injury patients were not el-
100b declined rapidly during the first 6 h after the evated and did not differ from control values
trauma. Long lasting post-traumatic elevated S- (Hermann et al., 2000). The release patterns of
100b values probably indicate either an ongoing NSE and S-100b is thought to reflect different
pathophysiological cascade that leads to second- types and dynamics of underlying pathophysiolo-
ary brain damage or the egress of S-100b either as gy between moderate to severe and mild TBI pa-
a result of necrosis or apoptosis of previously tients. Overall, patients with moderate to severe
damaged cells (Romner et al., 2000) or focal dam- head injury show a significantly higher and longer
age to the microvasculature. Further, it has re- release of both markers (Herrmann et al., 2000).
cently been suggested that lack of detectable In addition, recent studies have shown a corre-
concentrations of S-100b protein in serum pre- lation between the clinical outcome and CSF or
dicts a normal CT scan provided the blood sample serum concentrations of protein S-100b and NSE
is collected within the first 2 or 3 h after injury in patients with severe TBI (Skogseid et al., 1992;
(Ingebrigtsen et al., 2000). A comparative analysis Ross et al., 1996; Woertgen et al., 1997). Raabe
of the predictive value of the neurological status, et al. (1998) showed that excessive secondary in-
CT data, and NSE and S-100b serum concentra- crease in S-100b serum concentration relates
tions has shown that the initial (i.e., within 2–3 h of to severe brain damage associated with a fatal out-
injury) serum protein concentration of S-100b is come. The same group reported a significant cor-
the best predictor of outcome for long-term ne- relation between early S-100b values and
uropsychological disorders (Herrmann et al., the volume of cerebral contusions in patients with
2001). Advanced MRI techniques, such as quan- severe TBI but no association between NSE con-
titative MRI analysis or diffusion weighted or centration and contusion volume. However, there
284

is still some controversy concerning the clinical marker concentrations of S-100b are a conse-
significance of such data. For example, Skogseid quence of brain injury including mTBI (De Kruijk
et al. (1992) found a significant correlation between et al., 2001).
maximum NSE concentration and contusion vol- A major conclusion drawn by this review is that
ume. Alternatively, Woertgen et al. (1997) found determination of S-100b serum levels within 2–3 h
no correlation of either NSE or S-100b and the of head-injury may prove to be a useful indicator
severity of intracranial pathology as defined by the for damage to the brain after mTBI. But, impor-
Traumatic Coma Data Bank (TCDB) CT classifi- tantly, serum levels of S-100b also rise after frac-
cation. Further, Hermann et al. (1999, 2000) ture of bones, thoracic contusion, burns and even
showed that the severity of TBI is associated (1) minor bruising (Anderson et al., 2001). On the
with early post-traumatic release of both protein S- other hand, increased serum NSE levels do not
100b and NSE and (2) that the early kinetics of seem to be a specific sensitive marker for mTBI
neurobiochemical markers of brain damage after (De Kruijk et al., 2001). Given that S-100b is a
TBI do reflect intracranial pathology as demon- marker of damage to glial and/or Schwann cells
strated in cranial CT (Hermann et al., 1999, 2000). and NSE indicates damage to neurons, elevated
More recently, Pleines et al. have indicated that the plasma levels of S-100b after mTBI is a good in-
elevation of S-100b and NSE in CSF depends on dicator that damage which results in release of S-
the extent of the injury and that S-100bmay be a 100b is more likely to be localized in the white than
predictor of outcome after TBI, whereas NSE is a the grey matter of the brain. This conclusion is
better indicator of inflammatory responses (Pleines supported by the literature related to the effects of
et al., 2001). In this latter study, release patterns of rotational and acceleration/deceleration injuries to
both proteins did not differ between patients with the brain and allows development of the hypoth-
or without signs of focal neurological deficits. esis that TAI is an important pathophysiological
Herrmann et al. (2001) found that post-trau- cellular response during and following mTBI.
matic serum concentrations of NSE and S-100b However, since S-100b is released by glia than di-
not only reflected the overall severity of brain rectly from injured neurons it is implicit that the
trauma, as defined by GCS scores, but also subtle time course of release of detectable levels of S-100b
neuropsychological dysfunction. Neuropsycholog- in CSF is longer than that of a direct marker of
ical disorders, identified 2 weeks after injury, were neuronal injury. Siman et al. (2004) have recently
correlated with a significantly higher and longer reported, both in vitro and in vivo, elevated levels
lasting release of both brain proteins. Patients with of a-spectrin and calpain/caspase mediated amino-
neuropsychological disorders at 6 months after and carboxy-terminal breakdown products
head trauma, however, also exhibited significantly thereof. Moreover, 150-kDa a-spectrin fragments
higher NSE and S-100b serum concentrations dur- and proteins 14-3-z and 14-3-b were released by 6 h
ing the first 3 days after TBI (Herrmann et al., after both mild, moderate TBI and transient global
2001). ischemia in rat (Siman et al., 2004). Although a-
De Kruijk et al. (2001) investigated whether se- spectrin is widely distributed in many types of cell,
rum concentrations of NSE and S-100b in a group spectrin breakdown products have been reported
of patients with clearly defined mTBI were higher early after TAI in the subaxolemma region (Buki
than in serum of healthy controls. The mean serum et al., 1999), throughout the axoplasm within 2 h
concentrations of NSE did not provide a sensitive of injury, and has a biphasic presentation with
marker between mTBI patients and controls. On a second peak at 4–5 days after TBI in mouse
the contrary, the median serum S-100b concentra- (Saatman et al., 2003). In humans peak levels oc-
tion from 104 mTBI patients was significantly cur in CSF at 2–3 days after severe TBI (Farkas et
elevated compared with control, healthy patients al., 2005) and may fall over 4 days in patients with
(n ¼ 92). Neither did trauma to other parts of the a good outcome (D’Avella et al., 2004). Thus,
body induce an elevation of either S-100b or NSE. there is still a major requirement to define a
Thus, current opinion is that elevated serum marker of injury to neurons in mTBI.
285

Conclusions 9. Recent evidence has suggested that not all


injured axons proceed to secondary axo-
In mTBI tomy but that some demonstrate long-term
compaction of neurofilaments within their
1. Primary axotomy, or shearing of axons, cytoskeleton.
does not occur. 10. There is increasing evidence, from both
2. There is no support for the hypothesis that experimental, animal and clinical human
there is a rapid post-traumatic proteolysis of studies that a mild or severe sudden accel-
the entire axonal cytoskeleton in a signifi- eration–deceleration head trauma may re-
cant number of injured axons except in the sult in cells of the brain assuming a
most severe cases of diffuse head injury. vulnerable state for an unknown period.
3. mTBI may produce only a temporary elect- The fact that cells of the brain are in such a
rochemical disturbance of brain function vulnerable state may predispose the affected
caused by neuronal, chemical, and neuro- athlete to an increased risk of a more serious
electrical changes and without development and dangerous head injury if the athlete is
of overt pathology. exposed to a second head injury, for exam-
4. The great majority of injured axons in cases ple a Second Impact Syndrome, if allowed
of mTBI, concussion and more severe levels to return to the field of play during the same
of injury enter a ‘‘pathological cascade’’ of game.
events that culminate in ‘‘secondary axo-
tomy’’ at least 4 h and extend over many There are many Return-to-play Guidelines to
days after the initial trauma. help sport-physicians decide upon the initial man-
5. The use of modern, sensitive, techniques for agement and determine when an injured athlete
analysis of plasma components (e.g., plasma should return to play. However, considerable con-
levels of S-100b and NSE) allows demon- troversy exists and none of these guidelines has
stration of injury to axons in humans even been developed on the basis of specific, scientific
after mTBI and detection of damaged axons knowledge regarding the processes of either white
at earlier post-traumatic intervals than have matter injury or the recovery thereof. These guide-
been possible until very recently. lines rely first of all upon self-reported symptoms.
6. The time course of formation of axonal If the athlete is confused, the validity of the former
swellings is longer in humans than in the has to be questioned. Generally, most current
majority of animal experimental models — guidelines may allow the athlete to return to play
there is no evidence for recruitment at present after mTBI, even by the deepest grade I, in the
— it is just that reactive axons may be dem- same competition, if his/her clinical examination
onstrated up until 99 days after injury. results are apparently normal at rest and with ex-
7. There is now a clear consensus that axonal ertion. It is acknowledged that experts agree that
changes leading to the development of pa- no athlete who is still symptomatic should be al-
thology are initiated as a result of damage to lowed to return to competition and that ongoing
the axolemma. This damage leads to disrup- and repeated examination should be conducted
tion of ionic homeostatic mechanisms and/or after the injury.
a changed permeability of the axolemma. However, the present review provides strong
8. Responses by microtubules and neurofila- evidence that cells of the brain after mTBI may
ments differ in their time course both within remain alive but in a vulnerable state for an, as yet,
a single axon, between axons in different undetermined period. It is a major conclusion of
tracts within the CNS and between species. the present study that the brain of the injured
The time course for loss of microtubules, athlete may be in a vulnerable state, which may
compaction of neurofilaments and second- predispose the athlete to an increased risk of se-
ary axotomy is longest in humans. rious injury to the brain from a second head injury
286

(i.e., Second Impact Syndrome). In addition, since Adams, J.H., Graham, D.I., Murray, L.S., et al. (1982) Diffuse
brain responses may be blunted resulting in a axonal injury due to non-missile head in humans: an analysis
of 45 cases. Ann. Neurol., 12: 557–563.
longer reaction time to changes in the environ-
Anderson, R.E., Hansson, L.O., Nilsson, O., et al. (2001) High
ment, the injured athlete may be at increased risk serum S100B levels for trauma patients without head injuries.
of serious injury both to the brain and other parts Neurosurgery, 48: 1255–1260.
of the body. Bailes, J.E. and Cantu, R.C. (2001) Head injury in athletes.
Neurosurgery, 48(1): 26–42.
Bergsneider, M., Hovda, D.A., Lee, S.M., et al. (2000) Disso-
Therefore it is recommended that any con- ciation of cerebral glucose metabolism and level of con-
fused player, with or without amnesia, after sciousness during the period of metabolic depression
receiving either a blow or any significant ac- following human traumatic brain injury. J. Neurotrauma,
17(5): 389–401.
celeration–deceleration force to the head, a
Biasca, N. (2000) Review and consequences of the typical ice
grade I mTBI lesion, should be taken off the hockey related injuries in the Swiss Ice Hockey Association.
ice and should not be permitted to return to (SIHA) 1996–1998. In: Biasca N., Montag W.D. and Gerber
play for at least 72 h. Ch. (Eds.), Safety in Ice Hockey, Eighth International Sym-
posium of the International Ice Hockey federation IIHF. In-
ternational Ice Hockey Federation IIHF, Zurich, pp. 36–52.
Biasca, N., Battaglia, H. and Simmen, H.P. (1995) Analyse
typischer Eishockey-Verletzungen. SUVA Monographie Nr.
6, Schweizerische Unfallversicherungsanstalt SUVA, Abtei-
lung Unfallmedizin, CH-6002 Luzern, Switzerland.
Acknowledgements Biasca, N., Battaglia, H. and Tegner, Y. (1999) Diagnose und
Behandlungsvorschläge der Commotio cerebri bei Kontakt-
We would like to express our deep appreciation to und Kampf-sportarten: beispiel im Eishockey. Schweiz.
Prof. Patrick Bishop, Department of Kinesiology, Ärtzezeit., 80(10): 595–598.
Biasca, N. and Tegner, Y. (2001) International Sports Injury
University of Waterloo, Canada, for helping anal-
System — ISIS. Sports Medicine and Hockey; A summit for
yzing the video mechanisms of mTBI by the ice the NHL and Beyond, American Orthopedic Society for
hockey players. We would also like to thank Mr. Sports Medicine (AOSSM) and National Hockey League
Yelverton Tegner, MD, Associated Professor at Team Physician Society (NHLTPS), Hilton Toronto, Can-
the Institution of Health Sciences at the Luleå ada, August 24–26, pp. 15–36.
Biasca, N., Wirth, S. and Tegner, Y. (2002) The avoidability of
University of Technology, SE 961 36 Boden, Swe-
head and neck injuries in ice hockey: a historical review. Br.
den, and Mr. Reto Agosti, MD, Swiss Specialist of J. Sports Med., 36: 410–427.
Neurology, Director of the Headache Center Biberthaler, P., Mussack, T., Wiedemann, E., et al. (2001)
Clinic Hirslanden, Zurich, Switzerland, who as- Evaluation of S-100b as a specific marker for neuronal dam-
sisted us with suggestions and input to further im- age due to minor head trauma. World J. Surg., 25: 93–97.
Bigler, E.D. (1999) Neuroimaging in pediatric traumatic head
prove the quality of this manuscript. Special
injury: diagnostic considerations and relationships to ne-
thanks go to my resident Mr. Stephan Wirth, urobehavioral outcome. J. Head Trauma Rehabil., 14(4):
MD, Orthopedic University Hospital Zurich-Bal- 406–423.
grist, Zurich, Switzerland, who helped me editing Bishop, P.J. (1997) Impact performance characteristics of
the text and the figures. hockey helmets with liners of differing thickness. In: Ashare
A. (Ed.), Third International Symposium on Safety in Ice
Hockey. American Society for Testing and Materials, Phil-
adelphia, pp. 112–117.
Bishop, P.J. (2000) Biomechanics of sports protective equip-
References ment in ice hockey to prevent injuries. In: Biasca N., Montag
W.D. and Gerber Ch. (Eds.), Safety in Ice Hockey, Eight
Abdel-Dayem, H.M., Abu-Judeh, H., Kumar, M., et al. (1998) International Symposium of the International Ice Hockey
SPECT brain perfusion abnormalities in mild or moderate federation IIHF. International Ice Hockey Federation IIHF,
traumatic brain injury. Clin. Nucl. Med., 23(5): 309–317. Zurich, pp. 36–52.
Adams, J.H., Doyle, D., Ford, I., et al. (1989) Diffuse axonal Bishop, P.J. (2001) Protective equipment standards as a means
injury in head injury: definition, diagnosis and grading. His- of reducing injury risk. Sports Medicine and Hockey; A
topathology, 15: 49–59. summit for the NHL and Beyond, American Orthopedic
287

Society for Sports Medicine (AOSSM) and National Hockey De Kruijk, J.R., Leffers, P., Menheere, P.P., et al. (2001) S-
League Team Physician Society (NHLTPS), Hilton Toronto, 100B and neuron-specific enolase in serum of mild traumatic
Canada, August 24–26, p. 57. brain injury patients. Acta Neurol. Scand., 103: 175–179.
Bishop, P.J. and Arnold, J. (1993) The effectiveness of hockey Denny, B. and Russel, W.R. (1941) Experimental cerebral con-
helmets in limiting localized loading on the head. In: Castaldi cussion. Brain, 64: 93–164.
C.R., Bishop P.J. and Hoerner E.F. (Eds.), Safety in Ice Dick, R.W. (1997) Injury in collegiate ice hockey with an em-
Hockey: Second Volume, ASTM STP 1212. American Soci- phasis on injuries to the head. In: Ashare A. (Ed.), Third
ety for Testing and Materials, Philadelphia, pp. 175–182. International Symposium on Safety in Ice Hockey. American
Blumbergs, P.C. (1997) Pathology, Head Injury. In: Reilly P. Society for Testing and Materials, Philadelphia Abstract.
and Bullock R. (Eds.). Chapman and Hall, London, pp. Dixon, J.L. and Brodie, I.K.R. (1993) The new ISO Standards
39–70. for ice hockey helmets and face protectors: moving toward
Blumbergs, P.C., Scott, G., Manavis, J., et al. (1994) Staining of International Standards harmonization and conformity. In:
amyloid precursor protein to study axonal damage in mild Castaldi C.R., Bishop P.J. and Hoerner E.F. (Eds.), Safety in
head injury. Lancet, 344: 1055–1056. Ice Hockey: Second Volume, ASTM STP 1212. American
Blumbergs, P.C., Scott, G., Manavis, J., et al. (1995) Topog- Society for Testing and Materials, Philadelphia, pp. 192–213.
raphy of axonal injury as defined by amyloid precursor pro- Elson, L.M. and Ward, C.C. (1994) Mechanisms and patho-
tein and the sector scoring method in mild and severe closed physiology of mild head injury. Semin. Neurol., 14(1): 8–18.
head injury. J. Neurotrauma, 12: 565–572. Erb, D.E. and Povlishock, J.T. (1988) Axonal damage in severe
Brooks, W.M., Frieman, S.D. and Gasparovic, C. (2001) Mag- traumatic brain injury: an experimental study in the cat. Acta
netic resonance spectroscopy in traumatic brain injury. J. Neuropathol., 76: 347–358.
Head Trauma Rehabil., 16(2): 149–164. Farkas, O., Polgar, B., Szekeres-Bartho, J., et al. (2005) Spec-
Buki, A., Siman, R., Trojanowski, J.Q., et al. (1999) The role trin breakdown products in the cerebrospinal fluid in severe
of calpain-mediated spectrin proteolysis in traumatically in- head injury — preliminary observations. Acta Neurochir.,
duced axonal injury. J. Neuropathol. Exp. Neurol., 58: 147: 855–861.
365–375. Fineman, I., Hovda, D.A., Smith, M., et al. (1993) Concussive
Burke, C.J. (2001) Definition, incidence and initial assessment brain injury is associated with a prolonged accumulation of
of concussion. Sports Medicine and Hockey; A summit for calcium: a 45Ca autoradiographic study. Brain Res., 624:
the NHL and Beyond, American Orthopedic Society for 94–102.
Sports Medicine (August 24–26) (AOSSM) and National Galbraith, J.A., Thibault, L.E. and Matheson, R.A. (1993)
Hockey League Team Physician Society (NHLTPS), Hilton Mechanical and electrical responses of the squid giant axon
Toronto, Canada, pp. 107–108. to simple elongation. J. Biomech. Eng., 115: 13–22.
Cantu, R. (1992) Cerebral concussion in sport: management Garnett, M.R., Blamire, A.M., Corkill, R.G., et al. (2000) Early
and prevention. Sports Med., 14: 64–74. proton magnetic resonance spectroscopy in normal-appear-
Castaldi, C.R. (1993) The sports mouthguard: its use and mis- ing brain correlates with outcome in patients following trau-
use in ice hockey. In: Castaldi C.R., Bishop P.J. and Hoerner matic brain injury. Brain, 123: 2046–2054.
E.F. (Eds.), Safety in Ice Hockey: Second Volume, ASTM Gennarelli, T.A. (1991) Cerebral concussion and diffuse brain
STP 1212. American Society for Testing and Materials, Phil- injuries. In: Torg J.S. (Ed.), Athletic Injuries to the Head,
adelphia, pp. 164–174. Neck and Face (2nd ed.). Mosby Year Book, St. Louis, MO,
Cecil, K.M., Hills, E.C., Sandel, M.E., et al. (1998) Proton pp. 270–282.
magnetic resonance spectroscopy for detection of axonal in- Gennarelli, T.A. and Thibault, L.E. (1982) Biomechanics of
jury in the splenium of the corpus callosum of brain-injured acute subdural hematoma. J. Trauma, 22: 680–686.
patients. J. Neurosurg., 88: 795–801. Gennarelli, T.A., Thibault, L.E., Adams, J.H., et al. (1982)
Christman, C.W., Grady, M.S., Walker, S.A., et al. (1994) Ul- Diffuse axonal injury and traumatic coma in the primate.
trastructural studies of diffuse axonal injury in humans. J. Ann. Neurol., 12: 564–574.
Neurotrauma, 11: 173–186. Gennarelli, T.A., Thibault, L.E. and Graham, D.I. (1998) Dif-
Clark, J.M. (1974) Distribution of microglial clusters in the fuse axonal injury: an important form of traumatic brain
brain after head trauma injury. J. Neurol. Neurosurg. Psy- damage. Neuroscientist, 4(3): 202–215.
chiatry, 37: 463–474. Gennarelli, T.A., Thibault, L.E., Tipperman, R., et al. (1989)
Clayton, P. (1997) A review of head injury data in Canadian Axonal injury in the optic nerve: a model simulating diffuse
hockey league with an emphasis on the incidence of concus- axonal injury in man. J. Neurosurg., 71: 244–253.
sion. In: Ashare A. (Ed.), Third International Symposium on Gentleman, S.M., Roberts, G.W., Gennarelli, T.A., et al. (1995)
Safety in Ice Hockey. American Society for Testing and Ma- Axonal injury: a universal consequence of fatal closed head
terials, Philadelphia Abstract. injury? Acta Neuropathol., 89: 537–543.
D’Avella, D., Aguennouz, M., Angileri, F.F., et al. (2004) De- Giza, C.C. and Hovda, D.A. (2000) Ionic and metabolic con-
tection of a-II spectrin and breakdown products in humans sequences of concussion. In: Cantu R.C. (Ed.), Neurologic
after severe traumatic brain injury. Acad. Emerg. Med., 11: Athletic Head and Spine Injuries. WB Saunders Company,
515–520. New York, pp. 80–100.
288

Grady, M.S., Mclaughlin, M.R., Christman, C.W., et al. (1993) tomography and serum proteion S-100 measurements. J.
The use of antibodies targeted against the neurofilaments Clin. Neurosci., 4: 29–33.
subunits for the detection of diffuse axonal injury in humans. Ingebrigtsen, T., Waterloo, K., Jacobsen, E.A., et al. (1999)
J. Neuropathol. Exp. Neurol., 52: 143–152. Traumatic brain damage in minor head injury: relation of
Graham, D.I. and Gennarelli, T.A. (1997) Trauma. In: Graham serum S-100 protein measurements to magnetic resonance
D.I. and Lantos P.L. (Eds.), Chapter 5 in Greenfield’s Ne- imaging and neurobehavioral outcome. Neurosurgery, 45:
uropathology (6th ed.). Arnold, London, pp. 197–262. 468–476.
Graham, D.I., Mcintosh, T.K., Maxwell, W.L., et al. (2000) Jafari, S.S., Maxwell, W.L., Neilson, M., et al. (1997) Axonal
Recent advances in neurotrauma. J. Neuropathol. Exp. Ne- cytoskeletal changes after non-disruptive axonal injury. J.
urol., 59(8): 641–651. Neurocytol., 26: 207–221.
Gross, H., Kling, A., Henry, G., et al. (1996) Local cerebral Jafari, S.S., Neilson, M., Graham, D.I., et al. (1998) Axonal
glucose metabolism in patients with long-term behavioral cytoskeletal changes after non-disruptive axonal injury. II.
and cognitive deficits following mild traumatic brain injury. Intermediate sized axons. J. Neurotrauma, 15: 955–966.
J. Neuropsychiatry Clin. Neurosci., 8(3): 324–334. Katayama, Y., Becker, D.P., Tamura, T. and Hovda, D.A.
Hartmann, T. (2001) Cholesterol, Ab and Alzheimer’s disease. (1990) Massive increases in extracellular potassium and the
TINS, 24: S45–S48. indiscriminate release of glutamate following concussive
Hermann, M., Curio, N., Jost, S.T., et al. (1999) Protein S-100B brain injury. J. Neurosurg., 73: 889–900.
and neuron specific enolase as early neurobiochemical mark- Kawamata, T., Katayama, Y., Hovda, D.A., et al. (1992) Ad-
ers of the severity of traumatic brain injury. Restor. Neurol. ministration of excitatory amino acid antagonists via micro-
Neurosci., 14: 109–114. dialysis attenuated the increased in glucose utilization seen
Herrmann, M., Curio, N., Jost, S., et al. (2001) Release of bi- following concussive brain injury. J. Cereb. Blood Flow Me-
ochemical markers of damage to neuronal and glial brain tab., 12: 12–24.
tissue is associated with short and long term neuropsycho- Kelly, J.P. (1999) Traumatic brain injury and concussion in
logical outcome after traumatic brain injury. J. Neurol. Ne- sports. JAMA, 282: 989–991.
urosurg. Psychiatry, 70(1): 95–100. Kelly, J.P., Nichols, J.S., et al. (1991) Concussions in sports:
Hermann, M., Jost, S.T., Kutz, S., et al. (2000) Temporal pro- guidelines for the prevention of catastrophic outcome.
file of release of neurobiochemical markers of brain damage JAMA, 266: 2867–2869.
after traumatic brain injury is associated with intracranial Klatky, R., Furuya, Y., Valadka, A.B., Goodman, J.C. and
pathology as demonstrated in cranial computerized tomo- Robertson, C.S. (2002) Comparison of microdialysate argi-
graphy. J. Neurotrauma, 17(2): 113–122. nine and glutamate levels in severely head-injured patient.
Holbourn, A.H.S. (1943) Mechanics of head injury. Lancet, 2: Acta Neurochir. Suppl., 81: 347–349.
438–441. Langford, G.M. (1978) In vitro assembly of dogfish brain tub-
Hovda, D.A. (1996) Metabolic dysfunction. In: Narayan R.K., ulin and induction of coiled ribbon polymers by calcium.
Wiberger J.E. and Povlishock J.T. (Eds.), Neurotrauma. Exp. Cell Res., 111: 139–151.
McGraw-Hill, Inc, New York, pp. 1459–1478. Laprade, R.F. (2001) Ice hockey injuries at the collegiate level.
Hovda, D.A., et al. (1994) Long-term changes in metabolic Sports Medicine and Hockey; A summit for the NHL and
rates for glucose following mild, moderate and severe con- Beyond, American Orthopedic Society for Sports Medicine
cussive head injuries in adult rats. Soc. Neurosci. Abstract, (AOSSM) and National Hockey League Team Physician So-
20: 845. ciety (NHLTPS), Hilton Toronto, Canada, August 24–26,
Hovda, D.A., Prins, M., Becker, D.P., et al. (1999) Neurobi- pp. 37–45.
ology of concussion. In: Bailes J.E., Lovell M.R. and Ma- Levine, B., Cabezy, R., Mcintosh, A.R., et al. (2002) Functional
roon J.C. (Eds.), Sports-related Concussion. Quality Medical reorganization of memory after traumatic brain injury: a
Publishing, Inc., Pittsburgh, PA, pp. 13–51. study with H(2)(15)O positron emission tomography. J. Ne-
Ingebrigsten, T. (1998) Aspect on the management of minor urol. Neurosurg. Psychiatry, 73(2): 173–181.
head injury. Habilitation Work, Department of neurosur- Margulies, S.S. and Thibault, L.E. (1989) An analytical model
gery, Institute of clinical medicine. University of Tromso, of traumatic diffuse brain injury. J. Biomech. Eng., 111:
Norway. 241–249.
Ingebrigsten, T., Kongstad, B.R.P. and Langbakk, B. (1995) Maroon, J.C., Lovell, M.R., Norwig, J., et al. (2000) Cerebral
Increased serum concentrations of protein S-100 after minor concussion in athletes: evaluation and neuropsychological
head injury: a biochemical serum marker with prognostic testing. Neurosurgery, 47(3): 659–672.
value? J. Neurol. Neurosurg. Psychiatry, 59: 103–104. Matsumura, F. and Hayashi, M. (1976) Polymorphism of tub-
Ingebrigtsen, T., Romner, B., Marup-Jensen, S., et al. (2000) ulin assembly. In vitro formation of sheet, twisted ribbon and
The clinical value of serum S-100 protein measurements in microtubule. Biochim. Biophys. Acta, 453: 162–175.
minor head injury: a Scandinavian multicentre study. Brain Matsushita, Y., Shima, K., Katoh, H. and Nawashiro, H.
Inj., 14(12): 1047–1055. (2000) The synergistic effect of high K+ and hypoxia on
Ingebrigsten, T., Rommer, B. and Trumpy, J.H. (1997) Man- extracellular concentrations of neuroactive amino acid in
agement of minor head injury: the value of early computed hippocampus. Acta Neurochir. Suppl., 76: 247–249.
289

Maxwell, W.L. (1999) Cellular responses to ischaemic CNS in- Nevin, N.C. (1967) Neurophathological changes in the white
jury. In: Berry M. and Logan A. (Eds.), Pharmacological matter following head injury. J. Neuropathol. Exp. Neurol.,
strategies in the treatment of CNS injuries. CRC Press, Baton 26: 77–84.
Rouge, pp. 19–41. Ng, S.C., Poon, W.S., Chan, M.T., et al. (2000) Transcranial
Maxwell, W.L., Domleo, A., McColl, G., Jafari, S.S. and Gra- Doppler ultrasonography (TCD) in ventilated head injure
ham, D.I. (2003) Post-acute alterations in the axonal cyto- patients: correlation with stable xenon-enhanced CT. Acta
skeleton after traumatic axonal injury. J. Neurotrauma, 20: Neurochir. Suppl., 76: 479–482.
151–168. Odelgard, B. (1989) The development of head, face, and neck
Maxwell, W.L. and Graham, D.I. (1997) Loss of axonal mi- protectors for ice hockey players. In: Castaldi C.R. and Ho-
crotubules and neurofilaments after stretch-injury to guinea erner E.F. (Eds.), Safety in Ice Hockey: Second Volume,
pig optic nerve fibers. J. Neurotrauma, 14: 603–614. ASTM STP 1050. American Society for Testing and Mate-
Maxwell, W.L., Kosanlavit, R., McCreath, B.J., et al. (1999) rials, Philadelphia, pp. 220–234.
Freeze-fracture and cytochemical evidence for structural and Okonkwo, D.O., Buki, A., Siman, R. and Povlishock,
functional alteration in the axolemma and myelin sheath of J.T. (1999) Cyclosporin A limits calcium-induced axonal
adult guinea pig optic nerve fibers after stretch injury. J. Ne- damage following traumatic brain injury. Neuroreport, 10:
urotrauma, 16: 273–284. 353–358.
Maxwell, W.L., Mccreath, B.J., Graham, D.I., et al. (1995) Okonkwo, D.O. and Povlishock, J.T. (1999) An intrathecal
Cytochemical evidence for redistribution of membrane pump bolus of cyclosporin a before injury preserves mitochondrial
calcium-ATPase and ecto-Ca-ATPase activity, and calcium integrity and attenuates axonal disruption in traumatic brain
influx in myelinated nerve fibers of the optic nerve after injury. J. Cereb. Blood Flow Metab., 19: 443–451.
stretch injury. J. Neurocytol., 24: 925–942. Oppenheimer, D.R. (1968) Microscopic lesions in the brain
Maxwell, W.L., Povlishock, J.T. and Graham, D.I. (1997) A following head injury. J. Neurol. Neurosurg. Psychiatry, 31:
mechanistic analysis of nondisruptive axonal injury: a review. 299–306.
J. Neurotrauma, 14(7): 419–440. Pappius, H.M. (1981) Local cerebral glucose utilization
Maxwell, W.L., Whitfield, P., Suzen, B., et al. (1992) The in thermically traumatized rat brain. Ann. Neurol., 9:
cerebrovascular response to experimental lateral head accel- 484–491.
eration. Acta Neuropathol., 84: 289–296. Pashby, T. (1993) Eye protection in ice hockey: an historical
McGehee, D.V. (1996) Head injury in motor vehicle crashes: review. In: Castaldi C.R., Bishop P.J. and Hoerner E.F.
human factors, effects, and prevention. In: Rizzo M. and (Eds.), Safety in Ice Hockey: Second Volume, ASTM STP
Tranel D. (Eds.), Head Injury and Postconcussive Syndrome. 1212. American Society for Testing and Materials, Philadel-
Churchill Livingstone, London and New York, pp. 57–69. phia, pp. 159–163.
McIntosh, T.K., Faden, A.I., Bendall, M.R. and Vink, R. Pashby, T., Carson, J.D., Ordogh, D., et al. (2001) Eliminate head-
(1987) Traumatic brain injury in the rat: alterations in brain checking in ice hockey. Clin. J. Sport Med., 11(4): 211–213.
lactate and ph as characterized by H and P nuclear magnetic Peerless, S.J. and Rewcastle, N.B. (1967) Shear injuries of the
resonance. J. Neurochem., 49: 1530–1540. brain. J. Can. Med. Assoc., 96: 577–582.
McIntosh, T.K., Faden, A.I., Yamakami, I. and Vink, R. Pettus, E.H., Christman, C.W., Giebel, M.L., et al. (1994)
(1988) Magnesium deficiency exacerbates and pretreatment Traumatically induced altered membrane permeability: its
improves outcome following traumatic brain injury in the rat. relationship traumatically induced reactive axonal changes. J.
J. Neurotrauma, 5: 17–31. Neurotrauma, 11(5): 507–522.
McKeating, E.G., Andrew, P.J. and Mascia, L. (1998) Rela- Pettus, E.H. and Povlishock, J.T. (1996) Characterization of a
tionship of neuron specific enolase and protein S-100 con- distinct set of intra-axonal ultrastructural changes associated
centrations in systemic and jugular venous serum to injury with traumatically induced alteration in axolemma permea-
severity and outcome after traumatic brain injury. Acta Ne- bility. Brain Res., 772: 1–11.
urochir., 71: 117–119. Piccininni, P. (2001) Mouthguards and concussion prevention,
McKenzie, K.J., Mclellan, D.R., Gentleman, S.M., et al. (1996) International Symposium on Concussion in Sport. Br. J.
Is b-APP a marker of axonal damage in short-surviving head Sports Med., 35(5): 375.
injury? Acta Neuropathol., 92: 608–613. Pleines, U.E., Morganti-Kossmann, M.C., Rancan, M., et al.
Moore, B.W. (1965) A soluble protein characteristic of the (2001) S-100B reflects the extent of injury and outcome,
nervous system. Biochem. Biophys. Res. Commun., 19: whereas NSE is a better indicator of neuroinflammation in
739–744. patients with severe traumatic brain injury. J. Neurotrauma,
Muller, M., Schwerdtfegler, K., Maier, B., et al. (2001) Cerebral 18(5): 491–498.
blood flow velocity and inflammatory response after severe Povlishock, J.T. (1986) Traumatically induced axonal damage
traumatic brain injury. Eur. J. Ultrasound, 12(3): 203–208. without concomitant change in focally related neuronal so-
Nedd, K., Sfakianakis, G., Ganz, W., et al. (1993) 99mTc- mata and dendrites. Acta Neuropathol., 70: 53–59.
HMPAO SPECT of the brain in mild to moderate traumatic Povlishock, J.T. (1992) Traumatically induced axonal
brain injury patients: compared with CT, a prospective study. injury: pathogenesis and pathobiological implications. Brain
Brain Inj., 7(6): 469–479. Pathol., 2: 1–12.
290

Povlishock, J.T. (1993) Pathobiology of traumatically induced Samii, A., Badie, H., Fu, K., et al. (1999) Effects of an N-Type
axonal injury in animals and man. Ann. Emerg. Med., 22(6): calcium channel antagonist on calcium-45 accumulation fol-
980–986. lowing fluid-percussion injury. J. Neurotrauma, 16(10):
Povlishock, J.T., Becker, D.P., Cheng, C.L.Z., et al. (1983) 879–892.
Axonal changes in minor head injury. J. Neuropathol. Exp. Sherriff, F.E., Bridges, L.R., Gentleman, S.M., et al. (1994b)
Neurol., 42(3): 225–242. Markers of axonal injury in post mortem human brain. Acta
Povlishock, J.T. and Christman, C.W. (1995) The pathobiology Neuropathol., 88: 433–439.
of traumatically induced axonal injury in animals and hu- Sherriff, F.E., Bridges, L.R. and Sivologanathan, S. (1994a)
mans: a review of current thoughts. J. Neurotrauma, 12(4): Early detection of axonal injury after human head trauma
555–564. using immunocytochemistry for b-amyloid precursor protein.
Povlishock, J.T. and Jenkins, L.W. (1995) Are the pathobio- Acta Neuropathol., 87: 55–62.
logical changes evoked by traumatic brain injury immediate Siman, R., McIntosh, T.K., Soltesz, K.M., et al. (2004) Proteins
and irreversible? Brain Pathol., 5: 415–426. released from degenerating neurons are surrogate markers
Povlishock, J.T., Marmarou, A., McIntosh, T., et al. (1997) for acute brain damage. Neurobiol. Dis., 16: 311–320.
Impact acceleration injury in the rat: evidence for focal axo- Sinson, G., Bagley, L.J., Cecil, K.M., et al. (2001) Magnetiza-
lemmal and related neurofilament sidearm alteration. J. Ne- tion transfer imaging and proton MR spectroscopy in the
uropathol. Exp. Neurol., 56: 347–359. evaluation of axonal injury: correlation with clinical outcome
Povlishock, J.T. and Pettus, E.H. (1996) Traumatically induced after traumatic brain injury. Am. J. Neuroradiol., 22(1):
axonal damage: evidence for enduring changes in axolemmal 143–151.
permeability with associated cytoskeletal changes. Acta Ne- Skogseid, H.M., Nordby, H.K., Urdal, P., et al. (1992) In-
urochir., 66: 81–86. creased serum creatine kinase BB and neuron-specific enolase
Raabe, A., Grolms, C., Keller, M., et al. (1998) Correlations of following head injury indicates brain damage. Acta Ne-
computed tomography findings and serum brain damage urochir., 115: 106–111.
markers following severe head injury. Acta Neurochir., 140: Smith, D.H., Nonaka, M., Miller, R., et al. (2000) Immediate
787–792. coma following inertial brain injury dependent on axonal
Ranalli, D.N. (2000) Prevention of sports-related traumatic damage in the brainstem. J. Neurosurg., 93: 315–322.
dental injuries. Advances in Sports dentistry. Dent. Clin. Sports-Related Recurrent Brain Injuries-United States. (1997)
North Am., 44(1): 35–51. MMWR Morb. Mortal Wkly. Rep., 46: 224–227.
Reinert, M., Hoelper, B., Doppenberg, E., Zauner, A. and Stone, J.R., Singleton, R.H. and Povlishock, J.T. (2001) Intra-
Bullock, R. (2000) Substrate delivery and ionic balance dis- axonal neurofilament compaction does not evoke local ax-
turbance after severe human head injury. Acta Neurochir. onal swelling in all traumatically injured axons. Exp. Neurol.,
Suppl., 76: 439–444. 172: 320–331.
Ricker, J.H., Muller, R.A., Zafonte, R.D., et al. (2001) Verbal Stoner, L.J. and Kreating, M. (1993) Hockey equipment: safety
recall and recognition following traumatic brain injury: a or an illusion? In: Castaldi C.R., Bishop P.J. and Hoerner
(0–15)-water positron emission tomography study. J. Clin. E.F. (Eds.), Safety in Ice Hockey: Second Volume, ASTM
Exp. Neuropsychol., 23(2): 196–206. STP 1212. American Society for Testing and Materials, Phil-
Ringger, N.C., O’Steen, B.E., Brabham, J.G., et al. (2004) A adelphia, pp. 183–191.
novel marker for traumatic brain injury: CSF aII-spectrin Strich, S.J. (1961) Shearing of nerves fibers as a cause of brain
breakdown product levels. J. Neurotrauma, 21: 1443–1456. damage due to head injury: a pathological study of twenty
Roberts, W.O., Brust, J.D. and Leonard, B. (1999) Youth ice cases. Lancet, 2: 443–448.
hockey tournament injuries: rates and patterns compared to Strich, S.J. (1970) Lesions in the cerebral hemispheres after
season play. Med. Sci. Sports Exerc., 31(1): 46–51. blunt head injury. J. Clin. Pathol., 23(4): 166–171.
Romner, B., Ingebrigtsen, T., Kongstad, P., et al. (2000) Trau- Tegner, Y. (2000) Brain concussion in ice hockey: a suggestion
matic brain damage: serum S-100 protein measurements re- for uniform treatment. In: Biasca N., Montag W.D. and
lated to neuroradiological findings. J. Neurotrauma, 17(8): Gerber Ch. (Eds.), Safety in Ice Hockey, Eight International
641–647. Symposium of the International Ice Hockey federation IIHF.
Ross, S.A., Cunningham, R.T., Johnston, C.F., et al. (1996) International Ice Hockey Federation IIHF, Zurich, pp.
Neuron-specific enolase as an aid to outcome prediction in 86–89.
head injury. Br. J. Neurosurg., 10: 471–476. Tegner, Y. (2001) Concussion experiences: Swedish elite ice
Ross, D.T., Meaney, D.F., Sabol, M.K., et al. (1995) Distri- hockey league, International Symposium on Concussion in
bution of forebrain diffuse axonal injury following inertial Sport. Br. J. Sports Med., 35(5): 376.
closed head injury in miniature swine. Exp. Neurol., 126: Tegner, Y., Back, B., Gilenstam, K. and Holm, A. (2000)
291–299. Injury pattern in Swedish Elite Ice Hockey. Hygiae, 109(2):
Ruff, R.M., Crouch, J.A., Troster, A.I., et al. (1994) Selected 204.
cases of poor outcome following a minor brain trauma: Tegner, Y. and Lorentzon, R. (1996) Concussion among Swed-
comparing neuropsychological and positron emission tomo- ish elite ice hockey players. Br. J. Sports Med., 30(3):
graphy assessment. Brain Inj., 8(4): 297–308. 252–255.
291

Tomei, G., Spagnoli, D., Ducati, A., et al. (1990) Morphology Yamamoto, T., Rossi, S., Stiefel, M., et al. (1999) CSF and
and neurophysiology of focal axonal injury experimentally ECF glutamate concentrations in head injured patients. Acta
induced in the guinea pig optic nerve. Acta Neuropathol., 80: Neurochir. Suppl., 75: 17–19.
506–513. Yamazaki, Y., Yada, K., Morii, S., et al. (1995) Diagnostic
Woertgen, C.H., Rothoerl, R.D., Holzschuh, M., et al. (1997) significance of serum neuron-specific enolase and myelin ba-
Comparison of serial S-100 and NSE serum measurements sic protein assay in patients with acute head injury. Surg.
after severe head injury. Acta Neurochir., 139: 1161–1165. Neurol., 43: 267–270.
Yaghmai, A. and Povlishock, J.T. (1992) Traumatically in- Umile, E.M., Sandel, M.E., Alavi, A., et al. (2002) Dynamic
duced reactive change as visualized through the use of mono- imaging in mild traumatic brain injury: support for the the-
clonal antibodies targeted to the neurofilament subunits. J. ory of medial temporal vulnerability. Arch. Phys. Med. Re-
Neuropathol. Exp. Neurol., 51: 158–176. habil., 83(11): 1506–1513.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 20

Traumatic brain injury in infants: the phenomenon of


subdural hemorrhage with hemispheric hypodensity
(‘‘Big Black Brain’’)

Ann-Christine Duhaime and Susan Durham

Pediatric Neurosurgery, Children’s Hospital at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, NH 03756,
USA

Abstract: Clinical and experimental studies of traumatic brain injury during immaturity have been far less
numerous than those involving adults, and many questions remain about differences in injury responses
among patients of different ages. This chapter reviews a distinctive injury pattern common in infants, the
so-called ‘‘big black brain’’ response to acute subdural hematoma. The pathophysiology of this injury
remains incompletely understood. Insights from both clinical observation and experimental studies have
helped to clarify the probable causes of this injury pattern, which appears to require a combination of
stressors during a particular period of maturation.

Keywords: subdural hematoma; infant; black brain; child abuse; pathophysiology

Introduction how clinicians and scientists work synergistically


in head injury research.
There are a number of unique phenomena seen in
clinical practice that distinguish infant traumatic
brain injury from that occurring in older children Early clinical observations
or adults. One of the most dramatic of these is the
so-called ‘‘big black brain,’’ a pattern of tissue loss It has long been recognized that acute subdural
affecting the entire supratentorial hemisphere in hematoma in infancy could be associated with pro-
association with acute subdural hematoma. The found brain injury. The pioneering observations of
circumstances required to cause this injury pattern Guthkelch (1971) and Caffey (1972, 1974) linked
and the pathophysiology of the extensive paren- infantile subdural with a range of neurologic defi-
chymal destruction remain incompletely under- cits including chronic disability, coma, and death.
stood. This chapter will address insights gained These injuries were hypothesized to occur most of-
into this injury by both clinical observations and ten from violent manual shaking. The hypothesis
experimental strategies. Both approaches have that shaking caused these injuries was based on
contributed to advancing understanding of the re- statements from some perpetrators and witnesses
sponse of the infant to brain trauma, and illustrate who described shaking, the frequent paucity of vis-
ible external cranial injury in affected infants, an
association with distal metaphyseal and rib frac-
Corresponding author. Tel.: +1 603-653-9880; tures, and contemporaneous experimental findings
Fax: +1 603-650-0908; E-mail: acd@hitchcock.org on the role of rotational forces in subdural

DOI: 10.1016/S0079-6123(06)61020-0 293


294

hematoma and diffuse brain injuries (Ommaya and extensively distributed low density seen on the CT
Yarnell, 1962; Ommaya et al., 1968, 1973). The scan of such infants, the term ‘‘big black brain’’ has
forces and mechanisms necessary to cause acute been used to describe those with hypodensity in-
subdural hematoma in infancy remain uncertain, volving the entirety of one or both hemispheres
and many affected infants with accidental or in- (Duhaime et al., 1993; Graupman and Winston,
flicted events do, in fact, have evidence of head im- 2006). This nomenclature evolved from the use of
pact (Hahn et al., 1983; Duhaime et al., 1987, 1998; the term ‘‘black brain’’ to describe the CT appear-
Alexander et al., 1990; Ghahreman et al., 2005). In ance of diffuse bilateral hypodensity and loss of
this chapter we will focus on a specific pattern of gray-white differentiation, with or without relative
immature brain response to acute subdural hema- sparing of the thalami (‘‘reversal sign’’), seen most
toma, regardless of mechanism of injury. often in cases of severe or fatal pediatric head in-
Not all infants with acute subdural hematoma jury (Cohen et al., 1986; Whyte and Pascoe, 1989).
develop severe brain injury. The spectrum of clini- In a subset of infant subdurals, hypodensity is seen
cal outcomes ranges from no deficits to death which extends from the frontal pole to the occipital
(Ludwig and Warman, 1984; Duhaime et al., 1987, pole, crosses multiple vascular territories, and stops
1996; Gilles and Nelson, 1998; Ewing-Cobbs et al., abruptly at the tentorium. This pattern may be
1999; Ghahreman et al., 2005). As CT scanning present at presentation to the hospital or evolve
became widely available, patterns of brain damage over several days (Dias et al., 1998). In unilateral
emerged in the premorbid state and in survivors. cases, a contralateral frontal wedge-shaped region
Areas of hypodensity often were seen which might of low density also typically is seen, but the rest of
be patchy in distribution or could involve the entire the supratentorial compartment does not exhibit
supratentorial compartment unilaterally or bilater- hypodensity or loss of gray-white differentiation
ally (Zimmerman et al., 1979; Cohen et al., 1986; (Fig. 1). The hypodense hemisphere(s) typically go
Dias et al., 1998; Gilles and Nelson, 1998). Because on to sustain rapidly progressive atrophic changes
of the typical dark-appearing, homogeneous, and indicative of severe, diffuse brain destruction

Fig. 1. Two-year-old boy with a fall from a bunk bed resulting in acute left subdural hematoma. (A) Acute CT scan, with midline shift.
Hemispheres appear to have similar density at this time-point. The patient underwent emergency clot evacuation. (B) Four days post-
injury. The child has developed hypodensity and swelling of the entire left hemisphere. He underwent aggressive management of
intracranial pressure and survived with a right hemiparesis and hemianopsia.
295

Fig. 2. Twenty-three month old boy who sustained a witnessed accidental injury with an acute right subdural hematoma. He stood up
initially but then had a generalized seizure within seconds after impact. He received medical attention within minutes, and there was no
apnea or hypoventilation noted. (A) Acute CT scan. (B) CT scan after clot evacuation. There was no brain swelling at surgery. Note
lack of hypodensity, and bifrontal air. The patient regained consciousness promptly after surgery. Despite the symmetric radiologic
appearance of the hemispheres, the child was noted to have a left hemiparesis postoperatively. (C) MRI scan done on postoperative
day 2, FLAIR sequence. Note the high signal in the basal ganglia. (D) MRI, T2 weighted sequence, several months post-injury. Note
the widespread signal change and atrophy in the right hemisphere. (E) MRI, T1 sequence with contrast, 10 years after injury. There is
marked atrophy of the right hemisphere. The patient had a persistent hemiparesis and hemianopsia, developmental delays, and a
seizure disorder. He became seizure-free with no new deficits after hemispherectomy.

(Fig. 2). In children with subdural hematomas, the hypodensity correlates with worse acute clinical
bilateral pattern is seen about twice as commonly status and with worse prognosis (Duhaime et al.,
as the unilateral pattern, and occurs more com- 1996; Ewing-Cobbs et al., 1998; Gilles and Nelson,
monly in very young infants, while older infants 1998). Mortality in children with unilateral or bi-
and toddlers more often develop the unilateral lateral hemispheric involvement is 67% (Duhaime
form of ‘‘big black brain’’ (Duhaime et al., 1993; et al., 1993). Even many years after injury, survi-
Gilles and Nelson, 1998). The extent of the vors of bilateral ‘‘big black brain’’ remain blind,
296

non-ambulatory, nonverbal, and profoundly deve- of the basal ganglia can also been seen on MRI
lopmentally delayed. Those with the predominantly (Figs. 2 and 3). Therefore, the question has arisen as
unilateral form may make a better functional re- to whether this striking injury pattern reflects a
covery but remain severely impaired (Duhaime unique mechanism of injury, the presence of a par-
et al., 1996). ticular type of secondary injury, or a unique re-
While many workers have described the areas of sponse of the immature brain to trauma. Since
hypodensity seen on CT as ‘‘edema’’ or ‘‘infarct- bilateral ‘‘black brain’’ can occur from diffuse hypoxic-
ion,’’ these terms should be used with caution. ischemic insults as well as in the setting of subdural
This is because the pathophysiologic processes hematomas, we will focus on the unilateral pattern
these terms most often connote, in other ages and as the traumatic entity requiring a unique path-
with adult disorders, may not apply in this context. ophysiologic explanation. Both experimental and
For instance, excitotoxic lesions, to which the im- clinical investigations offer insights, as reviewed
mature brain may be particularly vulnerable, can below.
occur in the setting of normal vascular perfusion.
The pathology and radiology of this type of proc-
ess may look similar to that occurring from vas- Experimental models
cular occlusion, since both represent a mismatch
between metabolic demand and substrate delivery. The rodent subdural hematoma model most
If such a pathophysiology were in play, it might widely in use presently was first described by
not be necessary to postulate a mechanism requir- Miller et al. (1990). The model created a subdural
ing large vessel occlusion, such as strangulation, as hematoma by injecting autologous blood into the
some authors have done (Bird et al., 1987). Thus, subdural space of the cerebral convexity through a
care should be taken to describe findings rather small burr hole. Histopathology demonstrates a
than ascribing mechanisms that might be associ- bowl-shaped volume of brain damage underlying
ated with these radiologic findings in other ages the clot. By measuring brain metabolism and cere-
and clinical contexts (Gilles and Nelson, 1998). bral blood flow, investigators have demonstrated
Older children and adults with acute subdural that perfusion in the area of the clot is decreased,
hematoma may suffer extensive concomitant brain while metabolism is increased, creating a relative
damage, presumably due to the effects of primary mismatch between substrate delivery and demand.
mechanical brain injury and/or to secondary insults The perfusion abnormality appears to be mediated
including hypoxia, ischemia, or elevated intracra- by microvessel vasospasm rather than by large
nial pressure. However, the rapid appearance of vessel abnormalities (Inglis et al., 1990; Kuroda
extensive, diffuse supratentorial hypodensity with and Bullock, 1992). Microdialysis studies with the
loss of gray-white differentiation in the unilateral model have shown an increase in extracellular
or bilateral patterns described is seen routinely only glutamate, and treatment with glutamate receptor
in infants and toddler-aged children. The vast ma- blockers limits the volume of damage, suggesting
jority of children with the specific injury type re- that the increased metabolism seen under the
viewed in this paper are under 3 years of age. For hematoma is related to excitotoxic stress (Bullock
the sake of brevity we most often use the term et al., 1990, 1991; Inglis et al., 1992). Similar dam-
‘‘infants’’ in this chapter, but it should be under- age was not seen if a comparable volume of blood
stood that this phenomenon occurs in toddlers as was simply layered over the exposed cortical sur-
well. face in rodents and a clot left in place for several
More recent studies using magnetic resonance days (Duhaime et al., 1992). This demonstrates
imaging (MRI) have shown changes on diffusion- that the presence of blood alone is insufficient to
weighted imaging which corroborate the idea that cause damage, but that the blood must be injected
some form of hypoxic-ischemic injury or perfusion- into a closed space to cause injury.
demand mismatch is particularly common in in- While these studies are intriguing, the pathology
flicted injury (Ichord et al., 2007). Early involvement using the rodent subdural hematoma model does
297

Fig. 3. Two-year-old girl with injury, suspicious for inflicted trauma with acute right subdural hematoma. She had a seizure witnessed
in an outside emergency department, and was unresponsive with a dilated right pupil on presentation. The clot was evacuated and the
brain was noted to be slightly full at surgery, so a hemicraniectomy was performed. Intracranial pressure was measured and easily
controlled, and excellent brain perfusion was maintained. The patient was intubated but responsive and was able to follow commands.
(A) MRI, T2 sequence, done 1 day after surgery. Note symmetric appearance of brain parenchyma. (B) Diffusion-weighted sequence.
Note bright signal in basal ganglia and temporal and occipital cortex. (C) CT scan, postoperative day 3. Note development of
hypodensity of entire right hemisphere, with increased swelling. (D) MRI, diffusion sequence, one week postoperatively. Note bright
signal in entire hemisphere, as well as contralateral basal ganglia. (E) MR angiogram. All arteries appear patent. (F) MRI, fast spin-
echo T2 weighted sequence, 2 months post-injury. Note widespread damage to right hemisphere. The patient has a hemiparesis and
hemianopsia.

not reproduce the pathology of the ‘‘big black model also failed to reproduce the ‘‘big black
brain.’’ Injection of blood causes a localized lesion brain’’ phenomenon, as it too created a localized
rather than one involving the entire hemisphere. injury. However, unlike the rodent lesion, which
To learn whether the infantile pattern could be was largely cortical, the piglet lesion preferentially
reproduced by adapting the rodent model to an affected the underlying white matter. The authors
immature, gyrencephalic brain, Shaver et al. hypothesized that the white matter was affected
(1996) injected blood into the subdural space of because of its increased vulnerability during early
three-week-old piglets. Their model was modified life, when the metabolic rate is relatively high in
by use of a ‘‘cranial window’’ through the bone white matter but the vascular supply is immature
that allowed confirmation of a thin but extensive and may respond inadequately to increased de-
subdural clot overlying the hemisphere, compara- mand. It is for this reason that the white matter in
ble to that seen most often in human infants. This infants is thought to be preferentially vulnerable to
298

damage from perinatal hypoxic-ischemic insults We will now return to the clinical arena, where
and prematurity (Rorke, 1992). additional observations relevant to this issue have
Both the rodent and the piglet subdural injec- been made.
tion models fail to mimic the mechanical trauma
that occurs during the injury event in patients.
Since the presence of blood in the subdural space The role of apnea
was insufficient to cause the hemispheric damage
seen in infant subdural injuries, Duhaime et al. It has long been recognized that infants are sus-
(2000) performed a series of studies using piglets to ceptible to apnea from a variety of causes, includ-
isolate the effect of mechanical trauma on the ing head injury. That children with subdurals often
immature brain. This was done to test the hypo- present with hypoventilation or frank apnea is well
thesis that the infant brain might be particularly known (Johnson et al., 1995; Kemp et al., 2003;
vulnerable to damage from mechanical trauma Ichord et al., 2007). Therefore, it has been hypoth-
relative to other ages, thus explaining the extensive esized that hypoxia/ischemia, rather than trauma
hemispheric injuries seen with acute subdural hema- itself, is the cause of the diffuse hypodensity pat-
toma. The group developed a model of scaled cor- tern seen in the most severe of these injuries. The
tical impact that delivered a mechanically compa- fact that many of these infants have been the vic-
rable strain load to the same brain structures tims of inflicted trauma has been thought to
at different ages. These studies showed that rather contribute to this association. Specifically, it has
than being selectively vulnerable to mechanical been speculated that after the infant is injured, the
trauma, the infant brain was relatively resistant perpetrator does not seek immediate medical atten-
to injury from mechanical deformation (Duhaime tion, hoping the child will recover spontaneously.
et al., 2003). Therefore, another explanation for This leads to delay in care. If the child hypoven-
the unique pattern of extensive brain damage seen tilates during this delay period, diffuse hypoxic
with subdurals in infancy was needed. brain damage may result.
Other workers have attempted to more closely This scenario matches the histories obtained in
reproduce the mechanical forces that might be re- many cases, and likely contributes to the damage
sponsible for infant subdural hemorrhage using ani- seen in many infants. However, it is not a sufficient
mal models. Because of the long-held concept that explanation for all aspects of the ‘‘black brain’’
the injury might be caused by manual shaking of an phenomenon. This is because there are well-
infant, Smith et al. (1998) subjected rats to pro- documented accidental cases of subdural hema-
longed mechanical shaking. While subarachnoid toma in infants and toddlers that were witnessed
hemorrhage and surface contusions were found, and in which children got immediate medical
hemispheric tissue loss was not produced in this attention with no apparent apnea or hypoventila-
model. Anesthetized, immature sheep subjected to tion, but in which the phenomenon of hemispheric
30 min of intermittent, violent manual shaking hypodensity nonetheless appeared (Figs. 1 and 2).
exhibited similar finding to the shaken rats, with- The other limitation of apnea as the sole explana-
out hemispheric loss (Finnie et al., 2006). Piglets tion for the phenomenon is that it fails to account
subjected to a single or double high-velocity head for the fact that in one-third of the cases, the
rotation showed diffuse axonal injury and focal hypodensity is unilateral. One would generally
frontal subdural hemorrhage, but have not to date expect a diffuse insult like hypoxia to affect the
demonstrated the ‘‘big black brain’’ phenomenon brain symmetrically. The observation that one
(Raghupathi and Margulies, 2002; Raghupathi hemisphere can be totally destroyed while the
et al., 2004). other remains relatively preserved suggests that
It can be seen that in all these experimental more than one factor is in play. Gilles and others
models, elements of injury occur in isolation, have noted that in unilateral cases, the hypoden-
whereas in the clinical setting, multiple patho- sity occurs on the side of the subdural hematoma
physiologic stressors may occur in combination. (Duhaime et al., 1998; Gilles and Nelson, 1998;
299

Gilles et al., 2003). Therefore, it appears that some injury as determined by both immunohistochemi-
synergistic effect of the hemorrhage (or the forces stry and light microscopic findings. However,
that caused it) and a second insult may be needed. patients frequently did exhibit ‘‘ischemic’’ findings
(Geddes et al., 2001a, b). Additionally, damage at
the cervicomedullary junction was often encoun-
The role of seizures
tered, also raising the question whether apnea due
to damage in this location might contribute to the
Infants with head injury and other neurologic in-
findings; this has been hypothesized by others as
sults often present with seizures, which also may
well (Hadley et al., 1989; Johnson et al., 1995).
occur subclinically, especially in very young chil-
However, Geddes and colleagues did not address
dren. The incidence of clinical seizures in inflicted
the specific phenomenon of unilateral ‘‘big black
head injuries has been reported in 40–79% of pa-
brain’’ in their series.
tients (Ludwig and Warman, 1984; Johnson et al.,
Kohanek and colleagues have measured a wide
1995; Gilles and Nelson, 1998; Ghahreman et al.,
variety of cerebrospinal fluid markers from ventri-
2005; Bechtel et al., 2006). Clancy et al. (1988) have
culostomy samples from infants with traumatic
noted that up to 79% of seizures in neonates are
brain injury compared to those found in lumbar
subclinical, that is, are only detected on EEG re-
puncture samples from uninjured infants. They
cordings. This would suggest the possibility that
have reported higher levels of excitatory amino
even more seizure events may occur in infants with
acids and other markers of cellular damage in fluid
inflicted injury than are apparent clinically, espe-
from infants with inflicted injuries and in younger
cially in the youngest patients. Pharmacologic
infants compared to older children and those with
agents used for sedation and/or paralysis during
accidental mechanisms (Ruppell et al., 2001;
head trauma management in the intensive care unit
Berger et al., 2006). Whether these differences
could influence clinical epileptic events, with some
reflect an age effect or more extensive damage in
agents (such as midazolam) potentially protecting
the inflicted injury patients (primary or secondary)
against seizures, and others (narcotics, paralytics)
remains unclear.
potentially obscuring their detection. Seizures are
known to increase excitotoxic stress in animal
models and to be associated with increased meta-
Brain swelling, variability, and decompressive
bolic demand and worse outcome in human pa-
craniectomy
tients. Therefore, some workers have hypothesized
that the frequent occurrence of seizures in infants
Most babies with bilateral or unilateral hemi-
with traumatic brain injury may contribute to the
spheric hypodensity develop brain swelling and
pathophysiology of an infarct-like picture, such as
increased intracranial pressure. Survival is better
occurs in hemispheric hypodensity (Duhaime et al.,
in the youngest infants, probably because of their
1998). However, the exact contribution of seizures,
ability to split the sutures to relieve some of the
clinical or subclinical, to this phenomenon remains
pressure. Interestingly, in some cases brain swell-
unknown.
ing is not problematic, and why some children do
not develop significant swelling remains unknown
Neuropathology and clinical neurophysiology (see Figs. 1–3). It may be that acute injury factors
or genetic differences determine this variability.
As immunohistochemical techniques have become In recent years, hemicraniectomy has gained in-
available, additional detail about the neuropatho- creasing acceptance as a means to alleviate the
logy of fatal infant inflicted injuries has been brainstem compressive effects of brain swelling in
determined. Geddes reported that contrary to infants and young children (Cho et al., 1995). It is
older studies in which light microscopy was used not yet clear in which children and at what time-
to assess neuropathology, in her series most pa- point this should be undertaken, and the effect of
tients exhibited little in the way of diffuse axonal this practice on outcome has been difficult to
300

assess, although it does appear to be effective in organism has a limit to its ability to compensate,
lowering intracranial pressure (Taylor et al., 2001). and when this limit is reached, the resulting
At present it appears that even in those cases in decompensation is often precipitous.
which the hypodensity appears over several days, It has been demonstrated in a number of con-
the processes involved have already begun at prese- texts that the immature brain’s response to insult
ntation and do not appear reversible with current varies from the adult response. Its response to glo-
therapies (Duhaime et al., 1996; Dias et al., 1998; bal hypoxia-ischemia varies, with some ages and in
Graupman and Winston, 2006). Therefore, treat- some outcome measures having increased vulner-
ment strategies are aimed at protecting the un- ability to cell death and in others, increased resist-
damaged hemisphere. It has been hypothesized ance compared to the mature brain (Painter, 1995;
that the nearly universal finding of a wedge of Johnston et al., 2001; Vanucci and Hagberg, 2004).
damage in the contralateral medial frontal lobe Resistance to other insults seen in the context of
results from subfalcine herniation with trapping of trauma has been demonstrated by a number of ex-
the callosomarginal branch of the anterior cerebral perimental models cited above, including isolated
artery against the falx, resulting in focal infarction focal mechanical trauma and injected subdural
(Gilles and Nelson, 1998). In the setting of hemi- hematoma with increased intracranial pressure; in
craniectomy, this internal herniation is amelio- these contexts, immaturity confers protection.
rated. However, the authors have observed that In cases of infantile subdural hematoma with
some contralateral progression can occur despite hemispheric hypodensity, it appears likely that the
early relief of pressure, raising the possibility of a brain is subjected to a combination of stresses that
neurochemically-mediated process rather than a exceed its capacity to compensate. Most clinical
purely vascular occlusive one (Fig. 3). scenarios would point to a combination of local
perfusion decrement over the surface of the cere-
bral hemisphere related to the presence of subdural
Pathophysiology of the ‘‘Big Black Brain’’: blood, in combination with a second, more global
a hypothesis insult. This could include apnea/hypoxia, hyper-
carbia, hypotension, or increased metabolic de-
Both experimental and clinical evidences support mand from seizures. There may be additional in-
the idea that no single insult is sufficient to explain sults that are at present difficult to discern or
all cases of unilateral hemispheric hypodensity in measure. Why the process involves the entire hemi-
association with subdural hematoma in infants and sphere, even beyond the extent of visible subdural
toddlers. Not all children with subdural hemorrh- blood, remains incompletely understood. It ap-
age develop this finding; thus subdural blood itself pears to represent a diffuse decompensation re-
appears to be an insufficient cause. Global physio- sulting in widespread parenchymal death that
logic insults cannot fully explain the distribution in under certain circumstances may reach threshold
unilateral cases. Hemispheric hypodensity can be only on the side of the larger hemorrhage. Indi-
seen in nonaccidental and accidental injuries, with vidual differences with respect to cerebrovascular
apnea and with no apparent apnea, with clinical responses, acute injury cascades, inflammation,
seizures and without. Brain swelling usually but apoptotic pathways, or other neurochemical proc-
not always occurs. Atrophy is rapid and profound. esses likely influence why some patients have pro-
The phenomenon of subdural blood with unilateral found swelling and others do not.
hemispheric hypodensity has not yet been repro-
duced in an experimental model.
In many situations, infants and children demon- Conclusion
strate a remarkable ability to compensate for sys-
temic physiologic stresses. A common example is The unique infant pattern of unilateral hemi-
the child’s ability to maintain blood pressure in the spheric hypodensity associated with accidental
face of hypovolemia. However, the immature or inflicted acute subdural hematoma provides
301

insight into age-related differences in brain injury Clancy, R.R., Legido, A. and Lewis, D. (1988) Occult neonatal
response. This pattern of extensive and homoge- seizures. Epilepsia, 29: 256–261.
Cohen, R.A., Kaufman, R.A., Myers, P.A. and Towbin, R.B.
neous tissue destruction appears to require a com-
(1986) Cranial computed tomography in the abused child
bination of insults that overwhelm the immature with head injury. AJR, 146: 97–102.
brain’s ability to compensate and affects the entire Dias, M.S., Backstrom, J., Falk, M. and Li, V. (1998) Serial
hemisphere. Because the specific physiologic stres- radiography in the infant shaken impact syndrome. Pediatr.
ses vary from case to case, including hypoxia, Neurosurg., 29: 77–85.
hypotension, hypercarbia, and seizure activity, Duhaime, A.C., Bilaniuk, L. and Zimmerman, R. (1993) The
‘‘big black brain’’: radiographic changes after severe inflicted
different factors that decrease substrate delivery head injury in infancy. J. Neurotrauma, 10: S59.
or increase metabolic demand may be at play for Duhaime, A.C., Christian, C., Moss, E. and Seidl, T. (1996)
different patients. The observation that this pat- Long-term outcome in children with the shaking-impact
tern occurs uniquely in infants and toddlers more syndrome. Pediatr. Neurosurg., 24: 292–298.
Duhaime, A.C., Christian, C.W., Rorke, L.B. and Zimmerman,
likely reflects their age-dependent brain response
R.A. (1998) Nonaccidental head injury in infants: the
to combined stresses rather than reflecting a single, ‘‘Shaken Baby Syndrome.’’ New Engl. J. Med., 338:
specific mechanism or circumstance of injury. 1822–1829.
Duhaime, A.C., Gennarelli, L., Yachnis, A.T. and Ross, D.T.
(1992) Acute subdural hematoma: is the blood itself toxic?
References Pediatric Section, American Association of Neurological
Surgeons, Annual Meeting. Vancouver, B.C.
Alexander, R., Sato, Y., Smith, W. and Bennett, T. (1990) Duhaime, A.C., Gennarelli, T.G., Thibault, L.E., Bruce, D.A.,
Incidence of impact trauma with cranial injuries ascribed to Margulies, S.S. and Wiser, R. (1987) The shaken baby
shaking. Am. J. Dis. Child, 144: 724–726. syndrome: a clinical, pathological, and biomechanical study.
Bechtel, K., Stoessel, K., Leventhal, J.M., Ogle, E., Teague, B., J. Neurosurg., 66: 409–415.
Lavietes, S., Banyas, B., Allen, K., Dziura, J. and Duncan, C. Duhaime, A.C., Hunter, J.V., Grate, L.L., Kim, A., Demidenko,
(2006) Characteristics that distinguish accidental from abu- E., Golden, J.A. and Harris, C. (2003) Magnetic resonance
sive injury in hospitalized young children with head trauma. imaging studies of age-dependent responses to scaled focal
Pediatrics, 114: 165–168. brain injury in the piglet. J. Neurosurg., 99: 542–548.
Berger, P.R., Adelson, P., Richichi, R. and Kochanek, P.M. Duhaime, A.C., Margulies, S.S., Durham, S.R., O’Rourke,
(2006) Serum biomarkers after traumatic and hypoxemic M.M., Golden, J.A., Marwaha, S. and Raghupathi, R. (2000)
brain injuries: insight into the biochemical response of the Maturation-dependent response of the piglet brain to scaled
pediatric brain to inflected brain injury. Dev. Neurosci., 28: cortical impact. J. Neurosurg., 93: 455–462.
327–335. Ewing-Cobbs, L., Kramer, L., Prasad, M., Canales, D.N.,
Bird, C.R., McMahan, J.R., Gilles, F.H., Senac, M.O. and Louis, P.T., Fletcher, J.M., Vollero, H., Landry, S.H. and
Apthorp, J.S. (1987) Strangulation in child abuse: CT diag- Cheung, K. (1998) Neuroimaging, physical, and develop-
nosis. Radiology, 163: 373–375. mental findings after inflicted and noninflicted traumatic
Bullock, R., Butcher, S. and Mcculloch, J. (1990) Changes in brain injury in young children. Pediatrics, 102: 300–307.
extracellular glutamate concentration after acute subdural Ewing-Cobbs, L., Prasad, M., Kraer, L. and Landry, S. (1999)
haematoma in the rat: evidence for an ‘‘excitotoxic’’ mech- Inflicted traumatic brain injury: relationship of developmental
anism? Acta Neurochir. Suppl., 51: 274–276. outcome to severity of injury. Pediatr. Neurosurg., 31: 251–258.
Bullock, R., Butcher, S.P., Chen, M., Kendall, L. and Mccul- Finnie, J.W., Manavis, J. and Blumbergs, P.C. (2006) An ovine
loch, J. (1991) Correlation of the extracellular glutamate model of the shaken baby syndrome. International Neuro-
concentration with extent of blood flow reduction after sub- trauma Society 8th Annual Neurotrauma Symposium. Rotter-
dural hematoma in the rat. J. Neurosurg., 74: 794–802. dam, The Netherlands, International Neurotrauma Society.
Caffey, J. (1972) On the theory and practice of shaking infants: Geddes, J.F., Hackshaw, A.K., Vowles, G.H., Nickols, C.D.
its potential residual effects of permanent brain damage and and Whitwell, H.L. (2001a) Neuropathology of inflicted head
mental retardation. AJDC, 124: 161–169. injury in children: I. Patterns of brain damage. Brain, 124:
Caffey, J. (1974) The whiplash shaken infant syndrome: manual 1290–1298.
shaking by the extremities with whiplash-induced intracranial Geddes, J.F., Hackshaw, A.K., Vowles, G.H., Nickols, C.D.
and intraocular bleedings, linked with residual permanent and Whitwell, H.L. (2001b) Neuropathology of inflicted head
brain damage and mental retardation. Pediatrics, 54: injury in children: II. Microscopic brain injury in infants.
396–403. Brain, 124: 1299–1306.
Cho, D., Wang, Y. and Chi, C. (1995) Decompressive cranio- Ghahreman, A., Bhasin, V., Chaseling, R., Andrews, B. and
tomy for acute shaken/impact baby syndrome. Pediatr. Lang, E.W. (2005) Nonaccidental head injuries in children: a
Neurosurg., 23: 192–198. Sydney experience. J. Neurosurg., 103: 213–218.
302

Gilles, E.E., Mcgregor, M.L. and Levy-Clark, G. (2003) Retinal Ommaya, A.K., Faas, F. and Yarnell, P. (1968) Whiplash in-
hemorrhage asymmetry in inflicted head injury: a clue to jury and brain damage: an experimental study. JAMA, 204:
pathogenesis? J. Pediatr., 143: 494–499. 285–289.
Gilles, E.E. and Nelson Jr., M.D. (1998) Cerebral complica- Ommaya, A.K. and Yarnell, P. (1962) Subdural hematoma af-
tions of nonaccidental head injury in childhood. Pediatr. ter whiplash injury. Lancet, 2: 237–239.
Neurol., 19: 119–128. Painter, M.J. (1995) Animal models of perinatal asphyxia:
Graupman, P. and Winston, K.R. (2006) Nonaccidental head contributions, contradictions, and clinical relevance. Semin.
trauma as a cause of childhood death. J. Neurosurg. (4 Suppl. Pediatr. Neurol., 2: 37–56.
Pediatr.), 104: 245–250. Raghupathi, R. and Margulies, S.S. (2002) Traumatic axonal
Guthkelch, A.K. (1971) Infantile subdural hematoma and its injury after closed head injury in the neonatal pig. J. Neuro-
relationship to whiplash injuries. Br. Med. J., 2: 430–431. trauma, 19: 843–853.
Hadley, M.N., Sonntag, V.K.H., Rekate, H.L. and Murphy, A. Raghupathi, R., Mehr, M.F., Helfaer, M.A. and Margulies,
(1989) The infant whiplash-shake syndrome: a clinical and S.S. (2004) Traumatic axonal injury is exacerbated following
pathological study. Neurosurgery, 24: 536–540. repetitive closed head injury in the neonatal pig. J. Neuro-
Hahn, Y.S., Raimondi, A.J., Mclone, D.G. and Yamanouchi, trauma, 21: 307–316.
Y. (1983) Traumatic mechanisms of head injury in child Rorke, L.B. (1992) Anatomical features of the developing brain
abuse. Childs Brain, 10: 229–241. implicated in pathogenesis of hypoxic-ischemic injury. Brain
Ichord, R.N., Naim, M., Pollock, A.N., Nance, M.L., Margu- Pathol., 2: 211–221.
lies, S.S. and Christian, C.W. (2007) Hypoxic-ischemic injury Ruppell, R.A., Kochanek, P.M., Adelson, P.D., Rose, M.E.,
complicates inflicted and accidental traumatic brain injury in Wisniewski, S.R., Bell, M.J., Clark, R.S., Marion, D.W. and
young children: the role of diffusion-weighted imaging. J. Graham, S.H. (2001) Excitatory amino acid concentrations
Neurotrauma, 24(1): 106–118. in ventricular cerebrospinal fluid after severe traumatic brain
Inglis, F., Kuroda, Y. and Bullock, R. (1992) Glucose hyper- injury in infants and children: the role of child abuse. J.
metabolism after acute subdural hematoma is ameliorated by Pediatr., 138: 18–25.
a competitive NMDA antagonist. J. Neurotrauma, 9: 75–84. Shaver, E., Duhaime, A.C., Curtis, M., Gennarelli, L.M. and
Inglis, F.M., Bullock, R., Chen, D.I., Graham, D.I., Miller, Barrett, R. (1996) Experimental acute subdural hematoma in
J.D. and Mcculloch, J. (1990) Ischaemic brain damage asso- infant piglets. Pediatr. Neurosurg., 25: 123–129.
ciated with tissue hypermetabolism in acute subdural haem- Smith, S.L., Andrus, P.K., Gleason, D.D. and Hall, E.D. (1998)
atoma: reduction by a glutamate antagonist. Acta Neurochir. Infant rat model of the shaken baby syndrome: preliminary
Suppl., 51: 277–279. characterization and evidence for the role of free radicals in
Johnson, D.L., Boal, D. and Baule, R. (1995) Role of apnea in cortical hemorrhaging and progressive neuronal degenera-
nonaccidental head injury. J. Ped. Neurosurg., 23: 305–310. tion. J. Neurotrauma, 15: 693–705.
Johnston, M.V., Trescher, W.H., Ishida, A. and Nakajima, W. Taylor, A., Butt, W., Rosenfeld, J., Shann, F., Ditchfield, M.,
(2001) Neurobiology of hypoxic-ischemic injury in the de- Lewis, E., Klug, G., Wallace, D., Henning, R. and Tibbals,
veloping brain. Pediatr. Res., 49: 735–741. J. (2001) A randomized trial of very early decompressive
Kemp, A.M., Stoodley, N., Cobley, C., Coles, L. and Kemp, craniectomy in children with traumatic brain injury and
K.W. (2003) Apnoea and brain swelling in non-accidental sustained intracranial hypertension. Child Nerv. Syst., 17:
head injury. Arch. Dis. Child., 88: 472–476. 154–162.
Kuroda, Y. and Bullock, R. (1992) Local cerebral blood flow Vanucci, S.J. and Hagberg, H. (2004) Hypoxia-ischemia in the
mapping before and after removal of acute subdural hema- immature brain. J. Exp. Biol., 207(Pt 18): 3149–3154.
toma in the rat. Neurosurgery, 30: 687–691. Whyte, K.M. and Pascoe, M. (1989) Does ‘‘black’’ brain
Ludwig, S. and Warman, M. (1984) Shaken baby syndrome: a mean doom? Computed tomography in the prediction
review of 20 cases. Ann. Emerg. Med., 13: 104–107. of outcome in children with severe head injuries: ‘‘benign’’
Miller, J.D., Bullock, R., Graham, D.I., Chen, M.H. and Teas- vs. ‘‘malignant’’ brain swelling. Australas. Radiol., 33:
dale, G.M. (1990) Ischemic brain damage in a model of acute 344–347.
subdural hematoma. Neurosurgery, 27: 433–439. Zimmerman, R.A., Bilaniuk, L.T., Bruce, D., Schut, L., Uzzell,
Ommaya, A.K., Corrao, P. and Letcher, F.S. (1973) Head in- B. and Goldberg, H.I. (1979) Computed tomography of cra-
jury in the chimpanzee. Part 1: biodynamics of traumatic niocerebral injury in the abused child. Radiology, 130:
unconsciousness. J. Neurosurg., 39: 152–166. 687–690.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 21

Traumatic brain injury and Alzheimer’s disease:


a review

Corinna Van Den Heuvel1,, Emma Thornton1 and Robert Vink1,2

1
Discipline of Pathology, University of Adelaide, Adelaide, Australia
2
Centre for Neurological Diseases, The Hanson Institute, Adelaide, Australia

Abstract: In an effort to identify the factors that are involved in the pathogenesis of Alzheimer’s disease
(AD), epidemiological studies have featured prominently in contemporary research. Of those epidemio-
logical factors, accumulating evidence implicates traumatic brain injury (TBI) as a possible predisposing
factor in AD development. Exactly how TBI triggers the neurodegenerative cascade of events in AD
remains controversial. There has been extensive research directed towards understanding the potential
relationship between TBI and AD and the putative influence that apolipoprotein E (APOE) genotype has
on this relationship. The aim of the current paper is to provide a critical summary of the experimental and
human studies regarding the association between TBI, AD and APOE genotype. It will be shown that
despite significant discrepancies in the literature, there still appears to be an increasing trend to support the
hypothesis that TBI is a potential risk factor for AD. Furthermore, although it is known that APOE
genotype plays an important role in AD, its link to a deleterious outcome following TBI remains incon-
clusive and ambiguous.

Keywords: amyloid precursor protein; traumatic brain injury; apolipoprotein E; Alzheimer’s disease

Epidemiology of TBI and AD (AD) is the most common neurodegenerative dis-


order of modern societies accounting for 50–60%
Traumatic brain injury (TBI) is the leading cause of all age-related dementia (Andersen et al., 2006).
of death and disability in people under the age of Specifically, AD afflicts 8–10% of the population
45 years in industrialized countries. Studies in over the age of 65 and almost 50% over the age of
Australia, the United States, France and Spain 85 (Mattson, 1997). Over 24 million people world-
indicate that the incidence of death from TBI is wide are estimated to be currently suffering from
20–30 per 100,000 (Finfer and Cohen, 2001) with AD, and with the projected rise in the elderly
motor vehicle accidents accounting for the major- population over the coming decades, this has been
ity of fatal head injuries (Kraus, 1993). Those in- estimated to rise to 81 million by 2040 (Miller
dividuals who survive TBI are often left with et al., 2006). The possibility that TBI may predis-
permanent neurological deficits, which adversely pose a person to developing AD in later life has
affect their quality of life, and contributes to the significant social and medical implications, and
enormous social and economic costs of TBI that reinforces the need for preventative efforts and
are borne by communities. Alzheimer’s disease health service planning to cope with the potential
large increase in the number of AD patients (Lye
Corresponding author. Tel.: +61 8 8222 3370; Fax: +61 8 and Shores, 2000). It is therefore critical to estab-
8222 3392; E-mail: corinna.vandenheuvel@adelaide.edu.au lish whether any link between TBI and AD does

DOI: 10.1016/S0079-6123(06)61021-2 303


304

exist, and whether the mechanisms associated in all of these cell malfunctions (Mattson, 2004).
with this link can be exploited to develop an Moreover, Ab causes apoptotic cell death of neu-
interventional therapy. ronal cells in culture by the induction of caspases,
known instigators of apoptotic cell death (Awasthi
et al., 2005). Thus, when Ab is injected into the
The ‘Amyloid Cascade Hypothesis’ hippocampal region of rats, an almost complete
loss of CA1 neurones is observed (Miguel-Hidalgo
Neuropathologically, AD is characterized by the and Cacabelos, 1998).
presence of neurofibrillary tangles (NFTs) and
neuritic plaques. NFTs in the brain consist prima-
rily of hyperphosphorylated tau. Neuritic amyloid APP processing
plaques consist primarily of aggregated amyloid-b
peptides (Ab), a peptide of 40–42 amino acids, Neurotoxic Ab peptides are derived from enzymatic
which are surrounded by dystrophic neurites, mi- processing of the ubiquitously expressed, highly
croglia and reactive astrocytes (Selkoe, 2001). It is conserved type-1 transmembrane glycoprotein
now generally accepted that the development of called APP. Understanding the normal structure,
these two pathologies is central to the pathogenesis molecular function and processing of APP is there-
of AD and the leading ‘amyloid cascade hypoth- fore critical to unravelling the molecular basis of
esis’ suggests that it is the accumulation of Ab in AD. APP is found in all neurons and some glial cells
the brain which is the primary influence in AD in the central and peripheral nervous systems as well
(Hardy and Selkoe, 2002). Ab has been hypothe- as being expressed in platelets, endothelial cells and
sized to cause the pathologic and behavioural fibroblasts. In the brain, where it is constitutively
manifestations of AD, including neurofibrillary expressed, APP serves a synaptic function and is
tangle formation, neuronal degeneration, synaptic located predominantly in neuronal cell bodies, con-
dysfunction and loss as well as impaired memory centrated at axosomatic and other synaptic sites
(Janus et al., 2000). According to this theory, the (Beyreuther et al., 1993). After translation of APP,
accompanying NFT formation and neuronal loss which occurs within the endoplasmic reticulum, the
are downstream events resulting from an imbal- protein is transported through the secretory path-
ance between Ab production from its precursor way becoming N- and O-glycosylated and tyrosyl-
Amyloid Precursor Protein (APP). Evidence in sulphated while moving through the trans-Golgi
support of the hypothesis comes from the know- network (Tomita et al., 1998). Once mature, APP
ledge that genetic defects such as mutations in the can be processed by two mutually exclusive complex
APP gene, and the presenilin 1 and 2 genes, results pathways, either the non-amyloidogenic or the amy-
in aberrant amyloidogenic processing of APP loidogenic pathway (Fig. 1). The non-amyloidogenic
leading to Ab deposition and rare autosomal pathway accounts for the majority of APP process-
dominant forms of familial AD (Selkoe, 1997). ing (Suh and Checler, 2002), and results in a secreted
Also, active immunization against Ab is associated a form of APP (sAPPa) via a-secretase cleavage.
with reduced numbers of amyloid deposits in The b- and g-secretase pathway is responsible for
human AD brains (Hardy and Selkoe, 2002), producing secreted APPb (sAPPb) and the toxic Ab
while passive immunization with antibodies which is found within amyloid plaques in AD (Mills
against Ab results in neutralization of the patho- and Reiner, 1999). Thus, APP can result in both a
logic Ab resulting in rapid improvement in spatial beneficial or detrimental product depending on the
learning and memory, and restoration of blood- way in which it is post-translationally processed
brain barrier integrity in transgenic mice (Lee within cells.
et al., 2006). Degenerating neurones in AD display The secretases which play a crucial role in APP
impaired energy production, increased oxidative processing and cleavage have now been well chara-
damage and disruption of calcium homeostasis, cterised. The a-secretase includes members of the A
and importantly Ab has been shown to be involved disintegrin and metalloprotease (ADAM) family of
305

Fig. 1. Schematically illustrating the cleavage pathways available to amyloid precursor protein and the mutually exclusive nature of
the a- and b-secretase pathways.

proteases (Lammich et al., 1999), b-secretase is a but it also gives rise to sAPPa, which has been re-
type-1 membrane-spanning aspartic protease termed ported to have many neuroprotective and neurotro-
b-site APP-cleavage enzyme 1 (BACE1) (Vassar phic functions within the central nervous system (as
et al., 1999), and g-secretase comprises a complex of reviewed by Mattson et al. (1993)). The discovery
at least four proteins — presenilin 1 or 2, anterior that sAPPa has many beneficial functions, and the
pharynx defective homologue 1 (APH1), presenilin knowledge that production of sAPPa precludes the
enhancer 2 homologue (PEN2) and nicastrin formation of the toxic Ab, has led to an increased
(reviewed by Wilquet and De Strooper (2004)). awareness of the factors that govern its production.
Secretases are obvious targets for drug development These factors are the focus of many investigations
for the prevention and treatment of AD. Therapies that attempt to modulate APP processing toward
either aim to increase a-secretase activity, thereby production of sAPPa, thus slowing or inhibiting Ab
increasing non-amyloidogenic cleavage of APP, or production (Racchi et al., 1999). Stimulation of
inhibit the b- and g-secretase (Dewachter and Van glutamatergic G-protein-coupled receptors linked
Leuven, 2002; Arbel et al., 2005). g-Secretase inhib- to the phospholipase C/protein kinase C (PKC)
itors have also been produced (Dewachter and Van signalling system regulate and increase the release
Leuven, 2002) but these proteases have a wide- of sAPPa (Lee et al., 1997; Lee and Wurtman,
spread tissue distribution and their use in humans 2000). Recently it has been shown that neuronal
may be compromised by side effects resulting from adaptor proteins X 11a and X 11b interact with
blockade of g-secretase cleavage of Notch and other APP processing, possibly by modulating BACE
protein substrates (King et al., 2004). In contrast, cleavage of APP, and resulting in inhibition of
BACE inhibitors may prove beneficial in reducing Ab production (Miller et al., 2006). Furthermore,
Ab production without major side effects. Indeed SorLA/LR11, which is a sorting receptor, regulates
BACE deficient mice have reduced Ab production the intracellular transport and processing of APP in
and do not exhibit any overt abnormal phenotypes neurons thus inhibiting Ab production (Andersen
(Roberds et al., 2001). To date, drugs that target et al., 2006).
specific sites of this Ab cascade have only been
employed in cell culture and animal models of AD,
such that their potential in the clinic remains APP and Ab within damaged axons
unclear.
As indicated above, a-secretase processing not It is feasible that a relationship may exist between
only precludes the formation of Ab (Mattson, 1997) TBI and AD since TBI leads to overexpression of
306

APP within neuronal cell bodies (McKenzie et al., of Ab and subsequent AD. Similarly, excito-
1994; Bramlett et al., 1997; Van Den Heuvel et al., toxic neuronal injury and global ischaemia result
1999) and accumulation of APP within traumat- in accumulation and colocalization of activated
ically injured axons (Blumbergs et al., 1995). caspase-3, caspase-cleaved APP fragments and
Theoretically, the accumulation of APP results in apoptotic (TUNEL) labelling in affected hippo-
an increase in the substrate that could potentially campal neurons and plaque-associated neurites in
be processed to form amyloidogenic Ab. It has AD brains (Gervais et al., 1999). Therefore, the
therefore been hypothesized by a number of observations in TBI that excitotoxic neuronal
groups that the overexpression of APP may injury and global ischaemia resemble changes seen
exceed the limit of normal processing capacity, in dying neurons and plaque-associated neurites in
resulting in mismetabolization of APP into poten- AD brains (Masliah et al., 1998), supports the
tially amyloidogenic fragments (Graham et al., hypothesis that TBI induced caspase activation
1995, 1996). In support of this hypothesis, both may potentially result in increased APP proteolysis
axonal APP accumulation and long-term accumu- and may directly or indirectly influence Ab levels.
lation of Ab has been reported in axons following Evidence for the role of caspase-3 in APP cleavage
TBI (Iwata et al., 2002) suggesting that damaged and Ab production has come from recent studies
axons may provide a key source of Ab following examining the effects of caspase inhibition follow-
TBI (Uryu et al., 2004). Co-accumulation of Ab ing TBI in mice with the human Ab coding
with APP in swollen axons and neuronal cell sequence ‘knocked in’ to their endogenous APP
bodies has been found within days following gene. Caspase inhibitors prevented the elevated level
trauma in a pig model of diffuse axonal injury of Ab normally seen in these animals as well as re-
(Smith et al., 1999), and the same group also found ducing hippocampal cell death, although attenua-
widespread axonal Ab accumulation associated tion of cell death could not be completely attributed
with Ab plaques in brain-injured humans with to less Ab formation (Abrahamson et al., 2006).
diffuse axonal injury (Smith et al., 2003). It has Further human autopsy and experimental stud-
also been demonstrated that BACE and presenilin-1 ies are required to confirm these findings and to
co-localize with APP and Ab within the damaged ascertain the relevance of this co-accumulation
axons and that BACE and presenilin 1 mediate the within neuronal cell bodies and axons and to
Ab production from APP within the axonal better elucidate the exact mechanisms involved in
membrane (Kamal et al., 2001). This was later APP cleavage and subsequent deposition of Ab
confirmed by Chen et al. (2004) in a pig model who plaques.
demonstrated that the Ab accumulation in the
swollen axons persisted for 6 months following
injury in the subcortical white matter and basal Alzheimer’s pathology following fatal head injury in
ganglia. They concluded that TBI leading to humans
impaired axonal transport induces a long-term
pathological co-accumulation of APP with BACE, Preliminary evidence implicating a possible role
presenilin 1 and activated cysteine-dependent for TBI in the development of AD came from an
aspartate-specific proteases (caspases), thereby early case report documenting early-onset classic
providing a possible mechanism for APP cleavage AD pathology in a 38-year-old man who had
and production of Ab within axons following TBI. suffered a previous single episode of severe head
Following neuronal cell injury, activation of trauma 16 years ago (Rudelli et al., 1982). Since
caspase-3 (Raghupathi et al., 2000) occurs in par- then, studies of the brains of boxers suffering from
allel with increased APP accumulation and Ab dementia pugilistica have also demonstrated
production in several different experimental para- AD-like pathology with diffuse Ab plaque depo-
digms (Stone et al., 2002), suggesting a potential sition (Roberts et al., 1990). We can speculate that
association between caspase cleavage of APP, such Ab deposition resulted from repeated blows
caspase-mediated cell death, increased production to the head over a long period of time, and that
307

such events may also occur in the brains of head- Olsson et al. (2004) who reported elevated levels of
injured individuals. Histopathological studies of Ab1–42 and APP in ventricular CSF of severely
such individuals who died after suffering a single injured TBI patients. These groups postulate that
severe TBI demonstrate widespread cerebral Ab the increased levels of Ab1–42 in the CSF are
deposition in short (Roberts et al., 1991, 1994; directly related to the elevated Ab levels in the
Gentleman et al., 1997; Ikonomovic et al., 2004) brain (Raby et al., 1998). It has also been specu-
and long term (Clinton et al., 1991) survivors ir- lated that the rise in Ab1–42 in both brain and
respective of age. In contrast to these studies, CSF may be a direct result of the increased APP
Adle-Biassette et al. (1996) were unable to detect levels in neuronal cell bodies and axons after TBI
any Ab deposits in head injury cases of individuals (McKenzie et al., 1996). In direct contrast, a recent
below the age of 63 years, in a similar age range study of 29 TBI patients found a reduction in
and survival time to those studied by Roberts et al. Ab1–42 levels in CSF, which appeared to correlate
(1991). Also, the extensive non-selective autopsy with poor outcome after TBI (Franz et al., 2003).
investigation by Braak and Braak (1997) rarely Future studies are required to validate these CSF
detected Ab plaques in the younger subjects (be- findings and to resolve the contradictions. It is
low the age of 40 years). None of the 58 cases important to determine how relevant the CSF
examined in the 31–35 years age range, and only concentration of Ab1–42 is as a marker of poten-
two of the 83 subjects in the 36–40 years age range tial AD, and whether monitoring CSF levels might
had some Ab plaques. Furthermore, Ab deposits prove helpful in assessing the degree of neuronal
were detected in only 20 of the 207 people in their injury resulting from TBI. There is also the
fifth decade of life (Braak and Braak, 1997). potential of identifying those individuals poten-
Therefore, significant discrepancies exist in neuro- tially at risk of developing AD later in life (Raby
pathological studies. et al., 1998).
There also appears to be several morphological
similarities, as well as differences, between the Ab
plaques in AD and those found deposited in the Alzheimer’s pathology following experimental head
brain following TBI. In AD, there are numerous, injury
compact and hard core Ab deposits as well as
neurofibrillary tangles and neuropil threads, Studies using different forms of experimental TBI
whereas following TBI there appears to be a have given some insight into how brain injury may
higher incidence of diffuse Ab plaques (Clinton lead to AD, although results remain somewhat in-
et al., 1991; Roberts et al., 1991, 1994; Ikonomovic conclusive. In support of the argument that in-
et al., 2004). Exactly what this morphological creased APP levels following TBI may potentiate
difference in plaques means is unknown. One sim- AD pathology, experimental TBI in rats induced
ilarity between the deposits seen in TBI and AD is overexpression and accumulation of APP in the
that they are both composed primarily of Ab1–42. cerebral cortex and hippocampus, which subse-
These deposits appear much more abundantly quently led to neuronal degeneration in the CA3
than Ab1–40 deposits seen in TBI (Gentleman region of the hippocampus as early as 3 days post-
et al., 1997; Horsburgh et al., 2000a; Ikonomovic injury (Murakami et al., 1998). Also, a pig model
et al., 2004) or in AD (Iwatsubo et al., 1994). of rotational head injury developed rapid Ab
Another consequence of APP gene mutations is accumulation, manifested in axonal bulbs and
elevated Ab levels in the cerebrospinal fluid (CSF) diffuse plaques 3 days after trauma (Smith et al.,
in AD (Nakamura et al., 1994). Studies have since 1999). However, post-traumatic Ab deposition
aimed to determine if CSF levels of Ab are simi- has not been observed in the majority of non-
larly increased following TBI. Raby et al. (1998) transgenic animal studies where most failed
reported significantly elevated levels of Ab1–42 in to identify any Ab staining (Pierce et al., 1996,
the CSF in six patients (age 19–51 years) following 1998; Masumura et al., 2000; Laurer et al., 2001;
severe TBI, an observation later confirmed by Ciallella et al., 2002; Hamberger et al., 2003).
308

Experimental results from APP transgenic mice demonstrated in any of the described transgenic
also remain inconclusive. Transgenic mice with models, unlike human studies (Roberts et al., 1991,
AD-like pathology (platelet-derived-APP; PD-APP), 1994; Gentleman et al., 1997; Ikonomovic et al.,
which overexpress human APP by 10-fold and 2004). Secondly, an increased severity of injury
form amyloid plaques by 6 months of age, enable does not result in increased Ab deposition; if
the interaction between TBI and AD to be studied. anything it actually seems to correlate with re-
TBI in these animals resulted in a transient rise in duced Ab deposition or even resolution of already
Ab levels, particularly in the hippocampus. This established plaques, as reviewed by Szczygielski
elevation in Ab correlated with a much greater loss et al. (2005). This trend seems to be reversed in
of CA3 hippocampal neurones than injured wild humans, since both the risk (Plassman et al., 2000)
type mice, which also had a significant loss of and the post-traumatic Ab deposition increased
hippocampal neurones compared to uninjured with TBI severity (Roberts et al., 1994).
controls (Smith et al., 1998). However, TBI did
not lead to premature amyloid plaque formation
in these transgenic mice, and on long term exam- Epidemiological studies
ination there was a reported reduction in amyloid
plaques ipsilateral to the injury (Smith et al., 1998; The epidemiological studies that have reported on
Nakagawa et al., 1999). Paradoxically, TBI in- the relationship between TBI and AD are also
duced a regression of previously existing plaques contradictory, and accordingly, the association
in aged PD-APP-transgenic mice (Nakagawa still remains inconclusive. Some reports suggest a
et al., 2000). Thus, elevated Ab levels, which are positive association with AD (French et al., 1985;
expected to be seen in the APP-transgenic mice, Mortimer et al., 1985, 1991; Graves et al., 1990;
resulted in hippocampal neuronal death but the Van Duijn et al., 1992; Rasmusson et al., 1995;
TBI did not accelerate Ab deposition. Interest- Salib and Hillier, 1997) while other studies were
ingly, in transgenic mice that overexpress human unable to confirm head trauma as a risk factor
APP by only twofold, Ab deposition did not occur for AD (Katzman et al., 1989; Li et al., 1992;
following TBI and similar cognitive and motor Fratiglioni et al., 1993). With respect to the case-
deficits to wild-type mice were observed (Murai control studies, one of the earliest showed that
et al., 1998). In contrast, repetitive TBI in a patients with dementia of the Alzheimer type had a
Tg2576 APP-transgenic mice model did result in significantly greater incidence of antecedent head
greater Ab deposition and an increase in the pro- trauma (French et al., 1985). Subsequent pooled
duction of both soluble and insoluble cortical re-analysis of some of the case-control studies
Ab40 and Ab42, which may be accounted for by has shown positive associations between the de-
the higher levels of oxidative stress in repetitive velopment of AD and a history of head trauma,
TBI (Uryu et al., 2002). Normally, repetitive TBI particularly amongst males (Mortimer et al., 1991;
in animals does not lead to the formation of ne- Fleminger et al., 2003). Specifically, the meta-
urofibrillary tangles. Yet in a study by Yoshiyama analysis of seven case-control studies by Mortimer
et al. (2005), repetitive TBI in one of 12 mice et al. (1991) allowed for a more powerful statistical
resulted in the development of neurofibrillary investigation of the association between TBI and
tangles, which correlated with a remarkably poor AD and provided the first convincing evidence in
neurobehavioural score. Consequently the pres- support of a strong association between these two
ence of iron deposits, which also occurs in AD, pathologies. They achieved a high level of statis-
was thought to contribute to the formation of tical power (0.92) as opposed to the low mean
these neurofibrillary tangles (Yoshiyama et al., statistical power of the individual studies (0.22).
2005). The findings of the EURODERM re-analysis
The results of TBI studies in transgenic models highlighted the importance of designing studies
of AD are also in conflict with that seen in human with adequate statistical power and suggested that
TBI. Firstly, rapid Ab deposition has not been many of the early negative findings stem from the
309

fact that they suffered from insufficient statistical brain-injured veterans and 1228 age-matched non-
power (see review by Lye and Shores (2000)). This injured controls, follow up after 50 years revealed
is particularly important when the risk factor that moderate to severe head injuries in young men
under investigation, in these cases TBI, occurs in- may be associated with increased risk of AD and
frequently in the general population. Recent case- other dementias in late life. These findings were
control studies have refined their designs to avoid supported by the MIRAGE study, which is the
the methodological flaws found in earlier studies largest study to date (Guo et al., 2000). This study
and have shown significant associations between analysed 2233 definite and probable AD patients,
AD and TBI (Graves et al., 1990; Van Duijn et al., and 14,668 first-degree relatives and showed that
1992; Rasmusson et al., 1995; Salib and Hillier, head injury with loss of consciousness significantly
1997). An additional strength of the study by increased the AD risk. The magnitude of risk was
Rasmusson et al. (1995) was that it examined the proportional to injury severity and was heightened
association between AD and TBI of any severity, amongst first-degree relative of AD patients (Guo
concluding that even mild head injury may serve et al., 2000).
as a predisposing factor for some cases of AD. Several studies have shown that there is a
More recently, a detailed systematic review of greater risk of developing AD when head injury
case-control studies conducted over the past 10 has occurred in later life (within 10 years of onset
years sought to replicate those findings, and sup- of AD) as opposed to head injury occurring earlier
ported an association between head injury and in life (beyond 10 years of onset of AD) (Graves
AD, but only in males (Fleminger et al., 2003). The et al., 1990; Mortimer et al., 1991; Van Duijn et al.,
possible explanation for the gender differences in 1992). In contrast, other studies have shown that
the risk of AD following TBI may be attributed to head trauma occurring mainly in younger child-
the neuroprotective and neuroregenerative effects hood is associated with an increased risk of AD in
of the female hormones oestrogen and progester- later life (Schofield et al., 1997; Plassman et al.,
one (Stein, 2001), or it could be merely the fact 2000). After examining the incidence of AD pa-
that men typically suffer more severe injuries than thology in 58 consecutive patients with residual
women. The association between TBI and early closed TBI lesions and the frequency of TBI
onset of dementia was also demonstrated in a residuals in 57 age-matched autopsy controls,
recent population-based study of fall-related TBI, Jellinger et al. (2001) concluded that severe TBI
concluding that fall-related TBI may hasten, may have some influence on the development of
increase or even trigger the onset of AD (Luukinen AD irrespective of age when the TBI occurred. For
et al., 2005). an excellent detailed review of case-control studies
As with the case-control studies, there have been from 1984 to 1997 see Lye and Shores (2000).
conflicting reports in cohort studies. For example, A related hypothesis concerns the relationship
no significant association was found between head between TBI and AD focussing on the time delay
injury and the risk of developing AD in the early between TBI and the onset of AD. These studies
work (Katzman et al., 1989; Williams et al., 1991). have highlighted the importance of considering
Also, more recently a history of head trauma with more specific factors such as duration of loss of
unconsciousness was excluded as a risk factor for consciousness, severity of injury and the time of
AD in both the Rotterdam Study (Mehta et al., onset of AD symptoms. Cohort studies have sug-
1999) and the large EURODERM study (Launer gested that TBI may interact with other risk fac-
et al., 1999). The EURODERM study was a tors to hasten the onset of AD in persons
pooled analysis of four European population- susceptible to the disease (Sullivan et al., 1987;
based studies of individuals 65 years and older. Gedye et al., 1989; Schofield et al., 1997; Nemetz
Plassman et al. (2000) examined the association et al., 1999). Of 17 AD cases reported, there was a
between early adult head injury, as documented younger mean age of onset for cases with a history
by military hospital records, and dementia in late of head injury than for those cases without it
life. In the study of 548 world war II (US Navy) (Sullivan et al., 1987). In a larger study of 148
310

referral patients with confirmed probable AD, CNS (Nicoll, 1996). The e4 allele has been linked
Gedye et al. (1989) found that the mean age of AD to impaired branching and growth of neurites and
onset was significantly younger for those with a it is therefore suggested to exacerbate neurological
history of TBI. Also, in a slightly larger commu- disturbances (Kerr and Kraus, 1998). Numerous
nity based longitudinal study of aging in 271 par- studies have since confirmed this association by
ticipants in North Manhattan, participants who demonstrating that Ab deposition following head
suffered loss of consciousness exceeding 5 min fol- injury occurs more prominently in those who pos-
lowing a head trauma were at a significantly in- sess an APOE e4 allele. Also, clinical studies of
creased risk of earlier onset AD (Schofield et al., TBI have shown that possession of APOE e4 is
1997). Furthermore, among 1283 TBI cases in associated with a relatively poor outcome (Sorbi
Olmsted County, 31 patients developed AD. Given et al., 1995; Teasdale et al., 1997). In the study by
the incidence of AD in the community, this Teasdale et al. (1997), 57% of APOE e4 carriers
number was not unexpected, however the time be- had an unfavourable outcome (defined as dead,
tween TBI and onset of AD was less than expected vegetative state or severe disability) compared with
suggesting that TBI reduces the time of onset 27% of non-carriers of APOE e4. The relationship
of AD among persons at risk of developing AD between functional recovery in patients with head
(Nemetz et al., 1999). In contrast, Rasmusson injury and APOE e4 genotype was later confirmed
et al. (1995) did not find any effect of head injury in an independent study (Lichtman et al., 2000),
on age of onset of AD in a cohort of 68 AD cases, with rehabilitation outcome for TBI survivors be-
nor did the MIRAGE study (Guo et al., 2000). ing adversely affected in the presence of the APOE
allele. It was subsequently shown that APOEe4
individuals were 10 times more likely to develop
The influence of APOE genotype on TBI outcome AD after TBI than those who did not posses the
allele (Mayeux et al., 1995). A recent case study
Following reports that Ab is not found in all head- has demonstrated that possession of APOE e4 is
injured brains, it was speculated that it may only associated with a greater incidence of moderate/
be deposited in the brains of people who are ge- severe contusional injury and severe ischaemic
netically susceptible to developing AD, for exam- damage in fatal cases of TBI (Smith et al., 2006).
ple, those individuals possessing an apolipoprotein These authors postulate that APOE has a role in
E4 (APOE e4) allele. In 1993, it was demonstrated cerebrovascular and haematological mechanisms.
that the APOE e4 allele was a risk factor for AD Animal studies, using APOE knockout and
(Strittmatter et al., 1993) and the association bet- transgenic mice, have also provided further infor-
ween APOE e4 with late onset familial and spo- mation about APOE mechanisms and the response
radic AD has now been confirmed worldwide of the brain to injury (as reviewed by Horsburgh
(Corder et al., 1993; Saunders et al., 1993; Mayeux et al. (2000b)). Some studies have shown that the
et al., 1995; Nicoll et al., 1995; Horsburgh et al., APOE molecule may have a direct neurotoxic role
2000b). APOE protein has been co-localized with (Neve and Robakis, 1998), whereas others have
the neuropathological hallmarks of AD, including speculated that APOE e4 may directly interact
neurofibrillary tangles, Ab plaques and amyloid with Ab and impact on the metabolism of APP
angiopathy (Namba et al., 1991; Rebeck et al., (Growdon, 1998). Transgenic mice that express
1993). It is now known to be a major susceptibility human APOE e4 have poorer outcome and greater
factor associated with approximately 40–50% of mortality following TBI than mice expressing
sporadic and familial AD compared with 30% of human APOE e3 (Sabo et al., 1999). TBI mice
the normal population (Roses, 1996). expressing APOE e4 also had a decrease in neuro-
While it is known that APOE is a key factor protective sAPPa within the hippocampus,
involved in lipid transport within the human cen- whereas mice expressing APOE e3 had an increase
tral nervous system (CNS), the three alleles (2, 3 in sAPPa in the cortex. These studies concluded
and 4) have significantly varied effects within the that the alteration in APP metabolism contributed
311

to the increased mortality and poorer outcome in into release of Ab from axons, Ab plaque forma-
APOE e4 transgenic mice, and the greater recovery tion and toxicity remains unclear. Despite some
and less cortical damage in APOE e3 transgenic experimental and human studies suggesting that
mice (Ezra et al., 2003). In addition, transgenic APP overexpression following TBI may lead to Ab
APOE e4 mice, which also overexpress APP, had formation, there are numerous studies which have
accelerated deposition of Ab following injury, failed to demonstrate any such relationship.
suggesting APOE e4 may reduce the clearance of Accordingly, further investigation is required,
Ab, thereby favouring its deposition (Hartman particularly with larger prospective cohort studies
et al., 2002). of TBI survivors and larger autopsy series before
In contrast to the studies demonstrating an an association between TBI and onset of AD can
association between APOE e4 and an unfavoura- be definitively supported.
ble outcome following TBI, a number of recent
studies examining both severe and mild TBI have
failed to support such findings (Guo et al., 2000; References
Plassman et al., 2000; Jellinger et al., 2001;
Abrahamson, E.E., Ikonomovic, M.D., Ciallella, J.R., Hope,
Chamelian et al., 2004). Therefore there is no con- C.E., Paljug, W.R., Isanski, B.A., Flood, D.G., Clark, R.S.
clusive evidence linking APOE genotype with the and Dekosky, S.T. (2006) Caspase inhibition therapy abol-
development of AD following TBI (Jellinger, ishes brain trauma-induced increases in Abeta peptide: im-
2004). Further experimental studies and larger plications for clinical outcome. Exp. Neurol., 197: 437–450.
Adle-Biassette, H., Duyckaerts, C., Wasowicz, M., He, Y.,
clinicopathological and prospective human studies
Fornes, P., Foncin, J.F., Lecomte, D. and Hauw, J.J. (1996)
are warranted to clarify the relationship between BAP deposition and head trauma. Neurobiol. Aging, 17:
TBI, APOE genotype and AD. Additional studies 415–419.
with APOE transgenic mice are also expected to Andersen, O.M., Schmidt, V., Spoelgen, R., Gliemann, J.,
contribute to the greater understanding of the Behlke, J., Galatis, D., Mckinstry, W.J., Parker, M.W.,
Masters, C.L., Hyman, B.T., Cappai, R. and Willnow, T.E.
pathophysiological role of APOE in the brain.
(2006) Molecular dissection of the interaction between
amyloid precursor protein and its neuronal trafficking
receptor SorLA/LR11. Biochemistry, 45: 2618–2628.
Conclusion Arbel, M., Yacoby, I. and Solomon, B. (2005) Inhibition of
amyloid precursor protein processing by b-secretase through
site-directed antibodies. PNAS, 102: 7718–7723.
Although findings to date have been contradic-
Awasthi, A., Matsunaga, Y. and Yamada, T. (2005) Amyloid-
tory, there have been major advances from epide- beta causes apoptosis of neuronal cells via caspase cascade,
miological studies and both experimental and which can be prevented by amyloid-beta-derived short
human autopsy studies regarding the relationship peptides. Exp. Neurol., 196: 282–289.
between TBI and AD, and the influence that the Beyreuther, K., Pollwein, P., Multhaup, G., Monning, U.,
Konig, G., Dyrks, T., Schubert, D. and Masters, C.L. (1993)
APOE e4 genotype has on this putative associa-
Regulation and expression of the Alzheimer’s B/A4 amyloid
tion. Human studies have certainly supported the protein precursor in health, disease and Down’s Syndrome.
hypothesis that either repeated mild to moderate Ann. N.Y. Acad. Sci., 695: 91–102.
TBI, or moderate to severe single TBI with loss of Blumbergs, P.C., Scott, G., Manavis, J., Wainwright, H.,
consciousness, are risk factors for the subsequent Simpson, D.A. and Mclean, A.J. (1995) Topography of
axonal injury as defined by amyloid precursor protein and
development of AD. Moreover, those patients who
the sector scoring method in mild and severe closed head
have the APOE e4 are at greater risk of a poorer injury. J. Neurotrauma, 12: 565–572.
outcome following TBI. However, the exact mech- Braak, H. and Braak, E. (1997) Frequency of stages of
anism by which APOE genotype influences out- Alzheimer-related lesions in different age categories.
come and increases risk of AD following TBI is Neurobiol. Aging, 18: 351–357.
Bramlett, H.M., Kraydieh, S., Green, E.J. and Dietrich, W.D.
unclear. Experimental and human studies have
(1997) Temporal and regional patterns of axonal damage
also found accumulation of Ab and APP within following traumatic brain injury: a beta-amyloid precursor
damaged axons following TBI, but the exact im- protein immunocytochemical study in rats. J. Neuropathol.
plications of this and exactly how this translates Exp. Neurol., 56: 1132–1141.
312

Chamelian, L., Reis, M. and Feinstein, A. (2004) Six-month Ruffolo, S.C., Thornberry, N.A., Xanthoudakis, S., Zam-
recovery from mild to moderate traumatic brain injury: the boni, R.J., Roy, S. and Nicholson, D.W. (1999) Involvement
role of APOE-4 allele. Brain, 127: 2621–2628. of caspases in proteolytic cleavage of Alzheimer’s amyloid-
Chen, X.H., Siman, R., Iwata, A., Meaney, D.F., Trojanowski, beta precursor protein and amyloidogenic A beta peptide
J.Q. and Smith, D.H. (2004) Long-term accumulation of formation. Cell, 97: 395–406.
amyloid-beta, beta-secretase, presenilin-1, and caspase-3 in Graham, D.I., Gentleman, S.M., Lynch, A. and Roberts, G.W.
damaged axons following brain trauma. Am. J. Pathol., 165: (1995) Distribution of beta-amyloid protein in the brain
357–371. following severe head injury. Neuropathol. Appl. Neurobiol.,
Ciallella, J.R., Ikonomovic, M.D., Paljug, W.R., Wilbur, Y.I., 21: 27–34.
Dixon, C.E., Kochanek, P.M., Marion, D.W. and Dekosky, Graham, D.I., Gentleman, S.M., Nicoll, J.A., Royston, M.C.,
S.T. (2002) Changes in expression of amyloid precursor McKenzie, J.E., Roberts, G.W. and Griffin, W.S. (1996)
protein and interleukin-1beta after experimental traumatic Altered beta-APP metabolism after head injury and its
brain injury in rats. J. Neurotrauma, 19: 1555–1567. relationship to the aetiology of Alzheimer’s disease. Acta
Clinton, J., Ambler, M.W. and Roberts, G.W. (1991) Post Neurochir. Suppl. (Wien.), 66: 96–102.
traumatic Alzheimer’s disease: preponderance of a single Graves, A.B., White, E., Koepsell, T.D., Reifler, B.V., Van
plaque type. Neuropathol. Appl. Neurobiol., 17: 69–74. Belle, G., Larson, E.B. and Raskind, M. (1990) The associ-
Corder, E.H., Saunders, A.M., Strittmatter, W.J., Schmechal, ation between head trauma and Alzheimer’s disease. Am. J.
D.E., Gaskell, P.C., Small, G.W., Roses, A.D., Haines, J.L. Epidemiol., 131: 491–501.
and Pericak Vance, M.A. (1993) Gene dosage of apolipo- Growdon, J.H. (1998) Apolipoprotein E and Alzheimer’s
protein E type allele and the risk of Alzheimer’s disease in disease. Arch. Neurol., 44: 1053–1054.
late onset families. Science, 261: 921–923. Guo, Z., Cuppels, L.A., Kurz, A., Auerbach, S.H., Volicer, L.,
Dewachter, I. and Van Leuven, F. (2002) Secretases as targets Chui, H., Green, R.C., Sadovnick, A.D., Durara, R.,
for the treatment of Alzheimer’s disease: the prospects. Decarli, C., Johnson, K., Go, R.C., Growdon, J.H., Haines,
Lancet Neurol., 1: 409–416. J.L., Kukull, W.A. and Farrer, L.A. (2000) Head injury
Ezra, Y., Oron, L., Moskovich, L., Roses, A.D., Beni, S.M., and risk of AD in the MIRAGE study. Neurology, 54:
Shohami, E. and Michaelson, D.M. (2003) Apolipoprotein 1316–1323.
E4 decreases whereas apolipoprotein E3 increases the level of Hamberger, A., Huang, Y.L., Zhu, H., Bao, F., Ding, M.,
secreted amyloid precursor protein after closed head injury. Blennow, K., Olsson, A., Hansson, H.A., Viano, D. and
Neuroscience, 121: 315–325. Haglid, K.G. (2003) Redistribution of neurofilaments and
Finfer, S.R. and Cohen, J. (2001) Severe traumatic brain injury. accumulation of beta-amyloid protein after brain injury by
Resuscitation, 48: 77–90. rotational acceleration of the head. J. Neurotrauma, 20:
Fleminger, S., Oliver, D.L., Lovestone, S., Rabe-Hesketh, S. 169–178.
and Giora, A. (2003) Head injury as a risk factor for Hardy, J.A. and Selkoe, D.J. (2002) The amyloid hypothesis of
Alzheimer’s disease: the evidence 10 years on; a partial Alzheimer’s disease; progress and problems on the road to
replication. J. Neurol. Neurosurg. Psychiatry, 74: 857–862. therapeutics. Science, 297: 353–356.
Franz, G., Beer, R., Kampfl, A., Engelhardt, K., Schmutzhard, Hartman, R.E., Laurer, H., Longhi, L., Bales, K.R., Paul,
E., Ulmer, H. and Deisenhammer, F. (2003) Amyloid beta S.M., McIntosh, T.K. and Holtzman, D.M. (2002) Apolipo-
1–42 and tau in cerebrospinal fluid after severe traumatic protein E4 influences amyloid deposition but not cell loss
brain injury. Neurology, 60: 1457–1461. after traumatic brain injury in a mouse model of Alzheimer’s
Fratiglioni, L., Ahlbom, A., Viitanen, M. and Winblad, B. disease. J. Neurosci., 22: 10083–10087.
(1993) Risk factors for late-onset alzheimer’s disease: a pop- Horsburgh, K., Cole, G.M., Yang, F., Savage, M.J.,
ulation-based, case-control study. Ann. Neurol., 33: 258–266. Greenberg, B.D., Gentleman, S.M., Graham, D.I. and
French, L.R., Schuman, L.M., Mortimer, J.A., Hutton, J.T., Nicoll, J.A.R. (2000a) b-Amyloid (Ab)42(43), Ab42, Ab40
Boatman, R.A. and Christians, B. (1985) A case-control and apoE immunostaining of plaques in fatal head injury.
study of dementia of the Alzheimer type. Am. J. Epidemiol., Neuropath. Appl. Neurobiol., 26: 124–132.
121: 414–421. Horsburgh, K., Mccarron, M.O., White, F. and Nicoll, J.A.
Gedye, A., Beattie, B.L., Tuokko, H., Horton, A. and Korsarek, (2000b) The role of apolipoprotein E in Alzheimer’s disease,
E. (1989) Severe head injury hastens age of onset of acute brain injury and cerebrovascular disease: evidence of
Alzheimer’s disease. J. Am. Geriatr. Soc., 37: 970–973. common mechanisms and utility of animal models.
Gentleman, S.M., Greenberg, B.D., Savage, M.J., Noori, M., Neurobiol. Aging, 21: 245–255.
Newman, S.J., Roberts, G.W., Griffin, W.S. and Graham, D.I. Ikonomovic, M.D., Uryu, K., Abrahamson, E.E., Ciallella,
(1997) AB42 is the predominant form of amyloid-b-protein in J.R., Trojanowski, J.Q., Lee, V.M.-Y., Clark, R.S., Marion,
the brains of short-term survivors of head injury. Neuroreport, D.W., Wisniewski, S.R. and Dekosky, S.T. (2004)
8: 1519–1522. Alzheimer’s pathology in human temporal cortex surgically
Gervais, F.G., Xu, D., Robertson, G.S., Vaillancourt, J.P., Zhu, excised after severe brain injury. Exp. Neurol., 190: 192–203.
Y., Huang, J., Leblanc, A., Smith, D., Rigby, M., Shearman, Iwata, A., Chen, X.H., McIntosh, T.K., Browne, K.D. and
M.S., Clarke, E.E., Zheng, H., Van Der Ploeg, L.H., Smith, D.H. (2002) Long-term accumulation of amyloid-beta
313

in axons following brain trauma without persistent upregu- peptide (Ab) oligomers by passive immunization with a con-
lation of amyloid precursor protein genes. J. Neuropathol. formation-selective monoclonal antibody improves learning
Exp. Neurol., 61: 1056–1068. and memory in Ab precurosr protein (APP) transgenic mice.
Iwatsubo, T., Odaka, A., Suzuki, N., Mizusawa, H., Nukina, N. J. Biol. Chem., 281: 4292–4299.
and Ihara, Y. (1994) Visualization of AB42 and AB40 in Lee, J. and Wurtman, R.J. (2000) Regulation of APP synthesis
senile plaques with end specific AB monoclonals: evidence and secretion by neuroimmunophilin ligands and cycloox-
that an initially deposited species is AB42. Neuron, 13: 45–53. ygenase inhibitors. Ann. N.Y. Acad. Sci., 920: 261–268.
Janus, C., Pearson, J., Mclaurin, J., Mathews, P.M., Jiang, Y., Lee, R.K.K., Araki, W. and Wurtman, R.J. (1997) Stimulation
Schmidt, S.D., Chishti, M.A., Horne, P., Heslin, D., French, of amyloid precursor protein synthesis by adrenergic recep-
J., Mount, H.T., Nixon, R.A., Mercken, M., Bergeron, C., tors coupled to cAMP formation. Proc. Natl. Acad. Sci.
Fraser, P.E., St George-Hyslop, P. and Westaway, D. (2000) U.S.A., 94: 5422–5426.
A beta peptide immunization reduces behavioural impair- Li, G., Shen, Y.C., Li, Y.T., Chen, C.H., Zhau, Y.W. and
ment and plaques in a model of Alzheimer’s disease. Nature, Silverman, J.M. (1992) A case-control study of Alzheimer’s
408: 979–982. disease in China. Neurology, 42: 1481–1488.
Jellinger, K.A. (2004) Head injury and dementia. Curr. Opin. Lichtman, S.W., Seliger, G., Tycko, B. and Marder, K. (2000)
Neurol., 17: 719–723. Apolipoprotein E and functional recovery from brain injury
Jellinger, K.A., Paulus, W., Wrocklage, C. and Litvan, I. (2001) following post acute rehabilitation. Neurology, 55: 1536–1539.
Effects of closed traumatic brain injury and genetic factors Luukinen, H., Viramo, P., Herala, M., Kervinen, K.,
on the development of Alzheimer’s disease. Eur. J. Neurol., 8: Kesaniemi, Y.A., Savola, O., Winqvist, S., Jokelainen, J.
707–710. and Hillbom, M. (2005) Fall-related injuries and the risk of
Kamal, A., Almenar-Queralt, A., Leblanc, J.F., Roberts, E.A. dementia in elderly people: a population-based study. Eur. J.
and Goldstein, L.S. (2001) Kinesin-mediated axonal trans- Neurol., 12: 86–92.
port of a membrane compartment containing b-secretase and Lye, T.C. and Shores, A.E. (2000) Traumatic brain injury as a
presenilin-1 requires APP. Nature, 414: 643–648. risk factor for Alzheimer’s disease: a review. Neuropsychol.
Katzman, R., Aronson, M., Fuld, P., Kawas, C., Brown, T., Rev., 10: 115–129.
Morgenstern, H., Frishman, W., Gidez, L., Eder, H. and Masliah, E., Mallory, M., Alford, M. and Hansen, L.A. (1998)
Ooi, W.L. (1989) Development of dementing illness in an Caspase dependent DNA fragmentation might be associated
80-year-old volunteer cohort. Ann. Neurol., 25: 317–324. with excitotoxicity in Alzheimer’s disease. J. Neuropathol.
Kerr, M.E. and Kraus, M. (1998) Genetics and the central Exp. Neurol., 57: 1041–1052.
nervous system: apolipoprotein E and brain injury. AACN Masumura, M., Hata, R., Uramoto, H., Murayama, N.,
Clin. Issues, 9: 524–530. Ohno, T. and Sawada, T. (2000) Altered expression of
King, G.D., Cherian, K. and Turner, S.R. (2004) X11a impairs amyloid precursor proteins after traumatic brain injury in
g- but not b-cleavage of amyloid precursor protein. rats: in situ hybridization and immunochemical study.
J. Neurochem., 88: 971–982. J. Neurotrauma, 17: 123–134.
Kraus, J.F. (1993) Epidemiology of head injury. In: Cooper Mattson, M.P. (1997) Cellular actions of b-amyloid precursor
P.R. (Ed.), Head Injury (3rd ed). Williams and Wilkins, protein and it’s soluble and fibrillogenic derivatives. Physiol.
Baltimore. Rev., 77: 1081–1132.
Lammich, S., Kojro, E., Postina, R., Gilbert, S., Pfeiffer, R., Mattson, M.P. (2004) Pathways towards and away from
Jasionowski, M., Haass, C. and Fahrenholz, F. (1999) Con- Alzheimer’s disease. Nature, 430: 631–639.
stitutive and regulated a-secretase cleavage of Alzheimer’s Mattson, M.P., Cheng, B., Culwell, A.R., Esch, F.S.,
amyloid precursor protein by a disintegrin metalloprotease. Lieberburg, I. and Rydel, R.E. (1993) Evidence for excito-
Proc. Natl. Acad. Sci. U.S.A., 96: 3922–3927. protective and intraneuronal calcium-regulating roles for
Launer, L.J., Andersen, K., Dewey, M.E., Letenneur, L., secreted forms of the beta-amyloid precursor protein.
Ott, A., Amaducci, L.A., Brayne, C., Copeland, J.R., Dart- Neuron, 10: 243–254.
igues, J.F., Kragh-Sorensen, P., Lobo, A., Martinez-Lage, Mayeux, R., Ottman, R., Maestre, G., Ngai, C., Tang, M.X.,
J.M., Stijnen, T. and Hofman, A. (1999) Rates and risk Ginsberg, H., Chun, M., Tycko, B. and Shelanski, M. (1995)
factors for dementia and Alzheimer’s disease: results from Synergistic effects of traumatic head injury and apolipopro-
EURODEM pooled analyses. EURODEM Incidence tein E4 in patients with Alzheimer’s disease. Neurology, 45:
Research Group and Work Groups. European Studies of 555–557.
Dementia. Neurology, 52: 78–84. McKenzie, J.E., Gentleman, S.M., Roberts, G.W., Graham,
Laurer, H.L., Bareyre, F.M., Lee, V.M., Trojanowski, J.Q., D.I. and Royston, M.C. (1994) Increased numbers of beta
Longhi, L., Hoover, R., Saatman, K.E., Raghupathi, R., APP-immunoreactive neurons in the entorhinal cortex after
Hoshino, S., Grady, M.S. and McIntosh, T.K. (2001) Mild head injury. Neuroreport, 6: 161–164.
head injury increasing the brain’s vulnerability to a second McKenzie, K.J., Mclellan, D.R., Gentleman, S.M., Maxwell,
concussive impact. J. Neurosurg., 95: 859–870. W.L., Gennarelli, T.A. and Graham, D.I. (1996) Is X-APP a
Lee, E.B., Leng, L.Z., Zhang, B., Kwong, L., Trojanowski, marker of axonal damage in short-surviving head injury?
J.Q., Abel, T. and Lee, V.M.Y. (2006) Targeting amyloid-b Acta Neuropathol. (Berl.), 92: 608–613.
314

Mehta, K.M., Ott, A., Kalmijn, S., Slooter, A.J., Van Duijn, Nemetz, P.N., Leibson, C., Naessens, J.M., Beard, M.,
C.M., Hofman, A. and Breteler, M. (1999) Head trauma and Kokmen, E., Annegers, J.F. and Kurland, L.T. (1999) Trau-
risk of dementia and Alzheimer’s disease: the Rotterdam matic brain injury and time to onset of Alzheimer’s disease: a
study. Neurology, 53: 1959–1962. population-based study. Am. J. Epidemiol., 149: 32–40.
Miguel-Hidalgo, J.J. and Cacabelos, R. (1998) Beta-amyloid- Neve, R.L. and Robakis, N.K. (1998) Alzheimer’s disease: a
(1–40)-induced neurodegeneration in the rat hippocampal re-examination of the amyloid hypothesis. Trends Neurosci.,
neurons of the CA1 subfield. Acta Neuropathol. (Berl.), 95: 21: 15–19.
455–465. Nicoll, J.A. (1996) Genetics and head injury. Neuropathol.
Miller, C.C., Mcloughlin, D.M., Lau, K.F., Tennant, M.E. and Appl. Neurobiol., 22: 515–517.
Rogelj, B. (2006) The X11 proteins, A beta production and Nicoll, J.A.R., Roberts, G.W. and Graham, D.I. (1995) Apol-
Alzheimer’s disease. Trends Neurosci., 29: 280–285. ipoprotein E e4 allele is associated with deposition of amyloid
Mills, J. and Reiner, P.B. (1999) Regulation of amyloid b-protein following head injury. Nat. Med., 1: 135–137.
precursor protein cleavage. J. Neurochem., 72: 443–460. Olsson, A., Scsajbok, L., Ost, M., Hoglund, K., Nylen, K.,
Mortimer, J.A., French, L.R., Hutton, J.T. and Schuman, L.M. Rosengre, L., Nellgard, B. and Blennow, K. (2004) Marked
(1985) Head injury as a risk factor for Alzheimer’s disease. increase of beta-amyloid (1–42) and amyloid precursor
Neurology, 35: 264–267. protein in ventricular cerebrospinal fluid after severe trau-
Mortimer, J.A., Van Duijn, C.M., Chandra, V., Fratiglioni, L., matic brain injury. J. Neurol., 251: 870–876.
Graves, A.B., Heyman, A., Jorm, A.F., Kokmen, E., Kondo, Pierce, J.E., Smith, D.H., Trojanowski, J.Q. and McIntosh,
K. and Rocca, W.A. (1991) Head trauma as a risk factor T.K. (1998) Enduring cognitive, neurobehavioral and histo-
for Alzheimer’s disease: a collaborative re-analysis of case- pathological changes persist for up to one year following
control studies. EURODEM Risk Factors Research Group. severe experimental brain injury in rats. Neuroscience, 87:
Int. J. Epidemiol., 20(Suppl 2): S28–S35. 359–369.
Murai, H., Pierce, J.E.S., Raghupathi, R., Smith, D.H., Pierce, J.E., Trojanowski, J.Q., Graham, D.I., Smith, D.H. and
Saatman, K.E., Trojanowski, J.Q., Lee, V.M.Y., Loring, McIntosh, T.K. (1996) Immunohistochemical characteriza-
J.F., Eckman, C.B., Younkin, S.G. and McIntosh, T.K. (1998) tion of alterations in the distribution of amyloid precursor
Twofold overexpression of human b-amyloid precursor proteins and beta-amyloid peptide after experimental brain
proteins in transgenic mice does not affect the neuromotor, injury in the rat. J. Neurosci., 16: 1083–1090.
cognitive or neurodegenerative sequelae following experimen- Plassman, B.L., Havlik, R.J., Steffens, D.C., Helms, M.J.,
tal brain injury. J. Comp. Neurol., 392: 428–438. Newman, T.N., Drosdick, D., Phillips, C., Gau, B.A., Welsh-
Murakami, N., Yamaki, T., Iwamoto, Y., Sakakibara, T., Bohmer, K.A., Burke, J.R., Guralnik, J.M. and Breitner,
Kobori, N., Fushiki, S. and Ueda, S. (1998) Experimental J.C.S. (2000) Documented head injury in early adulthood and
brain injury induces expression of amyloid precursor protein, risk of Alzheimer’s disease and other dementias. Neurology,
which may be related to neuronal loss in the hippocampus. 55: 1158–1166.
J. Neurotrauma, 15: 993–1003. Raby, C.A., Morganti-Kossman, M.C., Kossman, T., Stahel,
Nakagawa, Y., Nakamura, M., McIntosh, T.K., Rodriguez, A., P.F., Watson, M.D., Evans, L.M., Mehta, P.D., Spiegel, K.,
Berlin, J.A., Smith, D.H., Saatman, K.E., Raghupathi, R., Kuo, Y., Roher, A.E. and Emmerling, M.R. (1998) Trau-
Clemens, J., Saido, T.C., Schmidt, M.L., Lee, V.M. and matic brain injury increases b-amyloid peptide 1–42 in
Trojanowski, J.Q. (1999) Traumatic brain injury in young, cerebrospinal fluid. J. Neurochem., 71: 2505–2509.
amyloid-beta peptide overexpressing transgenic mice induces Racchi, M., Solano, C.D., Sironi, M. and Govoni, S. (1999)
marked ipsilateral hippocampal atrophy and diminished Activity of a-secretase as the common final effector of protein
Abeta deposition during aging. J. Comp. Neurol., 411: kinase c-dependent and independent modulation of amyloid
390–398. precursor protein metabolism. J. Neurochem., 72: 2464–2470.
Nakagawa, Y., Reed, L., Nakamura, M., McIntosh, T.K., Raghupathi, R., Graham, D.I. and McIntosh, T.K. (2000)
Smith, D.H., Saatman, K.E., Raghupathi, R., Clemens, J., Apoptosis after traumatic brain injury. J. Neurotrauma, 17:
Saido, T.C., Lee, V.M. and Trojanowski, J.Q. (2000) Brain 927–938.
trauma in aged transgenic mice induces regression of Rasmusson, D.X., Brandt, J., Martin, D.B. and Folstein, M.F.
established abeta deposits. Exp. Neurol., 163: 244–252. (1995) Head injury as a risk factor in Alzheimer’s disease.
Nakamura, T., Shoji, M., Harigaya, Y., Watanabe, M., Brain Inj., 9: 213–219.
Hosoda, K., Cheung, T.T., Shaffer, L.M., Golde, T.E., Rebeck, G.W., Reiter, J.S., Strickland, D.K. and Hyman, B.T.
Younkin, L.H. and Younkin, S.G. (1994) Amyloid B levels in (1993) Apolipoprotein E in sporadic Alzheimer’s disease: all-
cerebrospinal fluid are elevated in early-onset Alzheimer’s elic variation and receptor interactions. Neuron, 11: 575–580.
disease. Ann. Neurol., 36: 903–911. Roberds, S.L., Anderson, J., Basi, G., Bienkowski, M.J.,
Namba, Y., Tomonaga, M., Kawasaki, H., Otomo, E. and Branstetter, D.G., Chen, K.S., Freedman, S.B., Frigon,
Ikeda, K. (1991) Apolipoprotein E immunoreactivity in cere- N.L., Games, N., Hu, G., Johnson-Wood, K., Kappenman,
bral amyloid deposits and neurofibrillary tangles in K.E., Kawabe, T.T., Kola, I., Kuehn, R., Lee, M., Liu, W.,
Alzheimer’s disease and kuru plaque amyloid in Creutz- Motter, R., Nichols, N.F., Power, M., Robertson, D.W.,
feldt-Jakob disease. Brain Res., 541: 163–166. Schenk, D., Schoor, M., Shopp, G.M., Shuck, M.E.,
315

Sinha, S., Svensson, K.A., Tatsuno, G., Tintrup, H., Wijs- (1998) Brain trauma induces massive hippocampal neuron
man, J., Wright, S. and Mcconlogue, L. (2001) BACE death linked to a surge in beta-amyloid levels in mice overex-
knockout mice are healthy despite lacking the primary b- pressing mutant amyloid precursor protein. Am. J. Pathol.,
secretase activity in brain: implications for Alzheimer’s dis- 153: 1005–1010.
ease therapeutics. Hum. Mol. Genet., 10: 1317–1324. Sorbi, S., Nacmias, N., Piacentini, S., Repice, A., Latorraca, S.,
Roberts, G.W., Allsop, D. and Bruton, C.J. (1990) The occult Forleo, P. and Amaducci, L. (1995) ApoE as a prognostic
aftermath of boxing. J. Neurol. Neurosurg. Psychiatry, 53: factor for post-traumatic coma. Nat. Med., 1: 852.
373–378. Stein, D.G. (2001) Brain damage, sex hormones and recovery: a
Roberts, G.W., Gentleman, S.M., Lynch, A. and Graham, D.I. new role for progesterone and estrogen? Trends Neurosci.,
(1991) beta A 4 amyloid protein deposition in brain after 24: 386–391.
head trauma. Lancet, 338: 1422–1423. Stone, J.R., Okonkwo, D.O., Singleton, R.H., Mutlu, L.K.,
Roberts, G.W., Gentleman, S.M., Lynch, A., Landon, M. and Helm, G.A. and Povlishock, J.T. (2002) Caspase-3-mediated
Graham, D.I. (1994) Beta amyloid protein deposition in the cleavage of amyloid precursor protein and formation of am-
brain after severe head injury: implications for the patho- yloid b peptide in traumatic axonal injury. J. Neurotrauma,
genesis of Alzheimer’s disease. J. Neurol. Neurosurg. 19: 601–614.
Psychiatry, 57: 419–425. Strittmatter, W.J., Saunders, A.M. and Schmechel, D. (1993)
Roses, A.D. (1996) Apolipoprotein E alleles as risk factors in Apolioprotein E high avidity binding to B-amyloid and
Alzheimer’s disease. Ann. Rev. Med., 47: 387–400. increased frequency of the E4 allele in late onset familial
Rudelli, R., Strom, J.O., Welch, P.T. and Ambler, M.W. (1982) Alzheimer’s disease. Proc. Natl. Acad. Sci. U.S.A., 90:
Post-traumatic premature Alzheimer’s disease. Arch. 1977–1981.
Neurol., 39: 570–575. Suh, Y.H. and Checler, F. (2002) Amyloid precursor protein,
Sabo, S.L., Lanier, L.M., Ikin, A.F., Khorkova, O., presenilins, and a-synuclein: molecular pathogenesis and
Sahasrabudhe, S., Greengard, P. and Buxbaum, J.D. (1999) pharmacological applications in Alzheimer’s disease. Pharm.
Regulation of X-amyloid secretion by FE65, an amyloid Rev., 54: 469–525.
protein precursor-binding protein. J. Biol. Chem., 274: Sullivan, P., Petitti, D. and Barbaccia, J. (1987) Head trauma
7952–7957. and age of onset of dementia of the Alzheimer type (Letter).
Salib, E. and Hillier, V. (1997) Head injury and the risk JAMA, 257: 2289–2290.
of Alzheimer’s disease: a case control study. Int. J. Geriat. Szczygielski, J., Mautes, A., Steudel, W.I., Falkai, P., Bayer, T.A.
Psychiatry, 12: 363–368. and Wirths, O. (2005) Traumatic brain injury: cause or risk
Saunders, A.M., Strittmatter, W.J. and Schmechel, D. (1993) of Alzheimer’s disease? A review of experimental studies.
Association of apolioprotein E allele e4 with late-onset J. Neural. Transm., 112: 1547–1564.
familial and sporadic Alzheimer’s disease. Proc. Natl. Acad. Teasdale, G.M., Nicoll, J.A.R., Murray, G. and Fiddes, M.
Sci. U.S.A., 90: 8098–8102. (1997) Association of apolipoprotein E polymorphism with
Schofield, P.W., Tang, M.X., Marder, K., Bell, K., Dooneief, outcome after head injury. Lancet, 350: 1069–1071.
G., Chun, M., Sano, M., Stern, Y. and Mayeux, R. (1997) Tomita, S., Kirino, Y. and Suzuki, T. (1998) Cleavage of
Alzheimer’s disease after remote head injury: an incidence Alzheimer’s amyloid precursor protein (APP) by secretases
study. J. Neurol. Neurosurg. Psychiatry, 62: 119–124. occurs after O-glycosylation of APP in the protein secretory
Selkoe, D.J. (1997) Alzheimer’s disease: genotypes, phenotypes, pathway. J. Biol. Chem., 273: 6277–6284.
and treatments. Science, 275: 630–631. Uryu, K., Chen, X.H. and Graham, D.J. (2004) Short-term
Selkoe, D.J. (2001) Alzheimer’s disease: genes, proteins, and accumulation of beta-amyloid in axonal pathology following
therapy. Physiol. Rev., 81: 741–766. traumatic brain injury in humans. Neurobiol. Aging, 25(Sup-
Smith, C., Graham, D.I., Murray, L.S., Stewart, J. and Nicoll, pl 1): P2–P250.
J.A.R. (2006) Association of APOe4 and cerebrovascular Uryu, K., Laurer, H., McIntosh, T.K., Pratico, D., Martinez,
pathology in traumatic brain injury. J. Neurol. Neurosurg. D., Leight, S., Lee, V.M.-Y. and Trojanowski, J.Q. (2002)
Psychiatry, 77: 363–366. Repetitive mild brain trauma accelerates Ab deposition, lipid
Smith, D.H., Chen, X.H., Iwata, A. and Graham, D.I. (2003) peroxidation, and cognitive impairment in a transgenic
Amyloid-X accumulation in axons after traumatic brain mouse model of Alzheimer amyloidosis. J. Neurosci., 22:
injury in humans. J. Neurosurg., 98: 1072–1077. 446–454.
Smith, D.H., Chen, X.H., Nonaka, M., Trojanowski, J.Q., Lee, Van Den Heuvel, C., Blumbergs, P.C., Finnie, J.W., Manavis,
V.M., Saatman, K.E., Leoni, M.J., Xu, B.N., Wolf, J.A. and J., Jones, N.R., Reilly, P.L. and Pereira, R.A. (1999) Upreg-
Meaney, D.F. (1999) Accumulation of amyloid beta and tau ulation of amyloid precursor protein messenger RNA in
and the formation of neurofilament inclusions following response to traumatic brain injury: an ovine head impact
diffuse brain injury in the pig. J. Neuropathol. Exp. Neurol., model. Exp. Neurol., 159: 441–450.
58: 982–992. Van Duijn, C.M., Tanja, T.A., Haaxma, R., Schulte, W., Saan,
Smith, D.H., Nakamura, M., McIntosh, T.K., Wang, J., R.J., Lameris, A.J., Antonides-Hendriks, G. and Hofman, A.
Rodriguez, A., Chen, X.H., Raghupathi, R., Saatman, K.E., (1992) Head trauma and the risk of Alzheimer’s disease. Am.
Clemens, J., Schmidt, M.L., Lee, V.M. and Trojanowski, J.Q. J. Epidemiol., 135: 775–782.
316

Vassar, R., Bennett, B.D., Babu-Khan, S., Kahn, S., Mendiaz, Wilquet, V. and De Strooper, B. (2004) Amyloid-X precursor
E.A., Denis, P., Teplow, D.B., Ross, S., Amarante, P., protein processing in neurodegeneration. Curr. Opin.
Loeloff, R., Luo, Y., Fisher, S., Fuller, J., Edenson, S., Neurobiol., 14: 582–588.
Lile, J., Jarosinski, M.A., Biere, A.L., Curran, E., Burgess, T., Yoshiyama, Y., Uryu, K., Higuchi, M., Longhi, L., Hoover, R.,
Louis, J., Collins, F., Treanor, J., Rogers, G. and Citron, M. Fujimoto, S., McIntosh, T., Lee, V.M. and Trojanowski, J.Q.
(1999) X-Secretase cleavage of Alzheimer’s amyloid precursor (2005) Enhanced neurofibrillary tangle formation, cerebral
protein by the transmembrane aspartic protease BACE. atrophy, and cognitive deficits induced by repetitive mild
Science, 286: 735–740. brain injury in a transgenic tauopathy mouse model.
Williams, D.B., Annegers, J.F., Kokmen, E., O’brien, P.C. and J. Neurotrauma, 22: 1134–1141.
Kurland, L. (1991) Brain injury and neurologic sequelae: a
cohort study of dementia, parkinsonism, amyotrophic lateral
sclerosis. Neurology, 41: 1554–1557.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 22

The neurotrophic protein S100B: value as a marker of


brain damage and possible therapeutic implications

Andrea Kleindienst1,, Felicitas Hesse1, M. Ross Bullock2 and Michael Buchfelder1

1
Department of Neurosurgery, Friedrich-Alexander-University, Erlangen-Nuremberg, Germany
2
Department of Neurosurgery, Virginia Commonwealth University Medical Center, Richmond, VA 23298-0631, USA

Abstract: We provide a critical analysis of the value of S100B as a marker of brain damage and possible
therapeutic implications. The early assessment of the injury severity and the consequent prognosis are of
major concern for physicians treating patients suffering from traumatic brain injury (TBI). A reliable
indicator to accurately determine the extent of the brain damage has to meet certain requirements: (i) to
originate in the central nervous system (CNS) with no contribution from extracerebral sources; (ii) a passive
release from damaged neurons and/or glial cells without any stimulated active release; (iii) a lack of specific
effects on neurons and/or glial cells interfering with the initial injury; (iv) an unlimited passage through the
blood-brain barrier (BBB). The measurement of putative biochemical markers, such as the S100B protein,
has been proposed in this role. Over the past decade, numerous studies have reported a positive correlation
of S100B serum levels with a poor outcome following TBI. However, some studies raise doubt whether the
serum measurement of S100B is a valid biochemical marker of brain damage. We summarize the specific
properties of S100B and analyze whether they support or counteract the necessary requirements to desig-
nate this protein as an indicator of brain damage. Finally, we report recent experimental findings suggesting
a possible therapeutic potential of S100B.

Keywords: S100B; TBI; neuroregeneration; brain damage; biomarker; neurotrophic factor

General features and origin of the S100B protein role of S100B while in the clinical setting the
measurement of S100B became more frequently
As early as 1965, a neurotrophic factor was puri- performed.
fied from bovine brain for the first time (Moore, The S100B protein belongs to a multigenic fam-
1965), and in 1978 this factor was demonstrated to ily of low molecular weight (9–13 kDa) calcium-
consist of two distinct proteins, S100b and S100a binding S100 proteins (Donato, 2001; Heizmann
(Isobe et al., 1978). After the identification of the et al., 2002). S100B is most abundant in glial cells
chromosomal localization of S100 proteins in of the central nervous system (CNS), predomi-
1995, the nomenclature was changed from S100b nately in astrocytes (Donato, 1986), and consti-
to S100B and from S100a to S100A1 (Schafer tutes 1–1.5 mg/mg of soluble protein (Matsutani
et al., 1995). In the following decade, experimental et al., 1985). S100 proteins contain no detectable
research was focused on identifying the specific carbohydrate, lipid, nucleic acid or phosphate, and
are dimers consisting of at least two types of sub-
Corresponding author. Tel.: +49-(0)-9131-85-34577; units of either identical or different amino acid
Fax: +49-(0)-9131-85-34551;
composition. S100 proteins are highly conserved in
E-mail: kleindienst@nch.imed.uni-erlangen.de amino acid composition among vertebrate species

DOI: 10.1016/S0079-6123(06)61022-4 317


318

(Donato, 1986), and comparison of the human and is severely compromised (Ellis et al., 1995) allow-
bovine S100A1 and the human and rat S100B ing the cell release of preformed S100B. Secondly,
subunit reveals an almost complete homology since an injury-induced ATP and glutamate release
(Isobe et al., 1984). has been shown also, the release of S100B might be
The extracerebral origin of S100B has also been in part due to an astrocyte receptor activation by
recognized in several studies and this data conflicts ATP and glutamate (Ciccarelli et al., 1999). The
with the interpretation of S100B serum levels fol- finding of an increased S100B release at 24 and
lowing traumatic brain injury (TBI). The S100B 48 h post-injury may thus imply that this is a long-
tissue distribution has been assessed using term metabolic response to brain damage, which
immunocytochemical approaches (Donato, 1991). promotes a ‘‘healing process’’ in injured neurons
Quantification in rat tissue found a S100B con- or astrocytes.
centration in brain tissue of 3000 ng per milligram
of soluble protein, in adipose tissue of 1000 ng, in
the skin of 80 ng, in the testes of 40 ng, and in all Cellular action of S100B
other tissue of around 1–2 ng (Zimmer and Van
Eldik, 1987). Thus, elevated S100B levels following Intracellularly, S100B is involved in signal trans-
injury may also come from either damaged skeletal duction via the inhibition of protein phoshorylat-
muscle (Arcuri et al., 2002) or adipose tissue ion, regulation of enzyme activity and by affecting
(Suzuki et al., 1984). the calcium homeostasis as well as the regulation
of cell morphology by interaction with elements of
the cytoplasmatic cytoskeleton (Zimmer and Van
Release mechanism of S100B Eldik, 1986; Rustandi et al., 1998; Wilder et al.,
1998). After release into the extracellular fluid,
Experimental findings demonstrate that S100B is S100B acts in an autocrine and paracrine manner.
actively secreted from astroglia although the exact In vitro studies demonstrate mitogenic properties
mechanism has yet not been identified. Under of S100B, such as an increased proliferative acti-
physiological circumstances, S100B release can be vity on melanoma cell lines at concentrations be-
stimulated by 5 HT 1a agonists in cultured rat as- low 50 nM and above 5 mM (Klein et al., 1989), as
trocytes (Shashoua et al., 1984; Van Eldik and well as on rat C6 glioma cells at 50 pM–1.5 nM
Zimmer, 1987; Whitaker-Azmitia et al., 1990), and (Selinfreund et al., 1991), preferably in its oxidized
is observed within a few minutes after activation state (Scotto et al., 1998). Further investigations
of A1 adenosine or mGlu3 metabotropic gluta- also confirmed a dose-dependent action of S100B
mate receptors and release may last up to 10 h exerting a neuroprotective and neurotrophic influ-
(Ciccarelli et al., 1999). In neuronal plus glial cul- ence at nanomolar concentrations (Rickmann
tures, the level of S100B in the growth medium was et al., 1995; Li et al., 1998; Huttunen et al.,
measurable (Willoughby et al., 2004). Following 2000), but at micromolar concentrations, an acti-
experimental TBI, applying a 50 ms strain (stretch) vation of the inducible nitric oxide (NO) synthase
on these cell cultures grown on deformable silicone was seen with subsequent NO generation, poten-
membranes (Ellis et al., 1995), there was a dra- tially leading to astrocytic death (Hu et al., 1997;
matic rise in S100B 15 s after injury (Willoughby Petrova et al., 2000).
et al., 2004), and thereafter a continued release of Different studies provide evidence for neurotro-
S100B until at least 48 h post-injury (Slemmer phic properties of S100B. In cultured mesencephal-
et al., 2002; Willoughby et al., 2004). ic rat neurons (Azmitia et al., 1990) and dorsal
The injury-induced S100B release is likely to be root ganglia (Van Eldik et al., 1991), S100B
the result of at least two possible processes. Firstly, has been shown to promote neurite outgrowth
electron microscopic studies of stretch-injured as- (Kligman and Marshak, 1985; Winningham-
trocyte cultures show that immediately after Major et al., 1989). In primary rat spinal cord
stretch-injury structural and membrane integrity culture, S100B rapidly promoted reassembly
319

and/or stabilization of the cytoskeletal system proton spectroscopy has been shown to identify
(Nishi et al., 1997), and prevented apoptosis in a brain metabolites like N-acteylaspartate (NAA),
neuroblastoma clonal cell line (Brewton et al., creatine, choline and lactate (Bruhn et al., 1989;
2001). Most importantly, however, after sciatic Frahm et al., 1989a–c). We demonstrated MR
nerve section in newborn rats, S100B rescued mo- proton spectroscopy to detect different concentra-
tor neuron death and preserved neuron diameter tions of aqueous solutions of S100B in vitro, with
(Iwasaki et al., 1997). a strong correlation between the S100B concen-
In addition to its trophic properties, a neuro- tration and the area under the curve of the re-
protective effect of S100B was found in vitro. spective S100B MR peak at 4.5 ppm (Kleindienst
S100B decreased neuronal cell death and mito- et al., 2005b).
chondrial dysfunction in rat hippocampal neurons Increased cerebral S100B levels following an in-
after both glucose deprivation, and increased in- traventricular S100B infusion in normal rats were
tracellular free calcium (Barger et al., 1995). S100B confirmed by MR proton spectroscopy while the
is, thus, thought to be involved in regulation of S100B serum concentration did not increase ac-
energy metabolism by stimulation of the fructose- cording to the notion that proteins do not cross an
1,6-biphosphate aldolase (Zimmer and Van Eldik, intact BBB (Kleindienst et al., 2005b). Following
1986) and phosphoglucomutase (Landar et al., controlled cortical impact injury in the rat, S100B
1996). S100B has also been implicated in cytosolic serum levels were elevated up to 48 h post-injury
Ca2+ buffering (Xiong et al., 2000). In cultures of (Rothoerl et al., 2000). In fluid percussion injury in
embryonal chick and neonatal rat neurons, S100B the rat, serum S100B levels increased immediately
was found to protect against glutamate- and after injury, peaked at 3 h and returned to normal
staurosporin-induced damage (Ahlemeyer et al., values at 24 h after injury (Kleindienst et al., 2004).
2000). The above-mentioned neuroprotective and This time profile of serum S100B levels parallels
neurotrophic properties of S100B have not been the BBB disruption found after experimental
elucidated after TBI. TBI (Povlishock et al., 1978). In contrast, cere-
bral S100B levels as quantified by MR proton
spectroscopy were raised slightly 3 h after injury,
Passage of S100B through the blood-brain barrier and increased significantly thereafter to more than
twice these values by day 5 (Kleindienst et al.,
While in cell cultures, the injury-induced S100B 2005b). Thus, no clear relationship exists between
release continues to increase up to 48 h (Slemmer the cerebral S100B dynamics and the serum S100B
et al., 2002; Willoughby et al., 2004), S100B serum levels.
levels in patients are highest directly after injury,
and become normalized within 24 h in a high per-
centage of cases, even in those patients with a bad Clinical studies
outcome (Jackson et al., 2000). The underlying
mechanism of the passage of S100B through the In 2003, a thorough review of the role of S100B as
blood-brain barrier (BBB) has not been clarified a marker of brain damage was published summa-
yet, nor do data exist about cerebral S100B levels rizing the results of 18 clinical studies in a total of
and their correlation to serum S100B levels. Since 1085 patients (Rothermundt et al., 2003). In 2004
one of the purposes of the BBB is to prevent pro- and 2005, another six studies comprising more
teins to enter the brain, there is reasonable doubt than 600 adult patients were performed supporting
whether S100B of cerebral origin may be able to the correlation of elevated serum levels of S100B
cross the intact BBB in the opposite direction. with a poor outcome after brain injury (Pelinka
The principle of spectroscopy is widely applied et al., 2004; Savola et al., 2004; Vos et al., 2004;
in chemistry for the analysis of molecules in solu- Wunderlich et al., 2004; Berger et al., 2005; Chen
tion, and MR spectroscopy can be used to identify and Zhu, 2005). The time profile of S100B re-
important molecules in living tissue. Recently, MR ported in the serum was variable, and even a
320

delayed increase of S100B serum levels on the 6th studies are consistent with the hypothesis that
day after injury has been reported and speculated S100B plays a role in lesion-induced collateral
to occur due to secondary brain cell damage sprouting and reactive synaptogenesis (Rickmann
(Raabe and Seifert, 2000). Measurements of et al., 1995; Li et al., 1998; McAdory et al., 1998;
cerebrospinal fluid S100B levels allow a true as- Huttunen et al., 2000), and that repair may occur
sessment of the cerebral S100B release following by interaction with growth factors (Gomide and
brain insults (Kleine et al., 2003; Petzold et al., Chadi, 1999).
2003; Hayakata et al., 2004; Shore et al., 2004), but The hippocampus is a region critical for learning
require for repetitive measurements either lumbar and memory. It displays increased susceptibility to
or ventricular cerebrospinal fluid drainage. injury, and cognitive impairment following injury
The importance of S100B in the acutely injured has been linked to hippocampal dysfunction
brain is not well known yet. In view of a substan- (Hicks et al., 1993). Recent findings of neurogen-
tial body of evidence demonstrating an association esis within the subgranular zone of the dentate
between S100B and bad outcome after TBI, it is gyrus of the hippocampus in adult mammals pro-
important to be aware that proof of association vides a possible source to replace lost neurons
is not a proof of causation, in science, and this is (Eriksson et al., 1998; Kornack and Rakic, 1999;
especially true of S100B. In contrast to TBI, fol- Gould and Gross, 2002). Indeed, neuronal pro-
lowing electroconvulsive therapy in patients, in- genitor cells within the hippocampus present in-
creased serum S100B levels were associated with creased cellular proliferation and subsequent
an improved cognitive performance (Agelink et al., neuron formation following experimental TBI
2001). Furthermore, strong evidence exists that in (Dash et al., 2001; Chirumamilla et al., 2002).
traumatized patients S100B serum levels are in- This adult neurogenesis has been found to be ac-
creased without concomitant brain damage tively regulated by hippocampal astrocytes, which
(Ashraf et al., 1999; Anderson et al., 2001a, b; are a cellular source for mitogenic/neurotrophic
Svenmarker et al., 2002; Kleine et al., 2003; factors and thus promote proliferation of adult
Pelinka et al., 2003; Nygren De Boussard et al., neural stem cells and instruct the fate commitment
2004). Finally, the exact function and effects of of developing neurons (Song et al., 2002). Conse-
increased cerebral levels of S100B after TBI are quently, the astrocytic neurotrophic protein S100B
not thoroughly understood, yet. is a potential candidate to increase this progenitor
cell proliferation and subsequent neuron forma-
tion following TBI, thereby enhancing hippocam-
Effect of S100B on recovery following experimental pal network repair and improving cognitive
TBI recovery.
This hypothesis of a neurogenic effect of S100B
Besides this experimental evidence for the benefi- has been examined by an intraventricular infusion
cial effect of S100B on neuronal maintenance, a of S100B following fluid percussion injury in the rat
specific role of S100B has also been proposed at a (Kleindienst et al., 2004). Functional recovery was
higher level in developmental plasticity (Marshak, assessed by the Morris water maze 5 weeks post-
1990), and in cell processes thought to be involved injury and revealed a significantly improved cogni-
in learning and memory, such as long-term po- tive performance following intraventricular S100B
tentiation (Fazeli et al., 1990). This has been con- infusion (Kleindienst et al., 2004). Within the
firmed by injection of S100B anti-serum into the hippocampus, S100B increased the proliferative re-
hemisphere of chicks causing amnesia for a passive sponse significantly by more than 40% on day 5
avoidance task (O’Dowd et al., 1997). Vice versa, post-injury (Fig. 1; Kleindienst et al., 2005a). S100B
S100B infused into the rat hippocampus, has been promoted the survival of the injury-induced pro-
shown to facilitate long-term memory for an in- genitor cell proliferation as well as the subsequent
hibitory avoidance task (Mello e Souza et al., differentiation into neurons (Fig. 2; Kleindienst
2000). Finally, light- and electron microscopic et al., 2005a).
321

Fig. 1. Enhancement of progenitor cell proliferation in the hippocampus following TBI and S100B treatment. The photomicrograph
shows the distribution of BromodeoxyUridine (BrdU) immunoreactive cells within the ipsilateral dentate gyrus following lateral fluid
percussion injury and an intraventricular S100B infusion. BrdU immunoreactive cells on day 5 post-injury are predominately identified
in the area of their origin, the subgranular zone. The arrows point to typical cluster of cells.

Thus, S100B has been demonstrated to increase interpretation of serum S100B levels following TBI
hippocampal stem/progenitor cell proliferation requires thorough knowledge of factors influencing
and neuronal differentiation following TBI, and the measured concentration. Firstly, one has to be
this enhanced neurogenesis is correlated with an aware of a contribution to serum S100B levels
improved cognitive recovery (Kleindienst et al., originating from extracerebral sources. Secondly,
2005a). These findings stress the importance of the injury-induced cerebral S100B release may be
astrocytic factors for neurogenesis and provide assumed to be a combination of a passive release by
compelling evidence for a therapeutic potential of damaged astrocytes and an active release by stimu-
S100B improving functional recovery following lated astrocytes initiating repair mechanisms, with
TBI. Further studies are needed to elucidate the both release patterns varying over time and in the
beneficial role of S100B on hippocampal network presence of secondary insults. Thirdly, the passage
repair, thus offering a potential therapy to aug- of cerebral S100B is modulated by the BBB on its
ment important innate repair mechanisms of the way into the extracerebral compartment, and se-
brain in order to promote memory consolidation rum S100B levels do not reflect the corresponding
in patients with brain insults. cerebral S100B release. Finally, a beneficial effect
of S100B on neuronal maintenance, neurogenesis
and cognitive performance has been demonstrated
Conclusion promoting repair mechanisms and functional re-
covery following TBI.
Taken together, although the desire for a marker of Supplementing measurements of S100B serum
brain damage is reasonable designating S100B for levels, we established MR proton spectroscopy to
this role warrants considerable simplification, and more accurately reflect cerebral S100B levels and
322

Fig. 2. Neuronal differentiation of progenitor cells in the ipsilateral dentate gyrus following TBI and S100B treatment. The confocal
photomicrograph shows the staining of the neuronal Marker NeuN (red) and BrdU immunoreactivity (green) in the dentate gyrus at 5
weeks following lateral fluid percussion injury and an intraventricular S100B infusion. The arrows point at progenitor cells demon-
strating a co-localization of NeuN and BrdU (orange) thereby indicating neuronal differentiation.

thus provide a clinical method to non-invasively electroconvulsive therapy, cognitive side effects, neuron spe-
monitor whole brain S100B levels repeatedly in cific enolase, and protein S-100. J. Neurol. Neurosurg. Psy-
patients with brain insults. The determination of chiatry, 71: 394–396.
Ahlemeyer, B., Beier, H., Semkova, I., Schaper, C. and Kriegl-
cerebral S100B levels may allow a more accurate stein, J. (2000) S-100beta protects cultured neurons against
estimation of the prognosis in head-injured pa- glutamate- and staurosporine-induced damage and is in-
tients. Moreover, we propose that S100B far from volved in the antiapoptotic action of the 5 HT(1A)-receptor
being a negative determinant of outcome, as sug- agonist, Bay  3702. Brain Res., 858: 121–128.
Anderson, R.E., Hansson, L.O., Nilsson, O., Dijlai-Merzoug,
gested previously in the human TBI and ischemia
R. and Settergren, G. (2001a) High serum S100B levels for
literature, may improve neurogenesis and func- trauma patients without head injuries. Neurosurgery, 48:
tional recovery following acute brain injury and 1255–1258; discussion 1258–1260.
may be in fact potentially a new treatment option, Anderson, R.E., Hansson, L.O., Nilsson, O., Liska, J., Setter-
which might improve outcome of patients with gren, G. and Vaage, J. (2001b) Increase in serum S100A1-B
and S100BB during cardiac surgery arises from extracerebral
many forms of acute brain damage.
sources. Ann. Thorac. Surg., 71: 1512–1517.
Arcuri, C., Giambanco, I., Bianchi, R. and Donato, R. (2002)
Annexin V, annexin VI, S100A1 and S100B in develop-
References ing and adult avian skeletal muscles. Neuroscience, 109:
371–388.
Agelink, M.W., Andrich, J., Postert, T., Wurzinger, U., Zeit, Ashraf, S., Bhattacharya, K., Tian, Y. and Watterson, K.
T., Klotz, P. and Przuntek, H. (2001) Relation between (1999) Cytokine and S100B levels in paediatric patients
323

undergoing corrective cardiac surgery with or without total NMR spectroscopy using stimulated echoes: initial applica-
circulatory arrest. Eur. J. Cardiothorac. Surg., 16: 32–37. tions to human brain in vivo. Magn. Reson. Med., 9: 79–93.
Azmitia, E.C., Dolan, K. and Whitaker-Azmitia, P.M. (1990) Frahm, J., Bruhn, H., Gyngell, M.L., Merboldt, K.D., Ha-
S-100B but not NGF, EGF, insulin or calmodulin is a CNS nicke, W. and Sauter, R. (1989b) Localized proton NMR
serotonergic growth factor. Brain Res., 516: 354–356. spectroscopy in different regions of the human brain in vivo.
Barger, S.W., Van Eldik, L.J. and Mattson, M.P. (1995) S100 Relaxation times and concentrations of cerebral metabolites.
beta protects hippocampal neurons from damage induced by Magn. Reson. Med., 11: 47–63.
glucose deprivation. Brain Res., 677: 167–170. Frahm, J., Michaelis, T., Merboldt, K.D., Hanicke, W., Gyng-
Berger, R.P., Adelson, P.D., Pierce, M.C., Dulani, T., Cassidy, ell, M.L., Chien, D. and Bruhn, H. (1989c) Localized NMR
L.D. and Kochanek, P.M. (2005) Serum neuron-specific spectroscopy in vivo. Progress and problems. NMR Biomed.,
enolase, S100B, and myelin basic protein concentrations after 2: 188–195.
inflicted and noninflicted traumatic brain injury in children. Gomide, V.C. and Chadi, G. (1999) The trophic factors S-
J. Neurosurg., 103: 61–68. 100beta and basic fibroblast growth factor are increased in
Brewton, L.S., Haddad, L. and Azmitia, E.C. (2001) Col- the forebrain reactive astrocytes of adult callosotomized rat.
chicine-induced cytoskeletal collapse and apoptosis in N-18 Brain Res., 835: 162–174.
neuroblastoma cultures is rapidly reversed by applied Gould, E. and Gross, C.G. (2002) Neurogenesis in adult mam-
S-100beta. Brain Res., 912: 9–16. mals: some progress and problems. J. Neurosci., 22: 619–623.
Bruhn, H., Frahm, J., Gyngell, M.L., Merboldt, K.D., Hayakata, T., Shiozaki, T., Tasaki, O., Ikegawa, H., Inoue, Y.,
Hanicke, W. and Sauter, R. (1989) Cerebral metabolism in Toshiyuki, F., Hosotubo, H., Kieko, F., Yamashita, T.,
man after acute stroke: new observations using localized Tanaka, H., Shimazu, T. and Sugimoto, H. (2004) Changes
proton NMR spectroscopy. Magn. Reson. Med., 9: 126–131. in CSF S100B and cytokine concentrations in early-phase
Chen, D.Q. and Zhu, L.L. (2005) Dynamic change of serum severe traumatic brain injury. Shock, 22: 102–107.
protein S100b and its clinical significance in patients with Heizmann, C.W., Fritz, G. and Schafer, B.W. (2002) S100 pro-
traumatic brain injury. Chin. J. Traumatol., 8: 245–248. teins: structure, functions and pathology. Front. Biosci., 7:
Chirumamilla, S., Sun, D., Bullock, M.R. and Colello, R.J. d1356–d1368.
(2002) Traumatic brain injury induced cell proliferation in Hicks, R.R., Smith, D.H., Lowenstein, D.H., Saint Marie, R.
the adult mammalian central nervous system. J. Neurotrau- and McIntosh, T.K. (1993) Mild experimental brain injury in
ma, 19: 693–703. the rat induces cognitive deficits associated with regional
Ciccarelli, R., Di Iorio, P., Bruno, V., Battaglia, G., D’Alimonte, neuronal loss in the hippocampus. J. Neurotrauma, 10:
I., D’Onofrio, M., Nicoletti, F. and Caciagli, F. (1999) Acti- 405–414.
vation of A(1) adenosine or mGlu3 metabotropic glutamate Hu, J., Ferreira, A. and Van Eldik, L.J. (1997) S100beta in-
receptors enhances the release of nerve growth factor and duces neuronal cell death through nitric oxide release from
S-100beta protein from cultured astrocytes. Glia, 27: 275–281. astrocytes. J. Neurochem., 69: 2294–2301.
Dash, P.K., Mach, S.A. and Moore, A.N. (2001) Enhanced Huttunen, H.J., Kuja-Panula, J., Sorci, G., Agneletti, A.L.,
neurogenesis in the rodent hippocampus following traumatic Donato, R. and Rauvala, H. (2000) Coregulation of neurite
brain injury. J. Neurosci. Res., 63: 313–319. outgrowth and cell survival by amphoterin and S100 proteins
Donato, R. (1986) S-100 proteins. Cell Calcium, 7: 123–145. through receptor for advanced glycation end products
Donato, R. (1991) Perspectives in S-100 protein biology. Re- (RAGE) activation. J. Biol. Chem., 275: 40096–40105.
view article. Cell Calcium, 12: 713–726. Isobe, T., Takahashi, K. and Okuyama, T. (1984) S100a0 (al-
Donato, R. (2001) S100: a multigenic family of calcium- pha alpha) protein is present in neurons of the central and
modulated proteins of the EF-hand type with intracellular peripheral nervous system. J. Neurochem., 43: 1494–1496.
and extracellular functional roles. Int. J. Biochem. Cell Biol., Isobe, T., Tsugita, A. and Okuyama, T. (1978) The amino acid
33: 637–668. sequence and the subunit structure of bovine brain S-100
Ellis, E.F., McKinney, J.S., Willoughby, K.A., Liang, S. and protein (PAP I-b). J. Neurochem., 30: 921–923.
Povlishock, J.T. (1995) A new model for rapid stretch- Iwasaki, Y., Shiojima, T. and Kinoshita, M. (1997) S100 beta
induced injury of cells in culture: characterization of the prevents the death of motor neurons in newborn rats after
model using astrocytes. J. Neurotrauma, 12: 325–339. sciatic nerve section. J. Neurol. Sci., 151: 7–12.
Eriksson, P.S., Perfilieva, E., Bjork-Eriksson, T., Alborn, A.M., Jackson, R.G., Samra, G.S., Radcliffe, J., Clark, G.H. and
Nordborg, C., Peterson, D.A. and Gage, F.H. (1998) Neuro- Price, C.P. (2000) The early fall in levels of S-100 beta in
genesis in the adult human hippocampus. Nat. Med., 4: traumatic brain injury. Clin. Chem. Lab. Med., 38:
1313–1317. 1165–1167.
Fazeli, M.S., Errington, M.L., Dolphin, A.C. and Bliss, T.V. Klein, J.R., Hoon, D.S., Nangauyan, J., Okun, E. and Coch-
(1990) Extracellular proteases and S100 protein in long-term ran, A.J. (1989) S-100 protein stimulates cellular prolifera-
potentiation in the dentate gyrus of the anaesthetized rat. tion. Cancer Immunol. Immunother., 29: 133–138.
Adv. Exp. Med. Biol., 268: 369–375. Kleindienst, A., Harvey, H.B., Rice, A.C., Muller, C., Hamm,
Frahm, J., Bruhn, H., Gyngell, M.L., Merboldt, K.D., Hanicke, R.J., Gaab, M.R. and Bullock, M.R. (2004) Intraventricular
W. and Sauter, R. (1989a) Localized high-resolution proton infusion of the neurotrophic protein S100B improves
324

cognitive recovery after fluid percussion injury in the rat. O’Dowd, B.S., Zhao, W.Q., Ng, K.T. and Robinson, S.R.
J. Neurotrauma, 21: 541–547. (1997) Chicks injected with antisera to either S-100 alpha or
Kleindienst, A., McGinn, M.J., Harvey, H.B., Colello, R.J., S-100 beta protein develop amnesia for a passive avoidance
Hamm, R.J. and Bullock, M.R. (2005a) Enhanced hippo- task. Neurobiol. Learn Mem., 67: 197–206.
campal neurogenesis by intraventricular S100B infusion is Pelinka, L.E., Kroepfl, A., Leixnering, M., Buchinger, W.,
associated with improved cognitive recovery after traumatic Raabe, A. and Redl, H. (2004) GFAP versus S100B in serum
brain injury. J. Neurotrauma, 22: 645–655. after traumatic brain injury: relationship to brain damage
Kleindienst, A., Tolias, C.M., Corwin, F.D., Muller, C., Mar- and outcome. J. Neurotrauma, 21: 1553–1561.
marou, A., Fatouros, P. and Bullock, M.R. (2005b) Assess- Pelinka, L.E., Szalay, L., Jafarmadar, M., Schmidhammer, R.,
ment of cerebral S100B levels by proton magnetic resonance Redl, H. and Bahrami, S. (2003) Circulating S100B is in-
spectroscopy after lateral fluid-percussion injury in the rat. creased after bilateral femur fracture without brain injury in
J. Neurosurg., 102: 1115–1121. the rat. Br. J. Anaesth., 91: 595–597.
Kleine, T.O., Benes, L. and Zofel, P. (2003) Studies of the brain Petrova, T.V., Hu, J. and Van Eldik, L.J. (2000) Modulation of
specificity of S100B and neuron-specific enolase (NSE) in glial activation by astrocyte-derived protein S100B: differen-
blood serum of acute care patients. Brain Res. Bull., 61: tial responses of astrocyte and microglial cultures. Brain
265–279. Res., 853: 74–80.
Kligman, D. and Marshak, D.R. (1985) Purification and chara- Petzold, A., Keir, G., Lim, D., Smith, M. and Thompson, E.J.
cterization of a neurite extension factor from bovine brain. (2003) Cerebrospinal fluid (CSF) and serum S100B: release
Proc. Natl. Acad. Sci. U.S.A., 82: 7136–7139. and wash-out pattern. Brain Res. Bull., 61: 281–285.
Kornack, D.R. and Rakic, P. (1999) Continuation of neuro- Povlishock, J.T., Becker, D.P., Sullivan, H.G. and Miller, J.G.
genesis in the hippocampus of the adult macaque monkey. (1978) Vascular permeability alterations to horseradish per-
Proc. Natl. Acad. Sci. U.S.A., 96: 5768–5773. oxidase in experimental brain injury. Brain Res., 153: 223–239.
Landar, A., Caddell, G., Chessher, J. and Zimmer, D.B. (1996) Raabe, A. and Seifert, V. (2000) Protein S-100B as a serum
Identification of an S100A1/S100B target protein: phospho- marker of brain damage in severe head injury: preliminary
glucomutase. Cell Calcium, 20: 279–285. results. Neurosurg. Rev., 23: 136–138.
Li, Y., Wang, J., Sheng, J.G., Liu, L., Barger, S.W., Jones, Rickmann, M., Wolff, J.R. and Meyer, D.L. (1995) Expression
R.A., Van Eldik, L.J., Mrak, R.E. and Griffin, W.S. (1998) of S100 protein in the vestibular nuclei during compensation
S100 beta increases levels of beta-amyloid precursor protein of unilateral labyrinthectomy symptoms. Brain Res., 688:
and its encoding mRNA in rat neuronal cultures. J. Neuro- 8–14.
chem., 71: 1421–1428. Rothermundt, M., Peters, M., Prehn, J.H. and Arolt, V. (2003)
Marshak, D.R. (1990) S100 beta as a neurotrophic factor. Prog. S100B in brain damage and neurodegeneration. Microsc.
Brain Res., 86: 169–181. Res. Tech., 60: 614–632.
Matsutani, T., Nagayoshi, M., Tamaru, M., Hirata, Y. and Rothoerl, R.D., Brawanski, A. and Woertgen, C. (2000) S-100B
Kato, K. (1985) Changes in the levels of neural cell specific protein serum levels after controlled cortical impact injury in
proteins in the developing rat brain. Neurochem. Res., 10: the rat. Acta Neurochir. (Wien.), 142: 199–203.
1155–1172. Rustandi, R.R., Drohat, A.C., Baldisseri, D.M., Wilder, P.T.
McAdory, B.S., Van Eldik, L.J. and Norden, J.J. (1998) S100B, and Weber, D.J. (1998) The Ca(2+)-dependent interaction
a neurotropic protein that modulates neuronal protein phos- of S100B(beta beta) with a peptide derived from p53. Bio-
phorylation, is upregulated during lesion-induced collateral chemistry, 37: 1951–1960.
sprouting and reactive synaptogenesis. Brain Res., 813: Savola, O., Pyhtinen, J., Leino, T.K., Siitonen, S., Niemela, O.
211–217. and Hillbom, M. (2004) Effects of head and extracranial in-
Mello e Souza, T., Rohden, A., Meinhardt, M., Goncalves, juries on serum protein S100B levels in trauma patients.
C.A. and Quillfeldt, J.A. (2000) S100B infusion into the rat J. Trauma, 56: 1229–1234; discussion 1234.
hippocampus facilitates memory for the inhibitory avoidance Schafer, B.W., Wicki, R., Engelkamp, D., Mattei, M.G. and
task but not for the open-field habituation. Physiol. Behav., Heizmann, C.W. (1995) Isolation of a YAC clone covering a
71: 29–33. cluster of nine S100 genes on human chromosome 1q21: ra-
Moore, B.W. (1965) A soluble protein characteristic of the tionale for a new nomenclature of the S100 calcium-binding
nervous system. Biochem. Biophys. Res. Commun., 19: protein family. Genomics, 25: 638–643.
739–744. Scotto, C., Mely, Y., Ohshima, H., Garin, J., Cochet, C.,
Nishi, M., Kawata, M. and Azmitia, E.C. (1997) S100beta Chambaz, E. and Baudier, J. (1998) Cysteine oxidation in the
promotes the extension of microtubule associated protein2 mitogenic S100B protein leads to changes in phosphorylation
(MAP2)-immunoreactive neurites retracted after colchicine by catalytic CKII-alpha subunit. J. Biol. Chem., 273:
treatment in rat spinal cord culture. Neurosci. Lett., 229: 3901–3908.
212–214. Selinfreund, R.H., Barger, S.W., Pledger, W.J. and Van Eldik,
Nygren De Boussard, C., Fredman, P., Lundin, A., Andersson, L.J. (1991) Neurotrophic protein S100 beta stimulates
K., Edman, G. and Borg, J. (2004) S100 in mild traumatic glial cell proliferation. Proc. Natl. Acad. Sci. U.S.A., 88:
brain injury. Brain Inj., 18: 671–683. 3554–3558.
325

Shashoua, V.E., Hesse, G.W. and Moore, B.W. (1984) Proteins Whitaker-Azmitia, P.M., Murphy, R. and Azmitia, E.C. (1990)
of the brain extracellular fluid: evidence for release of S-100 Stimulation of astroglial 5-HT1A receptors releases the se-
protein. J. Neurochem., 42: 1536–1541. rotonergic growth factor, protein S-100, and alters astroglial
Shore, P.M., Thomas, N.J., Clark, R.S., Adelson, P.D., morphology. Brain Res., 528: 155–158.
Wisniewski, S.R., Janesko, K.L., Bayir, H., Jackson, E.K. Wilder, P.T., Rustandi, R.R., Drohat, A.C. and Weber, D.J.
and Kochanek, P.M. (2004) Continuous versus intermittent (1998) S100B(betabeta) inhibits the protein kinase C-depend-
cerebrospinal fluid drainage after severe traumatic brain in- ent phosphorylation of a peptide derived from p53 in a
jury in children: effect on biochemical markers. J. Neuro- Ca2+dependent manner. Protein Sci., 7: 794–798.
trauma, 21: 1113–1122. Willoughby, K.A., Kleindienst, A., Muller, C., Chen, T., Muir,
Slemmer, J.E., Matser, E.J., De Zeeuw, C.I. and Weber, J.T. J.K. and Ellis, E.F. (2004) S100B protein is released by in
(2002) Repeated mild injury causes cumulative damage to vitro trauma and reduces delayed neuronal injury. J. Neuro-
hippocampal cells. Brain, 125: 2699–2709. chem., 91: 1284–1291.
Song, H., Stevens, C.F. and Gage, F.H. (2002) Astroglia induce Winningham-Major, F., Staecker, J.L., Barger, S.W., Coats, S.
neurogenesis from adult neural stem cells. Nature, 417: and Van Eldik, L.J. (1989) Neurite extension and neuronal
39–44. survival activities of recombinant S100 beta proteins that
Suzuki, F., Kato, K. and Nakajima, T. (1984) Hormonal reg- differ in the content and position of cysteine residues. J. Cell
ulation of adipose S-100 protein release. J. Neurochem., 43: Biol., 109: 3063–3071.
1336–1341. Wunderlich, M.T., Wallesch, C.W. and Goertler, M. (2004)
Svenmarker, S., Sandstrom, E., Karlsson, T. and Aberg, T. Release of neurobiochemical markers of brain damage is re-
(2002) Is there an association between release of protein lated to the neurovascular status on admission and the site of
S100B during cardiopulmonary bypass and memory distur- arterial occlusion in acute ischemic stroke. J. Neurol. Sci.,
bances? Scand. Cardiovasc. J., 36: 117–122. 227: 49–53.
Van Eldik, L.J., Christie-Pope, B., Bolin, L.M., Shooter, E.M. Xiong, Z., O’Hanlon, D., Becker, L.E., Roder, J., MacDonald,
and Whetsell Jr., W.O. (1991) Neurotrophic activity of S-100 J.F. and Marks, A. (2000) Enhanced calcium transients in
beta in cultures of dorsal root ganglia from embryonic chick glial cells in neonatal cerebellar cultures derived from S100B
and fetal rat. Brain Res., 542: 280–285. null mice. Exp. Cell Res., 257: 281–289.
Van Eldik, L.J. and Zimmer, D.B. (1987) Secretion of S-100 Zimmer, D.B. and Van Eldik, L.J. (1986) Identification of a
from rat C6 glioma cells. Brain Res., 436: 367–370. molecular target for the calcium-modulated protein S100.
Vos, P.E., Lamers, K.J., Hendriks, J.C., van Haaren, M., Fructose-1,6-bisphosphate aldolase. J. Biol. Chem., 261:
Beems, T., Zimmerman, C., van Geel, W., de Reus, H., Biert, 11424–11428.
J. and Verbeek, M.M. (2004) Glial and neuronal proteins in Zimmer, D.B. and Van Eldik, L.J. (1987) Tissue distribution of
serum predict outcome after severe traumatic brain injury. rat S100 alpha and S100 beta and S100-binding proteins. Am.
Neurology, 62: 1303–1310. J. Physiol., 252: C285–C289.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 23

Cerebellar injury: clinical relevance and potential in


traumatic brain injury research

Eugene Park1,2, Jinglu Ai1 and Andrew J. Baker1,2,

1
St. Michael’s Hospital, Trauma Research, Toronto, ON, M5B 1W8, Canada
2
University of Toronto, Institute of Medical Sciences, Toronto, ON, M5S 1A, Canada

Abstract: A treatment for traumatic brain injury (TBI) remains elusive despite compelling evidence from
animal models for a variety of therapeutic targets. Numerous animal models have been developed to
address the wide spectrum of mechanisms involved in the progression of secondary injury after TBI.
Evidence from well-established models such as the fluid percussion injury (FPI) device, cortical impact
model, and the impact acceleration model has demonstrated diffuse pathophysiological mechanisms
throughout various brain structures. More specifically, we have recently extended characterization of the
FPI model to include pathophysiological changes in the cerebellum following unilateral fluid percussion.
Data suggest that the cerebellum is susceptible to selective Purkinje cell loss as well as white matter
dysfunction. Despite the cerebellum’s low profile in TBI research, there is evidence to warrant further study
of the cerebellum to examine mechanisms of neuronal death and traumatic axonal injury. Furthermore,
evidence from clinical literature and basic science suggests that some components of TBI pathophysiology
have a basis in cerebellar dysfunction. This review highlights some of the recent findings in cerebellar
trauma and builds an argument for including the cerebellum as a model to assess mechanisms of secondary
injury and its potential contribution to the pathology of TBI.

Keywords: animal models; cerebellum; electrophysiology; FPI; TAI; TBI

Introduction injury mechanisms exists in variety of injured axon


subpopulations (reviewed by Buki and Povlishock
Intense scrutiny of neuronal cell death and trau- (2006)). These data support a need for in-depth
matic axonal injury (TAI) has clarified numerous scrutiny of mechanisms encompassing all regions
molecular and cellular events contributing to the susceptible to traumatic brain injury (TBI) in or-
progression of secondary injury cascades. As our der to formulate an accurate and thorough de-
understanding of molecular events continues to scription of pathophysiologic processes.
increase, our description of these events have be- The cerebellum’s role in TBI has received rela-
come increasingly complex. For example, all trau- tively limited scrutiny and characterization as
matically injured axons were previously thought to other structures including the brain stem, hippo-
undergo a series of pathophysiological events lead- campus, and cerebral cortex. It is becoming in-
ing to disconnection and bulb formation. How- creasingly evident that general mechanistic
ever, it has become evident that a spectrum of descriptions of cell death and axonal injury are
overly simplistic and do not accurately reflect the
Corresponding author. Tel.: +1-416-864-5510; Fax: +1-416- full spectrum of events occurring after TBI. Clini-
864-5512; E-mail: bakera@smh.toronto.on.ca cal cases of TBI and parallel research in animal

DOI: 10.1016/S0079-6123(06)61023-6 327


328

models have confirmed that the mechanisms of many of the motor, cognitive, and speech impair-
injury extend to the cerebellum thus building a ments described above are also observed in TBI
case for its relevance in neurotrauma. A thorough patients.
characterization of all affected structures through- There are many reviews on diagnoses, mecha-
out the brain, including the cerebellum, would nisms, outcome, epidemiology, cost analysis,
provide a better framework with which to optimize rehabilitation, etc., and specific case studies in
treatments for TBI. Furthermore, the potential for TBI involving cerebral injury. However there is
discovering or elucidating mechanisms of injury limited clinical literature with direct emphasis on
not yet described would also add to our existing cerebellar trauma. In 1917, Gordon Holmes
knowledge of secondary injury mechanisms. described observations of World War I soldiers
It may be argued that cerebellar trauma is of who had sustained gunshot wounds to the cere-
limited clinical importance; however, recent studies bellum. He described a drunken-sailor type gait,
have begun to elucidate roles in higher order cog- as well as the postural disturbances commonly
nitive function not previously appreciated in cere- associated with cerebellar dysfunction (Holmes,
bellar processing (Petrosini et al., 1998; Ramnani, 1917). A recent report of 26 patients who had
2006). These higher order functions as well as other sustained infratentorial injuries is the only clinical
neurological processing functions not previ- study to date to specifically address the issue of
ously known may account for some of the ob- direct trauma to the cerebellum (Nathoo et al.,
served behavioral abnormalities in people afflicted 2002). This particular type of injury, although rare
by TBI. In the present discussion we propose seve- in the civilian population, has been documented
ral lines of evidence to support the role of cere- predominantly in military literature (Rish et al.,
bellar injury as an important component of TBI, 1983; Brandvold et al., 1990). Moreover, the au-
and also several unique features that render the thors of this study note that cerebellar injury had
cerebellum a potentially useful structure in which no effect on outcome. It should be noted however,
to assess mechanisms of secondary injury after that the five point Glasgow Outcome Scale (GOS)
trauma. used in this particular study might not be an ad-
equately sensitive measure of functional and be-
havioral deficits to detect cerebellar dysfunction.
Clinical evidence of cerebellar trauma Furthermore, the GOS is primarily a measure of
global outcome and does not factor in the conse-
Clinical manifestation of cerebellar dysfunction quences of motor, speech, and cognitive impacts
include perturbations to stance and gait, character- on daily living.
ized by a loss of equilibrium, a wide-based stance, Despite limited literature on direct cerebellar
irregular steps, and lateral veering (Mariotti trauma there is a varied collection of literature in
et al., 2005). Cerebellar deficits can also give rise which the cerebellum has been referenced as ex-
to tremors (e.g., Parkinsonian), alterations to hibiting pathological changes including selective
speech resulting in slowed or slurred vocalization, cell loss, altered metabolism, and white matter in-
as well as cerebellar mutism (Gordon, 1996). jury after focal and diffuse TBI. For example,
Recent studies have also indicated an important Purkinje cell loss has been noted in the brains of
contribution of cerebellar function to visuo-motor boxers and implicated as a significant contributor
control (Glickstein, 2000). Patients with cerebellar to ataxias and Parkinsonian tremors (Guterman
ataxias exhibit ocular motor defects in target and Smith, 1987; Unterharnscheidt, 1995a, b).
fixation and coordinating head movements Another report cites hypertrophic olivary and
(Mariotti et al., 2005). Although these deficits are Purkinje cell degeneration following chronic TBI
generally not described in the context of trauma, (Anderson and Treip, 1973). Post-traumatic intra-
it is not unreasonable to draw inference to trau- cerebellar hemorrhage, though an unusual clinical
matic cerebellar injury which could result in simi- occurrence, has been observed after head trauma
lar pathological impairments, particularly since and is an important clinical entity with respect to
329

neurological outcome (Bostrom et al., 1992; Cerebellar injury in animal models


D’Avella et al., 2001). There have also been sev-
eral reports of cerebellar atrophy following TBI Purkinje cell vulnerability
(Krauss et al., 1995; Soto-Ares et al., 2001; Gale et
al., 2005). Furthermore, a subgroup of ataxic TBI Several studies in animal models of TBI have docu-
patients may have etiologic basis in cerebellar mented vulnerability of Purkinje cells following
dysfunction after injury (Chester and Reznick, forebrain injury. Both midline and unilateral fore-
1987; Mysiw et al., 1990). Crossed cerebellar brain FPI have been shown to result in significant
diaschisis (CCD), a well-documented phenome- and delayed cell death of Purkinje neurons accom-
non of altered or depressed metabolic flow and panied by the presence of activated microglia
activity in the hemisphere of the cerebellum cont- (Fukuda et al., 1996; Mautes et al., 1996). Other
ralateral to the side of cortical lesion or injury, has studies have supported these findings by demon-
also been documented after TBI (Alavi et al., strating the presence of FluorojadeB positive cells
1997). Ipsilateral metabolic changes as well as in the cerebellum, consistent with the morphology
hypermetabolic changes have also been docu- of Purkinje neurons (Sato et al., 2001; Hallam
mented (Shamoto and Chugani, 1997; Niimura et al., 2004). Using double label fluorescent
et al., 1999). A loss of afferent inputs from the immunohistochemistry, we recently demonstrated
cortico-ponto-cerebellar pathway is believed to selective vulnerability of Purkinje neurons in the
result in deactivation of the targets in the cerebel- posterior regions of the cerebellum following uni-
lum, although the exact physiological mechanism lateral FPI in the cerebrum (Park et al., 2006b). In
is not entirely understood (Feeney and Baron, this study we demonstrated that the majority of
1986). The occurrence of CCD underscores the Purkinje neurons were lost in the acute phase of
importance of synaptic connectivity between injury (24 h) while delayed cell loss occurred in the
the cerebrum and the cerebellum. This would sug- middle and anterior regions of the cerebellum at 7
gest TBI resulting in a loss of cortical synaptic and 14 days post-injury at higher grades of injury
input to the cerebellum that results in an unknown severity (Fig. 1).
volume of information processing that is not Examination of the coronal plane cerebellar
taking place. sections following forebrain FPI indicates no
The relationship between ischemia and cere- evidence of medio-lateral banding (unpublished
bellar injury is also of significance as Purkinje observations). Others, however, have reported
cells are highly susceptible to ischemic injury parasagittal banding patterns of Purkinje cell
(Bhatia et al., 1995; Welsh et al., 2002). Ischemia loss using the CCI model of trauma (Weber, J.,
is a common component of non-penetrating head personal communication), perhaps indicative of
injuries and an important contributor to second- variations in injury biomechanics to the cerebel-
ary injury mechanisms (Graham et al., 1978, lum. Although the mechanisms of Purkinje cell
1989). Examination of 151 cases of fatal head loss following trauma are still unknown, there
injury revealed that the majority of ischemic are several potential explanations to consider
damage occurred in the hippocampus (81% of with relevance to neurotrauma research. Presy-
patients); however, there was also a significant naptic hyperexcitability, differential gene expres-
proportion of patients with evidence of ischemic sion, pre- and post-synaptic cell survival, and
injury in the cerebellum (44%; Graham et al., neuronal–glial interactions are several possible
1978). Furthermore, TBI, including multisystem avenues to be explored. The cerebellum offers
trauma patients, often present with hypoxic- several unique features to address these mecha-
hypotensive or hemorrhagic complications nisms as contributors to secondary injury. Fur-
(Miller et al., 1978, 1981; The Brain Trauma thermore, these areas have a broad range of
Foundation, 2000) increasing the likelihood of application to the study of TBI in general and
global brain ischemia and subsequent cerebellar are not limited to analysis of cerebellum specific
injury. effects.
330

Fig. 1. Double label immunohistochemistry indicates selective Purkinje cell death in the cerebellum following forebrain fluid per-
cussion trauma: (a) Sham, (b) 1 day post-injury, (c) 14 days post-injury. Scale bar ¼ 50 mm. (d) Quantification of surviving Purkinje
cells from the posterior cerebellum following four grades of fluid percussion trauma indicates a dose–response effect at 1 day post-
injury (red asterisk). A significant decline in Purkinje cell numbers relative to sham animals is observed as early as 1 day post-injury in
the 2–2.5 atm injury groups (*). The cartoon (left) indicates the area of fluid percussion trauma (red arrow) and the region of Purkinje
cell quantification (red circle). (Adapted with permission from Park et al., 2006b.)

Presynaptic hyperexcitability presynaptic hyperexcitability in parallel fiber-


Purkinje cell synaptic connections (Ai and Baker,
The synaptic architecture of the Purkinje cells, 2002). Given that a single Purkinje cell receives
consisting of glutamatergic inputs from climbing input from 200,000 parallel fibers (Fox and
and parallel fibers, creates an environment in Barnard, 1957), the potential for a presynaptic
which the potential for synaptic mediated excito- excitotoxic event is high. In addition, a single
toxicity is high (for review see Slemmer et al., climbing fiber, originating from the inferior olive,
2005). Our laboratory has demonstrated that forms up to 1500 synaptic connections on the
direct cerebellar trauma results in delayed proximal dendrite of a Purkinje cell (Strata and
331

Rossi, 1998). Excitation of this afferent pathway Pre- and post-synaptic cell survival
with ibogaine administration results in excitotoxic
Purkinje cell death while ablation with 3-AP is The cerebellum is a potentially useful structure in
protective to Purkinje cells (O’Hearn and Molliver, which to assess the effects of neuronal cell loss in
1997). The pattern of cell death from climbing fiber pre- and post-synaptic targets following TBI. For
depolarization results in very distinct medio-lateral example, the lurcher mouse phenotype resulting
banding pattern of cell death likely corresponding from a mutation of the Grid2 gene for the
to the terminal afferent projections of the climbing d2-glutamate receptor (d2-GluR) results in chronic
fibers. However, there are factors beyond simple depolarization and loss of Purkinje cells (Zuo
afferent targets to consider in this discussion of et al., 1997). Subsequent to Purkinje neuron death
Purkinje cell vulnerability. is the loss of granule cells as well as retrograde loss
of inferior olive input (Heckroth and Eisenman,
1991; Heckroth, 1992; Zanjani et al., 1998).
Anterograde effects manifest as a loss of deep
Gene expression cerebellar nuclei (Heckroth, 1994). Whether these
effects are specific to the mutation of the d2-GluR
Despite a seemingly homogeneous and redundant or are a common response to Purkinje cell loss has
arrangement of cellular architecture throughout not been elucidated but may be a valuable area of
the cerebellum (Voogd and Glickstein, 1998; research to pursue. The extensive literature
Ramnani, 2006), there exist subtle differences describing afferent tracing to the cerebellum as
within cell populations that can give rise to vastly well as its relatively simple synaptic organization
different pathologies. In particular, differential makes it a suitable model in which to evaluate
expression of genes involved in metabolism and retrograde and anterograde fates of injured or
cell signaling have been demonstrated within sub- dying neurons. These studies could add to our
sets of Purkinje neurons with both medio-lateral understanding of how focal injuries affect distal
and anterior–posterior patterns of expression targets through retrograde or anterograde signa-
(reviewed by Herrup and Kuemerle (1997) and ling dysfunction or cell loss.
Sarna and Hawkes (2003)) (Fig. 2). Furthermore
patterned gene expression in Purkinje neurons
occurs independently of afferent synaptic organi- Neuronal– glial cell communication
zation. The effectiveness of neuroprotective strat-
egies targeting these differentially expressed gene The role of neuronal–glial communication follow-
products can be readily ascertained through histo- ing neurotrauma remains a controversial issue
logical examination for the presence or absence of with evidence to supporting glial scarring as
banding patterns (see O’Hearn and Molliver, inhibitor of endogenous repair mechanisms along
1997). There are numerous candidate proteins with compelling evidence to indicate neuroprotec-
that are expressed in distinct patterns within the tive roles as well (reviewed by Sofroniew (2005)).
cerebellum that are of interest to neurotrauma In the cerebellum, Bergmann glia residing in the
research including heat shock proteins, zebrin molecular layer form a complex functional and
expression, calcium binding proteins, and amino structural relationship with Purkinje cells
acid transporters (Herrup and Kuemerle, 1997). (reviewed by Bellamy (2006)). These specialized
There may also be others that have not yet been astrocytes form a sheath around the Purkinje
characterized to date. Also of importance is the neuron soma and synapses. This physical interac-
maintenance of the compartmentalized expression tion has functional implications as stimulation of
of these gene products across mammalian species climbing and parallel fibers have been shown to
permitting for a degree of consistency in cross- activate inward currents in Bergmann glia (Bergles
species comparisons of mechanism and role in et al., 1997; Clark and Barbour, 1997; Bellamy
TBI. and Ogden, 2005). These currents are in part
332

Fig. 2. Cartoon representation of cerebellar compartmentation in the mouse, as revealed by the expression of zebrin II in subsets of
Purkinje cells (Adapted with permission from Sillitoe and Hawkes, 2002): anterior, dorsal and posterior views are shown. The
cerebellar vermis is divided into 10 lobules (I–X). However, a more fundamental parcellation is into four transverse zones — anterior
(AZ), central (CZ), posterior (PZ), and nodular (NZ). The AZ and PZ are striped — all Purkinje cells in the CZ and NZ express zebrin
II uniformly (although stripes can be revealed here by using other markers, such as the small heat shock protein, HSP25 (Armstrong et
al., 2000)). Complementary views of a cerebellum whole mount immunoperoxidase stained for zebrin II are shown on the right.
(Adapted with permission from Sarna and Hawkes, 2003.)

contributed to by glial-expressed alpha-amino-3- represent a specialized form of astrocyte, they


hydroxy-5-methyl-4-isoxazole propionic acid maintain the characteristic reactive astrocyte res-
(AMPA) receptors, GluR1 and GluR4, as well as ponse to injury following trauma. We have demon-
glial glutamate transporters, GLAST and GLT-1 strated the presence of reactive astrocytes in
(Bellamy, 2006). Although the Bergmann glia regions of Purkinje cell loss following forebrain
333

Fig. 3. (a) Confocal image of sham tissue expression of GFAP in Bergmann glia astrocytes, in red, in close proximity to Purkinje cells,
labeled in green. (b) Following forebrain fluid percussion trauma there is a marked increased in GFAP immunoreactivity in regions of
cerebellar injury indicated by the loss of Purkinje cells and increase in GFAP immunoreactivity. Scale bar ¼ 50 mm. (Adapted with
permission from Park et al., 2006b.)

trauma as indicated by increased GFAP expression parallel accumulation in a subset of axons was ob-
(Fig. 3). The close anatomical coupling and func- served at 1 day post-injury while persistent calpain-
tional relationship between Purkinje cells and Berg- mediated degradation of aII-spectrin was observed
man glia presents an excellent opportunity to exa- up to 14 days post-injury. The acute degradation of
mine the changes in neuronal and glial heavy neurofilament chain (NF200) and prolonged
communication following TBI. Specifically, the degradation of calpain-mediated aII-spectrin
cerebellum is an ideal system in which to perform would suggest temporally preferential targeting of
patch-clamping experiments in brain slices, which calpain substrates, NF200 and aII-spectrin, in the
permits maintenance of cellular architecture cerebellum. Another possibility is that proteolysis
and synaptic connectivity. These features may be of these substrates occurred in subsets of axons
helpful in elucidating the changes in communica- with different modes of axonal injury progression.
tion and synaptic plasticity between glial cells and The observations of NF200 and aII-spectrin
neurons in pathophysiological states. degradation are not novel concepts in the study
of TAI. However, the results reveal a further level
Cerebellar white matter injury of complexity in TAI with respect to calpain’s
temporal substrate specificity.
Numerous animal models of TBI have reported Compound action potential recordings from cere-
evidence of TAI in the cerebellum (Lighthall et al., bellar white matter have also demonstrated the use-
1990; Shima and Marmarou, 1991; Foda and fulness of electrophysiological techniques in assess-
Marmarou, 1994; Hoshino et al., 2003). Informa- ment of white matter function after TBI. The results
tion regarding the functional consequences of indicate a consistent CAP response within selected
such injuries, however, is limited. We recently regions. Interestingly, recordings from the middle
characterized functional and structural changes in cerebellar lobe produce a primarily fast conducting
cerebellar white matter following forebrain FPI myelinated response whereas the posterior and ante-
and demonstrated significant and persistent deficits rior cerebellar lobes exhibit a two-peaked response
and pathological changes in the cerebellum (Park including both fast and slow myelinated and un-
et al., 2006a). Early neurofilament degradation and myelinated responses, respectively (Fig. 4). This
334

Fig. 4. (a) Compound action potentials were recorded from three regions of cerebellar white matter as indicated by the black-boxed
regions. (b) CAP waveforms were dependent on location of recording. Anterior and posterior waveforms consisted of distinct fast and
slow components. The middle cerebellar CAP response was primarily a fast myelinated signal. (c) CAP values from cerebellar white
matter following forebrain fluid percussion trauma indicated a significant decline in electrophysiological function (*) in the posterior
and middle regions of the cerebellum which persisted at 14 days post-injury. (Adapted with permission from Park et al., 2006a.)

presents an opportunity to examine the effects of Given the relative lack of electrophysiological data
TBI and potential therapeutic interventions on both available for TBI white matter injury studies, a
myelinated and demyelinated axon populations. A comparison of results between the cerebellum and
recent study demonstrated varying electrophysio- the corpus callosum may reveal whether these sus-
logical susceptibilities of these two axonal popu- ceptibilities are structure specific or are a general
lations in the corpus callosum (Reeves et al., 2005). principle of injured axons throughout the brain.
335

The cerebellum as a model of neurotrauma cause of morbidity and mortality in young adults.
Despite the prevalence of this silent epidemic,
A key question in the discussion of cerebellar there is little therapeutic benefit that has translated
injury and its application in neurotrauma research from the neuroprotective strategies developed in
is ultimately whether it is clinically relevant. We animal models of TBI (Bullock et al., 1999; Faden,
would argue ‘yes’ for the following reasons. There 2001; Narayan et al., 2002; Tolias and Bullock,
is sufficient evidence of clinical manifestation of 2004). The failure of clinical trials highlights a lack
cerebellar injury following TBI. In addition to the of complete understanding of the complexity of
role in motor coordination, the role of the cere- TBI pathophysiology. This includes optimizing
bellum in multiple higher order function is becom- therapeutic time windows and clarifying the mul-
ing more appreciated and hence a recognition of its tiple pathways leading to cell death. Addressing
potential importance if injured. The traumatized these issues will require multifaceted approaches
cerebellum offers numerous technical opportuni- and balancing the inhibition of secondary injury
ties to examine relevant areas of research related to mechanisms while not adversely affecting normal
mechanisms of cellular injury, neuronal–glial physiologic function (Faden, 2002; Ikonomidou
pathophysiological interactions, TAI, post-injury and Turski, 2002). We believe that this will be best
synaptic plasticity, selective neuronal vulnerability, achieved through complementation and compari-
and mechanisms of diffuse injury remote from the sons between existing and novel injury paradigms.
location of initial trauma. From an anatomical The cerebellum is one such novel area that remains
perspective, there are notable differences in human to be fully appreciated and described in the context
and rodent cerebellar placement that is likely to of TBI.
result in differences on the production of injury
biomechanics between these two species. However,
Abbreviations
there are structural similarities in the cerebellum,
such as the cellular organization and foliated seg-
AMPA alpha-amino-3-hydroxy-5-
ments that are maintained throughout all mam-
methyl-4-isoxazole propionic acid
malian species (Voogd and Glickstein, 1998). This
CCD crossed cerebellar diaschisis
gyrencephalic property is not maintained in the
FPI fluid percussion injury
cerebral cortex between species and represents an
GluR glutamate receptor
area in which the biomechanics of injury produc-
GOS Glasgow outcome scale
tion may differ significantly between higher and
NF200 heavy neurofilament chain
lower order species. Despite the pros and cons of
TAI traumatic axonal injury
anatomical similarity between species, the intent of
TBI traumatic brain injury
this discussion is to provide a rationale for inclu-
sion of the cerebellum not only for its clinical and
functional importance in TBI pathophysiology, Acknowledgment
but also as a structure in which to examine the
mechanistic phenomena of TBI mechanisms in We thank the Ontario Neurotrauma Foundation
general. We propose that comparison and com- for its funding support.
plement of studies across various injury models
will not only validate but also elucidate novel References
mechanism of secondary injury.
Ai, J. and Baker, A. (2002) Presynaptic hyperexcitability at
cerebellar synapses in traumatic injury rat. Neurosci. Lett.,
332: 155–158.
Conclusion
Alavi, A., Mirot, A., Newberg, A., Alves, W., Gosfield, T.,
Berlin, J., Reivich, M. and Gennarelli, T. (1997) Fluorine-18-
Traumatic brain injury continues to be a health FDG evaluation of crossed cerebellar diaschisis in head
care issue of epidemic proportions as a leading injury. J. Nucl. Med., 38: 1717–1720.
336

Anderson, J.R. and Treip, C.S. (1973) Hypertrophic olivary Fox, C.A. and Barnard, J.W. (1957) A quantitative study of the
degeneration and Purkinje cell degeneration in a case of long- Purkinje cell dendritic branchlets and their relationship to
standing head injury. J. Neurol. Neurosurg. Psychiatry, 36: afferent fibres. J. Anat., 91: 299–313.
826–832. Fukuda, K., Aihara, N., Sagar, S.M., Sharp, F.R., Pitts, L.H.,
Armstrong, C.L., Krueger-Naug, A.M., Currie, R.W. and Honkaniemi, J. and Noble, L.J. (1996) Purkinje cell vulner-
Hawkes, R. (2000) Constitutive expression of the 25-kDa ability to mild traumatic brain injury. J. Neurotrauma, 13:
heat shock protein Hsp25 reveals novel parasagittal bands of 255–266.
purkinje cells in the adult mouse cerebellar cortex. J. Comp. Gale, S.D., Baxter, L., Roundy, N. and Johnson, S.C. (2005)
Neurol., 416: 383–397. Traumatic brain injury and grey matter concentration: a
Bellamy, T.C. (2006) Interactions between Purkinje neurones preliminary voxel based morphometry study. J. Neurol.
and Bergmann glia. Cerebellum, 5: 116–126. Neurosurg. Psychiatry, 76: 984–988.
Bellamy, T.C. and Ogden, D. (2005) Short-term plasticity of Glickstein, M. (2000) How are visual areas of the brain con-
Bergmann glial cell extrasynaptic currents during parallel nected to motor areas for the sensory guidance of movement?
fiber stimulation in rat cerebellum. Glia, 52: 325–335. Trends Neurosci., 23: 613–617.
Bergles, D.E., Dzubay, J.A. and Jahr, C.E. (1997) Glutamate Gordon, N. (1996) Speech, language, and the cerebellum. Eur.
transporter currents in bergmann glial cells follow the time J. Disord. Commun., 31: 359–367.
course of extrasynaptic glutamate. Proc. Natl. Acad. Sci. Graham, D.I., Adams, J.H. and Doyle, D. (1978) Ischaemic
U.S.A., 94: 14821–14825. brain damage in fatal non-missile head injuries. J. Neurol.
Bhatia, K.P., Brown, P., Gregory, R., Lennox, G.G., Manji, Sci., 39: 213–234.
H., Thompson, P.D., Ellison, D.W. and Marsden, C.D. Graham, D.I., Ford, I., Adams, J.H., Doyle, D., Teasdale,
(1995) Progressive myoclonic ataxia associated with coeliac G.M., Lawrence, A.E. and Mclellan, D.R. (1989) Ischaemic
disease. The myoclonus is of cortical origin, but the patho- brain damage is still common in fatal non-missile head injury.
logy is in the cerebellum. Brain, 118(Pt 5): 1087–1093. J. Neurol. Neurosurg. Psychiatry, 52: 346–350.
Bostrom, K., Helander, C.G. and Lindgren, S. (1992) Blunt Guterman, A. and Smith, R.W. (1987) Neurological sequelae of
basal head trauma: aspects of unconsciousness. Acta Neuro- boxing. Sports Med., 4: 194–210.
chir. Suppl. (Wien.), 55: 25–28. Hallam, T.M., Floyd, C.L., Folkerts, M.M., Lee, L.L.,
Brandvold, B., Levi, L., Feinsod, M. and George, E.D. (1990) Gong, Q.Z., Lyeth, B.G., Muizelaar, J.P. and Berman,
Penetrating craniocerebral injuries in the Israeli involvement R.F. (2004) Comparison of behavioral deficits and acute
in the Lebanese conflict, 1982–1985: analysis of a less neuronal degeneration in rat lateral fluid percussion and
aggressive surgical approach. J. Neurosurg., 72: 15–21. weight-drop brain injury models. J. Neurotrauma, 21:
Buki, A. and Povlishock, J.T. (2006) All roads lead to discon- 521–539.
nection? Traumatic axonal injury revisited. Acta Neurochir. Heckroth, J.A. (1992) Development of glutamic acid dec-
(Wien.), 148: 181–194. arboxylase-immunoreactive elements in the cerebellar cortex
Bullock, M.R., Lyeth, B.G. and Muizelaar, J.P. (1999) Current of normal and lurcher mutant mice. J. Comp. Neurol., 315:
status of neuroprotection trials for traumatic brain injury: 85–97.
lessons from animal models and clinical studies. Neurosur- Heckroth, J.A. (1994) Quantitative morphological analysis of
gery, 45: 207–220. the cerebellar nuclei in normal and lurcher mutant mice. I:
Chester, C.S. and Reznick, B.R. (1987) Ataxia after severe head morphology and cell number. J. Comp. Neurol., 343:
injury: the pathological substrate. Ann. Neurol., 22: 77–79. 173–182.
Clark, B.A. and Barbour, B. (1997) Currents evoked in Berg- Heckroth, J.A. and Eisenman, L.M. (1991) Olivary morphol-
mann glial cells by parallel fibre stimulation in rat cerebellar ogy and olivocerebellar topography in adult lurcher mutant
slices. J. Physiol., 502(Pt 2): 335–350. mice. J. Comp. Neurol., 312: 641–651.
D’avella, D., Cacciola, F., Angileri, F.F., Cardali, S., La Rosa, Herrup, K. and Kuemerle, B. (1997) The compartmentalization
G., Germano, A. and Tomasello, F. (2001) Traumatic intra- of the cerebellum. Annu. Rev. Neurosci., 20: 61–90.
cerebellar hemorrhagic contusions and hematomas. J. Neuro- Holmes, G. (1917) The symptoms of acute cerebellar injuries
surg. Sci., 45: 29–37. due to gunshot wounds. Brain, 40: 461–535.
Faden, A.I. (2001) Neuroprotection and traumatic brain injury: Hoshino, S., Kobayashi, S., Furukawa, T., Asakura, T. and
the search continues. Arch. Neurol., 58: 1553–1555. Teramoto, A. (2003) Multiple immunostaining methods to
Faden, A.I. (2002) Neuroprotection and traumatic brain injury: detect traumatic axonal injury in the rat fluid-percussion
theoretical option or realistic proposition. Curr. Opin. Neurol., brain injury model. Neurol. Med. Chir. (Tokyo), 43:
15: 707–712. 165–174.
Feeney, D.M. and Baron, J.C. (1986) Diaschisis. Stroke, 17: Ikonomidou, C. and Turski, L. (2002) Why did NMDA recep-
817–830. tor antagonists fail clinical trials for stroke and traumatic
Foda, M.A. and Marmarou, A. (1994) A new model of diffuse brain injury? Lancet Neurol., 1: 383–386.
brain injury in rats. Part II: morphological characterization. Krauss, J.K., Wakhloo, A.K., Nobbe, F., Trankle, R.,
J. Neurosurg., 80: 301–313. Mundinger, F. and Seeger, W. (1995) Lesion of
337

dentatothalamic pathways in severe post-traumatic tremor. Ramnani, N. (2006) The primate cortico-cerebellar system:
Neurol. Res., 17: 409–416. anatomy and function. Nat. Rev. Neurosci., 7: 511–522.
Lighthall, J.W., Goshgarian, H.G. and Pinderski, C.R. (1990) Reeves, T.M., Phillips, L.L. and Povlishock, J.T. (2005) Mye-
Characterization of axonal injury produced by controlled linated and unmyelinated axons of the corpus callosum differ
cortical impact. J. Neurotrauma, 7: 65–76. in vulnerability and functional recovery following traumatic
Mariotti, C., Fancellu, R. and Di Donato, S. (2005) An brain injury. Exp. Neurol., 196: 126–137.
overview of the patient with ataxia. J. Neurol., 252: Rish, B.L., Dillon, J.D. and Weiss, G.H. (1983) Mortality fol-
511–518. lowing penetrating craniocerebral injuries: an analysis of the
Mautes, A.E., Fukuda, K. and Noble, L.J. (1996) Cellular re- deaths in the Vietnam Head Injury Registry population.
sponse in the cerebellum after midline traumatic brain injury J. Neurosurg., 59: 775–780.
in the rat. Neurosci. Lett., 214: 95–98. Sarna, J.R. and Hawkes, R. (2003) Patterned Purkinje cell
Miller, J.D., Butterworth, J.F., Gudeman, S.K., Faulkner, J.E., death in the cerebellum. Prog. Neurobiol., 70: 473–507.
Choi, S.C., Selhorst, J.B., Harbison, J.W., Lutz, H.A., Sato, M., Chang, E., Igarashi, T. and Noble, L.J. (2001) Neu-
Young, H.F. and Becker, D.P. (1981) Further experience in ronal injury and loss after traumatic brain injury: time course
the management of severe head injury. J. Neurosurg., 54: and regional variability. Brain Res., 917: 45–54.
289–299. Shamoto, H. and Chugani, H.T. (1997) Glucose metabolism in
Miller, J.D., Sweet, R.C., Narayan, R. and Becker, D.P. (1978) the human cerebellum: an analysis of crossed cerebellar di-
Early insults to the injured brain. JAMA, 240: 439–442. aschisis in children with unilateral cerebral injury. J. Child.
Mysiw, W.J., Corrigan, J.D. and Gribble, M.W. (1990) The Neurol., 12: 407–414.
ataxic subgroup: a discrete outcome after traumatic brain Shima, K. and Marmarou, A. (1991) Evaluation of brain-stem
injury. Brain Inj., 4: 247–255. dysfunction following severe fluid-percussion head injury to
Narayan, R.K., Michel, M.E., Ansell, B., Baethmann, A., the cat. J. Neurosurg., 74: 270–277.
Biegon, A., Bracken, M.B., Bullock, M.R., Choi, S.C., Sillitoe, R.V. and Hawkes, R. (2002) Whole-mount
Clifton, G.L., Contant, C.F., Coplin, W.M., Dietrich, W.D., immunohistochemistry: a high-throughput screen for pat-
Ghajar, J., Grady, S.M., Grossman, R.G., Hall, E.D., terning defects in the mouse cerebellum. J. Histochem.
Heetderks, W., Hovda, D.A., Jallo, J., Katz, R.L., Knoller, Cytochem., 50: 235–244.
N., Kochanek, P.M., Maas, A.I., Majde, J., Marion, D.W., Slemmer, J.E., De Zeeuw, C.I. and Weber, J.T. (2005) Don’t get
Marmarou, A., Marshall, L.F., McIntosh, T.K., Miller, E., too excited: mechanisms of glutamate-mediated Purkinje cell
Mohberg, N., Muizelaar, J.P., Pitts, L.H., Quinn, P., death. Prog. Brain Res., 148: 367–390.
Riesenfeld, G., Robertson, C.S., Strauss, K.I., Teasdale, G., Sofroniew, M.V. (2005) Reactive astrocytes in neural repair and
Temkin, N., Tuma, R., Wade, C., Walker, M.D., Weinrich, protection. Neuroscientist, 11: 400–407.
M., Whyte, J., Wilberger, J., Young, A.B. and Yurkewicz, L. Soto-Ares, G., Vinchon, M., Delmaire, C., Abecidan, E.,
(2002) Clinical trials in head injury. J. Neurotrauma, 19: Dhellemes, P. and Pruvo, J.P. (2001) Cerebellar atrophy after
503–557. severe traumatic head injury in children. Child’s Nerv. Syst.,
Nathoo, N., Chite, S.H., Edwards, P.J. and van Dellen, J.R. 17: 263–269.
(2002) Civilian infratentorial gunshot injuries: outcome anal- Strata, P. and Rossi, F. (1998) Plasticity of the olivocerebellar
ysis of 26 patients. Surg. Neurol., 58: 225–233. pathway. Trends Neurosci., 21: 407–413.
Niimura, K., Chugani, D.C., Muzik, O. and Chugani, H.T. The Brain Trauma Foundation. The American Association of
(1999) Cerebellar reorganization following cortical injury in Neurological Surgeons. The Joint Section on Neurotrauma
humans: effects of lesion size and age. Neurology, 52: and Critical Care. (2000) Resuscitation of blood pressure and
792–797. oxygenation. J. Neurotrauma, 17: 471–478.
O’Hearn, E. and Molliver, M.E. (1997) The olivocerebellar Tolias, C.M. and Bullock, M.R. (2004) Critical appraisal of
projection mediates ibogaine-induced degeneration of Pur- neuroprotection trials in head injury: what have we learned?
kinje cells: a model of indirect, trans-synaptic excitotoxicity. NeuroRx, 1: 71–79.
J. Neurosci., 17: 8828–8841. Unterharnscheidt, F. (1995a) A neurologist’s reflections
Park, E., Liu, E., Shek, M., Park, A. and Baker, A. (2006a) on boxing. I: impact mechanics in boxing and injuries other
Heavy neurofilament accumulation and alpha-spectrin than central nervous system damage. Rev. Neurol., 23:
degradation accompanies cerebellar white matter functional 661–674.
deficits following forebrain fluid percussion injury. Exp. Unterharnscheidt, F. (1995b) A neurologist’s reflections on
Neurol., in press, Oct. 26, Epub ahead of print. boxing. V: Concluding remarks. Rev. Neurol., 23:
Park, E., McKnight, S., Ai, J. and Baker, A.J. (2006b) Purkinje 1027–1032.
cell vulnerability to mild and severe forebrain head trauma. Voogd, J. and Glickstein, M. (1998) The anatomy of the cere-
J. Neuropathol. Exp. Neurol., 65: 226–234. bellum. Trends Neurosci., 21: 370–375.
Petrosini, L., Leggio, M.G. and Molinari, M. (1998) The cere- Welsh, J.P., Yuen, G., Placantonakis, D.G., Vu, T.Q., Haiss,
bellum in the spatial problem solving: a co-star or a guest F., O’Hearn, E., Molliver, M.E. and Aicher, S.A. (2002)
star? Prog. Neurobiol., 56: 191–210. Why do Purkinje cells die so easily after global brain
338

ischemia? Aldolase C, EAAT4, and the cerebellar contri- cells but protects inferior olivary neurons from target-related
bution to posthypoxic myoclonus. Adv. Neurol., 89: cell death. J. Neurosci., 18: 319–327.
331–359. Zuo, J., De Jager, P.L., Takahashi, K.A., Jiang, W., Linden,
Zanjani, H.S., Vogel, M.W., Martinou, J.C., Delhaye-Bou- D.J. and Heintz, N. (1997) Neurodegeneration in Lurcher
chaud, N. and Mariani, J. (1998) Postnatal expression of mice caused by mutation in delta2 glutamate receptor gene.
Hu-bcl-2 gene in Lurcher mutant mice fails to rescue Purkinje Nature, 388: 769–773.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 24

Sex differences in brain damage and recovery of


function: experimental and clinical findings

Donald G. Stein

Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA

Abstract: Until the last decade or so, there was very little systematic examination of sex differences in
recovery from brain injury — most of the work was anecdotal or based on very small studies comparing
males to females. This chapter reviews some of the physiological, morphological, and functional evidence
for sex differences in response to brain injury across the spectrum of development. It also examines more
recent data showing that fluctuations in hormonal status during the menstrual and estrous cycle can play a
determining role in functional outcome in both normal and brain-injured females, and that these hormonal
influences can be measured at both the cellular and behavioral levels.

Keywords: sex differences; brain damage; recovery; hormones; neurosteroids

Introduction animal studies females were excluded because of


concerns that their hormonal cycling would interfere
Over the last 30 years, development of some 40 with or influence drug metabolism and therefore
different compounds, and industry and government- ‘confound’ the interpretation of results. However,
supported clinical trials for those compounds, have there have been a number of highly publicized in-
failed to find any pharmacologic agent that safely itiatives calling for the systematic study
and effectively led to better functional outcomes of gender differences, as well as several edited
after traumatic brain injury (TBI). With the qual- volumes on sex differences and brain function
ified exception of tissue plasminogen activator (McIntosh, 1996; Kimura, 1999; Morrison, 2000),
(TPA), the search for an effective treatment for and the number of empirical studies examining
ischemic stroke yielded the same disappointing sex differences in brain injury has increased sub-
results. Throughout all these trials, no substantive, stantially in the last few years.
systematic attention was paid to whether there
were any differences in brain injury severity and
outcomes between males and females. Many of Traumatic brain injury is not a unitary event
the clinical trials did not include females and most of
the laboratory research has focused closely on male As most of the articles in this volume will show,
animals. In the human trials, women were often ex- TBI is not a simple event in which a sudden impact
cluded because investigators were worried about or penetration of the brain parenchyma kills a
possible side effects of new drugs on fecundity or certain number of cells. Rather it is a complex
hormonal cycling. Indeed, in both human and cascade of events that continues to unfold over
time and involves a relatively large number of
Corresponding author. Tel: +1-404-712-9704; Fax: +1-404- genomic, metabolic, cellular and anatomic changes
727-2388; E-mail: DSTEI04@emory.edu that can take months to years before complete

DOI: 10.1016/S0079-6123(06)61024-8 339


340

stabilization. To promote functional and adaptive Matters for Neuroscience,’’ Larry Cahill (2006)
central nervous system (CNS) repair in the acute points out that there are ‘copious’ sex influences
stage of injury, a number of steps are required. on brain function and anatomy that are often dis-
First, the initial progression of events causing regarded in neuroscience research. He lists five
cytotoxicity, inflammation, swelling and short- misconceptions concerning sex influences on brain:
and long-term degeneration of nerve and glial (1) sex differences are small and unreliable (there-
cells must be reduced. This process of repair en- fore not worth considering?); (2) average differ-
gages not just the damaged neurons, but the entire ences between the sexes are due to a few extreme
organism’s adaptive capacity. The prevention of cases in a distribution — this was the same argu-
further loss of vulnerable or injured nerve cells can ment presented in the early days of work on
play an important role in rebuilding damaged cir- recovery of function — i.e., extremely rare phe-
cuitry and in providing a physiological matrix for nomena are not worthy of serious study; (3) differ-
later, successful rehabilitation training and ther- ences within sex are more substantial than between
apy. Second, once the cascade of cytotoxic events sex and therefore can be dismissed as trivial; (4)
is controlled, endogenous, growth-promoting fac- any sex differences are likely to be explained by the
tors as well as the administration of exogenous, effects of estrogen, primarily during development;
trophic agents may then stimulate the formation and (5) if no sex differences in behavior exist, it
of new axons, dendrites and synapses or even en- must mean that the neural mechanisms underlying
hance neurogenesis in the damaged adult brain behavior(s) are identical for males and females,
(Brinton and Wang, 2006; Kobayashi et al., 2006; although a number of CNS recording and imaging
Sailor et al., 2006; Shankaran et al., 2006; Shivraj studies now show that this is not the case (Gur and
Sohur et al.; 2006, Taupin, 2006). Taken together, Gur, 2004; Rider and Abdou, 2004; Stein, 2004;
all these changes require considerable time and all Cahill, 2006).
occur in a complex genomic and proteomic milieu
that can be dramatically affected by a number of
contextual parameters. We now know that such Are there structural differences between the male
parameters include the subject’s health, sex and and female brain that can affect plasticity and
genomic and hormonal status during development outcome of injury?
and at the time of injury. It is important to note
here that there is no empirically based ‘principle’ As Cahill has noted, much of the earlier literature
of neuroscience claiming that unless recovery of on sex differences in vertebrate brain structure
function occurs within the first few weeks after an emphasized the locus of control for mating, vocali-
injury, there will never be any recovery at all (see zation (in songbirds), reproduction, and aggres-
Dancause et al. (2005) for review and discussion sion and territorial behaviors in the male and
on this issue). female nervous systems (Arnold et al., 2004;
For the purposes of this chapter, one of the Collaer et al., 2004). Following on the rapid de-
key questions to be asked is, ‘‘Are there, in fact, velopments in non-invasive imaging techniques, a
sex differences in the outcome of brain injury, and substantial number of recent experiments have
could they be traced to differences in hormonal studied sex differences in metabolic activity in a
status or factors related to steroid synthesis in the variety of brain areas as men and women go about
brain before, during, or after traumatic damage?’’ performing sensory, motor and cognitive tasks.
It has typically been assumed that sex differences This is perhaps the one area of clinical neuro-
in the cognitive, sensory, or motor performance of science showing the most interest in studying
males and females are due to the divergences in the whether sexual dimorphisms underlie the differ-
anatomical structure and neuronal connectivity of ences in male and female behaviors (see Gur and
the male and female brain. This is often referred Gur (2004) and Becker (2002) for comprehensive
to in the experimental literature as sexual dimor- reviews). For the present chapter and volume the
phism. In an excellent recent review, ‘‘Why Sex goal is to determine whether sexual dimorphisms
341

in the anatomy of the brain could account for stages of the estrous cycle) and measured dendritic
any beneficial or detrimental differences in the re- spine density and arborization (the major site of
sponse of the nervous system to traumatic injuries. excitatory activity) in the anterior cingulate cortex
to determine whether age, sex, and ovarian hor-
monal status would change the morphology of
Sex differences in the gross and cellular anatomy of these cells. In general, young males had greater
the brain spine densities and arborization than females,
although both sexes showed an expected decline
There are now many anatomical and physiological in the medial frontal cortex with age. However,
studies describing sex differences in structure/ the aged females showed less age-related drop-off
function relationships in the brain (Kimura, of spine density and arborization than the males.
1992; Shaywitz et al., 1995; Kimura, 1999; Cahill, This may be because the aged female rats, unlike
2006). Recently, Rabinowicz et al. (2002) exami- human females, continue to produce ovarian hor-
ned 86 different areas in the brains of 6 males mones, which could prevent some of the cell loss.
and 5 females, 12–24 years old. These investiga- It is also worth noting that the Juraska laboratory
tors measured cortical thickness, neuronal and claims that males also show an age-related loss of
glial packing densities (the volume of cells in a dendritic spines in the motor cortex while females
geometrically defined area), and neuronal and as- do not (unpublished meeting abstract).
trocyte sizes (the size of the ‘neuropil’). The Other investigators have reported sex differences
authors reported that females had more overall in the long-term synaptic potentiation (LTP) of
neuropil than males, but the males had higher dentate granule cells of the hippocampus. This
neuronal densities (more synaptic contacts /mm3). activity is thought to subserve hippocampally
The men had smaller but more numerous units, mediated learning and memory. For example,
while women had larger cells, especially in the Maren (1995) recorded LTP in young male and
left hemisphere (women are more frequently right- female rats after single-pulse stimulation of the
handed and have better language abilities). In perforant path. In this study, stimulation led to
another recent report, Goldstein et al. (2001) a greater magnitude of LTP in the male rats
used MRI to obtain T1-weighted three-dimen- compared to females and was attributed to the
sional images from 48 normal adults of similar males having higher levels of NMDA receptor
education, age, socioeconomic status, intelligence, activation than the females. In this context, since
and handedness. These investigators found that, in males have higher levels of LTP activation, and
general, women had larger cortical brain volumes since hippocampus and temporal cortex are
than men; however, the frontomedial cortex, the particularly susceptible to seizures, it would be in-
amygdala and the hypothalamus were larger in teresting to learn whether male or female patients
men. In particular, there was greater sexual dimor- with brain injuries have a higher incidence of
phism in those brain regions which, compared to post-traumatic epilepsy. There are two additional
homologous animal studies, show the greatest num- points to note in this context. First, Goldstein
bers of sex steroid receptors during development. et al. (2001) showed that when relative difference
One area of the cortex that can be affected by in overall brain size is taken into consideration,
sex, age, and hormonal milieu of the subject is the while the female hippocampus is larger than
medial frontal cortex, implicated by numerous that of the male, males have a bigger volume of
lesion studies in spatial learning and cognition the CA1 field and larger neurons than females.
(Markham and Juraska, 2002). The frontal cortex Second, females with very high levels of estrogen
and its related structures have also received during their menstrual cycle are more likely to
considerable attention in the functional recovery suffer from catamenial epilepsy; this could be
literature in both humans and laboratory animals. due to the greater sensitivity of the female hippo-
Markham et al. (2005) examined both young and campal cells to neurotransmitters, glucocorticoids,
aged male and female rats (the latter in various and NMDA receptor binding (see Herzog et al.
342

(2004) and Reddy (2004) for more discussion on are purportedly less susceptible to ischemic dam-
this issue). age of the CA1 region (He et al., 2002) than males.
Although there are currently no direct studies Conejo et al. suggest that the neuroprotection
on humans bearing directly on whether women may be conferred by the higher number of glial
might have a higher incidence of post-traumatic cells secreting neurotrophic factors, an idea which
epilepsy, there are some interesting clues. For is receiving growing experimental support. The
example, Smith et al. (2002), using transcranial higher numbers of glial cells present could also
magnetic stimulation to activate motor-evoked play a role in more efficient scavenging of cyto-
potentials by paired-pulse technique in the motor toxic agents such as excitatory amino acids and
cortex, evaluated the effects of ovarian hormones free radicals. Of the various types of glial cells
on cortical excitability in healthy human females found in the brain, astrocytes are the most active
(average age of 34 years) during the stages of their in producing neurosteroids such as pregnenalone,
menstrual cycle. The study sought to determine progesterone, dehydroepiandrosterone (DHEA),
whether levels of estrogen and progesterone could testosterone, estradiol, and others (Zwain and
affect cortical excitability in a way similar to what Yen, 1999). Recently, Cerghet et al. (2006) re-
glutamate agonists or benzodiazepines might do at ported that the density of oligodendrocytes and
the glutamate or GABAA receptor(s), respectively. the expression of myelin proteins in the corpus
When estrogen levels were high during the late callosum, fornix and spinal cord are 20–40%
follicular stage, the motor-evoked potentials were greater in male compared to female rats. But in
significantly higher as well. During the luteal phase the corpus callosum, the generation of new glia
of the cycle when progesterone was at its peak, and their apoptotic loss is twice as great in females
the motor-evoked potentials were significantly as in males. The authors took this to mean that
lower. These data were taken to show that sex the lifespan and turnover of oligodendrocytes in
hormones could induce either inhibition or acti- females is shorter, and this substantial sex dif-
vation of cerebrocortical activity as well as blood ference suggests that hormones are playing a criti-
flow, thus affecting drug metabolism and possibly cal role in this ‘turnover’ process which would
CNS mechanisms involved in cognition, per- include the synthesis of new myelin.
ception and the CNS response to injury. These To know whether the amounts of such neuro-
data can be taken to suggest that, at least for steroids differ between the sexes or whether the
females, any dynamic brain imaging studies should levels of neurosteroids or their receptors vary as a
take into consideration the patient’s hormonal function of the estrous cycle in human females
state during testing and evaluation. will require more research. A better understanding
of the role these factors play in mediating CNS
plasticity could help to explain why females
Sex differences in the brain may be present in glial might have more advantage in recovery from
as well as neuronal morphology TBI than males, especially if they have the capacity
to generate more myelin following a brain injury
Recently, Conejo et al. (2003) used GFAP- (Cerghet et al., 2006). In support of this hypothe-
immunoreactive labeling and unbiased stereologi- sis, it is interesting to note that after brain or spinal
cal counting to determine the numbers of reactive cord injury, the synthesis of pregnenalone, pro-
astrocytes in the CA and CA3 areas of the adult gesterone, and allopregnanolone increases two to
rat hippocampus in both males and females four times in the areas immediately adjacent to
(whose measurements were taken during the pro- the injury sites. Concomitant with this rise there
estrous phase of their cycle). Males had higher is a significant increase in glial hyperplasia and
numbers of astrocytes than females in the CA3 the expression of myelin basic protein and re-
area of the hippocampus, but the females had myelination in the areas surrounding the injury
higher numbers of those cells in the CA1 region. (di Michele et al., 2000). It is also worthwhile to
It is interesting to note that females in proestrus note that the expression of the myelin proteins can
343

be directly affected by treatment with progesterone the female cells underwent less apoptosis than
and some of its metabolites — with progesterone male cells and that this may have been due to the
having a direct stimulatory effect (Schumacher significantly higher levels of ERK1, ERK2, Bcl-2
et al., 2004; De Nicola et al., 2006; Labombarda and Akt, which helped to preserve the stability
et al., 2006; Magnaghi et al., 2006). of mitochondrial membranes and produced less
In the brain itself, McCarthy et al. (2002), using degradation over time. The authors state that,
both light and electron microscopy, looked at ‘‘these data are the first demonstration of sexual
the effects of the estrous cycle on changes in the dimorphism in neuronal cell signaling,’’ and fur-
activity of the TrkA receptor, a major receptor ther, that gender may provide a context in which
for neurotrophins in the brain which plays a part the same biological stimuli can lead to different
in neurite outgrowth in the dendritic fields of the outcomes in disease and behavior. For us, the fact
hippocampus. When estrogen levels were high, that hardly any molecular biological investiga-
there was a 16-fold increase in the number of tors consider sex as a variable in the in vitro phar-
TrkA-labeled astrocytes, but this number dropped macokinetic studies leads us to wonder how valid
off precipitously when the animals were ovariec- are many of the reports when it comes to defining
tomized. After estrogen replacement, there was the proper parameters for drug design, develop-
again a 12-fold increase. At the electron micro- ment and application to the treatment of brain
scope level the authors observed that the TrkA- injury and other organic disorders.
labeled cells were situated closest to dendrites and To emphasize the point that sex differences are
to unmyelinated axons. McCarthy and colleagues important in determining outcome of brain injury,
attributed the beneficial effects to estrogen-induced three other studies should be mentioned. First, it
activity helping the cells play a role in axonal is now known that small, sublethal ischemic insults
guidance and synaptic regeneration. This notion can protect from additional ischemic damage in
would appear to support the more recent findings the adult brain, a phenomenon known as ‘ischemic
described above. (For more detailed reviews of preconditioning’ (IPC) (Truettner et al., 2002).
the role of hormones and dendritic activity, see Truettner and colleagues created bilateral cere-
McEwen (1998) and Arnold et al. (2004).) bral artery occlusions to cause mild temporary
Much of contemporary neuroscience involves ischemia in rats. They then took tissue samples
the use of cell culture preparations, but researchers from the hippocampus, cortex, and striatum to
rarely, if ever, report the sex of the donor cells, examine the expression of genes thought to be
probably thinking that the sex origin of such cells implicated in neuroprotection after injury (e.g.,
would have no consequence for the measures they brain-derived neurotrophic factor (BDNF), nerve
are making. A more recent study (Zhang et al., growth factor (NGF), c-jun, and c-fos), and found
2003) now questions this assumption. Zhang et al. significant increases. The authors speculated that
compared cell survival and the levels of phospho- the increase in neurotrophic factors following the
kinases ERK1, ERK2, Akt and the ‘survival’ pro- pre-conditioning injury could help with protecting
tein Bcl-2 in vitro in embryonic (E19) rat cortical neurons from a second attack. What is particularly
neurons (cortical plate and ventricular zone) taken interesting about this finding is that others have
from male and female donors. To analyze cell shown that the extent of tolerance to hypoxia and
survival, the cells were double-labeled with MAP-2 ischemia in females varies according to the estrous
(microtubule-associated protein) and anti-GABA cycle and relative levels of estrogen or proges-
antibodies on day 14 in culture. The phospho- terone at the time of the insult.
kinases were evaluated with Western blots and Second, recent investigations (Kasischke et al.,
optical density. Cells taken from females had 1999; von Arnim et al., 2002) using mouse hippo-
a highly significant increase in survival compared campal slice preparation recordings show that
to cells taken from male donors, although there short-term hypoxia will suppress normal electrical
did not appear to be any differences in the number activity of neurons. The return of electrical activity
of GABA-ergic neurons. The authors suggest that is measured by population spike amplitudes (PSA)
344

and varies according to stage of estrus. In the performed the task as well as intact controls.
Kasischke et al. experiment, the best overall re- In another series of projects Goldman created
covery of the PSA was found in the males, but for the same injuries in males and females, tested the
females the worst recovery was observed during animals soon after surgery and then again over a
proestrus when estrogen was high, and the best year later for their learning abilities. At first test-
recovery was seen during diestrus when progester- ing, the very young toddler females performed
one was high. This means that if hormonal status much better than their male counterparts, but
and cycling is not considered when females are the differences between sexes disappeared by 18
included in brain injury studies, the reported months of age. For Goldman, this suggested that
findings must be considered inconclusive or in- males were initially worse because their orbito-
complete. As a case in point, in the later study by frontal cortex and its expected ‘functions’ matured
von Arnim et al. (2002), progesterone receptors more rapidly, so the loss of this structure produced
were down-regulated by preconditioning lesions the early deficits. We have already seen that even
(seen when progesterone levels are high and asso- in the embryonic stage of development, female
ciated with neuroprotection). The authors observe brain cells appear to have higher titers of neuro-
that, ‘‘Not only is net hypoxic tolerance gender trophic activity than males and this may confer
dependent but underlying mechanisms are gender some potential protection (and better behavioral
specific. Both the net difference in primary and performance) against neuronal loss following in-
induced hypoxic tolerance and the specificity jury during infancy (Zhang et al., 2003). Sub-
of the mechanisms conferring the hypoxic toler- sequent studies showed that there are neurosteroid
ance should be considered in the development of receptors in the developing and adult cortex which
future gender-specific therapeutic strategies’’ (p. 87 can play a role in mediating injury-induced neural
[italics added]). plasticity (Clark et al., 1988).
In a follow-up primate study, Clark and Gold-
man-Rakic (1989) gave male and female rhesus
The influence of sex differences and hormonal status monkeys orbitofrontal cortex lesions at about 50
on cerebral functions in normal and brain-damaged days of age followed by testosterone injections
subjects in five of the females. The injections were given
from birth to 46 days of age. These animals were
There is now a very substantial literature on the then compared to groups of intact monkeys given
role of sex hormones in the development and locus testosterone prenatally or at 540 and 675 days
of reproductive, maternal, social, and aggressive after birth. The animals were then tested behavi-
behaviors, to which is now being added a growing orally on several cognitive discrimination learning
number of studies examining sex differences in tasks. Interestingly, intact males learned the tasks
more complex cognitive and sensory functions. In somewhat better than the intact females but the
this area, especially in animal research, lesion ex- males with brain injury were more impaired on
periments combined with behavioral testing have object reversal learning (the task required being
long been used to exemplify dimorphic cognitive able to give up a previously learned strategy) than
responses to brain injury. One of the first to use the lesion females. When the brain-injured females
this technique in studying the development of sex were given testosterone, the advantage disap-
differences in cognition and recovery after brain peared and the lesion females performed as badly
injury was Goldman (1974). Goldman created as the injured males, again purportedly demon-
orbitofrontal cortical lesions in 50-day-old male strating either the neuroprotective effects of the
and female rhesus monkeys and then tested them female sex hormones in a brain injury condition
on an object discrimination learning task. At or, quite possibly, the detrimental effects of giving
about two-and-a-half months of age, the males testosterone to females.
with orbitofrontal cortical lesions were impaired in In adult laboratory rats, sex differences in spar-
learning but the females with the same surgery ing and recovery of function have also been found
345

after unilateral lesions of the entorhinal cortex, an Raz et al. (1995) examined 58 children (34 boys,
area of the brain thought to be implicated in 24 girls) 3 years or older who had suffered an in-
working and reference memory processing (Roof tracranial hemorrhage (ICH) and who were of
et al., 1993). These investigators looking at spatial 37 weeks of gestation or less at birth. The children
learning performance in both males and females were equated for a variety of factors such as age,
found that intact animals of both sexes performed race, mother’s IQ, parents’ education, and socio-
about the same on a task which required them to economic status. These children were compared
swim to a submerged platform that could be lo- on the Revised Wechsler Intelligence Test to high
cated only by using room cues as a spatial refer- birth risk infants without ICH. The girls signi-
ence. After brain injury, the male rats had much ficantly outperformed the boys when the extent
more difficulty remembering the location of the of insult was controlled for. The authors suggest
platform than did the females with the same dam- that even after the various controls for lesion size,
age; the latter did not differ from intact males or males seem to be more functionally affected by
females on performance of this task, even though early brain injury than females because their brains
the entorhinal cortex lesions were very large. As are generally more vulnerable to insult. They state
we learn more about sex differences in the out- that, ‘‘it is possible that regardless of developmen-
come of brain injury, it is becoming apparent that tal factors, the male brain may possess structural
there are no simple answers. Some studies show or physiological properties that could not only
females have an advantage while others report that affect the extent of cerebral damage, but might
males do better, especially when the lesions are also compromise functional recovery’’ (p. 964).
inflicted early in life while hormonal parameters A subsequent paper by Donders and Hoffman
are still being developed (Kolb and Cioe, 1996; (2002) also reported that girls with blunt TBI had
Forgie and Kolb, 1998). better verbal outcome scores on the California
Extremely low birth weight (ELBW) children Verbal Learning Test than boys, even after they
(o1 kg) also show sex differences in cognitive were fully equated on a variety of demographic,
abilities as early as 2 years of age (Hindmarsh premorbid measures, including basic intelligence
et al., 2000). The authors followed almost 400 pre- and post-injury, and severity and type of in-
ELBW children after discharge and tested them jury. After the injury, the boys could not remember
for cognitive abilities at 2 years of age using the as many words, used fewer adaptive learning strat-
Griffiths Mental Development Scales (locomotor, egies and needed more time than girls to work on
personal social, hearing and speech, eye and hand test problems. Following their review of the liter-
coordination, and ‘practical reasoning’). For the ature, Donders and Hoffman suggest that the out-
most part, the investigators found that, except for come of sex differences in pediatric TBI could be
locomotion, the female children were ‘significantly due to the neuroprotective effects of sex steroids,
superior’ in all areas of testing and regardless especially progesterone, although there are few, if
of the specific impairments manifested by the any, studies examining the role of hormonal treat-
boys and girls at time of testing. These data can be ments in survivors of pediatric TBI. Although
taken to support other findings showing that, in there are reports of females doing better, some
general, very young females perform better in studies draw the opposite conclusions. For exam-
language and social development and that this ple, Morrison et al. (2004) found no statistically
difference could be due to exposure to differential significant differences in outcome between boys
gonadal hormones early in life, which then leads to and girls with brain injury and there was even some
more rapid maturation and enhanced rate of func- trend toward the females being somewhat worse.
tion in cerebral structures of females. Although
other explanations may be offered, this may be one Sex differences in the brain-injured adult
of the reasons why males tend to have a higher
prevalence of developmental lags and learning dis- The influence of hormonal status (e.g., estrus) on
abilities (Arnold, 1996; Berk, 1997). cognitive performance in adult humans has been
346

more controversial than the findings in children, In an animal model of transient forebrain is-
perhaps because of the ways in which hormonal chemia, b-estradiol has been shown to exacerbate
assays are conducted and performance is measured the loss of neurons in the CA1 pyramidal cell
over the menstrual cycle (see Epting and Overman field. When rats were ovariectomized the ischemia
(1998) and Mumenthaler et al. (2001) for detailed caused a loss of 32% of pyramidal cells, but with
discussions on this issue). Data from laboratory estradiol replacement in intact and ovariectomi-
animals may be more precise and consistent be- zed rats the mean cell loss was 54% and 49%,
cause, among other things, it is easier to obtain a respectively (Harukuni et al., 2001). Would ad-
variety of carefully timed measures of hormonal ministration of progesterone alone or in combina-
fluctuations (see DeGraba and Pettigrew (2000) tion with the estradiol have prevented this loss?
for a discussion on some of the issues). In rats Estrogen is a proexcitatory agent and could
(Kinsley et al., 1999), females that had a number of have caused neuronal loss by increasing excitotoxi-
litters previously were compared to age-matched city and subsequent inflammation in neurons ren-
females that never had a litter and to foster dered particularly vulnerable by the ischemic
mothers who never delivered any litters but raised event. The hippocampus is also particularly sensi-
pups that were given to them. The multiparous tive to seizure activity, which could have triggered
females and foster mothers, with higher circula- the additional loss of the injured neurons. Proges-
ting levels of progesterone, performed much better terone, in contrast, has been shown to inhibit sei-
on a complex maze task than did the nulliparous zure activity, especially in women with catemenial
animals. epilepsy, and inhibition of progesterone metabo-
In rhesus monkeys (Lacreuse et al., 2001), spa- lism increases seizure activity (Herzog and Frye,
tial recognition memory performance also varies 2003).
across the menstrual cycle. Performance of the ani-
mals was measured on a number of learning tasks
and found to be better during the follicular and Do females generally have better outcomes after
luteal phases than during the peri-ovulatory phase, brain damage than males?
when estrogen levels were at their highest. This
finding fits with other data showing that high levels There is considerable controversy in the literature
of estradiol can disrupt learning and memory in over the issue of whether females recover better
female rats (Galea et al., 2001). Taken together, from TBI and stroke than males, though anecdotal
these data suggest that either reduced levels of est- reports claim an advantage for females. Some
rogen, or relatively higher levels of progesterone, clinical case-study reports suggest that women re-
or possibly both, are beneficial to learning. The cover better from stroke than men, while others
role of estrogen and related hormones in mediating claim the opposite or no differences between the
recovery after brain injury is becoming an increas- sexes. For example, a paper by Kertesz and
ingly studied issue, especially in light of the re- McCabe (1977) examined 23 males and 13 females
cently failed trials with estrogen in the treatment of and found no differences in recovery between the
stroke and Alzheimer’s disease in women. With sexes, whereas Basso et al. (1982) examined males
respect to CNS trauma and possibly late-life de- and females and found long-term aphasia severity
generative disorders such as Alzheimer’s, it may to be much more marked in males than in females,
also be important to determine first whether the even though initial impairments were judged to be
extent and severity of the diseases vary in relation the same. Groswasser et al. (1998) looked at the
to the normal hormonal status of the female. If outcome of ‘severe’ brain injuries in males and
natural or induced hormonal cycling does play a females (72 females, 262 males) and controlled for
role in the progression of injury or disease, then the age range and severity. Females apparently had
timing of drug therapy to the appropriate stage of better overall responses to rehabilitation therapy
estrus will need to be considered in the treatment than males when return-to-work was used as an
of adolescent learning and attention disorders. outcome measure.
347

Some reports contest the Groswasser and col- To add to this controversy, Ratcliffe et al.
leagues findings. Kraus et al. (2000) looked at gen- (2003) looked at the relationship between gender
der differences in outcome after TBI in two trauma and cognitive recovery one year after blunt TBI.
centers and found that mortality was substantially They selected 325 records from the TBI Model
higher in females than in males with the same ex- Systems National Database and used multivariate
tent of injury. The females who survived had analyses to evaluate the association between sex
poorer outcomes compared to males. Supporting and six cognitive measures of attention — working
this finding, Farace and Alves (2000) performed a memory, verbal memory, language, visual analytic
meta-analysis of previously published TBI studies skills, problem solving, and motor functioning.
to determine whether women did better than men Females represented 31% of the total number of
in TBI outcome on 20 different measures. These patients tested. The women showed better per-
investigators found that fatality rates for women formance on five of the six cognitive outcome
with TBI were much higher than for men (because measures. The authors point out that pre-injury
of body size women may suffer more severe inju- differences were not a factor in this design because
ries; there are also some data to suggest that they men did not perform better than women after in-
are more often hit by motor vehicles than injured jury on tasks like visual memory, on which in
while driving them). Poorer outcomes were also the normal condition they typically outperform
reported for women on measures of disability, females.
seizures, coma, etc. Farace and Alves mention The discrepancies among the different outcome
that sex differences in drug metabolism and other studies can be due to a number of factors, not the
variables could contribute to the negative findings. least of which is the way in which patients are
The report makes no mention of attempting to de- selected for studies. For instance, Obler et al.
termine the women’s hormonal status before, dur- (1995) reported in ‘early’ (i.e., 1970s) studies that
ing or after injury to see if these factors could have lesion site rather than gender was the determining
contributed to the more detrimental outcomes. factor in selecting patients and that this could have
Farace and Alves list a number of reasons why biased outcome results. We now know that the loci
females may have poorer outcomes than males, of brain injuries related to aphasia severity and
one of them being sex hormones. Recently, Farin disorders in linguistic abilities differ between men
et al. (2003) reported that women have signi- and women. According to Kimura (1999), the in-
ficantly greater frequencies of brain swelling than cidence and severity of speech disorders is much
men and worse outcomes than men if they are un- higher in women if damage is to the anterior cortex
der 50 years of age at the time of their injury. in the left hemisphere, while the reverse is true for
These results could have been due to higher levels men. Women also have higher incidence of manual
of estrogen relative to progesterone in the younger apraxias after lateral frontal cortical injuries, while
women at the time of injury, but this variable men show greater deficits if the damage is in the
was not studied (i.e., the authors did not mention posterior cortex. Could the outcome of these
whether some of the older women were on hor- studies be influenced by the women’s hormonal
mone replacement therapy (HRT) and whether the status at the time of their injury? There is some
younger women were on contraceptives, both of tantalizing but indirect evidence bearing on this
which could affect outcomes). It was interesting to question. Hausmann et al. (2002) used radio-
note that after 50 years of age, when both estrogen immunoassays taken every three days to evaluate
and progesterone are diminished, the amount of the hormonal status of healthy women volun-
brain swelling in older females dropped and was teers participating in a perceptual asymmetry
almost equivalent to that of the males. Another study using a variety of behavioral tasks that are
retrospective study by Coimbra et al. (2003) also taken to measure cerebral asymmetries. When
found no evidence of any sex differences in TBI the women were high in serum progesterone, there
outcomes between males and females on any of was less task-dependent cerebral asymmetry on
their measures of recovery or mortality. some of the behavioral tests, leading to what the
348

investigators called ‘interhemispheric decoupling,’ claimed, no differences between the sexes, then this
and this was interpreted to mean that progesterone would need to be determined in systematic experi-
mediates the extent of cerebral lateralization. ments controlling for at least some of these vari-
Therefore, as noted earlier, when functional MRIs ables before any firm conclusions can be drawn.
are used to evaluate structure–function/metabolic This highlights the validity of doing animal experi-
activities in the brains of women, it may be im- ments on brain injury outcomes; most of the vari-
portant to determine the subjects’ specific hormo- ables relevant to gender-specific issues can be
nal status at the time of testing in order to obtain controlled better in a laboratory setting, thereby
more reliable information concerning CNS organi- reducing the variability typical of uncontrolled
zation and whether there is, indeed, contextual clinical assessments.
and/or sex related cerebral asymmetry. Clearly, the issues surrounding sex differences
With respect to a number of earlier studies (and in brain injury are complex and more research is
in some current ones), Obler et al. (1995) note that needed to tease out whether sex differences can
many of the variables that could determine apha- play a role in normal cognitive functions as well as
sia severity were not published (e.g., gender distri- in TBI prognoses. Given the growing literature
bution, time since onset of the injury and testing, showing that hormones like progesterone, estrogen
specific locus of the injury, prior health status, or even testosterone can serve as treatments for
hearing status, medication history, etc.), and this TBI (see for example Wright et al. (2006)), it is
lack of information could certainly have contri- likely that the study of sex differences will begin to
buted to the variability in the findings. For pur- receive more attention if for no other reason than
poses of the present discussion, especially for to improve the quality and success of clinical trials
female subjects, hormonal status at the time of seeking treatments for the victims of acquired
injury is never mentioned. Moreover, it is highly brain injuries.
unlikely that the patients were queried as to
whether they were taking any kind of birth con-
trol medication or HRT prior to or at the time of Acknowledgment
their TBI. Such a chronic history of hormone
regulation could affect the outcome of the brain Thanks to Leslie McCann for editorial assistance.
injury, if, in fact, hormone levels were playing
any role in mediating inflammatory processes that References
accompany TBI (Stein, 2005). In general, and es-
pecially when people suffer from a severe brain von Arnim, C.A., Etrich, S.M., Timmler, M. and Riepe, M.W.
injury, there is also accompanying severe bodily (2002) Gender-dependent hypoxic tolerance mediated via
injury and shock. There is evidence that females, gender-specific mechanisms. J. Neurosci. Res., 68: 84–88.
Arnold, A.P. (1996) Genetically triggered sexual differentiation
especially when progesterone levels are high rela-
of brain and behavior. Horm. Behav., 30: 495–505.
tive to estrogen, maintain better immune function Arnold, A.P., Agate, R.J. and Carruth, L.C. (2004) Hormonal
activity and response to physiologic shock than and non-hormonal mechanisms of sexual differentiation of
males (Wichmann et al., 1996; Zellweger et al., the brain. In: Legato M.J. (Ed.), Principles of Gender-
1997; Kuebler et al., 2003). These better levels of Specific Medicine. Academic Press, New York, pp. 84–95.
Basso, A., Capitani, E. and Moraschini, S. (1982) Sex differ-
immunodepression may also benefit the response
ences in recovery from aphasia. Cortex, 18: 469–475.
to brain injury. Becker, J.B. (2002) Behavioral Endocrinology. MIT Press,
Especially in older women with stroke or injury, Cambridge, MA.
whether they are/were on HRT prior to or after Berk, L.E. (1997) Child Development. Allyn & Bacon, Boston.
the damage is never noted. This could be parti- Brinton, R.D. and Wang, J.M. (2006) Therapeutic potential of
neurogenesis for prevention and recovery from Alzheimer’s
cularly important if the older women were or were
disease: allopregnanolone as a proof of concept neurogenic
not on HRT at the time of their injury. All these agent. Curr. Alzheimer Res., 3: 185–190.
factors could contribute to differences in brain in- Cahill, L. (2006) Why sex matters for neuroscience. Nat. Rev.
jury outcome, and if there are, as some have in fact Neurosci., 7: 477–484.
349

Cerghet, M., Skoff, R.P., Bessert, D., Zhang, Z., Mullins, C. Goldman, P.L. (1974) An alternative to developmental plas-
and Ghandour, M.S. (2006) Proliferation and death of ticity: heterology of CNS structures in infants and adults.
oligodendrocytes and myelin proteins are differentially regu- In: Stein D.G., Rosen J.J. and Butters N. (Eds.), Plasticity
lated in male and female rodents. J. Neurosci., 26: 1439–1447. and Recovery of Function in the Central Nervous System.
Clark, A.S. and Goldman-Rakic, P.S. (1989) Gonadal hor- Academic Press, New York, pp. 149–174.
mones influence the emergence of cortical function in non- Goldstein, J.M., Seidman, L.J., Horton, N.J., Makris, N.,
human primates. Behav. Neurosci., 103: 1287–1295. Kennedy, D.N., Caviness Jr., V.S., Faraone, S.V. and
Clark, A.S., Maclusky, N.J. and Goldman-Rakic, P.S. (1988) Tsuang, M.T. (2001) Normal sexual dimorphism of the adult
Androgen binding and metabolism in the cerebral cortex of human brain assessed by in vivo magnetic resonance imaging.
the developing rhesus monkey. Endocrinology, 123: 932–940. Cereb. Cortex, 11: 490–497.
Coimbra, R., Hoyt, D.B., Potenza, B.M., Fortlage, D., Groswasser, Z., Cohen, M. and Keren, O. (1998) Female TBI
Hollingsworth-Fridlund, P. (2003) Does sexual dimorphism patients recover better than males. Brain Inj., 12: 805–808.
influence outcome of traumatic brain injury patients? The Gur, R.G. and Gur, R.E. (2004) Gender differences in the
answer is no! J. Trauma, 54: 689–700 functional organization of the brain. In: Legato M.J. (Ed.),
Collaer, M.L., Tory, H.O. and Valkenburgh, M.C. (2004) Do Principles of Gender-Specific Medicine, Vol. 1. Academic
sex steroid hormones contribute to sexual differentiation of Press, New York, pp. 63–70.
the human brain? In: Legato M.J. (Ed.), Principles of Gen- Harukuni, I., Hurn, P.D. and Crain, B.J. (2001) Deleterious
der-Specific Medicine, Vol. 1. Academic Press, New York, effect of beta-estradiol in a rat model of transient forebrain
pp. 71–83. ischemia. Brain Res., 900: 137–142.
Conejo, N.M., Gonzalez-Pardo, H., Pedraza, C., Navarro, Hausmann, M., Becker, C., Gather, U. and Gunturkun, O.
F.F., Vallejo, G. and Arias, J.L. (2003) Evidence for sexual (2002) Functional cerebral asymmetries during the menstrual
difference in astrocytes of adult rat hippocampus. Neurosci. cycle: a cross-sectional and longitudinal analysis. Neuro-
Lett., 339: 119–122. psychologia, 40: 808–816.
Dancause, N., Barbay, S., Frost, S.B., Plautz, E.J., Chen, D., He, Z., He, Y.J., Day, A.L. and Simpkins, J.W. (2002)
Zoubina, E.V., Stowe, A.M. and Nudo, R.J. (2005) Extensive Proestrus levels of estradiol during transient global cerebral
cortical rewiring after brain injury. J. Neurosci., 25: ischemia improves the histological outcome of the hippo-
10167–10179. campal CA1 region: perfusion-dependent and -independent
DeGraba, T.J. and Pettigrew, L.C. (2000) Why do neuro- mechanisms. J. Neurol. Sci., 193: 79–87.
protective drugs work in animals but not humans? Neurol. Herzog, A.G. and Frye, C.A. (2003) Seizure exacerbation as-
Clin., 18: 475–493. sociated with inhibition of progesterone metabolism. Ann.
De Nicola, A.F., Gonzalez, S.L., Labombarda, F., Deniselle, Neurol., 53: 390–391.
M.C., Garay, L., Guennoun, R. and Schumacher, M. (2006) Herzog, A.G., Harden, C.L., Liporace, J., Pennell, P., Schomer,
Progesterone treatment of spinal cord injury: effects on re- D.L., Sperling, M., Fowler, K., Nikolov, B., Shuman, S. and
ceptors, neurotrophins, and myelination. J. Mol. Neurosci., Newman, M. (2004) Frequency of catamenial seizure exacer-
28: 3–15. bation in women with localization-related epilepsy. Ann.
Donders, J. and Hoffman, N.M. (2002) Gender differences in Neurol., 56: 431–434.
learning and memory after pediatric traumatic brain injury. Hindmarsh, G.J., O’Callaghan, M.J., Mohay, H.A. and
Neuropsychology, 16: 491–499. Rogers, Y.M. (2000) Gender differences in cognitive abili-
Epting, L.K. and Overman, W.H. (1998) Sex-sensitive tasks in ties at 2 years in ELBW infants. Extremely low birth weight.
men and women: a search for performance fluctuations Early Hum. Dev., 60: 115–122.
across the menstrual cycle. Behav. Neurosci., 112: 1304–1317. Kasischke, K., Huber, R., Li, H., Timmler, M. and Riepe, M.W.
Farace, E. and Alves, W.M. (2000) Do women fare worse: a (1999) Primary hypoxic tolerance and chemical precondition-
metaanalysis of gender differences in traumatic brain injury ing during estrus cycle in mice. Stroke, 30: 1256–1262.
outcome. J. Neurosurg., 93: 539–545. Kertesz, A. and McCabe, P. (1977) Recovery patterns and
Farin, A., Deutsch, R., Biegon, A. and Marshall, L.F. (2003) prognosis in aphasia. Brain, 100(Pt 1): 1–18.
Sex-related differences in patients with severe head injury: Kimura, D. (1992) Sex differences in the brain. Sci. Am., 267:
greater susceptibility to brain swelling in female patients 50 118–125.
years of age and younger. J. Neurosurg., 98: 32–36. Kimura, D. (1999) Sex and Cognition. MIT University Press,
Forgie, M.L. and Kolb, B. (1998) Sex differences in the Cambridge, MA.
effects of frontal cortex injury: role of differential hormonal Kinsley, C.H., Madonia, L., Gifford, G.W., Tureski, K.,
experience in early development. Behav. Neurosci., 112: Griffin, G.R., Lowry, C., Williams, J., Collins, J., McLearie,
141–153. H. and Lambert, K.G. (1999) Motherhood improves learning
Galea, L.A., Wide, J.K., Paine, T.A., Holmes, M.M., Ormerod, and memory. Nature, 402: 137–138.
B.K. and Floresco, S.B. (2001) High levels of estradiol dis- Kobayashi, T., Ahlenius, H., Thored, P., Kobayashi, R.,
rupt conditioned place preference learning, stimulus response Kokaia, Z. and Lindvall, O. (2006) Intracerebral infusion
learning and reference memory but have limited effects on of glial cell line-derived neurotrophic factor promotes striatal
working memory. Behav. Brain Res., 126: 115–126. neurogenesis after stroke in adult rats. Stroke, 37: 2361–2367.
350

Kolb, B. and Cioe, J. (1996) Sex-related differences in cortical estradiol and progesterone levels across the menstrual cycle
function after medial frontal lesions in rats. Behav. Neuro- and human performance. Psychopharmacology (Berl.), 155:
sci., 110: 1271–1281. 198–203.
Kraus, J.F., Peek-Asa, C. and McArthur, D. (2000) The in- Obler, L.K., Goral, M. and Albert, M.L. (1995) Variability in
dependent effect of gender on outcomes following traumatic aphasia research: aphasia subject selection in group studies.
brain injury: a preliminary investigation. Neurosurg. Focus, Brain Lang., 48: 341–350.
8: e5. Rabinowicz, T., Petetot, J.M., Gartside, P.S., Sheyn, D., Sheyn,
Kuebler, J.F., Yokoyama, Y., Janar, D., Toth, B., Rue III, T. and De, C.M. (2002) Structure of the cerebral cortex in
L.W., Bland, K.I., Wang, P. and Chaudry, I.H. (2003) men and women. J. Neuropathol. Exp. Neurol., 61: 46–57.
Administration of progesterone after trauma and hemorr- Ratcliffe, J.J., Greenspan, A., Goldstein, F.C., Wright, D.W.,
hagic shock prevents hepalocellular injury. Arch. Surg., 138: Stringer, A., Bell, D., Bushnik, T., Englander, J., Hammond,
727–734. F., Novack, T. and Whyte, J. (2003) Gender and traumatic
Labombarda, F., Gonzalez, S., Gonzalez Deniselle, M.C., brain injury: do the sexes fare differently? Society for Experi-
Garay, L., Guennoun, R., Schumacher, M. and De Nicola, mental Medicine Abstract. Acad. Emerg. Med., 10: 491.
A.F. (2006) Progesterone increases the expression of myelin Raz, S., Lauterbach, M.D., Hopkins, T.L., Glogowski, B.K.,
basic protein and the number of cells showing NG2 immuno- Porter, C.L., Riggs, W.W. and Sander, C.J. (1995) A female
staining in the lesioned spinal cord. J. Neurotrauma, 23: advantage in cognitive recovery from early cerebral insult.
181–192. Dev. Psychol., 31: 958–966.
Lacreuse, A., Verreault, M. and Herndon, J.G. (2001) Fluctu- Reddy, D.S. (2004) Role of neurosteroids in catamenial epi-
ations in spatial recognition memory across the menstrual lepsy. Epilepsy Res., 62: 99–118.
cycle in female rhesus monkeys. Psychoneuroendocrinology, Rider, V.A. and Abdou, N.I. (2004) Sexual dimorphism and the
26: 623–639. immune system. In: Legato M.J. (Ed.), Principles of Gender-
Magnaghi, V., Veiga, S., Ballabio, M., Gonzalez, L.C., Garcia- Specific Medicine, Vol. 2. Academic Press, New York,
Segura, L.M. and Melcangi, R.C. (2006) Sex-dimorphic pp. 1071–1081.
effects of progesterone and its reduced metabolites on gene Roof, R.L., Duvdevani, R. and Stein, D.G. (1993) Gender in-
expression of myelin proteins by rat Schwann cells. J. Peri- fluences outcome of brain injury: progesterone plays a pro-
pher. Nerv. Syst., 11: 111–118. tective role. Brain Res., 607: 333–336.
Maren, S. (1995) Sexually dimorphic perforant path long-term Sailor, K.A., Ming, G.L. and Song, H. (2006) Neurogenesis as a
potentiation (LTP) in urethane-anesthetized rats. Neurosci. potential therapeutic strategy for neurodegenerative diseases.
Lett., 196: 177–180. Expert Opin. Biol. Ther., 6: 879–890.
Markham, J.A. and Juraska, J.M. (2002) Aging and sex influ- Schumacher, M., Guennoun, R., Robert, F., Carelli, C., Gago,
ence the anatomy of the rat anterior cingulate cortex. Neuro- N., Ghoumari, A., Gonzalez Deniselle, M.C., Gonzalez, S.L.,
biol. Aging, 23: 579–588. Ibanez, C., Labombarda, F., Coirini, H., Baulieu, E.E.
Markham, J.A., McKian, K.P., Stroup, T.S. and Juraska, J.M. and De Nicola, A.F. (2004) Local synthesis and dual actions
(2005) Sexually dimorphic aging of dendritic morphology in of progesterone in the nervous system: neuroprotection
CA1 of hippocampus. Hippocampus, 15: 97–103. and myelination. Growth Horm. IGF Res., 14(Suppl A):
McCarthy, J.B., Barker-Gibb, A.L., Alves, S.E. and Milner, T.A. S18–S33.
(2002) TrkA immunoreactive astrocytes in dendritic fields of Shankaran, M., King, C., Lee, J., Busch, R., Wolff, M. and
the hippocampal formation across estrous. Glia, 38: 36–44. Hellerstein, M.K. (2006) Discovery of novel hippocampal
McEwen, B.S. (1998) Multiple ovarian hormone effects on neurogenic agents by using an in vivo stable isotope labeling
brain structure and function. J. Gend. Specif. Med., 1: 33–41. technique. J. Pharmacol. Exp. Ther., 319: 1172–1181.
McIntosh, T.K. (1996) Neuropathological sequelae of trau- Shaywitz, B.A., Shaywitz, S.E., Pugh, K.R., Constable, R.T.,
matic brain injury: relationship to neurochemical and bio- Skudlarski, P., Fulbright, R.K., Bronen, R.A., Fletcher,
mechanical mechanisms. Lab. Invest., 74: 315–342. J.M., Shankweiler, D.P., Katz, L., et al. (1995) Sex differ-
di Michele, F., Lekieffre, D., Pasini, A., Bernardi, G., Benav- ences in the functional organization of the brain for language
ides, J. and Romeo, E. (2000) Increased neurosteroids syn- [see comments]. Nature, 373: 607–609.
thesis after brain and spinal cord injury in rats. Neurosci. Shivraj Sohur, U., Emsley, J.G., Mitchell, B.D. and Macklis,
Lett., 284: 65–68. J.D. (2006) Adult neurogenesis and cellular brain repair with
Morrison, M.F. (2000) Hormones, Gender, and the Aging neural progenitors, precursors and stem cells. Philos. Trans.
Brain: The Endocrine Basis of Geriatric Psychiatry. Cam- R. Soc. Lond. B. Biol. Sci., 361: 1477–1497.
bridge University Press, New York, NY. Smith, M.J., Adams, L.F., Schmidt, P.J., Rubinow, D.R.
Morrison, W.E., Arbelaez, J.J., Fackler, J.C., de Maio, A. and and Wassermann, E.M. (2002) Effects of ovarian hormones
Paidas, C.N. (2004) Gender and age effects on outcome after on human cortical excitability. Ann. Neurol., 51: 599–603.
pediatric traumatic brain injury. Pediatr. Crit. Care. Med., 5: Stein, D.G. (2004) Brain trauma, sex hormones, neuronal sur-
145–151. vival and recovery of function. In: Legato M. (Ed.), Princi-
Mumenthaler, M.S., O’Hara, R., Taylor, J.L., Friedman, L. ples of Gender-Specific Medicine, Vol. 1. Academic Press,
and Yesavage, J.A. (2001) Relationship between variations in New York, pp. 104–115.
351

Stein, D. G. (2005) The case for progesterone. In: Singh M. and Wright, D.W., Kellermann, A.L., Hertzberg, V.S., Clark, P.L.,
Simpkins J.W. (Eds.), The Future of Estrogen and Hormone Frankel, M., Goldstein, F.C., Salomone, J.P., Dent, L.L.,
Theraphy in Postmenopausal Women: What Basic Science Harris, O.A., Ander, D.S., Lowery, D.W., Patel, M.M.,
and Clinical Studies Teach Us, Vol. 1052. Annals of the New Denson, D.D., Gordon, A.B., Wald, M.M., Gupta, S.,
York Academy of Science, pp. 152–169. Hoffman, S.W., Stein, D.G. (2006) Progesterone for acute
Taupin, P. (2006) Adult neurogenesis in mammals. Curr. Opin. traumatic brain injury. Ann. Emerg. Med.
Mol. Ther., 8: 345–351. Zellweger, R., Wichmann, M.W., Ayala, A., Stein, S., Demaso,
Truettner, J., Busto, R., Zhao, W., Ginsberg, M.D. and Perez- C.M. and Chaudry, I.H. (1997) Females in proestrus state
Pinzon, M.A. (2002) Effect of ischemic preconditioning on maintain splenic immune functions and tolerate sepsis better
the expression of putative neuroprotective genes in the rat than males. Crit. Care Med., 25: 106–110.
brain. Brain Res. Mol. Brain Res., 103: 106–115. Zhang, L., Li, P.P., Feng, X., Barker, J.L., Smith, S.V. and
Wichmann, M.W., Zellweger, R., Demaso, C.M., Ayala, A. Rubinow, D.R. (2003) Sex-related differences in neuronal cell
and Chaudry, I.H. (1996) Enhanced immune responses in survival and signaling in rats. Neurosci. Lett., 337: 65–68.
females, as opposed to decreased responses in males follow- Zwain, I.H. and Yen, S.S. (1999) Neurosteroidogenesis in as-
ing haemorrhagic shock and resuscitation. Cytokine, 8: trocytes, oligodendrocytes, and neurons of cerebral cortex of
853–863. rat brain. Endocrinology, 140: 3843–3852.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 25

Heat acclimation: a unique model of physiologically


mediated global preconditioning against traumatic
brain injury

Na’ama A. Shein1,2, Michal Horowitz2 and Esther Shohami1,

1
Department of Pharmacology, Hebrew University, Jerusalem, Israel
2
Department of Physiology, Hebrew University, Jerusalem, Israel

Abstract: Sub-lethal exposure to practically any harmful stimulus has been shown to induce consequent
protection against more severe stress. This preconditioning (PC) effect may be achieved by exposure to
different stressors, indicating that the induction of tolerance involves activation of common protective
pathways. Chronic exposure to moderate heat (heat acclimation, HA) is a unique PC model, since this
global physiological adaptation, as opposed to discrete organ PC, has been shown to induce cross-tolerance
against other stressors, including closed head injury (CHI). HA animals show accelerated functional re-
covery after injury which is accompanied by reduced secondary brain damage. However, the precise
mechanisms underlying this phenomenon have not been thoroughly studied until recently. Here we will
address the concept of PC, highlighting the unique properties of HA as a model which can be used for the
study of endogenous protective pathways triggered by PC procedures. Several molecular mechanisms which
are suggested to mediate HA-induced neuroprotection will also be discussed, bringing to light their pote-
ntial contribution to the development of traumatic brain injury treatment strategies utilizing therapeutic
augmentation of endogenous defense mechanisms.

Keywords: closed head injury; erythropoietin; heat acclimation; hypoxia inducible factor-1; inflammation;
neuroprotection; preconditioning

The phenomenon of preconditioning which enhance intrinsic defensive ability. Conceiv-


ably, the coping ability of pre-exposed organisms
Throughout history, several scholars have appro- will consequently be augmented in the setting of
ached the notion that harmful substances or recurring challenges (Fig. 1).
stressors may also engulf beneficial effects. These Janoff (1964), first introduced the terms ‘pre-
apparently conflicting observations were linked conditioning’ (PC) and ‘tolerance’ to describe the
by early-time savants such as Hippocrates and phenomenon, setting the stage for numerous later
Paracelsus to the extent of exposure or the given studies which described many different PC models
dose, suggesting that sub-lethal subjection to a and demonstrated the beneficial effects of this
noxious insult may bring about the activation of protocol in providing consequent protection.
endogenous protective response mechanisms, Traditionally, PC protocols were aimed at pro-
viding enhanced defensive ability to a specific or-
Corresponding author. Tel.: +972-2-675-7513; Fax: +972-2- gan, with cardiac PC being the most extensively
675-8741; E-mail: esty@cc.huji.ac.il studied model. In the mid 1980s, it was initially

DOI: 10.1016/S0079-6123(06)61025-X 353


354

Non-preconditioned
Basal defensive ability

Overall
Endogenous protection activation outcome
Preconditioned

Deleterious
processes

time
Attenuated Time interval Severe Outcome
stress / harmful Hours-weeks stress/harmful measurement
stimuli exposure stimuli induction

Fig. 1. General model of preconditioning. Pre-exposure to attenuated stress/harmful stimuli enhances endogenous protective mecha-
nisms, leading to improved overall outcome following subsequent more severe insult. Note that the beneficial effect brought about by
preconditioning results from augmented defensive ability rather than a direct effect on the extent of deleterious processes.

reported that pre-exposure to an attenuated is- the central nervous system has been the subject of
chemic insult affords subsequent myocardial pro- extensive research. Ischemic PC, as well as PC by
tection (Murry et al., 1986). The beneficial effects exposure to hyperbaric oxygen provided protection
of myocardial ischemic PC were later shown to in various models of spinal cord injury (Toumpoulis
include the reduction of infarct size, a decline in and Anagnostopolus, 2003; Nie et al., 2006). Nu-
the occurrence of cardiac arrhythmias and accel- merous studies have revealed the profound effect of
erated functional recovery following ischemia PC by brief ischemic exposure on the outcome of
(Shiki and Hearse, 1987; Cleveland et al., 1996). brain ischemic injury. The beneficial consequences
In vitro experiments also indicated the protective of such procedures have been demonstrated in
potential of myocardial PC (Ikonomidis et al., different in vitro (Reshef et al., 1996; Liu et al.,
1994). 2000) as well as in vivo models (Sharp et al., 2004;
Since then, ischemic PC-induced cardio-protection Glantz et al., 2005, and see Dirnagl et al., 2003;
has been reproduced by numerous groups in Blanco et al., 2006 for review). The substantiation
various species including humans (Vohra and of PC-afforded neuroprotection in animal models
Galinanes, 2006). In addition, myocardial PC has consequently instigated a wide-scale research effort
shown to be clinically relevant in the setting of aimed at exploring methods of employing ischemic
myocardial infarction and in patients receiving PC in stroke therapy (Schaller, 2005).
repeated cycles of aortic clamping and reperfu- Interestingly, over the years, a body of evidence
sion prior to cardiopulmonary bypass (CPB) accumulated, indicating that the initial definition
(Yellon and Dana, 2000). Subsequently, the ben- of PC as the induction of tolerance toward a spe-
eficial effects of ischemic PC on lung preservation cific stressor by attenuated pre-exposure to the
in patients undergoing open heart surgery with same type of stress might have been too narrow.
CBP were also reported (see Luh and Yang, 2006 It has been shown that the effects of PC may
for review). be mimicked by exogenous treatments. PC-like
Similarly, the neuroprotective potential of is- effects have been achieved in various models by
chemic/hypoxic PC against ischemic injury within pharmacological interventions utilizing low doses
355

of otherwise harmful substances. Boeck et al. Individual thermoregulatory effectors display a


(2004) have shown that exposure to low doses of similar pattern of biphasic response during HA.
NMDA induces protection against the hippocam- Salivation, which is utilized by several rodent spe-
pal neurotoxicity of quinolic acid. Analogous ob- cies as the major pathway of evaporative cooling,
servations were reported while using PC with clearly demonstrates this phenomenon. While sa-
thrombin (Hua et al., 2003; Cannon et al., 2005). liva production is inadequate for sufficient heat
Importantly, Dahl and Balfour (1964) demon- dissipation at the onset of acclimation due to im-
strated that in rats, exposure to short periods of paired responsiveness of muscarinic receptors,
anoxia induces a ‘whole organism’ protective once the new acclimated homeostasis has been
effect, indicating that PC can be applied toward achieved, 3 weeks later, glandular efficiency is
conferring protection to organisms and not only to regained due to upregulation and a return of these
discrete organs. receptors to pre-acclimation affinity (Kloog et al.,
Taken together, these findings suggest that PC is 1985; Horowitz, 2002). Similarly, enhanced auto-
a global and well-preserved adaptation mechanism nomic sympathetic activity is utilized in order to
which is aimed at enabling dealing with stress. The compensate for the desensitization of cardiac
fact that this endogenous mechanism can be adrenergic receptors during short-term HA,
prompted, not only by exposure to stress but also whereas the HA heart maintains the ability to
via administration of exogenous agents, is of great produce adequate contractile force, to enhance
importance since it suggests that PC-like effects work efficiency and to enhance pressure in the
may be induced at will in a relevant clinical setting. face of reduced ATP utilization (Levi et al., 1993;
In addition, since global exposure to external am- Horowitz, 1998, 2002).
bient anoxia can cause PC of the entire organism, At the organism level, the HA phenotype has
it is conceivable that activation of the protective been characterized by reduced metabolic and heart
mechanisms which underlie PC-induced tolerance rates, decreased core body temperature, lower tem-
could be achieved by exposure to other types of perature thresholds for the activation of heat dissi-
environmental stress. pation mechanisms and an elevated temperature
threshold for the development of thermal injury.
Evidence now indicates that this acclimated pheno-
Long-term heat acclimation: physiologically type results from both post-translational modifica-
mediated global preconditioning tions as well as genomic responses. Maloyan et al.
(1999) showed that heat acclimated rats display
Heat acclimation (HA) is a conserved adaptive re- about three times more inducible heat shock protein
sponse to exposure to moderately high ambient 72 kDa than do non-acclimated counterparts, thus
temperature. Conceptually, the process may be showing better coping with heat stress without the
delineated as a transition from an early, transient, need for de novo HSP synthesis (which character-
‘‘inefficient’’ state to an ‘‘efficient’’ acclimated izes the heat shock response of non-acclimated
homeostatic state (Horowitz, 2003, 2007). animals). Alterations in genomic responses during
Long-term HA is achieved by a 3–4 week expo- the course of HA have been shown to include
sure to mild environmental heat. The acclimation changes in the expression pattern of genes which are
process is biphasic, consisting of two phases which involved in anti-apoptosis, antioxidation and heat
differ in terms of the controlling mechanisms for shock response (Horowitz et al., 2004).
heat dissipation. While increased excitability of the Altogether, these responses contribute to an over-
autonomic nervous system compensates for im- all expansion of the dynamic thermoregulatory
paired cellular performance during short-term range. Consequently, HA results in enhanced abil-
acclimation, the features of HA include enhanced ity of coping with exposure to environmental heat.
cellular function accompanied by decreased excit- Hence, in accordance with the terminology intro-
ability (Horowitz and Meiri, 1985; Horowitz, 1994 duced by Janoff (1964) HA may be viewed as a
and for review, see Horowitz, 2002, 2007). long-term physiologically mediated heat-PC.
356

An inseparable outcome of HA is that adjusting better functional recovery as well as reduced


to one stressor can, in addition to evolving primary secondary tissue damage. The motor ability of
adaptations, add to the amount of adjustment to the rats was evaluated according to a neurological
additional stressors. Such cross-reinforcement severity score (NSS) which is based on the pres-
raises the possibility of inducing adaptation to a ence of some reflexes and the ability to perform
stressor without previous exposure to that particu- motor and behavioral tasks such as beam walking,
lar one. beam balance and spontaneous locomotion. HA
Indeed, the ability of HA to induce ‘‘cross- rats showed significantly better motor function
tolerance’’ against a variety of stressors has been 48 h following injury as compared with normo-
reported. HA has been shown to convey improved thermic controls. Additionally, brain edema and
cardiac mechanical and metabolic performance blood-brain barrier disruption were reduced in the
and reduced injury upon ischemic-reperfusion in- acclimated group. These findings were subse-
sult to the heart. Levi et al. (1993) showed that quently reinforced in mice (Shein et al., 2005a),
following total ischemia, acclimated hearts dis- demonstrating significantly better motor function
played enhanced preservation of the cardiac ATP 24 h post-injury accompanied with a reduction in
pool and a delayed decline in intracellular pH. brain water accumulation in HA mice. These mice
HA has also been shown to induce cross-tolerance also perform better in a memory test when exam-
toward exposure to hyperbaric oxygen. Arieli et al. ined by the object recognition test (Ennaceur and
(2003) reported a delay in the onset of central Delacour, 1988). When tested 3 days following
nervous system oxygen toxicity following HA, CHI, HA mice spent most of their exploration
demonstrating that the latency to the appear- time at the new object while normothermic coun-
ance of evident toxicity was twice as long in heat terparts failed to prefer the ‘novel’ object over the
acclimated rats. familiar one.
Taken together, these findings indicate a wide Interestingly, it has also been shown that
scale protective effect which is induced by HA. neuroprotection may be achieved in vitro by ex-
This observation may be of particular importance posing cell cultures to serum derived from HA
in the setting of traumatic brain injury. A neuro- animals. Beit Yannai et al. (1998) demonstrated
protective effect elicited by HA will potentially that when exposing cultures of PC12 cells (a sym-
have a fundamental effect on functional recovery pathetic pheochromocytoma cell line widely used
upon subsequent injury. Additionally, it could as a neuronal cell model) to 1% serum from HA
provide valuable insight into common protective rats, a significant reduction in cell death occurs as
pathways which are shared by HA and post-injury compared with cultures exposed to serum taken
recovery mechanisms. from normothermic ones. Additionally, the HA
serum treated cultures showed increased neuro-
protection upon exposure to a free radical gener-
HA-induced neuroprotection in closed head injury ator causing oxidative stress.
Taken together, current data establishes that HA
The effects of HA on the outcome of closed head elicits a continuum of processes resulting in en-
injury (CHI) have been examined using a well- hanced neuroprotection. The fact that the beneficial
established CHI model (described by Shapira et al., effect can be demonstrated in different species as
1988 and Chen et al., 1996). The model reproduces well as in varying experimental models is in agree-
the post-traumatic sequence of events observed in ment with the observations pointing to the wide-
humans and consistently recreates several patho- scale nature of PC-induced tolerance. Although
physiological features such as edema formation, HA-induced neuroprotection was hypothesized to
blood-brain barrier disruption and functional dys- be facilitated via common protective pathways
function. shared by different PC paradigms (Horowitz,
Shohami et al. (1994b) demonstrated that when 2004), the underlying mechanisms remained poorly
CHI is induced in HA rats, they display faster and defined for some time. However, recent work has
357

provided new insight into several molecular proc- is difficult given the large number of scavengers
esses which may contribute to the overall functional and the fact that they interact with each other,
improvement. such that increase of one may lead to a decrease of
the other. However, this limitation can be over-
come by the cyclic voltammetry technique. Cyclic
Mechanisms of HA-induced neuroprotection
voltammetry was traditionally used in chemistry to
study electron transfer between molecules and was
The research of HA-induced neuroprotection is
shown initially by Kissinger et al. (1973) to be
generally aimed at identifying molecular effectors
applicable for evaluating antioxidant levels in
and signaling cascades which are involved in the
vivo. A new method was then developed, enabling
development of the protected phenotype. In the
the measurement of overall antioxidant activity
following sections we describe our own studies
based on the determination of the total reducing
which examined the effects of HA on pathways
power of the biological tissue or fluid (Kohen
which are known to either convey a well-established
et al., 1999; Kohen and Nyska, 2002).
protective effect or foster deleterious consequences
In our model of CHI, we have previously shown
in CHI, in an effort to examine whether these
the activation of the arachidonic acid metabolic
pathways may also play a role in the beneficial
cascade which produces free radicals (Shohami
outcome induced by HA. These include low mole-
et al., 1987, 1989) as well as enhanced cytokine
cular weight antioxidants and anti-inflammatory
production (Shohami et al., 1994a), a typical in-
capacity, hypoxia inducible factor-1, erythropoietin
flammatory response involving ROS. In addition,
receptor expression and signaling as well as acute
we have demonstrated that a marked decrease in
post-CHI inflammation and the expression of
LMWA concentrations occurs within minutes
neurotrophic factors.
following injury, indicating their immediate utili-
zation in an effort to overcome acute oxidative
Effect of HA on the ability to cope with oxidative stress. The detrimental role of ROS in early-CHI
stress: endogenous and post-CHI low-molecular pathology was also supported by the observation
weight antioxidant profile that ROS-neutralizing compounds are protective
when given soon after CHI (Beit-Yannai et al.,
Reactive oxygen species (ROS) have been exten- 1996).
sively studied and proposed as candidates for the We have examined the LMWA in the brains of
elicitation of pathological responses in the patho- HA rats, in an effort to correlate the observed
genesis of ischemia and trauma (e.g., Chan, protection with the ability of the brain to neutral-
2001; Kontos, 2001). A number of therapeutic ap- ize ROS. A cyclic voltammetry study performed
proaches, based on intervention by scavenging on the water-soluble fraction of LMWA showed
ROS, have been attempted both in experimental that the chemical nature of LMWA is not altered
models and in the clinical setting (for review, see by HA. Interestingly, the concentrations of the
Vink and Van Den Heuvel, 2004). reducing equivalents were significantly lower in the
Generally, several lines of defense have been HA group, i.e. the basal LMWA levels were found
developed by living cells in order to cope with to be lower in HA rats (Beit Yannai et al., 1997).
oxidative stress. These include preventive and re- However, while the relative changes in anodic
pair mechanisms as well as an antioxidant defense currents during the post-CHI period revealed
system, consisting of antioxidative enzymes and a decrease from basal (sham) levels in the normo-
low molecular weight antioxidants (LMWA). The thermic animals, this did not occur in the HA
main direct-acting LMWA found within the brain group which sustained levels higher than those
include the tripeptide glutathione (glu-cys-gly), toco- measured in sham controls, for up to a week
pherols (vitamin E), ascorbic acid (vitamin C), following trauma.
histidine-related compounds, melatonin, uric acid In light of these results we suggested that simi-
and lipoic acid. Evaluation of individual LMWA larly to observations in the ischemic heat
358

acclimated heart (Horowitz et al., 2004), in the an appropriate acclimation protocol, the loss of
setting of CHI long-term exposure to high ambient function strain was unable to acclimate to heat.
temperature provides a more efficient mechanism The contribution of HIF-1 targeted pathways
of dealing with ROS displayed by better ability of to HA-induced cardiac cross-tolerance has also
adjusting to the subsequent acute stress. recently been established. Maloyan et al. (2005)
reported an increase in HIF-1a protein levels and
HIF-1 DNA binding activity as well as enhanced
Involvement of hypoxia inducible factor-1 and target gene mRNA expression in the hearts of HA
erythropoietin signaling rats.
HIF-1 regulates the expression of erythropoietin
Oxygen deprivation is known to evoke a sequele of (Epo). Epo, traditionally recognized as the main
adaptive responses. These responses are aimed at erythropoietic cytokine, has been shown to be ex-
compensating for decreased aerobic ATP produc- tensively expressed within the brain (Buemi et al.,
tion and are regulated by the transcriptional acti- 2002). The cytokine as well as its specific receptor
vator hypoxia inducible factor-1 (HIF-1). HIF-1 (EpoR) are expressed by neuronal, glial and brain
regulates the expression of over 70 known target capillary endothelial cells and are upregulated by
genes, leading to increased tissue oxygen delivery ischemic as well as metabolic stress (Bernaudin
and ATP production (Semenza, 2004). The func- et al., 1999, 2000). Importantly, Epo has been
tional factor is a heterodimer, consisting of a shown to engulf neuroprotection in a large variety
constitutively expressed b subunit and an inducible of in vitro and in vivo models of brain injury as
a subunit, which undergo dimerization by means well as in human stroke patients (Morishita et al.,
of basic helix-loop-helix PAS domains. Interest- 1997; Agnello et al., 2002; Catania et al., 2002;
ingly, the accumulation of HIF-1a and the tran- Ehrenreich et al., 2002; Grasso et al., 2002).
scriptional activation of HIF-1 have recently been We have previously established the neuropro-
shown to also be triggered by non-oxygen-dependent tective effect of Epo in our model of CHI (Yatsiv
pathways including ROS, cytokines and several et al., 2005). When treated with recombinant
growth factors and hormones (Chandel et al., human Epo (rhEpo) 1 and 24 h following CHI,
2000; Haddad, 2002). The fact that many HIF-1 mice displayed lower motor deficits and acceler-
regulated genes are related to oxygen and energy ated restoration of cognitive function. In addition,
homeostasis, taken together with its importance in a reduction in the number of apoptotic neurons as
environmental adaptive processes led to the hypo- well as caspase-3 expression was found in the
thesis that this factor may also be involved in rhEpo-treated group. This was also accompanied
the metabolic responses during HA. Furthermore, by better preservation of axons at the trauma area
preliminary data from Maloyan et al. (2001) and and reduced activation of glial cells in the injured
Bromberg and Horowitz (2004) indicated that the hemisphere.
levels of HIF-1a are increased following HA. In light of this, we hypothesized that HIF-1 and
However, the role of HIF-1 in HA remained in turn, Epo signaling, may also play a part in the
unclear and it was not known until 2 years later benefits brought about by HA. We proceeded to
whether the increase in the levels of this factor are initially examine the basal and post-injury levels of
of functional importance or only an epipheno- HIF-1a in HA mice and normothermic counter-
menon. parts. The levels of HIF-1a were found to be much
The question of the involvement of HIF-1 in higher following HA and this upregulation was
HA was assessed by Treinin et al. (2003). Their sustained 4 h following injury (Shein et al., 2005a).
study elegantly demonstrated that HIF-1 is essen- Subsequently, the expression of Epo and EpoR
tial to the development of HA, using a ‘hif-1 loss of were evaluated. An increase in both basal and
function’ mutant strain of the C. elegans nema- post-injury receptor levels was found, but surpris-
tode. As opposed to wild type nematodes, which ingly this was not accompanied by upregulation of
demonstrated an acclimated phenotype following Epo itself. This was unexpected, given the large
359

number of studies which highlighted the associa- When we examined the levels of total and phos-
tion of HIF-1 upregulation and increased expres- phorylated Akt in HA mice and compared them
sion of Epo. However, several other reports have with the levels found in non-acclimated controls,
demonstrated that the behavior of HIF-1 target we observed an increase in post-injury phosphory-
genes varies among different genes and is also lated Akt in the HA group (Shein et al., 2005b).
greatly dependent on the experimental model used We may therefore conclude that HA has both
(Jones and Bergeron, 2001). In addition, HIF-1 constitutive and dynamic effects; namely estab-
activation which is not followed by increased lishing higher basal levels and rapidly elevated
Epo expression has previously been described post-injury levels of both HIF-1a and EpoR and
in the presence of pro-inflammatory cytokines that this in turn leads to increased post-CHI
(Hellwig-Burgel et al., 1999). enhancement of Akt phosphorylation.
The levels of EpoR are known to be upregulated
by ischemia and hypoxia (Siren et al., 2001; Genc
Effect of HA anti-inflammatory capacity and acute
et al., 2004). The established involvement of is-
post-CHI inflammation
chemia in CHI pathophysiology may therefore
explain the post-injury upregulation of this recep-
The process of acute inflammation, initiated follow-
tor. Since HA has been shown to mediate enhanced
ing CHI has been shown to involve the upregulation
heat tolerance as well as cross-tolerance via in-
of pro-inflammatory cytokines (Shohami et al.,
creased gene responsiveness (Horowitz et al., 2004),
1994a). These cytokines, namely tumor necrosis
we propose that it is likely that the marked increase
factor alpha (TNF-a) and interleukin-1b (IL-1b),
in EpoR in HA mice following injury is due to an
when present during the initial time period follow-
additive effect of both injury-induced ischemia and
ing CHI, have been implicated of involvement in
increased responsiveness resulting from HA.
blood-brain barrier disruption and associated with
Since the levels of EpoR were consistently
subsequent edema formation (Shohami et al., 1997;
higher following HA only, as well as in HA mice
Stahel et al., 2000). Knoblach and Faden (1998)
subjected to CHI, we calculated the overall recep-
have previously demonstrated that the post-injury
tor/ligand Epo ratio as a measure of overall Epo
expression of these deleterious effectors may be at-
signaling ability. This ratio was significantly higher
tenuated by pre-injury treatment with the anti-
in the HA group and led us to further examine
inflammatory cytokine interleukin-10 (IL-10). We
downstream intracellular signaling which is medi-
therefore conceived that elevated pre-injury levels
ated by this receptor.
of anti-inflammatory mediators may lead to
We chose to focus on the extent of phosphory-
decreased post-CHI acute inflammation in HA ani-
lation of Akt as an indicator of intracellular anti-
mals. Our current data indicates that HA mice
apoptotic Epo/EpoR signaling. Following EpoR
do indeed display both an increase in basal,
activation, Akt is phophorylated via the phospha-
pre-injury anti-inflammatory capacity as well as a
tidylinositol-3 kinase pathway. When phosphory-
subsequent decrease in post-injury expression of
lated, Akt subsequently acts as a kinase and
pro-inflammatory cytokine mRNA (Shein et al.,
phosphorylates pro-apoptotic mediators including
2006). Immunohistochemical findings now indicate
Bad, caspase-9, the forkhead transcription factor
that HA also enhances the presence of brain-derived
and glycogen synthase kinase 3b. Each of the
neurotrophic factor-positive microglia (Shein et al.,
cellular systems targeted by Akt is inactivated by
2006).
phosphorylation, resulting in a blockade of apo-
ptotic cell death. Additionally, Akt phosphorylat-
ion has been associated with neuroprotection and Summary
has been suggested as one of the molecular path-
ways mediating Epo-induced neuroprotection Current data suggests that the neuroprotective
(Digicaylioglu et al., 2001; Chong et al., 2002; effect of HA is likely to be mediated by a network
Kilic et al., 2006). of pathways which work in concert to yield overall
360

functional improvement. In light of the findings to adaptive mechanism as a model for enabling
date, it can be suggested that HA leads to both the better definition of the role of discrete endogenous
potentiation of endogenous protective pathways as neuroprotective pathways in the development of
well as to the attenuation of detrimental post- tolerance.
injury processes. Although some changes are con- HA is a unique model in terms of providing in-
stitutive and observed following the acclimation sight into physiologically induced neuroprotec-
period itself while others are dynamic and only tion. Unraveling the physiological mechanisms of
apparent after injury, they all eventually confer tolerance development could have profound
modifications of the post-injury state (Fig. 2). The implications for treatment in the pathophysiologi-
fact that the effects of HA can be demonstrated cal setting of brain trauma, since future inter-
individually for different molecular mediators sub- vention strategies may be aimed at enhancing
stantiates the importance of this physiological endogenous protective ability in a physiological-
mimetic manner.

Higher levels in HA Lower levels in HA

Abbreviations
HIF-1α
CHI closed head injury
Pre-CHI EpoR CPB cardiopulmonary bypass
Epo erythropoietin
IL-10/IL-4 EpoR erythropoietin receptor
HA long-term heat acclimation
HIF-1 hypoxia inducible factor-1
IL-1b interleukin-1beta
LMWA IL-10 interleukin-10
TNF-α LMWA low molecular weight antioxi-
HIF-1α
Post-CHI IL-1β dants
EpoR PC preconditioning
p-Akt ROS reactive oxygen species
TNFa tumor necrosis factor alpha.
Note, factors which are higher in HA (either pre or
post CHI) are associated with neuroprotection.
Factors which are lower in HA after CHI are
associated with damage

Fig. 2. A schematic overview summarizing current experimen- References


tal data for mechanisms suggested as being involved in HA-
induced neuroprotection in closed head injury (CHI). Follow- Agnello, D., Bigini, P., Villa, P., Mennini, T., Cerami, A.,
ing long-term heat acclimation (HA), the pre-injury levels of Brines, M.L. and Ghezzi, P. (2002) Erythropoietin exerts
hypoxia inducible factor-1alpha (HIF-1a), erythropoietin re- anti-inflammatory effect on the CNS in a model of experi-
ceptor (EpoR) and anti-inflammatory cytokines are increased. mental autoimmune encephalomyelitis. Brain Res., 952:
Higher levels of HIF-1a and EpoR are sustained in HA animals 128–134.
following injury as well and are complemented by an increase in Arieli, Y., Eynan, M., Gancz, H., Arieli, R. and Kashi, Y.
post-CHI low molecular weight antioxidants (LMWA) and (2003) Heat acclimation prolongs the time to central nervous
phosphorylated Akt (p-Akt) as well as a reduction in early post- system oxygen toxicity in the rat: possible involvement of
injury levels of pro-inflammatory cytokines. Abbreviations: HSP72. Brain Res., 962: 15–20.
CHI, closed head injury; HA, long-term heat acclimation; Beit-Yannai, E., Kohen, R., Horowitz, M., Trembovler, V. and
LMWA, low molecular weight antioxidants; IL-10, interleukin- Shohami, E. (1997) Changes in biological reducing activity in
10; IL-4, interleukin 4; TNF-a, tumor necrosis factor alpha; rat brain following closed head injury: a cyclic voltammetry
IL-1b, interleukin-1beta; HIF-1a, hypoxia inducible factor-1alpha; study in normal and acclimated rats. J. Cereb. Blood Flow
EpoR erythropoietin receptor; p-Akt, phosphorylated Akt. Metab., 17: 273–279.
361

Beit-Yannai, E., Trembovler, V., Horowitz, M., Lazarovici, P., Chong, Z.Z., Kang, J.Q. and Maiese, K. (2002) Hematopoietic
Kohen, R. and Shohami, E. (1998) Neuroprotection against factor erythropoietin fosters neuroprotection through novel
oxidative stress by serum from heat acclimated rats. Neuro- signal transduction cascades. J. Cereb. Blood Flow Metab.,
sci. Lett., 254: 89–92. 22: 503–514.
Beit-Yannai, E., Zhang, R., Trembovler, V., Samuni, A. and Cleveland Jr., J.C., Wollmering, M.M., Meidrum, D.R., Row-
Shohami, E. (1996) Cerbroprotective effect of stable nitrox- land, R.T., Rehring, T.F., Sheridan, B.C., Harken, A.H. and
ide radicals in closed head injury in the rat. Brain Res., 717: Banerjee, A. (1996) Ischemic preconditioning in human and
22–28. rat ventricle. Am. J. Physiol., 271: H178–H179.
Bernadin, M., Bellail, A., Marti, H.H., Yvon, A., Vivien, D., Dahl, N.A. and Balfour, W.M. (1964) Prolonged anoxic sur-
Duchatelle, I., Mackenzie, E.T. and Petit, E. (2000) Neurons vival due to anoxia pre-exposure: brain ATP, lactate, and
and astrocytes express Epo mRNA: oxygen-sensing mecha- pyruvate. Am. J. Physiol., 207: 452–456.
nisms that involve the redox-state of the brain. Glia, 30: Digicaylioglu, M., Bichet, S. and Marti, H.H. (2001) Erythro-
271–278. poietin-mediated neuroprotection involves cross-talk
Bernaudin, M., Marti, H.H., Roussel, S., Divoux, D., Nouve- between Jak-2 and NF-kappaB signaling cascades. Nature,
lot, A., MacKenzie, E.T. and Petit, E. (1999) A potential role 412: 641–647.
for erythropoietin in focal permanent cerebral ischemia in Dirnagl, U., Simon, R.P. and Hallenbeck, J.M. (2003) Ischemic
mice. J. Cereb. Blood Flow Metab., 19: 643–651. tolerance and endogenous neuroprotection. Trends Neuro-
Blanco, M., Lizasoain, I., Sobrino, T., Vivancos, J. and sci., 26: 248–254.
Castillo, J. (2006) Ischemic preconditioning: a novel target Ehrenreich, H., Hasselblatt, M., Dembowski, C., Cepek, L.,
for neuroprotective therapy. Cerebrovasc. Dis., 21(Suppl 2): Lewczuk, P., Stiefel, M., Rustenbeck, H.H., Breiter, N.,
38–47. Jacob, S., Knerlich, F., Bohn, M., Poser, W., Ruther, E.,
Boeck, C.R., Ganzella, M., Lottermann, A. and Vendite, D. Kochen, M., Gefeller, O., Gleiter, C., Wessel, T.C., De Ryck,
(2004) NMDA preconditioning protects against seizures and M., Itri, L., Prange, H., Cerami, A., Brines, M. and Siren,
hippocampal neurotoxicity induced by quinolinic acid in A.L. (2002) Erythropoietin therapy for acute stroke is both
mice. Epilepsia, 45: 745–750. safe and beneficial. Mol. Med., 8: 495–505.
Bromberg, Z. and Horowitz, M. (2004) Mus musculus: a model Ennaceur, A. and Delacour, J. (1988) A new one-trial test for
for studying the dynamics of heat acclimation and cross tol- neurobiological studies of memory in rats. 1: behavioral data.
erance between heat acclimation and oxygen deprivation. Behav. Brain Res., 31: 47–59.
Proceedings of the 1st Integrated meeting on Physiology and Genc, S., Koroglu, F. and Genc, K. (2004) Erythropoietin and
Pharmacology of Thermoregulation (PPTR 2004), October the nervous system. Brain Res., 1000: 19–31.
2004, Rhodes, Greece, p. 198. Glantz, L., Avramovich, A., Trembovler, V., Gurvitz, V., Kohen,
Buemi, M., Cavallaro, E., Floccari, F., Sturiale, A., Aloisi, C., R., Eidelman, L.A. and Shohami, E. (2005) Ischemic precon-
Trimarchi, M., Grasso, G., Corica, F. and Frisina, N. (2002) ditioning increases antioxidants in the brain and peripheral
Erythropoietin and the brain: from neurodevelopment to organs after cerebral ischemia. Exp. Neurol., 192: 117–124.
neuroprotection. Clin. Sci., 103: 275–282. Grasso, G., Buemi, M., Alafaci, C., Sfacteria, A., Passalacqua,
Cannon, J.R., Keep, R.F., Hua, Y., Richardson, R.J., M., Sturiale, A., Calapai, G., De Vico, G., Piedimonte, G.,
Schallert, T. and Xi, G. (2005) Thrombin preconditioning Salpietro, F.M. and Tomasello, F. (2002) Beneficial effects of
provides protection in a 6-hydroxydopamine Parkinson’s systemic administration of recombinant human erythropoie-
disease model. Neurosci. Lett., 373: 189–194. tin in rabbits subjected to subarachnoid hemorrhage. Proc.
Catania, M.A., Marciano, M.C., Parisi, A., Sturiale, A., Buemi, Natl. Acad. Sci. U.S.A., 99: 5627–5631.
M., Grasso, G., Squadrito, F., Caputi, A.P. and Calapai, G. Haddad, J.J. (2002) Recombinant human interleukin (IL)-1b-
(2002) Erythropoietin prevents cognition impairment in- mediated regulation of hypoxia inducible factor 1a (HIF-1a)
duced by transient brain ischemia in gerbils. Eur. J. Pharma- stabilization, nuclear translocation and activation requires an
col., 437: 147–150. antioxidant/reactive oxygen species (ROS)-sensitive mecha-
Chan, P.H. (2001) Reactive oxygen radicals in signaling and nism. Eur. Cytokine Netw., 13: 250–260.
damage in the ischemic brain. J. Cereb. Blood Flow Metab., Hellwig-Burgel, T., Rutkowski, K., Metzen, E., Fandrey, J. and
21: 2–14. Jelkmann, W. (1999) Interleukin-1beta and tumor necrosis
Chandel, N.S., McClintock, D.S., Feliciano, C.E., Wood, T.M., factor-alpha stimulate DNA binding of hypoxia-inducible
Melendez, J.A., Rodriguez, A.M. and Schumacker, P.T. factor-1. Blood, 94: 1561–1567.
(2000) Reactive oxygen species generated at mitochondrial Horowitz, M. (1994) Heat stress and heat acclimation: the cel-
complex III stabilize hypoxia inducible factor-1a during lular response-modifier of autonomic control. Integrative and
hypoxia: a mechanism of oxygen sensing. J. Biol. Chem., 275: cellular aspects of autonomic function. In: Pleschka K. and
25130–25138. Gerstberger R. (Eds.), Temperature and Osmoregulation.
Chen, Y., Constantini, S., Trembovler, V., Weinstock, M. and Libbey Eurotext, Paris, pp. 87–95.
Shohami, E. (1996) An experimental model of closed head Horowitz, M. (1998) Do cellular heat acclimation responses
injury in mice: pathophysiology, histopathology, and cogni- modulate central thermoregulatory activity? News Physiol.
tive deficits. J. Neurotrauma, 13: 557–568. Sci., 13: 218–225.
362

Horowitz, M. (2002) From molecular and cellular to integrative expression after experimental traumatic brain injury. Exp.
heat defense during exposure to chronic heat. Comp. Bio- Neurol., 153: 143–151.
chem. Physiol., 131: 475–483. Levi, E., Vivi, A., Hasin, Y., Tassini, M., Navon, G. and
Horowitz, M. (2003) Matching the heart to heat-induced cir- Horowitz, M. (1993) Heat acclimation improves cardiac
culatory load: heat-acclimatory responses. NIPS: News mechanics and metabolic performance during ischemia and
Physiol. Sci., 18: 215–221. reperfusion. J. Appl. Physiol., 75: 833–839.
Horowitz, M. (2007) Heat acclimation and cross-tolerance Liu, J., Ginis, I., Spatz, M. and Hallenbeck, J.M. (2000) Hypo-
against novel stressors: genomic-physiological linkage. In: xic preconditioning protects cultured neurons against hypo-
Sharma H.S. (Ed.), Progress in Brain Research: The Neuro- xic stress via TNF-alpha and ceramide. Am. J. Physiol. Cell
biology of Hyperthermia, Elsevier, Amsterdam, The Nether- Physiol., 278: C144–C153.
lands (in press). Luh, S. and Yang, P. (2006) Organ preconditioning: the past,
Horowitz, M., Eli-Berchoer, L., Wapinski, I., Friedman, N. and current status and related lung studies. J. Zhejiang Univ. Sci.
Kodesh, E. (2004) Stress-related genomic responses during B, 7: 331–341.
the course of heat acclimation and its association with Maloyan, A., Eli-Berchoer, L., Semenza, G.L., Gerstenblith,
ischemic-reperfusion cross-tolerance. J. Appl. Physiol., 97(4): G., Stern, M.D. and Horowitz, M. (2005) HIF-1alpha-
1496–1507. targeted pathways are activated by heat acclimation and
Horowitz, M. and Meiri, U. (1985) Altered responsiveness to contribute to acclimation-ischemic cross-tolerance in the
parasympathetic activation of submaxillary salivary gland in heart. Physiol. Genomics, 23(1): 79–88.
the heat acclimated rat. Comp. Biochem. Physiol., 80A: Maloyan, A., Palmon, A. and Horowitz, M. (1999) Heat accli-
57–60. mation increases the basal HSP72 level and alters its pro-
Hua, Y., Keep, R.F., Hoff, J.T. and Xi, G. (2003) Thrombin duction dynamics during heat stress. Am. J. Physiol., 276:
preconditioning attenuates brain edema induced by erythro- R1506–R1515.
cytes and iron. J. Cereb. Blood Flow Metab., 23: 1448–1454. Maloyan, A., Semenza, G.L., Gerstenblith, G., Stern, M. and
Ikonomidis, J.S., Tumiati, L.C., Weisel, R.D., Mickel, D.A.G. Horowitz, M. (2001) Heat-acclimation-ischemia cross tole-
and Li, R.K. (1994) Preconditioning human ventricular cardio- rance: does HIF-1 play a role? J. Mol. Cell Cardiol., 33: A72.
myocytes with brief periods of simulated ischemia. Cardiovasc. Morishita, E., Masuda, S., Nagao, M., Yasuda, Y. and Sasaki,
Res., 28: 623–634. R. (1997) Erythropoietin receptor is expressed in rat hippo-
Janoff, A. (1964) Alterations in lysosomes (intracellular en- campal and cerebral cortical neurons, and erythropoietin
zymes) during shock; effects of preconditioning (tolerance) prevents in vitro glutamate-induced neuronal death. Neuro-
and protective drugs. Int. Anesthesiol. Clin., 2: 251–259. science, 76: 105–116.
Jones, N.M. and Bergeron, M. (2001) Hypoxic preconditioning Murry, C.E., Jennings, R.B. and Reimer, K.A. (1986) Precon-
induces changes in HIF-1 target genes in neonatal rat brain. ditioning with ischemia: a delay of lethal cell injury in is-
J. Cereb. Blood Flow Metab., 21: 1105–1114. chemic myocardium. Circulation, 74: 1124–1136.
Kilic, E., Kilic, U., Wang, Y., Bassetti, C.L., Marti, H.H. and Nie, H., Xiong, L., Lao, N., Chen, S., Xu, N. and Zhu, Z. (2006)
Hermann, D.M. (2006) The phosphatidylinositol-3-kinase/ Hyperbaric oxygen preconditioning induces tolerance against
Akt pathway mediates VEGF’s neuroprotective activity and spinal cord ischemia by upregulation of antioxidant enzymes
induces blood brain barrier permeability after focal cerebral in rabbits. J. Cereb. Blood Flow Metab., 26: 666–674.
ischemia. FASEB J., 20(8): 1185–1187. Reshef, A., Sperling, O. and Zoref-Shani, E. (1996) Precondi-
Kissinger, P.T., Jonathan, B.H. and Adams, R.W. (1973) Voltam- tioning of primary rat neuronal cultures against ischemic in-
metry in brain tissue: a new neurophysiological measurement. jury: characterization of the ‘‘time window of protection.’’
Brain Res., 55: 209–213. Brain Res., 741: 252–257.
Kloog, Y., Horowitz, M., Meiri, U., Galron, R. and Avron, A. Schaller, B. (2005) Ischemic preconditioning as induction of
(1985) Regulation of submaxillary gland muscarinic recep- ischemic tolerance after transient ischemic attacks in human
tors during heat acclimation. Biochim. Biophys. Acta, 845: brain: its clinical relevance. Neurosci. Lett., 377: 206–211.
428–435. Semenza, G.L. (2004) Hydroxylation of HIF-1: oxygen sensing
Kohen, R., Beit Yannai, E., Berry, E.M. and Tirosh, O. (1999) in the molecular level. Physiology, 19: 176–182.
Overall low molecular antioxidant activity of biological fluids Shapira, Y., Shohami, E., Sidi, A., Soffer, D., Freeman, S. and
and tissues by cyclic voltammetry. Methods Enzymol., 300: Cotev, S. (1988) Experimental closed head injury in rats:
285–296. mechanical, pathophysiologic, and neurologic properties.
Kohen, R. and Nyska, A. (2002) Oxidation of biological sys- Crit. Care Med., 16: 258–265.
tems: oxidative stress phenomena, antioxidants, redox reac- Sharp, F.R., Ran, R., Lu, A., Tang, Y., Strauss, K.I., Glass, T.,
tions, and methods for their quantification. Toxicol. Pathol., Ardizzone, T. and Bernaudin, M. (2004) Hypoxic precondi-
30: 620–650. tioning protects against ischemic brain injury. NeuroRx, 1:
Kontos, H.A. (2001) Oxygen radicals in cerebral ischemia. 26–35.
Stroke, 11: 2712–2716. Shein, N.A., Doron, H., Alexandrovich, A.G., Horowitz, M.
Knoblach, S.M. and Faden, A.I. (1998) Interleukin-10 im- and Shohami, E. (2006) Heat acclimation increases pre-injury
proves outcome and alters proinflammatory cytokine anti-inflammatory capacity and leads to attenuation of
363

pro-inflammatory mRNA expression after closed head injury Siren, A.L., Knerlich, F., Poser, W., Gleiter, C.H., Bruck, W.
in mice, 5th Forum of European Neuroscience, Vienna, and Ehrenreich, H. (2001) Erythropoietin and erythropoietin
Austria. receptor in human ischemic/hypoxic brain. Acta Neuoro-
Shein, N.A., Horowitz, M., Alexandrovich, A.G., Tsenter, J. pathol. (Berl.), 101: 271–276.
and Shohami, E. (2005a) Heat acclimation increases hypoxia- Stahel, P.F., Shohami, E., Younis, F.M., Kariya, K., Otto, V.I.,
inducible factor 1alpha and erythropoietin receptor expres- Lenzlinger, P.M., Grosjean, M.B., Eugster, H.P., Trentz, O.,
sion: implication for neuroprotection after closed head injury Kossmann, T. and Morganti-Kossmann, M.C. (2000) Ex-
in mice. J. Cereb. Blood Flow Metab., 25: 1456–1465. perimental closed head injury: analysis of neurological out-
Shein, N.A., Horowitz, M., Alexandrovich, A.G., Tsenter, J. come, blood-brain barrier dysfunction, intracranial
and Shohami, E. (2005b) Akt-1 phosphorylation is increased neutrophil infiltration, and neuronal cell death in mice
and tumor necrosis factor is decreased after closed head in- deficient in genes for pro-inflammatory cytokines. J. Cereb.
jury in heat acclimated mice. J. Neurotrauma, 22(10): 1227. Blood Flow Metab., 20(2): 369–380.
Shiki, K. and Hearse, D.J. (1987) Preconditioning of ischemic Toumpoulis, I.K. and Anagnostopoulos, C.E. (2003) Early
myocardium: reperfusion induced arrhythmia. Am. J. Phy- ischemic preconditioning for spinal cord protection. Ann.
siol., 253: H1470–H1476. Thorac. Surg., 76: 1340–1341.
Shohami, E., Gallily, R., Mechoulam, R., Bass, R. and Treinin, M., Shliar, J., Jiang, H., Powell-Coffman, J.A.,
Ben-Hur, T. (1997) Cytokine production in the brain follow- Bromberg, Z. and Horowitz, M. (2003) HIF-1 is required
ing closed head injury: dexanabinol (HU-211) is a novel for heat acclimation in the nematode C. elegans. Physiol.
TNF-alpha inhibitor and an effective neuroprotectant. J. Genomics, 14: 17–24.
Neuroimmunol., 72: 169–177. Vink, R. and Van Den Heuvel, C. (2004) Recent advances in
Shohami, E., Novikov, M., Bass, R., Yamin, A. and Gallily, R. the development of multifactorial therapies for the treatment
(1994a) Closed head injury triggers early production of TNF of traumatic brain injury. Expert Opin. Investig. Drugs,
alpha and IL-6 by brain tissue. J. Cereb. Blood Flow Metab., 13(10): 1263–1274.
14(4): 615–619. Vohra, H.A. and Galinanes, M. (2006) Myocardial precondi-
Shohami, E., Novikov, M. and Horowitz, M. (1994b) Long tioning against ischemia-reperfusion apoptosis and necrosis
term exposure to heat reduces edema formation after closed in man. J. Surg. Res., 134(1): 138–144.
head injury in the rat. Acta Neurochir. Suppl. (Wien.), 60: Yatsiv, I., Grigoriadis, N., Simeonidou, C., Stahel, P.F.,
443–445. Schmidt, O.I., Alexandrovitch, A.G., Tsenter, J. and
Shohami, E., Shapira, Y., Sidi, A. and Cotev, S. (1987) Head Shohami, E. (2005) Erythropoietin is neuroprotective, im-
injury induces increased prostaglandin synthesis in rat brain. proves functional recovery, and reduces neuronal apoptosis
J. Cereb. Blood Flow Metab., 7: 58–63. and inflammation in a rodent model of experimental closed
Shohami, E., Shapira, Y., Yadid, G., Reisfeld, N. and Yedgar, head injury. FASEB J., 19: 1701–1703.
S. (1989) Brain phospholipase A2 is activated after experi- Yellon, D.M. and Dana, A. (2000) The preconditioning
mental closed head injury in the rat. J. Neurochem., 53: phenomenon: a tool for the scientist or a clinical reality.
1541–1546. Circ. Res., 87: 543–550.
This page intentionally left blank
SECTION VI

The Future of Neurotrauma: Developing Novel


Treatment Strategies
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 26

In vivo tracking of stem cells in brain and spinal cord


injury

Eva Sykova and Pavla Jendelova

Institute of Experimental Medicine ASCR, EU Centre of Excellence, Prague, Czech Republic; Center for Cell Therapy
and Tissue Repair, Charles University, Second Medical Faculty, Prague, Czech Republic; Department of Neuroscience,
Charles University, Second Medical Faculty, Prague, Czech Republic

Abstract: Cellular magnetic resonance (MR) imaging is a rapidly growing field that aims to visualize and
track cells in living organisms. Superparamagnetic iron oxide (SPIO) nanoparticles offer a sufficient signal
for T2 weighted MR images. We followed the fate of embryonic stem cells (ESCs) and bone marrow
mesenchymal stem cells (MSCs) labeled with iron oxide nanoparticles (Endorems) and human CD34+
cells labeled with magnetic MicroBeads (Miltenyi) in rats with a cortical or spinal cord lesion, models of
stroke and spinal cord injury (SCI), respectively. Cells were either grafted intracerebrally, contralaterally to
a cortical photochemical lesion, or injected intravenously. During the first post-transplantation week,
grafted MSCs or ESCs migrated to the lesion site in the cortex as well as in the spinal cord and were visible
in the lesion on MR images as a hypointensive signal, persisting for more than 30 days. In rats with an SCI,
we found an increase in functional recovery after the implantation of MSCs or a freshly prepared mono-
nuclear fraction of bone marrow cells (BMCs) or after an injection of granulocyte colony stimulating factor
(G-CSF). Morphometric measurements in the center of the lesions showed an increase in white matter
volume in cell-treated animals. Prussian blue staining confirmed a large number of iron-positive cells, and
the lesions were considerably smaller than in control animals. Additionally, we implanted hydrogels based
on poly-hydroxypropylmethacrylamide (HPMA) seeded with nanoparticle-labeled MSCs into hemisected
rat spinal cords. Hydrogels seeded with MSCs were visible on MR images as hypointense areas, and
subsequent Prussian blue histological staining confirmed positively stained cells within the hydrogels. To
obtain better results with cell labeling, new polycation-bound iron oxide superparamagnetic nanoparticles
(PC-SPIO) were developed. In comparison with Endorem, PC-SPIO demonstrated a more efficient intra-
cellular uptake into MSCs, with no decrease in cell viability. Our studies demonstrate that magnetic
resonance imaging (MRI) of grafted adult as well as ESCs labeled with iron oxide nanoparticles is a useful
method for evaluating cellular migration toward a lesion site.

Keywords: cell transplantation; iron oxide nanoparticles; magnetic resonance; MicroBeads; photochemical
lesion; scaffold; spinal cord injury

Introduction disorders of the central nervous system (CNS;


Park et al., 2002; McKay, 2004; Newman et al.,
Stem cells and progenitor cells are being explored 2004; Roitberg, 2004; Emsley et al., 2005; Fairless
in regenerative medicine for cell therapy in and Barnett, 2005; Pluchino et al., 2005; Zhao
et al., 2005; Uccelli et al., 2006). Crucial to the
Corresponding author. Tel.: +420-241062230; future success of cell transplantation in the clinical
Fax:+420-241062782; E-mail: sykova@biomed.cas.cz setting is the ability of transplanted cells to migrate

DOI: 10.1016/S0079-6123(06)61026-1 367


368

from the site of transplantation to the lesioned oxide nanoparticles, and cross-linked iron oxide
area and to survive, differentiate, and/or produce nanoparticles. Another approach for stabilizing
growth factors and cytokines for the prolonged iron oxide nanoparticles is the use of carboxylated
periods of time necessary for the patient to benefit polyamidoamine dendrimers (Bulte et al., 2001).
from their regenerative properties. Visualizing These highly soluble nanocomposites of iron ox-
transplanted cells in vivo is essential for preclini- ides and dendrimers are stable under a wide range
cal studies in rodents, and the magnetic tracking of of temperatures and pH and have an overall size of
cells appears to be a valuable tool for such studies. 20–30 nm.
Magnetic resonance (MR) imaging may also serve Iron oxide nanoparticles also require an appro-
to study cell migration to lesions, its timecourse, priate outer surface layer that induces the internal-
and how long the cells persist in the target region. ization of the particles into the cytoplasm,
Such information could help to elucidate the time so that they can be nonspecifically taken up by a
window during which the transplantation of ther- variety of cultured mammalian cells, regardless of
apeutic cells may be clinically effective, the number cell origin or animal species. In magnetodendrim-
of cells needed, and the optimal method of their ers, the highly charged carboxylated dendrimers
administration. For magnetic resonance imaging bind to multiple sites on the cell membrane, in-
(MRI) detection, cells can be labeled with MR ducing membrane bending followed by endocytosis
contrast agents in order to make them visible in (Zhang and Smith, 2000). In the case of anionic
vivo (Bulte et al., 2002; Jendelova et al., 2003, magnetic nanoparticles, the negative surface charge
2004; Sykova and Jendelova, 2005, 2006; Sykova induces an uptake three orders of magnitude
et al., 2006). greater than that of conventional dextran-coated
SPIO nanoparticles (Billotey et al., 2003). A
disadvantage of these particles, as well as of
Superparamagnetic cell labels the magnetodendrimers, is that they have not yet
been commercially developed. Another method is
Superparamagnetic iron oxide (SPIO) nanoparti- based on the mixing of a commercially available
cles were introduced as contrast agents shortly af- (U)SPIO formulation, Feridexs (Frank et al.,
ter the use of gadolinium-chelates (Mendonca Dias 2003) or Sinerems, and a commercially available
and Lauterbur, 1986; Renshaw et al., 1986; transfection agent, for example poly-L-lysine,
Pachernik et al., 2005). They are currently pre- Lipofectamin, or FugeneTM (Frank et al., 2003).
ferred as contrast agents primarily due to the fol- We have developed new polycation-bound iron
lowing properties: (a) they provide the greatest oxide superparamagnetic nanoparticles (PC-SPIO)
signal contrast change, in particular on T2 and that can be used for intracellular labeling (Horak
T2* weighted images; (b) they are composed of et al., 2006).
biodegradable iron; (c) their surface coating makes
them soluble and stable and allows for the chemi-
cal linkage of functional groups and ligands; and Cell labeling with dextran-coated SPIO
(d) they can be easily detected by both light and nanoparticles
electron microscopy (EM; Jendelova et al., 2003;
Bulte and Kraitchman, 2004). MR tracking of stem and progenitor cells in the
Iron oxide nanoparticle stabilization in order to lesioned CNS has been performed by several
prevent aggregation is most commonly accom- groups that utilized different methods of adminis-
plished by a surface coating of dextran. Dextran- tration. The first studies of imaging cell transplants
coated SPIO nanoparticles include the products labeled by superparamagnetic contrast agents in
Feridexs and Endorems, the ultrasmall super- rat brains were reported in 1992 (Norman et al.,
paramagnetic iron oxide (USPIO) nanoparticles 1992; Hawrylak et al., 1993). Rats received grafts
Combidexs and Sinerems, monocrystalline iron of fetal rat tissue prepared as cell suspensions and
369

labeled by incubation with reconstituted Sendai other sources of cells: (a) they are relatively easy to
viral envelopes containing iron oxide particles. isolate; (b) they can be used in autologous trans-
When magnetically labeled neurospheres were plantation protocols; and (c) they have already
transplanted into the ventricles of experimental been approved for the treatment of hematopoietic
allergic encephalomyelitis (EAE) rats at the peak diseases.
of their disease, the migration of glial precursors In our experiments, cell cultures of human MSCs
into white matter structures was observed on MR or rat MSCs were incubated 2–3 days in media
images (Bulte et al., 2003). Ferromagnetic-labeled containing Endorem. Nanoparticles were detected
neural progenitor cells were also transplanted into by staining for iron (Fig. 1B). Transmission elec-
the cisterna magna of rats that underwent experi- tron microscopy confirmed the presence of iron
mental MCAO (middle cerebral artery occlusion). oxide particles inside the cells, observed as large
MR images showed the migration of cells through- membrane-bound clusters scattered within the cell
out the ventricular system toward the ischemic cytoplasm (Fig. 1C). On the day that nanoparticles
brain parenchyma (Zhang et al., 2003). Olfactory were withdrawn, the efficiency of MSC labeling
ensheathing cells (OECs) labeled with magneto- (i.e., how many cells of the total number of analy-
dendrimers were implanted into the transected rat zed cells were labeled) was 50–70%.
spinal cord, and their distribution was followed in
vivo using MR imaging (Lee et al., 2004). In our
experiments (Jendelova et al., 2003, 2004), we have Tracking mesenchymal stem cells in an
shown that a suitable contrast agent for me- experimental model of stroke
senchymal stem cells (MSCs), embryonic stem
cells (ESCs), and OECs is a commercially avail- Endorem-labeled MSCs were co-labeled with
able contrast agent based on dextran-coated SPIO bromdeoxyuridine (BrdU) and grafted into rats
nanoparticles (Fig. 1A), Endorems (Guerbet, with a cortical photochemical lesion (Jendelova
France), which has also been approved as a blood et al., 2003). Rats were examined weekly for a
pool agent for human use. The contrast agent period of 3–7 weeks post-transplantation using a
Endorem can be easily incorporated by end- 4.7 T Bruker spectrometer. Single sagittal, coronal,
ocytosis, and all the cells survive and further di- and transversal images were obtained by a fast
vide in vitro. Therefore, Endorem uptake does not gradient echo sequence for localizing subsequent
need to be facilitated by a transfection agent, T2 weighted transversal images measured by a
which can damage large numbers of cells (Arbab standard turbospin echo sequence. The lesion was
et al., 2004). clearly visible on MR images 2 h after lesioning
Bone marrow-derived MSCs are adult stem cells as a hyperintense signal and remained visible
that reside within the bone marrow compartment. during the entire measurement period. No recog-
Recent data have presented evidence for their nizable hypointense signal in the lesion was
multilineage differentiation potential (Pittenger et detected during the first 2 days after implantation.
al., 1999). More recently, MSCs have been re- A decrease in the MR signal was found only at the
ported to have the ability to stimulate or partic- injection site in animals with cells injected contra-
ipate in the regeneration of diverse tissues and laterally to the lesion. One week after grafting,
organs, including the liver, myocardium, endothe- we observed a hypointense signal in the lesion,
lium, and CNS (Takahashi et al., 1999; Orlic et al., which intensified during the second and third
2001a, b; Jiang et al., 2002; Toma et al., 2002). In weeks (Fig. 1D). Histology confirmed that a large
addition, they can be genetically modified, and, number of Prussian blue-positive cells had entered
due to their migratory properties, they can serve as the lesion. No hypointense signal was found in
a carrier for drug delivery in tumor therapies other brain regions. The hypointense signal oc-
(Anderson et al., 2005). In cell therapy, bone mar- curred only in damaged areas populated with
row cells (BMCs) have some advantages over MSCs, and its intensity corresponded to Prussian
370

Fig. 1. (A) Scheme of an iron oxide nanoparticle. The contrast agent Endorem consists of a superparamagnetic Fe3O4 core coated by a
dextran shell. (B) Rat mesenchymal stem cells (MSCs) in culture labeled with superparamagnetic nanoparticles (blue dots). The cell
nuclei are counterstained with hematoxylin. (C) Transmission electron microphotograph of a cluster of iron nanoparticles surrounded
by a cell membrane. (D) An implant of Endorem-BrdU co-labeled MSCs (in the hemisphere contralateral to the lesion, arrow) and the
lesion (arrowhead) itself are both hypointense in an MR image taken 2 weeks after implantation. (E), (F) A hypointense signal (black
arrowhead) was observed in the lesion 6 days after the intravenous (i.v.) injection of Endorem-BrdU co-labeled MSCs, becoming more
hypointense and persisting for 47 days (F). (G, H) Massive invasion of rat MSCs (Prussian blue staining counterstained with
hematoxylin) into a photochemical lesion 7 weeks after i.v. injection into a rat with a photochemical lesion. (I) Serial section stained for
BrdU, 7 weeks after i.v. injection. (Adapted with permission from Jendelova et al. (2003).)

blue or BrdU staining. Only a few (less than 3%) the lesion, which densely populated the borders of
of the MSCs that migrated into the lesion ex- the lesion (Figs. 1G–I).
pressed the neuronal marker NeuN when tested 28 Only a few cells weakly stained for Prussian blue
days post-implantation. No GFAP-positive cells were found in photochemical lesions without any
were found in the lesion. implanted cells. The staining represents iron,
After the intravenous injection of MSCs, we which most likely originated in hemorrhages and
found a similar hypointense MR signal in the le- iron degradation products released from iron-
sion site. The signal was observed 6 days after cell containingproteins (such as hemoglobin, ferritin,
infusion and persisted for 7 weeks (Figs. 1E, F). and hemosiderin) and phagocytized by microglia/
Prussian blue and anti-BrdU staining confirmed macrophages. We did not observe any BrdU-
the presence of iron oxide-BrdU co-labeled cells in positive cells in the brains of nongrafted animals.
371

Tracking embryonic stem cells in an experimental very similar MR images to those obtained after the
model of stroke implantation of MSCs. In rats with a photochemi-
cal lesion and contralaterally injected cells, the cell
ESCs are pluripotent cell lines with a capacity for implants were visible as a hypointense area at the
self-renewal and broad differentiation plasticity. injection site. Two weeks after grafting, a hypoin-
They are derived from embryos and can be prop- tense signal was also observed in the corpus callo-
agated as a homogeneous, uncommitted cell pop- sum and in the lesion (Fig. 2B). At the same time,
ulation for an almost unlimited period of time histology showed that a large number of Prussian
without losing their pluripotency and their stable blue-positive cells had entered the lesion. Many
karyotype. Mouse ES cell-derived glial precursors, labeled cells were also detected in the corpus callo-
transplanted into rats with myelin disease, inter- sum, suggesting a migration from the contralateral
acted with the host neurons to produce myelin in hemisphere toward the lesion (Fig. 2C). When the
the brain and spinal cord (Brustle et al., 1999). Re- ESCs were injected intravenously into lesioned
tinoic acid-treated embryoid bodies from rats, we found a hypointense MR signal only at the
mouse ESCs, when transplanted into rat spinal site of the lesion. However, the extent of the dif-
cord 9 days after traumatic injury, differentiated ferentiation of eGFP ESCs labeled with nanopar-
into astrocytes, oligodendrocytes, and neurons and ticles, and found in the lesion, was much greater
promoted recovery (McDonald et al., 1999). than with grafted MSCs. Of all the eGFP ESCs
Bjorklund et al. (2002) reported that undifferenti- containing nanoparticles found in the lesion, 70%
ated mouse ESCs can become dopamine-producing of them proved to be astrocytes, very few (less than
neurons in the brain in a rat model of Parkinson’s 1%) were oligodendrocytes, and 5% of nanopar-
disease and can lead to partial functional recovery. ticle-labeled eGFP ESCs had differentiated into
We used mouse ESCs transfected with the pEG- neurons (Jendelova et al., 2004). Electron micro-
FP-C1 vector (Pachernik et al., 2005) and labeled scopy revealed mature astrocytes, neurons, and
with SPIO nanoparticles (Jendelova et al., 2004). oligodendrocytes in the lesion site containing iron
Since undifferentiated ESCs may form embryonic oxide nanoparticles, providing strong supporting
tumors when grafted into a host animal, we in- evidence that these cells differentiated in the lesion
duced neural differentiation by culturing enhanced from implanted embryonic cells (Figs. 2E, F).
green fluorescent protein (eGFP) ESCs in serum
containing DMEM (Dulbecco’s modified Eagle
medium)/F12 without leukemia inhibitory factor Tracking mesenchymal stem cells in a rat model of
(LIF) for 2 days and then transferred the cells into spinal cord injury
serum-free media supplemented with insulin, trans-
ferrin, selenium, and fibronectin for further culture Evaluating the effect of different BMC populations
(Pachernik et al., 2005). Feeder-free eGFP ESCs on morphological and functional recovery after spi-
were labeled with Endorem (112.4 mg/ml) during nal cord injury (SCI) is an important aspect of our
three passages (Fig. 2A). Cell counting of Prussian preclinical research. In rats with a balloon-induced
blue stained cells in suspension revealed that the spinal cord compression lesion, we studied the effect
labeling efficiency was 80%. Cells were trans- of an intravenous injection of Endorem-labeled
planted intracerebrally or intravenously on the 8th nonhematopoietic MSCs (Urdzikova et al., 2006).
day of differentiation into adult Wistar rats with a The results obtained were compared with those
cortical photochemical lesion 1 week after lesioning following the implantation of a freshly isolated
(Jendelova et al., 2004), and were detected by mononuclear fraction of bone marrow containing
staining for iron and by GFP fluorescence. When stromal cells, hematopoietic and nonhemato-
we implanted the ESCs 7 days post-lesion, we poietic stem and precursor cells, and lymphocytes
found a massive migration of Endorem-labeled (BMCs). Furthermore, we studied the effect of
GFP-positive cells into the lesion site regardless of granulocyte colony stimulating factor (G-CSF) mo-
the route of administration (Fig. 2D). We observed bilization of endogenous BMCs containing mainly
372

Fig. 2. (A) Trypsinized nanoparticle-labeled suspensions of ESCs after the third passage. (B) The cell implant (in the hemisphere
contralateral to the lesion) and the lesion are hypointense in MR images 2 weeks after implantation. A hypointensive signal is also
found in the corpus callosum. (C) Dense Prussian blue staining of the injection site in the contralateral hemisphere, the corpus
callosum, and the photochemical lesion, 4 weeks after grafting. (D) Invasion of GFP-labeled cells showing GFP-positive ESCs in the
lesion 4 weeks after the i.v. injection of eGFP ESCs (serial section to the slice shown in Fig. 2C). (E) Astrocyte from the brain tissue of
a rat with nanoparticle-labeled eGFP ESCs implanted contralaterally to the lesion; the nanoparticles are visible in small dense clusters
in the cell cytoplasm (arrowhead). (F) Neuron with nanoparticles marginated to the nuclear membrane. (Adapted with permission
from Jendelova et al. (2004).)

hematopoietic stem cells, along with progenitor after implantation using a standard whole body
cells and lymphocytes (Sasaki et al., 2001; Akiyama resonator. Functional status was assessed weekly
et al., 2002). for 5 weeks after spinal cord lesioning, using the
MSCs labeled with Endorem were injected in- Basso-Beattie-Bresnehan (BBB) locomotor rating
travenously into the femoral vein 1 week after le- score and the plantar test. Our data indicated that
sioning. MR images were taken ex vivo 4 weeks lesioned animals with grafted MSCs had higher
373

locomotor scores as indicated by their BBB scores staining confirmed a large number of positive
and showed better responses in sensitivity testing cells present in the lesion site (Fig. 3C). On serial
using the plantar test than did control animals. sections, Prussian blue-positive cells corresponded to
Particularly, the plantar test showed the recovery cells labeled with PKH 26, as seen in (Figs. 3D, E).
of sensitivity in the hind limbs. On MR images we To exclude the possibility that free nanopar-
observed the lesion as an inhomogeneity in the ticles were taken up by macrophages, we per-
tissue texture with a hyperintense signal only in the formed immunohistochemical and Prussian blue
area of the SCI (Fig. 3A). Images of longitudinal staining to co-localize iron in activated microglia/
spinal cord sections from animals grafted with macrophages. Although we found strong positive
nanoparticle-labeled MSCs showed the lesion as a staining for macrophages in the lesions using
dark hypointense area (Fig. 3B). Prussian blue ED1 antibody, Prussian blue staining did not,

Fig. 3. (A) Longitudinal MR image of a spinal cord lesion 5 weeks after induction. The formation of the lesion cavity is visible as a
strong hyperintensive signal (arrow). (B) Longitudinal MR image 4 weeks after MSC grafting. The lesion with nanoparticle-labeled
cells is visible as a dark hypointensive area (arrow). (C) Prussian blue staining of a lesion populated with nanoparticle-labeled MSCs
(blue dots). (D) Magnified region from Fig. C. (E) Intravenously injected MSCs were also detectable in the spinal cord lesion using the
membrane florescent dye PKH 26. Serial section to D. (F) Immunostaining with ED-1 antibody revealed a number of macrophages in
the lesion (white arrows). The majority of Prussian blue-positive cells did not co-localize with ED-1 staining; however, a few macro-
phages were Prussian blue-positive (black arrowheads). (Adapted with permission from Urdzikova et al. (2006).)
374

for the most part, co-localize with the ED1 staining SCF also had higher scores in BBB testing than
(Fig. 3F). did control animals and also showed a faster
Morphometric measurements of the spared recovery of sensitivity in their hind limbs using the
white and gray matter were performed in the cen- plantar test. However, the functional improvement
ter of the lesions. The spared cross-sectional area was more pronounced in MSC-treated rats
of the white matter, as well as the volume of spared (Figs. 4A, B). Morphological measurements of
white matter, was significantly greater in MSC- the spared cross-sectional area of the white matter
treated animals. The spared cross-sectional area of showed a statistically significant increase in groups
the gray matter was also significantly larger in treated with BMCs or G-SCF, compared to con-
MSC-treated animals. In further studies, lesioned trols, cranially to the lesion center; a statistically
animals grafted with BMCs or injected with G- significant increase in the volume of spared white

Fig. 4. (A) Behavioral open field BBB motor scores of MSC, BMC, and G-CSF treated rats were significantly higher than those of
saline-injected (control) animals, 14, 21, 28, and 35 days after SCI (po0.05). The scores of the MSC, BMC, and G-CSF treated animals
were not significantly different from one another at any time point. (B) Time course of the animals’ response to radiant heat measured
with the plantar test in treated and saline-injected rats. In all treated rats, the latency times decreased as their recovery progressed. The
most pronounced effect was seen in MSC treated rats. The latency time in the saline-injected (control) rats did not change during the 35
days survival period. Data are averaged between right and left hind limbs and expressed as mean7SEM. *po0.05 compared to control
group. (Adapted with permission from Urdzikova et al. (2006).)
375

Fig. 5. (A) The total volume of the white matter in 11 mm long segments of the spinal lesion in MSCs, BMCs, and G-CSF-treated and
saline-injected animals. (B) The total volume of gray matter in 11 mm long segments of the spinal lesion in MSCs, BMCs, and G-CSF-
treated and saline-injected animals. No statistically significant differences were observed between treated and saline-injected animals.
All data are presented as means7SEM. *po0.05 compared to saline-injected (control) rats. (Adapted with permission from Urdzikova
et al. (2006).)

matter was observed in the BMC-treated group. therefore developed new polycation-bound SPIO
The spared white matter volume in the G-CSF- nanoparticles (Fig. 6A; Horak et al., 2006). We
treated rats was also increased, but the increase did compared the influence of both types of nanopar-
not reach statistical significance (Fig. 5). ticles on cell viability and labeling efficiency in rat
and human MSCs. The PC-SPIO nanoparticle sus-
pension was used at a much lower iron concentra-
Stem cell labeling with polycation-bound tion per milliliter of culture media (15.4 mg/ml), and
nanoparticles the cells were incubated with PC-SPIO for 3 days.
The results were compared with Endorem labeling
In all the above-described experiments, dextran- (Tables 1 and 2). The measurements were per-
coated SPIO nanoparticles (the contrast agent formed on the day of withdrawal of the nanopar-
Endorem) were used as an intracellular label. ticles (day 3) and 4 days later (day 7).
However, the efficiency of the cell labeling was Labeling cells with PC-SPIO nanoparticles was
maximally 70%. In addition, on the day of particle more efficient than labeling with Endorem, i.e.,
withdrawal a decrease in cell viability (using the more cells from the total number of analyzed cells
WST1 colorimetric assay) was observed. We were labeled with PC-SPIO nanoparticles than
376

Fig. 6. (A) Schematic illustration of a polycation-bound superparamagnetic iron oxide nanoparticle. (B) Transmission electron
photomicrograph of MSCs labeled with PC-SPIO showing clusters of iron nanoparticles (arrows) in the cell cytoplasm, which are not
surrounded by a cell membrane. (C) Prussian blue staining of MSCs in culture labeled with PC-SPIO. (D, E) Axial and coronal (E) MR
images of a rat brain with 1000 cells labeled with PC-SPIO implanted to the left hemisphere and 1000 Endorem-labeled cells implanted
to the right hemisphere. MR images were taken 3 days after implantation.

Table 1. The percentage of cells labeled with nanoparticles af- In Endorem-labeled human MSCs, on day 3 the cell
ter 3 days incubation viability dropped to 50%; however, within 4 days it
Endorem-labeled PC-SPIO-labeled recovered to 87%. This transient decrease in cell
cells (%) cells (%) viability might be due to the relatively high con-
centration of iron in the culture media (112.4 g/ml),
Rat MSCs 59.3 92.2
Human MSCs 65.2 87.5
to which some cell cultures might be sensitive. The
average amount of iron present in rat MSCs
was determined by spectrophotometry after min-
with Endorem (Table 1; Fig. 6C). In Table 2 the eralization of iron-labeled cell suspensions. In PC-
cell viability of labeled cells is expressed as a per- SPIO-labeled cells, even though the concentration
centage of the number of viable unlabeled cells of iron in the culture media was 10 times lower
(taken as the control value and defined as 100%). (15.4 mg/ml culture media), the average amount of
377

Table 2. Cell viability (in %) after incubation with nanopar- stronger contrast change in the signal than do
ticles (day 3) and 4 days after withdrawal (day 7)
Endorem-labeled cells (Figs. 6D, E). Labeling with
Endorem-labeled PC-SPIO-labeled PC-SPIO thus enables the detection of a lower
cells (%) cells (%) number of cells in the tissue.
Day 3 Day 7 Day 3 Day 7

Rat MSCs 68.9 72.9 92.8 89.7 Cell labeling with magnetic MicroBeads
Human MSCs 49.0 87.0 97.2 98.9
The disadvantage of an intracellular label is that it
can affect cell metabolism and subsequently cell
iron was 38 pg/cell, while in Endorem-labeled cells viability. In addition, these labels are rather non-
only 17 pg/cell. Transmission electron photomicro- specific; they can be loaded by virtually any cell
graphs confirmed that more PC-SPIO nanoparticles present in the medium. The possibility of labeling
were taken up by the rat MSCs than were Endo- only selected cell types would therefore be very
rem nanoparticles (Fig. 6B). Moreover, Endorem useful. In addition, cells labeled and separated by
was observed as membrane-bound clusters within means of immunomagnetic selection would not
the cell cytoplasm, indicating an endocytotic proc- require in vitro culturing, since the label is at-
ess of nanoparticle uptake. The higher cellular tached during separation. This would also allow
uptake of PC-SPIO nanoparticles is possible due the immediate clinical use of labeled cells. The first
to the interaction of the surface coating with the experiments using contrast agents bound to an
negatively charged cell surface and subsequent antibody that can specifically bind to a single cell
endosomolytic uptake. Nanoparticles are trans- type were performed by Bulte et al. (1992). They
ported in endosomes and finally fused with lyso- described experiments with human lymphocytes
somes, a process during which the vesicle labeled with biotinylated anti-lymphocyte-directed
membranes disappear. monoclonal antibodies (mabs), to which streptavi-
To check the sensitivity of the MRI technique din and subsequently biotinylated dextran-mag-
and to mimic signal behavior in CNS tissue, sus- netite particles were coupled. We tested a new
pensions of unlabeled cells and cells labeled with type of specific cell labeling using commercially
PC-SPIO were imaged in vitro. MR images of available cell isolation kits for the magnetic sep-
1.7% gelatin phantoms containing iron-labeled aration of CD34+ cells. CD34+ cells are known as
MSCs were obtained using a 4.7 T Bruker spectro- hematopoietic progenitor cells. The cells were sep-
meter equipped with a standard resonator coil. arated by means of immunomagnetic selection
Even the sample containing the lowest concentra- with anti-CD34 antibodies. For sorting, a SPIO
tion (200 cells/ml, which corresponds on average to core coated with a polysaccharide that is linked to
2 cells per image voxel) provided visible contrast an antibody is bound to the respective cell
compared to a control phantom containing the (Fig. 7A). The size of the label is comparable to
same number of unlabeled cells. A similar set of commonly used superparamagnetic MR contrast
experiments was performed in earlier work agents; thus it can provide sufficient contrast on
(Jendelova et al., 2003), in which MR images of MR images.
gelatin phantoms showed a hypointense signal at Human CD34+ cells from peripheral blood
concentrations above 625 labeled cells/ml. were selected by CliniMACS CD34 Selection
For in vivo imaging, rats were examined 3 days Technology (Miltenyi). On EM images, we deter-
post-transplantation in an MR imager. Figure 6 mined that after cryopreservation, the nanoparti-
shows that PC-SPIO labeled cells are also clearly cles remained bound to the cell surface, and we
recognizable in vivo. The iron oxide-labeled cell observed several iron labels attached to the cell
implants are visible as a hypointense area at the surface (Fig. 7B). Purified CD34+ cells were im-
injection site. Cells labeled by PC-SPIO provide planted intracerebrally into rats with a cortical
378

Fig. 7. (A) Scheme of MicroBeads. A superparamagnetic iron oxide core coated with a polysaccharide is linked to an antibody, which
in turn is bound to a cell via an antigen–antibody complex. (B) Transmission electron microphotograph of MicroBeads binding to the
cell surface. (C) Human cells (positive staining for human nuclei) found in the lesion 4 weeks after the grafting of CD34+ cells. (D)
Staining with anti-CD34 revealed CD34+ cells in the subventricular zone. (Adapted with permission from Jendelova et al. (2005).)

photochemical lesion, contralaterally to the lesion hypointensity of the implant slightly decreased
(Jendelova et al., 2005; Sykova and Jendelova, during the first week and then remained without
2006). The average iron content per cell, deter- significant changes for the entire measurement pe-
mined by spectrometry, was 0.275 pg. This value is riod (4 weeks). During the second week, we ob-
lower by two orders of magnitude than in the served a weak hypointense signal in the lesion that
case of cell labeling using Endorem or PC-SPIO, persisted for the next 2 weeks. Prussian blue and
which enter the cell (17 or 38 pg, respectively); anti-human nuclei staining (Fig. 7C) confirmed
nevertheless, it still provides sufficient MR con- the presence of magnetically labeled human cells
trast (Jendelova et al., 2005). in the corpus callosum and in the lesion. CD34+
The cells were detected as a hypointense spot cells were detected not only in the corpus callosum
on T2 weighted images 24 h after grafting. The and in the lesion, but also in the subventricular
379

zone (Fig. 7D). Finally, human DNA was detected therefore serve as an alternative to the conven-
in the lesioned brain tissue using polymerase tional grafting of dissociated cells, benefiting from
chain reaction (PCR), confirming the presence of advances in surface chemistry and the cell–cell or
human cells. cell–matrix interactions that occur during deve-
lopment or regeneration.

Labeling of stem cells in polymer hydrogels as stem


cell carriers Discussion and conclusion

In the case of large lesions, cells alone are not able For clinical studies it is obvious that traditional
to repair the injury. It is necessary to bridge the histopathological methods for cell detection are not
gap left by the lost cell population in order to sufficient to inform us about the migration and fate
provide support for tissue restoration, reduce the of the grafted cells in the host tissue. Because of its
glial scar, and create a permissive environment for high spatial resolution, MR imaging is suitable for
cellular ingrowth and for the diffusion of neuro- imaging the distribution of magnetically labeled
active molecules. To bridge such a lesion, we have cells. Labeling methods that use the internalization
used biocompatible polymer hydrogels based on of iron oxide nanoparticles have some limitations.
poly-hydroxypropylmethacrylamide (HPMA) in To label cells with commercially available contrast
combination with stem cell grafting. Before trans- agents, such as Endorem, a relatively high concen-
plantation into a lesion, the hydrogels were seeded tration of iron in the culture media is necessary
in vitro with MSCs. In this case the hydrogels form (Jendelova et al., 2003, 2004). This may cause a
an inert environment, allowing for the free diffu- transient drop in cell viability, which can be de-
sion of intrinsic growth factors, in which the cells pendent on the type of labeled cell. For improved
start to differentiate and migrate. The inert and uptake of magnetic nanoparticles, the surface of
biocompatible environment of the hydrogels also the contrast agent needs to be optimized, so that it
provides an adequate standard background for can induce the internalization of the particles into
MR imaging of the cells (Sykova and Jendelova, the cytoplasm. One approach is to use internalizing
2005; Sykova et al., 2006). We employed these mabs. The mouse anti-Tfr mab OX-26 induces the
cell-polymer constructs in order to facilitate the internalization of Tft upon binding. Nanoparticles
regeneration of injured spinal cord (Lesny et al., have been conjugated to OX-26 to deliver them to
2006). The right half of a spinal cord segment was cells by receptor-mediated endocytosis (Bulte et al.,
removed by hemisection, and a block of HPMA 1999). A disadvantage of the use of internalizing
hydrogel seeded with Endorem-labeled MSCs was mabs, however, is that they are species-specific, and
inserted (Fig. 8A; Sykova and Jendelova, 2005). a newly synthesized antibody is required when per-
Six weeks after implantation, the hydrogel had forming studies in a different animal. In addition,
formed a continuous bridge between the hem- for eventual clinical use, there will be regulatory
isected spinal segments, reestablishing the anatom- issues regarding the use of a xenogeneic (i.e.,
ical continuity of the tissue. The hydrogel mouse) protein. Recently, a combination of dext-
was visible on MR images as a hypointense area ran-coated SPIO nanoparticles with a transfection
(Fig. 8B), and Prussian blue staining confirmed agent has been used (Kalish et al., 2003). When the
positively stained cells within the hydrogel complexes are added to a cell culture, the transfect-
(Fig. 8C). Immunostaining for blood vessels ion agent effectively transports the nanoparticles
(Reca-1, Abcam, UK) confirmed neovascularizat- into the cells through electrostatic interactions.
ion of the hydrogel implant (Fig. 8D). Staining for However, each combination of transfection agent
neurofilaments (NF160, Sigma, St. Louis, USA) and iron oxide nanoparticle has to be carefully tit-
showed axonal ingrowth into the hydrogel (Fig. rated and optimized for different cell cultures, since
8E; Sykova and Jendelova, 2005; Sykova et al., lower concentrations may result in insufficient cel-
2006). Hydrogels seeded with stem cells may lular uptake, whereas higher concentrations may
380

Fig. 8. (A) Endorem-labeled cells seeded into a hydrogel. (B) On MR images 6 weeks after implantation, the hydrogel was visible as a
hypointensive area. (C) Prussian blue staining confirmed the presence of Endorem-labeled cells in the hydrogel. (D) Neovascularization
of the implant (Reca-1). (E) Ingrowth of NF160-positive axons into the gel.
381

induce the precipitation of complexes or may be agents would enable us to follow the migration of
toxic to the cells. In addition, although it has been such cells when transplanted into humans, establish
reported that a poly-L-lysin-Feridex labeling com- the optimal number of transplanted cells, define
plex does not appear to affect the viability or pro- therapeutic windows, and monitor cell growth and
liferation of human MSCs, it was found that their possible side effects (malignancies). Currently, the
differentiation into chondrocytes was markedly in- described immunolabeling of specific cell types with
hibited (Kostura et al., 2004), while adipogenic and clinically approved MicroBeads may help to eluci-
osteogenic differentiation were not affected. PC- date the fate of implanted stem cells and evaluate
SPIO combine a low concentration of iron in the the effect of cell therapy in patients with various
cell culture media with the high efficiency of diseases of the brain or SCI.
transfection agents and thus have a broad poten-
tial for in vivo studies. However, for any intracel-
lular label, detailed studies examining the possible Abbreviations
biological side effects, which may vary among dif-
ferent cell types, are necessary. BBB Basso-Beattie-Bresnehan
On the other hand, particles that do not inter- BMCs bone marrow cells
nalize do not cause any metabolic alternations and BrdU bromdeoxyuridine
do not have any influence on cell viability or cell CNS central nervous system
proliferation (Jendelova et al., 2005). Their disad- DMEM Dulbecco’s modified Eagle medium
vantage is that particles that do not internalize and eGFP enhanced green fluorescent protein
thus stay attached to the outer cell membrane are EM electron microscopy
more likely to interfere with cell surface interac- ESCs embryonic stem cells
tions (including cell homing into tissues), may de- G-CSF granulocyte colony stimulating fac-
tach easily from the membrane, or may be tor
transferred to other cells. HPMA hydroxypropylmethacrylamide
MR tracking may serve in experimental models LIF leukemia inhibitory factor
to study how certain lesions target cell migration, Mab monoclonal antibody
at what speed the cells migrate, and for how long MCAO middle cerebral artery occlusion
they persist in the target organ. High contrast ef- MR magnetic resonance
fects on MR images are easily detected within an MRI magnetic resonance imaging
experimental time frame of 1–2 h per animal, MSCs mesenchymal stem cells
which is ideal for short and repetitive in vivo MRI. OECs olfactory ensheathing cells
In lesioned tissue, hemorrhage products give rise PCR polymerase chain reaction
to Prussian blue-positive deposits that are difficult PC-SPIO polycation-bound iron oxide super-
to distinguish from iron-containing nanoparticles paramagnetic nanoparticles
(Urdzikova et al., 2006). The hemorrhage degra- SCI spinal cord injury
dation products may also be partially localized to SPIO superparamagnetic iron oxide
macrophages because macrophages constitute the USPIO ultrasmall superparamagnetic iron
major cellular pathway for the redistribution of oxide
iron in mammals. Furthermore, hemorrhage con-
tributes to T2 weighted hypointensity, thus inter-
fering with the detection of labeled cells and
complicating the interpretation of MR images. Acknowledgment
Therefore, it is important to determine whether
cell labels remain co-localized with cell trans- This work was supported by grants from the
plants, especially under pathological conditions. Academy of Sciences of the Czech Republic
With proper attention to the limitations desc- AV0Z50390512, the Ministry of Education,
ribed above, labeling cells with superparamagnetic Youth, and Sports of the Czech Republic
382

1M0021620803, the National Grant Agency of the and myelination. Proc. Natl. Acad. Sci. U.S.A., 96:
Czech Republic GACR 309/06/1594, and the EC- 15256–15261.
FP6 project DiMI: LSHB-CT-2005-512146. Bulte, J.W., Zhang, S.C., van Gelderen, P., Herynek, V.,
Jordan, E.K., Janssen, C.H., Duncan, I.D. and Frank, J.A.
(2002) Magnetically labeled glial cells as cellular MR contrast
agents. Acad. Radiol., 9(Suppl 1): S148–S150.
Emsley, J.G., Mitchell, B.D., Kempermann, G. and Macklis,
References J.D. (2005) Adult neurogenesis and repair of the adult CNS
with neural progenitors, precursors, and stem cells. Prog.
Akiyama, Y., Radtke, C., Honmou, O. and Kocsis, J.D. (2002) Neurobiol., 75(5): 321–341.
Remyelination of the spinal cord following intravenous de- Fairless, R. and Barnett, S.C. (2005) Olfactory ensheathing
livery of bone marrow cells. Glia, 39(3): 229–236. cells: their role in central nervous system repair. Int. J. Bio-
Anderson, S.A., Glod, J., Arbab, A.S., Noel, M., Ashari, P., chem. Cell. Biol., 37(4): 693–699.
Fine, H.A. and Frank, J.A. (2005) Noninvasive MR imaging Frank, J.A., Miller, B.R., Arbab, A.S., Zywicke, H.A., Jordan,
of magnetically labeled stem cells to directly identify ne- E.K., Lewis, B.K., Bryant Jr., L.H. and Bulte, J.W. (2003)
ovasculature in a glioma model. Blood, 105(1): 420–425. Clinically applicable labeling of mammalian and stem cells by
Arbab, A.S., Yocum, G.T., Wilson, L.B., Parwana, A., Jordan, combining superparamagnetic iron oxides and transfection
E.K., Kalish, H. and Frank, J.A. (2004) Comparison of agents. Radiology, 228(2): 480–487.
transfection agents in forming complexes with ferumoxides, Hawrylak, N., Ghosh, P., Broadus, J., Schlueter, C., Green-
cell labeling efficiency, and cellular viability. Mol. Imaging, ough, W.T. and Lauterbur, P.C. (1993) Nuclear magnetic
3(1): 24–32. resonance (NMR) imaging of iron oxide-labeled neural
Billotey, C., Wilhelm, C., Devaud, M., Bacri, J.C., Bittoun, J. transplants. Exp. Neurol., 121: 81–92.
and Gazeau, F. (2003) Cell internalization of anionic ma- Horak, D., Sykova, E., Babic, M., Jendelova, P. and Hajek, M.
ghemite nanoparticles: quantitative effect on magnetic reso- (2006) Superparamagnetic iron oxide nanoparticles with
nance imaging. Magn. Reson. Med., 49(4): 646–654. modified surface and their use as probes for stem cell labe-
Bjorklund, L.M., Sanchez-Pernaute, R., Chung, S., Andersson, ling. Patent application no: PV 1006–120, Czech Republic.
T., Chen, I.Y., McNaught, K.S., Brownell, A.L., Jenkins, Jendelova, P., Herynek, V., De Croos, J., Glogarova, K.,
B.G., Wahlestedt, C., Kim, K.S. and Isacson, O. (2002) Em- Andersson, B., Hajek, M. and Sykova, E. (2003) Imaging
bryonic stem cells develop into functional dopaminergic neu- the fate of implanted bone marrow stromal cells labeled with
rons after transplantation in a Parkinson rat model. Proc. superparamagnetic nanoparticles. Magn. Reson. Med., 50:
Natl. Acad. Sci. U.S.A., 99(4): 2344–2349. 767–776.
Brustle, O., Jones, K.N., Learish, R.D., Karram, K., Choudh- Jendelova, P., Herynek, V., Urdzikova, L., Glogarova, K.,
ary, K., Wiestler, O.D., Duncan, I.D. and McKay, R.D. Kroupova, J., Bryja, V., Andersson, B., Burian, M., Hajek,
(1999) Embryonic stem cell-derived glial precursors: a source M. and Sykova, E. (2004) MR tracking of transplanted bone
of myelinating transplants. Science, 285(5428): 754–756. marrow and embryonic stem cells labeled by iron oxide
Bulte, J.W., Ben-Hur, T., Miller, B.R., Mizrachi-Kol, R., nanoparticles in rat brain and spinal cord. J. Neurosci. Res.,
Einstein, O., Reinhartz, E., Zywicke, H.A., Douglas, T. and 76(2): 232–243.
Frank, J.A. (2003) MR microscopy of magnetically labeled Jendelova, P., Herynek, V., Urdzikova, L., Glogarova, K., Rah-
neurospheres transplanted into the Lewis EAE rat brain. matova, S., Fales, I., Andersson, B., Prochazka, P., Zamecnik,
Magn. Reson. Med., 50(1): 201–205. J., Eckschlager, T., Kobylka, P., Hajek, M. and Sykova, E.
Bulte, J.W., Douglas, T., Witwer, B., Zhang, S.C., Strable, E., (2005) MR tracking of human CD34+ progenitor cells sep-
Lewis, B.K., Zywicke, H., Miller, B., van Gelderen, P., arated by means of immunomagnetic selection and trans-
Moskowitz, B.M., Duncan, I.D. and Frank, J.A. (2001) Mag- planted into injured rat brain. Cell Transplant., 14: 173–182.
netodendrimers allow endosomal magnetic labeling and in Jiang, Y., Jahagirdar, B.N., Reinhardt, R.L., Schwartz, R.E.,
vivo tracking of stem cells. Nat. Biotechnol., 19: 1141–1147. Keene, C.D., Ortiz-Gonzalez, X.R., Reyes, M., Lenvik, T.,
Bulte, J.W., Hoekstra, Y., Kamman, R.L., Magin, R.L., Webb, Lund, T., Blackstad, M., Du, J., Aldrich, S., Lisberg, A.,
A.G., Briggs, R.W., Go, K.G., Hulstaert, C.E., Miltenyi, S., Low, W.C., Largaespada, D.A. and Verfaillie, C.M. (2002)
The, T.H., et al. (1992) Specific MR imaging of human Pluripotency of mesenchymal stem cells derived from adult
lymphocytes by monoclonal antibody-guided dextran- marrow. Nature, 418(6893): 41–49.
magnetite particles. Magn. Reson. Med., 25(1): 148–157. Kalish, H., Arbab, A.S., Miller, B.R., Lewis, B.K., Zywicke,
Bulte, J.W. and Kraitchman, D.L. (2004) Iron oxide MR con- H.A., Bulte, J.W., Bryant Jr., L.H. and Frank, J.A. (2003)
trast agents for molecular and cellular imaging. NMR Bio- Combination of transfection agents and magnetic resonance
med., 17(7): 484–499. contrast agents for cellular imaging: relationship between re-
Bulte, J.W., Zhang, S.C., van Gelderen, P., Herynek, V., laxivities, electrostatic forces, and chemical composition.
Jordan, E.K., Duncan, I.D. and Frank, J.A. (1999) Neuro- Magn. Reson. Med., 50(2): 275–282.
transplantation of magnetically labeled oligodendrocyte Kostura, L., Kraitchman, D.L., Mackay, A.M., Pittenger, M.F.
progenitors: magnetic resonance tracking of cell migration and Bulte, J.W. (2004) Feridex labeling of mesenchymal stem
383

cells inhibits chondrogenesis but not adipogenesis or osteo- Pluchino, S., Zanotti, L., Deleidi, M. and Martino, G. (2005)
genesis. NMR Biomed., 17(7): 513–517. Neural stem cells and their use as therapeutic tool in neu-
Lee, I.H., Bulte, J.W., Schweinhardt, P., Douglas, T., Trifu- rological disorders. Brain Res. Brain Res. Rev., 48(2):
novski, A., Hofstetter, C., Olson, L. and Spenger, C. (2004) 211–219.
In vivo magnetic resonance tracking of olfactory ensheathing Renshaw, P.F., Owen, C.S., McLaughlin, A.C., Frey, T.G. and
glia grafted into the rat spinal cord. Exp. Neurol., 187(2): Leigh Jr., J.S. (1986) Ferromagnetic contrast agents: a new
509–516. approach. Magn. Reson. Med., 3(2): 217–225.
Lesny, P., Pradny, M., Jendelova, P., Michalek, J., Vacik, J. Roitberg, B. (2004) Transplantation for stroke. Neurol. Res.,
and Sykova, E. (2006) Macroporous hydrogels based on 26(3): 256–264.
2-hydroxyethyl methacrylate. Part 4: growth of rat bone Sasaki, M., Honmou, O., Akiyama, Y., Uede, T., Hashi, K. and
marrow stromal cells in three-dimensional hydrogels with Kocsis, J.D. (2001) Transplantation of an acutely isolated
positive and negative surface charges and in polyelectrolyte bone marrow fraction repairs demyelinated adult rat spinal
complexes. J. Mater. Sci. Mater. Med., 17: 829–833. cord axons. Glia, 35: 26–34.
McDonald, J.W., Liu, X.Z., Qu, Y., Liu, S., Mickey, S.K., Sykova, E. and Jendelova, P. (2005) Magnetic resonance
Turetsky, D., Gottlieb, D.I. and Choi, D.W. (1999) Trans- tracking of implanted adult and embryonic stem cells in in-
planted embryonic stem cells survive, differentiate and pro- jured brain and spinal cord. Ann. N.Y. Acad. Sci., 1049:
mote recovery in injured rat spinal cord. Nat. Med., 5(12): 146–160.
1410–1412. Sykova, E. and Jendelova, P. (2006) Magnetic resonance track-
McKay, R.D. (2004) Stem cell biology and neurodegenerative ing of transplanted stem cells in rat brain and spinal cord.
disease. Philos. Trans. R. Soc. Lond. B. Biol. Sci., 359(1445): Neurodegener. Dis., 3: 62–67.
851–856. Sykova, E., Jendelova, P., Urdzikova, L., Lesny, P. and Hejcl,
Mendonca Dias, M.H. and Lauterbur, P.C. (1986) Ferromag- A. (2006) Bone marrow stem cells and polymer hydrogels:
netic particles as contrast agents for magnetic resonance im- two strategies for spinal cord injury repair. Cell. Mol. Ne-
aging of liver and spleen. Magn. Reson. Med., 3(2): 328–330. urobiol., 26(7–8): 1111–1127.
Newman, M.B., Davis, C.D., Borlongan, C.V., Emerich, D. Takahashi, T., Kalka, C., Masuda, H., Chen, D., Silver, M.,
and Sanberg, P.R. (2004) Transplantation of human umbil- Kearney, M., Magner, M., Isner, J.M. and Asahara, T.
ical cord blood cells in the repair of CNS diseases. Expert (1999) Ischemia- and cytokine-induced mobilization of bone
Opin. Biol. Ther., 4(2): 121–130. marrow-derived endothelial progenitor cells for ne-
Norman, A.B., Thomas, S.R., Pratt, R.G., Lu, S.Y. and ovascularization. Nat. Med., 5(4): 434–438.
Norgren, R.B. (1992) Magnetic resonance imaging of neural Toma, C., Pittenger, M.F., Cahill, K.S., Byrne, B.J. and
transplants in rat brain using a superparamagnetic contrast Kessler, P.D. (2002) Human mesenchymal stem cells differ-
agent. Brain Res., 594: 279–283. entiate to a cardiomyocyte phenotype in the adult murine
Orlic, D., Kajstura, J., Chimenti, S., Bodine, D.M., Leri, A. and heart. Circulation, 105(1): 93–98.
Anversa, P. (2001a) Transplanted adult bone marrow cells Uccelli, A., Zappia, E., Benvenuto, F., Frassoni, F. and
repair myocardial infarcts in mice. Ann. N.Y. Acad. Sci., 938: Mancardi, G. (2006) Stem cells in inflammatory demyelina-
221–230. ting disorders: a dual role for immunosuppression and
Orlic, D., Kajstura, J., Chimenti, S., Jakoniuk, I., Anderson, neuroprotection. Expert Opin. Biol. Ther., 6(1): 17–22.
S.M., Li, B., Pickel, J., McKay, R., Nadal-Ginard, B., Urdzikova, L., Jendelova, P., Glogarova, K., Burian, M.,
Bodine, D.M., Leri, A. and Anversa, P. (2001b) Bone mar- Hajek, M. and Sykova, E. (2006) Transplantation of bone
row cells regenerate infarcted myocardium. Nature, marrow stem cells as well as mobilization by granulocyte-
410(6829): 701–705. colony stimulating factor promote recovery after spinal cord
Pachernik, J., Bryja, V., Esner, M., Kubala, L., Dvorak, P. and injury in rat. J. Neurotrauma, 23: 1379–1391.
Hampl, A. (2005) Neural differentiation of pluripotent Zhang, Z.G., Jiang, Q., Zhang, R., Zhang, L., Wang, L., Arni-
mouse embryonal carcinoma cells by retinoic acid: inhibi- ego, P., Ho, K.L. and Chopp, M. (2003) Magnetic resonance
tory effect of serum. Physiol. Res., 54(1): 115–122. imaging and neurosphere therapy of stroke in rat. Ann. Ne-
Park, K.I., Lachyankar, M., Nissim, S. and Snyder, E.Y. (2002) urol., 53(2): 259–263.
Neural stem cells for CNS repair: state of the art and future Zhang, Z.Y. and Smith, B.D. (2000) High-generation polycat-
directions. Adv. Exp. Med. Biol., 506(Pt B): 1291–1296. ionic dendrimers are unusually effective at disrupting anionic
Pittenger, M.F., Mackay, A.M., Beck, S.C., Jaiswal, R.K., vesicles: membrane bending model. Bioconjug. Chem., 11(6):
Douglas, R., Mosca, J.D., Moorman, M.A., Simonetti, 805–814.
D.W., Craig, S. and Marshak, D.R. (1999) Multilineage po- Zhao, C., Fancy, S.P., Magy, L., Urwin, J.E. and Franklin, R.J.
tential of adult human mesenchymal stem cells. Science, (2005) Stem cells, progenitors and myelin repair. J. Anat.,
284(5411): 143–147. 207(3): 251–258.
This page intentionally left blank
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 27

Intrathecal drug delivery strategy is safe and


efficacious for localized delivery to the spinal cord

Molly S. Shoichet1,2,3,, Charles H. Tator4, Peter Poon1,2, Catherine Kang1,2 and


M. Douglas Baumann1,2

1
Department of Chemical Engineering and Applied Chemistry, University of Toronto, Donnelly Center for Cellular and
Biomolecular Research, Toronto, ON, M5S 3E1, Canada
2
Institute of Biomaterials and Biomedical Engineering, 164 College St., Toronto, ON, M5S 3G9, Canada
3
Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, M5S 3H6, Canada
4
Krembil Neuroscience Centre, Toronto Western Research Institute, and Department of Surgery, University of Toronto,
399 Bathurst Street, Toronto, ON, M5 T 2S8, Canada

Abstract: Neuroprotective and neuroregenerative strategies for spinal cord injury repair are limited in part
by poor delivery techniques. A novel drug delivery system is being developed in our laboratory that can
provide localized release of therapeutically relevant molecules from an injectable hydrogel. Design criteria
were established for the hydrogel to be — injectable, fast-gelling, biocompatible, biodegradable and able to
release biologically active therapeutics when injected into the intrathecal space that surrounds the spinal
cord. This novel way of localized drug delivery to the spinal cord was tested first with a collagen gel and
then with a new hydrogel blend of hyaluronan and methylcellulose (HAMC). The underlying principle that
this novel methodology is both safe and able to provide localized delivery was proven with a fast gelling
collagen solution. Using a recombinant human epidermal growth factor, rhEGF, dispersed in collagen, we
demonstrated localized release to the injured spinal cord. We extended this technology to other fast-gelling
systems and found that HAMC was injectable due to the shear thinning property of hyaluronan (HA),
biocompatible and had some therapeutic benefit when injected into the intrathecal space using a com-
pression injury model in rats.

Keywords: spinal cord injury; localized drug delivery; hydrogels; hyaluronan; methyl cellulose; collagen;
epidermal growth factor

Introduction therapeutic strategies that have been investigated


hold great promise (Gorio et al., 2002), yet sys-
Of the several therapeutic strategies investigated temic delivery may cause side effects, and many
for spinal cord injury repair (Pearse et al., 2004; promising therapeutic proteins degrade when de-
Tsai et al., 2004; Ramer et al., 2005), only systemic livered systemically. Moreover, many therapeutic
delivery of methylprednisolone (MP) is used clini- molecules are unable to cross the blood-spinal
cally; however, results from its clinical trial have cord barrier (BSCB). These difficulties suggest that
been openly criticized (Hurlbert, 2001). Other local delivery strategies are required. Two in-
trathecal techniques have been used to test local-
Corresponding author. Tel.: +1-416-978-1460; Fax: +1-416-
ized delivery: (1) bolus injection, the effects of
which are short-lived because the therapeutic
978-4317; E-mail: molly@ecf.utoronto.ca

DOI: 10.1016/S0079-6123(06)61027-3 385


386

agent is washed away by the cerebrospinal fluid CSF volume, a volume which we expected to be
(CSF) flow (Terada et al., 2001; Yaksh et al., tolerated based on previous studies where liquids
2004); and (2) mini-pump delivery, which is inva- of a similar volume had been injected (Rieselbach
sive and can lead to complications due to infection et al., 1962). All animal procedures were per-
and/or blockage of the catheter (Jones and formed in accordance with the Guide to the Care
Tuszynski, 2001). A third extradural delivery strat- and Use of Experimental Animals (Canadian
egy is being investigated clinically for the delivery Council on Animal Care) and protocols were ap-
of rho-kinase inhibitors; however, in this methodo- proved by the Animal Care Committee of the Re-
logy where the therapeutic molecule is applied in a search Institute of the University Health Network.
fibrin glue to the external surface of the dura, the Importantly, injection into the intrathecal space
molecule must diffuse across the dura, the in- of both non-injured and spinal cord injured rats
trathecal space and then the spinal cord, resulting was tolerated and had no deleterious affects rela-
in only a very small fraction of the molecule tive to the injection of an artificial CSF (aCSF).
reaching the target tissue (Dergham et al., 2002; The safety of the delivery strategy methodology
McKerracher and Higuchi, 2006). was proven with a fast gelling collagen solution
(Jimenez Hamann et al., 2003) that was injected
into adult rats either non-injured or after acute
Injectable delivery strategy spinal cord compression injury. The compression
injury was achieved at thoracic level 2 (T2) using a
Given the limitations of current clinical strategies, modified aneurysm clip with a closing force rang-
we sought to develop an improved localized deli- ing from 35 to 56 g, corresponding to injuries
very strategy. As with any localized strategy, the ranging from moderate to severe (Rivlin and
therapeutic benefits should include decreased sys- Tator, 1977, 1978). Safety was assessed by histo-
temic toxicity and decreased dose applied. In the logy, immunohistochemistry and open-field motor
proposed model, we sought to develop a delivery function using the Basso, Beattie and Bresnahan
strategy that would localize the therapeutic mole- (BBB) scoring scale (Basso et al., 1996). This is a
cules at the site of injection in the intrathecal space 21-point scale that ranks no locomotion at 0 and
(or subarachnoid space) to localize delivery to the normal gait at 21. Neither healthy nor injured spi-
injured spinal cord. To this end, our goal was to nal cord tissue was affected by this injection strat-
develop a biocompatible, biodegradable, injecta- egy involving collagen gel. Similarly, locomotor
ble, fast gelling polymer within which therapeutic function was unaffected.
molecules could be dispersed and released. We
aimed to inject the drug delivery system via a
30 gauge needle into the intrathecal cavity and Localized release of bioactive factors
have it gel quickly (less than 1 min) in order to
localize release at this site (see Fig. 1 for concep- Having demonstrated the safety of this new in-
tual understanding of new strategy). trathecal delivery strategy (Jimenez Hamann et al.,
In the first set of studies we aimed to demon- 2003), our next goal was to demonstrate localized
strate safety because this was a completely new delivery and efficacy. Based on previous research
and novel approach toward drug delivery to the which demonstrated that mini-pump delivery into
spinal cord. Several questions arose in the deve- the subarachnoid space of EGF and FGF2 stimu-
lopment of this system, such as: What would hap- lated endogenous stem cell proliferation (Kojima
pen if we injected a polymeric hydrogel into the and Tator, 2000), we chose to study the effects of
intrathecal space? Would this strategy cause any their delivery by the injectable collagen system.
harm to either non-injured or spinal cord injured Using recombinant human (rh) EGF and rhFGF2,
rats? Given that the volume of CSF in the adult rat we first investigated the bioactivity and release
is 250 ml, we chose to investigate the injection of profile of each factor from the collagen gel in vitro.
20 ml, which represents less than 10% of the total Released EGF was bioactive for 14 days and
387

Fig. 1. Injectable delivery strategy to the intrathecal space of the spinal cord. A fine, 30 gauge needle pierces the dura of the spinal cord
and is used to deliver a fast gelling, biodegradable, biocompatible hydrogel for localized delivery of therapeutic molecules to the injured
spinal cord. (Adapted with permission, copyright Michael Corrin (2005).)

FGF2 for 56 days. Confident that our system al- spinal cord. Unlike rhEGF, rhFGF2 did not pene-
lowed release of bioactive factors, we investigated trate the injured (or non-injured) spinal cord tissue
the release of these factors in vivo (in the presence beyond the pia. It is not clear why EGF penetrated
of heparin as a stabilizer). Using a similar animal the tissue and FGF2 did not; however, this may
model — 35 g clip compression injury at T2 — we reflect the greater molecular weight of FGF2 re-
compared the delivery of rhEGF/rhFGF2 vs. lative to EGF or its charge. Importantly, release of
aCSF. We had determined that there was no EGF/FGF2 from the collagen did stimulate the
cross-reactivity between: (1) the rhEGF and the ependymal cells that line the central canal, which is
endogenous EGF in the rat; and (2) the rhFGF2 believed to be the source of endogenous stem cells
and endogenous FGF2, so that we could follow (Martens et al., 2002). There was also evidence of
the depth of penetration into the tissue by reduced cavitation around the site of injury in
immunohistochemistry. We determined that those animals injected with EGF/FGF2 relative to
rhEGF penetrated deeply into the injured spinal aCSF; however, there was no improvement in
cord tissue as evident at 30 min and 6 h after in- function. Stimulation of endogenous stem cells
jection of the collagen gel. At 1 and 7 days, it was was also observed in studies where EGF/FGF2
difficult to detect the rhEGF in the injured spinal was delivered by injection into the ventricles in the
cord due to either a fast release from the collagen brain (Martens et al., 2002).
gel or the limited sensitivity of the immunohisto- The novel injectable drug delivery system was
chemical method. Interestingly, in non-injured shown to be safe and to localize release of these
controls, rhEGF penetrated the spinal cord tissue, agents (Jimenez Hamann et al., 2005). This pro-
but the penetration was superficial and not deep. vided a new paradigm for drug delivery to the
The greater depth of penetration observed in the spinal cord that we have continued to investigate.
injured tissue likely reflects its greater permeabil- However, cellular build-up in the intrathecal space
ity. Figure 2 summarizes the depth of rhEGF pen- was observed when EGF/FGF2 was incorporated
etration observed in the injured and non-injured into the collagen gel, likely attracting fibroblasts
388

Fig. 2. Injection of rhEGF via the fast-gelling collagen gel to the (A) uninjured and (B) injured spinal cord demonstrates localized
delivery. Using immunohistochemical labeling against rhEGF, we observed a superficial penetration of rhEGF into the uninjured
spinal cord (A) and deep penetration into the injured spinal cord (B) as demarcated by the dotted lines. Penetration of rhEGF is
observed after 30 min and 6 h of injection, but not after 1 and 7 days. Figure is at 6 h after injection. (Adapted with permission from
Jimenez Hamann et al. (2005).)

and other cell types. To overcome this limitation thinning — that is it flows in the direction of stress
of the delivery vehicle, it was redesigned to be non- (Weiss, 2000) — and it is this unique property that
adhesive to cells while still meeting the other de- allows HAMC to meet the design criteria of in-
sign criteria of biocompatibility, biodegradability, jectable and fast gelling.
injectability, and fast gelation. Prior to initiating the in vivo study in rats,
HAMC was evaluated in vitro for degradation and
New fast-gelling blend shows therapeutic promise cell adhesion. HAMC was found to degrade (or
erode) within 14 days and was non-adhesive to
Of the many available chemical and physical gels cells. The lack of adhesion of 3T3 fibroblasts to
available, none met all of our design criteria, re- HAMC gels demonstrated the non-adhesive prop-
quiring us to develop a new gel system. While erty of HAMC in vitro.
physical gels are not as stable or robust as chemi- Thus this new fast-gelling, injectable HAMC
cal gels, we believed that they could be sufficiently was investigated as a drug delivery vehicle to the
stable in the intrathecal space. Both methylcellu- injured spinal cord. The first test was one of safety.
lose and hyaluronan met all of our design criteria, Since this was a new blend, we had to first demon-
except fast gelation. Methylcellulose (MC) is an strate that it would not cause any deleterious
inverse gelling polymer that forms a weak gel at effects in the rat animal model. Thus the biocom-
371C in water (Li et al., 2001), but does not gel fast patibility of HAMC within the intrathecal space
enough for our injectable drug delivery system. was examined in vivo in both uninjured and spinal
Hyaluronan (HA) does not gel on its own. How- cord injured rat animal models relative to control
ever, a blend of HA and MC (HAMC) gel quickly animals receiving aCSF injections. In this set of
and in fact form a gel at room temperature, likely studies HAMC was injected at T2 after a moderate
due to a ‘‘salting out’’ effect that HA has on MC clip compression injury using a 35 g clip, similar to
(Xu et al., 2004). the in vivo design with the injectable collagen sys-
In addition to promoting faster gelation of MC, tem. Injection of HAMC in spinal cord injured
HA is attractive because it is non-immunogenic, and uninjured rats were compared to a control
biocompatible (Vercruysse and Prestwich, 1998) injection of aCSF. The only difference other than
and known to promote wound healing by reducing the choice of material was the injection volume,
inflammation and minimizing tissue adhesion and where 10 ml was used instead of the 20 ml of col-
scar formation (Balazs, 1991). HA is also shear lagen injected.
389

Fig. 3. Representative histology sections of each experimental group stained with Luxol Fast Blue and counterstained with Hema-
toxylin and Eosin. Sections showing injection site in dura of: (A) uninjured rat injected with aCSF (arrow points to unsealed dura that
was punctured with needle); and (B) uninjured rat injected with HAMC (arrow points to resealed dura). (Scale bar ¼ 200 mm.)
(Adapted with permission from Gupta et al. (2006).)

It is important to realize that this new technique vs. 2.4270.35  105 mm2 for animals injected with
requires that the dura be punctured, which clini- HAMC. Thus, the inflammatory response at
cally can result in serious post-dural puncture 28 days after injection, was significantly reduced
headaches. Interestingly, in the spinal cord injured in those animals injected with HAMC relative to
animals that received HAMC, the dura was ob- aCSF controls (po0.05, n ¼ 6). This was an un-
served to re-seal whereas no re-sealing was ob- expected, yet beneficial effect of HAMC delivery,
served in animals that received aCSF (Fig. 3). This potentially due to the beneficial anti-inflammatory
key result is critical to the long-term application of effects attributed to HA in other tissues (Dougados,
this material clinically. After 28 days there was no 2000; Sheehan et al., 2003; Cabrera et al., 2004;
evidence of scar formation, arachnoiditis, syringo- Roth et al., 2005). The re-sealing of the dura pro-
myelia or of HAMC itself in the intrathecal space. vides another advantage over other delivery tech-
The benefits of HAMC were observed at the niques, such as bolus and intrathecal mini-pump
tissue level and the functional level. Both cavita- where the dura may not seal at the punctured site,
tion volume and inflammatory response were less resulting in the potential for CSF leakage and/or
in those animals injected with HAMC relative to infection.
aCSF. For example, the cavitation volume in an- The benefit or detriment of the inflammatory
imals injected with aCSF was 52.1720.1 mm3 response is widely debated in the neuroscience
whereas for HAMC the cavitation volume was community (Schwartz, 2003). For example,
36.676.0 mm3. While not statistically different (at Michael Schwartz is investigating the therapeutic
95% confidence), the trend of reduced cavitation benefit of macrophage delivery (Rapalino et al.,
for animals injected with HAMC is important and 1998; Knoller et al., 2005). In the case of HAMC
demonstrates that injection of HAMC is safe and delivery, the reduced inflammatory response ob-
affected neither the normal spinal cord nor the served, judged by ED-1 immunohistochemistry,
severity of injury. The inflammatory response was was thought to be beneficial and there was also
determined by quantification of ED-1 labeled improved locomotor function as described below.
macrophages/microglia based on pixel inten- To gain a greater perspective on the safety of
sity. For animals injected with aCSF, the area oc- HAMC delivery, the locomotor function of ani-
cupied was approximately 3.7971.39  105 mm2 mals was scored by the BBB and grid walk
390

A
21

18

15
BBB Score

12

0
0 5 10 15 20 25 30
Days after surgery
B
18

16

14

12
# of Foot Falls

10

0
0 5 10 15 20 25 30
Days after Surgery

Fig. 4. (A) BBB scores over 1 month to measure the effect of HAMC on animal function: (&) HAMC uninjured, (W) aCSF
uninjured, (’) HAMC injured, and (m) aCSF injured. The two uninjured groups scored normal (21 points) at all times; (B) % foot
falls of total steps per grid walk run for (&) HAMC uninjured, (W) aCSF uninjured, (’) HAMC injured, and (m) aCSF injured. The
two uninjured groups had virtually zero footfalls at all times. Data is shown as mean7standard deviation (uninjured animals: n ¼ 4;
injured animals: n ¼ 8). The asterisk (*) at day 7 in (A) indicates significant difference (p ¼ 0.035). (Adapted with permission from
Gupta et al. (2006).)

analysis. As shown in Fig. 4A, uninjured animals locomotor function relative to injection of
injected with HAMC or aCSF showed no differ- aCSF, and a statistically significant difference
ence in locomotor function, demonstrating was observed at day 7 (p ¼ 0.035). This sug-
safety of the new injectable HAMC material and gests a mild neuroprotective effect of HAMC
confirming safety of the methodology. Impor- (Rabchevsky et al., 1999) possibly due to the
tantly, injection of HAMC in spinal cord injured decreased inflammatory response observed by
animals showed a general trend of improved immunohistochemistry.
391

To confirm the functional benefit observed, grid Canadian Institute of Health Research, the Canada
walk analysis was completed following the meth- Foundation for Innovation and the Ontario
odology described by Metz et al. (2000), where Innovation Trust and the Ontario Branch of the
previously trained animals were scored on the Canadian Paraplegic Association for funding.
number of foot falls that occurred when walking
across a horizontal ladder. As shown in Fig. 4B, References
uninjured animals had nearly perfect scores of zero
foot falls throughout a run for both HAMC- Balazs, E.A. (1991) Cosmetic and pharmaceutical applications
injected and aCSF-injected animals, thereby corrob- of polymers. In: Gebelein C.G. (Ed.), Medical Applications
of Hyaluronan and its Derivatives. Plenum Press, New York,
orating the normal BBB scores shown (cf. Fig. 4A).
pp. 293–310.
It is important to note that uninjured animals Basso, D.M., Beattie, M.S. and Bresnahan, J.C. (1996) Graded
often miss 1–2 steps on the horizontal ladder (Metz histological and locomotor outcomes after spinal cord con-
et al., 2000; Hains et al., 2004), likely due to their tusion using the NYU weight-drop device versus transection.
carelessness when crossing the grid quickly. For Exp. Neurol., 139: 244–256.
Cabrera, P.V., Blanco, G., Alaniz, L., Greczanik, S., Garcia,
injured animals, the HAMC-injected animals had
M., Alvarez, E. and Hajos, S.E. (2004) CD44 and hyaluronic
fewer foot falls than aCSF-injected animals, but acid regulate in vivo iNOS expression and metalloproteinase
the difference between HAMC-injected and aCSF- activity in murine air-pouch inflammation. Inflamm. Res.,
injected animals was not statistically different. 53: 556–566.
Dergham, P., Ellezam, B., Essagian, C., Avedissian, H., Lubell,
W.D. and McKerracher, L. (2002) Rho signaling pathway
Conclusions and future outlook
targeted to promote spinal cord repair. J. Neurosci., 22:
We have demonstrated a new and safe way to 6570–6577.
Dougados, M. (2000) Sodium hyaluronate therapy in osteoar-
achieve localized delivery to the injured spinal
thritis: arguments for a potential beneficial structural effect.
cord. First with collagen and then with HAMC, Semin. Arthritis Rheum., 30: 19–25.
we demonstrated that the underlying methodology Gorio, A., Gokmen, N., Erbayraktar, S., Yilmaz, O., Mad-
is safe, having neither a negative impact on the aschi, L., Cichetti, C., Di Giulio, A.M., Vardar, E., Cerami,
tissue nor locomotor function. Moreover, there A. and Brines, M. (2002) Recombinant human erythropoietin
counteracts secondary injury and markedly enhances neuro-
may be a small therapeutic benefit of HAMC
logical recovery from experimental spinal cord trauma. Proc.
alone, indicating that this material may be useful Natl. Acad. Sci. U.S.A., 99: 9450–9455.
as a delivery and for its inherent ‘‘wound healing’’ Gupta, D., Tator, C.H. and Shoichet, M.S. (2006) Fast-gelling
properties. Lastly, we found that this delivery injectable blend of hyaluronan and methylcellulose for in-
strategy resulted in localized release of growth trathecal, localized delivery to the injured spinal cord. Bio-
materials, 27: 2370–2379.
factors into the injured spinal cord, thereby prov-
Hains, B.C., Saab, C.Y., Lo, A.C. and Waxman, S.G. (2004)
ing the hypothesis that this strategy could achieve Sodium channel blockade with phenytoin protects spinal
local release into the injured tissue. cord axons, enhances axonal conduction, and improves func-
In on-going studies we continue to examine the tional motor recovery after contusion SCI. Exp. Neurol., 188:
therapeutic benefit of the release of neuroprotec- 365–377.
Hurlbert, R.J. (2001) The role of steroids in acute spinal cord
tive and neuroregenerative factors to limit degen-
injury: an evidence-based analysis. Spine, 26: S39–S46.
eration and promote regeneration, respectively, Jimenez Hamann, M.C., Tator, C.H. and Shoichet, M.S. (2005)
following traumatic injury to the spinal cord. Lo- Injectable intrathecal delivery system for localized adminis-
calized release from hydrogel systems continues to tration of EGF and FGF-2 to the injured rat spinal cord.
be explored for its potential therapeutic benefit in Exp. Neurol., 194: 106–119.
Jimenez Hamann, M.C., Tsai, E.C., Tator, C.H. and Shoichet,
spinal cord injury.
M.S. (2003) Novel intrathecal delivery system for treatment
of spinal cord injury. Exp. Neurol., 182: 300–309.
Jones, L.L. and Tuszynski, M.H. (2001) Chronic intrathecal
Acknowledgments
infusions after spinal cord injury cause scarring and com-
pression. Microsc. Res. Tech., 54: 317–324.
We are grateful to Maria Jimenez-Hamann and Knoller, N., Auerbach, G., Fulga, V., Zelig, G., Attias, J.,
Dimpy Gupta for initial studies. We thank the Bakimer, R., Marder, J.B., Yoles, E., Belkin, M., Schwartz, M.
392

and Hadani, M. (2005) Clinical experience using incubated Rivlin, A.S. and Tator, C.H. (1977) Objective clinical assess-
autologous macrophages as a treatment for complete spinal ment of motor function after experimental spinal cord injury
cord injury: phase I study results. J. Neurosurg. Spine, 3: in the rat. J. Neurosurg., 47: 577–581.
173–181. Rivlin, A.S. and Tator, C.H. (1978) Effect of duration of acute
Kojima, A. and Tator, C.H. (2000) Epidermal growth factor spinal cord compression in a new acute cord injury model in
and fibroblast growth factor 2 cause proliferation of the rat. Surg. Neurol., 10: 38–43.
ependymal precursor cells in the adult rat spinal cord in Roth, A., Mollenhauer, J., Wagner, A., Fuhrmann, R., Straub,
vivo. J. Neuropathol. Exp. Neurol., 59: 687–697. A., Venbrocks, R.A., Petrow, P., Brauer, R., Schubert, H.,
Li, L., Thangamathesvaran, P.M., Yue, C.Y., Tam, K.C., Hu, Ozegowski, J., Peschel, G., Muller, P.J. and Kinne, R.W.
X. and Lam, Y.C. (2001) Gel network structure of methyl- (2005) Intra-articular injections of high-molecular-weight
cellulose in water. Langmuir, 17: 8062–8068. hyaluronic acid have biphasic effects on joint inflammation
Martens, D.J., Seaberg, R.M. and van der Kooy, D. (2002) In and destruction in rat antigen-induced arthritis. Arthritis
vivo infusions of exogenous growth factors into the fourth Res. Ther., 7: R677–R686.
ventricle of the adult mouse brain increase the proliferation of Schwartz, M. (2003) Macrophages and microglia in central
neural progenitors around the fourth ventricle and the central nervous system injury: are they helpful or harmful? J. Cereb.
canal of the spinal cord. Eur. J. Neurosci., 16: 1045–1057. Blood Flow Metab., 23, 385–394.
McKerracher, L. and Higuchi, H. (2006) Targeting Rho to Sheehan, K.M., DeLott, L.B., Day, S.M. and DeHeer, D.H.
stimulate repair after spinal cord injury. J. Neurotrauma, 23: (2003) Hyalgan has a dose-dependent differential effect on
309–317. macrophage proliferation and cell death. J. Orthop. Res., 21:
Metz, G.A., Merkler, D., Dietz, V., Schwab, M.E. and Fouad, 744–751.
K. (2000) Efficient testing of motor function in spinal cord Terada, H., Kazui, T., Takinami, M., Yamashita, K., Washi-
injured rats. Brain Res., 883: 165–177. yama, N. and Muhammad, B.A. (2001) J. Thorac. Card-
Pearse, D.D., Pereira, F.C., Marcillo, A.E., Bates, M.L., iovasc. Surg., 122: 979–985.
Berrocal, Y.A., Filbin, M.T. and Bunge, M.B. (2004) cAMP Tsai, E.C., Dalton, P.D., Shoichet, M.S. and Tator, C.H. (2004)
and Schwann cells promote axonal growth and functional Synthetic hydrogel guidance channels facilitate regeneration
recovery after spinal cord injury. Nat. Med., 10: 610–616. of adult rat brainstem motor axons after complete spinal
Rabchevsky, A.G., Fugaccia, I., Fletcher-Turner, A., Blades, cord transection. J. Neurotrauma, 21: 789–804.
D.A., Mattson, M.P. and Scheff, S.W. (1999) Basic fibroblast Vercruysse, K.P. and Prestwich, G.D. (1998) Hyaluronate de-
growth factor (bFGF) enhances tissue sparing and functional rivatives in drug delivery. Crit. Rev. Ther. Drug Carrier Syst.,
recovery following moderate spinal cord injury. J. Neuro- 15: 513–555.
trauma, 16: 817–830. Weiss, C. (2000) New frontiers in medical sciences: redefining
Ramer, L.M., Ramer, M.S. and Steeves, J.D. (2005) Setting the Hyaluronan. In: Weigel, G. A. a. P. H. (Eds.), Why Visco-
stage for functional repair of spinal cord injuries: a cast of elasticity is Important for the Medical Uses of Hyaluronan
thousands. Spinal Cord, 43: 134–161. and Hylans. Excerpta Medica, pp. 89–103.
Rapalino, O., Lazarov-Spiegler, O., Agranov, E., Velan, G.J., Xu, Y., Wang, C., Tam, K.C. and Li, L. (2004) Salt-assisted
Yoles, E., Fraidakis, M., Solomon, A., Gepstein, R., Katz, and salt-suppressed sol-gel transitions of methylcellulose in
A., Belkin, M., Hadani, M. and Schwartz, M. (1998) Im- water. Langmuir, 20: 646–652.
plantation of stimulated homologous macrophages results in Yaksh, T.L., Horais, K.A., Tozier, N., Rathbun, M., Richter,
partial recovery of paraplegic rats. Nat. Med., 4: 814–821. P., Rossi, S., Grafe, M., Tong, C., Meschter, C., Cline, J.M.
Rieselbach, R.E., Di Chiro, G., Freireich, E.J. and Rall, D.P. and Eisenach, J. (2004) Toxicol. Sci., 80: 322–334.
(1962) Subarachnoid distribution of drugs after lumbar in-
jection. N. Engl. J. Med., 267: 1273–1278.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 28

Decompression craniectomy after traumatic brain


injury: recent experimental results

Nikolaus Plesnila

Laboratory of Experimental Neurosurgery, Department of Neurosurgery and Institute for Surgical Research, University of
Munich Medical Center, GroX hadern, Marchioninistr 15, 81377 Munich, Germany

Abstract: Among the secondary events occurring after traumatic brain injury (TBI) pathologically in-
creased intracranial pressure (ICP) correlates most closely with poor outcome. In addition to infusion of
hypertonic solutions, e.g. mannitol, and other medical measures, decompression of the brain by surgical
removal of a portion of the cranium (craniectomy) has been used for many decades as an intuitive strategy
for the treatment of post-traumatic ICP increase. The lack of evidence-based clinical and controversial
experimental data, however, resulted in decompressive craniectomy to be recommended by most national
and international guidelines only as a third tier therapy for the treatment of pathologically elevated ICP.
Ongoing clinical trials on the use of decompressive craniectomy after TBI may clarify many aspects of the
clinical application of this technique, however, some important pathophysiological issues, e.g. the timing of
decompression craniectomy, its effect on brain edema formation, and its role for secondary brain damage,
are still widely discussed and can only be addressed in experimental settings. The aim of the current review
was therefore to summarize and discuss recent experimental data dealing with the use of decompression
craniectomy following TBI. The present results suggest that surgical decompression effectively prevents
secondary brain damage when performed early enough. Although caution should be taken when trans-
ferring conclusions drawn from experimental settings to the clinical situation, the current literature suggests
that the timing of decompression may be of utmost importance in order to exploit the full neuroprotective
potential of craniectomy following TBI.

Keywords: traumatic brain injury; decompression; craniectomy; intracranial pressure; mice; review

Introduction History of decompression craniectomy

Despite significant improvements in the manage- Opening of the cranial cavity is the most intuitive
ment of traumatic brain injury (TBI) within the treatment option for increased intracranial pres-
past two decades, brain edema formation resulting sure (ICP). Accordingly, as soon as increased ICP
in refractory intracranial hypertension remains the was considered an important component of the
leading cause of unfavorable outcome and death pathophysiology of TBI, i.e. at the beginning of the
among affected patients (Murray et al., 1999). last century, surgical decompression was used for
the treatment of patients suffering from the sequels
of severe TBI. The first report on the use of de-
compression craniectomy following TBI was pub-
Corresponding author. Tel.: +49-89-2180-76-535; Fax: +49- lished by the legendary Swiss surgeon and Noble
89-2180-76-532; E-mail: plesnila@med.uni-muenchen.de laureate for medicine Theodor Emil Kocher (1901).

DOI: 10.1016/S0079-6123(06)61028-5 393


394

The intense collaboration of Kocher with Harvey randomized, and controlled study design and in
Cushing resulted — among others — in the use of some cases an insufficient number of observations.
decompression craniectomy also for the treatment In additions to these limitations, there are also a
of other cerebral disorders, e.g., brain tumors and number of publications reporting no benefit from
vascular malformations (Cushing, 1905). decompressive craniectomy (Clark et al., 1968;
Munch et al., 2000; Soukiasian et al., 2002;
Messing-Junger et al., 2003). For example, a
Decompression craniectomy after TBI in man retrospective study comparing craniectomised
patients from one center with a matched control
Kocher’s initial positive experience with decom- population form the Traumatic Coma Data Bank
pression craniectomy was followed by a plethora of reported no differences between these two groups
controversial clinical findings on the use of surgical regarding ICP, therapy intensity level, and overall
decompression following brain trauma during the mortality (Munch et al., 2000), a finding also cor-
past 100 years (Diemath, 1966; Clark et al., 1968; roborated by others (Soukiasian et al., 2002). Ac-
Kerr, 1968; Kjellberg and Prieto, 1971; Ransohoff cordingly, despite a report of a randomized trial
and Benjamin, 1971; Ogawa et al., 1974; Venes demonstrating beneficial use of decompression
and Collins, 1975; Pereira et al., 1977; Makino and craniectomy in children (Taylor et al., 2001) and
Yamaura, 1979; Yamaura et al., 1979; Gerl and clear benefits of surgical decompression in stroke
Tavan, 1980). These early reports helped to deter- patients (Schwab et al., 1998; Fraser and Hartl,
mine the most effective surgical technique for de- 2005), the current literature does not provide
compression craniectomy in TBI patients. Another evidence-based proof for the general use of decom-
important finding was that decompression craniec- pression surgery following TBI in adults. As a re-
tomy, most effectively together with opening of the sult current American and European TBI
dura mater, lowers pathologically elevated ICP, a guidelines still recommend decompression craniec-
fact confirmed also by more recent studies (Dam tomy only as a ‘‘second tier’’ therapeutic option for
et al., 1996; Polin et al., 1997; Yoo et al., 1999; adult patients with TBI (Maas et al., 1997; The
Berger et al., 2002; Schneider et al., 2002; Ruf et al., Brain Trauma Foundation. The American Associ-
2003; Aarabi et al., 2006; Josan and Sgouros, 2006; ation of Neurological Surgeons. The Joint Section
Sahuquillo and Arikan, 2006). However, despite on Neurotrauma and Critical Care, 2000).
the positive effect of decompression craniectomy
on otherwise uncontrollable intracranial hyperten-
sion, the effect of surgical decompression on mor- Decompression craniectomy after experimental TBI
tality and overall functional outcome following
TBI remained controversial. For example, a recent Because heterogeneous patient populations and
review of 400 patients with severe TBI reported mechanisms of injury may have hampered the
that craniectomised patients may have benefited investigation of decompression craniectomy in the
from the procedure (Meier and Grawe, 2003). An- clinical environment, many researchers tried to
other study matching TBI patients with craniec- evaluate the therapeutic potential of decompres-
tomy with non-craniectomised controls from the sion craniectomy under more homogenous and
Traumatic Coma Data Bank reported good recov- controlled conditions, i.e. in experimental models
ery or moderate disability in 37% of decompressed of TBI. Most surprisingly also experimental data
patients as compared with only 16% in the historic on the therapeutic use of decompression craniec-
control group (Polin et al., 1997). Although a num- tomy after brain trauma were not univocal
ber of other studies support these findings (Kunze (Moody et al., 1968; Cooper et al., 1979; Gaab
et al., 1998; Guerra et al., 1999; De Luca et al., et al., 1979; Burkert and Plaumann, 1989; Rinaldi
2000; Kontopoulos et al., 2002; Schneider et al., et al., 1990). Although most authors agree with the
2002; Ziai et al., 2003), the general problem of all clinical finding that craniotomy lowers pathologi-
these investigations is the lack of a prospective, cally elevated ICP (Moody et al., 1968; Gaab et al.,
395

1979; Rinaldi et al., 1990), an increase in brain can only be avoided by prophylactic measures, sec-
edema formation and enhanced histopathological ondary injury is a dynamic process which develops
damage were observed after craniectomy in experi- gradually over several hours (maybe days) and
mental animals (Moody et al., 1968; Gaab et al., offers therefore the possibility for therapeutic inter-
1979; Rinaldi et al., 1990). Although it is difficult ventions (Murray et al., 1999; Hartl and Ougorets,
to retrospectively evaluate why experimental stud- 2004; Nortje and Menon, 2004). In case of experi-
ies show such heterogeneous results, the consider- mental traumatic contusions, as observed after the
ation of some pathophysiological concepts not controlled cortical impact (CCI) brain injury model,
available at the time when these experiments were these two components can easily be distinguished
performed may deepen our understanding on the by standard histopathological techniques, e.g. Nissl
use of decompression craniectomy after TBI. staining. Determination of the contusion volume
15 min after injury reveals a value of 20 mm3,
while 24 h after TBI the contusion volume plateaus
Pathophysiological considerations at 32 mm3. This indicates that the primary
contused tissue, which is necrotic already minutes
Experimental and clinical findings demonstrate that after TBI, i.e. it contains only pyknotic nuclei and
the pathophysiology of TBI consists of two com- does not show any perfusion, increases by 60%
ponents: one is determined by the immediate me- (Zweckberger et al., 2006). Determination of the
chanical damage to the brain by the traumatic contusion volume at intermediate time points, i.e.
insult and the other is determined by a plethora of 2, 6, and 12 h after TBI, shows a fast and linear
secondary events on the functional, neurochemical, expansion of contused tissue volume within the first
and molecular level, which ultimately result in addi- 12 h after trauma with only limited additional tissue
tional loss of initially viable brain tissue. While the loss from 12 to 24 h (Fig. 1). Since blood-brain
first, or primary injury component occurs within a barrier leakage, brain edema formation, and ICP
few milliseconds during the mechanical impact and increase show a very similar early time course

32 : p<0.05 vs 15min; n=7

30
Contusion volume (mm3)

28
+63%
26 +52%

24

22 +32%

20

18

16
0
0 2 6 12 24
Time after trauma (h)

Fig. 1. Secondary expansion of the volume of a traumatic cortical contusion after brain injury. As compared with the primary damage
measured immediately after the traumatic impact (0 h; controlled cortical impact in mice) secondary processes result in delayed and
progressive expansion of the contusion by more than 60% of its initial size during the first 24 h after experimental TBI. Adapted with
permission from Zweckberger et al. (2006).
396

(Zweckberger et al., 2003, 2006) it seem to be clear kinetic experimental TBI model, CCI, investigating
that secondary brain damage and its sequel, i.e. the maximal treatment potential of decompression
contusion expansion, is a linearly ongoing process craniectomy. This was achieved by comparing one
which starts immediately after TBI and levels off group of animals which was allowed to develop
approximately 12 h after injury. The consequence of post-traumatic ICP, while in a second group of
this finding is, that the later a therapeutic interven- animals pathological ICP increase was completely
tion starts the less effective it will be. In the case of prevented by a large parietal craniectomy (Fig. 2).
craniectomy this may indicate that delayed initiation Under these circumstances the contusion volume of
of decompression, i.e. at a time point when brain the not craniectomised animals increased by almost
edema and ICP already developed, may not have 60% within 24 h as compared with the primary
any beneficial potential. In the contrary, under cir- damage inflicted by the mechanical impact imme-
cumstances of massive brain edema formation and diately (15 min) after trauma (Fig. 3). In craniecto-
high ICP craniectomy, which among others lowers mised animals the secondary contusion expansion
vascular–interstitial pressure gradients, may also was completely blunted thereby demonstrating that
have adverse effects, e.g. an increased risk of inter- decompression craniectomy completely and effec-
nal or external herniation by enhanced vasogenic tively prevents the secondary loss of brain tissue
edema formation with subsequent cerebral ischemia following TBI.
or detrimental brain stem compression, respectively. Since two studies from the late 70 s using cold
Evaluating the protocols of the early experimen- lesion-induced brain injury reported up to seven-
tal studies investigating decompression craniectomy fold increase of brain edema volume 8 h following
following TBI (Moody et al., 1968; Cooper et al., decompressive craniectomy in dogs (Cooper et al.,
1979; Gaab et al., 1979; Burkert and Plaumann, 1979; Gaab et al., 1979), we performed wet–dry
1989; Rinaldi et al., 1990) it becomes clear that, ratio measurements for the determination of brain
most logically, a clinically relevant treatment sce- edema in our kinetic TBI model. Brain water con-
nario was chosen. In parallel to the situation in tent in mice increased dramatically by almost 3%
patients, animals were decompressed several hours (from 78 to 81%) following CCI. In craniectomised
after TBI when pathologically elevated ICP already animals surgical decompression prevented brain
developed. In view of our current understanding edema formation by more than 50%. Together
of the gradual and irreversible development of with the lack of secondary contusion expansion
secondary brain damage, the point in time of de- these findings indicate that approximately 50% of
compression in these studies may well have been post-traumatic brain edema formation following
chosen too late, i.e. at a time when the vicious CCI are primary, i.e. caused by the initial mechan-
circle of secondary brain damage already started ical impact, while only 50% are secondary and are
and sequels became irreversible. therefore amenable to therapeutic interventions.
Accordingly, the experimental data available up Finally, we were interested to evaluate the thera-
to the beginning of the new millennium did not peutic window of decompression craniectomy.
necessarily allow to draw definitive conclusions Control animals were traumatized but not craniec-
on the therapeutic potential of decompression tomised, while treated animals were craniectomised
craniectomy after TBI. immediately after TBI or with a delay of 1, 3, or 8 h.
Contusion volume in not craniectomised mice was
over 30 mm3 24 h after TBI indicating progressive
Experimental results on early decompression after secondary brain damage, while animals craniecto-
TBI mised immediately after trauma had contusion vol-
umes of 20 mm3, i.e. comparable with primary
Based on the above-mentioned pathophysiological contusions. If craniectomy was delayed by 1 or 3 h
understanding of the gradual and irreversible pro- a significant proportion of secondary brain damage
gression of parenchymal brain damage following was prevented, while decompression 8 h after TBI
TBI, we designed a series of experiments in a showed a marginal but statistically not significant
397

30 Mean +/- SD; n=3 p<0.001 vs. control


p<0.001 vs. 0 h (baseline)
25

20 Control
ICP (mmHg)

15

10

5
Craniectomy

0
0 1 2 3 6 12 24
Time after trauma (h)

Fig. 2. Intracranial pressure of craniectomised and not craniectomised mice after experimental TBI (CCI). While intracranial pressure
increases significantly in not decompressed animals (control; open circles; n ¼ 6), no significant ICP increase is observed in animals
receiving a large parietal craniectomy immediately after TBI (closed circles; n ¼ 6). Adapted with permission from Zweckberger et al.
(2003).

45
Mean +/- SD; n=7 Closed skull
40 p<0.05 vs. Control 24h Craniectomized

35
Contusion volume (mm3)

30

25

20

15

10

0
15 min 24 h 24 h

Fig. 3. Effect of decompression craniectomy on the secondary expansion of a traumatic cortical contusion. In not craniectomised mice
(gray bars) significant secondary expansion of a traumatic contusion (streaked area) is observed (compare also with Fig. 1). In contrast,
no secondary lesion expansion is present in craniectomised mice indicating that craniectomy completely prevents secondary brain
damage. Adapted with permission from Zweckberger et al. (2006).

effect. These results clearly indicate that the patho- effective treatment options, e.g. craniectomy, are
physiology of secondary lesion expansion follows a not or only marginally effective.
two step paradigm: a slow starting phase where Taken together, the recent literature indicates
treatment is effective which is ultimately followed that the timing of craniectomy is of major signifi-
by a self amplifying final phase when even the most cance for its therapeutic benefit: performed in the
398

presence of massive brain swelling decompression Berger, S., Schwarz, M. and Huth, R. (2002) Hypertonic saline
may be detrimental, but when performed early solution and decompressive craniectomy for treatment of
intracranial hypertension in pediatric severe traumatic brain
enough craniectomy may completely prevent seco-
injury. J. Trauma, 53: 558–563.
ndary brain damage (including secondary brain Burkert, W. and Plaumann, H. (1989) The value of large pres-
edema formation). sure-relieving trepanation in treatment of refractory brain
Although we are aware of the caution which edema. Animal experiment studies, initial clinical results.
should be taken when extrapolating experimental Zentralbl. Neurochir., 50: 106–108.
data to the human situation one has also to take Clark, K., Nash, T.M. and Hutchison, G.C. (1968) The failure
of circumferential craniotomy in acute traumatic cerebral
into consideration that pure physical phenomena swelling. J. Neurosurg., 29: 367–371.
like pressure changes in a closed cavity may not be Cooper, P.R., Hagler, H., Clark, W.K. and Barnett, P. (1979)
as different between species as compared with other Enhancement of experimental cerebral edema after decom-
parameters, e.g. gene expression or the initiation of pressive craniectomy: implications for the management of
severe head injuries. Neurosurgery, 4: 296–300.
intracellular signal transduction cascades. Accord-
Cushing, H. (1905) The establishment of cerebral hernia as a
ingly, our results suggest that early decompressive decompressive measure for inaccessible brain tumor: with the
craniectomy may have a clinical potential and description of intramuscular methods of making the bone
should clearly be favored in comparison with de- defect in temporal and occipital regions. Surg. Gynecol.
layed surgical decompression, which may even be Obstet., 1: 297–314.
Dam, H.P., Sizun, J., Person, H. and Besson, G. (1996) The
detrimental. In view of the present data we conclude
place of decompressive surgery in the treatment of uncon-
that timing seems to be of utmost importance and trollable post-traumatic intracranial hypertension in children.
that decompression has to be performed as early as Childs Nerv. Syst., 12: 270–275.
possible in order to exert its therapeutic potential. De Luca, G.P., Volpin, L., Fornezza, U., Cervellini, P.,
Zanusso, M., Casentini, L., Curri, D., Piacentino, M.,
Bozzato, G. and Colombo, F. (2000) The role of decompres-
Abbreviations sive craniectomy in the treatment of uncontrollable post-
traumatic intracranial hypertension. Acta Neurochir. Suppl.,
76: 401–404.
CCI controlled cortical impact Diemath, H.E. (1966) The value of bitemporal relief surgery in
ICP intracranial pressure severe closed skull brain trauma. Wien. Med. Wochenschr.,
TBI traumatic brain injury 116: 1043–1044.
Fraser, J.F. and Hartl, R. (2005) Decompressive craniectomy as
a therapeutic option in the treatment of hemispheric stroke.
Curr. Atheroscler. Rep., 7: 296–304.
Acknowledgments Gaab, M., Knoblich, O.E., Fuhrmeister, U., Pflughaupt,
K.W. and Dietrich, K. (1979) Comparison of the effects of
surgical decompression and resection of local edema in the
The excellent work of Klaus Zweckberger who
therapy of experimental brain trauma. Investigation of ICP,
performed most experiments investigating the role EEG and cerebral metabolism in cats. Childs Brain, 5:
of decompression craniectomy on secondary brain 484–498.
damage after TBI is highly appreciated. Special Gerl, A. and Tavan, S. (1980) Bilateral craniectomy in the
thanks also to Dr. Alexander Baethmann, who treatment of severe traumatic brain edema. Zentralbl.
Neurochir., 41: 125–138.
was always a source of inspiration and help during
Guerra, W.K., Gaab, M.R., Dietz, H., Mueller, J.U., Piek, J.
this project. and Fritsch, M.J. (1999) Surgical decompression for trau-
matic brain swelling: indications and results. J. Neurosurg.,
90: 187–196.
References Hartl, R. and Ougorets, I. (2004) Critical care of neurotrauma.
Curr. Neurol. Neurosci. Rep., 4: 481–488.
Aarabi, B., Hesdorffer, D.C., Ahn, E.S., Aresco, C., Scalea, Josan, V.A. and Sgouros, S. (2006) Early decompressive
T.M. and Eisenberg, H.M. (2006) Outcome following de- craniectomy may be effective in the treatment of refractory
compressive craniectomy for malignant swelling due to intracranial hypertension after traumatic brain injury. Childs
severe head injury. J. Neurosurg., 104: 469–479. Nerv. Syst., 22: 1268–1274.
399

Kerr, F.W. (1968) Radical decompression and dural grafting in Pereira, W.C., Neves, V.J. and Rodrigues, Y. (1977) Bifrontal
severe cerebral edema. Mayo Clin. Proc., 43: 852–864. decompressive craniotomy as the treatment for severe cere-
Kjellberg, R.N. and Prieto Jr., A. (1971) Bifrontal decompres- bral edema. Arq. Neuropsiquiatr., 35: 99–111.
sive craniotomy for massive cerebral edema. J. Neurosurg., Polin, R.S., Shaffrey, M.E., Bogaev, C.A., Tisdale, N.,
34: 488–493. Germanson, T., Bocchicchio, B. and Jane, J.A. (1997)
Kocher, T. (1901) Die Therapie des Hirndruckes. In: Hölder Decompressive bifrontal craniectomy in the treatment of se-
A. (Ed.), Hirnerschütterung, Hirndruck und chirurgische vere refractory posttraumatic cerebral edema. Neurosurgery,
Eingriffe bei Hirnkrankheiten. Hölder Verlag, Wien, pp. 41: 84–92.
262–266. Ransohoff, J. and Benjamin, V. (1971) Hemicraniectomy in the
Kontopoulos, V., Foroglou, N., Patsalas, J., Magras, J., Foro- treatment of acute subdural haematoma. J. Neurol. Ne-
glou, G., Yiannakou-Pephtoulidou, M., Sofianos, E., Anast- urosurg. Psychiatry, 34: 106.
assiou, H. and Tsaoussi, G. (2002) Decompressive Rinaldi, A., Mangiola, A., Anile, C., Maira, G., Amante, P.
craniectomy for the management of patients with refractory and Ferraresi, A. (1990) Hemodynamic effects of decom-
hypertension: should it be reconsidered? Acta Neurochir. pressive craniectomy in cold induced brain oedema. Acta
(Wien.), 144: 791–796. Neurochir. Suppl. (Wien.), 51: 394–396.
Kunze, E., Meixensberger, J., Janka, M., Sorensen, N. and Ruf, B., Heckmann, M., Schroth, I., Hugens-Penzel, M., Reiss,
Roosen, K. (1998) Decompressive craniectomy in patients I., Borkhardt, A., Gortner, L. and Jodicke, A. (2003) Early
with uncontrollable intracranial hypertension. Acta Neuro- decompressive craniectomy and duraplasty for refractory in-
chir. Suppl. (Wien.), 71: 16–18. tracranial hypertension in children: results of a pilot study.
Maas, A.I., Dearden, M., Teasdale, G.M., Braakman, R., Co- Crit. Care, 7: R133–R138.
hadon, F., Iannotti, F., Karimi, A., Lapierre, F., Murray, G., Sahuquillo, J. and Arikan, F. (2006) Decompressive craniec-
Ohman, J., Persson, L., Servadei, F., Stocchetti, N. and Un- tomy for the treatment of refractory high intracranial pres-
terberg, A. (1997) EBIC-guidelines for management of severe sure in traumatic brain injury. Cochrane Database Syst.
head injury in adults. European Brain Injury Consortium. Rev., CD003983.
Acta Neurochir. (Wien.), 139: 286–294. Schneider, G.H., Bardt, T., Lanksch, W.R. and Unterberg, A.
Makino, H. and Yamaura, A. (1979) Assessment of outcome (2002) Decompressive craniectomy following traumatic brain
following large decompressive craniectomy in management of injury: ICP, CPP and neurological outcome. Acta Neurochir.
serious cerebral contusion. A review of 207 cases. Acta Suppl., 81: 77–79.
Neurochir. Suppl. (Wien.), 28: 193–194. Schwab, S., Steiner, T., Aschoff, A., Schwarz, S., Steiner, H.H.,
Meier, U. and Grawe, A. (2003) The importance of decom- Jansen, O. and Hacke, W. (1998) Early hemicraniectomy in
pressive craniectomy for the management of severe head in- patients with complete middle cerebral artery infarction.
juries. Acta Neurochir. Suppl., 86: 367–371. Stroke, 29: 1888–1893.
Messing-Junger, A.M., Marzog, J., Wobker, G., Sabel, M. and Soukiasian, H.J., Hui, T., Avital, I., Eby, J., Thompson, R.,
Bock, W.J. (2003) Decompressive craniectomy in severe Kleisli, T., Margulies, D.R. and Cunneen, S. (2002) Decom-
brain injury. Zentralbl. Neurochir., 64: 171–177. pressive craniectomy in trauma patients with severe brain
Moody, R.A., Ruamsuke, S. and Mullan, S.F. (1968) An eval- injury. Am. Surg., 68: 1066–1071.
uation of decompression in experimental head injury. J. Taylor, A., Butt, W., Rosenfeld, J., Shann, F., Ditchfield, M.,
Neurosurg., 29: 586–590. Lewis, E., Klug, G., Wallace, D., Henning, R. and Tibballs, J.
Munch, E., Horn, P., Schurer, L., Piepgras, A., Paul, T. and (2001) A randomized trial of very early decompressive craniec-
Schmiedek, P. (2000) Management of severe traumatic brain tomy in children with traumatic brain injury and sustained
injury by decompressive craniectomy. Neurosurgery, 47: intracranial hypertension. Childs Nerv. Syst., 17: 154–162.
315–322. The Brain Trauma Foundation. The American Association of
Murray, G.D., Teasdale, G.M., Braakman, R., Cohadon, F., Neurological Surgeons. The Joint Section on Neurotrauma
Dearden, M., Iannotti, F., Karimi, A., Lapierre, F., Maas, and Critical Care. (2000) Critical pathway for the treatment
A., Ohman, J., Persson, L., Servadei, F., Stocchetti, N., of established intracranial hypertension. J. Neurotrauma, 17:
Trojanowski, T. and Unterberg, A. (1999) The European 537–538.
Brain Injury Consortium survey of head injuries. Acta Ne- Venes, J.L. and Collins, W.F. (1975) Bifrontal decompressive
urochir. (Wien.), 141: 223–236. craniectomy in the management of head trauma. J.
Nortje, J. and Menon, D.K. (2004) Traumatic brain injury: Neurosurg., 42: 429–433.
physiology, mechanisms, and outcome. Curr. Opin. Neurol., Yamaura, A., Uemura, K. and Makino, H. (1979) Large
17: 711–718. decompressive craniectomy in management of severe cere-
Ogawa, M., Minami, T., Katsurada, K. and Sugimoto, T. bral contusion. A review of 207 cases. Neurol. Med. Chir.
(1974) Evaluation of external cranial decompression for (Tokyo), 19: 717–728.
traumatic acute brain swelling. Med. J. Osaka Univ., 25: Yoo, D.S., Kim, D.S., Cho, K.S., Huh, P.W., Park, C.K. and
73–78. Kang, J.K. (1999) Ventricular pressure monitoring during
400

bilateral decompression with dural expansion. J. Neurosurg., decompressive craniectomy on secondary brain damage after
91: 953–959. controlled cortical impact in mice. J. Neurotrauma, 23:
Ziai, W.C., Port, J.D., Cowan, J.A., Garonzik, I.M., Bhardwaj, 1083–1093.
A. and Rigamonti, D. (2003) Decompressive craniectomy for Zweckberger, K., Stoffel, M., Baethmann, A. and Plesnila,
intractable cerebral edema: experience of a single center. J. N. (2003) Effect of decompression craniotomy on increase
Neurosurg. Anesthesiol., 15: 25–32. of contusion volume and functional outcome after con-
Zweckberger, K., Eros, C., Zimmermann, R., Kim, S.W., En- trolled cortical impact in mice. J. Neurotrauma, 20:
gel, D. and Plesnila, N. (2006) Effect of early and delayed 1307–1314.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 29

Novel neuroproteomic approaches to studying


traumatic brain injury

Andrew K. Ottens1,2,, Firas H. Kobeissy1,2, Brian F. Fuller1, Ming Chen Liu2,


Monika W. Oli3, Ronald L. Hayes2,3 and Kevin K.W. Wang1,2,3

1
Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research at the McKnight Brain Institute of the
University of Florida, PO Box 100256, Gainesville, FL 32610, USA
2
Department of Neuroscience, Center for Traumatic Brain Injury Studies at the McKnight Brain Institute of the University
of Florida, PO Box 100244, Gainesville, FL 32610, USA
3
Banyan Biomarkers, Inc., 12085 Research Dr., Alachua, FL 32615, USA

Abstract: Neuroproteomics entails wide-scope study of the nervous system proteome in both its content
and dynamics. The field employs high-end analytical mass spectrometry and novel high-throughput
antibody approaches to characterize as many proteins as possible. The most common application has been
differential analysis to identify a limited set of highly dynamic proteins associated with injury, disease, or
other altered states of the nervous system. Traumatic brain injury (TBI) is an important neurological
condition where neuroproteomics has revolutionized the characterization of protein dynamics, leading to
a greater understanding of post-injury biochemistry. Further, proteins of altered abundance or post-
translational modifications identified by neuroproteomic studies are candidate biochemical markers of TBI.
This chapter explores the use of neuroproteomics in the study of TBI and the validation of identified
putative biomarkers for subsequent clinical translation into novel injury diagnostics.

Keywords: proteomics; neuroproteomics; brain injury; neurotrauma; TBI

Introduction to TBI is estimated that between 1.6 and 3.8 million


sports-related TBI incidents actually occur in the
Traumatic brain injury (TBI) is neurotrauma US as compared with the 300,000 reported. TBI
caused by a mechanical force applied to the head costs the American economy an estimated $60 bil-
(Wang et al., 2004). There are approximately 1.4–2 lion per year in medical expenses and lost produc-
million TBI incidents annually in the United tivity (Finkelstein et al., 2006). All told, TBI
States, resulting in 1.1 million emergency depart- represents a major public health problem with
ment visits, 235,000 hospitalizations, 90,000 left significant societal and economic consequences
with long-term disabilities, and 50,000 deaths (Sosin et al., 1995; Wang et al., 2004; Yi and
(Langlois et al., 2006b; Ragnarsson, 2006). Yet Hazell, 2006). Despite this, there remains no ap-
the true burden of TBI may not be reflected in proved therapy for TBI (Narayan et al., 2002;
these numbers as many TBI incidents go unre- Wieloch and Nikolich, 2006).
ported or are not classified as TBI. For example it TBI results from a number of etiologies (Fig. 1)
primarily affecting male (78.8% of incidents) ado-
Corresponding author. Tel.: +1-352-392-8060; Fax: +1-352- lescents and young adults (31.7% of incidents are
392-2579; E-mail: aottens@mbi.ufl.edu among 15- to 24-year olds). In fact, TBI is

DOI: 10.1016/S0079-6123(06)61029-7 401


402

Fig. 1. Breakdown of reported TBI etiologies in the United States between 1995 and 2001 (Langlois et al., 2006a).

considered the leading cause of death and disabil- To this end, the present strategy is to develop
ity in children and young adults. A sharp rise in TBI diagnostics ahead of new therapy trials. For
TBI incidence among, typically young, American this purpose, it is important to have accurate
combat casualties recently added to this equation models of TBI that mimic human sequelae and
(Warden, 2006). Of Iraq and Afghanistan conflict outcomes (Narayan et al., 2002; Cernak, 2005).
casualties evacuated to the Walter Reed Army Several experimental TBI models have been intro-
Medical Center as of January 2006, 28% (1700 duced to investigate the pathobiology and mole-
cases) had TBI. An unfortunate benefit to in- cular dynamics of brain injury. Generally, a
creased incidents of military head trauma has been mechanical force is used to impact or torque the
a recent boost in funded research for development brain. The primary injury is comprised of contu-
of new neurotrauma diagnostics and therapies. sion and hemorrhage, characterized by rapid cell
This is an important opportunity as military death followed by diffuse axonal injury. Conse-
medicine has historically driven medical advance- quently, brain function is rapidly disrupted at the
ments in trauma, while progress in head injury site of injury and in distal regions interconnected
treatment has been slow to proceed (Narayan through white matter tracts, leading to likely def-
et al., 2002). icits in higher cognitive and vital sensory-motor
A major limitation is that TBI continues to be functions (Wieloch and Nikolich, 2006).
difficult to assess clinically, even with modern im- Secondary injury occurs in proximal brain re-
aging techniques such as magnetic resonance im- gions marked by massive edema, intracranial
aging (MRI) and computer tomography (CT), hemorrhages through blood-brain barrier (BBB)
which are limited by sensitivity, specificity and disruption, and further cell death and axonal in-
availability (Ottens et al., 2006). These diagnostics jury. Those biochemical events are mediated by
often do not provide reliable prognoses, particu- neurotoxic increases in excitatory amino acids
larly for mild TBI patients that account for 80% of (EAAs), ischemic factors (free radicals/oxidative
those who suffer lifelong impairments (Alexander, damage), proteolysis, and an inflammatory re-
1995). There is clearly a need for more defined TBI sponse mediated by microglia and astroglia acti-
diagnostics for patient management and progno- vation and proliferation (McIntosh et al., 1998;
sis. Likewise, these tools are needed as quantifiable Wieloch and Nikolich, 2006). A major causal fac-
outcome measures for development of new thera- tor for secondary degeneration is the disruption of
pies, a recognized deficiency of past TBI thera- intracellular calcium homeostasis following EAA
peutics studies (Denslow et al., 2003). glutamate overflow into extra-synaptic regions
403

(glutamate excitotoxicity), and the additional 2003; Kobeissy et al., 2006). A number of papers
glutamate release induced by a calcium influx. have been published recently discussing the appli-
These biochemical events can lead to prolonged cation of neuroproteomics in the field of neuro-
cell membrane depolarization, ATP depletion and trauma (Buonocore et al., 1999; Jenkins et al.,
subsequent ionic imbalance, manifested clinical as 2002; Satchell et al., 2003; Conti et al., 2004;
an increase in intracranial pressure (ICP) (Kupina Haskins et al., 2005; Kobeissy et al., 2006;
et al., 2003; Yi and Hazell, 2006). Furthermore, Kochanek et al., 2006). One major application of
secondary insults continue for days, potentially neuroproteomics lies in its ability to identify bio-
worsening a patient’s condition with unpredicted markers that are of great value in providing in-
results (coma or death). sights into injury severity, patient management,
Several studies have demonstrated that acti- and outcome. In this chapter we will explore the
vated cysteine proteases are major intracellular ef- application of neuroproteomics to TBI, discussing
fectors in neurotrauma pathology. These proteases the relevant preclinical animal models, the mass
act on a number of substrates including cytoske- spectrometry and antibody based neuroproteomic
letal, cytosolic, synaptic and membrane proteins, methods employed, and reviewing validation and
which upon proteolysis yield signature fragments translation of neuroproteomic data into viable
indicative of neuronal cell death dynamics (Farkas clinical biomarkers of TBI.
et al., 2005). Necrotic oncosis is mediated by cal-
pains activated by the inrush of calcium (Kupina
et al., 2003). Apoptosis is activated via more in- Modeling TBI in animals
volved caspase pathways during secondary injury
(Wennersten et al., 2003). Intrinsic and extrinsic Traumatic brain injury (TBI) has been modeled in
apoptotic effectors invoke caspase-8 or -9 activa- animals to replicate clinical trauma in a controlled
tion that activates the executioner caspases (pri- experimental setting. A variety of animal models
marily caspase-3) (Rink et al., 1995; Ottens et al., are available, and careful selection is required to
2006). Other molecular events occur tangential to determine which will work best with the scientific
cell death. Shifts in pro-survival and pro-death question at hand. A mechanical force is utilized to
signals (Rink et al., 1995; Shimamura et al., 2005) inflict injury, which must be controlled, reproduc-
can result in cells that survive but are susceptible ible and quantifiable. Invariably, the intensity of
to degeneration or later death. Another feature the mechanical force should dictate the severity of
of TBI is the marked long-term accumulation of injury. The injury should also be comparable to
proteins in damaged cells. Some of these proteins, the human injury, modeled with a direct correla-
such as synuclein (Syn), amyloid-b (Ab), and tion between injury and outcome.
neurofilament (NF), are major pathological com- The following section will cover the basic mecha-
ponents of neurodegenerative diseases such as nisms, advantages and limitations of current TBI
Alzheimer’s and Parkinson’s disease (Smith animal models. A summary of the models can be
et al., 2003). found in Fig. 2, which is based on a classification
A detailed understanding of molecular dynamics scheme from Ommaya (1995; see also Ommaya et
following TBI can be of high value to elucidate al., 2002) with mechanical paradigms added by
injury mechanism for diagnostics and treatments Cernak (2005) in his recent review.
(Shimamura et al., 2005). Recently, novel ne- Static and dynamic brain trauma differs from
uroproteomic approaches have been utilized to each other based on amplitude, duration, velocity
expand our understanding of TBI molecular mech- and acceleration. Static brain trauma is studied
anisms. Neuroproteomics is a new field that pro- utilizing models that have a defined amplitude and
vides a wide-scope, more global analysis of protein duration. A simple example of a static injury
dynamics to investigate differential alteration in model would be to crush a region of the brain
protein levels, modifications and functions post using a mechanical force (e.g., a vice) for a defined
neurotrauma (Jenkins et al., 2002; Denslow et al., period of time. If velocity and acceleration were
404

Fig. 2. Schematic representation of in vivo experimental models of traumatic brain injury.

factored into the static model then it would be- case special attention has been paid to the rela-
come dynamic. tionship between brain mass and acceleration in
Dynamic brain injury is the result of a mechan- order to regulate the inertial effects that determine
ical force with a well-defined amplitude, duration, the extent of damage. These models have utilized
velocity, and acceleration. Dynamic brain injury miniature swine, rabbits, rats and primates as test
can be sub-classified into indirect and direct inju- subjects. Unconstrained head motion is standard
ries. Indirect dynamic brain injury occurs from in acceleration models; however, it has been noted
blast, explosive phenomena in which the entire that unconstrained head motion can lead to sig-
body is affected by the kinetic energy released. nificant variability in the outcome. Non-human
Recent studies have noted that exposure to blast primates and miniature swine acceleration models
waves can cause peripheral blast trauma to the have been observed to produce injuries that are
brain without inflicting external cranial trauma strikingly similar to those found in human patients
(Saljo et al., 2000; Cernak et al., 2001). Animal (Povlishock et al., 1994). Often the result is profuse
models consist of exposing a test subject, often a axial injury that causes the test subject to go into a
rodent, to a blast wave with an over-pressurization prolonged coma. Rotational acceleration models
range of 154–340 kPa. Importantly, models of ro- are disadvantaged by the excessive cost involved in
dent blast exposure have produced similar out- the equipment and use of large animals, as well as
comes to studies of human blast injury patients the lack of adequate outcome measures of torque
(Cernak et al., 1999, 2000). induced brain injury.
Direct dynamic brain injury can be further clas- Impact dynamic head injury models reproduce
sified into acceleration and impact models. Accel- high energy concussive or penetrating injuries that
eration brain injury models study the effects of are either open or closed headed. Concussive
brain movement within the skull without direct closed head injury models are utilized in order to
contact with the brain tissue. Rapid head rotation replicate human concussive and diffuse brain in-
is most commonly used to simulate the effects of jury. There have been two controlled concussion
forcing the brain into the side of the skull. In this (CC) models put forth that utilize cats and rats as
405

test subjects (Tornheim and McLaurin, 1981; 1994; Povlishock et al., 1994), which allows for
Tornheim et al., 1990; Goldman et al., 1991). In finer control of the mechanical force than with
both, a force is applied to either the coronal struc- FPI. The use of pneumatics however practically
ture (cats) or the skull midline (rat). The cat model limits this model for use with rodents such as fer-
has been found to be suitable for the study of cere- rets, rats and mice. CCI is also considered to de-
bral contusion and resulting edema. The downside liver a more focused injury than FPI. The model
to the cat model is that the inflicted injury usually successfully replicates the clinical pathobiology of
results in hemorrhage, which limits the ability brain injury with skull deformation and cortical
of the researcher to use this model for the study compression. CCI is widely used for the study of
of molecular or cellular mechanisms related exclu- molecular and cellular mechanisms related to TBI
sively to the impact. The rat model has been a solid and is a useful tool in the development of novel
model for the study of mild to moderate injury therapies.
depending on the amount of force used. It also The weight drop model is another widely uti-
does not cause skull fracture or significant hem- lized direct impact model, simulating an impact
orrhage, and is comparable to similar injuries acceleration injury by dropping a brass weight
found in human TBI. These factors, as well as the onto a fixed target up against the surgically ex-
low cost in using rats, make this model a prime posed brain of a rat or mouse. In this model there
method for the study of both TBI mechanisms and is a direct correlation between the severity of in-
therapy. jury and the weight size and height from which it is
Open head contusion injuries apply a mechanical dropped (Marmarou et al., 1994; Piper et al., 1996;
force directly to the brain through a hole in the De Mulder et al., 2000). Historically, this model
skull (removed by craniotomy). An example is the has suffered from a few technical limitations. Al-
fluid percussion injury (FPI) model where a fluid though the weigh is dropped though a Plexiglas
pressure pulse is delivered to the revealed dura to tube, there is still a small amount of lateral mo-
cause brain deformation (Erb and Povlishock, tion. There is also the likelihood of a second im-
1988; Faden et al., 1989, 2003; McIntosh et al., pact as the weight rebounds, since it is dropped
1994; Conti et al., 1998; Albensi et al., 2000). FPI is freely. More recent improvements to the weight
a popular model for use with many different spe- drop model have utilized a laser guided, air-driven,
cies (rats, mice, cats, pigs, rabbits, dogs, and high-velocity impactor, which is more controlled
sheep). The model has been adapted to apply ei- and reproducible then earlier setups (Cernak et al.,
ther a central force to the vertex at the midline 2004). The model is able to replicate the biochem-
between bregma and lambda, or a lateral force over ical and neurological aspects of TBI in rats and
the left parietal bone between bregma and lambda mice as observed in clinical cases.
to produce different desired outcomes. Craniotomy High-velocity penetrating injury models have
position is critical for impairment determination been designed in order to study the effects of pro-
and reproducibility. While both central and lateral jectile injury to the brain. Initially models were
FPI have comparable pathobiology, it has been designed for use in cats and sheep (Carey et al.,
observed that central FPI causes death due to au- 1989, 1990; Finnie, 1993; Carey, 1995). In each
tonomic dysfunctions at hypothalamic levels. Thus, case, a small projectile (relative to the animal’s
lateral FPI can reproduce an injury with clinical size) was used to inflict a head wound on the
characteristics of contusion without skull fracture, anesthetized subject. Each of the models produced
but suffers from increased morbidity due to dis- sever tissue damage as well as brain displacement.
proportionate brainstem injury and limited control While these models are both useful in the histo-
of injury magnitude. pathological features of projectile wounds, they
Controlled cortical impact (CCI) is a more re- are not practical for molecular studies due to the
cent open-head concussive model that applies a use of large animal test subjects. Williams et al.
mechanical force to the intact dura by means of a (2005) recently developed a rat penetrating ballis-
pneumatic piston (Dixon et al., 1991; Gennarelli, tic brain injury (PBBI) model that mimics aspects
406

of a high energy bullet wound as demonstrated by Available technology has become quite good for
histopathological and physiological studies. analyzing relative abundance change (differential
Through the insertion of a rapidly inflatable analysis) among moderately or highly expressed
probe, they are able to reproduce the permanent proteins; however, extremes in dynamic range (1010
injury track left by a bullet, as well as the temporal orders) confound complete proteome coverage.
cavity generated by the dissipating energy from the Further, proteins encoded only by some 30,000
penetrating projectile. genes transcribe to multiple isoforms and can be
In all, there are a number of preclinical animal post-translationally modified by some 400 different
models that adequately reproduce facets of clinical processes (Morrison et al., 2002; Patton, 2002)
TBI, though the pathobiology of each model will producing a large array of functionally distinct
differ somewhat in mirroring the reality of human biomolecules. Though progress has been made in
TBI incidents. In the end, these models are useful characterizing the more common of these proc-
for generating preclinical tissue samples in a repro- esses, there remains no adequate means to capture
ducible, controlled manner for use in neuro- the fully functionalized state of proteins, let alone
proteomic studies reviewed in the following two in a wide-scale, high-throughput paradigm. To
sections. complicate matters further, any given proteome is
highly dynamic, able to shift in its protein compli-
ment in a matter of minutes to hours. As yet pro-
Neuroproteomic analysis with mass spectrometry in teomic experiments have been limited to temporal
TBI snap shots due to resource constraints.
In light of the challenges, progress has been
The introduction of peptide analysis by mass made in characterizing neuroproteome dynamics
spectrometry in combination with bioinformatics initiated by TBI — the primary motivators being
for data processing has revolutionized the field to identify biochemical markers for development
of proteomics. Figure 3 illustrates how the field of diagnostics and potential therapeutic avenues.
of proteomics (and genomics) has been driven Prominent protein dynamics are more easily trans-
by technology. By definition proteomics entails lated into clinical diagnostics; therefore, the capa-
wide-scale protein analysis made possible by bilities of current proteomic technologies have
high-throughput methods and instrumentation to coalesced well with the monitoring and prognostic
approach characterization of the complete pro- medical needs in neurotrauma (Denslow et al.,
teome, or more feasibly a distinct subset such as 2003; Wang et al., 2005).
the neuroproteome, the protein content of the A general platform has emerged for performing
central nervous system (CNS). What is also ap- mass spectrometry based differential proteomics
parent in Fig. 3 is that further, significant techno- for TBI research (Fig. 4), though the details for
logical advancements are poised to occur in each step do vary. The first important considera-
proteomics, which will continue to revolutionize tion is, what adjustments must be made to the
the field, bringing us closer to complete ne- proteomics experiment to suit the sample type
uroproteome characterization. (e.g., neuronal culture, brain tissue, cerebrospinal
Though mass spectrometers have been used to fluid (CSF)). Confounding factors introduced by
characterize macro biomolecules (e.g., polypep- the mechanism of insult and sample processing
tides) since the early 1990s (Kaufmann, 1995; must be controlled, generally by including
Nguyen et al., 1995), it has only been in the last matched naı̈ve or sham controls. Biological vari-
few years that instrument manufacturers have be- ability also will increase with sample heterogeneity
gun to cater to proteomic applications. Since, the (Molloy et al., 2003). Ideally, experiments should
proteomics market has swelled with a discernable be processed multiple times with separate samples,
rise in products and software and correlated pub- though this is often too costly; thus, subsequent
lications. Despite this, investigators remain limited validation of individual protein dynamics is often
in their ability to characterize a given proteome. a more manageable scenario.
407

Fig. 3. Timeline and publication history of proteomics vs. genomics. PubMed entries (searched on Oct. 4, 2006) for genomic studies
(search phrase ‘‘genome or genomics’’) and proteomic studies (search phrase ‘‘proteome or proteomics or 2D-PAGE’’) are normalized
against the number of all entries per publication year. The graph is overlaid by key technological advancements that have shaped each
field.

After extraction, proteins must be resolved from roughly 5000 proteins (gel spots in Fig. 4). The
one another. The most common approach to pro- neuroproteome contains more than this number of
teome separation in general and in TBI research proteins though, and does not resolve evenly
(Buonocore et al., 1999; Jenkins et al., 2002; across the separation space; thus, it is important
Satchell et al., 2003; Conti et al., 2004; Kochanek to have additional dimensions of separation, as
et al., 2006) has been two-dimensional polyacryl- afforded by peptide chromatography and mass
amide gel electrophoresis (2D-PAGE). Newer spectrometry.
multidimensional separation approaches have Mass spectrometry provides accurate and pre-
been introduced recently, overcoming some limi- cise mass-to-charge (m/z) information that can be
tation of 2D-PAGE (Ottens et al., 2006). For aligned against an established, species specific,
example, we have opted to use ion-exchange liq- protein database(s) that has been processed in
uid chromatography in tandem with gel elect- silico into sequence specific peptides. A general
rophoresis, in an approached called CAX-PAGE, caveat to mass spectrometry is that proteins must
to enhance differential comparison without hav- be fragmented into polypeptides (generally from 7
ing to mix samples and to extend the mass range to 20 amino acids) in order to permit accurate
(Ottens et al., 2005; Kobeissy et al., 2006). How- m/z assignment, though recent progress has been
ever, 2D-PAGE remains the most common ap- made to alleviate this limitation (Kelleher, 2004;
proach, having been established over 30 years Bogdanov and Smith, 2005). Nevertheless, mass
(Beranova-Giorgianni, 2003). 2D-PAGE involves spectrometry provides a degree of selectivity as yet
resolving proteins by their isoelectric point (pI) unrivalled for the identification of proteins on a
first and then by relative molecular mass (Mr). The large scale (technologies such as Edmund degrada-
peak capacity for each dimension (between 50 and tion or immunological assays are also selective,
100) is then multiplied to provide separation of but on a more limited scale). Time-of-flight mass
408

Fig. 4. Neuroproteomic analysis scheme using mass spectrometry. The general platform for neuroproteomic research is outlined in six
steps: (1) extract proteins from the biological sample and matched control; (2) resolve proteins via multidimensional separations; (3)
isolate target (e.g., differential) proteins and digest into tryptic fragments; (4) resolve peptides by chromatography (used with tandem
mass spectrometry); (5) analyze with mass spectrometry; and (6) search mass spectra against protein database to identify target
proteins.

spectrometers have often been used to identify peptide mass mapping (Aebersold and Mann,
proteins by precise and accurate measurement of 2003), multiple peptide m/z values (at least 4 or
peptide m/z values alone (no tandem mass spectra more) combined with the Mr and pI of the orig-
sequence information). In this approach, called inating protein (from 2D-PAGE) is selective
409

enough to provide identification. However, it is of- proteins, even following multidimensional protein
ten difficult to attain accurate measurement of 4 or separation. It therefore must be noted that it can
more peptides per protein, particularly for those of still be difficult to associate a single protein with
lower abundance. Conti et al. (2004) encountered a given differential gel spot or band even after
this limitation with lower abundant differential tandem mass spectrometry due to protein co-
proteins displayed on 2D-PAGE separation of migration. To help ascertain the differential nature
control and TBI CSF. Further, peptide mass map- of a protein, peptide tandem mass spectrometry
ping will not work well for proteins that have been data can also be quantitative. The number of
modified and therefore shifted in Mr or pI value. peptides per protein is a useful relative quantita-
The added selectivity afforded by tandem mass tive measure of differential abundance (Sanders
spectrometry alleviates these concerns where only et al., 2002; Peng et al., 2004; Ottens et al., 2005;
the sequence of the originating protein is needed to Kobeissy et al., 2006), e.g., six MAP2 peptides for
provide an accurate match to data of a single pep- naı̈ve indicates greater abundance than one pep-
tide (condoned when the data are of excellent spec- tide for TBI (Fig. 5). This information can be used
tral quality, though data for two or more peptides for data reduction in eliminating unlikely candi-
per protein are generally advised). Tandem mass date proteins, those with no apparent change or
spectrometry coupled with peptide chromatogra- opposite change in quantity, and to confirm the
phy is more resource intensive, and therefore is of- differential nature of the final protein list.
ten reserved for identification when peptide mass In all, mass spectrometry combined with protein
mapping fails, as used by Kochanek et al. (2006) and peptide separations is an effective platform
for differential analysis of control and TBI tissue. for identifying protein dynamics following TBI
Tandem mass spectrometry provides multidi- (Satchell et al., 2003; Conti et al., 2004; Haskins
mensional data via the use of gas-phase molecular et al., 2005; Kobeissy et al., 2006; Kochanak et al.,
fragmentation — traditionally through collision 2006). To date, a number of candidate biochemical
induced dissociation (Abersold and Mann, 2003), markers of TBI have been identified in a hand-
though more recent techniques such as electron ful of neuroproteomic studies, which are in the
capture dissociation (Cooper et al., 2005) and processes of validation and clinical translation.
electron transfer dissociation (Good and Coon, Characterization of global protein dynamics and
2006) have shown some improved functionality. pathways following neurotrauma is more elusive,
The intact peptide (parent ion) m/z value is analy- and will require more refined and exhaustive
zed in the first stage of mass spectral analysis. In proteomic experiments (further technological ad-
the second stage, the peptide ion is broken up at vancement will ultimately be required). Greater
integral points between amino acids into sequence protein and/or peptide separation, improved sen-
specific fragment ions (daughter ions), which are sitivity, and more refined m/z assignment of larger
mass analyzed to produce a tandem mass spectrum polypeptides will drive the neuroproteomics field
(Fig. 5). Fragmentation efficiency is not uniform forward, which will further the understanding of
between all amino acids of a peptide; however, the protein chemistry of brain injury and avenues
enough information is generally attained to corre- for new treatments.
late the data with a database derived peptide se-
quence having a matched parent ion m/z value.
Bioinformatic software is used to automatically TBI neuroproteomics by antibody arrays and high-
process tandem mass spectra against a protein da- throughput blotting
tabase(s). A score and/or a statistical measure (XC
and P (pep), respectively in Fig. 5) of how well the Alternative to the mass spectrometry-based pro-
data matches with the peptide sequence is pro- teomic approach, several antibody-based appro-
vided. The originating protein(s) is then listed with aches are also emerging through the availability
all associated peptides. As illustrated in Fig. 5, ex- of various antibody array or protein array plat-
cised gel bands or spots likely contain multiple forms (Zyomyx protein Biochips, BD PowerBlot
410

Fig. 5. Bioinformatics of mass spectrometry data. Illustrated is an example of the bioinformatic output of processed tandem mass
spectrometry data of one differential target from our recent TBI neuroproteomics study (Kobeissy et al., 2006). Corresponding naı̈ve
and TBI gel bands at position 23A (first differential band in the 23 protein fraction) were excised, digested and analyzed by reversed
phase liquid chromatography–tandem mass spectrometry. Peptide tandem mass spectra (peptide probability, po0.001) indicated the
presence of the protein MAP2. Protein coverage was lower in the TBI sample than in naı̈ve control (1 and 6 peptides, respectively),
correlating this protein with the less intense TBI gel band.

and BD Clontech antibody microarrays 500) labeled with green and red fluorescent Cyanine dye
(Moody et al., 2001; Graslund et al., 2002; James, (Cy-3 and Cy-5) pairs. An equal amount of each is
2002; Kusnezow and Hoheisel, 2002). The follow- mixed and applied to the slide. After washing, the
ing is a brief description of several of these tech- fluorescence intensity for Cy-3 and Cy-5 are then
nologies as applicable to TBI research, primarily respectively measured. The bound Cy-3 to Cy-5
biomarker development. ratio will then represent a relative differential
quantitative measure of each antigen. The method
was pioneered with a cytokine panel due to the
Antibody microarray multitude of cytokine antibodies available. Using
this kit, pro-inflammatory cytokines such as IL-1b
Antibody microarrays are modeled after DNA and TNF-alpha were found to be elevated in CSF
microarrays. A collection of highly specific pri- following TBI (Shiozaki et al., 2005), a general
mary antibodies (to different protein antigens) is indication for the involvement of CNS inflamma-
printed on a standard-size glass slide (e.g., BD- tion following neurotrauma. A drawback of this
Clontech microarray 500) (Gu et al., 2006). Bio- method is the technical challenge of even sample
logical samples (brain tissue lysate, CSF or serum/ labeling. As an alternative, it is also possible to
plasma) from control and treatment groups are construct a ‘‘sandwich format’’ antibody array in a
411

partitioned (e.g., 96-well) plate. In this case, two confirmed several novel TBI-linked proteolytic
highly specific primary antibodies are required to substrates (Fig. 6), including betaII-spectrin, stria-
detect each protein antigen. While the coating step tin, synaptotagmin-1, synaptojanin-1 and NSF
of the first set of antibodies (capture antibody) is (N-ethylmaleimide sensitive fusion protein) (Liu
the same as above, the proteins in the biological et al., 2006).
samples are not labeled or mixed. The second set In summary, antibody-based neuroproteomic
of detecting antibodies is applied to the plate, techniques offer several attractive features. First,
forming an antibody–antigen–antibody ‘‘sand- instrumentation requirements for antibody-based
wich’’. Finally, the abundance of the antigen is platforms are generally far less formidable in cost
quantified by measuring the amplifiable detection and operation than those for mass spectrometry
antibody in each well (Huang, 2001). based approaches. Second, since the target antigen
is known for each antibody probe, initial protein
identification is rapid and straightforward, and as
High-throughput immunoblotting a result the bioinformatic component of this ap-
proach is far less labor-intensive. Third, validation
Another novel technique called high-throughput is again relatively simple, since an antibody to the
immunoblotting (HTPI) has found some recent target is already available. One does need to be
utility for neuroproteomics research (Malakhov cautious about antibody cross reactivity to unre-
et al., 2002; Yoo et al., 2002; Lorenz et al., 2003). lated protein(s) that could give rise to false positive
This proteomic approach involves multiplexing identification. One major challenge of this ap-
traditional Western blot analysis. Using our own proach is the lack of available antibodies for all
recent TBI study as an example (Liu et al., 2006), 30,000 (approx.) proteins in the average mamma-
two pooled hippocampal lysate samples, naı̈ve lian proteome, let alone the multitude of post-
and CCI (200–1000 mg), were prepared and sub- translational forms, making this approach less
jected to two sets of five SDS-polyacrylamide gels than exhaustive (Kusnezow and Hoheisal, 2002).
(13  10 cm). Proteins in the gel were transferred to Across-species reactivity is another issue, i.e., can
a blotting membrane, which was then clamped to a an antibody raised against a human protein anti-
blotting manifold that isolates 40 longitudinal gen cross detect the rat protein homologue? Fur-
channels. A cocktail of between 4 and 6 antibodies thermore, the antibodies used are not standardized
was added to each channel and allowed to hybrid- in their affinity to target antigens; however, the
ize for 1 h at 371C. After washing, a secondary Human Proteome Organization (HUPO) is taking
antibody conjugated to Alexa680 fluorescent dye steps to standardize specific antibody sets for the
was added, then quantified by an Odyssey Infrared whole human proteome (Haab et al., 2005).
Imaging system (LI-COR). Each of the antibodies
per channel is well characterized and resolved from
one another by relative molecular mass, thus Validation of proteomic data for TBI biomarker
achieving coverage of 1000 proteins over five blots. development
As HTPI is still a Western blot in principle, it
is useful for detecting protein modifications that In recent years, the term proteomics is often men-
alter gel mobility (e.g., proteolytic breakdown tioned together with biomarker discovery, as pro-
products). Using HTPI, we identified more than teomic studies have the capability of identifying
40 decreased proteins following CCI relative to unique and unobvious protein biomarkers from
naı̈ve, many of which were seemingly subjected tissue or biofluids obtained from human diseased
to calpain and/or caspase proteolysis. Another patients or from animal models of disease and
positive feature is the ease for hit validation, as disorders. Biomarkers of acute brain injury have
the same antibodies used for HTPI can also be also attracted a lot of recent research inter-
used for subsequent conventional Western blot est (Wang et al., 2006). First, it is commonly
confirmation. In this manner, we successfully recognized that better patient monitoring and
412

Fig. 6. Example of TBI differential TBI proteome as analyzed by HTPI. HTPI Template A is shown for naı̈ve rat hippocampus (upper
panel) and the controlled cortical impact injury counterpart (lower panel). Molecular mass markers (lane 40) are indicated on the right.
Thirteen proteins in template A were reduced in average intensity (abundance) after TBI (solid boxes in upper panel). In addition,
several breakdown products were observed (dotted boxes in lower panel). Two proteins found to be up-regulated in TBI were CASK
(A3) and Psme3 (A29) (in dotted box). Adapted with permission from Liu et al. (2006).

management often leads to better clinical outcome. predictive of disease progression and treatment
Thus, a simple point-of-care (POC) CSF- or response (Lesko and Atkinson, 2001; Pineda et al.,
blood-based diagnostic test would be extremely 2004; Wang et al., 2006). In practice, proteins
useful, and significantly less expensive than any identified from differential neuroproteomic studies
brain image scanner (such as computed tomo- of brain injury (human or animal models) can
graphy). Second, numerous failures in clinical drug only be considered potential biomarkers for
trials for the treatment of ischemic strokes and brain injury, particularly if the goal is to develop
TBI over the last 10–15 years point to the difficulty a CSF- or blood-based brain injury biomarker
of translating preclinical successes to clinical suc- test. This is even truer of candidate proteins iden-
cesses. One remedy of the situation is the develop- tified from differential tissue lysate analysis, not in
ment of robust quantitative biomarkers that are biofluids.
413

Here are several criteria for an ideal biomarker phosphorylated form or breakdown products,
of brain injury: such as c-tau or alphaII-spectrin breakdown prod-
ucts (SBDPs) (Pineda et al., 2004). Afterwards,
i. Levels of the biomarker in a relevant biofluid
antigen affinity purification is highly recom-
(CSF and/or blood) correlate with disease
mended. Finally, some form of the antigen (puri-
severity, e.g., Glasgow Coma Score (GCS)
fied native protein or recombinant protein) must
for TBI.
be available for the development and testing of the
ii. Levels of the biomarker correlate with clin-
antibody, as well as for use to generate a standard
ical outcome, e.g., as measured by the Ex-
curve for absolute quantification.
tended Glasgow Outcome Scale (GOS-E).
What follows are iterations of trial and error to
iii. Biomarker signals are not confounded by
find the best antibody pair (capture and detection),
other non-brain injury neurological condi-
and the optimal assaying conditions to give the
tions.
best signal to noise ratio as well as the lowest de-
iv. Biomarker levels are responsive to therapeu-
tection limit. The term ELISA implies that the de-
tic interventions.
tection antibody is coupled to an enzyme-linked
To date, the most common validation technol- substrate to product system for signal amplifica-
ogy for neuroproteomics data to biomarker assay tion (e.g., alkaline phosphatase or horse radish
relies heavily on antibody assays, since antibodies peroxidase). An ideal biomarker ELISA assay
(both polyclonal and monoclonal) have the ability should strive to achieve high (490%) selectivity
to selectively identify and quantify a single protein (i.e., the ability to detect the presence of injury)
(antigen) of interest in a rapid, high-throughput and high (490%) specificity (i.e., the ability to
manner. Due to the maturity of immunological detect the absence of injury).
methods, antibodies-based diagnostic assays have A major challenge of brain injury biomarker
been refined almost to an art form, namely, en- assay development is the availability of high qual-
zyme-linked immunosorbant assays (ELISA). ity antibodies. On this front, there have been sig-
There are two forms of ELISAs: direct ELISA nificant developments in capturing and detecting
utilizes a single detection antibody; or sandwich agents beyond monoclonal antibodies, such as ap-
ELISA (swELISA) that requires the formation of tamers (Bunka and Stockley, 2006) and phage-
an immobilized capture antibody to protein anti- displayed single chain antibody fragments (Tomizaki
gen to detection antibody complex. For biomark- et al., 2005). Another major challenge is the de-
er-based diagnostic, swELISA is indeed the tection limits, since it is conceivable that the best
method of choice, since it provides antigen enrich- brain injury marker might be very low in abun-
ment that significantly improves signal fidelity. dance. ELISA sensitivity can be improved with an
The work flow for developing biomarker sw- enzymatic signal-enhancement method known as
ELISA assays is as follows. Once a putative bio- tyramide signal amplification (TSA). It is also
marker is identified and confirmed, one must apparent that technological advances in ELISA sen-
obtain a pair of compatible and high affinity anti- sitivity improvement will greatly benefit this field.
bodies. Antibodies from commercial sources are Animal models are commonly used for preclin-
explored first; however, custom-made antibodies ical biomarker validation, as they confer a number
are often needed. Custom antibodies are generally of advantages. One important advantage of pre-
raised against a peptide epitope-carrier protein clinical animal models is the ability to prioritize
complex, a recombinant protein or a fusion pro- the potential utility of biomarkers by use of rela-
tein (e.g., GST fusion protein) as the antigen, tively simple Western blot studies prior to com-
which is injected into host animals for an immune mitment of substantial time and resources to the
response (rabbit, chicken, goat, and mouse). Care development of more sensitive quantitative ap-
must be taken that the antibodies thus raised proaches such as ELISA. Employing a CCI model
will only recognize the protein isoform or post- of TBI, we capitalized on the ability to simultane-
translationally modified form of interest (e.g., ously characterize the appearance of biomarkers in
414

injured tissues as well as cerebrospinal fluid (Pike TBI. Thus, CCI reproduces important features of
et al., 2001, 2003). This often overlooked advan- calpain mediated pathology seen in human TBI.
tage allows the rigorous characterization of rela- The development of a clinical research platform
tionships between biomarker generation in injured for TBI poses considerable challenges. An impor-
brain tissue and appearance in other compart- tant limitation of clinical models, as typically im-
ments such as CSF and ultimately in blood. plemented, is the failure to incorporate systematic
Validations with preclinical animal models studies of the role of secondary insults and age,
make possible comparisons of distribution bioki- significant determinants of injury response in hu-
netics and the temporal profile of biomarker man TBI. Mild TBI and the frequent absence of
appearance in different, clinically relevant com- objective evidence of injury by computerized tomo-
partments such as CSF and blood, and their re- graphic studies or MRI are especially challenging.
lationship to the time of injury. These values can Neurological diagnosis of varying magnitudes of
include the half-life and time to maximal concen- mild or moderate TBI is unreliable, since the GCS
trations of biomarkers. A clear understanding of scale was developed primarily to assess more se-
the biokinetics of biomarkers is an important verely injured comatose patients. Outcome meas-
component and accurate interpretation of the ures are equally controversial and ambiguous.
clinical relevance of changes seen in biomarker However, greater progress has been made in estab-
levels. Apart from straightforward comparisons of lishing a clinical research platform for severe TBI.
appearance and temporal profile of biomarkers, Our program has recently completed the first
preclinical animal models provide important in- extensive examination of the potential clinical util-
formation on the potential relationship of bio- ity of biochemical markers of calpain and caspase-
marker levels to injury magnitudes, lesion volume 3 proteolysis in severe (GCS ¼ 4 8) TBI patients
and outcome. For example, our laboratory (Ringger (Pineda, 2006, in press). This research incorporated
et al., 2004) has confirmed that a biochemical important elements of a clinical research platform
marker of calpain proteolysis, the cytoskeletal pro- for biomarkers of TBI. Such a platform should be
tein alphaII-spectrin, when measured in CSF relia- able to test hypotheses about the potential clinical
bly correlated with the magnitude of CCI injury, utility and validity of biomarkers assessed, alone or
lesion volume and neurological outcome. in combination. Generally validation effects should
focus on whether biomarkers are reliably associ-
ated with the magnitude of brain injury, occurrence
Translation to clinical diagnostics of secondary insults and outcome. The platform
should allow rigorous determination of whether
The primary challenge facing translation of bio- appropriate markers can provide information criti-
markers to practical clinical diagnostics resides in cal for diagnosis, triage and management, as well
the approaches to confirm the clinical utility of as data on possible injury mechanisms and cellular
putative biomarkers of TBI identified and vali- localization of injury. In the research of Pineda
dated with preclinical animal models. Ideally, the (2006, in press), investigators were able to confirm
confirmation platform should integrate preclinical that a protein biomarker of calpain proteolysis
validation with clinical assessments of the poten- (degradation of alphaII-spectrin) measured in CSF
tial applicability of the candidate biomarkers. A was reliably associated with the magnitude of di-
direct comparison of biomarker generation bet- chotomized GCS on admission (GCS 3–6 vs. 6–8),
ween preclinical models and biomarker data from while a biomarker of caspase-3 alphaII-spectrin
human clinical studies allows investigators to gain proteolysis showed no such association. The bio-
considerable insight into the validity (or challenges marker of calpain proteolysis was also reliably as-
to the validity) of the employed preclinical animal sociated with secondary insult, (preadmission
models. For example, we have shown (unpublished hypotension) while a marker of caspase-3 pro-
data) similar profiles of alphaII-spectrin degrada- teolysis showed no significant relationship. The
tion by calpain in both CCI and severe human biomarker of calpain proteolysis, but not caspase-3
415

proteolysis was also differentially associated with In the near-term, whether by mass spectrometry or
outcome. Significantly higher level of a biomarker antibody-based arrays, neuroproteomics serves as
of calpain proteolysis was detected in patients within a high-throughput means to discover potentially
the first 24 h following injury who subsequently had novel biochemical markers of TBI. Significant ad-
a worse outcome (assessed by the dichotomized vances have also been made in developing strate-
GCS 6 months after injury). Importantly, calpain gies for preclinical and clinical validation of
proteolysis is primarily associated with oncotic biomarkers of TBI. While often limited by the fail-
necrosis while caspase-3 proteolysis is more promi- ure to incorporate the role of secondary insults, age
nently associated with apoptosis. Thus, these data and gender, preclinical animal models in combina-
collectively suggest that oncotic necrosis may more tion with neuroproteomic techniques have proven
importantly contribute to pathology in severe TBI invaluable for rapidly assessing the potential utility
than apoptosis. This hypothesis is consistent with of the relatively large numbers of biomarkers
the temporal profile of accumulation of these generated. While significant challenges remain in
biomarkers in CSF of severe TBI patients for the implementing preclinical platforms from mild
first 5 days following injury. Markers of calpain and moderate TBI, the establishment of effective
proteolysis were increased more than caspse-3 pro- platforms to validate biomarkers in severe TBI
teolysis, although caspase-3 proteolysis tended to be promises to define clinical useful subsets of bio-
more sustained. markers of TBI within the next few years. Looking
Our laboratory has recently begun an important forward, neuroproteomic technology will rapidly
series of studies that will significantly enhance our evolve to more effectively and more efficiently
ability to validate relationships between changes in characterize the neuroproteome as altered by TBI,
biomarkers and secondary insults following TBI. much as has been seen in the genomic revolution.
An important limitation in such studies has been This impending opportunity will confer unparal-
the absence of the ability to continuously record leled detail on the biochemical processes occurring
physiological data that would indicate the presence following TBI, and will inspire novel therapies tri-
of a secondary insult (e.g., reduced ICP, hypoten- als, to be evaluated with the biomarkers presently
sion, compromised tissue oxygenation). In fact, in development.
Hemphill et al. (2005) has recently documented
that studies conducted by collecting data of medi-
cal records, frequently missed secondary insults Disclaimer
documented by continuous recording of physio-
logical parameters. Thus, our program has recently KKW and RLH own stock, receive royalties from
initiated the systematic comparison of biomarker and are executive officers of Banyan Biomarkers
changes assessed from both CSF and blood with Inc. and as such may benefit financially as a result
physiological variables recorded continuously from of the outcomes of this research or work reported
severe TBI patient. The ultimate goal of these in this publication.
studies is to determine if changes in biomarker
levels can, in fact, predict onset of secondary in-
sults as well as document their occurrence. If so,
References
rapid assessment of biomarkers could provide criti-
cal information guiding management of severely Aebersold, R. and Mann, M. (2003) Mass spectrometry-based
injured TBI patients in the intensive care unit. proteomics. Nature, 422: 198–207.
Albensi, B.C., Knoblach, S.M., Chew, B.G., O’Reilly, M.P.,
Faden, A.I. and Pekar, J.J. (2000) Diffusion and high res-
olution MRI of traumatic brain injury in rats: time course
Closing remarks
and correlation with histology. Exp. Neurol., 162: 61–72.
Alexander, M.P. (1995) Mild traumatic brain injury: patho-
In conclusion, neuroproteomics has revolutionized physiology, natural history, and clinical management.
the exploration of protein dynamics following TBI. Neurology, 45: 1253–1260.
416

Beranova-Giorgianni, S. (2003) Proteome analysis by two-- Dixon, C.E., Clifton, G.L., Lighthall, J.W., Yaghmai, A.A. and
dimensional gel electrophoresis and mass spectrometry: Hayes, R.L. (1991) A controlled cortical impact model of
strengths and limitations. Trac-Trend Anal. Chem., 5: traumatic brain injury in the rat. J. Neurosci. Methods, 39:
273–281. 253–262.
Bogdanov, B. and Smith, R.D. (2005) Proteomics by FTICR Erb, D.E. and Povlishock, J.T. (1988) Axonal damage in severe
mass spectrometry: top down and bottom up. Mass Spect- traumatic brain injury: an experimental study in cat. Acta
rom. Rev., 24: 168–200. Neuropathol., 76: 347–358.
Bunka, D.H. and Stockley, P.G. (2006) Aptamers come of age Faden, A.I., Demediuk, P., Panter, S.S. and Vink, R. (1989)
— at last. Nat. Rev. Microbiol., 4: 588–596. The role of excitatory amino acids and NMDA receptors in
Buonocore, G., Liberatori, S., Bini, L., Mishra, O.P., Delivoria- traumatic brain injury. Science, 244: 798–800.
Papadopoulos, M., Pallini, V. and Bracci, R. (1999) Hypoxic Faden, A.I., Knoblach, S.M., Cernak, I., Fan, L., Vink, R.,
response of synaptosomal proteins in term guinea pig fetuses. Araldi, G.L., Fricke, S.T., Roth, B.L. and Kozikowski, A.P.
J. Neurochem., 73: 2139–2148. (2003) Novel diketopiperazine enhances motor and cognitive
Carey, M.E. (1995) Experimental missile wounding of the recovery after traumatic brain injury in rats and shows
brain. Neurosurg. Clin. N. Am., 6: 629–642. neuroprotection in vitro and in vivo. J. Cereb. Blood Flow
Carey, M.E., Sarna, G.S. and Farrell, J.B. (1990) Brain edema Metab., 23: 342–354.
after an experimental missile wound. Adv. Neurol., 52: Farkas, O., Polgar, B., Szekeres-Bartho, J., Doczi, T., Povli-
301–305. shock, J.T. and Buki, A. (2005) Spectrin breakdown products
Carey, M.E., Sarna, G.S., Farrell, J.B. and Happel, L.T. (1989) in the cerebrospinal fluid in severe head injury — preliminary
Experimental missile wound to the brain. J. Neurosurg., 71: observations. Acta Neurochir., 147: 855–861.
754–764. Finnie, J.W. (1993) Pathology of experimental traumatic
Cernak, I. (2005) Animal models of head trauma. NeuroRx, 2: craniocerebral missile injury. J. Comp. Pathol., 108: 93–101.
410–422. Finkelstein, E., Corso, P. and Miller, T. (2006) The Incidence
Cernak, I., Savic, J., Ignjatovic, D. and Jevtic, M. (1999) Blast and Burden of Injuries in the United States. Oxford Univer-
injury from explosive munitions. J. Trauma, 47: 96–104. sity Press, New York.
Cernak, I., Savic, V.J., Kotur, J., Prokic, V., Veljovic, M. and Gennarelli, T.A. (1994) Animate models of human head injury.
Grbovic, D. (2000) Characterization of plasma magnesium J. Neurotrauma, 11: 357–368.
concentration and oxidative stress following graded trau- Goldman, H., Hodgson, V., Morehead, M., Hazlett, J. and
matic brain injury in humans. J. Neurotrauma, 17: 53–68. Murphy, S. (1991) Cerebrovascular changes in a rat model of
Cernak, I., Vink, R., Zapple, D.N., Cruz, M.I., Ahmed, F., moderate closed-head injury. J. Neurotrauma, 8: 129–144.
Chang, T., Fricke, S.T. and Faden, A.I. (2004) The patho- Good, D.M. and Coon, J.J. (2006) Advancing proteomics with
biology of moderate diffuse traumatic brain injury as iden- ion/ion chemistry. Biotechniques, 40: 783–789.
tified using a new experimental model of injury in rats. Graslund, S., Falk, R., Brundell, E., Hooq, C. and Stahl, S.
Neurobiol. Dis., 17: 29–43. (2002) A high-stringency proteomics concept aimed for gen-
Cernak, I., Wang, Z., Jiang, J., Bian, X. and Savic, J. (2001) eration of antibodies specific for cDNA encoded proteins.
Ultrastructural and functional characteristics of blast injury- Biotechnol. Appl. Biochem., 35: 75–82.
induced neurotrauma. J. Trauma, 50: 695–706. Gu, Q., Sivanandam, T.M. and Kim, C.A. (2006) Signal sta-
Conti, A.C., Raghupathi, R., Trojanowski, J.Q. and McIntosh, bility of Cy3 and Cy5 on antibody microarrays. Proteome
T.K. (1998) Experimental brain injury induces regionally Sci., 4: 21.
distinct apoptosis during the acute and delayed post- Haab, B.B., Geierstanger, B.H., Michailidis, G., Vitzthum, F.,
traumatic period. J. Neurosci., 18: 5663–5672. Forrester, S., Okon, R., Saviranta, P., Brinker, A., Sorette,
Conti, A., Sanchez-Ruiz, Y., Bachi, A., Beretta, L., Grandi, E., M., Perlee, L., Suresh, S., Drwal, G., Adkins, J.N. and
Beltramo, M. and Alessio, M. (2004) Proteome study of Omenn, G.S. (2005) Immunoassay and antibody microarray
human cerebrospinal fluid following traumatic brain in- analysis of the HUPO Plasma Proteome Project reference
jury indicates fibrinogen degradation products as trauma- specimens: systematic variation between sample types and cali-
associated markers. J. Neurotrauma, 21: 854–863. bration of mass spectrometry data. Proteomics, 5: 3278–3291.
Cooper, H.J., Hakansson, K. and Marshall, A.G. (2005) The Haskins, W.E., Kobeissy, F.H., Wolper, R.A., Ottens, A.K.,
role of electron capture dissociation in biomolecular analysis. Kitlen, J.W., McClung, S.H., O’Steen, B.E., Chow, M.M.,
Mass Spectrom. Rev., 24: 201–222. Pineda, J.A., Denslow, N.D., Hayes, R.L. and Wang,
De Mulder, G., Van Rossem, K., Van Reempts, J., Borgers, M. K.K.W. (2005) Rapid discovery of putative protein biomark-
and Verlooy, J. (2000) Validation of a closed head injury ers of traumatic brain injury by SDS-PAGE-capillary liquid
model for use in long-term studies. Acta Neurochir. Suppl., chromatography-tandem mass spectrometry. J. Neurotrau-
76: 409–413. ma, 22: 629–644.
Denslow, N.D., Michel, M.E., Temple, M.D., Hsu, C.Y., Sa- Hemphill III, J.C., Barton, C.W., Marabito, D. and Manley,
atman, K. and Hayes, R.L. (2003) Application of proteomics G.T. (2005) Influence of data resolution and interpolation
technology to the field of neurotrauma. J. Neurotrauma, 20: method on assessment of secondary brain insults in neuro-
401–407. critical care. Physiol. Meas., 26: 373–386.
417

Huang, R.P. (2001) Simultaneous detection of multiple proteins immunoblotting. Ubiquitiin-like protein ISG15 modifies
with an array-based enzyme-linked immunosorbent assay key regulators of signal transduction. J. Biol. Chem., 278:
(ELISA) and enhanced chemiluminescence (ECL). Clin. 16608–16613.
Chem. Lab. Med., 39: 209–214. Marmarou, A., Foda, M.A., van den Brink, W., Campbell, J.,
James, P. (2002) Chips for proteomics: a new tool or just hype? Kita, H. and Demetriadou, K. (1994) A new model of diffuse
Biotechniques, Dec(Suppl): 4–13. brain injury in rats. Part I: Pathophysiology and biomechan-
Jenkins, L.W., Peters, G.W., Dixon, C.E., Zhang, X., Clark, ics. J. Neurosurg., 80: 291–300.
R.S.B., Skinner, J.C., Marion, D.W., Adelson, P.D. McIntosh, T.K., Juhler, M. and Wieloch, T. (1998) Novel
and Kochanek, P.M. (2002) Conventional and functional pharmacologic strategies in the treatment of experimental
proteomics using large format two-dimensional gel electro- traumatic brain injury. J. Neurotrauma, 15: 731–769.
phoresis 24 hours after controlled cortical impact in postnatal McIntosh, T.K., Yu, T. and Gennarelli, T.A. (1994) Alterations
day 17 rats. J. Neurotrauma, 19: 715–740. in regional brain catecholamine concentrations after experi-
Kaufmann, R. (1995) Matrix-assisted laser desorption ioniza- mental brain injury in the rat. J. Neurochem., 63: 1426–1433.
tion (MALDI) mass spectrometry: a novel analytical tool in Molloy, M.P., Brzenzinski, E.E., Hang, J., McDowell, M.T.
molecular biology and biotechnology. J. Biotechnol., 41: and VanBogelen, R.A. (2003) Overcoming technical varia-
155–175. tion and biological variation in quantitative proteomics. Pro-
Kelleher, N.L. (2004) Top-down proteomics. Anal. Chem., 76: teomics, 3: 1912–1919.
197A–203A. Moody, M.D., Van Arsdell, S.W., Murphy, K.P., Orencole,
Kobeissy, F.H., Ottens, A.K., Zhang, Z., Liu, M.C., Denslow, S.F. and Burns, C. (2001) Array-based ELISAs for high-
N.D., Dave, J.R., Tortella, F.C., Hayes, R.L. and Wang, throughput analysis of human cytokines. Biotechniques, 31:
K.K. (2006) Novel differential neuroproteomics analysis of 186–194.
traumatic brain injury in rats. Mol. Cell. Proteomics, 5: Morrison, R.S., Kinoshita, Y., Johnson, M.D., Uo, T., Ho,
1887–1898. J.T., McBee, J.K., Conrads, T.P. and Veenstra, T.D. (2002)
Kochanek, A.R., Kline, A.E., Gao, W.M., Chadha, M., Lai, Proteomic analysis in the neurosciences. Mol. Cell. Pro-
Y., Clark, R.S.B., Dixon, C.E. and Jenkins, L.W. (2006) Gel- teomics, 1: 553–560.
based hippocampal proteomic analysis 2 weeks following Narayan, R.K., Michel, M.E., Ansell, B., Baethmann, A.,
traumatic brain injury to immature rats using controlled Biegon, A., Bracken, M.B., Bullock, M.R., Choi, S.C.,
cortical impact. Dev. Neurosci., 28: 410–419. Clifton, G.L., Contant, C.F., Coplin, W.M., Dietrich, W.D.,
Kupina, N.C., Detloff, M.R., Bobrowski, W.F., Snyder, B.J. Ghajar, J., Grady, S.M., Grossman, R.G., Hall, E.D.,
and Hall, E.D. (2003) Cytoskeletal protein degradation and Heetderks, W., Hovda, D.A., Jallo, J., Katz, R.L., Knoller,
neurodegeneration evolves differently in males and females N., Kochanek, P.M., Maas, A.I., Majde, J., Marion, D.W.,
following experimental head injury. Exp. Neurol., 180: 55–73. Marmarou, A., Marshall, L.F., McIntosh, T.K., Miller, E.,
Kusnezow, W. and Hoheisel, J.D. (2002) Antibody microar- Mohberg, N., Muizelaar, J.P., Pitts, L.H., Quinn, P.,
rays: promises and problems, 33: S14–S23 Riesenfeld, G., Robertson, C.S., Strauss, K.I., Teasdale, G.,
Langlois, J.A., Rutland-Brown, W. and Thomas, K.E. (2006a) Temkin, N., Tuma, R., Wade, C., Walker, M.D., Weinrich,
Traumatic Brain Injury in the United States: Emergency De- M., Whyte, J., Wilberger, J., Young, A.B. and Yurkewicz, L.
partment Visits, Hospitalizations, and Deaths. Center for (2002) Clinical trials in head injury. J. Neurotrauma, 19:
Disease Control and Prevention, National Center for Injury 503–557.
Prevention and Control, Atlanta, GA. Nguyen, D.N., Becker, G.W. and Riggin, R.M. (1995) Protein
Langlois, J.A., Rutland-Brown, W. and Wald, M.M. (2006b) mass spectrometry: applications to analytical biotechnology.
The epidemiology and impact of traumatic brain injury: a J. Chromatogr. A, 705: 21–45.
brief overview. J. Head Trauma Rehabil., 21: 375–378. Ommaya, A.K. (1995) Head injury mechanisms and the con-
Lesko, L.J. and Atkinson Jr., A.J. (2001) Use of biomarkers cept of preventive management: a review and critical synthe-
and surrogate endpoints in drug development and regulatory sis. J. Neurotrauma, 12: 527–546.
decision making: criteria, validation, strategies. Annu. Rev. Ommaya, A.K., Goldsmith, W. and Thibault, L. (2002) Bio-
Pharmacol. Toxicol., 41: 347–366. mechanics and neuropathology of adult and paediatric head
Liu, M.C., Akle, V., Zheng, W.R., Dave, J.R., Tortella, F.C., injury. Br. J. Neurosurg., 16: 220–242.
Hayes, R.L. and Wang, K.K.W. (2006) Comparing calpain- Ottens, A.K., Kobeissy, F.H., Haskins, W.E., Golden, E.C.,
and caspase-3-degradation patterns in traumatic brain injury Zhang, Z.Q., Chen, S., Hayes, R.L., Wang, K.K.W. and
by differential proteome analysis. Biochem. J., 394: 715–725. Denslow, N.D. (2006) Neuroproteomics in neurotrauma.
Lorenz, P., Ruschpler, P., Koczan, D., Stiehl, P. and Thiesen, Mass Spectrom. Rev., 25: 380–408.
H.J. (2003) From transcriptome to proteome, differentially ex- Ottens, A.K., Kobeissy, F.H., Wolper, R.A., Haskins, W.E.,
pressed proteins identified in synovial tissue of patients suffer- Hayes, R.L., Denslow, N.D. and Wang, K.K.W. (2005) A
ing from rheumatoid arthritis and osteoarthritis by an initial multidimensional differential proteomic platform using dual
screen with a panel of 791 antibodies. Proteomics, 3: 991–1002. phase ion-exchange chromatography — polyacrylamide gel
Malakhov, M.P., Kim, K.I., Malakhova, O.A., Jacobs, B.S., electrophoresis/reverse phase liquid chromatography tandem
Borden, E.C. and Zhang, D.E. (2002) High-throughput mass spectrometry. Anal. Chem., 77: 4836–4845.
418

Patton, W.F. (2002) Detection technologies in proteome anal- (2005) Analysis of long-term gene expression in neurons of
ysis. J. Chromatogr. B, 771: 3–31. the hippocampal subfields following traumatic brain injury in
Peng, J., Kim, M.J., Cheng, D., Duong, D.M., Gygi, S.P. and rats. J. Neurosci., 131: 87–97.
Sheng, M. (2004) Semiquantitative proteomics analysis of rat Shiozaki, T., Hayakata, T., Tasaki, O., Hosotubo, H., Fuijita,
forebrain postsynaptic density fractions by mass spectrome- K., Mouri, T., Tajima, G., Kajino, K., Nakae, H., Tanaka,
try. J. Biol. Chem., 279: 21003–21011. H., Shimazu, T. and Sugimoto, H. (2005) Cerebrospinal fluid
Pike, B.R., Flint, J., Dave, J.R., Lu, X.C., Wang, K.K., concentrations of anti-inflammatory mediators in early-phase
Tortella, F.C. and Hayes, R.L. (2003) Accumulation of cal- severe traumatic brain injury. Shock, 23: 406–410.
pain and caspase-3 proteolytic fragments of brain-derived Smith, D.H., Uryu, K., Saatman, K.E., Trojanowski, J.Q. and
alphaII-spectrin in CSF after middle cerebral artery occlu- McIntosh, T.K. (2003) Protein accumulation in traumatic
sion in rats. J. Cereb. Blood Flow Metab., 24: 98–106. brain injury. Neuromolecular Med., 4: 59–72.
Pike, B.R., Flint, J., Dutta, S., Johnson, E., Wang, K.K.W. and Sosin, D.M., Sniezek, J.E. and Waxweiler, R.J. (1995) Trends in
Hayes, R.L. (2001) Accumulation of non-erythroid alphaII- death associated with traumatic brain injury, 1979 through
spectrin and calpain-cleaved alphaII-spectrin breakdown 1992. Success and failure. JAMA, 273: 1778–1780.
products in cerebrospinal fluid after TBI in rats. J. Neuroc- Tomizaki, K.Y., Usui, K. and Mihara, H. (2005) Protein-
hem., 78: 1297–1306. detecting microarrays: current accomplishments and require-
Pineda, J.A. (2006) Accumulation of alphaII-spectrin break- ments. Chembiochem, 6: 782–799.
down products in CSF of severely injured traumatic brain Tornheim, P.A., McDermott, F. and Shiguma, M. (1990) Effect
injury patients. J. Neurotrauma, in press. of experimental blunt head injury on acute regional cerebral
Pineda, J.A., Wang, K.K. and Hayes, R.L. (2004) Biomarkers blood flow and edema. Adv. Neurol., 52: 377–384.
of proteolytic damage following traumatic brain injury. Brain Tornheim, P.A. and McLaurin, R.L. (1981) Acute changes in
Pathol., 14: 202–209. regional brain water content following experimental closed
Piper, I.R., Thomson, D. and Miller, J.D. (1996) Monitoring head injury. J. Neurosurg., 55: 407–413.
weight drop velocity and foam stiffness as an aid to quality Wang, K.K.W., Larner, S.F., Robinson, G. and Hayes, R.L.
control of a rodent model of impact acceleration neurotrau- (2006) Neuroprotection targets after traumatic brain injury.
ma. J. Neurosci. Methods, 69: 171–174. Curr. Opin. Neurol., 19: 514–519.
Povlishock, J.T., Hayes, R.L., Michel, M.E. and McIntosh, Wang, K.K.W., Ottens, A., Haskins, W.E., Liu, M.C., Den-
T.K. (1994) Workshop on animal models of traumatic brain slow, N., Chen, S.S. and Hayes, R.L. (2004) Neuroproteomic
injury. J. Neurotrauma, 11: 723–732. studies of traumatic brain injury. In: Neuhold L.A. (Ed.),
Ragnarsson, K.T. (2006) Traumatic brain injury research since Human Brain Proteome — International Review of Neuro-
the 1998 NIH consensus conference. J. Head Trauma Re- biololgy, Vol. 61. Elsevier, San Diego, pp. 215–240.
habil., 21: 379–387. Wang, K.K.W., Ottens, A.K., Liu, M.C., Lewis, S.B., Meegan,
Ringger, N.C., O’Steen, B.E., Brabham, J.G., Siler, X., Pineda, C., Oli, M.W., Tortella, F.C. and Hayes, R.L. (2005) Proteo-
J., Wang, K.K.W. and Hayes, R.L. (2004) A novel marker mic identification of biomarkers of traumatic brain injury.
for traumatic brain injury: CSF alphaII-spectrin breakdown Expert Rev. Proteomics, 2: 603–614.
product levels. J. Neurotrauma, 21: 1443–1456. Warden, D. (2006) Military TBI during the Iraq and Afghani-
Rink, A., Fung, K.M., Trojanowski, J.Q., Lee, V.M., Neuge- stan wars. J. Head Trauma Rehabil., 21: 398–402.
bauer, E. and McIntosh, T.K. (1995) Evidence of apoptotic Wennersten, A., Holmin, S. and Mathiesen, T. (2003) Charac-
cell death after experimental traumatic brain injury in the rat. terization of Bax and Bcl-2 in apoptosis after experimental
Am. J. Pathol., 147: 1575–1583. traumatic brain injury in the rat. Acta Neuropathol., 105:
Saljo, A., Bao, F., Haglid, K.G. and Hansson, H.A. (2000) 281–288.
Blast exposure causes redistribution of phosphorylated ne- Wieloch, T. and Nikolich, K. (2006) Mechanisms of neural
urofilament subunits in neurons of the adult rat brain. J. plasticity following brain injury. Curr. Opin. Neurobiol., 16:
Neurotrauma, 17: 719–726. 258–264.
Sanders, S.L., Jeffings, J., Canutescu, A., Link, A.J. and Well, Williams, A.J., Hartings, J.A., Lu, X.C., Rolli, M.L., Dave,
P.A. (2002) Proteomics of the eukaryotic transcription ma- J.R. and Tortella, F.C. (2005) Characterization of a new rat
chinery: identification of proteins associated with compo- model of penetrating ballistic brain injury. J. Neurotrauma,
nents of yeast TFIID by multidimensional mass 22: 313–331.
spectrometry. Mol. Cell. Biol., 22: 4723–4738. Yi, J.H. and Hazell, A.S. (2006) Excitotoxic mechanisms and
Satchell, M.A., Zhang, X., Kochanek, P.M., Dixon, C.E., the role of astrocytic glutamate transporters in traumatic
Jenkins, L.W., Melick, J., Szabo, C. and Clark, R.S.B. (2003) brain injury. Neurochem. Int., 48: 394–403.
A dual role for poly-ADP-ribosylation in spatial memory Yoo, G.H., Piechocki, M.P., Ensley, J.F., Nguyen, T., Oliver,
acquisition after traumatic brain injury in mice involving J., Meng, H., Kewson, D., Shibuya, T.Y., Lonardo, F. and
NAD+ depletion and ribosylation of 14-3-3g. J. Neuroc- Tainsky, M.A. (2002) Docetaxel induced gene expression
hem., 85: 697–708. patterns in head and neck squamous cell carcinoma using
Shimamura, M., Garcia, J.M., Prough, D.S., Dewitt, D.S., cDNA microarray and PowerBlot. Clin. Cancer Res., 8:
Uchida, T., Shah, S.A., Avila, M.A. and Hellmich, H.L. 3910–3921.
Weber & Maas (Eds.)
Progress in Brain Research, Vol. 161
ISSN 0079-6123
Copyright r 2007 Elsevier B.V. All rights reserved

CHAPTER 30

Remyelination of the injured spinal cord

Masanori Sasaki1,2, Bingcang Li1,2,3, Karen L. Lankford1,2, Christine Radtke1,2,4 and


Jeffery D. Kocsis1,2,

1
Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine,
New Haven, CT 06510, USA
2
Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT 06516, USA
3
Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, P.R. China
4
Department of Plastic, Hand and Reconstructive Surgery, Medical School Hannover, Hannover, Germany

Abstract: Contusive spinal cord injury (SCI) can result in necrosis of the spinal cord, but often long white
matter tracts outside of the central necrotic core are demyelinated. One experimental strategy to improve
functional outcome following SCI is to transplant myelin-forming cells to remyelinate these axons and
improve conduction. This review focuses on transplantation studies using olfactory ensheathing cell (OEC)
to improve functional outcome in experimental models of SCI and demyelination. The biology of the OEC,
and recent experimental research and clinical studies using OECs as a potential cell therapy candidate are
discussed.

Keywords: spinal cord injury; remyelination; olfactory ensheathing cells

Introduction Cellular transplantation of appropriate cells into


experimental models of SCI can promote axonal
The clinical pathophysiology of contusive spinal regeneration, provide neuroprotective effects by
cord injury (SCI) is complex. A combination of secretion of neurotrophins and remyelinate axons.
hemorrhage, ischemia and/or edema develops re- One cell of particular interest as a cell therapy
sulting in necrosis with tissue loss (Schwab et al., candidate to both encourage axonal remyelination
2006, review). Additionally long tracts of the and regeneration is a specialized glial cell, the
injured spinal cord may survive, but become olfactory ensheathing cell (OEC). Adult olfactory
demyelinated. Although regeneration of spinal cord receptor neurons continually undergo turnover
axons is an ultimate objective, the presence in many from an endogenous progenitor pool, and their
patients with non-penetrating SCI of a population nascent axons grow through the olfactory nerves
of surviving axons that do not conduct impulse and cross the PNS–CNS interface, where they form
due to demyelination (Keirstead, 2005; Kocsis new synaptic connections in the olfactory bulb
and Sasaki, 2005, reviews), suggests a cell-based (Graziadei et al., 1978). OECs associate with olfac-
approach for remyelination as a potential strategy tory receptor neurons from their peripheral origin
for inducing recovery of function in SCI. One to their central projection in the outer nerve layer of
approach to this goal capitalizes on recent progress the olfactory bulb (Doucette, 1991). This putative
in the transplantation of cells into the injured CNS. support role of OECs in axonal growth within the
adult CNS has spawned extensive research to study
Corresponding author. Tel.: +1-(203)-937-3802; the potential of OEC transplants encouraging ax-
Fax: +1-(203)-937-3801; E-mail: Jeffery.Kocsis@yale.edu onal regeneration and functional recovery in SCI

DOI: 10.1016/S0079-6123(06)61030-3 419


420

models (Li et al., 1997, 1998; Ramon-Cueto et al., by transplanted cells is the X-irradiation/ethidium
1998, 2000; Imaizumi et al., 2000a, b). bromide model (X-EB). Ethidium bromide (EB), a
Transplantation of OECs after SCI is associated nucleic acid chelator, kills cells in the target zone
with functional improvement even when trans- (dorsal white matter) and focal X-irradiation to
plantation is delayed by weeks (Lu et al., 2002; blocks mitosis and kills oligodendrocyte progeni-
Keyvan-Fouladi et al., 2003). While the precise tors (Blakemore and Crang, 1985). In this model
mechanisms of the functional recovery after OEC system, endogenous repair in the center of the le-
transplantation are not fully understood, several sion is delayed by 6–8 weeks thus allowing a stable
mechanisms including elongative axonal regen- time window to assess transplanted cells (Honmou
eration, axonal sparing, sprouting and plasticity et al., 1996). Generally our analyses are carried out
associated with novel polysynaptic pathways, re- within 3 weeks after X-EB lesion induction, which
cruitment of endogenous SCs and remyelination is well within the time at which persistent demyeli-
have been proposed (Raisman, 2001; Bareyre nation is confirmed in a large number of control
et al., 2004; Sasaki et al., 2004; Keyvan-Fouladi studies (Blakemore and Crang, 1985; Franklin
et al., 2002). In animals with SCI and OEC trans- et al., 1996; Honmou et al., 1996; Kato et al.,
plants, myelinated axons spanning the lesion site 2000). The lesion induced by this procedure is
display a characteristic peripheral pattern of my- characterized by virtually complete loss of endog-
elination similar to that of Schwann cell (SC) my- enous glial elements (astrocytes and oligodendro-
elination (Franklin et al., 1996; Li et al., 1997, cytes) with preservation of axons (Fig. 1A). The
1998; Imaizumi et al., 1998, 2000b; Sasaki et al., higher power light micrograph of the lesion shows
2004). compact fields of demyelinated axons in close ap-
Remyelination by transplantation may be of position separated by areas of myelin debris and
particular importance because contusive SCI often macrophages (Figs. 1A2, A3). The lesion occupies
results in loss of function from demyelination nearly the entire dorso-ventral extent of the dorsal
induced by spinal cord trauma. OECs can form columns for 5–7 mm longitudinally.
myelin when transplanted into demyelinated spinal A large body of work supports the proposal
cord (Franklin et al., 1996; Imaizumi et al., 1998; that transplantation of OECs into various SCI
Sasaki et al., 2004). Moreover, human OECs can and demyelination models can promote axonal re-
form myelin in the immunosuprressed rat (Barnett generation, remyelination and functional recovery
et al., 2000; Kato et al., 2000) and OECs trans- (Franklin et al., 1996; Li et al., 1998; Ramon-Cueto
planted into the non-human primate can remyelin- et al., 1998; Imaizumi et al., 2000a, b; Ramon-
ate spinal cord axons (Radtke et al., 2004). Cueto et al., 2000; Lu et al., 2002; Keyvan-Fouladi
et al., 2003; Plant et al., 2003). Yet, there is an
important controversy as to whether the trans-
Remyelination in demyelinated spinal cord planted OECs associate with axons and form
peripheral myelin, as opposed to recruiting endog-
Three types of SCI models are commonly used enous SCs that form myelin (Takami et al., 2002;
in experimental studies in rodents: transection, Boyd et al., 2004). OECs can express a number of
compression and contusion (Rosenzweig and trophic factors, transcription factors and extracel-
McDonald, 2004, review). However, because of lular matrix molecules (Ramon-Cueto et al., 1998;
the complex pathophysiological conditions of SCI Chuah and West, 2002; Au and Roskams, 2003;
in these models, the study of remyelination is more Ramer et al., 2004), which could facilitate endog-
complex. Aside from the heterogeneity of the le- enous SC cell invasion, angiogenesis and activation
sion, endogenous remyelination by SCs occurs of progenitor cells to facilitate repair.
in these model systems thus making distinction of To address this issue, we used OECs from
endogenous remyelination from that of trans- GFP-expressing rats. After 3 weeks of transplan-
planted cells more complex. A model used by sev- tation into X-EB model, methylene blue/Azure II
eral groups including ours to study remyelination semithin plastic sections demonstrated extensive
Fig. 1. Light micrographs of transverse sections of the dorsal spinal cord stained with methylene blue/azure II showing the dorsal
funiculus in demyelinated (A) and OEC transplanted (B) rats. Examination at higher magnification show demyelinated (A2, A3) and
remyelinated (B2, B3) axons in the dorsal columns. Note the extensive myelination after transplantation of OEC (B). Many myelin-
forming cells were similar to peripheral myelin-forming cells, characterized by large nuclear and cytoplasmic regions (B3). A2, B2 and
A3, B3 were prepared from the boxed area in A1, B1 and A2, B2 respectively. Immuno-EM for GFP shows numerous GFP+ cells
remyelinating the demyelinated axons (C, D). Counterstaining for these sections was minimal, and dark staining shows electron dense
immunoperoxidase reaction product. Note the electron dense reaction product in the cytoplasm and nuclei of most cells forming
myelin (C, D). One cell associated with myelin in this field displays distinct reaction product, whereas an adjacent cell does not have
electron dense reaction product in its cytoplasm (D1). D2 is an enlargement of the box in D1. Note the dense reaction product in the
cytoplasm of the myelin-forming cell on the right and the basement membrane surrounding the cell. Scale bars ¼ 700 mm (A1, B1);
70 mm (A2, B2); 1 mm (A3, B3); 5 mm (C); 2 mm (D1); 0.4 (D2). C and D are modified with permission from Sasaki et al. (2006a).
422

remyelination in the demyelinated axons (Fig. 1B). cells within the dorsal column lesion formed that
To fully establish that the OECs derived from peripheral-type myelin. Immunostaining for P0 and
GFP rats were indeed responsible for the remyeli- NF in coronal section reveals a central NF+ axonal
nation, immuno-electron microscopy (EM) for core surrounded by P0-identified myelin, which is
GFP was performed. Intense reaction product wrapped by cytoplasm of a GFP-OEC (Fig. 2E),
was observed within the cytoplasm and nuclei indicating that transplanted OECs remyelinate the
of cell profiles surrounding myelinated axons demyelinated axons.
(Fig. 1C). To establish a first approximation of Moreover, we also transplanted highly purified
the relative contribution of donor cell to host cell OECs isolated from transgenic pigs expressing the
remyelination, quantitative analysis was performed alpha1, 2 fucosyltransferase gene (H-transferase or
on GFP immuno-EM experiments of two trans- HT) into a demyelinated lesion of the African
planted animals. A total of 136 axons were counted green monkey spinal cord (Radtke et al., 2004).
in the lesions, and of these 120 were GFP+ cells. Four weeks post-transplantation, robust remyeli-
Of the GFP+ cells, four (3.3%) showed no direct nation was found in 62.5% of the lesion sites,
contact with axons. The myelin-forming status of whereas there was virtually no remyelination in the
five GFP+ cells (4.2%) could not be determined non-transplanted controls. This was the first dem-
because of poor membrane preservation. Of the onstration that xenotransplantation of character-
remaining 111 GFP+ cells, 90 cells (81%; 90 of 111) ized OECs into the primate spinal cord results in
showed a distinct myelin structure. The remaining remyelination. This together with the immuno-
21 GFP+ cells (19%; 21 of 111) appeared to be in histochemical demonstration of the grafted cells
varying stages of loose wrapping or ensheathing the within the lesioned area confirmed that remyeli-
axons but did not show distinct compact myelin. nation was indeed achieved by OECs.
Within this same region, 16 myelinated axon pro-
files were detected, which were associated with my- Nodal reconstruction of remyelinated spinal cord
elin-forming cells that did not contain DAB axons
reaction product. An example of a non-GFP my-
elinated axon adjacent to a GFP+ myelinating cell The restoration of rapid and secure impulse con-
is shown in Figs. 1, D1 and D2. Note the presence duction after demyelination is dependent on the
of a basement membrane surrounding both myelin- acquisition of myelin sheaths and the clustering of
ated axons. In frozen sections, OECs from donor specific molecules within discrete domains of the
GFP rats (GFP-OEC) exhibited a robust distribu- myelinated axon membrane. In myelinated axons,
tion within the X-EB lesioned spinal cord dorsal voltage-gated sodium (Nav) channels are aggre-
columns. The GFP-OECs were easily distinguished gated in high density at nodes of Ranvier, whereas
at this time point by their green fluorescence Shaker-type potassium (Kv1) channels are sepa-
and were distributed longitudinally along the dor- rated from nodal Nav channels by septate-like
sal columns for 8 mm (Fig. 2A). GFP-OECs were paranodal junctions (Peles and Salzer, 2000;
mostly confined to the lesion zone of the dorsal Rasband and Trimmer, 2001; Girault and Peles,
funiculus. GFAP immunostaining indicated a near 2002). Of the seven Nav channel isoforms expressed
absence of astrocytes within the lesion site, but in- in nervous tissue (Goldin et al., 2000), Nav1.6 is the
tense GFAP staining was observed at the outer predominant one at mature nodes in both the PNS
boundary of the lesion zone (Figs. 2B, C). In con- and CNS (Caldwell et al., 2000; Boiko et al., 2001)
trast to GFAP immunolabeling, immunostaining following a transition from Nav1.2 (Boiko et al.,
for P0, a specific marker of peripheral myelin 2001; Kaplan et al., 2001; Jenkins and Bennett,
(Greenfield et al., 1973), was primarily localized 2002; Rios et al., 2003). The channel clustering
within the lesion and transplantation site (Figs. 2D, (Vabnick et al., 1997; Rasband et al., 1999) and the
E). Ringlet-like P0 immunostaining was associ- transition from Nav1.2 to Nav1.6 (Boiko et al.,
ated with GFP-OECs and was surrounded by 2001; Rios et al., 2003) are dependent on interac-
GFP+-OEC cytoplasm, indicating the transplanted tion of the axon with myelinating cells (Kaplan
423

Fig. 2. Sagittal frozen sections through the lesion site demonstrate the distribution of transplanted GFP-OEC. Transplanted cells are
primarily confined to the lesion site. Some cells migrated into the deep white matter. The dashed line demarcates lesion edge (A).
Coronal frozen sections in the lesion show the presence of GFP-OEC within a lesion site. Transplanted cells survived primarily in the
dorsal funiculus. There was little GFAP staining within the lesion zone (B). GFAP-positive cells were present at the peripheral margin
of the lesion. These results indicate that few astrocytes are present in the transplant region, and that there is a preponderance of GFP-
OEC in the lesion zone (B, C). P0-immunostaining (red) of the frozen coronal section reveals that most axons remyelinated by the
transplanted OECs are surrounded by peripheral type of myelin. Red-P0 rings are associated with green cellular elements, indicating
that transplanted OECs remyelinate the demyelinated axons (D). Expansion of a cell indicated by an arrow (D inset). P0 and
neurofilament (NF) staining at lesion boundary (dashed line) and transplant zone (E). Higher magnification showing neurofilament-
defined axon cores surrounded by P0 myelin rings enwrapped by GFP-OECs (E2). B–E are coronal. Scale bars: 1 mm (A); 400 mm (B);
30 mm (C); 10 mm (D); 20 mm (A, inset); 10 mm (B, inset); 30 mm (E1); 10 mm (E2); 2.5 mm (E2, inset). Modified with permission from
Sasaki et al. (2006a).
424

Fig. 3. Nodal structure of remyelinated axons (A, B) at 3 weeks after OEC transplantation. Sagittal section showing a field of
myelinated axons interspersed among donor OECs. Note the node structure in the center of the field (A). High-power electron
micrograph showing node and paranodal loops (B1). B2 is an enlargement of the boxed area in B1. Scale bars ¼ 5 mm (A); 1 mm (B1);
0.5 mm (B2). Modified with permission from Sasaki et al. (2006a).

et al., 1997; Eshed et al., 2005). Remyelinated beneath the myelin sheath in remyelinated axons,
axons display inappropriately short internodal suggesting that the transplanted GFP-OECs are
lengths (Gledhill and McDonald, 1977; Weiner competent to contribute to the specific clustering
et al., 1980; Blakemore and Murray, 1981; of Nav channels at nodes. As an additional deter-
Hildebrand et al., 1985), indicating that new nodes minant of the ability of axons myelinated by
are formed. Despite their location at formerly GFP-OECs to support the asymmetric organiza-
internodal sites, remyelinated PNS axons have tion of ion channels within remyelinated nodal
been shown to display high densities of Nav chan- regions, we examined the distribution of Kv1.2
nels at nodes (Novakovic et al., 1996, 1998) and in the juxtaparanodal region. Kv1.2, as well as
Kv1 aggregations within juxtaparanodal domains Kv1.1, form heteromultimers with Kv1.4 and Kvb2
(Rasband et al., 1998). The expression and organi- (Wang et al., 1993; Rasband et al., 1998) and
zation of specific isoforms of Nav and Kv1 channels have been shown previously to be aggregated in
in remyelinated CNS axons have not been exam- juxtaparanodal regions of most spinal cord axons
ined. Recently we reported that EM analysis of (Rasband et al., 1999; Rasband and Trimmer,
spinal cords performed at 3 weeks after GFP-OEC 2001). At 3 weeks (Figs. 4I–L), Kv1.2 is aggregated
transplantation demonstrated distinct nodes of within juxtaparanodal regions of the remyelinated
Ranvier (Fig. 3). Large cytoplasmic and nuclear axons, with some nodes exhibiting incursion of
compartments were present in cells associated Kv1.2 channels into adjacent paranodal regions.
with the myelin profiles (Fig. 3A). Longitudinal Kv1.2 was not observed within nodal areas in
sections of the dorsal columns revealed well these axons.
formed nodal and paranodal regions; paranodal
loops from adjacent myelin-forming cells were
readily recognized flanking nodes (Figs. 3B1, B2). Transected spinal cord
Also, in immunohistochemical analysis we obser-
ved Nav1.6 staining at most nodes, whereas detect- Distribution and myelin formation by transplanted
able Nav1.2 immunostaining was not apparent at GFP-OECs within the spinal cord
nodes (Fig. 4). In dorsal columns 3 weeks after
transplantation, virtually all nodes bounded by Spinal cord injuries without OEC transplants can
GFP-OEC myelin sheaths exhibited Nav1.6 stain- show limited SC-like myelination, presumably from
ing (Figs. 4A–D); similar to control spinal cord invasion of the injury site from endogenous SCs
axons, Nav1.2 immunolabeling was not observed at (Brook et al., 1998; Imaizumi et al., 2000a, b;
any nodes (Figs. 4E–H). The Nav1.6 labeling was Namiki et al., 2000; Takami et al., 2002) or possibly
localized to the nodal domain and was not obser- from precursor cells. The degree to which OECs
ved in paranodal or juxtaparanodal regions or can integrate into injury sites and survive and
425

Fig. 4. Nav1.2 and Nav1.6 at GFP-OEC nodes in remyelinated dorsal columns. At 3 weeks after transplantation, Nav1.6 clustering is
displayed at most Caspr-delimited (A–D) nodes formed by GFP-OECs at 3 weeks. In contrast, Caspr-delimited nodes formed by GFP-
OECs do not exhibit Nav1.2 immunostaining (E–H). Merged images of A–C and E–G are shown in D and H, respectively. Juxta-
paranodal Kv1.2 immunolabeling at 3 weeks after GFP-OEC transplantation dorsal columns. Paranodes display Caspr staining (I)
that is flanked by Kv1.2 aggregations within juxtaparanodal regions. Merged images of I–K are shown in L, respectively. Scale
bars ¼ 10 mm. Modified with permission from Sasaki et al. (2006a).

whether they form myelin or facilitate endogenous (Ramon-Cueto and Avila, 1998; Chuah and West,
myelin repair mechanisms was controversial. OECs 2002; Au and Roskams, 2003) that could facilitate
in culture are diverse and exhibit characteristics of endogenous cell invasion, as well as angiogenesis
astrocytes, SCs and oligodendrocytes. Moreover, and activation of progenitor cells. A recent study
they express a number of trophic factors, transcrip- was unable to find evidence of myelination in the
tion factors and extracellular matrix molecules compressed spinal cord by the OECs isolated from
426

Fig. 5. Montage image of sagittal frozen section showing distribution of GFP-OECs within and beyond the transection site (A).
Semithin plastic sections stained with methylene blue/Azure II through the transection site 5 weeks after transplantation of OECs.
Low-power micrograph showing completeness of the transection through the entire dorsal funiculus and beyond (B). EM from the
same lesion showing myelinated axons surrounded by a cellular element forming a tunnel (C). EM of anti-GFP immunoperoxidase
staining of OEC transplant (D). Reaction product can be seen in cytoplasmic regions of the myelin-forming cell but not in the myelin.
A crosscut section of axon showing cytoplastic reaction product. Enlargement of the boxed area is shown in D2. Note the presence of
extracellular fibrous elements in D1. Scale bars ¼ 250 mm (A); 0.75 mm (B); 5 mm (C); 1 mm (D1); 0.25 mm (D2). Modified with
permission from Sasaki et al. (2004).

embryonic day 18 rat, infected with a LacZ- rat. Five weeks after OEC transplantation, the
expressing retrovirus, and suggests that OEC trans- cells survived and distributed extensively within
plantation may facilitate endogenous SC invasion the transection site and more limitedly beyond
into the lesion site (Boyd et al., 2004). the lesion site (Fig. 5A1). High-power micro-
We transplanted genetically labeled GFP-OECs graphs (Fig. 5A2) of AnkyrinG and Caspr stain-
into a dorsal funiculus transection model in the ing demonstrate nodal and paranodal regions,
427

respectively, associated with some transplanted myelin (Fig. 5D). In a longitudinal section of a
GFP-OECs, suggesting forming a new node of myelinated axon, intense reaction product can be
Ranvier. Methylene blue/Azure II semithin plastic seen in the cytoplasm of the myelin-forming cell.
sections through this region of the lesion 5 weeks Large cytoplasmic and nuclear regions as well as
after transplantation revealed greater structural the presence of basal lamina and extracellular fi-
detail (Fig. 5B). As reported initially by Li et al. brils (Fig. 5D) indicate a peripheral pattern of
(1997), small groups of myelinated axons were myelination.
often surrounded by a non-myelinating cell form- Taken together, we conclude that transplanted
ing tube-like structures around the myelinated GFP-OECs integrate into the injury site of a dor-
axons. This organization is not observed after en- sal funiculus transection, distribute and associate
dogenous repair or after transplantation of SCs longitudinally with axons spanning the lesion site
(Imaizumi et al., 2000b). These profiles were ob- and do form myelin.
served in both dorsal and ventral regions of the
lesion zone (Fig. 5B). EM further indicates that
the myelinated axons in the lesion zone are sur- Improved hindlimb locomotor function
rounded by cytoplasmic extensions of cells form-
ing tunnels (Fig. 5C). These clusters of myelinated All OEC-transplanted animals in this model system
axons were variable from animal to animal and exhibited a gradual improvement in hindlimb
confined mostly to and near the lesion zone. Rings locomotor function during the 5-week recovery
of P0 labeling were observed in some regions of the period (Fig. 6). The sham injection group recov-
lesion and were often associated with GFP cellular ered to a Basso, Beattie and Bresnahan (BBB) lo-
elements. EM examination of anti-GFP immuno- comotor rating scale (Basso et al., 1995) of 9 and
peroxidase-reacted sections revealed that many did not display weight-bearing plantar stepping.
detectable GFP+ cells were in direct contact with However, the OEC transplant group recovered to
host axons. Reaction product was clearly evident 18 on the BBB score and displayed consistent
in the cytoplasm of many cells that formed well- weight-bearing plantar stepping. Statistical analysis
defined multi-laminate structures characteristic of indicated that the open-field locomotor scores at

Fig. 6. Open-field locomotor scores for OEC transplant (n ¼ 20) and sham injection (n ¼ 6) groups tested 1 week before and for 5
weeks after transplantation. Modified with permission from Sasaki et al. (2004).
428

3, 4 and 5 weeks after injury and OEC transplan-


tation was significantly higher than sham injection.

Neuroprotection of dorsal corticospinal tract

A recent study demonstrates that primary motor


cortex (M1) pyramidal neurons undergo apoptotic
cell death after axotomizing SCI (Hains et al.,
2003). Moreover, earlier work has shown that cor-
ticospinal neurons become atrophic after spinal
cord transection (McBride et al., 1989). To explore
the possibility that OECs are neuroprotective for
injured corticospinal tract (CST) neurons, we trans-
planted OECs into the dorsal transected spinal cord
(T9) and examined M1 to assess apoptosis and
neuronal loss at 1 and 4 weeks post-transplantation.
Triple labeling with Hoechst, Fluorogold (FG), and
terminal deoxynucleotidyltransferase-mediated de-
oxyuridine triphosphate–rhodamine nick end-labe-
ling (TUNEL) identified putatively axotomized
pyramidal cells in the SCI group, with and with-
out spinal OEC transplantation. The neuronal sub-
set undergoing apoptosis was readily identified in 1-
week tissue. Within these cells, nuclear morph-
ologies of both apoptotic and surrounding normal
neurons and glia could be identified (Figs. 7A, B).
Hoechst staining was observed in all cells and uni-
formly filled the spherical nuclear compartment. In
a proportion of cells undergoing apoptosis, Hoe- Fig. 7. Hoechst 33342, Fluorogold (FG), and TUNEL triple
chst staining revealed abnormal morphology, in- labeling of corticospinal neurons 1 week after injury. Hoechst
cluding the formation of chromatin condensates staining of non-TUNEL-positive (arrows with tails) and
within a less-distinct nucleus (Hains et al., 2003). TUNEL-positive (arrowheads) neurons with corresponding
FG-backfilling are shown. In SCI+FG+OEC animals (B),
Merging of Hoechst, FG and TUNEL staining fewer TUNEL-positive FG-backfilled neurons are observed
(Figs. 7A, B) showed that irregular nuclear frag- compared with SCI+FG+DMEM (A). Insets in A show two
ments identified by Hoechst staining overlap se- TUNEL-positive neurons exhibiting nuclear compartment-
curely with FG- and TUNEL-positive cells, alization and formation of nucleosomes, hallmarks of apoptosis.
permitting the confident designation of an apopto- Quantification of neurons that are both TUNEL- and FG-
positive (C) reveals that OEC transplantation significantly
tic status. The nuclear morphology of TUNEL- (*Po0.05) reduces apoptotic cell death at 1 week. No evidence
positive neurons was identical to that seen in our of death was observed at any other time-point. Scale
earlier paper (Hains et al., 2003) (Fig. 7A, insets). bars ¼ 125 mm in A, B; 20 mm in inset in A. Modified with per-
Apoptotic (FG- and TUNEL-positive cells) pyram- mission from Sasaki et al. (2006b).
idal neurons were observed both after SCI+
FG+DMEM (Fig. 7A, arrowheads) and in the neurons was significantly (Po0.05) reduced by
SCI+FG+OEC group (Fig. 7B, arrowheads), but 45% (16.476.40 vs. 36.175.69) in the OEC trans-
the number of apoptotic neurons was significantly plant group compared with the SCI+FG+
reduced in the SCI+FG+OEC group (Fig. 7C). DMEM group (Fig. 7C). At 4 weeks, both the
At 1 week after injury, the number of apoptotic SCI+FG+DMEM and SCI+FG+OEC groups
429

showed apoptotic activity comparable with sham OEC into spinal cord contusion injury
controls (0.9870.81 and 0.7570.56) (Fig. 7C).
We described the supraspinal effects of OEC trans- We also evaluated the in vivo fate of OECs trans-
plantation on apoptosis and cell survival of CST planted into the contused rat spinal cord with the
neurons within the M1 cortex after transection weight-drop device developed at New York Uni-
of their axons in the spinal cord. Our results indi- versity (NYU impactor) (Li et al., 2004). Trans-
cate that apoptosis of primary motor cortical neu- planted GFP-OECs were distributed widely within
rons is reduced and that cortical neuronal density is transplanted spinal cords. In sagittal sections,
increased after OEC transplantation. Enhanced GFP-OECs were widely distributed along the
levels of brain-derived neurotrophic factor (BDNF) length of the lesion (Fig. 8A). Immuno-EM using
were observed in the OEC transplanted lesion. an anti-GFP antibody revealed that the identified
Thus, transplantation of OECs into injured spinal OECs made direct and extensive contact with host
cord has a neuroprotective effect on corticospinal axons (Figs. 8B, C). Transplanted GFP-OECs pro-
neurons. The relative contribution of this effect to duced multi-laminate structures surrounding axons
the observed functional improvement after OEC in a one-to-one relationship with large amounts of
transplantation is uncertain, but this data indicates cytoplasm and basal lamina characteristic of pe-
that OEC transplantation results in a larger pool of ripheral myelin. The typical relationships of trans-
surviving corticospinal neurons. Thus, OEC trans- planted SCs and OECs with host axons were quite
plantation into the injured spinal cord has distant different. While the overwhelming majority of GFP
neuroprotective effects on descending cortical positive cells in both transplant conditions were in
projection neurons as well (Sasaki et al., 2006b). direct contact with host axons, GFP positive SCs

Fig. 8. Distribution of transplanted OECs into a contused rat spinal cord (A). Sagittal section of a segment of the spinal cord 8 mm
rostral and caudal to the injured area showed transplanted OECs concentrated near the center and distributed along the longitudinal
axes of the spinal cord at 3 weeks after transplantation (A), arrows indicated transplantation points. A2 and A3 are enlargements of
boxed areas in A1. Immuno EM for GFP revealed that many detectable GFP+ cells were in direct contact with host axons (Arrows).
Reaction product was clearly evident in the cytoplasm of many cells that formed well-defined multi-laminate structures characteristic
of myelin at 4 weeks after transplantation (8B, C). Scale bars ¼ 1 mm (A1); 100 mm (A2, A3); 5 mm (B); 1 mm (C).
430

cells were observed almost exclusively in one-to- strategy as an approach that may elicit some de-
one associations with host axons, with a high pro- gree of functional recovery, raising the possibility
portion of those contacts associated with myelina- that this approach might be useful in the treatment
tion, while GFP positive OECs typically wrapped of SCI. Experimental work indicates that cell
or engulfed several axons with a small percentage transplantation approaches can facilitate axonal
of contacts associated with myelination. regeneration, remyelination, neuroprotection and
possible neovascularization. Importantly, axons
remyelinated by transplanted OECs form appro-
Clinical perspectives
priate nodal sodium channel and conduction is
improved. Several clinical studies are ongoing us-
Clinical investigations in SCI using OEC trans-
ing cell therapy approaches for SCI (Senior, 2002;
plantation are in progress. A recent report tested
Dobkin et al., 2006). Future translational studies
the feasibility and safety of transplantation of
are highly expected to bridge the gap between
autologous OECs into the injured spinal cord in
basic and clinical research in OEC transplantation
human paraplegia in a single blind, Phase I clinical
for SCI.
trial (Feron et al., 2005). In this study, 1 year after
cell implantation, there were no medical, surgical
or other complications to indicate that the proce- Abbreviations
dure is unsafe. They conclude that transplantation
of autologous OECs into the injured spinal cord is BBB Basso, Bresnahan and Beattie (1995;
feasible and is safe up to 1 year post-implantation. i.e., and their rating scale for loco-
Long-term safety studies to compare neurological, motion)
functional and psychosocial outcomes are under- BDNF brain-derived neurotrophic factor
way. Furthermore, fetal brain tissue has been CNS central nervous system
transplanted into the lesions of more than 400 CST corticospinal tract
patients with SCI in China (Senior, 2002; Huang et DMEM Dulbecco’s modified Eagle’s medium
al., 2003). Dobkin et al. (2006) compared available EB ethidium bromide
reports from China with the experiences and EM electron microscopy
objective findings of patients who received preop- FG Fluorogold
erative and post-operative assessments before and K v1 Shaker-type potassium
up to 1 year after receiving cellular implants. They M1 primary motor cortex
concluded that the phenotype and the fate of the Nav voltage-gated sodium
transplanted cells, described as OECs in the China NYU New York University
study, are unknown. Perioperative morbidity and OEC olfactory ensheathing cells
lack of functional benefit were identified as the PNS peripheral nervous system
most serious clinical shortcomings. The proce- SC Schwann cells
dures observed did not attempt to meet interna- SCI spinal cord injury
tional standards for either a safety or efficacy trial. TUNEL terminal deoxynucleotidyltrans-
In the absence of a valid clinical trial protocol, ferase (TdT)-mediated deoxyuridine
Dobkin et al. (2006) suggest that physicians should triphosphate (dUTP)-rhodamine
not recommend this procedure to patients. nick end-labeling

Concluding remarks
Acknowledgments
Remyelination of the injured spinal cord is one of
the key elements for the functional recovery in SCI. This work was supported in part by the Depart-
Extensive experimental rodent models of SCI de- ment of Veterans Affairs, the NIH, and the
scribe the feasibility of OEC transplantation National Multiple Sclerosis Society. The Center
431

for Neuroscience and Regeneration Research is a Doucette, R. (1991) PNS-CNS transitional zone of the first
collaboration of the Paralyzed Veterans of America cranial nerve. J. Comp. Neurol., 312: 451–466.
and the United Spinal Association with Yale Uni- Eshed, Y., Feinberg, K., Poliak, S., Sabanay, H., Sarig-Nadir, O.,
Spiegel, I., Bermingham, J.R. and Peles, E. (2005) Gliomedin
versity. We thank Heather Mallozzi and Margaret mediates Schwann cell–axon interaction and the molecular as-
Borelli for excellent technical assistance. sembly of the nodes of Ranvier. Neuron, 47: 215–229.
Feron, F., Perry, C., Cochrane, J., Licina, P., Nowitzke, A.,
Urquhart, S., Geraghty, T. and Mackay-Sim, A. (2005) Au-
References tologous olfactory ensheathing cell transplantation in
human spinal cord injury. Brain, 128: 2951–2960.
Au, E. and Roskams, A.J. (2003) Olfactory ensheathing cells of Franklin, R.J., Gilson, J.M., Franceschini, I.A. and Barnett,
the lamina propria in vivo and in vitro. Glia, 41: 224–236. S.C. (1996) Schwann cell-like myelination following trans-
Bareyre, F.M., Kerschensteiner, M., Raineteau, O., Mettenlei- plantation of an olfactory bulb-ensheathing cell line into
ter, T.C., Weinmann, O. and Schwab, M.E. (2004) The areas of demyelination in the adult CNS. Glia, 17: 217–224.
injured spinal cord spontaneously forms a new intraspinal Girault, J.A. and Peles, E. (2002) Development of nodes of
circuit in adult rats. Nat. Neurosci., 10: 269–277. Ranvier. Curr. Opin. Neurobiol., 12: 476–485.
Barnett, S.C., Alexander, C.L., Iwashita, Y., Gilson, J.M., Gledhill, R.F. and McDonald, W.I. (1977) Morphological
Crowther, J., Clark, L., Dunn, L.T., Papanastassiou, V., characteristics of central demyelination and remyelination: a
Kennedy, P.G. and Franklin, R.J. (2000) Identification of a single-fiber study. Ann. Neurol., 1: 552–560.
human olfactory ensheathing cell that can effect transplant- Goldin, A.L., Barchi, R.L., Caldwell, J.H., Hofmann, F., Howe,
mediated remyelination of demyelinated CNS axons. Brain, J.R., Hunter, J.C., Kallen, R.G., Mandel, G., Meisler, M.H.,
123: 1581–1588. Netter, Y.B., Noda, M., Tamkun, M.M., Waxman, S.G.,
Basso, D.M., Beattie, M.S. and Bresnahan, J.C. (1995) A sen- Wood, J.N. and Catterall, W.A. (2000) Nomenclature of
sitive and reliable locomotor rating scale for open field testing voltage-dated sodium channels. Neuron, 28: 365–368.
in rats. J. Neurotrauma, 12: 1–21. Graziadei, P.P., Levine, R.R. and Graziadei, G.A. (1978)
Blakemore, W.F. and Crang, A.J. (1985) The use of cultured Regeneration of olfactory axons and synapse formation in
autologous Schwann cells to remyelinate areas of persistent the forebrain after bulbectomy in neonatal mice. Proc. Natl.
demyelination in the central nervous system. J. Neurol. Sci., Acad. Sci. U.S.A., 75: 5230–5234.
70: 207–223. Greenfield, S., Brostoff, S., Eylar, E.H. and Morell, P. (1973)
Blakemore, W.F. and Murray, J.A. (1981) Quantitative exam- Protein composition of myelin of the peripheral nervous sys-
ination of internodal length of remyelinated nerve fibres in tem. J. Neurochem., 20: 1207–1216.
the central nervous system. J. Neurol. Sci., 49: 273–284. Hains, B.C., Black, J.A. and Waxman, S.G. (2003) Primary
Boiko, T., Rasband, M.N., Levinson, S.R., Caldwell, J.H., cortical motor neurons undergo apoptosis following axoto-
Mandel, G., Timmer, J.S. and Matthews, G. (2001) Compact mizing spinal cord injury. J. Comp. Neurol., 462: 328–341.
myelin dictates the differential targeting of two sodium chan- Hildebrand, C., Kocsis, J.D., Berglund, S. and Waxman, S.G.
nel isoforms in the same axon. Neuron, 30: 91–104. (1985) Myelin sheath remodeling in regenerated rat sciatic
Boyd, J.G., Lee, J., Skihar, V., Doucette, R. and Kawaja, M.D. nerve. Brain Res., 358: 163–170.
(2004) LacZ-expressing olfactory ensheathing cells do not Honmou, O., Felts, P.A., Waxman, S.G. and Kocsis, J.D. (1996)
associate with myelinated axons after implantation into the Restoration of normal conduction properties in demyelinated
compressed spinal cord. Proc. Natl. Acad. Sci. U.S.A., 17:
spinal cord axons in the adult rat by transplantation of ex-
2162–2166.
ogenous Schwann cells. J. Neurosci., 16: 3199–3208.
Brook, G.A., Plate, D., Franzen, R., Martin, D., Moonen, G.,
Huang, H., Chen, L., Wang, H., Xiu, B., Wang, R., Zhang, J.,
Schoenen, J., Schmitt, A.B., Noth, J. and Nacimiento, W.
Zhang, F., Gu, Z., Li, Y., Song, Y., Hao, W., Pang, S. and
(1998) Spontaneous longitudinally orientated axonal regen-
Sun, J. (2003) Influence of patients’ age on functional recov-
eration is associated with the Schwann cell framework within
the lesion site following spinal cord compression injury of the ery after transplantation of olfactory ensheathing cells into
rat. J. Neurosci. Res., 53: 51–65. injured spinal cord injury. Chin. Med. J. (Engl.), 116:
Caldwell, J.H., Schaller, K.L., Lasher, R.S., Peles, E. and 1488–1491.
Levinson, S.R. (2000) Sodium channel Na (v) 1.6 is localized Imaizumi, T., Lankford, K.L., Waxman, S.G., Greer, C.A. and
at nodes of Ranvier, dendrites, and synapses. Proc. Natl. Kocsis, J.D. (1998) Transplanted olfactory ensheathing cells
Acad. Sci. U.S.A., 97: 5616–5620. remyelinate and enhance axonal conduction in the demyelin-
Chuah, M.I. and West, A.K. (2002) Cellular and molecular bi- ated dorsal columns of the rat spinal cord. J. Neurosci., 18:
ology of ensheathing cells. Microsc. Res. Tech., 58: 216–227. 6176–6185.
Dobkin, B.H., Curt, A. and Guest, J. (2006) Cellular trans- Imaizumi, T., Lankford, K.L., Burton, W.V., Fodor, W.L. and
plants in China: observational study from the largest human Kocsis, J.D. (2000a) Xenotransplantation of transgenic pig
experiment in chronic spinal cord injury. Neurorehabil. Neu- olfactory ensheathing cells promotes axonal regeneration in
ral Repair, 20: 5–13. rat spinal cord. Nat. Biotechnol., 18: 949–953.
432

Imaizumi, T., Lankford, K.L. and Kocsis, J.D. (2000b) Trans- Novakovic, S.D., Deerinck, T.J., Levinson, S.R., Shrager, P.
plantation of olfactory ensheathing cells or Schwann cells and Ellisman, M.N. (1996) Cluster of axonal Na+ channels
restores rapid and secure conduction across the transected adjacent to remyelinating Schwann cells. J. Neurocytol., 25:
spinal cord. Brain Res., 854: 70–78. 403–412.
Jenkins, S.M. and Bennett, V. (2002) Developing nodes of Novakovic, S.D., Levinson, S.R., Schachner, M. and Shrager, P.
Ranvier are defined by ankyrin-G clustering and are inde- (1998) Disruption and reorganization of sodium channels in
pendent of paranodal axoglial adhesion. Proc. Natl. Acad. experimental allergic neuritis. Muscle Nerve, 21: 1019–1032.
Sci. U.S.A., 99: 2303–2308. Peles, E. and Salzer, J.L. (2000) Molecular domains of myelin-
Kaplan, M.R., Cho, M.H., Ullian, E.M., Isom, L.L., Levinson, ated axons. Curr. Opin. Neurobiol., 10: 558–565.
S.R. and Barres, B.A. (2001) Differential control of clustering Plant, G.W., Christensen, C.L., Oudega, M. and Bunge, M.B.
of the sodium channels Na(v)1.2 and Na(v)1.6 at developing (2003) Delayed transplantation of olfactory ensheathing glia
CNS nodes of Ranvier. Neuron, 30: 105–109. promotes sparing/regeneration of supraspinal axons in the
Kaplan, M.R., Meyer-Franke, A., Lambert, S., Bennett, V., contused adult rat spinal cord. J. Neurotrauma, 20: 1–16.
Duncan, I.D., Levinson, S.R. and Barres, B.A. (1997) Radtke, C., Akiyama, Y., Brokaw, J., Lankford, K.L.,
Induction of sodium channel clustering by oligodendrocytes. Wewetzer, K., Fodor, W.L. and Kocsis, J.D. (2004)
Nature, 386: 724–728. Remyelination of the nonhuman primate spinal cord by
Kato, T., Honmou, O., Uede, T., Hashi, K. and Kocsis, J.D. transplantation of H-transferase transgenic adult pig
(2000) Transplantation of human olfactory ensheathing cells olfactory ensheathing cells. FASEB J., 18: 335–337.
elicits remyelination of demyelinated rat spinal cord. Glia, 30: Raisman, G. (2001) Olfactory ensheathing cells – another
209–218. miracle cure for spinal cord injury? Nat. Rev. Neurosci., 2:
Keirstead, H.S. (2005) Stem cells for the treatment of myelin 369–375.
loss. Trends Neurosci., 28: 677–683. Ramer, L.M., Au, E., Richter, M.W., Liu, J., Tetzlaff, W. and
Keyvan-Fouladi, N., Li, Y. and Raisman, G. (2002) How do Roskams, A.J. (2004) Peripheral olfactory ensheathing cells
transplanted olfactory ensheathing cells restore function? reduce scar and cavity formation and promote regeneration
Brain Res. Brain Res. Rev., 40: 325–327. after spinal cord injury. J. Comp. Neurol., 473: 1–15.
Keyvan-Fouladi, N., Raisman, G. and Li, Y. (2003) Functional Ramon-Cueto, A. and Avila, J. (1998) Olfactory ensheathing
repair of the corticospinal tract by delayed transplantation of glia: properties and function. Brain Res. Bull., 46: 175–187.
olfactory ensheathing cells in adult rats. J. Neurosci., 23: Ramon-Cueto, A., Cordero, M.I., Santos-Benito, F.F. and
9428–9434. Avila, J. (2000) Functional recovery of paraplegic rats and
Kocsis, J.D. and Sasaki, M. (2005) Transplantation of periph- motor axon regeneration in their spinal cords by olfactory
eral myelin-forming cells to repair demyelinated axons. ensheathing glia. Neuron, 25: 425–435.
In: Waxman S.G. (Ed.), Multiple Sclerosis as a Neuronal Ramon-Cueto, A., Plant, G.W., Avila, J. and Bunge, M.B.
Disease. Elsevier, San Diego, pp. 421–433. (1998) Long-distance axonal regeneration in the transected
Li, B., Lankford, K.L. and Kocsis, J.D. (2004) Distribution adult rat spinal cord is promoted by olfactory ensheathing
and migration of GFP-expressing mouse Schwann cells and glia transplants. J. Neurosci., 18: 3803–3815.
olfactory ensheathing cells transplanted into contused spinal Rasband, M.N., Peles, E., Trimmer, J.S., Levinson, S.R., Lux,
cord of the rat. Abstract of 34th Annual meeting, Society for S.E. and Shrager, P. (1999) Dependence of nodal sodium
Neuroscience. channel clustering on paranodal axoglial contact in the
Li, Y., Field, P.M. and Raisman, G. (1997) Repair of adult rat developing CNS. J. Neurosci., 19: 7516–7528.
corticospinal tract by transplants of olfactory ensheathing Rasband, M.N. and Trimmer, J.S. (2001) Developmental
cells. Science, 277: 2000–2002. clustering of ion channels at and near the node of Ranvier.
Li, Y., Field, P.M. and Raisman, G. (1998) Regeneration Dev. Biol., 236: 5–16.
of adult rat corticospinal axons induced by transplanted Rasband, M.N., Trimmer, J.S., Schwarz, T.L., Levinson, S.R.,
olfactory ensheathing cells. J. Neurosci., 18: 10514–10524. Ellisman, M.N., Schachner, M. and Schrager, P. (1998)
Lu, J., Feron, F., Mackay-Sim, A. and Waite, P.M. (2002) Potassium channel distribution, clustering, and function in
Olfactory ensheathing cells promote locomotor recovery remyelinating rat axons. J. Neurosci., 18: 36–47.
after delayed transplantation into transected spinal cord. Rios, J.C., Rubin, M., St Martin, M., Downey, R.T., Einheber,
Brain, 125: 14–21. S., Rosenbluth, J., Levinson, S.R., Bhat, M. and Salzer, J.L.
McBride, R.L., Feringa, E.R., Garver, M.K. and Williams Jr., (2003) Paranodal interactions regulate expression of sodium
J.K. (1989) Prelabeled red nucleus and sensorimotor cortex channel subtypes and provide a diffusion barrier for the node
neurons of the rat survive 10 and 20 weeks after spinal cord of Ranvier. J. Neurosci., 23: 7001–7011.
transection. J. Neuropathol. Exp. Neurol., 48: 568–576. Rosenzweig, E.S. and McDonald, J.W. (2004) Rodent models
Namiki, J., Kojima, A. and Tator, C.H. (2000) Effect of for treatment of spinal cord injury: research trends and
brain-derived neurotrophic factor, nerve growth factor, and progress toward useful repair. Curr. Opin. Neurol., 17(2):
neurotrophin-3 on functional recovery and regeneration after 121–131.
spinal cord injury in adult rats. J. Neurotrauma, 17: Sasaki, M., Black, J.A., Lankford, K.L., Tokuno, H.A.,
1219–1231. Waxman, S.G. and Kocsis, J.D. (2006a) Molecular
433

reconstruction of nodes of Ranvier after remyelination by Takami, T., Oudega, M., Bates, M.L., Wood, P.M., Kleitman,
transplanted olfactory ensheathing cells in the demyelinated N. and Bunge, M.B. (2002) Schwann cell but not olfactory
spinal cord. J. Neurosci., 26: 1803–1812. ensheathing glia transplants improve hindlimb locomotor
Sasaki, M., Hains, B.C., Lankford, K.L., Waxman, S.G. and performance in the moderately contused adult rat thoracic
Kocsis, J.D. (2006b) Protection of corticospinal tract neurons spinal cord. J. Neurosci., 22: 6670–6681.
after dorsal spinal cord transection and engraftment of Vabnick, I., Messing, A., Chiu, S.Y., Levinson, S.R., Schachner,
olfactory ensheathing cells. Glia, 53: 352–359. M., Roder, J., Li, C., Novakovic, S. and Shrager, P. (1997)
Sasaki, M., Lankford, K.L., Zemedkun, M. and Kocsis, J.D. Sodium channel distribution in axons of hypomyelinated and
(2004) Identified olfactory ensheathing cells transplanted into MAG null mutant mice. J. Neurosci. Res., 50: 321–336.
the transected dorsal funiculus bridge the lesion and form Wang, H., Kunkel, D.D., Martin, T.M., Schwartzkroin, P.A.
myelin. J. Neurosci., 24: 8485–8493. and Tempel, B.L. (1993) Heteromultimeric K+ channels in
Schwab, J.M., Brechtel, K., Mueller, C.A., Failli, V., Kaps, H.P., terminal and juxtaparanodal regions of neurons. Nature, 365:
Tuli, S.K. and Schluesener, H.J. (2006) Experimental strate- 75–79.
gies to promote spinal cord regeneration: an integrative per- Weiner, L.P., Waxman, S.G., Stohlman, S.A. and Kwan, A.
spective. Prog. Neurobiol., 78: 91–116. (1980) Remyelination following viral-inducted demyelina-
Senior, K. (2002) Olfactory ensheathing cells to be used in tion: ferric ion-ferrocyanide staining of nodes of Ranvier
spinal-cord repair trial. Lancet Neurol., 1: 269. within the CNS. Ann. Neurol., 8: 580–583.
This page intentionally left blank
Subject Index

4-aminopyridine (4-AP) 220, 221 astrocyte swelling 66, 189


a-CaMKII 153 astrocytes 21, 31, 34, 61–70, 72, 73, 83, 88, 89, 98,
aII-spectrin 333, 413, 414 103, 117, 121, 129, 158, 176, 187–190, 196,
acceleration input 29 227, 228, 317, 318, 320, 331–333, 342, 343,
acceleration/deceleration 68, 158, 264, 269–271, 371, 420, 422, 425
284 astrocytic cascades 34
acetylcholine 62, 274 reactive astrocytes 304
acidosis 61, 65, 66, 113, 120, 121 astroglia 61, 175, 282, 318, 402 (see also glial cells)
acute spinal cord injury study 217, 219, 222 athletes 253–258, 263, 280, 282
acute subdural hematoma (ASDH) 118, 239, 266, ATP 33, 34, 62–65, 69, 70, 72, 85, 87, 111,
293, 294, 296, 298, 300 (see also subdural 113–115, 117, 121, 122, 164, 279, 280, 318,
hematoma) 355, 356, 358, 403
Akt 69, 343, 359, 360 autophagy 82, 84, 85 (see also cell death)
Alzheimer’s disease 81, 253, 255, 303–311, 346 autoregulation 112, 113, 131, 210–212, 281, 282
(see also neurodegenerative disease) metabolic 112, 119
AMPA receptor 68, 70, 155, 159–162, 164, 279, pressure 112, 119
332 viscosity 112, 119
amyloid precursor protein (APP) 48, 86–89, 130, axolemma 52, 175, 271, 285
255, 272–274, 303–308, 310, 311 axonal damage 49, 90, 144, 267, 269, 271, 275, 278
amyloid-b peptides 304 axotomy 43, 48, 263
antibody microarray 410, 411 delayed 273, 274
aphasia 346–348 primary 285
apnea 281, 295, 298, 299, 300 secondary 263, 271, 273, 274, 277–279, 285
APOE genotype 303, 310, 311
apolipoprotein E 255, 303 BACE inhibitors 305
apolipoprotein E4 310 Bad 47, 359
apoptosis inducing factor (AIF) 84, 87 basal ganglia 99, 295–297, 306
apoptosis 35, 43–45, 47, 67, 81–88, 90, 121, 127, Bax 47, 84, 86
128, 218, 223, 224, 283, 319, 343, 355, 403, Bcl-2 47, 69, 70, 72, 84, 86, 343
415, 428, 429 (see apoptotic) Bergmann glia 331–333
apoptotic 34, 35, 43–48, 67, 81–87, 126, 128, 131, big black brain 293–295, 297–300
132, 135, 223, 300, 304, 342, 358, 359, 403, biochemical markers 130, 282, 317, 401, 406, 409,
428 414, 415
apparent diffusion co-efficient (ADC) mapping biomarkers 30, 35, 317, 401, 403, 411–415
236 biomechanical mechanisms 28
aquaporins 185 biomechanics 13, 18, 23–25, 27, 329, 335
arachidonic acid 175, 357 acceleration 17
arterio venous difference of oxygen (AVDO2) 112, deceleration 17
119, 120, 208, 210 deformations 14

435
436

force 14 gunshot wounds 4


shear strain 15 traffic injuries 4
strains 14 cell culture 63, 85, 175, 256, 305, 318, 319, 343,
stress 14 356, 369, 376, 379, 381
blood brain barrier (BBB) 88, 97–99, 102, 104, 115, cell death (CD) 13, 20, 21, 23, 24, 33–35, 43–48, 66,
117, 126, 136, 143, 144, 158, 186, 188, 190, 67, 72, 81–87, 114, 122, 125, 126, 128, 131,
237, 246, 282, 317, 319, 321, 356, 359, 395, 132, 135, 144, 147, 153, 164, 218, 255, 257,
402 263, 279, 304, 306, 319, 327, 329–331, 335,
blunt trauma 158, 268, 270 356, 359, 402, 403, 428 (see also apoptosis,
bone marrow stem cells (BMSCs) 225 necrosis, autophagy)
bone marrow stromal cells 217, 226, 227 cell membrane damage 19, 20
brain derived neurotrophic factor (BDNF) 62, cellular 43
229, 343, 359, 429 mechanisms 62, 147, 156, 157, 259, 405
brain edema 28, 86, 187, 235, 356, 393, 395, 396, morphology 61
398 response 13, 16, 18, 19, 21, 22, 43, 132, 177, 284
brain tissue oxygen tension 113, 207–209 cerebellar injury 327
brain water 185–189, 191, 192, 356, 396 cerebellum 44, 62, 65, 175, 256, 257, 327–329, 331,
bromdeoxyuridine (BrdU) 321, 322, 369, 370 333, 335
burst firing 195, 196, 200, 201 cerebral blood flow (CBF) 111, 112, 186, 208, 236,
263, 278, 281, 296
calcineurin 53, 88, 280 cerebral blood volume (CBV) 112, 113
calcium 31–34, 46, 47, 52, 53, 55, 62–64, 66, 68–70, cerebral contusion 27, 66, 235, 266, 283, 405
82, 87, 98, 114, 121, 135, 151, 153, 158, 218, cerebral cortex 48, 83, 130, 135, 145, 146, 165, 257,
274, 275, 279, 280, 304, 318, 319, 331, 402, 307, 327, 335
403 cerebral edema 102, 120, 185–188, 190, 246, 279
calcium ATPase 273, 279 (see also edema)
calcium dysregulation 46, 47, 52, 53 cytotoxic edema 185, 188–190, 192
calcium homeostasis 122, 279, 304, 318, 402 vasogenic edema 185, 188, 190–192
calcium oscillations 62 cerebral energy metabolism 114, 115
calcium signaling 62 cerebral ischemia 65–67, 87, 120, 127, 130, 136,
calcium waves 62 396 (see also ischemia)
intracellular Ca2+ 46, 68 cerebral metabolic rate of oxygen (CMRO2) 112,
intracellular stores 62 119, 120, 208, 210, 281
calpain 45–47, 49, 52, 53, 81, 82, 85–88, 91, cerebral oxygen tension 111
98, 131, 135, 218, 279, 280, 333, 403, 411, cerebral oxygen transport 111, 118
414 cerebral oxygenation 121, 207, 209
calpain inhibitors 87, 88, 91 cerebral perfusion pressure 112, 120, 207
calpain proteolysis 414, 415 cerebral tissue PO2 111, 113, 121
caspases 45–47, 81, 84, 85, 122, 218, 304, 306, 403 cerebrospinal fluid (CSF) 98, 119, 185, 266, 299,
caspase-3 33, 47, 67, 83–85, 87, 89, 132, 306, 307, 320, 386, 406, 414
358, 403, 414, 415 cerebrovascular permeability 237, 238
caspase-12 83, 84 cerebrovascular reactivity 112
caspase-3 proteolysis 414, 415 carbon dioxide (CO2) reactivity 112
caspase-inhibitor 85 Cethrins 217, 223, 224, 229 (see also Rho
causes of neurotrauma 3–6 antagonist)
accidents 4 c-fos 343
falls 4 CGRP 99, 101, 102, 196
firearm injuries 5 child abuse 253, 254, 293
437

chloride 98 diffuse axonal injury (DAI) 17, 27, 31, 49, 53, 55,
chondroitin sulfate proteoglycan (CSPG) 132 126, 264, 266, 298, 306, 402
citric acid cycle 111, 115, 117, 118 diffuse head injuries 270
c-jun 343 diffuse injuries 17, 27, 29, 43, 44, 55, 56, 90, 335
cognitive dysfunction 253–255, 257 (see also DTBI)
cognitive performance 320, 321, 345 diffuse traumatic brain injury (DTBI) 43–49, 53,
collagen gel 385–388 55, 56 (see also diffuse injuries)
community costs 6 drug delivery 369, 385–388
computer tomography (CT) 119, 402 delivery strategies 385
concussions 254, 264 injectable delivery 386
controlled cortical impact (CCI) 67, 86, 88, 90, intrathecal drug delivery 385
127, 271, 319, 395, 405, 412 dynamic brain injury 404
contusion volume 136, 283, 284, 395, 396 dynamic loading 15, 16, 265
contusions 17, 27, 44, 45, 98, 120, 207, 235,
237–239, 241, 266, 270, 283, 298, 395, 396 edema 28, 65, 66, 86, 88, 97–99, 101–103, 119, 120,
corpus callosum 55, 126, 266, 267, 334, 342, 371, 126, 131, 136, 185–192, 235, 237–240, 246,
378 296, 356, 359, 393, 395, 396, 402, 405, 419
cortical circuits 200 (see also cerebral edema)
cortical contusion 144, 270 cytotoxic edema 65, 98, 119, 120, 126, 185, 188,
craniectomy 212, 299, 393–398 189, 190, 192
critical time frames 244 vasogenic edema 97–99, 102, 104, 185, 190–192,
crossed cerebellar diaschisis (CCD) 329 237, 238, 396
cross-tolerance 256, 257, 353, 356, 358, 359 electrophysiology 62, 143, 144, 156, 158, 159, 165,
CSF production 185–187, 192 327
CT scan 4, 6, 119, 120, 239, 243, 244, 246–248, 270, energy 45, 65, 82, 84, 85, 87, 90, 111, 114, 115, 117,
283, 294 118, 121, 268–270, 280, 281, 304, 319, 358,
0 0
cyclic guanosine 3 ,5 -monophosphate (cGMP) 404, 406
171, 173 energy metabolism 111, 114, 118, 121, 319
cGMP synthesis 174 enzyme-linked immunosorbant assays (ELISA) 413
cyclosporine A (CsA) 81, 88 epidemiology 3, 264, 303, 328
cysteine proteases 43, 45, 46, 52, 53, EURODERM study 309
403 (see also calpains and caspases) MIRAGE study 309, 310
cytochrome C 46, 52, 83, 122, 223 epidermal growth factor 385
cytokines 87, 89, 127, 176, 225, 279, 358, 359, 368, erythropoietin (Epo) 353, 357, 358
410 European Union Directive 244, 247
IL-1b 127 excitatory amino acid (EAA) 46, 90, 97, 175, 279,
IL-6 127 299, 342, 402
TNFa 127 excitatory neurotransmitters 279
cytoskeleton 31, 46, 52, 53, 87, 272, 274, 275, 277, excitotoxicity 33, 65, 67, 218, 223, 330, 346, 403
279, 285, 318 experimental models 13, 27–29, 90, 115, 187, 188,
222, 248, 253–256, 285, 296–298, 300, 356,
decompression craniectomy 299, 393–398 357, 381, 394, 419
dementia pugilistica 254, 255, 258, 306 cellular injury models 14
demyelination 125, 128, 131, 132, 220, 221, 227, compression injury 18, 385–388
228, 419, 420, 422 contusion 18
depolarization 33, 55, 62, 67, 68, 155–157, 164, fluid percussion injury 18, 72, 144, 271, 277, 319,
274, 276, 278–280, 331, 403 320, 327, 405
dexanabinol 244, 246, 247 of stroke 369
438

piglet subdural injection models 298 GLUT transporters 115


rodent subdural hematoma model 296 GLUT-1 115
weight drop 18, 30, 155, 156, 254, 255, 257, 405, GLUT-3 115
429 glutamate 17, 32, 46, 64–65, 67, 117, 148, 159–162,
extracellular signal regulated kinase (ERK) 218, 223, 254, 279, 296, 318, 319, 332, 342,
34, 69 402, 403
ERK1 343 glutamate receptors 31, 32, 63, 65, 160, 162, 279,
ERK2 343 318
N-methyl D-aspartate receptors (NMDAR)
Fampridines 220, 221, 229 (see also 31–35, 159, 160, 161
4-aminopyridine or 4-AP) a-Amino-3-hydroxy-5-methyl-4-isoxazole-
fas ligand 84 propionic acid receptors (AMPAR) 32, 33,
field extracellular postsynaptic potentials (fEPSP) 160–162, 164
147 metabotropic glutamate receptors (mGluRs) 32,
finite element analysis (FEA) 13, 21, 23, 24 63, 160, 318
fluid percussion injury 18, 144, 271, 277, 279, 319, glutamate transporters 65, 332
320, 327, 405 (see also lateral fluid GLT-1 65
percussion injury) GLAST 65
focal head injuries (FHI) 270 glycolysis 65, 84, 111, 115–117, 121, 279,
focal injuries 16, 27, 44, 270, 331 280
free radicals 35, 46, 47, 85, 136, 159, 177, 279, 342, GM-1 218, 222, 228
357, 402 granulocyte colony stimulating factor (G-CSF)
funding 7, 8, 24, 259, 391 367, 371, 375
fura-2 158 growth factor 135, 226, 320, 358, 368, 379, 385,
391
g-aminobutyric acid (GABA) 33, 62, 146, 148, 150,
156, 162, 163, 171 Hagen–Poiseuille equation 112, 119
GABAA receptor 147, 149–151, 158, 162, 342 head injury 4, 6, 17, 18, 30, 47, 87, 88, 111,
gap junctions 63, 66, 72, 73 118–122, 257, 264–268, 271, 274, 280–284,
gene expression 30, 34, 47, 98, 100, 329, 331, 293, 294, 298, 299, 306, 307, 309, 310, 329,
398 356–357, 402, 404
GFP 371, 420, 422, 427, 429, 430 minor 6
Glasgow Coma Score (GCS) 281, 413 moderate 6
Glasgow outcome scale (GOS) 240, 328 severe 6
Glasgow coma scale (GCS) 240 heat acclimation 257, 353–360
glia 30, 33, 34, 62–64, 67, 81–83, 86, 98, 103, 117, heat shock proteins 331, 355
118, 144, 158, 160, 186, 225, 256, 284, helmets 7, 14, 97, 263, 264
331–333, 342, 428 bicycle helmets 7
glial cells 30, 61, 64, 98, 114, 144, 160, 228, hockey helmets 268
236, 279, 283, 304, 317, 333, 340, motorcycle helmets 7
342, 358 hematoma 16, 27, 44, 66, 67, 118–120, 209, 210,
glial fibrillary acidic protein (GFAP) 61, 64, 67, 228, 239, 266, 268, 270, 293–296, 298, 300
134, 196, 282, 422 hemicraniectomy 299, 300
glial scar 64, 226, 331, 379 high-throughput immunoblotting 411
glial swelling 44, 66, 98, 126 hippocampal 20, 63, 66, 85, 86, 144–148, 156,
glucose 65–67, 111, 115, 117, 120, 122, 278, 280, 160, 162, 164, 253, 256, 304, 306, 308,
281, 319 319–321, 341, 343, 355, 411
glucose metabolism 120, 278, 281 (see also hippocampus)
439

hippocampus 44, 45, 48, 62, 64, 67, 72, 83, 86, 89, incidence of spinal cord injury 4
99, 101, 126, 136, 145–147, 151, 153, 161, incidence of traumatic brain injury 4
162, 225, 254–257, 307, 308, 310, 320, 321, infancy 293, 294, 298, 344
327, 329, 341–343, 346 inflammation 98, 127, 128, 135, 160, 176, 178, 188,
CA1 145 218, 224, 300, 340, 346, 353, 357, 359, 388,
CA3 145 410
dentate gyrus (DG) 145 neurogenic inflammation 97, 100–104
perforant pathway 153 inflammatory processes 127, 130, 134, 348
histopathology of cerebral contusion 235 inflammatory response 45, 46, 101, 128, 130, 136,
hormonal cycling 339, 346 176, 177, 224, 284, 357, 389, 390, 402
hormonal status 339–341, 344–348 informed consent 243–248
hyaluronan and methylcellulose (HAMC) 385, inhibitory postsynaptic current (sIPSC) 150,
388–391 156
hyaluronan 385, 388 inositol-1,4,5 trisphosphate (IP3) 32, 34, 68,
hydrogel 18, 367, 379, 385, 386, 391 69, 72
hydrolytic enzymes 43, 47, 56 IP3 receptor 63
hyperglycolysis 120, 280 intensive care 6, 118, 243, 249, 281, 299, 415
hyperventilation 98, 112–114, 119, 120, interleukin-1 127
210–212 interleukin-1b (IL-1b) 223, 359
hypotension and hypoxia 118 international neurotrauma society (INTS) 3, 8
hypothermia 81, 90, 91, 98, 135, 136, 211, 212, 247 intracranial pressure (ICP) 6, 28, 49, 66, 98, 112,
hypoxia 65–67, 90, 102, 115, 118, 119, 121, 118, 120, 186, 207, 235, 281, 296, 299, 300,
122, 210, 211, 296, 298, 300, 301, 343, 393, 403
357–359 monitoring 111
hypoxia inducible factor-1 353, 357, 358 ionic fluxes 278, 281
ionic homeostasis 31, 33, 45, 66, 82, 111, 144, 164,
ice hockey 263, 265, 267–271, 275, 286 218, 263
ICP and cerebral circulation 119, 120 ischemia 64–68, 72, 87, 88, 102, 120, 121, 127, 130,
immunoreactivity 64, 67, 87, 89, 99, 100, 103, 177, 136, 158, 207, 209, 210, 222, 236, 254, 256,
255 282, 284, 296, 298, 300, 322, 329, 343, 346,
impact loading 16, 266 354, 356, 357, 359, 396, 419
impulsive loading 16, 266, 267 ischemic preconditioning (IPC) 256, 343
in vitro models 18, 21, 24, 27–30, 34, 35, 68, 143,
144, 158, 253, 257 jugular bulb oximetry 208
cell/substrate stretch model 30 jugular bulb oxygen saturation (SjvO2) 111, 120,
scratch model/laceration 30 121, 207–212
weight drop/compression 30
in vitro TBI models 14, 18, 159 kainate receptors 148
(see also experimental models)
in vivo animal models 17, 278 laceration 30, 83, 97, 131, 266
(see also experimental models) lactate 65, 84, 111, 113, 115, 117, 120, 121, 210,
in vivo imaging 35, 377, 378 211, 280, 319
in vivo models 17, 18, 21, 27–29, 31, 33, 143, 144, lactate-to-pyruvate ratio 121, 211
253, 354, 358 laser doppler flowmetry 111
cortical impact model 29, 86 lateral fluid percussion injury (LFPI) 72, 144, 145,
fluid percussion model 29 147, 148, 150–153, 155, 156, 164, 279 (see
incapacity 244, 246 also fluid precursion injury)
acute incapacity 247 lesion progression 131, 235
440

loads 14–19, 266 mild head injury 6, 274, 309


dynamic loading 15, 16, 265 mild injury 73, 144, 158, 160, 253, 254, 263
impact loading 16, 266 miniature inhibitory postsynaptic currents
impulsive loading 16, 266, 267 (mIPSCs) 150, 151, 156
static loading 15 minocycline 217, 223
long-term depression (LTD) 151, 153 minor traumatic brain injury (mTBI) 263 (see also
long-term potentiation (LTP) 149, 151–153, 161, mild injury)
257, 320, 341 minor head injury 6 (see also mild head injury)
low molecular weight antioxidants (LMWA) mitochondria 33, 34, 44, 52, 68, 72, 84–86, 90,
357 113–115, 117, 118, 121, 271, 275, 279, 280
lucifer yellow 19, 153 electron transport chain 85, 117
lysosomal enzymes 47, 48 (see also hydrolytic mitochondrial function 111, 121, 122, 159
enzymes) mitochondrial permeability transition pore
cathepsin 47 (MPTP) 84, 122
mitogen activated protein kinase (MAPK) 33–35,
macrophage 98, 128, 130, 134, 176–178, 224, 225, 69
227, 370, 373, 381, 389, 420 MK-801 159
magnesium 31, 98, 102, 103 Monro–Kellie doctrine 119
magnetic resonance imaging (MRI) 127, 134, 227, morphological injury 97
228, 235, 270, 283, 296, 341, 348, 367, 368, morris water maze (MWM) 145, 254, 255, 320
377, 381, 402, 414 multi modal monitoring 207
diffusion-weighted MR 236 myelin 129, 136, 137, 220, 223–226, 228, 274,
marker (see biomarker) 282, 342, 371, 420–427, 429
mass spectrometry 401, 403, 406, 407, 409, 411, (see also remyelination, demyelination)
415
tandem mass spectrometry 409, 410 Na+-K+ ATPase 65, 164
matrix metalloproteases 85 Na+/K+ ATPase activity 164
mechanical permeability 32 nanoparticles 369, 373, 375, 377, 381
mechanical response 13–16, 21, 23, 24 iron oxide 367, 368, 371, 379
mechanoporation 32, 48, 49, 84, 88, 98 near infra red spectroscopy (NIRS) 111, 207,
dextrans 48, 50, 51, 53 209–211
horseradish peroxidase 48 necrosis (see necrotic)
membrane resealing 43, 49 aponecrosis 45
mediators 61, 87, 98, 102, 130, 224, 226, 279, 359, necrotic 34, 43–49, 67, 82, 85–88, 90, 126, 218, 235,
360 238–241, 395, 403, 419
metabolic cascade 278, 357 nerve growth factor (NGF) 226, 343
metabotropic glutamate receptors (mGluRs) 32, NeuN 322, 370
34, 63, 69, 70, 160, 318 neural progenitor cells (NPCs) 225, 226, 369
mGluR1 160 neural stem cells (NSCs) 136, 225, 226, 320
mGluR5 160 neurodegenerative disease 45, 125, 253, 255,
methylcellulose 385, 388 256, 403 (see also Alzheimer’s disease,
methylprednisolone 135, 178, 217, 218, 221, 222, Parkinson’s disease)
228, 229, 385 neuroexcitation 46
MicroBeads 367, 377, 378, 381 neurofibrillary tangles 304, 307, 308, 310
microdialysis 46, 111, 120, 121, 175, 211, 278, 296 neurofilaments 46, 88, 271–274, 277, 278, 280, 285,
microglia 31, 35, 89, 98, 130, 176, 196, 304, 329, 379
359, 370, 373, 389, 402 neurogenesis 225, 226, 320–322, 340
microtubules 271–274, 277, 279, 280, 285 neurokinin A (NKA) 99–102
441

neurokinin receptors 100 (see also tachykinin patch clamping 158, 333
receptors) pathogenesis 45, 51, 53, 56, 67, 83, 87, 89, 130, 187,
neuron specific enolase (NSE) 68, 256, 282, 283 190, 303, 304, 357
neuronal cascades 33 pathomechanisms 125, 126, 128, 131, 136
neuronal transmitter 62, 171, 173 pathophysiology 13, 23, 61, 65, 70, 73, 97, 99, 111,
neuropeptides 97–102, 104 118, 125–127, 131, 185, 195, 235, 275, 283,
neuroprotection 3, 61, 64, 217, 223, 342–344, 353, 293, 296, 299, 300, 327, 335, 359, 393, 395,
354, 356–360, 428, 430 397, 419
neuroproteomics 401, 403, 409, 411, 413, 415 patient protection 243, 248
neurotransmitter receptors 62, 158 peptide chromatography 407, 409
NF200 333 peroxynitrite (ONOO) 175
NF-kappaB 130 pH 66, 72, 117, 120, 356, 368
nicotinamide adenine dinucleotide brain pH 65, 121, 209
phosphate diaphorase (NADPHd) pH paradox 121
171–173, 177 pharmacological therapies 157, 218, 229, 248
nitric oxide 46, 88, 89, 98, 101, 113, 136, 171–179, phospholipases 68, 279
223 phospholipase A2 280
nitric oxide synthase (NOS) 46, 88, 171, 318 phospholipase C 32, 63, 280, 305
brain nitric oxide synthase (bNOS) 172 polypharmacia 91
calcium-dependent nitric oxide synthase (cNOS) poly-hydroxypropylmethacrylamide 367, 379
176 population spike amplitudes (PSA) 343, 344
inducible NOS (iNOS) 136, 176 positron emission tomography (PET) 120, 207,
neuronal nitric oxide synthase (nNOS) 175 208, 210–212, 278
NK1 receptor antagonists 103, 104 post-tetanic potentiation (PTP) 151
NMDA antagonists 34 potassium channel blockers 217, 221
NMDA receptor 46, 64, 68, 84, 148, 159, 160, 162, potassium channels 220, 221, 227, 422, 424
175, 279, 341 antagonism of 220
N-methyl-D-aspartate (NMDA) 148, 175, 279 potassium 33, 98, 117, 217, 218, 220, 279
nodal reconstruction 422 preconditioning 256, 257, 344, 353, 355
nogo 223, 224 ischemic preconditioning 256, 343
anti-NOGO monoclonal antibodies 217 presenilin 304, 305, 306
pressure changes 28, 398
olfactory ensheathing cells (OECs) 217, 225, pressure volume index 119
228, 369, 419, 420, 422, 424, 425, presynaptic hyperexcitability 329–331
428–430 primary damage 13, 19, 23, 395, 396
oligodendrocytes 35, 125, 128, 132, 176, 224, 226, primary phase 14, 19
227, 342, 371, 420, 425 progenitor cells 136, 225, 226, 320, 367–369, 372,
oligodendroglial precursor cells (OPCs) 225, 227 377, 420, 425
osmotic potential 239 programmed cell death 82, 126 (see also apoptosis)
oxidative phosphorylation 65, 111, 115, 117, 118, progressive injury 125, 127, 128, 130, 132, 135,
280, 281 136
oxidative stress 97, 122, 308, 356, 357 propidium iodide 70, 72, 158, 256
oxygen and hemoglobin 113, 114 prostaglandins 113, 136, 279
proteases 43, 45, 46, 52, 53, 55, 82, 84, 86, 100, 122,
pain 99, 101, 104, 134, 135, 195–201, 229 280, 305, 306, 403
Parkinson’s disease 81, 130, 371, 403 (see also protein kinase C (PKC) 46, 160, 162, 305
neurodegenerative disease) proteomics 401, 406, 411
PARP-1 86 (see also neuroproteomics)
442

proxy consent 243, 244, 247 sexual dimorphism 340, 341, 343
Prussian blue staining 367, 372, 373, 376, 379, 380 shaken baby syndrome 254
purinergic receptors 69, 72 signal transduction 171, 173, 174, 318, 398
P2X receptors 69 small interfering RNA (siRNA) 189
P2Y receptors 69 sodium 32–34, 55, 61, 65, 68–70, 98, 135, 195–200,
Purkinje cell 327–333 218, 221, 227, 422
sodium channel 32, 135, 196, 221, 430
randomized controlled trial (RCT) 219, 243,244 Nav1.3 sodium channel 195, 197–199, 201
enrollment criteria 244 sodium/calcium exchangers 55, 67
reactive oxygen species (ROS) 34, 84, 86, 87, 90, spectrin 46, 47, 52, 87, 88, 273, 274, 280
218, 357, 358 sports 3, 24, 253, 254, 263–265, 269, 275,
rehabilitation 97, 202, 217, 219, 220, 310, 328, 340, 401
346 American football 264
remyelination 131, 132, 134, 226, 342, 419, 420, boxing 268
422, 430 ice hockey 263
repeated injuries in vitro 256 return-to-play guidelines 263, 285
repeated TBI 253, 254, 257, 258 (see also repetitive soccer 264
traumatic brain injury) spreading depression 30, 279
repetitive traumatic brain injury, 253 static injury 403
Rho 223, 224, 386 stem cells 136, 217, 225–227, 320, 367, 369, 371,
Rho antagonist 217, 224 372, 379, 381, 387
risk–benefit ratio 244, 246, 248 bone marrow mesenchymal stem cells 367
road traffic accident mortality 5 embryonic stem cells 367, 369, 371
rotational head injury 307 strain-injury 68 (see also stretch)
strains 14, 16, 17, 19, 21, 23, 28, 55
S-100B protein 256, 317, 320 stretch 30–32, 35, 68, 143, 144, 151, 158–164, 253,
S100B 317–322 256, 257, 271, 275, 277, 279, 318
neurotrophic properties of S100B 318 stretch injury 35, 143, 158–162, 164, 253, 256, 271,
neuroprotective effect of S100B 319 277, 279, 318
S-100a protein 282 striatum 191, 343
Schwann cells 132, 225, 226, 228, 282, 284 stroke 31, 89, 97, 103, 136, 185, 188, 217, 248,
second impact syndrome 285, 286 339, 346, 348, 354, 358, 367, 369, 371,
second messengers 72 394, 412
secondary insults 118, 119, 126, 207, 254, 281, 296, subcellular 43–46, 51, 52, 55, 56, 153
321, 403, 414, 415 subdural hematoma (SDH) 27, 66, 67, 118, 120,
secondary phase 14, 21, 23 210, 239, 266, 280, 293–296, 298, 300 (see
secretases 304, 305 also acute subdural hematoma)
a-secretase 304, 305 substance P 97–104, 196
b-secretase 304, 305 superoxide 46, 175, 177
g-secretase 304, 305 surgery 178, 218, 219, 224, 238–241, 344,
seizure threshold 143, 156 354, 394
seizures 64, 145, 299, 300, 341, 347 surgical decompression 217–219, 393, 394,
sex differences 339 396, 398
sex steroids 86, 345 surgical treatment 217, 219, 235, 239
estradiol 342, 346 synaptic circuits 143, 144
estrogen 86, 309, 340–344, 346, 348 synaptic plasticity 32, 69, 148, 149, 151–153, 257,
progesterone 309, 342, 344–346, 348 333, 335 (see also long-term potentiation,
testosterone 86, 342, 344, 348 long-term depression)
443

tachykinin receptors 100, 103 (see also neurokinin transplantation 136, 226–228, 367–369, 377, 379,
receptors) 419–422, 424, 426, 427–430
tensile strain 68, 156, 158, 266–268, 273, 274, 276 cellular transplantation 136, 419
(see also stretch) OEC transplantation 228, 420, 426, 428–430
thalamus 44, 48, 130, 144, 195, 199–201 traumatic axonal injury (TAI) 48, 85, 271, 327
ventroposterior lateral (VPL) nucleus 195, treatment strategies 14, 17, 90, 111, 125, 300, 353
199 TrkA receptor 343
VPL neurons 195, 199–201 (see also tumor necrosis factor (TNF) 84, 89, 177, 223, 359
ventroposterior lateral nucleus) tumor necrosis factor alpha (TNF-a) 127, 223,
therapeutic window 33, 88–90, 218, 227, 246, 381, 225, 229, 359, 360, 410
396 TUNEL 82, 87, 89, 306, 428
therapies 27, 34, 52, 97, 127, 137, 178, 217, 218, two-dimensional polyacrylamide gel
220, 229, 300, 305, 369, 402, 405, 415 electrophoresis (2D-PAGE) 407, 408, 409
time frames 243–245
time window 243, 244, 246–248, 335, 368, 420 voltage gated calcium channels 32, 55
(see also critical time frames) voltage-gated sodium channels 32, 195, 196, 422
tissue deformation 15, 16, 28, 31 Nav1.3 195, 197–199, 201
tissue oxygen response 211
tissue strain 18, 28 weight drop model 155, 156, 254, 255, 257, 405,
tissue tolerances 14, 19, 21 406
tolerance criteria 13, 14, 18, 23, 24 whole-cell patch-clamp recordings 198
transcranial Doppler flowmetry 111 whole-cell patch voltage-clamp recordings 148
This page intentionally left blank

Potrebbero piacerti anche