Sei sulla pagina 1di 438

List of Contributors

A. Arenas, Department of Rehabilitation Medicine, University of Miami School of Medicine, P.O. Box
016960 (D-461), Miami, FL 33101, USA
K. Anderson, Reeve-Irvine Research Center, 1216 GNRF, University of California, Irvine, Irvine, CA
92697-4292, USA
H.W.G. Baker, University of Melbourne Department of Obstetrics and Gynaecology Melbourne IVF
Reproductive Services, Royal Women’s Hospital, Carlton, Vic. 3058, Australia
M.S. Beattie, Department of Neuroscience, laboratory of CNS Repair and Spinal Trauma and Repair
Laboratories, The Ohio State University College of Medicine and Pulic Health, Columbus, OH, USA
L.A. Birder, Departments of Medicine and Pharmacology, University of Pittsburgh School of Medicine,
Pittsburgh, PA 15261, USA
A.F. Brading, Oxford Continence Group, University Department of Pharmacology, Mansfield Road,
Oxford OX1 3QT, UK
J.C. Bresnahan, Department of Neuroscience, Laboratory of CNS Repair and Spinal Trauma and Repair
Laboratories, The Ohio State University College of Medicine and Public Health, Columbus, OH, USA
J.A. Brock, Spinal Injuries Research Centre, Prince of Wales Medical Research Institute, Gate 1, Barker
Street, Randwick, NSW 2031, Australia
A. Brown, Biotherapeutics Research Group, The Spinal Cord Injury Team, Robarts Research Institute and
The Graduate Program in Neuroscience, The University of Western Ontario, P.O. Box 5015, 100 Perth
Drive, London, ON N6A 5K8, Canada
D.J. Brown, Victorian Spinal Cord Service, Austin Health, Heidelberg, Vic., Australia
E.A.L. Chung, St Mark’s Hospital, Northwick Park, Watford Road, Harrow, Middlesex, HA1 3UJ, UK
V.E. Claydon, International Collaboration on Repair Discoveries (ICORD), University of British Co-
lumbia, Vancouver, BC V6 T 1Z4, Canada
H.L. Collins, Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201,
USA
M.D. Craggs, Centre for Spinal Research, Functional Assessment and Restoration, London Spinal Cord
Injuries Unit, Royal National Orthopaedic Hospital NHS Trust, Brockley Hill, Stanmore, Middlesex
HA7 4LP, UK
W.C. de Groat, Departments of Pharmacology and Urology, University of Pittsburgh School of Medicine,
Pittsburgh, PA, USA
G.A. Dekaban, Spinal Cord Injury Team, BioTherapeutics Research Group, Robarts Research Institute,
100 Perth Drive, P.O. Box 5015, London, ON N6A 5K8, Canada
S.E. DiCarlo, Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201,
USA
J.W. Downie, Department of Urology and Pharmacology, Faculty of Medicine, Dalhousie University,
5850 College St., Halifax, NS B3 H 1X5, Canada
S.L. Elliott, Departments of Psychiatry and Urology, University of British Columbia, British Columbia
Center for Sexual Medicine, Echelon-5, 855 West 12th Avenue, Vancouver, Vancouver Sperm Retrieval
Clinic, Vancouver Hospital and G.F. Strong Rehabilitation Centre, Vancouver, BC, Canada

v
vi

G.S. Emch, Department of Neuroscience, Georgetown University Medical Center, TRB EP04, Washing-
ton, DC 20057, USA
A.V. Emmanuel, St Mark’s Hospital, Northwick Park, Watford Road, Harrow, Middlesex, HA1 3UJ, UK
P. Enck, Department of Psychosomatic Medicine, University Hospitals Tuingen, Schaffhausenstr 113,
72072 Tubingen, Germany
F.A. Frizelle, Colorectal Unit, Department of Surgery, Christchurch Hospital and Burwood Spinal Unit,
Christchurch, New Zealand
R.A. Gaunt, Department of Biomedical Engineering and Center for Neuroscience, University of Alberta,
507 HMRC, Edmonton, AB T6G 2S2, Canada
I. Greving, Department of Internal Medicine, Elisabeth Hospital, Gelsenkirchen, Germany
D. Gris, Spinal Cord Injury Team, BioTherapeutics Research Group, Robarts Research Institute, 100
Perth Drive, P.O. Box 5015, London, ON N6A 5K8, Canada
L.A. Havton, Department of Neurology, David Geffen School of Medicine at University of California Los
Angeles, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
S.T. Hill, Victorian Spinal Cord Service, Austin Health, Heidelberg and Melbourne IVF Reproductive
Services, Royal Women’s Hospital, Melbourne, Vic., Australia
T.X. Hoang, Department of Neurology, David Geffen School of Medicine at University of California Los
Angeles, 710 Westwood Plaza, Los Angeles, CA 90095-1769, USA
C.H. Hubscher, Department of Anatomical Sciences and Neurobiology, University of Louisville School of
Medicine, Louisville, KY 40292, USA
J.E. Jacob, Biotherapeutics Research Group, The Spinal Cord Injury Team, Robarts Research Institute
and The Graduate Program in Neuroscience, The University of Western Ontario, P.O. Box 5015, 100
Perth Drive, London, ON N6A 5K8, Canada
R.D. Johnson, Department of Physiological Sciences, College of Veterinary Medicine and the McKnight
Brain Institute, University of Florida, Gainesville, FL 32610-0144, USA
A.K. Karlsson, Spinal Injuries Unit, Sahlgrenska University Hospital, Institute of Clinical Neuroscience,
Sahlgrenska Academy, S 413 45 Goteborg, Sweden
J.R. Keast, Pain Management Research Institute, University of Sydney at Royal North Shore Hospital, St
Leonards, NSW, Australia
S. Klosterhalfen, Institute of Medical Psychology, University Hospitals Dusseldorf, Germany
A. Krassioukov, International Collaboration on Repair Discoveries (ICORD), Division of Physical Med-
icine, School of Rehabilitation and Department of Medicine, University of British Columbia, Vancou-
ver, BC V6 T 1Z4, Canada and Department of Physical Medicine and Rehabilitation, University of
Western Ontario, London, ON, Canada
G.M. Leedy, Division of Social Work, University of Wyoming, Laramie, WY, USA
I.J. Llewellyn-Smith, Cardiovascular Medicine and Centre for Neuroscience, Flinders University, Bedford
Park, SA 5042, Australia
A.C. Lynch, Colorectal Unit, Department of Surgery, Christchurch Hospital and Burwood Spinal Unit,
Christchurch, New Zealand
D.R. Marsh, Department of Anatomy and Cell Biology, Dalhousie University, Halifax, NS, Canada
C.J. Mathias, Neurovascular Medicine Unit, Faculty of Medicine, Imperial College London at St. Mary’s
Hospital, London W2 1NY, UK, Autonomic Unit, National Hospital for Neurology and Neurosurgery,
Queen Square, and Institute of Neurology, University College London, London, UK
E.M. McLachlan, Spinal Injuries Research Centre, Prince of Wales Medical Research Institute, Gate 1,
Barker Street, Randwick, NSW 2031, Australia
Y.S. Nout, Department of Neuroscience, Laboratory of CNS Repair and Spinal Trauma and Repair
Laboratories, The Ohio State University College of Medicine and Public Health, Columbus, OH, USA
vii

P.J. Potter, Regional Spinal Cord Injury Rehabilitation Program, and Physical Medicine and Rehabil-
itation, St. Joseph’s Health Center, The University of Western Ontario, London, ON, Canada
A. Prochazka, Department of Biomedical Engineering and Center for Neuroscience, University of Alberta,
507 HMRC, Edmonton, AB T6G 2S2, Canada
A.G. Rabchevsky, University of Kentucky, Spinal Cord & Brain Injury Research Center and Department
of Physiology, 741 South Limestone Street, B 371 BBSRB, Lexington, KY 40536-0509, USA
T. Ramalingam, Specialist Registrar in Colorectal Surgery, Headquarters Army Medical Directorate,
Former Army Staff College, Camberley, Surrey GU16 4NP, UK
D.W. Rodenbaugh, Department of Molecular and Integrative Physiology, University of Michigan, Ann
Arbor, MI 48109, USA
L.P. Schramm, Departments of Biomedical Engineering and Neuroscience, The Johns Hopkins University
School of Medicine, 606 Traylor Building, 720 Rutland Avenue, Baltimore, MD 21205, USA
S.J. Shefchyk, Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB
R3E 3J7, Canada
M.L. Sipski, Veterans Administration Rehabilitation Research and Development, Center of Excellence in
Functional Recovery and Spinal Cord Injury, Miami, FL 33101, USA
M.A. Vizzard, Departments of Neurology and Anatomy and Neurobiology, University of Vermont College
of Medicine, Burlington, VT 05405, USA
L.C. Weaver, Spinal Cord Injury Laboratory, BioTherapeutics Research Group, Robarts Research In-
stitute, 100 Perth Drive, P.O. Box 5015, London, ON N6A 5K8, Canada
B. Wietek, Department of Radiology, University Hospitals Tubingen, Germany
J.R. Wrathall, Department of Neuroscience, Georgetown University Medical Center, TRB EP04, Wash-
ington, DC 20057, USA
N. Yoshimura, Departments of Pharmacology and Urology, University of Pittsburgh School of Medicine,
Pittsburgh, PA, USA
N.D.T. Zinck, Department of Pharmacology, Faculty of Medicine, Dalhousie University, 5850 College St.,
Halifax, NS B3 H 1X5, Canada
Dedication

‘‘Spinal cord injury is a ferocious assault on the body that leaves havoc in its wake. Paralysis is
certainly part of its legacy, but there are other equally devastating consequences including
autonomic dysfunction: compromised cardiovascular, bowel, bladder, and sexual function.
Treatments and cures for these losses would greatly improve the quality of life for all of us living
with spinal cord injury. I am hopeful that the multi-faceted and collaborative approach to spinal
cord repair evidenced by this book and its contributors means that there will be effective
therapies for autonomic dysfunction in the not too distant future.’’
Christopher Reeve, September 30, 2004

Christopher Reeve sent this endorsement to us only 10 days before his death. His passionate advocacy for
research that would better the lives of all who have suffered spinal cord injury has affected all of us who
work in this field. We dedicate this book to Christopher Reeve and to all, who like him, strive to overcome
the tragedy of spinal cord injury. As Canadian editors, we particularly would like to thank and
acknowledge the efforts of our advocates, Mr. Rick Hansen and Ms. Barbara Turnbull for their tireless
efforts to bring awareness, expertise and funding to the field of spinal cord injury research, in all of its
dimensions. Finally, we dedicate this book to everyone who has sustained a spinal cord injury and lives
courageously, hoping that the efforts of science will bring timely rewards.

Lynne Weaver and Canio Polosa


on behalf of the contributors,
March 1, 2005

ix
Foreword

Autonomic dysfunction after spinal cord injury: the perspective of a person with a spinal cord injury.

‘‘Something’s wrong. I’m hot. No, wait, now I’m cold. But why am I sweating? I never sweat. My
legs won’t stop jumping. Now my hands are in tight fists and my torso is tight as well. Why does
my scalp itch? Oh no, the headache is starting. I’m getting dysreflexic. What is causing it? My
catheter seems okay, I don’t feel any kinks. But why isn’t there any urine in the leg bag? Oh no, is
the catheter clogged up? The headache is getting worse. It feels like a nail is being hammered into
my head. What am I going to do? It’s going to get worse. I know it, I know what’s going to
happen. But I’m all alone. I can’t reach anyone to ask for help. How am I going to fix this? I’m
going to have to drive myself to the doctor. I’m trying to adjust everything, but it’s not working.
My head, it hurts so bad. I can’t think straight. Everything looks a little bit blurry. It’s hard to
breathe now, the spasms are so severe. I feel like I’m going to throw up. Why is this happening to
me? Why? I’m crying now, I’m so afraid. I know what can happen if I don’t stop this. My heart is
beating wildly, my head is hurting more and more. It feels like it’s going to explode. I have to
hurry to the doctor before it’s too late. I could die from this y’’

This is what happened to me a few years ago during one of my worst attacks of autonomic dysreflexia.
Fortunately, I was able to make it to the doctor’s office and convince him of the urgency of the situation. He
changed the catheter, which was indeed clogged with sediment, and within a matter of seconds after the
volume of my bladder was reduced, the grossly obvious symptoms vanished. There are after effects from
such an episode, however. The biggest being extreme exhaustion, which is not trivial when you’re paralyzed.
For a little background on my spinal cord injury, I’m classified as a C5 ASIA grade B and was injured in
a motor vehicle accident in December of 1988. The most prominent source of my autonomic dysreflexia
stems from my bladder. While in the hospital, I tried various methods of managing my bladder and finally
settled on using a suprapubic catheter. This provides me with the greatest level of independence, comfort,
and reliability. I do use an anti-cholinergic medication to reduce spasticity in my bladder. This is a very
important point to stress. If I miss my medication or am delayed in taking it, I experience a continuous state
of mild dysreflexia. That consists of increased spasticity in my body, hot/cold flashes, and an itchy scalp. I
do not develop the severe headache, however, but I also do not know what fluctuations are occurring with
my blood pressure when in that state. These symptoms disappear shortly after I resume the medication.
Just as every spinal cord injury is different, the primary stimulus and pattern of symptoms of autonomic
dysreflexia experienced by each individual are different. The two most common stimuli appear to be
bladder or bowel distension. One anecdotal observation is that the onset and intensity of the symptoms
seem to occur faster and more severely with increasing time post-injury. I have experienced this first-hand
and have been told the same thing by many other people with spinal cord injury. Is this truly common
among the majority of people who develop autonomic dysreflexia? If so, what is the underlying biological
mechanism? And, what long-term damage develops in people with chronic spinal cord injury who repeat-
edly experience episodes of dysreflexia? These are but a few of the problems that need to be addressed in the
scientific and clinical settings.

xi
xii

Aside from acute autonomic dysreflexia episodes, there are many autonomic dysfunctions that present
difficulties that people with any level of spinal cord injury have to deal with on a daily basis. Some of the
most prominent problems include impairments in bladder and bowel control, sexual function, body tem-
perature regulation, cardiovascular control, and metabolism. Any of these dysfunctions can significantly
reduce a person’s quality of life.
Now, in addition to being a quadriplegic, I am also a scientist and, when I first entered the field of spinal
cord injury science in 2000, I observed that research regarding autonomic dysfunctions resulting from
spinal cord injury was not very prevalent. Yet these are problems that everybody with cord injury ex-
periences to some degree. This is a perplexing paradox. In an effort to address this issue, I conducted a
study to determine what areas of functional recovery were most important to people living with spinal cord
injury. Regaining bladder/bowel function and eliminating autonomic dysreflexia was the first or second
highest priority for approximately 40% of quadriplegics and paraplegics and, similarly, regaining sexual
function was the first or second highest priority to 28.3% of quadriplegics and 45.5% of paraplegics
(Anderson, 2004). These results demonstrate that research regarding autonomic dysfunctions is extremely
important. To that end, this book has been written about what is already known and to serve as a platform
for fueling future research.
After all, it is all of these autonomic functions that we take for granted when we have them and that
dominate our lives when we lose them.

References

Anderson, K.D. (2004) Targeting recovery: priorities of the spinal cord injured population. J Neurotrauma, 21: 1371–1383.

Kim Anderson
Contents

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

Dedication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix

Foreword by Kim Anderson . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xvii

Overview: Autonomic dysfunction in spinal cord injury: clinical presentation of symptoms and
signs
A-.K. Karlsson (Goteborg, Sweden) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1

Section I. Anatomical Changes Mediating Autonomic Dysfunction After Cord Injury

1. Effects of spinal cord injury on synaptic inputs to sympathetic preganglionic neurons


I.J. Llewellyn-Smith, L.C. Weaver and J.R. Keast (Bedford Park, SA, London, ON,
Canada and St. Leonards, NSW, Australia) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11

2. Spinal sympathetic interneurons: Their identification and roles after spinal cord injury
L.P. Schramm (Baltimore, MD, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27

3. Which pathways must be spared in the injured human spinal cord to retain cardiovascular
control?
A. Krassioukov (Vancouver, BC, Canada) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39

Section II. Urinary Bladder Dysfunction

4. Disordered control of the urinary bladder after human spinal cord injury: what are the
problems?
P.J. Potter (London, ON, Canada) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51

5. Mechanisms underlying the recovery of lower urinary tract function following spinal cord
injury
W.C. de Groat and N.Yoshimura (Pittsburgh, PA, USA). . . . . . . . . . . . . . . . . . . . . 59

6. Spinal mechanisms contributing to urethral striated sphincter control during continence and
micturition: ‘‘How good things might go bad’’
S.J. Shefchyk (Winnipeg, MB, Canada) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85

xiii
xiv

7. Neurochemical plasticity and the role of neurotrophic factors in bladder reflex pathways
after spinal cord injury
M.A. Vizzard (Burlington, VT, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97

8. Effect of injury severity on lower urinary tract function after experimental spinal
cord injury
J.R. Wrathall and G.S. Emch (Washington, DC, USA) . . . . . . . . . . . . . . . . . . . . . . 117

9. Role of the urothelium in urinary bladder dysfunction following spinal cord injury
L.A. Birder (Pittsburgh, PA, USA). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135

10. Plasticity in the injured spinal cord: can we use it to advantage to reestablish effective
bladder voiding and continence?
N.D.T. Zinck and J.W. Downie (Halifax, NS, Canada) . . . . . . . . . . . . . . . . . . . . . . 147

11. Control of urinary bladder function with devices: successes and failures
R.A. Gaunt and A. Prochazka (Edmonton, AB, Canada). . . . . . . . . . . . . . . . . . . . . 163

12. Novel repair strategies to restore bladder function following cauda equina/ conus medullaris
injuries
T.X. Hoang and L.A. Havton (Los Angeles, CA, USA) . . . . . . . . . . . . . . . . . . . . . . 195

13. Pelvic somato-visceral reflexes after spinal cord injury: measures of functional loss and
partial preservation
M.D. Craggs (Stanmore, UK) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205

Section III. Cardiovascular Dysfunction

14. The clinical problems in cardiovascular control following spinal cord injury: an overview
A. Krassioukov and V.E. Claydon (Vancouver, BC, Canada) . . . . . . . . . . . . . . . . . . 223

15. Orthostatic hypotension and paroxysmal hypertension in humans with high spinal cord
injury
C.J. Mathias (London, UK). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 231

16. Autonomic dysreflexia after spinal cord injury: central mechanisms and strategies for
prevention
L.C. Weaver, D.R. Marsh, D. Gris, A. Brown and G.A. Dekaban (London, ON,
Canada and Halifax, NS, Canada) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 245

17. Segmental organization of spinal reflexes mediating autonomic dysreflexia after spinal cord
injury
A.G. Rabchevsky (Lexington, KY, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265

18. Spinal cord injury alters cardiac electrophysiology and increases the susceptibility to
ventricular arrhythmias
H.L. Collins, D.W. Rodenbaugh and S.E. DiCarlo (Detroit and Ann Arbor,
MI, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 275
xv

19. Adaptations of peripheral vasoconstrictor pathways after spinal cord injury


E.M. McLachlan and J.A. Brock (Randwick, NSW, Australia). . . . . . . . . . . . . . . . . 289

20. Genetic approaches to autonomic dysreflexia


A. Brown and J.E. Jacob (London, ON, Canada) . . . . . . . . . . . . . . . . . . . . . . . . . . 299

Section IV. Bowel Dysfunction

21. Gastrointestinal symptoms related to autonomic dysfunction following spinal cord injury
E.A.L. Chung and A.V. Emmanuel (Harrow, UK) . . . . . . . . . . . . . . . . . . . . . . . . . 317

22. Colorectal motility and defecation after spinal cord injury in humans
A.C. Lynch and F.A. Frizelle (Christchurch, New Zealand) . . . . . . . . . . . . . . . . . . . 335

23. Mechanisms controlling normal defecation and the potential effects of spinal cord injury
A.F. Brading and T. Ramalingam (Oxford and Camberley, UK) . . . . . . . . . . . . . . . 345

24. Alterations in eliminative and sexual reflexes after spinal cord injury: defecatory function
and development of spasticity in pelvic floor musculature
Y.S. Nout, G.M. Leedy, M.S. Beattie and J.C. Bresnahan (Columbus, OH and
Laramie, WY, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 359

25. Upper and lower gastrointestinal motor and sensory dysfunction after human spinal cord
injury
P. Enck, I. Greving, S. Klosterhalfen and B. Wietek (Tubingen, Gelsenkirchen and
Dusseldorf, Germany) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 373

Section V. Sexual Dysfunction

26. Problems of sexual function after spinal cord injury


S.L. Elliott (Vancouver, BC, Canada) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 387

27. Ascending spinal pathways from sexual organs: effects of chronic spinal lesions
C.H. Hubscher (Louisville, KY, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 401

28. Descending pathways modulating the spinal circuitry for ejaculation: effects of chronic
spinal cord injury
R.D. Johnson (Gainesville, FL, USA). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 415

29. Male fertility and sexual function after spinal cord injury
D.J. Brown, S.T. Hill and H.W.G. Baker (Heidelberg, Melbourne and Carlton,
Australia) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 427

30. Female sexual function after spinal cord injury


M.L. Sipski and A. Arenas (Miami, FL, USA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . 441

Subject Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449


Introduction

This book was inspired by a gathering of basic and clinical scientists, and healthcare providers, for a
workshop on autonomic dysfunction after spinal cord injury held in Banff, Alberta, Canada in July, 2003.
The discussions in this meeting highlighted the contrast between the high priority assigned by people with
spinal cord injury to finding a cure for their autonomic dysfunctions, and the limited awareness of these
issues or attention given to them by the scientific and medical community, other than care providers who
interact with cord-injured people regularly. Research is needed to gain greater understanding of the
mechanisms of these problems and to develop treatments and prevention strategies for them. To provide a
foundation for such endeavours, this book contains a compilation of what is known about bladder,
cardiovascular, bowel and sexual dysfunction after spinal cord injury, as it relates to the changes within the
autonomic nervous system control of these functions.
The book is organized into sections that focus on each of the affected visceral functions: urinary bladder,
cardiovascular, gastrointestinal and sexual. The book begins with a description of the time course of
autonomic dysfunctions and their ramifications from the first hours after a spinal cord injury to the more
stable chronic states. The next section contains three chapters that address anatomical findings that may
provide some of the foundation for autonomic dysfunctions in many of the systems. The system-specific
chapters then follow in four sections. Each section begins with a chapter or two defining the clinical
problems experienced by people with cord injury. The following chapters present research, basic and
clinical, that address the autonomic dysfunctions.
We have noted themes that transcend the different sections and can pertain to bladder, bowel, cardio-
vascular and sexual functions. For example, sprouting of axons, including the central processes of sensory
neurons, within the injured spinal cord can be advantageous or detrimental, depending on the amount,
location and potential for new contacts of this sprouting. This may also pertain to changes in the
autonomic ganglia outside the central nervous system. Another theme is loss of coordination and balance
of parasympathetic, sympathetic and somatic systems in the absence of modulatory influences from
supraspinal neuronal systems. Bladder dyssynergia and autonomic dysreflexia, with its episodic hyperten-
sion, have much in common. Lack of coordinated control of pelvic neurons leads to failure of defaecation
and ejaculation. Spinal cord injury affects more than spinal neurons; it impacts on peripheral ganglia and
target tissues such as blood vessels and the wall of the urinary bladder. Growth factors that one would
think should be advantageous to repair of the injured spinal cord, may actually promote development of
circuits that impair rather than support recovered function. Many other themes also thread through this
book.
Finally, we would like to acknowledge several people who have helped us during the preparation of this
book. Ms. Bibi Pettypiece organized the meeting in Banff that started this project and was in commu-
nication with all of the contributing authors to coordinate the details that can so easily become a burden.
Her assistance was invaluable. Ms. Eilis Hamilton applied her considerable skill with graphic presentation
to many of the illustrations in this book, adding to their clarity. The authors who have benefited from her
assistance are very appreciative. Mr. Tom Merriweather and Ms. Maureen Twaig from Progress in Brain
Research, Elsevier, have been encouraging and helpful throughout our effort. Lastly, we realize that we
have not included all of the work that has been done on the subject of autonomic dysfunction after spinal

xvii
xviii

cord injury. Excellent work is being done in addition to that described in these chapters, and we apologize
to those who did not have the opportunity to contribute to this book.

Lynne Weaver
Canio Polosa
Editors
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

Overview

Autonomic dysfunction in spinal cord injury: clinical


presentation of symptoms and signs

Ann-Katrin Karlsson

Spinal Injuries Unit, Sahlgrenska University Hospital, Institute of Clinical Neuroscience, Sahlgrenska Academy, S 413 45
Goteborg, Sweden

Abstract: Spinal cord injury and especially cervical spinal cord injury implies serious disturbances in
autonomic nervous system function. The clinical effects of these disturbances are striking. In the acute
phase, the autonomic imbalance and its effect on cardiovascular, respiratory system and temperature
regulation may be life threatening. Serious complications such as over-hydration with the risk of pulmo-
nary edema or hyponatremia are seen. The cord-injured person suffers from autonomic nervous system
dysfunction also affecting bladder and bowel control, renal and sexual function. Paralytic ileus may cause
vomiting and aspiration, which in turn interferes with respiratory function in those with cervical spinal cord
injury. The cord-injured person is at risk to develop pressure sores from the moment of the accident. Two to
three months post-injury the cord-injured person with a lesion level above the fifth thoracic segment may
develop autonomic dysreflexia, characterised by sympathetically mediated vasoconstriction in muscular,
skin, renal and presumably gastrointestinal vascular beds induced by an afferent peripheral stimulation
below lesion level. The reaction might cause cerebrovascular complications and has effects on metabolism.
Some of the autonomic disturbances are transient and a new balance is reached months post-injury, while
others persist for life.

Spinal cord injury, in a moment, dramatically with spinal cord injury consider the disturbances in
changes the life of the affected person. The loss of autonomic nervous system function even more
control of skeletal muscle, as well as of sensations devastating than the loss of motor and sensory
from below the injury, together with the impair- function.
ment of thermoregulation, urinary bladder and
bowel function produce a profound deterioration
in the quality of life for people after spinal cord The moment of the accident
injury. Tetraplegic subjects rank improvement in
hand function as the most important factor to en- A cervical spinal cord injury may be life threaten-
hance quality of life (Anderson, 2004). However, ing. When the level of lesion is above the third
paraplegic subjects give normal sexual function the cervical segment (C3), the injured person needs
highest priority and, when the first and second immediate assistance of respiration due to the loss
choice was combined, recovery of normal bladder of the supraspinal excitatory drive of the phrenic
and bowel function were given the highest priority motor neurons located at C3–C4. Even when the
in both groups of subjects. This shows that people level of the lesion is below C3, the cord-injured
person may suffer from life-threatening conditions
Corresponding author. Tel.:+46 31 3421000; due to autonomic nervous system dysfunction.
Fax: +46 31 415835; E-mail: ann-katrin.karlsson@neuro.gu.se Cardiac arrest may be one of the causes of death in

DOI: 10.1016/S0079-6123(05)52034-X 1
2

the first few minutes following a cervical spinal if the injured person lies more than 2 h in the same
cord injury due to the disruption of central sym- position on a hard spine board, he/she is at risk of
pathetic control and the concomitant unopposed developing pressure sores. This risk is attributed to
vagal outflow. The incidence of this complication the loss of sensory inputs to the brain from below
is difficult to estimate, although some data indicate the level of the lesion, but this may not be the
a decreasing incidence during the last decades. The whole explanation since unconscious patients
number of patients who reach hospital alive has without spinal cord injury do not seem to be at
increased more than twofold from the 1940s to the as high risk of developing pressure sore as are the
late 1970s. This probably reflects improved skill in cord injury patients, conscious or unconscious.
treatment of a potential spinal cord injury by the Regulation of blood flow in the skin exposed to
first aid team, since unskilled handling of an un- pressure seems to be deranged in the decentralized
stable neck might result in an ascending neurolog- areas of the body, even though subcutaneous
ical level of the lesion, making it life threatening. adipose tissue blood flow during resting conditions
shows no difference when compared to that in
able-bodied people (Karlsson et al., 1997a).
In the emergency room

When arriving at the hospital the person with a Blood pressure control and the syndrome of
cervical spinal cord injury presents with the fol- inappropriate anti-diuretic hormone secretion
lowing symptoms: flaccid paresis, exclusively dia-
phragmatic respiration, low blood pressure and After the extent of the injury is visualized radio-
mostly bradycardia. It is well recognized that spi- graphically, the injured patient is transported ei-
nal cord injury implies inability to empty the blad- ther to an operating theatre, an intensive care unit
der voluntary and, accordingly, an indwelling or to a spinal cord injury unit. Careful monitoring
catheter is always placed in the urinary bladder of blood pressure, heart rate, respiratory rate and
in the emergency room when a spinal cord injury is body temperature begins. A mean arterial blood
suspected. At this stage, new risks appear. Because pressure above 80 mmHg is recommended (Hadley,
the spinal cord injury is often part of a multitrau- 2002), and this level is sometimes maintained
ma the low blood pressure after cervical spinal by intravenous fluid supply and/or by the use of
cord injury may be misinterpreted as consequent pressor agents. Urine output in the cord-injured
to extensive blood loss. A treatment with rapid patient usually is low during the first days post-
infusion of intravenous fluids might lead to pul- injury, probably due to an inappropriate secretion
monary edema. On the other hand, the cord-in- of anti-diuretic hormone. Three to six days post-
jured person may be bleeding in the abdomen and injury urine output reaches 5–6 l/day; the accu-
this may be difficult to diagnose because of the mulated water is excreted. If urine output is strictly
pre-existing low blood pressure, absence of tonic monitored daily, this polyuria may be misinter-
contraction of the abdominal muscles (guarding preted as a sign of inability to concentrate the
reaction) and absence of pain. For this and other urine, and if the loss of water is fully substituted by
reasons, the cord-injured person needs to be care- intravenous fluids, or vasopressin is given, hypo-
fully investigated by computerized tomography natremia may develop. Some years ago a 28-year-
scanning and magnetic resonance imaging. An un- old man sustained a C7 spinal cord injury resulting
stable fracture in the spinal column with the risk of in tetraplegia and developed a serious hypo-
deterioration of the neurological outcome some- natremia with concomitant loss of vision (Karlsson
times results in placing the patient on a hard table, and Krassioukov, 2004). His cervical fracture was
a so-called ‘‘spine board.’’ Then the patient can be stabilized surgically and during the day of surgery
moved from the emergency room to X-ray or to he received 8.6 l of fluid intravenously. When his
the intensive care unit without having to be moved urine output some days later increased to 6 l, it was
from bed to examination tables and back. However, misinterpreted as inability to concentrate the urine
3

and vasopressin was given. Serum sodium concen- such as simultaneous injuries in the chest. How-
tration decreased to 121 mmol/l (at the lowest lev- ever, the 40% loss of vital capacity, due to the
el), and he was treated by restriction of fluid intake paralysis of the inter-costal muscles, is also of im-
and by mineral corticoids. His sodium level nor- portance. The loss of sympathetically mediated
malized slowly. As soon as his water intake ex- bronchial dilatation may add further to the risk of
ceeded 0.7 l/day his serum level of sodium developing respiratory failure. Inhalation of
decreased. As a result of these problems, he had bronchodilators is usually used in the acute phase
a partial loss of vision that was permanent. Urine following spinal cord injury. In spite of this treat-
output must be calculated for a longer period than ment, excessive mucus production and stagnation
the previous day to avoid this risk of overcom- of secretion is seen. The autonomic nervous system
pensation. imbalance might be life threatening during this
condition, since a person with cervical injury who
has a tendency to hypoxia might sustain a severe
Bradycardia
bradycardia or heart arrest during tracheal suc-
tioning. Irritation of the trachea is a heavy stim-
Another effect of the loss of supraspinal control of
ulus of vagal outflow even in able-bodied people
the sympathetic nervous system is the bradycardia
and the reaction in cord-injured patients might be
that is seen sometimes during the first 2–3 weeks
an exaggeration of this reaction due to the loss of
post-injury. An example of this is a man, who at
supraspinal control of the sympathetic nervous
the age of 39 years sustained a spinal cord injury at
system. Pretreatment with anti-cholinergic drugs is
the C5 level, an ASIA C type injury. He was
sometimes needed before tracheal suctioning.
treated in the intensive care unit for 2 days and his
condition was uncomplicated except for brad-
ycardia. At the spinal cord injury unit, he showed
Temperature regulation
signs of a decreased arterial oxygen tension a tra-
cheal suction induced a cardiac arrest. He was
The respiratory problems might lead to pneumo-
transferred to the intensive care unit, where he
nia with high fever. Then the cord-injured patient
stayed for 12 days. He had a prolonged period of
is faced with another effect of autonomic distur-
bradycardia with a mean heart rate of 48 bpm that
bances: that is the inability to lose excess heat by
lasted for 2–3 weeks. The cardiologist was con-
sweating. This inability might be life threatening
sulted and he prescribed a 24 h electrocardiogram
during high fever or in an extremely warm climate.
recording. The recording showed sinus brad-
Several years ago a young man was treated at Sa-
ycardia with a mean rate of 48 bpm and the in-
hlgrenska University Hospital. He had sustained a
stallation of a pacemaker was suggested. However,
C4 spinal cord injury and had an aspiration of
a week later the bradycardia resolved spontane-
fluid into his lungs during the transport to hospi-
ously and during the next 2 weeks the heart rate
tal. He developed pneumonia and his temperature
increased to a mean of 57 bpm. This imbalance in
increased from 41.0 to 42.41 C and, at this high
the acute phase seems to be replaced by a new
temperature, he died of a cardiac arrest.
balance later since, when measuring heart rate
Even though central temperature control is un-
variability in the chronic phase, no difference is
affected by cervical spinal cord injury, we some-
found when comparing cord-injured subjects to
times see a prolonged period of increased
able-bodied subjects (Gao et al., 2002).
temperature in newly injured patient. Careful ex-
amination reveals no signs of infection or inflam-
Respiratory system mation and 3–6 weeks post-injury, the temperature
normalizes. Some people with cervical spinal cord
In the first weeks post-injury there is a risk of res- injury complain of feeling very cold. This is very
piratory failure in the cervical spinal cord injury marked after a shower, when the patient some-
patient. This is sometimes due to obvious reasons times needs a heating quilt or a heater to feel
4

comfortable. The ability to increase temperature needs to be turned every second hour in order to
by shivering is lost below the lesion level and may avoid skin problems. What makes the skin more
explain some of the sensation. However, some vulnerable to pressure during fever is not known.
patients suffer from coldness all the time. Another problem with the skin, seen during the
first months post-injury, is acne vulgaris that
sometimes flares up in the person with cervical in-
Blood pressure and mobilization
jury. This condition is not life threatening and oc-
curs above as well as below lesion level. Whether
When the fracture is stabilized and the neurological
this is due solely to hormonal disturbances elicited
level of lesion is stable, the person with spinal cord
by a stress reaction or to a combination with au-
injury needs to be mobilized. The low blood pres-
tonomic dysfunction is not known.
sure and the inability to increase blood pressure by
vasoconstriction below lesion level make mobiliza-
tion of the person with cervical injury difficult. Urinary system, bladder control
It has to be done gradually by tilting the patient
101/day while blood pressure and neurological sta- The autonomic nervous system dysfunction in-
tus are continuously monitored. When the patient volves the urinary system during the initial post-
tolerates a 401 tilt, they are usually able to sit in a injury stage of ‘‘spinal shock’’ and for the lifetime
wheelchair. Age at injury seems to affect the ability of the person. The dysfunction entails loss of con-
to mobilize the patients, since the elderly usually trol of the urinary outlet and, during spinal shock,
need more time to become mobilized. Whether this loss of sensation from the bladder wall making the
is due to a lower tolerance to low blood pressure or patient at risk of over-distension of the bladder.
to greater decreases in blood pressure during mo- During spinal shock the bladder is atonic irrespec-
bilization is not known. The renin–angiotensin sys- tive of level of lesion. When the stage of spinal
tem plays a role in blood pressure control in shock is past, which may take 3–4 months, people
cervical spinal cord injury (Johnson et al., 1971; with cervical or thoracic lesions develop a spinal
Sutters et al., 1992). This was clearly demonstrated reflex bladder that expels urine under high intra-
when we treated a man in his 40 s with a cervical vesical pressure at a certain amount of bladder
injury who had suffered from renal failure prior to filling, a condition categorized as upper motor
his injury. His mobilization was prolonged and he neuron lesion. People with lower lumbar and/or
suffered from symptoms of low blood pressure for sacral levels of lesion retain an atonic bladder, a
several months after his injury. lower motor neuron lesion. Regulation of bladder
emptying appears rather robust and might be nor-
malized even if the person suffers from some de-
Skin and sensation
gree of paresis and loss of sensation. In a
retrospective chart review of 249 patients with up-
Very few patients are able to lie or sit more than 4 h
per motor neuron lesions who had been treated at
in the same position without getting redness in the
the Spinal Cord Injury Unit in Göteborg,
skin of areas used for body support; this is the first
we found that almost 30% of the individuals
sign of pressure sores. The patient needs to be
recovered normal micturition and most of them
turned every fourth hour even during a skull trac-
had injuries classified as ASIA C and D (Karin
tion period. Four to five caregivers are needed to do
Pettersson, personal communication).
a safe log-roll of the patient. When the patient is
mobilized to a wheel chair sitting position, selection
of cushions is of great importance in order to avoid Urinary system, renal function
pressure sores. The risk of developing pressure sores
persists in the cord-injured person and is increased Renal dysfunction has previously been the major
during severe infections with increased body tem- cause of death following spinal cord injury. The
perature. Under these circumstances the patient mortality rate due to renal failure has decreased
5

from about 40% in the late 1940s (Whiteneck aspiration and the influence on respiration are
et al., 1992) to 3–5% during the last decade (Webb controlled. The pathogenesis of this temporary
et al., 1984). This dramatic change is probably due paralytic ileus is unclear and the time frame for
to improvements in bladder emptying regime as return of bowel activity does not correspond to
well as to the introduction of antibiotics to treat return of reflex activity in the bladder or return of
urinary tract infections. Even better, it seems that tonus and reflexes in the skeleto-muscular system.
renal function has a capacity to improve during Even though a program for bowel emptying is in-
the first years post-injury. In a retrospective chart troduced, the evacuation of stool may be protract-
review, we found that glomerular filtration rate ed. This might be due to the new balance of
was low in those with cervical spinal cord injury in parasympathetic control, with an intact innerva-
the first months after injury. However, at a follow- tion of the ascending colon via the vagus nerve and
up 2–3 years later we found that the glomerular the loss of supraspinal control of the sacral para-
filtration rate had increased, at least in the group sympathetic supply to the colon.
who emptied the bladder by clean intermittent When the bowel program has started the cord-
catheterization (Karin Pettersson, personal com- injured person may face new problems. There is a
munication). risk of developing anal incontinence, and the risk
is highest with lesions in the lower lumbar level
due to a low tonus in the external anal sphincter.
Gastrointestinal system This is perhaps one of the most devastating effects
of the injury to the spinal cord and may be one
The gastrointestinal system is also affected by the important reason why cord-injured people do not
spinal cord injury. Newly injured patients are at return to work after their injury.
increased risk of developing stress related gastric The higher tonus and the uninhibited activity in
ulcers and are regularly offered anti-acid treat- the anal sphincter after cervical and thoracic le-
ment. It might be that the unopposed vagal out- sions might give rise to severe pain that seems to
flow plays a role and increases the risk of ulcer originate from the anal sphincter. The pain is
formation. Other problems from the intestinal sys- made worse by anal fissures and hemorrhoids, and
tem are obvious to the patient soon after injury. this pain, as well as anal incontinence and consti-
The bowel is silent and the voluntary control of pation, might later lead to colostomy. However,
bowel emptying is lost. This paralytic ileus ceases the pain problem is not always resolved even
within 1–2 weeks, but if liquids or solid food is though the rectum and anal region are bypassed.
given prior to this there is a clear risk of a pro-
longed period of paralytic ileus, with the concom-
itant risk of nausea and vomiting. A patient who is Sexual function
placed in skull traction is hard to manage properly
during vomiting and there is a great risk of aspi- Some cord-injured men already in the intensive
ration. Furthermore, a paralytic ileus could give care unit ask about their ability to have an active
rise to a meteoristic abdomen, which might inter- sex life and become fathers. The erectile dysfunc-
fere with respiration by interference with the tion in men after spinal cord injury has different
breathing movement of the diaphragm. characteristics depending on level of lesion, and
Programs for bowel emptying must be intro- mainly follows the pattern of bladder dysfunction.
duced and we choose a rather conservative way of The person with an upper motor neuron lesion
treatment initially: no ingestion of food or drink usually has the capacity for reflex erection by tac-
until the bowel has been emptied. In people with tile stimulation. The person with a lower motor
lesions at the cervical and thoracic level this takes neuron lesion has loss of all erectile function. The
about 3–6 days, but in the low lumbar lesion level capacity of psychogenic erection is lost in all cord-
the emptying might be further delayed by several injured men with a complete lesion. Retrograde
days. By this regime, the risk of vomiting and ejaculation is the rule when there is an ejaculation
6

at all. Today, we can offer drugs and different sympathetic neurons may be asymptomatic and
stimulations in order to improve erectile function much more frequent than previously known. Con-
and to produce an anterograde ejaculation. Vibra- tinuous measurement of noradrenalin in blood
tion and electro-stimulation need careful monitor- samples collected every 30 min in 24 h revealed
ing of blood pressure since these methods readily several peaks even in subjects who were asympto-
evoke autonomic dysreflexia. By the means of vi- matic (Karlsson et al., 1997b).
bration and electro-stimulation in combination The reaction is triggered by distension of hollow
with insemination or in vitro fertilization, men af- organs below lesion level such as the urinary blad-
ter spinal cord injury can become fathers. At our der, the gall bladder, the renal pelvis and ureters or
Spinal Cord Injury Unit, with 35–50 newly injured the gastrointestinal system. A bone fracture below
patients admitted every year, we have about 75 lesion level may also give rise to the reaction.
children who have cord-injured fathers (Agneta When the triggering factor is withdrawn, the blood
Siösteen, personal communication). pressure returns to normal. However, if the cord-
Sexual function is impaired also in women. The injured person or the treating staff is unfamiliar
female analogue to erectile dysfunction, that is, with the reaction, there is a serious risk of com-
loss of lubrication, needs treatment. Fertility is plications as intracranial hemorrhage or cerebral
unaffected, but the autonomic disturbances make infarction. In our clinical practice there have been
the woman with spinal cord injury at increased two cases in the last 2 years. The first was a woman
risk of urinary incontinence, urinary tract infection in her 50s who sustained a C4–C5 level spinal cord
and pressure sores during child bearing. They are injury and tetraplegia a few years ago. She was
also at risk of developing severe autonomic discharged to home and some months later be-
dysreflexia during labor. came severely constipated and developed an auto-
nomic dysreflexia reaction. She was treated at a
local hospital where there was inadequate knowl-
Autonomic dysreflexia edge of autonomic dysreflexia. The blood pressure
stayed very high and the patient developed a cer-
Two to four months post-injury the person with ebral infarction. Another case was seen recently. A
cervical spinal cord injury may suddenly experi- person with cervical spinal cord injury who had
ence a flushing in the face and complain of severe been injured for more than 30 years was treated
headache. The blood pressure increases from 100/60 for a serious infection. He developed an abscess in
to 240/120 mmHg. When looking for triggering the abdomen and suffered from a prolonged ep-
factors, the staff might find an obstruction of the isode of dysreflexia. During this period we meas-
urinary outlet. When urine passes again the blood ured his cerebral blood flow and found signs of
pressure returns to normal. This reaction, the so- decreased perfusion in parts of the brain, indicat-
called autonomic dysreflexia reaction, is seen in ing vasoconstriction. We speculated that this vaso-
cord-injured people with a lesion level above T6. constriction might have been an autoregulatory
The clinical reaction is not an all-or-none reaction response of the cerebral vessels in the presence of
but graded; in mild cases the person just feels a greatly increased systemic arterial pressure.
small chill. Investigations have shown that the re- The autonomic dysreflexia reaction has other
action is caused by a severe vasoconstriction below side effects and can influence metabolism. Region-
lesion level in skin, muscular (Karlsson et al., al investigation of adipose tissue metabolism
1998) and renal vascular beds (Gao et al., 2002). above and below lesion level showed an increase
Presumably also the splanchnic/gastrointestinal in glycerol release — activation of lipolysis —
vascular bed is involved. The reaction is mediat- during induced dysreflexia, that is, during sympa-
ed by the sympathetic nervous system as shown by thetic activation below lesion level (Karlsson et al.,
a profound increase in noradrenalin spillover be- 1998). Whether this is of importance for the insulin
low lesion level (Karlsson et al., 1998; Gao et al., resistance sometimes seen after spinal cord injury
2002). The peripheral afferent stimulation of the (Karlsson, 1999) is not known.
7

Another associated risk with the autonomic Long-term effects


dysreflexia reaction is the vasoconstriction in the
renal vascular bed (Gao et al., 2002). This sympa- Gradually after spinal cord injury some risks seem
thetic activation below lesion level might contrib- to diminish, others persist and new ones develop.
ute to the development of renal failure. Previously, By 15–20 years post-injury new complications
many tetraplegic patients used bladder tapping to from the circulatory system are sometimes seen.
condom drainage as a method of bladder empty- Following large meals the cord-injured person with
ing. Bladder tapping is known to induce auto- a cervical lesion is sometimes unable to sit upright
nomic dysreflexia every time it is performed. The due to rapid fall in blood pressure. Whether this is
introduction of intermittent catheterization may merely an effect of duration of injury or a marker
thus have the advantage of maintaining renal of concomitant disease is not known. The muscle
function both by the intermittent total emptying of and skin vascular beds are unable to vasoconstrict
the bladder, with markedly lower risk of urinary during the post-prandial increase in blood flow in
tract infections, and by the absence of triggering the intestinal vascular bed, but this limitation is
factors for renal vasoconstriction. present since the onset of injury and therefore,
does not solely explain the new complication.
People with cervical spinal cord injury sometimes
experience low urine output during the daytime
Visceral sensation
when they are in sitting position. During the night
there is a huge urine flow, making the patient at risk
The loss of sensation includes not only skin and
of incontinence or over-distension of the bladder.
joints but also loss of visceral sensation. This is of
importance during pathological processes in the
internal organs, and also seems to include loss of
Time course of autonomic nervous system changes
normal sensation of hunger and satiety, a condi-
— transitional stage?
tion that might contribute to disturbances in body
weight sometimes seen in tetraplegic people. A
Bradycardia lasts for few weeks. Paralytic ileus
weight gain implies the risk of diabetes, impaired
lasts for 1–2 weeks. Signs of the inappropriate an-
glucose control and increased levels of serum lip-
ti-diuretic hormone secretion syndrome appear
ids. A higher risk in cord-injured than in able-
during the first week. The risk of respiratory fail-
bodied subjects has been reported (Duckworth
ure is great during the first weeks after injury. The
et al., 1983; Bauman and Spungen, 2001), although
bladder is flaccid for 3–4 months. Body tempera-
a study that controlled for inheritance by compar-
ture is increased for 3–4 weeks. The risk of deep
ing cord-injured subjects to their siblings showed
vein thrombosis is increased for weeks or months.
no difference in glucose tolerance evaluated by the
There are many signs of a transitional stage in
hyper-insulinemic, normo-glycemic clamp method
autonomic nervous system dysfunction following
(Karlsson, 1999).
spinal cord injury. However, as described above,
the time frame is highly divergent. Some of the
changes are attributed to the spinal shock, or more
Thrombo-emboli precisely to the recovery from spinal shock. How-
ever, our knowledge of the pathology behind spi-
During the first months post-injury the cord-in- nal shock is sparse, and it seems that the return of
jured person is at increased risk of developing deep reflex activity in the spinal cord follows a different
vein thrombosis. Treatment with anti-coagulants time course. Muscular tone and tendon reflexes
is given for 3–6 months. The role of the autonomic appear within 6–8 weeks, whereas reflexes for
nervous system in this increased risk is unclear. bladder emptying may return much later, up to
Later in life, cord-injured people appear not to 3–4 months post-injury. The imbalance in heart
have increased risk of deep vein thrombosis. rate regulation has a time course of its own. We
8

have some knowledge from animal and human Karlsson, A.K. (1999) Insulin resistance and sympathetic func-
research regarding the plasticity of spinal neural tion in high spinal cord injury. Spinal Cord, 37: 494–500.
Krassioukov, A.V., Bunge, R.P., Pucket, W.R. and Bygrave,
circuits after injury (Krassioukov and Weaver,
M.A. (1999) The changes in human spinal sympathetic pre-
1995; Krassioukov et al., 1999) but the clinical ganglionic neurons after spinal cord injury. Spinal Cord, 37:
implications of these findings are unclear. 6–13.
After spinal cord injury, new balances are cre- Karlsson, A.K., Elam, M., Friberg, P., Biering-Sorensen, F.,
ated in the autonomic nervous system and we need Sullivan, L. and Lonnroth, P. (1997a) Regulation of lipolysis
to create more knowledge about how this is done. by the sympathetic nervous system: a microdialysis study in
normal and spinal cord-injured subjects. Metabolism, 46:
We also have to develop more understanding of 388–394.
the overall disturbances in the autonomic nervous Karlsson, A.K., Elam, M., Friberg, P., Sullivan, L., Attvall, S.
system of cord-injured people since these distur- and Lonnroth, P. (1997b) Peripheral afferent stimulation of
bances have a profound impact on their life. decentralized sympathetic neurons activates lipolysis in spinal
cord-injured subjects. Metabolism, 46: 1465–1469.
Karlsson, A.K., Friberg, P., Lonnroth, P., Sullivan, L. and
Elam, M. (1998) Regional sympathetic function in high spi-
References nal cord injury during mental stress and autonomic dysre-
flexia. Brain, 121: 1711–1719.
Anderson, K.D. (2004) Targeting recovery: Priorities of the spi- Karlsson, A.K. and Krassioukov, A.V. (2004) Hyponatremia-
nal cord-injured population. J. Neurotrauma, 21: 1371–1383. induced transient visual disturbances in acute spinal cord in-
Bauman, W.A. and Spungen, A.M. (2001) Carbohydrate and jury. Spinal Cord, 42(3): 204–207.
lipid metabolism in chronic spinal cord injury. J. Spinal Cord Krassioukov, A.V. and Weaver, L.C. (1995) Reflex and mor-
Med., 24: 266–277. phological changes in spinal preganglionic neurons after cord
Duckworth, W.C., Jallepalli, P. and Solomon, S.S. (1983) Glu- injury in rats. Clin. Exp. Hypertens., 17: 361–373.
cose intolerance in spinal cord injury. Arch. Phys. Med. Re- Sutters, M., Wakefield, C., O’Neil, K., Appleyard, M., Frankel,
habil., 64: 107–110. H., Mathias, C.J. and Peart, W.S. (1992) The cardiovascular,
Gao, S.A., Ambring, A., Lambert, G. and Karlsson, A.K. endocrine and renal response of tetraplegic and paraplegic
(2002) Autonomic control of the heart and renal vascular bed subjects to dietary sodium restriction. J. Physiol., 457:
during autonomic dysreflexia in high spinal cord injury. Clin. 515–523.
Auton. Res., 12: 457–464. Webb, D.R., Fitzpatrick, J.M. and O’Flynn, J.D. (1984) A 15-
Hadley, M.N., Walters, B.C., Grabb, P.A., Oyesiku, N.M., year follow-up of 406 consecutive spinal cord injuries. Br. J.
Przbylski, G.J., Resnick, D.K. and Ryken, T.C. (2002) Man- Urol., 56: 614–617.
agement of acute spinal cord injuries in an intensive care unit Whiteneck, G.G., Charlifue, S.W., Frankel, H.L., Fraser, M.H.,
or other monitored settings. Neurosurgery, 50: S51–S57. Gardner, B.P., Gerhart, K.A., Krishnan, K.R., Menter, R.R.,
Johnson, R.H., Park, D.M. and Frankel, H.L. (1971) Ortho- Nuseibeh, I. and Short, D.J. (1992) Mortality, morbidity, and
static hypotension and the renin-angiotensin system in par- psychosocial outcomes of persons spinal cord injured more
aplegia. Paraplegia, 9: 146–152. than 20 years ago. Paraplegia, 30: 617–630.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 1

Effects of spinal cord injury on synaptic inputs to


sympathetic preganglionic neurons

Ida J. Llewellyn-Smith1,, Lynne C. Weaver2 and Janet R. Keast3

1
Cardiovascular Medicine and Centre for Neuroscience, Flinders University, Bedford Park, SA 5042, Australia
2
Spinal Cord Injury Laboratory, BioTherapeutics Research Group, Robarts Research Institute, London, ON, Canada
3
Pain Management Research Institute, University of Sydney at Royal North Shore Hospital, St Leonards,
NSW, Australia

Abstract: Spinal cord injuries often lead to disorders in the control of autonomic function, including
problems with blood pressure regulation, voiding, defecation and reproduction. The root cause of all these
problems is the destruction of brain pathways that control spinal autonomic neurons lying caudal to the
lesion. Changes induced by spinal cord injuries have been most extensively studied in sympathetic pre-
ganglionic neurons, cholinergic autonomic neurons with cell bodies in the lateral horn of thoracic and
upper lumbar spinal cord that are the sources of sympathetic outflow. After an injury, sympathetic pre-
ganglionic neurons in mid-thoracic cord show plastic changes in their morphology. There is also extensive
loss of synaptic input from the brain, leaving these neurons profoundly denervated in the acute phase of
injury. Our recent studies on sympathetic preganglionic neurons in lower thoracic and upper lumbar cord
that regulate the pelvic viscera suggest that these neurons are not so severely affected by spinal cord injury.
Spinal interneurons appear to contribute most of the synaptic input to these neurons so that injury does not
result in extensive denervation. Since intraspinal circuitry remains intact after injury, drug treatments
targeting these neurons should help to normalize sympathetically mediated pelvic visceral reflexes. Fur-
thermore, sympathetic pelvic visceral control may be more easily restored after an injury because it is less
dependent on the re-establishment of direct synaptic input from regrowing brain axons.

Every year, tens of thousands of people worldwide and severity of these problems is dependent
suffer a spinal cord injury, often with devastating on the level and completeness of the injury. Two
consequences. Injured people can lose mobility cardiovascular consequences of spinal cord injury
because they become partially or totally paralyzed are resting or postural hypotension and autonomic
and experience persistent pain and spasticity. As dysreflexia, a condition in which strokes or death
this volume documents, their quality of life can can occur when noxious or innocuous sensory
also be significantly damaged by injury-induced stimuli entering the cord below the injury reflexly
disorders in the control of autonomic function, induce episodes of hypertension. Spinal cord
including problems with blood pressure regulation, injury can also produce a variety of difficulties
voiding, defecation and reproduction. The presence that impair the voiding of urine, including
detrusor hyperreflexia, detrusor sphincter dyssy-
Corresponding author. Tel.: +61-8-8204-4456; nergia and detrusor areflexia. Fecal incontinence
Fax: +61-8-8204-5268;
and constipation are also outcomes of spinal cord
E-mail: Ida.Llewellyn-Smith@flinders.edu.au injury; and inability to achieve psychogenic and

DOI: 10.1016/S0079-6123(05)52001-6 11
12

reflexogenic erections and ejaculatory dysfunction dorsolateral funiculus in the white matter; sympa-
in men and failure of vaginal lubrication in women thetic preganglionic neurons in the dorsolateral
have a significant impact on sexual life after injury. funiculus are more common in the rostral than the
The root cause of all these autonomic sequelae caudal thoracic cord (Strack et al., 1988). A small
of spinal cord injury is the disruption of the brain proportion of sympathetic preganglionic neurons
pathways that control spinal autonomic neurons. are associated with the bundles of dendrites that
Subsequently, when axons conveying supraspinal course mediolaterally between the intermediolateral
drive are dying or dead, neuronal circuits caudal to cell column and central canal. These sympathetic
the injury are able to reorganize. Primary afferent preganglionic neurons have spindle-shaped cell
neurons sprout (Christensen and Hulsebosch, bodies and comprise the intercalated nucleus. The
1997; Krenz and Weaver, 1998b; Krenz et al., somata and dendrites of the sympathetic pregangl-
1999; Wong et al., 2000; Weaver et al., 2001; On- ionic neurons in the intercalated nucleus are ori-
darza et al., 2003) and spinal autonomic interneu- ented parallel to the dendritic bundles with which
rons can become key regulators of some of the they are associated. A final concentration of sym-
information that is conveyed from the spinal cord pathetic preganglionic cell bodies, which are usually
to the periphery (see Schramm, this volume). fusiform in shape, occurs in the central autonomic
Changes also occur in the synaptic circuitry con- area dorsal to the central canal. Somata occupying
trolling sympathetic preganglionic neurons that this position are most frequently encountered at the
have axons providing central drive to all sympa- caudal end of the sympathetic preganglionic neuron
thetic postganglionic neuron in paravertebral and distribution, i.e., in the lowest thoracic and upper
prevertebral ganglia and to chromaffin cells in the lumbar segments.
adrenal medulla.

Morphological changes after spinal cord injury


Location and morphology of sympathetic
preganglionic neurons Spinal cord injury can evoke significant changes in
sympathetic preganglionic neurons, as well as de-
Sympathetic preganglionic neurons are small- to priving them of input from the brain. While neu-
medium-sized cholinergic neurons and their cell rons at the site of a cord injury can be killed,
bodies are located in the thoracic and upper lumbar sympathetic preganglionic neurons that lie distant
spinal cord in four distinct subnuclei within the from the lesion site may also be affected, at least in
lateral horn (Cabot, 1990). The majority of sym- the acute phase of injury. Within 3 days of a com-
pathetic preganglionic somata occur in the inter- plete spinal cord transection at thoracic segment 4/5,
mediolateral cell column, which lies at the border the dendrites of sympathoadrenal preganglionic
between the grey and white matter of the spinal neurons in the intermediolateral cell column of
cord. In the intermediolateral cell column, the cell mid-thoracic cord have retracted to about one-
bodies of sympathetic preganglionic neurons are third of their original length and the diameter of
spindle-shaped or fusiform and occur in groups or their cell bodies has decreased to about 60% of
‘‘nests’’ that are spaced at short intervals along the that in intact cord (Llewellyn-Smith and Weaver,
grey–white boundary. Most of the dendrites of the 2001). This reduction in soma size and dendrite
neurons in the intermediolateral cell column run length is less pronounced 7 days after transection;
rostrally or caudally and can be hundreds of and, by 14 days post-operatively, the sympathetic
micrometers long. In addition, some sympathetic preganglionic neurons and their dendrites are not
preganglionic neurons in the intermediolateral significantly different in size from those in intact
cell column have dendrites that are oriented med- cord (Krassioukov and Weaver, 1996; Krenz and
iolaterally, traveling either into the dorsolateral Weaver, 1998a). The shrinkage and regrowth of
funiculus or toward the central canal. Other sym- sympathetic preganglionic somata and dendrites
pathetic preganglionic somata are situated in the correlate with the degeneration and clearance from
13

the intermediolateral cell column of axons de- the intermediolateral cell column, around it in
tached from their cell bodies by the transection. spinal cord laminae V and VII and in lamina X
Many degenerating profiles of severed axons can dorsal to the central canal (Cabot et al., 1994; Joshi
be seen ultrastructurally at 3 days after injury, but et al., 1995; Clarke et al., 1998; Cano et al., 2001;
virtually none are present at 14 days (Weaver et al., Deuchars et al., 2001; Tang et al., 2004). The axons
1997). Although new synapses form on sympatho- of presympathetic supraspinal and intraspinal neu-
adrenal preganglionic neurons after a spinal cord rons contain a diverse array of neurotransmitters,
injury (Weaver et al., 1997), reinnervation by ax- many of which have been shown to have direct
ons of spinal neurons is unlikely to be the spur for effects on sympathetic preganglionic neurons.
regrowth of dendrites. Even at 14 days after a Neurons in the brain are probably the exclusive
complete transection, sympathetic preganglionic source of monoamine innervation, whereas both
neurons in the mid-thoracic cord are profoundly supraspinal and intraspinal neurons contribute ax-
denervated. The density of synapses on their cell ons containing amino acids and neuropeptides.
bodies has been cut to half of that in intact cord
and their axodendritic input is reduced by 70%
(Llewellyn-Smith and Weaver, 2001). Whether or Amino acids
not sympathetic preganglionic neurons continue to
be reinnervated after 2 weeks of injury has not Glutamate, g-aminobutyric acid (GABA) and
been studied ultrastructurally. However, anastomo- glycine, all produce fast synaptic responses in sym-
sing networks of axons immunoreactive for growth- pathetic preganglionic neurons (e.g., Mo and Dun,
associated protein 43, a marker for developing and 1987a, b; Inokuchi et al., 1992a, b; Krupp and Feltz,
regenerating axons, are present at least as long as 6 1995; Krupp et al., 1997) and are considered the
weeks after spinal cord injury (Cassam et al., 1999). main fast-acting transmitters regulating their activ-
Continuing reorganization of synaptic circuitry ity (Dampney, 1994). Axons immunoreactive for
controlling the activity of sympathetic preganglion- these amino acids synapse on sympathetic pregangl-
ic neurons may underlie the increasing severity of ionic neurons (Bacon and Smith, 1988; Bogan et al.,
attacks of autonomic dysreflexia (Maiorov et al., 1989; Cabot et al., 1992) and quantitative ultra-
1997a, b; Marsh and Weaver, 2004). structural studies have demonstrated that synaptic
vesicles containing at least one type of amino acid
are present in virtually all of the axons that provide
Innervation of sympathetic preganglionic neurons in input to these neurons (Llewellyn-Smith et al., 1992,
intact and injured cord 1995b, 1998). Brainstem neurons innervate sympa-
thetic preganglionic neurons monosynaptically
In intact cord, sympathetic preganglionic neurons (Zagon and Smith, 1993; Deuchars et al., 1995,
are innervated by both supraspinal and intraspinal 1997); and these spinally projecting neurons contain
neurons. Virus tracing studies have been particu- markers for glutamate and GABA axons, including
larly useful for revealing the locations of presym- phosphate activated glutaminase, vesicular gluta-
pathetic neurons, i.e., those that are directly mate transporter 2 and glutamic acid decarboxylase
antecedent to sympathetic preganglionic neurons (Minson et al., 1991; Stornetta et al., 2002, 2004).
and are likely to be involved in regulating their Furthermore, immunoreactivity for glutamate or
activity. Supraspinal inputs to sympathetic pre- GABA occurs in boutons in the intermediolateral
ganglionic neurons come from five main brain re- cell column that have been anterogradely labeled
gions, including the rostral ventrolateral medulla, from the medulla (Llewellyn-Smith et al., 1995b).
the rostral ventromedial medulla, the caudal raphe Although neurons in the brain provide many of the
nuclei, the A5 region and the paraventricular nu- glutamate- and GABA-immunoreactive axons in
cleus of the hypothalamus (Strack et al., 1989; the intermediolateral cell column, spinal cord injury
Sved et al., 2001). Spinal neurons that project to does not deprive sympathetic preganglionic neurons
sympathetic preganglionic neurons occur within of amino acid-containing inputs. Ultrastructural
14

studies at times when severed supraspinal axons are immunoreactive for dopamine b-hydroxylase
degenerating or have just been removed from below (Cassam et al., 1997), the enzyme that produces
a lesion show that glutamate- and GABA-immuno- noradrenaline from dopamine; and we have de-
reactive synaptic contacts persist on sympathetic scribed axons caudal to a 2-week transection that
preganglionic neurons caudal to either 3- or 7-day contain tyrosine hydroxylase and form synapses
complete spinal cord transections (Llewellyn-Smith in the intermediolateral cell column (Fig. 2B;
et al., 1997; Llewellyn-Smith and Weaver, 2001). Llewellyn-Smith et al., 1995a). Hence, the cat-
Hence, intraspinal neurons as well as supraspinal echolamine enzyme-immunoreactive fibers present
neurons provide amino acid-containing inputs to at 2 weeks may arise from these neurons. Some
sympathetic preganglionic neurons. immunoreactivity may also be present in the non-
terminal portions of severed axons since, at 2
weeks, degenerating terminals cannot be found in
Monoamines the intermediolateral cell column at the ultrastruc-
tural level (Weaver et al., 1997). Determining
Adrenaline and noradrenaline evoke both whether the spinal neurons that express catechola-
excitatory and inhibitory responses in sympathet- mine enzymes at 2 weeks after transection synthe-
ic preganglionic neurons (Coote et al., 1981; size dopamine, adrenaline or noradrenaline will
Kadzielawa, 1983; Ma and Dun, 1985a; Miyazaki require different experimental strategies, such as
et al., 1989; Lewis and Coote, 1990a). At the light fluorescence histochemistry, or multiple-label
microscope level, sympathetic preganglionic neu- immunofluorescence for investigating coexistence
rons at all levels of intact cord are surrounded by of relevant synthetic enzymes or amine transport-
networks of nerve fibers immunoreactive for en- ers. A more detailed anatomical analysis of
zymes of catecholamine synthesis, such as tyrosine 11-week transected cord will also be needed to as-
hydroxylase (Fig. 1A) and phenylethanolamine certain whether there are any enzyme-immunore-
N-methyltransferase (Fig. 1B). In intact cord, phe- active fibers present at the chronic stage of injury.
nylethanolamine N-methyltransferase-immunore- Pharmacological and physiological studies indi-
active axons have been confirmed to synapse on cate that, in general, serotonin (5-hydroxytrypta-
sympathetic preganglionic neurons at the electron mine (5-HT)) has a sympathoexcitatory action on
microscope level (Milner et al., 1988; Bernstein- sympathetic nerve activity and directly on sympa-
Goral and Bohn, 1989) and we have found thetic preganglionic neurons (e.g., Ma and Dun,
that sympathetic preganglionic neurons receive 1986; Yusof and Coote, 1988; Lewis and Coote,
synapses from axons with immunoreactivity for 1990b; Pickering et al., 1994). Serotonergic axons,
tyrosine hydroxylase (Fig. 2A). All of this cat- marked by either immunoreactivity for 5-HT or
echolamine input to sympathetic preganglionic the serotonin transporter, also form a dense plexus
neurons probably originates in the brain (see be- of axons around sympathetic preganglionic neu-
low). Major sources are the C1 adrenergic neurons rons (Figs. 3A and C) and synapses by 5-HT-
of the rostral ventrolateral medulla and the nor- immunoreactive axons have been demonstrated on
adrenergic neurons of the A5 group (Jansen et al., sympathetic preganglionic neurons that project to
1995). The supraspinal origin of catecholamine in- the superior cervical ganglion and adrenal medulla
put is supported by the disappearance of immuno- (Bacon and Smith, 1988; Vera et al., 1990; Jensen
reactivity for catecholamine-synthesizing enzymes et al., 1995). Retrograde and viral tracing studies
caudal to a complete spinal cord transection. indicate that the serotonergic axons surrounding
Staining for tyrosine hydroxylase and phenyl- sympathetic preganglionic neurons arise from rap-
ethanolamine N-methyltransferase is substantially he neurons, mainly those in the medullary nuclei
reduced at 2 weeks post-operatively (Figs. 1C and (Loewy and McKellar, 1981; Bowker et al., 1982;
D) and is absent by 11 weeks (Figs. 1E and F). Jansen et al., 1995). Very rare 5-HT-immunoreac-
Interestingly, after spinal cord injury, some neurons tive neurons have been detected in the spinal cord
in the intermediate grey of the spinal cord become (Newton et al., 1986). However, these are unlikely
15

Fig. 1. Axons containing immunoreactivity for the catecholamine synthesizing enzymes, tyrosine hydroxylase (TH) and phenyl-
ethanolamine N-methyltransferase (PNMT), disappear from the intermediolateral cell column caudal to a complete spinal cord
transection (TX). (A, B) Intact cord. (C, D) 2-week transected cord. (E, F) 11-week transected cord. (A, C, E) Stained for tyrosine
hydroxylase and (B, D, F) stained for phenylethanolamine N-methyltransferase (PNMT). Transections were located in caudal thoracic
segment 4/rostral thoracic segment 5. All micrographs show the intermediolateral cell column in thoracic segment 6. Bars, 100 mm.

to nnervate sympathetic preganglionic neurons be- Neuropeptides


cause 5-HT-immunoreactive and serotonin trans-
porter-immunoreactive axons are absent from Axons containing a substantial array of neuro-
autonomic areas caudal to a 2-week transection peptides have been demonstrated by light micros-
(Figs. 3B and D). Hence, severed serotonergic ax- copy in regions of the cord where the cell bodies
ons disappear from the cord before axons that are and dendrites of sympathetic preganglionic neu-
immunoreactive for catecholamine enzymes. rons are located (Table 1) and a number of these
16

Fig. 2. Axons immunoreactive for tyrosine hydroxylase form synapses in intact and transected cord. (A) In the intermediolateral cell
column of intact cord, a tyrosine hydroxylase-immunoreactive varicosity (TH) forms a synapse (arrowhead) on a dendrite that
contains cholera toxin B subunit (CTB) retrogradely transported from the adrenal medulla. An adjacent non-immunoreactive var-
icosity (asterisk) synapses (arrowheads) on the same dendrite. (B) In the intermediolateral cell column of thoracic segment 8, a tyrosine
hydroxylase-immunoreactive varicosity (TH) forms a synapse (arrowheads) on a dendrite 2 weeks after a complete spinal cord
transection (TX) in caudal thoracic segment 4/rostral thoracic segment 5. An adjacent non-immunoreactive varicosity (asterisk)
synapses (arrowhead) on the same dendrite. Bars, 500 nm.

Fig. 3. Axons containing the serotonergic markers, 5-HT or the serotonin transporter, disappear from the intermediolateral cell
column caudal to a complete spinal cord transection (TX). (A, C) Intact cord. (B, D) 2-week transected cord. (A, B) Stained for 5-HT
and (C, D) stained for the serotonin transporter (SERT). Transections were located in caudal thoracic segment 4/rostral thoracic
segment 5. All micrographs show the intermediolateral cell column in thoracic segment 6. Bar in (D) applies to (A—D), 100 mm.
17

Table 1. Neuropeptide immunoreactivity identified in axons in autonomic regions of the thoracic and upper lumbar spinal cord

Angiotensin II Neurotensin
Avian pancreatic polypeptide (APP) Nociceptin
Calcitonin gene-related peptide (CGRP) Orexina
Cholecystokinin (CCK) Oxytocin
Cocaine and amphetamine regulated transcript (CART)a Pituitary adenylate cyclase activating polypeptide (PACAP)a
Corticotropin releasing factor (CRF) Somatostatin
Enkephalina Substance Pa
Galanin Thyrotropin releasing hormone
Neuropeptide Y (NPY)a Vasoactive intestinal peptide (VIP)
Neurophysin Vasopressin
a
Axons containing these neuropeptides have been shown to form synapses either in the intermediolateral cell column or on identified
sympathetic preganglionic neurons.

have been shown to synapse on sympathetic pre- acetyltransferase-immunoreactive (i.e., choliner-


ganglionic neurons (Bacon and Smith, 1988; Vera gic) neurons in the intermediolateral cell column
et al., 1990; Llewellyn-Smith et al., 1991; Pilowsky (Llewellyn-Smith and Weaver, 2004). Hence, some
et al., 1992). When applied to sympathetic pre- of the axons containing these neuropeptides come
ganglionic neurons, many of these neuropeptides from spinal interneurons to innervate the choli-
evoke synaptic responses (e.g., Ma and Dun, nergic sympathetic preganglionic neurons. The
1985b; Dun and Mo, 1988; Kolaj et al., 1997; source of the neuropeptide Y-immunoreactive ax-
Lai et al., 1997; Antunes et al., 2001; van den Top ons that persist in the lateral horn may be the
et al., 2003). Some of the neuropeptide-immuno- neurons in laminae V and VII in intact cord that
reactive axons supplying sympathetic preganglion- express neuropeptide Y mRNA (Minson et al.,
ic neurons arise exclusively from neurons in the 2001). The intraspinal enkephalin innervation of
brain, including those containing oxytocin, vaso- autonomic regions may arise from small enkepha-
pressin and orexin. However, other neuropeptides, lin-immunoreactive neurons in lamina X that
such as substance P, enkephalin and neuropeptide we have detected in sections from intact cord
Y, occur in autonomic areas of intact and fixed with high concentrations of glutaraldehyde
transected cord (Figs. 4–6), suggesting that both (Llewellyn-Smith and Keast, unpublished obser-
supraspinal and intraspinal neurons supply the vations), as neurons in this location are known to
lateral horn. Substance P is co-localized with se- communicate with sympathetic preganglionic neu-
rotonin in brainstem neurons that innervate the rons (Cano et al., 2001; Tang et al., 2004). The cell
spinal cord (Sasek et al., 1990). A large subset bodies of origin of the intraspinal substance P in-
of spinally projecting cardiovascular neurons in put have yet to be defined.
the medulla contain preproenkephalin mRNA
(Stornetta et al., 1999) and immunoreactivity for
neuropeptide Y and mRNA for preproneuropep- Rostrocaudal differences in sympathetic
tide Y have been identified in medullospinal neu- preganglionic neurons and their innervation
rons (Minson et al., 1994; Stornetta et al., 1999).
Nevertheless, complete spinal cord transection The sympathetic nervous system was original-
does not destroy all varicose axons in the inter- ly thought to act in an undifferentiated way to
mediolateral cell column immunoreactive for sub- allow an animal to respond appropriately to life-
stance P, enkephalin or neuropeptide Y (Davis threatening situations. However, over the past two
and Cabot, 1984; Romagnano et al., 1987; Cassam decades, it has become increasingly clear that cen-
et al., 1997) and we have shown that, caudal to a 7- tral control of sympathetic outflow is differential,
day complete transection, axons containing each permitting specific functional groups of sympathetic
of these neuropeptides form synapses on choline preganglionic neurons to respond in different ways
18

Fig. 4. Caudal to a complete spinal cord transection (TX), axons containing the neuropeptides, substance P, enkephalin and ne-
uropeptide Y, are still present in the intermediolateral cell column of mid-thoracic spinal cord segments. (A, C, E) Intact cord. (B, D,
F) 2-week transected cord. (A, B) Stained for substance P (SP), (C, D) stained for enkephalin (ENK) and (E, F) stained for
neuropeptide Y (NPY). Transections were located in caudal thoracic segment 4/rostral thoracic segment 5. Micrographs show the
intermediolateral cell column from thoracic segments 7, 8 or 9. Bars, 100 mm.

to the same homeostatic challenge (reviewed by the outcomes of spinal cord injury will vary de-
Morrison, 2001). Differences in the spatial ar- pending on the functional group of sympathetic
rangement of sympathetic preganglionic neurons preganglionic neurons that are deprived of their
and in their innervation are likely to be the ana- supraspinal input.
tomical basis for these differentiated physiological Sympathetic preganglionic neurons are topo-
responses. The differences in the innervation of graphically organized along the rostrocaudal axis
sympathetic preganglionic neurons suggest that of the spinal cord (Strack et al., 1988). Preganglionic
19

Fig. 5. Caudal to a complete spinal cord transection (TX), axons containing the neuropeptides, substance P, enkephalin and ne-
uropeptide Y, are still present in the intermediolateral cell column of lower thoracic and upper lumbar segments. (A, C, E) Intact cord.
(B, D, F) 2-week transected cord. (A, B) Stained for substance P (SP), (C, D) stained for enkephalin (ENK) and (E, F) stained for
neuropeptide Y (NPY). Transections were located in caudal thoracic segment 4/rostral thoracic segment 5. Micrographs show the
intermediolateral cell column from thoracic segment 13 or lumbar segment 1. Bars, 100 mm.

neurons in the rostral thoracic cord supply rostral and adrenaline from chromaffin cells. The caudal
sympathetic ganglia (e.g., the superior cervical end of the range includes sympathetic pregangl-
ganglion and the stellate ganglion) and participate ionic neurons involved in regulating the activity of
in the regulation of targets in the upper body, like organs in the lower body, like the urinary bladder,
pupils, salivary glands and the heart. The mid- lower bowel and reproductive organs. Despite this
thoracic cord contains sympathetic preganglionic general topographical organization, the distribu-
neurons that project to the celiac ganglion as part tions of sympathetic preganglionic neurons pro-
of the circuitry controlling mesenteric vasculature, jecting to different target ganglia or adrenal
gut motility and gut secretion as well as sympa- chromaffin cells overlap so that sympathetic pre-
thetic preganglionic neurons projecting to the ad- ganglionic neurons of different functions are in-
renal medulla to regulate release of noradrenaline termixed within each spinal segment. For example,
20

Fig. 6. Caudal to a complete spinal cord transection (TX), axons containing the neuropeptides, substance P, enkephalin and ne-
uropeptide Y, are still present in the central autonomic area of upper lumbar segments. (A, C, E) Intact cord. (B, D, F) 2-week
transected cord. (A, B) Stained for substance P (SP), (C, D) stained for enkephalin (ENK) and (F) stained for neuropeptide Y (NPY).
Transections were located in caudal thoracic segment 4/rostral thoracic segment 5. All micrographs show the central autonomic area in
lumbar segment 2. cc, central canal. Bars, 100 mm.

in the rat, thoracic segment 6 contains a mixture of rostrocaudal arrangement of their cell bodies. In
sympathetic preganglionic neurons that send ax- autonomic areas of cat thoracolumbar cord, nerve
ons to the superior cervical ganglion, stellate gan- fibers immunoreactive for 5-HT, substance P,
glion, celiac ganglion or adrenal medulla. somatostatin, oxytocin, neurotensin or neurophy-
Although not very well studied, rostrocaudal sin show a non-uniform rostrocaudal distribution
differences in the innervation of sympathetic pre- (Krukoff et al., 1985) as do 5-HT-immunoreactive
ganglionic neurons parallel the target-based axons in rabbit intermediolateral cell column
21

(Jensen et al., 1995). In rats, oxytocin-immunore- axons at the light microscopic level (Figs. 5C,
active axons closely appose sympathetic pregangl- and 6C). Similarly, dense baskets were present
ionic neurons retrogradely labeled from the around retrogradely labeled neurons in cords at
cervical sympathetic trunk; but sympathoadrenal 2 and 11 weeks after transection (e.g., Figs. 7 and 8).
preganglionic neurons are not innervated by ox- These observations imply that most of the
ytocin fibers (Holets and Elde, 1982; Appel and enkephalin input to pelvic visceral sympathetic
Elde, 1988). Inputs to choline acetyltransferase- preganglionic neurons comes from spinal neurons
immunoreactive sympathetic preganglionic neu- below the transection. Furthermore, the density of
rons that express Fos in response to hypotension the enkephalin innervation of neurons projecting
also show a heterogenous pattern of innervation to the major pelvic ganglion suggests that inter-
by some types of axons (Minson et al., 2002). neurons are likely to provide the predominant in-
Although apposing almost all Fos-positive sym- put to sympathetic preganglionic neurons that
pathetic preganglionic neurons in upper and control the pelvic viscera.
middle thoracic cord, neuropeptide Y or phenyl- At the electron microscope level, we observed
ethanolamine N-methyltransferase-immunoreac- that the density of synapses on sympathetic pre-
tive axons avoid significant proportions of these ganglionic neurons projecting to the major pelvic
neurons in lower thoracic segments. More than ganglion did not appear to differ in intact and
half of the hypotension-sensitive sympathetic transected cord, although this observation was not
preganglionic neurons in the middle and lower quantified. This conclusion was supported by
thoracic cord lacked appositions from galanin- quantification of enkephalin-immunoreactive in-
immunoreactive axons, whereas some choline ace- put to these neurons. In intact cord, sympathetic
tyltransferase-positive, Fos-negative neurons in preganglionic neurons that projected to the major
the lumbar cord lay in dense baskets of galanin- pelvic ganglion received many synapses from
positive fibers. enkephalin-immunoreactive axon terminals. In
Recently, we have examined enkephalin- the intermediolateral cell column, 52% of the
immunoreactive inputs to sympathetic pregangl- synaptic input to retrogradely labeled cell bodies
ionic neurons retrogradely labeled with cholera was enkephalin-immunoreactive. Furthermore,
toxin B subunit from the major pelvic ganglion this enkephalin innervation was targeted to cell
(Llewellyn-Smith et al., 2005), which contains bodies in preference to dendrites since only 29% of
sympathetic and parasympathetic preganglionic the input to retrogradely labeled dendrites was
neurons innervating the urinary bladder, lower enkephalin positive. In the 2-week transected cord,
bowel and reproductive organs. This work has re- enkephalin occurred in 65% of the varicosities that
vealed a striking difference between the reaction to synapsed on sympathetic preganglionic somata
spinal cord injury of these neurons, which have that projected to the major pelvic ganglion from
somata in thoracic segment 12 to lumbar segment the intermediolateral cell column. The proportion-
2, and more rostral sympathetic preganglionic al change in input between cell bodies in intact and
neurons. In contrast to choline acetyltransferase- transected cord was not statistically significant.
immunoreactive sympathetic preganglionic neu- However, the increase in enkephalin input from
rons in thoracic segment 8 (Llewellyn-Smith and 52% to 65% suggests a small loss of synapses due
Weaver, 2001), sympathetic preganglionic neurons to transection. This loss might have been revealed
that are in circuits controlling pelvic viscera appear if data had been collected from a larger number of
to retain most of their innervation after a complete rats. These data indicate that the enkephalin input
spinal cord transection. In intact cord, we found to pelvic visceral sympathetic preganglionic neu-
that sympathetic preganglionic neurons projecting rons is not significantly affected by transection,
to the major pelvic ganglion from the intermedio- due to the fact that it is predominantly intraspinal.
lateral cell column, the intercalated nucleus and Since pelvic visceral sympathetic preganglionic
central autonomic area were surrounded by very neurons are not substantially denervated after spi-
dense baskets of enkephalin-immunoreactive nal cord injury, their somata and dendrites may
22

Fig. 7. In transected cord, enkephalin-immunoreactive axons closely appose sympathetic preganglionic neurons projecting to the
major pelvic ganglion. Transections (TX) were located in caudal thoracic segment 4/rostral thoracic segment 5. (A) A sympathetic
preganglionic neuron (asterisk) that has retrogradely transported cholera toxin B subunit (CTB) from the major pelvic ganglion
(MPG) lies at the lateral edge of the intermediolateral cell column (IML) in 11-week transected cord. A host of enkephalin (ENK)-
immunoreactive varicosities form close appositions on the sympathetic preganglionic neuron. A retrogradely labeled dendrite in the
white matter (WM) also receives many close appositions from enkephalin-containing terminals. Some appositions are indicated by
arrowheads. Bar, 20 mm. (B) Retrogradely labeled sympathetic preganglionic neurons (asterisks) in the central autonomic area lie
within a very dense network of enkephalin (ENK)-immunoreactive axons. Bar, 50 mm.

not undergo the shrinkage and regrowth that we affected, they should be more easily restored after
have previously documented in more rostral sym- an injury because there will be less dependence on
pathetic preganglionic neurons immediately after the re-establishment of direct synaptic input from
injury. However, further studies are needed to ex- regrowing supraspinal axons. It will be interesting to
plore this possibility. see whether spinal interneurons are equally impor-
The dominance of intraspinal pathways in the tant in the regulation of parasympathetic pregangl-
control of sympathetic preganglionic neurons sup- ionic neurons, which project to the major pelvic
plying the major pelvic ganglion has important im- ganglion from the lower lumbar and upper sacral
plications for the restoration of pelvic visceral cord and are also critical for pelvic visceral function.
function after spinal cord injury. Since intraspinal
circuits controlling pelvic visceral sympathetic pre- Acknowledgments
ganglionic neurons are relatively unaffected by spi-
nal cord injury, drug treatments that target this Project Grants (#229907 to ILS and #000044 to
persistent circuitry should help to normalize sym- JRK) and Research Fellowships (#229921 to ILS
pathetically mediated pelvic visceral reflexes. Fur- and #358709 to JRK) from the National Health
thermore, since sympathetic components are less and Medical Research Council of Australia, grants
23

Fig. 8. In transected cord, enkephalin-immunoreactive axons synapse on sympathetic preganglionic neurons projecting to the major
pelvic ganglion. In the central autonomic area of 2-week transected cord, an enkephalin-immunoreactive varicosity (ENK) forms a
synapse (arrowheads) on a dendrite that contains a crystal due to retrograde transport of cholera toxin B subunit (CTB) from the
major pelvic ganglion (MPG). The transection was located in caudal thoracic segment 4/rostral thoracic segment 5. Bar, 500 nm.

from the National Heart Foundation of Australia Bowker, R.M., Westlund, K.N., Sullivan, M.C. and Coulter,
(#G98A0097 and #G00A0512 to ILS), a Visiting J.D. (1982) Organization of descending serotonergic projec-
tions to the spinal cord. Prog. Brain Res., 57: 239–265.
Scientist Award from the Heart and Stroke Foun-
Cabot, J.B. (1990) Sympathetic preganglionic neurons: Cyto-
dation of Canada (ILS), Ontario Heart and Stroke architecture, ultrastructure and biophysical properties. In:
Foundation (LCW) and the Canadian Institutes of Loewy A.D. and Spyer K.M. (Eds.), Central Regulation of
Health Research (LCW) supported this work. Autonomic Functions. Oxford University Press, pp. 44–67.
Carolyn Martin, Natalie Fenwick and Lee Travis Cabot, J.B., Alessi, V. and Bushnell, A. (1992) Glycine-like
provided expert technical assistance. immunoreactive input to sympathetic preganglionic neurons.
Brain Res., 571: 1–18.
Cabot, J.B., Alessi, V., Carroll, J. and Ligorio, M. (1994) Spinal
cord lamina V and lamina VII interneuronal projections to
References sympathetic preganglionic neurons. J. Comp. Neurol., 347:
515–530.
Antunes, V.R., Brailoiu, G.C., Kwok, E.H., Scruggs, P. and Cano, G., Sved, A.F., Rinaman, L., Rabin, B.S. and Card, J.P.
Dun, N.J. (2001) Orexins/hypocretins excite rat sympathetic (2001) Characterization of the central nervous system inner-
preganglionic neurons in vivo and in vitro. Am. J. Physiol. vation of the rat spleen using viral transneuronal tracing.
Regul. Integr. Comp. Physiol., 281: R1801–R1807. J. Comp. Neurol., 439: 1–18.
Appel, N.M. and Elde, R.P. (1988) The intermediolateral cell Cassam, A.K., Llewellyn-Smith, I.J. and Weaver, L.C. (1997)
column of the thoracic spinal cord is comprised of target- Catecholamine enzymes and neuropeptides are expressed in
specific subnuclei: evidence from retrograde transport studies fibres and somata in the intermediate gray matter in chronic
and immunohistochemistry. J. Neurosci., 8: 1767–1775. spinal rats. Neuroscience, 78: 829–841.
Bacon, S.J. and Smith, A.D. (1988) Preganglionic sympathetic Cassam, A.K., Rogers, K.A. and Weaver, L.C. (1999) Co-lo-
neurones innervating the rat adrenal medulla: immunocyto- calization of substance P and dopamine beta-hydroxylase
chemical evidence of synaptic input from nerve terminals with growth-associated protein-43 is lost caudal to a spinal
containing substance P, GABA or 5-hydroxytryptamine. cord transection. Neuroscience, 88: 1275–1288.
J. Auton. Nerv. Syst., 24: 97–122. Christensen, M.D. and Hulsebosch, C.E. (1997) Spinal cord
Bernstein-Goral, H. and Bohn, M.C. (1989) Phenylethanola- injury and anti-NGF treatment results in changes in CGRP
mine N-methyltransferase-immunoreactive terminals synapse density and distribution in the dorsal horn in the rat. Exp.
on adrenal preganglionic neurons in the rat spinal cord. Ne- Neurol., 147: 463–475.
uroscience, 32: 521–537. Clarke, H.A., Dekaban, G.A. and Weaver, L.C. (1998) Iden-
Bogan, N., Mennone, A. and Cabot, J.B. (1989) Light micro- tification of lamina V and VII interneurons presynaptic to
scopic and ultrastructural localization of GABA-like adrenal sympathetic preganglionic neurons in rats using a
immunoreactive input to retrogradely labeled sympathetic recombinant herpes simplex virus type 1. Neuroscience, 85:
preganglionic neurons. Brain Res., 505: 257–270. 863–872.
24

Coote, J.H., Macleod, V.H., Fleetwood-Walker, S.M. and afferents, by alpha-methylnoradrenaline and endogenous
Gilbey, M.P. (1981) The response of individual sympathetic catecholamines. Neuropharmacology, 22: 3–17.
preganglionic neurons to microiontophoretically applied en- Kolaj, M., Shefchyk, S.J. and Renaud, L.P. (1997) Two con-
dogenous monoamines. Brain Res., 215: 135–145. ductances mediate thyrotropin-releasing-hormone-induced
Dampney, R.A.L. (1994) Functional organization of central depolarization of neonatal rat spinal preganglionic and lat-
pathways regulating the cardiovascular system. Physiol. eral horn neurons. J. Neurophysiol., 78: 1726–1729.
Rev., 74: 323–364. Krassioukov, A.V. and Weaver, L.C. (1996) Morphological
Davis, B.M. and Cabot, J.B. (1984) Substance P-containing changes in sympathetic preganglionic neurons after spinal
pathways to avian sympathetic preganglionic neurons: evi- cord injury in rats. Neuroscience, 70: 211–225.
dence for major spinalspinal circuitry. J. Neurosci., 4: Krenz, N.R., Meakin, S.O., Krassioukov, A.V. and Weaver,
2145–2159. L.C. (1999) Neutralizing intraspinal nerve growth factor
Deuchars, S.A., Brooke, R.E., Frater, B. and Deuchars, J. blocks autonomic dysreflexia caused by spinal cord injury.
(2001) Properties of interneurones in the intermediolateral J. Neurosci., 19: 7405–7414.
cell column of the rat spinal cord: role of the potassium Krenz, N.R. and Weaver, L.C. (1998a) Changes in the mor-
channel subunit Kv3.1. Neuroscience, 106: 433–446. phology of sympathetic preganglionic neurons parallel the
Deuchars, S.A., Morrison, S.F. and Gilbey, M.P. (1995) development of autonomic dysreflexia after spinal cord injury
Medullary-evoked EPSPs in neonatal rat sympathetic in rats. Neurosci. Lett., 243: 61–64.
preganglionic neurones in vitro. J. Physiol., 487(Part 2): Krenz, N.R. and Weaver, L.C. (1998b) Sprouting of primary
453–463. afferent fibers after spinal cord transection in the rat.
Deuchars, S.A., Spyer, K.M. and Gilbey, M.P. (1997) Stimu- Neuroscience, 85: 443–458.
lation within the rostral ventrolateral medulla can evoke Krukoff, T.L., Ciriello, J. and Calaresu, F.R. (1985) Segmental
monosynaptic GABAergic IPSPs in sympathetic pregangl- distribution of peptide- and 5HT-like immunoreactivity in
ionic neurons in vitro. J. Neurophysiol., 77: 229–235. nerve terminals and fibres of the thoracolumbar sympathetic
Dun, N.J. and Mo, N. (1988) In vitro effects of substance P on nuclei of the cat. J. Comp. Neurol., 240: 103–116.
neonatal rat sympathetic preganglionic neurones. J. Physiol., Krupp, J., Bordey, A. and Feltz, P. (1997) Electrophysiological
399: 321–333. evidence for multiple glycinergic inputs to neonatal rat sym-
Holets, V. and Elde, R. (1982) The differential distribution and pathetic preganglionic neurons in vitro. Eur. J. Neurosci., 9:
relationship of serotonergic and peptidergic fibers to sym- 1711–1719.
pathoadrenal neurons in the intermediolateral cell column of Krupp, J. and Feltz, P. (1995) Excitatory postsynaptic currents
the rat: a combined retrograde axonal transport and and glutamate receptors in neonatal rat sympathetic pregangl-
immunofluorescence study. Neuroscience, 7: 1155–1174. ionic neurons in vitro. J. Neurophysiol., 72(4): 1503–1512.
Inokuchi, H., Yoshimura, M., Trzebski, A., Polosa, C. and Lai, C.C., Wu, S.Y., Lin, H.H. and Dun, N.J. (1997) Excitatory
Nishi, S. (1992a) Fast inhibitory postsynaptic potentials and action of pituitary adenylate cyclase activating polypeptide
responses to inhibitory amino acids of sympathetic pregangl- on rat sympathetic preganglionic neurons in vivo and in
ionic neurons in the adult cat. J. Auton. Nerv. Syst., 41: vitro. Brain Res., 748: 189–194.
53–60. Lewis, D.I. and Coote, J.H. (1990a) Excitation and inhibition
Inokuchi, H., Yoshimura, M., Yamada, S., Polosa, C. and of rat sympathetic preganglionic neurons by catecholamines.
Nishi, S. (1992b) Fast excitatory postsynaptic potentials and Brain Res., 530: 229–234.
the responses to excitant amino acids of sympathetic Lewis, D.I. and Coote, J.H. (1990b) The influence of 5-hydro-
preganglionic neurons in the slice of the cat spinal cord. xytryptamine agonists and antagonists on identified sympa-
Neuroscience, 46: 657–667. thetic preganglionic neurons in the rat, in vivo. Br. J.
Jansen, A.S.P., Wessendorf, M.W. and Loewy, A.D. (1995) Pharmacol., 99: 667–672.
Transneuronal labeling of CNS neuropeptide and monoa- Llewellyn-Smith, I.J., Arnolda, L.F., Pilowsky, P.M.,
mine neurons after pseudorabies virus injections into the Chalmers, J.P. and Minson, J.B. (1998) GABA- and
stellate ganglion. Brain Res., 683: 1–24. glutamate-immunoreactive synapses on sympathetic pre-
Jensen, I., Llewellyn-Smith, I.J., Minson, J.B., Pilowsky, P.M. ganglionic neurons projecting to the superior cervical gan-
and Chalmers, J.P. (1995) Serotonin inputs to rabbit sympa- glion. J. Auton. Nerv. Syst., 71: 96–110.
thetic preganglionic neurons projecting to the superior cer- Llewellyn-Smith, I.J., Cassam, A.K., Krenz, N.R.,
vical ganglion or adrenal medulla. J. Comp. Neurol., 353: Krassioukov, A.V., Minson, J.B., Pilowsky, P.M., Arnolda,
427–438. L.F., Chalmers, J.P. and Weaver, L.C. (1995a) Do sympa-
Joshi, S., LeVatte, M.A., Dekaban, G.A. and Weaver, L.C. thetic preganglionic neurons receive synapses from intraspi-
(1995) Identification of spinal interneurons antecedent to ad- nal catecholamine neurons? Soc. Neurosci. Abstr., 21: 1401.
renal sympathetic preganglionic neurons using trans-synaptic Llewellyn-Smith, I.J., Cassam, A.K., Krenz, N.R., Krassiou-
transport of herpes simplex virus type 1. Neuroscience, 65: kov, A.V. and Weaver, L.C. (1997) Glutamate- and GABA-
893–903. immunoreactive synapses on sympathetic preganglionic neu-
Kadzielawa, K. (1983) Inhibition of the activity of sympathetic rons caudal to a spinal cord transection in rats. Neuroscience,
preganglionic neurones and neurones activated by visceral 80: 1225–1235.
25

Llewellyn-Smith, I.J., DiCarlo, S.E., Collins, H.L. and Keast, Minson, J.B., Llewellyn-Smith, I.J. and Arnolda, L.F. (2001)
J.R. (2005) Enkephalin-immunoreactive interneurons exten- Neuropeptide Y mRNA expression in interneurons in rat
sively innervate sympathetic preganglionic neurons regulat- spinal cord. Auton. Neurosci., 93: 14–20.
ing the pelvic viscera. J. Comp. Neurol., 488: 278–289. Minson, J.B., Llewellyn-Smith, I.J., Pilowsky, P.M. and
Llewellyn-Smith, I.J., Minson, J.B., Pilowsky, P.M., Arnolda, Chalmers, J.P. (1994) Bulbospinal neuropeptide Y-immuno-
L.F. and Chalmers, J.P. (1995b) The one hundred percent reactive neurons in the rat: comparison with adrenaline-syn-
hypothesis: glutamate or GABA in synapses on sympathetic thesising neurons. J. Auton. Nerv. Syst., 47: 233–243.
preganglionic neurons. Clin. Exp. Hypertens., 17: 323–334. Minson, J., Pilowsky, P., Llewellyn-Smith, I., Kaneko, T.,
Llewellyn-Smith, I.J., Minson, J.B., Pilowsky, P.M. and Kapoor, V. and Chalmers, J. (1991) Glutamate in spinally
Chalmers, J.P. (1991) There are few catecholamine- or ne- projecting neurons of the rostral ventral medulla. Brain Res.,
uropeptide Y-containing synapses in the intermediolateral 555: 326–331.
cell column of rat thoracic spinal cord. Clin. Exp. Pharmacol. Miyazaki, T., Coote, J.H. and Dun, N.J. (1989) Excitatory and
Physiol., 18: 111–115. inhibitory effects of epinephrine on neonatal rat sympathetic
Llewellyn-Smith, I.J., Phend, K.D., Minson, J.B., Pilowsky, preganglionic neurons in vitro. Brain Res., 497: 108–116.
P.M. and Chalmers, J.P. (1992) Glutamate immunoreactive Mo, N. and Dun, N.J. (1987a) Excitatory postsynaptic poten-
synapses on retrogradely labelled sympathetic neurons in rat tials in neonatal rat sympathetic preganglionic neurons: pos-
thoracic spinal cord. Brain Res., 581: 67–80. sible mediation by NMDA receptors. Neurosci. Lett., 77:
Llewellyn-Smith, I.J. and Weaver, L.C. (2001) Changes in 327–332.
synaptic inputs to sympathetic preganglionic neurons after Mo, N. and Dun, N.J. (1987b) Is glycine an inhibitory trans-
spinal cord injury. J. Comp. Neurol., 435: 226–240. mitter in rat lateral horn cells? Brain Res., 400: 139–144.
Llewellyn-Smith, I.J. and Weaver, L.C. (2004) Interneuronal Morrison, S.F. (2001) Differential control of sympathetic out-
inputs to sympathetic preganglionic neurons: evidence from flow. Am. J. Physiol. Regul. Integr. Comp. Physiol., 281:
transected spinal cord. In: Dun N.J., Machado B.H. and R683–R698.
Pilowsky P.M. (Eds.), Neural Mechanisms of Cardiovascular Newton, B.W., Maley, B.E. and Hamill, R.W. (1986)
Regulation. Kluwer, pp. 265–283. Immunohistochemical demonstration of serotonin neurons
Loewy, A.D. and McKellar, S. (1981) Serotonergic projections in autonomic regions of the rat spinal cord. Brain Res., 376:
from the ventral medulla to the intermediolateral cell column 155–163.
in the rat. Brain Res., 211: 146–152. Ondarza, A.B., Ye, Z. and Hulsebosch, C.E. (2003) Direct ev-
Ma, R.C. and Dun, N.J. (1985a) Norepinephrine depolarizes idence of primary afferent sprouting in distant segments fol-
lateral horn cells of neonatal rat spinal cord in vitro. Neuro- lowing spinal cord injury in the rat: colocalization of GAP-43
sci. Lett., 60: 163–168. and CGRP. Exp. Neurol., 184: 373–380.
Ma, R.C. and Dun, N.J. (1985b) Vasopressin depolarizes lat- Pickering, A.E., Spanswick, D. and Logan, S.D. (1994)
eral horn cells of the neonatal rat spinal cord in vitro. Brain 5-Hydoxytryptamine evokes depolarizations and membrane
Res., 348: 36–43. potential oscillations in rat sympathetic preganglionic neu-
Ma, R.C. and Dun, N.J. (1986) Excitation of lateral horn neu- rones. J. Physiol., 480: 109–121.
rons of the neonatal rat spinal cord by 5-hydroxytryptamine. Pilowsky, P., Llewellyn-Smith, I.J., Lipski, J. and Chalmers, J.
Dev. Brain Res., 24: 89–98. (1992) Substance P immunoreactive boutons form synapses
Maiorov, D.N., Krenz, N.R., Krassioukov, A.V. and Weaver, with feline sympathetic preganglionic neurons. J. Comp. Ne-
L.C. (1997a) Role of spinal NMDA and AMPA receptors in urol., 320: 121–135.
episodic hypertension in conscious spinal rats. Am. J. Romagnano, M.A., Braiman, J., Loomis, M. and Hamill, R.W.
Physiol., 273: H1266–H1274. (1987) Enkephalin fibers in autonomic nuclear regions: in-
Maiorov, D.N., Weaver, L.C. and Krassioukov, A.V. (1997b) traspinal vs. supraspinal origin. J. Comp. Neurol., 266:
Relationship between sympathetic activity and arterial pres- 319–331.
sure in conscious spinal rats. Am. J. Physiol., 272: Sasek, C.A., Wessendorf, M.W. and Helke, C.J. (1990) Evi-
H625–H631. dence for co-existence of thyrotropin-releasing hormone,
Marsh, D.R. and Weaver, L.C. (2004) Autonomic dysreflexia, substance P and serotonin in ventral medullary neurons that
induced by noxious or innocuous stimulation, does not de- project to the intermediolateral cell column in the rat. Ne-
pend on changes in dorsal horn substance P. J. Neurotrauma, uroscience, 35: 105–119.
21: 817–828. Stornetta, R.L., Akey, P.J. and Guyenet, P.G. (1999) Location
Milner, T.A., Morrison, S.F., Abate, C. and Reis, D.J. (1988) and electrophysiological characterization of rostral medul-
Phenylethanolamine N-methyltransferase-containing termi- lary adrenergic neurons that contain neuropeptide Y mRNA
nals synapse directly on sympathetic preganglionic neurons in rat medulla. J. Comp. Neurol., 415: 482–500.
in the rat. Brain Res., 448: 205–222. Stornetta, R.L., McQuiston, T.J. and Guyenet, P.G. (2004)
Minson, J.B., Arnolda, L.F. and Llewellyn-Smith, I.J. (2002) GABAergic and glycinergic presympathetic neurons of rat
Neurochemistry of nerve fibers apposing sympathetic pre- medulla oblongata identified by retrograde transport of
ganglionic neurons activated by sustained hypotension. pseudorabies virus and in situ hybridization. J. Comp Ne-
J. Comp. Neurol., 449: 307–318. urol., 479: 257–270.
26

Stornetta, R.L., Sevigny, C.P., Schreihofer, A.M., Rosin, D.L. phalin-, and serotonin-immunoreactive terminals and retro-
and Guyenet, P.G. (2002) Vesicular glutamate transporter gradely labeled sympathetic preganglionic neurons in the rat:
DNPI/VGLUT2 is expressed by both C1 adrenergic and a study using a double-peroxidase procedure. Synapse, 6:
nonaminergic presympathetic vasomotor neurons of the rat 221–229.
medulla. J. Comp. Neurol., 444: 207–220. Weaver, L.C., Cassam, A.K., Krassioukov, A.V. and Lle-
Strack, A.M., Sawyer, W.B., Hughes, J.H., Platt, K.B. and wellyn-Smith, I.J. (1997) Changes in immunoreactivity for
Loewy, A.D. (1989) A general pattern of CNS innervation of growth associated protein-43 suggest reorganization of
the sympathetic outflow demonstrated by transneuronal synapses on spinal sympathetic neurons after cord transec-
pseudorabies viral infections. Brain Res., 491: 156–162. tion. Neuroscience, 81: 535–551.
Strack, A.M., Sawyer, W.B., Marubio, L.M. and Loewy, A.D. Weaver, L.C., Verghese, P., Bruce, J.C., Fehlings, M.G., Krenz,
(1988) Spinal origin of sympathetic preganglionic neurons in N.R. and Marsh, D.R. (2001) Autonomic dysreflexia and
the rat. Brain Res., 455: 187–191. primary afferent sprouting after clip-compression injury of
Sved, A.F., Cano, G. and Card, J.P. (2001) Neuroanatomical the rat spinal cord. J. Neurotrauma, 18: 1107–1119.
specificity of the circuits controlling sympathetic outflow to Wong, S.T., Atkinson, B.A. and Weaver, L.C. (2000) Confocal
different targets. Clin. Exp. Pharmacol. Physiol., 28: microscopic analysis reveals sprouting of primary afferent
115–119. fibres in rat dorsal horn after spinal cord injury. Neurosci.
Tang, X., Neckel, N.D. and Schramm, L.P. (2004) Spinal in- Lett., 296: 65–68.
terneurons infected by renal injection of pseudorabies virus in Yusof, A.P.M. and Coote, J.H. (1988) Excitatory and inhibi-
the rat. Brain Res., 1004: 1–7. tory actions of intrathecally administered 5-hydroxytrypta-
van den Top, M., Nolan, M.F., Lee, K., Richardson, P.J., mine on sympathetic nerve activity in the rat. J. Auton. Nerv.
Buijs, R.M., Davies, C.H. and Spanswick, D. (2003) Orexins Syst., 22: 229–236.
induce increased excitability and synchronisation of rat sym- Zagon, A. and Smith, A.D. (1993) Monosynaptic projections
pathetic preganglionic neurones. J. Physiol., 549: 809–821. from the rostral ventrolateral medulla oblongata to identified
Vera, P.L., Holets, V.R. and Miller, K.E. (1990) Ultrastructural sympathetic preganglionic neurons. Neuroscience, 54:
evidence of synaptic contacts between substance P-, enke- 729–743.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 2

Spinal sympathetic interneurons: Their identification


and roles after spinal cord injury

Lawrence P. Schramm

Departments of Biomedical Engineering and Neuroscience, The Johns Hopkins University School of Medicine, 606 Traylor
Building, 720 Rutland Avenue, Baltimore, MD 21205, USA

Abstract: Primary afferent neurons rarely, if ever, synapse on the sympathetic preganglionic neurons that
regulate the cardiovascular system, nor do sympathetic preganglionic neurons normally exhibit sponta-
neous activity in the absence of excitatory inputs. Therefore, after serious spinal cord injury ‘‘spinal
sympathetic interneurons’’ provide the sole excitatory and inhibitory inputs to sympathetic preganglionic
neurons. Few studies have addressed the anatomy and physiology of spinal sympathetic interneurons, to a
great extent because they are difficult to identify. Therefore, this chapter begins with descriptions of both
neurophysiological and neuroanatomical criteria for identifying spinal sympathetic interneurons, and it
discusses the advantages and disadvantages of each. Spinal sympathetic interneurons also have been little
studied because their importance in intact animals has been unknown, whereas the roles of direct pro-
jections from the brain to sympathetic preganglionic neurons are better known. This chapter presents
evidence that spinal sympathetic interneurons play only a minor role in sympathetic regulation when the
spinal cord is intact. However, they play an important role after spinal cord injury, both in generating
ongoing activity in sympathetic nerves and in mediating segmental and intersegmental sympathetic reflexes.
The spinal sympathetic interneurons that most directly influence the activity of sympathetic preganglionic
neurons after spinal cord injury are located close to their associated sympathetic preganglionic neurons,
and the inputs from distant segments that mediate multisegmental reflexes are relayed to sympathetic
preganglionic neurons multisynaptically via spinal sympathetic interneurons. Finally, spinal sympathetic
interneurons are more likely to be excited and less likely to be inhibited by both noxious and innocuous
somatic stimuli after chronic spinal transection. The onset of this hyperexcitability corresponds to mor-
phological changes in both sympathetic preganglionic neurons and primary afferents, and it may reflect the
pathophysiological processes that lead to autonomic dysreflexia and the hypertensive crises that may occur
with it in people after chronic spinal injury.

Introduction maker potentials, and under ordinary


circumstances their activity is determined by
Sympathetic preganglionic neurons are the final synaptic inputs from the brain and spinal cord
neurons within the central nervous system that (see Laskey and Polosa, 1988, for review). The
regulate sympathetic output to nearly every tissue regulation of the activity of sympathetic pregangl-
and organ. Like somatic motoneurons, sympa- ionic neurons by brainstem systems has been ex-
thetic preganglionic neurons do not exhibit pace- tensively investigated (Laskey and Polosa, 1988;
Cabot, 1996; Blessing, 1997). However, less is
Corresponding author. Tel.: +410-955-3026; known about the regulation of sympathetic activ-
Fax: +410-955-9826; E-mail: LSCHRAMM@bme.jhu.edu ity after spinal cord injury, when inputs from the

DOI: 10.1016/S0079-6123(05)52002-8 27
28

brainstem are lost. Sympathetic activity after after spinal cord injury and because they may play
spinal cord injury is enigmatic because it ranges positive roles in the recovery of autonomic function
from abnormally low, leading to bouts of hypo- or pathological roles in mediating autonomic dys-
tension, to abnormally high, leading to hyperten- function during spinal cord repair and regeneration.
sive crises (Mathias and Frankel, 1992). One The anatomy and physiology of spinal sympa-
characteristic upon which there appears to be lit- thetic systems have been comprehensively re-
tle disagreement is that few, if any, spinal primary viewed (Laskey and Polosa, 1988; Cabot, 1996;
afferents synapse directly upon sympathetic pre- Weaver and Polosa, 1997). Therefore, this chapter
ganglionic neurons (Laskey and Polosa, 1988). will concentrate on recent studies of the anatomy
Therefore, by definition, after a complete spinal and physiology of spinal sympathetic interneurons
cord transection spinal interneurons convey all in rats with intact, acutely transected, and chron-
spinal inputs to sympathetic preganglionic neu- ically transected spinal cords. I begin by reviewing
rons, whether these inputs are derived from pri- the methods used to identify and characterize spi-
mary afferents or from intraspinal sources of nal sympathetic interneurons.
ongoing sympathetic activity.
For the purposes of this chapter, I define spinal Spinal sympathetic interneurons are identified both
interneurons as all spinal neurons other than (1) physiologically and anatomically
somatic motoneurons and (2) autonomic pregangl-
ionic neurons. I define spinal sympathetic inter- Ideally, we could identify each spinal sympathetic
neurons as spinal interneurons with connections interneuron, whether characterized physiologically
that can directly or indirectly affect sympathetic or anatomically, by tracing its axon to synapses
activity. Some spinal neurons that play no role in upon sympathetic preganglionic neurons. However,
sources of sympathetic activity in intact spinal this is possible under only special conditions, usu-
cords may participate in those sources after spinal ally in vitro (see, for example, Deuchars et al.,
cord injury. These neurons may, themselves, be 2001). In all other cases, the ‘‘sympathetic’’ nature
under tonic descending inhibition when the spinal of spinal sympathetic interneurons must either be
cord is intact, or their connections to sympathetic inferred from their neurophysiological properties
preganglionic neurons may be via other interneu- or by tracing their connections to sympathetic
rons that are tonically inhibited. Such neurons preganglionic neurons using specialized, trans-
would be classified as spinal sympathetic interneu- synaptic, retrograde labeling methods.
rons after, but not before, spinal cord injury. Gebber and colleagues pioneered neurophysio-
That spinal sympathetic interneurons have been logical identification of spinal sympathetic inter-
little studied is understandable for two reasons. neurons by identifying spinal neurons with
First, as discussed below, they are unique neither discharge patterns that were correlated with the
in their neurotransmitters nor their morphology. discharge patterns in either pre- or postganglionic
Thus, they are not readily identified. Second, Miller sympathetic axons (Gebber and McCall, 1976;
et al. (2001) found very few spinal interneurons Barman and Gebber, 1984). These investigators
with activities correlated with ongoing renal sym- cross-correlated the ongoing activity of single spi-
pathetic nerve activity in rats with intact spinal nal interneurons and the ongoing activity in sym-
cords. This observation suggests that spinal sym- pathetic nerves. Neurons with activities either
pathetic interneurons play a minor role in the reg- positively or negatively correlated with sympathet-
ulation of sympathetic activity in animals with ic nerve activity were defined as spinal sympathetic
intact spinal cords. Therefore, these interneurons interneurons. This remains the only neurophysio-
have not attracted attention in studies of normal, logical method for identifying spinal sympathetic
autonomic regulation of the circulation. In recent interneurons (Chau et al., 1997, 2000; Miller et al.,
years, however, spinal sympathetic interneurons 2001; Tang et al., 2003).
have attracted more attention because they may Neurophysiological identification of spinal sym-
play important roles in autonomic dysfunction pathetic interneurons has two drawbacks. First,
29

how does one distinguish between spinal sympa-


thetic interneurons and sympathetic preganglionic
neurons? The activities of both types of neurons
could be correlated with sympathetic nerve activ-
ity. Second, how does one distinguish between
spinal sympathetic interneurons and other inter-
neurons which share inputs with sympathetic pre-
ganglionic neurons but which are not involved in
sympathetic processing?
Distinguishing between spinal sympathetic in-
terneurons and sympathetic preganglionic neurons
is the easier of these problems. As shown by
Gebber and McCall (1976), sympathetic pregangl-
ionic neurons rarely discharge at rates exceeding
20 Hz. Therefore, the minimum interspike interval
for sympathetic preganglionic neurons is approx-
imately 50 ms. The discharge patterns of spinal
interneurons, on the other hand, usually include
bursts of action potentials with interspike intervals
of 20 ms or less. Therefore, with the relatively mi-
nor risk of misidentifying some spinal sympathetic
interneurons with low discharge rates, spinal sym-
pathetic interneurons can be distinguished from
sympathetic preganglionic neurons by the presence
of short interspike intervals in their discharge pat-
terns. An additional criterion for some spinal sym- Fig. 1. Neurophysiological identification of spinal sympathetic
pathetic interneurons is their dorsal location in the interneurons by cross-correlation. Upper panel: renal sympa-
spinal cord. Sympathetic preganglionic neurons thetic nerve activity (RSNA, upper trace) and simultaneously
are never located within spinal laminae I–V. recorded occurrences of action potentials in a spinal interneu-
ron (lower trace). Lower panel: cross-correlation between a
Therefore, sympathetically correlated neurons lo- 10 min recording of spinal neuronal action potentials and si-
cated in the dorsal laminae of the spinal cord are multaneously recorded RSNA. From Krassioukov et al. (2002),
very likely to be spinal sympathetic interneurons. with permission.
Figure 1 illustrates the neurophysiological iden-
tification of a putative spinal sympathetic inter- these activities was calculated (Fig. 1, lower panel,
neuron. The ongoing activity of a spinal neuron dark trace). Zero time on the correlogram was the
was recorded at a depth of 300 mm from the dorsal instant at which the interneuron began a burst of
surface of the 10th thoracic (T10) spinal segment of activity. The sharp positive peak in the correlo-
an anesthetized rat, acutely spinally transected at gram, approximately 75 ms after the onset of the
the 3rd cervical segment (C3). The neuron was burst, indicated that bursts of renal sympathetic
identified as an interneuron both by its dorsal po- nerve activity regularly lagged the onset of bursts
sition and by the presence of bursts of activity with of activity of the interneuron by 75 ms. To gauge
interspike intervals of 10 ms. At the temporal res- the significance of the correlation, the interdis-
olution of Fig. 1, these bursts are visible as darker charge intervals of the interneuron’s ongoing
action potential indicators (upper panel, lower activity were shuffled 10 times to generate 10 ‘‘dum-
trace). Ongoing renal sympathetic nerve activity my’’ cross-correlations with renal sympathetic nerve
(Fig. 1, upper panel, upper trace) was recorded activity (Fig. 1, lower panel, lighter traces). The
simultaneously with the ongoing activity of the positive correlation between the interneuron’s actu-
interneuron, and the cross-correlation between al ongoing activity and ongoing renal sympathetic
30

nerve activity was so much larger than the enve- activity. The polarities of somatically evoked re-
lope of the 10 dummy correlations that the prob- sponses of uncorrelated interneurons were much
ability that this relationship could have occurred less likely to match those of simultaneous respons-
by chance was very small. es in renal sympathetic nerve activity. Further-
A more difficult problem than distinguishing more, the excitatory fields of uncorrelated neurons
spinal sympathetic interneurons from sympathetic were significantly larger than those of correlated
preganglionic neurons is distinguishing spinal neurons, and they were often larger than the ex-
sympathetic interneurons from interneurons that citatory fields for renal sympathetic nerve activity.
are only coincidently correlated with sympathetic Excitatory fields are defined as the area of body
nerve activity. An example of such a coincidence surface from which stimulation of sensory recep-
would be the case in which both sympathetic pre- tors evoked excitation of the neuron.
ganglionic neurons and interneurons were driven Once identified neurophysiologically, spinal
by a common synaptic input. Indeed, this distinc- sympathetic interneurons can be anatomically lo-
tion cannot be made unambiguously using neuro- cated and morphologically characterized either by
physiological techniques. Nevertheless, confidence intracellular labeling (Deuchars et al., 2001) or by
in identifying spinal sympathetic interneurons is the juxtacellular labeling method (Pinault, 1996;
possible when (1) bursts of ongoing or evoked ac- Schreihofer and Guyenet, 1997; Tang et al., 2003).
tivity of a sympathetically correlated interneuron The juxtacellular method involves approaching the
usually lead ongoing or evoked bursts of sympa- soma or proximal dendrites of a spinal sympa-
thetic nerve activity by an interval consistent with thetic interneuron very closely and passing positive
the calculated conduction time from the interneu- current pulses into it through a biocytin-filled elec-
ron to the recording site on the sympathetic nerve trode. The current apparently electroporates (gen-
and (2) evoked excitatory and inhibitory responses erates temporary pores in) the neuron’s membrane
of interneurons to applied stimuli are correlated and carries biocytin into the cell. Biocytin rapidly
with responses in sympathetic nerve activity to the diffuses throughout the neuron’s soma and dend-
same stimuli. rites. Labeled neurons are identified and recon-
Figure 1 illustrates a case in which the first of structed histologically after treatment with a
these criteria was met. The 75 ms lag between streptavidin-conjugated chromogen. Although this
bursts of ongoing activity of the interneuron and method has been used to visualize spinal sympa-
bursts of ongoing sympathetic nerve activity rep- thetic interneurons (Tang et al., 2003), it suffers
resents an aggregate conduction velocity of ap- from two drawbacks. First, respiratory and vas-
proximately 0.5 m/s, which is consistent with the cular movements of the spinal cord often prevent
expected conduction velocity of the largely un- one from approaching interneurons closely enough
myelinated axons of the renal sympathetic nerve. to label them without injuring them. Second, al-
Subsequently, this neuron also met the second cri- though the somas and dendrites of labeled spinal
terion. Pinch of the left flank, within the region of sympathetic interneurons are well demonstrated
the T10 dermatome, excited both the activity of by juxtacellular labeling, axons are never observed.
this neuron and renal sympathetic nerve activity, Axons of spinal sympathetic interneurons can be
whereas pinch of the left hip and left shoulder demonstrated by intracellular labeling. To date,
caused decreases in both this neuron’s activity and however, intracellular labeling of spinal sympa-
renal sympathetic nerve activity (data not shown). thetic interneurons has been accomplished only in
Chau et al. (1997) found that the polarities vitro (Deuchars et al., 2001).
(direction in which firing frequency changed, up Although neurophysiological studies permit
or down) of somatically evoked responses of the functional characterization of spinal sympathetic
majority of interneurons with ongoing activities interneurons, correlation methods, alone, cannot
positively correlated with renal sympathetic nerve unequivocally identify spinal sympathetic interneu-
activity matched the polarities of simultaneously rons. Spinal sympathetic interneurons can be more
evoked responses in renal sympathetic nerve definitively identified by retrograde, trans-synaptic
31

tracing from sympathetic preganglionic neurons. infection and the perfusion of the animal. This
In an ingenious series of experiments, Cabot and interval usually ranges between 3 and 6 days. For
colleagues (1994) simultaneously injected the beta identification of spinal sympathetic interneurons,
subunit of cholera toxin (cholera toxin B) and infected rats are kept for approximately 72 h before
wheat germ agglutinin into the superior cervical perfusion. Rats kept for this time manifest no
ganglion of rats. Both the cholera toxin B and the visible symptoms of the infection.
wheat germ agglutinin were transported from the When virus is injected into the adrenal gland,
ganglion to the somas and dendrites of sympa- both preganglionic neurons projecting directly to
thetic preganglionic neurons with synapses in that adrenal chromaffin cells and postganglionic neu-
ganglion. However, only the wheat germ aggluti- rons projecting to both adrenal medullary and ad-
nin was transported further in the retrograde di- renal cortical blood vessels are infected. Therefore,
rection, across the synapses made by spinal the spinal sympathetic interneurons infected by
sympathetic interneurons on sympathetic pre- injection of virus into the adrenal gland may be-
ganglionic neurons, thereby labeling the spinal long to at least two classes of interneurons, neu-
sympathetic interneurons. Thus, sympathetic pre- rons involved in overall metabolic regulation and
ganglionic neurons were identified by their com- neurons involved in the regulation of the adrenal
bined labeling with cholera toxin B and wheat circulation. As discussed below, the distinction
germ agglutinin. Spinal sympathetic interneurons between these classes of interneurons may be of
were identified by their labeling with wheat germ limited importance because it is likely that neither
agglutinin but not cholera toxin B. Although these play an important role in animals with intact spi-
were landmark experiments, they were hampered nal cords. After spinal cord lesions, most stimuli
by faint labeling of spinal sympathetic interneu- that activate one class of adrenal spinal sympa-
rons, due in large part to restricted, retrograde, thetic interneurons are likely to activate both.
trans-synaptic transport of wheat germ agglutinin As in the case of the cholera toxin B and wheat
from sympathetic preganglionic neurons. germ agglutinin experiments described above, viral
More recently, spinal sympathetic interneurons methods also require distinguishing between sym-
have been identified by the retrograde, trans-synap- pathetic preganglionic neurons and spinal sympa-
tic transport of herpes viruses (Strack et al., 1989a, thetic interneurons. Sympathetic preganglionic
b; Schramm et al., 1993; Clarke et al., 1998; Tang neurons can be identified because, in addition to
et al., 2004). Herpes simplex and pseudorabies vi- being immunohistochemically labeled for the virus,
rus are two herpes viruses that are rapidly taken up they also label positively for choline acetyl transf-
by the axons of sympathetic postganglionic neu- erase, a synthetic enzyme for acetyl choline found
rons and by the axons of preganglionic neurons in relatively few spinal neurons other than sympa-
projecting to the adrenal medulla. Virus is trans- thetic preganglionic neurons and somatic motoneu-
ported back to the somas of these neurons where it rons. Thus, spinal neurons that co-label for virus
replicates and moves trans-synaptically to the neu- and choline acetyl transferase can be identified as
rons’ synaptic antecedents. Thus, virus taken up sympathetic preganglionic neurons, and neurons
from a peripheral organ or tissue by sympathetic that are infected but do not co-label for choline
postganglionic neurons infects the sympathetic pre- acetyl transferase can be identified as spinal sym-
ganglionic neurons that synapse on those neurons. pathetic interneurons. Sympathetic preganglionic
Virus replicates in the sympathetic preganglionic neurons and somatic motoneurons can be distin-
neurons, and spinal and brainstem interneurons guished by their differential, dorsoventral locations.
that synapse on infected sympathetic preganglionic An alternative method for identifying sympathetic
neurons are infected by further retrograde, trans- preganglionic neurons depends on their propensity
synaptic movement of virus. Antibodies to the vi- for transporting retrograde tracers from the circu-
ruses are used to label infected neurons. The ap- lation to their somas and dendrites (Fig. 2). In this
proximate number of synapses traversed by the method, a large quantity (8–12 mg/kg) of a con-
virus can be controlled by the interval between the ventional retrograde tracer such as Fluorogolds
32

Fig. 2. Anatomical identification of interneurons using pseudorabies virus and Fluorogolds. Left panel: ultraviolet illumination.
Sympathetic preganglionic neuron (white arrow) identified by fluorescence of intraperitoneally injected Fluorogolds. Right panel:
under illumination for the chromogen used to identify pseudorabies virus, both the sympathetic preganglionic neuron (white arrow)
and a spinal sympathetic interneuron (gray arrow) are visible as gray neurons. The spinal sympathetic interneuron is definitively
identified by its absence under ultraviolet illumination. After Tang et al. (2004), with permission.

is injected either intraperitoneally or subcutaneously Spinal interneurons play a more important role in
(Anderson and Edwards, 1994). Approximately 1 generating sympathetic activity after spinal cord
week post-injection, most peripherally projecting lesions in rats
neurons (such as autonomic preganglionic neurons
and somatic motoneurons) are labeled with the Although most investigators have found that ac-
tracer and can be detected under ultraviolet illu- tivity is reduced in sympathetic nerves of unanest-
mination. Although the labeling of somatic moto- hetized people (Wallin, 1986) and rats
neurons is highly variable by tracers administered (Krassioukov and Weaver, 1995; Randall et al.,
intraperitoneally, the labeling of autonomic pre- 2005) after spinal cord transection, many investi-
ganglionic neurons is more uniform. A potential gators report that detectable levels of ongoing ac-
drawback of this method is that freshly adminis- tivity remain in some nerves. Therefore, spinal
tered Fluorogolds appears to interfere with some sympathetic interneurons must provide ongoing
viral tracing methods (Strack and Loewy; excitatory input to sympathetic preganglionic neu-
Schramm, unpublished data). In our hands, how- rons in the absence of pathways from the brain-
ever, pseudorabies virus can be safely injected 1 stem sympatho-excitatory systems. Although in
week after treatment with this retrograde tracer. anesthetized, surgically prepared rats with acute
The major drawback of the viral tracing meth- spinal transections, sympathetic activity is sub-
ods is that infection by the virus may be capri- stantially reduced in some nerves, it is maintained
cious. Within a population of identically treated, or even increased in others (Meckler and Weaver,
virus-injected animals, some may not exhibit any 1985; Taylor and Schramm, 1987).
infection, some may exhibit infections that appear The observations of decreased activity in some
highly specific (infecting only sympathetic pre- nerves are easily explained by the decrease in sup-
ganglionic neurons and spinal sympathetic inter- raspinal drive to some sympathetic preganglionic
neurons) and some may exhibit infections that neurons after spinal cord injury. Maintenance —
destroy many neurons. A second drawback is that and even increases — in sympathetic activity after
the number of synapses retrogradely traversed by spinal cord injury are less easily explained. Very
the virus can only be estimated from the survival likely, sympathetic preganglionic neurons whose
time. Finally, the viral infection often initiates an activity was either not diminished or was increased
immune response that, itself, could alter the fur- after spinal transection received little drive from
ther transport of the virus. brainstem circuits before transection. Alternatively,
33

brainstem sources of activity for these neurons ongoing activities of almost 50% of the interneu-
were replaced by even more powerful intraspinal rons recorded at T10 were correlated to ongoing
sources after transection. In either case, it also is renal sympathetic nerve activity, the activities of
likely that potentially excitatory spinal inputs to only 16% of interneurons at T8 were correlated
these sympathetic preganglionic neurons were with renal sympathetic nerve activity. The activi-
under tonic inhibition from supraspinal systems. ties of no interneurons at T2, T13 or L2 were cor-
Thus, I propose that spinal transection abolishes related with renal sympathetic nerve activity. In
descending excitation, either directly to sympa- unpublished studies (Chau and Schramm), this
thetic preganglionic neurons or indirectly to spinal exploration extended to C2 and L5 without detect-
sympathetic interneurons. However, it also abol- ing additional interneurons correlated with renal
ishes descending inhibition of spinal systems with sympathetic nerve activity. Because anatomical
excitatory inputs to sympathetic preganglionic data indicate that the sympathetic preganglionic
neurons. Ruggiero et al. (1997a, b) provided clear neurons that are most likely to generate renal
evidence that acute spinal transection releases the sympathetic nerve activity lie in 8th through 12th
activities of many dorsal horn and intermediate thoracic segments (Tang et al., 2004), these data
zone neurons from inhibition. They found that show that circuits in distant spinal segments play
acute cervical spinal cord transection in anestheti- little role in generating ongoing renal sympathetic
zed rats and pigs significantly increased the nerve activity. Whether a similar degree of longi-
number of neurons expressing the c-fos gene in tudinal specificity exists for cardiac and pelvic
many dorsal horn laminae and in lamina VII of the sympathetic nerves remains to be determined.
thoracic spinal cord.
Based on these observations, Miller et al. (2001) Long propriospinal pathways affecting sympathetic
predicted that spinal neurons with ongoing activ- activity are multisynaptic
ities correlated with renal sympathetic nerve ac-
tivity would be relatively rare in rats with intact Although distant spinal segments appear to play
spinal cords because spinal circuits that might ex- little or no role in generating ongoing sympathetic
cite sympathetic preganglionic neurons would be activity in a given segment after spinal transection,
under tonic, supraspinal inhibition. As noted sympathetic reflexes can be evoked by stimulating
above, this prediction was confirmed by their ob- afferents to distant segments (see Weaver and
servation that the activities of only one-fifth as Polosa, 1997, for review). To what extent are the
many spinal interneurons were correlated with re- sympathetic reflexes elicited from distant segments
nal sympathetic nerve activity in rats with intact mediated by monosynaptic projections to sympa-
spinal cords as were correlated in rats with acutely thetic preganglionic neurons? Cabot et al. (1994)
transected spinal cords. noted that spinal sympathetic interneurons retro-
gradely labeled by injecting wheat germ agglutinin
The generation of ongoing sympathetic activity after into the superior cervical ganglion exhibited ‘‘a
spinal transection is localized to a restricted number strict segmental organization’’ with respect to their
of spinal segments associated sympathetic preganglionic neurons. In
other words, wheat germ agglutinin-labeled neu-
As described above, after acute spinal transection, rons (spinal sympathetic interneurons) were not
activity persists in some sympathetic nerves. found in segments that did not contain cholera
To what extent is this ongoing activity generated toxin B-labeled neurons (sympathetic preganglion-
locally, and to what extent does it represent activ- ic neurons).
ity common to the entire spinal cord? Chau et al. These observations were confirmed in the renal
(1997) searched the spinal cord from T2 to the 2nd sympathetic system using the retrograde transport
lumbar (L2) segment for interneurons with activ- of pseudorabies virus. Tang et al. (2004) found
ities correlated to renal sympathetic nerve activity that between caudal cervical and caudal lumbar
in rats with acute spinal transections. Although the segments, infected spinal sympathetic interneurons
34

were located only in segments of caudal thoracic adrenal sympathetic preganglionic neurons were
and rostral lumbar segments, the segments in located across the entire mediolateral span of lamina
which infected sympathetic preganglionic neurons VII, and spinal sympathetic interneurons were
were also located. The first infected thoracic similarly distributed, usually intercalated among
interneurons appear 68–72 h after injection of the sympathetic preganglionic neurons. Pseudorabies
pseudorabies virus into the kidney. This delay is virus injected into the kidney of the rat also
identical to that required to infect brainstem neu- infected sympathetic preganglionic neurons locat-
rons that have known, monosynaptic projections ed across the entire intermediate zone of the spinal
to sympathetic preganglionic neurons. Apparently cord between the lateral funiculus and lamina X
in this model, the time required for retrograde (Tang et al., 2004). The majority of spinal sympa-
transport of pseudorabies virus from its uptake thetic interneurons labeled in those experiments
at a synapse to the soma of the next neuron is brief were similarly distributed.
compared to the time necessary for enough Although most anatomically identified spinal
replication to occur for the virus to be visible sympathetic interneurons have been detected
immunohistochemically in that newly infected among, or just dorsal to, populations of sympa-
neuron. Similarly, the transport time is brief com- thetic preganglionic neurons, small numbers iden-
pared to the time required for replication to in- tified after renal injections of pseudorabies virus
crease the intracellular concentration of virus for were located (in descending order of density) in
retrograde infection of a neuron’s synaptic ante- lamina IV, II, and I (Tang et al., 2004). Interest-
cedents. Because neurons as far rostral as the ingly, spinal sympathetic interneurons identified
paraventricular nucleus of the hypothalamus can by their positive cross-correlations with renal sym-
be infected in as little as 72 h, the absence of spinal pathetic nerve activity were distributed somewhat
sympathetic interneurons in caudal cervical, ros- more widely than anatomically identified spinal
tral thoracic and caudal lumbar spinal cord that sympathetic interneurons, for instance in the me-
lack infected sympathetic preganglionic neurons dial portions of laminae I, II, and III (Chau et al.,
strongly suggests that long propriospinal inputs to 2000; Tang et al., 2003). The wider distribution of
sympathetic preganglionic neurons infected from neurophysiologically identified spinal sympathetic
renal injections are multisynaptic. interneurons was not surprising. Anatomically
The majority of spinal sympathetic interneurons identified spinal sympathetic interneurons were
projecting monosynaptically to sympathetic pre- visualized using a relatively short, post-infection
ganglionic neurons are located either among or survival time (72 h). As discussed above, during
just dorsal to their functionally related popula- that time, pseudorabies virus would have been un-
tions of sympathetic preganglionic neurons. Not likely to have traversed more than the two
only are the longitudinal distributions of spinal synapses between the renal sympathetic postgangl-
sympathetic interneurons and their related sympa- ionic neurons and the first spinal sympathetic in-
thetic preganglionic neurons similar, but the den- terneurons presynaptic to infected sympathetic
sities of spinal sympathetic interneurons are preganglionic neurons. Spinal sympathetic inter-
greatest in or near the spinal laminae that contain neurons identified by cross-correlation, on the
their associated sympathetic preganglionic neu- other hand, could have been located in spinal cir-
rons. Thus, Cabot et al. (1994) localized spinal cuits many synapses removed from sympathetic
sympathetic interneurons to the sympathetic pre- preganglionic neurons and could, therefore, be ex-
ganglionic neuron-rich lateral portion of lamina pected to be located more remotely.
VII and the reticulated (lateral) portion of lamina The locations of spinal sympathetic interneu-
V, just dorsal to the intermediolateral column. rons with respect to sympathetic preganglionic
Clarke et al. (1998) used the retrograde transport neurons may be more important than their loca-
of modified Herpes simplex virus to identify spinal tions with respect to their inputs. Histological re-
sympathetic interneurons that were presynaptic to construction of spinal sympathetic interneurons
adrenal sympathetic preganglionic neurons. Infected (Deuchars et al., 2001; Tang et al., 2003) indicated
35

that the dendritic trees of these neurons often ex-


tended hundreds of microns in two, and sometimes
three, dimensions. Tang et al. (2004) concluded
that the dendrites of some individual spinal sym-
pathetic interneurons were so extensive that they
could receive not only primary afferent inputs but
inputs from a variety of descending or propriospi-
nal pathways as well.

Spinal sympathetic interneurons in rats are more


likely to be excited and less likely to be inhibited by
somatic stimuli after chronic spinal cord transection Fig. 3. Responses of spinal sympathetic interneurons to so-
matic stimulation 1 month after T3 spinal cord transection.
Because the severity of autonomic dysreflexia Upper panel: schematic drawing of the cutaneous regions from
increases with time after spinal cord injury which responses of spinal sympathetically correlated interneu-
(Krassioukov and Weaver, 1995; Krassioukov rons were elicited. Lower panel: representative rate meter re-
sponses of a sympathetically correlated neuron to noxious (left)
et al., 2003), we have supplemented the studies of and innocuous (right) stimulation of cutaneous regions 1–5 in a
rats with acutely transected spinal cords described rat chronically transected at T3. From Krassioukov et al.
above with studies after chronic, T3, spinal tran- (2002), with permission.
section. In both chronically and acutely spinally
transected rats, Krassioukov et al. (2002) identified
spinal sympathetic interneurons in the T10 segment of somatic regions that project to caudal lumbar
by cross-correlation with renal sympathetic nerve spinal cord (Fig. 3, regions 4 and 5) decreased on-
activity. They compared the responses in activity going renal sympathetic nerve activity. The ongo-
of spinal sympathetic interneurons to somatic ing activities of a majority of T10 spinal
stimulation in those two populations. To stand- sympathetic interneurons were inhibited by stim-
ardize stimulation sites, the left body wall was di- ulation of these regions. One month after spinal
vided into five regions, beginning at approximately cord transection, both noxious and innocuous
the T8 dermatome and ending at the left hip and stimulation of regions 1, 3, and 5 were significantly
hindlimb (Fig. 3). more likely to increase the activities of spinal sym-
Two types of stimuli were delivered to these re- pathetic interneurons than in the acutely trans-
gions, a 10-s pinch with toothed forceps (noxious) ected state, and innocuous stimulation of regions 1
and 10 s of brushing with a cotton applicator (in- and 5 was less likely to decrease their activities.
nocuous). Responses in the activities of spinal Although autonomic dysreflexia may occur in
sympathetic interneurons and renal sympathetic the acute stage of spinal cord injury (Krassioukov
nerve activity observed in acutely spinally trans- et al., 2003), it is far more common in the chronic
ected rats by Krassioukov et al. (2002) corre- stage in both humans (Mathias and Frankel, 1992)
sponded closely to those reported previously in and rats (Krassioukov and Weaver, 1995). In rats,
rats with acutely transected spinal cord (Chau the onset of autonomic dysreflexia correlated well
et al., 1997, 2000). Both noxious and innocuous with morphological changes in sympathetic pre-
stimulation of somatic regions projecting to caudal ganglionic neurons (Krenz and Weaver, 1998b)
thoracic spinal cord (Fig. 3, regions 1–3), increased and with increases in sprouting of primary afferent
the magnitudes of bursts in ongoing renal sympa- axons (Krenz and Weaver, 1998a; Wong et al.,
thetic nerve activity. Responses in the activities of 2000). Some of these axons appeared to synapse
spinal sympathetic interneurons were more varia- on neurons appropriately positioned to be spinal
ble. Nevertheless, the majority of T10 spinal sym- sympathetic interneurons (Wong et al., 2000). The
pathetic interneurons were excited by stimulation electrophysiological experiments described above
of regions 1–3. Noxious and innocuous stimulation provided a neurophysiological correlation to both
36

the morphological changes in sympathetic pre- References


ganglionic neurons and primary afferents exhibit-
ed in rats after spinal injury and to the increased Anderson, C.R. and Edwards, S.L. (1994) Intraperitoneal injec-
tions of Fluorogold reliably labels all sympathetic pregangl-
sympathetic reactivity to somatic stimuli experi-
ionic neurons in the rat. J. Neurosci. Methods, 53: 137–141.
enced by spinally injured patients. Barman, S.M. and Gebber, G.L. (1984) Spinal interneurons
with sympathetic nerve-related activity. Am. J. Physiol., 247:
R761–R767.
Summary Blessing, W.W. (1997) The Lower Brainstem and Bodily Home-
ostasis. Oxford University Press, New York.
Cabot, J.B. (1996) Some principles of the spinal organization of
Physiological data suggest that spinal sympathetic the sympathetic preganglionic outflow. Prog. Brain Res., 107:
interneurons play a minimal role in generating and 29–42.
modulating renal sympathetic activity in rats with Cabot, J.B., Alessi, V., Carroll, J. and Ligorio, M. (1994) Spinal
intact spinal cords. Similar data suggest that spinal cord lamina V and lamina VII interneuronal projections in
sympathetic interneurons are released from nor- sympathetic preganglionic neurons. J. Comp. Neurol., 347:
515–530.
mally occurring, descending, tonic inhibition after Chau, D., Johns, D.G. and Schramm, L.P. (2000) Ongoing and
spinal cord injury. Spinal sympathetic interneu- stimulus-evoked activity of sympathetically correlated neu-
rons then play a significant (although not a func- rons in the intermediate zone and dorsal horn of acutely
tionally adaptive) role in both generating ongoing spinalized rats. J. Neurophysiol., 83: 2699–2707.
sympathetic activity and participating in spinal Chau, D., Kim, N. and Schramm, L.P. (1997) Sympathetically
correlated activity of dorsal horn neurons in spinally
sympathetic reflexes. Most spinal sympathetic in- transected rats. J. Neurophysiol., 77: 2966–2974.
terneurons are located close to their associated Clarke, H.A., Dekaban, G.A. and Weave, L.C. (1998) Identi-
sympathetic preganglionic neurons, and the ongo- fication of lamina V and VII interneurons presynaptic to
ing activity in individual sympathetic nerves ap- adrenal sympathetic preganglionic neurons in rats using a
pears to be generated within restricted numbers of recombinant herpes simplex virus type 1. Neuroscience, 85:
863–872.
spinal segments. Although stimulation of afferents Deuchars, S.A., Brooke, R.E., Frater, B. and Deuchars, J.
to distant spinal segments can affect the activity of (2001) Properties of interneurones in the intermediolateral
sympathetic preganglionic neurons, multisynaptic cell column of the rat spinal cord: role of the potassium
(rather than monosynaptic) pathways mediate channel subunit KV3. 1. Neuroscience,, 106: 433–446.
these effects. Finally, 1 month after spinal cord Gebber, G.L. and McCall, R.B. (1976) Identification and dis-
charge patterns of spinal sympathetic interneurons. Am. J.
transection at rostral thoracic level, spinal sympa-
Physiol., 231: 722–723.
thetic interneurons at T10 are more likely to be Krassioukov, A.V. and Weaver, L.C. (1995) Episodic hyper-
excited by both innocuous and noxious stimula- tension due to autonomic dysreflexia in acute and chronic
tion of more widespread cutaneous regions. Col- spinal cord-injured rats. Am. J. Physiol., 268: H2077–H2083.
lectively, these observations provide anatomical Krassioukov, A.V., Furlan, J.C. and Fehlings, M.G. (2003)
Autonomic dysreflexia in acute spinal cord injury: an under-
and physiological substrates for both the genera-
recognized clinical entity. J. Neurotrauma,, 20: 707–716.
tion of ongoing sympathetic activity and the in- Krassioukov, A.V., Johns, D.G. and Schramm, L.P. (2002)
tense, dysfunctional, sympathetic responses to Sensitivity of sympathetically correlated spinal interneurons,
both noxious and innocuous stimuli observed in renal sympathetic nerve activity, and arterial pressure to so-
humans after spinal cord injury. matic and visceral stimuli after chronic spinal injury. J. Ne-
urotrauma,, 19: 1521–1529.
Krenz, N.R. and Weaver, L.C. (1998a) Sprouting of primary
afferent fibers after spinal cord transection in the rat. Ne-
Acknowledgment uroscience, 85: 443–458.
Krenz, N.R. and Weaver, L.C. (1998b) Changes in the mor-
Research co-authored by Lawrence Schramm and phology of sympathetic preganglionic neurons parallel the
development of autonomic dysreflexia after spinal cord injury
the preparation of this chapter were supported by
in rats. Neurosci. Lett., 243: 61–64.
NIH grant HL16315. Critical editorial assistance Laskey, W. and Polosa, C. (1988) Characteristics of the sym-
and advice were provided by Dr. Baohan Pan, pathetic preganglionic neuron and its synaptic input. Prog.
M.D., Ph.D. and Diana C. Schramm, M.A. Neurobiol., 31: 41–87.
37

Mathias, C.J. and Frankel, H.L. (1992) Autonomic disturbanc- Schreihofer, A.M. and Guyenet, P.G. (1997) Identification of
es in spinal cord lesions. In: Bannister R. and Mathias C.J. C1 presympathetic neurons in rat rostral ventrolateral me-
(Eds.), Autonomic Failure, A Textbook of Clinical Disorders dulla by juxtacellular labeling in vivo. J. Comp. Neurol., 387:
of the Autonomic Nervous System. Oxford University Press, 524–536.
New York, pp. 839–881. Strack, A.M., Sawyer, W.B., Platt, K.B. and Loewy, A.D.
Meckler, R.L. and Weaver, L.C. (1985) Splenic, renal, and (1989a) CNS cell groups regulating the sympathetic outflow
cardiac nerves have unequal dependence upon tonic supra- to adrenal gland as revealed by transneuronal cell body labe-
spinal inputs. Brain Res., 338: 123–135. ling with pseudorabies virus. Brain Res., 491: 274–296.
Miller, C.O., Johns, D.G. and Schramm, L.P. (2001) Spinal Strack, A.M., Sawyer, W.B., Hughes, J.H., Platt, K.B. and
interneurons play a minor role in generating ongoing renal Loewy, A.D. (1989b) A general pattern of CNS innervation
sympathetic nerve activity in spinally intact rats. Brain Res., of the sympathetic outflow demonstrated by transneuronal
918: 101–106. pseudorabies viral infections. Brain Res., 491: 156–162.
Pinault, D. (1996) A novel single-cell staining procedure per- Tang, X., Neckel, N.D. and Schramm, L.P. (2003) Locations
formed in vivo under electrophysiological control: morpho- and morphologies of sympathetically correlated neurons in
functional features of juxtacellularly labeled thalamic cells the T10 spinal segment of the rat. Brain Res., 976: 185–193.
and other central neurons with biocytin or neurobiotin. Tang, X., Neckel, N.D. and Schramm, L.P. (2004) Spinal in-
J. Neurosci. Meth., 65: 113–136. terneurons infected by renal injection of pseudorabies virus in
Randall, D.C., Baldridg, B.R., Zimmerman, E.E, Carroll, J.J., the rat. Brain Res., 1004: 1–7.
Speakman, R.O., Brown, D.R., Taylor, R.F., Patwardhan, Taylor, R.F. and Schramm, L.P. (1987) Differential effects of
A. and Burgess, D.E. (Oct. 21, 2004; Epub ahead of print) spinal transection on sympathetic nerve activities in rats. Am.
Blood pressure power within frequency range around 0.4 Hz J. Physiol., 253: R611–R618.
in rat conforms to self-similar scaling following spinal cord Wallin, G. (1986) Abnormalities of sympathetic regulation after
transection. Am. J. Physiol. Regul. Integr. Comp. Physiol. cervical cord lesions. Acta Neurochir. Suppl. (Wien), 36:
Ruggiero, D.A., Sica, A.L., Anwar, M., Frasie, I., Gootman, N. 123–124.
and Gootman, P.M. (1997a) Induction of c-fos gene expression Weaver, L.C. and Polosa, C. (1997) Spinal cord circuits pro-
by spinal cord transection in Sus scrofa. Brain Res., 763: 21–29. viding control of sympathetic preganglionic neurons. In: Jor-
Ruggiero, D.A., Anwar, M., Kim, J., Sica, A.L., Gootman, dan D. (Ed.), The Autonomic Nervous System: Central
A.L. and Gootman, P.M. (1997b) Induction of c-fos gene Nervous Control of Autonomic Function. Harwood Aca-
expression by spinal cord transection in the rat. Brain Res., demic Press, Amsterdam, pp. 29–61.
763: 301–305. Wong, S.T., Atkinson, B.A. and Weaver, L.C. (2000) Confocal
Schramm, L.P., Strack, A.M., Platt, K.B. and Loewy, A.D. microscopic analysis reveals sprouting of primary afferent
(1993) Peripheral and central pathways regulating the kidney: fibres in rat dorsal horn after spinal cord injury. Neurosci.
a study using pseudorabies virus. Brain Res., 616: 251–262. Lett., 296: 65–68.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Published by Elsevier B.V.

CHAPTER 3

Which pathways must be spared in the injured human


spinal cord to retain cardiovascular control?

Andrei Krassioukov

International Collaboration on Repair Discoveries (ICORD) and School of Rehabilitation, University of British Columbia,
Vancouver, BC, Canada and Department of Physical Medicine and Rehabilitation, University of Western Ontario,
London, ON, Canada

Abstract: Cardiovascular abnormalities following spinal cord injury are attributed to autonomic instability
caused by a combination of changes occurring within the spinal cord, including loss of descending au-
tonomic control and plastic changes within spinal and peripheral circuits. Previous animal studies have
shown that localized disruption of the descending vasomotor pathways results in cardiovascular changes
similar to those observed following cord injury. However, the location of these pathways in humans is
uncertain. This chapter presents clinical and histopathological findings from individuals with spinal cord
injury that associates a common area of white matter destruction with severe cardiovascular symptoms.
These data provide evidence that descending vasomotor pathways in the human spinal cord project through
the dorsal aspects of the lateral funiculus.

Introduction cord-injured individuals (Piepmeier et al., 1985;


Lehmann et al., 1987; Atkinson and Atkinson, 1996).
People with cervical or high thoracic spinal cord Likewise, the severity of autonomic dysreflexia
injury face life-long abnormalities in systemic ar- correlates with completeness of spinal injury
terial pressure control (Mathias and Frankel, as assessed by the American Spinal Injury
1992; Karlsson, 1999; Teasell et al., 2000). In gen- Association (ASIA) score: only 27% of incom-
eral, their basal systemic arterial pressure is lower plete quadriplegics present signs of dysreflexia in
than normal and is complicated by orthostatic comparison with 91% of complete quadriplegics
intolerance (Mathias and Frankel, 1992; Cariga (Curt et al., 1997).
et al., 2002). In addition, these cord-injured people Determinations of plasma catecholamine levels
experience transient episodes of hypertension, in cord-injured individuals, and other evidence,
known as ‘‘autonomic dysreflexia’’ that are often suggest that a decrease in sympathetic neuronal
associated with disturbances in heart rate and activity is the main cause of the hypotension and
rhythm (Krassioukov et al., 2003; Clydon et al., postural intolerance (Figoni, 1984; Mathias, 1995;
2005). The severity of spinal cord injury varies Karlsson et al., 1998; Gao et al., 2002). The de-
between individuals and impacts greatly upon creased sympathetic activity, in turn, presumably
cardiovascular control. For example, hypotension results from damage to the spinal pathways that
affects between 20% and 30% of all spinal carry facilitatory input from the lower brainstem
to the sympathetic preganglionic neurons. The de-
Corresponding author. Tel.: +603 822 2673 (off.); struction of these descending vasomotor pathways,
resulting in the loss of excitatory supraspinal input
+604 822 9305 (lab); Fax: +604 822 2924;
E-mail: krassioukov@icord.org to the spinal sympathetic preganglionic neurons,

DOI: 10.1016/S0079-6123(05)52003-X 39
40

is currently considered the major factor for the


persistent lack of sympathetic tone after spinal
cord injury (Mathias and Frankel, 1992; Atkinson
and Atkinson, 1996). Damage to the spinal path-
ways that carry inhibitory input from the lower
brainstem to the sympathetic preganglionic neu-
rons may have a role in autonomic dysreflexia, by
allowing exaggerated activity in the spinal reflex
circuits, caudal to the lesion, that connect spinal
afferent projections to preganglionic neurons. Fig. 1. Schematic diagrams of the possible localization of de-
scending vasomotor pathways within the spinal cord as previ-
Damage to the pathways from the lower brain- ously reported in experimental animals (A) and human
stem to sympathetic preganglionic neurons has a investigations (B). These pathways were localized within dif-
central role in generating the abnormal systemic ferent regions of the white matter in primates (Kerr and
arterial pressure control, typical of cervical or high Alexander, 1964) (arrow 1), in cats and rodents (Lebedev et al.,
thoracic spinal cord injury. Therefore, these path- 1986; Reis et al., 1988; Ruggiero et al., 1989) (arrow 2), and in
humans (Nathan and Smith, 1987) (arrow 3). (B) The boxed
ways are a high priority target for repair, regen- areas indicate the two potential localizations of descending
erative and neuroprotective treatment. Knowledge vasomotor pathways in man, which we examined in the present
of the localization of these pathways in the human investigation: the dorsal aspect of lateral funiculus (Area I), and
spinal cord is therefore essential. This chapter de- white matter adjacent to dorsolateral aspects of the intermedio-
scribes an approach to obtaining such knowledge. lateral cell column (Area II). (From Furlan et al. (2003), with
permission from J. Neurotrauma.)
Previous work in experimental animals, using
electrical stimulation or lesions, led to the conclu-
symptoms had the greatest damage in Area I. The
sion that the pathways for cardiovascular control
remainder of the cases, who had only minor car-
run in the dorsal aspect of the lateral funiculus of
diovascular symptoms, or no symptoms at all, had
the spinal cord white matter (Kerr and Alexander,
significantly less damage in this area. The degree
1964; Illert and Gabriel, 1972; Foreman and
of damage to Area II did not correlate well with
Wurster, 1973; Lebedev et al., 1986). Henceforth,
the extent of cardiovascular dysfunction in these
this area of the white matter will be referred to as
individuals.
Area I (Fig. 1). By contrast, a study in patients
On the basis of these data it may be concluded
undergoing limited cordotomy for the relief of
that, in humans, the pathways from the lower
chronic pain resistant to medical treatment, has
brainstem to the sympathetic preganglionic neu-
suggested that these pathways run in the white
rons run in the dorsal aspect of the lateral fun-
matter adjacent to the dorsolateral aspect of the
iculus of the white matter. Since this is the general
intermediolateral cell column (Nathan and Smith,
region where previous work on experimental an-
1987). Henceforth, this area of the white matter
imals (rat, cat, dog, others) had localized similar
will be referred to as Area II (Fig. 1).
pathways, this study further confirms the relevance
This chapter describes a retrospective study of
to humans of animal model studies of autonomic
cases of spinal cord injury from which detailed
dysfunctions after spinal cord injury.
clinical records and spinal cord specimens were
available. The cases with the most severe cardio-
vascular symptoms were identified. It was hypoth-
esized that this case subset would also have the Study groups
most severe damage to the pathways from lower
brainstem to the sympathetic preganglionic neu- We retrospectively reviewed the charts of the spi-
rons. The extent and severity of white matter nal cord injury cases included in this study and
damage was estimated using stains for myelin and collected data on age and gender, causes of spinal
for an axoplasmic marker. This study has shown cord injury, neurological assessment (including
that the group with the most severe cardiovascular severity and level of injury), cardiovascular
41

parameters, and clinical history predating the A total of seven cases with spinal cord injury
spinal cord injury, for example, pre-existing (two females and five males, aged 31–82 years with
cardiovascular disease. Detailed information on a mean of 60.0 years), and five individuals with
cardiovascular parameters was collected during intact central nervous system (two females and
the acute stage of injury in all individuals. We also three males, aged 30–73 years with a mean of 51.4
searched for evidence of episodes of autonomic years) were analyzed. Individuals from the control
dysreflexia in these individuals. Heart rate and group were comparable to the cord injury group
blood pressure had been evaluated in each patient with regard to age (P ¼ 0.42) and gender
hourly for the first 2 weeks and then every 2 h until (P ¼ 1.0). All spinal cord-injured individuals had
discharge. Daily averages were calculated from a cervical injury. Neurological evaluation using the
all measurements available for each day in the ASIA scale showed that individuals in Group 1
patients’ charts during a 5-week post-injury period. had a more severe cord injury (Table 1).
These cases of cord injury all had a cervical injury
and were assigned to one of the two groups: cases Cardiovascular parameters
that developed severe cardiovascular dysfunction
during the acute post-injury period (Group 1), and There were significant differences in the cardio-
cases with no or minor cardiovascular dysfunction vascular parameters between individuals in
in this period (Group 2). The control group Groups 1 and 2. Severe hypotension, bradycardia,
(Group 3) included five cases with intact central and episodes of autonomic dysreflexia, which are
nervous system. Neurological evaluation of the signs of disrupted supraspinal cardiovascular con-
severity of cord injury was conducted through as- trol, were prominent among the cases in Group 1.
sessment of motor and sensory impairments ac- Severe hypotension (neurogenic shock) in the early
cording to the ASIA scoring system (Maynard Jr. post-injury period required the administration of
et al., 1997). The ASIA Grade A represents the vasopressive agents to all individuals in Group 1.
most severe, complete injury with complete motor Intravenous dopamine was administered, on aver-
and sensory impairment, and ASIA Grade D age, for 774.1 days (1–19 days) in this group. In
characterizes minor, incomplete cord injury (mild contrast, only one individual in Group 2 required
motor dysfunction and no sensory loss). infusion of dopamine for a period of 11 h. After

Table 1. Clinical and neurological data in spinal cord-injured individuals with severe (Group 1) or minor cardiovascular dysfunction
(Group 2) and in control cases (Group 3)

Groups Cases Gender Age (years) ASIA grade Level of SCI Cause of SCI Time from SCI to death

1 1 Female 31 A C 2,3 Diving accident 9 months


2 Male 66 B C 4,5 Fall 6 months
3 Female 43 A C 5,6 Motor vehicle accident 3.5 months
4 Male 66 A C 6,7 Spontaneous epidural hemorrhage 5 months
2 5 Male 65 C C 1,2 Motor vehicle accident 36 months
6 Male 67 A C 2,3 Fall 3.5 months
7 Male 82 B C 5,6 Spontaneous epidural hemorrhage 5 weeks
3 8 Male 30 N/A N/A N/A N/A
9 Male 53 N/A N/A N/A N/A
10 Female 37 N/A N/A N/A N/A
11 Male 73 N/A N/A N/A N/A
12 Female 64 N/A N/A N/A N/A

N/A, not applicable.


Adapted from Furlan et al. (2003), with permission from J. Neurotrauma.
42

the first 2 days, the cases in Group 2 had normal for examination. In Group 3, the third thoracic
arterial pressures, no bradycardia and no episodes segment was examined. Two sets of alternate spi-
of autonomic dysreflexia. Although individuals in nal cord sections (5–8 mm) were obtained and
Group 1 were treated with vasopressor agents in stained for: (1) general histology and myelin pres-
the early stage of cord injury, their systolic and ervation using hematoxylin and eosin and luxol
diastolic blood pressures were significantly lower fast blue; and (2) axonal preservation using
than those of Group 2 during the first 4 weeks post immunocytochemical staining for neurofilament
injury. Individuals in Group 1, and some of Group 200 (Sigma, 1:200).
2 also had bradycardia during the first 2 days after Sections were viewed with bright field illumina-
injury. Then, the mean daily heart rate in Group 1 tion (Axioscope, Zeiss), and the extent of injury
remained significantly lower than the heart rate in and axonal degeneration was examined using the
Group 2 for the following 4 weeks after injury. By Northern Eclipse imaging software (Version 6.0,
the end of the 4th week, the cases in Group 1 had Empix Imaging Inc). In sections stained for myelin
recovered cardiovascular function and the two with luxol fast blue, the total area of demyelina-
groups did not differ with respect to heart rate and tion (Fig. 2, pink areas within the white matter)
blood pressure. was measured and presented as a fraction (percent
Three individuals in Group 1 (Cases 1, 3, and 4 and standard error of the mean) of the total sur-
of Table 1) developed episodes of autonomic face area of the spinal cord section (including
dysreflexia during their stay at the hospital. A white and gray matter). In sections stained for ne-
typical episode of dysreflexia occurred in Case 1 at urofilament 200, axonal counts were conducted in
day 4. This was her first episode of dysreflexia. selected areas of white matter (see below).
During this episode her systolic and diastolic blood Axonal preservation was examined within the
pressures reached 180 and 100 mmHg, respectively, two areas that have been previously suggested to
from a resting mean pressure of 90 mmHg. This contain descending vasomotor pathways: the dor-
episode was accompanied by an increased heart sal aspects of the lateral funiculus (Area I in Fig.
rate to 80 beats per minute (from a resting rate of 1B), and the white matter adjacent to the dorso-
55 beats per minute). Pounding headache, double lateral portions of the intermediolateral column
vision and anxiety were the major complaints (Area II in Fig. 1B) (Kerr and Alexander, 1964;
during this episode. Elevation of the head and Foreman and Wurster, 1973; Lebedev et al., 1986;
analgesic medication were effective in managing Nathan and Smith, 1987). We also examined the
this episode. No episodes of autonomic dysreflexia extent of axonal preservation within the dorsal
were reported in individuals in Group 2. Pre- columns and the corticospinal tracts in each case
existing systemic hypertension was established in (Fig. 3). Sections stained for neurofilament 200
Case 4 (Group 1) and Case 5 (Group 2) prior to were examined under low magnification (  1.25)
spinal cord injury. Although daily recording of and areas of interests were identified (Fig. 3). For
cardiovascular parameters was not performed in each section, using high magnification (  20) im-
individuals from Group 3, we found no clinical ages, at least three representative fields from the
data to suggest cardiovascular abnormalities. chosen areas were examined (Fig. 3a–3d). Finally,
the axonal counts were conducted using the
Northern Eclipse software. Axonal preservation
Histopathological findings was expressed as the mean number of preserved
axons per 10,000 mm27SEM.
The spinal cord tissue from the cases included in Cross-sectional analysis of spinal cord sections
this study was fixed with 10% buffered formalin from the high thoracic cord, caudal to the injury
for 2 weeks and paraffin embedded. In no case did site, revealed that the extent of white matter de-
the postmortem interval exceed 24 h. In each cord generation was 24.6572.1% (range: 19.29–29.66%)
injury case, at least one segment caudal to the level in Group 1, and 8.271.2% (range: 6.06–10.15%)
of injury (upper thoracic segments) was selected in Group 2 (Fig. 2). This showed that spinal cords
43

Fig. 2. Myelin staining with Luxol Fast Blue of spinal cord sections from the high thoracic spinal cord. (A) Spinal cord injury case who
developed severe cardiovascular complications (Group 1); (B) spinal cord injury case with no significant cardiovascular dysfunction
(Group 2); and (C) individual with intact CNS (control case, Group 3). Calibration bar is 2 mm. A well-defined butterfly shaped area of
the gray matter is present in all sections. Myelin-containing white matter is stained blue. Areas of axonal degeneration and myelin loss
(pink areas within the white matter) are present in sections from Cases 3 and 7. (D) Average values of white matter degeneration
(expressed as a percent of total spinal cord area) in spinal cord sections from individuals in Group 1 were significantly greater than
those of individuals from Group 2. (From Furlan et al. (2003), with permission from J. Neurotrauma.)

from cases of Group 1 with severe cardiovascular similar (P ¼ 0.167). There were fewer preserved
dysfunction after cord injury had more extensive axons per 10,000 mm2 in Area I (dorsal aspects of
areas of white matter degeneration than spinal the lateral funiculus) in individuals from Group 1
cords from cases who had no or minor cardiovas- (2075) than in individuals from Group 2 (52715;
cular dysfunction after injury (Po0.002). P ¼ 0.029) and Group 3 (6573; Po0.001). Also,
Axons within the spinal cord were unequiv- the number of preserved axons within the Area I in
ocally identified in spinal cord sections using Group 2 was significantly less than in Group 3
immunohistochemistry for neurofilament 200 and (P ¼ 0.034).
bright field microscopy (Fig. 3B). In control cases, The number of preserved axons per 10,000 mm2
axons were evenly distributed throughout the white within Area II (white matter adjacent to the
matter. However, there was a striking difference in dorsolateral aspects of the intermediolateral cell
axonal preservation in different regions of the spi- column) in Group 1 (5977) was significantly re-
nal cord from individuals with spinal cord injury. duced in comparison with Group 2 (93713;
The number of preserved axons per 10,000 mm2 P ¼ 0.028) and Group 3 (109715; P ¼ 0.013).
within the corticospinal tract in Group 1 (2175) There were no significant differences in axonal
was significantly lower than in Group 2 (88712; counts within Area II between Groups 2 and 3
Po0.001) and Group 3 (117714; Po0.001). (P ¼ 0.357).
There was no significant difference between the
number of axons within the corticospinal tract of Discussion
individuals from Groups 2 and 3 (P ¼ 0.184).
The number of preserved axons per 10,000 mm2 Previous investigations have demonstrated that
within the dorsal column in Group 1 (100713), hypotension, bradycardia, and autonomic dysre-
Group 2 (131714), and Group 3 (136714) was flexia occur more frequently in individuals with
44

Fig. 3. (A) Staining of axons by immunocytochemistry for neurofilament 200 (NF200) in low power (  1.25) photomicrograph of a
spinal cord from a cord-injured individual with severe cardiovascular dysfunction (Case 4). Calibration bar is 1 mm. The squares
indicate the areas of the spinal cord in which preserved axons were counted. The four areas examined in this study for axonal
preservation were the following: dorsal column (DC), Area I, lateral corticospinal tracts (CST), and Area II. (B) High magnification
(  20) of different areas of the spinal cord stained with NF200 from three representative cases, one from each of the groups. Brown-
stained dots represent cross-sections of spinal axons immunocytochemically identified with NF200. There was a significant axonal loss
within Area 1 (panel b-1) and the CST (panel c-1) in all individuals from Group 1. (From Furlan et al. (2003), with permission from
J. Neurotrauma.)

severe cervical spinal cord injury (Lehmann et al., and the difference in incidence and severity of car-
1987; Mathias and Frankel, 1992; Noreau et al., diovascular symptoms in the two groups. In addi-
2000; Silver, 2000). This study has demonstrated a tion, the histology shows that the axon loss was
relationship between the location and severity of not homogeneously distributed across the area of
pathology in the spinal cord and cardiovascular the sections. The dorsal columns had very little
dysfunction in human cases of spinal cord injury. axon loss. Area I and the corticospinal tract
The histological analysis demonstrates that Group showed the greatest loss. Area II had an interme-
1, with significant cardiovascular dysfunction, had diate amount of loss.
greater myelin and axon loss than the cases in We expected that the severe cardiovascular
Group 2, who had insignificant cardiovascular symptoms of Group 1 would be associated with
dysfunction. By these criteria, injury was more se- a very large axon loss in the area traversed by the
vere in Group 1 than in Group 2. This matches the descending vasomotor pathways. Area I had a loss
higher ASIA grade for Group 1 than for Group 2, of 70% of axons in Group 1 cases, whereas this
45

area in Group 2 was decreased by 20%. In con- within the human spinal cord (Bunge et al., 1993;
trast, Group 1 lost 20% of axons in Area II vs. the Hayes and Kakulas, 1997; Puckett et al., 1997;
loss of 15% by Group 2, changes that were rela- Kakulas, 1999). Moreover, some histopathological
tively similar. Accordingly, the cardiovascular dys- findings in humans are significantly different from
function in Group 1 and the lack of dysfunction in those observed in animal models of cord injury
Group 2 correlate best with the axonal losses in (Puckett et al., 1997). Therefore, to extrapolate
Area I. Thus, Area I, the dorsal area of the dorso- information from animal models to human disor-
lateral funiculus, seems a more likely candidate ders, it is essential to compare findings from an-
than the more ventral Area II as the site of the imal and human studies.
cardiovascular pathways. This view is consistent Using cervical electrical stimulation and selec-
with previous work by Fehlings and Tator (1995) tive lesions, numerous investigators have reported
demonstrating a relationship between loss of func- that the descending vasomotor pathways are lo-
tion and axonal loss after spinal cord injury (SCI). calized within extensive areas from ventral to dor-
They showed that inclined plane score varied log- sal in the peripheral aspects of the lateral funiculus
arithmically with number of axons at the injury in cats and monkeys (Kerr and Alexander, 1964;
site, such that loss of greater than 50% of axons is Illert and Gabriel, 1972; Foreman and Wurster,
required for a significant drop in neurological 1973). Barman and Wurster (1975) demonstrated
function. that the descending sympathetic pathways are
These observations contradict the conclusions situated on the surface of the dorsolateral
reached by Nathan and Smith (1987) that, in funiculus and are organized in a dorsal-to-ventral
humans, the descending vasomotor pathways are manner based on electrical stimulation in dogs.
localized to the white matter adjacent to the Lebedev et al. (1986) carried out an electrophys-
dorsolateral aspect of the intermediolateral cell iological study before and after the dorsolateral
column (Area II of the present study). These funiculus transection showing that descending
conclusions were based on the analysis of vasomotor pathways are situated within the
postmortem spinal cord sections from patients dorsal parts of the lateral funiculus in cats, the
who underwent antero-lateral cordotomies for area which corresponds to Area I in our
control of intractable pain, and subsequently, investigation.
developed cardiovascular symptoms such as The knowledge of the area greatly responsible
hypotension and orthostatic intolerance. However, for vasomotor control has significant relevance to
careful analysis of their data showed that the recovery from spinal cord injury. Area I is su-
antero-lateral cordotomy resulted also in partial perficial and easily accessible from the dorsolat-
destruction of the lateral funiculus (Area I of the eral surface of the spinal cord. It is conceivable
present study). In other words, the cases in the that topical treatments could be applied to this
report by Nathan and Smith (1987) had damage in region. Thus, an avenue for future studies is an
both areas examined in the present study. There- exploration of techniques for delivering treatment
fore, it cannot be excluded that the relevant to this region. An additional question, raised by
damage was to Area I. this study, is whether it is possible to distinguish
Much of our present understanding of the anatomically, within Area I, the region of faci-
pathophysiology of central nervous system litatory pathways responsible for the maintenance
(CNS) disorders, including spinal cord injury, is of blood pressure and the orthostatic tolerance,
based on extrapolations from animal models from the location of inhibitory pathways that
(Krassioukov and Weaver, 1996; Krenz and limit the spinal reflexes responsible for dysreflexia.
Weaver, 1998; Maiorov et al., 1998; Osborn This investigation documents that anatomical
et al., 1989). Although considerable clinical data studies of cases of spinal cord injury can provide
are available on neurological function after human crucial information that may assist with treatment
spinal cord injury, only a limited number of stud- of the disabling cardiovascular consequences of
ies have been directed to histopathological changes spinal cord injury.
46

Acknowledgments Furlan, J.C., Fehlings, M.G., Shannon, P., Norenberg, M.D.


and Krassioukov, A.V. (2003) Descending vasomotor path-
This study was conducted with the support of a ways in humans: correlation between axonal preservation
and cardiovascular dysfunction after spinal cord injury.
Christopher Reeve Paralysis Foundation grant J. Neurotrauma, 20(12): 1351–1363.
(KB2-0003-1), a Cervical Spine Research Society Gao, S.A., Ambring, A., Lambert, G. and Karlsson, A.K.
grant, support from the Canadian Syringomyelia (2002) Autonomic control of the heart and renal vascular bed
Network, and a grant from the Heart and Stroke during autonomic dysreflexia in high spinal cord injury. Clin.
Foundation of Ontario (NA4951) awarded to Auton. Res., 12(6): 457–464.
Hayes, K.C. and Kakulas, B.A. (1997) Neuropathology of hu-
Dr. A. Krassioukov. Dr. J. Furlan (Toronto, man spinal cord injury sustained in sports-related activities.
ON) was a postdoctoral fellow who conducted a J. Neurotrauma, 14(4): 235–248.
major part of the histopathological analysis. The Illert, M. and Gabriel, M. (1972) Descending pathways in the
author also would like to acknowledge Dr. A. cervical cord of cats affecting blood pressure and sympathetic
Marcillo (Miami, FL), Mrs. Lorraine Yamamoto activity. Pflugers Arch., 335: 109–124.
Kakulas, B.A. (1999) A review of the neuropathology of human
(Burlington, ON), and Mrs. Lynda Rickards, R.N.
spinal cord injury with emphasis on special features. J. Spinal
(Toronto, ON) for their assistance and support Cord Med., 22: 119–124.
during the project. Karlsson, A.K. (1999) Autonomic dysreflexia. Spinal Cord, 37:
383–391.
Karlsson, A.K., Friberg, P., Lonnroth, P., Sullivan, L. and
Elam, M. (1998) Regional sympathetic function in high
References spinal cord injury during mental stress and autonomic
dysreflexia. Brain, 121: 1711–1719.
Atkinson, P.P. and Atkinson, J.L.D. (1996) Spinal shock. Kerr, F.W.L. and Alexander, S. (1964) Descending autonomic
Mayo Clin. Proc., 71: 384–389. pathways in the spinal cord. Arch. Neurol., 10: 249–261.
Barman, S.M. and Wurster, R.D. (1975) Visceromotor organ- Krassioukov, A.V., Furlan, J.C. and Fehlings, M.G. (2003)
ization within descending spinal sympathetic pathways in the Autonomic dysreflexia in acute spinal cord injury: an under-
dog. Circ. Res., 37: 209–214. recognized clinical entity. J. Neurotrauma, 20(8): 707–716.
Bunge, R.P., Puckett, W.R., Becerra, J.L., Marcillo, A. and Krassioukov, A.V. and Weaver, L.C. (1996) Morphological
Quencer, R.M. (1993) Observation on the pathology of hu- changes in sympathetic preganglionic neurons after spinal
man spinal cord injury. A review and classification of 22 new cord injury in rats. Neuroscience, 70: 211–226.
cases with details from a case of chronic cord compression Krenz, N.R. and Weaver, L.C. (1998) Sprouting of primary
with extensive focal demyelination. In: Seil F.J. (Ed.) Ad- afferent fibers after spinal cord transection in the rat. Ne-
vances in Neurology, Vol. 59. Raven Press Ltd., New York. uroscience, 85: 443–458.
Cariga, P., Catley, M., Mathias, C.J., Savic, G., Frankel, H.L. Lebedev, V.P., Krasyukov(Krassioukov), A.V. and Nikitin,
and Ellaway, P.H. (2002) Organisation of the sympathetic S.A. (1986) Electrophysiological study of sympathoexcitatory
skin response in spinal cord injury. J. Neurol. Neurosurg. structures of the bulbar ventrolateral surface as related to
Psychiat., 72(3): 356–360. vasomotor regulation. Neuroscience, 17(1): 189–203.
Clydon, V., Elliott, S., Sheel, W. and Krassioukov, A.V. (2005) Lehmann, K.G., Lane, J.G., Piepmeier, J.M. and Batsford,
Incidence of cardiac dysrhythmias during sperm retrieval in W.P. (1987) Cardiovascular abnormalities accompanying
men with spinal cord injury. Spinal Cord (submitted). acute spinal cord injury in humans: incidence, time course
Curt, A., Nitsche, B., Rodic, B., Schurch, B. and Dietz, V. and severity. J. Am. Coll. Cardiol., 10(1): 46–52.
(1997) Assessment of autonomic dysreflexia in patients with Maiorov, D.N., Fehlings, M.G. and Krassioukov, A.V. (1998)
spinal cord injury. J. Neurol., Neurosurg. Psychiat., 62: Relationship between severity of spinal cord injury and ab-
473–477. normalities in neurogenic cardiovascular control in conscious
Fehlings, M.G. and Tator, C.H. (1995) The relationships rats. J. Neurotrauma,, 15(5): 365–374.
among the severity of spinal cord injury, residual neurolog- Mathias, C.J. (1995) Orthostatic hypotension: causes, mecha-
ical function, axon counts and counts of retrogradely labeled nisms, and influencing factors. Neurology, 45(Suppl 5):
neurons after experimental spinal cord injury. Exp. Neurol., S6–S11.
132: 220–228. Mathias, C.J. and Frankel, H.L. (1992) The cardiovascular
Figoni, S.F. (1984) Cardiovascular and haemodynamic re- system in tetraplegia and paraplegia. In: Frankel H.L. (Ed.)
sponses to tilting and to standing in tetraplegic patients. Handbook of Clinical Neurology, Vol. 17(25). Elsevier,
Paraplegia, 22: 99–109. Amsterdam, pp. 435–456.
Foreman, R.D. and Wurster, R.D. (1973) Localization and Maynard Jr., F.M., Bracken, M.B., Creasey, G., Ditunno Jr.,
functional characteristics of descending sympathetic spinal J.F., Donovan, W.H., Ducker, T.B., Garber, S.L., Marino,
pathways. Am. J. Physiol., 225(1): 212–217. R.J., Stover, S.L., Tator, C.H., Waters, R.L., Wilberger, J.E.
47

and Young, W. (1997) International standards for neurolog- Puckett, W.R., Hiester, E.D., Norenberg, M.D., Marcillo, A.E.
ical and functional classification of spinal cord injury. Amer- and Bunge, R.P. (1997) The astroglial response to Wallerian
ican Spinal Injury Association. Spinal Cord, 35(5): 266–274. degeneration after spinal cord injury in humans. Exp. Ne-
Nathan, P.W. and Smith, M.C. (1987) The location of de- urol., 148(2): 424–432.
scending fibres to sympathetic preganglionic vasomotor and Reis, D.J., Morrison, S. and Ruggiero, D.A. (1988) The C1 area
sudomotor neurons in man. J. Neurol., Neurosurg. Psychiat., of the brainstem in tonic and reflex control of blood pressure.
50: 1253–1262. Hypertension, 11: I8–I13.
Noreau, L., Proulx, P., Gagnon, L., Drolet, M. and Laramee, Ruggiero, D.A., Cravo, S., Arango, V. and Reis, D.J. (1989)
M.T. (2000) Secondary impairments after spinal cord injury: Central control of the circulation by the rostral ventrolateral
a population-based study. Am. J. Phys. Med. Rehabil., 79(6): reticular nucleus: anatomical substrates. Prog. Brain Res., 81:
526–535. 49–79.
Osborn, J.W., Taylor, R.F. and Schramm, L.P. (1989) Deter- Silver, J.R. (2000) Early autonomic dysreflexia. Spinal Cord,
minants of arterial pressure after chronic spinal transection in 38: 229–233.
rats. Am. J. Physiol., 256: R666–R673. Teasell, R., Arnold, A.P., Krassioukov, A.V. and Delaney,
Piepmeier, J.M., Lehmann, K.B. and Lane, J.G. (1985) Cardi- G.A. (2000) Cardiovascular consequences of loss of supra-
ovascular instability following acute cervical spinal cord spinal control of the sympathetic nervous system following
trauma. Cent. Nerv. Syst. Trauma, 2(3): 153–160. spinal cord injuries. Arch. Phys. Med. Rehabil., 81: 506–516.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Published by Elsevier B.V.

CHAPTER 4

Disordered control of the urinary bladder after


human spinal cord injury: what are the problems?

Patrick J. Potter

Regional Spinal Cord Injury Rehabilitation Program, and Physical Medicine and Rehabilitation,
St. Joseph’s Health Center, The University of Western Ontario, London, ON, Canada

Abstract: Spinal cord injury has a profound impact on the storage and voiding functions of the urinary
bladder. Loss of autonomic and somatic control mechanisms leads to hypo- or hyperactivity of the bladder
wall and sphincters causing problems that range from incontinence to complete loss of the capacity to
empty the bladder. This chapter outlines the types of bladder dysfunction that occur after spinal cord
injury, their relative prevalence and current practices used to manage the problems. With all the inter-
ventions that are available, management of bladder function often still remains a compromise, as the
medications and physical interventions available may stimulate or block components of the voiding reflex,
but are often not fully restorative in this effort.

Introduction and autonomic functions usually do not recover


completely (Menter and Hudson, 1995; Wolfe
Urinary bladder control is dependent upon coor- et al., 2002; Potter et al., 2004). When only
dinated interaction between the somatic and au- partial resolution of the impairment in bladder
tonomic nervous systems (Burns et al., 2001; de control occurs, then accommodation to the altered
Groat and Yoshimura, 2001). When the supraspi- physiological condition requires interventions that
nal, coordinated control of these systems is lost, range from pharmacological treatments to appli-
the resulting bladder dysfunction is termed ‘‘ne- ances and mechanical or electrical devices, as pre-
urogenic bladder impairment’’ or a neurogenic sented in the following chapters. For previous
bladder. The presence of a neurogenic urinary reviews of the neurogenic bladder after spinal cord
bladder is extremely common after spinal cord in- injury see Perkash (2004) and Burns et al. (2001).
jury (Waites et al., 1993; Cardenas and Hooton,
1995; Vines, 1996; Sapounzi-Krepia et al., 1998;
Stover et al., 1989; Chen et al., 1999; Wolfe et al., Brief overview of normal bladder function
2002). In general, this dysfunction is not unique to
spinal cord injury as it can arise from disrupted The urinary bladder is a fluid reservoir that nor-
peripheral or central nervous system control. For mally empties completely in a well-controlled man-
example, a neurogenic bladder can occur in dis- ner. All of the clinical problems encountered after
eases such as multiple sclerosis (Anderson et al., spinal cord injury are manifestations of impairment
1976). Although some neurological recovery may in these two basic functions of the bladder: storage
occur following a spinal cord injury, motor, sensory and emptying. The lower urinary tract is made up
of the bladder, internal sphincter, external sphinc-
Corresponding author. Tel.:+519 685 4080; ter and urethra. The bladder wall is composed of
Fax: +519 685 4081; E-mail: patrick.potter@sjhc.london.on.ca smooth muscle, termed the detrusor muscle, and

DOI: 10.1016/S0079-6123(05)52004-1 51
52

has a base or trigone, a body and a neck. Para- spinal cord and in the bladder lead to malfunctions
sympathetic control of the detrusor muscle origi- in these reflexes and in the storage and voiding
nates from preganglionic axons in the pelvic nerve functions of the urinary bladder.
with cell bodies in the 2nd to 4th sacral (S) spinal
cord segments. Detrusor contraction is mediated
Clinical presentations of bladder dysfunction after
primarily by parasympathetic stimulation. Sympa-
spinal cord injury
thetic control of this muscle comes from the hypo-
gastric nerve that contains axons of preganglionic
Neurogenic bladder impairments depend greatly
neurons that are located in the 10th thoracic (T10)
upon the level and extent of central nervous system
to 2nd lumbar (L2) spinal cord segments. Sensory
injury. The lower levels of injury, such as at the level
innervation of the bladder wall travels to the S2–S4
of the conus medullaris, are more likely to result in
spinal segments via the pelvic nerve. The internal
a flaccid bladder. Thoracic and cervical level inju-
bladder sphincter is made of smooth muscle and is
ries commonly generate mixed pictures of detrusor
located at the junction of the bladder neck and
hyperactivity, sphincter spasticity and lack of coor-
urethra. This sphincter receives parasympathetic
dination between the detrusor and sphincters that is
and sympathetic innervation like that of the de-
termed dyssynergia. Because the urinary bladder is
trusor. However, in this muscle, sympathetic stim-
innervated bilaterally, hemi-cord impairments such
ulation causes contraction. The external sphincter
as the Brown Sequard Syndrome often do not result
is striated muscle that surrounds the urethra and is
in significant bladder dysfunction. Furthermore, in-
controlled by somatic innervation from the S2–S4
juries that spare the central portion of the cord re-
spinal segments that reach this sphincter via the
sult in relative sparing of bladder function.
pudendal nerve. Details of the anatomy and phar-
Issues of bladder dysfunction relate to four major
macology of these pathways are presented in later
problems: (1) inadequate or excessive detrusor
chapters and in reviews by de Groat and
function, (2) inadequate or excessive sphincter func-
Yoshimura (2001) and Burns et al. (2001).
tion, (3) dyssynergy between detrusor and sphincter
To promote the storage function of the urinary
actions and (4) impaired ability to sense the bladder
bladder, sympathetic innervation plays two key
(Lisenemayer and Oakley, 2003). Approaches to
roles. First, through a-adrenergic receptors, the
treatment can therefore be based on manipulation
neck and internal sphincter of the bladder are con-
of these functions. Often combinations of ap-
tracted to close the bladder outlet (Ek et al., 1977).
proaches are required and the type of bladder man-
Next, via b-adrenergic receptors, the body of the
agement may change through a cord-injured
bladder relaxes. Normally, filling of the bladder
person’s life. For example, during acute care im-
occurs with minimal increases in pressure. Voiding
mediately after injury, a Foley catheter is often in-
the bladder is a coordinated process that involves
serted to drain the flaccid bladder. As some degree
contraction of the detrusor muscle with concomi-
of continence develops with time after injury, this
tant relaxation of the striated muscle of the urethra
approach would likely change to intermittent cath-
and pelvic floor and relaxation of internal and ex-
eterization during rehabilitation. After discharge
ternal sphincters. This requires integrated control
from the hospital, some people who are able to re-
from pontine centers in the brain and sacral spinal
gain an active lifestyle, even including participation
neurons (see de Groat and Yoshimura, 2001).
in sports, rely on condom drainage into a leg bag
When the bladder volume increases, sensory input
that obviates the need for strictly timed procedures
from the bladder wall to the sacral spinal neurons
such as intermittent catheterization.
increases until the threshold for the micturition re-
flex is reached and reflex voiding can be initiated.
This process in the able-bodied person is under Inadequate detrusor function
voluntary control and is accomplished by well-
regulated and integrated autonomic and somatic Inadequate contraction of the detrusor muscle
reflexes. After spinal cord injury, changes in the is often associated with spinal cord injuries that
53

impair the distal conus medullaris region of the of the detrusor muscle is extremely variable be-
spinal cord. These very low injuries mimic lower tween individuals. Clinical approaches to treat this
motor neuron impairment such as found in problem include anticholinergic (anti-muscarinic)
peripheral neuropathies, resulting in absent or sig- medication if excessive intravesical pressures pre-
nificantly decreased detrusor contraction. There- vail. If the detrusor muscle cannot be relaxed ad-
fore the approach to remedy this problem is to equately with such medication to provide
augment emptying. Such augmentation can be in continence between intermittent catherizations,
the form of cholinergic muscarinic receptor stim- an indwelling catheter or attached device such as
ulation such as the oral administration of bethane- a condom catheter is necessary. If, due to lack of
col. Bethanecol must be taken every 4–6 h, and a sensation, voiding cannot be managed effectively
bladder response occurs 30 min to 1 h after taking or conveniently, an external appliance (condom
the drug. Therefore, drug use has to be timed drainage) may be used in males. However similar
to coordinate with bladder fullness. Mechanical devices are notoriously difficult to maintain in fe-
techniques used by some include increasing intra- males, resulting, instead, in the use of an indwell-
abdominal pressure with external mechanical pres- ing Foley or suprapubic (inserted through the
sure such as the Credé maneuver that utilizes for- lower abdominal wall) catheter. Decreasing de-
ward flexion over the subject’s hand as it presses trusor contractions may also be accomplished by
into the abdomen to facilitate voiding. Often emp- chemically blocking C-fiber bladder afferent ne-
tying with this procedure is incomplete, and to urotransmission with intravesical vanilloids such
prevent urinary tract infections from occurring as as capsaicin or resiniferatoxin or by intravesical
a consequence of the residual urine in the bladder, administration of anticholinergics. Another in-
the Credé maneuver may be combined with one travesical approach under investigation is injec-
catheterization per day to empty the bladder com- tion of botulinum toxin into the detrusor muscle to
pletely. This procedure is not successful in people cause relaxation (Reitz et al., 2004). Intravesical
with detrusor–sphincter dyssynergia as it also can administration of medication is more invasive than
cause contraction of the sphincters, blocking the oral medications but does offer options when oral
outflow of urine (Chancellor et al., 1990). The anticholinergic drugs are not effective. When an
most common approach to the management of an intravesical route is used for treatment, effects are
inadequate detrusor response is intermittent cath- temporary and repeated treatments are necessary.
eterization. In some cases of significantly de- The ideal time frames for repeated intravesical
creased detrusor function, spontaneous detrusor drug administration are not well established. More
contractions may occur that fail to empty the invasive approaches for reducing detrusor hyper-
bladder but are a cause of incontinence. In such activity include denervation procedures such as
cases, an anticholinergic (anti-muscarinic) drug sacral rhizotomy, a procedure that must be viewed
such as oxybutin and intermittent catheterization cautiously as it is irreversible. To address reduced
may be combined. bladder capacity due to detrusor hyperreflexia, the
bladder size and capacity may be increased by a
surgical augmentation cystoplasty using a piece
Excessive detrusor function of bowel.

Increased detrusor tone or spasticity (detrusor


hyperreflexia) is part of the upper motor neuron Inadequate sphincter function
syndrome. In this situation, the detrusor muscle is
considered to be ‘‘unstable,’’ contracting at lower Inadequate sphincter function, whether associated
bladder volumes and often producing excessive with inadequate or excessive detrusor function,
intravesical pressures. Detrusor hyperreflexia of- results in incontinence. Sphincter tone can be en-
ten, but not always, occurs with thoracic and cer- hanced by blocking muscarinic cholinergic recep-
vical cord injuries, and the extent of hyperactivity tors or by stimulating b-adrenergic receptors. Of
54

these two possible approaches, blocking mu- adrenergic antagonist as discussed above, and ap-
scarinic cholinergic receptors is usually the supe- plying a device for collection of urine such as a
rior, although combination therapy may be condom catheter. Alternatively, detrusor contrac-
utilized. In this condition, the goal is to restore tion may be blocked pharmacologically and blad-
the storage function of the bladder. Once this is der emptying accomplished by intermittent
accomplished, if the person cannot initiate void- catheterization. Often these approaches are only
ing, then emptying is done by intermittent cathe- partially successful and a compromise in bladder
terization. If continence cannot be maintained management is reached within the tolerance limits
with drugs, then voiding into an appliance such as for side effects of the medication. Either the person
a condom catheter is possible, and cholinergic voids more frequently, and experiences urgency, or
muscarinic receptor agonists such as bethanecol catheterizes more frequently. The most common
can be utilized to facilitate voiding. Operative ap- approach in males is to reduce sphincter tone and
proaches to enhance the usefulness of the bladder apply an external device.
neck in maintaining continence include surgically
modifying the bladder neck or implantation of an
Impaired ability to sense the bladder
artificial sphincter. The most common approach
for dealing with inadequate sphincter tone is to
At this time, 4-aminopyridine is the only pharma-
enhance the contraction pharmacologically and
cological agent that has been demonstrated to en-
utilize intermittent catheterization for bladder
hance electrical conduction in the spinal cord,
emptying.
enhancing sensation of bladder contraction and
fullness in some individuals (Potter et al., 1998). In
Excessive sphincter tone the absence of such sensation, management must
be accomplished by systems that continuously
a-2 receptor sympathetic adrenergic blockade is drain the urine such as condom drainage, indwell-
the mainstay of pharmacological management of ing catheters (suprapubic or Foley) or diapers or
the contracted bladder neck, to allow emptying in methods that employ timed, regular emptying such
the presence of excessive sphincter tone. Originally as intermittent catheterization.
developed to treat hypertension, this group of
medications has evolved, through several genera-
Infection can be a consequence of all
tions, to a family of drugs that can be taken once
management systems
per day and that have infrequent and less severe
side effects such as hypotension. Other approaches
Infection is a problem, secondary to almost all
to the spastic sphincter include sphincterotomy
methods of managing the neurogenic bladder after
and pudendal nerve section.
spinal cord injury (Bennett et al., 1995; Stover
et al., 1989; Esclarin De Ruz et al., 2000). As the
Detrusor-sphincter dyssynergia prevalence of resistant bacteria increases (Waites
et al., 2000; Siroky, 2002) commonly used, inex-
After spinal cord injury, the clinical presentation pensive antibiotics become ineffective. The fre-
of a person can be an inability to empty the blad- quency of urinary tract infections may necessitate
der either spontaneously or by self-initiated void- antibiotic prophylaxis (Galloway, 1997; Waites
ing. With these symptoms, urodynamic studies are et al., 2001; Morton et al., 2002). Alternative prep-
required to ascertain whether the impairment arations such as cranberry juice that contribute to
stems from inadequate detrusor function, exces- maintenance of the integrity of the bladder urot-
sive sphincter activity or dyssynergy between the helium as a barrier to bacteria then become more
two muscle groups. Approaches to treating dyssy- important considerations for long-term prophy-
nergia usually involve decreasing bladder neck re- laxis (Reid et al., 2003). For the cord-injured per-
sistance with a drug such as an a-2 receptor son, often the most sought after management
55

strategy is that which, in their mind, most mimics strictures, bladder diverticuli, chronic cystitis and
‘‘normal function’’ (Jamil, 2001). increased incidence of bladder cancer. These prev-
alence studies reveal that, although we are well
aware of the high incidence of neurogenic bladder,
Incidence and prevalence of urinary bladder we are still limited in our ability to manage its
dysfunction after cord injury consequences. For example, incomplete emptying
is associated with high residual urine volume,
Most people, during the first days after spinal cord which is a risk factor for incontinence and
injury, have evidence of a neurogenic bladder. In infection (Shekelle et al., 1999; Trautner and
people with incomplete injury, the majority of re- Darouiche, 2002). Recognizing that continence is
covery of bladder function is evident in the first the first issue associated with a neurogenic bladder,
6–9 months and improvement can continue for pain and infection are equally important long-term
2 years after injury. The negative consequences of sequelae (Post et al., 1998). Although continence
the neurogenic bladder to the health and quality of may be controlled with devices, sepsis, pain and
life for cord-injured people are decreasing with incontinence may result from recurrent urinary
current improvements in management and under- tract infections.
standing of the causes of the problems. In one of
the early papers on urological aspects of rehabil-
itation, Bors (1951), described up to 80% mortal- Conclusion
ity of spinal cord injured soldiers in World War I,
before they were able to return to the United The consequences of spinal cord injury to the
States. By the time of World War II the survival function of the urinary bladder are severe and play
rate had increased to 88%. Bors attributed this a serious role in the health and well-being of the
improvement to greater understanding of the cord-injured person for life. For the bladder to be
pathophysiology of the neurogenic bladder and an effective reservoir, we fully utilize reflex con-
the advent of antibiotics. Mortality due to uro- traction of the sphincter and reflex detrusor relax-
logical causes is now estimated to be o3% (Jamil, ation. For the bladder to empty, these processes
2001). The current focus of modern rehabilitation must be reversed. After minor impairments void-
medicine and research is directed toward issues of ing may still be possible but with greater effort,
morbidity and not mortality. The prevalence of a incontinence, incomplete emptying, increased fre-
neurogenic bladder after spinal cord injury is high quency, urgency or hesitancy. Given that we often
(Anson and Shepard, 1996; Noreau et al., 2000). A cannot fully reverse the effects of impaired neuro-
study by the Model Spinal Cord Injury Systems of logical control of bladder function, even with the
Care determined that 81% of persons with spinal extensive array of available medication, the most
cord injury reported some degree of impaired effective approach to the management of ne-
bladder function (McKinley et al., 1999). Even urogenic bladder remains to find the best balance
more significant are the secondary sequelae, in- between a person’s need for emptying their blad-
cluding frequent urinary tract infections, pain sec- der, their tolerance for medication, assistive de-
ondary to urinary tract infection and pain vices and appliances, and the social consequences
secondary to indwelling devices such as Foley of maintaining continence (Stover et al., 1989;
catheters. Cardenas and Hooton, 1995, Liguori et al., 1997;
In a survey of Spinal Cord Injured persons Yavuzer et al., 2000; Boschen et al., 2003).
(Wolfe et al., 2002; Potter et al., 2004) regarding Although restoration of normal function is the
the long-term sequelae of spinal cord injury, uro- ultimate goal of research, development of superior
logical problems had a high prevalence. The ‘‘neu- management methods is a high priority as well.
rological impairment’’ of bladder function does Newer generations of pharmacological agents are
not appear to change with time, but time and ag- being developed to provide better therapeutic re-
ing result in secondary problems such as urethral sponses with less side effects. Using strategies such
56

as administering medications directly into the Esclarin De Ruz, A., Garcia Leoni, E. and Herruzo Cabrera, R.
bladder can minimize side effects of drugs. Meth- (2000) Epidemiology and risk factors for urinary tract infec-
ods for intermittent catheterization have been im- tion in patients with spinal cord injury. J. Urol., 164:
1285–1289.
proved by devices such as hydrophilic catheters Galloway, A. (1997) Prevention of urinary tract infection in
that can be inserted with much less friction than patients with spinal cord injury—a microbiological review.
conventional catheters (Hedlund et al., 2001; Spinal Cord, 35: 198–204.
Vapnek et al., 2003). Probiotic treatment, the in- Hedlund, H., Hjelmais, K., Jonsson, O., Klarslov, P. and
travesicular administration of healthy bacteria to Talja, M. (2001) Hydrophilic versus non-coated catheters for
prevent infection, may reduce the need for antibi- intermittent catheterization. Scand. J. Urol. Nephrol., 35:
49–53.
otics. Research must be directed toward finding a Jamil, F. (2001) Towards a catheter free status in neurogenic
cure for the bladder dysfunction after cord injury, bladder dysfunction: a review of bladder management op-
but also must address the important objective of tions in spinal cord injury. Spinal Cord, 39(7): 355–361.
providing better management of the neurogenic Liguori, P.A., Cardenas, D.D. and Ullrich, P. (1997) Social and
bladder. functional variables associated with urinary tract infections
in persons with spinal cord injury. Arch. Phys. Med. Rehab.,
78: 156–160.
Lisenemayer, T.A. and Oakley, A. (2003) Accuracy of individ-
References uals with spinal cord injury at predicting urinary tract infec-
tions based on their symptoms. J. Spinal Cord Med. Winter,
Anderson, J.T. and Bradley, W.E. (1976) Bladder and urethral 26(4): 352–357.
innervation in multiple sclerosis. Br. J. Urol., 48: 239–243. McKinley, W.O., Jackson, A.B., Cardenas, D.D. and De Vivo,
Anson, C.A. and Shepherd, C. (1996) Incidence of secondary M.J. (1999) Long-term medical complications after traumatic
complications in spinal cord injury. Int. J. Rehab. Res., 19: spinal cord injury: a regional model systems analysis. Arch.
55–66. Phys. Med. Rehab., 80(11): 1402–1410.
Bennett, C.J., Young, M.N. and Darrington, H. (1995) Differ- Menter, R.R. and Hudson, L.M. (1995) Effects of age at injury
ences in urinary tract infections in male and female spinal and the aging process. In: Stover S.L., DeLisa J.A. and
cord injury patients on intermittent catheterization. Paraple- Whiteneck G.G. (Eds.), Spinal Cord Injury Clinical Out-
gia, 33: 69–72. comes from the Model Systems. Aspen Publishers, Gait-
Bors, E. (1951) Urologic aspects of rehabilitation in SCI. hersburg, MD, pp. 272–288.
JAMA, 146: 225–229. Morton, S.C., Shekelle, P.G., Adams, J.L., Bennett, C.,
Boschen, K.A., Tonack, M. and Gargaro, J. (2003) Long-term Dobkin, B.H., Montgomerie, J. and Vickrey, B.G. (2002)
adjustment and community reintegration following spinal
Antimicrobial prophylaxis for urinary tract infection in per-
cord injury. Int. J. Rehab. Res., 6(3): 157–164.
sons with spinal cord dysfunction. Arch. Phys. Med. Rehab.,
Burns, A.S., Rivas, D.A. and Ditunno, J.F. (2001). The man-
83: 129–138.
agement of the neurogenic bladder and sexual dysfunction
Noreau, L., Proulx, P., Gagnon, L., Drolet, M. and Laramee,
after spinal cord injury, Spine, 26: S129–S136.
M.T. (2000) Secondary impairments after spinal cord injury:
Cardenas, D.D. and Hooton, T.M. (1995) Urinary tract infec-
a population-based study. Am. J. Phys. Med. Rehab., 79:
tion in persons with spinal cord injury. Arch. Phys. Med.
Rehab., 76: 272–280. 526–535.
Chancellor, M.B., Kaplan, S.A. and Blaivas, J.G. (1990) De- Perkash, I. (2004) Donald Munro Lecture 2003. Neurogenic
trusor-external sphincter dyssynergia. In: Block, G. and bladder: past, present and future. J. Spinal Cord Med., 27:
Whelan, J. (Eds.), Ciba Foundation Symposium. Vol. 51, 383–386.
Neurobiology of Continence. Wiley, Chichester, West Sus- Post, M.W., de Witte, L.P., van Asbeck, F.W., van Dijk, A.J.
sex, pp. 195–213. and Schrijvers, A.J. (1998) Predictors of health status and life
Chen, D., Apple Jr., D.F., Hudson, L.M. and Bode, R. (1999) satisfaction in spinal cord injury. Arch. Phys. Med. Rehab.,
Medical complications during acute rehabilitation following 79(4): 395–401.
spinal cord injury—current experience of the Model Systems. Potter, P.J., Hayes, K.C., Segal, J.L., Hsieh, J.T.C., Brunnermann,
Arch. Phys. Med. Rehab., 80(11): 1397–1401. S.R., Delaney, G.A., Tierny, D.S. and Mason, D. (1998)
de Groat, W.C. and Yoshimura, N. (2001) Pharmacology of the Randomized double-blind crossover trial of fampridine-SR
lower urinary tract. Annu. Rev. Pharmacol. Toxicol., 41: (sustained release 4-aminopyridine) in patients with incom-
691–721. plete spinal cord injury. J. Neurotrauma, 15(10): 837–849.
Ek, A., Alm, P., Andersson, K.E. and Persson, C.G. (1977) Potter, P.J., Wolfe, D.L., Burkell, T.A. and Hayes, K.C. (2004)
Adrenergic and cholinergic nerves of the human urethra and Challenges in educating individuals with spinal cord injury to
urinary bladder: a histochemical study. Acta. Physiol. reduce secondary conditions. Topics in Spinal Cord Injury
Scand., 99(3): 345–352. Rehab., 10: 30–40.
57

Reid, G., Potter, P.J., Lam, D., Warren, D., Borrie, M. and Vapnek, J.M., Maynard, F.M. and Kim, J. (2003) A prospec-
Hayes, K.C. (2003) Cranberry juice to reduce Biofilms and tive randomized trial of the LoFric hydrophilic coated cath-
infection in geriatric and spinal cord injured patients with eter versus conventional plastic catheter for clean
dysfunctional bladders. Natraceuticals & Food, 8: 23–28. intermittent catheterization. J. Urol., 169(3): 994–998.
Reitz, A., Stohrer, M., Kramer, G., Delpopo, G., Chartier- Vines, C.L. (1996) Identifying Secondary Conditions in Arkan-
Kastler, E., Panneck, J., Burgdofer, H., Gocking, K., sas with Spinal Cord Injuries. Arkansas Spinal Cord Com-
Madersbacher, H., Scchumacker, S., Richter, R., von Tobel, mission, Little Rock, AR.
J. and Schurch, B. (2004) European experience of 200 cases Waites, K., Chen, Y., De Vivo, M.J., Canupp, K.C. and Moser,
treated with botulinum-A toxin injections into the detrusor S.A. (2000) Antimicrobial resistance in gram negative bacte-
muscle for urinary incontinence due to neurogenic detrusor ria isolated from the urinary tract in community-residing
overactivity. Eur. Urol., 4: 510–515. persons with spinal cord injury. Arch. Phys. Med. Rehab.,
Sapounzi-Krepia, D., Soumilas, A., Papadakis, N., Sapkas, G., 81: 764–769.
Nomicos, J., Theodossopoulou, E. and Dimitriadou, A. Waites, K.B., Canupp, K.C., Chen, Y., De Vivo, M.J. and
(1998) Post traumatic paraplegics living in Athens: the impact Moser, S.A. (2001) Bacteremia after spinal cord injury in
of pressure sores and UTIs on everyday life activities. Spinal initial versus subsequent hospitalizations. J. Spinal Cord
Cord, 36: 432–437. Med., 24(2): 96–100.
Shekelle, P.G., Morton, S.C., Adams, J.L., Clark, K.A., Waites, K.B., Canupp, K.C. and De Vivo, M.J. (1993)
Pathak, M. and Vickrey, B.G. (1999) Systematic review of Epidemiology and risk factors for urinary tract infection
risk factors for urinary tract infection in adults with spinal following spinal cord injury. Arch. Phys. Med. Rehab., 74(7):
cord dysfunction. J. Spinal Cord Med., 22: 258–272. 691–695.
Siroky, M.B. (2002) Pathogenesis of bacteriuria and infection in Wolfe, D.L., Potter, P.J., Jutai, J.W., Beggs, C., Colwell, S.,
the spinal cord injured patient. Am. J. Med., 113(1A): Orenczuk, S., Regan, M.A., Tonack, M., Trujillo, S. and
67S–79S. Zarnowiecki, E. (2002) Long-term physical impact of spinal
Stover, S.L., Lloyd, L.K., Waites, K.B. and Jackson, A.B. cord injury: the role of rehabilitation in educating consumers
(1989) Urinary tract infection in spinal cord injury. Arch. to reduce complications over the long-term. J. Spinal Cord
Phys. Med. Rehab., 70: 47–54. Med., 22(Suppl. 1): S50.
Trautner, B.W. and Darouiche, R.O. (2002) Prevention of Yavuzer, G., Gok, H., Tuncer, S., Soygur, T., Arikan, N. and
urinary tract infection in patients with spinal cord injury. Arasil, T. (2000) Compliance with bladder management in
J. Spinal Cord Med., 25: 277–283. spinal cord injury patients. Spinal Cord, 38(12): 762–765.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 5

Mechanisms underlying the recovery of lower urinary


tract function following spinal cord injury

William C. de Groat and Naoki Yoshimura

Departments of Pharmacology and Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA

Abstract: The lower urinary tract has two main functions, the storage and periodic expulsion of urine,
which are regulated by a complex neural control system in the brain and lumbosacral spinal cord. This
neural system coordinates the activity of two functional units in the lower urinary tract: (1) a reservoir (the
urinary bladder) and (2) an outlet (consisting of bladder neck, urethra and striated muscles of the pelvic
floor). During urine storage the outlet is closed and the bladder is quiescent, thereby maintaining a low
intravesical pressure over a wide range of bladder volumes. During micturition the outlet relaxes and the
bladder contracts to promote the release of urine. This reciprocal relationship between bladder and outlet is
generated by visceral reflex circuits, some of which are under voluntary control. Experimental studies in
animals indicate that the micturition reflex is mediated by a spinobulbospinal pathway passing through a
coordination center (the pontine micturition center) located in the rostral brainstem. This reflex pathway is
in turn modulated by higher centers in the cerebral cortex that are presumably involved in the voluntary
control of micturition. Spinal cord injury at cervical or thoracic levels disrupts voluntary control of voiding
as well as the normal reflex pathways that coordinate bladder and sphincter functions. Following spinal cord
injury, the bladder is initially areflexic but then becomes hyperreflexic due to the emergence of a spinal
micturition reflex pathway. Studies in animals indicate that the recovery of bladder function after spinal
cord injury is dependent in part on plasticity of bladder afferent pathways and the unmasking of reflexes
triggered by capsaicin-sensitive C-fiber bladder afferent neurons. The plasticity is associated with changes in
the properties of ion channels and electrical excitability of afferent neurons, and appears to be mediated in
part by neurotrophic factors released in the spinal cord and the peripheral target organs.

Introduction certain level of function even after elimination of


extrinsic neural input.
The functions of the lower urinary tract to store The lower urinary tract is also unusual with
and periodically release urine are dependent upon regard to its pattern of activity and the complexity
neural circuits located in the brain, spinal cord and of its neural regulation. For example, the urinary
peripheral ganglia (Barrington, 1925; Kuru, 1965; bladder has two principal modes of operation:
de Groat et al., 1993; Morrison et al., 2002). storage and elimination. Thus many of the neural
This dependence on central nervous control circuits exhibit switch-like or phasic patterns of
distinguishes the lower urinary tract from many activity (de Groat, 1975) in contrast to tonic
other visceral structures (e.g., the gastrointestinal patterns occurring in autonomic pathways to
tract and cardiovascular system) that maintain a cardiovascular organs. In addition, micturition is
under voluntary control and depends upon learned
Corresponding author. Tel.: +412 648 9357; behavior that develops during maturation of the
Fax: +412 648 1945; E-mail: degroat@server.pharm.pitt.edu nervous system, whereas many other visceral

DOI: 10.1016/S0079-6123(05)52005-3 59
60

functions are regulated involuntarily. Micturition sympathetic (hypogastric nerves and sympathetic
also depends on the integration of autonomic and chain) (Fig. 1) (de Groat et al., 1993).
somatic efferent mechanisms within the lumbosacral
spinal cord (Chancellor and Yoshimura, 2002;
Morrison et al., 2002). This is necessary during Parasympathetic pathways
urine storage and elimination to coordinate the
activity of visceral organs (the bladder and The sacral, parasympathetic, efferent pathway
urethra) with that of urethral striated muscles. provides the major excitatory input to the blad-
The dependence of lower urinary tract functions der and consists of spinal preganglionic neurons
on complex central neural networks renders these (Morgan et al., 1979) with cell bodies, in most of
functions susceptible to a variety of neurological the species studied, situated in the gray matter of
disorders (Torrens and Morrison, 1987; Chancellor the sacral spinal segments. In some species, for
and Yoshimura, 2002). This chapter will review example, the rat, the cell bodies span the caudal
studies in animals and humans that have provided lumbar and rostral sacral segments. Regardless of
insights into the neural control of the lower the actual location of the cell bodies, this group of
urinary tract and the disruption of this control neurons will be called the ‘‘sacral parasympathetic
by spinal cord injury. nucleus’’ in this chapter. These neurons send axons
to peripheral ganglion cells that, in turn, innervate
the bladder and urethral smooth muscle (Fig. 1).
Anatomy and innervation Transmission in bladder ganglia is mediated by
acetylcholine that excites the ganglion cells by act-
The storage and periodic elimination of urine are ing on nicotinic cholinergic receptors, whereas
regulated by the activity of two functional units in parasympathetic neuroeffector transmission in the
the lower urinary tract: (1) a reservoir (the bladder) bladder is mediated by acetylcholine acting on
and (2) an outlet (consisting of bladder neck, muscarinic receptors (de Groat and Yoshimura,
urethra and striated muscles of the pelvic floor). 2001; Andersson and Arner, 2004). Both M2 and
Under normal conditions, the urinary bladder and M3 muscarinic receptor subtypes are expressed in
outlet exhibit a reciprocal relationship. During bladder smooth muscle; however, examination of
urine storage, the bladder neck and proximal subtype-selective muscarinic receptor antagonists
urethra are closed; and the bladder smooth muscle and studies of muscarinic receptor knockout mice
is quiescent, allowing intravesical pressure to have revealed that the M3 subtype is the principal
remain low over a wide range of bladder volumes. receptor involved in excitatory transmission.
During voluntary micturition, the initial event is a In bladders of various animals, stimulation
reduction of intraurethral pressure, which reflects of parasympathetic nerves also produces a
a relaxation of the pelvic floor and the periurethral non-cholinergic contraction that is resistant to
striated muscles, and an opening of the bladder atropine and other muscarinic receptor blocking
neck (Chancellor and Yoshimura, 2002). The agents. Adenosine triphosphate (ATP) has been
changes in the urethra are followed in a few identified as the excitatory transmitter mediating
seconds by a detrusor contraction and a rise in the non-cholinergic contraction (Ralevic and
intravesical pressure that is maintained until the Burnstock, 1998; Burnstock, 2001). ATP excites
bladder empties. Reflex inhibition of the smooth the bladder smooth muscle by acting on P2X
and striated muscles of the urethra also contrib- purinergic receptors that are ligand-gated ion
utes to the reduction of outlet resistance during channels. Among the seven types of P2X recep-
micturition. These changes are coordinated by three tors that have been identified, P2X1 is the major
sets of nerves emerging from the thoracolumbar subtype expressed in the rat and also in the human
and sacral levels of the spinal cord: (1) sacral bladder smooth muscle. Although purinergic
parasympathetic (pelvic nerves), (2) sacral somatic excitatory transmission is not important in the
(pudendal nerves) and (3) thoracolumbar normal human bladder, it appears to be involved
61

Fig. 1. Diagram showing the sympathetic, parasympathetic and somatic innervation of the urogenital tract of the male cat. Sym-
pathetic preganglionic pathways emerge from the lumbar spinal cord and pass to the sympathetic chain ganglia (SCG) and then via the
inferior splanchnic nerves (ISN) to the inferior mesenteric ganglia (IMG). Preganglionic and postganglionic sympathetic axons then
travel in the hypogastric nerve (HGN) to the pelvic plexus and the urogenital organs. Parasympathetic preganglionic axons that
originate in the sacral spinal cord pass in the pelvic nerve to ganglion cells in the pelvic plexus and to distal ganglia in the organs. Sacral
somatic pathways are contained in the pudendal nerve, which provides an innervation to the penis, the ischiocavernosus (IC),
bulbocavernosus (BC) and external urethral sphincter (EUS) muscles. The pudendal and pelvic nerves also receive postganglionic
axons from the caudal SCG. These three sets of nerves contain afferent axons from the lumbosacral dorsal root ganglia. Abbreviations:
U, ureter; PG, prostate gland; VD, vas deferens.

in bladders in patients with pathological condi- receptor antagonists or by desensitization of P2X


tions such as chronic urethral outlet obstruction or purinergic receptors, indicating that acetylcholine
interstitial cystitis (Palea et al., 1993; O’Reilly or ATP is involved in excitatory transmission to
et al., 2002). urethral smooth muscle (Zoubek et al., 1993).
Parasympathetic pathways to the urethra induce
relaxation during voiding. In various species the Sympathetic pathways
relaxation is not affected by muscarinic antago-
nists and therefore is not mediated by acetylcho- Sympathetic preganglionic pathways that arise
line. However inhibitors of nitric oxide synthase from the 11th thoracic (T11) to 2nd lumbar (L2)
block the relaxation in vivo during reflex voiding spinal segments pass to the sympathetic chain
or block the relaxation of urethral smooth muscle ganglia (SCG) and then to prevertebral ganglia in
strips induced in vitro by electrical stimulation of the superior hypogastric and pelvic plexuses
intramural nerves indicating that nitric oxide is the (Fig. 1) and also to short adrenergic neurons in
inhibitory transmitter involved in relaxation the bladder and urethra (de Groat et al., 1993;
(Burnett et al., 1997; Ho et al., 1999; Morrison Delancey et al., 2002). Sympathetic postganglionic
et al., 2002). In some species, neurally evoked con- nerves that release norepinephrine provide an
tractions of the urethra are reduced by muscarinic excitatory input to smooth muscle of the urethra
62

and bladder base, an inhibitory input to smooth et al., 2003, 2004). Studies of the biomechanical
muscle in the bladder body as well as inhibitory properties of the intact female rat urethra in vitro
and facilitatory inputs to vesical parasympathetic have confirmed the large contribution of striated
ganglia (Andersson, 1993; de Groat and Booth, muscle activity and nicotinic receptor mechanisms
1993). Radioligand receptor binding studies to the contractions of the mid-urethra (Jankowski
showed that a-adrenergic receptors are concen- et al., 2004).
trated in the bladder base and proximal urethra,
whereas b-adrenergic receptors are most promi-
nent in the bladder body (Andersson, 1993). These Afferent pathways
observations are consistent with pharmacological
studies showing that sympathetic nerve stimula- Afferent axons innervating the urinary tract are
tion or exogenous catecholamines produce b- present in the three sets of nerves that innervate
adrenergic receptor-mediated inhibition of the the lower urinary tract (Janig and Morrison, 1986;
body and a-adrenergic receptor-mediated contrac- de Groat et al., 1993; Bahns et al., 1998; Morrison
tion of the base, dome and urethra. Molecular and et al., 2002). The most important afferents for in-
contractility studies have shown that b3-adrenergic itiating micturition are those passing through the
receptors elicit inhibition and a1-adrenergic recep- pelvic nerve to the sacral spinal cord. These affer-
tors elicit contractions. The a1A-adrenergic recep- ents are small myelinated (Ad) and unmyelinated
tor subtype is most prominent in the normal (C) fibers that convey information from receptors
bladders but the a1D-subtype is upregulated in in the bladder wall to second-order neurons in the
bladders from patients with outlet obstruction, spinal cord. Ad bladder afferents in the cat re-
raising the possibility that a1-adrenergic receptor spond in a graded manner to passive distension as
excitatory mechanisms in the bladder might con- well as active contraction of the bladder and ex-
tribute to irritative lower urinary tract symptoms hibit intravesical pressure thresholds in the range
in patients with obstruction (de Groat and of 5–15 mmHg, similar to the pressures at which
Yoshimura, 2001; Morrison et al., 2002). humans report the first sensation of bladder filling
(Chancellor and Yoshimura, 2002; Morrison et al.,
2002). These fibers also code for noxious stimuli in
Somatic pathways the bladder. On the other hand, C-fiber bladder
afferents in the cat have high thresholds and com-
The external urethral sphincter, which is composed monly do not respond to even high levels of in-
of striated muscle, receives a somatic cholinergic travesical pressure (Habler et al., 1990). However,
innervation via the pudendal nerve from anterior activity in some of these afferents is unmasked or
horn cells in the third and fourth sacral segments enhanced by chemical irritation of the bladder
(Fig. 1). Branches of the pudendal nerve and other mucosa. These findings indicate that C-fiber affer-
sacral somatic nerves also carry efferent impulses ents in the cat have specialized functions, such as
to muscles of the pelvic floor and proprioceptive the signaling of inflammatory or noxious events in
afferent signals from these muscles as well as sen- the lower urinary tract. Nociceptive and me-
sory information from the urethra. Analysis of chanoceptive information is also carried in the
urethral closure mechanisms in the female rat dur- hypogastric nerves to the thoracolumbar segments
ing bladder distension-evoked and sneeze-induced of the spinal cord (Bahns et al., 1998).
stress conditions revealed that the major rise in In rats, A- and C-fiber bladder afferents are not
urethral pressure occurred in the mid-urethra and distinguishable on the basis of stimulus modality;
was mediated by efferent pathways in the puden- thus both types of afferents consist of mechano-
dal nerve to the external urethral sphincter as well and chemo-sensitive populations (Sengupta and
as pathways in nerves to the iliococcygeus and Gebhart, 1994; Morrison et al., 1999; Shea et al.,
pubococcygeus muscles, but not by pathways in 2000; Rong et al., 2002). C-fiber afferents that
the sympathetic or parasympathetic nerves (Kamo respond only to bladder filling have also been
63

identified in the rat bladder and appear to be contain peptides: calcitonin gene-related peptide,
volume receptors possibly sensitive to stretch of vasoactive intestinal polypeptide, pituitary-adenyl
the mucosa. C-fiber afferents are sensitive to the cyclase activating polypeptide (PACAP), tachy-
neurotoxins, capsaicin and resiniferatoxin as well kinins, galanin and opioid peptides (de Groat,
as to other substances such as tachykinins, nitric 1987; Keast and de Groat, 1992; Maggi, 1993;
oxide, ATP, prostaglandins and neurotrophic fac- Morrison et al., 2002). Nerves containing these
tors released into the bladder by afferent nerves, peptides are common in the bladder, in the sub-
urothelial cells and inflammatory cells (Vizzard mucosal and epithelial layers, and around blood
et al., 1995; Lee et al., 2000; Chuang et al., 2001; vessels (de Groat and Yoshimura, 2001). In the
Yoshimura et al., 2001a; Morrison et al., 2002). spinal cord, peptidergic nerve terminals have a
These substances can sensitize the afferent nerves distribution very similar to the distribution of
and change their response to mechanical stimuli. pelvic nerve afferents labeled with horseradish
The properties of lumbosacral dorsal root gan- peroxidase (Kawatani et al., 1985; de Groat, 1987).
glion cells innervating the bladder, urethra and Peptidergic bladder afferent neurons in the rat also
external urethral sphincter in the rat have been express TrkA, a high-affinity receptor for nerve
studied with patch clamp recording techniques in growth factor (NGF) (McMahon et al., 1994) and
combination with axonal tracing methods to receptors for capsaicin (TRPV1) (Yoshimura
identify the different populations of neurons et al., 1996, 1998a, 2003) and tachykinins (NK-2
(Yoshimura et al., 1996, 2001b, 2003; Yoshimura and NK-3 receptors) (Morrison et al., 2002;
and de Groat, 1997, 1999; Black et al., 2003). Sculptoreanu and de Groat, 2003). Capsaicin, a
Based on responsiveness to capsaicin it is estimated neurotoxin that can release peptides from afferent
that approximately 70% of bladder afferent terminals, produces inflammatory responses, in-
neurons in the rat are of the C-fiber type. These cluding plasma extravasation and vasodilatation,
neurons exhibit high threshold, tetrodotoxin- when applied locally to the bladder in experimen-
resistant sodium channels and action potentials. tal animals (Maggi, 1993). These findings suggest
They show phasic firing (one to two spikes) in re- that the neuropeptides may be important trans-
sponse to prolonged depolarizing current pulses. mitters in the afferent pathways from the lower
Approximately 90% of the bladder C-fiber afferent urinary tract. Tachykinins may also act back on
neurons also are excited by ATP, which induces a afferent terminals in an auto-feedback manner
depolarization and firing by activating P2X3 or to modulate the excitability of the terminals
P2X2/3 receptors (Zhong et al., 2003). These (Morrison et al., 2002; Sculptoreanu and de Groat,
neurons express isolectin-B4 binding, which is 2003).
commonly used as a marker for ATP responsive
sensory neurons. A-fiber afferent neurons are Urothelial– afferent interactions
resistant to capsaicin and ATP, and exhibit low
threshold tetrodotoxin-sensitive sodium channels Recent studies have revealed that the urothelium,
and action potentials and tonic firing (multiple which has been traditionally viewed as a passive
spikes) to depolarizing current pulses. C-fiber barrier at the bladder luminal surface (Lavelle
bladder afferent neurons also express a slowly et al., 2000; Lewis, 2000), also has specialized
decaying A-type K+ current that controls spike sensory and signaling properties that allow
threshold and firing frequency (Yoshimura et al., urothelial cells to respond to their chemical and
1996, 2003). Suppression of this K+ current induces physical environment and to engage in reciprocal
hyperexcitability of bladder afferent neurons. These chemical communication with neighboring nerves
properties of dorsal root ganglion cells are in the bladder wall (Ferguson et al., 1997; Birder
consistent with the different properties of A- and et al., 1998, 2001, 2002, 2003; Cockayne et al.,
C-fiber afferent receptors in the bladder. 2000). These properties include: (1) expression of
Immunohistochemical studies have shown that a nicotinic, muscarinic, tachykinin, adrenergic and
large percentage of bladder afferent neurons capsaicin (TRPV1) receptors, (2) responsiveness to
64

transmitters released from sensory nerves, (3) close


physical association with afferent nerves and (4)
ability to release chemical mediators such as ATP
and nitric oxide that can regulate the activity of
adjacent nerves and thereby trigger local vascular
changes and/or reflex bladder contractions. The
role of ATP in urothelial–afferent communication
has attracted considerable attention because blad-
der distension releases ATP from the urothelium
and intravesical administration of ATP induces
bladder hyperactivity, an effect blocked by admin-
istration of P2X purinergic receptor antagonists
that suppress the excitatory action of ATP on
bladder afferent neurons (Morrison et al., 2002).
Mice in which the P2X3 receptor was knocked out
exhibited hypoactive bladder activity and ineffi-
cient voiding (Cockayne et al., 2000), suggesting
that activation of P2X3 receptors on bladder
afferent nerves by ATP released from the urothe-
lium is essential for normal bladder function. The
chapter by Birder (this volume) presents a more
detailed discussion of urothelial–afferent interac-
tions and changes in the urothelium after spinal
Fig. 2. Combined cystometrograms and sphincter electromyo-
cord injury. grams (EMG) comparing reflex voiding responses in an infant
(A) and in a paraplegic subject (C) with a voluntary voiding
Reflex mechanisms controlling the lower response in an able-bodied adult (B). The abscissa in all records
urinary tract represents bladder volume in milliliters and the ordinates rep-
resent bladder pressure in cm H2O and electrical activity of the
EMG recording. On the left side of each trace the arrows in-
The neural pathways controlling lower urinary dicate the start of a slow infusion of fluid into the bladder
tract function are organized as simple on–off (bladder filling). Vertical dashed lines indicate the start of
switching circuits that maintain a reciprocal rela- sphincter relaxation that precedes by a few seconds the bladder
tionship between the urinary bladder and urethral contraction in A and B. (B) Note that a voluntary cessation of
voiding (stop) is associated with an initial increase in sphincter
outlet (de Groat et al., 1981, 1993) (Fig. 2). The
EMG followed by a reciprocal relaxation of the bladder. A
principal reflex components of these switching resumption of voiding is again associated with sphincter relax-
circuits are listed in Table 1 and illustrated in ation and a delayed increase in bladder pressure. On the other
Fig. 3. Intravesical pressure measurements during hand, in the paraplegic subject (C) the reciprocal relationship
bladder filling in both humans and animals reveal between bladder and sphincter is abolished. During bladder
filling, transient uninhibited bladder contractions occur in as-
low and slowly increasing bladder pressures when
sociation with sphincter activity. Further filling leads to more
bladder volume is below the threshold for inducing prolonged and simultaneous contractions of the bladder and
voiding (Fig. 2). The accommodation of the blad- sphincter (bladder-sphincter dyssynergia). Loss of the recipro-
der to increasing volumes of urine is primarily a cal relationship between bladder and sphincter in paraplegic
passive phenomenon dependent upon the intrinsic people interferes with bladder emptying.
properties of the vesical smooth muscle and qui-
escence of the parasympathetic efferent pathway. urethra (Table 1, Fig. 3) (de Groat and Lalley,
In addition, in some species urine storage is also 1972; de Groat et al., 1981). During bladder filling
facilitated by sympathetic reflexes that mediate the activity of the sphincter electromyogram
an inhibition of bladder activity, closure of the (EMG) increases (Fig. 2), reflecting an increase
bladder neck and contraction of the proximal in efferent firing in the pudendal nerve and an
65

Table 1. Reflexes to the lower urinary tract

Afferent Pathway Efferent Pathway Central Pathway

Urine storage

Low-level vesical afferent activity (pelvic nerve) 1. External sphincter contraction (somatic nerves) Spinal reflexes
2. Internal sphincter contraction (sympathetic nerves)
3. Detrusor inhibition (sympathetic nerves)
4. Ganglionic inhibition (sympathetic nerves)
5. Sacral parasympathetic outflow inactive

Micturition

High-level vesical afferent activity (pelvic nerve) 1. Inhibition of external sphincter activity S-B-S reflexa
2. Inhibition of sympathetic outflow S-B-S reflex
3. Activation of parasympathetic outflow to the bladder S-B-S reflex
4. Activation of parasympathetic outflow to the urethra Spinal reflex

a
S-B-S reflex, spinobulbospinal reflex

increase in outlet resistance that contributes to the tracing, measurements of gene expression and
maintenance of urinary continence. patch-clamp recording in spinal cord slice prepa-
The storage phase of the urinary bladder can be rations have recently provided many new insights
switched to the voiding phase either involuntarily into the morphological and electrophysiological
(reflexly) or voluntarily (Fig. 2). The reflex switch properties of these reflex components. Neurotropic
is readily demonstrated in the human infant viruses, such as pseudorabies virus, have been
(Fig. 2A) or in the anesthetized animal when the particularly useful since they can be injected into a
volume of urine exceeds the micturition threshold. target organ (urinary bladder, urethra and urethral
At this point, increased afferent firing from tension sphincter) and then move intraaxonally from the
receptors in the bladder reverses the pattern of periphery to the central nervous system, where
efferent outflow, producing firing in the sacral they replicate and then pass retrogradely across
parasympathetic pathways and inhibition of synapses to infect second- and third-order neurons
sympathetic and somatic pathways. The expulsion in the neural pathways (Vizzard et al., 1995;
phase consists of an initial relaxation of the Nadelhaft and Vera, 1996; Sugaya et al., 1997).
urethral sphincter (Fig. 2) followed in a few Since pseudorabies virus can be transported across
seconds by a contraction of the bladder, and an many synapses, it could sequentially infect all of
increase in bladder pressure and flow of urine. the neurons that connect directly or indirectly to
Relaxation of the urethral outlet is mediated by the lower urinary tract (Fig. 4).
activation of a parasympathetic reflex pathway to
the urethra that triggers the release of nitric oxide, Anatomy of the spinal cord
an inhibitory transmitter, as well as by removal of
adrenergic and somatic cholinergic excitatory The spinal cord gray matter is divided into three
inputs to the urethra. Secondary reflexes elicited general regions: (1) the dorsal horn that contains
by flow of urine through the urethra facilitate interneurons that process sensory input, (2) the
bladder emptying. ventral horn that contains motoneurons and (3)
The reflex circuitry controlling micturition the intermediate region, located between the dorsal
consists of four basic components: spinal efferent and ventral horns, that contains interneurons and
neurons, spinal interneurons, primary afferent autonomic preganglionic neurons. These regions
neurons and neurons in the brain that modulate are further subdivided into layers or laminae that
spinal reflex pathways. Transneuronal virus are numbered, starting with the superficial layer of
66

Fig. 3. Diagram showing neural circuits controlling continence and micturition. (A) Urine storage reflexes. During the storage of
urine, distention of the bladder produces low level vesical afferent firing, which in turn stimulates (1) the sympathetic outflow to the
bladder outlet (base and urethra) and (2) pudendal outflow to the external urethral sphincter. These responses occur by spinal reflex
pathways and represent guarding reflexes, which promote continence. Sympathetic firing also inhibits the detrusor muscle and mod-
ulates transmission in bladder ganglia. A region in the rostral pons (the pontine storage center) increases external urethral sphincter
activity. (B) Voiding reflexes. During elimination of urine, intense bladder afferent firing activates spinobulbospinal reflex pathways
passing through the pontine micturition center, which stimulate the parasympathetic outflow to the bladder and internal sphincter
smooth muscle and inhibit the sympathetic and pudendal outflow to the urethral outlet. Ascending afferent input from the spinal cord
may pass through relay neurons in the periaqueductal gray (PAG) before reaching the pontine micturition center.

the dorsal horn (lamina I) and extending to the intermediolateral gray matter (laminae V–VII) in
ventral horn (lamina IX), and the commissure the sacral (in the rat, also caudal lumbar) segments
connecting the two sides of the spinal cord (lamina of the spinal cord (Fig. 4) (Morgan et al., 1981; de
X) (Fig. 5D). Groat et al., 1982; Araki and de Groat, 1997;
Miura et al., 2001a), whereas sympathetic pre-
Efferent pathways ganglionic neurons are located in both medial
(lamina X) and lateral sites (laminae V–VII) in the
Parasympathetic preganglionic neurons innervat- intermediate gray matter of the rostral lumbar
ing the lower urinary tract are located in the spinal cord. Parasympathetic preganglionic
67

Fig. 4. Transneuronal virus tracing of the central pathways controlling the urinary bladder of the rat. Injection of pseudorabies virus
into the wall of the urinary bladder leads to retrograde transport of virus (dashed arrows) and sequential infection of postganglionic
neurons, preganglionic neurons and then various central neural circuits synaptically linked to the preganglionic neurons. Normal
synaptic connections are indicated by solid arrows. At long survival times virus can be detected with immunocytochemical techniques
in neurons at specific sites throughout the spinal cord and brain extending to the pontine micturition center in the pons (i.e.,
Barrington’s nucleus) and to the cerebral cortex. Other sites in the brain labeled by virus are: (1) the paraventricular nucleus (PVN),
medial preoptic area (MPOA) and periventricular nucleus (Peri V.N.) of the hypothalamus, (2) periaqueductal gray (PAG), (3) locus
coeruleus (LC) and subcoeruleus, (4) red nucleus, (5) medullary raphe nuclei and (6) the noradrenergic cell group designated A5. Sixth
lumbar (L6) spinal cord section showing on the left side the distribution of virus labeled parasympathetic preganglionic neurons (&)
and interneurons (K) in the region of the parasympathetic nucleus, the dorsal commissure (DCM) and the superficial laminae of the
dorsal horn (DH), 72 h after injection of the virus into the bladder. The right side shows the entire population of preganglionic neurons
(PGN)(&) labeled by axonal tracing with the fluorescent dye (Fluorogold), injected into the pelvic ganglia and the distribution of
virus-labeled bladder PGN (’). Composite diagram of neurons in 12 spinal sections (42 mm each).

neurons send dendrites to discrete regions of the 1990; Sasaki, 1994). This dendritic distribution of
spinal cord including: (1) the lateral and dorsal sphincter motoneurons is similar to that of sacral
lateral funiculus, (2) lamina I on the lateral edge of preganglionic neurons indicating that these two
the dorsal horn, (3) the dorsal gray commissure populations of neurons may receive synaptic
(lamina X) and (4) gray matter and lateral fun- inputs from the same interneuronal sites and
iculus ventral to the autonomic nucleus (Morgan fiber tracts in the spinal cord.
et al., 1993). As discussed below, this dendritic
structure very likely indicates the origin of impor- Afferent projections in the spinal cord
tant synaptic inputs to these cells. Pudendal moto-
neurons innervating the external urethral sphincter Afferent pathways from the lower urinary tract
in the cat are located in the ventrolateral division project to discrete regions of the dorsal horn that
of Onuf’s nucleus and send dendritic projections contain the interneurons as well as the soma and/
into (1) the lateral funiculus, (2) lamina X (3) in- or dendrites of efferent neurons innervating the
termediolateral gray matter and (4) rostrocaudally lower urinary tract. Pelvic nerve afferent pathways
within the nucleus (Thor et al., 1989; Beattie et al., from the urinary bladder of the cat and rat project
68

Fig. 5. Comparison of the distribution of bladder afferent projections to the 6th lumbar (L6) spinal cord of the rat (A) with the
distribution of c-fos positive cells in the L6 spinal segment following chemical irritation of the lower urinary tract of the rat (B) and the
distribution of interneurons in the L6 spinal cord labeled by transneuronal transport of pseudorabies virus injected into the urinary
bladder (C). Afferents labeled by wheatgerm agglutinin-horseradish peroxidase (WGA-HRP) injected into the urinary bladder. C-fos
immunoreactivity is present in the nuclei of cells. DH, dorsal horn; SPN, sacral parasympathetic nucleus; CC central canal. (D)
Drawing shows the laminar organization of the cat spinal cord.

into Lissauer’s tract at the apex of the dorsal horn Spinal interneurons
and then pass rostrocaudally giving off collaterals
that extend laterally and medially through the su- As shown in Fig. 5C, interneurons retrogradely
perficial layer of the dorsal horn (lamina I) into the labeled by injection of pseudorabies virus into the
deeper layers (laminae V–VII and X) at the base of urinary bladder of the rat are located in the regions
the dorsal horn (Fig. 5A) (Morgan et al., 1981; of the spinal cord receiving afferent input from the
Steers et al., 1991). The lateral pathway, which is bladder (Nadelhaft and Vera, 1995; Sugaya et al.,
the most prominent projection, terminates in the 1997). Interneuronal locations also overlap in
region of the sacral parasympathetic nucleus many sites with the dendritic distribution of the
(SPN) and also sends some axons to the dorsal efferent neurons. A similar distribution of labeled
commissure (Fig. 5A). Pudendal afferent pathways interneurons has been noted following injections
from the urethra and urethral sphincter exhibit a of virus into the urethra (Vizzard et al., 1995) or
similar pattern of termination in the sacral spinal the external urethral sphincter, indicating a prom-
cord (Thor et al., 1989). The overlap of bladder inent overlap of the interneuronal pathways con-
and urethral afferents in the lateral dorsal horn trolling the various target organs of the lower
and dorsal commissure indicates these regions are urinary tract.
likely to be important sites of viscerosomatic in- The spinal neurons involved in processing af-
tegration and be involved in coordinating bladder ferent input from the lower urinary tract have been
and sphincter activity. identified by the expression of the immediate early
69

gene, c-fos (Fig. 5B). In the rat, noxious or non- group (Fig. 4). Several regions in the hypothalamus
noxious stimulation of the bladder and urethra and the cerebral cortex also exhibited virus-infected
increases the levels of Fos protein, primarily in the cells. Neurons in the cortex were located primarily
dorsal commissure, the superficial dorsal horn and in the medial frontal cortex. Other anatomical
in the area of the sacral parasympathetic nucleus studies in which anterograde tracer substances
(Fig. 5B) (Birder and de Groat, 1993; Birder et al., were injected into brain areas and then identified
1999). Some of these interneurons send long pro- in terminals in the spinal cord are consistent with
jections to the brain, whereas others make local the virus tracing data (Morrison et al., 2002).
connections in the spinal cord and participate in
segmental spinal reflexes.
Patch clamp recordings from parasympathetic Organization of urine storage and voiding reflexes
preganglionic neurons in the neonatal rat spinal
slice preparation have revealed that interneurons Sympathetic storage pathway
located immediately dorsal and medial to the
parasympathetic nucleus make direct monosynap- The integrity of the sympathetic input to the lower
tic connections with the preganglionic neurons urinary tract is not essential for the performance of
(Araki, 1994; Araki and de Groat, 1996, 1997). micturition (Torrens and Morrison, 1986; de
Microstimulation of interneurons in both loca- Groat et al., 1993). However, physiologic experi-
tions elicits glutamatergic, N-methyl-D-aspartic ments in animals indicate that during bladder
acid (NMDA) and non-NMDA excitatory postsy- filling, the sympathetic system does provide a tonic
naptic currents in the preganglionic neurons. Stim- inhibitory input to the bladder as well as an
ulation of a subpopulation of medial interneurons excitatory input to the urethra. This sympathetic
elicits gamma-amino butyric acid (GABA)ergic input is physiologically significant since surgical
and glycinergic inhibitory postsynaptic currents. interruption or pharmacologic blockade of the
Thus local interneurons are likely to play an im- sympathetic innervation can reduce urethral
portant role in both excitatory and inhibitory re- outflow resistance, reduce bladder capacity and
flex pathways controlling the preganglionic increase the frequency and amplitude of bladder
outflow to the lower urinary tract. Glutamatergic contractions recorded under constant volume
excitatory currents have also been elicited in pre- conditions.
ganglionic neurons by stimulation of the projec- Sympathetic reflex activity is elicited by a
tions from lamina X and the lateral funiculus sacrolumbar, intersegmental spinal reflex pathway
(Miura et al., 2001a, 2003). that is triggered by vesical afferent activity in the
pelvic nerves (Fig. 3A, Table 1) (de Groat and
Lalley, 1972). The reflex pathway is inhibited when
Pathways in the brain bladder pressure is raised to the threshold for
producing micturition. This inhibitory response is
The neurons in the brain that control the lower abolished by transection of the spinal cord at the
urinary tract have been studied with a variety of lower thoracic level, indicating that it originates at
anatomical tracing techniques in several species a supraspinal site, possibly the pontine micturition
(Morrison et al., 2002). In the rat, transneuronal center. Thus, the vesicosympathetic reflex repre-
virus tracing methods have identified many popu- sents a negative feedback mechanism, whereby an
lations of neurons that are involved in the control increase in bladder pressure tends to increase in-
of bladder, urethra and the urethral sphincter hibitory input to vesical ganglia and smooth mus-
including: Barrington’s nucleus (the pontine mi- cle, thus allowing the bladder to accommodate
cturition center), medullary raphe nucleus which large volumes (Fig. 3A). Increased sympathetic
contains serotonergic neurons, the locus coeruleus excitatory input to the bladder base and urethra
which contains noradrenergic neurons, per- would complement these mechanisms by increas-
iaqueductal gray and the A5 noradrenergic cell ing outflow resistance.
70

Urethral sphincter storage pathway through the pontine micturition center (Fig. 3B).
The pathway functions as on–off switch (de Groat,
Motoneurons innervating the striated muscles of 1975) that is activated by a critical level of afferent
the urethral sphincter exhibit a tonic discharge activity arising from tension receptors in the blad-
that increases during bladder filling. This activity der, and is in turn modulated by inhibitory and ex-
is mediated in part by low-level afferent input from citatory influences from areas of the brain rostral to
the bladder (Fig. 3A, Table 1). During micturition the pons (e.g., diencephalon and cerebral cortex)
the firing of sphincter motoneurons is inhibited. (Torrens and Morrison, 1986; de Groat et al., 1993).
This inhibition is dependent in part on supraspinal In contrast to the reflex control of the bladder,
mechanisms since it is not as prominent in chronic the parasympathetic control of the urethra in the
spinal animals. Electrical stimulation of the pon- rat appears to be dependent on pathways organ-
tine micturition center induces sphincter relaxation ized in the spinal cord (Table 1) that are modulated
suggesting that bulbospinal pathways from the by input from the brain. Nitric oxide-mediated
pons may be responsible for maintaining the nor- relaxation of the urethra that occurs in response to
mal reciprocal relationship between bladder and bladder distension is reduced but not eliminated by
sphincter (Holstege et al., 1986; Shefchyk, 1989; acute transection of the spinal cord (Kakizaki
Mallory et al., 1991). et al., 1997, Cheng et al., 1997). The reflex
relaxation of the urethral smooth muscle is very
prominent in chronic spinal-cord-transected rats.
Spinobulbospinal parasympathetic micturition Electrophysiological studies in cats and rats
pathway have confirmed that the parasympathetic efferent
outflow to the urinary bladder is activated by a
Micturition is mediated by activation of the sacral long latency supraspinal reflex pathway (de Groat
parasympathetic efferent pathway to the bladder et al., 1981, 1982, 1993; Mallory et al., 1989;
and the urethra as well as reciprocal inhibition of Cheng et al., 1999). In cats, recordings from sacral
the somatic pathway to the urethral sphincter parasympathetic preganglionic neurons innervat-
(Table 1, Fig. 3B). Studies in animals using brain ing the urinary bladder show that reflex firing oc-
lesioning techniques revealed that neurons in the curs with a long latency (65–100 ms) following
brainstem at the level of the inferior colliculus (i.e., stimulation of myelinated (Ad) vesical afferents in
the pontine micturition center) have an essential the pelvic nerve (Fig. 6). Afferent stimulation also
role in the control of the parasympathetic compo- evokes negative field potentials in the rostral pons
nent of micturition (Barrington, 1925; Kuru, 1965; at latencies of 30–40 ms, whereas electrical stimu-
Torrens and Morrison, 1986; Mallory et al., 1991; lation in the pons excites sacral preganglionic neu-
de Groat et al., 1993). Removal of areas of the rons at latencies of 45–60 ms. The sum of the
brain above the inferior colliculus by inter- latencies for the spinobulbar and bulbospinal
collicular decerebration usually facilitates micturit- components of the reflex pathway approximates
ion by elimination of inhibitory inputs from more the latency for the entire reflex. In cats, it is be-
rostral centers (Yokoyama et al., 2000). However, lieved that the ascending afferent pathways from
transections at any point below the colliculi abol- the spinal cord project to a relay station in the
ish micturition. Bilateral lesions in the rostral periaqueductal gray (PAG), which then connects
pons, in the region of the pontine micturition cen- to the pontine micturition center (Fig. 3B) (Blok
ter in cats, also abolish micturition (Barrington, and Holstege, 1994; Blok et al., 1995; Blok, 2002).
1925), whereas electrical or chemical stimulation at
these sites triggers bladder contractions and mi-
cturition (Kuru, 1965; Sugaya et al., 1987; Kruse Pontine micturition center
et al., 1990; Mallory et al., 1991; Noto et al., 1991b).
These observations led to the concept of a spino- Physiological and anatomical experiments have
bulbospinal micturition reflex pathway that passes provided substantial support for the concept that
71

Fig. 6. Diagram showing the organization of the parasympathetic excitatory reflex pathway to the detrusor muscle. Scheme is based on
electrophysiological studies in cats. In animals with an intact spinal cord, micturition is initiated by a supraspinal reflex pathway
passing through a center in the brain stem. The pathway is triggered by myelinated afferents (A-d fibers), which are connected to the
tension receptors in the bladder wall. Injury to the spinal cord above the sacral segments interrupts the connections between the brain
and spinal autonomic centers and initially blocks micturition. However, over a period of several weeks following cord injury, a spinal
reflex mechanism emerges, which is triggered by unmyelinated vesical afferents (C-fibers); the A-fiber-afferent inputs are ineffective.
The C-fiber reflex pathway is usually weak or undetectable in animals with an intact nervous system. Stimulation of the C-fiber bladder
afferents by instillation of ice water into the bladder (cold stimulation) activates voiding responses in people with spinal cord injury.
Capsaicin (20–30 mg, subcutaneously) blocks the C-fiber reflex in chronic spinal cats, but does not block micturition reflexes in intact
cats. Intravesical capsaicin also suppresses detrusor hyper-reflexia and cold-evoked reflexes in people with neurogenic bladder dys-
function.

neuronal circuitry in the pontine micturition cen- putative inhibitory transmitters into the pontine
ter functions as a switch in the micturition reflex micturition center of the cat can increase the vol-
pathway. The switch seems to regulate bladder ume threshold for inducing micturition and in
capacity and also coordinate the activity of the high doses completely block reflex voiding, indi-
bladder and external urethral sphincter. Electrical cating that synapses in this region are important
or chemical stimulation in the pontine micturition for regulating the set point for reflex voiding and
center of the rat, cat and dog induces: (1) a sup- also are an essential link in the reflex pathway
pression of urethral EMG, (2) firing of sacral (Mallory et al., 1991). Brain imaging studies in
preganglionic neurons, (3) bladder contractions humans using positron emission tomography
and (4) release of urine (Torrens and Morrison, (PET) or functional magnetic resonance imaging
1986; Mallory et al., 1989; de Groat et al., 1993; have identified increased neuronal activity in the
Blok, 2002). On the other hand, microinjections of pontine micturition center and PAG during
72

voiding (Blok et al., 1997, 1998; Athwal et al., Yoshimura, 2001). Glutamic acid, which is the
1999, 2001). major excitatory transmitter in the central nervous
system, has an important role in the control of the
micturition reflex. Experiments in rats indicate
Suprapontine control of micturition
that glutamatergic transmission in the spinal cord
is essential for bladder and urethral reflexes and
The organization of suprapontine pathways con-
for the spinal processing of afferent input from the
trolling micturition is less well defined, despite the
bladder. Both NMDA and a-amino-3-hydroxy-
fact that there is a large body of literature dealing
5-methyl-4-isoxazoleproprionic acid (AMPA)/
with the responses of the lower urinary tract to
kainate receptors are involved in glutamatergic
lesions or electrical stimulation of the brain (de
transmission in the micturition reflex pathway
Groat et al., 1993). In brief, it appears that the
(Yoshiyama et al., 1993, 1994, 1995, 1997; Sugaya
voluntary control of micturition is dependent
and de Groat, 1994; Matsumoto et al., 1995a, b).
upon (1) connections between the frontal cortex
A study using spinal slice preparations from neo-
and the septal and the preoptic regions of the
natal rats also revealed that sacral preganglionic
hypothalamus and (2) connections between the
neurons directly receive glutamatergic excitatory
paracentral lobule and the brain stem and spinal
inputs through NMDA and AMPA/kainate re-
cord (Torrens and Morrison, 1986; de Groat et al.,
ceptors from spinal interneurons in the region of
1993). Lesions to these areas of cortex resulting
the sacral parasympathetic nucleus (Araki and de
from tumors, aneurysms or cerebrovascular
Groat, 1996). Thus, it is likely that glutamatergic
disease, appear to remove inhibitory control over
transmission is important at various sites in the
the anterior hypothalamic area that normally
micturition reflex pathway.
provides an excitatory input to micturition
The spinobulbospinal micturition reflex path-
centers in the brainstem (Yokoyama et al., 2000).
way controlling bladder and urethral reflexes is
Human PET scan studies have revealed that two
also modulated by various neurotransmitters such
cortical areas (the right dorsolateral prefrontal
as norepinephrine (Yoshimura et al., 1990a, b;
cortex and the anterior cingulate gyrus) were
Ishizuka et al., 1996), dopamine (Yoshimura et al.,
active during voiding (Blok et al., 1997, 1998;
1993, 1998b; Seki et al., 2001), 5-hydroxytrypta-
Blok, 2002). The hypothalamus including the
mine (Thor et al., 1990; Steers et al., 1992a; Espey
preoptic area as well as the pons and the PAG
and Downie, 1995; Danuser and Thor, 1996),
also showed activity in concert with voluntary
gamma aminobutyric acid (Steers et al., 1992b;
micturition. Other PET studies that examined the
Igawa et al., 1993; Araki, 1994), acetylcholine
changes in brain activity during filling of the
(Sugaya et al., 1987; Ishiura et al., 2001) and
bladder revealed that increased activity occurred in
neuropeptides, tachykinins, enkephalins (Booth
the PAG, the midline pons, the mid-cingulate
et al., 1985; Noto et al., 1991a), vasoactive intesti-
gyrus and bilaterally in the frontal lobes (Athwal
nal polypeptide (de Groat et al., 1990) and PACAP
et al., 1999, 2001; Matsuura et al., 2002). These
(Ishizuka et al., 1995), acting through different re-
results are consistent with the notion that the PAG
ceptor subtypes (de Groat and Yoshimura, 2001).
receives information about bladder fullness and
then relays this information to other brain areas
involved in the control of bladder storage.
Neurogenic dysfunction of the lower urinary tract

Supraspinal and spinal neurotransmitters Neurogenic disturbances of micturition can be


controlling micturition classified into two general categories: failure to
store and failure to eliminate urine (Wein, 2002).
Various neurotransmitters at the spinal and Problems with storage occur with differing degrees
supraspinal level are involved in regulation of of severity, ranging from reduced bladder capacity
micturition and continence (de Groat and and frequency of urination to urgency and
73

incontinence. A common finding is that disorders powerful and cause urinary incontinence. In addi-
affecting the brain, particularly suprapontine areas, tion, the bladder is usually only emptied partially
produce hyperactive or uninhibited bladders. owing to development of simultaneous contrac-
Cerebrovascular accidents, Parkinson’s disease, tions of the bladder and the striated urethral
tumors or demyelinating diseases are common sphincter (detrusor–sphincter dyssynergia, Fig. 2C).
causes of this problem (Betts, 1999; Sakakibara Inefficient voiding may also be due to unsustained
and Fowler, 1999; Wein, 2002). bladder contractions.
Failure to eliminate urine occurs under various In normal micturition, activation of the pontine
conditions that interrupt the detrusor to detrusor micturition center simultaneously induces bladder
excitatory reflex pathway or that interfere with the contractions and a suppression of sphincter activ-
coordination between detrusor and sphincters ity. A complete suprasacral lesion interrupts this
(Chancellor and Yoshimura, 2002; Wein, 2002). coordination between the bladder and the striated
Areflexic bladders can occur with (1) damage to sphincter (Fig. 2C). Thus, cord-injured people with
the pelvic nerve or the sacral spinal cord (lower chronic upper motoneuron lesions exhibit (1)
motor neuron lesions), (2) lesions of the afferent detrusor hyperreflexia, (2) coordinated relaxation
pathways (e.g., diabetes, tabes dorsalis, pernicious of urethral sphincter smooth muscle and (3)
anemia, herniated intervertebral disc) or (3) the dyssynergic contraction of urethral sphincter
acute stage of spinal cord injury rostral to the striated muscle (detrusor–sphincter dyssynergia).
sacral segments (an upper motor neuron lesion) All patterns of detrusor hyperreflexia associated
(Fam and Yalla, 1988; Chancellor and Blaivas, with detrusor-sphincter dyssynergia lead to high
1996; Yoshimura, 1999). intravesical pressure and/or severe bladder
trabeculation with the formation of diverticula,
which often induce vesicoureteral reflux and dete-
Spinal cord injury rostral to the lumbosacral level rioration of the upper urinary tract (Fam and
Yalla, 1988; Chancellor and Blaivas, 1996).
The upper motoneuron type of spinal cord injury People with spinal cord injury at the level of T6
initially leads to a phase of spinal shock that is or higher, often exhibit autonomic dysreflexia,
followed by a recovery phase during which neu- which is characterized by arterial pressor responses
rological changes emerge. During the period of induced by stimuli below the level of spinal cord
spinal shock immediately after spinal cord injury, lesion such as bladder distension, fecal impaction
there is a flaccid paralysis and absence of reflex or bladder inflammation (Fam and Yalla, 1988).
activity below the level of lesion; thus the urinary These stimuli cause excitation of sympathetic
bladder becomes areflexic. However, activity of pathways and induce arteriolar vasoconstriction
striated and smooth muscle sphincters rapidly re- and hypertension as well as piloerection and
covers after suprasacral injuries. Because sphincter sweating below the level of injury.
tone is present, urinary retention develops and
cord-injured patients have to be treated with in-
termittent or continuous catheterization to elimi- Spinal cord injury at or below the sacral level
nate urine from the urinary bladder. Following the
spinal shock phase, reflex detrusor activity reap- In the lower motoneuron type of spinal cord
pears after 2–12 weeks in most cases (Fam and injury, complete lesions of the sacral cord or the
Yalla, 1988; Chancellor and Blaivas, 1996). cauda equina usually result in flaccidity of the
During the recovery phase, the detrusor devel- bladder and its outlet. The bladder becomes
ops involuntary reflex contractions (neurogenic areflexic, thereby bladder compliance and bladder
detrusor overactivity) in response to visceral stim- capacity are increased. Pressures in the striated
uli such as bladder filling or suprapubic manual urethral sphincter are decreased. When the lesion
compression. After the recovery period, these extends to the thoracolumbar spinal cord, the
involuntary bladder contractions become more sympathetic outflow to the internal smooth muscle
74

sphincter of the urethra is also damaged, and the although voiding contractions in spinal cord-
bladder neck becomes incompetent in association injured rats were still triggered by capsaicin-
with hypoactive detrusor and striated sphincter resistant Ad-fiber afferents (Cheng et al., 1995;
(Fam and Yalla, 1988; Chancellor and Blaivas, Cheng and de Groat, 2004). Capsaicin treatment
1996; Yoshimura, 1999). also eliminated detrusor-sphincter dyssynergia in
anesthetized chronic spinal rats. Clinical studies
demonstrated that C-fiber afferents innervating
Changes in functions of bladder afferent pathways the bladder are involved in detrusor hyperreflexia
after spinal cord injury and autonomic dysreflexia in spinal cord-injured
people (Geirsson et al., 1995; Igawa et al., 1996,
A slow recovery of lower urinary tract function 2003; Cruz et al., 1997) and detrusor hyperreflexia
following spinal cord injury is also observed in in people with multiple sclerosis (Fowler et al.,
animals such as cats and rats (de Groat et al., 1992; Szallasi and Fowler, 2002). Taken together,
1990; Kruse et al., 1993; de Groat, 1995). Com- it is clear that the functional properties of C-fiber
plete transection of the thoracic spinal cord ini- afferents in the bladder are altered following spinal
tially produces detrusor areflexia in both species, cord injury, thereby inducing hyperreflexic bladder
followed by the emergence of detrusor hype- activity in both humans and animals (Fig. 6).
rreflexia with detrusor-sphincter dyssynergia. Other evidence of a reorganization of C-
Electrophysiological and pharmacological studies fiber-mediated reflex pathways in subjects with
have shown that the reflex pathways controlling suprasacral spinal cord injury was obtained in
the lower urinary tract are markedly different be- studies of the cold stimulation-evoked voiding re-
tween spinal intact and spinal-injured animals. In flex. Instillation of cold water into the bladder in
spinal intact cats and rats, the micturition reflex is these subjects induces reflex voiding (the Bors Ice
mediated by a long-latency supraspinal reflex Water Test) (Bors and Comarr, 1971; Geirsson
pathway, passing through the pons, that is acti- et al., 1993, 1995). This reflex does not occur in
vated by myelinated Ad-fiber bladder afferents normal subjects, except for infants (Geirsson et al.,
(de Groat et al., 1981, 1993, 1998; Mallory et al., 1994). It has been shown in the cat that C-fiber
1989; Yoshimura, 1999). However, in chronic spi- bladder afferents are responsible for cold-induced
nal cats the afferent limb of the micturition reflex bladder reflexes (Fall et al., 1990). Intravesical ad-
consists of unmyelinated C-fiber afferents (Fig. 6). ministration of capsaicin to paraplegic people
It has also been demonstrated that in chronic spi- blocks the cold-induced bladder reflexes, indicat-
nal cats (3–6 weeks after injury), subcutaneously ing that they are mediated by C-fiber afferents
administered capsaicin, a C-fiber neurotoxin, com- (Geirsson et al., 1995, Igawa et al., 1996, 2003).
pletely blocked reflex bladder contractions induced Studies in rats revealed that cold stimulation in-
by bladder distention, whereas capsaicin had no duced detrusor-sphincter dyssynergia and that
inhibitory effects on reflex bladder contractions in capsaicin treatment prevented it (Cheng et al.,
spinal intact cats (de Groat et al., 1990; Cheng 1997). Thus, cold- and capsaicin-sensitive C-fiber
et al., 1999). Thus, it is plausible that C-fiber bladder afferents can evoke detrusor hyperreflexia
bladder afferents that usually do not respond to and detrusor-sphincter dyssynergia. These re-
bladder distention (i.e., silent C-fibers) (Habler sponses are facilitated after the elimination of
et al., 1990) become mechano-sensitive and initiate supraspinal controls by spinal cord injury (Fig. 6).
automatic micturition after spinal cord injury.
Increased excitability of C-fiber afferents after
spinal cord injury has also been demonstrated in Changes in the firing properties of bladder afferent
rats in which detrusor hyperreflexia was identified neurons following spinal cord injury
during cystometrograms as non-voiding bladder
contractions prior to micturition. The non-voiding The mechanisms for inducing hyperexcitability of
contractions were suppressed by capsaicin, C-fiber bladder afferents were investigated by
75

whole-cell patch-clamp recording in dissociated channels from the tetrodotoxin-resistant type to


dorsal root ganglion neurons innervating the rat the tetrodotoxin-sensitive type. Since tetrodotoxin-
urinary bladder (Yoshimura and de Groat, 1997). sensitive Na+ currents have a lower threshold for
Chronic spinal cord injury in rats produced hyper- activation than tetrodotoxin-resistant currents, it
trophy of bladder afferent neurons as reflected by is reasonable to assume that these changes in ex-
an increase in cell diameter and cell input capac- pression of Na+ channels in bladder afferent neu-
itance. This confirmed earlier findings that bladder rons after spinal cord injury contribute to a low
afferent neurons in the L6-S1 (1st sacral) dorsal threshold for spike activation in these neurons.
root ganglia undergo somal hypertrophy (45–50% In chronic spinal cord-transected rats, bladder
increase in cross-sectional area) in chronic spinal afferent neurons with tetrodotoxin-sensitive spikes
cord-transected rats (Kruse et al., 1995). In addi- exhibited no apparent membrane potential relax-
tion to neuronal hypertrophy, bladder afferent ation when the neurons were gradually depolar-
neurons in chronic spinal rats increased their ex- ized by injecting inward currents. In these neurons,
citability. In contrast to the majority (approxi- voltage responses induced by current injections
mately 70%) of bladder afferent neurons from were not altered by application of 4-aminopyri-
spinal cord intact rats that exhibited high thresh- dine, a K+ channel blocker, although the neurons
old tetrodotoxin-resistant humped action poten- with tetrodotoxin-resistant spikes still had 4-ami-
tials (Yoshimura et al., 1996), 60% of bladder nopyridine-sensitive membrane potential relaxa-
afferent neurons in chronic spinal cord-transected tion during depolarization as found in spinal cord
rats exhibited tetrodotoxin-sensitive low-threshold intact rats. The phenomenon of membrane poten-
action potentials. The mean threshold for spike tial relaxation is due to slowly inactivating IA cur-
activation in cord-transected rats ( 25.5 mV) was rents that can be elicited by depolarization from
21% lower than in intact animals ( 20 mV). the resting membrane potential (Yoshimura et al.,
1996; Yoshimura, 1999). Therefore it is likely that
Plasticity in Na+ and K+ channels of bladder following spinal cord injury A-type K+ channels
afferent neurons following spinal cord injury are suppressed in parallel with an increased ex-
pression of tetrodotoxin-sensitive Na+ currents,
The alteration of electrophysiological properties in thereby increasing excitability of bladder afferent
bladder afferent neurons after spinal cord injury neurons. Since tetrodotoxin-resistant Na+ cur-
was also reflected in changes in density of different rents and IA currents are preferentially expressed
types of Na+ currents (Yoshimura and de Groat, in small-sized C-fiber afferent neurons in spinal
1997; Black et al., 2003). Consistent with the in- cord intact rats (Gold et al., 1996a; Yoshimura
crement in the proportion of neurons with tetrodo- et al., 1996), the changes in these channels after
toxin-sensitive spikes, the number of bladder spinal cord injury must occur primarily in C-fiber
afferent neurons that predominantly expressed bladder afferent neurons and contribute to in-
tetrodotoxin-sensitive Na+ currents (60–100% of creased cell excitability of these neurons.
total Na+ currents) also increased. The density of Immunohistochemical studies using antibodies to
tetrodotoxin-sensitive Na+ currents in bladder af- tetrodotoxin-resistant Na+ channel protein (Nav
ferent neurons significantly increased from 32.1 to 1.8) revealed that tetrodotoxin-resistant Na+
80.6 pA/pF, while tetrodotoxin-resistant current channels are located not only in small-sized dor-
density decreased from 60.5 to 17.9 pA/pF fol- sal root ganglion cell bodies, but also in superficial
lowing spinal cord injury. In addition, an increase laminae of the dorsal horn of the spinal cord
in tetrodotoxin-sensitive Na+ currents was detect- (Novakovic et al., 1998; Yoshimura et al., 2001).
ed in some bladder afferent neurons that still re- Thus changes occurring in afferent cell bodies fol-
tained a predominance of tetrodotoxin-resistant lowing spinal cord injury may also occur at affer-
currents (450% of total Na+ currents) after spi- ent axons and/or receptors in the bladder and thus
nal cord injury. These data indicate that spinal contribute to the emergence of the C-fiber-
cord injury induces a switch in expression of Na+ mediated spinal micturition reflex.
76

and detrusor-sphincter dyssynergia after spinal


cord injury (Fig. 7).
In addition, we have recently found that in-
creased NGF in the spinal cord after spinal cord
injury is also responsible for inducing hyperexcit-
ability of C-fiber bladder afferent pathways, and
that intrathecal application of NGF antibodies,
which neutralized NGF in the spinal cord, sup-
pressed detrusor hyperreflexia and detrusor-
sphincter dyssynergia in spinal cord injured rats
(Seki et al., 2002, 2004a, b). Intrathecal adminis-
Fig. 7. Diagram of hypothetical mechanisms inducing lower
urinary tract dysfunction following spinal cord injury (SCI).
tration of NGF antibodies also reportedly blocked
The subsequent events occurring after SCI are indicated by the autonomic dysreflexia in paraplegic rats (Krenz
numbers 1–7. Injury to the spinal cord (1) causes detrusor- et al., 1999). Thus, NGF and its receptors in the
sphincter dyssynergia (DSD) (2) leading to functional obstruc- bladder and/or the spinal cord are potential targets
tion of urethra. Increased urethral resistance induces bladder for new therapies to suppress detrusor hype-
hypertrophy (3), resulting in increased levels of nerve growth
factor (NGF, 4) in the bladder smooth muscle. The nerve
rreflexia and detrusor-sphincter dyssynergia after
growth factor (NGF) level in the spinal cord (5) is also in- spinal cord injury (Fig. 7).
creased after SCI. Increased NGF in the bladder and spinal
cord is transported to bladder afferent pathways (6), followed
by hyperexcitability of bladder afferent pathways (7). Hyper- Spinal mechanisms involving vasoactive intestinal
excitability of bladder afferent pathways causes or enhances
neurogenic detrusor overactivity (8) and DSD (2).
polypeptide (VIP) and pituitary adenylate cyclase
activating polypeptide (PACAP)

Role of neurotrophic factors Vasoactive intestinal polypeptide (VIP) and


pituitary adenylate cyclase activating polypeptide
Nerve growth factor (NGF) has been implicated as (PACAP) are contained in afferent neurons inner-
a chemical mediator of pathology-induced changes vating the urinary bladder of the cat and rat
in C-fiber afferent nerve excitability and reflex (Kawatani et al., 1985; see Vizzard, this volume,
bladder activity (Yoshimura, 1999; Vizzard, 2000). for references). In the cat sacral spinal cord, VIP
It has been demonstrated that chronic administra- is present exclusively in C-fiber afferent axons
tion of NGF into the bladder of rats induced (Morgan et al., 1999), and is located in afferent ter-
bladder hyperactivity and increased the firing fre- minals projecting to the sacral parasympathetic nu-
quency of dissociated bladder afferent neurons cleus (Kawatani et al., 1985). In chronic spinal cats,
(Yoshimura et al., 1999), and that the production VIP-immunoreactivity is distributed over a wider
of neurotrophic factors including NGF increased area of the lateral dorsal horn in the sacral spinal
in the bladder after spinal cord injury (Vizzard, cord, suggesting C-fiber afferent axonal sprouting
2000, this volume). It has also been shown that the after spinal injury (Thor et al., 1986). In addition,
bladder hyperactivity and hypertrophy of afferent the effects of intrathecal administration of VIP are
and efferent neurons innervating the hypertrophic changed. In normal cats, VIP inhibits the micturit-
bladder in rats with partial urethral obstruction ion reflex, whereas in paraplegic cats VIP facilitates
was antagonized in part by systemic autoimmuni- the micturition reflex (de Groat et al., 1990). These
zation against NGF (Steers et al., 1996). Thus it findings suggest that the action of a putative C-fiber
seems that target organ–neural interactions medi- afferent transmitter may underlie the emergence of
ated by neurotrophic factors such as NGF pro- C-fiber bladder reflexes in the paraplegic cat.
duced in the hypertrophied bladder muscle may In normal and spinal cord-injured rats, VIP and
contribute to changes in C-fiber bladder afferent PACAP, another member of the secretin/glucagon/
pathways that underlie the detrusor hyperreflexia VIP peptide family, facilitated the micturition
77

reflex by actions on the spinal cord (Ishizuka et al., mRNAs levels of GluR-A and GluR-B AMPA
1995; Yoshiyama and de Groat, 1997; Vizzard, receptor subunits and NR1, but not NR2 NMDA
this volume). Patch-clamp studies in the neonatal receptor subunits (Shibata et al., 1999). On the
rat spinal slice preparation revealed that PACAP other hand, motoneurons in the urethral sphincter
has a direct excitatory action on parasympathetic nucleus express all four AMPA receptor subunits
preganglionic neurons due in part to blockade of (GluR-A, -B, -C and -D) in conjunction with
K+ channels and also has an indirect action med- moderate amounts of NR2A and NR2B as well as
itated by activation of excitatory interneurons high levels of NR1 receptor subunits. It seems
(Miura et al., 2001b). PACAP increased the fre- likely that this difference in expression accounts
quency of spontaneous excitatory postsynaptic for the different sensitivity of bladder and sphinc-
potentials as well as spontaneous firing and de- ter reflexes to glutamatergic antagonists.
creased the threshold for action potential genera-
tion. PACAP also increased the number and Spinal tachykinin mechanisms
frequency of action potentials elicited by depolar-
izing current pulses. PACAP levels in bladder Tachykinins, such as substance P and neurokinin
afferent neurons and in spinal cord projections are A, that are released at C-fiber afferent terminals
upregulated after spinal cord injury (Zvarova can act in the bladder wall to modulate the excit-
et al., 2005; Vizzard, this volume). These findings ability of afferent nerves and induce bladder con-
suggest that putative C-fiber afferent transmitters, tractions (de Groat et al., 1993; Morrison et al.,
such as VIP in the cat or PACAP in the rat, may 2002), and are also thought to be involved in the
underlie the emergence of C-fiber afferent evoked spinal cord as mediators of excitatory transmission
bladder reflexes after spinal cord injury. between primary afferent nerves and second-order
spinal neurons that express NK1 receptors. Al-
Spinal glutamatergic mechanisms though some studies have reported an increase in
bladder capacity and a decrease in maximal void-
Glutamic acid plays an essential role as an exci- ing pressure after intrathecal administration of
tatory transmitter in the spinal reflex pathways NK1 receptor antagonists in normal conscious or
controlling bladder and external urethral sphincter anesthetized rats, other experiments have failed to
activity in both normal and spinal cord-injured detect a change in bladder function after admin-
rats. A study of the effect of selective antagonists istration of these agents in normal rats (de Groat
revealed that NMDA and non-NMDA glut- and Yoshimura, 2001). On the other hand, a re-
amatergic (AMPA) receptors are involved in the duction in detrusor hyperreflexia in spinal cord-
supraspinal and spinal reflex pathways controlling injured rats has been noted after administration of
voiding, and that AMPA receptor mechanisms are NK-1 and NK-2 tachykinin receptor antagonists
most important. Intrathecal or intravenous ad- (Abdel-Gawad et al., 2001). The role of NK1 re-
ministration of NMDA or AMPA antagonists in ceptor-expressing spinal neurons in C-fiber-affer-
urethane-anesthetized rats depressed bladder con- ent mediated bladder activity was also evaluated
tractions and electromyographic activity of the by destroying these neurons in the L6-S1 spinal
external urethral sphincter (Yoshiyama et al., cord by intrathecal administration of a ribosome-
1993, 1995). In spinal cord injured-rats, external inactivating toxin, saporin conjugated with a
urethral sphincter electromyographic activity was specific NK1 receptor ligand that promotes the
more sensitive than bladder reflexes to glut- binding and internalization of the toxin in NK1
amatergic antagonists (Yoshiyama et al., 1993, receptor-expressing neurons. In treated animals,
1997), raising the possibility that the two reflex NK1 receptor immunoreactivity was reduced in
pathways have different types of receptors. This lamina I of the spinal cord and the bladder
was confirmed with in situ hybridization tech- hyperactivity induced by intravesical instillation
niques which revealed that sacral parasympathetic of capsaicin was reduced, but cystometric param-
preganglionic neurons in the rat express high eters in awake rats were not changed (Seki et al.,
78

2005). Preliminary experiments in spinal cord-in- functions are regulated by a complex neural con-
jured rats indicate that detrusor hyperreflexia is trol system located in the brain and spinal cord.
also reduced by saporin treatment (Seki et al., This control system performs like a simple switch-
2004a). These studies raise the possibility that NK1 ing circuit to maintain a reciprocal relationship
receptor antagonists might be useful clinically in between the reservoir (bladder) and outlet compo-
treating bladder dysfunction in people with spinal nents (urethra and urethral sphincter) of the uri-
cord injury. nary tract. Spinal cord injury disrupts voluntary
control and the normal reflex pathways that coor-
Peripheral muscarinic mechanisms dinate bladder and sphincter function. Studies in
animals indicate that recovery of bladder function
Muscarinic receptor antagonists are the first line following spinal cord injury is dependent upon the
therapy for detrusor hyperreflexia induced by spi- reorganization of reflex pathways in both the pe-
nal cord injury because bladder contractions are ripheral and central nervous system. Part of this
induced by activation of postjunctional muscarinic reorganization may be influenced by neural-target
receptors in the detrusor muscle by acetylcholine organ interactions that are mediated by neurotro-
released from parasympathetic postganglionic phic factors released by the peripheral organs.
nerves (Kim et al., 1997; Stohrer et al., 1999;
Chapple, 2000). Although the M2 receptor is the
predominant subtype in the bladder (approximate- Acknowledgments
ly 80%), the contractions of the bladder are me-
diated by the M3 receptor subtype, which is The authors’ research is supported by NIH
therefore considered the primary target of drug research grants (DK49430, DK 57267, P01 HD
therapy (Hegde and Eglen, 1999; Chapple, 2000). 39768).
However, other receptors may play a role in trig-
gering bladder activity after spinal cord injury. References
M1 muscarinic receptors are located prejunc-
tionally on cholinergic nerve terminals in the blad- Abdel-Gawad, M., Dion, S.B. and Elhilali, M.M. (2001)
der (Somogyi and de Groat, 1999). Activation of Evidence of a peripheral role of neurokinins in detrusor
these receptors facilitates ACh release during the hyperreflexia: a further study of selective tachykinin antag-
prolonged, high-frequency nerve firing that would onists in chronic spinal injured rats. J. Urol., 165: 1739–1744.
Andersson, K.E. (1993) Pharmacology of lower urinary tract
occur during voiding. This amplification mecha- smooth muscle and penile erection tissues. Pharmacol. Rev.,
nism through pre-junctional M1 receptors is up- 45: 253–308.
regulated in spinal cord injured rats, suggesting Andersson, K.E. and Arner, A. (2004) Urinary bladder con-
that detrusor hyperreflexia occurring after spinal traction and relaxation: physiology and pathology. Physiol.
cord injury may be due in part to an enhancement Rev., 84: 935–986.
Araki, I. (1994) Inhibitory postsynaptic currents and the effects
of ACh release from parasympathetic nerve ter- of GABA on visually identified sacral parasympathetic
minals (Somogyi et al., 1998; Somogyi and de preganglionic neurons in neonatal rats. J. Neurophysiol.,
Groat, 1999). Activation of postjunctional M2 re- 72: 2903–2910.
ceptors also elicits a direct contraction of the rat Araki, I. and de Groat, W.C. (1996) Unitary excitatory synaptic
bladder smooth muscle after spinal cord injury currents in preganglionic neurons mediated by two distinct
groups of interneurons in neonatal rat sacral parasympa-
(Braverman et al., 1999). Thus, antimuscarinic thetic nucleus. J. Neurophysiol., 76: 215–226.
agents may act by multiple mechanisms to reduce Araki, I. and de Groat, W.C. (1997) Developmental synaptic
detrusor hyperreflexia. depression underlying reorganization of visceral reflex path-
ways in the spinal cord. J. Neurosci., 17: 8402–8407.
Athwal, B.S., Berkley, K.J., Brennan, A., Craggs, M., Fowler,
Conclusions
C.J., Hussain, I., Sakakibara, R. and Frackowiak, R.S.J.
(1999) Brain activity associated with the urge to void and
The lower urinary tract has two main functions: bladder fill volume in normal men: preliminary data from a
storage and periodic elimination of urine. These PET study. BJU Int., 84: 148–149.
79

Athwal, B.S., Berkley, K.J., Hussain, I., Brennan, A., Craggs, Blok, B.F. and Holstege, G. (1994) Direct projections from the
M., Sakakibara, R., Frackowiak, R.S.J. and Fowler, C.J. periaqueductal gray to the pontine micturition center
(2001) Brain responses to changes in bladder volume and (M-region). An anterograde and retrograde tracing study in
urge to void in healthy men. Brain, 124(2): 369–377. the cat. Neurosci. Lett., 166: 93–96.
Bahns, E., Ernsberger, U., Janig, W. and Nelke, A. (1998) Blok, B.F., Sturms, L.M. and Holstege, G. (1998) Brain
Functional characteristics of lumbar visceral afferent fibers activation during micturition in women. Brain,
from the urinary bladder and urethra in the cat. Eur. J. 121(Part 11): 2033–2042.
Physiol., 407: 510. Blok, B.F., Willemsen, A.T. and Holstege, G. (1997) A PET
Barrington, F.J.F. (1925) The effect of lesions of the hind-and study on brain control of micturition in humans. Brain, 120:
mid-brain on micturition in the cat. J. Exp. Physiol., 15: 111–121.
81–102. Blok, B.F.M. (2002) Brain control of the lower urinary tract.
Beattie, B.S., Li, Q., Leedy, G. and Bresnahan, J.C. (1990) Scand. J. Urol. Nephrol., 36(Suppl. 210): 11–15.
Motoneurons innervating the external anal and urethral Booth, A.M., Hisamitsu, T., Kawatani, M. and de Groat, W.C.
sphincters of the female cat have different patterns of den- (1985) Regulation of urinary bladder capacity by endogenous
dritic arborization. Neurosci. Lett., 111: 69–74. opioid peptides. J. Urol., 133: 339–342.
Betts, C.D. (1999) Bladder and sexual function in multiple Bors, E. and Comarr, A.E. (1971) Neurological Urology:
sclerosis. In: Fowler C.J. (Ed.), Neurology of Bladder, Bowel, Physiology of Micturition, Its Neurological Disorders and
and Sexual Dysfunction. Butterworth-Heinemann, Boston, Sequelae. University Park Press, Baltimore.
pp. 289–308. Braverman, A.S., Legos, J., Young, W., Luthin, G. and
Birder, L.A., Apodaca, G., de Groat, W.C. and Kanai, A.J. Ruggieri, M. (1999) M2 receptors in genito-urinary smooth
(1998) Adrenergic- and capsaicin-evoked nitric oxide release muscle pathology. Life Sci., 64: 429–436.
from urothelium and afferent nerves in urinary bladder. Am. Burnett, A.L., Calvin, D.C., Chamness, S.L., Liu, J.X., Nelson,
J. Physiol., 275: F226–F229. R.J., Klein, S.L., Dawson, V.L., Dawson, T.M. and Snyder,
Birder, L.A., Barrick, S.R., Roppolo, J.R., Kanai, A.J., de S.H. (1997) Urinary bladder-urethral sphincter dysfunction
Groat, W.C., Kiss, S. and Buffington, C.A. (2003) Feline in mice with targeted disruption of neuronal nitric oxide
interstitial cystitis results in mechanical hypersensitivity and synthase models idiopathic voiding disorders in humans.
altered ATP release from bladder urothelium. Am. J. Nat. Med., 3: 571–574.
Physiol., 285: F423–F429. Burnstock, G. (2001) Purinergic signaling in the lower urinary
Birder, L.A. and de Groat, W.C. (1993) Induction of c-fos gene tract. In: Abbracchio M.P. and Williams M. (Eds.), Handbook
expression in spinal neurons of the rat by nociceptive and of Experimental Pharmacology. Springer, Berlin, pp. 423–515.
non-nociceptive stimulation of the lower urinary tract. Am. J. Chancellor, M.B. and Blaivas, J.G. (1996) Spinal cord injury.
Physiol., 265: R643–R648. In: Chancellor M.B. and Blaivas J.G. (Eds.), Practical Ne-
Birder, L.A., Kanai, A.J., de Groat, W.C., Kiss, S., Nealen, uro-Urology. Genitourinary Complications in Neurologic
M.L., Burke, N.E., Dineley, K.E., Watkins, S., Reynolds, I.J. Disease. Butterworth-Heinemann, Stoneham, pp. 99–118.
and Caterina, M.J. (2001) Vanilloid receptor expression sug- Chancellor, M.B. and Yoshimura, N. (2002) Physiology and
gests a sensory role for urinary bladder epithelial cells. Proc. pharmacology of the bladder and urethra. In: Walsh P.C.,
Natl. Acad. Sci., U. S. A., 98: 13396–13401. Retik A.B., Vaughn E.D. and Wein A.J. (Eds.), Campbell’s
Birder, L.A., Nakamura, Y., Kiss, S., Nealen, M.L., Barrick, Urology, Chapter 23. W.B. Saunders Co., Philadelphia,
S., Kanai, A.J., Wang, E., Ruiz, G., de Groat, W.C., pp. 831–886.
Apodaca, G., Watkins, S. and Caterina, M.J. (2002) Altered Chapple, C.R. (2000) Muscarinic receptor antagonists in the
urinary bladder function in mice lacking the vanilloid recep- treatment of overactive bladder. Urology, 55: 33–46.
tor TRPV1. Nat. Neurosci., 5: 856–860. Cheng, C.L., Chai, C.Y. and de Groat, W.C. (1997) Detrusor-
Birder, L.A., Roppolo, J.R., Erickson, V.L. and de Groat, sphincter dyssynergia induced by cold stimulation of the
W.C. (1999) Increased c-fos expression in spinal lumbosacral urinary bladder of rats. Am. J. Physiol., 272: R1271–R1282.
projection and preganglionic neurons after irritation of the Cheng, C.L. and de Groat, W.C. (2004) The role of capsaicin-
lower urinary tract in the rat. Brain Res., 834: 55–65. sensitive afferent fibers in the lower urinary tract dysfunction
Black, J.A., Cummins, T.R., Yoshimura, N., de Groat, W.C. induced by chronic spinal cord injury in rats. Exp. Neurol.,
and Waxman, S.G. (2003) Tetrodotoxin-resistant sodium 187: 445–454.
channels Nav 1.8/SNS and Nav 1.9/NaN in afferent neurons Cheng, C.L., Liu, J.C., Chang, S.Y., Ma, C.P. and de Groat,
innervating urinary bladder in control and spinal cord in- W.C. (1999) Effect of capsaicin on the micturition reflex in
jured rats. Brain Res., 963: 132–138. normal and chronic spinal cord-injured cats. Am. J. Physiol.,
Blok, B.F., De Weerd, H. and Holstege, G. (1995) Ultrastruc- 277: R786–R794.
tural evidence for a paucity of projections from the Cheng, C.L., Ma, C.P. and de Groat, W.C. (1995) Effect of
lumbosacral cord to the pontine micturition center or M- capsaicin on micturition and associated reflexes in chronic
region in the cat: a new concept for the organization of the spinal rats. Brain Res., 678: 40–48.
micturition reflex with the periaqueductal gray as central re- Chuang, Y.C., Fraser, M.O., Yu, Y., Beckel, J.M., Seki, S.,
lay. J. Comp. Neurol., 359: 300–309. Nakanishi, Y., Yokoyama, H., Chancellor, M.B.,
80

Yoshimura, N. and de Groat, W.C. (2001) Analysis of the Delancey, J., Gosling, J., Creed, K., Dixon, J., Delmas, V.,
afferent limb of the vesicovascular reflex using neurotoxins, Landon, D. and Norton, P. (2002) Gross anatomy and cell
resiniferatoxin and capsaicin. Am. J. Physiol., 281: biology of the lower urinary tract. In: Abrams P., Cardozo
R1302–R1310. L., Khoury S. and Wein A. (Eds.), Incontinence. Health
Cockayne, D.A., Hamilton, S.G., Zhu, Q.M., Dunn, P.M., Publications, Ltd., Jersey, UK, pp. 17–82.
Zhong, Y., Novakovic, S., Malmberg, A.B., Cain, G., Espey, M.J. and Downie, J.W. (1995) Serotonergic modulation
Berson, A., Kassotakis, L., Hedley, L., Lachnit, W.G., of cat bladder function before and after spinal transection.
Burnstock, G., McMahon, S.B. and Ford, A.P. (2000) Eur. J. Pharmacol., 287: 173–177.
Urinary bladder hyporeflexia and reduced pain-related Fall, M., Lindstrom, S. and Mazieres, L. (1990) A bladder-to-
behaviour in P2X3-deficient mice. Nature, 407: 1011–1015. bladder cooling reflex in the cat. J. Physiol. (Lond.), 427:
Cruz, F., Guimaraes, M., Silva, C., Rio, M.E., Coimbra, A. and 281–300.
Reis, M. (1997) Desensitization of bladder sensory fibers by Fam, B. and Yalla, S.V. (1988) Vesicourethral dysfunction in
intravesical capsaicin has long lasting clinical and urody- spinal cord injury and its management. Semin. Neurol., 8:
namic effects in patients with hyperactive or hypersensitive 150–155.
bladder dysfunction. J. Urol., 157: 585–589. Ferguson, D.R., Kennedy, I. and Burton, T.J. (1997) ATP is
Danuser, H. and Thor, K.B. (1996) Spinal 5-HT2 receptor- released from rabbit urinary bladder cells by hydrostatic
mediated facilitation of pudendal nerve reflexes in the anaes- pressure changes–a possible sensory mechanism? J. Physiol.
thetized cat. Br. J. Pharmacol., 118: 150–154. (Lond.), 505: 503.
de Groat, W.C. (1975) Nervous control of the urinary bladder Fowler, C.J., Jewkes, D., McDonald, W.I., Lynn, B. and de
of the cat. Brain Res., 87: 201–211. Groat, W.C. (1992) Intravesical capsaicin for neurogenic
de Groat, W.C. (1987) Neuropeptides in pelvic afferent path- bladder dysfunction. Lancet, 339: 1239.
ways. Experientia, 43: 801–813. Geirsson, G., Fall, M. and Lindstrom, S. (1993) The ice-water
de Groat, W.C. (1995) Mechanisms underlying the recovery of test–a simple and valuable supplement to routine cystometry.
lower urinary tract function following spinal cord injury. Br. J. Urol., 71: 681–685.
Paraplegia, 33: 493–505. Geirsson, G., Fall, M. and Sullivan, L. (1995) Clinical and
de Groat, W.C., Araki, I., Vizzard, M.A., Yoshiyama, M., urodynamic effects of intravesical capsaicin treatment in pa-
Yoshimura, N., Sugaya, K., Tai, C. and Roppolo, J.R. (1998) tients with chronic traumatic spinal detrusor hyperreflexia. J.
Developmental and injury induced plasticity in the micturit- Urol., 154: 1825–1829.
ion reflex pathway. Behav. Brain Res., 92: 127–140. Geirsson, G., Lindstrom, S., Fall, M., Gladh, G., Hermansson,
de Groat, W.C. and Booth, A.M. (1993) Synaptic transmission G. and Hjalmas, K. (1994) Positive bladder cooling test
in pelvic ganglia. In: Maggi C.A. (Ed.), The Autonomic in neurologically normal young children. J. Urol., 151:
Nervous System, Vol. 3, Nervous Control of the Urogenital 446–448.
System, 1st Ed. Harwood Academic Publishers, London, Gold, M.S., Shuster, M.J. and Levine, J.D. (1996) Character-
pp. 291–347. ization of six voltage-gated K+ currents in adult rat sensory
de Groat, W.C., Booth, A.M., Milne, R.J. and Roppolo, J.R. neurons. J. Neurophysiol., 75: 2629–2646.
(1982) Parasympathetic preganglionic neurons in the sacral Habler, H.J., Janig, W. and Koltzenburg, M. (1990) Activation
spinal cord. J. Auton. Nerv. Syst., 5: 23–43. of unmyelinated afferent fibres by mechanical stimuli and
de Groat, W.C., Booth, A.M. and Yoshimura, N. (1993) inflammation of the urinary bladder in the cat. J. Physiol.
Neurophysiology of micturition and its modification in an- (Lond.), 425: 545–562.
imal models of human disease. In: Maggi C.A. (Ed.), The Hegde, S.S. and Eglen, R.M. (1999) Muscarinic receptor sub-
Autonomic Nervous System, Vol. 3, Chapter 8, Nervous types modulating smooth muscle contractility in the urinary
Control of the Urogenital System. Harwood Academic Pub- bladder. Life Sci., 64: 419–428.
lishers, London, UK, pp. 227–289. Ho, K.M., Ny, L., McMurray, G., Andersson, K.E., Brading,
de Groat, W.C., Kawatani, M., Hisamitsu, T., Cheng, C.L., A.F. and Noble, J.G. (1999) Co-localization of carbon mon-
Ma, C.P., Thor, K., Steers, W. and Roppolo, J.R. (1990) oxide and nitric oxide synthesizing enzymes in the human
Mechanisms underlying the recovery of urinary bladder urethral sphincter. J. Urol., 161: 1968–1972.
function following spinal cord injury. J. Auton. Nerv. Syst., Holstege, G., Griffiths, D., deWall, H. and Dalm, E. (1986)
30(Suppl.): S71–S77. Anatomical and physiological observations on supraspinal
de Groat, W.C. and Lalley, P.M. (1972) Reflex firing in the control of bladder and urethral sphincter muscles in cats. J.
lumbar sympathetic outflow to activation of vesical afferent Comp. Neurol., 250: 449.
fibres. J. Physiol. (Lond), 226: 289–309. Igawa, Y., Komiyama, I., Nishizawa, O. and Ogawa, A. (1996)
de Groat, W.C., Nadelhaft, I., Milne, R.J., Booth, A.M., Intravesical capsaicin inhibits autonomic dysreflexia in pa-
Morgan, C. and Thor, K. (1981) Organization of the sacral tients with spinal cord injury. Neurourol. Urodyn., 15: 374.
parasympathetic reflex pathways to the urinary bladder and Igawa, Y., Mattiasson, A. and Andersson, K.E. (1993) Effects
large intestine. J. Auton. Nerv. Syst., 3: 135–160. of GABA-receptor stimulation and blockade on micturition
de Groat, W.C. and Yoshimura, N. (2001) Pharmacology of the in normal rats and rats with bladder outflow obstruction. J.
lower urinary tract. Ann. Rev. Pharm. Toxicol., 41: 691–721. Urol., 150: 537–542.
81

Igawa, Y., Satoh, T., Mizusawa, H., Seki, S., Kato, H., Kruse, M.N., Noto, H., Roppolo, J.R. and de Groat, W.C.
Ishizuka, O. and Nishizawa, O. (2003) The role of capsaicin- (1990) Pontine control of the urinary bladder and external
sensitive afferents in autonomic dysreflexia in patients with urethral sphincter in the rat. Brain Res., 532: 182–190.
spinal cord injury. BJU Int., 91: 637–641. Kuru, M. (1965) Nervous control of micturition. Physiol. Rev.,
Ishiura, Y., Yoshiyama, M., Yokoyama, O., Namiki, M. and de 45: 425–494.
Groat, W.C. (2001) Central muscarinic mechanisms regulat- Lavelle, J.P., Meyers, S.A., Ruiz, W.G., Buffington, C.A.,
ing voiding in rats. J. Pharmacol. Exp. Ther., 297: 933–939. Zeidel, M.L. and Apodaca, G. (2000) Urothelial pathophys-
Ishizuka, O., Alm, P., Larsson, B., Mattiasson, A. and And- iological changes in feline interstitial cystitis: a human model.
ersson, K.E. (1995) Facilitatory effect of pituitary adenylate Am. J. Physiol., 278: F540–F553.
cyclase activating polypeptide on micturition in normal, con- Lee, H.Y., Bardini, M. and Burnstock, G. (2000) Distribution
scious rats. Neuroscience, 66: 1009–1014. of P2X receptors in the urinary bladder and the ureter of the
Ishizuka, O., Mattiasson, A. and Andersson, K.E. (1996) Role rat. J. Urol,, 163: 2002–2007.
of spinal and peripheral alpha 2 adrenoceptors in micturition Lewis, S.A. (2000) Everything you wanted to know about the
in normal conscious rats. J. Urol., 156: 1853–1857. bladder epithelium but were afraid to ask. Am. J. Physiol.,
Janig, W. and Morrison, J.F. (1986) Functional properties of 278: 867–874.
spinal visceral afferents supplying abdominal and pelvic or- Maggi, C.A. (1993) The dual sensory and efferent functions of the
gans, with special emphasis on visceral nociception. Prog. capsaicin-sensitive primary sensory neurons in the urinary blad-
Brain. Res., 67: 87–114. der and urethra. In: Maggi C.A. (Ed.), The Autonomic Nervous
Jankowski, R.J., Prantil, R.L., Fraser, M.O., Chancellor, M.B., System, Vol. 3, Chapter 13, Nervous Control of the Urogenital
de Groat, W.C., Huard, J. and Vorp, D.A. (2004) Develop- System. Harwood Academic Publishers, London, pp. 383–422.
ment of an experimental system for the study of urethral Mallory, B., Steers, W.D. and de Groat, W.C. (1989) Electro-
biomechanical function. Am. J. Physiol., 286: F225–F232. physiological study of micturition reflexes in rats. Am. J.
Kakizaki, H., Fraser, M.O. and de Groat, W.C. (1997) Reflex Physiol., 257: R410–R421.
pathways controlling urethral striated and smooth muscle Mallory, B.S., Roppolo, J.R. and de Groat, W.C. (1991) Phar-
function in the male rat. Am. J. Physiol., 272: R1647–R1656. macological modulation of the pontine micturition center.
Kamo, I., Cannon, T.W., Conway, D.A., Torimoto, K., Brain Res., 546: 310–320.
Chancellor, M.B., de Groat, W.C. and Yoshimura, N. Matsumoto, G., Hisamitsu, T. and de Groat, W.C. (1995a)
(2004) The role of bladder-to urethral reflexes in urinary Non-NMDA glutamatergic excitatory transmission in the
continence mechanisms in rats. Am. J. Physiol., 287: descending limb of the spinobulbospinal micturition reflex
F434–F441. pathway of the rat. Brain Res., 693: 246–250.
Kamo, I., Torimoto, K., Chancellor, M.B., de Groat, W.C. and Matsumoto, G., Hisamitsu, T. and de Groat, W.C. (1995b)
Yoshimura, N. (2003) Urethral closure mechanisms under Role of glutamate and NMDA receptors in the descending
sneeze-induced stress condition in rats: a new animal model limb of the spinobulbospinal micturition reflex pathway of
for evaluation of stress urinary incontinence. Am. J. Physiol., the rat. Neurosci. Lett., 18: 58–61.
285: R356–R365. Matsuura, S., Kakizaki, H., Mitsui, T., Shiga, T., Tamaki, N.
Kawatani, M., Erdman, S. and de Groat, W.C. (1985) Vaso- and Koyanagi, T. (2002) Human brain region response to
active intestinal polypeptide and substance P in afferent distention or cold stimulation of the bladder: a positron
pathways to the sacral spinal cord of the cat. J. Comp. emission tomography study. J. Urol., 168: 2035–2039.
Neurol., 241: 327–347. McMahon, S.B., Armanini, M.P., Ling, L.H. and Phillips, H.S.
Keast, J.R. and de Groat, W.C. (1992) Segmental distribution (1994) Expression and coexpression of Trk receptors in sub-
and peptide content of primary afferent neurons innervating populations of adult primary sensory neurons projecting to
the urogenital organs and colon of male rats. J. Comp. identified peripheral targets. Neuron, 12: 1161–1171.
Neurol., 319: 615–623. Miura, A., Kawatani, M. and de Groat, W.C. (2001a) Excita-
Kim, Y.H., Bird, E.T., Priebe, M. and Boone, T.B. (1997) The tory synaptic currents in lumbosacral parasympathetic pre-
role of oxybutynin in spinal cord injured patients with ganglionic neurons elicited from the lateral funiculus. J.
indwelling catheters. J. Urol., 158: 2083–2086. Neurophysiol., 86: 1587–1593.
Krenz, N.R., Meakin, S.O., Krassioukov, A.V. and Weaver, Miura, A., Kawatani, M. and de Groat, W.C. (2001b) Effect of
L.C. (1999) Neutralizing intraspinal nerve growth factor pituitary adenylate cyclase activating polypeptide on lumbo-
blocks autonomic dysreflexia caused by spinal cord injury. J. sacral preganglionic neurons in the neonatal rat spinal cord.
Neurosci., 19: 7405–7414. Brain Res., 895: 223–232.
Kruse, M.N., Belton, A.L. and de Groat, W.C. (1993) Changes Miura, A., Kawatani, M. and de Groat, W.C. (2003) Excitatory
in bladder and external urethral sphincter function after synaptic currents in lumbosacral parasympathetic pregangl-
spinal cord injury in the rat. Am. J. Physiol., 264: ionic neurons evoked by stimulation of the dorsal commis-
R1157–R1163. sure. J. Neurophysiol., 89: 382–389.
Kruse, M.N., Bray, L.A. and de Groat, W.C. (1995) Influence Morgan, C., Nadelhaft, I. and de Groat, W.C. (1979) Location
of spinal cord injury on the morphology of bladder afferent of bladder preganglionic neurons within the sacral parasym-
and efferent neurons. J. Auton. Nerv. Syst., 54: 215–224. pathetic nucleus of the cat. Neurosci. Lett., 14: 189–194.
82

Morgan, C., Nadelhaft, I. and de Groat, W.C. (1981) The dis- Sasaki, M. (1994) Morphological analysis of external urethral
tribution of visceral primary afferents from the pelvic nerve and external anal sphincter motoneurones of cat. J. Comp.
to Lissauer’s tract and the spinal gray matter and its rela- Neurol., 349: 269–287.
tionship to the sacral parasympathetic nucleus. J. Comp. Sculptoreanu, A. and de Groat, W.C. (2003) Protein kinase C is
Neurol., 201: 415–440. involved in neurokinin receptor modulation of N- and L-type
Morgan, C.W., de Groat, W.C., Felkins, L.A. and Zhang, S.J. Ca2+ channels in DRG neurons of the adult rat. J. Neuro-
(1993) Intracellular injection of neurobiotin or horseradish physiol., 90: 21–31.
peroxidase reveals separate types of preganglionic neurons in Seki, S., Erickson, K.A., Seki, M., Nishizawa, O., Chancellor,
the sacral parasympathetic nucleus of the cat. J. Comp. Ne- M.B., de Groat, W.C. and Yoshimura, N. (2004a) Targeting
urol., 331: 161–182. spinal neurokinin I receptor-expressing neurons for the treat-
Morgan, C.W., Ohara, P.T. and Scott, D.E. (1999) Vasoactive ment of neurogenic detrusor overactivity in spinal cord
intestinal polypeptide in sacral primary sensory pathways in injury. J. Urol., 171: 142–143.
the cat. J. Comp. Neurol., 407: 381–394. Seki, S., Erickson, K.E., Seki, M., Nishizawa, O., Igawa, Y.,
Morrison, J., Steers, W.D., Brading, A., Blok, B., Fry, C., Ogawa, T., de Groat, W.C., Chancellor, M.B. and Yos-
de Groat, W.C., Kakizaki, H., Levin, R. and Thor, K. (2002) himura, N. (2005) Elimination of lamina I spinal neurons
Neurophysiology and neuropharmacology. In: Abrams P., expressing neurokinin 1 receptor using [Sar9, Met (O2)11]
Cardozo L., Khoury S. and Wein A. (Eds.), Incontinence. substance P-saporin reduces bladder overactivity and spinal
Health Publications Ltd., Jersey, pp. 83–164. c-fos expression induced by intravesical instillation of caps-
Morrison, J., Wen, J. and Kibble, A. (1999) Activation of pelvic aicin. Am. J. Physiol., 288: F466–F473.
afferent nerves from the rat bladder during filling. Scand. Seki, S., Igawa, Y., Kaidoh, K., Ishizuka, O., Nishizawa, O.
J. Urol. Nephrol., 33(Suppl. 201): 73–75. and Andersson, K.E. (2001) Role of dopamine D1 and D2
Nadelhaft, I. and Vera, P.L. (1996) Neurons in the rat brain receptors in the micturition reflex in conscious rats.
and spinal cord labeled after pseudorabies virus injected into Neurourol Urodyn., 20: 105–113.
the external urethral sphincter. J. Comp. Neurol., 375: Seki, S., Sasaki, K., Fraser, M.O., Igawa, Y., Nishizawa, O.,
502–517. Chancellor, M.B., de Groat, W.C. and Yoshimura, N. (2002)
Noto, H., Roppolo, J.R., de Groat, W.C., Nishizawa, O., Immunoneutralization of nerve growth factor in lumbosacral
Sugaya, K. and Tsuchida, S. (1991a) Opioid modulation of spinal cord reduces bladder hyperreflexia in spinal cord in-
the micturition reflex at the level of the pontine micturition jured rats. J. Urol., 168: 2269–2274.
center. Urol. Int., 47(Suppl. 1): 19–22. Seki, S., Sasaki, K., Igawa, Y., Nishizawa, O., Chancellor,
Noto, H., Roppolo, J.R., Steers, W.D. and de Groat, W.C. M.B., de Groat, W.C. and Yoshimura, N. (2004b) Suppres-
(1991b) Electrophysiological analysis of the ascending and sion of detrusor-sphincter dyssynergia by immunoneutrali-
descending components of the micturition reflex pathway in zation of nerve growth factor in lumbosacral spinal cord in
the rat. Brain Res., 549: 95–105. spinal cord injury rats. J. Urol., 171: 478–482.
Novakovic, S.D., Tzoumaka, E., McGivern, J.G., Haraguchi, Sengupta, J.N. and Gebhart, G.F. (1994) Mechanosensitive
M., Sangameswaran, L., Gogas, K.R., Eglen, R.M. and properties of pelvic nerve afferent fibers innervating the uri-
Hunter, J.C. (1998) Distribution of the tetrodotoxin-resistant nary bladder of the rat. J. Neurophysiol., 72: 2420–2430.
sodium channel PN3 in rat sensory neurons in normal and Shea, V.K., Cai, R., Crepps, B., Mason, J.L. and Perl, E.R.
neuropathic conditions. J. Neurosci., 18: 2174–2187. (2000) Sensory fibers of the pelvic nerve innervating the rat’s
O’Reilly, B.A., Kosaka, A.H., Knight, G.F., Chang, T.K., urinary bladder. J. Neurophysiol., 84: 1924–1933.
Ford, A.P., Rymer, J.M., Popert, R., Burnstock, G. and Shefchyk, S.J. (1989) The effects of lumbosacral deafferentation
McMahon, S.B. (2002) P2X receptors and their role in female on pontine micturition centre-evoked voiding in the decere-
idiopathic detrusor instability. J. Urol., 167: 157–164. brate cat. Neurosci. Lett., 99: 175–180.
Palea, S., Artibani, W., Ostardo, E., Trist, D.G. and Pietra, C. Shibata, T., Watanabe, M., Ichikawa, R., Inoue, Y. and
(1993) Evidence for purinergic neurotransmission in the hu- Koyanagi, T. (1999) Different expressions of a-amino-3-
man urinary bladder affected by interstitial cystitis. J. Urol., hydroxy-5-methyl-4-isoxazole propionic acid and N-methyl-
150: 2007–2012. D-aspartate receptor subunit mRNAs between visceomotor
Ralevic, V. and Burnstock, G. (1998) Receptors for purines and and somatomotor neurons of the rat lumbosacral spinal cord.
pyrimidines. Physiol. Rev., 50: 413. J. Comp. Neurol., 404: 172–182.
Rong, W., Spyer, K.M. and Burnstock, G. (2002) Activation Somogyi, G.T. and de Groat, W.C. (1999) Function, signal
and sensitisation of low and high threshold afferent fibres transduction mechanisms and plasticity of presynaptic
mediated by P2X receptors in the mouse urinary bladder. muscarinic receptors in the urinary bladder. Life Sci., 64:
J. Physiol., 541: 591–600. 411–418.
Sakakibara, R. and Fowler, C.J. (1999) Cerebral control of Somogyi, G.T., Zernova, G.V., Yoshiyama, M., Yamamoto, T.
bladder, bowel and sexual function and effects of brain dis- and de Groat, W.C. (1998) Frequency dependence of mu-
ease. In: Fowler C.J. (Ed.), Neurology of Bladder, Bowel and scarinic facilitation of transmitter release in urinary bladder
Sexual Dysfunction. Butterworth-Heinemann, Boston, strips from neurally intact or chronic spinal cord transected
pp. 229–244. rats. Br. J. Pharmacol., 125: 241–246.
83

Steers, W.D., Albo, M. and van Asselt, E. (1992a) Effects on Wein, A.J. (2002) Pathophysiology and categorization of
serotonergic agonists on micturition and sexual function in voiding dysfunction. In: Walsh P.C., Retik A.B., Vaughn
the rat. J. Drug. Dev. Res., 27: 361–375. E.D. and Wein A.J. (Eds.), Campbell’s Urology, Chapter 24.
Steers, W.D., Ciambotti, J., Etzel, B., Erdman, S. and de Groat, W.B. Saunders Co., Philadelphia, pp. 887–899.
W.C. (1991) Alterations in afferent pathways from the Yokoyama, O., Yoshiyama, M., Namiki, M. and de Groat,
urinary bladder of the rat in response to partial urethral W.C. (2000) Role of the forebrain in bladder overactivity
obstruction. J. Comp. Neurol., 310: 401–410. following cerebral infarction in the rat. Exp. Neurol., 163:
Steers, W.D., Creedon, D.J. and Tuttle, J.B. (1996) Immunity 469–476.
to nerve growth factor prevents afferent plasticity following Yoshimura, N. (1999) Bladder afferent pathway and spinal
urinary bladder hypertrophy. J. Urol., 155: 379–385. cord injury: possible mechanisms inducing hyperreflexia of
Steers, W.D., Meythaler, J.M., Haworth, C., Herrell, D. and the urinary bladder. Prog. Neurobiol., 57: 583–606.
Park, T.S. (1992b) Effects of acute bolus and chronic con- Yoshimura, N., Bennet, N.E., Phelan, M.W. and de Groat,
tinuous intrathecal baclofen on genitourinary dysfunction W.C. (1999) Effects of chronic nerve growth factor treatment
due to spinal cord pathology. J. Urol., 148: 1849–1855. on rat urinary bladder and bladder afferent neurons. Soc.
Stohrer, M., Madersbacher, H., Richter, R., Wehnert, J. and Neurosci. Abstr., 25: 1946.
Dreikorn, K. (1999) Efficacy and safety of propiverine in Yoshimura, N. and de Groat, W.C. (1997) Plasticity of Na+
SCI-patients suffering from detrusor hyperreflexia — a dou- channels in afferent neurones innervating rat urinary bladder
ble-blind, placebo-controlled clinical trial. Spinal Cord, 37: following spinal cord injury. J. Physiol., 503: 269–276.
196–200. Yoshimura, N. and de Groat, W.C. (1999) Increased excitabil-
Sugaya, K. and de Groat, W.C. (1994) Effects of MK-801 and ity of afferent neurons innervating rat urinary bladder after
CNQX, glutamate receptor antagonists, on bladder activity chronic bladder inflammation. J. Neurosci., 19: 4644–4653.
in neonatal rats. Brain Res., 640: 1–10. Yoshimura, N., Erdman, S.L., Snider, M.W. and de Groat,
Sugaya, K., Matsuyama, K., Takakusaki, K. and Mori, S. W.C. (1998a) Effects of spinal cord injury on neurofilament
(1987) Electrical and chemical stimulations of the pontine immunoreactivity and capsaicin sensitivity in rat dorsal root
micturition center. Neurosci. Lett., 80: 197–201. ganglion neurons innervating the urinary bladder. Neurosci.,
Sugaya, K., Roppolo, J.R., Yoshimura, N., Card, J.P. and de 83: 633–643.
Groat, W.C. (1997) The central neural pathways involved in Yoshimura, N., Mizuta, E., Kuno, S., Sasa, M. and Yoshida,
micturition in the neonatal rat as revealed by the injection of O. (1993) The dopamine D1 receptor agonist SKF 38393
pseudorabies virus into the urinary bladder. Neurosci. Lett., suppresses detrusor hyperreflexia in the monkey with
223: 197–200. parkinsonism induced by 1-methyl-4-phenyl-1,2,3,6-tetra-
Szallasi, A. and Fowler, C.J. (2002) After a decade of hydropyridine (MPTP). Neuropharmacology, 32: 315–321.
intravesical vanilloid therapy: still more questions than Yoshimura, N., Mizuta, E., Yoshida, O. and Kuno, S. (1998b)
answers. Lancet. Neurol., 1: 167–172. Therapeutic effects of dopamine D1/D2 receptor agonists on
Thor, K., Kawatani, M. and de Groat, W.C. (1986) Plasticity in detrusor hyperreflexia in 1-methyl-4-phenyl-1,2,3,6-tetra-
the reflex pathways to the lower urinary tract of the cat during hydropyridine-lesioned parkinsonian monkeys. J. Pharma-
postnatal development and following spinal cord injury. In: col. Exp. Ther., 286: 228–233.
Goldberger M., Gorio A. and Murray M. (Eds.) Development Yoshimura, N., Sasa, M., Yoshida, O. and Takaori, S. (1990a)
and Plasticity of the Mammalian Spinal Cord, Fidia Research Mediation of micturition reflex by central norepinephrine
Series, Vol. III. Fidia Press, Padova, Italy, pp. 65–81. from the locus coeruleus in the cat. J. Urol., 143: 840–843.
Thor, K.B., Hisamitsu, T. and de Groat, W.C. (1990) Un- Yoshimura, N., Sasa, M., Yoshida, O. and Takaori, S. (1990b)
masking of a neonatal somatovesical reflex in adult cats by Alpha 1-adrenergic receptor-mediated excitation from the
the serotonin autoreceptor agonist 5-methoxy-N,N-dimethyl- locus coeruleus of the sacral parasympathetic preganglionic
tryptamine. Dev. Brain Res., 54: 35–42. neuron. Life Sci., 47: 789–797.
Thor, K.B., Morgan, C., Nadelhaft, I., Houston, M. and de Yoshimura, N., Seki, S. and de Groat, W.C. (2001a) Nitric
Groat, W.C. (1989) Organization of afferent and efferent oxide modulates Ca2+ channels in dorsal root ganglion
pathways in the pudendal nerve of the female cat. J. Comp. neurons innervating rat urinary bladder. J. Neurophysiol.,
Neurol., 288: 263–279. 86: 304–311.
Torrens, M. and Morrison, J. (1987) The Physiology of the Yoshimura, N., Seki, S., Erickson, K.A., Erickson, V.L.,
Lower Urinary Tract. Springer, New York, pp. 1–355. Chancellor, M.B. and de Groat, W.C. (2003) Histological
Vizzard, M.A. (2000) Changes in urinary bladder neurotrophic and electrical properties of rat dorsal root ganglion neurons
factor mRNA and NGF protein following urinary bladder innervating the lower urinary tract. J. Neurosci., 23:
dysfunction. Exp. Neurol., 161: 273–284. 4355–4361.
Vizzard, M.A., Erickson, V.L., Card, J.P., Roppolo, J.R. and Yoshimura, N., Seki, S., Novakovic, S.D., Tzoumaka, E.,
de Groat, W.C. (1995) Transneuronal labeling of neurons in Erickson, V.L., Erickson, K.A., Chancellor, M.B. and de
the adult rat brainstem and spinal cord after injection of Groat, W.C. (2001b) The role of the tetrodotoxin-resistant
pseudorabies virus into the urethra. J. Comp. Neurol., 355: sodium channel Nav1.8 (PN3/SNS) in a rat model of visceral
629–640. pain. J. Neurosci., 21: 8690–8696.
84

Yoshimura, N., White, G., Weight, F.F. and de Groat, W.C. Yoshiyama, M., Roppolo, J.R. and de Groat, W.C. (1997)
(1996) Different types of Na+ and A-type K+ currents in Effects of LY215490, a competitive alpha-amino-3-hydroxy-
dorsal root ganglion neurones innervating the rat urinary 5-methylisoxazole-4-propionic acid (AMPA) receptor antag-
bladder. J. Physiol. (Lond.), 494: 1–16. onist, on the micturition reflex in the rat. J. Pharmacol. Exp.
Yoshiyama, M. and de Groat, W.C. (1997) Effects of in- Ther., 280: 894–904.
trathecal administration of pituitary adenylate cyclase acti- Zhong, Y., Banning, A.S., Cockayne, D.A., Ford, A.P., Burn-
vating polypeptide (PACAP) on the lower urinary tract in the stock, G. and McMahon, S.B. (2003) Bladder and cutaneous
rat. Society for Neuroscience Abstracts, 23: 1523. sensory neurons of the rat express different functional P2X
Yoshiyama, M., Roppolo, J.R. and de Groat, W.C. (1993) Ef- receptors. Neuroscience, 120: 667–675.
fects of MK-801 on the micturition reflex in the rat — pos- Zoubek, J.A., Somogyi, G.T. and de Groat, W.C. (1993) A
sible sites of action. J Pharmacol. Exp. Ther., 265: 844–850. comparison of inhibitory effects of neuropeptide Y on rat
Yoshiyama, M., Roppolo, J.R. and de Groat, W.C. (1994) Al- urinary bladder, urethra and vas deferens. Am. J. Physiol.,
terations by urethane of glutamatergic control of micturition. 265: R537–R543.
Eur. J. Pharmacol., 264: 417–425. Zvarova, K., Dunleavy, J.D. and Vizzard, M. (2005) Changes
Yoshiyama, M., Roppolo, J.R. and de Groat, W.C. (1995) In- in pituitary adenylate cyclase activating polypeptide (PA-
teractions between NMDA and AMPA/kainate receptors in the CAP) expression in urinary bladder pathways after spinal
control of micturition in the rat. Eur. J. Pharmacol., 287: 73–78. cord injury (SCI). Exp. Neurol., 192: 46–59.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 6

Spinal mechanisms contributing to urethral striated


sphincter control during continence and micturition:
‘‘How good things might go bad’’

Susan J. Shefchyk

Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 3J7, Canada

Abstract: The external urethral sphincter motoneurons in the sacral ventral horn control the striated
external urethral sphincter muscles that circle the urethra. Activity in these motoneurons and muscle
normally contribute to continence but during micturition, when urine must pass through the urethra, the
motoneurons and striated muscle must be silenced. Following injury to descending pathways in the spinal
cord, the ability to inhibit sphincter activity is disrupted or lost, resulting in bladder–sphincter dyssynergia
and functional obstruction of the urethra during voiding. This chapter will first review the various reflex
pathways and neuronal properties that contribute to continence, and which must be modulated during
micturition in the spinal intact animal. A discussion about how the dyssynergia seen with spinal cord injury
may be produced will then be presented.

Discussions about the neural control of the lower the urethra is open to allow the passage of
urinary tract usually concentrate on the autonomic urine from the contracting bladder. The sacral
control of the bladder and urethra smooth muscle ventral horn motoneurons innervating the striated
with less focus on the role of the striated urethral sphincter muscle fibres are thus subject to differ-
sphincter muscle. However, the coordination ential control during bladder filling and micturit-
between autonomic and somatic motor systems ion. The manifestation of abnormal micturition
is fundamental to both continence and micturition. patterns involving both autonomic and sphincter
While the striated sphincter is not under auto- somatic systems creates major challenges in
nomic control per se, the convergence and coor- clinical management, particularly in individuals
dination of various autonomic and somatic with spinal cord and supra-spinal disease or
sphincter reflex pathways unite the two systems damage.
closely. For the purpose of this review, the discus- We will see that the neural control of sphincter
sion will be limited to the spinal neural control motoneuron output is not unlike that of limb
of the striated external urethral sphincter muscle. motoneuron, including the interplay of descending
Contraction of the sphincter muscle contributes and segmental reflex pathways as well as major
to continence by closing the urethra. In contrast, contributions from the anatomical and electrical
sphincter muscle activity is suppressed during properties of the motoneurons themselves. The
micturition when the sacral parasympathetic abnormal striated sphincter activity, commonly
bladder preganglionic neurons are activated and referred to as sphincter dyssynergia (Andersen and
Bradley, 1976; Blaivas et al., 1981; Koyanagi et al.,
Corresponding author. Tel.: +204-789-3736; 1982; Kruse et al., 1993; Pikov and Wrathall,
Fax: +204-789-3930; E-mail: sjs@scrc.umanitoba.ca 2001), occurs as a consequence of various spinal

DOI: 10.1016/S0079-6123(05)52006-5 85
86

cord and supra-spinal lesions. This review will interneurons such as Renshaw cells (Hultborn
address the mechanism(s) contributing to the et al., 1988; Alvarez et al., 1999). This lack of an
recruitment of the sphincter motoneurons and anatomical substrate for recurrent inhibition is
muscles during continence, the de-recruitment of consistent with the absence of any evidence for
these motoneurons and muscle during micturition, functional recurrent inhibition of the sphincter
and the possible underlying factors responsible for motoneurons (Mackel, 1979). Although a variety
the abnormal sphincter motor patterns observed of anatomical and electrophysiological evidence
following spinal cord injury. By first reviewing shows descending inputs from various supra-
what is known about the control of the urethral spinal regions (Mackel, 1979; Nakagawa, 1980;
sphincter reflex, a groundwork for a discussion Holstege et al., 1987; Hermann et al., 1998), the
about how good things can go bad can begin. functional contributions of these descending path-
ways to the dynamic control of the sphincter
motoneurons is not clear. Figure 1 summarizes the
Continence major inputs to the sphincter motoneurons.
The location and morphology of the ventral
In humans, as well as cats and rats, continence is horn motoneurons, sometimes referred to as
facilitated by closure of the urethra achieved via external urethral sphincter pudendal motoneu-
contraction of the smooth muscle of the bladder rons, innervating the external striated sphincter
neck and urethra and activity of the striated muscle in humans, cats, rats and monkeys have
sphincter muscles surrounding the urethra been described in some detail (Onuf, 1899;
(reviewed in de Groat et al., 2001). In addition, Schroder, 1981; Roppolo et al., 1985; McKenna
the muscles of the pelvic floor may also contribute and Nadelhaft, 1986; Beattie et al., 1990; Sasaki,
to continence. The source of excitation to the 1994; Pullen et al., 1997). As mentioned earlier,
sphincter motoneurons has been the subject of these motoneurons have axon collaterals that
investigation for over half a century. Much of the project to the region of the cell somas (Sasaki,
focus has been on the segmental sensory inputs to 1994). Studies have also shown that the dendrites
the sphincter motoneurons, a sensory input that of these motoneurons form bundles that travel
is largely excitatory and associated with perineal rostral-caudally between clusters of sphincter
and pudendal cutaneous, urethral (Bradley and motoneuron somas (Beattie et al., 1990). There is
Teague, 1972, 1977; Mackel, 1979; Fedirchuk no evidence to date of gap junctions or coupling
et al., 1992; Shefchyk and Buss, 1998; Buss and between dendrites or somas.
Shefchyk, 1999) and pelvic visceral (Garry et al., The passive electrical membrane properties of
1959; McMahon et al., 1982) afferents. In contrast cat sphincter motoneurons have been described in
to hindlimb motoneurons, there appears to be no detail by several laboratories (Hochman et al.,
significant monosynaptic afferent input to sphinc- 1991; Sasaki, 1991; Shimoda et al., 1992). Sphincter
ter motoneurons (Jankowska et al., 1978; Mackel, motoneurons have a very high membrane input
1979; Fedirchuk et al., 1992; Buss and Shefchyk, resistance and low rheobase making them easily
1999), and direct feedback from the muscle itself recruited. They also display a depolarizing ‘‘sag’’
is almost nonexistent (Chennells et al., 1960; in the membrane response to hyperpolarization,
Lassmann, 1984). In addition, there is no electro- which can contribute to rebound excitation fol-
physiological evidence in sphincter motoneurons lowing hyperpolarization. Their action potentials
for any crossed or reciprocal inhibition as de- are followed by relatively short duration after
scribed for other sacral motoneuron populations hyperpolarizations, suggesting that they are well
(Jankowska et al., 1978). Sphincter motoneurons suited to fire tonically. These properties are con-
have axon collaterals (Sasaki, 1994), but these sistent with the need for a continuous motoneuron
appear to terminate directly in the region of the output to drive the sphincter muscles tonically
sphincter motor nucleus and do not project during the long periods of bladder filling (Garry
medially to the region of recurrent inhibitory et al., 1959; McMahon et al., 1982).
87

Fig. 1. A summary of the descending and segmental sensory systems described anatomically or physiologically as having inputs to the
external urethral sphincter motoneurons. The size of the arrow reflects the hypothesized functional strength of the input to the
motoneurons.

For over 20 years now non-linear membrane dorsal horn neurons (Russo et al., 1997; Morisset
responses to depolarizing current injection, and Nagy, 1999; Derjean et al., 2003) and sacral
referred to as plateau potentials or bistable mem- preganglionic neurons (Derjean et al., 2005). Other
brane properties, have been described and studied putative transmitter systems, including substance
in spinal hindlimb motoneurons and interneurons P and acetylcholine, have also been implicated in
in a variety of vertebrates (Hultborn and Kiehn, the facilitation of plateau properties (Delgado-
1992; Morisset and Nagy, 1999). Similar motor Lezama et al., 1997; Russo et al., 1997). On the
responses have been documented in humans other hand, metabotropic GABAergic receptors
(Collins et al., 2001). When expressed, these prop- are thought to suppress plateaux and bistable
erties function to amplify or prolong a neuron’s properties (Derjean et al., 2003).
output in response to a brief excitatory input. More recently, for the first time in cat sphincter
Furthermore, the expression of these properties motoneurons, the presence of similar active mem-
may be controlled by a balance of metabotropic brane properties that are sensitive to neuromodu-
excitatory and inhibitory neuromodulatory sub- latory systems have been described (Paroschy and
stances. For instance, serotonin and noradrenalin Shefchyk, 2000). Paroschy and Shefchyk (2000)
have been shown to facilitate plateau property reported that brief trains of perineal or pudendal
expression in ventral horn motoneurons afferent stimulation produced sustained firing in
(Hounsgaard et al., 1988; Russo et al., 1998), cat sphincter motoneurons. This type of sustained
and activation of metabotropic glutamate type 1 response had been noted years ago by McMahon
receptors can facilitate their expression in deep et al. (1982). Using intracellular recordings,
88

Paroschy and Shefchyk (2000) showed that a brief which for this purpose more closely mimics human
intracellular depolarizing current injection could micturition.
produce a train of action potentials characterized It has been recognized that when the bladder
by an accelerating firing rate, or a membrane de- contracts during micturition and urine flow begins,
polarization, that persisted well beyond the period the urethra is unobstructed to facilitate the flow of
of current injection. As with cat hindlimb moto- fluid out of the body. For many years, a variety of
neurons, the expression of these non-linear re- reflex feedback loops recruited during the bladder
sponses to depolarizing current injection was contraction and initial flow of urine into the ure-
facilitated by the intravenous administration of thra (Barrington, 1914; Garry et al., 1959) were
serotonin precursors or noradrenalin. Paroschy conceived as responsible for the decrease in sphinc-
and Shefchyk (2000) concluded that at least some ter activity. However, it was not until examination
proportion of sphincter motoneurons could alter of the pattern of sphincter activity changes during
their excitability and firing characteristics (i.e., filling and micturition in the de-afferented animal
express non-linear responses manifested as persist- was done (Shefchyk, 1989) did it become evident
ent firing or sustained membrane depolarization), that a central micturition circuitry contributed to
and that such properties could contribute to the the sphincter silencing in the absence of sensory
tonic activity of the sphincter motoneurons during feedback. That is not to say that segmental reflexes
continence. This non-linear property may reduce originating from the bladder and urethra do not
the need for a continuous synaptic excitation of contribute to the sphincter silencing, but that a
the sphincter motoneuron to maintain a given central circuitry is also available.
muscle force because it could enhance and extend
any periodic excitatory actions of segmental or
descending synaptic inputs during bladder filling. Direct inhibition of sphincter motoneurons
during micturition

Micturition The initial question raised was whether the


absence of tonic or evoked sphincter activity in
During micturition, the external urethral sphincter the cat model during the void was due to a periodic
muscle relaxes and the urethra is opened allowing absence of excitation or an active inhibition of the
urine to flow freely. When the sacral parasympa- motoneurons by the micturition circuitry. Shimoda
thetic bladder preganglionic neurons are recruited, et al. (1992) and Fedirchuk et al. (1993), using
a parallel system is engaged to ensure that sphincter intracellular recordings from sphincter motoneurons
motoneurons do not fire and that the sphincter during distension- and brainstem-evoked micturition
muscle activity ceases for a period of time during showed that the membrane of the urethra sphincter
the void. In humans (Andersen and Bradley, 1976; motoneurons hyperpolarized during micturition
Blaivas et al., 1981; Dyro and Yalla, 1986) and when the firing of sphincter motor axons, recorded
cats (Barrington, 1914; Rampal and Mignard, in the pudendal nerve, and the sphincter muscle
1979b; Sackman and Sims, 1990), there is normally electromyogram were silenced. Furthermore,
a complete silencing of sphincter muscle activity during this hyperpolarization an increase in
during micturition while in the rat, the sphincter membrane conductance could be detected and
displays a pattern of rhythmic bursting during the intracellular injection of chloride ions could
bladder contraction (Kakizaki et al., 1997; Streng reverse the hyperpolarization (Fedirchuk et al.,
et al., 2004). The mechanism responsible for this 1993). Together, these data lead to the suggestion
rhythmic activity in the rat has not been identified, that an active inhibitory chloride conductance at
but while it does not appear to be a detriment to the motoneuron membrane was contributing to
bladder emptying in the rat, such patterns in the inhibition of the motoneurons (Fedirchuk
humans or cats are considered pathological. This et al., 1993; Shefchyk, 1998). Shefchyk et al. (1998)
review will focus on results from the cat model, went on to show that in the cat, the sphincter
89

motoneuron inhibition was sensitive to the neurons in the dorsal commissure of the cat sacral
glycinergic antagonist, strychnine, and that the spinal cord. More recently, Sie et al. (2001)
motoneuron somas and proximal dendrites were extended this observation to show that some of
immunopositive for gephyrin, a protein that has these spinal interneurons were glycinergic. Based
been shown to be associated with glycinergic on the facts that these interneurons were positive
receptors in the spinal cord (Fyffe et al., 1995). for GABA and glycine and that micro-stimulation
Immunohistochemical evidence for GABAergic in the area of these neurons decreased intra-ure-
terminals on cat sphincter motoneurons has also thral pressure (Blok et al., 1998), it was hypoth-
been obtained (Ramirez-Leon et al., 1994), but a esized that these neurons mediated the inhibition
functional examination of the GABAergic actions of the sphincter motoneurons during micturition.
directly on the motoneurons or sphincter activity Buss and Shefchyk (2003), using extracellular
has not been reported. Unfortunately, in the rat recordings from single units in the dorsal commis-
the sphincter system has not been subject to a sure and deep dorsal horn, identified a group of
similar examination, and it is not known whether neurons that were excited during reflex distension-
the rhythmic sphincter muscle activity during evoked or pontine micturition center-evoked void
micturition is due to: (1) a periodic inhibition of responses, but not by bladder distension alone.
sphincter motoneurons during voiding, (2) the These neurons may be those described by Blok and
expression of intrinsic oscillatory properties in the co-workers. In addition, the anatomical identifi-
motoneurons during voiding, or (3) a combination cation within this region of a variety of neurons
of both tonic inhibition of some motoneurons as in linked to the sphincter motoneurons has been
the cat and human, with only a subpopulation described using viral tracing methods (Nadelhaft
phasically active during voiding. and Vera, 1996; for review of sacral interneurons,
The membrane hyperpolarization observed in see Shefchyk, 2001).
the cat sphincter motoneurons during micturition
functions to decrease the excitability of the cells by
moving the membrane potential away from the Evidence for premotoneuronal inhibition
firing threshold. As the expression of the non- in excitatory reflex pathways to
linear membrane properties described by Paroschy sphincter motoneurons
and Shefchyk (2000) is voltage-dependent (thresh-
old around 43 mV), such membrane hyperpolar- Fedirchuk et al. (1994) observed that polysynaptic
ization could function to turn off any expressed excitatory postsynaptic potentials in hindlimb
non-linear responses (see Fig. 6 in Paroschy and motoneurons, within the first sacral segment, were
Shefchyk, 2000). The fact that perineal, pudendal diminished in amplitude, or completely sup-
(Fedirchuk et al., 1994) and urethral afferent- pressed, during voiding in the absence of a postsy-
evoked (Buss and Shefchyk, 1999) excitatory naptic motoneuron membrane hyperpolarization.
postsynaptic potentials in sphincter motoneurons This led to the hypothesis that the excitatory path-
are decreased in amplitude during voiding, when ways from the perineal and pudendal afferents
the membrane conductance of the sphincter moto- may be gated out during micturition at sites pre-
neurons increases is consistent with a conductance motoneuronal, that is, at the segmental excitatory
shunting effect to diminish the excitatory poten- interneurons mediating the excitation or at the
tials. Such postsynaptic inhibition in the spinal primary afferent terminals themselves.
cord is usually mediated by local interneurons. The possibility that perineal and pudendal pri-
Evidence for the location and identity of spinal mary afferents were subjected to presynaptic inhi-
inhibitory interneurons that may be part of the bition, or primary afferent depolarization, was
circuitry activated during micturition was provid- examined by monitoring the excitability of single
ed by Blok et al. (1997), who reported a descend- identified afferents in the dorsal horn of the sacral
ing projection from the brainstem pontine spinal segments during micturition (Angel et al.,
micturition center to a population of GABAergic 1994; Buss and Shefchyk, 1999). Angel et al. (1994)
90

observed that while bladder distension in the ab- differed in their activity during micturition. One
sence of micturition did not produce excitability group, already mentioned earlier, were recruited
changes in pudendal or perineal afferents, during during micturition, and may be the inhibitory
distension-evoked micturition reflexes, over one- neurons we are discussing. A second population,
third of the afferents tested displayed an increase which were excited by perineal and pudendal af-
in excitability during the void. It was concluded ferents were inhibited during micturition and
that primary afferent depolarization could be de- could be the excitatory interneurons receiving in-
creasing transmitter release from these afferents hibition during micturition (Buss and Shefchyk,
and diminishing the efficacy of the afferent exci- 2003).
tatory actions during voiding. Buss and Shefchyk As summarized in Fig. 2, the central micturition
(1999) examined the perineal and pudendal affer- circuitry has access to a circuitry of spinal neurons
ents separately from the urethral pudendal affer- that coordinate various inhibitory pathways dur-
ents during micturition evoked by both distension ing micturition. The need to ensure the suppres-
of the bladder and by electrical stimulation of the sion of sphincter motoneuron output must be
pontine micturition center. The results obtained a high priority for the system as reflected in the
during both types of evoked micturition were sim- redundancy in the mechanisms responsible for this
ilar: 50% of the dorsal penile/clitoral afferents un- inhibition. The relative contributions of each of
derwent primary afferent depolarization during these inhibitory systems is not known at this time,
the void, while almost 60% of the urethral affer- but it can by hypothesized that disruption of any
ents examined showed excitability increases during of these components may contribute to undesired
voiding. Biphasic changes in excitability were not- motoneuron hyperactivity and dyssynergia during
ed in 4/11 afferents, that is, they underwent pri- bladder emptying.
mary afferent depolarization early during the void
then showed a decrease in excitability as the
sphincter activity returned following the void. It So, how might good things go bad?
was hypothesized that the spinal circuitry and in-
terneurons mediating presynaptic inhibition of the Depending upon the extent of damage to the white
perineal, pudendal and urethral sacral afferents matter of the spinal cord, a variety of changes may
was organized in a way to recognize the different occur in lower urinary tract function. With supra-
functions of the perineal/pudendal versus urethral sacral injuries, bladder and sphincter dyssynergia
afferents during the voiding cycle. Specifically, is commonly encountered and has been character-
Buss and Shefchyk (2003) proposed the existence ized in humans (Andersen and Bradley, 1976), as
of at least two subgroups of interneurons recruited well as cat (Rampal and Mignard, 1976b) and ro-
during micturition to accomplish this selective dent (Pikov and Wrathall, 2001) models. Although
sensory modulation. The GABA-containing spinal the details may vary somewhat, the common fea-
neurons that Blok and co-workers (Blok et al., ture of the dyssynergia is the presence of activity in
1997,1998; Sie et al., 2001) described would be ex- the sphincter muscle during bladder contractions,
cellent candidates for mediating the presynaptic an activity that effectively limits the amount of
inhibition of the primary afferents. urine that can be expelled. Various attempts to
In addition to presynaptic inhibition of primary decrease the neural drive to the sphincter muscle,
afferents, it is also possible that the excitatory in- including denervating the muscle physically, or
terneurons interposed between the primary affer- chemically with botulinum toxin (Smith et al.,
ents and the sphincter motoneurons could be the 2002), are directed to the problem of too much
targets of direct inhibitory inputs during micturit- sphincter tone and reflex activity. But, such ap-
ion. Buss and Shefchyk (2003) found two popula- proaches are obviously limited and do not address
tions of neurons in the sacral dorsal commissure of the basic issue, that is, the identity of the mech-
the cat that responded to electrical stimulation anisms contributing to the inappropriate sphincter
of perineal, pudendal and urethral afferents, but motoneuron activity.
91

Fig. 2. Summary of the inhibitory mechanisms thought to contribute to the suppression of sphincter motoneuron activity during
micturition in the spinal intact animal.

In humans and some of the animal models stud- disrupted. If we consider the factors discussed so
ied, the loss of descending pathways controlling far in this chapter, we can identify some potential
motor neurons, below the spinal cord lesion, results mechanisms for the abnormal sphincter activity
in loss of voluntary striated muscle control. How- during micturition following spinal cord injury.
ever, this is often accompanied by hyper-reflexia and The loss of descending pathways that facilitate
spasms in the ‘‘paralyzed’’ muscles. For limb motor spinal inhibitory circuits may be part of the prob-
systems, the spasms and hyperactivity appear to in- lem. The loss of descending pathways that have
volve changes in the control of transmission through tonic inhibitory control over all excitatory seg-
excitatory segmental reflex pathways and adapta- mental reflex pathways, including those to the
tions in the intrinsic properties of the motoneurons sphincter motoneurons, might contribute to in-
themselves (for discussion, see Gorassini et al., creased reflex activation of the sphincter during
2004). Even with incomplete lesions of the spinal both continence and micturition. This increased
white matter, in particular the dorsolateral fun- activity would demand even more powerful inhi-
iculus, the release of a variety of segmental excita- bition to prevent the recruitment of sphincter
tory reflexes in motor systems below the level of the motoneurons during micturition. Furthermore, a
lesion (Cavallari and Petersson, 1989) may contrib- decrease in excitability of a subset of interneurons
ute to increased reflex motor output. The striated specifically mediating the micturition-related
sphincter muscle appears to be no different from postsynaptic inhibition of the sphincter motoneu-
limb muscles in terms of the increased tone and rons or of the excitatory interneurons interposed
reflex activity following cord injury. Not only is between segmental afferents and the motoneurons,
there an apparent increase in excitability and output could lead to the sphincter dyssynergia during mi-
of the sphincter motoneurons in general, but in cturition. We are addressing excitability changes in
addition the mechanisms normally used by the the neurons responsible for postsynaptic inhibi-
central micturition circuitry to inhibit sphincter tion, and these same concerns may be raised for
activity appear to be either absent or greatly the spinal interneurons mediating presynaptic
92

inhibition of the primary afferents. It has been contributing to excitability changes (Derjean
documented that spinal cord presynaptic inhibito- et al., 2003, 2005). These exciting possibilities
ry pathways are disrupted with loss of descending remain to be tested. Whether such systems can
systems (Carpenter et al., 1963; Hongo and enhance the expression of non-linear membrane
Jankowska, 1967), and this could contribute to properties and/or membrane excitability in the
increased reflex transmission through many affer- neurons remains to be tested in both spinal cord
ent systems. This possibility has been addressed in intact and lesioned animal models. Furthermore,
studies using baclofen, an agonist of spinal GABA we must look beyond the motoneurons themselves.
ergic systems, to treat spinal cord-injured subjects It is known that unidentified interneurons can ex-
with sphincter hyperactivity and dyssynergia press plasticity in their firing characteristics, in-
(Hachen and Krucker, 1977). Although the target cluding membrane oscillations and plateau
may be afferent excitatory pathways to the sphinc- potentials/enhanced firing (Derjean et al., 2003,
ter system, the clinical results revealed that this 2005). It would be exciting to determine if only
approach was not selective and can also decrease excitatory interneurons have this capability or if
bladder afferent transmission to the point of com- inhibitory spinal neurons also express such plas-
promising the bladder contractions (Steers, 1989). ticity and by what mechanisms. If inhibitory neu-
Turning now to the possible effects of loss of rons can, then we might develop strategies to
neuromodulators released from descending sys- ensure their expression should it be diminished or
tems, we will consider the modulation of intrinsic lost following spinal cord injury. Regardless of
electrical properties and excitability changes in whether we are considering motoneurons or inter-
spinal interneuron and sphincter motoneuron sys- neurons, the reality of the presence of plasticity of
tems involved in continence and micturition. It has firing pattern and excitability should now be care-
been established in rat tail motoneurons that fully addressed in both intact and chronic lesion
changes in various membrane currents occur fol- models for the spinal neurons involved in lower
lowing lower lumbar spinal cord injury and that urinary tract function. We need to determine: (1) if
these changes may contribute to an increased re- such properties are expressed in all spinal cell
cruitment of motoneurons and spastic motor ac- types; (2) what ion channels and second messenger
tivity in the tail musculature (Li et al., 2004). systems mediate the plasticity; and (3) which
Gorassini and co-workers have described results neuromodulators facilitate and inhibit the chan-
suggestive of similar mechanisms in limb moto- nels and systems involved in the expression of the
neurons of spinal cord injured people (Gorassini property. This knowledge may provide some ex-
et al., 2004). With the loss of descending serotonergic citing new insights into selectivity and organiza-
and adrenergic systems to the sphincter motoneu- tion of the systems and cellular mechanism.
rons known to promote non-linear properties and In summary, the control of the external urethral
increased motoneuron output (Paroschy and sphincter is not simple. The integration of various
Shefchyk, 2000), one might predict decreased, segmental inputs, interneuron activity and moto-
not increased sphincter reflex activity. However, neuron properties must be considered when ad-
it is known that substances intrinsic to the spinal dressing the changes in sphincter motor function
cord may also facilitate the expression of similar following spinal cord injury. Although the sphinc-
membrane currents and excitability changes. For ter system differs in many ways from limb muscle
instance, acetylcholine can produce plateau poten- control, there are similarities that are significant
tials (Svirskis and Hounsgaard, 1998) and a strong and we may do well to consider the well-developed
intrinsic cholinergic source to the sphincter moto- literature on limb reflex control and spinal cord
neurons may be directly from their own axon injury. The circuitry controlling the sphincter is
collaterals (Sasaki, 1994). Furthermore, changes in largely intrinsic to the spinal cord, and thus may
glutamatergic inputs and receptors following cord still be available for manipulation and participa-
injury (see Llewellyn-Smith et al., 1997), may tion in the normalization of lower urinary tract
result in glutamatergic metabotropic receptors function following spinal cord injury.
93

References Delgado-Lezama, R., Perrier, J.-F., Nederfaard, S., Svirskis, C.


and Hounsgaard, J. (1997) Metabotropic synaptic regulation
Alvarez, F.J., Dewey, D.E., McMillin, P. and Fyffe, R.E. (1999) of intrinsic response properties of turtle spinal motoneu-
Distribution of cholinergic contacts on Renshaw cells in the rones. J. Physiol., 504: 97–102.
rat spinal cord: a light microscopic study. J. Physiol., Derjean, D., Bertrand, S., Le Masson, G., Landry, M., Moris-
515(Part 3): 787–797. set, V. and Nagy, F. (2003) Dynamic balance of metabo-
Andersen, J.T. and Bradley, W.E. (1976) The syndrome of tropic inputs causes dorsal horn neurons to switch functional
detrusor-sphincter dyssynergia. J. Urol., 116: 493–495. states. Nat. Neurosci., 6: 274–281.
Angel, M.J., Fyda, D., McCrea, D.A. and Shefchyk, S.J. (1994) Derjean, D., Bertrand, S., Nagy, F. and Shefchyk, S.J. (2005)
Primary afferent depolarization of cat pudendal afferents Plateau potentials and membrane oscillations in parasympa-
during micturition and segmental afferent stimulation. thetic preganglionic neurons and intermediolateral neurons in
J. Physiol., 479: 451–461. the rat lumbo-sacral spinal cord. J. Physiol., 563.2: 583–596.
Barrington, F.J.F. (1914) The nervous mechanism of micturit- Dyro, F.M. and Yalla, S.V. (1986) Refractoriness of urethral
ion. Q. J. Exp. Physiol., 8: 33–71. striated sphincter during voiding: studies with afferent pudendal
Beattie, B.S., Li, Q., Leedy, G. and Bresnahan, J.C. (1990) reflex arc stimulation in male subjects. J. Urol., 135: 732–736.
Motoneurons innervating the external anal and urethral Fedirchuk, B., Downie, J.W. and Shefchyk, S.J. (1994) Reduc-
sphincters of the female cat have different patterns of tion of perineal evoked excitatory postsynaptic potentials in
dendritic arborization. Neurosci. Lett., 111: 69–74. cat lumbar and sacral motoneurons during micturition.
Blaivas, J.G., Sinha, H.P., Zayed, A.A.H. and Labib, K.B. J. Neurosci., 14: 6153–6159.
(1981) Detrusor-external sphincter dyssynergia: A detailed Fedirchuk, B., Hochman, S. and Shefchyk, S.J. (1992) An in-
electromyographic study. J. Urol., 125: 545–548. tracellular study of perineal and hindlimb afferent inputs
Blok, B.F.M., de Weerd, H. and Holstege, G. (1997) The pon- onto sphincter motoneurons in the decerebrate cat. Exp.
tine micturition center projects to sacral cord GABA Brain Res., 89: 511–516.
immunoreactive neurons in the cat. Neurosci. Lett., 233: Fedirchuk, B. and Shefchyk, S.J. (1993) Membrane potential
109–112. changes in sphincter motoneurons during micturition in the
Blok, B.F.M., van Maarseveen, J.T.P.W. and Holstege, G. decerebrate cat. J. Neurosci., 13: 3090–3094.
(1998) Electrical stimulation of the sacral dorsal gray Fedirchuk, B., Song, L., Downie, J.W. and Shefchyk, S.J.
commisure evoked relaxation of the external urethral (1992) Spinal distribution of extracellular field potentials
sphincter in the cat. Neurosci. Lett., 249: 68–70. generated by electrical stimulation of pudendal and perineal
Bradley, W.E. and Teague, C.T. (1972) Electrophysiology of afferents in the cat. Exp. Brain Res., 89: 517–520.
pelvic and pudendal nerves in the cat. Exp. Neurol., 35: Fyffe, R.E.W., Alvarez, F.J., Harrington, D. and Dewey, D.E.
378–393. (1995) Expression of glycine receptors by identified alpha and
Bradley, W.E. and Teague, C.T. (1977) Synaptic events in gamma motoneurones. In: Taylor A., Gladden M.H. and
pudendal motoneurons of the cat. Exp. Neurol., 56: 237–240. Durbaba R. (Eds.), Alpha and Gamma Motor Systems. Ple-
Buss, R.R. and Shefchyk, S.J. (1999) Excitability changes in num Press, New York, pp. 421–428.
sacral afferents innervating the urethra, perineum and hind- Garry, R.C., Roberts, T.D.M. and Todd, J.K. (1959) Reflexes
limb skin of the cat during micturition. J. Physiol., 514: involving the external urethral sphincter in the cat.
593–607. J. Physiol., 149: 653–665.
Buss, R.R. and Shefchyk, S.J. (2003) Sacral dorsal horn neu- Gorassini, M.A., Knash, M.E., Harvey, P.J., Bennett, D.J. and
rone activity during micturition in the cat. J. Physiol., 551: Yang, J.F. (2004) Role of motoneurons in the generation of
387–396. muscle spasms after spinal cord injury. Brain., 127: 2247–2258.
Carpenter, D., Lundberg, A. and Norsell, U. (1963) Primary Hachen, H.J. and Krucker, V. (1977) Clinical and laboratory
afferent depolarization evoked from the sensorimotor cortex. assessment of the efficacy of Baclofen (Lioresal(R)) on
Acta Physiol. Scand., 59: 126–142. urethral sphincter spacticity in patients with traumatic
Cavallari, P. and Petersson, L.G. (1989) Tonic suppression of paraplegia. Eur. Urol., 3: 237–240.
reflex transmission in low spinal cats. Exp. Brain Res., 77: Hermann, G.E., Bresnahan, J.C., Holmes, G.M., Rogers, R.C.
201–212. and Beattie, M.S. (1998) Descending projections from the
Chennells, M., Floyd, W.F. and Gould, R.P. (1960) Muscle nucleus raphe obscurus to pudendal motoneurons in the male
spindles in the external anal spincter of the cat. J. Physiol., rat. J. Comp. Neurol., 397: 458–474.
151: 23P–25P. Hochman, S., Fedirchuk, B. and Shefchyk, S.J. (1991)
Collins, D.F., Burke, D. and Gandevia, S.C. (2001) Large Membrane electrical properties of external urethral and
involuntary forces consistent with plateau-like behavior of external anal sphincter somatic motoneurons in the
human motoneurons. J. Neurosci., 21: 4059–4065. decerebrate cat. Neurosci. Lett., 127: 87–90.
De Groat, W.C., Fraser, M.O., Yoshiyama, M., Smerin, S., Holstege, G. and Tan, J. (1987) Supraspinal control of
Tai, C., Chancellor, M.B., Yoshimura, N. and Roppolo, J.R. motoneurons innervating the striated muscles of the pelvic
(2001) Neural control of the urethra. Scand. J. Urol. floor including urethral and anal sphincters in the cat. Brain,
Nephrol. Suppl., 207: 35–43. 110: 1323–1344.
94

Hongo, T. and Jankowska, E. (1967) Effects from the Nakagawa, S. (1980) Onuf’s nucleus of the sacral cord in a
sensorimotor cortex on the spinal cord in cats with transected South American monkey (Saimiri): its location and bilateral
pyramids. Exp. Brain Res., 3: 117–134. cortical input from area 4. Brain Res., 191: 337–344.
Hounsgaard, J., Hultborn, H., Jespersen, B. and Kiehn, O. Onuf, B. (1899) Notes on the arrangement and function of the
(1988) Bistability of a-motoneurones in the decerebrate cat cell groups in the sacral region of the spinal cord. J. Nerv.
and in the acute spinal cat after intravenous 5-hydro- Ment. Dis., 26: 498–504.
xytryptophan. J. Physiol., 405: 345–367. Paroschy, K.L. and Shefchyk, S.J. (2000) Non-linear membrane
Hultborn, H. and Kiehn, O. (1992) Neuromodulation of ver- properties of sacral sphincter motoneurones in the decere-
tebrate motor neuron membrane properties. Curr. Opin. brate cat. J. Physiol., 523: 741–753.
Neurobiol., 2: 770–775. Pikov, V. and Wrathall, J.R. (2001) Coordination of the blad-
Hultborn, H., Lipski, J., Mackel, R. and Wigstrom, H. (1988) der detrusor and the external urethral sphincter in a rat
Distribution of recurrent inhibition within a motor nucleus. model of spinal cord injury: effect of injury severity.
I. Contribution from slow and fast motor units to the exci- J. Neurosci., 21: 559–569.
tation of Renshaw cells. Acta Physiol. Scand., 134: 347–361. Pullen, A.H., Tucker, D. and Martin, J.E. (1997) Morpholog-
Jankowska, E., Padel, Y. and Zarzecki, P. (1978) Crossed ical and morphometric characterisation of Onuf’s nucleus in
disynaptic inhibition of sacral motoneurones. J. Physiol., the spinal cord in man. J. Anat., 191: 201–213.
285: 425–444. Ramirez-Leon, V., Ulfhake, B., Arvidsson, U., Verhofstad,
Kakizaki, H., Fraser, M.O. and de Groat, W.C. (1997) Reflex A.A.J., Visser, T.J. and Hokfelt, T. (1994) Serotoninergic,
pathways controlling urethral striated and smooth muscle peptidergic and GABAergic innervation of the ventrolateral
function in the male rat. Am. J. Physiol., 272: R1647–R1656. and dorsolateral motor nuclei in the cat s1/s2 segments: an
Koyanagi, T., Arikado, K., Takamatsu, T. and Tsuji, I. (1982) immunofluorescence study. J. Chem. Neuroanat., 7: 87–103.
Experience with electromyography of the external urethral Roppolo, J.R., Nadelhaft, I. and de Groat, W.C. (1985) The
sphincter in spinal cord injury patients. J. Urol., 127: organization of pudendal motoneurons and primary afferent
272–276. projections in the spinal cord of the Rhesus monkey revealed
Kruse, M.N., Belton, A.L. and de Groat, W.C. (1993) Changes by horseradish peroxidase. J. Comp. Neurol., 234: 475–488.
in bladder and external urethral sphincter function after Russo, R.E., Nagy, F. and Hounsgaard, J. (1997) Modulation
spinal cord injury in the rat. Am. J. Physiol., 264: of plateau properties in dorsal horn neurones in a slice prep-
R1157–R1163. aration of the turtle spinal cord. J. Physiol., 499: 459–474.
Lassmann, G. (1984) Muscle spindles and encapsulated nerve Russo, R.E., Nagy, F. and Hounsgaard, J. (1998) Inhibitory
endings in external urethral sphincter. Acta Neuropathol. control of plateau properties in dorsal horn neurones in the
(Berl.), 63: 344–346. turtle spinal cord in vitro. J. Physiol., 506: 795–808.
Li, Y., Li, X., Harvey, P.J. and Bennett, D.J. (2004) Effects of Sackman, J.E. and Sims, M.H. (1990) Electromyographic eval-
baclofen on spinal reflexes and persistent inward currents in uation of the external urethral sphincter during cystometry in
motoneurons of chronic spinal rats with spasticity. J. Neuro- male cats. Am. J. Vet. Res., 51: 1237–1241.
physiol., 92: 2694–2703. Sasaki, M. (1991) Membrane properties of external urethral
Llewellyn-Smith, I.J., Cassam, A.K., Krenz, N.R., Krassioukov, and external anal sphincter motoneurones in the cat.
A.V. and Weaver, L.C. (1997) Glutamate- and GABA- J. Physiol., 440: 345–366.
immunoreactive synapses on sympathetic preganglionic neu- Sasaki, M. (1994) Morphological analysis of external urethral
rons caudal to a spinal cord transection in rats. Neuroscience, and external anal sphincter motoneurones of cat. J. Comp.
80: 1225–1235. Neurol., 349: 269–287.
Mackel, R. (1979) Segmental and descending control of the Schroder, H.D. (1981) Onuf’s nucleus X: a morphological study
external urethral and anal sphincters in the cat. J. Physiol., of a human spinal nucleus. Anat. Embryol., 162: 443–453.
294: 105–122. Shefchyk, S.J. (1989) The effects of lumbosacral deafferentation
McKenna, K.E. and Nadelhaft, I. (1986) The organization of on pontine micturition center-evoked voiding in the decere-
the pudendal nerve in the -male and female rat. J. Comp. brate cat. Neurosci. Lett., 99: 175–180.
Neurol., 248: 532–549. Shefchyk, S.J. (1998) Modulation of excitatory perineal reflexes
McMahon, S.B., Morrison, J.F.B. and Spillane, K. (1982) An and sacral striated sphincter motoneurons during micturition
electrophysiological study of somatic and visceral conver- in the cat. In: Rudomin P., Romo R. and Mendell L.M.
gence in the reflex control of the external sphincters. (Eds.), Presynaptic Inhibition and Neural Control. Oxford
J. Physiol., 328: 379–387. University Press, Oxford, pp. 398–406.
Morisset, V. and Nagy, F. (1999) Ionic basis for plateau po- Shefchyk, S.J. (2001) Sacral spinal interneurones and the con-
tentials in deep dorsal horn neurons of the rat spinal cord. trol of urinary bladder and urethral striated sphincter muscle
J. Neurosci., 19: 7309–7316. function. J. Physiol., 533: 57–63.
Nadelhaft, I. and Vera, P. (1996) Neurons in the rat brain and Shefchyk, S.J. and Buss, R.R. (1998) Urethral pudendal affer-
spinal cord labeled after pseudorabies virus injected into the ent-evoked bladder and sphincter reflexes in decerebrate and
external urethral sphincter. J. Comp. Neurol., 375: 502–517. acute spinal cats. Neurosci. Lett., 244: 137–140.
95

Shefchyk, S.J., Espey, M.J., Carr, P., Nance, D., Sawchuk, M. Smith, C.P., Somogyi, T. and Chancellor, M.B. (2002) Emerg-
and Buss, R. (1998) Evidence for a strychnine-sensitive ing role of botulinum toxin in the treatment of neurogenic
mechanism and glycine receptors involved in the control of and non-neurogenic voiding dysfunction. Curr. Urol. Rep.,
urethral sphincter activity during micturition in the cat. Exp. 3: 382–387.
Brain Res., 119: 297–306. Steers, W.D. (1989) Intrathecal baclofen administration —
Shimoda, N., Takakusaki, K., Nishizawa, O., Tsuchida, S. and insights into the regulation of genitourinary function and
Mori, S. (1992) The changes in the activity of pudendal therapy. J. Urol., 141: 142.
motoneurons in relation to reflex micturition evoked in Streng, T., Santti, R., Andersson, K.-E. and Talo, A. (2004)
decerebrate cats. Neurosci. Lett., 135: 175–178. The role of the rhabdosphincter in female rat voiding. BJU
Sie, J.A., Blok, B.F., de Weerd, H. and Holstege, G. (2001) Int., 94: 138–142.
Ultrastructural evidence for direct projections from the pon- Svirskis, G. and Hounsgaard, J. (1998) Transmitter regulation
tine micturition center to glycine-immunoreactive neurons in of plateau properties in turtle motoneurons. J. Neuro-
the sacral dorsal gray commissure in the cat. J. Comp. physiol., 79: 45–50.
Neurol., 429: 631–637.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 7

Neurochemical plasticity and the role of


neurotrophic factors in bladder reflex pathways after
spinal cord injury

Margaret A. Vizzard

Departments of Neurology and Anatomy and Neurobiology, University of Vermont College of Medicine, Burlington,
VT 05405, USA

Abstract: Transection of the spinal cord that interrupts the spinobulbospinal micturition reflex pathway,
abolishes voluntary voiding and initially produces an areflexic bladder with complete urinary retention.
However, depending upon the species, reflex bladder activity slowly recovers over the course of weeks or
months. In chronic spinal animals, reflex mechanisms in the lumbosacral spinal cord are capable of du-
plicating many of the functions performed by reflex pathways in animals with an intact spinal cord and can
induce bladder hyperreflexia. However, the bladder does not empty efficiently due to a loss of blad-
der–sphincter coordination (bladder–sphincter dyssynergia). In contrast to normal animals in which the
sphincter relaxes during voiding, animals with a spinal cord injury exhibit sphincter contractions during
voiding, an increase in urethral outlet resistance, urinary retention, bladder hyperreflexia, bladder over-
distension, and an increase in bladder afferent cell size. Changes in electrophysiological or neurochemical
properties of bladder afferent cells in the dorsal root ganglia and of spinal pathways could contribute to the
emergence of the spinal micturition reflex, bladder hyperreflexia and changes in the pharmacologic re-
sponses of reflex pathways in the lumbosacral spinal cord after spinal cord injury. Urinary bladder hype-
rreflexia after spinal cord injury may reflect a change in the balance of neuroactive compounds in bladder
reflex pathways. This review will detail: (1) changes in the neurochemical phenotype of bladder afferent
neurons and of spinal neurons mediating micturition reflexes after spinal cord injury, with an emphasis on
three neuroactive compounds, neuronal nitric oxide synthase (nNOS), galanin, and pituitary adenylate
cyclase activating polypeptide (PACAP); (2) possible functional consequences on bladder reflexes of
changes in spinal cord neurochemistry after spinal cord injury, and (3) the potential role of neurotrophic
factors expressed in the urinary bladder or spinal cord after spinal cord injury in mediating these neuro-
chemical changes.

Neural control of micturition and the smooth and striated muscle of the urethral
sphincters (Kuru, 1965; Klück, 1980; de Groat and
The storage and periodic elimination of urine re- Steers, 1990) (Fig. 1). Three neural pathways reg-
quires a complex neural control system that coor- ulate the lower urinary tract (Fig. 1): (1) sacral
dinates the activities of a variety of effector organs parasympathetic (pelvic) nerves provide excitatory
including the smooth muscle of the urinary bladder input to the bladder; (2) thoracolumbar sympathetic
(hypogastric) nerves provide an inhibitory input to
Corresponding author. Tel.: +802-656-3209; the bladder and an excitatory input to the bladder
Fax: +802-656-8704; E-mail: margaret.vizzard@uvm.edu neck and urethra; and (3) sacral somatic (pudendal)

DOI: 10.1016/S0079-6123(05)52007-7 97
98

Fig. 1. Neuroanatomy of micturition reflex circuitry. Postganglionic neurons that innervate the urinary bladder may be located in
intramural ganglia within the detrusor wall or in some species (e.g., rattus norvegicus), postganglionic neurons are located in pelvic
ganglia located in close proximity to the urinary bladder. From Fernandez (2002) copyright 2002 with permission.

nerves which innervate the striated muscles of the pathways are inactive (Kuru, 1965; de Groat and
sphincters and pelvic floor (Kuru, 1965; Klück, Kruse, 1993; de Groat et al., 1993; de Groat
1980; de Groat and Steers, 1990; Middleton and et al., 1997). During reflex or voluntary micturition,
Keast, 2004). Each of these sets of nerves contains the activity patterns are reversed such that para-
afferent (sensory) as well as efferent (motor) axons sympathetic pathways are excited and somatosym-
(Morrison, 1987; Lincoln and Burnstock, 1993). pathetic pathways are inhibited thereby promoting
The central neural pathways controlling the urine flow (Middleton and Keast, 2004) (Fig. 2).
lower urinary tract exhibit ‘‘all-or-none’’ or The lower urinary tract reflex mechanisms, or-
‘‘switch-like’’ characteristics reflecting the storage ganized at the level of the lumbosacral spinal cord,
and elimination functions of the lower urinary tract are modulated predominantly by supraspinal con-
(de Groat and Kruse, 1993; de Groat et al., 1993; trols (Kuru, 1965; de Groat, 1975; de Groat and
de Groat et al., 1997) (Fig. 2). During urine storage, Kruse, 1993; de Groat et al., 1993; de Groat et al.,
somatic and sympathetic pathways to the sphinc- 1997; Middleton and Keast, 2004). These mecha-
ters and sympathetic inhibitory inputs to the blad- nisms can be summarized as follows: (1) storage re-
der are tonically active, whereas parasympathetic flexes (parasympathetic and somatic) are organized
99

Fig. 2. Diagram illustrating the switch-like properties of the micturition reflexes. During urine storage, a low level of bladder afferent
activity activates efferent input to the external urethral sphincter (sympathetic and somatic nerves). A high level of afferent activity
induced by urinary bladder distention activates the switching circuit in the central nervous system resulting in the activation of efferent
pathways to the urinary bladder detrusor muscle (parasympathetic nerves), inhibition of efferent outflow to the sphincter and urine
outflow. From de Groat (1993) copyright 1993 with permission.

at the spinal level; (2) elimination reflexes (para- Emergence of automatic micturition after spinal
sympathetic) are organized at a supraspinal site in cord injury may be dependent on multiple factors,
the pons; and (3) spinal storage reflexes are mod- including: (1) elimination of bulbospinal inhibitory
ulated by inputs from the rostral pons. pathways; (2) strengthening of existing synapses, or
formation of new synaptic connections due to ax-
Spinal voiding reflexes after spinal cord injury onal sprouting in the spinal bladder reflex path-
ways; (3) changes in synthesis, release, or action of
Spinal cord injury above the lumbosacral spinal cord neurotransmitters in bladder reflex pathways;
(upper motoneuron injury) alters the coordination (4) alteration in afferent input from peripheral or-
between urinary bladder and sphincter and gans; or (5) central (spinal cord) and peripheral
chronically impairs micturition in humans, and in (urinary bladder) changes in the expression of
experimental animals (Kuru, 1965; de Groat et al., neurotrophic factors, which, in turn, would act by
1993). Spinal cord injury produces an initial period their influence on factors 2–5. A number of labo-
of urinary bladder areflexia that persists for several ratories have demonstrated electrical (Yoshimura,
weeks to months depending on the species. This 1999) and organization changes (Vizzard, 2000b)
period is followed by the emergence of a micturition in the central and peripheral components of the
reflex at the spinal level and the appearance of spon- micturition reflex pathways after spinal cord injury
taneous, involuntary bladder contractions (de Groat that may underlie the emergence of the spinal mi-
et al., 1993). However, the micturition reflex in cturition reflex with associated bladder dysfunction.
chronically spinalized animals is characterized by si- The changes that occur in spinal voiding reflexes
multaneous bladder and external urethral sphincter after spinal cord injury appear to be similar in
contractions (bladder–sphincter dyssynergia) leading humans and experimental animals and are begin-
to inefficient bladder emptying, large residual urine ning to provide important insights into a variety of
volumes and bladder hypertrophy (de Groat et al., neurogenic disorders of the lower urinary tract
1993). It has been suggested that bladder–sphincter (de Groat and Kruse, 1993; de Groat et al., 1993).
dyssynergia results from the loss of brainstem A major breakthrough has been the recognition
coordination mechanisms (de Groat et al., 1993). that C-fiber bladder afferents can reflexly trigger
100

bladder hyperactivity (de Groat et al., 1981, 1990, 1993; Vizzard et al., 1995b; de Groat et al., 1997)
1993). In cats with transected spinal cords, the have been used to examine the properties of uro-
properties of C-fiber bladder afferents are altered, genital afferent pathways in the lumbosacral spinal
so that they become mechanosensitive and re- cord (L6–S1). Anterograde transganglionic tracing
spond to bladder distension (de Groat et al., 1981, using horseradish peroxidase or wheat germ ag-
1990, 1993). In chronic spinal cord injury, C-fiber glutinin-conjugated horseradish peroxidase re-
afferent evoked bladder reflexes emerge. However, vealed that bladder afferents in rats pass through
in cats with an intact spinal cord, myelinated (A-q) the dorsal roots into Lissauer’s tract at the apex of
afferents activate the micturition reflex (de Groat the dorsal horn and then give off collaterals which
and Ryall, 1969; de Groat, 1975; de Groat et al., extend ventromedially and ventrolaterally along
1993). de Groat and colleagues (de Groat et al., the superficial layers of the dorsal horn to the
1990, 1993; de Groat and Kruse, 1993) have dem- dorsal commissure and to the area of the sacral
onstrated that systemically administered capsaicin, parasympathetic nucleus (laminae V–VII), which
a C-fiber neurotoxin, blocked bladder hype- contains preganglionic parasympathetic neurons.
rreflexia in the chronic paraplegic cat but was These afferents do not extend into the center of the
without effect in cats with intact spinal cords. In dorsal horn or into the ventral horn. The most
the rat, both the spinal and supraspinal micturit- prominent pathway is located in lamina I on the
ion reflexes are activated by capsaicin-resistant Aq lateral edge of the dorsal horn in a region termed
afferents (Mallory et al., 1989); by contrast, caps- the lateral collateral pathway of Lissauer’s tract.
aicin-sensitive afferents do appear to modulate Afferents from the uterine cervix, and from the
micturition under certain conditions (Maggi, 1991, urethra and external urethral sphincter, also
1993). In rats, C-fiber bladder afferents are not project heavily into the lateral collateral pathway.
necessary for eliciting bladder reflexes after spinal In contrast, afferents from the clitoris or penis
cord injury but do contribute to the appearance of project almost exclusively to the dorsal commis-
non-voiding bladder contractions (i.e., increases in sure. Thus, it has been concluded that excretory
bladder pressure not associated with release of reflexes depend on spinal processing in the regions
urine) after spinal cord injury (Cheng et al., 1995). of the lateral collateral pathway and sacral
The mechanisms underlying the emergence of the parasympathetic nucleus, whereas sexual reflexes
C-fiber evoked reflex are unknown. However, recent are processed in the dorsal commissure.
experiments have begun to examine changes in the The neuroactive compounds in the afferent
electrical properties of afferent neurons innervating pathways from the urogenital tract have been
the urinary bladder of the adult rat before and after examined using histochemical techniques (Donovan
spinal cord injury (Yoshimura, 1999). These studies et al., 1983; de Groat et al., 1986; Keast and de
suggest an ionic mechanism underlying the relative Groat, 1992; Vizzard et al., 1993b, c, 1994b;
inexcitability of C-fiber bladder afferents in normal Vizzard and de Groat, 1996). Bladder afferents
animals and the increased excitability of these af- contain a variety of neuropeptides, including
ferents after spinal cord injury (Yoshimura, 1999). calcitonin gene-related peptide, substance P,
Changes in the electrophysiological properties of vasoactive intestinal polypeptide, cholecystokinin,
bladder afferent neurons after spinal cord injury and enkephalins (Donovan et al., 1983; de Groat
may occur concomitantly with the changes in ne- et al., 1986; Vizzard, 2000d, 2001). Multiple
urochemical properties reviewed here. neuropeptides are present in the same cells sug-
gesting that transmission at afferent terminals in
Neurochemistry and morphology of afferent and the spinal cord and in the target organs is likely to
spinal pathways to the urogenital tract be complex and involve multiple neurotransmitters.
Our recent studies (Zvarova et al., 2004, 2005)
Axonal tracing and immunocytochemical tech- have also demonstrated that bladder afferent cells
niques (de Groat et al., 1981, 1986; Donovan et al., express pituitary adenylate cyclase-activating
1983; Steers et al., 1991a; de Groat and Kruse, polypeptide (PACAP) and galanin and that this
101

expression is increased after chronic cyclophosph- 1991; Vizzard et al., 1994a). In both the rat and
amide-induced cystitis or spinal cord injury (see cat, NADPH-d is present in a prominent afferent
below). With the exception of calcitonin gene- bundle projecting from Lissauer’s tract to the
related peptide, all of these substances are predom- region of the parasympathetic nucleus (Vizzard
inantly expressed in small (presumably C-fiber) et al., 1993a, c, 1994a, 1995a). This afferent path-
afferents (Donovan et al., 1983; de Groat et al., way closely resembles the central projections of the
1986; Keast and de Groat, 1992; Vizzard et al., afferent neurons innervating the pelvic viscera
1993b, c, 1994b; Vizzard and de Groat, 1996). (Steers et al., 1991a). In the cat, the NADPH-d
afferent pathways closely resemble the vasoactive
Neurochemical plasticity in bladder afferent cells in intestinal polypeptide-containing afferent projec-
dorsal root ganglia after spinal cord injury tions to the sacral spinal cord (Basbaum and
Glazer, 1983; Honda et al., 1983; Kawatani et al.,
The neuroactive compounds in the afferent path- 1985).
ways from the lower urinary tract and those in the Although NADPH-d was present in the primary
central pathways (interneurons, preganglionic neu- afferent neurons (Aimi et al., 1991; Vizzard et al.,
rons) exhibit either excitatory or inhibitory ac- 1993b, 1994b) and in their central projections in
tions. Pathological conditions can alter the known the rat (Vizzard et al., 1993a, c) and the cat
balance of these neuroactive compounds either in (Vizzard et al., 1994a, c), nNOS-IR was not iden-
the periphery or in the central pathways, conceiv- tified (Vizzard et al., 1994c, 1995a). These data
ably shifting the balance to a hyper- or hypo-active indicate that in pelvic afferent neurons in normal
state. Urinary bladder hyperreflexia, after spinal rats, NADPH-d is not a marker for nNOS and
cord injury (upper motoneuron injury), may reflect that NO is not a transmitter, or that immunore-
this change in the balance of neuroactive com- activity for nNOS is not as sensitive as NADPH-d
pounds in bladder reflex pathways. histochemistry. A similar situation exists for so-
matic afferent neurons in the adjacent lumbar
Nitric oxide (L4–L5) dorsal root ganglia that contain relatively
large numbers of NADPH-d positive neurons
Neuronal nitric oxide synthase (Ruda et al., 1994) but few nNOS-IR neurons in
Previous histochemical and pharmacological stud- rats (Verge et al., 1992; Zhang et al., 1993). However,
ies have raised the possibility that nitric oxide (NO) following sciatic nerve injury (Verge et al., 1992;
is a transmitter in autonomic reflex pathways. Zhang et al., 1993), pelvic nerve injury (Vizzard
In the rat, nicotinamide adenine dinucleotide et al., 1995a), or chronic bladder irritation
phosphate diaphorase (NADPH-d) activity (a pre- (Vizzard and de Groat, 1996) the levels of nNOS
sumed indicator of the presence of neuronal nitric protein or nNOS mRNA are markedly increased
oxide synthase) (Dawson et al., 1991; Hope et al., in the lumbosacral dorsal root ganglia. Increased
1991) and neuronal nitric oxide synthase (nNOS) NADPH-d staining or nNOS-IR has also
immunoreactivity (IR) have been identified in been noted following axotomy in motoneurons
sympathetic (Valtshanoff et al., 1992) and para- (Wu et al., 1994) and in some parasympathetic
sympathetic preganglionic neurons (Vizzard et al., preganglionic neurons (Vizzard et al., 1993a, 1995a).
1993c, 1995a; Saito et al., 1994) in the spinal cord Thus, expression of nNOS is plastic and neurons
and in some parasympathetic postganglionic neu- that do not normally synthesize nNOS can synthe-
rons in the peripheral ganglia. In cats, NADPH-d size the protein after injury or chemical stimulation.
activity is present in sympathetic preganglionic The possibility that nNOS participates in the
neurons but not in parasympathetic preganglionic pathophysiology of spinal cord injury has recently
neurons (Vizzard et al., 1994c). NADPH-d activity been investigated (Guizar-Sahagun et al., 1996;
is also present in a large percentage of visceral Sharma et al., 1996). Changes in the expression of
afferent neurons in dorsal root ganglia at various nNOS or NADPH-d activity in the myenteric
levels of the spinal cord of the rat (Aimi et al., plexus or thoracic spinal cord after spinal cord
102

injury have been demonstrated (Guizar-Sahagun parasympathetic nucleus in the L6–S1 spinal seg-
et al., 1996). Acute (1 day) spinal cord injury re- ments were significantly increased. In the L6–S1
sulted in increased numbers of NADPH-d positive sacral parasympathetic nucleus of animals with
cell bodies in the myenteric plexus. However, no intact spinal cords, an average of 5.270.4 cell
increase in NADPH-d positive cell bodies in the profiles/section (L6) and 4.870.6 cell profiles/sec-
myenteric plexus was observed after chronic (10 tion (S1) were nNOS-IR, whereas in animals with
weeks) injury (Guizar-Sahagun et al., 1996). Focal spinal cord injury, the L6–S1 sacral parasympa-
trauma to the dorsal horn of the thoracic thetic nucleus had an average of 9.370.9 cell pro-
(T10–T11) spinal cord of the rat significantly in- files/section (L6) and 10.370.4 cell profiles/section
creased the numbers of nNOS-IR spinal neurons (S1) that were nNOS-IR (Vizzard, 1997). In con-
in the perifocal T9 and T12 segments of the spinal trast, no difference in the numbers of nNOS-IR
cord (Sharma et al., 1996). Topical application of cells in the region of the intermediolateral cell nu-
nNOS antiserum, 2 min after injury, prevented the cleus in the L1–L2 segments was detected after
upregulation of nNOS-IR (Sharma et al., 1996). spinal cord injury (8 cell profiles/section in L1
These studies have raised the possibility that: (1) and 6 cell profiles/section in L2) (Vizzard, 1997).
nNOS participates in the pathogenesis of second- Following complete spinal cord transection, the
ary spinal damage after spinal cord injury and (2) number of nNOS-IR neurons increased approxi-
changes in nNOS expression in the gastrointestinal mately 30–70-fold in L6 and S1 dorsal root ganglia
tract may be relevant to its reduced motility after and 2–4-fold in L1 and L2 dorsal root ganglia.
spinal cord injury. However, there was not a significant change in the
numbers of nNOS-IR cells in the L5 dorsal root
ganglia (Vizzard, 1997). After spinal cord injury,
nNOS expression in lower urinary tract pathways the increase in nNOS-IR in the L6 and S1 dorsal
after spinal cord transection at the 8th thoracic root ganglia was highly significant (pp0.001;
segment 12–19 nNOS-IR cell profiles/section) as was the
Following spinal cord injury, nNOS-IR fibers were increase in nNOS-IR in the L1 and L2 dorsal root
detected along the lateral edge of the dorsal horn ganglia (p p 0.001; 15–40 cell profiles/section)
extending from Lissauer’s tract to the region of the (Vizzard, 1997). Following spinal cord injury,
sacral parasympathetic nucleus in the lumbar (L6) nNOS-IR was not restricted to the small and the
and sacral (S1) spinal segments (Vizzard, 1997). medium sized dorsal root ganglia neurons as noted
These fibers were not present in the adjacent spinal in control animals. After spinal cord injury,
segments (L4, L5, or S2) nor were they present at nNOS-IR was present in the medium (25–30 mm)
the rostral lumbar (L1–L2) spinal levels before or and in the large (430 mm) sized dorsal root
after spinal cord injury. The nNOS-IR fiber stain- ganglion neurons although occasional small
ing in the lateral collateral pathway was not dorsal root ganglion cells still exhibited nNOS-
present in every transverse section suggesting that IR (Vizzard, 1997).
the nNOS-IR fibers may occur intermittently To determine if the increase in nNOS-IR in
along the rostral–caudal axis as noted for viscer- dorsal root ganglion neurons was occurring after
al afferent projections labeled with wheat germ spinal cord injury in the urinary bladder afferent
agglutinin horseradish peroxidase or horseradish neurons, fluorogold was injected into the urinary
peroxidase (Steers et al., 1991a). The general lo- bladder of spinal cord injured animals 5–7 days
cation of the nNOS-IR fibers in lamina I and their prior to euthanasia. In the L6 and S1 dorsal root
selective segmental distribution are very similar to ganglia of spinal cord injured animals, an average
the central projections of the visceral afferents in of 31.373.0 cell profiles/section and 17.571.4 cell
the pelvic nerve, designated the lateral collateral profiles/section, respectively, were fluorogold-labe-
pathway of Lissauer’s tract (de Groat et al., 1986). led after injection of dye into the bladder (Vizzard,
Following spinal cord injury, the numbers 1997). In these ganglia, an average of 41.277.8%
of nNOS-IR cells in the region of the sacral (L6) and 36.370.9% (S1), respectively, of
103

fluorogold-labeled bladder afferent neurons were this hyperreflexia at the spinal cord level. The
nNOS-IR (Vizzard, 1997). In contrast, no fluoro- present results indicate that NO may have addi-
gold-labeled bladder afferent neurons were nNOS- tional functions in bladder primary afferent path-
IR in spinal cord intact animals. At rostral lumbar ways following spinal cord injury. It is possible
levels, a higher percentage of bladder afferents that NO may play a role in the sensitization of the
normally express nNOS-IR in comparison to bladder afferents or in changes in the central
lumbosacral dorsal root ganglia (5%). Follow- processing of afferent input that could contribute
ing spinal cord injury, a significantly greater per- to the pathologically induced alterations in lower
centage of dye-labeled bladder afferents in the L1 urinary tract function (de Groat et al., 1993;
and L2 dorsal root ganglia exhibited nNOS–IR de Groat and Kruse, 1993).
compared to spinal cord intact animals and the
percentage of fluorogold-labeled cells that were
Pituitary adenylate cyclase activating
nNOS-IR (20–55%) was similar to the percent-
polypeptide (PACAP)
age of fluorogold nNOS-IR cells in the L6–S1
dorsal root ganglia (Vizzard, 1997).
PACAP belongs to the vasoactive intestinal
polypeptide/secretin/ glucagon family of bioactive
peptides, and was isolated from the hypothalamus
Role of nitric oxide in lower urinary tract pathways
based on its stimulation of anterior pituitary
after spinal cord injury
adenylyl cyclase activity (Arimura, 1998). Two
The function of the NO formed by the enhanced
p-amidated forms of PACAP arise from alterna-
expression of nNOS in bladder afferent cells and
tive post-translational processing; PACAP38 has
lumbosacral preganglionic neurons following
38 amino acid residues [proPACAP(131–168)],
spinal cord injury is uncertain. However, the role
while PACAP27 corresponds to the N-terminus
of NO in the lower urinary tract after spinal cord
of PACAP38 [proPACAP(131–157)]. PACAP27
injury may be similar to its suggested role follow-
exhibits 68% homology with vasoactive intestinal
ing chronic chemical irritation/inflammation of the
polypeptide (Kimura et al., 1990). The relative
urinary bladder (Kakizaki and de Groat, 1996;
levels of the two forms are tissue-specific, although
Vizzard and de Groat, 1996). Although NO does
PACAP38 predominates in most tissues (Arimura,
not appear to be involved in the normal micturit-
1998). The rat PACAP precursor protein consists
ion reflex in the rat (Rice, 1995; Kakizaki and
of 175 amino acid residues (Kimura et al., 1990;
de Groat, 1996; Vizzard and de Groat, 1996) NO
Arimura, 1998; Braas et al., 1998); PACAP38 is
does appear to play a role in the facilitation of the
identical among mammalian species, suggesting
micturition reflex by noxious chemical irritation of
similar physiologically important roles for this
the bladder (Rice, 1995; Kakizaki and de Groat,
peptide. These peptides are abundantly expressed
1996). Bladder hyperreflexia induced by either
and have diverse functions as regulators, signaling
acetic acid (0.1%) (Kakizaki and de Groat, 1996)
modulators, and trophic factors in the nervous and
or turpentine (Rice, 1995) was partially antago-
endocrine systems (Arimura, 1998).
nized by intrathecal spinal cord administration of
NOS inhibitors. This suggests that NO is involved
at the spinal level in the facilitation of the mi- PACAP expression in lower urinary tract pathways
cturition reflex by nociceptive bladder afferents. after spinal cord transection at the 8th thoracic
The spinal micturition reflex pathway can also segment
produce bladder hyperreflexia in paraplegic and In rats with an intact spinal cord, PACAP is
urethral obstructed animals. Thus, NO may also expressed in nerve fibers in the superficial laminae
be involved in the facilitation of the spinal of the dorsal horn and dorsal commissure in all
micturition reflex following spinal cord injury. thoracic, lumbar, and sacral segments examined.
Increased expression of nNOS in lumbosacral Some PACAP staining in the intact spinal cord
preganglionic neurons could also contribute to was unique to specific levels with PACAP staining
104

being present in the lateral horn in L1–L2, and (Zvarova et al., 2005). At various times after
L6–S1 spinal segments. spinal cord injury (48 h to 6 weeks), PACAP–IR
At 6 weeks after spinal cord injury, PACAP–IR was significantly (pp0.001) increased in the rostral
increased in several regions in the rostral lumbar lumbar (L1–L2) and lumbosacral (L6–S1) dorsal
L1–L2 spinal cord compared to that in rats with root ganglia (Zvarova et al., 2005). Both small
intact spinal cords. The density of PACAP–IR (16.873.5 mm) and medium (24.072.0 mm) sized
increased in the superficial laminae (I–II) of the dorsal root ganglion cells expressed PACAP–IR in
dorsal horn having a denser distribution through- animals with intact or injured spinal cords.
out the entire medial (3.0-fold increase) to the lat- PACAP–IR was occasionally observed in larger
eral (7.0-fold increase) extent of the laminae (X30 mm) sized dorsal root ganglion cells. No
(Zvarova et al., 2005). Increased (17.0-fold in- change in numbers of cells expressing PACAP–IR
crease) PACAP–IR fiber staining was also present, was observed in the L4–L5 dorsal root ganglia at
after spinal cord injury, in a small fiber bundle any time after spinal cord injury.
extending laterally from Lissauer’s tract (LT) in To determine if the increase in PACAP–IR in
lamina I into the dorsolateral funiculus (Zvarova the lumbosacral dorsal root ganglia neurons after
et al., 2005). No dramatic changes in PACAP–IR cord injury was occurring in bladder afferent cells,
were observed in the region of the intermediolat- Fast Blue (FB) was injected into the urinary blad-
eral nucleus following spinal cord injury. der to label bladder afferent cells retrogradely in
PACAP–IR was unchanged in the L4–L5 the L1, L2, L6, S1 dorsal root ganglia (Fig.
segments after spinal cord injury in every region 4A–4C) (Zvarova et al., 2005). In animals with
examined: dorsal horn, dorsal commissure, or intact spinal cords, approximately 45% of bladder
lateral horn regions. In contrast, significant afferent cells in the L6–S1 dorsal root ganglia ex-
changes in PACAP–IR were detected in the hibited PACAP–IR (Fig. 4D). A similar percent-
L6–S1 spinal cord after spinal cord injury (Fig. age (40%) of bladder afferent cells in rostral
3B,D). In the L6 spinal segment, PACAP–IR was lumbar dorsal root ganglia (L1–L2) of control an-
dramatically increased in the dorsal horn (1.4–7.4- imals also exhibited PACAP–IR. After spinal
fold increase), dorsal commissure (11.7-fold in- cord injury (6 weeks), the percentage of bladder
crease), sacral parasympathetic nucleus (15.0-fold afferent cells exhibiting PACAP–IR significantly
increase), and lateral collateral pathway (17.0-fold (p p 0.001) increased in the L6 (88.872.2%) and
increase) (Fig. 3E) (Zvarova et al., 2005). Changes S1 dorsal root ganglia (80.272.5%) and in the
in PACAP–IR in the S1 segment were comparable L1–L2 dorsal root ganglia (L1, 74.873.5%; L2,
to those in the L6 segment after spinal cord injury 69.573.2%) (Fig. 4D) (Zvarova et al., 2005).
(Fig. 3D) (Zvarova et al., 2005). In some trans- Increases in the percentage of bladder afferent cells
verse sections of the L6–S1 spinal cord, PACA- expressing PACAP–IR after spinal cord injury were
P–IR axons in the lateral collateral pathway observed at the earliest time point after spinal cord
terminated at the base of the dorsal horn (Fig. 3B) injury (48 h) and were maintained up to 6 weeks
whereas in others, they extended medially toward after spinal cord injury with little variation with
the central canal in distinct bundles through time after injury (Fig. 4D) (Zvarova et al., 2005).
laminae V–VII (Fig. 3D) (Zvarova et al., 2005).
In contrast to PACAP–IR in the spinal cord, PACAP neuronal functions in the lower urinary
PACAP–IR in the dorsal root ganglia (L1–S1) was tract
expressed by neuronal cell bodies and fibers PACAP have diverse functions in the endocrine,
throughout each dorsal root ganglion examined. nervous, gastrointestinal, and cardiovascular sys-
In control animals, PACAP–IR was present in tems (Braas and May, 1996; Arimura, 1998). High
small numbers of cells in the L1–S1 dorsal root levels of PACAP and vasoactive intestinal poly-
ganglia. The number of PACAP–IR cells among peptide expression have been identified in many
the dorsal root ganglia examined was comparable CNS neurons and in sensory and autonomic gan-
(range 20–24 PACAP–IR cell profiles/section) glia (Sundler et al., 1996; Brandenburg et al., 1997;
Fig. 3. Fluorescence photomicrographs showing PACAP–IR in the dorsolateral quadrant of L6 (A,B) and S1 (C,D) spinal segments in control animals (A, C) and after
spinal cord injury (SCI, 6 weeks, B, D). Increased density of PACAP–IR was observed in the medial to lateral extent of the superficial laminae (I–II) of the dorsal horn
(DH) following SCI (A vs. B). Increased PACAP–IR was present in a fiber bundle (B) extending from Lissauer’s tract in lamina I along the lateral edge of the DH to the
region of the sacral parasympathetic nucleus (SPN) (lateral collateral pathway of Lissauer, LCP). Although this fiber bundle was present in control tissue sections, the
staining was less intense (C) and was less frequently observed in transverse sections compared to that after SCI. Faint PACAP–IR was present in the region of the SPN in
control sections (A,C) and was increased after SCI (B,D). Some PACAP–IR fibers in the LCP appeared to terminate in the region of the SPN, whereas others projected
medially toward the central canal (D, arrows). CC, central canal; DCM, dorsal commissure. Calibration bar represents 125 mm. (E). Histogram summarizing changes in
PACAP staining density in specific regions of the L6 spinal cord segment after spinal cord injury (SCI, 6 weeks). The spinal cord inset depicts the areas analyzed: medial
dorsal horn (MDH), lateral dorsal horn (LDH), lateral collateral pathway of Lissauer (LCP), sacral parasympathetic nucleus (SPN), dorsal commissure (DCM), and
ventral horn (VH). The density of PACAP–IR was significantly increased in the LDH, MDH, SPN, DCM, and LCP of the L6 spinal cord segment. Similar changes were
observed in the S1 spinal cord segment. *pp0.001. Reprinted from Zvarova et al. (2005) copyright 2005 with permission from Elsevier.

105
106
Fig. 4. Effect of spinal cord injury on PACAP expression by dorsal root ganglion cells. PACAP–IR in the L6 dorsal root ganglion (DRG) after spinal cord injury (SCI)
(A, B, C). (A) Fast Blue (FB) labeled bladder afferent cells in a L6 DRG section after SCI. (B) Same L6 DRG section shown in (A) immunostained for PACAP–IR.
PACAP–IR was primarily located in small and medium sized DRG cells. Bladder afferent cells expressing PACAP–IR are indicated by white arrows (A, B). (C) Merged
image of panels (A, B) with FB cells pseudocolored blue and PACAP–IR cells pseudocolored red. FB cells (presumptive bladder afferents) expressing PACAP–IR appear
pinkish-purple (white arrows). Some PACAP–IR cells do not show FB (red cells, yellow arrows). (D) After SCI, a significantly greater percentage (85%) of FB-labeled
bladder afferent cells expressed PACAP–IR at all time points examined; however, not all bladder afferent cells expressed PACAP after SCI. In addition, not all
PACAP–IR in the L6 DRG is accounted for by bladder afferent cells (B,C, yellow arrows). Calibration bar represents 40 mm in (A, B, C). *pp0.001. Reprinted from
Zvarova et al. (2005) copyright 2005 with permission from Elsevier.
107

Arimura, 1998; Braas et al., 1998). In neurons, 2000d) and spinal cord injury (Zvarova et al.,
PACAP facilitates calcium ion flux, induces mem- 2005) may represent a principal component of
brane depolarization, increases spike frequency, bladder hyperreflexia, by increasing excitability of
activates potently adenylyl cyclase and phospholi- sensory neurons in the bladder reflex arc.
pase C signaling, and stimulates neurotransmitter
secretion (May and Braas, 1995; Braas and May,
Galanin
1996, 1999; May et al., 1998; Beaudet et al., 2000).
Several immunocytochemical studies using a spe-
Previous studies have demonstrated that galanin
cific monoclonal antibody against PACAP
has a potent neuromodulatory action on the isolat-
demonstrate widespread PACAP–IR in nerve
ed human detrusor muscle where galanin suppresses
fibers along the rat urinary tract including the
the cholinergic component of the response to elec-
bladder smooth muscle, suburothelial plexuses,
tric field stimulation (Maggi et al., 1987). Thus, an
and blood vessels (Fahrenkrug and Hannibal,
inhibitory action for galanin on neurotransmitter
1998). Neonatal capsaicin (C-fiber neurotoxin)
release has been suggested in smooth muscle tissues
treatment significantly reduced this distribution
and may also pertain to the urinary bladder (Maggi
in adults (Fahrenkrug and Hannibal, 1998) sug-
et al., 1987). Galanin-IR was expressed in identical
gesting that the majority of the fibers are derived
spinal cord regions in animals with intact or injured
from small sensory neurons. Dorsal root ganglia
spinal cords. However, the intensity and the overall
with high PACAP expression demonstrate dra-
distribution of the staining were increased in specific
matic neurochemical plasticity during altered
spinal cord segments and regions after cord injury
physiological states (Vizzard, 2000d; Zvarova
(Zvarova et al., 2004). Increases in galanin expres-
et al., 2005). Many studies have demonstrated
sion in bladder afferent cells in the dorsal root gan-
changes in PACAP expression in sensory neurons
glia may therefore act to oppose the actions of
following nerve injury (i.e., axotomy) (Zhang
PACAP and nNOS, whereas a decrease would re-
et al., 1996; Larsen et al., 1997). A limited number
inforce these actions in micturition reflex pathways
of studies have examined PACAP expression fol-
after spinal cord injury (Vizzard, 1997; Vizzard
lowing the induction of inflammatory states. Pre-
et al., 2003). Significant changes in galanin expres-
vious studies have suggested an association of
sion were found after spinal cord injury in specific
PACAP expression with inflammation in sensory
regions of the L1, L2, L4, and S1 spinal segments,
neurons following either somatic (hindpaw) or oc-
suggesting a modulatory role for galanin in spinal
ular inflammation (Wang et al., 1996; Zhang et al.,
micturition reflex pathways.
1998). Our laboratory was the first to demonstrate
an up-regulation of PACAP levels in bladder
afferent cells and spinal cord projections follow- Galanin expression in lower urinary tract pathways
ing cyclophosphamide-induced cystitis (Vizzard, after spinal cord transection at the 8th thoracic
2000d) or spinal cord injury (Zvarova et al., 2005). segment
Cyclophosphamide-induced cystitis is character- Galanin-IR was expressed in identical spinal cord
ized by an increased frequency of voiding in awake regions in animals with intact or injured spinal
rats and by urinary bladder overactivity in anest- cords. However, the intensity and the overall dis-
hetized rats (Lecci et al., 1994). Spinal cord injury tribution of the staining were increased in specific
rostral to the lumbosacral spinal cord results in spinal cord segments and regions after cord injury
bladder hyperreflexia and bladder–sphincter dys- (Zvarova et al., 2004). In the intact spinal cord,
synergia (Kruse et al., 1993; Vizzard, 1997, 2000a, galanin-IR was present in nerve fibers but not in
b). As studies have shown that PACAP facilitates neuronal cell bodies as in the injured spinal cord.
spontaneous bladder contractions in control ani- In the L1 spinal cord segments, the density of
mals (Ishizuka et al., 1995), the observed increase galanin-IR was significantly decreased (pp0.001)
in PACAP expression in bladder afferent cells and in the superficial laminae (I–II) (2-fold) of the
spinal cord projections during cystitis (Vizzard, dorsal horn and in the lateral collateral pathway
108

following spinal cord injury. In contrast, in the L2 Role of neurotrophic factors in neuronal
spinal segments the only changes in galanin-IR were plasticity and lower urinary tract dysfunction after
increases in the intermediolateral nucleus (Zvarova spinal cord injury
et al., 2004). In the L4–L6 segments galanin-IR was
increased in the dorsal commissure region of the L4 Neurotrophic factors
segment after cord injury. In the S1 spinal segment,
galanin-IR was increased in the dorsal commissure A potential mechanism underlying the neurochem-
(1.2-fold), lateral collateral pathway (1.4-fold), and ical changes (Vizzard and de Groat, 1996; Vizzard,
sacral parasympathetic nucleus (1.4-fold). 2000b, c, d) in bladder afferent neurons after spinal
In contrast to galanin-IR in the spinal cord, cord injury (described above) may involve ne-
galanin-IR in the dorsal root ganglia (L1–S1) was urotrophic factors expressed in the urinary bladder
expressed consistently by both neuronal cell bodies or spinal cord or changes in neural activity
and fibers (Fig. 5A1, B1). In control animals, (Vizzard, 2000a). The concept of trophic interac-
galanin-IR was present in modest numbers of cells tions between nerve cells and their targets is clearly
in the L1–S1 dorsal root ganglia (Fig. 5A1, B1, C) demonstrated during embryonic or postnatal
(Zvarova et al., 2004). The number of galanin-IR development (Oppenheim et al., 1991). Recent
cells among the dorsal root ganglia examined was experiments from several laboratories have
comparable (range 12–29 galanin-IR cell profiles/ demonstrated the influence of target organ–neuron
section). At 6 weeks after spinal cord injury, interactions in the adult animal (Steers and
galanin-IR was significantly (pp0.001) increased de Groat, 1988; Steers et al., 1991a, b; Tuttle and
in the rostral lumbar (L1) and sacral (S1) dorsal Steers, 1992; Tuttle et al., 1994; Vizzard, 2000a).
root ganglia (Fig. 5A). Both small (17.574.2 mm) A large number of studies have demonstrated
and medium (23.573.5 mm) sized dorsal root that pathological changes in a target organ after
ganglion cells expressed galanin-IR in control spinal cord injury can alter the neurochemical
animals following spinal cord injury. No change in (Vizzard, 1997; Yoshimura, 1999; Vizzard et al.,
numbers of cells expressing galanin-IR was 2003), electrical (Yoshimura, 1999), and organiza-
observed in the L2, L4–L6 dorsal root ganglia tional (Vizzard, 2000b) properties of micturition
following spinal cord injury (Fig. 5C) (Zvarova reflex pathways. A possible mechanism underlying
et al., 2004). We examined galanin-IR after acute these changes may involve neurotrophic factors or
spinal cord injury (o1 week) to determine if we neural activity arising in the bladder. Previous
had missed an earlier increase in galanin expres- experiments have demonstrated target organ to
sion that might have returned to control levels by 6 neuron interactions in the adult animal (Steers and
weeks after spinal cord injury. The number of de Groat, 1988; Steers et al., 1991a, b,1996; Tuttle
galanin-IR cells in the dorsal root ganglia after et al., 1994; Zvara et al., 2002). Furthermore,
acute spinal cord injury (o1 week) was not a recent study from this laboratory has demon-
different from control (Fig. 5C). strated changes in mRNA or protein expression
To determine if galanin-IR was expressed in of neurotrophic factors in the urinary bladder after
bladder afferent cells, F B dye was injected into the complete spinal cord injury, including nerve
urinary bladder to retrogradely label bladder growth factor, brain-derived neurotrophic factor,
afferent cells in the L1, L2, L6, S1 dorsal root glial-derived neurotrophic factor, neurotrophin-3
ganglia (Fig. 5A2, B2). In control animals, and -4 (Vizzard, 2000a). Both acute and chronic
approximately 1.5% of bladder afferent cells in spinal cord injury (4–6 weeks) resulted in signifi-
the L1, L2, L6, or S1 dorsal root ganglia exhibited cant increases in nerve growth factor, brain-derived
galanin-IR (Fig. 5B2, 5D) (Zvarova et al., 2004). neurotrophic factor, glial-derived neurotrophic
Following spinal cord injury (6 weeks), the factor, neurotrophin-3 and -4 transcript expres-
percentage of bladder afferent cells exhibiting sion as well as in increased nerve growth factor
galanin-IR significantly increased in the L1–L2, protein expression in urinary bladder (Vizzard,
L6, and S1 dorsal root ganglia (Fig. 5D). 2000a).
109

Fig. 5. (A, B) Bladder afferent cells in lumbosacral dorsal root ganglia (DRG) express galanin (Gal)-immunoreactivity (IR). (A1, B1).
Fluorescence photographs of Gal-IR cells (arrows and arrowheads) in the L1 DRG from control (A1) and spinal cord injured (SCI)
(B1) rats. Bladder afferent cells in the DRG were labeled by retrograde transport of Fast Blue (FB; A2, B2). Some bladder afferent cells
express Gal-IR before (A1, A2, arrowheads) and after SCI (B1, B2, arrowheads). Note that not all bladder afferent cells express Gal-
IR (B2, arrow) and not all Gal-IR cells are bladder afferent cells (A1, B1; arrows). Calibration bar represents 100 mm. (C) Histogram
depicting the number of Gal-IR DRG cells per section in DRG examined in control (spinal intact) and SCI rats 6 weeks or less than 1
week (o week) after SCI. No changes in the numbers of Gal-IR cells in DRG examined were observed less than 1 week after SCI. In
contrast, significant increases in the numbers of Gal-IR cells in the L1 and S1 DRG were observed 6 weeks after SCI. (D) Histogram
depicting the percentage of bladder afferent cells in the DRG expressing Gal-IR in control or SCI rats. Six weeks after SCI, the
percentage of bladder afferent cells expressing Gal-IR significantly increased in all DRG examined. *pp0.001. Reprinted from
Zvarova et al. (2004) copyright 2004 with permission.

It has also been reported that nerve growth fac- growth factor content in spinal segments immedi-
tor levels increase in the transected spinal cord ately rostral to the T8 transection site (Fig. 6A).
(Krenz and Weaver, 2000; Brown et al., 2004) after However, some of the spinal segments caudal to
spinal cord injury. Our work (Zvarova et al., 2004) the transection site (T9–T10; T13–L1; L6–S1)
also demonstrated a significant increase in nerve exhibit decreased nerve growth factor protein
110

Fig. 6. (A). Changes in total spinal cord nerve growth factor (NGF) as detected with a NGF ELISA (Enzyme Linked Immunosorbant
Assay) after spinal cord injury (SCI) (o 1 week or 6 weeks). The line drawing at the top represents the spinal cord and the vertical line
indicates the position of the spinal cord transection at (T8). A significant increase in total NGF in the T7–T8 spinal segments was
present 6 weeks after SCI. Significant decreases in total NGF in the T9–T10, T13–L1, and L6–S1 spinal segments were present after
acute or chronic SCI compared to control values. *pp0.001. (B) Changes in total spinal cord brain-derived neurotrophic factor
(BDNF) as detected with a BDNF ELISA after SCI o1 week or 6 weeks. The line drawing at the top represents the spinal cord and the
vertical line indicates the position of the spinal cord transection at (T8). A significant increase in total BDNF from the T7–T8, T9–T10,
T13–L1, and L6–S1 spinal segments were detected after acute (o1 week) or chronic (6 weeks) SCI. Significant increases in spinal cord
BDNF were also seen acutely after SCI in the T11–T12 and L4–L5 spinal segments but not after chronic SCI. *pp0.001. Reprinted
from Zvarova et al. (2004) copyright 2004 with permission.
111

content with acute or chronic spinal cord injury At sites of tissue injury, inflammation or target
(Fig. 6A). In contrast, brain-derived neurotrophic organ hypertrophy, cytokines and growth factors
factor protein content significantly increased in the are up-regulated and this can result in the up-
majority of spinal segments examined (Fig. 6B). regulation of nerve growth factor (Lewin and
No spinal segments exhibited a decrease in brain- Mendell, 1993; Woolf et al., 1997) (Fig. 7). Nerve
derived neurotrophic factor protein content after growth factor activates TrkA receptors on axon
spinal cord injury (Fig. 6B). Thus, bladder afferent terminals in the urinary bladder or spinal cord re-
neurons may have at least two potential sources of sulting in internalization and retrograde transport
increased brain-derived neurotrophic factor fol- of activated TrkA (Kuruvilla et al., 2004) to affer-
lowing spinal cord injury: (1) central terminals in ent cells in the dorsal root ganglia. Excess nerve
the spinal cord and (2) peripheral terminals in the growth factor within the dorsal root ganglia may
urinary bladder (Vizzard, 2000a). The results of induce increased production of neuropeptides
these studies may focus new attention on the po- (i.e., substance P, calcitonin gene-related peptide
tential role of brain-derived neurotrophic factor in and PACAP) in sensory neurons (Gary and
micturition reflex plasticity after spinal cord injury. Hargreaves, 1992; Woolf et al., 1997) (Fig. 7). An
A model of nerve growth factor-dependent increase in the levels of neuroactive compounds
sensory consequences of tissue damage and in- (e.g., enkephalin (Lewin and Mendell, 1993),
flammation in the somatic system has been pro- dynorphin (Ruda et al., 1988), calcitonin gene-
posed (Lewin and Mendell, 1993). This proposed related peptide (Gary and Hargreaves, 1992; Woolf
scheme has been modified for this review to dem- et al., 1997; Vizzard, 2001), substance P (Ruda
onstrate how an excess of nerve growth factor or et al., 1988; Gary and Hargreaves, 1992; Lewin and
another neurotrophic factor in the urinary bladder Mendell, 1993; Vizzard, 2001), neuropeptide Y
or spinal cord could alter lower urinary tract (Lewin and Mendell, 1993), nNOS (Vizzard et al.,
pathways after spinal cord injury, and is shown in 1995a; Vizzard and de Groat, 1996; Vizzard, 1997),
Fig. 7. In addition to peripheral afferent changes and PACAP (Jongsma et al., 2000; Vizzard, 2000d)
after spinal cord injury, it is clear that central reflex following noxious peripheral stimulation, cyclophos-
mechanisms are also changed. phamide-induced cystitis (Vizzard, 2000c, d, 2001;

Fig. 7. Proposed involvement of urinary bladder or spinal cord neurotrophic factors in plasticity of micturition reflexes after spinal
cord injury (SCI).
112

Vizzard and de Groat, 1996), or spinal cord injury mentorship and support. Gratitude is also ex-
(Vizzard and de Groat, 1996; Vizzard, 2000b) has pressed to Dr. Victor May and Dr. Karen Braas,
also been demonstrated in dorsal root ganglion cells University of Vermont, for encouraging me to
as well as in spinal cord neurons. Furthermore, in- explore the role of PACAP in lower urinary tract
travesical administration of exogenous nerve growth reflexes. Many former and current members of
factor in animals may facilitate afferent firing and my laboratory have contributed to the studies
induce bladder hyperreflexia that is blocked by anti- discussed in this review; including Dr. Li-ya Qiao,
nerve growth factor treatment (Dmitrieva Dr. Katarina Zvarova, Mr. Dana J. Dunleavy,
et al., 1997). Over-expression of nerve growth fac- Ms. Elaine Murray, and Ms. Susan Malley.
tor in bladder smooth muscle in spontaneously hy-
pertensive rats leads to bladder hyperinnervation References
and bladder overactivity (Clemow et al., 1998).
Immunoneutralization of nerve growth factor in the Aimi, Y., Fujimura, M., Vincent, S.R. and Kimura, H. (1991)
lumbosacral spinal cord suppresses detrusor–sphinc- Localization of NADPH-diaphorase containing neurons in
ter dyssynergia in spinal cord-injured rats (Seki et sensory ganglia of the rat. J. Comp. Neurol., 306: 382–392.
Arimura, A. (1998) Perspectives on pituitary adenylate cyclase
al., 2004). Thus, changes in the expression of ne- activating polypeptide (PACAP) in the neuroendocrine, en-
urotrophic factors after spinal cord injury may result docrine, and nervous systems. Jpn. J. Physiol., 48: 301–331.
in changes in the neurochemical phenotype of blad- Basbaum, A.I. and Glazer, E.J. (1983) Immunoreactive vaso-
der afferent cells in dorsal root ganglia as well as active intestinal polypeptide is concentrated in the sacral spi-
nal cord: a possible marker for pelvic visceral afferent fibers.
contribute to urinary bladder dysfunction (Fig. 7).
Somatosens. Res., 1: 69–82.
Beaudet, M.M., Parsons, R.L., Braas, K.M. and May, V.
Conclusions (2000) Mechanisms mediating pituitary adenylate cyclase-ac-
tivating polypeptide depolarization of rat sympathetic neu-
Spinal cord injury rostral to the lumbosacral spinal rons. J. Neurosci., 20: 7353–7361.
cord (upper motoneuron injury) results in dramatic Braas, K.M. and May, V. (1996) Pituitary adenylate cyclase-ac-
tivating polypeptides, PACAP-38 and PACAP-27, regulation of
changes in the neurochemistry of peripheral and sympathetic neuron catecholamine, and neuropeptide Y expres-
central micturition reflex pathways. Changes in the sion through activation of type I PACAP/VIP receptor iso-
neurochemical properties of these neurons after spi- forms. Ann. N. Y. Acad. Sci., 805: 204–216 discussion 217–218.
nal cord injury may be mediated by pathological Braas, K.M. and May, V. (1999) Pituitary adenylate cyclase-
changes in the target organ (i.e., urinary bladder) or activating polypeptides directly stimulate sympathetic neuron
neuropeptide Y release through PAC(1) receptor isoform
spinal cord and contribute to urinary bladder hype- activation of specific intracellular signaling pathways. J. Biol.
rreflexia. Neurotrophic factors expressed in the Chem., 274: 27702–27710.
hypertrophied urinary bladder or spinal cord after Braas, K.M., May, V., Harakall, S.A., Hardwick, J.C. and
spinal cord injury are likely to contribute to this Parsons, R.L. (1998) Pituitary adenylate cyclase-activating
polypeptide expression and modulation of neuronal excitabil-
neurochemical plasticity. Distinguishing between
ity in guinea pig cardiac ganglia. J. Neurosci., 18: 9766–9779.
beneficial or detrimental effects of these neuroactive Brandenburg, C.A., May, V. and Braas, K.M. (1997) Identi-
compounds and neurotrophic factors in the context fication of endogenous sympathetic neuron pituitary adeny-
of micturition reflexes or regenerative responses will late cyclase-activating polypeptide (PACAP): depolarization
be a challenge but is essential to understanding the regulates production and secretion through induction of
effects of therapies directed at blocking the effects of multiple propeptide transcripts. J. Neurosci., 17: 4045–4055.
Brown, A., Ricci, M.J. and Weaver, L.C. (2004) NGF message
neuroactive compounds or neurotrophic factors. and protein distribution in the injured rat spinal cord. Exp.
Neurol., 188: 115–127.
Acknowledgments Cheng, C.L., Ma, C.P. and de Groat, W.C. (1995) Effect of
capsaicin on micturition and associated reflexes in chronic
spinal rats. Brain Res., 678: 40–48.
This work was supported by NIH grants
Clemow, D.B., Steers, W.D., McCarty, R. and Tuttle, J.B.
DK051369, NS040796, DK065989, DK060481. (1998) Altered regulation of bladder nerve growth factor and
Gratitude is expressed to Dr. William C. de Groat, neurally mediated hyperactive voiding. Am. J. Physiol., 44:
University of Pittsburgh, for his tremendous R1279–R1286.
113

Dawson, T.M., Bredt, D.S., Fotuhi, M., Hwang, P.M. and Gary, M.B. and Hargreaves, K.M. (1992) Enhanced release of
Snyder, S.H. (1991) Nitric oxide synthase and neuronal immunoreactive CGRP and substance P from spinal dorsal
NADPH diaphorase are identical in brain and peripheral horn slices occurs during carrageenan inflammation. Brain
tissues. Proc. Natl. Acad. Sci., 88: 7797–7801. Res., 582: 139–142.
de Groat, W.C. (1975) Nervous control of the urinary bladder Guizar-Sahagun, G., Garcia-Lopez, P., Espitia, A.L., Mendez,
of the cat. Brain Res., 87: 201–211. S., Castaneda-Hernandez, G., Madrazo, I. and Franco-
de Groat, W.C. (1993) Anatomy and physiology of the lower Bourland, R.E. (1996) Histochemical evidence of the in-
urinary tract. Urol. Clin. North Am., 20: 383–401. creased expression of nicotinamide adenine dinucleotide
de Groat, W.C., Booth, A.M. and Yoshimura, N. (1993) phosphate-dependent diaphorase in neurons of the myenter-
Neurophysiology of micturition and its modification in ic plexus after acute spinal cord injury in adult rats. Neurosci.
animal models of human disease. In: Maggi C.A. (Ed.) The Lett., 206: 185–188.
Autonomic Nervous System, Vol. 3. Harwood Academic Honda, C.N., Rethelyi, M. and Petrusz, P. (1983) Preferential
Publishers, London, pp. 227–290. immunohistochemical localization of vasoactive intestinal po-
de Groat, W.C., Kawatani, M., Hisamisu, T., Cheng, C.-L., lypeptide (VIP) in the sacral spinal cord of the cat: light and
Ma, C.P., Thor, K., Steers, W. and Roppolo, J.R. (1990) electron microscopic observations. J. Neurosci., 3: 2183–2196.
Mechanisms underlying the recovery of urinary bladder Hope, B.T., Michael, G.J., Jnigge, K.M. and Vincent, S.R.
function following spinal cord injury. J. Auton. Nerv. Syst., (1991) Neuronal NADPH diaphorase is a nitric oxide synt-
30: S71–S78. hase. Proc. Natl. Acad. Sci., 88: 2811–2814.
de Groat, W.C., Kawatani, M., Hisamitsu, T., Booth, A.M., Ishizuka, O., Alm, P., Larsson, B., Mattiasson, A. and
Roppolo, J.R., Thor, K., Tuttle, P. and Nage, J. (1986) Andersson, K.E. (1995) Facilitatory effect of pituitary adeny-
Neural control of micturition: the role of neuropeptides. late cyclase-activating polypeptide on micturition in normal,
J. Auton. Nerv. Syst., 9(Suppl): 369–387. conscious rats. Neuroscience, 66: 1009–1014.
de Groat, W.C. and Kruse, M.N. (1993) Central processing Jongsma, H., Danielsen, N., Sundler, F. and Kanje, M. (2000)
and morphological plasticity in lumbosacral afferent path- Alteration of PACAP distribution and PACAP receptor
ways from the lower urinary tract. In: Mayer E.A. and binding in the rat sensory nervous system following sciatic
Raybould H.E. (Eds.) Basic and Clinical Aspects of nerve transection. Brain Res., 853: 186–196.
Chronic Abdominal Pain. Pain Research and Clinical Kakizaki, H. and de Groat, W.C. (1996) Role of nitric oxide in
Management, Vol. 9. Elsevier Science Publishers, Amster- the facilitation of the micturition reflex by bladder irritation.
dam, pp. 219–236. J. Urol., 155: 355–360.
de Groat, W.C., Kruse, M.N., Vizzard, M.A., Cheng, C.-L., Kawatani, M., Lowe, I., Nadelhaft, I., Morgan, C. and
Araki, I. and Yoshimura, N. (1997) Modification of urinary de Groat, W.C. (1985) Vasoactive intestinal polypeptide
bladder function after spinal cord injury. In: Seil F.J. (Ed.) and substance P in primary afferent pathways to the sacral
Advances in Neurology, Vol. 72. Raven Press, New York, spinal cord of the cat. J. Comp. Neurol., 241: 311–316.
pp. 347–364. Keast, J.R. and de Groat, W.C. (1992) Segmental distribution
de Groat, W.C., Nadelhaft, I., Milne, R.J., Booth, A.M., and peptide content of primary afferent neurons innervating
Morgan, C. and Thor, K. (1981) Organization of the sacral the urogenital organs and colon of male rats. J. Comp.
parasympathetic reflex pathways to the urinary bladder and Neurol., 319(4): 615–623.
large intestine. J. Auton. Nerv. Syst., 3: 135–160. Kimura, C., Ohkubo, S., Ogi, K., Hosoya, M., Itoh, Y., Onda,
de Groat, W.C. and Ryall, R.W. (1969) Reflexes to sacral H., Miyata, A., Jiang, L., Dahl, R.R., Stibbs, H.H., Arimura,
parasympathetic neurones concerned with micturition in the A. and Fujino, M. (1990) A novel peptide which stimulates
cat. J. Physiol. (London), 200: 87–108. adenylate cyclase: molecular cloning and characterization
de Groat, W.C. and Steers, W.D. (1990) Autonomic regulation of the ovine and human cDNAs. Biochem. Biophys. Res.
of the urinary bladder and sex organs. In: Loewy A.D. and Commun., 166: 81–89.
Spyer K.M. (Eds.), Central Regulation of Autonomic Func- Klück, P. (1980) The autonomic innervation of the human uri-
tions. Oxford University Press, London, pp. 310–333. nary bladder neck and urethra: a histochemical study. Anat.
Dmitrieva, N., Shelton, D., Rice, A.S.C. and McMahon, S.B. Rec., 198: 439–447.
(1997) The role of nerve growth factor in a model of visceral Krenz, N.R. and Weaver, L.C. (2000) Nerve growth factor in
inflammation. Neuroscience, 78: 449–459. glia and inflammatory cells of the injured rat spinal cord.
Donovan, M.K., Winternitz, S.R. and Wyss, J.M. (1983) An J. Neurochem., 74: 730–739.
analysis of the sensory innervation of the urinary system of Kruse, M.N., Bray, L.A. and de Groat, W.C. (1993) Influence
the rat. Brain Res. Bull., 11: 321–324. of spinal cord injury on the morphology of bladder afferent
Fahrenkrug, J. and Hannibal, J. (1998) Pituitary adenylate and efferent neurons. J. Auton. Nerv. Syst., 54: 215–224.
cyclase activating polypeptide immunoreactivity in capsaicin- Kuru, M. (1965) Nervous control of micturition. Physiol. Rev.,
sensitive nerve fibres supplying the rat urinary tract. Neuro- 45: 425–494.
science, 83: 1261–1272. Kuruvilla, R., Zweifel, L.S., Glebova, N.O., Lonze, B.E., Valdez,
Fernandez, O. (2002) Mechanisms and current treatments of uro- G., Ye, H. and Ginty, D.D. (2004) A neurotrophin signaling
genital dysfunction in multiple sclerosis. J. Neurol., 249: 1–8. cascade coordinates sympathetic neuron development through
114

differential control of TrkA trafficking and retrograde signa- Ruda, M.A., Besse, D., Inagaki, S., DeLeon, M. and Ren, K.
ling. Cell, 118: 243–255. (1994) Nitric oxide expression and regulation in the dorsal
Larsen, J.O., Hannibal, J., Knudsen, S.M. and Fahrenkrug, J. root ganglion and spinal cord. In: Chiueh C.C., Gilbert D.L.
(1997) Expression of pituitary adenylate cyclase-activating and Colton C.A. (Eds.), The Neurobiology of NO and OH.
polypeptide (PACAP) in the mesencephalic trigeminal New York Academy of Sciences, New York, pp. 181–200.
nucleus of the rat after transection of the masseteric nerve. Ruda, M.A., Iadarola, M.J., Cohen, L.V. and Young II, W.S.
Mol. Brain Res., 46: 109–117. (1988) In situ hybridization, histochemistry and
Lecci, A., Giulani, S., Santiciolo, P. and Maggi, C.A. (1994) immunocytochemistry reveal an increase in spinal dynorphin
Involvement of spinal tachykinin NK1 and NK2 receptors in biosynthesis in a rat model of peripheral inflammation and
detrusor hyperreflexia during chemical cystitis in anaesthe- hyperalgesia. Proc. Natl. Acad. Sci., 85: 622–626.
tized rats. Eur. J. Pharm., 259: 129–135. Saito, S., Kidd, G.J., Trapp, B.D., Dawson, T.M., Bredt, D.S.,
Lewin, G.R. and Mendell, L. (1993) Nerve growth factor and Wilson, D.A., Traystman, R.J., Snyder, S.H. and Hanley,
nociception. Trends Neurosci., 16: 353–359. D.F. (1994) Rat spinal cord neurons contain nitric oxide
Lincoln, J. and Burnstock, G. (1993) Autonomic innervation of synthase. Neuroscience, 59: 447–456.
the urinary bladder and urethra. In: Maggi C.A. (Ed.) The Seki, S., Sasaki, K., Igawa, Y., Nishizawa, O., Chancellor,
Autonomic Nervous System, Vol. 3. Harwood Academic M.B., De Groat, W.C. and Yoshimura, N. (2004) Suppres-
Publishers, London, pp. 33–68. sion of detrusor–sphincter dyssynergia by immunoneutrali-
Maggi, C.A. (1991) The role of neuropeptides in the regulation of zation of nerve growth factor in lumbosacral spinal cord in
the micturition reflex: An update. Gen. Pharmacol., 22: 1–24. spinal cord injured rats. J. Urol., 171: 478–482.
Maggi, C.A. (1993) The dual, sensory and efferent function of Sharma, H.S., Westman, J., Olsson, Y. and Alm, P. (1996)
the capsaicin-sensitive primary sensory nerves in the bladder Involvement of nitric oxide in acute spinal cord injury: an
and urethra. In: Maggi C.A. (Ed.) The Autonomic Nervous immunocytochemical study using light and electron
System, Vol. 3. Nervous Control of the Urogenital System. microscopy in the rat. Neurosci. Res., 24: 373–384.
Harwood Academic Publishers, London. Steers, W.D., Ciambotti, J., Etzel, B., Erdman, S. and de Groat,
Maggi, C.A., Santicioli, P., Patacchini, R., Turini, D., Barbanti, W.C. (1991a) Alterations in afferent pathways from the uri-
G., Beneforti, P., Giuliani, S. and Meli, A. (1987) Galanin: a nary bladder of the rat in response to partial urethral
potent modulator of excitatory neurotransmission in the hu- obstruction. J. Comp. Neurol., 310: 1–10.
man urinary bladder. Eur. J. Pharmacol., 143: 135–137. Steers, W.D., Creedon, D.J. and Tuttle, J.B. (1996) Immunity
Mallory, B., Steers, W.D. and de Groat, W.C. (1989) Electro- to nerve growth factor prevents afferent plasticity following
physiological study of micturition reflexes in rats. Am. urinary bladder hypertrophy. J. Urol., 155: 379–385.
J. Physiol., 257: R410–R421. Steers, W.D. and de Groat, W.C. (1988) Effect of bladder outlet
May, V., Beaudet, M.M., Parsons, R.L., Hardwick, J.C., obstruction on micturition reflex pathways in the rat.
Gauthier, E.A., Durda, J.P. and Braas, K.M. (1998) Mech- J. Urol., 140: 864–871.
anisms of pituitary adenylate cyclase activating polypeptide Steers, W.D., Kolbeck, S., Creedon, D. and Tuttle, J.B. (1991b)
(PACAP)-induced depolarization of sympathetic superior Nerve growth factor in the urinary bladder of the adult reg-
cervical ganglion (SCG) neurons. Ann. N. Y. Acad. Sci., 865: ulates neuronal form and function. J. Clin. Invest., 88:
164–175. 1709–1715.
May, V. and Braas, K.M. (1995) Pituitary adenylate cyclase- Sundler, F., Ekblad, E., Hannibal, J., Moller, K., Zhang, Y.Z.,
activating polypeptide (PACAP) regulation of sympathetic Mulder, H., Elsas, T., Grunditz, T., Danielsen, N., Fahrenkrug,
neuron neuropeptide Y and catecholamine expression. J. and Uddman, R. (1996) Pituitary adenylate cyclase-acti-
J. Neurochem., 65: 978–987. vating peptide in sensory and autonomic ganglia: localization
Middleton, J.W. and Keast, J.R. (2004) Artificial autonomic and regulation. Ann. N.Y. Acad. Sci., 805: 410–426.
reflexes: using functional electrical stimulation to mimic Tuttle, J.B. and Steers, W.D. (1992) Nerve growth factor re-
bladder reflexes after injury or disease. Auton. Neurosci., sponsiveness of cultured pelvic ganglion neurons from the
113: 3–15. adult rat. Brain Res., 588: 29–40.
Morrison, J.F.B. (1987) Reflex control of the lower urinary Tuttle, J.B., Steers, W.D., Albo, M. and Nataluk, E. (1994)
tract. In: Torrens M. and Morrison J.F.B. (Eds.), The Phys- Neural input regulates tissue NGF and growth of the adult
iology of the Lower Urinary Tract. Springer, New York. rat urinary bladder. J. Auton. Nerv. Syst., 49: 147–158.
Oppenheim, R.W., Prevette, D., Qin-Wei, Y., Collins, R. and Valtshanoff, J.G., Weinberg, R.J. and Rustioni, A. (1992)
MacDonald, J. (1991) Control of embryonic motoneuron NADPH diaphorase in the spinal cord of rats. J. Comp.
survival in vivo by ciliary neurotrophic factor. Science, 251: Neurol., 321: 209–222.
1616–1618. Verge, V.M.K., Xu, Z., Xu, X.J., Wiesenfeld-Hallin, Z. and
Rice, A.S.C. (1995) Topical spinal administration of a nitric Hökfelt, T. (1992) Marked increase in nitric oxide synthase
oxide synthase inhibitor prevents the hyper-reflexia associat- mRNA in dorsal root ganglia after peripheral axotomy: in
ed with a rat model of persistent visceral pain. Neurosci. situ hybridization and functional studies. Proc. Natl. Acad.
Lett., 187: 111–114. Sci., 89: 11618–11621.
115

Vizzard, M.A. (1997) Increased expression of neuronal nitric Vizzard, M.A., Erickson, V.L., Card, J.P., Roppolo, J.R. and
oxide synthase in bladder afferent and spinal neurons de Groat, W.C. (1995b) Transneuronal labeling of neurons in
following spinal cord injury. Dev. Neurosci., 19: 232–246. the adult rat brainstem and spinal cord after injection of
Vizzard, M.A. (2000a) Changes in urinary bladder neurotro- pseudorabies virus into the urethra. J Comp. Neurol., 355:
phic factor mRNA and NGF protein following urinary blad- 629–640.
der dysfunction. Exp. Neurol., 161: 273–284. Vizzard, M.A., Qiao, L. and Zvara, P. (2003) Neurotrophic
Vizzard, M.A. (2000b) Increased expression of spinal cord Fos mechanisms in bladder overactivity after spinal cord injury.
protein induced by bladder stimulation after spinal cord Auton. Neurosci. Basic Clin., 106: 14.
injury. Am. J. Physiol., 279: R295–R305. Wang, Z.-Y., Alm, P. and Hakanson, R. (1996) PACAP occurs
Vizzard, M.A. (2000c) Increased expression of spinal Fos in sensory nerve fibers and participates in ocular inflamma-
protein in lower urinary tract pathways induced by bladder tion in the rabbit. Ann. N.Y. Acad. Sci., 805: 779–783.
distension following chronic cystitis. Am. J. Physiol., 279: Woolf, C.J., Allchorne, A., Safieh-Garabedian, B. and Poole, S.
R295–R305. (1997) Cytokines, nerve growth factor and inflammatory
Vizzard, M.A. (2000d) Up-regulation of pituitary adenylate hyperalgesia: the contribution of tumour necrosis factor. Br.
cyclase-activating polypeptide in urinary bladder pathways J. Pharm., 121: 417–424.
after chronic cystitis. J. Comp. Neurol., 420: 335–348. Wu, W., Liuzzi, F.J., Schinco, F.P., Depto, A.S., Li, Y., Mong,
Vizzard, M.A. (2001) Alterations in neuropeptide expression in J.A., Dawson, T.M. and Snyder, S.H. (1994) Neuronal nitric
lumbosacral bladder pathways following chronic cystitis. oxide synthase is induced in spinal neurons by traumatic in-
J. Chem. Neuroanat., 21: 125–138. jury. Neuroscience, 61: 719–726.
Vizzard, M.A. and de Groat, W.C. (1996) Increased expression Yoshimura, N. (1999) Bladder afferent pathways and spinal
of neuronal nitric oxide synthase (NOS) in bladder afferent cord injury: possible mechanisms inducing hyperreflexia of
pathways following chronic bladder irritation. J. Comp. the urinary bladder. Prog. Neurobiol., 57: 583–606.
Neurol., 370: 191–202. Zhang, X., Verge, V., Wiesenfeld-Hallin, Z., Ju, G., Bredt,
Vizzard, M.A., Erdman, S.L. and de Groat, W.C. (1993a) The D.S., Snyder, S.H. and Hökfelt, T. (1993) Nitric oxide
effect of rhizotomy on NADPH diaphorase staining in the synthase-like immunoreactivity in lumbar dorsal root ganglia
lumbar spinal cord of the rat. Brain Res., 607: 349–353. and spinal cord of rat and monkey and effect of peripheral
Vizzard, M.A., Erdman, S.L. and de Groat, W.C. (1993b) axotomy. J. Comp. Neurol., 335: 563–575.
Localization of NADPH-diaphorase in bladder afferent and Zhang, Y.Z., Danielsen, N., Sundler, F. and Mulder, H. (1998)
postganglionic efferent neurons of the rat. J. Auton. Nerv. Pituitary adenylate cyclase-activating peptide is upregulated
Syst., 44: 85–90. in sensory neurons by inflammation. Neuroreport, 9:
Vizzard, M.A., Erdman, S.L. and de Groat, W.C. (1993c) 2833–2836.
Localization of NADPH-diaphorase in pelvic afferent and Zhang, Y.-Z., Hannibal, J., Zhao, Q., Moller, K., Danielsen,
efferent pathways of the rat. Neurosci Lett., 152: 72–76. N., Fahrenkrug, J. and Sundler, F. (1996) Pituitary adenylate
Vizzard, M.A., Erdman, S.L. and de Groat, W.C. (1995a) In- cyclase activating peptide expression in the rat dorsal root
creased expression of neuronal nitric oxide synthase (NOS) in ganglia: up-regulation after peripheral nerve injury. Neuro-
visceral neurons after nerve injury. J. Neurosci., 15: 4033–4045. science, 74: 1099–1110.
Vizzard, M.A., Erdman, S.L., Erickson, V.L., Stewart, R.J., Zvara, P., Kliment, J., DeRoss, A.L., Irwin, B.H., Malley, S.E.,
Roppolo, J.R. and de Groat, W.C. (1994a) Localization of Plante, M.K. and Vizzard, M.A. (2002) Differential expres-
NADPH-diaphorase in the lumbosacral spinal cord and sion of bladder neurotrophic factor mRNA in male and fe-
dorsal root ganglia of the cat. J. Comp. Neurol., 339: 62–75. male rats after bladder outflow obstruction. J. Urol., 168:
Vizzard, M.A., Erdman, S.L., Förstermann, U. and de Groat, 2682–2688.
W.C. (1994b) Differential distribution of nitric oxide synt- Zvarova, K., Dunleavy, J.D. and Vizzard, M.A. (2005) Chang-
hase in neural pathways to the urogenital organs (urethra, es in pituitary adenylate cyclase activating polypeptide
penis, urinary bladder) of the rat. Brain Res., 646: 279–291. (PACAP) expression in urinary bladder pathways after spi-
Vizzard, M.A., Erdman, S.L., Roppolo, J.R., Förstermann, U. nal cord injury (SCI). Exp. Neurol., 192(1): 46–59.
and de Groat, W.C. (1994c) Differential localization of neu- Zvarova, K., Murray, E. and Vizzard, M.A. (2004) Changes in
ronal nitric oxide synthase immunoreactivity and NADPH- galanin immunoreactivity in rat lumbosacral spinal cord and
diaphorase activity in the cat spinal cord. Cell Tiss. Res., 278: dorsal root ganglia after spinal cord injury. J. Comp. Ne-
299–309. urol., 475: 590–603.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 8

Effect of injury severity on lower urinary tract


function after experimental spinal cord injury

Jean R. Wrathall and Gregory S. Emch

Department of Neuroscience, Georgetown University Medical Center, TRB EP04, Washington, DC 20057, USA

Abstract: Lower urinary tract dysfunction is a serious burden for patients following spinal cord injury.
Patients are usually limited to treatment with urinary drainage catheters, which can lead to repeated urinary
tract infections and lower quality of life. Most of the information previously obtained regarding lower
urinary tract function after spinal cord injury has been in completely transected animals. After thoracic
transection in the rat, plasticity of local lumbosacral spinal circuitry establishes a ‘‘reflex bladder,’’ which
results in partial recovery of micturition, albeit with reduced voiding efficiency. Since at least half of cord-
injured patients exhibit neurologically incomplete injury, rat models of clinically relevant incomplete con-
tusion injury have been developed. With respect to lower urinary tract function, recent anatomical and
physiological studies have been performed after incomplete thoracic contusion injury. The results show
greater recovery of lower urinary tract function that varies inversely with the severity of the initial trauma
and is positively correlated with time after injury. Recovery, as measured by coordination of the bladder
with the external urethral sphincter, occurs between 1 and 4 weeks after spinal cord injury. It is associated
with normalization of: serotonin immunoreactivity and glutamate receptor subunit mRNA expression in
the dorsolateral nucleus that innervates the external urethral sphincter muscle, the response to glut-
amatergic pharmacological probes administered at the lumbosacral spinal cord level, and c-Fos activation
patterns in the lumbar spinal cord. Understanding the mechanisms involved in this recovery will provide a
basis for enhancing lower urinary tract function in patients after incomplete spinal cord injury.

Introduction often with resistant organisms (Trautner and


Darouiche, 2002; Garcia Leoni and Esclarin De
Spinal cord injury results in lower urinary tract Ruz, 2003). Recently, there have been advances in
dysfunction that contributes to patients’ morbidity the treatment of other types of lower urinary tract
and mortality and profoundly limits their quality disorders, such as stress incontinence (Thor, 2003).
of life. Currently, there are few options to treat These have been based on understanding the ne-
lower urinary tract dysfunction after spinal cord uroanatomical pathways and transmitter systems
injury. Most patients are limited to the use of uri- involved in lower urinary tract function. With re-
nary drainage catheters transiently or permanent- spect to spinal cord injury, detailed studies of the
ly. The altered voiding dynamics, repeated use of changes that occur after complete spinal cord
catheters, and frequent exposure to antibiotic transection have been performed and are reviewed
agents predispose individuals with spinal cord in- elsewhere in this book. However, at least half of all
jury to recurrent episodes of urinary tract infection spinal cord injury patients have incomplete injuries
(Bracken et al., 1990), and much less is known
Corresponding author. Tel.: +202-687-1196; about the alterations in lower urinary tract func-
Fax: +202-687-0617; E-mail: wrathalj@georgetown.edu tion after incomplete spinal cord injury. Recent

DOI: 10.1016/S0079-6123(05)52008-9 117


118

studies from our laboratory, reviewed below, dem-


onstrate that the effect of incomplete spinal cord Pontine micturition
injury on lower urinary tract function depends centers
upon the severity of the injury. Under certain con-
ditions considerable recovery of lower urinary
tract function can occur (Pikov and Wrathall,
2001). Understanding the mechanisms involved in
this recovery may provide a basis for new thera-
peutic approaches to enhance lower urinary tract
function after clinical spinal cord injury. CRF GLU

Bladder
Normal Control of lower urinary tract function and detrusor SPN
effect of spinal cord transection 5-HT GLU

EUS
The bladder and external urethral sphincter com- DL
prise a coordinated visceral system that has been
well studied both in normal and spinal transected Fig. 1. Spinal cord and brainstem control of micturition in the
animals (Tiseo and Yaksh, 1990; de Groat, 1995; rat. Neurons innervating the bladder detrusor reside in the
sacral parasympathetic nucleus (SPN) at spinal segments
Morrison, 1997; de Groat et al., 1998). Urine stor-
L6–S1, while those innervating the external urethral sphincter
age and voiding are the two main functions of the (EUS) muscle are part of the dorsolateral nucleus (DL) in the
lower urinary tract. Urine storage and release from spinal cord at L6/S1. Afferents from the bladder convey pres-
the bladder depend on sympathetic and parasym- sure information to the pontine micturition centers, which
pathetic innervation, respectively. The pre-gang- stimulate the sacral parasympathetic nucleus to cause bladder
contraction and the dorsolateral nucleus to activate, intermit-
lionic sympathetic neurons are located in the rostral
tently, the external urethral sphincter in a coordinated fashion.
segments of the lumbar spinal cord and the pre- Both afferent and efferent pathways are glutamatergic (GLU).
ganglionic parasympathetic neurons in the rostral Serotonin (5-HT) and corticotrophin releasing hormone (CRF)
portion of the sacral spinal cord (de Groat, 1995). are also involved in descending motor control pathways.
The urethral sphincter is responsible for outlet re-
sistance during storage. Micturition in rats is pro- tegmentum of the pons and the periaqueductal
duced by coordinated contraction of the smooth gray are considered to be important in triggering
muscle of the urinary bladder and contraction of the the initiation of voiding (Kruse et al., 1990, 1991).
striated muscle of the external urethral sphincter The external urethral sphincter-controlling motor
intermittently, in 4–6 Hz frequency bursts, which neurons, located in the dorsolateral nucleus of
appears to facilitate voiding (Mersdorf et al., 1993). L6-S1 (Schroder, 1980), receive direct and indirect
If external urethral sphincter activity is prevented by (via spinal interneurons) supraspinal projections
neuromuscular blockade, efficient voiding is abol- mostly from the pontine micturition center
ished (Maggi et al., 1986a; Mersdorf et al., 1993; (Vizzard et al., 1995; Nadelhaft and Vera, 1996;
Kakizaki et al., 1997). As shown in Fig. 1 effective Marson, 1997), the D-region just ventral to
voiding, therefore, requires interaction of spinal au- Barrington’s nucleus (Ding et al., 1995), and the
tonomic reflexes with supraspinal micturition-con- ventrolateral pontine periaqueductal gray (Marson,
trolling centers to coordinate urine expulsion from 1997; Ding et al., 1998; Matsuura et al., 1998).
the bladder with low-frequency intermittent con- Other brainstem nuclei with identified connec-
tractions of the somatically innervated external ure- tions to bladder and external urethral sphincter
thral sphincter muscle (Holstege et al., 1986; pathways are the raphe magnus, raphe pallidus,
Holstege and Tan, 1987; Kakizaki et al., 1997). parapyramidal medullary reticular formation,
Among the brainstem centers involved in the subcoeruleus pars alpha, locus coeruleus, and the
control of micturition, the pontine micturition A5 and A7 nuclei (Vizzard et al., 1995; Marson,
center (Barrington’s nucleus) in the dorsolateral 1997). Of these, cells in the raphe nuclei and
119

nucleus paragigantocellularis in the medullary re- ‘‘complete’’ may have some residual connections
ticular formation produce serotonin (5-hydroxy- with supraspinal control centers. For example,
tryptamine, 5-HT) (Marson, 1997). Thus, 5-HT is with respect to lower urinary tract function, 15%
a marker of direct supraspinal projections to the of people classified as complete can consciously
dorsolateral nucleus (Ramirez-Leon et al., 1994; detect bladder filling and/or electrical stimulation
Tang et al., 1998). Because of the importance of indicating the persistence of afferent connections
these descending control pathways, a lesion of the with the cerebral cortex (Wyndaele, 1991). Because
spinal cord above the lumbar level would be ex- of the importance of incomplete spinal cord injury,
pected to leave spinal micturition reflexes intact rat models of clinically relevant incomplete con-
and compromise lower urinary tract function by tusion injury have been developed and character-
affecting bladder–external urethral sphincter sy- ized (e.g., Wrathall et al., 1985; Gruner, 1992). In
nergy of activation (Chancellor et al., 1990). these models, spinal cord injury is produced by the
Previous studies of complete spinal cord transec- impact of a weight onto the exposed dura after a
tion in the cat (de Groat, 1990), and in the rat laminectomy, usually at a mid-thoracic location.
(Kruse et al., 1993; Mimata et al., 1993), have The impact produces mechanical destruction of
shown that lower urinary tract function varies with tissue and hemorrhage that is maximal in the cen-
time after injury. Initially, during the phase of spinal tral gray matter and the white matter just above it
shock, the bladder is areflexic and urinary retention in the dorsal funiculus (Noble and Wrathall,
occurs (Hassouna et al., 1984). During this stage, 1989). However, a peripheral rim of white matter
the bladder becomes tonically overdistended and is spared, the thickness of which depends on the
noncompliant. Then the spinal reflex activity reap- severity of the impact. A 10 g weight impacting
pears (Tiseo and Yaksh, 1990), in an exaggerated onto the exposed dura at thoracic level T8 will
(spastic) mode with hyperreflexic bladder detrusor spare nearly a complete peripheral rim if dropped
muscle contractions (Osborn et al., 1990). This is from 12.5 or 25 mm, but only residual white matter
believed to be due to a lack of supraspinal inhibition in the most ventrolateral region remains after a
and/or an increase of afferent signaling (Cheng 50 mm impact, as shown in Fig. 2. However, the
et al., 1995) as well as plasticity of afferents (Kruse spared white matter is far from normal, as there is
et al., 1995) resulting from the enlarged bladder. a preferential loss of the larger axons from even
The external urethral sphincter, which in uninjured the most peripheral regions and the myelination
animals is under supraspinal control and works in and glial microenvironment remain abnormal
synergy with the detrusor muscle, becomes contin- chronically for at least 2 months after spinal cord
uously active and therefore dyssynergic with the injury (Wrathall et al., 1998; Rosenberg et al.,
emerging automatic bladder contraction reflex 2005). Thus, there is complete loss of long de-
(Schalow et al., 1995). Thus, although spinal cir- scending and ascending axons in some regions of
cuits alone are capable of establishing automatic the spinal cord at the injury epicenter and partial
bladder control after transection (de Groat et al., loss in other regions of white matter.
1998), detrusor–external urethral sphincter coordi- There is now considerable data documenting re-
nation that is mediated via a spino-bulbo-spinal re- covery of hind limb sensory and motor function
flex (Holstege et al., 1986; de Groat, 1990) does not after experimental contusion of the thoracic spinal
recover after complete spinal cord transection. cord (Wrathall, 1994). The extent of recovery is
inversely related to the severity of the initial trauma
Incomplete spinal cord injury and lower urinary and, for the most part, positively correlated with
tract function time after injury. The recovery phase following in-
itial hind limb areflexia and complete paralysis is
Large-scale clinical trials demonstrate that at least characterized by the return of segmental reflexes in
half of cord-injured people have neurologically a modified state and increasingly effective use of the
incomplete injury (Bracken et al., 1990). Further, hind limbs in coordinated movements that are
a significant proportion of those classified as known to mediate postural control, swimming
120

Fig. 2. White matter sparing at the spinal cord injury epicenter. Top, bottom left: Photomicrographs of representative sections through
the lesion epicenter from one rat of each of the three T8 spinal cord injury (SCI) groups and from an uninjured control stained with
eriochrome cyanine to label myelin. The dorsal, lateral, and ventral funicular white matter of the normal spinal cord is heavily stained,
whereas little myelin staining is seen in the gray matter. The cross-sectional profiles of the injured spinal cords are reduced in diameter.
The center of the injured cords contains cavities and an abnormal loose network of cells, but no myelin staining is apparent.
A peripheral rim of residual white matter is seen. Myelin staining is present but reduced compared with normal white matter, consistent
with the chronic hypo-myelination of residual axons. Bottom right: The average areas of myelinated white matter from the ventral and
lateral funicular zones at the lesion epicenter in the injury groups. SCI height indicates height from which weight is dropped, i.e.,
severity of injury. * Indicates a significant difference from the 12.5 mm weight drop group, based on p.o.0.05 in Tukey’s post hoc test
after ANOVA. Scale bar, 250 mm. Taken from Pikov and Wrathall (2001) with permission.

movements and elements of locomotion. This re- The difference in function between these spinal
covery phase plateaus at 3–4 weeks in the adult rat cord-injured animals at a few days after injury and
with the standard tests revealing no significant ad- 4 weeks later is remarkable — from almost com-
ditional recovery between 4 and 8 weeks (Noble plete paralysis to quite effective, albeit still abnor-
and Wrathall, 1989) or even by 6 months (J.R. mal, use of their hind limbs. Thus, considerable
Wrathall, unpublished). With respect to open field natural recovery of hind limb sensory-motor func-
locomotion after an incomplete contusion, rats tion occurs in rats that retain only 10–20% of spi-
show a stereotypical pattern of recovery of loco- nal cord tissue at the injury epicenter.
motion consisting of early, intermediate, and late These spinal cord-injured rats also demonstrate
stages (Basso et al., 1995). During the first stage abnormalities of lower urinary tract function and
there is increasing joint movement in the hind are initially unable to urinate. A ‘‘reflex bladder’’
limbs. In the second, rats become capable of plan- develops with time as seen after spinal cord tran-
tar stepping and bearing weight on their hind limbs. section. However, depending on the severity of the
In the last phase of recovery, exhibited by the least lesion, coordinated function of the bladder and the
severely injured rats, there is consistent coordinated external urethral sphincter may also recover after
weight-bearing locomotion with increasingly nor- incomplete spinal cord injury (Pikov et al., 1998;
mal positioning of the paws, the trunk, and the tail. Pikov, 2000; Pikov and Wrathall, 2001, 2002).
121

Evaluating recovery of lower urinary tract function


after experimental contusion spinal cord injury

Our initial interest in recovery of lower urinary


tract function stemmed from the observation that
the length of time required for manual expression
of the bladder after contusion injury appeared to
be reduced with acute treatments that enhanced
white matter sparing and recovery of hind limb
function. Groups of rats treated with the gluta-
mate receptor antagonist NBQX demonstrated a
dose-related sparing of white matter at the injury
epicenter and a dose-related decrease in the
number of days required for them to acquire a
reflex bladder (Wrathall et al., 1994). Similarly,
when a sodium channel blocker was used to reduce
axonal loss, the number of days required to attain Fig. 3. Time course of recovery of spontaneous voiding. The
a reflex bladder after spinal cord injury was re- bars on the curve show the urine volumes (71 standard error of
duced (Teng and Wrathall, 1997). the mean) manually expressed every 12 h from the urinary
bladder, plotted against time after spinal cord injury (SCI). The
Evaluating the volume of urine expressed over
volume collected increased between days 0 and 4, presumably
time after three different severities of thoracic con- due to an increase in urine production during the immediate
tusion injury revealed an initial increase in expressed post-operative period. The expressed volume then decreased
volume followed by a decrease, as the spinal bladder after day 4, presumably due to recovery of spontaneous void-
reflex was established (Pikov and Wrathall, 2001). ing. After day 4, progressively less urine was expressed from the
bladders of the rats with the (milder) 12.5 mm weight drop cord
However, more severe spinal cord injury, associated
injury, indicating a faster recovery of spontaneous voiding in
with greater loss of white matter at the injury site, as this group than in the other two more severely injured groups.
shown in Fig. 2, was reflected in a slower establish- Taken from Pikov and Wrathall (2001) with permission.
ment of spontaneous reflex voiding (Fig. 3).
To study recovery of lower urinary tract func- In initial studies average values of external ure-
tion further, we used a urodynamic procedure that thral sphincter spiking activity during bladder fill-
allows a rapid collection of data over a large ing and voiding were calculated, and threshold (at
number of voiding cycles (Maggi et al., 1986b). As the initiation of contraction) and maximal in-
shown in Fig. 4, bladder intravesical pressure was travesical pressures during voiding were measured
recorded with a transurethral bladder catheter for each voiding cycle over a 20-min period in each
(polyethylene-50) during continuous perfusion of the animals (Pikov et al., 1998). The most useful
with warm saline (0.22 ml/min). During the blad- measure was found to be the change in external
der detrusor contractions, fluid was released by urethral sphincter spiking activity calculated from
flowing around the catheter in the urethra. The the raw electromyography data. This was obtained
signal from the pressure transducer was amplified, by counting the number of peaks above the base-
sampled at 1 kHz and acquired on-line using Bio- line at 100 ms intervals with a custom-written peak
Bench 1.0 software (National Instruments, Austin, detection macro in Microsoft Excel (Pikov and
TX). For electromyography, two fine (50 mm) Wrathall, 2001). The change in external urethral
epoxy-coated platinum–iridium wire electrodes sphincter spiking activity was measured during
were placed percutaneously in the sphincter area bladder filling and emptying as illustrated in
of the urethra to record external urethral sphincter Fig. 5. Although the catheter and electrodes were
electrical activity. The electromyographic activity inserted under anesthesia, the rats were then al-
was pre-amplified, sampled at 1 kHz, and acquired lowed to recover so that the urodynamic record-
on-line simultaneously with intravesical pressure. ings were done on awake restrained animals
122

Fig. 4. Experimental design for urodynamic recordings. A transurethral bladder catheter (polyethylene-50) is implanted in an anest-
hetized rat. After recovery from anesthesia, urodynamic recordings are performed under light anesthesia. The catheter is connected to
an infusion pump and a pressure transducer. Bladder intravesical pressure is recorded during continuous perfusion with room
temperature saline (CMG, cystometrogram). During bladder contractions, fluid is released by flowing around the catheter in the
urethra. For electromyography (EMG), two fine platinum wire electrodes are placed percutaneously in the sphincter area of the urethra
to record external urethral sphincter (EUS) electrical activity. The EMG activity is amplified, sampled at 1 kHz, and acquired on-line
simultaneously with intravesical pressure. Taken from Pikov et al. (1998) with permission.

because anesthesia markedly reduces the efficiency In order to investigate anatomical evidence of
of voiding (Yoshiyama et al., 1994, 1999). supraspinal involvement in lower urinary tract func-
Comparing urodynamic measures of normal rats tion, pseudorabies virus was injected into the blad-
to those after thoracic contusion or complete tran- der wall in normal and spinal cord-injured animals.
section injury, we found that rats after both tran- The rats were allowed to survive long enough for
section and contusion spinal cord injury showed transneuronal tracing to the brainstem. As shown in
evidence of reflexive bladder contractions in week 2 Fig. 6, labeling was present in the pontine micturit-
after injury, but only the contused groups demon- ion center and in the periaqueductal gray. As might
strated some recovery of coincidental activation of be expected, there was much lower labeling in the
the external urethral sphincter (Pikov et al., 1998). contused cord-injured animal as compared to unin-
Bladder weight was measured, showing a six-fold jured control, illustrating an anatomical basis for
increase at week 1, an eight-fold increase at week 2 the reduced supraspinal control of lower urinary
after incomplete contusion, and an 11.6-fold in- tract function in rats after injury.
crease in cord-transected animals. Voided volume,
or the amount that is released from the bladder in
each contraction, was found to decrease in both Effect of injury severity on chronic lower urinary
contused and transected animals. Voiding efficiency tract function after incomplete spinal cord injury
(volume voided/capacity  100), due to increase in
bladder capacity and decrease in voided volume was More information was obtained from an injury
very low (2.8–3.5%) in both groups of cord-injured dose–response study in which groups of rats were
animals as compared to uninjured controls (58%). subjected to spinal cord injury with the widely
The inter-contraction interval (time between de- used Multicenter Animal Spinal Cord Injury Study
trusor contractions) was also lower in the spinal cord (MASCIS) injury device (Gruner, 1992) and pro-
injury groups (40–70 vs. 115 s in control animals). duced by the impact of a 10 g weight dropped from
Fig. 5. Urodynamic analysis of detrusor–external urethral sphincter coordination in representative uninjured animals (A, D, G and J), and animals with 12.5 mm weight
drop (B, E, H and K) and 50 mm weight drop (C, F, I and L) spinal cord injury (SCI) at 8 weeks. (A–C) Bladder intravesical pressure (IVP) recordings during one voiding
cycle. Solid horizontal line in A and B indicates the duration of stream-like voiding, and the dashed line in C indicates the drop-by-drop voiding. (D–F) external urethral
sphincter EMG recordings, showing activation of external urethral sphincter EMG in relation to the voiding cycle in uninjured and 12.5 mm SCI animals but not in the
50 mm SCI animal. (G–I) Power spectrum analysis of external urethral sphincter EMG activity as a function of time. A broad band of frequencies (5–40 Hz) shows an
increased power during the voiding phase in uninjured and 12.5 mm cord-injured animals but not in the 50 mm cord-injured animal. (J–L) Peak detection analysis of the
external urethral sphincter spiking activity (ESA). Peaks were detected in 1 ds (ds ¼ 101 s) intervals. An increase in spiking activity occurred at the time corresponding to
the voiding phase in uninjured and 12.5 mm cord-injured animals, but there was no change in the level of spiking activity in the 50 mm cord-injured animal. Taken from
Pikov and Wrathall (2001) with permission.

123
124

Fig. 6. Pseudorabies virus tracing from the bladder. (A and B) Micrographs of Pseudorabies virus-labeled neurons in the pontine
micturition center of a normal rat (A) and a rat on day 12 after a 10 g  25 mm weight drop contusion (B). The large neurons of the
mesencephalic nucleus of the fifth cranial nerve on the left of each field demonstrate some nonspecific staining. Bar ¼ 100 mm. (C–F)
Schematized images of Pseudorabies virus retrograde transneuronal labeling from the bladder. Dorsolateral tegmentum (C and D) and
periaqueductal gray (E and F) in uninjured (C and E) and SCI animals (D and F). Abbreviations: Mes5-mesencephalic trigeminal
nucleus, PAG-periaqueductal gray, SCP-superior cerebellar peduncle. Taken from Pikov et al. (1998) with permission.

a height of 12.5, 25 or 50 mm onto the dura after a bladder pressure during urodynamic evaluation. In
laminectomy at T8 (Pikov and Wrathall, 2001). contrast the 12.5 mm group, although showing
Table 1 shows a comparison of bladder weights, similar tendencies, was not significantly different
volumes and pressure (intravesical pressure) at 8 from controls in these parameters of lower urinary
weeks after injury. Compared to uninjured con- tract function.
trols, the 25 and 50 mm groups demonstrated sig- As shown in Fig. 7A, the 12.5 mm spinal cord-
nificantly greater bladder weight and volume injured group recovered bladder–external urethral
chronically. The most severely injured (50 mm) sphincter coordination (as measured by the change
group also had significantly decreased maximal in external urethral sphincter spiking activity) by 8
125

Table 1. Changes in lower urinary tract parameters 8 weeks after spinal cord injury

Parameter Spinal cord injury severity (weight drop in mm)

0 (n ¼ 6) 12.5 (n ¼ 7) 25 (n ¼ 4) 50 (n ¼ 7)

Bladder weight (g) 0.1170.01 0.2370.09 0.37*70.13 0.41*70.18


Bladder volume (ml) 3.871.5 9.176.0 14.9*76.2 26.1**78.1
Intravesical pressure amplitude (mmHg) 25.477.7 19.477.6 14.173.0 13.0*75.1

The number of animals in each group is shown within parentheses. Urodynamic evaluation was performed at 8 weeks after SCI or laminectomy. The
bladder was then weighed after blot-drying, and its length and width were measured to calculate volume. The amplitude of the bladder pressure was
calculated as the difference between the maximal intravesicular pressure during voiding and the pressure just before voiding was initiated. Mean
values7standard error are presented for each measurement. *po0.05, indicates significantly different from values in control rats (0 weight drop). **,
significantly different from both the control and 12.5 group. Data from Pikov and Wrathall (2001).

Fig. 7. Recovery of detrusor–external urethral sphincter coordination at 8 weeks after spinal cord injury (SCI) depends upon injury
severity (A), and is correlated to white matter sparing at the epicenter (B), to the amount of serotonin immunoreactivity (5HT-IR) in
the dorsolateral nucleus (C), and to CRF immunoreactivity (CRF-IR) in the sacral parasympathetic nucleus (SPN; D). In panel A, SCI
height indicates distance of weight drop, i.e., severity of spinal cord injury. Bars represent means and standard errors. Vertical axis is
change in external urethral sphincter spiking activity (sESA) during voiding. N ¼ 6, 7, 5 and 7 for the 0, 12.5, 25, and 50 mm groups,
respectively (ds ¼ 101 s). Correlation coefficients are shown at the top left corner of B–D. In A, symbols indicate a significant
difference from the uninjured group (*) or from both the uninjured and 12.5 mm groups (**). (po0.001). Taken from Pikov and
Wrathall (2001) with permission. Data from Pikov and Wrathall (2001).

weeks to an extent statistically indistinguishable well as chronic white matter sparing at the injury
from uninjured controls, whereas the 25 and 50 mm epicenter (Fig. 7B). Correlation with spared de-
spinal cord-injured groups did not. Furthermore, scending control pathways from the brainstem was
at 8 weeks there was a significant correlation be- indicated by quantification of the relative immuno-
tween the degree of recovery of lower urinary tract reactivity for 5-HT associated with the dorsolateral
function in terms of bladder–external urethral nucleus motor neurons that innervate the external
sphincter coordination and the initial impact as urethral sphincter (Fig. 7C), and corticotrophin
126

Table 2. Expression of NMDA (NR1, NR2A, NR2B) and AMPA (GluR1, GluR2, GluR3, GluR4) subunit mRNA in dorsolateral
nucleus motoneurons at 8 weeks after spinal cord injury

Subunit Spinal cord injury severity (weight drop in mm)

0 ðn ¼ 6Þ 12.5 ðn ¼ 7Þ 25 ðn ¼ 3Þ 50 ðn ¼ 5Þ

NR1 39.478.7 43.776.0 43.176.4 45.175.3


NR2A 6.871.5 9.972.7 12.4*74.1 12.3*72.9
NR2B 1.670.4 2.370.5 2.270.5 2.270.2
GluR1 1.370.2 1.670.5 1.670.4 2.070.6
GluR2 4.671.1 6.372.2 10.5*71.6 8.2*72.6
GluR3 3.272.1 5.871.3 4.971.6 5.972.6
GluR4 2.470.6 2.970.6 2.070.3 2.370.2

The data are presented as the number of grains per square micrometer of cell area (mean 7 standard error). Significant difference from the uninjured
group is indicated by asterisk (*) and bold font and is based on po0.05 in Tukey’s post hoc test after ANOVA.

releasing factor immunoreactivity associated with c-Fos, which is expressed in neurons after intense
the sacral parasympathetic nucleus that innervates or prolonged activation (Rinaman et al., 1993), in
the bladder detrusor muscle (Fig. 7D). response to bladder filling in normal and spinal
Glutamate receptors are utilized in spinal circuits cord-injured rats at 8 weeks after injury (Emch
controlling the detrusor and external urethral et al., 2003). Rats that had not recovered normal
sphincter (Matsumoto et al., 1995a, b; Iwabuchi, levels of external urethral sphincter spiking activity
1997), and thus changes in properties of these re- (25 mm injury group) demonstrated extensive
ceptors may be involved in altered lower urinary c-Fos activation (Fig. 8), as previously reported
tract function after spinal cord injury. Comparing for rats after complete transection (Vizzard, 2000).
groups of rats that did or did not recover blad- In contrast, the mild (12.5 mm) group that recov-
der–external urethral sphincter coordination at 8 ered bladder–external urethral sphincter coordina-
weeks after spinal cord injury showed significant tion had a c-Fos activation pattern at 8 weeks that
differences in the expression of mRNAs for gluta- was similar to uninjured controls. Thus, aspects of
mate receptors in the dorsolateral nucleus motoneu- recovered lower urinary tract function after this
rons that innervate the external urethral sphincter severity of spinal cord injury include normal blad-
(Table 2). The 25 and 50 mm groups that did not der–external urethral sphincter coordination, nor-
recover normal external urethral sphincter spiking mal expression of glutamate receptor mRNAs in
activity also exhibited abnormally high expression of dorsolateral nucleus neurons and a normal local
NR2A and GluR2, as determined by in situ hybrid- segmental response to bladder stimulation as in-
ization autoradiography. Assuming these changes in dicated by c-Fos expression.
chronic mRNA levels produce altered functional
glutamate receptors, these alterations may be related
to the aberrant hyperactivity of these motoneurons What occurs during recovery of bladder–external
as in the spontaneous spastic activity seen chroni- urethral sphincter coordination after mild
cally in lumbosacral somatic motoneurons after contusion injury?
spinal cord injury (Hiersemenzel et al., 2000; Little
et al., 2000). In contrast the 12.5 mm group that did In order to study the mechanisms that may be in-
recover normal external urethral sphincter spiking volved in this recovery of lower urinary tract func-
activity expressed normal levels of the receptor sub- tion we compared animals after mild spinal cord
unit mRNAs in the dorsolateral nucleus motoneu- injury at 5 days after injury (non-recovered; sub-
rons that innervate the external urethral sphincter. acute), when the bladder reflex can be detected
Recently, we investigated, by immunohistoche- without any coordinated activation of the external
mical identification, the proto-oncogene product urethral sphincter, to animals at 8 weeks (recovered;
127

A B

L6 spinal cord
55 +
50
Fos count/10 um section

45 +
40
35
30
25 +
*
20
15
10
5
0
C SPN DCM MDH LDH 12.5mm 8wk 25mm 8wk

Fig. 8. After spinal cord injury c-Fos immunoreactivity in the L6 spinal cord segment is altered in animals with abnormal lower
urinary tract function and returns to normal when animals recover detrusor–external urethral sphincter coordination. The bladders of
lightly anesthetized rats were catheterized and room temperature saline was perfused continuously for 2 h to stimulate the voiding
reflex. Spinal cords were sectioned and c-Fos immunohistochemistry was performed with methods adapted from Emch et al. (2001). In
animals with abnormal detrusor–external urethral sphincter coordination as measured in our EMG preparation, the pattern of c-Fos
expression was altered in 4 areas of the L6 spinal cord: the sacral parasympathetic nucleus (SPN), the dorsal gray commissure (DCM),
the medial dorsal horn (MDH), and the lateral dorsal horn (LDH). Increases in c-Fos expression normalized by 8 weeks after injury in
animals that recovered lower urinary tract function, i.e. the mild injury group (10 g  12.5 mm weight drop). The pattern of c-Fos
expression in animals that did not recover lower urinary tract function, i.e. the moderate injury group (10 g  25 mm weight drop), did
not normalize. (A and B) Sections of the L6 DCM tissue stained for c-Fos-IR in a control rat (A) and in a rat 8 weeks after the injury
produced by a 10 g weight dropped 25 mm (B). Note the large number of immunoreactive cells in B. (C) Histograms of c-Fos counts in
the four L6 areas listed above. Open bars indicate controls. Gray and black bars show counts in the same regions in rats injured with
the 12.5 and 50 mm weight drop, respectively. +, significantly different from both control and the 12.5 mm injury group. *Significantly
different from control only.

chronic), when the external urethral sphincter about 30% at the earlier time point (subacute) with
spiking activity is indistinguishable from uninjured some recovery by 8 weeks (chronic), when the
controls (Fig. 9). Bladder weight at both subacute change in intravesical pressure was not significantly
and chronic time points was higher than in unin- different from that in uninjured animals. The in-
jured animals, and there was no difference in blad- crease and decrease in intravesical pressure during
der weight between these two time points. The detrusor contraction and relaxation, respectively,
change in bladder pressure during contraction occurred more slowly in injured than in uninjured
(change in intravesical pressure) was decreased by animals. The external urethral sphincter spiking
128

To test the hypothesis that altered glutamate


receptor function is involved with altered lower
urinary tract function after spinal cord injury, the
intravesical pressure and external urethral sphinc-
ter spiking activity were measured during urody-
namic evaluation in the presence of the NMDA
receptor antagonist 3(2-carboxypiperazin-4-yl)-
propyl-1-phosphonic acid (CPP) and the (R,S)-2-
amino-3-(3-hydroxy-5-methyl-4-isoxazolyl)propionic
acid (AMPA) receptor antagonist 2,3-Dihydro-
xy-6-nitro-7-sulphamoylbenzo(f)-quinoxaline 6-
Nitro-7-sulphamoylbenzo(f)-quinoxaline-2,3-dione
(NBQX) (Pikov and Wrathall, 2002). The drugs
were given intrathecally to the lumbosacral spinal
cord with increasing dosages administered during
the urodynamic procedures. The range of doses
was chosen so as not to affect the detrusor itself.
We found that external urethral sphincter spik-
ing activity was mildly affected by low doses of
either drug and was dramatically inhibited by high
doses. The mean values of intravesical pressure
change were unaffected by either drug except for
Fig. 9. Time course of recovery of detrusor and external ure-
high doses of CPP in the uninjured and chronic
thral sphincter muscle function after mild spinal cord injury.
The increase in intravesical pressure (dIVP, top panel) and in groups. In contrast, the external urethral sphincter
external urethral sphincter spiking activity (dESA, bottom pan- spiking activity was decreased with each drug in a
el) during voiding was measured in uninjured rats and in rats at dose-dependent manner, with maximal inhibition
5 days (subacute) and 8 weeks (chronic) after spinal cord injury of external urethral sphincter spiking activity
produced by dropping at 10 g weight 12.5 mm unto the dura at
(60–70%) seen at the highest doses used. The in-
T8 (ds ¼ 101 s). Symbols indicate a significant difference from
the uninjured group (*) or from both the uninjured and chronic hibitory dose (ID)50 values for individual animals
groups (**). Adapted from Pikov and Wrathall (2002) with were used to calculate the average ID50 values
permission. for NBQX and CPP. There was no effect of
injury on the ID50 of NBQX between experimental
activity was significantly inhibited at the subacute groups. In contrast, the subacute group exhibited
time point (5 days) and recovered by 8 weeks after a 50% lower ID50 for CPP than uninjured
this mild spinal cord injury (Fig. 9). controls. By 8 weeks, this effect was no longer
In examining the dorsolateral nucleus motoneu- evident.
rons we found that 5-HT immunoreactivity at 5 Our finding of changes only in NMDA receptor
days is significantly below that of uninjured con- function is consistent with evidence suggesting that
trols, and, by 8 weeks, it has recovered to normal NMDA and non-NMDA receptors are part of
levels. This suggests that sprouting of raphespinal parallel, but functionally separate, synaptic circuits
fibers spared by the injury may be involved in the that are important in micturition (Yoshiyama
recovery of external urethral sphincter spiking ac- et al., 1995). An intriguing hypothesis to explain
tivity shown in Fig. 9. In contrast, corticotrophin our results is an alteration in NMDA receptor sub-
releasing factor associated with the sacral para- unit composition during recovery of lower urinary
sympathetic nucleus was about 60% of the normal tract function. CPP, a potent NMDA receptor an-
at both 5 days and 8 weeks, consistent with the tagonist, has different affinities to NMDA recep-
ability of a reflex bladder to be established in the tors depending upon the NR2 subunit present as
absence of supraspinal connections. part of the receptor complex, ranking in the order
129

NR2A4NR2B4NR2D while the affinity to may serve as the basis for novel pharmacological
glutamate is in the opposite order (Monaghan strategies to enhance functional recovery in the
et al., 1998). Thus, increased sensitivity to CPP subacute period after spinal cord injury.
could be due to a shift toward a higher proportion Subsequent studies have confirmed that at 5 days
of NR2A at 5 days after spinal cord injury. If such after a mild spinal cord injury, NMDA receptors
receptor composition shifts are confirmed and are on dorsolateral nucleus motoneurons may contain
typical after spinal cord injury, this information a higher proportion of NR2A than after recovery

Fig. 10. (Upper panel) Photomicrograph depicting Neutral Red-stained dorsolateral (DL) nucleus motoneuronal cell bodies with
overlying NR2A mRNA grains. Uninjured animals exhibit normal levels of mRNA, whereas 5 days after a MASCIS mild contusion
injury (12.5 mm weight drop), the number of grains are increased. Scale bar ¼ 5 mm. (Lower panel) Quantitative comparison of in situ
hybridization net grain counts for the NMDA subunits NR1, NR2A, NR2B, and the AMPA subunit GluR2 in the DL nucleus. Data
are expressed as net grain counts/mm2 cell body. NR2A and GluR2 net grain counts from injured animals are significantly higher than
uninjured controls, po0.01, whereas those for NR1 and NR2B do not differ significantly between injured and uninjured animals.
Symbols represent data from individual rats; the bar represents the mean value for the group.
130

at 8 weeks. Slides containing the dorsolateral nu- 22.5


cleus from animals at 5 days after mild MASCIS 20.0
contusion injury and from uninjured control ani- 17.5
mals were hybridized with 35S-ATP labeled anti- 15.0
sense oligonucleotides to the NMDA subunit

dESA
12.5
mRNA for NR1, NR2A, and NR2B and the
AMPA subunit mRNA for GluR2. The density of
10.0 ∗
7.5
the grains over the dorsolateral nucleus neurons 5.0 ∗ ∗
was then calculated as a ratio of grains/mm2 and 2.5 ∗
was corrected for background by subtracting the
0.0
density of grains from adjacent areas devoid of tis-

k
s

k
8w
ol

1w

1w

4w

4w

8w
sue. The results (Fig. 10) indicate that the mRNAs

tr
on

m
m

m
m

m
m
for GluR2 and NR2A are abnormally expressed

.5

25

.5

25

.5

25
12

12

12
(upregulated) at 5 days as found chronically in
more severely injured groups as shown in Table 2. Fig. 11. Recovery of detrusor–external urethral sphincter co-
GluR2 is generally found as part of functional ordination occurs by 4 weeks after mild spinal cord injury. At 1
AMPA receptors but the presence of NR2A in week after cord injury both mild (12.5 mm) and moderate
NMDA receptors is associated with altered recep- (25 mm) injury groups exhibit changes in external urethral
sphincter spiking activity (dESA) during bladder contraction
tor sensitivity as mentioned above. Our current
that are significantly decreased from uninjured controls. By 4
data suggest that altered sensitivity of dorsolateral weeks, animals in the mild group exhibit dESA that have re-
nucleus motoneurons may be a general occurrence covered to control levels. *po0.001, ANOVA with Tukey’s
in the subacute period after spinal cord injury but is post hoc testing.
normalized if and when recovery of bladder–exter-
nal urethral sphincter coordination occurs.
Recently, we have undertaken studies to further to the lumbar spinal cord (Lecci et al., 1992).
define the time of recovery of lower urinary tract Administration of 5-HT antagonists, such as
function after mild spinal cord injury as measured WAY-100635 increases bladder capacity and in-
by the normalization of external urethral sphincter hibits the voiding reflex (Kakizaki et al., 1997;
spiking activity. We have found that complete re- Testa et al., 1999). In fact, modulation of the se-
covery is consistently found by 4 weeks after spinal rotonergic influence on micturition is under clin-
cord injury, as shown in Fig. 11. Further, this func- ical investigation for application in stress-induced
tional recovery is mirrored by recovery of 5-HT urinary incontinence. The drug duloxetine, an in-
immunoreactivity of the dorsolateral nucleus moto- hibitor of both serotonin and norepinephrine re-
neurons (Fig. 12). Are these correlations indications uptake appears especially promising for reducing
of a functional connection? Future studies will fo- stress-induced urinary incontinence by facilitating
cus on this question, further testing the temporal external urethral sphincter activity (Thor, 2003).
relationship between recovery of external urethral We speculate that pharmacological support of
sphincter spiking activity and 5-HT immunoreac- serotonergic neurotransmission after incomplete
tivity and extending the studies to pharmacological spinal cord injury may modulate the extent of ex-
evaluation of the functional role of the serotonin ternal urethral sphincter activation and thus nor-
system in recovery of lower urinary tract function. malize the bladder–sphincter dyssynergia for
The serotonin system has been shown to alter efficient voiding.
the micturition reflex in both normal rats and
those after a complete spinal cord transection.
Administration of 8-OH-DPAT (a selective Conclusion
5-HT1A agonist) facilitates the voiding reflex re-
gardless of whether drug administration is intra- On the basis of our studies, we postulate that spinal
peritoneal, intracerebroventricular or intrathecal cord injury initially affects the lower urinary tract
131

Fig. 12. 5-HT immunoreactivity in the dorsolateral (DL) nucleus recovers by 4 weeks after mild SCI. 5-HT pixel density was quantified
in the DL nucleus using METAMORPHs software in controls (A), at 1 week (B), and at 4 weeks (C) after mild and moderate spinal
cord injury. # indicates DL motor neurons in A–C. (D) 5-HT pixel density is significantly decreased in both injury groups at 1 week
post-injury (*po0.001), but normalizes by 4 weeks in the mild group. Bars represent mean+SEM of 5-HT pixel density with N ¼ 6 for
controls and 5 for each injury group. ANOVA with Tukey’s post hoc testing.

function similarly after a wide range of severities of interneuronal alterations that results in the devel-
thoracic spinal cord injury ranging from a mild opment of a reflex bladder (Kruse et al., 1993;
contusion through a complete surgical transection. de Groat, 1995), as described elsewhere in this
The loss of normal descending control inhibits volume. We postulate that the same mechanisms
normal micturition requiring manual bladder occur after incomplete contusion injury in the first
expression in the rat or catheterization for a week(s) after spinal cord injury. However, the
cord-injured person. The bladder responds by sparing of key long tract axons then allows a sec-
enlarging, and in transection models this is known ond form of recovery to occur, which is only seen
to initiate the afferent plasticity and subsequent with incomplete spinal cord injury. Changes occur
132

in some spared descending control pathways for de Groat, W.C., Araki, I., Vizzard, M.A., Yoshiyama, M.,
lower urinary tract function, such as the raphespi- Yoshimura, N., Sugaya, K., Tai, C. and Roppolo, J.R. (1998)
nal system, that allow simultaneous activation of Developmental and injury induced plasticity in the micturit-
ion reflex pathway. Behav. Brain Res., 92: 127–140.
the external urethral sphincter with bladder con- Ding, Y.Q., Takada, M., Tokuno, H. and Mizuno, N. (1995)
traction. With time this coordination improves Direct projections from the dorsolateral pontine tegmentum
and some of the initial changes associated with loss to pudendal motoneurons innervating the external urethral
of lower urinary tract function normalize, such as sphincter muscle in the rat. J. Comp. Neurol., 357: 318–330.
altered glutamate receptor subunit expression on Ding, Y.Q., Wang, D., Nie, H., Guan, Z.L., Lu, B.Z. and Li,
J.S. (1998) Direct projections from the periaqueductal gray to
dorsolateral nucleus motoneurons, 5-HT immuno- pontine micturition center neurons projecting to the lumbo-
reactivity in the dorsolateral nucleus, and perhaps sacral cord segments: an electron microscopic study in the
increased c-Fos activation patterns. rat. Neurosci. Lett., 242: 97–100.
A challenge for future studies will be identifying Emch, G., Lund, IV., Gandy, V., Lytle, J.M. and Wrathall, J.R.
the key causal factors in this secondary recovery (2003) Glutamate receptor plasticity during recovery of lower
urinary tract function after spinal cord injury. Soc. Neurosci.
phase that produces bladder–external urethral
Abs., 954: 914.
sphincter coordination. With this information, Emch, G.S., Hermann, G.E. and Rogers, R.C. (2001) TNF-
we may learn how to increase the efficiency of alpha-induced c-Fos generation in the nucleus of the solitary
voiding and thus reduce the long-term deleterious tract is blocked by NBQX and MK-801. Am. J. Physiol.
effects of abnormal lower urinary tract function Regul. Integr. Comp. Physiol., 281: R1394–R1400.
Garcia, Leoni, M.E. and Esclarin De Ruz, A. (2003) Manage-
after incomplete spinal cord injury.
ment of urinary tract infection in patients with spinal cord
injuries. Clin. Microbiol. Infect., 9: 780–785.
Gruner, J.A. (1992) A monitored contusion model of spinal
Acknowledgments cord injury in the rat. J. Neurotrauma, 9: 123–126 discussion
126–128.
Hassouna, M., Galeano, C., Abdel-Rahman, M. and Elhilali,
Studies reviewed in this chapter were supported by M. (1984) Vesicourethral motility following acute spinal cord
NIH PO1 NS28130, RO1 NS 35647, and RO1 transection in the cat. J. Urol., 131: 370–373.
NS37733. Hiersemenzel, L.P., Curt, A. and Dietz, V. (2000) From spinal
shock to spasticity: neuronal adaptations to a spinal cord
injury. Neurology, 54: 1574–1582.
Holstege, G., Griffiths, D., de Wall, H. and Dalm, E. (1986)
References Anatomical and physiological observations on supraspinal
control of bladder and urethral sphincter muscles in the cat.
Basso, D.M., Beattie, M.S. and Bresnahan, J.C. (1995) A sen- J. Comp. Neurol., 250: 449–461.
sitive and reliable locomotor rating scale for open field testing Holstege, G. and Tan, J. (1987) Supraspinal control of moto-
in rats. J. Neurotrauma, 12: 1–21. neurons innervating the striated muscles of the pelvic floor
Bracken, M.B., Shepard, M.J., Collins, W.F., Holford, T.R., including urethral and anal sphincters in the cat. Brain,
Young, W., Baskin, D.S., Eisenberg, H.M., Flamm, E., 110(Part 5): 1323–1344.
Leo-Summers, L., Maroon, J., et al. (1990) A randomized, Iwabuchi, N. (1997) Sacral glutamatergic transmission in the
controlled trial of methylprednisolone or naloxone in the descending limb of the micturition reflex in the cat. Fukuoka
treatment of acute spinal-cord injury. Results of the Second Igaku Zasshi., 88: 30–38.
National Acute Spinal Cord Injury Study. N. Engl. J. Med., Kakizaki, H., Fraser, M.O. and De Groat, W.C. (1997) Reflex
322: 1405–1411. pathways controlling urethral striated and smooth muscle
Chancellor, M.B., Kaplan, S.A. and Blaivas, J.G. (1990) De- function in the male rat. Am. J. Physiol., 272: R1647–R1656.
trusor–external sphincter dyssynergia. Ciba Found. Symp., Kruse, M.N., Belton, A.L. and de Groat, W.C. (1993) Changes
151: 195–206 discussion 207–113. in bladder and external urethral sphincter function after spi-
Cheng, C.L., Ma, C.P. and de Groat, W.C. (1995) Effect of nal cord injury in the rat. Am. J. Physiol., 264: R1157–R1163.
capsaicin on micturition and associated reflexes in chronic Kruse, M.N., Bray, L.A. and de Groat, W.C. (1995) Influence of
spinal rats. Brain Res., 678: 40–48. spinal cord injury on the morphology of bladder afferent and
de Groat, W.C. (1990) Central neural control of the lower uri- efferent neurons. J. Auton. Nervous System, 54: 215–224.
nary tract. Ciba Found. Symp., 151: 27–44 discussion 44–56. Kruse, M.N., Mallory, B.S., Noto, H., Roppolo, J.R. and de
de Groat, W.C. (1995) Mechanisms underlying the recovery of Groat, W.C. (1991) Properties of the descending limb of the
lower urinary tract function following spinal cord injury. spinobulbospinal micturition reflex pathway in the cat. Brain
Paraplegia, 33: 493–505. Res., 556: 6–12.
133

Kruse, M.N., Noto, H., Roppolo, J.R. and de Groat, W.C. Osborn, J.W., Taylor, R.F. and Schramm, L.P. (1990) Chronic
(1990) Pontine control of the urinary bladder and external cervical spinal cord injury and autonomic hyperreflexia in
urethral sphincter in the rat. Brain Res., 532: 182–190. rats. Am. J. Physiol., 258: R169–R174.
Lecci, A., Giuliani, S., Santicioli, P. and Maggi, C.A. (1992) Pikov, V. (2000) Recovery of the detrusor–external urethral
Involvement of 5-hydroxytryptamine 1A receptors in the sphincter coordination after incomplete spinal cord injury.
modulation of micturition reflexes in the anesthetized rat. Cell Biol., Georgetown University, Washington, DC.
J. Pharmacol. Exp. Ther., 262: 181–189. Pikov, V., Gillis, R.A., Jasmin, L. and Wrathall, J.R. (1998)
Little, J.W., Burns, S.P., James, J.J. and Stiens, S.A. (2000) Assessment of lower urinary tract functional deficit in rats
Neurologic recovery and neurologic decline after spinal cord with contusive spinal cord injury. J. Neurotrauma, 15:
injury. Phys. Med. Rehabil. Clin. N. Am., 11: 73–89. 375–386.
Maggi, C.A., Giuliani, S., Santicioli, P. and Meli, A. (1986a) Pikov, V. and Wrathall, J.R. (2001) Coordination of the blad-
Analysis of factors involved in determining urinary bladder der detrusor and the external urethral sphincter in a rat
voiding cycle in urethan-anesthetized rats. Am. J. Physiol., model of spinal cord injury: effect of injury severity. J. Ne-
251: R250–R257. urosci., 21: 559–569.
Maggi, C.A., Santicioli, P. and Meli, A. (1986b) The nonstop Pikov, V. and Wrathall, J.R. (2002) Altered glutamate receptor
transvesical cystometrogram in urethane-anesthetized rats: a function during recovery of bladder detrusor–external ure-
simple procedure for quantitative studies on the various thral sphincter coordination in a rat model of spinal cord
phases of urinary bladder voiding cycle. J. Pharmacol. Meth., injury. J. Pharmacol. Exp. Ther., 300: 421–427.
15: 157–167. Ramirez-Leon, V., Ulfhake, B., Arvidsson, U., Verhofstad,
Marson, L. (1997) Identification of central nervous system neu- A.A., Visser, T.J. and Hokfelt, T. (1994) Serotoninergic,
rons that innervate the bladder body, bladder base, or ex- peptidergic and GABAergic innervation of the ventrolateral
ternal urethral sphincter of female rats: a transneuronal and dorsolateral motor nuclei in the cat S1/S2 segments:
tracing study using pseudorabies virus. J. Comp. Neurol., an immunofluorescence study. J. Chem. Neuroanat., 7:
389: 584–602. 87–103.
Matsumoto, G., Hisamitsu, T. and de Groat, W.C. (1995a) Rinaman, L., Verbalis, J.G., Stricker, E.M. and Hoffman, G.E.
Non-NMDA glutamatergic excitatory transmission in the (1993) Distribution and neurochemical phenotypes of caudal
descending limb of the spinobulbospinal micturition reflex medullary neurons activated to express cFos following pe-
pathway of the rat. Brain Res., 693: 246–250. ripheral administration of cholecystokinin. J. Comp. Neurol.,
Matsumoto, G., Hisamitsu, T. and de Groat, W.C. (1995b) 338: 475–490.
Role of glutamate and NMDA receptors in the descending Rosenberg, L.J., Zai, L.J. and Wrathall, J.R. (2005) Chronic
limb of the spinobulbospinal micturition reflex pathway of alterations in the cellular composition of spinal cord white
the rat. Neurosci. Lett., 183: 58–61. matter following contusion injury. Glia, 49: 107–120.
Matsuura, S., Allen, G.V. and Downie, J.W. (1998) Volume- Schalow, G., Bersch, U., Michel, D. and Koch, H.G. (1995)
evoked micturition reflex is mediated by the ventrolateral Detrusor–sphincteric dyssynergia in humans with spinal cord
periaqueductal gray in anesthetized rats. Am. J. Physiol., 275: lesions may be caused by a loss of stable phase relations
R2049–R2055. between and within oscillatory firing neuronal networks of
Mersdorf, A., Schmidt, R.A. and Tanagho, E.A. (1993) Uro- the sacral micturition center. J. Auton. Nervous System, 52:
dynamic evaluation and electrical and pharmacologic neuro- 181–202.
stimulation. The rat model. Urol. Res., 21: 199–209. Schroder, H.D. (1980) Organization of the motoneurons in-
Mimata, H., Satoh, F., Tanigawa, T., Nomura, Y. and Ogata, nervating the pelvic muscles of the male rat. J. Comp. Ne-
J. (1993) Changes of rat urinary bladder during acute phase urol., 192: 567–587.
of spinal cord injury. Urol. Int., 51: 89–93. Tang, Y., Rampin, O., Calas, A., Facchinetti, P. and Giuliano,
Monaghan, D.T., Andaloro, V.J. and Skifter, D.A. (1998) Mo- F. (1998) Oxytocinergic and serotonergic innervation of
lecular determinants of NMDA receptor pharmacological identified lumbosacral nuclei controlling penile erection in
diversity. Prog. Brain Res., 116: 171–190. the male rat. Neuroscience, 82: 241–254.
Morrison, J. (1997) Central nervous control of the bladder. In: Teng, Y.D. and Wrathall, J.R. (1997) Local blockade of sodium
Jordan D. (Ed.), Central Nervous Control of Autonomic channels by tetrodotoxin ameliorates tissue loss and long-
Function. Harwood Academic Publishers, Amsterdam, term functional deficits resulting from experimental spinal
pp. 129–149. cord injury. J. Neurosci., 17: 4359–4366.
Nadelhaft, I. and Vera, P.L. (1996) Neurons in the rat brain Testa, R., Guarneri, L., Poggesi, E., Angelico, P., Velasco, C.,
and spinal cord labeled after pseudorabies virus injected into Ibba, M., Cilia, A., Motta, G., Riva, C. and Leonardi, A.
the external urethral sphincter. J. Comp. Neurol., 375: (1999) Effect of several 5-hydroxytryptamine(1A) receptor
502–517. ligands on the micturition reflex in rats: comparison with
Noble, L.J. and Wrathall, J.R. (1989) Correlative analyses WAY 100635. J. Pharmacol. Exp. Ther., 290: 1258–1269.
of lesion development and functional status after graded Thor, K.B. (2003) Serotonin and norepinephrine involvement in
spinal cord contusive injuries in the rat. Exp. Neurol., 103: efferent pathways to the urethral rhabdosphincter: implica-
34–40. tions for treating stress urinary incontinence. Urology, 62: 3–9.
134

Tiseo, P.J. and Yaksh, T.L. (1990) The spinal pharmacology of after spinal cord trauma with the AMPA/kainate antagonist
urinary function: studies on urinary continence in the un- NBQX. J. Neurosci., 14: 6598–6607.
anaesthetized rat. Ciba Found. Symp., 151: 91–104 discus- Wrathall, J.R., Li, W. and Hudson, L.D. (1998) Myelin gene
sion 104–109. expression after experimental contusive spinal cord injury.
Trautner, B.W. and Darouiche, R.O. (2002) Prevention of uri- J. Neurosci., 18: 8780–8793.
nary tract infection in patients with spinal cord injury. Wrathall, J.R., Pettegrew, R.K. and Harvey, F. (1985) Spinal
J. Spinal Cord Med., 25: 277–283. cord contusion in the rat: production of graded, reproduc-
Vizzard, M.A. (2000) Increased expression of spinal cord Fos ible, injury groups. Exp. Neurol., 88: 108–122.
protein induced by bladder stimulation after spinal cord in- Wyndaele, J.J. (1991) Investigation of the afferent nerves of the
jury. Am. J. Physiol. Regul. Integr. Comp. Physiol., 279: lower urinary tract in patients with ‘complete’ and ‘incom-
R295–R305. plete’ spinal cord injury. Paraplegia, 29: 490–494.
Vizzard, M.A., Erickson, V.L., Card, J.P., Roppolo, J.R. and de Yoshiyama, M., Nezu, F.M., Yokoyama, O., de Groat, W.C.
Groat, W.C. (1995) Transneuronal labeling of neurons in the and Chancellor, M.B. (1999) Changes in micturition after
adult rat brainstem and spinal cord after injection of pseudo- spinal cord injury in conscious rats. Urology, 54: 929–933.
rabies virus into the urethra. J. Comp. Neurol., 355: 629–640. Yoshiyama, M., Roppolo, J.R. and de Groat, W.C. (1994) Al-
Wrathall, J.R. (1994) Behavioral analysis in experimental spinal teration by urethane of glutamatergic control of micturition.
cord trauma. In: Salzman S. and Faden A. (Eds.), Neuro- Eur. J. Pharmacol., 264: 417–425.
biology of Central Nervous System Trauma. Oxford Uni- Yoshiyama, M., Roppolo, J.R. and de Groat, W.C. (1995) In-
versity Press, NY, pp. 79–85. teractions between NMDA and AMPA/kainate receptors in
Wrathall, J.R., Choiniere, D. and Teng, Y.D. (1994) Dose-de- the control of micturition in the rat. Eur. J. Pharmacol., 287:
pendent reduction of tissue loss and functional impairment 73–78.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 9

Role of the urothelium in urinary bladder


dysfunction following spinal cord injury

Lori A. Birder

Departments of Medicine and Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA

Abstract: A consequence of spinal cord injury is a change in bladder reflex pathways resulting in the
emergence of detrusor hyperreflexia and increased activity of the urethral sphincter. A basis for some of
these alterations could be changes in the environment of bladder sensory nerve endings at the target organ.
Recent evidence suggests that the urothelium (the lining of the urinary bladder) plays a prominent role in
modulating bladder sensory nerve ending excitability. It is conceivable that factors and processes affecting
the plasticity of bladder neurons after spinal cord injury may be partly due to changes occurring in the
urothelium. Although the urothelium has classically been thought of as a passive barrier to ions/solutes, a
number of novel properties have been recently attributed to these cells. Our work and that of others clearly
demonstrates that the urothelium exhibits both ‘‘sensor’’ (expression of sensor molecules or response to
thermal, mechanical and chemical stimuli) as well as ‘‘transducer’’ (release of factors/transmitters) prop-
erties. Taken together, these and other findings discussed in this chapter suggest a sensory function for the
urothelium and that alterations in urothelial properties may contribute to afferent abnormalities following
spinal cord injury.

The urothelium: an effective barrier against solutes accelerated proliferation can occur in pathology.
and pathogens For example, using a model which creates a selec-
tive injury of apical urothelial cells (protamine
The bladder urothelium is a specialized lining of sulfate), it has been shown that, in response to
the urinary tract, extending from the renal pelvis injury, the urothelium undergoes both functional
to the urethra. The urothelium is composed of at and structural changes in order to restore the bar-
least three layers: a basal cell layer attached to a rier (Lavelle et al., 2002).
basement membrane, an intermediate layer and a The umbrella cells function as a barrier against
superficial apical layer with large (diameters of most substances found in urine thus protecting the
25–250 mm) hexagonal ‘‘umbrella’’ cells (Lewis, underlying tissues (Negrete et al., 1996; Zeidel,
2000; Acharya et al., 2004). It has been reported 1996; Lewis, 2000; Apodaca, 2004). When this
for some species that both the umbrella and per- function is compromised during injury or inflam-
haps intermediate cells may have projections to the mation, it can result in the passage of toxic sub-
basement membrane (Martin, 1972; Hicks, 1975; stances into the underlying tissue (neural/muscle
Apodaca, 2004). The basal cells, which are thought layers) resulting in urgency, frequency and dysuria.
to be precursors for other cell types, normally ex- The superficial or umbrella cells play a prominent
hibit a low (3–6 months) turnover rate; however, role in maintaining this barrier, and exhibit a
number of unique properties including specialized
Corresponding author. Tel.: +412-383-7368; membrane lipids, asymmetric unit membrane par-
Fax: +412-648-7197; E-mail: lbirder@pitt.edu ticles and a plasmalemma with stiff plaques (Lewis,

DOI: 10.1016/S0079-6123(05)52009-0 135


136

2000; Hu et al., 2002; Apodaca, 2004). These cells properties that could allow reciprocal communi-
are also interconnected with extensive junctional cation with neighboring urothelial cells as well
complexes which include cytoskeletal elements, as nerve fibers or other cell types (i.e., immune,
and cytoplasmic and transmembrane proteins, myofibroblasts, inflammatory) in the bladder wall.
some of which play a role in cell–cell adhesion Recent studies have shown that afferent as well as
(Lewis, 2000; Acharya et al., 2004; Apodaca, autonomic axons are located in close proximity to
2004). This ‘‘water-tight’’ function of the apical the urothelium (Birder et al., 2001; Beckel et al.,
cell membrane is partly due to the above-mentioned 2004). Peptide- and TRPV1-immunoreactive nerve
specialized lipid molecules and uroplakin proteins, fibers have been found localized throughout the
a major protein component of the apical cell urinary bladder musculature and in a plexus
membrane, which reduce the permeability of beneath, and extending into, the urothelium. This
the urothelium to small molecules (water, urea, suggests that the release of a number of mediators
protons). Tight junction complexes are thought to from the urothelium could alter bladder nerve
reduce the movement of ions and solutes between excitability and, in turn, release of mediators from
cells (Tammela et al., 1993; Lewis, 2000; Wang nearby bladder nerves may also impact urothelial
et al., 2003; Apodaca, 2004). function. In support of this idea is evidence that
ATP (released from urothelial cells during stretch)
can activate a population of suburothelial bladder
Sensor and transducer functions of urothelium afferents expressing P2X3 purine receptors (Ferguson
et al., 1997; Burnstock, 2001), signaling changes in
Urothelial cells exhibit a number of properties bladder fullness and pain (Vlaskaovaka et al.,
similar to neurons (nociceptors/mechanorecep- 2001). Accordingly, P2X3 null mice exhibit urinary
tors): both types of cells use diverse signal-trans- bladder hyporeflexia, suggesting that this receptor
duction mechanisms to detect physiological and neural–epithelial interactions are necessary for
stimuli. Examples of ‘‘sensor molecules’’ (i.e., re- normal bladder function (Cockayne et al., 2000).
ceptors/ion channels) associated with neurons that Thus, the activation of bladder nerves and
have been thus far identified in urothelium include urothelial cells could modulate urinary bladder
receptors for: bradykinin (Chopra et al., 2005), function directly or indirectly via the release
neurotrophins (trkA and p75) (Wolf-Johnston of chemical factors in the urothelial layer. This
et al., 2004), purines (P2X and P2Y) (Lee et al., type of regulation may be similar to the epithelial-
2000; Birder et al., 2004a; Sun and Chai, 2004; dependent secretion of chemical factors in airway
Tempest et al., 2004), norepinephrine (a- and b-) epithelium thought to modulate submucosal
(Birder et al., 1998, 2002b), acetylcholine (mu- nerves and bronchial smooth muscle tone
scarinic and nicotinic) (Beckel et al., 2002, 2004; (Homolya et al., 2000; Jallat-Daloz et al., 2001).
Chess-Williams, 2002). Other sensor molecules The cellular mechanism(s) by which stretch
identified in urothelial cells are protease-activated evokes the release of ATP from epithelial, end-
receptors (D’Andrea et al., 2003), mechanosensi- othelial or other cell types is unclear. One hypoth-
tive Na+ channels (Lewis and Hanrahan, 1985; esis is that cellular distension causes intracellular
Wellner and Isenberg, 1993; Smith et al., 1998; vesicles rich in ATP to fuse with the urothelial cell
Carattino et al., 2005) and a number of transient membrane promoting ATP release and an increase
receptor potential (TRP) channels (TRPV1, in umbrella cell surface area during bladder filling.
TRPV2, TRPV4, TRPM8) (Birder et al., 2001, This mechanism would allow the bladder to ex-
2002a; Barrick et al., 2003; Stein et al., 2004). pand its epithelial surface area as urine accumu-
The ability of urothelial cells to express ‘‘sensor lates. Consistent with this possibility, mechanical
molecules’’ and release chemical mediators (nitric distension of the excised bladder has been shown
oxide, adenosine triphosphate (ATP), acetylcho- to trigger an increase in the membrane capacitance
line, substance P, prostaglandins) suggests that of the apical urothelial surface (Truschel et al.,
these cells exhibit specialized sensory and signaling 2002).
137

Impact of spinal cord injury on urothelial cell barrier prevents the ‘‘acute’’ cord injury-induced disrup-
function and morphology tions in both epithelial morphology and barrier
function (Figs. 2A–D). These and other findings
Spinal cord injury (transection of the spinal cord suggest the involvement of the autonomic nervous
in rats at level T8-9) resulted in changes in both system in the acute effects of cord injury on
urothelial morphology and barrier function the bladder urothelium. Studies have also shown
(Apodaca et al., 2003). At 24 h post spinal cord that the release of stress hormones (i.e., nor-
injury, there is a decrease in transepithelial resist- epinephrine) can disrupt the urothelial tight junc-
ance and an increase in permeability to water and tions with loss of urothelial cells (Veranic and
urea. The urothelium also exhibits a number of Jezernik, 2000). Thus, it is possible that transec-
regions which lack umbrella cells (Figs. 1E and F). tion of the spinal cord could induce the release of
The alterations in ultrastructure and function ‘‘stress-hormones’’ which could contribute to the
worsen within a few days following spinal cord changes observed in the urothelium at early time
injury, when significant disruptions in the urothe- points following spinal cord injury. We have found
lium are observed (Figs. 1G and H) correlating that intravesical administration of norepinephrine
with decreased transepithelial resistance and in- significantly altered epithelial permeability
creased permeabilities. Although some of these (decrease in transepithelial resistance) compared
changes could be the downstream consequence of to controls (Birder et al., unpublished results).
barrier disruption, some of the alterations could be Although the mechanism for these changes is
due to cord injury-induced urinary retention and under investigation, one possibility is adrenergic-
bladder overdistension. The bladders were not induced release of a soluble factor such as nitric
cannulated to avoid catheter-induced injury/in- oxide from urothelial cells (Birder et al., 2002b),
flammation, but were manually expressed several which in excess levels can alter the barrier function
times a day. Following recovery of the spinal reflex of the urothelium (Truschel et al., 2002). Alterna-
pathways and emergence of automatic micturition tively, cord injury-dependent neurotransmitter re-
14–28 days after spinal cord injury, barrier func- lease from efferent nerves could stimulate mast cell
tion was re-established, although the morphology release of histamine, bradykinin, prostaglandins,
of the urothelium was altered and the superficial leukotrienes and proteases, all of which could
urothelial cells were smaller (Figs. 1I–L). stimulate urothelial cells and contribute to tissue
Examination of urothelial morphology and damage and inflammation. In addition to the re-
function at earlier time points (1–2 h after tran- lease of catecholamines from bladder efferent
section) revealed significant changes in urothelial nerves, other modulators released from immune
morphology including areas of urothelium which cells might also play a role in the loss of barrier
lacked apical cells (Figs. 1C and D). These findings function after spinal cord injury.
correlated with decreased transepithelial resist- Reports suggest that capsaicin-sensitive nerves
ance, which suggests disruption of the tight junc- may contribute to mucosal protection following
tions and cell–cell contact. In contrast, only minor injury or inflammation (Abdel-Salam et al., 1999).
alterations in urea and water permeabilities oc- The neurotoxin capsaicin was therefore used to
curred at this time. The reason the permeabilities evaluate the involvement of capsaicin-sensitive
did not change is unknown. One possibility is the bladder afferents in changes in mucosa ultrastruc-
underlying cells still provide an adequate barrier ture and permeability after acute spinal cord injury
during the acute phase of spinal cord injury. (Figs. 2E–H). In these studies, capsaicin-pretreat-
A number of mechanisms may contribute to ment did not prevent functional changes but en-
these acute urothelial changes including increased hanced the susceptibility of the mucosa to injury
autonomic activity (i.e., release of catecholamines by decreasing transepithelial resistance compared
from stimulated efferent nerves) following spinal to untreated cord-injured animals or capsaicin-
cord injury. We have shown that pretreatment with treated controls. It has been suggested that the
the ganglionic blocking agent, hexamethonium, effect of capsaicin may be due to alterations in
138

Fig. 1. Scanning electron micrograph (SEM) of urothelial images taken from control and at various time points following spinal cord
injury (SCI). (A and B), Images taken from sham-treated animals; or 2 h (C and D), 24 h (E and F), 3 days (G and H), 14 days (I and J)
or 28 days (K and L) following SCI. Panels on the right depict a higher magnification view of inset regions shown on the left. (With
permission from Apodaca et al., Am. J. Physiol., 2003.) Arrows in F indicate small shrunken cells associated with epithelium.
139

Fig. 2. Scanning electron micrograph (SEM) of images depicting effects of either hexamethonium (50 mM) or capsaicin (100 mg/kg s.c.,
4 days prior) on acute changes in urothelial ultrastructure 2 h following spinal cord injury (SCI). (A and B) pretreatment with
hexamethonium before SCI; (C and D) hexamethonium pretreatment prior to sham surgery; (E and F) capsaicin pretreatment (4 days
prior) then SCI; (G and H) capsaicin pretreatment then sham surgery. Panels on the right depict a higher magnification view of inset
regions shown on the left. (With permission from Apodaca et al., Am. J. Physiol., 2003.)

substance P/calcitonin gene-related peptide con- leading to increased permeability may not be
tent in capsaicin-sensitive nerves (Szolcsanyi and evident using the employed morphological
Bartho, 2001). Although we did not detect a approaches.
significant difference in the surface architecture Thus, in addition to affecting the bladder de-
between treated animals and controls, dilation of trusor muscle and its innervation (de Groat, 1995),
extracellular spaces or alterations in tight junctions spinal cord injury also leads to a rapid disruption
140

of urothelial barrier function. This is evident by a been shown in sensory nerves that ATP can po-
loss of cell–cell interactions, decreased transepi- tentiate the response of vanilloid receptors (caps-
thelial resistance and increased water and urea aicin, protons and moderate heat act on vanilloid
permeabilities. Our results indicate that release of receptors) by lowering the threshold for responses
neurotransmitters by bladder efferent nerves is at to these stimuli (Tominaga et al., 2001). This rep-
least partially responsible for the disruption of the resents a novel mechanism through which the large
urothelium. Although the neurotransmitters and/ amounts of ATP released from damaged or sen-
or inflammatory mediators that disrupt barrier sitized cells in response to injury or inflammation
function are not well characterized, our observa- may trigger increased excitability of afferent
tions indicate that bladder nerves play an important nerves. These findings have clinical significance
role in regulating and maintaining barrier function. and suggest that alterations in afferents or epithe-
lial cells in pelvic viscera may contribute to the
sensory abnormalities in a number of pelvic dis-
Impact of spinal cord injury on urothelial cell sensor orders, including interstitial cystitis, a chronic
and transducer properties painful condition of the urinary bladder (Nickel,
2003). In a comparable disease in cats, termed fe-
Sensitization of urothelial cells and afferents can line interstitial cystitis (Buffington et al., 1999) we
be triggered by various mediators (nerve growth reported alterations in stretch-evoked release of
factor, ATP, nitric oxide, prostaglandins) released urothelium-derived ATP (Birder et al., 2003), con-
by both neuronal and non-neuronal cells (urothe- sistent with the augmented release of ATP from
lial cells, fibroblasts, mast cells) located near the urothelial cells from some patients with interstitial
luminal surface of the bladder. Increased endog- cystitis (Sun et al., 2001). We have recently found
enous levels of nerve growth factor and/or similar results in chronic spinal cord-injured cats
urothelial receptors for nerve growth factor (p75; (Birder et al., unpublished results), in which the
trkA) have been detected in the target organ augmented stretch-evoked ATP release from urothe-
(smooth muscle and urothelium) in a number of lial cells may contribute to bladder hyperreflexia.
bladder pathologies (Steers et al., 1991; Vizzard, The urothelium maintains a tight barrier to ion/
2000; Jallat-Daloz et al., 2001; Wolf-Johnston solute flux and augmented release of mediators
et al., 2003; Wolf-Johnston et al., 2004). More- such as nitric oxide from urothelial cells and/or
over, altered nerve growth factor levels (even in the nearby bladder nerves may play a role in the
absence of inflammation) have been linked to maintenance and regulation of this urothelial bar-
changes in the properties of afferent pathways rier function. Our previous studies have demon-
(Kornblum and Johnson, 1982; Dmitrieva and strated an upregulation in inducible nitric oxide
McMahon, 1996; Lamb et al., 2004). Thus, nerve synthase and elevated basal levels of nitric oxide
growth factor may play a significant role in en- measured in the bladder mucosa in cats with in-
hancing the sensitivity of a number of ‘‘sensor terstitial cystitis (Birder et al., 2005). Similar find-
molecules’’ within both the urothelium and senso- ings have been obtained clinically, as some patients
ry neurons. These findings suggest that targeting with classic interstitial cystitis also demonstrate
nerve growth factor and/or nerve growth factor elevated release of nitric oxide (Hosseini et al.,
signaling mechanisms may provide important in- 2004). As increased nitric oxide has been linked to
sight into new therapies for urinary bladder dys- cellular damage and alterations in epithelial bar-
function caused by inflammation or injury. rier function (Salzman et al., 1995; Arkovitz et al.,
Another important component of the injury/in- 1997), and it has been demonstrated that feline
flammatory response is ATP release from various interstitial cystitis is accompanied by changes in
cell types including the urothelium, which can in- bladder permeability and urothelial ultrastructure
itiate painful sensations by exciting purinergic (Lavelle et al., 2000), we examined whether nitric
(P2X) receptors on sensory fibers (Cockayne oxide levels might also be altered in rats following
et al., 2000; Burnstock, 2001). Recently, it has spinal cord injury, which results in changes in both
141

urothelial morphology and function (Apodaca of events that are thought to be part of symptoms
et al., 2003). In urothelium from normal rats, associated with urinary tract infections.
basal nitric oxide release (measured from the urinary Taken together, modification of the urothelium
bladder mucosal surface) remained undetectable and/or loss of epithelial integrity in a number of
(o10 nM nitric oxide release). However, after bladder pathologies could result in passage of tox-
chronic spinal cord injury, we detected elevated ic/irritating urinary constituents through the epi-
levels of basal nitric oxide release (200–500 nM) thelium leading to changes in the properties of
recorded from the mucosal surface of the urinary sensory pathways.
bladder (Truschel et al., 2001).
To evaluate the impact of elevated mucosal ni-
Therapeutic options for spinal cord injury that could
tric oxide levels on epithelial function, we exam-
target the urothelium
ined the effects of prolonged exposure to high
concentrations (2.5–5 mM) nitric oxide donors
An emerging body of evidence indicates that
(S-nitroso-N-acetyl penicillamine or sodium nit-
urothelial cells exhibit ‘‘polymodal’’ properties,
roprusside) on cultured urothelial cells. It has been
i.e., can be activated by chemical, thermal or me-
previously shown that cultured urothelial cells ex-
chanical stimuli, and that their activation can po-
hibit properties similar to native urothelium
tentially evoke the release of a myriad of
(Truschel et al., 1999). Normally, these urothelial
transmitters which can impact afferent activity
cultures exhibit a high transepithelial resistance
and ultimately bladder function. While the urot-
and low urea and water permeabilities. However,
helium has been historically viewed as primarily a
the administration of high concentrations of nitric
‘‘barrier,’’ it is becoming increasingly clear that it
oxide resulted in a significant decrease in transep-
is a responsive structure capable of detecting phys-
ithelial resistance (90% decrease as well as 3–5-
iological and chemical stimuli, and of releasing a
fold increase in permeability to water and urea), as
number of signaling molecules. The following is a
compared to controls (Truschel et al., 2001). This
summary of various therapies, most given intrave-
response was reversible upon washout of the nitric
sically, which are traditionally thought to target
oxide donor. Similar findings were obtained using
bladder nerves. It is conceivable that a number of
excised urinary bladder from rodents, in which
these treatments could also target urothelial re-
application of high concentrations of nitric oxide
ceptors and/or release mechanisms.
also decreased transepithelial resistance by 60%
compared to untreated control (Truschel et al.,
2001). Although their mechanism is unknown, Intravesical vanilloid compounds
these effects are reminiscent of similar observa-
tions in epithelia of other organs (lung, gut) in One example of a urothelial ‘‘neuronal-like’’ sen-
which excess production of nitric oxide has been sor molecule is the TRP channel TRPV1, known
linked to changes in epithelial integrity (Ding to play an important role in nociception and in
et al., 2004; Han et al., 2004). urinary bladder function (Szallasi, 2001). It is well
Disruption of epithelial integrity in some blad- established that the painful sensations induced by
der pathologies may also be due to substances such capsaicin, the pungent substance in hot peppers,
as antiproliferative factor, which has been shown are caused by stimulation of vanilloid receptor-1
to be secreted by bladder epithelial cells from in- (TRPV1), an ion channel protein which is activat-
terstitial cystitis patients and can inhibit epithelial ed by vanilloid compounds such as capsaicin,
proliferation thereby adversely affecting barrier moderate heat and protons (Caterina et al., 1997;
function (Keay et al., 1999, 2004). Uropathogenic Caterina, 2001). TRPV1 is highly expressed in
Escherichia coli can also bind to uroplakin pro- urinary bladder unmyelinated axons (C-fiber) that
teins present on the apical surface of superficial detect bladder distension or the presence of irritant
umbrella cells (Schilling and Hultgren, 2002). This chemicals (Chancellor and de Groat, 1999).
is thought to be an initial step leading to a cascade Intravesical instillation of vanilloid compounds
142

Fig. 3. (A) Confocal image of basal cells depicting TRPV1-immunoreactivity (cy-3, red) and cytokeratin-17, a marker for these cells
(Fluorscein isothiocyanate or FITC, green) immunoreactivity. (B) Confocal image of urinary bladder urothelium reveals TRPV1-
positive (cy-3, red) nerve fibers located in close proximity to basal urothelial cells (FITC, green). Punctate TRPV1 staining in urothelial
cells was electronically subtracted to facilitate imaging of the TRPV1-IR nerve fiber. (With permission from Birder et al., Proc. Natl.
Acad. Sci. USA, 2001.)

such as capsaicin or resiniferatoxin, which leads to ATP release and membrane capacitance as well as
desensitization of bladder nerves, has been shown a decrease in hypotonic or stretch-evoked ATP
to improve voiding efficiency significantly in cord- release from cultured TRPV1 null urothelial cells.
injured animals as well as in patients with detrusor Thus, the functional significance of these receptors
hyperactivity (Szallasi and Fowler, 2002; Kim in the bladder extends beyond pain sensation to
et al., 2003). include participation in normal bladder function.
One of the more remarkable findings in our own These receptors are also essential for normal me-
studies is that TRPV1 is not only expressed by chanically evoked, purinergic signaling by the
afferent nerves that form close contact with urothelium. In addition to the known effects on
urothelial cells but also by the urothelial cells bladder nerves, intravesical use of vanilloids could
themselves (Fig. 3) (Birder et al., 2001). Further, also target TRPV1 on urothelial cells, where
TRPV1 receptor expression correlates with sensi- persistent activation of urothelial TRPV1 might
tivity to vanilloid compounds, as exogenous ap- lead to receptor desensitization or depletion of
plication of capsaicin or resiniferatoxin increases urothelial-derived transmitters.
intracellular calcium and evokes the release of
transmitters (nitric oxide, ATP) in cultured ur- Antimuscarinic drugs
othelial cells. These responses are dependent upon
TRPV1 expression (Birder et al., 2001, 2002a). In Antimuscarinic drugs are widely regarded as a
neurons, TRPV1 is thought to integrate/amplify standard treatment in patients with neurogenic
the response to various stimuli and thus plays an lower urinary tract dysfunction (Andersson and
essential role in the development of inflammation- Yoshida, 2003). By targeting muscarinic receptors
induced hyperalgesia. Thus, it seems likely that on bladder smooth muscle, these agents prevent
urothelial-TRPV1 might participate in a similar receptor stimulation by acetylcholine released
manner, in the detection of irritant stimuli follow- from bladder efferent nerves and promote in-
ing bladder inflammation or infection. creased bladder capacity. However, these drugs are
While anatomically normal, TRPV1 null mice ex- thought to be effective during bladder storage when
hibited a number of alterations in bladder function parasympathetic nerves are silent. Since various
including a reduction of in vitro, stretch-evoked stimuli have been shown to release acetylcholine
143

from urothelial cells (Andersson and Yoshida, Diagnostic test for spinal cord injury: the ice
2003; Beckel et al., 2004), it is postulated that this water test
release from non-neural stores (i.e., urothelium)
could also contribute to detrusor overactivity It has been reported that intravesical instillation of
(Andersson and Yoshida, 2003). Thus, the effec- cold solutions can unmask the presence of de-
tiveness of some of these agents may be partly due trusor reflex activity in people with spinal cord
to targeting urothelial receptors and/or release injury (Balmaseda et al., 1988). This ‘‘bladder
mechanisms. Although muscarinic receptor sub- cooling reflex’’ is thought to be due to activation of
types have been detected on urothelial cells a subset of cold-sensitive C-fiber-type bladder
(Hawthorn et al., 2000; Beckel et al., 2004), a role afferents, which are sensitive to both cold temper-
for these receptors in bladder function has not yet atures and menthol (Jiang et al., 2002).
been established. Taken together, these data sug- Some TRP ion channels can be activated by a
gest that the bladder urothelium may be an addi- wide range of temperatures as well as by natural
tional source of acetylcholine that influences products (capsaicin, menthol), which can elicit
bladder contractility by modulating smooth mus- sensations of hot or cold (Patapoutian et al.,
cle tone and afferent activity. 2003). In contrast to TRPV1, which is a detector
of warm temperatures, TRPM8 has been shown to
be activated by cold temperatures as well as by
Botulinum toxin cooling agents (menthol) and is expressed in a
subset of sensory neurons (Clapham, 2003). Both
Recent studies have demonstrated that in- of these TRP channels are also expressed in blad-
tradetrusor injection of botulinum neurotoxin type der urothelium, suggesting that the urothelium can
A (Botox) is an effective therapy in a number of express a range of thermoreceptors underlying
lower urinary tract disturbances including the both ‘‘cold’’ and ‘‘heat’’ stimuli. While the func-
severe incontinence due to neurogenic detrusor tional role of these thermosensitive channels in
overactivity of spinal cord injury patients (Harper urothelium remains to be clarified, it seems likely
et al., 2004; Reitz and Schurch, 2004). Following that a primary role for these proteins may be
injection, the toxin binds to bladder cholinergic to recognize noxious stimuli in the bladder. For
nerve terminals and cleaves the protein, SNAP25, example, noxious cold solutions instilled into the
necessary for exocytosis and release of acetylcho- urinary bladder could, via stimulation of urothelial
line (Harper et al., 2004). In patients with spinal TRP channels, augment the release of urothelial-
cord injury, this treatment can lead to a significant derived mediators, thereby altering bladder affer-
reduction in episodes of incontinence and an in- ent excitability. Further studies are needed to
crease in maximum bladder capacity. elucidate fully the role of these TRP channels in
There is evidence that Botox can suppress the urothelium and influence on bladder function.
release of a number of mediators (acetylcholine,
ATP and neuropeptides) from both neural and
non-neural cells (Morris et al., 2001). Recent stud- Conclusion
ies have demonstrated that this agent can effec-
tively reduce both chemically and mechanically There is considerable interest in the putative role
evoked ATP release from cultured urothelial cells of urothelial receptors/ion channels and release
(Barrick et al., 2004). Alterations in the release of mechanisms in bladder function. By targeting
ATP or other transmitters from the urothelium various urothelial sensor molecules and/or modu-
could have a profound impact on neural excita- lating the release of transmitters/inflammatory me-
bility. Taken together, these findings suggest that diators, it may be possible to modulate afferent
targeting neural and non-neural release mechanisms activity and prevent the disruption of the urothe-
may be effective for the treatment of bladder lium that accompanies spinal cord injury and oth-
hyperreactivity in spinal cord injury. er bladder conditions. These results highlight the
144

need for additional studies in order to establish the Birder, L., Apodaca, G., de Groat, W.C. and Kanai, A.J. (1998)
physiological relevance of these urothelial targets. Adrenergic- and capsaicin-evoked nitric oxide release from
urothelium and afferent nerves in urinary bladder. Am. J.
Physiol., 275: F226–F229.
Acknowledgments Birder, L., Barrick, S.R., Roppolo, J.R., Kanai, A.J., de Groat,
W.C., Kiss, S. and Buffington, C.A. (2003) Feline interstitial
I thank Drs. A. Kanai and W. C. de Groat for cystitis results in mechanical hypersensitivity and altered
ATP release from bladder urothelium. Am. J. Physiol., 285:
critical comments and suggestions during prepa-
F423–F429.
ration of this chapter. The electron micrographs Birder, L., Kanai, A.J., de Groat, W.C., Kiss, S., Nealen, M.L.,
were prepared by W. Giovani Ruiz. This work was Burke, N.E., Dineley, K.E., Watkins, S., Reynolds, I.J. and
supported by grants to Lori A. Birder from the Caterina, M.J. (2001) Vanilloid receptor expression suggests
NIH (RO1-DK-54824 and RO1-DK-57284). a sensory role for urinary bladder epithelial cells. Proc. Natl.
Acad. Sci. USA, 98: 13396–13401.
Birder, L., Nakamura, Y., Kiss, S., Nealen, M.L., Barrick,
References S.R., Kanai, A.J., Wang, E., Ruiz, W.G., de Groat, W.C.,
Apodaca, G., Watkins, S. and Caterina, M.J. (2002a) Altered
Abdel-Salam, O.M., Debrecini, A., Mozsik, G. and Szolcsanyi, J. urinary bladder function in mice lacking the vanilloid
(1999) Capsaicin-sensitive afferent sensory nerves in modu- receptor TRPV1. Nat. Neurosci., 5: 856–860.
lating gastric mucosal defense against noxious agents. Birder, L., Nealen, M.L., Kiss, S., de Groat, W.C., Caterina,
J. Physiol. Paris, 93: 443–454. M.J., Wang, E., Apodaca, G. and Kanai, A.J. (2002b)
Acharya, P., Beckel, J., Ruiz, W.G., Wang, E., Rojas, R., Beta-adrenoceptor agonists stimulate endothelial nitric oxide
Birder, L. and Apodaca, G. (2004) Distribution of the tight synthase in rat urinary bladder urothelial cells. J. Neurosci.,
junction proteins ZO-1, occludin, and claudin-4, -8, and -12 22: 8063–8070.
in bladder epithelium. Am. J. Physiol., 287: F305–F318. Birder, L., Ruan, H.Z., Chopra, B., Xiang, Z., Barrick, S.R.,
Andersson, K.E. and Yoshida, M. (2003) Antimuscarinics and Buffington, C.A., Roppolo, J.R., Ford, A.P., de Groat, W.C.
the overactive detrusor-which is the main mechanism of ac- and Burnstock, G. (2004a) Alterations in P2X and P2Y
tion? Eur. Urol., 43: 1–5. purinergic receptor expression in urinary bladder from
Apodaca, G. (2004) The uroepithelium: not just a passive bar-
normal cats and cats with interstitial cystitis. Am. J. Physiol.,
rier. Traffic, 5: 1–12.
287: F1084–F1091.
Apodaca, G., Kiss, S., Ruiz, W.G., Meyers, S., Zeidel, M.L.
Birder, L., Wolf-Johnston, A.S., Buffington, C.A., Roppolo,
and Birder, L. (2003) Disruption of bladder epithelium bar-
J.R., de Groat, W.C. and Kanai, A.J. (2005) Altered
rier function after spinal cord injury. Am. J. Physiol., 284:
inducible nitric oxide synthase expression and nitric oxide
F966–F976.
Arkovitz, M.S., Wispe, J.R., Garcia, V.F. and Szabo, C. (1997) production in urinary bladder from cats with feline interstitial
Selective inhibition of the inducible isoform of nitric oxide cystitis. J. Urol., 173: 625–629.
synthase prevents pulmonary transvascular flux during acute Buffington, C.A., Chew, D.J. and Woodworth, B.E. (1999)
endotoxemia. J. Pediatr. Surg., 31: 1009–1015. Feline interstitial cystitis. J. Am. Vet. Med. Assoc., 215:
Balmaseda, M.T., Reynolds, H.T. and Gordon, C. (1988) The 682–687.
value of the ice water test in the management of the neuro- Burnstock, G. (2001) Purine-mediated signalling in pain
genic bladder. Am. J. Physiol. Med. Rehabil., 67: 225–227. and visceral perception. Trends Pharmacol. Sci., 22: 182–188.
Barrick, S.R., de Groat, W.C. and Birder, L.A. (2004) Carattino, M.D., Sheng, S. and Kleyman, T.R. (2005) Muta-
Regulation of chemical and mechanical-evoked ATP release tions in the pore region modify epithelial sodium channel
from urinary bladder urothelium by botulinum toxin type A. gating by shear stress. J. Biol. Chem., 280: 4393–4401.
Soc. Neurosci. Abstr., 26: 541.5. Caterina, M.J. (2001) The vanilloid receptor: a molecular
Barrick, S.R., Lee, H., Meyers, S., Caterina, M.J., Kanai, A.J., gateway to the pain pathway. Ann. Rev. Neurosci., 24:
Zeidel, M.L., Chopra, B., de Groat, W.C. and Birder, L. 602–607.
(2003) Expression and function of TRPV4 in urinary bladder Caterina, M.J., Schumacher, M.A., Tominaga, M., Rosen,
urothelium. Soc. Neurosci. Abstr., 26: 608.6. T.A., Levine, J.D. and Julius, D. (1997) The capsaicin
Beckel, J., Barrick, S.R., Keast, J.R., Meyers, S., Kanai, A.J., de receptor: a heat activated ion channel in the pain pathway.
Groat, W.C., Zeidel, M.L. and Birder, L. (2004) Expression Nature, 389: 816–824.
and function of urothelial muscarinic receptors and interac- Chancellor, M.B. and de Groat, W.C. (1999) Intravesical caps-
tions with bladder nerves. Soc. Neurosci. Abstr., 26: 846.23. aicin and resiniferatoxin therapy: spicing up the ways to treat
Beckel, J., Birder, L., Kiss, S., Kanai, A.J., Lee, S.J., the overactive bladder. J. Urol., 162: 3–11.
Yoshiyama, M. and de Groat, W.C. (2002) Expression of Chess-Williams, R. (2002) Muscarinic receptors of the urinary
nicotinic acetylcholine receptors in rat urothelium. Soc. bladder: detrusor, urothelial and prejunctional. Auton.
Neurosci. Abstr., 25: 538.10. Autacoid. Pharmacol., 22: 133–145.
145

Chopra, B., Barrick, S.R., Meyers, S., Beckel, J., Zeidel, M.L., Jiang, C.H., Mazieres, L. and Lindstrom, S. (2002) Cold- and
Ford, A.P., de Groat, W.C. and Birder, L. (2005) Expression menthol-sensitive C afferents of cat urinary bladder. J.
and function of bradykinin B1/B2 receptors in normal and in- Physiol., 543(1): 211–220.
flamed rat urinary bladder urothelium. J. Physiol., 562: 859–871. Keay, S., Warren, J.W., Zhang, C.O., Tu, L.M., Gordon, D.A.
Clapham, D.E. (2003) TRP channels as cellular sensors. Nat. and Whitmore, K.E. (1999) Antiproliferative activity is
Rev., 426: 517–524. present in bladder but not renal pelvic urine from intersti-
Cockayne, D.A., Hamilton, S.G., Zhu, Q.M., Dunn, P.M., tial cystitis patients. J. Urol., 162: 1487–1489.
Zhong, Y., Novakovic, S., Malmberg, A.B., Cain, G., Berson, Keay, S.K., Szekely, Z., Conrads, T.P., Veenstra, T.D., Barchi,
A., Kassotakis, L., Hedley, L., Lachnit, W.G., Burnstock, J.J., Zhang, C.O., Koch, K.R. and Michejda, C.J. (2004) An
G., McMahon, S.B. and Ford, A.P.D.W. (2000) Urinary antiproliferative factor from interstitial cystitis patients is a
bladder hyporeflexia and reduced pain-related behaviour in frizzled 8 protein-related sialoglycopeptide. Proc. Natl. Acad.
P2X(3)-deficient mice. Nature, 407: 1011–1015. Sci. USA, 101: 11803–11808.
D’Andrea, M.R., Saban, M.R., Nguyen, N.B., Andrade-Gordon, Kim, J.H., Rivas, D.A., Shenot, P.J., Green, B., Kennelly, M.,
P. and Saban, R. (2003) Expression of protease-activated Erickson, J.R., O’Leary, M., Yoshimura, N. and Chancellor,
receptor-1, -2, -3 and -5 in control and experimentally M.B. (2003) Intravesical resiniferatoxin for refractory
inflamed mouse bladder. Am. J. Pathol., 162: 907–923. detrusor hyperreflexia: a multicenter, blinded, randomized,
de Groat, W.C. (1995) Mechanisms underlying the recovery of placebo-controlled trial. J. Spinal Cord Med., 26: 358–363.
lower urinary tract function following spinal cord injury. Kornblum, H.I. and Johnson, E.M. (1982) Time and dose
Paraplegia, 33: 493–505. dependencies of effects of nerve growth factor on sympa-
Ding, L.A., Li, J.S., Li, Y.S., Zhu, N.T., Lui, F.N. and Tan, L. thetic and sensory neurons in neonatal rats. Brain Res.,
(2004) Intestinal barrier damage caused by trauma and lip- 234: 41–51.
opolysaccharide. W. J. Gastroentero., 10: 2373–2378. Lamb, K., Gebhart, G.F. and Bielefeldt, K. (2004) Increased
Dmitrieva, N. and McMahon, S.B. (1996) Sensitization of nerve growth factor expression triggers bladder overactivity.
visceral afferents by nerve growth factor in the adult rat. J. Pain, 5: 150–156.
Pain, 66: 87–97. Lavelle, J., Meyers, S., Ramage, R., Bastacky, S., Doty, D.,
Ferguson, D.R., Kennedy, I. and Burton, T.J. (1997) ATP is Apodaca, G. and Zeidel, M.L. (2002) Bladder permeability
released from rabbit urinary bladder epithelial cells by hy- barrier: recovery from selective injury of surface epithelial
drostatic pressure changes — a possible sensory mechanism? cells. Am. J. Physiol., 283: F242–F253.
J. Physiol., 505(2): 503–511. Lavelle, J., Meyers, S.A., Ruiz, W.G., Buffington, C.A., Zeidel,
Han, X., Fink, M.P., Uchiyama, T., Yang, R. and Delude, R.L. M.L. and Apodaca, G. (2000) Urothelial pathophysiological
(2004) Increased iNOS activity is essential for pulmonary changes in feline interstitial cystitis: a human model. Am. J.
epithelial tight junction dysfunction in endotoxemic mice. Physiol., 278: F540–F553.
Am. J. Physiol., 286: L259–L267. Lee, H.Y., Bardini, M. and Burnstock, G. (2000) Distribution
Harper, M., Fowler, C.J. and Dasgupta, P. (2004) Botulinum of P2X receptors in the urinary bladder and the ureter of the
toxin and its applications in the lower urinary tract. BJU Int., rat. J. Urol., 163: 2002–2007.
93: 702–706. Lewis, S.A. (2000) Everything you wanted to know about the
Hawthorn, M.H., Chapple, C.R., Cock, M. and Chess- bladder epithelium but were afraid to ask. Am. J. Physiol.,
Williams, R. (2000) Urothelium-derived inhibitory factor(s) 278: F867–F874.
influences on detrusor muscle contractility in vitro. Br. J. Lewis, S.A. and Hanrahan, J.W. (1985) Apical and basolateral
Pharmacol., 129: 416–419. membrane ionic channels in rabbit urinary bladder epitheli-
Hicks, M. (1975) The mammalian urinary bladder: an accom- um. Pflugers Arch., 405: S83–S88.
modating organ. Biol. Rev., 50: 215–246. Martin, B.F. (1972) Cell replacement and differentiation in
Homolya, L., Steinberg, T.H. and Boucher, R.C. (2000) Cell to transitional epithelium: a histological and autoradiographic
cell communication in response to mechanical stress via study of the guinea-pig bladder and ureter. J. Anat., 112:
bilateral release of ATP and UTP in polarized epithelia. 433–455.
J. Cell Biol., 150: 1349–1360. Morris, J.L., Jobling, P. and Gibbins, I.L. (2001) Differential
Hosseini, A., Ehren, I. and Wiklund, N.P. (2004) Nitric oxide as inhibition by botulinum neurotoxin A of cotransmitters re-
an objective marker for evaluation of treatment response in leased from autonomic vasodilator neurons. Am. J. Physiol.,
patients with classic interstitial cystitis. J. Urol., 172: 2261–2265. 281: H2124–H2127.
Hu, P., Meyers, S., Liang, F.X., Deng, F.M., Kachar, B., Zeidel, Negrete, H.O., Lavelle, J., Berg, J., Lewis, S.A. and Zeidel,
M.L. and Sun, T.T. (2002) Role of membrane proteins in M.L. (1996) Permeability properties of the intact mammalian
permeability barrier function: uroplakin ablation elevates bladder epithelium. Am. J. Physiol., 271: F886–F894.
urothelial permeability. Am. J. Physiol., 283: F1200–F1207. Nickel, J.C. (2003) Interstitial cystitis — an elusive clinical
Jallat-Daloz, I., Cognard, J.L., Badet, J.M. and Regnard, J. target? J. Urol., 170: 816–817.
(2001) Neural-epithelial cell interplay: in vitro evidence that Patapoutian, A., Peier, A.P., Story, G.M. and Viswanath, V.
vagal mediators increase PGE2 production by human nasal (2003) ThermoTRPs and beyond: mechanisms of tempera-
epithelial cells. Allergy Asthma Proc., 22: 17–23. ture sensation. Nat. Rev. Neurosci., 4: 529–539.
146

Reitz, A. and Schurch, B. (2004) Intravesical therapy options for Tominaga, M., Wada, M. and Masu, M. (2001) Potentiation
neurogenic detrusor overactivity. Spinal Cord, 42: 267–272. of capsaicin receptor activation by metabotropic ATP
Salzman, A.L., Menconi, M.J., Unno, N., Ezzell, R.M., Casey, receptors: a possible mechanism for ATP-evoked pain and
D.M., Gonzalez, P.K. and Fink, M.P. (1995) Nitric oxide hypersensitivity. Proc. Natl. Acad. Sci. USA, 29: 820–825.
dilates tight junctions and depletes ATP in cultured Caco- Truschel, S.T., Ruiz, W.G., Kanai, A.J., Kiss, S., Meyers, S.,
2BBe intestinal epithelial monolayers. Am. J. Physiol., 268: Apodaca, G., de Groat, W.C., Maggi, C.A., Zeidel, M.L. and
G361–G373. Birder, L. (2001) Involvement of nitric oxide (NO) in bladder
Schilling, J. and Hultgren, S. (2002) Recent advances into the afferent and urothelial abnormalities following chronic spinal
pathogenesis of recurrent urinary tract infections: the bladder cord injury. Soc. Neurosci. Abstr., 24: 842.9.
as a reservoir for uropathogenic Escherichia coli. Int. J. Truschel, S.T., Ruiz, W.G., Shulman, T., Pilewski, J., Sun,
Antimicrob. Agents, 19: 457–460. T.T., Zeidel, M.L. and Apodaca, G. (1999) Primary uroep-
Smith, P.R., Mackler, S.A., Weiser, P.C., Brooker, D.R., Ahn, ithelial cultures. A model system to analyze umbrella cell
Y.J., Harte, B.J., McNulty, K.A. and Kleyman, T.R. (1998) barrier function. J. Biol. Chem., 274: 15020–15029.
Expression and localization of epithelial sodium channel in Truschel, S.T., Wang, E., Ruiz, W.G., Leung, S.M., Rojas, R.,
mammalian urinary bladder. Am. J. Physiol., 274: F91–F96. Lavelle, J., Zeidel, M.L., Stoffer, D. and Apodaca, G. (2002)
Steers, W.D., Ciambotti, J., Etzel, B., Erdman, S. and de Groat, Stretch-regulated exocytosis/endocytosis in bladder umbrella
W.C. (1991) Alterations in afferent pathways from the cells. Mol. Biol. Cell, 13: 830–846.
urinary bladder of the rat in response to partial urethral Veranic, P. and Jezernik, K. (2000) The response of junctional
obstruction. J. Comp. Biol., 310: 401–410. complexes to induced desquamation in mouse bladder urot-
Stein, R.J., Santos, S., Nagatomi, J., Hayashi, Y., Minnery, helium. Biol. Cell., 92: 105–113.
B.S., Xavier, M., Patel, A.S., Nelson, J.B., Futrell, W.J., Vizzard, M.A. (2000) Changes in urinary bladder neurotrophic
Yoshimura, N., Chancellor, M.B. and deMiguel, F. (2004) factor mRNA and NGF protein following urinary bladder
Cool (TRPM8) and hot (TRPV1) receptors in the bladder dysfunction. Exp. Neurol., 161: 273–284.
and male genital tract. J. Urol., 172: 1175–1178. Vlaskaovaka, M., Kasakov, L., Rong, W.F., Bodin, R., Bardini,
Sun, Y. and Chai, T.C. (2004) Up-regulation of P2X3 receptor M., Cockayne, D.A., Ford, A.P.D.W. and Burnstock, G.
during stretch of bladder urothelial cells from patients with (2001) P2X3 knockout mice reveal a major sensory role for
interstitial cystitis. J. Urol., 171: 448–452. urothelially released ATP. J. Neurosci., 21: 5670–5677.
Sun, Y., Keay, S., DeDeyne, P.G. and Chai, T.C. (2001) Wang, E., Lee, J.M., Johnson, J.P., Kleyman, T., Bridges, R.
Augmented stretch activated adenosine triphosphate release and Apodaca, G. (2003) Hydrostatic pressure-regulated ion
from bladder uroepithelial cells in patients with interstitial transport in bladder uroepithelium. Am. J. Physiol., 285:
cystitis. J. Urol., 166: 1951–1956. F651–F663.
Szallasi, A. (2001) Vanilloid receptor ligands: hopes and Wellner, M.C. and Isenberg, G. (1993) Properties of stretch-
realities for the future. Drugs Aging, 18: 561–573. activated channels in myocytes from the guinea pig urinary
Szallasi, A. and Fowler, C.J. (2002) After a decade of bladder. J. Physiol., 466: 213–227.
intravesical vanilloid therapy: still more questions than Wolf-Johnston, A.S., Beckel, J.M., Barrick, S.R., Kanai, A.J.,
answers. Lancet Neurol., 1: 167–172. de Groat, W.C. and Birder, L. (2004) Induction of urinary
Szolcsanyi, J. and Bartho, L. (2001) Capsaicin-sensitive affer- cystitis results in increased expression of nerve growth factor
ents and their role in protection: an update. J. Physiol. Paris, and its receptors. Soc. Neurosci. Abstr., 27: 742.16.
95: 181–188. Wolf-Johnston, A.S., Buffington, C.A., Roppolo, J.R., Tai, C.,
Tammela, T., Wein, A.J., Monson, F.C. and Levin, R.M. Kanai, A.J., de Groat, W.C. and Birder, L. (2003) Increased
(1993) Urothelial permeability of the isolated whole bladder. NGF and TRPV1 expression in urinary bladder and sensory
Neurourol. Urodynam., 12: 39–47. neurons from cats with feline interstitial cystitis. Soc.
Tempest, H.V., Dixon, A.K., Turner, W.H., Elneil, S., Sellers, Neurosci. Abstr., 27: 608.4.
L.A. and Ferguson, D.R. (2004) P2X and P2Y receptor Zeidel, M.L. (1996) Low permeabilities of apical membranes of
expression in human bladder urothelium and changes in barrier epithelia: what makes water-tight membranes water-
interstitial cystitis. BJU Int., 93: 1344–1388. tight? Am. J. Physiol., 271: F243–F245.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 10

Plasticity in the injured spinal cord: can we use it


to advantage to reestablish effective bladder voiding
and continence?

Natasha D.T. Zinck1, and John W. Downie1,2

1
Department of Pharmacology, Faculty of Medicine, Dalhousie University, 5850 College St., Halifax, NS B3H 1X5,
Canada
2
Department of Urology, Faculty of Medicine, Dalhousie University, 5850 College St., Halifax, NS B3H 1X5, Canada

Abstract: Micturition is coordinated at the level of the spinal cord and the brainstem. Spinal cord injury
therefore directly interrupts spinal neuronal pathways to the brainstem and results in bladder areflexia.
Some time after injury, however, dyssynergic bladder and sphincter function emerges. The changes me-
diating the appearance of bladder function after spinal cord injury are currently unknown. Primary afferent
neurons have been shown to sprout in response to spinal cord injury. Sprouting primary afferents have been
linked to the pathophysiology of centrally manifested disorders, such as autonomic dysreflexia and ne-
uropathic pain. It is proposed that sprouting of bladder primary afferents contributes to disordered bladder
functioning after spinal cord injury. During development of the central nervous system, the levels of specific
neuronal growth-promoting and guidance molecules are high. After spinal cord injury, some of these
molecules are upregulated in the bladder and spinal cord, suggesting that axonal outgrowth is occurring.
Sprouting in lumbosacral spinal cord is likely not restricted to neurons involved in the micturition reflex.
Furthermore, sprouting of some afferents may be contributing to bladder function after injury, whereas
sprouting of others might be hindering emergence of function. Thus selective manipulation of sprouting
targeting afferents that are contributing to emergence of bladder function after injury is critical. Further
research regarding the role that neuronal sprouting plays in the emergence of bladder function may con-
tribute to improved treatment of bladder dyssynergia after spinal cord injury.

Introduction neurons exist within an environment rich in mole-


cules that are important for axonal guidance,
Plasticity within the spinal cord outgrowth and targeting (Drescher et al., 1997;
Dickson and Senti, 2002). These molecules act by
Axonal outgrowth and pathfinding are limited either attracting or repelling the leading edge of
within the adult central nervous system (CNS). growing axons (the growth cone), thus directing
During development, however, axonal outgrowth developing axons to their appropriate targets
and pathfinding are critical for a properly wired, (Gallo and Letourneau, 2004; Gordon-Weeks,
and thus, functioning nervous system (Crowley 2004). It has been proposed that there is a de-
et al., 1994; Maier et al., 1999). Developing crease or absence in growth-promoting molecules
once adulthood is reached, and that this is why the
Corresponding author. Tel.: +(902) 494-3459; adult CNS is unable to promote neuronal plastic-
Fax: +(902) 494-1388; E-mail: nzinck@dal.ca ity. For this reason, much research has focused on

DOI: 10.1016/S0079-6123(05)52010-7 147


148

reproducing the molecular environment that is Axonal remodeling occurs normally within the
present during development with the hopes of aid- developing micturition reflex
ing neuronal repair and regrowth after CNS injury.
On the other hand, CNS plasticity may also have The micturition reflex is subject to extensive re-
negative consequences. There is increasing evidence modeling during development. For approximately
that upregulation of key elements involved in aid- the first 3 weeks of life, micturition in rats and cats
ing axonal outgrowth contributes to the pathogen- is evoked by stimulation of somatic perineal af-
esis of centrally manifested disorders. These include ferents, via licking of the perineum by the mother
neuropathic pain (Theodosiou et al., 1999) as well (Maggi et al., 1986; Thor et al., 1989). The effe-
as autonomic dysreflexia (Krenz et al., 1999). Spi- rent limb, through the pelvic nerve, stimulates
nal cord injury, depending on its severity, affects detrusor contraction, thereby facilitating bladder
many centrally mediated visceral functions, includ- emptying. This somatic-bladder reflex is a spinal
ing bladder function. Normal micturition requires reflex that becomes progressively weaker through-
supraspinal integration at the level of the pontine out postnatal life to a point where there appears to
micturition center (Barrington, 1915). Suprasacral be a switch from spinally mediated to supraspin-
spinal cord injury disconnects the parasympathetic ally mediated micturition (de Groat et al., 1998).
spinal outflow from the pons rendering the bladder Micturition in these animals, as in adults, is in
areflexic. Bladder function is novel in that micturit- response to bladder stretch, having both afferent
ion emerges some time after spinal cord injury and efferent limbs in the pelvic nerve. This switch
without neuronal integration at the level of the is thought to be accompanied by major changes in
brainstem (Yoshiyama et al., 1999). However, post neuronal circuits used to elicit micturition.
spinal cord injury, voiding is dysfunctional due to Developing nervous systems undergo a great
lack of coordination between the detrusor muscle deal of synaptic strengthening and neuronal re-
and the external urethral sphincter, clinically finement. One process thought to play a major role
termed detrusor–sphincter dyssynergia (Kruse during development of the micturition reflex is
et al., 1993). For spinal cord-injured patients, this synaptic competition. The numbers of synapses on
results in hyperactive inefficient bladder function. spinal neurons derived from spinal and supraspinal
Treatments for these patients focus on alleviating sources change throughout development (Fig. 1).
hyperactive bladder dysfunction pharmacologically It is thought that synaptic input to the para-
via non-selective anti-muscarinic agents (Pannek sympathetic preganglionic nucleus from supraspi-
et al., 2000), reducing dyssynergy surgically nal centers increases during postnatal development
through external sphincterotomy (Reynard et al., and out competes sacral interneurons for the same
2003) and generating voiding on demand by sacral target (de Groat, 2002). This may explain the loss
anterior root stimulation (Schumacher et al., 1999). of spinally mediated micturition and the emer-
Spinal cord injury has been shown to induce gence of supraspinally mediated micturition dur-
neuronal sprouting within the spinal cord. Plastic- ing development.
ity of neurons within the micturition reflex circuit Coincidently, spinal cord injury in adult animals
is a possible mechanism by which bladder function triggers a switch back to micturition mediated via
emerges after spinal cord injury. Continued re- a spinal reflex (Kakizaki and de Groat, 1997;
search regarding the role that neuronal sprouting Shefchyk and Buss, 1998) thereby making synaptic
plays in bladder function after spinal cord injury competition an hypothesis not only for the emer-
will aid in the development of treatment methods gence of brain stem-mediated micturition during
which target the cause of bladder dysfunction after development, but also for the emergence of spinal
injury rather than its symptoms. In this chapter, reflex micturition after spinal cord injury.
we will discuss the evidence for neuronal plasticity If the alternative neuronal pathways for eliciting
occurring in the micturition reflex path and how bladder contraction already exist in the spinal
this may mediate the emergence of bladder dys- cord, why does it take so long for a spinal reflex to
function after spinal cord injury. emerge after spinal cord injury? Assuming that
149

transection (Yoshiyama et al., 1999) and in humans,


depending on the severity of the injury, it may not
emerge for months (Weld and Dmochowski, 2000).
This delay in emergence of the bladder function
implies that other changes associated with neurons
in the micturition reflex, both phenotypic and
anatomical, may be involved in emergence of
bladder function after spinal cord injury.

Bladder primary afferents and spinal cord injury

General characteristics

Retrograde tracers such as cholera toxin B subunit


and horseradish peroxidase have proven useful for
illustrating the pattern of bladder primary afferent
termination in the spinal cord (Morgan et al.,
1981; Nadelhaft and Booth, 1984; Wang et al.,
1998). Bladder primary afferents enter Lissauer’s
tract from which two major tracts form the path-
ways of entry into the gray matter of the dorsal
horn. The lateral collateral pathway extends from
superficial dorsal horn (lamina I and II) through
the dorsolateral funiculus and terminates densely
in the area of the parasympathetic preganglionic
nucleus. A few terminations are also seen to enter
the dorsal gray commissure from this pathway.
A second and less dense pathway, the medial col-
lateral pathway, sends fibers from the dorsomedial
border of the dorsal horn in laminae I and II
Fig. 1. Synaptic competition may explain the switch from spi- to lamina X. Terminating fibers from this pathway
nal to brainstem control of micturition during development. It can also be seen in medial laminae V and VI.
is proposed that sacral preganglionic neurons (PGN) in neon-
ates receive input from sacral interneurons (INT) as well as
Bladder primary afferents exhibit a periodical
from descending fibers from the brain stem. In young neonates rostrocaudal termination pattern within the
a spinal reflex elicits micturition because sacral interneurons spinal cord.
outcompete any descending modulation by the brainstem. As Bladder primary afferents consist of small un-
the neonate ages the number of synapses on preganglionic neu- myelinated C-fibers and thinly myelinated A-delta
rons from sacral interneurons decreases, and the number from
the brain stem increases. The strong synaptic input from
fibers. These subtypes can be divided further into
the brainstem is thought to underlie supraspinally controlled peptidergic and non-peptidergic groups. Pep-
micturition in the older neonate and throughout adulthood. tidergic fibers are characterized by the presence
From de Groat (2002). of calcitonin gene-related peptide and substance P.
Non-peptidergic primary afferents contain fluoride
spinal shock accounts for bladder areflexia during resistant acid phosphatase activity and are able to
the first few days after injury, reflex mediated bind the plant isolectin B4 (IB4) (Stucky and
micturition should occur soon after resolution of Lewin, 1999). Regionally, peptidergic afferents
areflexia. However, micturition does not emerge terminate in lamina I as well as lamina II outer,
in rats until around 2 weeks after spinal cord whereas non-peptidergic afferents terminate in
150

lamina II inner. 86% of all bladder primary affer- the density and distribution of trk receptors
ents innervating the bladder body are peptidergic thought to play a role in growth-promoting signa-
fibers (Yoshimura et al., 2003). The greatest pro- ling (Qiao and Vizzard, 2002) among L6/S1 dorsal
portion of non-peptidergic afferent innervation is root ganglion cells.
to the distal urethra where close to 30% of sensory
innervation is from IB4 positive fibers.
Sprouting and the factors implicated in this response
Further characterization of these two groups
reveals that peptidergic and non-peptidergic fibers
As well as phenotypic changes, anatomical reor-
possess different trophic factor receptors and
ganization of bladder primary afferents may con-
therefore respond to distinct neurotrophic factors.
tribute to emergence of bladder function after
Peptidergic primary afferents contain the trkA nerve
spinal cord injury.
growth factor receptor while non-peptidergics
do not contain trk receptors but contain glial
derived neurotrophic factor receptors Ret and Neurotrophic factors
GRFalpha1. Neuronal plasticity that follows spinal cord injury
is thought to be regulated largely by neurotrophic
factors. If neuronal plasticity is involved in the
Phenotypic changes emergence of bladder function after spinal cord
injury, then changes in trophic factor levels at both
After spinal cord injury, bladder primary afferents organ and spinal levels are likely to be involved.
may undergo changes in phenotype as well as a Nerve growth factor provides trophic support to
sprouting response. In the cat, under normal the majority of bladder primary afferents and
conditions, bladder sensory information travels to chemically increases the sensitivity of primary affe-
the spinal cord via mechano-sensitive A-delta rents (Lamb et al., 2004). Several studies have in-
fibers (Janig and Morrison, 1986). After spinal vestigated the role that nerve growth factor may
cord injury, sensory information is conveyed by play in bladder function after spinal cord injury.
what were once mechano-insensitive C-fibers Increases in spinal nerve growth factor have
(de Groat et al., 1990). This increase in C-fiber been linked to the development of disorders such
afferent excitability may be mediated by a decrease as neuropathic pain and autonomic dysreflexia. In
in tetrodotoxin-resistant and an increase in both disorders, nerve growth factor is increased
tetrodotoxin-sensitive Na+ channel expression predominantly within small diameter primary affe-
(Yoshimura and de Groat, 1997; Waxman et al., rent neurons. Neuronal nerve growth factor is in-
1999). A decrease in Nav 1.8 channels, a subtype of creased in lumbar and sacral (L6/S1) dorsal root
tetrodotoxin-resistant Na+ channels, is associated ganglia and L6 spinal cord after thoracic spinal
with bursting behavior of cerebellar Purkinje cord injury (Seki et al., 2002). Schwann cells,
neurons (Renganathan et al., 2003) as well as astrocytes and other microglia also upregulate
spontaneous activity of dorsal root ganglion their expression of nerve growth factor after spinal
cells (Renganathan et al., 2001). Decreased ion cord injury (Krenz and Weaver, 2000). In models
conductance across A-type potassium channels is of neuropathic pain, expression of nerve growth
also thought to contribute to increased afferent factor by glial cells sensitizes primary afferent
excitability (Sculptoreanu et al., 2004). nociceptors leading to hyperalgesia and allodynia.
Although overall afferent number remains the The spinal cord is not the only source of nerve
same, there is an increase in proportion of growth factor. In fact, the bladder has increased
myelinatated fibers (Yoshimura et al., 1998) as levels of nerve growth factor mRNA acutely after
well as dorsal root ganglion cell body size (Yu spinal cord injury. Upregulation of nerve growth
et al., 2003) among bladder primary afferents after factor protein and increased expression of its re-
spinal cord injury. Other injury-induced changes ceptor, trkA, also occur in the bladder and dorsal
to bladder afferent phenotype include changes in root ganglia 6 weeks post injury (Vizzard, 2000;
151

Qiao and Vizzard, 2002), well after the emergence unpublished data). Furthermore, administration
of voiding function in the rat. Thus bladder- of antibodies against nerve growth factor after
derived nerve growth factor may facilitate changes spinal cord injury leads to a decrease in density
that occur in bladder function at chronic time- and distribution of these peptidergic fibers in the
points after spinal cord injury but not the initial dorsal horn (Christensen and Hulsebosch, 1997).
emergence of bladder function. Nerve growth fac- Therefore increases in L6/S1 spinal calcitonin
tor acts on peptidergic C-fibers and has been gene-related peptide may be the consequence of
shown to induce the expression and secretion of peptidergic fiber sprouting elicited by increased
calcitonin gene-related peptide (Bowles et al., nerve growth factor. At both acute (3 days)
2004). Increased calcitonin gene-related peptide (N. Zinck, V. Rafuse, J. Downie, unpublished
distribution within the L6/S1 spinal cord segments data) and chronic (6 weeks) (Vizzard and Boyle,
has been demonstrated in spinal cord injury rats at 1999) time-points post spinal cord injury growth
timepoints before the emergence of bladder func- associated protein-43 is increased within L6/S1 spi-
tion (Fig. 2) (N. Zinck, V. Rafuse and J. Downie, nal cord segments, suggesting synaptic remodeling

Fig. 2. Calcitonin gene-related peptide-immunoreactive primary afferents sprout in rat lumbosacral spinal cord after spinal cord
injury. Inset shows the approximate level of the longitudinal sections. Increases in density and distribution of fiber terminations is seen
in rats 8 days post spinal cord transection at T10 when compared to non-injured (control) rats. Arrowhead indicates filling in of gaps
between primary afferent termination bundles. SPN, sacral parasympathetic nucleus; DCG, dorsal commissural nucleus. (N.D.T.
Zinck, V.F. Rafuse, J.W. Downie, unpublished.)
152

is occurring both before and after emergence of the presence of nerve growth factor. Because nerve
bladder function. growth factor-stimulated brain-derived neurotro-
Although previous literature suggested that phic factor upregulation is associated with inflam-
non-peptidergic afferents do not sprout in re- matory processes in the peripheral nervous system,
sponse to injury (Belyantseva and Lewin, 1999), central inflammatory processes after spinal cord
recent studies report increased fiber branching of injury may not be contributing to increases in brain-
IB4 positive neurons within the dorsal root ganglia derived neurotrophic factor at the spinal level.
after spinal nerve transection (Li and Zhou, 2001).
Bladder mRNA levels of glial-derived neurotro- Cellular adhesion molecules
phic factor are also increased after spinal cord in- A critical aspect of neuronal outgrowth is appro-
jury in the rat (Vizzard, 2000) although direct priate synapse formation, facilitated largely by
studies of IB4 positive fiber density or distribution changes in adhesivity of growth cones. Several
in L6/S1 spinal cord segments after spinal cord factors have been implicated in mediating adhe-
injury have not been reported. Furthermore, un- sion of individual axon terminals to neighboring
like nerve growth factor, no studies have been cells or to the extracellular matrix. Three major
conducted that selectively target non-peptidergic players in the cell adhesion molecule family are
afferents via application of glial-derived neurotro- N-cadherin, the Ig cell adhesion molecules, L1 and
phic factor or antibodies to glial-derived neurotro- neural cell adhesion molecule (NCAM) (Kiryushko
phic factor in lumbosacral spinal cord making it et al., 2004). Homophilic binding of these members
difficult to determine what role this neurotrophin is necessary for proper adhesion and synapse
plays in the micturition reflex. formation throughout the development of the
Brain-derived neurotrophic factor is a member central nervous system.
of the nerve growth factor family and is also NCAM levels are greatly upregulated at critical
associated with small to medium diameter timepoints in neuronal development (Bruses et al.,
peptidergic primary afferents that terminate in 2002). Alternative splicing of the same gene results
lamina I and II of the dorsal horn (Luo et al., in three different isoforms of NCAM that have
2001). It is upregulated and synthesized in response been implicated in the creation of neuronal
to peripheral inflammation (Apfel et al., 1996). In networks during development and after injury in
fact, increases in nerve growth factor have been the adult nervous system (Kiss and Muller, 2001).
linked to increased expression of brain-derived Named by their molecular weights, NCAM-180
neurotrophic factor in trkA-containing sensory and -140 span the cell membrane whereas NCAM-
afferents and to heightened pain sensitivity associ- 120 has only an extracellular domain. NCAM can
ated with neuropathic pain (Obata et al., 2003). be separated from other members of the cell
Increased expression of brain-derived neurotrophic adhesion molecule family by their ability to bind
factor occurs throughout the spinal cord, including homopolymers of the carbohydrate polysialic acid.
L6/S1, after complete thoracic spinal cord injury. Polysialic acid-bound NCAM facilitates a decrease
Upregulation of trk B within bladder-specific in homophilic NCAM binding and thus a decrease
dorsal root ganglion neurons also occurs after in cellular adhesion and an increase in axonal
spinal cord injury (Qiao and Vizzard, 2002). defasciculation (Tang and Landmesser, 1993;
Unlike other neurotrophic factors studied, Monnier et al., 2001). Polysialic acid has been
increased spinal brain-derived neurotrophic factor shown to increase after central (Bonfanti et al.,
expression occurs both acutely and chronically 1996) and peripheral nerve (Franz et al., 2005)
after spinal cord injury. In fact, in the lumbosacral injury. Increases in polysialic acid have been asso-
cord, brain-derived neurotrophic factor upregula- ciated with regeneration of injured neurons of the
tion precedes that of nerve growth factor (Zvarova hippocampus (Aubert et al., 1998). During the first
et al., 2004), implying that in spinal cord injury, few weeks after birth neonatal rats use a spinal
the increased brain-derived neurotrophic factor pathway to elicit micturition. Approximately three
expression in primary afferents does not require weeks after birth bladder emptying is controlled by
153

supraspinal neurons. Polysialic acid-NCAM levels activation. NCAM is not only unique in its ability
are elevated in L6/S1 spinal cord segments of to bind polysialic acid, but has been shown to act
postnatal day 6 (P6) rats (Fig. 3) (N. Zinck, V. as a co-receptor with GFRa1, a glycosylpho-
Rafuse, J. Downie, unpublished data) when com- sphatidylinositol-linked receptor for glial-derived
pared to adult control, as suggested by the level of neurotrophic factor (Zhou et al., 2003). Glial-de-
smearing in lane one (P6) compared to lane two rived neurotrophic factor binding to this receptor
(control). This is a time when rat pups still use a complex has been associated with stimulating ax-
spinal reflex to elicit micturition implying that po- onal growth in vitro (Paratcha et al., 2003). Inter-
lysialic acid may have a role in the plasticity that action of brain-derived neurotrophic factor with
accompanies the transition from spinal to supra- its receptor trkB may be, in part, mediated by the
spinal micturition in rats. presence of polysialic acid-NCAM, as selective
NCAM has recently been shown to interact with enzymatic removal of polysialic acid decreases
brain-derived neurotrophic factor and glial-de- trkB phosphorylation (Vutskits et al., 2001). Be-
rived neurotrophic factor. NCAM, depending on cause both polysialic acid and NCAM have been
the isoform, signals through two main pathways, shown to interact with neurotrophic factors that
the Fyn/FAK pathway or through the fibroblast are upregulated in lumbosacral spinal cord after
growth factor receptor, both of which converge spinal cord injury (Zvarova et al., 2004), polysialic
upstream of the mitogen-activated protein kinase acid-NCAM may play a significant role in the
kinase MEK (Kiryushko et al., 2004). Signaling emergence of the spinal micturition reflex.
via the fibroblast growth factor receptor also in-
duces release of intracellular calcium stores, which
Inhibitors of neuronal outgrowth after injury
has been shown to mediate axonal outgrowth by a
The central nervous system contains several
variety of mechanisms, including by growth asso-
known inhibitors of neuronal sprouting. These
ciated protein-43 dependent phosphorylation and
molecules are expressed to stop neuronal out-
growth and aberrant synapse formation at the end
of development and are often upregulated after
nerve injury. Injury to the central nervous system
causes the formation of a glial scar that over ex-
presses both myelin-dependent and -independent
inhibitors of neuronal extension thereby impeding
neuronal sprouting and regeneration. Myelin-de-
pendent inhibitors of sprouting include Nogo-A,
myelin associated glycoprotein and oligodendro-
cyte myelin glycoprotein (Grados-Munro and
Fournier, 2003) whereas myelin-independent inhi-
bitors of sprouting are members of the proteo-
glycan family including chondroitin sulfate
Fig. 3. Neural cell adhesion molecule (NCAM) in neonatal proteoglycans (Bovolenta and Fernaud-Espinosa,
spinal cord is highly polysialated. Illustration shows a western 2000). Important signaling pathways of key mol-
blot of NCAM isoforms in rat L6/S1 spinal cord. Arrowheads ecules that inhibit sprouting will be discussed in
point out the level of three NCAM isoforms (NCAM-120, the section ‘‘Manipulation of neuronal sprouting’’.
NCAM-140, NCAM-180). The level of polysialation can be
inferred by the smearing of the NCAM bands due to variable
polysialic acid (PSA) binding to NCAM. At post-natal day 6 Do interneurons play a role in bladder function after
(P6) rats contain all three isoforms as well as showing smearing
spinal cord injury?
between these bands. Adult rats (control) have weak to no ex-
pression of NCAM-180 and little PSA expression as indicated
by a lack of smearing between bands. (N.D.T. Zinck, V.F. There are many neurons in the lumbosacral spinal
Rafuse, J.W. Downie, unpublished.) cord that respond to bladder afferent stimulation
154

(McMahon and Morrison, 1982; Honda, 1985; It is likely that spinal interneurons undergo
Coonan and Downie, 1999). Some may participate physiological and structural changes after spinal
in ascending transmission of bladder-related ac- cord injury and that these changes are contributing
tivity, either as part of the micturition reflex path- to emergence of micturition after injury. However,
way or in pain-related pathways (McMahon and until precise methods are developed to study these
Morrison, 1982; Milne et al., 1982; Ding et al., interneurons specifically, the contribution of spi-
1994). Also, activation of spinal interneurons is an nal bladder interneurons to bladder function after
important component of efficient micturition both spinal cord injury will remain elusive.
for bladder contraction and sphincter inhibition
(Shefchyk, 2001). These interneuron pools have Repairing the injured spinal cord to improve
not been well localized. However, it appears from bladder function
virus tracing and immediate early gene expression
studies that the dorsal gray commissure and the Cellular implants
region of the parasympathetic preganglionic nu-
cleus are important locations of bladder and Stem cells have been touted as a major aid in the
sphincter-related interneurons (Nadelhaft et al., treatment of many diseases, including Parkinson’s
1992; Nadelhaft and Vera, 1996; Marson, 1997; disease, diabetes and amyotrophic lateral sclerosis.
Grill et al., 1998; Vera and Nadelhaft, 2000). Because stem cells have the ability to differentiate
There is some suggestion of synaptic reorgani- into various tissue types, they also have been tested
zation in the lumbosacral spinal cord after spinal for a role in repairing the damaged spinal cord af-
cord injury in rats (Yu et al., 2003). On the other ter injury. Stem cells injected into severely injured
hand, evidence for change in the pelvic afferent rat spinal cord have shown cellular differentiation,
terminal arbor in the spinal cord is lacking (Kruse resulting in increased axonal regeneration and sig-
et al., 1995). There is evidence for reorganization nificant improvement in motor function below the
of motor neuronal pathways after spinal cord in- site of injury (McDonald et al., 1999). Few studies,
jury to facilitate hind limb locomotion (Grasso however, have addressed the impact of stem cell
et al., 2004). Thus one possibility is that spinal implantation on visceral function after spinal cord
interneuron reorganization may underlie the emer- injury. Injections of neural stem cells in the injured
gence of bladder activity after spinal cord injury. rat spinal cord has improved lower urinary tract
A second possibility is that emergence of blad- function by increasing voiding efficiency, although
der function after spinal cord injury is a matter of has not improved bladder–sphincter dyssynergia
developing access to an existing spinal circuit sub- (Mitsui et al., 2003).
serving micturition. The existence of such a circuit, Implantation of nerve grafts with cells genetically
analogous to the spinal pattern generator for lo- engineered to secrete growth factors has become a
comotion, is implied by the finding that micturit- common animal model to treat spinal cord injury.
ion can be evoked by perineal or urethral nerve The ability of the peripheral nervous system to
stimulation in some circumstances (Shefchyk and regenerate after nerve injury is far greater than that
Buss, 1998; Boggs et al., 2004). One problem with of the CNS. Because of this ability of peripheral
this circuit is that it appears not to be activated by nerves to regenerate, nerve grafts implanted in the
bladder distension and thus coordinated micturit- injured spinal cord are often made from peripheral
ion never emerges in spinal cord-injured cats. It is nerve tissue or peripheral nervous system specific
possible that sprouting of urethral or perineal af- cell types. For example, implantation of a Schwann
ferents is hindering the use of this circuit at later cell graft, secreting trkB activating neurotrophins,
time points post spinal cord injury. Thus encour- brain-derived neurotrophic factor and neurotro-
aging the sprouting of bladder afferents to target phin-3, into a severely contused rat spinal cord has
the spinal micturition circuit while suppressing the provided restoration of bladder function (Sakamoto
perineal inputs to the circuit may be an appropri- et al., 2002). Not every growth factor will have
ate approach to restoration of function. positive effects on bladder function. For example,
155

implantation of peripheral nerve grafts secreting cord injury, no definitive studies have been con-
acidic fibroblast growth factor in a patient with a ducted. The last decade has provided a wealth of
complete hemisection of the thoracic spinal cord knowledge with respect to specific extracellular
improved motor recovery although bladder function signaling pathways involved in neurite outgrowth
was unchanged (Cheng et al., 2004). Because differ- and extension. With a molecular knowledge of key
ent motor and visceral systems are reliant on specific elements responsible for axon regeneration, new
growth factors, it is likely that if we are to signif- targets have been uncovered for experimental ma-
icantly improve micturition by the addition of nipulation. The function of the lower urinary tract
neuronal growth factors, then we need first to could benefit from such investigation, providing
establish which trophic factors are pertinent to the new avenues of research that have the potential to
micturition reflex. answer questions regarding the neuronal plasticity
Olfactory ensheathing cells, as the name implies, of the micturition reflex after spinal cord injury.
ensheath fibers as they travel from the olfactory Neurotrophic factors, especially nerve growth fac-
bulb to the peripheral nervous system. These cells tor, have been shown to play a major role in bladder
are thought to provide a permissive environment function after spinal cord injury, thereby making
from the CNS to the peripheral nervous system by possible methods for interfering with signaling path-
remaining in contact with the olfactory nerve and ways of these molecules invaluable. Intrathecal ad-
inhibiting astrocytes from blocking entry into the ministration of immuno-neutralizing nerve growth
CNS. Injection of olfactory ensheathing cells into factor antibody into the L6/S1 spinal cord segments
deep dorsal horn regions after transection of dorsal results in a marked decrease in the number of non-
roots promotes afferent reentry into the dorsal spi- voiding bladder contractions, as well as increasing
nal cord and also facilitates the emergence of blad- voiding efficiency in rats with complete spinal cord
der function (Pascual et al., 2002). Although injury (Fig. 4). Electromyographic recording of
deafferentation is not a model of spinal cord inju- external urethral sphincter muscles in spinal rats
ry, these findings provide justification for further treated with this antibody also shows a decrease in
investigation on the effects that olfactory ensheath- detrusor–sphincter dyssynergia (Seki et al., 2004).
ing cells may have on spinally mediated bladder These results are very similar to the effects of
function. Recently, many investigators studying subcutaneous administration of capsaicin to spinal
bladder incontinence are turning to gene therapy cord-injured rats. Because capsaicin is selectively
as a possible way to treat urological dysfunction. neurotoxic to C-fibers, upregulation of nerve growth
This technique involves the use of a viral vector that factor is likely to occur in small diameter primary
encodes a particular gene of interest and is injected afferents (Cheng and de Groat, 2004).
into the bladder wall. Transport of the vector to the Saporin-tagged IB4 injected intrathecally at the
bladder afferent neurons in dorsal root ganglia as level of L6/S1 in a recent experiment caused selec-
well as to the spinal cord is accomplished due to tive reduction of IB4-positive (non-peptidergic) af-
retrograde virion transmission. This technique has ferents in L6 and a decrease in bladder overactivity
previously been used to transport molecules like in response to inflammation (Vulchanova et al.,
nerve growth factor as well as pre-proenkephalin to 2001; Nishiguchi et al., 2004). It was not clear
bladder primary afferents to treat bladder afferent which visceral population of IB4-positive afferents
neuropathy associated with diabetes (Goins et al., were destroyed after application of the cytotoxin
2001; Yoshimura et al., 2001). This may also be an saporin in this experiment, so the role of bladder
interesting avenue of research for treating bladder specific non-peptidergic fibers in detrusor overacti-
dysfunction after spinal cord injury. vity after inflammation is unclear. Injections of
saporin-tagged IB4 in the bladder wall or pelvic
Manipulation of neuronal sprouting nerve would aid in clarifying these results. Recent-
ly, saporin-tagged IB4 that had been injected into
Although it has been implied that neurons con- the sciatic nerve was detected in the spinal cord
trolling the micturition reflex sprout after spinal indicating that these more selective methods are
156

Fig. 4. Antagonism of nerve growth factor (NGF) normalizes aberrant bladder function after spinal cord injury. Bladder function is
abnormal 10 days after a mid-thoracic spinal cord injury and the administration of vehicle i.t. (A). However, 14 days after i.t.
administration of 10 ug NGF-Ab the non-voiding contractions are fewer and smaller (B). Arrows indicate voiding episodes. The
micturition pattern in panel B closely resembles that seen in uninjured rats. Infusion of saline ¼ 0.04 ml/min. From Seki et al. (2002)
with permission.

possible (Vulchanova et al., 2001; Tarpley et al., are elevated to a point at which myelin-associated
2004). The role that IB4 positive bladder afferents glycoprotein signaling is overridden. When cAMP
play in bladder overactivity associated with spinal levels decline, myelin-associated glycoprotein is
cord injury, however, has not yet been investigated. then able to promote growth cone collapse, ending
As described earlier, the peripheral nervous axonal outgrowth during development. Exogenous
system has a far greater ability to regenerate after elevation of cAMP (Neumann et al., 2002) or
neuronal assault than the CNS. The most studied vaccination against myelin (Huang et al., 1999) in
of all myelin-associated inhibitors is Nogo. The models of spinal cord injury has improved neuronal
receptor for Nogo, NgR, forms a receptor complex regenerative ability as well as increased functional
with the low-affinity neurotrophin receptor p75. recovery. In addition, inhibiting protein kinase A, a
Myelin-associated glycoprotein and oligodendro- downstream regulator of cAMP, decreases the
cyte myelin glycoprotein, along with Nogo, ability of peripheral nerve grafts to extend axons
mediate their inhibitory actions on neuronal out- into host spinal cord (Cai et al., 2001).
growth through the NgR-p75 receptor complex Signaling through NgR-p75 activates other
(Wang et al., 2002). The antagonist to the Nogo downstream effectors important in signaling inhi-
receptor, NEP (1–40), and exogenous addition of bition of neuronal outgrowth. A small group of
neurotrophins (Cai et al., 1999) have both signi- GTPases, known as Rho GTPases, are activated in
ficantly improved axon re-growth after nerve injury response to Nogo- and myelin-associated glyco-
(GrandPre et al., 2002). p75 receptor knockouts protein binding to the p75 receptor. It is becoming
have shown similar results (Wang et al., 2002). An increasingly evident that the low-affinity receptor
overview of signaling pathways that are critical in for all neurotrophins, p75, has a major role in reg-
regulating neuronal extension is presented in Fig. 5. ulation of neuronal extension and survival. Con-
During development, myelin-associated glyco- version of RhoGDP to its active GTP-bound state is
protein appears to play a key role in the switch thought to be mediated by a number of GTPase-
from neurite extension to arrest. Cyclic adenosine activating proteins, guanine nucleotide exchange
monophosphate (cAMP) levels appear to be critical factors, and guanine nucleotide dissociation inhib-
in this switch. During development, cAMP levels itor (GDI). Rho-GDI interacts directly with
157

Fig. 5. Major signaling pathways of myelin associated inhibitors of neuronal outgrowth. Myelin associated glycoprotein (MAG),
Nogo and oligodendrocyte myelin glycoprotein signal through the Nogo receptor (NgR)-p75 receptor complex to activate Rho and
Rho-kinase. Neurotrophic factors (NT) stimulate neuronal regeneration through increases in cAMP levels. MAG signaling is able to
inhibit cAMP induced regeneration via signaling through an inhibitory G protein. Arrest of axonal remodeling during development is
likely mediated by MAG signaling. From Grados-Munro and Fournier (2003) with permission.

the intracellular domain of the p75 receptor as well as the myosin binding subunit of myosin
(Yamashita and Tohyama, 2003) and thus is light chain phosphatase induces actomyosin as-
thought to play a key role in Rho signaling. In its sembly and growth cone collapse. Inhibition of
active state, RhoGTP acts to stabilize actin polym- Rho-kinase signaling with the specific Rho-kinase
erization, thereby facilitating growth cone collapse inhibitor, Y–27632 results in increased axonal ex-
and inhibiting axonal sprouting through activation tension and growth cone formation within dorsal
of several effector molecules (Bishop and Hall, root ganglia neurons (Borisoff et al., 2003).
2000). One of the most studied Rho effector mol- Most research directed at restoring function
ecules involved in actin reorganization is a serine after spinal cord injury seeks to enhance survival
threonine kinase known as Rho-kinase. Rho- and extension of injured and uninjured neurons.
kinase regulates axonal outgrowth via two main Although neuronal sprouting and synapse creation
pathways. Phosphorylation of myosin light chain is essential for reestablishing damaged connections
158

between neurons, it is critical that new synapses undergo. Some of these changes may play a role in
are functionally relevant. Increasing synaptogene- the emergence of bladder function after spinal cord
sis has the potential to do harm as well as good as injury. However, uncontrolled plasticity does not
seen in neuropathic pain and autonomic dysre- appear to provide functional outcomes that are
flexia. Upregulation of nerve growth factor has favorable to a spinal cord-injured person. Great
been linked to bladder hyperactivity after spinal advances have been made in unraveling the mo-
cord injury (Seki et al., 2002). Thus, if plasticity is lecular mechanisms of neuronal plasticity. These
mediating emergence of micturition after spinal findings have already contributed to a better
cord injury, then perhaps the goal for intervention understanding of how bladder afferents respond
in this process should be to maximize functional to spinal cord injury and how bladder function
outcome by selectively targeting sprouting to pro- might be dictated by that response. Because
duce efficient micturiton and avoid unwanted side bladder activity after spinal cord injury is
effects. Several molecules in the CNS (e.g. netrin, dysfunctional, specifically targeting bladder affer-
semaphorins and slits) are implicated in mecha- ent outgrowth may serve as a potential therapy for
nisms in guidance of growth cones to appropriate restoring ‘‘normal’’ bladder voiding and conti-
targets and formation of appropriate synapses (for nence. Although this presents a difficult task,
review see Dickson and Senti, 2002) However, research will be facilitated by the plethora of new
specific targeting of sprouting neurons to produce molecular techniques and the creation of knockout
efficient, non-dyssynergic function after spinal and transgenic animals. In conclusion, plasticity of
cord injury is currently not feasible because the bladder primary afferents after spinal cord injury is
intricate molecular interactions involved are not associated with bladder dysfunction, and therefore
well understood. The most practical strategy, for this dysfunction may be considered to be an
the present, may be to reduce neuronal sprouting unwanted consequence of plasticity. We suggest
in lumbosacral spinal cord in an attempt to alle- that, with more research, uncontrolled plasticity
viate bladder hyperreflexia. may be directed toward the more positive outcome
Current technology has provided ways to ma- of normal bladder function after spinal cord injury.
nipulate the level of polysialic acid-neural cell ad-
hesion molecule thereby creating possible methods References
to explore sprouting responses of bladder neurons
after spinal cord injury. NCAM knockout mice Apfel, S.C., Wright, D.E., Wiideman, A.M., Dormia, C., Snider,
W.D. and Kessler, J.A. (1996) Nerve growth factor regulates
have a nervous system completely stripped of the expression of brain-derived neurotrophic factor mRNA in
NCAM and polysialic acid (Cremer et al., 1994), the peripheral nervous system. Mol. Cell. Neurosci., 7: 134–142.
thereby having severe consequences in synapto- Aubert, J., Ridet, J., Schachner, M., Rougon, G. and Gage, F.
genesis-driven processes like long term potentiat- (1998) Expression of L1 and PSA during sprouting and re-
generation in adult hippocampal formation. J. Comp. Ne-
ion (Muller et al., 2000; Bukalo et al., 2004). In
urol., 399: 1–19.
addition, a specific enzyme endoneuraminidase has Barrington, F. (1915) The nervous mechanism of micturition.
been used to cleave the polysialic acid moiety from Quart. J. Exp. Physiol., 8: 33–71.
NCAM. In vivo injections of endoneuraminidase Belyantseva, I.A. and Lewin, G.R. (1999) Stability and plasticity
delays collateral sprouting and decreases branching of primary afferent projections following nerve regeneration
of corticospinal axons in rat pups (Daston et al., and central degeneration. Eur. J. Neurosci., 11: 457–468.
Bishop, A.L. and Hall, A. (2000) Rho GTPases and their ef-
1996), as well as decreases axonal plasticity in the fector proteins. Biochem. J., 348(Part 2): 241–255.
arcuate nucleus of adult rats (Hoyk et al., 2001). Boggs, J.W., Wenzel, B.J., Gustafson, K.J., and Grill, W.M.
(2004) A spinal micturition reflex mediated by afferents in the
Can neuronal plasticity play a part in restoring deep perineal nerve. J. Neurophysiol., in press.
Bonfanti, L., Merighi, A. and Theodosis, D.T. (1996) Dorsal
bladder function after spinal cord injury?
rhizotomy induces transient expression of the highly sialylated
isoform of the neural cell adhesion molecule in neurons and
This chapter has outlined several plastic changes astrocytes of the adult rat spinal cord. Neuroscience, 74:
that neurons involved in the micturition reflex may 619–623.
159

Borisoff, J.F., Chan, C.C., Hiebert, G.W., Oschipok, L., Daston, M.M., Bastmeyer, M., Rutishauser, U. and O’Leary,
Robertson, G.S., Zamboni, R., Steeves, J.D. and Tetzlaff, D.D. (1996) Spatially restricted increase in polysialic acid
W. (2003) Suppression of Rho-kinase activity promotes enhances corticospinal axon branching related to target rec-
axonal growth on inhibitory CNS substrates. Mol. Cell. ognition and innervation. J. Neurosci., 16: 5488–5497.
Neurosci., 22: 405–416. de Groat, W. (2002) Plasticity of bladder reflex pathways dur-
Bovolenta, P. and Fernaud-Espinosa, I. (2000) Nervous system ing postnatal development. Physiol. Behav., 77: 689–692.
proteoglycans as modulators of neurite outgrowth. Prog. de Groat, W., Kawatani, M., Hisamitsu, T., Cheng, C., Ma, C.,
Neurobiol., 61: 113–132. Thor, K., Steers, W. and Roppolo, J. (1990) Mechanisms
Bowles, W.R., Sabino, M., Harding-Rose, C. and Hargreaves, underlying the recovery of urinary bladder function following
K.M. (2004) Nerve growth factor treatment enhances release spinal cord injury. J. Auton. Nerv. Sys., 30: S71–S78.
of immunoreactive calcitonin gene-related peptide but not de Groat, W.C., Araki, I., Vizzard, M.A., Yoshiyama, M.,
substance P from spinal dorsal horn slices in rats. Neurosci. Yoshimura, N., Sugaya, K., Tai, C. and Roppolo, J.R. (1998)
Lett., 363: 239–242. Developmental and injury induced plasticity in the micturit-
Bruses, J.L., Chauvet, N., Rubio, M.E. and Rutishauser, U. ion reflex pathway. Behav. Brain Res., 92: 127–140.
(2002) Polysialic acid and the formation of oculomotor Dickson, B.J. and Senti, K.A. (2002) Axon guidance: growth
synapses on chick ciliary neurons. J. Comp. Neurol., 446: cones make an unexpected turn. Curr. Biol., 12: R218–R220.
244–256. Ding, Y.-Q., Qin, B.-Z., Li, J.-S. and Mizuno, N. (1994) In-
Bukalo, O., Fentrop, N., Lee, A.Y., Salmen, B., Law, J.W., duction of c-fos-like protein in the spinoparabrachial tract-
Wotjak, C.T., Schweizer, M., Dityatev, A. and Schachner, neurons locating within the sacral parasympathetic nucleus in
M. (2004) Conditional ablation of the neural cell adhesion the rat. Brain Res., 659: 283–286.
molecule reduces precision of spatial learning, long-term po- Drescher, U., Faissner, A., Klein, R., Rathjen, F.G. and
tentiation, and depression in the CA1 subfield of mouse hip- Sturmer, C. (1997) Axonal growth and pathfinding: from
pocampus. J. Neurosci., 24: 1565–1577. phenomena to molecules. Cell. Tissue Res., 290: 187–188.
Cai, D., Qiu, J., Cao, Z., McAtee, M., Bregman, B.S. and Franz, C., Rutishauser, U. and Rafuse, V. (2005) Polysialyated
Filbin, M.T. (2001) Neuronal cyclic AMP controls the de- neural cell adhesion molecule (PSA-NCAM) is necessary for
velopmental loss in ability of axons to regenerate. J. Neuro- selective targeting of regenerating motor neurons. J. Neuro-
sci., 21: 4731–4739. sci., 25: 2081–2091.
Cai, D., Shen, Y., De Bellard, M., Tang, S. and Filbin, M.T. Gallo, G. and Letourneau, P.C. (2004) Regulation of growth cone
(1999) Prior exposure to neurotrophins blocks inhibition of actin filaments by guidance cues. J. Neurobiol., 58: 92–102.
axonal regeneration by MAG and myelin via a cAMP-de- Goins, W.F., Yoshimura, N., Phelan, M.W., Yokoyama, T.,
pendent mechanism. Neuron, 22: 89–101. Fraser, M.O., Ozawa, H., Bennett, N.J., de Groat, W.C.,
Cheng, C.L. and de Groat, W.C. (2004) The role of capsaicin- Glorioso, J.C. and Chancellor, M.B. (2001) Herpes simplex
sensitive afferent fibers in the lower urinary tract dysfunction virus mediated nerve growth factor expression in bladder and
induced by chronic spinal cord injury in rats. Exp. Neurol., afferent neurons: potential treatment for diabetic bladder
187: 445–454. dysfunction. J. Urol., 165: 1748–1754.
Cheng, H., Liao, K.K., Liao, S.F., Chuang, T.Y. and Gordon-Weeks, P.R. (2004) Microtubules and growth cone
Shih, Y.H. (2004) Spinal cord repair with acidic fibroblast function. J. Neurobiol., 58: 70–83.
growth factor as a treatment for a patient with chronic par- Grados-Munro, E.M. and Fournier, A.E. (2003) Myelin-asso-
aplegia. Spine, 29: E284–E288. ciated inhibitors of axon regeneration. J. Neurosci. Res., 74:
Christensen, M.D. and Hulsebosch, C.E. (1997) Spinal cord 479–485.
injury and anti-NGF treatment results in changes in CGRP GrandPre, T., Li, S. and Strittmatter, S.M. (2002) Nogo-66
density and distribution in the dorsal horn in the rat. Exp. receptor antagonist peptide promotes axonal regeneration.
Neurol., 147: 463–475. Nature, 417: 547–551.
Coonan, E.M., Downie, J.W. and Du, H.-J. (1999) Sacral spi- Grasso, R., Ivanenko, Y.P., Zago, M., Molinari, M.,
nal cord neurons responsive to bladder pelvic and perineal Scivoletto, G., Castellano, V., Macellari, V. and Lacquaniti,
inputs in cats. Neurosci. Lett., 260: 137–140. F. (2004) Distributed plasticity of locomotor pattern gener-
Cremer, H., Lange, R., Christoph, A., Plomann, M., Vopper, ators in spinal cord injured patients. Brain, 127: 1019–1034.
G., Roes, J., Brown, R., Baldwin, S., Kraemer, P. and Scheff, Grill, W., Wang, B., Hadziefendie, S. and Haxhiu, M. (1998)
S. (1994) Inactivation of the N-CAM gene in mice results in Identification of the spinal neural network involved in coordi-
size reduction of the olfactory bulb and deficits in spatial nation of micturition in the male cat. Brain Res., 796: 150–160.
learning. Nature, 367: 455–459. Honda, C.N. (1985) Visceral and somatic afferent convergence
Crowley, C., Spencer, S.D., Nishimura, M.C., Chen, K.S., onto neurons near the central canal in the sacral spinal cord
Pitts-Meek, S., Armanini, M.P., Ling, L.H., MacMahon, of the cat. J. Neurophysiol., 53: 1059–1078.
S.B., Shelton, D.L. and Levinson, A.D. (1994) Mice lacking Hoyk, Z., Parducz, A. and Theodosis, D.T. (2001) The highly
nerve growth factor display perinatal loss of sensory and sialylated isoform of the neural cell adhesion molecule is re-
sympathetic neurons yet develop basal forebrain cholinergic quired for estradiol-induced morphological synaptic plasticity
neurons. Cell, 76: 1001–1011. in the adult arcuate nucleus. Eur. J. Neurosci., 13: 649–656.
160

Huang, D.W., McKerracher, L., Braun, P.E. and David, S. (1999) McMahon, S. and Morrison, J. (1982) Two groups of spinal
A therapeutic vaccine approach to stimulate axon regeneration interneurons that respond to stimulation of the viscera of the
in the adult mammalian spinal cord. Neuron, 24: 639–647. cat. J. Physiol., 322: 21–34.
Janig, W. and Morrison, J.F. (1986) Functional properties of McMahon, S.B. and Morrison, J.F.B. (1982) Spinal neurones
spinal visceral afferents supplying abdominal and pelvic or- with long projections activated from the abdominal viscera of
gans, with special emphasis on visceral nociception. Prog. the cat. J. Physiol., 322: 1–20.
Brain Res., 67: 87–114. Milne, R.J., Foreman, R.D. and Willis, W.D. (1982) Responses
Kakizaki, H. and de Groat, W.C. (1997) Reorganization of of primate spinothalamic neurons located in the sacral
somato-urethral reflexes following spinal cord injury in the intermediomedial gray (Stilling’s nucleus) to proprioceptive
rat. J. Urol., 158: 1562–1567. input from the tail. Brain Res., 234: 227–236.
Kiryushko, D., Berezin, V. and Bock, E. (2004) Regulators of Mitsui, T., Kakizaki, H., Tanaka, H., Shibata, T., Matsuoka, I.
neurite outgrowth: role of cell adhesion molecules. Ann. NY and Koyanagi, T. (2003) Immortalized neural stem cells
Acad. Sci., 1014: 140–154. transplanted into the injured spinal cord promote recovery of
Kiss, J.Z. and Muller, D. (2001) Contribution of the neural cell voiding function in the rat. J. Urol., 170: 1421–1425.
adhesion molecule to neuronal and synaptic plasticity. Rev. Monnier, P.P., Beck, S.G., Bolz, J. and Henke-Fahle, S. (2001)
Neurosci., 12: 297–310. The polysialic acid moiety of the neural cell adhesion
Krenz, N.R., Meakin, S.O., Krassioukov, A.V. and Weaver, molecule is involved in intraretinal guidance of retinal
L.C. (1999) Neutralizing intraspinal nerve growth factor ganglion cell axons. Dev. Biol., 229: 1–14.
blocks autonomic dysreflexia caused by spinal cord injury. Morgan, C., Nadelhaft, I. and de Groat, W.C. (1981) The dis-
J. Neurosci., 19: 7405–7414. tribution of visceral primary afferents from the pelvic nerve
Krenz, N.R. and Weaver, L.C. (2000) Nerve growth factor in to Lissauer’s tract and the spinal gray matter and its rela-
glia and inflammatory cells of the injured rat spinal cord. tionship to the sacral parasympathetic nucleus. J. Comp.
J. Neurochem., 74: 730–739. Neurol., 201: 415–440.
Kruse, M.N., Belton, A.L. and de Groat, W.C. (1993) Changes Muller, D., Djebbara-Hannas, Z., Jourdain, P., Vutskits, L.,
in bladder and externalurethral sphincter function after spi- Durbec, P., Rougon, G. and Kiss, J.Z. (2000) Brain-derived
nal cord injury in the rat. Am. J. Physiol., 264: R1157–R1163. neurotrophic factor restores long-term potentiation in poly-
Kruse, M.N., Bray, L.A. and de Groat, W.C. (1995) Influence sialic acid-neural cell adhesion molecule-deficient hippocam-
of spinal cord injury on the morphology of bladder afferent pus. Proc. Natl. Acad. Sci. USA, 97: 4315–4320.
and efferent neurons. J. Auton. Nerv. Syst., 54: 215–224. Nadelhaft, I. and Booth, A.M. (1984) The location and
Lamb, K., Gebhart, G.F. and Bielefeldt, K. (2004) Increased morphology of preganglionic neurons and the distribution
nerve growth factor expression triggers bladder overactivity. of visceral afferents from the rat pelvic nerve: a horseradish
J. Pain., 5: 150–156. peroxidase study. J. Comp. Neurol., 226: 238–245.
Li, L. and Zhou, X.F. (2001) Pericellular Griffonia simplicifolia Nadelhaft, I. and Vera, P.L. (1996) Neurons in the rat brain and
I isolectin B4-binding ring structures in the dorsal root gan- spinal cord labeled after pseudorabies virus injected into the
glia following peripheral nerve injury in rats. J. Comp. external urethral sphincter. J. Comp. Neurol., 375: 502–517.
Neurol., 439: 259–274. Nadelhaft, I., Vera, P.L., Card, J.P. and Miselis, R.R. (1992)
Luo, X.G., Rush, R.A. and Zhou, X.F. (2001) Ultrastructural Central nervous system neurons labelled following the injec-
localization of brain-derived neurotrophic factor in rat pri- tion of pseudorabies virus into the rat urinary bladder.
mary sensory neurons. Neurosci. Res., 39: 377–384. Neurosci. Lett., 143: 271–274.
Maggi, C.A., Santicioli, P. and Meli, A. (1986) Postnatal de- Neumann, S., Bradke, F., Tessier-Lavigne, M. and Basbaum,
velopment of micturition reflex in rats. Am. J. Physiol., 250: A.I. (2002) Regeneration of sensory axons within the injured
R926–R931. spinal cord induced by intraganglionic cAMP elevation.
Maier, D.L., Mani, S., Donovan, S.L., Soppet, D., Tessarollo, Neuron, 34: 885–893.
L., McCasland, J.S. and Meiri, K.F. (1999) Disrupted cor- Nishiguchi, J., Sasaki, K., Seki, S., Chancellor, M.B., Erickson,
tical map and absence of cortical barrels in growth-associated K.A., de Groat, W.C., Kumon, H. and Yoshimura, N. (2004)
protein (GAP)-43 knockout mice. Proc. Natl. Acad. Sci. Effects of isolectin B4-conjugated saporin, a targeting cyto-
USA, 96: 9397–9402. toxin, on bladder overactivity induced by bladder irritation.
Marson, L. (1997) Identification of central nervous system neu- Eur. J. Neurosci., 20: 474–482.
rons that innervate the bladder body, bladder base, or ex- Obata, K., Yamanaka, H., Dai, Y., Tachibana, T., Fukuoka,
ternal urethral sphincter of female rats: a transneuronal T., Tokunaga, A., Yoshikawa, H. and Noguchi, K. (2003)
tracing study using pseudorabies virus. J. Comp. Neurol., Differential activation of extracellular signal-regulated pro-
389: 584–602. tein kinase in primary afferent neurons regulates brain-de-
McDonald, J.W., Liu, X.Z., Qu, Y., Liu, S., Mickey, S.K., rived neurotrophic factor expression after peripheral
Turetsky, D., Gottlieb, D.I. and Choi, D.W. (1999) inflammation and nerve injury. J. Neurosci., 23: 4117–4126.
Transplanted embryonic stem cells survive, differentiate and Pannek, J., Sommerfeld, H.J., Botel, U. and Senge, T. (2000)
promote recovery in injured rat spinal cord. Nat. Med., 5: Combined intravesical and oral oxybutynin chloride in adult
1410–1412. patients with spinal cord injury. Urology, 55: 358–362.
161

Paratcha, G., Ledda, F. and Ibanez, C.F. (2003) The neural cell Stucky, C.L. and Lewin, G.R. (1999) Isolectin B(4)-positive and
adhesion molecule NCAM is an alternative signaling recep- -negative nociceptors are functionally distinct. J. Neurosci.,
tor for GDNF family ligands. Cell, 113: 867–879. 19: 6497–6505.
Pascual, J.I., Gudino-Cabrera, G., Insausti, R. and Nieto- Tang, J. and Landmesser, L. (1993) Reduction of intramuscular
Sampedro, M. (2002) Spinal implants of olfactory ensheath- nerve branching and synaptogenesis is correlated with de-
ing cells promote axon regeneration and bladder activity after creased motoneuron survival. J. Neurosci., 13: 3095–3103.
bilateral lumbosacral dorsal rhizotomy in the adult rat. Tarpley, J.W., Kohler, M.G. and Martin, W.J. (2004) The be-
J. Urol., 167: 1522–1526. havioral and neuroanatomical effects of IB(4)-saporin treat-
Qiao, L. and Vizzard, M.A. (2002) Up-regulation of tyrosine ment in rat models of nociceptive and neuropathic pain.
kinase (Trka, Trkb) receptor expression and phosphorylation Brain Res., 1029: 65–76.
in lumbosacral dorsal root ganglia after chronic spinal cord Theodosiou, M., Rush, R.A., Zhou, X.F., Hu, D., Walker, J.S.
(T8-T10) injury. J. Comp. Neurol., 449: 217–230. and Tracey, D.J. (1999) Hyperalgesia due to nerve damage:
Renganathan, M., Cummins, T.R. and Waxman, S.G. (2001) role of nerve growth factor. Pain, 81: 245–255.
Contribution of Na(v)1.8 sodium channels to action potential Thor, K.B., Blais, D.P. and de Groat, W.C. (1989) Behavioral
electrogenesis in DRG neurons. J. Neurophysiol., 86: analysis of the postnatal development of micturition in kit-
629–640. tens. Brain Res. Dev. Brain Res., 46: 137–144.
Renganathan, M., Gelderblom, M., Black, J.A. and Waxman, Vera, P.L. and Nadelhaft, I. (2000) Anatomical evidence for
S.G. (2003) Expression of Nav1.8 sodium channels perturbs two spinal ‘afferent-interneuron-efferent’ reflex pathways in-
the firing patterns of cerebellar Purkinje cells. Brain Res., volved in micturition in the rat: a ‘pelvic nerve’ reflex path-
959: 235–242. way and a ‘sacrolumbar intersegmental’ reflex pathway.
Reynard, J.M., Vass, J., Sullivan, M.E. and Mamas, M. (2003) Brain Res., 883: 107–118.
Sphincterotomy and the treatment of detrusor-sphincter Vizzard, M.A. (2000) Changes in urinary bladder neurotrophic
dyssynergia: current status, future prospects. Spinal Cord, factor mRNA and NGF protein following urinary bladder
41: 1–11. dysfunction. Exp. Neurol., 161: 273–284.
Sakamoto, K., Uvelius, B., Khan, T. and Damaser, M.S. (2002) Vizzard, M.A. and Boyle, M.M. (1999) Increased expression of
Preliminary study of a genetically engineered spinal cord im- growth-associated protein (GAP-43) in lower urinary tract
plant on urinary bladder after experimental spinal cord injury pathways following cyclophosphamide (CYP)-induced cysti-
in rats. J. Rehabil. Res. Dev., 39: 347–357. tis. Brain Res., 844: 174–187.
Schumacher, S., Bross, S., Scheepe, J.R., Alken, P. and June- Vulchanova, L., Olson, T.H., Stone, L.S., Riedl, M.S., Elde, R.
mann, K.P. (1999) Sacral anterior root stimulation and pos- and Honda, C.N. (2001) Cytotoxic targeting of isolectin IB4-
terior rhizotomy in spastic neuropathic bladder. Restor. binding sensory neurons. Neuroscience, 108: 143–155.
Neurol. Neurosci., 14: 195–199. Vutskits, L., Djebbara-Hannas, Z., Zhang, H., Paccaud, J.P.,
Sculptoreanu, A., Yoshimura, N. and de Groat, W. Durbec, P., Rougon, G., Muller, D. and Kiss, J.Z. (2001) PSA-
(2004) KW-7158 [(2S)-(+)-3,3,3-trifluoro-2-hydroxy-2-me- NCAM modulates BDNF-dependent survival and differentia-
thyl-N-(5,5,10-trioxo-4,10-dihydrothieno[3,2-c][1]benzothiepin- tion of cortical neurons. Eur. J. Neurosci,, 13: 1391–1402.
9-yl)propanamide] enhances A-type K+ currents in neurons Wang, H.F., Shortland, P., Park, M.J. and Grant, G. (1998)
of the dorsal root ganglion of the adult rat. J. Pharmacol. Retrograde and transganglionic transport of horseradish
Exp. Ther., 310: 159–168. peroxidase-conjugated cholera toxin B subunit, wheatgerm
Seki, S., Sasaki, K., Fraser, M.O., Igawa, Y., Nishizawa, O., agglutinin and isolectin B4 from Griffonia simplicifolia I in
Chancellor, M.B., de Groat, W.C. and Yoshimura, N. (2002) primary afferent neurons innervating the rat urinary bladder.
Immunoneutralization of nerve growth factor in lumbosacral Neuroscience, 87: 275–288.
spinal cord reduces bladder hyperreflexia in spinal cord in- Wang, K.C., Kim, J.A., Sivasankaran, R., Segal, R. and He, Z.
jured rats. J. Urol., 168: 2269–2274. (2002) P75 interacts with the Nogo receptor as a co-receptor
Seki, S., Sasaki, K., Igawa, Y., Nishizawa, O., Chancellor, for Nogo, MAG and OMgp. Nature, 420: 74–78.
M.B., De Groat, W.C. and Yoshimura, N. (2004) Suppres- Waxman, S.G., Cummins, T.R., Dib-Hajj, S., Fjell, J. and
sion of detrusor-sphincter dyssynergia by immunoneutrali- Black, J.A. (1999) Sodium channels, excitability of primary
zation of nerve growth factor in lumbosacral spinal cord in sensory neurons, and the molecular basis of pain. Muscle
spinal cord injured rats. J. Urol., 171: 478–482. Nerve, 22: 1177–1187.
Shefchyk, S. and Buss, R. (1998) Urethral pudendal afferent- Weld, K.J. and Dmochowski, R.R. (2000) Association of level
evoked bladder and sphincter reflexes in decerabrate and of injury and bladder behavior in patients with post-trau-
acute spinal cats. Neurosci. Lett., 244: 137–140. matic spinal cord injury. Urology, 55: 490–494.
Shefchyk, S.J. (2001) Sacral spinal interneurones and the con- Yamashita, T. and Tohyama, M. (2003) The p75 receptor acts
trol of urinary bladder and urethral striated sphincter muscle as a displacement factor that releases Rho from Rho-GDI.
function. J. Physiol., 533: 57–63. Nat. Neurosci., 6: 461–467.
Shefchyk, S.J. and Buss, R.R. (1998) Urethral pudendal affer- Yoshimura, N. and de Groat, W.C. (1997) Plasticity of Na+
ent-evoked bladder and sphincter reflexes in decerebrate and channels in afferent neurones innervating rat urinary bladder
acute spinal cats. Neurosci. Lett., 244: 137–140. following spinal cord injury. J. Physiol., 503(Part 2): 269–276.
162

Yoshimura, N., Erdman, S.L., Snider, M.W. and de Groat, Yoshiyama, M., Nezu, F.M., Yokoyama, O., de Groat, W.C.
W.C. (1998) Effects of spinal cord injury on neurofilament and Chancellor, M.B. (1999) Changes in micturition after
immunoreactivity and capsaicin sensitivity in rat dorsal root spinal cord injury in conscious rats. Urology, 54: 929–933.
ganglion neurons innervating the urinary bladder. Neurosci- Yu, X., Xu, L., Zhang, X.D. and Cui, F.Z. (2003) Effect of
ence, 83: 633–643. spinal cord injury on urinary bladder spinal neural pathway:
Yoshimura, N., Franks, M.E., Sasaki, K., Goins, W.F., Goss, a retrograde transneuronal tracing study with pseudorabies
J., Yokoyama, T., Fraser, M.O., Seki, S., Fink, J., Glorioso, virus. Urology, 62: 755–759.
J., de Groat, W.C. and Chancellor, M.B. (2001) Gene ther- Zhou, F.Q., Zhong, J. and Snider, W.D. (2003) Extra-
apy of bladder pain with herpes simplex virus (HSV) vectors cellular crosstalk: when GDNF meets N-CAM. Cell,
expressing preproenkephalin (PPE). Urology, 57: 116. 113: 814–815.
Yoshimura, N., Seki, S., Erickson, K.A., Erickson, V.L., Chan- Zvarova, K., Murray, E. and Vizzard, M.A. (2004) Changes in
cellor, M.B. and de Groat, W.C. (2003) Histological and galanin immunoreactivity in rat lumbosacral spinal cord and
electrical properties of rat dorsal root ganglion neurons in- dorsal root ganglia after spinal cord injury. J. Comp.
nervating the lower urinary tract. J. Neurosci., 23: 4355–4361. Neurol., 475: 590–603.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 11

Control of urinary bladder function with devices:


successes and failures

Robert A. Gaunt and Arthur Prochazka

Department of Biomedical Engineering and Center for Neuroscience, University of Alberta, 507 HMRC, Edmonton,
AB T6G 2S2, Canada

Abstract: The management of urinary tract dysfunction is crucial for the health and well-being of people
with spinal cord injury. Devices, specifically catheters, play an important role in the daily regime of bladder
management for most people with spinal cord injury. However, the high incidence of complications as-
sociated with the use of catheters, and the fact that the spinal segments involved in lower urinary tract
control remain intact in most cord-injured people, continue to motivate research into devices that could
harness the nervous system to provide greater control over lower urinary tract function. Mechanical devices
discussed in this review include catheters, artificial urethral sphincters, urethral stents and intraurethral
pumps. Additionally, many attempts to restore control of the lower urinary tract with electrical stimulation
have been made. Stimulation sites have included: inside the bladder, bladder wall, thigh, pelvic floor, dorsal
penile nerve, pelvic nerve, tibial nerve, sacral roots, sacral nerves and spinal cord. Catheters and sacral root
stimulators are two techniques whose efficacy is well established. Some approaches have proven less
successful and others are still in the development stage. Modifications to sacral root stimulation including
posterior root stimulation, anodal blockade and high-frequency blockade as well as new techniques in-
cluding intraspinal microstimulation, urethral afferent stimulation and injectable microstimulators are also
discussed. No single device has yet restored the control and function of the lower urinary tract to the pre-
injury state, but new techniques are bringing this possibility closer to reality.

Introduction spinal cord injury and account for the second


highest number of bed-days for readmitted pa-
People with spinal cord injury face many chal- tients (Savic et al., 2000; Middleton et al., 2004).
lenging medical problems. Inadequate post-injury While management of lower urinary tract dys-
management of lower urinary tract dysfunction function with devices, primarily catheters, has re-
can lead to many complications including renal duced mortality after spinal cord injury, the high
failure. This used to be the leading cause of death incidence of complications is largely due to the
after spinal cord injury, but has dropped to fourth limited success that these devices or other treat-
position in recent decades (Frankel et al., 1998) ment modalities have had in restoring normal
with improved treatment methods (Jamil, 2001). function to the neurogenic bladder. In addition to
However, complications of the genitourinary sys- these clinical considerations, effective management
tem, primarily urinary tract infections, are the of lower urinary tract dysfunction is generally
most common cause of rehospitalization after outranked in its importance to patients only by the
desire for hand function in people with quadriple-
Corresponding author. Tel.: +780-492-3783; gia and sex function in people with paraplegia
Fax: +780-495-1617; E-mail: arthur.prochazka@ualberta.ca (Anderson, 2004). These factors provide an

DOI: 10.1016/S0079-6123(05)52011-9 163


164

impetus to develop improved methods of manag- co-contractions of the external urethral sphincter.
ing lower urinary tract dysfunction after spinal This combination, termed detrusor–sphincter dys-
cord injury. synergia (Andersen and Bradley, 1976; Blaivas et al.,
1981), leads to incontinence, inefficient voiding
with high residual volumes and high intravesical
Lower urinary tract control pressures which in turn leads to ureteric reflux and
upper urinary tract deterioration.
The lower urinary tract has two functions: storing
urine (continence) and voiding urine (micturition).
The lower urinary tract is innervated by the so-
matic nervous system and both the sympathetic Why devices?
and parasympathetic branches of the autonomic
nervous system. Efferent parasympathetic inner- Any treatment for lower urinary tract dysfunction
vation of the detrusor, the muscular layer of the after spinal cord injury should create a bladder
bladder, arises from preganglionic neurons in the capable of storing large volumes of urine at low
sacral (S) parasympathetic nucleus in spinal seg- pressure, prevent incontinent episodes and allow
ments S2–S4. The preganglionic neurons send ax- periodic evacuation of urine at low pressure. Sur-
ons via the pelvic nerve to the pelvic plexus where gical treatments, such as bladder augmentation
they synapse with ganglionic neurons. Afferent in- using a section of intestine, ameliorate the problem
nervation of the bladder is also primarily via the of hyperreflexia and low storage volume (Hollander
pelvic nerve. Efferent somatic innervation of the and Diokno, 1993), while sphincterotomies
external urethral sphincter arises from motoneu- (cutting into the external urethral sphincter) im-
rons in Onuf’s nucleus in spinal segments S1–S3. prove detrusor–sphincter dyssynergia (Reynard
These efferent axons as well as the afferents of the et al., 2003). Anticholinergic medications are fre-
external urethral sphincter and urethra travel via quently used to relax the hyperreflexive bladder
the pudendal nerve. As the bladder fills during the but have undesirable side effects including a dry
storage phase, stretch-sensitive mechanoreceptors mouth and blurred vision (Wein, 1998). These
in the bladder wall transmit a sense of fullness to treatments address the symptoms of the ne-
both spinal and supraspinal centers. Once the urogenic bladder so that storage and evacuation
decision to void is reached, the external urethral of urine is achieved without upper urinary tract
sphincter is voluntarily relaxed and parasympa- damage, but they do not address the fundamental
thetic activity causes detrusor contractions. This loss of control associated with spinal cord injury.
synergistic activity, coordinated by the pontine mi- Devices present attractive alternatives to the
cturition center, results in micturition (Barrington, management of lower urinary tract dysfunction
1921, 1925). More details on the anatomy and after spinal cord injury as they attempt, at least
physiology of the lower urinary tract can be found partly, to restore the control of the neurogenic
in de Groat (1993) and de Groat et al. (2001). bladder. Additionally, while devices are locally in-
After spinal cord injury, supraspinal coordina- vasive to varying degrees, they do not generally
tion from the pontine micturition center is lost cause systemic complications as do pharmacolog-
leading to lower urinary tract dysfunction. Sacral ical treatments. Surgical procedures such as the
spinal cord or cauda equina lesions generally lead ones described above are usually irreversible and
to an areflexive bladder and sphincter paralysis. subsequently limit patients to a specified course of
Suprasacral lesions, however, spare sacral spinal treatment while possibly excluding new tech-
reflexes, and after a period of shock reflexive blad- niques. The devices described in this review, and
der contractions often occur at low bladder vol- those under development do not generally cause
umes. This condition, called detrusor hyperreflexia irreversible changes and therefore do not prevent
or neurogenic detrusor overactivity (reviewed in patients from taking advantage of improved treat-
Yoshimura, 1999), is often accompanied by reflexive ments in the future.
165

Many review articles have been published that (Comarr, 1972; Lapides et al., 1972), and this
focus on devices for bladder control (Schmidt, technique, along with generally improved medical
1983; Talalla et al., 1987; Lee, 1997; Rijkhoff et al., care, has caused urinary tract dysfunction to fall
1997b; Grill et al., 2001; Groen and Bosch, 2001; from the primary cause of death (22%) for pa-
Jamil, 2001; Jezernik et al., 2002; Van Kerrebroeck, tients injured between 1943 and 1972 to the fourth
2002; Middleton and Keast, 2004; Rijkhoff, 2004b; most common cause of death (9%) for patients
van Balken et al., 2004), so no attempt will be injured between 1973 and 1990 (Frankel et al.,
made to provide detailed descriptions of each of 1998).
these methods here. Rather, we will summarize the Chronically indwelling urethral and suprapubic
methods that have been devised over the years for catheters, condom catheters and clean intermittent
device-based management of the neurogenic blad- catheterization are common forms of catheteriza-
der secondary to spinal cord injury and summarize tion currently used in the management of spinal
current research on those devices and methods cord injury patients. Each method has its own ad-
that are likely to affect the field in the future. Ad- vantages and disadvantages (reviewed in Selzman
ditionally, we will attempt to identify the reasons and Hampel, 1993), but clean intermittent cathe-
that many methods and devices have ultimately terization is the form of bladder management
been unsuccessful, sometimes in spite of good clin- least likely to lead to complications (Weld and
ical results. Finally, we will summarize the prob- Dmochowski, 2000). Clean intermittent catheter-
lems that we feel should be addressed to improve ization is generally the most prescribed form
the effectiveness and adoption of devices in the of bladder management at hospital discharge
management of the neurogenic bladder. Both me- (Cardenas et al., 1995), and although a number
chanical and electrical devices will be described as of reports suggest that there is a trend for some
they have met with different levels of success and people to switch to other methods (Cardenas et al.,
failure and have the potential to offer solutions to 1995; Weld and Dmochowski, 2000), a more re-
a variety of problems faced by people with spinal cent study suggests that this trend may be revers-
cord injury. ing (Hansen et al., 2004). However, only 30% of
cord-injured people using clean intermittent cath-
eterization remain free of urinary tract infections.
Clean intermittent catheterization requires good
Mechanical devices for control of the lower hand function, preventing people with tetraplegia
urinary tract and impaired hand function as well as some people
with paraplegia from performing this procedure
Catheters themselves (Selzman and Hampel, 1993; Dahlberg
et al., 2004).
The use of catheters to manage urinary retention The critical role of catheter technology and
dates back to ancient Egypt (reviewed in Nacey techniques in the management of lower urinary
and Delahunt, 1993). During World War I, up tract dysfunction after spinal cord injury cannot be
to 80% of patients with spinal cord injury died overstated. The simplicity and clinical efficacy of
shortly after injury due to complications arising catheters in increasing life expectancy in people
from the neurogenic bladder (Kennedy, 1946). with cord injury make them arguably the single
However, improved management of the lower most important device for these people. However,
urinary tract using catheters during World War II the high incidence of urinary tract infections and
(Kennedy, 1946) and especially Guttmann’s other complications associated with catheter
technique of sterile intermittent catheterization use presents a continuous burden on patients and
(Guttmann and Frankel, 1966) helped reduce this the medical system. This, and the desire of people
figure significantly. Sterile intermittent catheteriza- with spinal cord injury for improved methods
tion was eventually modified to non-sterile clean in- (Anderson, 2004), is a motivation for new device
termittent catheterization for reasons of practicality development.
166

Artificial sphincters for patients with detrusor–sphincter dyssynergia.


Sphincterotomies are generally irreversible and
The concept of an artificial urethral sphincter was can cause hemorrhage, erectile dysfunction, blad-
first proposed by Foley (1947) to treat urinary in- der neck stenosis or stricture (reviewed in Reynard
continence. The artificial urethral sphincter devel- et al., 2003). Urethral stents are inserted into the
oped by Scott, Bradley and Timm (Scott et al., urethra and mechanically hold the external ure-
1974; Timm et al., 1974) has developed into the thral sphincter open. After sphincterotomy, or im-
commercially available AMS 800 artificial sphinc- plantation of a urethral stent, most cord-injured
ter (American Medical Systems, Minnetonka, people must wear a collection device such as a
MN, USA) (reviewed in Hajivassiliou, 1998). The condom catheter as the continence mechanism of
AMS 800 uses a pump to deflate a cuff placed the urethra is defeated.
around the bladder neck or urethra by transferring Several different urethral stent designs have
fluid to a pressure-regulated reservoir. The cuff re- been tested in various trials including the
inflates automatically over a period of several UroLumes (American Medical Systems, Minne-
minutes. Of reported studies using the AMS 800 tonka, MN, USA) (Chancellor et al., 1999b),
including 2606 subjects, 73% achieved full conti- Memokaths (Doctors & Engineers A/S Ltd.,
nence, 14% experienced device failure, 4.5% ex- Kvistgaard, Denmark) (Low and McRae, 1998;
perienced infections and 11.7% experienced Hamid et al., 2003), Memotherms (Bard Corp.,
urethral erosion from excessive pressure placed Covington, GA, USA) (Juan Garcia et al., 1999)
on the urethra by the cuff (Hajivassiliou, 1998). and Ultraflexs (Boston Scientific Corp., Natick,
Artificial urethral sphincters are primarily used MA, USA) (Chartier-Kastler et al., 2000). The
to treat patients with post-prostatectomy inconti- UroLume, Memotherm and Ultraflex are flexible
nence, but have been successful in managing wire mesh tubes while the Memokath is a helically
incontinence with other etiologies as well (Petrou wound wire. The devices are inserted into the ure-
et al., 2000). It was originally suggested that thra and positioned in the region of the external
detrusor hyperreflexia was a contraindication for urethral sphincter where the wire becomes largely
artificial urethral sphincter implantation as high covered by urothelium over time. The UroLume is
intravesicular pressures may cause deflation of the best studied of these devices and has similar
the pressure-regulated cuff (Scott et al., 1974). results, in terms of urodynamic parameters and
However, artificial urethral sphincters have been incidence of urinary tract infection to sphincter-
implanted in spinal cord injury patients with an otomies, but requires less hospitalization and is
overall success rate of 70% (Light and Scott, 1983), potentially reversible (Chancellor et al., 1999a). A
though device removal due to infections was high 5-year multi-center trial of the UroLume in 160
(24%). Currently, artificial urethral sphincters are cord-injured subjects showed that the treatment
not commonly used to manage incontinence after was successful in 84%, while 15% required ex-
spinal cord injury, but can be useful in people with plantation. Complications such as device migra-
lesions leading to a flaccid bladder and sphincter. tion were most common in the first 3 months
(Chancellor et al., 1999b). Although explantation
of the stent was possible, it has presented a variety
Urethral stents of challenges (Chancellor et al., 1999b; Wilson et al.,
2002). The Memokath was found to be suitable
Urethral stents were first developed to treat ure- for short-term implantation only as most devices
thral strictures, but shortly after, their use in spinal fail within 2 years (Hamid et al., 2003) and com-
cord injury patients with detrusor–sphincter dys- plications including migration, autonomic dysre-
synergia leading to hydronephrosis and vesicoure- flexia and stone formation on the stent can occur
teric reflux was described (Shaw et al., 1990). (Low and McRae, 1998). However, explantation
Urethral stents were proposed as an alternative to of this device is much simpler than the UroLume
sphincterotomies, the primary surgical treatment due to its helical design and thermosensitive
167

material which, when cooled with saline, becomes


soft and uncoils, making this device useful for
acute management of detrusor–sphincter dyssy-
nergia (Hamid et al., 2003).
Urethral stents represent a clinically successful
device for management of detrusor–sphincter
dyssynergia in people with spinal cord injury. Al-
though stents do not restore normal control of the
sphincter, their efficacy, simplicity and potential
reversibility makes them an attractive option for
people who would otherwise receive an irreversible
sphincterotomy (Chancellor et al., 1999b).

Intraurethral pump

In 1997, Nativ et al. (1997) described a device in-


corporating a miniature valve and pump that
could be inserted into the urethra to control both
continence and voiding in women. The In-FlowTM
intraurethral pump (SRS Medical Systems, Inc.,
Billerica, MA, USA) is designed to manage chron-
ic urinary retention caused by an atonic bladder or
urethral dysfunction. The device secures itself in
the urethra by means of flexible fins that open in
the bladder and a flange at the external urethral
meatus (see Fig. 1). The device is controlled by a
remote activator that is placed over the pubic area
and is magnetically coupled to the pump. Once
activated, the turbine actively pumps urine out of Fig. 1. The In-FlowTM intraurethral pump. (A) Photograph
the bladder at a rate of 6–12 ml/s until the bladder showing the unfolded petals that secure the device in the ure-
is empty. The device is easily inserted by a phy- thra and prevent migration. (B) Diagram showing the place-
sician and can be removed by the patients if they ment of the device in the urethra. Adapted from Madjar et al.
(1999) and Schurch et al. (1999).
wish. The device is designed to be replaced every
month, but successful usage to an average of 90
days has been reported, at which time the device
can become fouled by salt deposits (Madjar et al., reported success rates of 50% with average follow-
1999). up times of 3 and 7.6 months respectively. Most of
In a study of 18 women with spinal cord injury those patients that adopted this device for long-
and hyporeflexive bladders, only six continued to term usage were previously dependent on clean
use the device at follow-up (mean 9.6 months) intermittent catheterization and preferred the con-
(Schurch et al., 1999). Discomfort, incontinence, venience of this device. Intraurethral pumps are
urinary tract infections, technical failures, urethral very interesting from a technical viewpoint and
dilation and the possibility of long-term urethral further investigation with clearer indications for
damage were cited as reasons why this device was use, such as complete spinal cord injury, atonic
unsuitable for chronic use. Studies in 60 (Mazouni bladder and previous dependence on clean inter-
et al., 2004) and 92 (Madjar et al., 1999) patients mittent catheterization may improve the success
with voiding dysfunction from various etiologies rate among people with spinal cord injury.
168

Electrical stimulation devices for control of the is below the threshold required to elicit bladder
lower urinary tract contractions directly via stimulation of the efferent
portion of the pelvic nerve or of the detrusor
While mechanical devices are necessarily limited to myocytes themselves. Acute studies in rats and
treating symptoms of the neurogenic bladder, elec- cats have confirmed the hypothesis that in-
trical stimulation techniques allow devices to be travesical electrical stimulation acts by stimulat-
created that can exert control over spared muscles ing stretch-sensitive mechanoreceptors in the wall
and their neural control systems. Electrical cur- of the bladder that reflexively cause contractions
rent, passed between two electrodes, can be used to of the bladder (Ebner et al., 1992).
generate action potentials in surviving neurons in Few reports of intravesical electrical stimulation
the spinal cord or peripheral nerve below the le- studies in people with spinal cord injury exist, but
sion in spinal cord injury patients. These artificial- one dealing specifically with subjects with incom-
ly generated action potentials can lead directly to plete spinal cord injury reported improvements in
muscular contraction or they can modulate the bladder sensation, detrusor contraction and resid-
activity of neuronal networks and reflex pathways ual volumes in almost all subjects (Madersbacher
(termed neuromodulation). et al., 1982). A retrospective study on the effec-
The discussion below of devices and techniques tiveness of intravesical electrical stimulation for
that have been developed to control the lower people with spinal cord injury by the same author
urinary tract is organized by the location of stim- indicated that one third of the subjects experienced
ulation electrodes rather than by the neurophys- improvements in sensation, detrusor contractility
iological mechanisms on which the devices operate and voluntary control. This occurred only in in-
or by their intended function. Five primary loca- dividuals with preserved pain sensation in the
tions can be identified where electrical stimulation S2–S4 dermatomes (Madersbacher, 1990). This
electrodes can be placed: on or in the bladder, on would seem to be the only predictor of the efficacy
the skin, peripheral nerve, sacral roots and in the of this therapy. Additionally, patients require
spinal cord itself. Figure 2 shows the various stim- many hours of treatment before the effectiveness
ulation locations for devices discussed throughout of intravesical electrical stimulation can begin to
this review. be evaluated and the positive results reported by
some investigators (Kaplan, 2000) have not been
repeatable by others (Decter, 2000). While in-
Electrical stimulation of the bladder travesical electrical stimulation has been used to
treat patients with spinal cord injury, recent stud-
Intravesical stimulation ies have focused on children with underactive
Intravesical electrical stimulation was the first bladders (Gladh et al., 2003). Intravesical electrical
attempt at treating bladder dysfunction using elec- stimulation seems ultimately unattractive as a clin-
trical stimulation. In 1878, M.H. Saxtorph de- ical technique to improve micturition in people
scribed a technique in which stimulation between a with spinal cord injury as it only seems to work in
catheter-mounted electrode, passed into the blad- some with incomplete spinal cord injury, requires
der to act as the cathode (see Fig. 2A), and a sup- long treatments before effectiveness can be evalu-
rapubically placed indifferent electrode, was used ated and the results have not been repeatable
to treat urinary retention caused by an underactive among investigators.
bladder (reviewed in Madersbacher, 1990). In-
travesical electrical stimulation is essentially a ne-
uromodulation therapy intended to reinforce the Bladder wall stimulation
weak functioning of existing neural micturition Electrical stimulation of the exterior surface of the
pathways by stimulating mechanoreceptors in the bladder (see Fig. 2B) was first studied in the early
bladder wall to facilitate reflex bladder contrac- 1950s (Boyce et al., 1964). This marked the begin-
tions and improve sensation. Electrical stimulation ning of the development of electrical stimulation
169

Fig. 2. Electrode locations for controlling the lower urinary tract. The locations are numbered primarily by the order in which they are
discussed in the text. The location for a posterior rhizotomy is also indicated. (A) Intravesical, (B) bladder wall, (C) Thigh, (D) pelvic
floor, (E) dorsal penile nerve, (F) tibial nerve, (G) pelvic nerve, (H) intradural sacral anterior root, (I) extradural mixed sacral root, (J)
intradural sacral posterior root, (K) sacral nerve, (L) spinal cord, (M) intraurethral, (N) pudendal nerve, (O) sacrum.

devices to elicit voiding directly, in response to the enough to generate useful bladder contractions
high morbidity and mortality associated with cath- spread to surrounding structures causing co-
eterization (Bradley et al., 1962). Several groups activation of the external urethral sphincter and
developed implanted stimulators inductively pelvic floor musculature. It was noted that the
coupled to external transmitters with variations canine bladder is primarily an abdominal organ,
in the design, placement and number of electrodes whereas the human bladder is a pelvic organ and is
(Bradley et al., 1962; Hald et al., 1967; Stenberg in close proximity to the pelvic floor musculature,
et al., 1967; Susset and Boctor, 1967; Merrill and increasing pelvic floor susceptibility to contraction
Conway, 1974; Magasi and Simon, 1986). Initial by current spread (Bradley et al., 1963).
animal experiments demonstrated that dogs with Because of this problem, experimental and clinical
spinal cord transections were able to void regularly work was then directed toward obtaining sufficient
using the implanted stimulators without requiring contraction of the bladder while limiting current
additional procedures (Bradley et al., 1962, 1963; spread. Tape electrodes and more powerful stimula-
Kantrowitz and Schamaun, 1963). However, re- tors successfully elicited micturition, but infection
sults in spinal cord injury patients implanted with and technical failures prevented evaluation of their
these stimulators were much less successful (Bradley long-term effect (Bradley et al., 1963). Experience
et al., 1963; Hald et al., 1967; Stenberg et al., 1967; with the Avco stimulator, in which individual wires
Susset and Boctor, 1967; Merrill and Conway, were embedded into the bladder wall, were also
1974). The primary reason that these people were hampered by activation of urethral and pelvic floor
unable to void was that stimulation currents high musculature (Hald et al., 1967; Stenberg et al.,
170

1967). Another stimulator design, the Mentor


bladder stimulator, used two helical wire electrodes
sewn into the bladder wall. This was successful in
two of five people with upper motoneuron lesions,
but required subarachnoid injections of phenol
to abolish electrically induced detrusor–sphincter
dyssynergia that otherwise prevented micturition
(Merrill and Conway, 1974). Susset and Boctor
(1967) reported a successful implant that incorpo-
rated eight disc electrodes around the dome of
the bladder in a person with a complete lower
motoneuron lesion. These investigators considered
upper motoneuron lesions to be a contraindication
for implantation of these systems due to the un-
wanted activation of sphincter and pelvic floor mus-
cles. The most successful report of bladder wall
stimulation was made by Magasi and Simon (1986)
in which 29 of 32 subjects with neurogenic bladder
paralysis attained complete voiding with eight disc
electrodes implanted around the bladder (see
Fig. 3). However, the concomitant sphincter acti-
vation reported by most investigators, lead and
electrode breakage, receiver malfunction, bladder
perforation and pain caused failure in most human
studies. With the success of sacral root stimulation
(see below) for restoring micturition in people with
upper motoneuron lesions, and the multiple diffi-
culties in achieving successful clinical results with
bladder wall stimulation, recent work in this area
has focused on people with lower motoneuron
lesions who cannot benefit from sacral root stim-
ulation (Walter et al., 1999).

Transcutaneous electrical stimulation


Fig. 3. The bladder wall stimulator used by Magasi and Simon
Thigh stimulation (1986). (A) The intended positioning of electrodes on the blad-
der. (B) Actual positions of electrodes around the bladder in
In 1986 it was reported that electrical stimulation
one female subject. Reprinted from Magasi and Simon (1986)
through surface electrodes over the thigh muscles with permission from S. Karger AG, Basel.
(see Fig. 2C) could cause changes in the urody-
namic parameters of spinal cord injury patients spasticity in cord-injured people, noted that 16 of
(Wheeler et al., 1986). Stimulation was applied 32 subjects became continent (Shindo and Jones,
through bilateral quadriceps surface electrodes on a 1987) perhaps indicating a suppression of detrusor
daily basis for 4–8 weeks. Some people exhibited hyperreflexia. A more recent study examining uro-
persistent increases in bladder capacity and/or re- dynamic changes in response to thigh muscle stim-
ductions in bladder pressure, while others experi- ulation showed that 8 of 14 subjects, including one
enced the opposite result. Another study, examining person with spinal cord injury and neurogenic de-
hamstring and quadriceps stimulation to reduce trusor overactivity, increased their bladder volumes
171

by 450% (Okada et al., 1998). However, no meth- results in people with spinal cord injury for sup-
ods of identifying those people likely to respond pressing hyperreflexive bladder contractions are
positively to treatment exist. None of these studies mixed (reviewed in Previnaire et al., 1998). Given
noted any adverse side effects from the treatment. the side effects of maximal functional electrical
The effects of electrical stimulation of the thigh stimulation, including physical discomfort in people
muscles on the bladder may be mediated by limb with incomplete spinal cord injury, possibly limiting
afferents known to inhibit bladder contractions to the stimulation current to non-therapeutic levels
prevent leakage during physical activity (Fall and (Previnaire et al., 1998), as well as psychological
Lindstrom, 1991), although other mechanisms discomfort (van Balken et al., 2004), anticholinergic
have been proposed (Okada et al., 1998). Carry- medications are often a more practical method to
over, observed with thigh stimulation, has also manage neurogenic detrusor overactivity.
been observed with other electrical stimulation
techniques (Fall and Lindstrom, 1991), and may
be at least partially explained by mechanisms such Dorsal penile nerve stimulation
as those proposed by Vodovnik (1981). Despite the Stimulation of the dorsal penile nerve or clitoral
simplicity of this approach, efficacy in some people nerve can inhibit detrusor activity. These nerves
and lack of adverse side effects, few studies of form the most superficial branch of the pudendal
stimulation of the thigh muscles have been report- nerve and are therefore easily accessible. Detrusor
ed, and this technique does not appear to be widely inhibition by this means was first demonstrated sci-
used in practice. This is likely because many pa- entifically using mechanical stimulation (penile
tients show no improvement, and those that may squeeze) to suppress ongoing bladder contractions
cannot be identified prior to treatment. Addition- (Kondo et al., 1982). This effect has also been
ally, treatment requires a significant time invest- demonstrated using electrical stimulation with bi-
ment and most people can achieve effective polar surface electrodes placed on the penis
suppression of hyperreflexive bladder contractions (Nakamura and Sakurai, 1984) (see Fig. 2E). Giv-
with anticholinergic medications. en the success of this simple technique in inhibiting
bladder contractions, its potential in treating sub-
jects with detrusor hyperreflexia secondary to spinal
Pelvic floor maximal functional electrical cord injury has been examined by several groups. In
stimulation one study of six spinal cord injury subjects with
On the basis of a previous observation, Moore and complete and incomplete cervical and thoracic le-
Schofield (1967) decided to test the effectiveness of sions, inhibition of detrusor contractions during
electrically induced maximal contraction of the pel- bladder filling was demonstrated in all subjects
vic floor musculature (see Fig. 2D) to treat female (Wheeler et al., 1992). Continuous stimulation at
patients with stress incontinence. Some people re- five pulses per second was sufficient to increase the
ported being cured after a single session and more volume at which reflexive bladder contractions first
reported a reduction in symptoms. Maximal func- occurred during a cystometrogram by an average of
tional electrical stimulation may involve the use of 76% (range 26% to 150%) without side effects.
surface, vaginal, anal, penile, percutaneous or a Continuous stimulation of the genital nerves, how-
combination of such electrodes to stimulate the ever, may pose practical challenges for the design of
pelvic floor musculature and pudendal nerve at the a neuroprosthesis and would preclude measurement
maximum tolerable threshold for subjects. This of bladder activity using peripheral nerve recording
treatment can lead to long-lasting bladder inhibi- techniques (Jezernik et al., 2000). Kirkham et al.
tion in subjects with non-neurogenic bladder (2001) and Dalmose et al. (2003) therefore exam-
overactivity (Fall and Lindstrom, 1991). While ined whether conditional stimulation of the dorsal
maximal functional electrical stimulation is used penile nerve was sufficient to effect clinically useful
in some clinical settings for treating incontinence inhibition of the detrusor. Stimulation lasting 1 min
in many patient groups (Geirsson and Fall, 1997), was initiated by a rise in bladder pressure of 10 cm
172

H2O during a cystometrogram and successfully in- required to cause detrusor inhibition with bipolar
hibited bladder contractions while increasing blad- percutaneous anal sphincter stimulation could be
der capacity by 144% (7127%) in all six spinal reduced by changing the cathode to a surface elec-
cord injury subjects studied (Kirkham et al., 2001). trode positioned over the posterior tibial nerve
The effects of conditional stimulation on detrusor (McGuire et al., 1983). In the same study, similar
inhibition are robust among both male (Kirkham results were observed with percutaneous tibial
et al., 2001; Dalmose et al., 2003) and female nerve stimulation alone (see Fig. 2F). This target
(Dalmose et al., 2003) patients with a wide range of was chosen as it is the acupuncture point used to
injury levels. In addition to inhibiting hyperreflexive inhibit bladder contractions in Chinese medicine.
bladder contractions, dorsal penile nerve stimula- This technique was successful in improving conti-
tion can reduce blood pressure in people with high nence in 19 of 22 subjects, including four with
level spinal cord injuries (Lee et al., 2003). This may spinal cord injury, although bladder contractions
reduce the risks associated with autonomic dysre- returned immediately once stimulation ceased.
flexia often triggered by a full bladder or bowel. More recent work with tibial nerve stimulation
Dorsal penile nerve stimulation is an active area includes evaluation of the commercially available
of neuroprosthesis development due to the relative Urgent PC device (CystoMedix, Andover, MN,
simplicity of the technique and its reliability and USA), formerly the Urosurge SANS device given
efficacy in people with spinal cord injury. Two re- FDA approval in 2000 (Govier et al., 2001;
cent reports describe attempts at moving dorsal van Balken et al., 2001; Vandoninck et al., 2003).
penile nerve stimulation from the laboratory to Encouraging results were reported in patients with
clinical use. Lee and Creasey (2002) describe the overactive bladders although no spinal cord injury
application of a surface stimulation system to a subjects were included in these studies. Two small-
person with an incomplete cervical (C)6 injury scale studies evaluating tibial nerve stimulation in
who experienced episodes of incontinence after subjects with spinal cord injury also reported re-
sensing his full bladder, but before he was able to ductions in incontinence caused by neurogenic de-
catheterize himself. Figure 4 shows the effect that trusor overactivity, although the results in the
conditional dorsal penile nerve stimulation had on subset of people with spinal cord injury are not
bladder contractions in this subject. During home stated in one study (Amarenco et al., 2003) and the
use for 3 weeks, this man applied stimulation when other is a report from a single patient (Andrews
he sensed his bladder was full, allowing him time and Reynard, 2003). Given the technical simplicity
to perform successful catheterization. He contin- of this technique and its potential to suppress ne-
ued to use the system after the trial was over be- urogenic detrusor overactivity, further experi-
cause of its success and his confidence in it. ments to determine efficacy in larger groups of
Additionally, Fjorback et al. (2003) developed a spinal cord injury subjects would help determine if
portable device that measured bladder pressure this technique should be pursued.
and automatically stimulated the dorsal penile
nerve to inhibit bladder contractions. This device Pelvic nerve stimulation
used a catheter to measure bladder pressure, and Stimulation of the nerve supply to the bladder
as such was impractical clinically, but did serve to presents some potential advantages over bladder
demonstrate the feasibility of a closed-loop system wall stimulation for people with spinal cord injury.
to treat neurogenic detrusor overactivity. Stimulation of the pelvic nerve (see Fig. 2G), which
contains the preganglionic parasympathetic fibers
innervating the detrusor, should cause contraction
Stimulation of peripheral nerve of the entire detrusor at a much lower current than
bladder wall stimulation (Hald, 1969). Pelvic nerve
Tibial nerve stimulation stimulation was shown to elicit bladder contrac-
In 1983, a report of investigations on nonhuman tions in dogs, but co-activation of the sphincters
primates demonstrated that the amount of current prevented good micturition, especially in male dogs
173

Fig. 4. Bladder pressure recording with and without stimulation during provocation of the bladder with rapid infusions of saline.
Reproducible reflexive bladder contractions were caused by rapid infusions of 60 ml of saline (C and D) and were abolished by
withdrawal of the saline. Further provocations (E and F) resulted in small reflexive bladder contractions that were immediately
abolished by stimulation of the dorsal penile nerve. During F and G, bladder pressure increases are hyperreflexive contractions caused
by the high volume. The two stimulation periods shown by open arrows at the onset and termination of G indicate patient-initiated
stimulation in response to sensation of bladder fullness. These suppressions of reflexive bladder contractions were better than the
previous ones. Without stimulation, bladder contractions are not suppressed (H). Reprinted from Lee and Creasey (2002) with
permission from the American Congress of Rehabilitation Medicine and the American Academy of Physical Medicine and Reha-
bilitation.

(Holmquist and Olin, 1968a, b). Sphincter activa- Tanagho and Schmidt’s group (Heine et al., 1977;
tion was likely a reflex caused by stimulation of Schmidt et al., 1979). These investigators reported
bladder afferents in the pelvic nerve. In addition, positive results using a sacral anterior root
chronic stimulation was found to cause fibrosis of stimulator to elicit voiding in spinalized animals.
the pelvic nerve leading to reductions in bladder This was followed by successful outcomes in hu-
response over time (Hald, 1969). Application in mans with spinal cord injury (Brindley et al., 1982;
humans was also frustrating because of the struc- Tanagho et al., 1989). Brindley’s device was
ture of the pelvic nerve. Whereas a distinct pelvic commercialized as the Finetech–Brindley Bladder
nerve exists in the cat and dog, parasympathetic System (Finetech Medical Ltd., Welwyn Garden
innervation of the human bladder is distributed City, UK) and has been implanted in over 2500
from the pelvic plexus arising from the pelvic people, in some cases for over 20 years (Rijkhoff,
nerves shortly after their exit from the sacral fora- 2004b). This system has been described and
mina (Wozniak and Skowronska, 1967), making reviewed in detail in a number of articles, so only
placement of electrodes very difficult (Susset and a summary of the device will be presented here
Boctor, 1967; Hald, 1969). Despite this, brief re- (Brindley, 1977; Brindley et al., 1982; Brindley
ports describing mixed results with pelvic nerve et al., 1986; Creasey, 1993; Egon et al., 1998).
stimulation in humans appeared in the 1970s The two prerequisites for implantation of sacral
(Burghele, 1973; Kaeckenbeeck, 1979 cited in anterior root stimulators are intact parasympathetic
Rijkhoff et al., 1997b), but no further reports ex- preganglionic neurons and a detrusor that is able to
ist to our knowledge. contract (Creasey, 1993). Electrodes can either be
implanted intradurally (see Fig. 2H) on the S2–S4
anterior roots (Brindley et al., 1982) or extradurally
Stimulation of sacral roots and nerves (see Fig. 2I) on the mixed sacral roots within the
spinal canal (Sauerwein et al., 1990; Lee, 1997). In
Sacral root stimulation either case, the procedure is usually combined with
The first electrical stimulation technique to devel- sacral posterior rhizotomy to abolish hyperreflexive
op into a commercially available device for blad- bladder and sphincter contractions, autonomic
der emptying in cord-injured people began with dysreflexia triggered by bladder fullness and pain
the work of Brindley (1977) as well as that of in patients with incomplete lesions (Creasey, 1993;
174

patients are continent, have increased bladder ca-


pacity, are able to void using their stimulator with
residual volumes o30 ml and are freed from cath-
eter usage leading to a great reduction in urinary
tract infections (Van Kerrebroeck et al., 1993). Ad-
ditionally, patients have reported beneficial stimula-
tor-driven erections and defecation (Brindley et al.,
1986; Van Kerrebroeck et al., 1993; Egon et al.,
1998).
One limitation of this device is that electrical
stimulation of the sacral roots, in addition to pro-
ducing sustained increases in bladder pressure, ac-
tivates the external urethral sphincter due to the
presence of both small diameter parasympathetic
preganglionic fibers and large diameter somatic fib-
ers in the sacral anterior roots (Brindley, 1977).
Since large fibers have lower thresholds of electrical
stimulation, excitation of the parasympathetic pre-
ganglionic fibers is accompanied by excitation of
the somatic fibers, leading to external urethral
sphincter contraction and urethral occlusion. The
Finetech–Brindley sacral anterior root stimulator
circumvents this problem by utilizing the difference
in the relaxation time of the detrusor and the
sphincter (Brindley et al., 1982). A train of electrical
stimuli is applied for 3–9 s, allowing bladder pres-
sure to rise behind the closed sphincter. Upon ces-
sation of stimulation, the striated sphincter relaxes
quickly while bladder pressure is transiently main-
tained allowing post-stimulus voiding (Brindley
et al., 1982) (see Fig. 6). Despite the supranormal
bladder pressures that occur with this technique, no
evidence of vesicoureteric reflux or hydronephrosis
Fig. 5. The components of the Finetech–Brindley sacral ante- has been found (Creasey, 1993). Sacral roots also
rior root stimulator. (A) Intradural electrodes and leads. (B) contain fibers innervating the musculature of the
Extradural electrodes and leads. (C) 2 and 3 channel implan- legs, and leg movement during stimulation can be
table receiver blocks. These components are implanted subcu-
cumbersome to some patients.
taneously and connect to the electrode leads. (D) The external
components of the device including the stimulator and trans- Given the proven benefits to people and the
mission block that is placed on the skin over the receiver block. large number of other neuroprosthetic devices
Adapted from Egon et al. (1998). implanted in patients (Rijkhoff, 2004b), one might
ask why more Finetech–Brindley sacral anterior
root stimulators have not been implanted. This is
Brindley, 1994). After electrode placement, leads likely due in part to the unwillingness of people to
are tunneled subcutaneously to an implantable re- undergo the irreversible posterior rhizotomy,
ceiver, activated by an external controller through a which, in addition to its very great benefits, abol-
radiofrequency link (Brindley et al., 1982). Figure 5 ishes reflex erection, defecation and micturition as
shows the components of this system. After im- well as any remaining perineal sensation. Implan-
plantation and posterior rhizotomy, the majority of tation of this system is also technically demanding
175

the sacral nerves through the sacral foramina were


developed (Schmidt et al., 1990) making the im-
plant procedure faster and less invasive than spinal
implantation of extradural electrodes. Since this
time, sacral nerve neuromodulation for treating
non-neurogenic bladder dysfunction including in-
continence, urgency–frequency and urinary reten-
tion have been well studied (Bosch and Groen,
2000; Siegel et al., 2000). A sacral nerve stimulator
based on this work has been commercialized
by Medtronic as the InterStims (Medtronic,
Minneapolis, MN, USA) and implanted in more
Fig. 6. Urine flow rate in a subject voiding his bladder using the than 10,000 people (Rijkhoff, 2004b). The InterS-
post-stimulus voiding technique. During stimulation, the flow tim consists of a battery-powered implantable
rate drops to nearly zero as contraction of the sphincter oc- stimulator connected to a single quadripolar elec-
cludes the urethra. Reprinted with permission from Brindley
et al. (1982).
trode usually inserted through the S3 sacral for-
amen to lie next to the S3 spinal nerve (see Fig. 2K
and Fig. 7). Sacral neuromodulation is only effec-
tive in a subset of patients with the above-men-
and attempts to market the device in the United tioned bladder dysfunctions, so all patients are
States as the VocareTM Bladder System by Ne- initially evaluated with a percutaneous electrode
uroControl Corp. (Cleveland, OH, USA) were connected to an external stimulator to assess their
ultimately unsuccessful for commercial and regu- response to this treatment before permanent im-
latory reasons (Hall, 2003), despite evidence that plantation (Bosch and Groen, 2000; Siegel et al.,
long-term use could realize a reduction in costs for 2000). Other reviews provide more detail about
management of lower urinary tract dysfunction this technology and its history (Schmidt, 1988;
(Creasey and Dahlberg, 2001). Commercialization Groen and Bosch, 2001; Jezernik et al., 2002; Van
in the USA has recommenced through NDI Med- Kerrebroeck, 2002; Middleton and Keast, 2004;
ical (Cleveland, OH, USA). Rijkhoff, 2004b; van Balken et al., 2004).
With the effectiveness of sacral nerve neuromod-
ulation, a number of investigators have examined
Sacral nerve neuromodulation this modality for treating neurogenic detrusor over-
In the early 1980s, Tanagho and Schmidt began activity in small-scale studies of subjects with spinal
implanting extradural sacral root stimulators in cord injury. Improvements in incontinence and in-
patients (Tanagho and Schmidt, 1988; Tanagho creases in maximal cystometric capacity have been
et al., 1989). It was found that continence could be demonstrated in subjects with incomplete spinal
controlled by low-frequency, low-amplitude stim- cord injury (Ishigooka et al., 1998; Chartier-Kastler
ulation to maintain sphincter contraction without et al., 2001; Hohenfellner et al., 2001). However, S3
concomitant detrusor contraction (Tanagho and sacral nerve neuromodulation in subjects with
Schmidt, 1988). As contractions of the detrusor complete spinal cord injury has been generally less
are inhibited by contractions of the sphincter, it effective (Chartier-Kastler et al., 2001) or had no
was also noted that stimulation causing sphincter effect at all (Hohenfellner et al., 2001; Schurch
contraction could inhibit detrusor activity leading et al., 2003) leading to the suggestion that intact
to improvements in continence in a range of ne- spinobulbospinal pathways contribute to the suc-
urogenic and non-neurogenic bladder conditions cess of sacral neuromodulation (Schurch et al.,
(Tanagho and Schmidt, 1988; Tanagho et al., 2003). On the other hand, stimulation of the mixed
1989). This neuromodulatory aspect of these im- S2 root extradurally using the Finetech–Brindley
plants was pursued and techniques for accessing stimulator without posterior rhizotomy, has
176

standing (Davis et al., 1994, 1997) a functional


electrical stimulator (FES 24-A; NeoPraxis Pty.
Ltd., Lane Cove, NSW, Australia) that included
electrodes intended to restore bladder function was
implanted in a thoracic (T)10 paraplegic (Davis
et al., 1999). Three pairs of electrodes intended to
elicit bladder contractions were inserted bilaterally
through the sacral foramina targeting the S2–S4
spinal nerves. An epidural electrode, intended to
suppress detrusor hyperreflexia, was implanted on
the conus medullaris to obviate the need for dorsal
rhizotomies (Davis et al., 2001). Increases in blad-
der pressure and some voiding was reported with
Fig. 7. Insertion of the quadripolar electrode through the S3
intermittent stimulation, but few data were pre-
sacral foramen to lie next to the S3 nerve. This electrode in-
sertion can be performed percutaneously to test the acute re- sented (Davis et al., 1999, 2001). Two additional
sponse of a patient to neuromodulation. Reprinted with the subjects were implanted with similar systems, but
permission of Medtronic, Inc. r 2005. bladder contractions could not be elicited (Smith
et al., 2002; Benda et al., 2003). However, in these
subjects, external urethral sphincter activity caused
successfully suppressed hyperreflexive bladder con- by low-frequency stimulation of the sacral nerves
tractions in people with complete spinal cord injury was reduced by selective high-frequency blockade
and neurogenic detrusor overactivity (Kirkham of the large somatic fibers (Shaker et al., 1998;
et al., 2002). The differing results observed using Benda et al., 2003). A conference report intro-
the S2 and S3 spinal nerves for neuromodulation duced the Minax system, a subset of the Praxis
in subjects with complete spinal cord injury may FES 24 stimulator specifically for bladder control
indicate a fundamental difference in the neural (Houdayer et al., 2002), but no further reports
pathways being excited in these two cases: namely have been published on either the Minax or Praxis
that S3 neuromodulation has a larger supraspinal systems and the company NeoPraxis appears to
component than S2 neuromodulation. have become inactive.
These results suggest that the success of neuro-
modulatory techniques for people with spinal cord
injury may depend on the completeness of the in- Stimulation of the spinal cord
jury as well as the specific location of the elec-
trodes. Given the commercial availability of the During the late 1960s and early 1970s, experiments
InterStim and the straightforward and well-estab- were performed by Nashold and Friedman in an-
lished evaluation techniques, sacral neuromodula- imals (Nashold et al., 1971) and humans (Nashold
tion will likely continue to be investigated for its et al., 1972) to test the efficacy of deep stimulation
usefulness in treating cord-injured people with ne- of the spinal cord (see Fig. 2L) to restore micturit-
urogenic detrusor overactivity. ion after spinal cord injury. These experiments
were conducted based on earlier studies demon-
strating that electrical stimulation of the cut sacral
NeoPraxis Praxis/Minax spinal cord could elicit bladder contractions
In 1983, a multi-channel implantable neuropros- (Stewart, 1899). It was proposed that electrical
thesis targeting sacral roots and nerves, based up- stimulation of the presumed sacral micturition
on existing cochlear stimulation technology from center (Kuru, 1965) would be capable of causing
Cochlear Ltd. (Lane Cove, NSW, Australia), was coordinated micturition after spinal cord injury.
proposed (reviewed in Davis et al., 2001). After Two electrodes, forming a bipolar pair, were im-
successful implantation of a 22 channel system for planted in the intermediolateral gray matter of the
177

sacral spinal cord (Nashold et al., 1971; Friedman


et al., 1972). Optimal rostrocaudal electrode place-
ment was determined by monitoring the change in
bladder pressure during stimulation of the dorsal
surface of the sacral spinal cord. The location
causing the greatest increase in bladder pressure
was selected as the location for implantation of the
penetrating electrodes. Friedman et al. (1972) re-
ported that six of 11 animals with intact spinal
cords and five of nine animals with transected spi-
nal cords voided during acute experimentation.
Additionally, in chronic experiments, stimulation
in five of six animals with intact spinal cords and
five of 10 animals with transected spinal cords
produced voiding. In animals where voiding did
not occur, bladder pressures elicited by electrical
stimulation were low, possibly indicating poor
Fig. 8. Diagram showing the location of the penetrating elec-
electrode placement. While some reduction in pel-
trodes implanted into the spinal cord. Reprinted with permis-
vic floor electromyogram, suggesting coordinated sion from Nashold et al. (1972).
sphincter inhibition, was occasionally noted, stim-
ulus spread resulting in activation of the sphincter were achieved. To overcome this, partial transure-
was also observed (Friedman et al., 1972). Studies thral sphincterotomies were performed that al-
conducted by another group, utilizing a variety of lowed voiding in two of the male patients without
electrode designs and stimulation parameters, con- causing incontinence. The female subject was able
cluded that sphincter motoneurons in the spinal to void with an intact sphincter. This general pat-
cord were always stimulated with the sacral para- tern was reported in most patients in a 10-year
sympathetic nucleus but that post-stimulus void- review of the technique (Nashold et al., 1981).
ing could be used successfully to empty the bladder Good voiding was achieved in 10 of 13 females
(Jonas et al., 1975; Jonas and Tanagho, 1975). subjects, but in only 5 of 14 male subjects. Clinical
Based on these results, 27 patients, 17 in the results from Duke University, where the technique
USA, nine in France and one in Sweden, were was developed, reported success in six of seven fe-
implanted with penetrating spinal cord electrodes male subjects and four of seven male subjects
(see Fig. 8) beginning in 1970 (Nashold et al., (Nashold et al., 1981). Of the male subjects, two
1981). These patients are believed to be the only voided successfully after bladder neck resections or
people in the world implanted with electrodes tar- partial sphincterotomies but two others failed to
geting intraspinal structures. The implanted device void even after these procedures were performed.
consisted of two electrodes, connected to a subcu- However, two male subjects did achieve concom-
taneous radiofrequency receiver that could be ac- itant relaxation of the urethra leading to complete
tivated by a handheld stimulator placed over the bladder evacuation without sphincterotomy, sug-
skin. The report on the initial four patients (three gesting that at least in some patients, spinal cord
male, one female) with electrodes implanted at the stimulation can elicit coordinated voiding (Grimes
S1 level eventually showed good voiding in three et al., 1975). Ultimately, 60% of the subjects ob-
patients (Nashold et al., 1972). Unlike some of the tained clinically good micturition with low residual
animal work, electrical stimulation did not gener- volumes, reductions in urinary tract infections,
ally cause concomitant relaxation of the urethra in increases in bladder capacity and freedom from
these patients. Rather, a spastic external urethral catheterization. Reductions in spasticity, as well
sphincter prevented micturition in the male subjects as erections in some male patients, and defecation
even though large increases in bladder pressure in some female patients were reported. However,
178

autonomic and motor responses including sweat- activated during stimulation, resulting in the func-
ing and lower limb movement often accompanied tional, but non-physiological, post-stimulus void-
stimulation of the spinal cord (Nashold et al., ing pattern associated with this implant. Rather
1981). than abandoning stimulation of sacral roots be-
Despite these reasonably good clinical results, cause of these limitations however, several tech-
no further implants using this procedure were per- niques are being developed to address these issues.
formed. There were several reasons for the aban- Three promising techniques are discussed here.
donment of this approach. One reason is that the Additional techniques to reduce or eliminate stim-
procedure, which involves highly invasive surgery, ulation-induced sphincter contractions are re-
was unsuccessful in 40% of the subjects (Nashold viewed by Rijkhoff et al. (1997b).
et al., 1981) presumably due to ineffective elec-
trode placement. Since neither coordinated mi-
cturition nor selective stimulation of the bladder Sacral posterior and anterior root stimulation
were achieved in most patients, this procedure One of the primary benefits of posterior rhizotomy
offered no advantages over Brindley’s sacral ante- in spinal cord injury is the abolition of neurogenic
rior root stimulator system (1982) but added the detrusor overactivity allowing low-pressure stor-
complication of a more unpredictable electrode age of urine and an increase in bladder capacity.
placement compared to sacral root electrode im- Since inhibition of reflexive bladder contractions
plantation. The inability to achieve stimulation of has been demonstrated in some people with spinal
the bladder without concomitant sphincter activity cord injury using sacral nerve neuromodulation
was likely due to stimulus spread and the close techniques (Ishigooka et al., 1998; Chartier-
proximity of the sacral parasympathetic nucleus Kastler et al., 2001; Hohenfellner et al., 2001), ne-
and Onuf’s nucleus (Kuru, 1965; de Araujo et al., uromodulation of sacral roots was investigated in
1982). five subjects with complete spinal cord injury and
neurogenic detrusor overactivity who underwent
implantation of a Finetech–Brindley system with-
Future devices for electrical control of the bladder out the usual posterior rhizotomy (Kirkham et al.,
2002). Electrodes were implanted extradurally on
Modifications of sacral root stimulators the mixed sacral roots in four subjects and in-
tradurally on anterior, posterior (see Fig. 2J) and
The Finetech–Brindley sacral anterior root mixed roots in the remaining subject. Since pos-
stimulator has proven to be the only commercial- terior rhizotomy was not performed, this system
ly successful electrical stimulation device to restore was referred to as a ‘‘Sacral Posterior and Anterior
voiding in people with spinal cord injury. Despite Root Stimulator’’ (Kirkham et al., 2002). In the
its proven efficacy (Van Kerrebroeck et al., 1993; three subjects that exhibited neurogenic detrusor
Brindley, 1994) and low risk of complications and overactivity after implantation, stimulation with
technical failures (Brindley, 1995), two issues exist, small pulse widths successfully inhibited hyperre-
that if overcome, could help improve the function flexive bladder contractions and increased bladder
and acceptance of this device. The posterior capacity to a level similar to that obtained with
rhizotomy that is performed in conjunction with anticholinergic medication. However, intermittent
the implantation of the Finetech–Brindley sacral stimulation at larger pulse widths to induce void-
anterior root stimulator eliminates neurogenic de- ing was unsuccessful, in spite of large increases in
trusor overactivity, detrusor–sphincter dyssynergia bladder pressure, because of detrusor–sphincter
and autonomic dysreflexia triggered by bladder dyssynergia between stimulation periods. Less
afferents, but irreversibly eliminates reflex erec- than 50% of the bladder volume was voided.
tions, reflex defecation, reflex micturition and any The sacral posterior and anterior root stimula-
remaining perineal sensation. The second issue is tor system is an important next step in the devel-
that both detrusor and sphincter efferent fibers are opment of sacral root stimulation in that
179

neurogenic detrusor overactivity, previously


abolished by posterior rhizotomy can be eliminat-
ed by posterior root neuromodulation. However,
detrusor–sphincter dyssynergia, also previously
abolished by posterior rhizotomy, prevents blad-
der voiding in this system. Clinical use of the sacral
posterior and anterior root stimulator system
will require additional techniques to inhibit
Fig. 9. Electrical current under the anode hyperpolarizes the
detrusor–sphincter dyssynergia without posterior
membrane of large fibers at lower stimulation amplitudes than
rhizotomy to allow effective voiding. smaller fibers. This section of hyperpolarized membrane blocks
the transmission of action potentials generated at the cathode
Selective anodal block preventing activation of the sphincter while allowing activation
Anodal blocking offers a method to block action of the bladder. Action potentials propagate and are blocked in
the large fibers in both directions. The tripolar cuff design
potential propagation in large fibers, effectively
minimizes current spread and I1 and I2 are the two independent
allowing selective stimulation of the small para- current sources sharing a common cathode. Reprinted with
sympathetic fibers innervating the bladder. Selec- permission from Rijkhoff et al. (1997a).
tive stimulation of these fibers would reduce the
concomitant contraction of the sphincter and low-
er limbs that presently occurs with the Fine- Several issues remain with anodal blocking
tech–Brindley sacral anterior root stimulator. This however. Anodal blocking waveforms being ex-
could restore a more physiological voiding pattern. amined currently are monophasic and require
Anodal blocking takes advantage of the fact that pulse widths of approximately 600 ms in humans
axons are hyperpolarized under the anode, reduc- (Rijkhoff et al., 1998). Depending on the currents
ing their excitability. Since larger diameter axons required, long duration pulses can lead to irre-
have a lower threshold for electrical activation, versible electrochemical reactions at the electrodes
they can be selectively hyperpolarized (Accornero and eventual nerve damage (McCreery et al.,
et al., 1977; Rijkhoff et al., 1994a). Stimulation at 1990). However, methods to increase safety by re-
the cathode excites both large somatic fibers and ducing the charge per phase used to achieve anodal
the smaller parasympathetic fibers, but action po- block are being examined (Vuckovic and Rijkhoff,
tential propagation in the somatic fibers is blocked 2004). Additionally, implantable stimulators capa-
at the hyperpolarized portion of membrane, al- ble of producing the waveforms generally required
lowing selective transmission in the parasympa- for anodal blocking do not exist, although they are
thetic fibers (see Fig. 9). Brindley and Craggs under development (Bugbee et al., 2001; Rijkhoff,
(1980) successfully tested this technique in animals 2004a). Although anodal blocking allows selective
to achieve selective parasympathetic fiber stimula- activation of the bladder, posterior rhizotomies
tion with a sacral anterior root stimulator, but it may still be required to allow voiding. In one an-
did not work well enough in humans for regular imal study, anodal blocking without posterior
use (Brindley et al., 1982). More recently, anodal rhizotomy never resulted in voiding because of
blocking has been examined in modeling studies to reflexes that increased intraurethral pressure. In
determine stimulation parameters (Fang and the same experiment however, continuous voiding
Mortimer, 1991; Rijkhoff et al., 1994a). Addition- was achieved once a posterior rhizotomy was per-
ally, both animal studies (Fang and Mortimer, formed (Grunewald et al., 1998). In another ani-
1991; Koldewijn et al., 1994; Rijkhoff et al., 1994b; mal study, complete voiding was achieved using
Grunewald et al., 1998) and intraoperative hu- anodal blocking without posterior rhizotomy
mans studies (Rijkhoff et al., 1997a, 1998) of (Koldewijn et al., 1994). Even if posterior rhizo-
anodal blocking have demonstrated large decreas- tomy is required to allow voiding when using
es in sphincter and leg activity while producing anodal blocking, this technique may allow a more
bladder contractions. physiological continuous voiding pattern and may
180

reduce unwanted leg movements that currently


occur with sacral anterior root stimulators.

High-frequency blockade
Another technique to prevent external urethral
sphincter activation with sacral anterior root
stimulators uses high-frequency stimulation to
block action potential propagation in somatic
fibers. Sawan, Elhilali and colleagues have suc-
cessfully stimulated mixed sacral roots with high-
frequency (600 Hz), low-amplitude pulses to block
urethral sphincter efferent activity while superim-
posing low-frequency, high-amplitude pulses to
activate parasympathetic preganglionic efferents
that in turn generate bladder contractions
(Shaker et al., 1998; Abdel-Gawad et al., 2001) (see
Fig. 10). High-frequency stimulation has been
shown to block action potential propagation by
hyperpolarizing axons and maintaining them in
their refractory period (Solomonow et al., 1983).
High-frequency, low-amplitude stimulation hyper-
polarizes large fibers but the stimulation amplitude
is not high enough to affect the smaller parasym-
pathetic fibers. In a study of 12 chronically
implanted dogs, all voided with o20% residual
urine and seven of the group voided with o10%
residual urine (Abdel-Gawad et al., 2001). This
system represents a potential advantage over the Fig. 10. High-frequency block of large fibers allowing selective
current Finetech–Brindley sacral anterior root activation of the bladder by sacral ventral root stimulation.
stimulator where the bladder and sphincter contract Low-frequency pulses lead to activation of large and small
simultaneously and voiding occurs post-stimulus. fibers, while high-frequency pulses block action potential prop-
agation in large fibers selectively. (A) High-frequency, low-am-
This device, including a proposed method of plitude pulses superimposed on low-frequency, high-amplitude
neuromodulation to inhibit the hyperreflexive pulses. LFA – low-frequency amplitude, LFP – low-frequency
bladder by stimulation of the sacral nerves, has period, LFW – low-frequency pulse width, HFA – high-fre-
been patented (Sawan and Elhilali, 2002). While quency amplitude, HFP – high-frequency period, HFW – high-
this group has not published results regarding the frequency pulse width. (B) Difference between low-frequency
only stimulation and combined low- and high-frequency stim-
proposed neuromodulatory action of their device, ulation. Intraurethral pressure and sphincter EMG were reduced
proof of principle has been demonstrated in cord- while bladder pressure was maintained when the selective stim-
injured people with a similar device (Kirkham ulation (high-frequency, low-amplitude) waveform was utilized.
et al., 2002). Neuromodulation could remove the Reprinted from Boyer et al. (2000) with permission from the
necessity for posterior rhizotomy in sacral root IEEE.
stimulators, especially since successful voiding
without posterior rhizotomy has been achieved et al., 2003). Although no reports in humans exist
with this stimulation paradigm, at least in animals from Sawan and Elhilali’s group, Victhom Human
(Abdel-Gawad et al., 2001). A brief report from Bionics (Saint-Augustin-de-Desmaures, Quebec,
another group using the same stimulation paradigm Canada) has licensed the technology and is con-
suggests that it is also effective in humans (Benda tinuing development of the device.
181

Intraspinal microstimulation must be offered, especially since an intraspinal


microstimulation implant would likely be at least as
Since Nashold and Friedman’s original work difficult to perform as a sacral anterior root stimula-
on spinal cord stimulation to evoke micturition tor implant. One advantage of intraspinal micro-
(Nashold et al., 1971; Nashold et al., 1972), interest stimulation is that bladder contractions can be
has persisted in this technique. In these experi- evoked without concomitant sphincter contractions
ments, electrodes were on the order of 0.3–0.4 mm (Carter et al., 1995; Grill et al., 1999). However,
in diameter with 0.5–1.0 mm long exposed tips more importantly, intraspinal microstimulation
(Nashold et al., 1971; Jonas et al., 1975) leading to allows the possibility of activating sacral intern-
geometric electrode surface areas of 0.5–1.4 mm2. euronal networks that produce coordinated mi-
Stimulus spread from these comparatively large cturition or some part thereof (Nashold et al., 1971;
electrodes between the adjacent sacral parasympa- Grimes et al., 1975; Grill, 2000).
thetic nucleus and Onuf’s nucleus (Kuru, 1965; Networks of interneurons responding to pelvic
de Araujo et al., 1982) was the likely cause of ob- and pudendal afferents and receiving projections
served concomitant bladder and sphincter contrac- from the pontine micturition center exist in various
tions (Nashold et al., 1972). Electrode development regions around the central canal, in the dorsal gray
has improved the ability to selectively stimulate commissure and in the intermediolateral cell column
specific regions within the spinal cord (Prochazka of the sacral spinal cord (reviewed in de Groat et al.,
et al., 1976; Mushahwar et al., 2000; McCreery 1996; Shefchyk, 2001) and are active during mi-
et al., 2004). Currently, electrode arrays for chronic cturition (Grill et al., 1998; Buss and Shefchyk,
intraspinal microstimulation use microwire elec- 2003). One group of interneurons, located in the
trodes (see Fig. 11A) or silicon substrate micro- dorsal gray commissure, are of particular interest in
electrodes manufactured using photolithographic relation to inhibition of the external urethral sphinc-
processes (see Fig. 11B). Microwire-based elec- ter as the interneurons contain inhibitory neuro-
trodes use iridium and platinum–iridium alloy transmitter, receive direct projections from the
wires 20–30 mm in diameter with 20–100 mm long pontine micturition center and are believed to
exposed tips resulting in geometric electrode sur- project to Onuf’s nucleus (Blok et al., 1997; Sie
face areas of 1600–10,000 mm2 (Mushahwar et al., et al., 2001). Since Onuf’s nucleus does not receive
2000). Silicon substrate microelectrodes used for inhibitory projections from supraspinal centers, in-
chronic intraspinal microstimulation implants can hibitory interneurons in the dorsal gray commissure
have multiple stimulation sites at various depths may mediate voluntary relaxation of the sphincter
per penetrating shank with electrode surface areas (Blok, 2002). This view is supported by the finding
around 2000 mm2 (McCreery et al., 2004). These that electrical stimulation in the dorsal gray com-
electrode surface areas are 50–300 times smaller missure produced active and sustained decreases in
than the smallest electrodes used in the first spinal urethral pressure in spinally intact cats (Blok et al.,
cord stimulation experiments (Nashold et al., 1971; 1998; McCreery et al., 2004). Additionally, some
Jonas et al., 1975) and allow selective stimulation voiding can occur when electrodes around the cen-
of the sacral parasympathetic nucleus without con- tral canal are stimulated (Grill et al., 1999). The
comitant sphincter activation (Carter et al., 1995; ability to actively inhibit urethral activity is not cur-
Grill et al., 1999). Although electrode arrays can rently possible with sacral root stimulation. These
cause inflammatory reactions, glial scarring and results demonstrate the potential of intraspinal mi-
neural death around the implantation site, this can crostimulation to achieve coordinated micturition
be minimized (McCreery et al., 2004) and would through bladder excitation and sphincter inhibition.
presumably be just as safe or safer in long-term Current research on intraspinal microstimulation
use as the larger electrodes implanted in humans is therefore focused on simultaneous stimulation of
(Nashold et al., 1981). the sacral parasympathetic nucleus to produce blad-
If intraspinal microstimulation is to be clinically der contractions and the dorsal gray commissure
useful, clear advantages over sacral root stimulation to actively relax the urethra (see Fig. 11C). This
182

Fig. 11. Electrode designs and target locations (in the cat) for intraspinal microstimulation. (A) Microwire electrode array with
electrode tips placed in protective tubing. (B) Multi-site penetrating silicon electrode array. (C) Electrode targets in the cat sacral spinal
cord. Electrical stimulation in the dorsal gray commissure, which contains interneurons with inhibitory projections to sphincter
motoneurons, can elicit relaxation of the external urethral sphincter. Electrical stimulation of sacral parasympathetic nucleus, which
contains bladder preganglionic neurons, can elicit sustained increases in bladder pressure. Adapted from Prochazka et al. (2002a) and
McCreery et al. (2004).

approach has produced sustained high-pressure absence of anesthesia. Additionally, electrodes tar-
bladder contractions, coordinated increases in blad- geting the sacral parasympathetic nucleus produced
der pressure and decreases in urethral pressure and similar increases in bladder pressure using the same
occasional incomplete voiding (Prochazka et al., stimulation parameters, before and after complete
2003b), but so far it has proven unreliable. One spinal cord transection. However, bladder pressure
possible reason is that in addition to the interneu- increases produced by stimulation in the sacral para-
rons in the dorsal gray commissure that inhibit sympathetic nucleus and urethral pressure decreases
sphincter motoneurons, there are other interneurons produced by stimulation in the dorsal gray commis-
in this same region that have been shown to decrease sure may not be sufficient to produce micturition. In
their firing rate during micturition, and may be part some cases, simultaneous intraurethral pressure re-
of pathways with excitatory connections to sphincter cordings in the vicinity of the external urethral
motoneurons (Buss and Shefchyk, 2003). If this is sphincter and bladder pressure recordings, have in-
the case, electrical stimulation in parts of the dorsal dicated that voiding should occur (Prochazka et al.,
gray commissure may, in fact, activate more excita- 2003a) (see Fig. 12), but once the urethral pressure
tory interneurons than inhibitory interneurons, and catheter was removed to unblock the urethra, stim-
thereby cause contraction, rather than relaxation of ulation through the same electrodes did not elicit
the external urethral sphincter. It is also unknown voiding. In this case, high pressure in the bladder
whether intraspinal microstimulation can affect the neck or distal urethra may have prevented voiding.
activity of the smooth muscle internal urethral
sphincter, which is the primary mechanism to main-
tain continence until the bladder is very full or at Urethral afferent stimulation
high pressure. These results were obtained in an
awake animal, suggesting that previous results Electrical stimulation of afferent branches of the
in anesthetized animals may also hold true in the pudendal nerve, specifically the dorsal penile
183

incomplete. Despite the fact that only incomplete


voiding has been demonstrated, this stimulation
technique is very intriguing from the perspective of
neuroprosthesis device development. So far, it is
the only way in which bladder contractions can be
generated without invasive implantations of elec-
trodes targeting the bladder, sacral roots or spinal
cord itself. However, even more interesting is the
apparent ability of urethral afferent stimulation to
evoke coordinated micturition after complete spi-
nal cord injury (Shefchyk and Buss, 1998).

Fig. 12. Bladder and urethral pressure changes in response to


electrical stimulation through three electrodes: two targeting the
Microstimulators
sacral parasympathetic nucleus and one targeting the dorsal
gray commissure. During stimulation, immediate increases in Implanted neuroprostheses discussed in this review
bladder pressure and decreases in urethral pressure were have all used the same basic set of components. An
achieved. The pressures measured in the bladder and urethra implanted stimulator, either battery powered or
became essentially equal (within the calibration error of the
transducers) during stimulation indicating that voiding might
inductively coupled to an external power source, is
occur in the absence of the urethral catheter. However, once the implanted subcutaneously in a convenient location
catheter was removed to unblock the urethra, stimulation such as the abdominal or chest region and long
through the same electrodes did not induce voiding. Adapted leads connect the electrodes to the stimulator. This
from Prochazka et al. (2003a). design can lead to time-consuming surgical proce-
dures as well as technical failures including lead
nerve, has been shown to inhibit hyperreflexive breakage and connector failure (Brindley, 1995).
bladder contractions occurring after spinal cord Microstimulators such as the BION (Advanced
injury. However, electrical stimulation of urethral Bionics, Valencia, CA) provide an alternative ap-
afferents, also forming part of the pudendal nerve, proach. BIONs are self-contained, injectable mi-
has been shown to elicit bladder contractions crostimulators that are programmed and powered
as well as relaxation of the sphincter. These spinal by inductive coupling. A single external transmit-
reflexes were first described by Barrington in ter coil can communicate with up to 256 BIONs,
spinal cord-transected cats (Barrington, 1914, each of which can deliver current controlled pulses
1941), and more recently have been investigated with a pulse width range of 4–512 ms and ampli-
in cord-transected cats (Shefchyk and Buss, 1998; tude range of 0–30 mA up to frequencies of 50
Gustafson et al., 2003) and humans with spinal pulses per second in a package 2 mm in diameter
cord injury (Gustafson et al., 2003, 2004). These and 16 mm long (Loeb et al., 2001) (see Fig. 13).
reflexes are presumed to facilitate voiding by pos- A more recent version of the BION includes a
itive feedback from afferents sensitive to urethral lithium-ion battery to power the device during
dilation (Shafik et al., 2003a, b). In humans, ure- normal use, but is programmed and recharged us-
thral afferents were electrically stimulated using a ing the external coil. This device is larger (3.3 mm
catheter-mounted electrode (Gustafson et al., diameter, 27 mm long) (Groen et al., 2004), but
2003) passed into the urethra (see Fig. 2M). In allows subjects to be free of the external coil and
subjects with complete spinal cord injury, bladder associated hardware during daily activities, at least
contractions reaching 70 cm H2O as well as void- in applications not requiring phasic control.
ing could be achieved if the bladder volume was Two studies have evaluated the use of BIONs
above a threshold value (Gustafson et al., 2004). for treating bladder dysfunction using the induc-
Below this threshold, bladder contractions could tively powered (Buller et al., 2002) and battery-
not be generated and voiding was therefore powered (Groen et al., 2004) systems. Although
184

of magnetic stimulation to exert control over lower


urinary tract function in spinal cord injury subjects
but have arrived at very different results (Lin et al.,
1997; Bycroft et al., 2004). In the first study it was
concluded that repetitive magnetic stimulation
Fig. 13. A glass encapsulated BION. Adapted from Loeb et al. (15–30 Hz) of the sacral nerves, achieved by plac-
(2001). ing the stimulation coil over the sacrum (see Fig.
2O), caused direct activation of parasympathetic
preganglionic neurons innervating the bladder
final reports have yet to be published, both studies leading to increases in bladder pressure. Fatigue
have shown positive results using pudendal nerve of the external urethral sphincter and intermittent
(see Fig. 2N) stimulation to treat overactive blad- stimulation to allow post-stimulus voiding were
der incontinence. BIONs have not been clinically the two methods postulated to explain the voiding
tested in spinal cord injury patients for bladder that occurred, even though many subjects had re-
control, but a number of stimulation techniques ceived sphincterotomies (Lin et al., 1997). The
presented in this review could be adapted to use more recent study suggests that magnetic stimula-
BIONs. tion of the sacral nerves causes direct inhibition of
bladder contractions in cord-injured people with
MiniatURO neurogenic detrusor overactivity. It also stated
that the previously described bladder contractions
The miniatURO (Biocontrol Medical Ltd., were rebounds occurring at the end of stimulation
Yahud, Israel) is a closed-loop implantable as a result of releasing the detrusor from direct
electrical stimulation system for treating mixed inhibition (Bycroft et al., 2004).
urinary incontinence. The device consists of an Whatever the effect of magnetic stimulation on
intra-abdominal pressure sensor, pelvic floor stim- bladder function in cord-injured people, this tech-
ulation electrode and a stimulator with integrated nique holds promise for a device-based therapy for
control system. Stimulation is triggered by in- bladder control. Current magnetic stimulation
creases in intra-abdominal pressure. This device systems are large and would not be suitable for
has been tested in short-term settings where ab- chronic use. However, the potential benefits of
dominal pressure was measured rectally, stimulat- this completely noninvasive technique, and its
ing electrodes were introduced percutaneously and demonstrated efficacy for treating neurogenic de-
the stimulator was worn externally. All subjects in trusor overactivity and/or eliciting partial voiding,
a group of 16 with stress incontinence responded deserve further investigation.
positively and became dry or had reduced episodes
of incontinence (Nissenkorn et al., 2004). While
this device has not been tested in cord-injured Successes and failures of devices for bladder control
people, there is reason to believe that this kind of
device might be effective in the spinal cord injury A wide variety of techniques and devices have been
population (Fjorback et al., 2003). developed to manage the significant problems as-
sociated with lower urinary tract dysfunction after
spinal cord injury. While devices such as the Fine-
Transcutaneous magnetic stimulation tech–Brindley sacral anterior root stimulator can
truly be said to exert control over the lower uri-
Transcutaneous magnetic stimulation of the nerv- nary tract, other devices, including catheters mere-
ous system is a noninvasive method of activating ly manage the symptoms associated with lower
neural tissue and has therefore become a useful urinary tract dysfunction after spinal cord injury.
technique for human experimentation and clinical However, the ultimate goal of any treatment
diagnostics. Two studies have examined the ability modality, including pharmacological and surgical
185

Table 1. Summary of mechanical devices used for lower urinary tract management after spinal cord injury

Device Efficacy Advantages Disadvantages Current status

Catheter Very good Simple, inexpensive Urinary tract infections, inconvenient Clinical
Artificial sphincter Mixed Proven design Infection, urethral erosion Limited use
Urethral stent Good Reversible alternative to sphincterotomy Not for long-term implantation Some clinical
Intraurethral pump Good Simple implant Regular replacement, discomfort Investigational

methods, is to create a bladder capable of storing cord injury. Many techniques have been presented
large volumes of urine at low pressure, while pre- in this review that vary significantly in their ability
venting incontinent episodes and allowing periodic to establish a high-volume, low-pressure bladder
evacuation of that urine at low pressure. If a and to produce low-pressure voiding. Improving
treatment achieves its intended function, then the continence, storage volume and storage pressure
merits of one technique versus another depend on by inhibiting hyperreflexive bladder contractions
issues such as adverse side effects, procedural re- has been the specific aim of a number of devices.
versibility, device cost, ease of use and ease of im- A brief summary of the efficacy, main advantages
plantation. and disadvantages and current status of these
devices is presented in Table 2, while a similar
summary for devices intended to evoke bladder
Device efficacy, advantages and disadvantages emptying is presented in Table 3. Of the electrical
stimulation techniques intended to inhibit hyper-
Two classes of devices have been described in this reflexive bladder contractions, both dorsal penile
review: mechanical devices and electrical stimula- nerve stimulation and tibial nerve stimulation have
tion devices. The mechanical devices discussed shown promising results. However, these methods
range from the very simple to the very complex require additional study with the use of more
and address voiding dysfunction, incontinence and practical devices that are suitable for chronic
detrusor–sphincter dyssynergia. A brief summary use. Bladder wall, pelvic nerve and spinal
of the efficacy, main advantages and disadvantages cord stimulation are all methods to elicit voiding
and current status of these devices is presented in that have been tested in humans with spinal cord
Table 1. Several general comments about the use injury and subsequently abandoned. However,
of mechanical devices can be made. Catheters and bladder wall stimulation and spinal cord stimula-
the In-Flow intraurethral pump do not require tion continue to be explored in the laboratory
implantation. They are also commercially availa- setting.
ble, allowing easy adoption by clinicians. Artificial
urethral sphincters and urethral stents do require
surgery, but this is relatively simple. However, Clinical success of devices in managing dysfunction
none of these devices offer the potential to restore after spinal cord injury
lower urinary tract function to the pre-injury state.
There are also significant side effects due to the Catheters and urethral stents, as well as dorsal pe-
chronic presence of foreign materials in the lower nile nerve, tibial nerve, spinal cord and sacral root
urinary tract. Finally, no mechanical device is stimulation have all been shown to be effective in
able to affect neurogenic detrusor overactivity, so managing various aspects of lower urinary tract
pharmacological or surgical treatments must be dysfunction after spinal cord injury. However, the
used to establish a high-volume, low-pressure success of these devices from a clinical perspective
bladder. varies significantly. The clinical success of catheters
Electrical stimulation devices, while not cura- remains foremost. Of the other effective techniques,
tive, offer the unique potential to restore normal only the Finetech–Brindley sacral anterior root
function to the lower urinary tract after spinal stimulator could be considered a proven clinical
186

Table 2. Summary of electrical stimulation devices used to control continence after spinal cord injury

Device Efficacy Advantages Disadvantages Current status

Pelvic floor maximal Mixed Long-lasting Physical and Limited use


functional electrical improvements psychological
stimulation discomfort
Inconsistent results
Thigh stimulation Mixed Non-invasive No predictors for Limited use
success
Long-lasting Time-consuming
improvements treatment
Dorsal penile nerve Promising Non-invasive Not well studied Investigational
surface stimulation
Tibial nerve stimulation Promising Simple treatment Permanent device Investigational
procedure required
Not well studied
Sacral neuro- Mixed Minimally invasive May not work in Investigational
modulation complete injuries
Proven design and Inconsistent results, Not
implant procedure well studied
Commercially available

success with over 2500 systems implanted (Rijkhoff, that eventual regenerative therapies will likely be
2004b). This system is successful because it is the combined with neuroprostheses to maximize func-
only device that reliably evokes complete bladder tional recovery (Prochazka et al., 2002b), new and
evacuation, significantly reducing the complications improved devices are required to restore control of
associated with catheterization. When combined the lower urinary tract after spinal cord injury.
with sacral posterior rhizotomy, as it nearly always The feasibility and efficacy of a number of me-
is, the Finetech–Brindley sacral anterior root chanical and electrical devices for treating lower
stimulator provides a complete system for lower urinary tract dysfunction has been clearly demon-
urinary tract control after spinal cord injury. How- strated in spinal cord injury patients. However, no
ever, the consequences of irreversible rhizotomy, clinical device can be said to have solved the
the technically demanding implant procedure and problem of bladder control as low-pressure phys-
commercial and regulatory issues have limited the iological voiding can not yet be produced and no
availability of this device as well as the acceptance device has successfully incorporated methods to
of it by patients and clinicians. With this in mind, produce both voiding and suppression of ne-
given the large number of people that could benefit urogenic detrusor overactivity. While future im-
from such a device, and a generally increasing clin- proved biomaterials will undoubtedly reduce some
ical acceptance of neuroprostheses in general, the of the side effects associated with the use of
Finetech–Brindley sacral anterior root stimulator mechanical devices (Beiko et al., 2004), these are
has had a limited impact on the spinal cord injury unlikely to achieve complete control over the lower
population throughout most of the world. urinary tract. Electrical stimulation devices on the
other hand have demonstrated the ability to achieve
significant control over neurogenic detrusor
The future of devices for bladder control overactivity and voiding. Adaptations of the Fine-
tech–Brindley sacral anterior root stimulator, in-
Progress is being made on regeneration of the spi- cluding posterior root stimulation and anodal
nal cord, but a complete biological cure for spinal blocking or high-frequency blockade of somatic fib-
cord injury is unlikely to be developed in the near ers, offer the possibility of inhibiting hyperreflexive
future (Fawcett, 2002). In light of this, and the fact bladder contractions and producing physiological
187

Table 3. Summary of electrical stimulation devices used to control voiding after spinal cord injury

Device Efficacy Advantage Disadvantages Current status

Intravesical electrical Mixed Long-lasting results Ineffective in many Limited use


stimulation patients
Can improve voluntary No predictors for
voiding in incomplete success
injuries
Time-consuming
treatment
Bladder wall Mixed Simple surgical Sphincter and pelvic Abandoned except for
stimulation approach floor contraction lower motoneuron
lesions
Electrode–bladder
interface failure
Pelvic nerve stimulation Poor Sphincter and pelvic Abandoned
floor contraction
Difficult surgical
approach
Nerve unsuitable for
stimulation
Sacral root stimulation Very good Voiding without Posterior rhizotomy Clinical
catheterization required
Low residual volume Demanding surgical
implant
Proven design and Unphysiological
implant procedure voiding
Long-term usage with
few failures
Commercially available
Spinal cord stimulation Good Voiding without Sphincter contraction Originally abandoned,
catheterization currently investigational
Low residual volume Sphincterotomy
required in males
Long-term usage Difficult electrode
placement 40% failure
rate

voiding without the currently requisite posterior in their current form would be even more difficult
rhizotomy. Combining such methods may signifi- than implantation of sacral root stimulators. Unless
cantly improve future electrical stimulation devices simpler electrode configurations are developed, this
for bladder control. may limit clinical use of intraspinal microstimula-
Other electrical stimulation techniques currently tion even if it can be shown to be reliable and ef-
under investigation also show promise. Intraspinal fective in animals. Stimulation of the dorsal penile
microstimulation has the potential to utilize remain- nerve and urethral afferents are among the most
ing spinal cord networks and can achieve selective interesting techniques currently being investigated
stimulation of the bladder as well as active inhibi- to restore control of the lower urinary tract from a
tion of the sphincter, albeit not reliably enough to neuroprosthetic device development perspective.
produce consistent voiding. Intraspinal microstim- They offer the potential of a minimally invasive
ulation research has yet to address the problem of implant to suppress neurogenic detrusor overactiv-
neurogenic detrusor overactivity and surgical im- ity and produce voiding, perhaps using micro-
plantation of intraspinal microstimulation devices stimulators. If these techniques are to have an
188

impact on the clinical management of lower urinary urinary tract function is to be developed. Perhaps the
tract dysfunction after spinal cord injury, they must most difficult and crucial problem is achieving re-
provide clear improvements in treatment over what laxation of the sphincter to allow physiological void-
can be currently accomplished with catheters, phar- ing. It has been said that ‘‘The key to control of the
macological therapies and surgical intervention. bladder lies in control of the sphincter’’ (Schmidt,
An ideal device for controlling lower urinary 1986). While this comment was originally made in
tract function after spinal cord injury can be pos- reference to the ability of sphincter activity to mod-
tulated based on the material presented in this ulate bladder contractility, it is equally true that the
review. Above all, the device must suppress ne- ability to control the sphincter and eliminate un-
urogenic detrusor overactivity and allow user- wanted activity remains the final piece of the puzzle
controlled, continuous, low-pressure voiding with that must be put in place for bladder control neuro-
a low residual volume. The device should not re- prostheses to be completely effective. A number of
quire additional pharmacological or surgical pro- techniques are being investigated to accomplish this,
cedures to operate successfully and would ideally but no single one has completely achieved this goal.
require only minimally invasive surgery. Although In conclusion, while many devices and techniques
not discussed in this review, an implant to restore have been tried and ultimately abandoned, the suc-
lower urinary tract function should ultimately cesses clearly show the immense benefits that can be
provide means to restore bowel and sex function as achieved with the use of devices in the control of the
well. Finally, any procedure must be completely dysfunctional bladder
reversible so that people are not committed to a
particular device for the rest of their lives and are
able to take advantage of future devices. Acknowledgments

The authors would like to gratefully acknowledge


Conclusions Dr. Jonathan Norton for his helpful comments in
the preparation of this article and Jan Kowalczewski
In this review we have discussed devices and tech- for preparing Fig. 2. The work on bladder control
niques aimed at restoring normal function to the devices in Arthur Prochazka’s laboratory is sup-
lower urinary tract after spinal cord injury. Some of ported by NIH-NINDS contract N01-NS-2-2342.
these devices and techniques have ultimately proven R.G. is supported by the Alberta Heritage Foun-
to be unsuccessful for reasons such as insufficient dation for Medical Research.
efficacy, unacceptable treatment procedures, side ef-
fects and technical failures. Other devices, most no-
tably catheters and sacral root stimulation, have References
been successful. Over 20 years of experience with
Abdel-Gawad, M., Boyer, S., Sawan, M. and Elhilali, M.M.
sacral root stimulation to restore voiding clearly in- (2001) Reduction of bladder outlet resistance by selective
dicates the advantages that people with spinal cord stimulation of the ventral sacral root using high frequency
injury can expect to achieve with neuroprostheses. blockade: a chronic study in spinal cord transected dogs.
However, detrusor–sphincter dyssynergia must be J. Urol., 166: 728–733.
overcome before neuroprostheses can gain more Accornero, N., Bini, G., Lenzi, G.L. and Manfredi, M. (1977)
Selective activation of peripheral nerve fibre groups of
widespread use. Electrical stimulation techniques to different diameter by triangular shaped stimulus pulses.
suppress neurogenic detrusor overactivity, allowing J. Physiol., 273: 539–560.
efficient bladder filling and storage, have been dem- Amarenco, G., Ismael, S.S., Even-Schneider, A., Raibaut, P.,
onstrated in some cord-injured people. But relatively Demaille-Wlodyka, S., Parratte, B. and Kerdraon, J. (2003)
Urodynamic effect of acute transcutaneous posterior tibial
few studies have been conducted and many details
nerve stimulation in overactive bladder. J. Urol., 169:
regarding these techniques have yet to be elucidated. 2210–2215.
These, or other similar techniques, must be success- Andersen, J.T. and Bradley, W.E. (1976) The syndrome of de-
ful if a neuroprosthesis for complete control of lower trusor–sphincter dyssynergia. J. Urol., 116: 493–495.
189

Anderson, K.D. (2004) Targeting recovery: priorities of the Brindley, G.S. (1994) The first 500 patients with sacral anterior
spinal cord-injured population. J. Neurotrauma, 21: root stimulator implants: general description. Paraplegia, 32:
1371–1383. 795–805.
Andrews, B.J. and Reynard, J.M. (2003) Transcutaneous pos- Brindley, G.S. (1995) The first 500 sacral anterior root stimula-
terior tibial nerve stimulation for treatment of detrusor tors: implant failures and their repair. Paraplegia, 33: 5–9.
hyperreflexia in spinal cord injury. J. Urol., 170: 926. Brindley, G.S. and Craggs, M.D. (1980) A technique for anod-
Barrington, F.J.F. (1914) The nervous mechanism of micturit- ally blocking large nerve fibres through chronically implanted
ion. Q. J. Exp. Physiol., 8: 33–71. electrodes. J. Neurol. Neurosurg. Psychiat., 43: 1083–1090.
Barrington, F.J.F. (1921) The relation of the hind-brain to mi- Brindley, G.S., Polkey, C.E. and Rushton, D.N. (1982) Sacral
cturition. Brain, 4: 23–53. anterior root stimulators for bladder control in paraplegia.
Barrington, F.J.F. (1925) The effect of lesions on hind- and Paraplegia, 20: 365–381.
mid-brain on micturition in the cat. Q. J. Exp. Physiol., 15: Brindley, G.S., Polkey, C.E., Rushton, D.N. and Cardozo, L.
81–102. (1986) Sacral anterior root stimulators for bladder control in
Barrington, F.J.F. (1941) The component reflexes of micturit- paraplegia: the first 50 cases. J. Neurol. Neurosurg. Psychiat.,
ion in the cat. Part III. Brain, 64: 239–243. 49: 1104–1114.
Beiko, D.T., Knudsen, B.E., Watterson, J.D., Cadieux, P.A., Bugbee, M., Donaldson, N.N., Lickel, A., Rijkhoff, N.J. and
Reid, G. and Denstedt, J.D. (2004) Urinary tract biomate- Taylor, J. (2001) An implant for chronic selective stimulation
rials. J. Urol., 171: 2438–2444. of nerves. Med. Eng. Phys., 23: 29–36.
Benda, B.J., Houdayer, T., Creasey, G., Betz, R.R., Smith, Buller, J., Cundiff, G., Noel, K., Van Rooyen, J., Leffler, K.,
B.T., Johnston, T.E. and Davis, R. (2003) The Praxis FES Ellerkman, M. and Bent, A. (2002) RF BION(TM): an in-
system and bladder/bowel management in patients with jectable microstimulator for the treatment of overactive blad-
spinal cord injury. In: Meadows P. (Ed.), IFESS Conference der disorders in adult females. Eur. Urol. Suppl., 1: 40.
Proceedings, Brisbane, Australia. Burghele, T. (1973) Electrostimulation of the neurogenic uri-
Blaivas, J.G., Sinha, H.P., Zayed, A.A. and Labib, K.B. (1981) nary bladder. In: Lutzeyer W. and Melchior H. (Eds.), Uro-
Detrusor–external sphincter dyssynergia: a detailed elect- dynamics: Upper and Lower Urinary Tract. Springer, Berlin,
romyographic study. J. Urol., 125: 545–548. pp. 319–322.
Blok, B.F. (2002) Central pathways controlling micturition and Buss, R.R. and Shefchyk, S.J. (2003) Sacral dorsal horn neurone
urinary continence. Urology, 59: 13–17. activity during micturition in the cat. J. Physiol., 551: 387–396.
Blok, B.F., de Weerd, H. and Holstege, G. (1997) The pontine Bycroft, J.A., Craggs, M.D., Sheriff, M., Knight, S. and Shah,
micturition center projects to sacral cord GABA immunore- P.J. (2004) Does magnetic stimulation of sacral nerve roots
active neurons in the cat. Neurosci. Lett., 233: 109–112. cause contraction or suppression of the bladder? Neurourol.
Blok, B.F., van Maarseveen, J.T. and Holstege, G. (1998) Elec- Urodyn., 23: 241–245.
trical stimulation of the sacral dorsal gray commissure evokes
Cardenas, D.D., Farrell-Roberts, L., Sipski, M.L. and Rubner,
relaxation of the external urethral sphincter in the cat. Ne- D. (1995) Management of gastrointestinal, genitourinary,
urosci. Lett., 249: 68–70. and sexual function. In: Stover S.L., DeLisa J.A. and
Bosch, J.L. and Groen, J. (2000) Sacral nerve neuromodulation Whiteneck G.G. (Eds.), Spinal Cord Injury: Clinical Out-
in the treatment of patients with refractory motor urge in- comes from the Model Systems. Aspen Publishers Inc., Gait-
continence: long-term results of a prospective longitudinal hersburg, pp. 120–144.
study. J. Urol., 163: 1219–1222. Carter, R.R., McCreery, D.B., Woodford, B.J., Bullara, L.A.
Boyce, W.H., Lathem, J.E. and Hunt, L.D. (1964) Research and Agnew, W.F. (1995) Micturition control by microstim-
related to the development of an artificial electrical stimula- ulation of the sacral spinal cord of the cat: acute studies.
tor for the paralyzed human bladder: a review. J. Urol., 91: Rehab. Eng., IEEE Trans., 3: 206–214.
41–51. Chancellor, M.B., Bennett, C., Simoneau, A.R., Finocchiaro,
Boyer, S., Sawan, M., Abdel-Gawad, M., Robin, S. and M.V., Kline, C., Bennett, J.K., Foote, J.E., Green, B.G.,
Elhilali, M.M. (2000) Implantable selective stimulator to im- Martin, S.H., Killoran, R.W., Crewalk, J.A. and Rivas, D.A.
prove bladder voiding: design and chronic experiments in (1999a) Sphincteric stent versus external sphincterotomy in
dogs. IEEE Trans. Rehab. Eng., 8: 464–470. spinal cord injured men: prospective randomized multicenter
Bradley, W.E., Chou, S.N. and French, L.A. (1963) Further trial. J. Urol., 161: 1893–1898.
experience with the radio transmitter receiver unit for the Chancellor, M.B., Gajewski, J., Ackman, C.F., Appell, R.A.,
neurogenic bladder. J. Neurosurg., 20: 953–960. Bennett, J., Binard, J., Boone, T.B., Chetner, M.P., Crewalk,
Bradley, W.E., Wittmers, L.E., Chou, S.N. and French, L.A. J.A., Defalco, A., Foote, J., Green, B., Juma, S., Jung, S.Y.,
(1962) Use of a radio transmitter receiver unit for the treat- Linsenmeyer, T.A., MacMillan, R., Mayo, M., Ozawa, H.,
ment of neurogenic bladder. A preliminary report. J. Ne- Roehrborn, C.G., Shenot, P.J., Stone, A., Vazquez, A.,
urosurg., 19: 782–786. Killorin, W. and Rivas, D.A. (1999b) Long-term followup of
Brindley, G.S. (1977) An implant to empty the bladder or the North American multicenter UroLume trial for the treat-
close the urethra. J. Neurol. Neurosurg. Psychiat., 40: ment of external detrusor–sphincter dyssynergia. J. Urol.,
358–369. 161: 1545–1550.
190

Chartier-Kastler, E.J., Denys, P., Chancellor, M.B., Haertig, Ebner, A., Jiang, C. and Lindstrom, S. (1992) Intravesical elec-
A., Bussel, B. and Richard, F. (2001) Urodynamic monitor- trical stimulation — an experimental analysis of the mech-
ing during percutaneous sacral nerve neurostimulation in anism of action. J. Urol., 148: 920–924.
patients with neurogenic detrusor hyperreflexia. Neurourol. Egon, G., Barat, M., Colombel, P., Visentin, C., Isambert, J.L.
Urodyn., 20: 61–71. and Guerin, J. (1998) Implantation of anterior sacral root
Chartier-Kastler, E.J., Thomas, L., Bussel, B., Chancellor, stimulators combined with posterior sacral rhizotomy in spi-
M.B., Richard, F. and Denys, P. (2000) A urethral stent for nal injury patients. World J. Urol., 16: 342–349.
the treatment of detrusor-striated sphincter dyssynergia. BJU Fall, M. and Lindstrom, S. (1991) Electrical stimulation. A
Int., 86: 52–57. physiologic approach to the treatment of urinary inconti-
Comarr, A.E. (1972) Intermittent catheterization for the trau- nence. Urol. Clin. North Am., 18: 393–407.
matic cord bladder patient. J. Urol., 108: 79–81. Fang, Z.P. and Mortimer, J.T. (1991) Selective activation of
Creasey, G.H. (1993) Electrical stimulation of sacral roots for small motor axons by quasi-trapezoidal current pulses. IEEE
micturition after spinal cord injury. Urol. Clin. North Am., Trans. Biomed. Eng., 38: 168–174.
20: 505–515. Fawcett, J. (2002) Repair of spinal cord injuries: where are we,
Creasey, G.H. and Dahlberg, J.E. (2001) Economic conse- where are we going? Spinal Cord, 40: 615–623.
quences of an implanted neuroprosthesis for bladder and Fjorback, M.V., Hansen, J., Dalmose, A.L., Rijkhoff, N.J.M.
bowel management. Arch. Phys. Med. Rehab., 82: and Sinkjaer, T. (2003) A portable device for experimental
1520–1525. treatment of neurogenic detrusor overactivity. Neuromodu-
Dahlberg, A., Perttila, I., Wuokko, E. and Ala-Opas, M. (2004) lation, 6: 158–165.
Bladder management in persons with spinal cord lesion. Spi- Foley, F.E.B. (1947) An artificial sphincter: a new device and
nal Cord, 42: 694–698. operation for control of enuresis and urinary incontinence.
Dalmose, A.L., Rijkhoff, N.J., Kirkeby, H.J., Nohr, M., J. Urol., 58: 250–259.
Sinkjaer, T. and Djurhuus, J.C. (2003) Conditional stimula- Frankel, H.L., Coll, J.R., Charlifue, S.W., Whiteneck, G.G.,
tion of the dorsal penile/clitoral nerve may increase cysto- Gardner, B.P., Jamous, M.A., Krishnan, K.R., Nuseibeh, I.,
metric capacity in patients with spinal cord injury. Savic, G. and Sett, P. (1998) Long-term survival in spinal cord
Neurourol. Urodyn., 22: 130–137. injury: a fifty year investigation. Spinal Cord, 36: 266–274.
Davis, R., Houdayer, T., Andrews, B. and Barriskill, A. (1999) Friedman, H., Nashold Jr., B.S. and Senechal, P. (1972) Spinal
Paraplegia: prolonged standing using closed-loop functional cord stimulation and bladder function in normal and para-
electrical stimulation and Andrews ankle-foot orthosis. Artif. plegic animals. J. Neurosurg., 36: 430–437.
Organs, 23: 418–420. Geirsson, G. and Fall, M. (1997) Maximal functional electrical
Davis, R., Houdayer, T., Andrews, B., Emmons, S. and stimulation in routine practice. Neurourol. Urodyn., 16:
Patrick, J. (1997) Paraplegia: prolonged closed-loop stand- 559–565.
ing with implanted nucleus FES-22 stimulator and Andrews’ Gladh, G., Mattsson, S. and Lindstrom, S. (2003) In-
foot-ankle orthosis. Stereotact. Funct. Neurosurg., 69: travesical electrical stimulation in the treatment of micturit-
281–287. ion dysfunction in children. Neurourol. Urodyn., 22:
Davis, R., MacFarland, W.C. and Emmons, S.E. (1994) Initial 233–242.
results of the nucleus FES-22-implanted system for limb Govier, F.E., Litwiller, S., Nitti, V., Kreder Jr., K.J. and
movement in paraplegia. Stereotact. Funct. Neurosurg., 63: Rosenblatt, P. (2001) Percutaneous afferent neuromodulation
192–197. for the refractory overactive bladder: results of a multicenter
Davis, R., Patrick, J. and Barriskill, A. (2001) Development of study. J. Urol., 165: 1193–1198.
functional electrical stimulators utilizing cochlear implant Grill, W.M. (2000) Electrical activation of spinal neural circuits:
technology. Med. Eng. Phys., 23: 61–68. application to motor-system neural prostheses. Neuromod-
de Araujo, C.G., Schmidt, R.A. and Tanagho, E.A. (1982) ulation, 3: 97–106.
Neural pathways to lower urinary tract identified by retro- Grill, W.M., Bhadra, N. and Wang, B. (1999) Bladder and
grade axonal transport of horseradish peroxidase. Urology, urethral pressures evoked by microstimulation of the sacral
19: 290–295. spinal cord in cats. Brain Res., 836: 19–30.
de Groat, W.C. (1993) Anatomy and physiology of the lower Grill, W.M., Craggs, M.D., Foreman, R.D., Ludlow, C.L. and
urinary tract. Urol. Clin. North Am., 20: 383–401. Buller, J.L. (2001) Emerging clinical applications of electrical
de Groat, W.C., Fraser, M.O., Yoshiyama, M., Smerin, S., Tai, stimulation: opportunities for restoration of function. J. Re-
C., Chancellor, M.B., Yoshimura, N. and Roppolo, J.R. hab. Res. Dev., 38: 641–653.
(2001) Neural control of the urethra. Scand. J. Urol. Ne- Grill, W.M., Wang, B., Hadziefendic, S. and Haxhiu, M.A.
phrol. Suppl. 207: 35–43; discussion 106–125. (1998) Identification of the spinal neural network involved in
de Groat, W.C., Vizzard, M.A., Araki, I. and Roppolo, J. (1996) coordination of micturition in the male cat. Brain Res., 796:
Spinal interneurons and preganglionic neurons in sacral au- 150–160.
tonomic reflex pathways. Prog. Brain Res., 107: 97–111. Grimes, J.H., Nashold, B.S. and Anderson, E.E. (1975) Clinical
Decter, R.M. (2000) Intravesical electrical stimulation of the application of electronic bladder stimulation in paraplegics.
bladder: con. Urology, 56: 5–8. J. Urol., 113: 338–340.
191

Groen, J., Amiel, C. and Bosch, R. (2004) Chronic puden- Holmquist, B. and Olin, T. (1968b) Electromicturition in male
dal nerve neuromodulation using an implantable mini- dogs at pelvic nerve stimulation. An urethrocystographic
stimulator in women with idiopathic refractory detrusor study. Scand. J. Urol. Nephrol., 2: 115–127.
overactivity incontinence. International Continence Society, Houdayer, T., Barriskill, A., Davis, R., Milijasevic, Z. and
Paris. Cousins, R. (2002) The Praxis and Minax FES systems for
Groen, J. and Bosch, J.L. (2001) Neuromodulation techniques functional restoration in SCI. Proceedings of the IEEE 28th
in the treatment of the overactive bladder. BJU Int., 87: Annual Northeast Bioengineering Conference, Philadelphia,
723–731. PA, pp. 189–190.
Grunewald, V., Bhadra, N., Creasey, G.H. and Mortimer, J.T. Ishigooka, M., Suzuki, Y., Hashimoto, T., Sasagawa, I., Nakada,
(1998) Functional conditions of micturition induced by T. and Handa, Y. (1998) A new technique for sacral nerve
selective sacral anterior root stimulation: experimental stimulation: a percutaneous method for urinary incontinence
results in a canine animal model. World J. Urol., 16: caused by spinal cord injury. Br. J. Urol., 81: 315–318.
329–336. Jamil, F. (2001) Towards a catheter free status in neurogenic
Gustafson, K.J., Creasey, G.H. and Grill, W.M. (2003) A cath- bladder dysfunction: a review of bladder management op-
eter based method to activate urethral sensory nerve fibers. tions in spinal cord injury (SCI). Spinal Cord, 39: 355–361.
J. Urol., 170: 126–129. Jezernik, S., Craggs, M., Grill, W.M., Creasey, G. and Rijkhoff,
Gustafson, K.J., Creasey, G.H. and Grill, W.M. (2004) A ure- N.J. (2002) Electrical stimulation for the treatment of bladder
thral afferent mediated excitatory bladder reflex exists in hu- dysfunction: current status and future possibilities. Neurol.
mans. Neurosci. Lett., 360: 9–12. Res., 24: 413–430.
Guttmann, L. and Frankel, H. (1966) The value of intermittent Jezernik, S., Wen, J.G., Rijkhoff, N.J., Djurhuus, J.C. and
catheterisation in the early management of traumatic para- Sinkjaer, T. (2000) Analysis of bladder related nerve cuff
plegia and tetraplegia. Paraplegia, 4: 63–84. electrode recordings from preganglionic pelvic nerve and
Hajivassiliou, C.A. (1998) The development and evolution of sacral roots in pigs. J. Urol., 163: 1309–1314.
artificial urethral sphincters. J. Med. Eng. Technol., 22: Jonas, U., Heine, J.P. and Tanagho, E.A. (1975) Studies on
154–159. feasibility of urinary bladder evacuation by direct spinal cord
Hald, T. (1969) Neurogenic dysfunction of the urinary bladder. stimulation. I. Parameters of most effective stimulation. In-
An experimental and clinical study with special reference to vest. Urol., 13: 142–150.
the ability of electrical stimulation to establish voluntary mi- Jonas, U. and Tanagho, E.A. (1975) Studies on feasibility of
cturition. Dan. Med. Bull., 16(Suppl 5): 1+. urinary bladder evacuation by direct spinal cord stimulation
Hald, T., Meier, W., Khalili, A., Agrawal, G., Benton, J.G. and II Poststimulus voiding: A way to overcome outflow resist-
Kantrowitz, A. (1967) Clinical experience with a radio-linked ance. Invest. Urol., 13: 151–153.
bladder stimulator. J. Urol., 97: 73–78. Juan Garcia, F.J., Salvador, S., Montoto, A., Lion, S., Balvis,
Hall, S.W. (2003) Commercializing neuroprostheses: the busi- B., Rodriguez, A., Fernandez, M. and Sanchez, J. (1999) In-
ness of putting the brain back in business. Molecular Biology. traurethral stent prosthesis in spinal cord injured patients
Princeton University, Princeton, p. 185. with sphincter dyssynergia. Spinal Cord, 37: 54–57.
Hamid, R., Arya, M., Wood, S., Patel, H.R. and Shah, P.J. Kantrowitz, A. and Schamaun, M. (1963) Paraplegic dogs: uri-
(2003) The use of the Memokath stent in the treatment of nary bladder evacuation with direct electric stimulation. Sci-
detrusor sphincter dyssynergia in spinal cord injury patients: ence, 139: 115–116.
a single-centre seven-year experience. Eur. Urol., 43: Kaplan, W.E. (2000) Intravesical electrical stimulation of the
539–543. bladder: pro. Urology, 56: 2–4.
Hansen, R.B., Biering-Sorensen, F. and Kristensen, J.K. (2004) Kennedy, R.H. (1946) The new viewpoint toward spinal cord
Bladder emptying over a period of 10–45 years after a trau- injuries. Ann. Surg., 124: 1057–1065.
matic spinal cord injury. Spinal Cord, 42: 631–637. Kirkham, A.P., Knight, S.L., Craggs, M.D., Casey, A.T. and
Heine, J.P., Schmidt, R.A. and Tanagho, E.A. (1977) Intraspi- Shah, P.J. (2002) Neuromodulation through sacral nerve
nal sacral root stimulation for controlled micturition. Invest. roots 2 to 4 with a Finetech–Brindley sacral posterior and
Urol., 15: 78–82. anterior root stimulator. Spinal Cord, 40: 272–281.
Hohenfellner, M., Humke, J., Hampel, C., Dahms, S., Matzel, Kirkham, A.P., Shah, N.C., Knight, S.L., Shah, P.J. and Crag-
K., Roth, S., Thuroff, J.W. and Schultz-Lampel, D. (2001) gs, M.D. (2001) The acute effects of continuous and condi-
Chronic sacral neuromodulation for treatment of neurogenic tional neuromodulation on the bladder in spinal cord injury.
bladder dysfunction: long-term results with unilateral im- Spinal Cord, 39: 420–428.
plants. Urology, 58: 887–892. Koldewijn, E.L., Rijkhoff, N.J., van Kerrebroeck, E.V., De-
Hollander, J.B. and Diokno, A.C. (1993) Urinary diversion and bruyne, F.M. and Wijkstra, H. (1994) Selective sacral root
reconstruction in the patient with spinal cord injury. Urol. stimulation for bladder control: acute experiments in an an-
Clin. North Am., 20: 465–474. imal model. J. Urol., 151: 1674–1679.
Holmquist, B. and Olin, T. (1968a) Electromicturition in female Kondo, A., Otani, T. and Takita, T. (1982) Suppression of
dogs at pelvic nerve stimulation. An urethrocystographic bladder instability by penile squeeze. Br. J. Urol., 54:
study. Scand. J. Urol. Nephrol., 2: 129–135. 360–362.
192

Kuru, M. (1965) Nervous control of micturition. Physiol. Rev., Merrill, D.C. and Conway, C.J. (1974) Clinical experience with
45: 425–494. the Mentor bladder stimulation. I. Patients with upper motor
Lapides, J., Diokno, A.C., Silber, S.J. and Lowe, B.S. (1972) neuron lesions. J. Urol., 112: 52–56.
Clean, intermittent self-catheterization in the treatment of Middleton, J.W. and Keast, J.R. (2004) Artificial autonomic
urinary tract disease. J. Urol., 107: 458–461. reflexes: using functional electrical stimulation to mimic blad-
Lee, T. (1997) Electrical stimulation techniques to restore blad- der reflexes after injury or disease. Auton Neurosci, 113: 3–15.
der function. Neurosurg. Quart., 7: 264–272. Middleton, J.W., Lim, K., Taylor, L., Soden, R. and Rutkowski,
Lee, Y.H. and Creasey, G.H. (2002) Self-controlled dorsal pe- S. (2004) Patterns of morbidity and rehospitalisation follow-
nile nerve stimulation to inhibit bladder hyperreflexia in in- ing spinal cord injury. Spinal Cord, 42: 359–367.
complete spinal cord injury: a case report. Arch. Phys. Med. Moore, T. and Schofield, P.F. (1967) Treatment of stress in-
Rehab., 83: 273–277. continence by maximum perineal electrical stimulation. Br.
Lee, Y.H., Creasey, G.H., Lim, H., Song, J., Song, K. and Kim, Med. J., 3: 150–151.
J. (2003) Detrusor and blood pressure responses to dorsal Mushahwar, V.K., Collins, D.F. and Prochazka, A. (2000)
penile nerve stimulation during hyperreflexic contraction of Spinal cord microstimulation generates functional limb
the bladder in patients with cervical cord injury. Arch. Phys. movements in chronically implanted cats. Exp. Neurol.,
Med. Rehab., 84: 136–140. 163: 422–429.
Light, J.K. and Scott, F.B. (1983) Use of the artificial urinary Nacey, J. and Delahunt, B. (1993) The evolution and develop-
sphincter in spinal cord injury patients. J. Urol., 130: ment of the urinary catheter. Aust. N. Z. J. Surg., 63:
1127–1129. 815–819.
Lin, V.W., Wolfe, V., Frost, F.S. and Perkash, I. (1997) Mi- Nakamura, M. and Sakurai, T. (1984) Bladder inhibition by
cturition by functional magnetic stimulation. J. Spinal Cord penile electrical stimulation. Br. J. Urol., 56: 413–415.
Med., 20: 218–226. Nashold Jr., B.S., Friedman, H. and Boyarsky, S. (1971) Elec-
Loeb, G.E., Peck, R.A., Moore, W.H. and Hood, K. (2001) trical activation of micturition by spinal cord stimulation.
BION system for distributed neural prosthetic interfaces. J. Surg. Res., 11: 144–147.
Med. Eng. Phys., 23: 9–18. Nashold Jr., B.S., Friedman, H., Glenn, J.F., Grimes, J.H.,
Low, A.I. and McRae, P.J. (1998) Use of the Memokath for Barry, W.F. and Avery, R. (1972) Electromicturition in par-
detrusor–sphincter dyssynergia after spinal cord injury — a aplegia. Implantation of a spinal neuroprosthesis. Arch.
cautionary tale. Spinal Cord, 36: 39–44. Surg., 104: 195–202.
Madersbacher, H. (1990) Intravesical electrical stimulation for Nashold Jr., B.S., Friedman, H. and Grimes, J. (1981) Electrical
the rehabilitation of the neuropathic bladder. Paraplegia, 28: stimulation of the conus medullaris to control the bladder in
349–352. the paraplegic patient. A 10-year review. Appl. Neurophysiol.,
Madersbacher, H., Pauer, W. and Reiner, E. (1982) Rehabil- 44: 225–232.
itation of micturition by transurethral electrostimulation of Nativ, O., Moskowitz, B., Issaq, E., Condrea, A., Kastin, A.,
the bladder in patients with incomplete spinal cord lesions. Halachmi, S., Burbara, J., Madjar, S. and Beyar, M. (1997) A
Paraplegia, 20: 191–195. new intraurethral sphincter prosthesis with a self contained
Madjar, S., Sabo, E., Halachmi, S., Wald, M., Issaq, E., urinary pump. ASAIO J., 43: 197–203.
Moskovitz, B., Beyar, M. and Nativ, O. (1999) A remote Nissenkorn, I., Shalev, M., Radziszewski, P., Dobronski, P.,
controlled intraurethral insert for artificial voiding: a new Borkowski, A. and De Jong, P.R. (2004) Patient-adjusted
concept for treating women with voiding dysfunction. intermittent electrostimulation for treating stress and urge
J. Urol., 161: 895–898. urinary incontinence. BJU Int., 94: 105–109.
Magasi, P. and Simon, Z. (1986) Electrical stimulation of the Okada, N., Igawa, Y., Ogawa, A. and Nishizawa, O. (1998)
bladder and gravidity. Urol. Int., 41: 241–245. Transcutaneous electrical stimulation of thigh muscles in the
Mazouni, C., Karsenty, G., Bladou, F. and Serment, G. (2004) treatment of detrusor overactivity. Br. J. Urol., 81: 560–564.
Urethral device in women with chronic urinary retention: an Petrou, S.P., Elliott, D.S. and Barrett, D.M. (2000) Artificial
alternative to self-catheterization? Eur. J. Obstet. Gynecol. urethral sphincter for incontinence. Urology, 56: 353–359.
Reprod. Biol., 115: 80–84. Previnaire, J.G., Soler, J.M. and Perrigot, M. (1998) Is there a
McCreery, D., Pikov, V., Lossinsky, A., Bullara, L. and place for pudendal nerve maximal electrical stimulation for
Agnew, W. (2004) Arrays for chronic functional microstim- the treatment of detrusor hyperreflexia in spinal cord injury
ulation of the lumbosacral spinal cord. IEEE Trans. Neural. patients? Spinal Cord, 36: 100–103.
Syst. Rehab. Eng., 12: 195–207. Prochazka, A., Gaunt, R.G., Mushahwar, V., Downie, J.W.
McCreery, D.B., Agnew, W.F., Yuen, T.G. and Bullara, L. and Shefchyk, S.J. (2003a) Functional microstimulation of
(1990) Charge density and charge per phase as cofactors in the lumbosacral spinal cord: Quarterly Report #6. NIH-NI-
neural injury induced by electrical stimulation. IEEE Trans. NDS contract N01-NS-2-2342.
Biomed. Eng., 37: 996–1001. Prochazka, A., Mushahwar, V., Downie, J.W. and Shefchyk,
McGuire, E.J., Zhang, S.C., Horwinski, E.R. and Lytton, B. S.J. (2002a) Functional microstimulation of the lumbosacral
(1983) Treatment of motor and sensory detrusor instability spinal cord: Quarterly Report #1. NIH-NINDS contract
by electrical stimulation. J. Urol., 129: 78–79. N01-NS-2-2342.
193

Prochazka, A., Mushahwar, V., Downie, J.W., Shefchyk, S.J. Schmidt, R.A. (1986) Advances in genitourinary neurostimula-
and Gaunt, R.G. (2003b) Functional microstimulation of the tion. Neurosurgery, 19: 1041–1044.
lumbosacral spinal cord: Quarterly Report #5. NIH-NINDS Schmidt, R.A. (1988) Applications of neurostimulation in urol-
contract N01-NS-2-2342. ogy. Neurourol. Urodyn., 7: 585–592.
Prochazka, A., Mushahwar, V. and Yakovenko, S. (2002b) Schmidt, R.A., Bruschini, H. and Tanagho, E.A. (1979) Uri-
Activation and coordination of spinal motoneuron pools af- nary bladder and sphincter responses to stimulation of dorsal
ter spinal cord injury. Prog. Brain Res., 137: 109–124. and ventral sacral roots. Invest. Urol., 16: 300–304.
Prochazka, A., Westerman, R.A. and Ziccone, S.P. (1976) Dis- Schmidt, R.A., Senn, E. and Tanagho, E.A. (1990) Functional
charges of single hindlimb afferents in the freely moving cat. evaluation of sacral nerve root integrity. Report of a tech-
J. Neurophysiol., 39: 1090–1104. nique. Urology, 35: 388–392.
Reynard, J.M., Vass, J., Sullivan, M.E. and Mamas, M. (2003) Schurch, B., Reilly, I., Reitz, A. and Curt, A. (2003) Electro-
Sphincterotomy and the treatment of detrusor–sphincter physiological recordings during the peripheral nerve evalua-
dyssynergia: current status, future prospects. Spinal Cord, 41: tion (PNE) test in complete spinal cord injury patients.
1–11. World J. Urol., 20: 319–322.
Rijkhoff, N.J. (2004a) Emerging FES applications for control Schurch, B., Suter, S. and Dubs, M. (1999) Intraurethral
of the urinary bladder. In: Horch K.W. and Dhillon sphincter prosthesis to treat hyporeflexic bladders in women:
G.S. (Eds.) Neuroprosthetics: Theory and Practice, Vol. 2. does it work? BJU Int., 84: 789–794.
World Scientific Publishing Co. Ltd, Singapore, pp. Scott, F.B., Bradley, W.E. and Timm, G.W. (1974) Treatment
1054–1066. of urinary incontinence by an implantable prosthetic urinary
Rijkhoff, N.J. (2004b) Neuroprostheses to treat neurogenic sphincter. J. Urol., 112: 75–80.
bladder dysfunction: current status and future perspectives. Selzman, A.A. and Hampel, N. (1993) Urologic complications
Childs Nerv. Syst., 20: 75–86. of spinal cord injury. Urol. Clin. North Am., 20: 453–464.
Rijkhoff, N.J., Holsheimer, J., Koldewijn, E.L., Struijk, J.J., Shafik, A., el-Sibai, O. and Ahmed, I. (2003a) Effect of urethral
van Kerrebroeck, P.E., Debruyne, F.M. and Wijkstra, H. dilation on vesical motor activity: identification of the
(1994a) Selective stimulation of sacral nerve roots for bladder urethrovesical reflex and its role in voiding. J. Urol., 169:
control: a study by computer modeling. IEEE Trans. Bio- 1017–1019.
med. Eng., 41: 413–424. Shafik, A., Shafik, A.A., El-Sibai, O. and Ahmed, I. (2003b)
Rijkhoff, N.J., Koldewijn, E.L., Van Kerrebroeck, P.E., Role of positive urethrovesical feedback in vesical evacua-
Debruyne, F.M. and Wijkstra, H. (1994b) Acute animal stud- tion. The concept of a second micturition reflex: the
ies on the use of anodal block to reduce urethral resistance in urethrovesical reflex. World J. Urol., 21: 167–170.
sacral root stimulation. IEEE Trans. Rehab. Eng., 2: 92–99. Shaker, H.S., Tu, L.M., Robin, S., Arabi, K., Hassouna, M.,
Rijkhoff, N.J., Wijkstra, H., van Kerrebroeck, P.E. and Sawan, M. and Elhilali, M.M. (1998) Reduction of bladder
Debruyne, F.M. (1997a) Selective detrusor activation by outlet resistance by selective sacral root stimulation using
electrical sacral nerve root stimulation in spinal cord injury. high-frequency blockade in dogs: an acute study. J. Urol.,
J. Urol., 157: 1504–1508. 160: 901–907.
Rijkhoff, N.J., Wijkstra, H., van Kerrebroeck, P.E. and Shaw, P.J., Milroy, E.J., Timoney, A.G., el Din, A. and
Debruyne, F.M. (1997b) Urinary bladder control by electri- Mitchell, N. (1990) Permanent external striated sphincter
cal stimulation: review of electrical stimulation techniques in stents in patients with spinal injuries. Br. J. Urol., 66: 297–302.
spinal cord injury. Neurourol. Urodyn., 16: 39–53. Shefchyk, S.J. (2001) Sacral spinal interneurones and the con-
Rijkhoff, N.J., Wijkstra, H., van Kerrebroeck, P.E. and trol of urinary bladder and urethral striated sphincter muscle
Debruyne, F.M. (1998) Selective detrusor activation by function. J. Physiol., 533: 57–63.
sacral ventral nerve-root stimulation: results of intraoperative Shefchyk, S.J. and Buss, R.R. (1998) Urethral pudendal affer-
testing in humans during implantation of a Finetech–Brindley ent-evoked bladder and sphincter reflexes in decerebrate and
system. World J. Urol., 16: 337–341. acute spinal cats. Neurosci. Lett., 244: 137–140.
Sauerwein, D., Ingunza, W., Fischer, J., Madersbacher, H., Shindo, N. and Jones, R. (1987) Reciprocal patterned electrical
Polkey, C.E., Brindley, G.S., Colombel, P. and Teddy, P. stimulation of the lower limbs in severe spasticity. Physio-
(1990) Extradural implantation of sacral anterior root therapy, 73: 579–582.
stimulators. J. Neurol. Neurosurg. Psychiat., 53: 681–684. Sie, J.A., Blok, B.F., de Weerd, H. and Holstege, G. (2001)
Savic, G., Short, D.J., Weitzenkamp, D., Charlifue, S. and Ultrastructural evidence for direct projections from the pon-
Gardner, B.P. (2000) Hospital readmissions in people with tine micturition center to glycine-immunoreactive neurons in
chronic spinal cord injury. Spinal Cord, 38: 371–377. the sacral dorsal gray commissure in the cat. J. Comp. Ne-
Sawan, M. and Elhilali, M.M. (2002) Electronic stimulator im- urol., 429: 631–637.
plant for modulating and synchronizing bladder and sphinc- Siegel, S.W., Catanzaro, F., Dijkema, H.E., Elhilali, M.M.,
ter function. US Pat. 6,393,323. McGill University, Gestion Fowler, C.J., Gajewski, J.B., Hassouna, M.M., Janknegt,
Univalor. R.A., Jonas, U., van Kerrebroeck, P.E., Lycklama a
Schmidt, R.A. (1983) Neural prostheses and bladder control. Nijeholt, A.A., Oleson, K.A. and Schmidt, R.A. (2000)
IEEE Eng. Med. Biol. Mag., 2: 31–36. Long-term results of a multicenter study on sacral nerve
194

stimulation for treatment of urinary urge incontinence, Van Kerrebroeck, P.E., Koldewijn, E.L. and Debruyne, F.M.
urgency-frequency, and retention. Urology, 56: 87–91. (1993) Worldwide experience with the Finetech–Brindley
Smith, B.T., Betz, R.R., Johnston, T., Benda, B., Mulcahey, sacral anterior root stimulator. Neurourol. Urodyn., 12:
M.J., Davis, R. and Barriskill, A. (2002) Initial experiences 497–503.
with the Praxis multi-functional implantable FES system for Vandoninck, V., van Balken, M.R., Finazzi Agro, E., Petta, F.,
individuals with spinal cord injury. FESnet http: Micali, F., Heesakkers, J.P., Debruyne, F.M., Kiemeney,
//fesnet.eng.gla.ac.uk/conference/x_smithb.pdf.PDF. L.A. and Bemelmans, B.L. (2003) Percutaneous tibial nerve
Solomonow, M., Eldred, E., Lyman, J. and Foster, J. (1983) stimulation in the treatment of overactive bladder: urody-
Control of muscle contractile force through indirect high- namic data. Neurourol. Urodyn., 22: 227–232.
frequency stimulation. Am. J. Phys. Med., 62: 71–82. Vodovnik, L. (1981) Therapeutic effects of functional electrical
Stenberg, C.C., Burnette, H.W. and Bunts, R.C. (1967) Elec- stimulation of extremities. Med. Biol. Eng. Comput., 19:
trical stimulation of human neurogenic bladders: experience 470–478.
with 4 patients. J. Urol., 97: 79–84. Vuckovic, A. and Rijkhoff, N.J. (2004) Different pulse shapes
Stewart, C.C. (1899) On the course of impulses to and from the for selective large fibre block in sacral nerve roots using a
cat’s bladder. Am. J. Physiol., 2: 182–202. technique of anodal block: an experimental study. Med. Biol.
Susset, J.G. and Boctor, Z.N. (1967) Implantable electrical Eng. Comput., 42: 817–824.
vesical stimulator: clinical experience. J. Urol., 98: 673–678. Walter, J.S., Wheeler, J.S., Cai, W., King, W.W. and Wurster,
Talalla, A., Bloom, J.W. and Quang, N. (1987) FES for blad- R.D. (1999) Evaluation of a suture electrode for direct blad-
der: direct or indirect means? Pacing Clin. Electrophysiol., der stimulation in a lower motor neuron lesioned animal
10: 240–245. model. IEEE Trans. Rehab. Eng., 7: 159–166.
Tanagho, E.A. and Schmidt, R.A. (1988) Electrical stimulation Wein, A.J. (1998) Pharmacologic options for the overactive
in the clinical management of the neurogenic bladder. bladder. Urology, 51: 43–47.
J. Urol., 140: 1331–1339. Weld, K.J. and Dmochowski, R.R. (2000) Effect of bladder
Tanagho, E.A., Schmidt, R.A. and Orvis, B.R. (1989) Neural management on urological complications in spinal cord in-
stimulation for control of voiding dysfunction: a preliminary jured patients. J. Urol., 163: 768–772.
report in 22 patients with serious neuropathic voiding disor- Wheeler Jr., J.S., Robinson, C.J., Culkin, D.J. and Bolan, J.M.
ders. J. Urol., 142: 340–345. (1986) The effect of thigh muscle reconditioning by electrical
Timm, G.W., Bradley, W.E. and Scott, F.B. (1974) Experi- stimulation on urodynamic activity in SCI patients. J. Am.
mental evaluation of an implantable externally controllable Paraplegia Soc., 9: 16–23.
urinary sphincter. Invest. Urol., 11: 326–330. Wheeler Jr., J.S., Walter, J.S. and Zaszczurynski, P.J. (1992)
van Balken, M.R., Vandoninck, V., Gisolf, K.W., Vergunst, H., Bladder inhibition by penile nerve stimulation in spinal cord
Kiemeney, L.A., Debruyne, F.M. and Bemelmans, B.L. (2001) injury patients. J. Urol., 147: 100–103.
Posterior tibial nerve stimulation as neuromodulative treat- Wilson, T.S., Lemack, G.E. and Dmochowski, R.R. (2002)
ment of lower urinary tract dysfunction. J. Urol., 166: 914–918. UroLume stents: lessons learned. J. Urol., 167: 2477–2480.
van Balken, M.R., Vergunst, H. and Bemelmans, B.L. (2004) Wozniak, W. and Skowronska, U. (1967) Comparative anat-
The use of electrical devices for the treatment of bladder omy of pelvic plexus in cat, dog, rabbit, macaque and man.
dysfunction: a review of methods. J. Urol., 172: 846–851. Anat. Anz., 120: 457–473.
Van Kerrebroeck, P.E. (2002) Neuromodulation and other Yoshimura, N. (1999) Bladder afferent pathway and spinal
electrostimulatory techniques. Scand. J. Urol. Nephrol., Sup- cord injury: possible mechanisms inducing hyperreflexia of
pl. 210: 82–86. the urinary bladder. Prog. Neurobiol., 57: 583–606.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 12

Novel repair strategies to restore bladder function


following cauda equina/conus medullaris injuries

Thao X. Hoang and Leif A. Havton

Department of Neurology, David Geffen School of Medicine at University of California Los Angeles, 710 Westwood Plaza,
Los Angeles, CA 90095-1769, USA

Abstract: Trauma to the thoracolumbar junction or lumbosacral spine may result in a conus medullaris or
cauda equina syndrome. In both conditions, symptoms typically include paraparesis or paraplegia, sensory
impairment, pain, as well as bladder, bowel, and sexual dysfunctions. We present in this review a series of
neural repair strategies that have been developed to address the unique features and challenges of subjects
with a conus medullaris or cauda equina syndrome. We address, in particular, neural repair strategies that
may have a translational research potential to restore bladder function. Recent animal injury models have
suggested that a progressive retrograde death of both autonomic and motor neurons may contribute to the
neurological deficits in subjects with conus medullaris and cauda equina injuries. For subjects with acute
injuries, we present novel strategies to promote neuroprotection, axonal regeneration, and functional
reinnervation of the lower urinary tract. For subjects with chronic injuries, we discuss new approaches to
replace lost autonomic and motor neurons. A brief discussion on a variety of outcome measures that may
be suitable to evaluate the function of the lower urinary tract in rodent neural repair models is also
provided.

Introduction dysfunction. Bladder dysfunction may include loss


of voluntary micturition, urethral closure impair-
In humans, trauma to the spine at the thoraco- ment, and an areflexic bladder (Pavlakis et al.,
lumbar junction or to the lumbosacral spine may 1983; Fowler et al., 1984; Light et al., 1993;
result in a conus medullaris or cauda equina syn- Fowler, 1999).
drome. According to the International Standards Occasionally, sacral segments may show pre-
for Neurological and Functional Classification of served reflexes, e.g., the micturition and bulbocav-
Spinal Cord Injury, a conus medullaris syndrome ernosus reflexes. Such reflex sparing may be
consists of an injury to both the sacral cord encountered when the injury affects the most ros-
(conus) and lumbar nerve roots within the spinal tral part of the sacral cord with sparing of the most
canal, while a cauda equina syndrome consists of caudal portion of the sacral spinal cord. In de-
an injury to the lumbosacral nerve roots below the tailed neurologic evaluations of patients with
tip of the conus medullaris (Maynard et al., 1997). conus medullaris and cauda equina lesions, find-
Clinically, injury to the conus medullaris and ca- ings typically include an absent or substantially
uda equina may cause paraparesis or paraplegia, diminished bulbocavernosus reflex, a bladder de-
sensory disturbance, as well as bladder and bowel trusor areflexia on cystometrogram examinations,
and neuropathic perineal electromyographic
Corresponding author. Tel.: +310-206-6500; changes indicated by polyphasic waves, fibrilla-
Fax: +310-794-9486; E-mail: lhavton@mednet.ucla.edu tion potentials, and positive sharp waves (Pavlakis

DOI: 10.1016/S0079-6123(05)52012-0 195


196

et al., 1983). However, a conventional urodynamic and peripheral portions of the nervous system may
evaluation alone is unable to distinguish between a vary between patients with a conus medullaris
pure conus medullaris lesion and a conus lesion syndrome. While the literature on postmortem
with a concomitant cauda equina involvement pathology of traumatic conus medullaris and
(Beric and Light, 1992). cauda equina injuries is scant, certain types of
Conus medullaris and cauda equina injuries are pathologies may be more prevalent in association
complex lesions, which may present with a com- with certain types of trauma. For instance,
bination of pathologies, including intramedullary lumbosacral root avulsion injuries may develop
damage as well as with stretching, tearing, crush, more frequently in association with high-energy
laceration, and avulsion of proximal and distal trauma, e.g., high velocity motor vehicle accidents
portions of nerve roots as a result of, e.g., trau- (Moschilla et al., 2001; Hans et al., 2004; Lang
matic falls, motor vehicle accidents, acts of vio- et al., 2004; Monga et al., 2004). However, animal
lence, herniated discs, and sports injuries (Fig. 1). models to study conus medullaris and cauda
The relative proportion of lesions to the central equina injuries have been relatively sparse.

Effects of axonal lesions on efferent spinal


cord neurons

Injuries to the conus medullaris and lumbosacral


nerve roots may cause central and/or peripheral
lesions to the axons of efferent spinal cord moto-
neurons and preganglionic parasympathetic neu-
rons, as well as to the primary afferents located in
the dorsal roots. Factors such as species, age,
location, and severity of injury may influence the
retrograde response of an axotomized neuron. In
particular, the location of the axonal lesion greatly
affects the fate of preganglionic parasympathetic
neurons and motoneurons. In rats, cats, and
primates, a peripheral nerve transection, spinal
nerve transection or limb amputation resulted in
no or limited motoneuron loss (Carlson et al.,
1979; Johnson et al., 1991; Vanden Noven et al.,
1993; Piehl et al., 1995; Anneser et al., 2000; Wu
and Kaas, 2000; Ma et al., 2001). Similarly,
preganglionic parasympathetic neurons showed
no detectable loss at 2–4 weeks after a peripheral
nerve injury performed by a surgical removal of
the major pelvic ganglion (Vizzard et al., 1995).
In contrast, a cervical or lumbar ventral root
Fig. 1. Magnetic resonance imaging (MRI) study of the lower avulsion injury, alone or combined with a seg-
thoracic and upper lumbar spine of a young adult male fol- mental dorsal root lesion, resulted in a progressive
lowing an acute traumatic injury from a motor vehicle accident. and marked retrograde death of motoneurons in
The MRI scan shows a severe dislocation at the T12–L1 ver- the adult rat (Wu, 1993; Koliatsos et al., 1994;
tebral level with a posterior displacement of the lumbar column,
Novikov et al., 1995, 2000; Kishino et al., 1997;
rupture of the T12–L1 intervertebral disc, and an L1 vertebral
fracture. There is a near obliteration of the spinal canal with Martin et al., 1999) and the adult cat (Holmberg
crush injury to the conus medullaris. SC ¼ spinal cord. and Kellerth, 2000). A more proximal lesion, such
197

as an intramedullary transection of the ventral parasympathetic neurons of the intermediolateral


funiculus, also induced a retrograde death of many nucleus and the somatic motoneurons of the
motoneurons in the adult cat (Lindå et al., 1992). dorsolateral and dorsomedial nuclei innervating
Less information is available on the response of the external urethral and anal sphincters, respec-
autonomic efferent neurons to lesions to the cen- tively (Schrøder, 1980). We demonstrated that an
tral and most proximal portions of their axons. L5–S2 ventral root avulsion injury causes a pro-
gressive and parallel death of axotomized pre-
ganglionic parasympathetic neurons and
A model to study cauda equina/conus medullaris motoneurons (Fig. 3; Hoang et al., 2003).
injuries in the rat Already at 1 week after the lesion, there was a
significant loss of 12% of the preganglionic para-
We have recently developed a conus medullaris sympathetic neurons and 17% of the motoneu-
and cauda equina injury model in the adult rat to rons. The number of undetectable neurons
study the retrograde effects of a proximal lumbo- gradually increased to 78% of the preganglionic
sacral ventral root lesion on axotomized pregangl- parasympathetic neurons and 84% of the moto-
ionic parasympathetic neurons and motoneurons neurons by 6 weeks postoperatively.
with pelvic targets, including the external urethral
and anal sphincters (Hoang et al., 2003). In this
model, we performed a unilateral ventral root av-
ulsion injury of the lumbar (L) and sacral (S), L5,
L6, S1, and S2 segments (Fig. 2). These segmental
levels were selected to include primarily the L6 and
S1 segments, which contain the preganglionic

Fig. 3. Immunohistochemical light stable detection of prelabe-


led Fluorogold (FG, Fluorochrome, Denver, CO) shows close
to a 50% loss of the preganglionic parasympathetic neurons
(PPN) and motoneurons (MN) on the injured side compared to
the contralateral non-lesioned side at 2 weeks after an L5–S2
ventral root avulsion injury in the adult rat (a–d). Rats were
injected intraperitoneally with 400 ml of 0.5% FG at 1 week
prior to the ventral root avulsion injury to label all pregangl-
ionic parasympathetic neurons and motoneurons. Following an
intravascular perfusion with a 4% paraformaldehyde solution,
Fig. 2. Schematic drawing of the unilateral lumbosacral ventral 40 mm spinal cord frozen sections of the L6 and S1 segments
root avulsion injury model. Four consecutive lumbosacral ven- were processed for immunohistochemistry using an antibody
tral roots (L5–S2) are avulsed at the ventral root exit from the against FG (Fluorochrome, Denver, CO) to enhance detection
spinal cord surface. Note that the dorsal roots remain intact. of the fluorescent retrograde tracer. Scale bar ¼ 50 mm for a–d.
198

In our lumbosacral ventral root avulsion model, neurons following proximal lesions to their axons.
we detected nuclear chromatin condensation, The mechanisms for cell death may also be age-
apoptotic bodies, and the activation of caspase-3 dependent, as treatment with caspase inhibitors
in the cytoplasm and nuclei of both pregangl- protected axotomy-induced motoneuron death in
ionic parasympathetic neurons and motoneurons, neonatal rats but not in adult rats (Chan et al.,
suggesting that apoptosis contributes to the retro- 2001).
grade cell death of both preganglionic parasym-
pathetic neurons and motoneurons (Fig. 4; Hoang
et al., 2003). Our findings are in agreement with Expression of nitric oxide synthase in axotomized
other studies that show evidence for apoptotic cell spinal cord neurons
death mechanisms following a sciatic nerve injury
in the adult rat. For instance, axotomized moto- A ventral root transection injury that does not lead
neurons demonstrated fragmentation of nuclear to any significant motoneuron loss, does not in-
DNA, early accumulation of single-strand breaks duce any nitric oxide synthase expression in axo-
(Martin et al., 1999; Liu and Martin, 2001), acti- tomized rat motoneurons (Wu, 1993). However, a
vation of caspase-3, nuclear accumulation of the ventral root avulsion injury induces the expression
apoptosis-inducing factor p53, and increased of nitric oxide synthase in axotomized motoneu-
detection of apoptosis-associated Bax (Martin rons, which are destined to undergo retrograde
and Liu, 2002). However, a ventral root avulsion degeneration and death (Wu, 1993; Novikov et al.,
injury at the cervical level in adult mice induced 1995). An additional association between the in-
ultrastructural changes in motoneurons, including jury-induced expression of nitric oxide synthase
disruption, perturbation, and depletion of organ- and motoneuron death was demonstrated in a
elles, suggestive of a necrotic process (Li et al., pharmacological study, which showed that treat-
1998). Interestingly, nuclear pyknosis, a finding ment with nitroarginine, a specific nitric oxide
suggestive of apoptosis, was also seen in lesioned synthase inhibitor, reduces motoneuron loss fol-
motoneurons at later stages (Li et al., 1998). lowing a spinal nerve root lesion in the rat (Wu
Therefore, it seems possible that both apoptotic and Li, 1993).
and necrotic mechanisms may be involved in In contrast to motoneurons innervating the up-
the retrograde cell death of efferent spinal cord per and lower extremities, about half of the normal
population of preganglionic parasympathetic neu-
rons stain for nicotinamide adenine dinucleotide
phosphate hydrogen (NADPH) diaphorase or
express neuronal nitric oxide synthase in the rat
conus medullaris (Vizzard et al., 1993a; Saito et al.,
1994). A marked increase in the number of
NADPH diaphorase positive preganglionic para-
sympathetic neurons was detected at 1–3 weeks
after a combined ventral and dorsal rhizotomy
(Vizzard et al., 1993b). The number of pregangl-
ionic parasympathetic neurons expressing neuron-
al nitric oxide synthase also increased in the
Fig. 4. Immunohistochemistry for light stable detection of ac- absence of any detectable cell death, at 2–4 weeks
tivated caspase-3 (Promega, Madison, WI) demonstrates pres- following a surgical resection of the major pelvic
ence of the activated caspase-3 in a subset of the remaining ganglion (Vizzard et al., 1995). However, the
parasympathetic preganglionic neurons and motoneurons on
expression of neuronal nitric oxide synthase
the lesioned side at 2 weeks after the avulsion injury (arrows, a,
b). High magnification micrograph demonstrates activated ca- in preganglionic parasympathetic neurons under-
spase-3 being located primarily within the soma (b). Scale going a progressive cell death from a lumbosacral
bar ¼ 300 mm for a; 50 mm for b. ventral root avulsion is not yet known. Because
199

of the above differences between motoneurons ability to regenerate axons into a peripheral nerve
and preganglionic parasympathetic neurons in graft implanted into the spinal cord for up to 3
neuronal nitric oxide synthase expression under weeks, after a cervical spinal nerve avulsion injury
normal conditions, and in response to injury, we in the adult rat (Wu et al., 2004).
speculate that the role of nitric oxide synthase
in preganglionic parasympathetic neurons and
motoneurons may differ. Changes in levels of neurotrophins and their
receptors after injury

Acute surgical interventions to protect neurons from It has been assumed that the implanted avulsed
cell death nerve roots may produce factors that promote
neuroprotection. Previous studies have shown that
Previous investigations have demonstrated that a following a peripheral nerve injury, the distal nerve
surgical implantation of peripheral nervous tissue segment demonstrates a proliferation of Schwann
into the brain or spinal cord may induce regener- cells and an increased production of growth fac-
ation of central axons into the implanted graft tors, including glial-derived neurotrophic factor,
(Richardson et al., 1980; David and Aguayo, 1981; nerve growth factor, brain-derived neurotrophic
Benfey and Aguayo, 1982; Campbell et al., 1992; factor, and neurotrophin-4/5 (Salzer and Bunge,
Anderson et al., 1998). It has been recognized that 1980; Meyer et al., 1992; Funakoshi et al., 1993;
the regeneration of central axons into the implant- Naveilhan et al., 1997). It is therefore possible that
ed peripheral nerve graft was increased when the the neuroprotective effect of the root implant may
implantation was combined with an axonal injury be exerted by either one such neurotrophic factor
(Richardson and Issa, 1984). or by a combination of growth factors. Spinal cord
Attempts to reduce a ventral root avulsion- neurons may demonstrate injury-induced changes
induced death of motoneurons using implanted in their gene expression for trophic factor recep-
grafts have had variable results. A surgical im- tors. Following a ventral root avulsion in the adult
plantation of a peripheral nervous system graft rat, the axotomized motoneurons showed an
into the cervical spinal cord of rats following an increase in their mRNA expression for glial-
avulsion injury of the C7 spinal roots resulted in derived neurotrophic factor receptors, such as gly-
an inhibition of the injury-induced expression of cosyl-phosphatidylinositol-linked protein receptor
nitric oxide synthase in axotomized motoneuron (GFRa-1) and tyrosine kinase receptor c-Retp-
and an increased survival of the motoneurons (Wu roto-oncogene (c-RET), and the leukemia inhibi-
et al., 1994). A similar inhibition of nitric oxide tory factor receptor, but a decrease in mRNA
synthase and protection of motoneurons against a expression for tyrosine kinase B (trkB) and trkC
cervical spinal root avulsion-induced retrograde receptors (Hammarberg et al., 2000). From this
death has also been described following an acute perspective it is of interest to note that treatments
implantation of the avulsed ventral root into the using glial-derived neurotrophic factor are neuro-
spinal cord in adult rats (Chai et al., 2000). On the protective for axotomized motoneurons (Li et al.,
other hand, Novikova et al. (1997) reported that 1995; Watabe et al., 2000; Natsume et al., 2002).
neither a peripheral nerve graft nor a combina- Also, treatment with brain-derived neurotrophic
tion of the graft and an embryonic spinal cord factor, a trkB agonist, following a ventral root
implanted into the dorsolateral funiculus of the avulsion injury blocked the expression of nitric
injured lumbar spinal cord could prevent the oxide synthase in axotomized motoneurons
retrograde motoneuron degeneration, and death (Novikov et al., 1997) and reduced the retrograde
induced by ventral root avulsion in the adult rat. A motoneuron death (Kishino et al., 1997; Novikov
recent study investigating the therapeutic window et al., 1997). In contrast, ciliary neurotrophic fac-
of opportunity for nerve graft implants demon- tor had no detectable protective effect on axo-
strated that lesioned motoneurons retain their tomized motoneurons after a ventral root avulsion
200

injury in adult rats (Novikov et al., 1995). with proximal stumps only was present, nerve
Although extensive information is available on transfers using a femoral or intercostal nerve were
the gene expression of growth factor receptors in performed when an intradural lesion with distal
motoneurons and on the effects of treatment stumps was present, and an intrapelvic repair with
with a wide variety of neurotrophic factors on nerve grafts was performed when an intrapelvic
motoneurons, corresponding information for au- lesion was present. The study concluded that sub-
tonomic neurons, specifically preganglionic para- jects with lesions to the proximal nerve roots and
sympathetic neurons innervating pelvic organs, is cauda equina may recover basic lower extremity
presently very sparse or lacking. functions of unsupported standing and walking
following a surgical reconstruction of spinal
nerves, repair of ventral roots, and nerve transfers
Functional reinnervation of denervated targets using (Lang et al., 2004). No subjects with bladder and
surgical interventions bowel dysfunctions were included in the series,
however.
A ventral root avulsion injury followed by an Surgical attempts involving peripheral nerve or
acute surgical implantation of the avulsed ventral nerve root grafting techniques to promote a func-
root into the spinal cord results in regenerative tional reinnervation of the lower urinary tract fol-
growth of axons by motoneurons, innervation of lowing a spinal cord injury, have been sparse in the
the implanted ventral root, and functional rein- literature. However, an interesting proof-of-prin-
nervation of skeletal muscle in rats (Carlstedt ciple demonstration was provided in a cat model,
et al., 1986; Smith and Kodama, 1991; Chai et al., where a skin–CNS–bladder reflex pathway for mi-
2000; Gu et al., 2004), cats (Cullheim et al., 1989; cturition was constructed following a spinal cord
Hoffmann et al., 1996), and primates (Carlstedt injury by performing a microanastomosis of the
et al., 1993; Hallin et al., 1999). L7–S1 ventral root, while leaving the dorsal root
The above encouraging studies have also been intact (Xiao et al., 1999). The cross-wired somato-
translated into the clinic for the repair of avulsed autonomic bladder reflex was effective in initiating
nerve roots in humans with brachial plexus injuries bladder contractions and coordinated voiding in
(Carlstedt and Norén, 1995; Carlstedt et al., 1995, cats with an intact spinal cord, as well as inducing
2000). A review of the first human cases suggested bladder contractions after an acute spinal cord
that an early intervention with a short lag time transection injury (Xiao et al., 1999). These results
between the injury and restorative surgery is an are of particular interest for future possible
important factor for reestablishment of muscle lumbosacral ventral root repair interventions, in
function, primarily in the most proximal upper part because the findings show that somatic moto-
extremity muscle groups (Carlstedt et al., 2000). neurons can innervate a parasympathetic nerve
In a recent study, the therapeutic opportunities and functionally innervate the bladder.
and clinical outcomes were investigated in 10 pa-
tients after lumbosacral plexus injuries and surgi-
cal interventions (Lang et al., 2004). Most patients Strategies to treat chronic conus medullaris and
had suffered a severe traumatic injury with a pelvic cauda equina injuries
fracture and a traction injury to the lumbosacral
plexus. Clinically, lower extremity weakness and Because of the progressive and parallel retrograde
dysfunction were present. In this series of cases, cell death of both preganglionic parasympathetic
one of four defined surgical strategies was applied neurons and motoneurons that follows lesions to
for each patient. Specifically, an intradural repair their axons at the ventral root exit zone (Hoang
of ventral roots with nerve grafts was performed et al., 2003), neuroprotective strategies and at-
when an intradural lesion with both stumps was tempts to induce axonal regeneration in lesioned
present, an intradural–extradural repair with nerve efferent spinal cord neurons are likely to have a
grafts was performed when an intradural lesion specific time window of therapeutic opportunity.
201

Therefore, subjects with a chronic conus medulla- motoneurons normally exhibit a cholinergic
ris syndrome face a different set of challenges, as phenotype (Barber et al., 1984). It is likely that a
the lesioned efferent neurons in the conus medul- cell replacement strategy for the treatment of a
laris may have become largely depleted. In this chronic conus medullaris and cauda equina injury
setting, alternative treatment strategies need to be will also need to be combined with other thera-
considered. Given the injury-induced neuronal peutic interventions, especially with a peripheral
degeneration and death of preganglionic parasym- nerve or root grafting procedure to provide a con-
pathetic neurons and motoneurons, a cell replace- duit for the extension of central axons of grafted
ment approach appears attractive. cells into the peripheral nervous system and for
Much interest has been generated for the po- ultimate functional reinnervation of pelvic targets,
tential use of pluripotent and multipotent stem e.g., the lower urinary tract.
cells in the treatment of neurological disorders,
including spinal cord injury. Multipotent neural
Assessments of lower urinary tract function
stem cells have been isolated from both the fetal
following acute and chronic treatment interventions
and adult human nervous system (Johansson et al.,
1999; Vescovi et al., 1999; Villa et al., 2000; Palmer
In future animal studies on neural repair following
et al., 2001; Westerlund et al., 2003). Although
a traumatic injury to the conus medullaris and/or
neural stem cells have demonstrated their ability to
cauda equina, it is important to be able to assess
differentiate into both neurons and glial cells in
the effects of the treatment intervention. In rodent
vitro, most stem cells are restricted to a glial
studies, a variety of functional and end-organ as-
lineage and very few stem cells become neurons
sessment methods are available and used to eval-
when transplanted into the spinal cord (McDonald
uate the lower urinary tract under normal and
et al., 1999; Cao et al., 2001). For some neural
pathological conditions. These methods include
repair strategies, the apparent preferred develop-
bladder expressions to assess for urinary retention,
ment of neural stem cells into a glial lineage may
behavioral studies on voiding patterns, urodynam-
be a good feature, for instance when remyelination
ic studies, metabolic cage assessments, as well as
of lesioned ascending and descending white matter
postmortem bladder weight and size measure-
tracts is desired. However, the limited yield of
ments (Maggi et al., 1986; Chancellor et al.,
neurons derived from transplanted neural stem
1994; Pikov et al., 1998; Kerns et al., 2000; Pikov
cells into the spinal cord presents a potential
and Wrathall, 2001).
problem for purposes of replacing degenerated
and lost spinal cord neurons in subjects with a
conus medullaris syndrome. Conclusions
Interestingly, recent studies have demonstrated
that an in vitro priming procedure of fetal human Conus medullaris and cauda equina injuries com-
neural stem cells can markedly increase the per- monly result in severe neurological impairments,
centage of stem cells that differentiate into neurons including paralysis, sensory disturbance, pain, as
when transplanted into the spinal cord (Wu et al., well as bladder, bowel, and sexual dysfunctions. In
2002). A large portion of the primed cells, which recent years, a more comprehensive understanding
were grafted into the spinal cord, demonstrated of the underlying neurobiology and pathophysiology
immunoreactivity for choline acetyltransferase has been emerging. A post-traumatic progressive
(Wu et al., 2002). The ability to influence the loss of efferent somatic and autonomic neurons is
differentiation of grafted cells into choline acetylt- an important effect of conus medullaris and cauda
ransferase-immunoreactive neurons in the spinal equina injuries in experimental models. Early
cord is of particular interest when considering cell interventions to decrease the retrograde death of
replacement strategies for the treatment of conus axotomized neurons are likely to remain important
medullaris and cauda equina injuries, as both in future translational research efforts. For acute
preganglionic parasympathetic neurons and and subacute injuries, surgical strategies, where
202

peripheral nerve grafts or lesioned nerve roots are Cao, Q.L., Zhang, Y.P., Howard, R.M., Walters, W.M.,
implanted into the lumbosacral spinal cord, may Tsoulfas, P. and Whittemore, S.R. (2001) Pluripotent stem
also be applied in select subjects to promote func- cells engrafted into the normal or lesioned adult rat spinal
cord are restricted to a glial lineage. Exp. Neurol., 167:
tional reinnervation of peripheral targets, includ- 48–58.
ing the lower urinary tract. A combination of Carlson, J., Lais, A.C. and Dyck, P.J. (1979) Axonal atrophy
medical and surgical treatments to increase neuro- from permanent peripheral axotomy in adult cat. J. Ne-
protection, axonal regeneration and peripheral uropathol Exp. Neurol., 38(6): 579–585.
target reinnervation may also be considered. In Carlstedt, T., Anand, P., Hallin, R.G., Misra, P.V., Norén, G.
and Seferlis, T. (2000) Spinal root repair and reimplantation
chronic injuries, cell replacement strategies, rather of avulsed ventral roots into the spinal cord after brachial
than neuroprotective approaches, may become plexus injury. J. Neurosurg., 93: 237–247.
more viable options, as many efferent spinal cord Carlstedt, T., Grane, P., Hallin, R.G. and Norén, G. (1995)
neurons may already have undergone retrograde Return of function after spinal cord implantation of avulsed
cell death. However, cell replacement strategies roots. Lancet, 346: 1323–1325.
Carlstedt, T., Lindå, H., Cullheim, S. and Risling, M. (1986)
may still need to be combined with e.g., a periph-
Reinnervation of hindlimb muscles after ventral root avuls-
eral nerve or nerve root grafting procedure to fa- ion and implantation in the lumbar spinal cord in the adult
cilitate and promote functional reinnervation of rat. Acta Physiol. Scand., 128: 645–646.
peripheral targets. Carlstedt, T. and Norén, G. (1995) Repair of ruptured
spinal nerve roots in a brachial plexus lesion. Case report.
J. Neurosurg., 82(4): 661–663.
Acknowledgments Carlstedt, T.P., Hallin, R.G., Hedström, K.G. and Nilsson-
Remahl, I.A.M. (1993) Functional recovery in primates with
brachial plexus injury after spinal cord implantation of avu-
Supported by research grants from NIH
lsed ventral roots. J. Neurol. Neurosurg. Psychiat., 56:
(NS042719), The Paralysis Project of America, 649–654.
The State of California Roman Reed Bill, and Chai, H., Wu, W., So, K.-F. and Yip, H.K. (2000) Survival and
SCORE. regeneration of motoneurons in adult rats by reimplantation
of ventral root following spinal root avulsion. Neuroreport,
11: 1249–1252.
References Chan, Y.M., Wu, W., Yip, H.K., So, K.F. and Oppenheim,
R.W. (2001) Caspase inhibitors promote the survival of avu-
Anderson, P.N., Campbell, G., Zhang, Y. and Lieberman, A.R. lsed spinal motoneurons in neonatal rats. Neuroreport, 12:
(1998) Cellular and molecular correlates of the regeneration 541–545.
of adult mammalian CNS axons into peripheral nerve grafts. Chancellor, M.B., Rivas, D.A., Huang, B., Kelly, G. and
Prog. Brain Res., 117: 211–232. Salzman, S.K. (1994) Micturition patterns after spinal
Anneser, J.M.H., Berhele, A., Borasio, G.D., Castro-Lopez, trauma as a measure of autonomic functional recovery.
J.M., Zieglgänsberger, W. and Tölle, T.R. (2000) Axotomy of J. Urol., 151(1): 250–254.
the sciatic nerve differentially affects expression of metabo- Cullheim, S., Carlstedt, T., Lindå, H., Risling, M. and Ulfhake,
tropic glutamate receptor mRNA in adult motoneurons. B. (1989) Motoneurons reinnervate skeletal muscle after ven-
Brain Res., 868: 215–221. tral root implantation into spinal cord of the cat. Neurosci-
Barber, R.P., Phelps, P.E., Houser, C.R., Crawford, G.D., ence, 29: 725–734.
Salvaterra, P.M. and Vaughn, J.E. (1984) The morphology David, S. and Aguayo, A.J. (1981) Axonal elongation into
and distribution of neurons containing choline acetyltransf- peripheral nervous system ‘‘bridges’’ after central nervous
erase in the adult rat spinal cord: an immunohistochemical system injury in rats. Science, 214: 931–933.
study. J. Comp. Neurol., 229: 329–346. Fowler, C.J. (1999) Neurological disorders of micturition and
Benfey, M. and Aguayo, A.J. (1982) Extensive elongation of their treatment. Brain, 122: 1213–1231.
axons from rat brain into peripheral nerve grafts. Nature, Fowler, C.J., Kirby, R.S., Harrison, M.J., Milroy, E.J. and
296: 150–152. Turner-Warwick, R. (1984) Individual motor unit analysis in
Beric, A. and Light, J.K. (1992) Detrusor function with lesions the diagnosis of disorders of urethral sphincter innervation.
of the conus medullaris. J. Urol., 148: 104–106. J. Neurol. Neurosurg. Psychiat., 47(6): 637–641.
Campbell, G., Lieberman, A.R., Anderson, P.N. and Funakoshi, H., Frisén, J., Barbany, G., Timmusk, T.,
Turmaine, M. (1992) Regeneration of adult rat CNS axons Zachrisson, O., Verge, V.M. and Person, H. (1993)
into peripheral nerve autografts: ultrastructural studies of the Differential expression of mRNA for neurotrophins and
early stages of axonal sprouting and regenerative axonal their receptors after axotomy of the sciatic nerve. J. Cell
growth. J. Neurocytol., 21(11): 755–787. Biol., 123: 455–465.
203

Gu, H.-Y., Chai, H., Zhang, J.-Y., Yao, Z.-B., Zhou, L.-H., Light, J.K., Beric, A. and Petronic, I. (1993) Detrusor
Wong, W.-M., Bruce, I. and Wu, W.-T. (2004) Survival, dysfunction with lesions of the cauda equina, with special
regeneration and functional recovery of motoneurons in emphasis on the bladder neck. J. Urol., 149(3): 539–542.
adult rats by implantation of ventral root following spinal Lindå, H., Cullheim, S. and Risling, M. (1992) A light and
root avulsion. Eur. J. Neurosci., 19: 2123–2131. electron microscopic study of intracellularly HRP-labeled
Hallin, R.G., Carlstedt, T., Nilsson-Remahl, I. and Risling, M. lumbar motoneurons after intramedullary axotomy in the
(1999) Spinal cord implantation of avulsed ventral roots in adult cat. J. Comp. Neurol., 318(2): 188–208.
primates; correlation between restored motor function and Liu, Z. and Martin, L.J. (2001) Motor neurons rapidly accu-
morphology. Exp. Brain Res., 124: 304–310. mulate DNA single-strand breaks after in vitro exposure to
Hammarberg, H., Piehl, F., Risling, M. and Cullheim, S. (2000) nitric oxide and peroxynitrite and in vivo axotomy. J. Comp.
Differential regulation of trophic factor receptor mRNAs in Neurol., 432: 35–60.
spinal motoneuons after sciatic nerve transection and ventral Ma, J., Novikov, L.N., Wiberg, M. and Kellerth, J.-O. (2001)
root avulsion in the rat. J. Comp. Neurol., 426: 587–601. Delayed loss of spinal motoneurons after peripheral nerve
Hans, F.J., Reinges, M.H. and Krings, T. (2004) Lumbar nerve injury in adult rats: a quantitative morphological study. Exp.
root avulsion following balanced fast field echo MRI. Brain Res., 139: 216–223.
Neuroradiology, 46: 144–147. Maggi, C.A., Santicioli, P. and Meli, A. (1986) The nonstop
Hoang, T.X., Nieto, J.H., Tillakaratne, N.J.K. and Havton, transvesical cystometrogram in urethane-anesthetized rats: a
L.A. (2003) Autonomic and motor neuron death is progres- simple procedure for quantitative studies on the various
sive and parallel in a lumbosacral ventral root avulsion model phases of urinary bladder voiding cycle. J. Pharmacol. Meth-
of cauda equine injury. J. Comp. Neurol., 426: 587–601. ods, 15(2): 157–167.
Hoffmann, C.F.E., Marani, E., Van Dijk, J.G., Kamp, W.V.D. Martin, L.J., Kaiser, A. and Price, A.C. (1999) Motor neuron
and Thoemer, R.T.W.M. (1996) Reinnervation of avulsed degeneration after sciatic nerve avulsion in adult rat evolves
and reimplanted ventral rootlets in the cervical spinal cord of with oxidative stress and is apoptosis. J. Neurobiol., 40:
the cat. J. Neurosurg., 84: 234–243. 185–201.
Holmberg, P. and Kellerth, J.O. (2000) Do synaptic rearrange- Martin, L.J. and Liu, Z. (2002) Injury-induced spinal
ments underlie compensatory reflex enhancement in spinal motor neuron apoptosis preceeded by DNA single-strand
motoneurons after partial cell loss? Synapse, 38(4): 384–391. breaks and is p53- and Bax-dependent. J. Neurobiol.,
Johnson, I.P., Sears, T.A. and Hunter, A.S. (1991) Retrograde 50: 181–197.
response to axotomy of motoneurons in the thoracic spinal Maynard, F.M., Braken, M.B., Creasey, G., Ditunno, J.F.,
cord of the aging cat. Neurobiol. Aging, 12(2): 151–160. Donovan, W.H., Ducker, T.B., Garber, S.L., Marino, R.J.,
Johansson, C.B., Svensson, M., Wallstedt, L., Janson, A.M. Stover, S.L., Tator, C.H., Waters, R.L., Wilberger, J.E. and
and Frisén, J. (1999) Neural stem cells in the adult human Young, W. (1997) International Standards for Neurological
brain. Exp. Cell Res., 253: 733–736. and Functional Classification of Spinal Cord Injury. Spinal
Kerns, J.M., Damaser, M.S., Kane, J.M., Sakamoto, K., Cord, 35: 266–274.
Benson, J.T., Shott, S. and Brubaker, L. (2000) Effects of McDonald, J.W., Liu, X.Z., Qu, Y., Liu, S., Mickey, S.K.,
pudendal nerve injury in the female rat. Neurourol. Urodyn., Turetsky, D., Gottlieb, D.I. and Choi, D.W. (1999) Trans-
19(1): 53–69. planted embryonic stem cells survive, differentiate and pro-
Kishino, A., Ishige, Y., Tatsuno, T., Nakayama, C. and mote recovery in injured rat spinal cord. Nat. Med., 5:
Noguchi, H. (1997) BDNF prevents and reverses adult rat 1410–1412.
motor neuron degeneration and induces axonal outgrowth. Meyer, M., Matsuoka, I., Wetmore, C., Olson, L. and Thoenen,
Exp. Neurol., 144(2): 273–286. H. (1992) Enhanced synthesis of brain-derived neurotrophic
Koliatsos, V.E., Price, W.L., Pardo, C.A. and Price, D.L. factor in the lesioned peripheral nerve: different mechanisms
(1994) Ventral root avulsion: an experimental model of death are responsible for the regulation of BDNF and NGF
of adult motor neurons. J. Comp. Neurol., 342(1): 35–44. mRNA. J. Cell Biol., 119: 45–54.
Lang, E.M., Borges, J. and Carlstedt, T. (2004) Surgical treat- Monga, P., Ahmed, A., Gupta, G.R. and Rao, K.S. (2004)
ment of lumbosacral plexus injuries. J. Neurosurg. (Spine 1), Traumatic lumbar nerve root avulsion: evaluation using
1: 64–71. electrodiagnostic studies and magnetic resonance myelo-
Li, L., Houenou, L.J., Wu, W., Lei, M., Prevette, D.M. and graphy. J. Trauma, 56: 182–184.
Oppenheim, R.W. (1998) Characterization of spinal moto- Moschilla, G., Song, S. and Chakera, T. (2001) Post-traumatic
neuron degeneration following different types of peripheral lumber nerve avulsion. Australas. Radiol., 45: 281–284.
nerve injury in neonatal and adult mice. J. Comp. Neurol., Natsume, A., Mata, M., Wolfe, D., Oligino, T., Goss, J.,
396: 158–168. Huang, S., Glorioso, J. and Fink, D.J. (2002) Bcl-2 and
Li, L., Wu, W., Lin, L.F., Lei, M., Oppenheim, R.W. and GDNF delivered by HSV-mediated gene transfer after spinal
Houenou, L.J. (1995) Rescue of adult mouse motoneurons root avulsion provide a synergistic effect. J. Neurotrauma,
from injury-induced cell death by glial cell line-derived 19(1): 61–68.
neurotrophic factor. Proc. Natl. Acad. Sci., 92(21): Naveilhan, P., ElShamy, W.M. and Ernfors, P. (1997) Differ-
9771–9775. ential regulation of mRNAs for GDNF and its receptors Ret
204

and GDNFR alpha after sciatic nerve lesion in the mouse. despite prolonged absence of functional synaptic contact with
Eur. J. Neurosci., 9(7): 1450–1460. muscle. Exp. Neurol., 123: 147–156.
Novikov, L., Novikova, L. and Kellerth, J.-O. (1995) Brain- Vescovi, A.L., Parati, E.A., Gritti, A., Poulin, P., Ferrario, M.,
derived neurotrophic factor promotes survival and blocks Wanke, E., Frolichstal-Schoeller, P., Cova, L., Arcellana-
nitric oxide synthase expression in adult rat spinal motoneu- Panlilio, M., Colombo, A. and Galli, R. (1999) Isolation and
rons after ventral root avulsion. Neurosci. Lett., 200: 45–48. cloning of multipotential stem cells from the embryonic hu-
Novikov, L., Novikova, L. and Kellerth, J.-O. (1997) Brain- man CNS and establishment of transplantable human neural
derived neurotrophic factor promotes axonal regeneration stem cell lines by epigenetic stimulation. Exp. Neurol., 56(1):
and long-term survival of adult rat spinal motoneurons in 71–83.
vivo. Neuroscience, 79: 765–774. Villa, A., Snyder, E.Y., Vescovi, A. and Martinez-Serrano, A.
Novikov, L.N., Novikova, L.N., Holmberg, P. and Kellerth, (2000) Establishment and properties of a growth factor-
J.-O. (2000) Exogenous brain-derived neurotrophic factor dependent, perpetual neural stem cell line from the human
regulates the synaptic composition of axonally lesioned and CNS. Exp. Neurol., 161: 67–84.
normal adult rat motoneurons. Neuroscience, 100(1): 171–181. Vizzard, M.A., Erdman, S.L. and de Groat, W.C. (1993a)
Novikova, L., Novikov, L. and Kellerth, J.-O. (1997) Effects of Localization of NADPH-diaphorase in pelvic afferent and
neurotransplants and BDNF on the survival and regenera- efferent pathways of the rat. Neurosci. Lett., 152: 72–76.
tion of injured adult spinal motoneurons. Eur. J. Neurosci., Vizzard, M.A., Erdman, S.L. and de Groat, W.C. (1993b) The
9: 2774–2777. effect of rhizotomy on NADPH diaphorase staining in the
Palmer, T.D., Schwartz, P.H., Taupin, P., Kaspar, B., Stein, lumbar spinal cord of the rat. Brain Res., 607: 349–353.
S.A. and Gage, F.H. (2001) Progenitor cells from human Vizzard, M.A., Erdman, S.L. and de Groat, W.C. (1995) In-
brain after death. Nature, 411: 42–43. creased expression of neuronal nitric oxide synthase (NOS) in
Pavlakis, A.J., Siroky, M.B., Goldstein, I. and Krane, R.J. visceral neurons after nerve injury. J. Neurosci., 15: 4033–4045.
(1983) Neurourologic findings in conus medullaris and cauda Watabe, K., Ohashi, T., Sakamoto, T., Kawazoe, Y.,
equina injury. Arch. Neurol., 40: 570–573. Takeshima, T., Oyanagi, K., Inoue, K., Eto, Y. and
Piehl, F., Tabar, G. and Cullheim, S. (1995) Expression of Kim, S.U. (2000) Rescue of lesioned adult rat spinal moto-
NMDA receptor mRNAs in rat motoneurons is down-reg- neurons by adenoviral gene transfer of glial cell line-derived
ulated after axotomy. Eur. J. Neurosci., 7: 2101–2110. neurotrophic factor. J. Neurosci. Res., 60(4): 511–519.
Pikov, V., Gillis, A., Jasmin, L. and Wrathall, J.R. (1998) As- Westerlund, U., Moe, M.C., Varghese, M., Berg-Johnsen, J.,
sessment of lower urinary tract functional deficit in rats with Ohlsson, M., Langmoen, I.A. and Svensson, M. (2003) Stem
contusive spinal cord injury. J. Neurotrauma, 15(5): 375–386. cells from the adult human brain develop into functional
Pikov, V. and Wrathall, J.R. (2001) Coordination of the neurons in culture. Exp. Cell Res., 289: 378–383.
bladder detrusor and the external urethral sphincter in a rat Wu, C.W. and Kaas, J.H. (2000) Spinal cord atrophy and re-
model of spinal cord injury: effect of injury severity. organization of motoneuron connections following long-
J. Neurosci., 21(2): 559–569. standing limb loss in primates. Neuron, 28: 967–978.
Richardson, P.M. and Issa, V.M. (1984) Peripheral injury Wu, P., Tarasenko, Y.I., Gu, Y., Huang, L.-Y.M., Coggeshall,
enhances central regeneration of primary sensory neurons. R.E. and Yu, Y. (2002) Region-specific generation of
Nature, 309: 791–793. cholinergic neurons from fetal human neural stem cells graft-
Richardson, P.M., McGuinness, U.M. and Aguayo, A.J. (1980) ed in adult rat. Nat. Neurosci., 5: 1271–1278.
Axons from CNS neurons regenerate into PNS grafts. Wu, W. (1993) Expression of nitric-oxide synthase (NOS) in
Nature, 284(5753): 264–265. injured CNS neurons as shown by NADPH diaphorase his-
Saito, S., Kidd, G.J., Trapp, B.D., Dawson, T.M., Bredt, D.S., tochemistry. Exp. Neurol., 120: 153–159.
Wilson, D.A., Traystman, R.J., Snyder, S.H. and Hanley, Wu, W., Chai, H., Zhang, J., Gu, H., Xie, Y. and Zhou, L.
D.F. (1994) Rat spinal cord neurons contain nitric oxide (2004) Delayed implantation of a peripheral nerve graft re-
synthase. Neuroscience, 59(2): 447–456. duces motoneurons survival but does not affect regeneration
Salzer, J.L. and Bunge, R.P. (1980) Studies of Schwann cell following spinal root avulsion in adult rats. J. Neurotrauma,
proliferation I. An analysis in tissue culture of proliferation 21: 1050–1058.
in development, Wallerian degeneration, and direct injury. Wu, W., Han, K., Li, L. and Schinco, F.P. (1994) Implantation
J. Cell Biol., 84: 739–752. of PNS graft inhibits the induction of neuronal nitric oxide
Schrøder, H.D. (1980) Organization of the motoneurons synthase and enhances the survival of spinal motoneurons
innervating the pelvic muscles of the male rat. J. Comp. following root avulsion. Exp. Neurol., 129: 335–339.
Neurol., 192(3): 567–587. Wu, W. and Li, L. (1993) Inhibition of nitric oxide synthase
Smith, K.J. and Kodama, R.T. (1991) Reinnervation of den- reduces motoneuron death due to spinal root avulsion.
ervated skeletal muscle by central neurons regenerating via Neurosci. Lett., 153(2): 121–124.
ventral roots implanted into the spinal cord. Brain Res., 551: Xiao, C.-G., de Groat, W.C., Godec, C.J., Dai, C. and Xiao, Q.
221–229. (1999) ‘‘Skin–CNS–bladder’’ reflex pathway for micturition
Vanden Noven, S., Wallace, N., Muccio, D., Turtz, A. and after spinal cord injury and its underlying mechanisms.
Pinter, M.J. (1993) Adult spinal motoneurons remain viable J. Urol., 162: 936–942.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 13

Pelvic somato-visceral reflexes after spinal cord


injury: measures of functional loss and
partial preservation

Michael D. Craggs

Centre for Spinal Research, Functional Assessment and Restoration, London Spinal Cord Injuries Unit, Royal National
Orthopaedic Hospital NHS Trust, Brockley Hill, Stanmore, Middlesex HA7 4LP, UK

Abstract: For people with spinal cord injuries, the impact of bladder, bowel and sexual problems on quality
of life and lost opportunities can be devastating. Supra-sacral spinal lesions can cause incontinence by
interrupting those pathways that normally coordinate the function of the bladder, bowel and sphincters.
From a scientific perspective, neural control of the pelvic organs is one of the most intriguing in the body,
involving both somatic and autonomic pathways participating in an exquisitely fine integration of lumbo-
sacral reflexes. This chapter aims to review briefly those aspects of neural control of the pelvic organs that
are amenable to neurophysiological examination in man. More specifically, it will focus in greater detail on
the interactions of somatic and autonomic lumbo-sacral pathways responsible for coordinating the bladder
and sphincters. Where appropriate, it will make comparisons with those controlling the bowel. It will
describe how measurement of pelvic floor and sphincter reflexes can be used to assess the modulatory
effects of sacral autonomic pathways on sacral somatic reflexes and vice versa including the so-called
‘‘guarding reflex’’ and vesical inhibitory reflexes. Aberrant activity following spinal cord injury (SCI), such
as bladder hyperreflexia and sphincter dyssynergia, will be discussed in relation to these reflexes. The effects
of volitional modulation of pelvic floor reflexes in people with both complete and incomplete lesions will be
described. Finally, the chapter will address the possible utility of neurophysiological measures for com-
plementing the established neurological classification and the assessment of somatic sensory-motor im-
pairment in SCI.

Introduction Supra-sacral spinal lesions cause incontinence


by interrupting those pathways that normally co-
For people with spinal cord injuries, the impact of ordinate the bladder, bowel and sphincter func-
bladder, bowel and sexual problems on quality of tions. Such lesions are conventionally termed
life and lost opportunities can be devastating. The upper motor neuron lesions. Lesions of the sacral
social stigma of urinary and fecal incontinence cord or nerve roots, for example, cauda equina
make these problems some of the most seriously (lumbar-sacral nerve roots) damage or disease, are
debilitating conditions needing long-term care and conventionally termed lower motor neuron lesions
management in spinal cord injury (SCI). as they disconnect bladder, part of the bowel and
their sphincters from their source of innervation in
Corresponding author. Tel./fax: +44 (0)20 8909 5343 (Hos- the spinal cord. Depending on their completeness,
pital), +44 (0)20 7679 9379 (University); these lesions can result in total loss of reflex con-
E-mail: michael.craggs@ucl.ac.uk trol of the bladder, bowel and sphincters, again

DOI: 10.1016/S0079-6123(05)52013-2 205


206

leading to serious problems of storage and void- the bladder or rectum, respectively. Although the
ing. sensations of fullness and the strong desire to mi-
From a scientific perspective, neural control of cturate or defecate are sometimes difficult to sup-
the pelvic organs is one of the most intriguing in press, micturition and defecation can be suspended
the body, involving both somatic and autonomic for some length of time, and continence main-
pathways participating in an exquisitely fine inte- tained even when the bladder and rectum are very
gration of lumbo-sacral reflexes. Volition can play full. The mechanisms of control in man are
a significant part in this modulation. In adult man, brought about through a balance of modulatory
segmental reflexes are under fine tuning by mod- influences at many levels in the neuraxis from
ulatory pathways from various levels of the brain, brain to the sacral (S) cord and pelvic plexus.
from brain stem to cerebral cortex. For the lower
urinary tract, precise coordination between the
Peripheral innervation
bladder detrusor muscle, urethral smooth muscles
and voluntary muscle of the urethral sphincter is
The bladder and colo-rectum, together with their
essential for maintaining continence over long pe-
respective sphincters, show many similarities in
riods of time and voiding efficiently when neces-
muscular organization and peripheral extrinsic
sary.
nerve supply. However, their functions are very
Through a comprehensive knowledge of the
different and depend on their unique neuro-ana-
physiological and neurophysiological mechanisms
tomical specialization. The colo-rectum has a well-
controlling the pelvic organs, we can hope to de-
developed enteric (intrinsic) nervous system that
velop more reliable and sensitive diagnostic tools
coordinates motility of its smooth muscle, secre-
to assess new interventions for treating pelvic dys-
tion and absorption and is modulated by extrinsic
function.
nerves. The bladder, in contrast, is controlled al-
This chapter aims to review briefly those aspects
most exclusively by extrinsic nerves so that con-
of neural control of the pelvic organs that are
traction of its smooth muscle, the detrusor, is a
amenable to neurophysiological examination in
coordinated event for efficient emptying. The de-
man. More specifically, it will focus in greater de-
trusor remains quiescent at all other times while
tail on the interactions of somatic and autonomic
the bladder is filling.
lumbo-sacral pathways responsible for coordinat-
ing the bladder and sphincters, but make compar-
isons, where appropriate, with those controlling The bowel
the bowel. It will examine the modulatory effects
of sacral autonomic pathways on sacral somatic The intrinsic nerve supply to the colo-rectum is
reflexes and vice versa. It will compare these inter- derived from the myenteric and submucosal plex-
actions using neurophysiological measures follow- uses. These plexuses have a complex structural or-
ing SCI and show the effects of volitional ganization and involve the interactions of many
modulation in people with both complete and in- different neurotransmitter pathways, including the
complete lesions. Finally, it will discuss the possi- modulating influences of the extrinsic nerve supply
ble utility of such neurophysiological measures for with its sympathetic and parasympathetic nerves
complementing the established neurological clas- (Burnstock, 1990). The plexuses in the rectum and
sification and the assessment of somatic sensory- distal colon are dense and irregular compared to
motor impairment in SCI. those in the more proximal colon. Sacral para-
sympathetic innervation of the rectum and distal
Neural control of the pelvic viscera and pelvic floor colon is clearly an important aspect of the extrinsic
control of defecation. Outlet function is also very
Normal micturition and defecation are initiated specialized (Mathers, 1992). The ano-rectum is
voluntarily at an appropriate and socially conven- able to distinguish solid, liquid and gas content by
ient time when sensory signals indicate fullness in a complex sampling mechanism and to facilitate
207

selective passage of gas when appropriate, whereas no muscle spindles and, is therefore quite distinct
urethro-vesical muscle function is very well coor- from all other striated muscles of the pelvic floor
dinated, so that even the smallest leakage of urine (Schrøder and Reske-Nielsen, 1983). The other
is prevented at inappropriate times. muscles of the pelvic floor are innervated by
branches of the sacral anterior roots. Of these
muscles, the levator ani group comprises a mixture
The bladder
of slow-twitch and fast-twitch fibers that function-
ally provide sustained tone and phasic anti-stress
The bladder detrusor smooth muscle and smooth
contractions respectively to give peri-urethral and
muscle of the distal rectal wall receive parasympa-
peri-anal occlusion.
thetic innervation from preganglionic neurons in
the intermediolateral column of the S2–S4 sacral
The sensory pathways
segments of the spinal cord via post-ganglionic
neurons originating in ganglia of the pelvic plexus
Pelvic visceral nerves are the main sensory path-
or in the bladder wall itself (Burnstock, 1990). In-
ways from the bladder and rectum, but some sen-
teracting at the peripheral ganglia are sympathetic
sory information is also conveyed in the
efferent fibers of the hypogastric nerve plexus (in-
sympathetic hypogastric nerves to the thoraco-
ferior mesenteric ganglia). The sympathetic path-
lumbar spinal cord, especially from the colorec-
way has its origin in the intermediolateral column
tum, bladder neck and proximal urethra (Jänig
of the 9th thoracic (T) to 2nd lumbar (L) segments
and Morrison, 1986). Sensory information from
of the spinal cord. The preganglionic axons synapse
the distal urethra, anal canal and perineum is car-
first in the pre-vertebral ganglia or lumbosacral
ried almost exclusively by the pudendal nerves.
sympathetic chain ganglia (De Groat, 1997).
Essential to proper coordination of the bladder,
bowel and sphincters is the reflex circuitry of the
The pelvic floor lumbo-sacral spinal segments. Modulation is by
supra-spinal pathways from the brain-stem and
Muscles of the pelvic floor, including the peri-ure- diencephalon together with volitional pathways
thral and peri-anal muscles (for example, levator from the cerebral cortex (Craggs and Vaizey,
ani and pubo-rectalis) and urethral and anal stri- 1999).
ated sphincters, are innervated from the sacral
cord. However, only the specialized sphincter mus- Coordination of the lower urinary tract
cles receive their peripheral innervation via the
pudendal nerves. All of these somatic neural path- The coordinated function of the urinary bladder
ways arise from motor neurons in Onuf’s nucleus, and urethral sphincters depends on the complete
a specialized group of anterior horn cells spanning integrity of central and peripheral neural circuitry
the second, third and fourth sacral spinal seg- in a complex control system located in the brain
ments. Unlike the anal canal, the urethral wall and spinal cord. These functions have been inves-
contains a predominantly slow-twitch striated tigated extensively in recent times through a com-
muscle sphincter known as the rhabdosphincter. bination of urodynamics, neurophysiology and
This sphincter is innervated by small motor neu- sophisticated imaging techniques (Blok and Will-
rons in Onuf’s nucleus that is situated close to the emsen, 1997).
intermediolateral neurons of the preganglionic In mature adults, this integrated control system
parasympathetic pathway to the bladder detrusor is believed to act like a switching circuit (De
muscle. This close anatomic relationship has func- Groat, 1997) but under close cognitive and voli-
tional significance for close somato-visceral inte- tional supervision (Blok and Willemsen, 1997).
gration in the lower urinary tract (Mundy and Ascending afferent activity from the lower urinary
Thomas, 1994). Interestingly, unlike the external tract is routed both to those parts of the brain-
anal sphincter, the rhabdosphincter is said to have detecting sensation and to the pontine micturition
208

center through relay neurons in the peri-aqueduct- enabling urine to be expelled quickly and efficient-
al gray of the brain stem. This center appears to ly from the bladder until empty. The detrusor
maintain an appropriate reciprocal relationship contraction relies on the descending drive from the
between lumbo-sacral reflexes so as to coordinate pontine medial nucleus to the parasympathetic
the bladder and the urethral sphincters during motoneurons in the sacral cord being turned ‘‘on’’
storage and voiding (Yoshimura et al., 2004). (Blok et al., 1997), while activity from the pontine
lateral nucleus modulating the ‘‘guarding reflex’’ is
switched off.
Storage reflexes

During bladder filling, a number of lumbo-sacral Coordination of the bowel


reflex pathways appear to be active, ensuring com-
petent urethral closure together with detrusor in- Although we have considerable knowledge about
hibition to maintain continence. As the bladder the control of the urinary bladder and its sphinc-
slowly fills and stores urine, any tendency for ters, much less is known about the precise role of
spontaneous reflex contractions of the detrusor spinal reflex mechanisms and central modulatory
smooth muscle in the bladder wall, due to rising effects on bowel function. It is the enteric nervous
pelvic afferent nerve activity, is inhibited by a system, both in the colon and rectum, which plays
combination of sympathetically mediated reflexes a major role in the coordination of these parts of
suppressing detrusor activity and contracting the the bowel. Unlike the bladder, the rectum is only a
smooth muscle of the bladder neck and proximal very temporary reservoir for the feces propelled
urethra (Yoshimura et al., 2004). As the bladder into it by peristalsis from the colon. Furthermore,
volume increases, the striated urethral sphincter the anal canal has a very distinct internal sphincter
also contracts automatically via a pelvic afferent to comprising smooth muscle, which provides conti-
pudendal efferent reflex to prevent leakage, a nence, but which relaxes automatically when the
mechanism sometimes known as the ‘‘guarding rectum fills. This recto-anal response is not a spinal
reflex’’ (Park et al., 1997). Evidence for this mech- reflex, but appears to depend on a local nitric ox-
anism is seen as a build-up in the sphincter elect- ide-mediated mechanism (Burleigh, 1992). Howev-
romyogram (EMG) during bladder filling. Such er, whereas the enteric nervous system plays the
pudendal activity probably further inhibits any major role in coordinating ano-rectal function, the
tendency for aberrant parasympathetic activity extrinsic autonomic influences of the sacral para-
(Craggs and MacFarlane, 1999). When the bladder sympathetic pathway through the pelvic nerves and
reaches its maximum capacity and there is a strong lumbar sympathetic pathways via the inferior mes-
desire to void, then voluntary control of the stri- enteric plexus and hypogastric nerves, together
ated urethral sphincter and other pelvic floor mus- with their central connections via the brain stem,
cles can come into play to preserve continence do play an important part in the modulation of
until a suitable place for micturition is found. these intrinsic effects in the normal bowel (Craggs
and Vaizey, 1999). We do know that experimen-
tally, direct sympathetic stimulation decreases con-
Voiding reflexes tractility and motility of the colorectum and
constriction of the internal anal sphincter, where-
Voiding, heralded by an abrupt cessation of the as parasympathetic stimulation has the opposite
striated sphincter EMG, is brought about by four effects (see Brading and Ramalingam, this volume).
synergistic reflexes (Yoshimura et al., 2004). Two
of these cause relaxation of the bladder neck
smooth muscle and striated urethral sphincter, re- Storage and voiding reflexes
spectively before a third reciprocal action gives a
powerful detrusor contraction, sustained by a The maintenance of fecal continence is a product
fourth, a urethral to bladder facilitatory reflex, of stool consistency, colo-rectal activity and the
209

synchronous relationship between the external and latency of around 90 ms (Basinski et al., 2003)
internal anal sphincters (Engel et al., 1995). The suggesting that the guarding reflex probably in-
internal anal sphincter maintains a constant tone volves many interneurons in its arc and could even
and prevents stool leakage during everyday activ- engage a supra-spinal pathway (Park et al., 1997).
ities. The external anal sphincter acts like an
‘‘emergency brake’’ being used voluntarily and
maximally when there is a feeling of impending
defecation, thus allowing time to organize a suit- Neurophysiological measures of sacral reflexes
able situation for toiletting. Interestingly, we can
observe an anal sphincter ‘‘guarding reflex’’ during Routine clinical neurophysiological testing of pel-
distension of the rectum (Chung et al., 2004) par- vic floor or sphincter muscles can be done by di-
alleling that seen during filling of the bladder. rectly examining various sacral reflexes involving
the bulbocavernosus, ischiocavernosus or sphinc-
ter muscles. Diagnostically, examination of these
The guarding reflex reflexes by concentric needle electromyography or,
less specifically, with surface electrodes, is advo-
Electromyographic evidence for a guarding reflex cated for all people presenting with sacral dys-
can be seen during the normal urodynamic test, function (Fowler et al., 2002). For convenience,
that is, as the bladder reaches its full capacity (end the pudendo-anal reflex (PAR) has proved to be
fill volume, EFV) the striated sphincter EMG sig- one of the easiest and most useful sacral somatic
nal reaches its maximum peak-to-peak amplitude. reflexes to test (Fig. 1). The normal pudendal-anal
To reiterate, the normal guarding reflex seems to reflex can be easily facilitated by voluntary con-
function to automatically enhance sphincter clo- tractions of the pelvic floor (Fig. 1C).
sure, and in the process, helps to inhibit unwanted
activity of the bladder, and perhaps also the bowel,
so helping to prevent incontinence probably
through contraction-activated pudendal afferents. Pudendal urethral and anal reflexes
It may be that, part of a so-called guarding re-
flex could also involve smooth muscle responses in Interestingly, the pudendal-anal reflex often mir-
the bladder neck, proximal urethra and internal rors the pudendo-urethral reflex and can be used
anal sphincter. However, whereas in striated mus- as its surrogate (Podnar and Vodusek, 2001). It is
cle, electromyographic signals are proven to be known that the motoneurones for both the anal
real and related to muscle action potentials, in and urethral sphincters are sited close together in
smooth muscle, this relationship is much more Onuf’s nucleus in humans, which is in contrast to
controversial because of the presence of large most other non-primate animals (Blok, 2002). Op-
movement artifact (Craggs, 1998). Therefore, at timal recruitment of the pudendal-anal reflex is
this time we can only resort to measuring the slow achieved by using a condition-test paradigm to
pressure changes associated with autonomic func- stimulate the dorsal penile or clitoral nerves (ex-
tions of the urethra and anal canal. clusively pudendal sensory nerves) with pairs of
As described previously, the peripheral afferent electrical pulses through skin electrodes. The reflex
limb of the guarding reflex is conveyed by sensory is recorded as a compound motor-evoked poten-
afferent fibers in the pelvic nerves projecting from tial of the sphincter. By using a pressure sensor
the bladder or bowel to the sacral cord, whereas with integral ring electrodes to record the pressure
the efferent limb is via the pudendal motor nerves and surface-evoked motor potentials simultane-
to the pelvic floor and sphincters from Onuf’s nu- ously in the anal canal (Fig. 1A), it has been shown
cleus in the sacral ventral cord. Interestingly, re- that the best recruitment of Onuf’s motoneurones
cent neurophysiological testing of the pathways of is achieved when the double-pulse interval is be-
a similar reflex (bladder-anal) has revealed a long tween 2–4 ms (Fig. 1B) (Rodi and Vodusek, 1995).
210

A C
Pudendo -Anal

100 µV
Reflex
V

Anal
Sphincter

250 V
10 cmH2 O
Pressure

probe
Electrical Stimulation
Anal

(+ voluntary sphincter contraction)


Of the Pudendal
Afferent Nerves
DPN Stimulation
0.3s (Dorsal Penile Nerve) Paired pulses 100ms
ms
2ms pulse interval
Paired pulses 200s
200 s
@ 15mA (2 x PAR Threshold)

B
n = 10 The effect of paired pulse
0 ms separation on EAS amplitude
1 ms
Pudendal Anal Reflex

150
EMG Amplitude (V)
EMG Amplitude µV
V

2 ms
100µV
100 V

4 ms 100
8 ms
16 ms 50
32 ms
64 ms 0.00
0
Paired Pulse Separation (msec)
0 0.05 0.10 s Paired-pulse interval ms

Fig. 1. The Pudendo-Anal Reflex (PAR). A. Stimulation and recording setup for studies of this reflex. Pudendal afferent fibers in the
dorsal penile (or dorsal clitoral) nerves are stimulated to evoke the reflex. B. Paired-pulse stimulation at a 2–4 ms interval elicits an
optimal-compound motor-evoked potential response from the anal sphincter. C. The response is further facilitated by voluntary
contraction of the sphincter muscles.

Pelvo-pudendal reflex integration Aberrant somato-visceral reflexes following SCI

By combining pudendal-anal reflex measurement Damage to the spino-bulbo-spinal pathways (con-


with urodynamics (or proctodynamics), it is pos- necting the lumbar-sacral segments with the brain-
sible to demonstrate facilitatory effects of the pel- stem) in SCI, whether complete or incomplete, can
vic afferent activity from the bladder (or rectum) cause serious disruption to coordination of the
on the sphincters. In a recent study on bladder and pelvic organs and sphincters leading to un-inhib-
bowel filling, it was shown that modulation of the ited pelvic (parasympathetic) reflexes. In the blad-
reflex, reflecting the effects of a guarding reflex, der, this results in what is commonly termed
increased for both organs at end fill volume (EFV) detrusor hyperreflexia or neurogenic detrusor
(Chung et al., 2004). As expected, the pudendal- over-activity (Fig. 3). However, it is not clear
anal reflex was suppressed during bladder empty- whether over-activity is a regular feature seen in
ing in healthy subjects (Fig. 2). This latter finding the rectum or descending colon following SCI; in
confirmed an earlier study (Dyro and Yalla, 1986) fact, the common occurrence of constipation
and correlated well with the standard electromyo- might indicate otherwise although drugs used to
graphic findings seen with filling and voiding suppress the overactive bladder may be relevant in
cystometrograms. this context.
211

Fig. 2. Modulation of the normal PAR during cystometry. For these studies, the PAR can be conveniently used as a surrogate for the
pudendo-urethral reflex (see explanation in the text). A. During filling cystometry, the sphincter EMG and PAR increase progressively
as EFV is reached, demonstrating the presence of a ‘‘guarding reflex’’. During voiding, the PAR and sphincter EMG are markedly
suppressed, consistent with the need for unobstructed bladder emptying. B. A graphical presentation of the responses of peak-to-peak
PAR in five healthy control subjects (mean 7 SD) at EFV and during voiding. For comparison across the group, the responses for
each subject were normalized to the size of the PAR when the bladder was empty.

Detrusor hyperreflexia is often exacerbated by Efferent effects


uncoordinated viscero-somatic reflexes giving im-
paired vesico-urethral function (detrusor–sphinc- By cutting off the descending excitatory modulat-
ter dyssynergia), leading to obstructed voiding, ing influences from the lateral pontine nucleus to
high bladder pressures and incontinence (Has- Onuf’s nucleus in the sacral cord, we could expect
souna et al., 2004). If left untreated, it is these high poor or absent sphincter guarding reflexes and the
pressures that can lead to renal failure. The usual loss of reflex inhibition of detrusor activity during
management for the neurogenic bladder is a com- the filling phase (Park et al., 1997). Conversely, if
bination of anti-cholinergic drugs to suppress the proper activation of the medial pontine nucleus
over-activity and clean intermittent catheterization were dysfunctional, then the drive necessary for
to empty the bladder. bladder emptying would also be missing.
It appears that a combination of these effects
and the loss of voluntary descending control of the
Afferent effects sphincters and facilitation of the guarding reflex
could be to disinhibit visceral Ad afferent activity,
For the lower urinary tract, the effect of a com- allowing unfettered sacral hyperreflexia to take
plete spinal cord lesion is to block ascending af- over reducing bladder capacity. In addition, the
ferent activity from the bladder or urethra, loss of any descending inhibitory pathways from
reaching the brain-stem peri-aqueductal gray dur- the brain stem, normally keeping reflexes mediated
ing filling and prevent proper pontine coordina- by bladder C-fiber afferents in check, could also
tion of the bladder and sphincters. In fact, all make the bladder more sensitive to distension,
sensations from the lower urinary tract, bowel, again tending to drive the bladder toward lower
pelvic floor and sphincters would be absent as a capacity (Yoshimura, 1999) [see De Groat and
result of such a lesion and this would include all Yoshimura, this volume]. The exact mechanism of
other proprioceptive pathways normally linking C-fiber hyper-excitability after SCI is not entirely
the pelvic floor more directly with the somato- clear. It could be due to a change in the mechanical
sensory cortex. The loss of pelvic sensation is, sensitivity of the sensory endings, but more likely
perhaps, one of the most disabling features of SCI. it involves reorganization in the spinal neural
212
A B C
Detrusor Hyperreflexia

<F>

DRG Detrusor Pressure

Urine Flow

Pelvic Intraurethral Pressure


nerves
Pudendal
nerves Sphincter EMG Sphincter Dyssynergia

Fig. 3. Reflexes of the lower urinary tract after SCI. A. Aberrant pelvic reflexes causing detrusor hyperreflexia and detrusor–external sphincter dyssynergia. B. Cystometry
in a cord-injured person with a complete injury, showing the high detrusor pressures generated during sphincter dyssynergia measured by electromyography. C. A
simultaneous X-ray frame showing the moment of detrusor–sphincter dysynergia.
213

circuitry (Shefchyk, 2002). Perhaps there is prolif- Spinal Injuries Association, ASIA A) spinal cord
eration of sacral spinal connections of these affer- lesion, but it is often present but variable in people
ent pathways, brought about by new occupation with incomplete lesions (Siroky and Krane, 1982).
of vacant excitatory post-synaptic receptor sites in Therefore, it is believed that the partial preserva-
the sacral segmental pelvic reflex pathway. The tion or loss of the guarding reflex, in response to
vacancy might follow degeneration of the excita- bladder filling, would be an indication of the com-
tory pathway from the medial pontine nucleus, pleteness of spinal injury.
which under normal bladder filling conditions
would be silent but comes into play to activate
The bladder guarding reflex
the detrusor for voiding. Speculatively, these re-
ceptor sites in the sacral cord could be taken over
Recent studies of neurourological function (Balasu-
by collateral sprouting of the bladder afferents.
bramaniam et al., 2004) have confirmed that at
bladder EFV (defined as the volume at which void-
Inhibitory effects ing or neurogenic detrusor overactivity, NDO, oc-
curs), the guarding reflex, measured by the
Whatever the precise mechanisms generating de- modulation of the PAR (see Fig. 2), is absent or
trusor hyperreflexia, it has been known for many very weak in most people with a neurologically de-
years from studies in experimental animal models fined complete supra-sacral spinal cord lesion
and clinically that pudendal afferent stimulation (Fig. 5). In people with incomplete lesions (ASIA
can inhibit bladder reflexes (Lindstrom et al., 1983; B-D), the guarding reflex is often preserved but very
Fall et al., 1991). For example, in recent studies it variable. It has also been demonstrated that a weak
has been shown that bladder activity can be pro- guarding reflex is often associated with low bladder
foundly suppressed by magnetic stimulation of the capacity, perhaps as a result of weak pudendal in-
pudendal afferents in the spinal roots (Sheriff hibition on aberrant detrusor contractions.
et al., 1996) or by electrical stimulation of the In contrast to healthy volunteers who have a
dorsal penile nerves in healthy volunteers, in sub- very suppressed level of PAR during voiding, most
jects with idiopathic detrusor over-activity as well people with a complete or incomplete supra-sacral
as in subjects with complete spinal cord lesions lesion have an exaggerated pudendal reflex as ex-
(Shah et al., 1998) (Fig. 4B). This suggests that pected during vesico-sphincter dyssynergia (Dyro
reflexes involving somato-visceral inhibitory inter- and Yalla, 1986; Sethi et al., 1989).
actions are present at the local sacral segmental
level whether or not the spinal cord is intact
The bowel guarding reflex
(Fig. 4A) (Craggs and MacFarlane, 1999). More
importantly for people with spinal cord injuries,
A recent addition to this study has interestingly
such stimulation (popularly referred to as neuro-
discovered that whilst an analogous bowel ‘‘guard-
modulation) not only inhibits detrusor hype-
ing reflex’’ during rectal filling exists, it appears to
rreflexia but also increases bladder capacity
be present irrespective of SCI (Chung et al., 2004).
significantly (Kirkham et al., 2001), which could
Perhaps this is a reflection of the greater depend-
provide a useful alternative treatment to drugs.
ence on automatic control in bowel function; the
guarding reflex requiring only sacral segmental
The guarding reflex after SCI control. The external anal sphincter is the auto-
matic braking system in the proper coordination
Synthesis of the guarding reflex appears to require of the bowel and sphincters for continence, but in
involvement of the pontine micturition centre as proper bowel function, continence is a differential
well as the integrity of supra-sacral pathways. The process depending on whether the rectal content is
guarding reflex is said to be absent in over 85% of gas, solid or liquid. In man, it is social circum-
cord-injured people with a complete (American stances and voluntary control of the sphincters
A B
140

Pressure
cm H2O
Bladder
0

Stimulation

C
Neuromodulation
200

Bladder Capacity
Mean % Change
Electrical
Stimulation
of Pudendal
Afferent
100
Pathways
at the Dorsal
Penile Nerves
0
Serial Cystometrograms

Fig. 4. Controlling detrusor hyperreflexia by non-invasive neuromodulation through pudendal afferent pathways. (A) By stimulating
the dorsal penile (or clitoral) nerves with electrical pulses between 10–20 per second and above twice the threshold for the pudendo-
anal reflex it is possible to profoundly suppress detrusor hyperreflexia. (B) The upper trace shows the effect of continuous stimulation
of the dorsal penile nerves on the bladder pressure rise associated with a detrusor hyperreflexia contraction provoked at the middle
arrow. Control hyperreflexic contractions provoked at the other arrows can be seen before and after stimulation. Again this response is
flanked by control provocations. (C) Repeated cystometrograms with continuous neuromodulation (shaded area) demonstrating
significant increases in bladder volume when compared to control fills. Following stimulation the bladder takes some time to restore to
its smaller capacity probably as a result of stretching of the bladder wall during the period of neuromodulation.

A B
End Fill Volume ASIA E – Non SCI ASIA A – Complete SCI
Voiding or Overactivity ASIA B-D – Incomplete SCI
1.0
(standardised to an empty
normalised PAR +/-SD

0.6
*
Mean peak-peak PAREFV

* 0.5
0.5
(mV mean +/-SD)

0.4

** 0.3
0.0 0.2
bladder=0)

0.1
0
-0.5
-0.1 200 400 600 800
-0.2
-1.0 -0.3
Non-SCI Incomplete Complete Mean Bladder EFV (mL mean+/-SD)
Subjects SCI SCI
Subjects Subjects

Fig. 5. Changes in the bladder guarding reflex after SCI. For an explanation of the relationship between the measured pudendo-anal
reflex during bladder filling and the guarding reflex refer to Fig. 2 Panel A shows the changes in the peak-to-peak amplitude of the
normalized PAR at EFV and voiding (or NDO in the case of the two SCI groups). Subjects with a complete spinal lesion have little or
no guarding reflex (white bars) compared to subjects without SCI, whereas subjects with incomplete injuries have a very variable reflex.
This variability probably reflects the wide range of neurological impairment in incomplete subjects. In subjects with a complete injury,
the PAR change (striped bars) reflects the presences of sphincter dyssynergia. B. Subjects with a complete lesion present with the
smallest guarding reflex and bladder capacity suggesting that there is little reflex inhibition of the detrusor (that is, neurogenic detrusor
over-activity occurs at low bladder volumes) (Balasubramaniam et al., 2004).
215

that determine the final emergency brake for con- their pelvic floor and sphincter reflexes including
tinence. the guarding reflex.
For definitions of neurological grading see ap-
pendix [American Spinal Injuries Association/In-
Volitional effects on sphincter reflexes after SCI ternational Medical Society of Paraplegia (ASIA/
IMSOP), 1996] adapted from an original classifi-
Although most of the neural circuits involved in cation scheme by Frankel et al. (1969).
the normal control of the bladder and bowel are
autonomic, continence is very much a function of
Neurophysiological measures of volition on
volitional control. Voluntary contraction of the
pelvic sphincters
pelvic floor muscles plays an important role in
normal continence mechanisms particularly during
Measuring the effects of volition on the pelvic
postponement of voiding. When the bladder or
sphincters in some cord-injured people, for exam-
bowel reach their near-maximal capacity at EFV,
ple, those with a putative complete injury (as de-
not only does contraction of the sphincters prevent
termined by the ASIA classification), is difficult.
inadvertent leaking, but such contractions proba-
To overcome these difficulties, a recent study
bly inhibit the parasympathetic reflex pathways
(Balasubramaniam et al., 2005) has tested subjects
within the spinal cord to suppress premature void-
by presenting them with a visual signal instructing
ing contractions. This mechanism could operate
them to make a voluntary squeeze of their anal
directly via descending cortico-spinal pathways
sphincter (or attempt to do so) and hold it for the
leading to inhibition of visceral reflexes. Indirectly,
duration of the signal (Fig. 6). Clearly, for those
there could be facilitation of the motor component
with a complete lesion, there was no internal sen-
of the guarding reflex that in turn activates pu-
sory feedback, but by making their anal sphincter
dendal afferents to suppress the visceral reflexes.
EMG audible, they could hear whether they were
Such a mechanism would be similar to that envis-
successful at the task. The EMG was integrated to
aged for electrical neuromodulation (Craggs and
determine the mean peak amplitude. During the
McFarlane, 1999). Interestingly, the sensations of
attempted contraction, the computer controlling
a full bladder and the strong desire to void can
the experiment elicited a pudendal-anal reflex. The
also be suppressed by voluntary contractions of
test was repeated ten times and averaged puden-
the pelvic floor, or again, by therapeutic neuro-
dal-anal reflexes and integrated EMGs were de-
modulation (Oliver et al., 2003).
termined for each subject with an empty bladder
It remains to be determined whether people with
and rectum.
an incomplete SCI and some volitional control
The results compared non-SCI subjects with co-
over their pelvic floor can also voluntarily inhibit
horts of complete and incomplete chronic spinal
their detrusor hyperreflexia and suppress the desire
cord injuries. The study concluded that both the
to void.
pudendal-anal reflex and integrated EMG corre-
lated reasonably well with ASIA grading taken at
the same time, however, it was the pudendal-anal
Neurological grading and pelvic floor reflexes
reflex measurement that correlated more system-
atically with the neurological assessment of injury.
In complete supra-sacral SCI (ASIA A), we would
expect all volitional effects and sensations related
to pelvic function to be lost. In addition, we might Sensitivity of sacral reflex measurement and
also expect a very poor or absent guarding reflex neurological assessment in SCI
during bladder filling (see Fig. 5). On the other
hand, in people with incomplete lesions (ASIA B- Routine assessment of patients with SCI currently
D), the picture would be much more variable with relies on subjective clinical measurement using the
some preservation of voluntary modulation of ASIA classification and Impairment Score. It is
216

A Bladder Empty

PAR
EMG

PAR 50µV
+ Squeeze
150ms
EMG 500µV
+ Squeeze
3s
Integrated
EMG

Squeeze

B Integrated EMG C PAR Response


400
% Change in Integrated

% Change in peak to

200
PAR peak to peak

300
amplitude
amplitude
EMG peak

200
100

100

0
0
Non-SCI Incomplet Complete Non-SCI Incomplet Complete
Subjects e SCI SCI Subjects e SCI SCI
Subjects Subjects Subjects Subjects

Fig. 6. Neurophysiological measures of volition on the pelvic sphincters. A. During voluntary contractions of the pelvic floor muscles
and sphincters (squeeze) the PAR is facilitated. B and C show the change in anal sphincter EMG and PAR amplitude, respectively,
during voluntary anal squeeze (Balasubramaniam et al., 2005).

not always consistently rated and neither does it the tests will be for assessing the outcome of ther-
assess autonomic function. However, recent so- apeutic interventions for functional restoration.
mato-sensory (Krassioukov et al., 1999) and so-
mato-motor (Smith et al., 2000) testing are
introducing more objective neurophysiological Conclusions
measures into the evaluation of people with in-
complete SCI. As described here, there is little Regardless of the approach for functional resto-
doubt that objective measures of pelvic function ration, whether by promoting natural recovery
through neurophysiological testing of somato-vis- with neuro-protection, tapping into cortical plas-
ceral reflexes could also help to evaluate auto- ticity (Belci et al., 2004), implanting novel devices
nomic function; there appears to be a good to stimulate residual neural pathways (Grill et al.,
correlation with the standard neurological assess- 2001; Craggs, 2004), or ultimately developing a
ment of injury. It remains to be seen how sensitive ‘‘cure’’ through functional neural repair (Ramer
217

et al., 2005), there is going to be a need to assess level and includes the
carefully the benefits through more sensitive meas- sacral segments S4–S5.
ures of autonomic neural function. C Incomplete Motor function is
By using a combination of urodynamic and preserved below the
proctodynamic techniques, ideally with simultane- neurological level, and
ous video-imaging, together with the sort of ob- more than half of key
jective neurophysiological measures described in muscles below the
this chapter, we can look forward to developing neurological level have
reliable protocols for sensitive assessment of au- a muscle grade less
tonomic function of the pelvic organs. Such proto- than 3.
cols (together with those for assessing other D Incomplete Motor function is
autonomic physiological disturbances in SCI) will preserved below the
help us to characterize all autonomic dysfunction neurological level, and
more objectively. These can then be combined with at least half of key
the other well-established neurological and neuro- muscles below the
physiological assessments to give a much more neurological level have
comprehensive picture of SCI and the effect of in- a muscle grade of 3 or
terventions. The ultimate goal for all those con- more.
cerned with autonomic function of the bladder and E Normal Motor and sensory
bowel will be to develop a robust impairment function are normal.
grading and scoring system for pelvic dysfunction.

Acknowledgments
References
I am grateful to Vernie Balasubramaniam for al-
ASIA/IMSOP. (1996) International Standards for Neurological
lowing me to use some of the data obtained in her and Functional Classification of Spinal Cord Injury-Revised.
research studies funded by the International Spinal American Spinal Injuries Association, Chicago, IL.
Research Trust, UK. Also, I thank Joe Wood- Balasubramaniam, A.V., Chung, E.A.L., Bycroft, J.A., Knight,
house and Alex Chung, two of my Clinical Re- S.L., Gall, A., Middleton, F.R.I., Fowler, C.J. and Craggs,
M.D. (2005). Modulation of Pelvic Floor Reflexes by Voli-
search Fellows, for reading the various draft
tion in Spinal Cord Injury. Proceedings of the Urological
manuscripts and making some very helpful com- Research Society, Royal College of Surgeons, London.
ments and suggestions whilst writing this chapter. Balasubramaniam, V., Bycroft, J., Wood, S., Middleton,
F.R.I., Fowler, C.J. and Craggs, M.D. (2004) Urinary guard-
ing reflex: Aberrant in spinal cord injury? Brit. J. Urol. Int.,
93(Suppl 4): 4.
Appendix Basinski, C., Fuller, E., Brizendine, E.G. and Benson, J.T.
(2003) Bladder-anal reflex. Neurourol. Urodyn., 22: 683–686.
Belci, M., Catley, M., Husain, M., Frankel, H.L. and Davey,
American Spinal Injuries Association Classifica-
N.J. (2004) Magnetic brain stimulation can improve clinical
tion Grades and Impairment Scale (1996). outcome in incomplete spinal cord injured patients. Spinal
Cord, 42: 417–419.
Grade Completeness Description
Blok, B.F. (2002) Central pathways controlling micturition and
A Complete No sensory or motor urinary continence. Urology, 59(Suppl 1): 13–17.
function preserved in Blok, B.F.F., Willemsen, A.T.M. and Hostege, G. (1997) A
the sacral segments PET study on brain control of micturition in humans. Brain,
S4–S5. 120: 111–121.
Burleigh, D.E. (1992) Pharmacology of the internal anal
B Incomplete Sensory but not motor
sphincter. In: Henry M.M. and Swash M. (Eds.), Colopr-
function is preserved octology and the Pelvic Floor. Butterworth-Heinemann,
below the neurological Oxford, pp. 37–53.
218

Burnstock, G. (1990) Innervation of the bladder and bowel. In: Cervero F. and Morrison J.F.B. (Eds.) Visceral Sensation,
Block G. and Whelan J. (Eds.) Neurobiology of Inconti- Progress in Brain Research, Vol. 69. Elsevier, Amsterdam,
nence, Ciba Foundation Symposium, Vol. 151. Wiley, Chich- pp. 87–114.
ester, pp. 2–26.
Chung, E.A.L., Balasubramaniam, A.V., Middleton, F., Craggs, Kirkham, A.P.S., Shah, N.C., Knight, S.L., Shah, P.J.R. and
Craggs, M.D. (2001) The acute effects of continuous and
M.D. and Emmanuel, A.V. (2004) Relationship between
bladder and bowel filling and the guarding reflex mechanism. conditional neuromodulation on the bladder in spinal cord
Brit. J. Urol. Int., 94: 1176. injury. Spinal Cord, 39: 420–428.
Craggs, M.D. (1998) The elusive electromyogram: Fact vs ar- Krassioukov, A., Wolfe, D.L., Hsieh, J.T., Hayes, K.C. and
Durham, C.E. (1999) Quantitative sensory testing in patients
tifact. Editorial comment. Neurourol. Urodyn., 17: 80–82.
Craggs, M.D. (2004) Future prospects for complete restoration with incomplete spinal cord injury. Arch. Phys. Med. Re-
of bladder function by neuroprostheses. In: Corcos J. and habil., 80: 1258–1263.
Lindstrom, S., Fall, M., Carlsson, C.A. and Erlandson, B.E.
Schick E. (Eds.), The Textbook of Neurogenic Bladder,
Adults and Children. Martin Dunitz, London, pp. 625–635 (1983) The neurophysiological basis of bladder inhibition in
Ch. 56. response to intravaginal electrical ‘‘stimulation’’. J. Urol.,
Craggs, M.D. and MacFarlane, J. (1999) Neuromodulation of 129: 405–410.
Mathers, S. (1992) Neural control of the pelvic sphincters. In:
the lower urinary tract. Exp. Physiol., 84: 149–160.
Craggs, M.D. and Vaizey, C.J. (1999) Neurophysiology of the Henry M.M. and Swash M. (Eds.), Coloproctology and the
bladder and bowel. In: Fowler C.J. (Ed.), Neurology of Pelvic Floor. Butterworth-Heinemann, Oxford, pp. 61–71.
Bowel, Bladder and Sex, (Blue Books of Practical Neurolo- Mundy, A.R. and Thomas, P.J. (1994) Clinical physiology
of the bladder, urethra, and pelvic floor. In: Mundy A.R.,
gy). Butterworth-Heinemann, Oxford, pp. 19–32.
De Groat, W.C. (1997) Central nervous system control of mi- Stephenson T.P. and Wein A.J. (Eds.), Urodynamics –
cturition. In: O’Donnell P. (Ed.), Urinary Incontinence. Mo- Principles, Practice, and Application. Churchill-Livingstone,
sby, St. Louis, pp. 33–47. Edinburgh, pp. 15–27.
Oliver, S.E., Fowler, C.J., Mundy, A.R. and Craggs, M.D.
Dyro, F.M. and Yalla, S.V. (1986) Refractoriness of urethral
striated sphincter during voiding: Studies with afferent pu- (2003) Measuring the sensations of urge and bladder filling
dendal reflex arc stimulation in male subjects. J. Urol., 135: during cystometry in urge incontinence and the effects of
neuromodulation. Neurourol. Urodynam., 33: 7–16.
732–736.
Engel, A.F., Kamm, M.A., Bartram, C.I. and Nicholls, R.J. Park, L.M., Bloom, D.A. and McGuire, E.J. (1997) The guard-
(1995) Relationship of symptoms in faecal incontinence to ing reflex revisited. Brit. J. Urol., 80: 940–945.
specific sphincter abnormalities. Int. J. Colorectal Dis., 10: Podnar, S. and Vodusek, D.B. (2001) Protocol for clinical ne-
urophysiologic examination of the pelvic floor. Neurourol.
152–155.
Fall, M. and Lindstrom, S. (1991) Electrical stimulation. A Urodynam., 20: 669–682.
physiologic approach to the treatment of urinary inconti- Ramer, L.M., Ramer, M.S. and Steeves, J.D. (2005) Setting the
nence. Urol. Clin. North Am., 18: 393–407. stage for functional repair of spinal cord injuries: A cast of
thousands. Spinal Cord, March 2005, 43(3): 134–161.
Fowler, C.J., Benson, J.T., Craggs, M.D., Vodusek, D.B.,
Yang, C.C. and Podnar, S. (2002) Clinical neurophysiology. Rodi, Z. and Vodusek, D.B. (1995) The sacral reflex studies:
In: Abrams P., Cardozo L., Khoury S. and Wein A. (Eds.), Single versus double pulse electrical stimulation. Neurourol.
Urodynam., 14: 496.
Incontinence (International Consultation on Incontinence
ICS/WHO). Health Publication Limited, Plymouth, Schrøder, H.D. and Reske-Nielsen, E. (1983) Fiber S types in
pp. 391–424. the striated urethral and anal sphincters. Acta Neuropathol.,
Frankel, H.D.H., Hancock, D.O., Hyslop, G., Melzak, J., 60: 278–282.
Michaelis, L.S., Ungar, G.H., Vernon, J.D.S. and Walsh, Sethi, R.K., Bauer, S.B., Dyro, F.M. and Krarup, C. (1989)
J.J. (1969) The value of postural reduction in the initial Modulation of the bulbocavernosus reflex during voiding:
management of closed injuries of the spine with paraplegia Loss of inhibition in upper motor neuron lesions. Muscle
and tetraplegia. Paraplegia, 7: 179–192. Nerve, 12: 892–897.
Grill, W.M., Craggs, M.D., Foreman, R.D., Ludlow, C.L. and Shah, N., Edhem, I., Knight, S.L., Shah, P.J.R. and Craggs,
Buller, J.R. (2001) Emerging clinical applications of electrical M.D. (1998) Acute suppression of provoked detrusor hype-
stimulation: Opportunities for restoration of function. J. Re- rreflexia by electrical stimulation of the dorsal penile nerve.
habil. Res. Dev., 38(6): 641–653. Eur. Urol., 33(Suppl 1): 60.
Hassouna, M., Elmayergi, N. and Abdelhady, M. (2004) Shefchyk, S.J. (2002) Spinal cord neural organization control-
Pathophysiology of spinal cord injury. In: Corcos J. and ling the urinary bladder and striated sphincter. Prog. Brain
Schick E. (Eds.), Textbook of the Neurogenic Bladder. Mar- Res., 137: 71–82.
tin-Dunitz, London, pp. 178–191 Ch. 14. Sheriff, M.K.M., Shah, P.J.R., Fowler, C.J., Mundy, A.R. and
Jänig, W. and Morrison, J.F.B. (1986) Functional properties of Craggs, M.D. (1996) Neuromodulation of detrusor hyper-
spinal visceral afferents supplying abdominal and pelvic or- reflexia by functional magnetic stimulation of the sacral
gans with special emphasis on visceral nociception. In: roots. Brit. J. Urol., 78: 39–46.
219

Siroky, M.B. and Krane, R.J. (1982) Neurologic aspects of de- Yoshimura, N. (1999) Bladder afferent pathway and spinal
trusor-sphincter dyssynergia, with reference to the guarding cord injury: Possible mechanisms including hyperreflexia of
reflex. J. Urol., 127: 953–957. the urinary bladder. Prog. Neurobiol., 57: 583–606.
Smith, H.C., Savic, G., Frankel, H.L., Ellaway, P.H., Maskill, Yoshimura, N., Seki, S. and Chancellor, M.B. (2004) Integrated
D.W., Jamous, M.A. and Davey, N.J. (2000) Corticospinal physiology of the lower urinary tract. In: Corcos J. and
function studied over time following incomplete spinal cord Schick E. (Eds.), Textbook of the Neurogenic Bladder. Mar-
injury. Spinal Cord, 38: 292–300. tin-Dunitz, London, pp. 73–87 Ch. 6.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 14

The clinical problems in cardiovascular control


following spinal cord injury: an overview

Andrei Krassioukov1,2,3,4, and Victoria E. Claydon1

1
International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC V6T 1Z4,
Canada
2
Division of Physical Medicine and Rehabilitation, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
3
School of Rehabilitation, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
4
Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada

Abstract: On a daily basis, individuals with cervical and upper thoracic spinal cord injury face the challenge
of managing their unstable blood pressure, which frequently results in persistent hypotension and/or
episodes of uncontrolled hypertension. This chapter will focus on the clinical issues related to abnormal
cardiovascular control in individuals with spinal cord injury, which include neurogenic shock, autonomic
dysreflexia and orthostatic hypotension. Blood pressure control depends upon tonic activation of sym-
pathetic preganglionic neurons by descending input from the supraspinal structures (Calaresu and Yardley,
1988). Following spinal cord injury, these pathways are disrupted, and thus spinal circuits are solely
responsible for the generation of sympathetic activity (Osborn et al., 1989; Maiorov et al., 1997). This
results in a variety of cardiovascular abnormalities that have been well documented in human studies, as
well as in animal models (Osborn et al., 1990; Mathias and Frankel, 1992a, b; Krassioukov and Weaver,
1995; Maiorov et al., 1997, 1998; Teasell et al., 2000). However, the recognition and management of these
cardiovascular dysfunctions following spinal cord injury represent challenging clinical issues. Moreover,
cardiovascular disorders in the acute and chronic stages of spinal cord injury are among the most common
causes of death in individuals with spinal cord injury (DeVivo et al., 1999).

Pathophysiology of cardiovascular dysfunction after icantly to our present understanding of the patho-
spinal cord injury physiology of autonomic dysreflexia and abnor-
mal cardiovascular control after spinal cord
The first description of one of the most common injury (Osborn, Taylor and Schramm, 1989, 1990;
autonomic disturbances in individuals with spinal Mathias and Frankel, 1992a; Krassioukov and
cord injury, known as autonomic dysreflexia, ap- Weaver, 1995; Maiorov, Weaver and Krassioukov,
peared in 1860 (Hilton, 1860). However, it was not 1997; Maiorov, Fehlings and Krassioukov, 1998;
until 1947 that Guttman and Whitteridge de- Krassioukov et al., 2002). Recently, numerous
scribed the possible mechanisms responsible for elements within autonomic circuits were identified
the development of this condition (Guttman and that could contribute to abnormal cardiovascular
Whitteridge, 1947). Both clinical observations and control after spinal cord injury: (a) descending
experimental animal data have contributed signif- vasomotor (sympathoexcitatory) pathways, (b)
sympathetic preganglionic neurons, (c) spinal in-
Corresponding author. Tel.: +001 604 822 2673; terneurons, (d) spinal afferents, and (e) peripheral
Fax: +001 604 822 2924; E-mail: krassioukov@icord.org neurovascular mechanisms. Spinal cord injury

DOI: 10.1016/S0079-6123(05)52014-4 223


224

disrupts the descending sympathoexcitatory path- reported that severe hypotension was present in all
ways provided by medullary neurons located with- 31 tetraplegic subjects assessed with severe spinal
in the rostroventrolateral medulla. These neurons cord injury, half of whom required pressor therapy
provide tonic input to the sympathetic pregangl- in order to maintain arterial blood pressure (Glenn
ionic neurons located within the lateral horns of and Bergman, 1997). In addition to the pro-
the spinal grey matter of the thoracic and upper nounced hypotension described, many patients
lumbar spinal segments (T1–L2). Disruption of with acute spinal cord injury experience severe ab-
these descending cardiovascular pathways results normalities in heart rate. Bradycardia was report-
in at least four phenomena: initial sympathetic ed in 64–77% of patients with cervical spinal cord
hypoactivity; alterations in the morphology of injury during the acute post-injury stage, and was
sympathetic preganglionic neurons; plastic chang- more severe and frequent within the first 5 weeks
es within the spinal circuits (including sprouting after injury (Piepmeier et al., 1985; Winslow et al.,
and the potential formation of inappropriate 1986; Lehmann et al., 1987). In contrast, when the
synaptic connections); and the development of injury is in the mid-thoracic spinal cord, leaving
changes in sympathetic neurovascular transmission cardiac sympathetic neurons under brainstem con-
and smooth muscle responsiveness (Krassioukov trol, and vagal and sympathetic influences more in
et al., 1999; Teasell et al., 2000; Yeoh et al., 2004). balance, bradycardia is a less severe problem.
The initial sympathetic hypoactivity results in low Furlan et al. (2003) and colleagues reported that
resting blood pressure, loss of blood pressure the hypotension and bradycardia observed initially
homeostasis and disturbed reflex control (Mathias after injury persisted in the individuals with more
and Frankel, 1992b). However, with time follow- severe injury of the descending cardiovascular
ing spinal cord injury, the loss of descending in- autonomic pathways. Moreover, all individuals
hibitory pathways and plastic changes within the in this group required vasopressor therapy in order
spinal cord, coupled with peripheral neurovascular to maintain systolic arterial blood pressure above
changes, are likely to predispose to episodes of 90 mmHg. In contrast, individuals with less severe
extreme hypertension, associated with autonomic injury to the descending cardiovascular pathways
dysreflexia, that frequently develop in both acute tended to show higher levels of blood pressure and
and chronic stages of spinal injury. heart rate, although minor and short-term hypo-
tension and low heart rates were occasionally
observed.
The acute post-injury period and neurogenic shock In addition to neurogenic shock, the acute phase
of spinal cord injury is also associated with ‘‘spinal
Acute spinal cord injury in humans, especially at shock’’ (Nacimiento and Noth, 1999; Ditunno
the cervical level, results in severe hypotension and et al., 2004). Some authors use these terms inter-
persistent bradycardia that are common compo- changeably, however, it is important to recognize
nents of the phenomenon known as neurogenic that these are two clinically important and distinct
shock (Atkinson and Atkinson, 1996). This event conditions. Neurogenic shock is characterized
is more profound and long lasting in humans after by changes occurring in blood pressure control
spinal cord injury than in experimental animals. following spinal cord injury, whereas spinal shock
Moreover, clinical observations strongly suggest is characterized by a marked reduction or aboli-
that the extent to which prolonged and severe hy- tion of sensory, motor or reflex function of the
potension requiring vasopressive therapy occurs is spinal cord below the level of injury (Ditunno
associated with the severity of the spinal cord in- et al., 2004). Clinically, spinal shock in humans
jury and cervical or thoracic location of the injury, can persist for days to weeks, with a mean dura-
and can last up to 5 weeks after injury (Mathias tion of between 4 and 6 weeks after the injury.
and Frankel, 1992b; Atkinson and Atkinson, 1996; Traditional views of the clinical course of the re-
Vale et al., 1997; Nacimiento and Noth, 1999; covery of spinal shock were related to the emer-
Hadley et al., 2002a). In one study, Glenn et al. gence of certain groups of reflexes. For example,
225

some considered that spinal shock had ended when (Mathias and Frankel, 1992a; DeVivo et al.,
the appearance of initial reflexes, such as the 1999). Untreated episodes of autonomic dysreflex-
bulbocavernosus reflex occurred in the first few ia may have serious consequences, including in-
days after spinal cord injury, others with the tracranial hemorrhage, retinal detachments,
recovery of deep tendon reflexes at 2 weeks post seizures and death (Yarkony et al., 1986; Pine
injury, while some groups classified the end of et al., 1991; Eltorai et al., 1992). Different noxious
spinal shock as when the bladder voiding reflexes and non-noxious stimuli such as bowel and blad-
recover after approximately 2 months. For further der distension, spasticity and pressure sores may
details, see Ditunno et al. (2004). provoke the sudden increases in arterial blood
pressure of autonomic dysreflexia (Teasell et al.,
2000). Dysreflexia is three times more prevalent in
Management of the acute period of spinal
quadriplegics with complete injury, in comparison
cord injury
to those with incomplete injury (Curt et al., 1997).
Finally, autonomic dysreflexia, even in quadriple-
Unfortunately, there are no prospective controlled
gics, is not always severe, and may be character-
studies on the effects of hypotension upon the
ized only by sweating and piloerection, or may
outcome following acute spinal cord injury in hu-
even be asymptomatic (Kirshblum et al., 2002).
mans. However, the occurrence of hypotension in
These cardiovascular abnormalities are attrib-
the acute period following traumatic cervical or
uted to autonomic instability, caused by changes
upper thoracic spinal cord injury or severe head
occurring within the spinal autonomic circuits in
injury has been shown to be associated with worse
both the acute and chronic stages following spinal
outcomes (King et al., 2000). Recent guidelines on
cord injury (Mathias and Frankel, 1992a; Teasell
arterial blood pressure management after acute
et al., 2000). The destruction of the descending
spinal cord injury (Hadley, 2002b) recommend
vasomotor pathways results in the loss of excita-
maintenance of mean arterial blood pressure at a
tory supraspinal input to the sympathetic pre-
level of at least 85–90 mmHg for the first week
ganglionic neurons, and is currently considered as
after acute spinal cord injury to ensure adequate
the major factor underlying the persistent arterial
spinal cord perfusion. Prompt treatment of hypo-
hypotension and lack of sympathetic tone seen af-
tension with restoration of blood volume (using
ter high spinal cord injury (Furlan et al., 2003).
both colloid and albumin) and vasopressive ther-
It is important to note, however, that although
apy with dopamine, and the a-adrenergic agonist,
autonomic dysreflexia occurs more often in the
phenylephrine were recommended to maintain the
chronic stage of spinal cord injury at or above the
mean blood pressure.
6th thoracic segment, there is clinical evidence of
early episodes of autonomic dysreflexia in the first
Autonomic dysreflexia days and weeks after the injury (Silver, 2000;
Krassioukov et al., 2003). In fact, it seems likely
Individuals with a cervical or high thoracic spinal that autonomic dysreflexia is under-recognized in
cord injury face life-long abnormalities of blood the acute phase of spinal cord injury. We recently
pressure control (Mathias and Frankel, 1992a; conducted a study in order to determine the inci-
Teasell et al., 2000). In general, the resting arterial dence and clinical associations of early autonomic
blood pressure in these individuals is lower than dysreflexia in individuals with acute traumatic spi-
in able-bodied subjects, often with disabling nal cord injury (Krassioukov et al., 2003). Among
episodes of orthostatic hypotension. However, 58 patients with acute traumatic spinal cord injury,
life-threatening episodes of autonomic dysreflexia, three individuals showed early autonomic dysre-
characterized by extreme hypertension accompa- flexia and all the three had complete cervical
nied by a pounding headache, slow heart rate and tetraplegia. The trigger mechanisms for autonomic
upper body flushing may also occur, where systolic dysreflexia were somatic pain, fecal impaction
blood pressure can reach up to 300 mmHg and abdominal distension. The earliest episode of
226

autonomic dysreflexia occurred on the 4th day post upright posture (orthostatic hypotension), partic-
injury. Patients with severe cervical spinal cord ularly in the acute phase of injury (Mathias, 1995;
injury are particularly susceptible to early onset of Cariga et al., 2002). The symptoms of orthostatic
autonomic dysreflexia (Krassioukov et al., 2003). hypotension in spinal cord injured individuals are
similar to those seen in other populations experi-
encing orthostatic hypotension, and are associated
Management of autonomic dysreflexia
with cerebral hypoperfusion (Cleophas et al.,
1986). The symptoms commonly include fatigue
In patients with spinal cord injury, appropriate
or weakness, light-headedness, dizziness, blurred
bladder and bowel routines, in addition to the
vision, dyspnea and restlessness (Frisbie and
prevention of pressure sore development, are the
Steele, 1997; Sclater and Alagiakrishnan, 2004).
most effective measures for prevention of auto-
In one study (Illman et al., 2000), 41.1% of spinal
nomic dysreflexia. However, for each individual,
cord injured individuals who developed orthostatic
the identification and elimination of specific trig-
hypotension were asymptomatic despite significant
gers for autonomic dysreflexia should also be em-
blood pressure falls. Concerning the incidence and
ployed to manage and prevent episodes of
prevalence of orthostatic hypotension in this pop-
autonomic dysreflexia (Mathias and Frankel,
ulation, orthostatic maneuvers performed during
1992a; Teasell et al., 2000). The initial manage-
physiotherapy and mobilization are reported to
ment of an episode of autonomic dysreflexia
induce blood pressure changes, diagnostic of
should involve placing the patient in an upright
orthostatic hypotension, in 74% of spinal cord
position in order to provoke an orthostatic reduc-
injured individuals, suggesting that orthostatic
tion in blood pressure, and the loosening of any
hypotension is a common phenomenon among
tight clothing. Throughout the episode, the blood
the spinal cord injured population (Illman et al.,
pressure should be checked at 5 min intervals. It is
2000).
then necessary to search for and eliminate the pre-
Orthostatic hypotension following spinal cord
cipitating stimulus, which is most commonly (in
injury appears to be related to excessive pooling of
85% of cases) related to either bladder distention
blood in the viscera and dependent extremities,
or bowel impaction (Mathias and Frankel, 1992a;
presumably due in part to the absence or low level
Teasell et al., 2000). The use of antihypertensive
of efferent sympathetic nervous activity and to the
drugs should be considered as a last resort, but
loss of the reflex vasoconstrictor effect of arterial
may be necessary if the blood pressure remains
baroreceptor unloading by the sympathetic pre-
elevated after following the aforementioned steps.
ganglionic neurons below the lesion (Mathias,
1995). This is likely to be compounded by the loss
Orthostatic hypotension of lower extremity muscle function that is known
to be important in counteracting venous pooling in
Low arterial blood pressure is a problem in both the upright position. The resultant excessive ve-
acute and chronic high-level spinal cord injured nous pooling in the lower extremities, and reduced
patients. Indeed, Mathias and colleagues noted blood volume in the intrathoracic veins leads to a
that there was an inverse linear relationship reduced pressure in the large veins draining into
between the level of spinal cord injury and resting the atria of the heart (Jacobsen et al., 1992; Faghri
blood pressure (Mathias and Frankel, 1992a; et al., 2001). This in turn results in a decrease in
Mathias, 1995). This lower resting blood pressure ventricular end-diastolic filling pressure and stroke
is thought to be secondary to a reduction in sym- volume (Ten Harkel et al., 1994), leading to a
pathetic nervous activity below the level of the decrease in cardiac output and arterial pressure.
spinal cord injury. Tachycardia may occur as a consequence of
In addition to the low resting blood pressure, reduced cardiac parasympathetic (vagal) activity,
many individuals with high spinal cord injury also reflexly induced by unloading the arterial bar-
experience a further drop in blood pressure in the oreceptors, but this response is not usually
227

sufficient to compensate for the decreased stroke cerebral ischemia. The major goals of non-phar-
volume, and blood pressure remains low. macologic therapy for orthostatic hypotension are
It is likely, however, that there are additional the expansion of blood volume and avoidance of
factors that may predispose spinal cord injured excessive venous pooling (Oldenburg et al., 2002).
individuals to orthostatic hypotension. Individuals Shannon et al. reported that drinking water prior
with spinal cord injury are reported to have im- to meals may reduce the risk of developing post-
paired baroreflex function (Wecht et al., 2003), prandial orthostatic hypotension (Shannon et al.,
smaller plasma volumes due to hyponatremia 2002). Plasma volume expansion by increased salt
(Frisbie and Steele, 1997), and possible cardiovas- intake is also recommended, particularly in the
cular deconditioning, at least in the early period mornings, at which time individuals with spinal
following spinal cord injury, due to prolonged pe- cord injury are reported to be more prone to hy-
riods of bed rest (Vaziri, 2003). Any combination potension (Oldenburg et al., 2002). It is also rec-
of these additional factors following spinal cord ommended to sleep with the head of the bed raised
injury would be likely to further increase the like- by 15–301, which increases plasma volume, possi-
lihood and severity of orthostatic hypotension. bly by activation of the renin–angiotensin–aldos-
Orthostatic hypotension is usually primarily as- terone system, and thus reducing the excretion of
sociated with the acute phase of spinal cord injury sodium during the night (Oldenburg et al., 2002).
and improves over time. Although the reasons for Another commonly used measure to prevent or-
this improvement have not been clearly estab- thostatic hypotension in individuals with spinal
lished, the view that orthostatic hypotension is cord injury is the use of abdominal and lower ex-
only a temporary problem for spinal cord injured tremity compression bandages to prevent pooling
individuals may not be entirely accurate. Indeed, of the blood in these regions (Tanaka et al., 1997).
there is some evidence to suggest that, in some Pharmacological treatment for orthostatic
cord-injured people, these troublesome episodes of hypotension may be used as an alternative or
orthostatic hypotension can persist for many adjunct, but is usually only considered when
years, and may even become worse with time non-pharmacological management fails (Mukand
(Frisbie and Steele, 1997). It may be, however, that et al., 2001; Oldenburg et al., 2002; Nieshoff
with time, spinal cord injured individuals become et al., 2004). The physiological targets for phar-
more tolerant to these hypotensive episodes. There macological therapy are essentially the same as for
is some evidence to suggest that people with spinal non-pharmacological therapy: expansion of the
cord injury are able to tolerate profound hypo- circulating blood volume and prevention of
tension without symptoms (Illman et al., 2000), venous pooling (Oldenburg et al., 2002). One of
and that this is related to alterations in cerebral the most commonly used agents is fludrocortisone,
autoregulation such that cerebral blood flow a mineralocorticoid that leads to sodium and
(Gonzalez et al., 1991) and/or oxygenation water retention (Groomes and Huang, 1991;
(Houtman et al., 2000) are maintained, even in Barber et al., 2000), thus causing expansion of
the face of low cerebral perfusion pressures. the blood volume, and might also increase
a-adrenoceptor sensitivity. Side effects of this
treatment include hypokalemia and excessive wa-
Management of orthostatic hypotension ter retention, which limits its use in the elderly and
in individuals with congestive heart failure. The
Recognition of the symptoms of orthostatic most commonly prescribed agent for the manage-
hypotension (lightheadedness, dizziness, blurred ment of orthostatic hypotension following after
vision, fatigue and others) is crucial in the man- spinal cord injury is midodrine (Barber et al., 2000;
agement of individuals with spinal cord injury. In Mukand et al., 2001; Sclater and Alagiakrishnan,
emergency situations, it is imperative to return the 2004). Midodrine is rapidly absorbed after oral
patient to a horizontal position and elevate the administration with peak serum levels occurring in
lower extremities in order to prevent prolonged approximately 30 min. In the body it is metabolized
228

to desglymidodrine, an active a-1 adrenoreceptor spinal cord injury. Arch. Phys. Med. Rehabil., 80(11):
agonist. Side effects of midodrine include pruritus, 1411–1419.
piloerection and urinary retention (Mukand et al., Ditunno, J.F., Little, J.W., Tessler, A. and Burns, A.S. (2004)
Spinal shock revisited: a four-phase model. Spinal Cord,
2001). Patients with autonomic dysreflexia could 42(7): 383–395.
be very sensitive to catecholamines and midodrine Eltorai, I., Kim, R., Vulpe, M., Kasravi, H. and Ho, W. (1992)
as a result of changes in peripheral vascular Fatal cerebral hemorrhage due to autonomic dysreflexia in a
neurotransmission and vascular responsiveness. tetraplegic patient: case report and review. Paraplegia, 30:
Therefore, treatment should be started with low 355–360.
Faghri, P.D., Yount, J.P., Pesce, W.J., Seetharama, S. and
doses, and gradually titrated to the effective dose Votto, J.J. (2001) Circulatory hypokinesis and functional
(Mukand et al., 2001). electric stimulation during standing in persons with spinal
In conclusion, cardiovascular dysfunction is a cord injury. Arch. Phys. Med. Rehabil., 82(11): 1587–1595.
serious problem after spinal cord injury that varies Frisbie, J.H. and Steele, D.J.R. (1997) Postural hypotension
with the severity and location of injury and with and abnormalities of salt and water metabolism in myelopa-
thy patients. Spinal Cord, 35(5): 303–307.
the time after injury. The degree of cardiovascular
Furlan, J.C., Fehlings, M.G., Shannon, P., Norenberg, M.D.
dysfunction after spinal cord injury correlates well and Krassioukov, A.V. (2003) Descending vasomotor path-
with the severity of injury to the spinal motor and ways in humans: correlation between axonal preservation
sensory pathways. Currently, treatments for and cardiovascular dysfunction after spinal cord injury.
cardiovascular problems after cord injury can J. Neurotrauma, 20(12): 1351–1363.
Glenn, M.B. and Bergman, S.B. (1997) Cardiovascular changes
address only the adverse symptoms. The unique
following spinal cord injury. Topics in Spinal Cord Injury
pathways in the spinal cord and organization of Rehabil., 2(4): 47–53.
spinal circuits controlling cardiovascular function Gonzalez, F., Chang, J.Y., Banovac, K., Messina, D.,
must be understood more completely in humans to Martinez-Arizala, A. and Kelle, R.E. (1991) Autoregulation
permit specific targeting of cardiovascular of cerebral blood flow in patients with orthostatic hypoten-
dysfunctions by future strategies for protection sion after spinal cord injury. Paraplegia, 29: 1–7.
Groomes, T.E. and Huang, C.T. (1991) Orthostatic hypoten-
and repair of the injured cord. sion after spinal cord injury: treatment with fludrocortisone
and ergotamine. Arch. Phys. Med. Rehabil., 72(1): 56–58.
Guttman, F.L. and Whitteridge, D. (1947) Effects of bladder
References distention on autonomic mechanisms after spinal cord inju-
ries. Brain, 70: 361–404.
Atkinson, P.P. and Atkinson, J.L.D. (1996) Spinal shock. Hadley, M. (2002a) Blood pressure management after acute
Mayo Clin. Proc., 71: 384–389. spinal cord injury. Neurosurgery, 50(3): S58–S62.
Barber, D.B., Rogers, S.J., Fredrickson, M.D. and Able, A.C. Hadley, M.N., Walters, B.C., Grabb, P.A., Oyesiku, N.M.,
(2000) Midodrine hydrochloride and the treatment of ortho- Przybylski, G.J., Resnick, D.K. and Rylen, T.C. (2002b)
static hypotension in tetraplegia: two cases and a review of Guidelines for the management of acute cervical spine and
the literature. Spinal Cord, 38(2): 109–111 Calaresu, F.R. spinal cord injuries. Neurosurgery, 50(Suppl 3): S1–S199.
and Yardley, C.P. (1988) Medullary basal sympathetic tone. Hilton, J. (1860) Pain and therapeutic influence of mechanical
Ann. Rev. Physiol., 50: 511–524. and physiological rest in accidents and surgical diseases.
Cariga, P., Ahmed, S., Mathias, C.J. and Gardner, B.P. (2002) Lancet, 2: 401–404.
The prevalence and association of neck (coat-hanger) pain Houtman, S., Colier, W.N., Oeseburg, B. and Hopman, M.T.
and orthostatic (postural) hypotension in human spinal cord (2000) Systemic circulation and cerebral oxygenation during
injury. Spinal Cord, 40(2): 77–82. head-up tilt in spinal cord injured individuals. Spinal Cord,
Cleophas, T.J.M., Kauw, F.H.W., Bijl, C., Meijers, J. and 38: 158–163.
Stapper, G. (1986) Effects of beta-adrenergic-receptor ago- Illman, A., Stiller, K. and Williams, M. (2000) The prevalence
nists and antagonists in diabetics with symptoms of postural of orthostatic hypotension during physiotherapy treatment in
hypotension — a double-blind, placebo-controlled study. patients with an acute spinal cord injury. Spinal Cord, 38(12):
ANGIA, 37(11): 855–862. 741–747.
Curt, A.A., Nitsche, B., Rodic, B., Schurch, B. and Dietz, V. Jacobsen, T.N., Nielsen, H.V., Kassis, E. and Amtorp, O.
(1997) Assessment of autonomic dysreflexia in patients with (1992) Subcutaneous and skeletal-muscle vascular-responses
spinal cord injury. J. Neurol. Neurosurg. Psychiatry., 62: in human limbs to lower-body negative-pressure. Acta
473–477. Physiol. Scand., 144(3): 247–252.
DeVivo, M.J., Krause, J.S. and Lammertse, D.P. (1999) Recent King, B.S., Gupta, R. and Narayan, R.K. (2000) The early
trends in mortality and causes of death among persons with assessment and intensive care unit management of patients
229

with severe traumatic brain and spinal cord injuries. Surg. Oldenburg, O., Kribben, A., Baumgart, D., Philipp, T., Erbel,
Clin. North Am., 80(3): 855–870. R. and Cohen, M.V. (2002) Treatment of orthostatic hypo-
Kirshblum, S.C., House, J.G. and O’Connor, K.C. (2002) Silent tension. Curr. Opin. Pharmacol., 2(6): 740–747.
autonomic dysreflexia during a routine bowel program in Osborn, J.W., Taylor, R.F. and Schramm, L.P. (1989) Deter-
persons with traumatic spinal cord injury: a preliminary minants of arterial pressure after chronic spinal transection in
study. Arch. Phys. Med. Rehabil., 83: 1774–1776. rats. Am. J. Physiol., 256: R666–R673.
Krassioukov, A.V., Bunge, R.P., Puckett, W.R. and Bygrave, Osborn, J.W., Taylor, R.F. and Schramm, L.P. (1990) Chronic
M.A. (1999) The changes in human spinal cord sympathetic cervical spinal cord injury and autonomic hyperreflexia in
preganglionic neurons after spinal cord injury. Spinal Cord, rats. Am. J. Physiol., 258: R169–R174.
37: 6–13. Piepmeier, J.M., Lehmann, K.B. and Lane, J.G. (1985) Cardi-
Krassioukov, A.V., Furlan, J.C. and Fehlings, M.G. (2003) ovascular instability following acute cervical spinal cord
Autonomic dysreflexia in acute spinal cord injury: an under- trauma. Central Nervous System Trauma, 2(3): 153–160.
recognized clinical entity. J. Neurotrauma, 20(8): 707–716. Pine, Z.M., Miller, S.D. and Alonso, J.A. (1991) Atrial fibril-
Krassioukov, A.V., Johns, D.G. and Schramm, L.P. (2002) lation associated with autonomic dysreflexia. Am. J. Phys.
Sensitivity of sympathetically correlated spinal interneurons, Med. Rehabil., 70(5): 271–273.
renal sympathetic nerve activity, and arterial pressure to so- Sclater, A. and Alagiakrishnan, K. (2004) Orthostatic hypo-
matic and visceral stimuli after chronic spinal injury. J. Ne- tension — a primary care primer for assessment and treat-
urotrauma, 19(12): 1521–1529. ment. Geriatrics, 59(8): 22–27.
Krassioukov, A.V. and Weaver, L.C. (1995) Episodic hyper- Shannon, J.R., Diedrich, A., Biaggioni, I., Tank, J., Robertson,
tension due to autonomic dysreflexia in acute and chronic R.M., Robertson, D. and Jordan, J. (2002) Water drinking as
spinal cord-injured rats. Am. J. Physiol., 268: H2077–H2083. a treatment for orthostatic syndromes. Am. J. Med., 112(5):
Lehmann, K.G., Lane, J.G., Piepmeier, J.M. and Batsford, 355–360.
W.P. (1987) Cardiovascular abnormalities accompanying Silver, J.R. (2000) Early autonomic dysreflexia. Spinal Cord,
acute spinal cord injury in humans: incidence, time course 38: 229–233.
and severity. J. Am. Coll. Cardiol., 10(1): 46–52. Tanaka, H., Yamaguchi, H. and Tamai, H. (1997) Treatment of
Maiorov, D.N., Fehlings, M.G. and Krassioukov, A.V. (1998) orthostatic intolerance with inflatable abdominal band. Lan-
Relationship between severity of spinal cord injury and ab- cet, 349(9046): 175.
normalities in neurogenic cardiovascular control in conscious Teasell, R., Arnold, A.P., Krassioukov, A.V. and Delaney,
rats. J. Neurotrauma, 15(5): 365–374. G.A. (2000) Cardiovascular consequences of loss of supra-
Maiorov, D.N., Weaver, L.C. and Krassioukov, A.V. (1997) spinal control of the sympathetic nervous system following
Relationship between sympathetic activity and arterial pres- spinal cord injuries. Arch. Phys. Med. Rehabil., 81: 506–516.
sure in conscious spinal rats. Am. J. Physiol., 272: Ten Harkel, A.D., van Lieshout, J.J. and Wieling, W. (1994)
H625–H631. Effects of leg muscle pumping and tensing on orthostatic
Mathias, C.J. (1995) Orthostatic hypotension: causes, mecha- arterial pressure: a study in normal subjects and patients with
nisms, and influencing factors. Neurology, 45(Suppl 5): autonomic failure. Clin. Sci., 87(5): 553–558.
S6–S11. Vale, F.L., Burns, J., Jackson, A.B. and Hadley, M.N. (1997)
Mathias, C.J. and Frankel, H.L. (1992a) Autonomic distur- Combined medical and surgical treatment after acute spinal
bances in spinal cord lesions. In: Bannister R. and Mathias cord injury: results of a prospective pilot study to assess the
C.J. (Eds.) Autonomic Failure, a Textbook of Clinical Dis- merits of aggressive medical resuscitation and blood pressure
orders of the Autonomic Nervous System, Vol. 3(43). Oxford management. J. Neurosurg., 87(2): 239–246.
Medical Publications, Oxford, UK, pp. 839–881. Vaziri, N.D. (2003) Nitric oxide in microgravity — induced
Mathias, C.J. and Frankel, H.L. (1992b) The cardiovascular orthostatic intolerance: relevance to spinal cord injury.
system in tetraplegia and paraplegia. In: Frankel H.L. (Ed.) J. Spinal Cord Med., 26(1): 5–11.
Handbook of Clinical Neurology, Vol. 17(25). Elsevier Sci- Wecht, J.M., De Meersman, R.E., Weir, J.P., Spungen, A.M.
ence Publishers, B.V., Amsterdam, pp. 435–456. and Bauman, W.A. (2003) Cardiac autonomic responses to
Mukand, J., Karlin, L., Barrs, K. and Lublin, P. (2001) Mid- progressive head-up tilt in individuals with paraplegia. Clin.
odrine for the management of orthostatic hypotension in Auton. Res., 13(6): 433–438.
patients with spinal cord injury: a case report. Arch. Phys. Winslow, E.B., Lesch, M., Talano, J.V. and Meyer Jr., P.R.
Med. Rehabil., 82(5): 694–696. (1986) Spinal cord injuries associated with cardiopulmonary
Nacimiento, W. and Noth, J. (1999) What, if anything, is spinal complications. Spine, 11(8): 809–812.
shock? Arch. Neurol., 56(8): 1033–1035. Yarkony, G.M., Katz, R.T. and Wu, Y. (1986) Seizures sec-
Nieshoff, E.C., Birk, T.J., Birk, C.A., Hinderer, S.R. and ondary to autonomic dysreflexia. Arch. Phys. Med. Rehabil.,
Yavuzer, G. (2004) Double-blinded, placebo-controlled trial 67: 834–835.
of midodrine for exercise performance enhancement in Yeoh, M., McLachlan, E.M. and Brock, J.A. (2004) Tail arteries
Tetraplegia: a pilot study. J. Spinal Cord Med., 27(3): from chronically spinalized rats have potentiated responses to
219–225. nerve stimulation in vitro. J. Physiol., 556: 545–555.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 15

Orthostatic hypotension and paroxysmal


hypertension in humans with high spinal cord injury

Christopher J. Mathias1,2,

1
Neurovascular Medicine Unit, Faculty of Medicine, Imperial College London at St Mary’s Hospital, London W2 1NY, UK
2
Autonomic Unit, National Hospital for Neurology and Neurosurgery, Queen Square, and Institute of Neurology,
University College London, London, UK

Abstract: The spinal cord is essential for normal autonomic nervous system regulation of the cardiovas-
cular system as the preganglionic neurons controlling the heart and blood vessels originate in the tho-
racolumbar spinal segments. The site and extent of a spinal cord injury determine the degree of autonomic
involvement in cardiovascular dysfunction after the injury. After complete cervical cord lesions the entire
sympathetic outflow is separated from cerebral control; this may cause orthostatic hypotension. Commonly
after traumatic injuries to the spinal cord, one or more segments are totally destroyed. However, the distal
portion of the spinal cord often retains function and activation of spinal cord reflexes working independ-
ently of the brain can result in paroxysmal hypertension. This chapter will focus on orthostatic hypotension
and paroxysmal hypertension in cord-injured people with lesions affecting the cervical and upper thoracic
spinal cord. Conditions promoting these abnormalities in blood pressure will be elaborated. Possible
mechanisms for the hypo- and hypertension will be discussed, as will strategies for managing these prob-
lems.

Introduction are totally destroyed. However, the distal portion


of the spinal cord often retains function and ac-
The spinal cord is essential for normal functioning tivation of spinal cord reflexes working independ-
of the autonomic nervous system, as the entire ently of the brain can result in paroxysmal
sympathetic outflow (from the 1st thoracic (T1) to hypertension. This chapter will focus on ortho-
upper lumbar (L2/3)) and a proportion of the static hypotension and paroxysmal hypertension
parasympathetic outflow (the sacral parasympa- in cord-injured people with lesions affecting the
thetic) travel in the spinal cord before they reach cervical and upper thoracic spinal cord.
their target organs. In spinal cord injuries, there-
fore, there are varying degrees of autonomic in-
volvement, depending upon the site and extent of Basal blood pressure
the lesion. In complete tetraplegics with cervical
cord lesions the entire sympathetic outflow is sep- In recently injured tetraplegics the basal supine
arated from cerebral control; this may cause or- level of blood pressure usually is lower than nor-
thostatic hypotension. Commonly after traumatic mal (mean arterial pressure: 57 mmHg in tetra-
injuries to the spinal cord, one or more segments plegics and 82 mmHg in normal subjects; Mathias
et al., 1979a). It is dependent upon a number of
Corresponding author. Tel.: +0207 886 1468; factors, including complicating trauma and drug
Fax: +0207 996 1540; E-mail: c.mathias@imperial.ac.uk therapy. The lower levels of blood pressure appear

DOI: 10.1016/S0079-6123(05)52015-6 231


232

secondary to diminution in sympathetic nervous salt excretion as do normal subjects (Sutters et al.,
activity (Stjernberg et al., 1986). It is unlikely that 1992). In tetraplegics, recumbency may induce a
skeletal muscle paralysis alone is the explanation, diuresis but not a natriuresis; this differs from
as tetraplegics due to poliomyelitis often have subjects with primary autonomic failure, who also
normal or even higher levels of blood pressure. have nocturnal polyuria (Mathias et al., 1986), and
In recently injured tetraplegics the basal heart in whom recumbency causes both diuresis and
rate is usually below 100 beats/min, unlike low natriuresis (Kooner et al., 1987). The difference
spinal cord injuries in whom the heart rate often is may relate to the ability of tetraplegics to mount
higher. This probably is due to a reduction in an adequate hormonal response to oppose natriur-
neural and hormonal sympathetic chronotropic esis, unlike those in autonomic failure in whom
influences in high lesions. The efferent parasym- these responses often are muted. These observa-
pathetic cardiac pathways, however, are intact and tions have practical importance, as a period of re-
the absence of sympathetic activation may predis- cumbency in people with high spinal lesions will
pose susceptible patients to vagal over-activity. often result in accentuation of orthostatic hypo-
This may result in bradycardia and cardiac arrest, tension. This may be reduced or prevented by
as may occur during tracheal stimulation (Frankel head-up tilt.
et al., 1975; Mathias, 1976)
In the chronic stage, the supine blood pressure
in people with high lesions is lower than in normal Orthostatic hypotension
subjects (Frankel et al., 1972). Non-invasive am-
bulatory 24 h recordings confirm loss of nocturnal Subjects with high spinal cord lesions are prone to
circadian fall in blood pressure, which occurs in orthostatic (postural) hypotension when changing
normal subjects (Nitsche et al., 1996). Basal levels from the horizontal to upright position (Fig. 1).
of plasma noradrenaline and adrenaline in tetra- Orthostatic hypotension is defined as a decrease in
plegics are low, consistent with a diminished systolic blood pressure of more than 20 mmHg, or
peripheral sympathetic activity, caused by absence a fall in diastolic blood pressure of more than
of tonic supraspinal drive. This has been con- 10 mmHg, while upright or during head-up tilt to
firmed by measuring skin and muscle sympathetic 60o, for at least 3 min (Schatz et al., 1996). The fall
nerve activity using microneurography (Stjernberg in blood pressure is accompanied by a variety of
et al., 1986). Subjects with spinal cord injury, symptoms (Table 1); these can vary in nature and
however, are prone to renal damage and renal intensity and are not necessarily related to the
failure, as complications of lower urinary tract degree of hypotension.
dysfunction (see also Karlsson, this volume) may During head-up postural change there usually is
account for sustained hypertension in some. an immediate fall in blood pressure, often to ex-
In the absence of adequate resting sympathetic tremely low levels. There usually is no loss of con-
tone a number of secondary mechanisms, particu- sciousness, except in recently injured tetraplegics
larly hormonal, attempt to maintain blood pressure. or in chronic tetraplegics, following a period of
An important component is the renin–angioten- recumbency. This tolerance to a low cerebral per-
sin–aldosterone system, through the direct pressor fusion pressure is similar to that observed in pa-
effects of angiotensin-II and the salt-retaining ef- tients with primary autonomic failure, who have
fects of aldosterone. Drugs that interfere with the the ability to autoregulate their cerebral circula-
system, such as the angiotensin-converting enzyme tion at considerably lower perfusion pressure levels
inhibitor, captopril, substantially lower supine than normal subjects (Goadsby, 2002).
blood pressure in tetraplegics. Small doses of di- Following the initial fall in blood pressure, the
uretics, that cause salt loss and lower intravascular subsequent responses vary. In some subjects,
fluid volume, may cause a marked fall in supine blood pressure continues to fall as head-up tilt is
blood pressure. A low salt diet lowers blood pres- maintained. There is no rise in levels of plasma
sure, despite the ability of tetraplegics to reduce noradrenaline following head-up postural change,
233

Table 1. Some of the symptoms resulting from orthostatic hy-


potension and impaired perfusion of various organs

Cerebral hypoperfusion
Dizziness
Visual disturbances
Blurred
Tunnel
Scotoma
Greying out
Blacking out
Colour defects
Loss of consciousness
Cognitive deficits
Muscle hypoperfusion
Paracervical and suboccipital (‘coat-hanger’) ache
Subclavian steal-like syndrome
Renal hypoperfusion
Oliguria
Non-specific
Weakness, lethargy, fatigue

Adapted from Mathias (2003a). With permission.


Fig. 1. Top panels: blood pressure (BP) and heart rate (HR) in
a tetraplegic before and after head-up tilt, in the early stages of
rehabilitation, when there were few muscle spasms and minimal from the adrenal cortex. The salt and water
autonomic dysreflexia. Bottom panels: Blood pressure (BP) and retaining effects of aldosterone increase intra-
heart rate (HR) in a tetraplegic before, during, and after head- vascular volume. These various actions of the
up tilt to 45o. Blood pressure promptly falls but with partial renin–angiotensin–aldosterone system help raise
recovery, which in this case is linked to skeletal muscle spasms
blood pressure. During tilt there also may be
(S) inducing spinal sympathetic activity. Some of the later os-
cillations may be due to a rise in plasma renin, which was activation of spinal reflexes from stimulation
measured where there are interruptions in the blood pressure of skin, skeletal muscles or viscera, as part of
records. In the later phases of tilt, skeletal muscle spasms occur autonomic dysreflexia.
more frequently and further elevate the blood pressure. On re- During head-up postural change the fall in
turn to the horizontal, blood pressure rises rapidly above the
blood pressure is accompanied by a reduction in
previous level, and then slowly returns to a steady value. Heart
rate usually moves in the opposite direction to blood pressure, central venous pressure, stroke volume, and car-
except during muscle spasms, when there is an increase. From diac output, which is probably the result of venous
Mathias and Frankel (1988). With Permission. pooling, diminished venous return, and the inabil-
ity to increase sympathetic cardiac inotropic
which is consistent with an inability to increase activity. Venous pooling may cause cyanotic dis-
sympathetic nervous activity reflexly in response to coloration of the legs and may account for ankle
postural change, as is expected. If tilt is prolonged, oedema. Urine volume is usually reduced, often to
blood pressure tends to partly recover. This low levels and may raise the question of urinary
recovery may in part be related to activation of outflow tract obstruction. Oliguria is due to the
the renin–angiotensin–aldosterone system. Release fall in blood pressure, which reduces renal plasma
of renin appears independent of sympathetic flow and glomerular filtration rate. Increased levels
stimulation and is probably secondary to renal of the antidiuretic hormone, vasopressin, also
baroreceptor stimulation from the fall in renal per- probably contribute. In subjects with high spinal
fusion pressure (Mathias et al., 1975). Renin results lesions there is an exaggerated rise in vasopressin
in formation of the peptide angiotensin II, a pow- levels during head-up tilt when compared to nor-
erful direct-acting vasoconstrictor. Angiotensin II mal subjects.
also facilitates peripheral noradrenaline release and During head-up postural change there often is a
activity, and stimulates the release of aldosterone rapid rise in heart rate, which is inversely related to
234

the fall in blood pressure. This is due to with- Table 2. Some of the non-pharmacological approaches used in
drawal of vagal tone presumably due to unloading the management of orthostatic hypotension
of baroreceptor afferents; it is markedly attenuat- To be avoided
ed although not abolished by atropine. Prop- Sudden head-up postural change (especially on waking)
ranolol also reduces the heart rate rise during tilt, Prolonged recumbency
suggesting that beta-adrenoceptor stimulation par- Straining during micturition and defecation
High environmental temperature (including hot baths)
tially contributes. Despite marked orthostatic hy- Drugs with vasodepressor properties
potension, the heart rate does not usually rise
above 100 beats/min. This is different from pa- To be introduced
Head-up tilt during sleep
tients with an intact sympathetic nervous system, High salt intake
in ‘shock’ with a similarly low level of blood pres- Adopting different body positions
sure, in whom heart rate usually rises considerably
To be considered
above 100 beats/min. Elastic stockings
The clinical problems resulting from orthostatic Thigh cuffs
hypotension in people with high spinal lesions are Abdominal binders
not usually as severe and prolonged as the prob- Water ingestion
lems in subjects with primary autonomic failure.
Adapted from Mathias (2003b). With permission.
There usually is no loss of consciousness, except in
recently injured tetraplegics, in the early stages of
rehabilitation, or in chronic tetraplegics following blood flow at lower perfusion pressures probably
a period of recumbency. This tolerance to a low also contributes. Some of the newer treatments to
cerebral perfusion pressure is similar to that ob- improve motor function, such as functional elec-
served in patients with primary autonomic failure, trical stimulation, may reduce orthostatic hypo-
who have greater autoregulation of their cerebral tension (Sampson et al., 2000).
circulation as described above (Goadsby, 2002). Awareness of the many factors that lower blood
Subjects may be affected in various ways, as out- pressure is important. These include simple meas-
lined in Table 1. There may be transient dizziness ures such as avoiding rapid postural change, es-
when changing to the upright position, impaired pecially in the morning when getting out of bed.
concentration and attention (Critchley and Mathias, The supine blood pressure often is lowest in the
2003), or a ‘coat-hanger’ neck ache (Cariga et al., morning, especially in those in whom nocturnal
2002). In some, arm movement that increases polyuria reduces extracellular fluid volume. Pro-
upper limb blood flow may cause a subclavian longed bed rest and recumbency, especially post-
steal-like effect, and cause symptoms of cerebral operatively, should be avoided. Straining during
hypoperfusion by reducing blood flow to the brain micturition and bowel movement should be avoid-
stem. Tiredness and lethargy may be related to a ed. The blood pressure may fall to extremely low
low level of blood pressure, as has been noted in levels during straining, similar to that observed
primary autonomic failure (Mathias et al., 1999). during the Valsalva manoeuvre even when
Management consists largely of non-pharmaco- intrathoracic pressure is elevated only to 20 or
logical measures, based on knowledge of patho- 30 mmHg (van Lieshout et al., 1991). In hot
physiological processes (Table 2) Symptoms of weather, body temperature may rise as thermo-
orthostatic hypotension often are diminished with regulatory mechanisms such as sweating are im-
frequent postural change to the head-up position, paired; this may further increase vasodilatation
along with elevation of the head end of the bed at and worsen orthostatic hypotension. In chronic
night. This may activate the renin–angiotensin–al- autonomic failure, ingestion of alcohol or large
dosterone axis, causing vasoconstriction and plas- meals, especially those containing a high carbohy-
ma volume expansion, which helps buffer the fall drate content (Mathias et al., 1989), may cause
in blood pressure during head-up postural change. supine postprandial hypotension and aggravate
An improved ability to autoregulate cerebral orthostatic hypotension post-meal (Mathias et al.,
235

1991; Chaudhuri et al., 1994). In tetraplegics, this Table 3. Outline of the major actions by which a variety of
post-meal hypotension does not seem to occur to drugs may reduce orthostatic hypotension
the same extent; there is a modest fall in blood Reducing salt loss/plasma volume expansion
pressure accompanied by an elevation in heart rate Mineralocorticoids (fludrocortisone)
(Baliga et al., 1997). Levels of forearm venous Reducing nocturnal polyuria
plasma noradrenaline do not change, excluding a V2-receptor agonists (desmopressin)
Vasoconstriction — sympathetic
generalized increase in sympathetic nerve activity. On resistance vessels (ephedrine, midodrine, phenylephrine,
The mechanisms responsible for this difference in noradrenaline, clonidine, tyramine with monoamine oxidase
tetraplegics are unclear and could include activa- inhibitors, yohimbine, L-dihydroxyphenylserine)
tion of pressor reflexes from the gastrointestinal On capacitance vessels (dihydroergotamine)
tract (Mathias and Bannister, 2002). Arm exercise Vasoconstriction — non-sympathomimetic
V1 receptor agents — terlipressin
may induce hypotension in subjects with high le- Ganglionic nicotinic-receptor stimulation
sions (King et al., 1992; 1994). Cholinesterase inhibitors — pyridostigminc
In some cord-injured people, preventing venous Preventing vasodilatation
pooling when upright by using abdominal binders, Prostaglandin synthetase inhibitors (indomethacin,
thigh cuffs, and lower limb elastic stockings may flurbiprofen)
Dopamine receptor blockade (metoclopramide,
be beneficial. Each has its limitations. Recent ob- domperidone)
servations indicate that ingestion of 500 ml of wa- Beta-adrenoceptor blockade (propranolol)
ter raises supine blood pressure substantially and Preventing postprandial hypotension
improves orthostatic hypotension in primary au- Adenosine receptor blockade (caffeine)
tonomic failure (Cariga et al., 2001; Mathias and Peptide release inhibitors (somatostatin analogue: octreotide)
Increasing cardiac output
Young, 2004); it also appears to raise blood pres- Beta-blockers with intrinsic sympathomimetic activity
sure in people with high lesions (Tank et al., 2003). (pindolol, xamoterol)
Activation of spinal sympathetic reflexes, by in- Dopamine agonists (ibopamine)
duction of muscle spasms or tapping of the ante- Increasing red cell mass
rior abdominal wall suprapubically to contract the Erythropoietin
urinary bladder, may cause autonomic dysreflexia Adapted from Mathias (2003b). With permission.
and thus elevate blood pressure.
A range of drugs is used to reduce orthostatic
hypotension in primary autonomic failure (Table 3); Paroxysmal hypertension
some of these drugs have been used with success
in spinal injuries. However, subjects with high Subjects with high spinal lesions may have an ex-
lesions, unlike those with autonomic failure, are aggerated rise in blood pressure in response to
prone to paroxysms of hypertension, the severity of stimuli that originate below the level of the lesion
which may be exacerbated by such drugs. Drugs (Fig. 2). These may arise from skin, abdominal and
with short half-lives are preferable and ideally pelvic viscera, or by contraction of skeletal mus-
should be used only for limited periods, such as in cles. There usually is a fall in heart rate because of
the early stages of rehabilitation. Ephedrine in a increased vagal activity. In recently injured tetra-
dose of 15 mg, half an hour before postural change plegics there usually is no change in blood pressure
often is of value. The alpha-adrenoceptor agonist or heart rate during such stimulation (Fig. 3), in
midodrine may have a role (Barber et al., 2000; contrast to the chronic stage when spinal cord re-
Mukand et al., 2001). In the majority of patients, flexes have recovered. The cardiovascular effects
however, drugs are not needed. Although ortho- are part of the syndrome of autonomic dysreflexia.
static hypotension in people with high lesions is Other features include sweating in skin around and
primarily due to impaired sympathetic activation, in above the level of the lesion, contraction of the
clinical practice it is important to be aware that a urinary bladder and rectum, penile erection, seminal
variety of non-neurogenic factors (Table 4) may fluid emission and skeletal muscle spasms — com-
contribute. ponents of the ‘mass reflex’ described by Head and
236

Table 4. Examples of non-neurogenic causes of orthostatic hypotension

Low intravascular volume


Blood/plasma loss Haemorrhage, burns
Fluid/electrolyte deficiency
Diminished intake Vomiting
Loss from gut Diarrhea
Loss from kidney Salt losing nephropathy, diuretics
Endocrine deficiency Adrenal insufficiency (Addison’s disease)
Cardiac insufficiency
Myocardial Myocarditis
Impaired ventricular filling Atrial myxoma, constrictive pericarditis
Impaired output Aortic stenosis
Vasodilatation
Endogenous Hyperpyrexia
Exogenous Drugs such as glyceryl trinitrate (GTN)
Alcohol
Excessive heat

Fig. 2. Blood pressure (BP), heart rate (HR), intravesical pressure (IVP), plasma noradrenaline (NA) and adrenaline (A) levels in a
tetraplegic subject before, during and after bladder stimulation induced by suprapubic percussion of the anterior abdominal wall. The
rise in BP is accompanied by a fall in heart rate as a result of increased vagal activity in response to the rise in blood pressure. There is a
rise in levels of plasma NA (open histograms), but not A (filled histograms) suggesting an increase in sympathetic neural activity
independently of adrenomedullary activation. From Mathias and Frankel (1986). With permission.

Riddoch in 1917 (Table 5). The rise in blood pres- due to activation of spinal cardiac reflexes when the
sure, first reported by Guttmann and Whitteridge lesion is rostral to the cardiac sympathetic
in 1947, is secondary to vasoconstriction in resist- preganglionic neurons (Corbett et al., 1975). The
ance vessels and capacitance vessels (Corbett et al., rapid rise in blood pressure after stimulation is
1971). Stroke volume and cardiac output increase suggestive of reflex sympathetic activity, through
237

Fig. 3. Average levels of mean blood pressure (MBP), heart rate (HR), plasma noradrenaline (NA, continuous line) and adrenaline
(A, interrupted line) in recently injured and chronic tetraplegics before, during and after bladder stimulation (BS). The bars indicate
7SEM. No changes occur in the recently injured tetraplegics, unlike the chronic tetraplegics in whom MBP and plasma NA levels rise
and HR falls. There are no changes in plasma A levels. From Mathias et al. (1979a). With permission.

Table 5. Clinical manifestations of autonomic dysreflexia the isolated spinal cord. This interpretation is sup-
ported by the fact that plasma noradrenaline, but
Paraesthesiae in neck, shoulders and arms not adrenaline, levels increase during dysreflexia
Fullness in head
Hot ears
(Mathias et al., 1976a). Adrenomedullary secretion
Throbbing headache, especially in the occipital and frontal does not contribute to this elevation in blood
regions pressure. This differs markedly from the extremely
Tightness in chest and dyspnea high levels of plasma catecholamines often found
Hypertension and bradycardia in pheochromocytoma where there also is blood
Occasionally cardiac dysrhythmias
Pupillary dilatation
pressure lability. During autonomic dysreflexia, cir-
Pallor initially, followed by flushing of face and neck and culating levels of other vasoconstrictor substances,
sweating in areas above and around the lesion (above lesion) such as renin (and by inference angiotensin-II lev-
Cold peripheries; piloerection (below lesion) els), aldosterone, vasopressin, and atrial natriuretic
Contraction of urinary bladder and large bowelaa –4 peptide remain unchanged or fall (Mathias et al.,
Penile erection and seminal fluid emissiona
1981; Krum et al., 1992). Whether levels of other
Adapted from Mathias and Frankel (2002). vasoconstrictor peptides such as neuropeptide Y
a
May occur as part of the ‘mass’ reflex. (NPY) and endothelin rise in man is not known.
238

The rise in blood pressure and the widespread are likely to include peripheral neural and vascular
involvement of the vasculature below the lesion, changes, as well as central nervous system plastic-
despite a modest and often localized stimulus only ity (Weaver, 2002), and need further investigation,
involving a few segments, suggest the spread of in the light of clinical and recent experimental
neuronal impulses intraspinally and/or extraspin- data (see Weaver et al.; McLachlan and Brock;
ally. In tetraplegics, microneurography indicates Rabchevsky; Schramm, this volume).
only a moderate and transient rise in muscle sym- The exaggerated pressor responses to stimuli
pathetic nerve activity during autonomic dysre- causing autonomic dysreflexia do not occur in le-
flexia (Stjernberg et al., 1986) with no correlation sions below the fifth thoracic segment (Fig. 4). This
between the cardiac cycle and muscle sympathetic indicates that the sympathetic neural outflow below
nerve discharge as occurs normally. Hyperactivity T5, which incorporates neural control of the large
of target organs innervated by the autonomic splanchnic circulatory bed, is of major importance
nervous system has been demonstrated by re- in generating the blood pressure response during
sponses of the dorsal foot vein of tetraplegics to dysreflexia and also in maintaining blood pressure
local intravenous noradrenaline (Arnold et al., homeostasis. Mild episodes of autonomic dysreflex-
1995). The exaggerated blood pressure response to ia probably occur intermittently through the day
various stimuli, including to noradrenaline infused in response to various stimuli, but often are not
intravenously (Mathias et al., 1976b), suggests su- noticed and may be of little consequence. Severe
persensitivity of adrenoceptors, or that other autonomic dysreflexia is of major clinical impor-
mechanisms are responsible for the enhanced vas- tance. Hypertension, especially linked to bladder
cular response (see also McLachlan and Brock, contractions and voiding, may not be accompa-
this volume). Overall, the results of physiological nied by symptoms (silent autonomic dysreflexia;
and pharmacological studies, in conjunction with Linsenmeyer et al., 1996). When autonomic dysre-
the neurohormonal observations, indicate that au- flexia is prolonged there may be considerable
tonomic dysreflexia is a more appropriate term
than autonomic hyperreflexia. However, data ob-
tained from indirect techniques, such as total and
regional body noradrenaline spillover, indicate a
marked (332% total and 15-fold in the leg) in-
crease during bladder stimulation, and suggest
that in people with high lesions, greater quantities
of noradrenaline may be released per impulse dur-
ing dysreflexia (Karlsson et al., 1998; Gao et al.,
2002). This suggestion is based on the assumption
that the sparse firing of muscle sympathetic
fibres recorded by Stjernberg et al. (1986) during
dysreflexia can be generalized to reflect the entire
sympathetic outflow below the lesion. The spill-
over data of Gao et al. (2002) were interpreted as
possible evidence for a significant increase in sym-
pathetic nerve firing during dysreflexia, as has been
documented in the rat (Maiorov et al., 1997). Fig. 4. Changes in mean blood pressure (DMBP) and heart rate
However, these findings could indicate greater (DHR) in subjects with spinal cord lesions at different levels
numbers of axons firing, higher firing rates, greater (cervical and thoracic) after bladder stimulation induced by
suprapubic percussion of the anterior abdominal wall. In the
release of transmitter per impulse, or reduced
cervical and high thoracic lesions there is a marked elevation in
noradrenaline reuptake by presynaptic terminals. blood pressure and a fall in heart rate. In lesions below T5 there
The mechanisms accounting for the substantial are minimal cardiovascular changes. From Mathias and Frankel
rise in blood pressure during autonomic dysreflexia (1986). With permission.
239

morbidity, as a result of excessive sweating over Table 6. Causes of autonomic dysreflexia


the head and neck, and a throbbing headache. The Abdominal or pelvic visceral stimulation
latter is often, but not always, related to the level Ureter
of blood pressure and may be dependent on dis- Calculus
tension of pain-sensitive cranial blood vessels. Urinary bladder
With recurrent episodes of dysreflexia, headache Distension by blocked catheter or discoordinated bladder
Infection
may become a particularly severe symptom despite Irritation by calculus, catheter or bladder washout
modest elevations in blood pressure. Other com- Rectum and anus
plications of vasospasm and hypertension during Enemas
autonomic dysreflexia include myocardial failure Fecal retention
and neurological complications such as epileptic Anal fissure
Gastrointestinal organs
seizures, visual defects, and cerebral hemorrhage. Gastric dilatation
These may result in extensive and permanent Gastric ulceration
neurological deficits, or even death. Cholecystitis or cholelithiasis
The key factor in the management of autonomic Uterus
dysreflexia is to determine the provoking cause Contraction during pregnancy
Menstruation, occasionally
(Table 6), and rectify it. To lower blood pressure
rapidly, head-up tilt (which causes venous pooling) Cutaneous stimulation
initially may be used. A range of drugs, that act Pressure sores
Infected ingrowing toenails
through the proposed mechanisms responsible for Burns
autonomic dysreflexia, have been tried (Table 7).
Preventing afferent stimulation, for instance by the Skeletal muscle spasms
Especially in limbs with contractures
use of a local anaesthetic, such as lignocaine in the
urinary bladder, can be effective. Drugs that act Miscellaneous
partially (reserpine) or entirely (spinal anaesthet- Intrathecal neogstimine
Electroejaculatory procedures
ics) on the spinal cord are of particular use in se- Ejaculation
vere episodes of dysreflexia. Some may be given Vaginal dilatation
intrathecally (Middleton et al., 1996). The gangl- Urethra — insertion of catheter or abscess
ionic blocker, hexamethonium, was successfully Fracture of bones
used in the past, but, like other drugs that reduce
Adapted from Mathias and Frankel (2002). With permission.
sympathetic efferent activity, may cause profound
hypotension. The a2-adrenoceptor agonist, cloni-
dine, does not lower supine blood pressure in

Table 7. Some of the drugs used in the management of autonomic dysreflexia classified according
to their major site of action on the reflex arc and target organs

Afferent Topical lignocaine


Spinal cord Clonidinea
Reserpinea
Spinal anaesthetics
Efferent Sympathetic ganglia Hexamethonium
Sympathetic nerve terminals Guanethidine
a-adrenoceptors Phenoxybenzamine
Target Organs Blood vessels Glyceryl trinitrate
Nifedipine
Sweat glands Pro-Banthinea

Adapted from Mathias and Frankel (2002). With permission.


a
These drugs have multiple effects, some of which are peripheral
240

Fig. 5. Ambulatory blood pressure measurements demonstrating (A), a normal profile with a preserved physiological fall in blood
pressure at night between midnight and 04.00 h, (B) multiple episodes of autonomic dysreflexia in a complete tetraplegic and (C) the
same subject as in (B) after effective treatment of autonomic dysreflexia with the antihypertensive drug nifedipine. Despite improve-
ments in (C), the loss of the circadian regulation of blood pressure persists. In each panel, the top half shows systolic, mean, and
diastolic pressure (in mmHg, from top to bottom) and the bottom half shows heart rate in beats/min (bpm). From Curt et al. (1997).
241

Fig. 6. Blood pressure (BP) and heart rate (HR) in a tetraplegic in the supine position before, during, and after bladder stimulation
(BS) by suprapubic percussion of the anterior abdominal wall, a procedure that induces hypertension. Sublingual glyceryl trinitrate
(GTN) (0.5 mg for 3.5 min) rapidly reverses the hypertension, elevates the heart rate and then causes substantial hypotension. Levels of
plasma renin activity (PRA) rise as a result of the fall in blood pressure. From Mathias and Frankel (1988).

people with high lesions (Reid et al., 1977), but successfully used during surgery. During labour,
reduces hypertension during autonomic dysreflexia severe hypertension may occur and should be
(Mathias et al., 1979b). Alpha-adrenoceptor avoided, ideally by using a spinal anaesthetic.
blockers, such as phenoxybenzamine, and the se- Despite recognizing the cause, it may be ex-
lective blockers prazosin and terazosin are useful tremely difficult to resolve problems that include
in autonomic dysreflexia due to bladder outflow severe skeletal muscle spasms or recurrent urinary
obstruction, as they relax the smooth muscle of the bladder infection. Long-term drug therapy for au-
urinary sphincter (Vaidyanathan et al., 1998). tonomic dysreflexia in such subjects is often only
Drugs acting directly on blood vessels, such as partially successful and may result in undesirable
glyceryl trinitrate and calcium channel blockers, side effects. In severe cases, surgical procedures on
such as nifedipine are effective (Thyberg et al., 1994; the spinal cord, such as rhizotomy and cordotomy,
Curt et al., 1997) (Fig. 5), and have the advantage of or peripheral procedures, such as sacral and hypo-
being given sublingually. They also have the poten- gastric neurotomy, may need to be considered.
tial to lower blood pressure substantially (Fig. 6). Non-surgical approaches, such as subarachnoid
In some subjects, autonomic dysreflexia may be a block with alcohol or phenol, have also been uti-
major and recurring problem because of difficulty in lised. These procedures, however, usually abolish
either defining or resolving the precipitating cause. spinal reflex activity and result in flaccidity of
Unusual examples of the former are gastric ulcer- skeletal muscles, and bladder and bowel atony,
ation or cholecystitis, which are difficult to detect with their attendant disadvantages.
because of lack of pain. A common example, which
may be overlooked, is an anal fissure. Paroxysmal
hypertension can be a particular problem during
surgery, especially if either the urinary bladder or References
the large bowel is involved. In these patients hyper-
tension is often controlled successfully by deepening Arnold, J.M., Feng, Q.P., Delaney, G.A. and Teasell, R.W.
the level of spinal or general inhalation anesthesia, (1995) Autonomic dysreflexia in tetraplegic patients: evidence
for alpha-adrenoceptor hyper-responsiveness. Clin. Auton.
along with an increase in positive pressure ventila- Res., 5: 267–270.
tion (Welply et al., 1975). Short-acting ganglionic Baliga, R.R., Catz, A.B., Watson, L.P., Short, D.J., Frankel,
blockers or direct-acting vasodilators have also been H.L. and Mathias, C.J. (1997) Cardiovascular and hormonal
242

responses to food ingestion in humans with spinal cord tran- King, M.L., Freeman, D.M., Pellicone, J.T., Wanstall, E.R.
section. Clin. Auton. Res., 7: 137–141. and Bhansali, L.D. (1992) Exertional hypotension in thoracic
Barber, D.B., Rogers, S.J., Fredrickson, M.D. and Able, A.C. spinal cord injury: case report. Paraplegia, 30: 261–266.
(2000) Midodrine hydrochloride and the treatment of ortho- King, M.L., Lichtman, S.W., Pellicone, J.T., Close, R.J. and
static hypotension in tetraplegia: two cases and a review of Lisanti, P. (1994) Exertional hypotension in spinal cord in-
the literature. Spinal Cord, 38: 109–111. jury. Chest, 106: 1166–1171.
Cariga, P., Ahmed, S., Mathias, C.J. and Gardner, B.P. (2002) Kooner, J.S., da Costa, D.F., Frankel, H.L., Bannister, R.,
The prevalence and association of neck (coat-hanger) pain Peart, W.S. and Mathias, C.J. (1987) Recumbency induces
and orthostatic (postural) hypotension in human spinal cord hypertension, diuresis and natriuresis in autonomic failure,
injury. Spinal Cord, 40: 77–82. but diuresis alone in tetraplegia. J. Hypertens., 5: 327–329.
Cariga, P. and Mathias, C.J. (2001) Haemodynamics of the Krum, H., Louis, W.J., Brown, D.J., Clarke, S.J., Fleming, J.A.
pressor effect of oral water in human sympathetic denerva- and Howes, L.G. (1992) Cardiovascular and vasoactive hor-
tion due to autonomic failure. Clin. Sci., 101: 313–319. mone responses to bladder distension in spinal and normal
Chaudhuri, K.R., Maule, S., Thomaides, T., Pavitt, D. and man. Paraplegia, 30: 348–354.
Mathias, C.J. (1994) Alcohol ingestion lowers supine blood Linsenmeyer, T.A., Campagnolo, D.I. and Chou, I.H. (1996)
pressure, causes splanchnic vasodilatation and worsens post- Silent autonomic dysreflexia during voiding in men with spi-
ural hypotension in primary autonomic failure. J. Neurol., nal cord injuries. J. Urol., 155: 519–522.
241: 145–152. Maiorov, D.N., Weaver, L.C. and Krassioukov, A.V. (1997)
Corbett, J.L., Debarge, O., Frankel, H.L. and Mathias, C.J. Relationship between sympathetic activity and arterial pres-
(1975) Cardiovascular responses in tetraplegic man to muscle sure in conscious spinal rats. Am. J. Physiol., 272: H625–H631.
spasm, bladder percussion and head-up tilt. Clin. Exp. Mathias, C.J. (1976) Bradycardia and cardiac arrest during
Pharmacol. Physiol., 2(Suppl): 189–193. tracheal suction — mechanisms in tetraplegic patients. Eur. J.
Corbett, J.L., Frankel, H.L. and Harris, P.J. (1971) Cardio- Intens. Care Med., 2: 147–156.
vascular reflex responses to cutaneous and visceral stimuli in Mathias, C.J. (2003a) Autonomic diseases — clinical features
spinal man. J. Physiol., 215(2): 395–409. and laboratory evaluation. J. Neurol. Neurosurg. Psychiat.,
Critchley, H.D. and Mathias, C.J. (2003) Blood pressure, at- 74: 31–41.
tention and cognition: drivers and air traffic controllers. Clin. Mathias, C.J. (2003b) Autonomic diseases — management.
Auton. Res., 13: 399–401. J. Neurol. Neurosurg. Psychiat., 74: 42–47.
Curt, A., Nitsche, B., Rodic, B., Schurch, B. and Dietz, V. Mathias, C.J. and Bannister, R. (2002) Postprandial hypoten-
(1997) Assessment of autonomic dysreflexia in patients with sion in autonomic disorders. In: Mathias C.J. and Bannister
spinal cord injury. J. Neurol. Neurosurg. Psychiat., 62: R. (Eds.), Autonomic Failure: A Textbook of Clinical Dis-
473–477. orders of the Autonomic Nervous System (4th ed.). Oxford
Frankel, H.L., Mathias, C.J. and Spalding, J.M.K. (1975) University Press, Oxford, pp. 283–295.
Mechanisms of reflex cardiac arrest in tetraplegic patients. Mathias, C.J., Christensen, N.J., Corbett, J.L., Frankel, H.L.,
Lancet, ii: 1183–1185. Goodwin, T.J. and Peart, W.S. (1975) Plasma catechola-
Frankel, H.L., Michaelis, L.S., Golding, D.R. and Beral, V. mines, plasma renin activity and plasma aldosterone in tet-
(1972) The blood pressure in paraplegia-1. Paraplegia, 10: raplegic man, horizontal and tilted. Clin. Sci. Mol. Med., 49:
193–198. 291–299.
Gao, S.A., Ambring, A., Lambert, G. and Karlsson, A.-K. Mathias, C.J., Christensen, N.J., Corbett, J.L., Frankel, H.L.
(2002) Autonomic control of the heart and renal vascular bed and Spalding, J.M.K. (1976a) Plasma catecholamines during
during autonomic dysreflexia in high spinal cord injury. Clin. paroxysmal neurogenic hypertension in quadriplegic man.
Auton. Res., 12: 457–464. Circ. Res., 39: 204–208.
Goadsby, P.J. (2002) Autoregulation and autonomic control of Mathias, C.J., Christensen, N.J., Frankel, H.L. and Spalding,
the cerebral circulation: implications and pathophysiology. J.M.K. (1979a) Cardiovascular control in recently injured
In: Mathias C.J. and Bannister R. (Eds.), Autonomic failure. tetraplegics in spinal shock. Q. J. Med. NS, 48: 273–287.
A Textbook of Clinical Disorders of the Autonomic Nervous Mathias, C.J., da Costa, D.F., Fosbraey, P., Bannister, R.,
System (4th ed). Oxford University Press, Oxford, pp. 85–91. Wood, S.M., Bloom, S.R. and Christensen, N.J. (1989) Car-
Guttmann, L. and Whitteridge, D. (1947) Effects of bladder diovascular, biochemical and hormonal changes during food
distension on autonomic mechanisms after spinal cord injury. induced hypotension in chronic autonomic failure. J. Neurol.
Brain, 70: 361–404. Sci., 94: 255–269.
Head, H. and Riddoch, G. (1917) The autonomic bladder, ex- Mathias, C.J., Fosbraey, P., da Costa, D.F., Thornley, A. and
cessive sweating and some other reflex conditions in gross Bannister, R. (1986) The effect of desmopressin on nocturnal
injuries of the spinal cord. Brain, 40: 188–263. polyuria, overnight weight loss and morning postural hypo-
Karlsson, A.-K., Friberg, P., Lonnroth, P., Sullivan, L. and tension in patients with autonomic failure. Br. Med. J., 293:
Elam, M. (1998) Regional sympathetic function in high 353–354.
spinal cord injury during mental stress and autonomic Mathias, C.J. and Frankel, H. (2002) Autonomic disturbances
dysreflexia. Brain, 121: 1711–1719. in spinal cord lesions. In: Mathias C.J. and Bannister R.
243

(Eds.), Autonomic Failure: A Textbook of Clinical Disorders Reid, J.L., Wing, L.M.H., Mathias, C.J., Frankel, H.L. and
of the Autonomic Nervous System (4th Ed.). Oxford Uni- Neill, E. (1977) The central hypotensive effect of clonidine:
versity Press, Oxford, pp. 494–513. studies in tetraplegic subjects. Clin. Pharmacol. Ther., 21:
Mathias, C.J. and Frankel, H.L. (1986) The neurological and 375–381.
hormonal control of blood vessels and heart in spinal man. Sampson, E.E., Burnham, R.S. and Andrews, B.J. (2000) Func-
J. Auton. Nerv. System.(Suppl): 457–464. tional electrical stimulation effect on orthostatic hypotension
Mathias, C.J. and Frankel, H.L. (1988) Cardiovascular control after spinal cord injury. Arch. Phys. Med. Rehabil., 81:
in spinal man. Ann. Rev. Physiol., 50: 577–592. 139–143.
Mathias, C.J., Frankel, H.L., Christensen, N.J. and Spalding, Schatz, I.J., Bannister, R., Freeman, R.L., Jankovic, J., Koller,
J.M.K. (1976b) Enhanced pressor response to noradrenaline W.C., Low, P.A., Mathias, C.J., Polinsky, R.J., Quinn, N.P.,
in patients with cervical spinal cord transaction. Brain, 99: Robertson, D. and Streeten, D.H.P. (1996) Consensus state-
757–770. ment on the definition of orthostatic hypotension, pure au-
Mathias, C.J., Frankel, H.L., Davies, I.B., James, V.H.T. and tonomic failure and multiple system atrophy. Clin. Auton.
Peart, W.S. (1981) Renin and aldosterone release during sym- Res., 6: 125–126.
pathetic stimulation in tetraplegia. Clin. Sci., 60: 399–604. Stjernberg, L., Blumberg, H. and Wallin, B.G. (1986) Sympa-
Mathias, C.J., Holly, E., Armstrong, E., Shareef, M. and Ban- thetic activity in man after spinal cord injury: outflow to
nister, R. (1991) The influence of food on postural hypoten- muscle below the lesion. Brain, 109: 695–715.
sion in three groups with chronic autonomic failure: clinical Sutters, M., Wakefield, C., O’Neil, K., Appleyard, M., Frankel,
and therapeutic implications. J. Neurol. Neurosurg. Psych- H.L., Mathias, C.J. and Peart, W.S. (1992) The cardiovas-
iat., 54: 726–730. cular, endocrine and renal response to tetraplegic and par-
Mathias, C.J., Mallipeddi, R. and Bleasdale-Barr, K. (1999) aplegic subjects to dietary sodium restriction. J. Physiol.
Symptoms associated with orthostatic hypotension in pure (London), 457: 515–523.
autonomic failure and multiple system atrophy. J. Neurol., Tank, J., Schroeder, C., Stoffels, M., Diedrich, A., Sharma,
246: 893–898. A.M., Luft, F.C. and Jordan, J. (2003) Pressor effect of water
Mathias, C.J., Reid, J.L., Wing, L.M.H., Frankel, H.L. and drinking in tetraplegic patients may be a spinal reflex. Hy-
Christensen, N.J. (1979b) Antihypertensive effects of cloni- pertension, 41: 1234–1239.
dine in tetraplegic subjects devoid of central sympathetic Thyberg, M., Ertzgaard, P., Gylling, M. and Granerus, G.
control. Clin. Sci., 57: 425s–428s. (1994) Effect of nifedipine on cystometry-induced elevation
Mathias, C.J. and Young, T.M. (2004) Water drinking in the of blood pressure in patients with a reflex urinary bladder
management of orthostatic intolerance due to orthostatic after a high level spinal cord injury. Paraplegia, 32: 308–313.
hypotension, vasovagal syncope and the postural tachycardia Vaidyanathan, S., Soni, B.M., Sett, P., Watt, J.W., Oo, T. and
syndrome. Eur. J. Neurol., 11: 613–619. Bingley, J. (1998) Pathophysiology of autonomic dysreflexia:
Middleton, J.W., Siddall, P.J., Walker, S., Molloy, A.R. and long-term treatment with terazosin in adult and paediatric
Rutkowski, S.B. (1996) Intrathecal clonidine and baclofen in spinal cord injury patients manifesting recurrent dysreflexic
the management of spasticity and neuropathic pain following episodes. Spinal Cord, 36: 761–770.
spinal cord injury: a case study. Arch. Phys. Med. Rehabil., van Lieshout, J.J., lmholz, B.P.M., Wesseling, K.H., Speelman,
77: 824–826. J.D. and Wieling, W. (1991) Singing-induced hypotension: a
Mukand, J., Karlin, L., Barrs, K. and Lublin, P. (2001) Mid- complication of high spinal cord lesion. Neth. J. Med., 38:
odrine for the management of orthostatic hypotension in 75–79.
patients with spinal cord injury: A case report. Arch. Phys. Weaver, L.C. (2002) What causes autonomic dysreflexia after
Med. Rehabil., 82: 694–696. spinal cord injury? Clin. Auton. Res., 12: 424–426.
Nitsche, B., Perschak, H., Curt, A. and Dietz, V. (1996) Loss of Welply, N., Mathias, C.J. and Frankel, H.L. (1975) Circulatory
circadian blood pressure variability in complete tetraplegia. reflexes in tetraplegics during artificial ventilation and during
J. Hum. Hypertens., 10: 311–317. general anaesthesia. Paraplegia, 13: 172–182.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 16

Autonomic dysreflexia after spinal cord injury:


central mechanisms and strategies for prevention

Lynne C. Weaver1,, Daniel R. Marsh2, Denis Gris1, Arthur Brown1 and Gregory
A. Dekaban1

1
Spinal Cord Injury Team, BioTherapeutics Research Group, Robarts Research Institute, 100 Perth Drive, P.O. Box 5015,
London, ON N6A 5K8, Canada
2
Department of Anatomy and Cell Biology, Dalhousie University, Halifax, NS, Canada

Abstract: Spinal reflexes dominate cardiovascular control after spinal cord injury (SCI). These reflexes are
no longer restrained by descending control and they can be impacted by degenerative and plastic changes
within the injured cord. Autonomic dysreflexia is a condition of episodic hypertension that stems from
spinal reflexes initiated by sensory input entering the spinal cord caudal to the site of injury. This hy-
pertension greatly detracts from the quality of life for people with cord injury and can be life-threatening.
Changes in the spinal cord contribute substantially to the development of this condition. Rodent models
are ideal for investigating these changes. Within the spinal cord, injury-induced plasticity leads to nerve
growth factor (NGF)-dependent enlargement of the central arbor of a sub-population of sensory neurons.
This enlarged arbor can provide increased afferent input to the spinal reflex, intensifying autonomic
dysreflexia. Treatments such as antibodies against NGF can limit this afferent sprouting, and diminish the
magnitude of dysreflexia. To assess treatments, a compression model of SCI that leads to progressive
secondary damage, and also to some white matter sparing, is very useful. The types of spinal reflexes that
likely mediate autonomic dysreflexia are highly susceptible to inhibitory influences of bulbospinal pathways
traversing the white matter. Compression models of cord injury reveal that treatments that spare white
matter axons also markedly reduce autonomic dysreflexia. One such treatment is an antibody to the
integrin CD11d expressed by inflammatory leukocytes that enter the cord acutely after injury and cause
significant secondary damage. This antibody blocks integrin-mediated leukocyte entry, resulting in greatly
reduced white-matter damage and decreased autonomic dysreflexia after cord injury. Understanding the
mechanisms for autonomic dysreflexia will provide us with strategies for treatments that, if given early after
cord injury, can prevent this serious disorder from developing.

Introduction to autonomic dysreflexia entirely devoted to autonomic dysfunction after


cord injury, contains many chapters written by
In 2001, my co-authors and I published a chapter investigators quoted in that 2001 review. As the
reviewing the central mechanisms for autonomic work of others is presented elsewhere in this book,
dysreflexia in a volume of Progress in Brain Re- this chapter will focus on the 10 years of investi-
search that focused on spinal cord injury (SCI) gations of autonomic dysreflexia from our labo-
(Weaver et al., 2001a). The present volume, ratory that began in 1995.
After SCI, large increases in arterial pressure
Corresponding author. Tel.: +519 663 3776; (AP) can develop in response to sensory input en-
Fax: +519 663 3789; E-mail: lcweaver@robarts.ca tering the spinal cord below the level of the lesion

DOI: 10.1016/S0079-6123(05)52016-8 245


246

(Mathias and Frankel, 1992a, b; Lee et al., 1994; This chapter will review studies in rats that
Krassioukov and Weaver, 1995; Maiorov et al., demonstrated some of the changes within the
1998). This hypertension, part of a condition spinal cord that can contribute to the development
termed autonomic dysreflexia, occurs in 50–90% of autonomic dysreflexia, and experiments that
of people with tetraplegia or high paraplegia show the potential for treatments to prevent the
(Corbett et al., 1975; Erickson, 1980; Lindan et development of dysreflexia.
al., 1980; Mathias and Frankel, 1992a; Lee et al.,
1994; Giannantoni et al., 1998; Vaidyanathan
et al., 1998; Karlsson, 1999; Krassioukov et al., Rodent models of autonomic dysreflexia
2003). The hypertension can result in debilitating
headaches, seizures, strokes and even death. As Rats and mice readily develop autonomic dysre-
blood pressure control normally depends upon flexia after SCI (Osborn et al., 1990; Maiorov
supraspinal regulation of sympathetic neurons et al., 1997a, b, 1998, 2001; Landrum et al., 1998;
(Calaresu and Yardley, 1988), SCI leads to a con- Jacob et al., 2001, 2003). In response to distension
dition in which central nervous system (CNS) of the urinary bladder or colon, exaggerated in-
regulation of AP is dominated by excitatory spinal creases in AP are apparent within a day of a cord
reflexes. This excitation is prominent, in part, transection at the upper thoracic (T) segments
because cord injury blocks descending bulbospinal (T2–4) and these responses augment with time af-
inhibition of spinal sympathetic reflexes (Sato and ter injury. This progression after cord transection
Schmidt, 1973; Dembowsky et al., 1978, 1980); the in rats was followed for up to 5 weeks in our
unchecked activity of these reflexes leads to laboratory. A conscious, unrestrained rat with an
autonomic dysreflexia. intact CNS has a transient, 5–15 mmHg increase in
Dysreflexia occurs after injury at or rostral to mean arterial pressure (MAP) upon colon disten-
the 6th thoracic spinal segment, because injury at sion, accompanied by an increase in heart rate
these levels leaves the sympathetic control of the (HR) (Krassioukov and Weaver, 1995; Maiorov
extensive abdominal circulation amenable to un- et al., 1997b). In this chapter, changes in blood
restrained spinal reflexes (Mathias and Frankel, pressure will be expressed as changes in MAP,
1992a). These reflexes can be caused by innocuous unless otherwise indicated. The first day after
or noxious stimulation of the skin, pressure sores, cord transection, the increase in AP, in response
distension or inflammation of the urinary bladder to visceral stimulation, can be as great as
or gastrointestinal tract and also by muscle spasms 4172 mmHg, but within the next few days, these
that often develop after SCI (Guttman and responses become smaller, perhaps associated with
Whitteridge, 1947; Corbett et al., 1975; Mathias degenerative changes in the spinal sympathetic
and Frankel, 1992a; Blackmer, 2003). They can be preganglionic neurons caudal to the injury (see the
initiated by routine daily procedures such as blad- following sections) (Krassioukov and Weaver,
der catheterization and bowel evacuation. Dysre- 1995; Maiorov et al., 1997a, b). Then, in the en-
flexia is not always severe and may include only suing weeks, the increases in AP during dysreflexia
sweating, flushed skin above the segment of injury, become larger, reaching values as high as
piloerection and small increases in AP. However, 5274 mmHg by 5 weeks after SCI. The large re-
this condition can become uncontrolled, leading to sponse on the first day likely reflects the full ca-
life-threatening hypertension (Shea et al., 1973; pacity of the spinal reflex, without descending
Naftchi, 1990; Mathias and Frankel, 1992c; Lee inhibitory restraint (Dembowsky et al., 1980), and
et al., 1994; Naftchi and Richardson, 1997; before the secondary effects of cord injury have
Giannantoni et al., 1998; Blackmer, 2003). Mech- impacted greatly. After this time, the magnitude of
anisms for autonomic dysreflexia may involve af- the hypertension appears to become a function of
ferent, interneuronal and efferent components of many changes ongoing in the spinal cord and
the spinal sympathetic reflex and the vasculature, vasculature, some degenerative and others, plastic
as also discussed in other chapters of this book. responses to the injury. These intraspinal changes,
247

and their relationship to this time course of auto-


nomic dysreflexia will be discussed later in this
chapter.
Autonomic dysreflexia is readily apparent in
rats when SCI is caused by a method that repro-
duces the typical clinical injury. This method en-
tails brief (60 s) extradural compression of the cord
with a calibrated, modified aneurysm clip. The
clip-compression model mimics the key patho-
physiological features of human SCI. This model
produces mechanical injury (primary injury) and
secondary damage by a variety of well-character-
ized mechanisms including microvasculature dis-
ruption, hemorrhage, ischemia, increases in
intracellular calcium, calpain activation, progres-
sive axonal injury and glutamate toxicity (Wallace
and Tator, 1986; Wallace et al., 1986; Fehlings
et al., 1989; Koyanagi et al., 1993a, b; Agrawal
and Fehlings, 1996, 1997a, b; Agrawal et al., 1998;
Schumacher et al., 1999). Marked autonomic
dysreflexia is readily evoked after severe (50 g)
clip-compression cord injury at T2–4 in rats and
mice (Maiorov et al., 1998; Mayorov et al., 2001;
Weaver et al., 2001b; Jacob et al., 2003) (Fig. 1A).
Distension of the colon with a small balloon-
tipped catheter or cutaeous stimulation in con-
scious rats at two weeks after injury causes pressor
responses that continue to increase in magnitude
until 6 weeks after cord injury, when they appear
to plateau (Gris et al., 2004; Marsh and Weaver,
2004). Dysreflexia after clip-compression injury
can be evoked by visceral or cutaneous stimuli that
are noxious or non-noxious. Gently stroking the
skin of the back caudal to the segment of injury
causes pressor responses of 2375 mmHg, whereas
Fig. 1. The MAP, pulsatile AP and HR in two rats, 2 weeks
pinching the skin increases pressure by up to after clip-compression SCI: after intrathecal treatment with
3272 mmHg. Likewise, distension of the colon to control immunoglobulin G (IgG) (A) and after intrathecal
an extent that would be noxious in the intact an- treatment with trkA-IgG (B). Colon distension for 60 s (onset
imal increases AP by 4274 mmHg, whereas a and duration marked with a thick line) stimulated a large in-
crease in AP and MAP and a decrease in HR in the control
milder distension, that would not be noxious, still
IgG-treated rat. In contrast, colon distension caused only a
increases AP by 2473 mmHg. In a conscious rat modest decrease in AP and MAP after treatment with trkA-
with an intact CNS, this non-noxious cutaneous or IgG. Reprinted with permission from Marsh et al. (2002).
visceral stimulation would cause either no change
in AP or a transient (5–10 s) increase of abnormal, or dysreflexic responses, as they are
5–10 mmHg. These findings refute the idea that more than twice the magnitude of those in an
autonomic dysreflexia is initiated only by noxious intact rat.
stimuli. The responses to the non-noxious stimuli The large increase in noradrenaline spill-over re-
are smaller than those to noxious stimuli, but are ported during dysreflexia is consistent with intense
248

sympathetic firing relative to baseline values with time after injury, from 400 to 450 beats/min
(Karlsson et al., 1998). Direct measurements of in intact rats or cord-injured rats up to a week
visceral vasomotor sympathetic firing can be made after injury, to rates of 500–570 beats/min at 2–6
in animals. The renal nerve has mostly vasomotor weeks after injury. In one study, we attributed this
sympathetic axons, and renal sympathetic firing tachycardia to increased afferent input to upper
recorded in conscious rats, after cord transection thoracic preganglionic neurons, resulting from
at T4, increases massively during episodes of au- sprouting of primary afferent fibers in the dorsal
tonomic dysreflexia (Maiorov et al., 1998). The horn (Krenz et al., 1999). This input, if tonically
increases are at least sixfold above baseline firing active, could contribute to enhanced sympathetic
and, during the first week after cord transection, drive to the heart. Others have also reported in-
they parallel the increase in amplitude of the blood creases in HR after cord transection at T4
pressure responses. Recordings at later times have (Rodenbaugh et al., 2003), and have suggested
not been reported. These spinal reflexes are much several mechanisms for this tachycardia (see
larger than they would be if the nervous system Collins et al., this volume).
were intact (Coote et al., 1969; Koizumi and
Brooks, 1972; Dembowsky et al., 1980) and reflect
loss of descending inhibition and, with time, plas- Reorganization of the injured spinal cord
tic changes within the spinal cord.
Basal firing of the renal sympathetic nerve is Caudal to the SCI, the loss of supraspinal input
greatly reduced by cord transection so that the may make the sympathetic preganglionic neuron
nerve targets are possibly approaching the state of more sensitive to spinal afferent input, not only
‘‘denervation’’ postulated by McLachlan and because of the loss of tonically active inhibitory
Brock (see this volume and Yeoh et al., 2004). supraspinal influences, but also because, in the
The low firing rate certainly could prompt signif- absence of descending excitatory synaptic input,
icant changes in the peripheral sympathetic system the remaining excitatory synapses within spinal
and vascular targets. However, even the low basal reflex circuits have a more dominant influence. As
sympathetic firing in the first week after T4 tran- reviewed by Llewellyn-Smith et al. (this volume),
section appears to make a contribution to vaso- glutamatergic inputs to preganglionic neurons are
motor control, as ganglionic blockade decreases largely lost within a few days of cord transection
resting AP by 4577 mmHg (Maiorov et al., (Llewellyn-Smith and Weaver, 2001), yet, at the
1997b). This lowered sympathetic activity becomes same time, the neurons respond vigorously to
the norm after cord injury, and, if this activity is reflex inputs, suggesting that a few dominant
greatly increased by a spinal reflex, coupled with a synapses can activate the neurons. Reorganization
more reactive vasculature and changes in periph- of the spinal pathways controlling sympathetic
eral neurotransmission, large hypertensive re- preganglionic neurons after the loss of supraspinal
sponses can occur. input is likely to change the makeup of inputs to
Spinal cord transection or compression also has these neurons (Krassioukov and Weaver, 1996;
significant initial effects on resting AP, decreasing Cassam et al., 1997, 1999; Weaver et al., 2001b).
it by 2374 mmHg during the first 24 h after cord We searched for changes in glutamatergic spinal
injury (Maiorov et al., 1997b). This decrease is reflex transmission via N-methyl-D-aspartate
transient and by 1 week after injury AP returns to (NMDA) and a-amino-3-hydroxy-5-methyl-4-
normal values (10173 mmHg). This range of AP isoxazolpropionic acid (AMPA) receptors during
is stable for at least 6 weeks (Krassioukov and the first 2 weeks after spinal cord transection
Weaver, 1995; Maiorov et al., 1997a; Krenz et al., (Maiorov et al., 1997a). Using selective receptor
1999; Weaver et al., 2001b; Marsh et al., 2002; Gris antagonists, these two receptors appeared to con-
et al., 2004; Marsh and Weaver, 2004). In contrast, tribute about equally (35% each) to the reflex
HR initially is not altered by cord transection or excitation causing autonomic dysreflexia, at 2 and
clip-compression at T4, but gradually increases 16 days after the injury, demonstrating no change
249

with time in the roles of these receptors during the interneurons after cord injury does not appear to
initial period after injury. increase and, therefore, an increased number of
The enhanced transmission through the spinal synapses from interneurons to preganglionic neu-
reflex circuits suggested by the recording of renal rons does not mediate the time-dependent increas-
nerve responses (Maiorov et al., 1997b) could be es in the spinal sympathetic reflexes and pressor
mediated by changes in the afferent, interneuronal responses. The interneurons may still contribute if
or preganglionic neuronal components of the arc. they are more excited by an afferent input to the
Earlier studies lead to the hypothesis that synaptic spinal cord, and provide increased preganglionic
input to preganglionic neurons undergoes plastic excitation through temporal rather than spatial
reorganization after cord injury, resulting in summation. Consistent with this view, an electro-
replacement of inputs from bulbospinal neurons physiological study of responses of sympathetical-
with those from interneurons (Krassioukov and ly correlated spinal interneurons to cutaneous
Weaver, 1996; Weaver et al., 1997). Thoracic inputs, after spinal cord transection, showed that
preganglionic neurons undergo a transient degen- the receptive fields of individual interneurons to
erative response, characterized by retraction of this input enlarge with time after spinal cord
dendrites and shrinkage of the soma, during the transection, increasing their capacity to provide
first week after cord transection (the period of excitatory input to the preganglionic neurons
synaptic loss), and then re-establish a normal (Krassioukov et al., 2002) (see also Schramm, this
dendritic arbor and soma size within 2–4 weeks volume). Moreover, the gray matter containing
after the injury (Krassioukov and Weaver, 1996; thoracic preganglionic neurons, and interneurons
Krenz and Weaver, 1998a; Llewellyn-Smith and providing input to them, develops a network of
Weaver, 2001). This acute degenerative response fibers immunoreactive to growth-associated
of the preganglionic neurons with later morpho- protein-43 that increases in density between 2
logical recovery was also found in samples of in- and 4 weeks after cord transection (Weaver et al.,
jured human spinal cord (Krassioukov et al., 1997). This protein was also present in putative
1999). These observations led us to propose a growth cones and in cell bodies that likely were
model in which synapses from supraspinal neurons interneurons. These data suggest that, although
were replaced with those from spinal circuits, the thoracic preganglionic neurons themselves did
leading to exaggerated spinal reflex control of not receive new synaptic inputs, growth within the
preganglionic neurons. Immediately after cord local neuropil likely generated new inputs to other
injury, the preganglionics are intact and descend- neurons antecedent to the preganglionics.
ing inhibition is lost, leading to robust spinal
reflex-mediated pressor responses. Then, pregangl-
ionic neurons undergo a degenerative response Changes in the primary afferent arbor contributing
that is maximal at about 1 week, the time when to autonomic dysreflexia
autonomic dysreflexia is decreased in magnitude.
Finally, as the preganglionic neurons recover from In the dorsal horn, the arbors of small-diameter
the atrophy and have a normal soma size and primary afferent neurons can enlarge greatly in
dendritic tree, with potentially new synaptic inputs rats and mice after SCI (Krenz and Weaver,
from interneurons, autonomic dysreflexia begins 1998b; Krenz et al., 1999; Wong et al., 2000; Jacob
to increase in magnitude. Although this was an et al., 2001, 2003; Weaver et al., 2001b) (Fig. 2)
appealing model, an electron microscopic study potentially leading to increased reflex excitation of
(Llewellyn-Smith and Weaver, 2001) demonstrated preganglionic neurons, via interneuronal path-
that the loss of bulbospinal synaptic input to ways. These afferent neurons that are calcitonin
thoracic preganglionic neurons reduces the num- gene-related peptide (CGRP)-immunoreactive,
ber of synapses on these neurons by 50–70%, and and the interneurons in laminae III–VII activated
this loss is not replaced by intraspinal inputs. by these afferent projections, make up the path-
Accordingly, the number of synapses from the way, mediating spinal reflex excitation of
250

Fig. 2. Digital photomicrographs of CGRP-immunoreactive (left, red) and substance P-immunoreactive (right, green) afferent fibers in
the dorsal horn of sham-injured rats (A and B) and of rats with clip-compression SCI at T4 (C-H). Examples are shown at 2 weeks after
sham or SCI. Tissue sections (30 mm) from T12 to L1 were processed for double immunofluorescence for CGRP (red) and substance P
(green) and visualized with a 10  objective (A–D, calibration bar ¼ 100 mm), a 20  objective (E–G, calibration bar ¼ 50 mm), or with
confocal microscopy (H, calibration bar ¼ 10 mm). Co-localization of CGRP and substance P in afferent fibers is indicated by yellow.
Reprinted with permission from Marsh and Weaver (2004).
251

preganglionic neurons (Sato and Schmidt, 1973; dysreflexia in response to colon distension and no
Cabot et al., 1994; Joshi et al., 1995; Clarke et al., change in the CGRP-immunoreactive afferent
1998). After cord transection or compression in arbor. In contrast, after a more severe (50 g) inju-
rats and some strains of mice, the 2-week time ry, autonomic dysreflexia develops consistently
course for the increased arbor size correlates with and the CGRP-immunoreactive arbor in the lower
the gradual increase in the magnitude of auto- thoracic and lumbar spinal cord enlarges. The dif-
nomic dysreflexia (described earlier) and may also ference between the transection and compression
be a factor contributing to the increase. After injury model is the sparing of a limited number of
transection injury, the size of the increase in the white-matter axons that pass through the lesion
CGRP-immunoreactive afferent arbor correlates site. If the compression is only moderate, and
with the magnitude of dysreflexia (Krenz et al., many axons are spared, sufficient bulbospinal in-
1999; Jacob et al., 2001, 2003). This afferent arbor put remains to prevent the development of
remains enlarged at 1 month after cord injury, a dysreflexia and afferent sprouting. However, after
time when autonomic dysreflexia is well developed severe compression injury, the small sparing of
in rats (Krassioukov and Weaver, 1995). The descending fibers is not adequate to block dysre-
‘‘enlarged’’ CGRP-immunoreactive afferent arbor flexia and the sprouting response does develop,
might reflect only increased CGRP expression in albeit modestly.
the fibers rather than a sprouting or growth The small diameter C-type sensory neurons in
response. However, a confocal microscopic the dorsal root ganglia contain a population that
analysis of changes in the afferent arbor after T4 express CGRP, and a subset of neurons within this
cord transection, using retrograde transport of population also express substance P (Price, 1985;
wheat germ agglutinin, revealed a true increase in Ju et al., 1987). In addition to the small-diameter
the size of the arbor of small diameter fibers in the neurons, CGRP is also expressed in some of the
lumbar spinal cord (Wong et al., 2000). Likewise, larger sensory neurons (McCarthy and Lawson,
transport of the b subunit of cholera toxin after 1990; Lawson et al., 1993, 1996). The plasticity in
cord transection also revealed expansion of the the CGRP-immunoreactive afferent arbor initiat-
arbor of larger afferent fibers in the lumbar cord ed by cord injury appears to target only the pop-
(Krenz and Weaver, 1998b). Together, these find- ulation that expresses CGRP without substance P
ings demonstrate that the increase in CGRP after (Marsh and Weaver, 2004) (Fig. 2). The absence of
cord injury reflects a growth response, not simply an sprouting of substance P-containing afferent fibers
increase in gene expression. The changes in the in the dorsal horn, in the presence of increased
larger diameter fibers probably underlie the re- densities of CGRP-containing fibers, shows the
sponses to non-noxious cutaneous and visceral stim- selectivity of this response, suggesting that the
ulation described above (Marsh and Weaver, 2004). larger fibers play a greater role in dysreflexia.
The relationship between changes in the afferent Finally, as also discussed by Rabchevsky (this vol-
arbor and the magnitude of dysreflexia is less ro- ume), increases or decreases in the afferent arbor
bust in the clip-compression cord-injury model are only relevant to inputs entering the spinal cord
that spares some descending pathways. Enlarge- at the same segmental region. For example, be-
ment of the CGRP-immunoreactive primary af- cause of the segmental arrangement of the primary
ferent arbor is also characteristic of this model, but afferent projections to the spinal cord (Vizzard
changes are smaller than after cord transection, et al., 2000), changes in the lumbosacral arbor will
whereas the magnitude of dysreflexia can also be impact on responses to colon or bladder distension
as great (Krenz and Weaver, 1998b; Krenz et al., but not on those to cutaneous stimulation of the
1999; Weaver et al., 2001b). Injury to the cord mid-thoracic trunk. Likewise, changes in the mid-
must be severe for enlargement of the primary af- thoracic region do not impact on responses to co-
ferent arbor and autonomic dysreflexia to occur. lon distension (Cameron et al., 2003; Gris et al.,
At 2 weeks after moderate/mild (20 g) clip-com- 2005). In our studies, the afferent arbor was par-
pression cord injury at T4, rats have no autonomic ticularly increased in the lower lumbar segments
252

after cord transection or compression (Krenz and T4 SCI, we have confirmed that NGF levels in the
Weaver, 1998b; Krenz et al., 1999; Jacob et al., injured rat spinal cord are significantly greater in
2001, 2003; Weaver et al., 2001b), making afferent and adjacent to the injury (T3–5) than rostral to
plasticity more of an issue to responses initiated the injury (T1 and 2) or in the lumbar cord. NGF
from the pelvic organs or hind limbs. at the injury site was approximately twofold great-
Enlargement of the afferent arbor depends upon er than the content in this region in un-injured rats
actions of nerve growth factor (NGF) in the in- (1.570.8 pg/mg). Immunocytochemistry on spinal
jured rat spinal cord. Intrathecal delivery of a cord sections revealed increased NGF-immunore-
highly selective neutralizing antibody to NGF for activity in cells and fibers in the dorsal root
2 weeks after cord transection at T4 completely entry zone, and in astrocytes, microglia and
blocked the sprouting of the small-diameter affer- leptomeningeal cells of cord-injured rats (Krenz
ent fibers in the dorsal horn (Krenz et al., 1999). and Weaver, 2000). These were prevalent in
Delivery of the antibody subcutaneously to pe- segments T3–8.
ripheral targets of the sensory neurons had no ef- A second study of NGF in the injured spinal
fect, demonstrating that the afferent sprouting was cord extended these observations further by
not caused by a target-derived source of NGF. searching for cells that produced NGF, i.e., con-
Blockade of the sprouting response was due to the tained messenger RNA (mRNA) for NGF. In ad-
neutralizing effects of the anti-NGF antibody on dition, double-labeling immunocytochemistry was
the trkA-expressing central arbors of these sensory used to identify, with greater certainty, the cells
neurons. Autonomic dysreflexia was measured in containing this protein (Brown et al., 2004). We
the same rats and the increases in AP caused by also compared the NGF profiles in spinal cords
visceral stimulation were reduced by 43% in the with transection or compression injuries. In intact
(intrathecal) antibody-treated rats. This reduced spinal cords, NGF mRNA was in leptomeningeal
the dysreflexia to the magnitude of the spinal reflex cells and neurons of the intermediate gray matter,
that is present in the conscious rat within 48 h of whereas NGF protein was only in the leptome-
cord transection, before enlargement of the affer- ninges (Fig. 3). At 3–7 days after transection or
ent arbor could occur (Maiorov et al., 1997a). clip-compression injury NGF mRNA and protein
Similarly, intrathecal delivery, for 2 weeks after were expressed in the lesion and throughout the
clip-compression injury at T4, of a trkA-IgG fu- intermediate gray matter and the white matter
sion protein that sequesters NGF decreased rostral and caudal to the injury site. The NGF
dysreflexia in rats by 30% (Marsh et al., 2002) mRNA and protein in spinal cord sections adja-
(Fig. 1B). These data led to two conclusions. First, cent to each other were expressed in ramified mi-
afferent sprouting in the spinal cord dorsal horn is croglia, astrocytes, intermediate gray neurons, pial
clearly associated with the time-dependent increase cells, leptomeningeal cells and Schwann cells in the
in hypertensive responses to sensory stimulation, lateral white matter and lesion site (Fig. 4).
characteristic of autonomic dysreflexia. Second, Rounded macrophages in the lesion contained
the sprouting is caused by an intraspinal action of immunoreactivity for NGF, but the cells express-
NGF, presumably at the central arbors of the sen- ing NGF mRNA were not macrophages, and did
sory neurons containing CGRP. not appear to contain the protein. These findings
These studies suggest that NGF is a culprit demonstrated that a variety of cells within the
in the development of autonomic dysreflexia. injured spinal cord could produce NGF, but that
Normally, the spinal cord contains very little macrophages are not among them. Macrophages
NGF but NGF levels within a few segments of a may have contained NGF because they phagocy-
cord injury site have been reported to increase to a tosed it.
peak at 1 week post injury, remaining increased for The studies described above show a strong
4 weeks (Bakhit et al., 1991). Using a two-site relationship between NGF and changes in the
enzyme-linked immunosorbant assay (ELISA) to afferent arbor that can contribute to dysreflexia.
analyze spinal cord homogenates from rats with a The effectiveness of the anti-NGF antibody and
253

Fig. 3. The NGF mRNA expression in the injured and un-injured (control) spinal cord. Hybridization signals visualized by alkaline
phosphatase are illustrated in longitudinal sections of the 5th thoracic spinal cord segment (T5). The lateral edge of the spinal cord is at
the top of each photomicrograph. In the un-injured control rats, NGF mRNA was intense in leptomeningeal cells at the edge of the un-
injured spinal cord and in gray-matter cells with morphology like neurons (A–C). Processing serial sections to detect either NGF
mRNA (C) or NeuN-immunoreactivity, confirmed that these gray-matter cells were neurons (D). The leptomeningeal cells at the edge
of the un-injured spinal cord had hybridization signal but none was present in white matter cells (B). NGF mRNA expression increased
after T4 transection-SCI (E–G). At one day, little NGF mRNA was present with the exception of patches of signal in the le-
ptomeningeal cells of the pia mater (E). At 3 days, expression of NGF mRNA was robust in white and gray matter and in the
leptomeninges, especially in segments close to the lesion (F). The NGF mRNA signals were still present but less abundant at 7 days
after SCI (G). Panels H–J show cells expressing NGF mRNA 7 days after SCI (at higher magnification). Within the lesion site, cells
were typically round and clustered (H), whereas those in the white matter were long and spindle shaped (I) and gray-matter cells
expressing NGF mRNA were oval and neuron-like in morphology or small and spindle-shaped (J); d, day; wm, white matter; gm, gray
matter; con, control. The scale bars on A and B are 50 mm; the bar on C also refers to D and is 50 mm; the bar for E–G, shown in G is
250 mm; the bar for H–J, shown in J is 50 mm. Reprinted from Brown et al. (2004) with permission from Elsevier.

the trkA-IgG fusion protein in limiting dysreflexia response to cord injury begins within minutes
shows that this strategy might be useful clinically and evolves for days, spreading throughout the
to prevent the development of dysreflexia. The damaged cord and into adjacent, non-injured
stimulus for the increased intraspinal NGF is now regions (Blight, 1985; Tator and Fehlings, 1991;
in question. The inflammatory response to trau- Young, 1993; Popovich et al., 1997; Taoka and
matic injury of the spinal cord is likely to promote Okajima, 1998). First, pro-inflammatory chemo-
the production of NGF. The inflammatory kines and cytokines are released into the injured
254

Fig. 4. Identification of cells immunoreactive for the astrocytes marker glial fibrillary acidic protein (GFAP) and NGF in longitudinal
sections of the T5 spinal cord. Cells in the white matter are shown at 7 days after T4 transection-SCI (transect, A–C) or T4 clip-
compression SCI (clip, D–F). Immunoreactivity in the lesion is shown at 7 days after clip-SCI (G–I). Immunoreactivity for NGF (A, D
and G) and for GFAP (B, E and H) colocalized in the astrocytic glia limitans at the pial border of the white matter (C, arrow), and in
white-matter astrocytes (C, and F arrowheads). As few GFAP-immunoreactive astrocytes are in the lesion center, only small areas of
colocalization were present in the lesion (I, arrowhead). Scale bar in I refers to all panels and is 50 mm. Reprinted from Brown et al.
(2004) with permission from Elsevier.

area by astrocytes, microglia and endothelial cells. and NGF expression and/or signaling are clearly
Next, activation and proliferation of glia (gliosis) related. Interleukin-6 (IL-6) and IL-1b, two
occur, and neutrophils and monocyte/macrophages cytokines found in the injured cord (Streit et al.,
enter the spinal cord from the circulation (Popovich 1998; Hayashi et al., 2000), have well-documented
et al., 1997; Taoka et al., 1997; Leskovar et al., actions to increase the expression of NGF in the
2000). Details of the inflammation in the injured CNS (Bandtlow et al., 1990; Spranger et al., 1990;
spinal cord will be presented below. Inflammation Saporito et al., 1993) and in Schwann cells of
255

peripheral nerves (Heumann et al., 1987a, b; and intensified necrosis and apoptosis of neurons
Lindholm et al., 1987). In response to cord and glia, events that contribute to increases in
injury, microglial NGF mRNA transcription and lesion size (Taoka and Okajima, 1998; Bethea and
NGF protein release (Heese et al., 1998a, b) are Dietrich, 2002). Neutrophils and hematogenous
stimulated synergistically by IL-1 and tumor ne- macrophages invade the spinal cord, peaking at
crosis factor (TNF)-a. The central processes of 12 h and 5–7 days after SCI, respectively
sensory neurons have IL-6 receptors that would be (Popovich et al., 1997; Taoka et al., 1997;
exposed to IL-6 in the injured cord, leading to Leskovar et al., 2000), and releasing pro-inflam-
stimulation of NGF expression. matory cytokines and reactive oxygen and nitro-
The NGF also has pro-inflammatory actions gen species (Taoka and Okajima, 1998; Popovich
(LaSala et al., 2000). The high-affinity trkA re- et al., 1999). These cytotoxic substances can fur-
ceptor is expressed on monocytes, B-lymphocytes ther damage the white and gray matter and
and T-lymphocytes. Although the signaling caused contribute to scar formation (Jones et al., 2002).
by NGF on these receptors is not fully understood, The progression of secondary damage deterio-
NGF promotes expression of Bcl-2 in monocytes rates neurological function, and renders the spinal
and mast cells, conferring resistance to cell death cord less amenable to repair strategies such as cell
(Bullock and Johnson, 1996; LaSala et al., 2000; replacement or therapeutic gene transfer. There-
Saragovi and Gehring, 2000). The NGF also can fore, preventing this progression is of key impor-
facilitate inflammation by promoting differentia- tance. No practical way to control the early
tion of myeloid progenitor cells, inducing prolif- inflammatory response has yet been devised,
eration and maturation of B-lymphocytes and despite our understanding of its destructive role
stimulating the release of inflammatory cytokines in the final outcome of an injury. We postulated
such as TNF-a from basophils and mast cells. that a successful anti-inflammatory treatment
In summary, inflammation can promote NGF must selectively disrupt the early, destructive,
expression and NGF can promote the process of leukocyte-mediated actions in the injured cord,
inflammation. Clearly, the relationship between while leaving an opportunity for later regenerative
inflammation and NGF could contribute to the interventions and wound-healing responses. After
NGF-dependent increases in primary afferent SCI, intraspinal leukocyte infiltration first requires
arbors in the spinal cord and the development of leukocyte tethering, by selectins, to the surface of
autonomic dysreflexia. endothelial cells (Bevilacqua, 1993). This is
followed by the interaction of endothelial cell-
adhesion molecules with integrins on the
Inflammation, secondary damage and autonomic leukocyte surface (Neish et al., 1995; Shanley
dysreflexia et al., 1998), facilitating leukocyte extravasation
through the blood-CNS barrier. To prevent this
Whereas blocking inflammation after SCI may interaction, we used a monoclonal antibody
limit the production of NGF, it also has a host of (mAb) to the CD11d subunit of the CD11d/
neuroprotective actions that make this issue a high CD18 integrin (Grayson et al., 1999; Van der
priority in devising strategies to prevent the devel- Vieren et al., 1999) (ICOS Corporation, Bothell,
opment of autonomic dysreflexia as well as the WA). This mAb treatment substantially decreases
other autonomic, sensory and motor dysfunctions the numbers of neutrophils and macrophages at
after cord injury. Secondary damage after an SCI the lesion at 3 days after SCI (Mabon et al., 2000;
is caused, in part, by ischemia, cellular and tissue Saville et al., 2004). It also markedly reduces oxi-
edema, amino acid excitotoxicity and oxidative dative damage to the injured spinal cord (Bao
damage. The extent of this damage directly corre- et al., 2004a, b). The anti-CD11d mAb was inject-
lates with the magnitude of autonomic dysreflexia ed intravenously in three consecutive doses at 2, 24
(Weaver et al., 2001b). These changes often relate to and 48 h after clip-compression cord injury at T4,
early inflammation and lead to myelin degradation and effects were assessed on autonomic dysreflexia
256

and on the spinal cord lesion at 2 and 6 weeks after control rats more often included arrhythmias
the injury (Gris et al., 2004) (Figs. 5 and 6). (Fig. 5). AP responses of the treated rats remained
At 2 weeks after cord injury, colon distension smaller than those of the control rats at this time.
increased AP and decreased HR for the duration Again, the mean changes in AP of the control rats
of stimulus (Fig. 5). The anti-CD11d mAb treat- (40 mmHg) were significantly greater than those
ment markedly reduced these increases in AP. of the treated rats (28 mmHg). At this time, as at
Mean increases during the 1 min stimulation peri- the earlier 2-week analysis, the anti-CD11d treat-
od were reduced significantly from 30 mmHg in ment did not alter resting AP (110 mmHg) or
the control rats to 20 mmHg in the treated rats. HR (500 beats/min). Additional experiments
As the normal reflex response to this stimulus in an were done to determine the effect of delaying the
intact rat is an increase of 15 mmHg in AP onset of treatment to 6 h after compression injury.
(Maiorov et al., 1997b), this treatment affected The second and third injections of anti-CD11d
much of the reflex increment attributable to cord antibody were administered at 24 and 48 h, re-
injury. The average decreases in HR during the spectively. This treatment also decreased dysre-
1 min colon distension in control rats (65 beats/ flexia by 50% at 6 weeks after cord injury,
min) tended to be greater than in treated rats (45 demonstrating that a clinically relevant dosing
beats/min), possibly because of the greater arterial schedule still is highly effective.
baroreceptor activation by the larger pressor re- The basis for the improved autonomic outcome
sponses. The treatment had little impact on resting after this selective anti-inflammatory treatment
AP (100 mmHg) or HR (500 beats/min). At 6 was sparing of the tissue surrounding the lesion ar-
weeks after compression cord injury, the increases ea. The spinal cords had increased areas of compact
in AP during dysreflexia were greater than at 2 myelin within 2–3 mm of the injury site at 2 and 6
weeks and HR responses to colon distension in the weeks after injury (Fig. 6). Neurofilament in the

Fig. 5. Anti-CD11d mAb treatment reduces the magnitude of autonomic dysreflexia. Changes in pulsatile AP, MAP and HR are
shown during a 2 ml balloon distension of the colon in control (A and D) and mAb-treated rats (B and E) at 2 weeks (top row) and 6
weeks (bottom row) after SCI at T4. The large increase in pressure evoked at 6 weeks was often associated with arrhythmias (D).
Average change in MAP was assessed at 2 (C) and 6 (F) weeks after cord injury in control rats (& C, n ¼ 14; F, n ¼ 6) and mAb-
treated rats (’ C, n ¼ 10; F, n ¼ 7). The dotted lines on the bar graphs show the approximate change in pressure that would be elicited
by such stimulation in a rat with an intact spinal cord (Maiorov et al., 1997a). bpm, beats per minute; *Po0.05 compared to control
rats. Reprinted with permission from Gris et al. (2004), copyright 2004 by the Society for Neuroscience.
Fig. 6. Anti-CD11d mAb-treatment increases myelin in the injured cord after SCI. Solochrome cyanin-stained sections taken at 6 weeks after T4 SCI are shown at the
lesion epicenter and at 2.0 mm caudal to the epicenter in control and mAb-treated rats (a). Arrows on photomicrographs at the epicenter indicate patches of intact dark
blue compact myelin. Note the more intact neuropil at 2 mm caudal to the injury site in the rat treated with anti-CD11d mAb, reflected by more abundant compact myelin
and a smaller cavitation than in the control rat. Calibration bar equals 200 mm and applies to all photomicrographs. Treatment effects on normalized areas of compact
myelin are illustrated after SCI at T4 (b, 2 weeks; c, 6 weeks). Myelin was stained with luxol fast blue in b and solochrome cyanin in c. & control rats (b, n ¼ 10; c, n ¼ 5).
’ mAb-treated rats (b, n ¼ 8; c, n ¼ 7). *Po0.05 compared to control rats. +shortest distance where area is larger than area at epicenter, Po0.05. Reprinted with
permission from Gris et al. (2004) copyright 2004 by the Society for Neuroscience.

257
258

Fig. 7. Longitudinal section of rat lumbar (L2) spinal cord illustrating fibers immunoreactive for 5-HT (serotonin, green) streaming
from the spared white matter bundle at the edge of the spinal cord caudal to a clip-compression SCI at the 12th thoracic segment. This
section shows the abundance of these fibers at 4 weeks after cord injury in a rat that had been treated with the anti-CD11d mAb. Fibers
extend to the edge of the gray matter (bottom of illustration) and appear to surround sympathetic preganglionic neurons in the
intermediolateral cell column. The inset shows these lateral horn neurons stained for neurofilament immunoreactivity (red) with fibers
and possible terminals on their somata. This response suggests that axons spared by the anti-CD11d mAb treatment produced
collateral sprouting branches directed toward the preganglionic neurons and possibly innervating them. Calibration bar ¼ 50 mm (inset
10 mm).
259

gray and white matter was also significantly This approach has significant clinical promise as it
increased, indicative of neuronal sparing. The also has beneficial effects on motor function and
improvement in dysreflexia evoked by colon the development of neuropathic pain (Gris et al.,
distension did not relate to effects on the CGRP- 2004; Oatway et al., 2005).
immunoreactive primary afferent arbor. No treat- In conclusion, SCI leads to a host of changes
ment effects on this arbor were found in the dorsal within the injured cord that promote pathological
horn of the lumbar spinal cord, the segments secondary outcomes such as autonomic dysreflex-
mediating the afferent input from colon distension ia. Although neurotrophic factors are useful and
(Gris et al., 2005). Remarkably, the afferent arbor probably necessary for the process of regeneration
at T9, caudal to the injury site, actually had a of injured tracts and neurons, NGF does not ap-
greater area in antibody-treated than control rats. pear to be a good candidate for this task as it
This presumably reflected the tissue sparing in the clearly contributes to secondary maladaptive dis-
dorsal horn as the treated rats had almost intact orders. Likewise, inflammation is a necessary part
dorsal horns, whereas the controls had significant of healing but, in the CNS, inflammation can
secondary tissue damage that extended from T4 to cause great damage that is not readily reversed.
T9. This change would not have impacted on Therefore, the inflammatory response to cord
dysreflexia initiated from the colon but might have injury also is an ideal target for manipulation to
affected dysreflexia evoked from the mid-thoracic maintain its useful functions while limiting its
region. This relationship was not assessed in our capacity to cause destructive or maladaptive
study, as we did not anticipate the effect near the responses. Autonomic dysreflexia is one of many
lesion. However, other than effects due to tissue secondary consequences of cord injury such as
sparing, the anti-integrin antibody treatment bladder and sexual dysfunction, chronic pain,
did not alter the CGRP-immunoreactive arbor. and muscle spasticity. Understanding the mecha-
Instead effects appeared to be caused by retention nisms of autonomic dysreflexia and development
of some descending control of spinal neurons. of treatments to prevent its development may im-
In a different study of effects of the anti-CD11d pact on many of these disabling secondary
antibody treatment (Oatway et al., 2005), we disorders.
found that it led to extensive sprouting of de-
scending serotonergic axons that passed through
the margins of the lesion. These axons were par-
ticularly attracted to the intermediolateral cell col-
umn, forming masses of projections to this area Acknowledgments
(Fig. 7). The projections provided restored, even
enhanced, bulbospinal serotonergic inputs to the This research was supported by grants from the
preganglionic neurons caudal to the injury site. As Canadian Institutes of Health Research, The
this study concerned only the injury and responses Heart and Stroke Foundation of Ontario
of the lower thoracic and lumbar cord, it would (T4053) and The Ontario Neurotrauma Founda-
not have affected autonomic dysreflexia and we tion. The authors wish to acknowledge that part of
can only speculate that the same phenomenon the research reviewed in this chapter was conduct-
would occur after a mid-thoracic injury, leading to ed by previous postdoctoral fellows and graduate
enhanced serotonergic input to all preganglionic students, Dr. Andrei Krassioukov, Dr. Dmitry
neurons caudal to the injury site. In summary, Mayorov, Dr. Natalie Krenz, Dr. Sharon Wong,
blocking inflammation with a selective, transient Mr. Aly Cassam, Ms. Mary-Jo Ricci and Mr.
approach, which does not impair later wound Mark Oatway. We also are indebted to our past
healing actions of leukocytes, markedly decreased and present excellent technical staff, Mrs. Barbara
the development of autonomic dysreflexia, by Atkinson, Ms. Yuhua Chen, Ms. Eilis Hamilton,
mechanisms that appear to be related to preserva- Ms. Leyana Saville and Mrs. Carmen Simedrea
tion or sprouting of spared descending pathways. who have been essential to this research.
260

References Cameron, A.A., Smith, G.M., Randall, D.C., Brown, D.R. and
Rabchevsky, A.G. (2003) Effects of nerve growth factor over-
Agrawal, S.K. and Fehlings, M.G. (1996) Mechanisms of expression on autonomic dysreflexia after spinal cord injury.
secondary injury to spinal cord axons in vitro role Na+, J. Neurotrauma, 20: 1086.
Na+–K+–ATPase, the Na+–H+ exchanger, and the Cassam, A.K., Llewellyn-Smith, I.J. and Weaver, L.C. (1997)
Na+–Ca+ exchanger. J. Neurosci., 16(2): 545–552. Catecholamine enzymes and neuropeptides are expressed in
Agrawal, S.K. and Fehlings, M.G. (1997a) Role of NMDA and fibres and somata in the intermediate grey matter in chronic
non-NMDA ionotropic glutamate receptors in traumatic spinal rats. Neuroscience, 78: 829–841.
spinal cord axonal injury. J. Neurosci., 17(3): 1055–1063. Cassam, A.K., Rogers, K.A. and Weaver, L.C. (1999) Co-
Agrawal, S.K. and Fehlings, M.G. (1997b) The effect of the localization of substance P and dopamine b-hydroxylase with
sodium channel blocker QX-314 on recovery after acute growth associated protein-43 is lost caudal to a spinal cord
spinal cord injury. J. Neurotrauma, 14: 81–88. transection. Neuroscience, 88: 1275–1288.
Agrawal, S.K., Theriault, E. and Fehlings, M.G. (1998) Role of Clarke, H.A., Dekaban, G.A. and Weaver, L.C. (1998) Iden-
group I metabotropic glutamate receptors in traumatic spinal tification of lamina V and VII interneurons presynaptic to
cord white matter injury. J. Neurotrauma, 15: 929–941. adrenal sympathetic preganglionic neurons in rats using a
Bakhit, C., Armanini, M., Wong, W.L.T., Bennett, G.L. and recombinant herpes simplex virus type 1. Neuroscience, 85:
Wrathall, J.R. (1991) Increase in nerve growth factor-like 863–872.
immunoreactivity and decrease in choline acetyltransferase Coote, J.H., Downman, C.B.B. and Weber, W.V. (1969) Reflex
following contusive spinal cord injury. Brain Res., 554: discharges into thoracic white rami elicited by somatic and
264–271. visceral afferent excitation. J. Physiol., 202: 147–159.
Bandtlow, C.E., Meyer, M., Lindholm, D., Spranger, M., Corbett, J.L., Debarge, O., Frankel, H.L. and Mathias, C.
Heumann, R. and Thoenen, H. (1990) Regional and cellular (1975) Cardiovascular responses in tetraplegic man to muscle
codistribution of interleukin 1 beta and nerve growth factor spasm, bladder percussion and head-up tilt. Clin. Exp.
mRNA in the adult rat brain: possible relationship to the Pharmacol. Physiol., Suppl., 2: 189–193.
regulation of nerve growth factor synthesis. J. Cell Biol., 111: Dembowsky, K., Czachurski, J., Amendt, K. and Seller, H.
1701–1711. (1978) Tonic, supraspinal, monoaminergic inhibition on spi-
Bao, F., Chen, Y., Dekaban, G.A. and Weaver, L.C. (2004a) nal somato-sympathetic reflexes. Pflugers Arch., 373: R76.
An anti-CD11d integrin antibody reduces cyclooxygenase-2 Dembowsky, K., Czachurski, J., Amendt, K. and Seller, H.
expression and protein and DNA oxidation after spinal cord (1980) Tonic descending inhibition of the spinal somato-
injury in rats. J. Neurochem., 90: 1194–1204. sympathetic reflex from the lower brain stem. J. Auton. Nerv.
Bao, F., Chen, Y., Dekaban, G.A. and Weaver, L.C. (2004b) Syst., 2: 157–182.
Early anti-inflammatory treatment reduces lipid peroxidation Erickson, R.P. (1980) Autonomic hyperreflexia: pathophysiol-
and protein nitration after spinal cord injury in rats. ogy and medical management. Arch. Phys. Med. Rehabil.,
J. Neurochem., 88: 1335–1344. 61: 431–440.
Bethea, J.R. and Dietrich, W.D. (2002) Targeting the host in- Fehlings, M.G., Tator, C.H. and Linden, R.D. (1989) The ef-
flammatory response in traumatic spinal cord injury. Curr. fect of nimodipine and dextran on axonal function and blood
Opin. Neurol., 15(3): 355–360. flow following experimental spinal cord injury. J. Neurosurg.,
Bevilacqua, M.P. (1993) Endothelial-leukocyte adhesion mol- 71: 403–416.
ecules. Ann. Rev. Immunol., 11: 767–804. Giannantoni, A., Di, S.S., Scivoletto, G., Mollo, A., Silecchia,
Blackmer, J. (2003) Rehabilitation medicine: 1. Autonomic A., Fuoco, U. and Vespasiani, G. (1998) Autonomic
dysreflexia. CMAJ, 169: 931–935. dysreflexia during urodynamics. Spinal Cord, 36: 756–760.
Blight, A.R. (1985) Delayed demyelination and macrophage Grayson, M.H., Van der Vieren, M., Sterbinsky, S.A., Gallantin,
invasion: a candidate for ‘‘secondary’’ cell damage in spinal W.M., Hoffman, P., Staunton, D. and Bochner, B.S. (1999)
cord injury. Central Nervous System Trauma, 2: 299–315. alphadbeta2 integrin is a ligand for vascular cell adhesion
Brown, A., Ricci, M.-J. and Weaver, L.C. (2004) NGF message molecule-1. Int. Arch. Allergy Immunol., 118(2–4): 263–264.
and protein distribution in the injured rat spinal cord. Exp. Gris, D., Marsh, D.R., Oatway, M.A., Chen, Y., Hamilton,
Neurol., 188: 115–127. E.F., Dekaban, G.A. and Weaver, L.C. (2004) Transient
Bullock, E.D. and Johnson, E.M. (1996) Nerve growth factor blockade of the CD11d/CD18 integrin reduces secondary
induces the expression of certain cytokine genes and bcl-2 in damage after spinal cord injury, improving sensory, auto-
mast cells. Potential role in survival promotion. J. Biol. nomic, and motor function. J. Neurosci., 24: 4043–4051.
Chem., 271: 27500–27508. Gris, D., Marsh, D.R., Dekaban, G.A. and Weaver, L.C.
Cabot, J., Alessi, V., Carroll, J. and Ligorio, M. (1994) Spinal (2005). Comparison of methylprednisolone and anti-CD11d
cord lamina V and lamina VII interneuronal projections to antibody treatments on autonomic dysreflexia after spinal
sympathetic preganglionic neurons. J. Comp. Neurol., 347: cord injury. Exp. Neurol.
515–530. Guttman, F.L. and Whitteridge, D. (1947) Effects of bladder
Calaresu, F.R. and Yardley, C.P. (1988) Medullary basal sym- distention on autonomic mechanisms after spinal cord inju-
pathetic tone. Ann. Rev. Physiol., 50: 511–524. ries. Brain, 70: 361–404.
261

Hayashi, M., Ueyama, T., Nemoto, K., Tamaki, T. and Koyanagi, I., Tator, C.H. and Lea, P.J. (1993a) Three-dimen-
Senba, E. (2000) Sequential mRNA expression for sional analysis of the vascular system in the rat spinal cord
immediate early genes, cytokines, and neurotrophins in with scanning electron microscopy of vascular corrosion
spinal cord injury. J. Neurotrauma, 17: 203–218. casts. Part 2: Acute spinal cord injury. Neurosurgery, 33:
Heese, K., Fiebich, B.L., Bauer, J. and Otten, U. (1998a) 285–291.
NF-kappaB modulates lipopolysaccharide-induced micro- Koyanagi, I., Tator, C.H. and Theriault, E. (1993b) Silicone
glial nerve growth factor expression. Glia, 22: 401–407. rubber microangiography of acute spinal cord injury in the
Heese, K., Hock, C. and Otten, U. (1998b) Inflammatory sig- rat. Neurosurgery, 32: 260–268.
nals induce neurotrophin expression in human microglial Krassioukov, A.V., Bunge, M.B., Puckett, W.R. and Bygrave,
cells. J. Neurochem., 70: 699–707. M.A. (1999) The changes in human spinal sympathetic
Heumann, R., Korsching, S., Bandtlow, C. and Thoenen, H. preganglionic neurons after spinal cord injury. Spinal Cord,
(1987a) Changes of nerve growth factor synthesis in non- 37: 6–13.
neuronal cells in response to sciatic nerve transection. J. Cell Krassioukov, A.V., Furlan, J.C. and Fehlings, M.G. (2003)
Biol., 104: 1623–1631. Autonomic dysreflexia in acute spinal cord injury: an under-
Heumann, R., Lindholm, D., Bandtlow, C., Meyer, M., recognized clinical entity. J. Neurotrauma, 20: 707–716.
Radeke, M.J., Misko, T.P., Shooter, E. and Thoenen, H. Krassioukov, A.V., Johns, D.G. and Schramm, L.P. (2002)
(1987b) Differential regulation of mRNA encoding nerve Sensitivity of sympathetically correlated spinal interneurons,
growth factor and its receptor in rat sciatic nerve during renal sympathetic nerve activity, and arterial pressure to so-
development, degeneration, and regeneration: role of matic and visceral stimuli after chronic spinal injury.
macrophages. Proc. Natl. Acad. Sci. USA, 84: 8735–8739. J. Neurotrauma, 19: 1521–1529.
Jacob, J.E., Gris, P., Fehlings, M.G., Weaver, L.C. and Brown, Krassioukov, A.V. and Weaver, L.C. (1995) Episodic
A. (2003) Autonomic dysreflexia after spinal cord transection hypertension due to autonomic dysreflexia in acute and
and compression in 129Sv, C57BL and wallerian degenera- chronic spinal cord-injured rats. Am. J. Physiol., 268:
tion slow mutant mice. Exp. Neurol., 183: 136–146. H2077–H2083.
Jacob, J.E., Pniak, A., Weaver, L.C. and Brown, A. (2001) Krassioukov, A.V. and Weaver, L.C. (1996) Morphological
Autonomic dysreflexia in a mouse model of spinal cord changes in sympathetic preganglionic neurons after spinal
injury. Neuroscience, 108: 687–693. cord injury in rats. Neuroscience, 70: 211–226.
Jones, L.L., Yamaguchi, Y., Stallcup, W.B. and Tuszynski, Krenz, N.R., Meakin, S.O., Krassioukov, A.V. and Weaver,
M.H. (2002) NG2 is a major chondroitin sulfate proteogly- L.C. (1999) Neutralizing intraspinal nerve growth factor
can produced after spinal cord injury and is expressed by blocks autonomic dysreflexia caused by spinal cord injury.
macrophages and oligodendrocyte progenitors. J. Neurosci., J. Neurosci., 19(17): 7405–7414.
22: 2792–2803. Krenz, N.R. and Weaver, L.C. (1998a) Changes in the mor-
Joshi, S., LeVatte, M.A., Dekaban, G.A. and Weaver, L.C. (1995) phology of sympathetic preganglionic neurons parallel the
Identification of spinal interneurons antecedent to adrenal development of autonomic dysreflexia after spinal cord injury
sympathetic preganglionic neurons using trans-synaptic trans- in rats. Neurosci. Lett., 243: 61–64.
port of herpes simplex virus type 1. Neuroscience, 65: 893–903. Krenz, N.R. and Weaver, L.C. (1998b) Sprouting of primary
Ju, G., Hokfelt, T., Brodin, E., Fahrenkrug, J., Fischer, J.A., afferent fibers after spinal cord transection in the rat.
Frey, P., Elde, R.P. and Brown, J.C. (1987) Primary sensory Neuroscience, 85: 443–458.
neurons of the rat showing calcitonin gene-related peptide Krenz, N.R. and Weaver, L.C. (2000) Nerve growth factor in
immunoreactivity and their relation to substance P-, glia and inflammatory cells of the injured rat spinal cord.
somatostatin-, galanin-, vasoactive intestinal polypeptide- J. Neurochem., 74: 730–739.
and cholecystokinin-immunoreactive ganglion cells. Cell Landrum, L.M., Thompson, G.M. and Blair, R.W. (1998) Does
Tissue Res., 247: 417–431. postsynaptic a1-adrenergic receptor supersensitivity contrib-
Karlsson, A.-K. (1999) Autonomic dysreflexia. Spinal Cord, 37: ute to autonomic dysreflexia? Am. J. Physiol., 274:
383–391. 1090–1098.
Karlsson, A.-K., Friden, P.M., Lonnroth, P., Sullivan, L. and LaSala, A., Corinti, S., Federici, M., Saragovi, H.U. and
Elam, M. (1998) Regional sympathetic function in high Girolomoni, G. (2000) Ligand activation of nerve growth
spinal cord injury during mental stress and autonomic factor receptor TrkA protects monocytes from apoptosis.
dysreflexia. Brain, 121: 1711–1719. J. Leukoc. Biol., 68: 104–110.
Koizumi, K. and Brooks, C.M. (1972) The integration of Lawson, S.N., McCarthy, P.W. and Prabhakar, E. (1996)
autonomic system reactions: a discussion of autonomic Electrophysiological properties of neurones with CGRP-like
reflexes, their control and their association with somatic immunoreactivity in rat dorsal root ganglia. J. Comp. Ne-
reflexes. In: Adrian R.H., Helmrich E., Holzer H., Jung R., urol., 365: 355–366.
Kramer K., Krayer O., Lynen F., Miescher P.A., Rasmussen Lawson, S.N., Perry, M.J., Prabhakar, E. and McCarthy, P.W.
H., Renold A.E., Trendelenburg U., Ullrich K., Vogt W. and (1993) Primary sensory neurones: neurofilament,
Weber A. (Eds.), Reviews of Physiology: Biochemistry and neuropeptides, and conduction velocity. Brain Res. Bull.,
Experimental Pharmacology. Springer, Berlin, pp. 1–68. 30: 239–243.
262

Lee, B.Y., Karmakar, M.G., Herz, B.L. and Sturgill, R.A. Mayorov, D.N., Adams, M.A. and Krassioukov, A.V. (2001)
(1994) Autonomic dysreflexia revisited. J. Spinal Cord Med., Telemetric blood pressure monitoring in conscious rats be-
18: 75–87. fore and after compression injury of spinal cord. J. Neuro-
Leskovar, A., Moriarty, L.J., Turek, J.J., Schoenlein, I.A. and trauma, 18: 727–736.
Borgens, R.B. (2000) The macrophage in acute neural injury: McCarthy, P.W. and Lawson, S.N. (1990) Cell type and con-
changes in cell numbers over time and levels of cytokine duction velocity of rat primary sensory neurons with ca-
production in mammalian central and peripheral nervous lcitonin gene-related peptide-like immunoreactivity.
systems. J. Exp. Biol., 203(Part 12): 1783–1795. Neuroscience, 34: 623–632.
Lindan, R., Joiner, E., Freehafer, A.A. and Hazel, C. (1980) Naftchi, N.E. (1990) Mechanism of autonomic dysreflexia;
Incidence and clinical features of autonomic dysreflexia in contributions of cathecholamine and peptide neurotransmit-
patients with spinal cord injury. Paraplegia, 18: 285–292. ters. Ann. NY Acad. Sci., 579: 133–148.
Lindholm, D., Heumann, R., Meyer, M. and Thoenen, H. (1987) Naftchi, N.E. and Richardson, J.S. (1997) Autonomic dysreflexia:
Interleukin-1 regulates synthesis of nerve growth factor in pharmacological management of hypertensive crises in spinal
non-neuronal cells of rat sciatic nerve. Nature, 330: 658–659. cord injured patients. J. Spinal. Cord. Med., 20: 355–360.
Llewellyn-Smith, I.J. and Weaver, L.C. (2001) Changes in Neish, A.S., Read, M.A., Thanos, D., Pine, R., Maniatis, T.
synaptic inputs to sympathetic preganglionic neurons after and Collins, T. (1995) Endothelial interferon regulatory fac-
spinal cord injury. J. Comp. Neurol., 435: 226–240. tor 1 cooperates with NF-kappa B as a transcriptional ac-
Mabon, P.J., Weaver, L.C. and Dekaban, G.A. (2000) Inhibi- tivator of vascular cell adhesion molecule 1. Mol. Cell Biol.,
tion of monocyte/macrophage migration to a spinal cord in- 15(5): 2558–2569.
jury site by an antibody to the integrin alphaD: a potential Oatway, M.A., Chen, Y., Bruce, J.C., Dekaban, G.A. and
new anti-inflammatory treatment. Exp. Neurol., 166: 52–64. Weaver, L.C. (2005) An anti-CD11d integrin antibody treat-
Maiorov, D.N., Fehlings, M.G. and Krassioukov, A.V. (1998) ment restores normal serotonergic projections to the dorsal,
Relationship between severity of spinal cord injury and ab- intermediate and ventral horns of the injured spinal cord.
normalities in neurogenic cardiovascular control in conscious J. Neurosci., 25: 637–647.
rats. J. Neurotrauma, 15(5): 365–374. Osborn, J.W., Taylor, R.F. and Schramm, L.P. (1990) Chronic
Maiorov, D.N., Krenz, N.R., Krassioukov, A.V. and Weaver, cervical spinal cord injury and autonomic hyperreflexia in
L.C. (1997a) Role of spinal NMDA and AMPA receptors in rats. Am. J. Physiol., 258: R169–R174.
episodic hypertension in conscious spinal rats. Am. J. Popovich, P.G., Guan, Z., Wei, P., Huitinga, I., van Rooijen,
Physiol., 273: H1266–H1274. N. and Stokes, B.T. (1999) Depletion of hematogenous ma-
Maiorov, D.N., Weaver, L.C. and Krassioukov, A.V. (1997b) crophages promotes partial hindlimb recovery and neuroan-
Relationship between sympathetic activity and arterial pres- atomical repair after experimental spinal cord injury. Exp.
sure in conscious spinal rats. Am. J. Physiol., 272: Neurol., 158: 351–365.
H625–H631. Popovich, P.G., Wei, P. and Stokes, B.T. (1997) Cellular in-
Marsh, D.R. and Weaver, L.C. (2004) Autonomic dysreflexia, flammatory response after spinal cord injury in Sprague–Da-
induced by noxious or innocuous stimulation, does not de- wley and Lewis rats. J. Comp. Neurol., 377: 443–464.
pend on changes in dorsal horn substance P. J. Neurotrauma, Price, J. (1985) An immunohistochemical and quantitative ex-
21: 817–828. amination of dorsal root ganglion neuronal subpopulations.
Marsh, D.R., Wong, S.T., Meakin, S.O., MacDonald, J.I.S., J. Neurosci., 5(8): 2051–2059.
Hamilton, E.F. and Weaver, L.C. (2002) Neutralizing intra- Rodenbaugh, D.W., Collins, H.L. and DiCarlo, S.E. (2003)
spinal NGF with a trkA-IgG fusion protein blocks develop- Increased susceptibility to ventricular arrhythmias in hyper-
ment of autonomic dysreflexia in a clip-compression model of tensive paraplegic rats. Clin. Exp. Hypertens., 25: 349–358.
spinal cord injury. J. Neurotrauma, 19: 1531–1542. Saporito, M.S., Wilcox, H.M., Hartpence, K.C., Lewis, M.E.,
Mathias, C.J. and Frankel, H.L. (1992a) Autonomic distur- Vaught, J.L. and Carswell, S. (1993) Pharmacological induc-
bances in spinal cord lesions. In: Mathias C.J. and Bannister tion of nerve growth factor mRNA in adult rat brain. Exp.
R. (Eds.) Autonomic Failure A Textbook of Clinical Disor- Neurol., 123: 295–302.
ders of the Autonomic Nervous System, Vol. 4(51). Oxford Saragovi, H.U. and Gehring, K. (2000) Development of phar-
University Press, Oxford, pp. 494–513. macological agents for targeting neurotrophins and their re-
Mathias, C.J. and Frankel, H.L. (1992b) Autonomic distur- ceptors. Trends Pharmacol. Sci., 21: 93–98.
bances in spinal cord lesions. In: Bannister R. and Mathias Sato, A. and Schmidt, R.F. (1973) Somatosympathetic reflexes:
C.J. (Eds.) Autonomic Failure, a Textbook of Clinical Dis- afferent fibers, central pathways, discharge characteristics.
orders of the Autonomic Nervous System, Vol. 3(43). Oxford Physiol. Rev., 53(4): 916–947.
Medical Publications, Oxford, UK, pp. 839–881. Saville, L.R., Pospisil, C.H., Mawhinney, L.A., Bao, F.,
Mathias, C.J. and Frankel, H.L. (1992c) The cardiovascular Simedria, F.C., Peters, A.A., O’Connell, P.J., Weaver, L.C.
system in tetraplegia and paraplegia. In: Frankel H.L. (Ed.) and Dekaban, G.A. (2004) A monoclonal antibody to
Handbook of Clinical Neurology, Vol. 17(25). Elsevier CD11d reduces the inflammatory infiltrate into the injured
Science Publishers, B.V., Amsterdam, The Netherlands, spinal cord: a potential neuroprotective treatment. J. Ne-
pp. 435–456. uroimmunol., 156: 42–57.
263

Schumacher, P.A., Eubanks, J.H. and Fehlings, M.G. (1999) W.M. and Staunton, D.E. (1999) The leukocyte integrin
Increased calpain I-mediated proteolysis and preferential loss aDb2 binds VCAM-1: evidence for a binding interface be-
of dephosphorylated NF200, following traumatic spinal cord tween I domain and VCAM-1. J. Immunol., 163: 1984–1990.
injury. Neuroscience, 91: 733–744. Vizzard, M.A., Brisson, M. and De Groat, W.C. (2000)
Shanley, T.P., Warner, R.L., Crouch, L.D., Dietsch, G.N., Transneuronal labeling of neurons in the adult rat central
Clark, D.L., O’Brien, M.M., Gallatin, W.M. and Ward, P.A. nervous system following inoculation of pseudorabies virus
(1998) Requirements for ad in IgG immune complex-induced into the colon. Cell Tissue Res., 299(1): 9–26.
rat lung injury. J. Immunol., 160: 1014–1020. Wallace, M.C. and Tator, C.H. (1986) Spinal cord blood flow
Shea, J.D., Gioffre, R., Carrion, H. and Small, M.P. (1973) measured with microspheres following spinal cord injury in
Autonomic hyperreflexia in spinal cord injury. Southern the rat. Can. J. Neurol. Sci., 13: 91–96.
Med. J., 66(8): 869–872. Wallace, M.C., Tator, C.H. and Frazee, P. (1986) Relationship
Spranger, M., Lindholm, D., Bandtlow, C., Heumann, R., between posttraumatic ischemia and hemorrhage in the in-
Gnahn, H., Naher-Noe, M. and Thoenen, H. (1990) Regu- jured rat spinal cord as shown by colloidal carbon angio-
lation of nerve growth factor (NGF) synthesis in the rat graphy. Neurosurgery, 18: 433–439.
central nervous system: comparison between the effects of Weaver, L.C., Cassam, A.K., Krassioukov, A.V. and
interleukin-1 and various growth factors in astrocyte cultures Llewellyn-Smith, I.J. (1997) Changes in immunoreactivity
and in vivo. Eur. J. Neurosci., 2: 69–76. for growth associated protein-43 suggest reorganization of
Streit, W.J., Semple-Rowland, S.L., Hurley, S.D., Miller, R.C., synapses on spinal sympathetic neurons after cord transec-
Popovich, P.G. and Stokes, B.T. (1998) Cytokine mRNA tion. Neuroscience, 81: 535–551.
profiles in contused spinal cord and axotomized facial nu- Weaver, L.C., Marsh, D.R., Gris, D., Meakin, S.O. and
cleus suggest a beneficial role for inflammation and gliosis. Dekaban, G.A. (2001a) Central mechanisms for autonomic
Exp. Neurol., 152: 74–87. dysreflexia after spinal cord injury. In: McKerracher L.,
Taoka, Y. and Okajima, K. (1998) Spinal cord injury in the rat. Douchet D. and Rossignol S. (Eds.) Progress in Brain Re-
Prog. Neurobiol., 56: 341–358. search, Vol. 7. Elsevier Science B.V., Amsterdam, pp. 83–95.
Taoka, Y., Okajima, K., Uchiba, M., Murakami, K., Weaver, L.C., Verghese, P., Bruce, J.C., Fehlings, M.G., Krenz,
Kushimoto, S., Johno, M., Naruo, M., Okabe, H. and N.R. and Marsh, D.R. (2001b) Autonomic dysreflexia and
Takatsuki, K. (1997) Role of neutrophils in spinal cord injury primary afferent sprouting after clip-compression injury of
in the rat. Neuroscience, 79(4): 1177–1182. the rat spinal cord. J. Neurotrauma, 18: 1107–1119.
Tator, C.H. and Fehlings, M.G. (1991) Review of the secondary Wong, S.T., Atkinson, B.A. and Weaver, L.C. (2000) Confocal
injury theory of acute spinal cord trauma with emphasis on microscopic analysis of sprouting small-diameter primary af-
vascular mechanisms. J. Neurosurg., 75: 15–26. ferent fibres after spinal cord injury. Neurosci. Lett., 296:
Vaidyanathan, S., Soni, B.M., Sett, P., Watt, J.W.H., Oo, T. 65–68.
and Bingley, J. (1998) Pathophysiology of autonomic Yeoh, M., McLachlan, E.M. and Brock, J.A. (2004) Chronic
dysreflexia: long-term treatment with terazosin in adult and decentralization potentiates neurovascular transmission in
paediatric spinal cord injury patients manifesting recurrent the isolated rat tail artery, mimicking the effects of spinal
dysreflexic epidsodes. Spinal Cord, 36: 761–770. transection. J. Physiol., 561: 583–596.
Van der Vieren, M., Crowe, D.T., Hoekstra, D., Vazeux, R., Young, W. (1993) Secondary injury mechanisms in acute spinal
Hoffman, P.A., Grayson, M.H., Bochner, B.S., Gallatin, cord injury. J. Emerg. Med., 11: 13–22.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

Chapter 17

Segmental organization of spinal reflexes mediating


autonomic dysreflexia after spinal cord injury

Alexander G. Rabchevsky

University of Kentucky, Spinal Cord & Brain Injury Research Center and Department of Physiology, 741 South Limestone
Street, B371 BBSRB, Lexington, KY 40536-0509, USA

Abstract: Spinal cord injuries above mid-thoracic levels can lead to a potentially life-threatening hyper-
tensive condition termed autonomic dysreflexia that is often triggered by distension of pelvic viscera
(bladder or bowel). This syndrome is characterized by episodic hypertension due to sudden, massive
discharge of sympathetic preganglionic neurons in the thoracolumbar spinal cord. This hypertension is
usually accompanied by bradycardia, particularly if the injury is caudal to the 2nd to 4th thoracic spinal
segments. The development of autonomic dysreflexia is correlated with aberrant sprouting of peptidergic
afferent fibers into the spinal cord below the injury. In particular, sprouting of nerve growth factor-
responsive afferent fibers has been shown to have a major influence on dysreflexia, perhaps by amplifying
the activation of disinhibited sympathetic neurons. Using a model of noxious bowel distension after com-
plete thoracic spinal transection at the 4th thoracic segment in rats, we selectively altered C-fiber sprouting,
at specified spinal levels caudal to the injury, with microinjections of adenovirus encoding the growth-
promoting nerve growth factor or the growth-inhibitory semaphorin 3A. This was followed by assessment
of physiological responses to colorectal distension and subsequent histology. Additionally, anterograde
tract tracers were injected into the lumbosacral region to compare the extent of labeled propriospinal
rostral projections in uninjured cords to those in cords after complete 4th thoracic transection. In summary,
overexpression of chemorepulsive semaphorin 3A impeded C-fiber sprouting in lumbosacral segments and
mitigated hypertensive autonomic dysreflexia, whereas the opposite results were obtained with nerve
growth factor overexpression. Furthermore, compared to naı̈ve rats, there were significantly more labeled
lumbosacral propriospinal projections rostrally after thoracic injury. Collectively, our findings suggest that
distension of pelvic viscera increases the excitation of expanded afferent terminals in the disinhibited
lumbosacral spinal cord. This, in turn, triggers excitation and sprouting of local propriospinal neurons to
relay visceral sensory stimuli and amplify the activation of sympathetic preganglionic neurons in the
thoracolumbar cord, to enhance transmission in the spinal viscero-sympathetic reflex pathway. These
responses are manifested as autonomic dysreflexia.

Autonomic dysreflexia is a clinical syndrome that after complete injuries (Snow et al., 1978). The
develops following spinal cord injury above the condition is commonly triggered by distension of
sixth thoracic (T) spinal segment (T6). It is present the pelvic viscera (bowel and bladder) and is man-
after complete as well as incomplete injuries ifested by often debilitating hypertension and
(Karlsson, 1999) with an incidence of up to 70% sweating, dizziness, nausea, and often severe head-
aches (Snow et al., 1978; Lindan et al., 1980;
Corresponding author. Tel.: +(859) 323-0267; Harati, 1997; Karlsson, 1999). A reflex brad-
Fax: +(859) 257-5737; E-mail: AGRab@uky.edu ycardia often accompanies these episodes of

DOI: 10.1016/S0079-6123(05)52017-X 265


266

hypertension, particularly if the injury is caudal to 2004) and the subsequent sprouting of calcitonin
the spinal segments providing sympathetic control gene-related peptide (CGRP)+ afferent fibers in
of the heart (T1–T4). During distension of the the thoracolumbar spinal cord (Krenz and Weav-
bladder or bowel, the consequent afferent barrage er, 1998a; Krenz et al., 1999; Weaver et al., 2001;
into the dorsal horn of the lumbosacral spinal cord Marsh et al., 2002). It is important to note that
ultimately results in massive sympathetic reflex CGRP immunoreactivity has been reported in Ab,
discharge below the level of the injury. This results Ad, and C fiber afferent projections (McCarthy
in vasoconstriction of the muscular, splanchnic, and Lawson, 1990; Lawson et al., 1993; Lawson
and cutaneous vascular beds (reviewed in Karlsson, et al., 1996). Because these CGRP+ fibers also can
1999). The resultant paroxysmal hypertension pro- be labeled with various neurotransmitter markers,
duces a baroreceptor-mediated reflex bradycardia the identity of specific afferent fiber populations
accompanied by withdrawal of sympathetic activ- that sprout after spinal cord injury remains
ity above the lesion level with resultant vasodila- uncertain. For example, it is reported that sub-
tation that produces adverse symptoms such as populations of CGRP+ fibers sprouting distal to
headaches and skin flushing. spinal cord injury sites are nociceptive primary af-
The dysreflexic hypertension stems, in part, ferents. This could be the anatomical substrate for
from injury-induced loss of tonic control of sym- the development and maintenance of chronic pain
pathetic preganglionic neurons in the intermedio- syndromes after spinal cord injury (Ondarza et al.,
lateral cell column of the thoracolumbar spinal 2003). Conversely, there is evidence for a lack of
cord by medullo-spinal neurons in the rostral and significant substance P+ fiber sprouting in parallel
caudal ventrolateral medulla (Finestone and Teasell, with increased CGRP+ fiber sprouting in a rat
1993; Zagon and Smith, 1993). Anatomical and model of autonomic dysreflexia (Marsh and Weav-
physiological changes that occur in sympathetic er, 2004). Since substance P content can be used to
preganglionic neurons and sympathetically related distinguish subpopulations of unmyelinated C
interneurons as descending inputs degenerate fibers from myelinated Ad fibers, the authors
following high thoracic spinal cord transection suggest that sprouting of Ab and Ad fibers, and
have been the subject of considerable attention not C fibers, contributed to increased sympathetic
(Weaver et al., 1997; Krenz and Weaver, 1998b; outflow after injury. Accordingly, non-noxious
Klimaschewski, 2001). Additionally, the influence cutaneous and colonic stimulation below the
of injury-induced sprouting of primary afferent injury was also reported to elicit dysreflexic
fibers into the thoracolumbar spinal cord has been hypertension, albeit to a lesser degree than with
the focus of investigations (Krenz and Weaver, noxious stimulation.
1998a; Weaver et al., 2001). Although autonomic To investigate further the role of nerve growth
dyreflexia can be elicited by even non-noxious factor and CGRP+ afferent fiber sprouting in the
stimuli below the injury level (Marsh and Weaver, development of autonomic dysreflexia, we precise-
2004), a model for consistently inducing autonom- ly manipulated injury-induced CGRP+ C-fiber
ic dysreflexia employs colorectal distension in spi- sprouting in the dorsal horns with bilateral
nal-injured rats. This model is designed to mimic microinjections of a well-characterized replica-
the common clinical manifestation of noxious fecal tion-defective, temperature-sensitive recombinant
impaction (Krassioukov and Weaver, 1995). We adenovirus encoding growth-promoting nerve
have used this preparation to investigate the growth factor (Romero et al., 2001). The princi-
undefined relationships among visceral sensory pal goal was to target growth factor overexpres-
afferents, lumbosacral relay neurons, and sympa- sion to selected regions of the dorsal gray matter of
thetic preganglionic neurons that trigger dysre- the spinal cord, caudal to a complete T4 transec-
flexic hypertension. tion, to identify sites instrumental to augmenting
A contributing factor underlying autonomic dysreflexic responses to colorectal distension 2
dysreflexia is the injury-induced elevation in spi- weeks after injury (Maiorov et al., 1998). Once the
nal levels of nerve growth factor (Brown et al., critical spinal levels were identified, with subsequent
267

immunohistochemical analyses for CGRP+ affer- of CGRP+ primary afferent fibers into the
ent fiber sprouting (Krenz et al., 1999; Weaver mid-thoracic spinal cord dorsal horns compared
et al., 2001), we used recombinant adenovirus to to control injured rats injected with green fluores-
overexpress C-fiber growth-inhibitory semaphorin cent protein adenovirus at T5/T6 (n ¼ 3). Notably,
3A (Tang et al., 2004) after injury in an attempt to we found that sympathetic preganglionic neurons,
prevent sprouting and mitigate dysreflexic hyper- pre-labeled with FluoroGold (Anderson and
tension. Regarding the aforementioned variability Edwards, 1994), were completely surrounded by
in phenotypes of post-traumatic sprouting afferent sprouting CGRP+ fibers (not shown). Physiolog-
fibers, Tang et al. (2004) showed that overexpres- ical recordings of mean arterial blood pressure and
sion of nerve growth factor within the spinal cord heart rate in response to colorectal distension
directly increased sprouting of CGRP+ axons showed that evoked hypertension was no greater
without altering neuropeptide expression or compared to injured controls (Fig. 1), even though
sprouting of other sensory axon populations. nerve growth factor induced robust afferent fiber
These populations included myelinated and un- sprouting into the intermediolateral cell column.
myelinated, glial cell line-derived neurotrophic Since this implied that neither the actions of nerve
factor-responsive subpopulations of nociceptive growth factor nor sprouting CGRP+ fibers directly
axons. Moreover, overexpression of semaphorin influenced the sympathetic discharge in response to
3A, a repulsive guidance molecule for nerve growth colorectal distension, we chose to similarly manip-
factor-responsive C-fibers, reduced sprouting of ulate injury-induced sprouting at more caudal levels,
CGRP+ and substance P+ axons compared with based on the evidence that sensory input from the
overexpression of control green fluorescent protein. pelvic viscera enter the spinal cord caudal to T5/T6.
The sympathetic component of the intermedio- Electrophysiological (Al-Chaer et al., 1997) and
lateral cell column extends from T1 to L2, and anatomical studies (Birder et al., 1991; Keast and
primary afferent sprouting has been reported both De Groat, 1992; Vizzard, 2000) report that the
close to and distant from a T4 transection (Krenz primary afferents supplying the pelvic viscera
and Weaver, 1998a; Krenz et al., 1999). We first (bladder, distal colon, rectum) in rats run in the
tested whether increasing primary afferent sprout- pelvic nerve and distribute mainly to the L6 and S1
ing just caudal to at T4 transection would augment dorsal root ganglia and spinal cord segments. A
hypertension evoked by colorectal distension smaller percentage of afferents run in the hypogas-
(Cameron et al., 2004). Two weeks after we over- tric nerve to the T13 and L1 dorsal root ganglia and
expressed nerve growth factor in the T5/T6 dorsal spinal cord segments, and neurons responsive to
horns (n ¼ 5), there was copious central sprouting colorectal distension are located in the superficial

Fig. 1. Overexpression of nerve growth factor (NGF) in lumbosacral spinal levels augments CGRP+ afferent fiber sprouting 2 weeks
post-injury. Photomicrographs showing CGRP+ staining in S1 spinal segments injected with adenovirus expressing control green
fluorescent protein (GFP Adts; left column) or NGF Adts (right column). Note the robust CGRP+ afferent fiber sprouting throughout
the S1 injection site in response to NGF overexpression compared to GFP controls. Scale bars ¼ 100 mm.
268

dorsal horns of the T13–L2 spinal segments (Ness primarily by visceral afferent activation of prop-
and Gebhart, 1988, 1989). Within the L6/S1 spinal riospinal interneurons that project from the
regions, however, lamina X and V–VIII contain lumbosacral to the thoracic spinal cord.
most of the neurons responding to visceral stim- Therefore, we injected T4 spinal-transected rats
ulation (Al-Chaer et al., 1997). These projection with adenovirus encoding nerve growth factor into
neurons respond physiologically to colorectal dis- the T13/L1 (n ¼ 8) or L6/S1 (n ¼ 9) segments ver-
tension (Martinez et al., 1998; Landrum et al., sus green fluorescent protein controls (n ¼ 7 per
2002), as well as to cutaneous stimulation. level) to augment sprouting specifically and, po-
Two groups of lumbosacral spinal interneurons tentially, to augment dysreflexic hypertension
relay visceral information rostrally to supraspinal (Cameron et al., 2004). After 2 weeks of nerve
targets, each through a separate pathway. One growth factor overexpression within either caudal
group of neurons resides primarily in the dorsal spinal level, hypertension induced by colorectal
commissure of L6/S1 segments (Willis et al., 1999; distension was greater in magnitude (35 mmHg)
Vizzard et al., 2000) and projects axons in the me- than in green fluorescent protein-injected injured
dial part of the dorsal columns (Hirshberg et al., controls (20 mmHg) (Fig. 1). Conversely, L6/S1
1996; Wang et al., 1999). Most of these neurons overexpression of semaphorin 3A, a chemorepul-
respond physiologically to colorectal distension sive factor for nerve growth factor-responsive pri-
and their axons terminate in the medullary gracile mary afferent fibers, led to a marked reduction in
nucleus (Al-Chaer et al., 1996, 1999). The other evoked hypertension compared to cord-injured
group of neurons lies in both the dorsal commis- controls (10 mmHg) (not shown).
sure and lateral parasympathetic nucleus and their Subsequent histological processing of the lum-
axons project through the ventrolateral white mat- bosacral spinal cord confirmed that overexpression
ter to the thalamus (Ness and Gebhart, 1987). of nerve growth factor in the S1 segment elicited
These neurons are activated by colorectal disten- profuse sprouting of CGRP+ fibers 2 weeks after
sion as well as somatic (cutaneous) stimulation. T4 cord transection compared to modest sprouting
Propriospinal neurons connect lamina X/dorsal in response to control green fluorescent protein
commissure at multiple rostro-caudal levels of the overexpression (Fig. 2). In contrast, overexpres-
cord (Petkó and Antal, 2000) and injections of sion of semaphorin 3A in the S1 segment caused
anterograde tract tracers into lamina X/dorsal diminished sprouting compared to controls (not
commissure in the lumbosacral segments (L6–S2) shown). Linear regression analysis of percent
labels terminals in lamina X throughout the cord CGRP+ fiber area covered in the dorsal horns
up to cervical levels (Matsushita, 1998). These plotted against reflex-induced hypertensive chang-
studies show that long-ranging propriospinal neu- es revealed a positive correlation between the ex-
rons exist in the dorsal commissure/deep dorsal tent of C-fiber sprouting into the lumbosacral
horn of the lumbosacral spinal cord. Such relays spinal level and the severity of autonomic dysre-
extending the entire length of the cord could po- flexia among the three treatment groups (Cameron
tentially send off collaterals after spinal cord in- et al., 2004). This suggests that, in this model of
jury and influence the activity of sympathetic autonomic dysreflexia induced by noxious colo-
preganglionic neurons in the intermediolateral cell rectal distension, nerve growth factor-induced
column. Since the number of activated neurons sprouting of primary afferents in the region where
expressing c-Fos in response to colorectal disten- colonic afferents enter the spinal cord causes ab-
sion is much greater after a chronic spinal cord normal activation of sympathetic preganglionic
transection than after an acute injury (Landrum neurons in the thoracolumbar spinal cord via pu-
et al., 2002), it suggests that larger numbers of tative propriospinal pathways (Matsushita, 1998;
lumbosacral propriospinal neurons can be activat- Petkó and Antal, 2000).
ed by visceral stimuli following injury. In view of These results are consistent with a nerve growth
these findings and our preliminary results, we rea- factor-dependent increase in the number of pri-
soned that autonomic dysreflexia may be mediated mary afferent terminals. This may amplify the
269

Fig. 2. Physiological responses to colorectal distension (CRD) 2 weeks after T4 spinal transection. Illustrative traces of pulsatile
arterial pressure (PAP), mean arterial pressure (MAP), and heart rate (HR) before, during, and after 1 min CRD with rectal balloon
catheter inflation (indicated by arrows) from injured rats with bilateral L6/S1 injections of (A) adenovirus expressing control green
fluorescent protein (GFP Adts) or (B) nerve growth factor (NGF Adts). Note that both injured groups show autonomic dysreflexia
with hypertension accompanied by bradycardia, but the severity of hypertension is almost twofold greater and more prolonged with
nerve growth factor overexpression.

synaptic action of colonic afferents, during colo- 2002). It seems likely that nerve growth factor-
rectal distension, on lumbosacral dorsal horn neu- mediated plasticity of CGRP+ afferents causes
rons that project axons rostrally in the gray matter hyperactivity of lumbosacral propriospinal neu-
and dorsal columns (Wang et al., 1999; Willis rons and, as a result, excitation of sympathetically
et al., 1999) and may connect with, and influence correlated interneurons in the intermediolateral cell
the activity of, preganglionic sympathetic neurons column. In support of this, colorectal distension in
in the intermediolateral cell column (Llewellyn- acute and chronic spinal-transected rats activates
Smith and Weaver, 2001). Intrathecal delivery of sympathetically correlated neurons in the T10 seg-
nerve growth factor antibody following spinal ment, most likely via sacral–thoracic interneurons
cord injury reduces the elevated nerve growth fac- (Chau et al., 2000; Krassioukov et al., 2002).
tor protein levels in L6 and S1 dorsal root ganglia We investigated the potential influence of
and spinal cord of cord-injured rats (Seki et al., intraspinal sprouting of the axons of lumbosacral
270

projection interneurons for activating sympathet- entry of the visceral afferents in the hypogastric
ically correlated interneurons after complete high nerve, alone or in addition to T4 transection, pro-
thoracic spinal cord injury. To do this we injected duced a colorectal distension-induced rise in mean
the anterograde tracer biotinylated dextran amine arterial pressure with the same magnitude as T4-
(BDA) into the S1 dorsal horn of acute T4 spinal- and L5-transected animals (10 mmHg above
transected animals and of uninjured controls baseline) (Cameron et al., 2004). This finding in-
(Cameron et al., 2004). Two weeks later, there dicates that the upper lumbar sympathetic neu-
were conspicuously more labeled ipsilateral (and rons, that are known to constrict hindlimb and
contralateral) propriospinal projections in the ros- visceral blood vessels (Baron et al., 1985; Bahr
tral thoracic gray matter in the injured spinal cords et al., 1986), are not sufficient to induce significant
(Fig. 3). In fact, many of these projections were dysreflexic hypertension. However, activation of
found in proximity to FluoroGold-labeled sympa- the entire sympathetic column after injury does
thetic preganglionic neurons (Fig. 4). Additionally, not appear necessary for eliciting modest hyper-
we found that transection of the L5 spinal cord tension during distension of the pelvic viscera.
segment alone, or in addition to T4, markedly re- Notably, experimental autonomic dysreflexia was
duced colorectal distension-induced hypertension. not completely eliminated by impeding CGRP+
This suggests that relay neurons arising in the L6/ fiber sprouting with semaphorin 3A overexpression,
S1 segments are necessary for increased hyperten- similar to previous findings in which endogenous
sion elicited by pelvic visceral distension. Moreo- nerve growth factor was immunologically neutral-
ver, transection of the T11 cord segment above the ized after complete T4 spinal transection

Fig. 3. Injections of BDA tracer into L6/S1 spinal levels label


more ascending projections after T4 transection. Longitudinal
and horizontal sections at thoracolumbar spinal levels of (A)
non-transected or (B) T4-transected rats approximately 5 mm
rostral to 200 nl injections of BDA into the left L6/S1 dorsal
commissure. After 2 weeks to allow tracer transport and verify Fig. 4. High magnification, dual immunofluorescent images at
physiologically that the injured rats had developed autonomic the mid-thoracic spinal level 2 weeks after T4 transection dem-
dysreflexia, histological processing revealed significantly more onstrate close proximity of (A) FluoroGold-labeled sympathet-
BDA-labeled projections (data not shown) within ipsilateral ic preganglionic neurons (arrows) in the intermediolateral cell
gray matter extending to rostral levels of the thoracic spinal column and (B) BDA-labeled fibers originating from lumbosa-
cord. Scale bar ¼ 0.5 mm. cral projection interneurons. Scale bar ¼ 50 mm.
271

(Krenz et al., 1999; Marsh et al., 2002). Since the interneurons (Fig. 5). We hypothesize that in-
high-affinity nerve growth factor trkA receptor is creased nerve growth factor-mediated sprouting of
located on cholinergic propriospinal neurons in sacral nociceptive C-fibers triggered by complete
the deep dorsal horn of the rat spinal cord thoracic spinal cord transection (de Groat et al.,
(Michael et al., 1997), it is possible that, in addi- 1990) drives larger numbers of propriospinal pro-
tion to altering primary afferent plasticity, spinal jection neurons located in the dorsal intercom-
cord injury provokes both nerve growth factor- missural nucleus and amplifies the information
dependent and -independent reorganization of relayed from colonic afferents during colorectal
propriospinal pathways as well. Such changes distension to lumbosacral dorsal horn neurons.
may provide a neural substrate for the amplifica- These, in turn, convey the signal to rostral sym-
tion of minor sensory signals entering the spinal pathetically correlated neurons (Chau et al., 2000;
cord, resulting in the synchronous discharge of the Tang et al., 2003) that activate the sympathetic
preganglionic sympathetic column. preganglionic neurons to elicit hypertension (Fig.
In our attempts to identify neuronal substrates 5). Since larger numbers of lumbosacral proprio-
for the autonomic hyperactivity after complete spinal neurons are activated by visceral stimuli in
spinal cord injury, we have proposed a model more chronic stages of spinal cord injury (Landrum
(Cameron et al., 2004) which involves the sprout- et al., 2002), this implies that acute functional re-
ing of CGRP+ visceral primary afferent C-fibers organization occurs, which results in signal ampli-
and of the axons of lumbosacral projection fication by spinal circuitry that has presumably

Fig. 5. Schematic illustration depicting the etiology of autonomic dysreflexia evoked by pelvic visceral distension and other stimuli.
Following complete spinal cord injury above the T6 level, sympathetic preganglionic neurons (blue) are released from descending
medullo-spinal control (dashed arrow) and autonomic spinal reflexes are rendered hyperactive. Consequently, pelvic visceral sensory
input (yellow) is relayed by propriospinal neurons (green) projecting from the dorsal gray commissure at the lumbosacral level to
sympathetic preganglionic neurons and/or sympathetically correlated interneurons located in the thoracolumbar cord (blue). Post-
traumatic C-fiber sprouting into the lumbosacral cord (yellow) further amplifies the central signals (green) to elicit hypertension,
ultimately causing profound peripheral vasoconstriction of splanchnic, muscle, and cutaneous vascular beds. Subsequent stimulation
of aortic depressor nerve (ADN) and carotid sinus nerve (CSN) baroreceptor afferents of the petrosal ganglion (red) is conveyed to the
nucleus tractus solitarius that elicits bradycardia via activation of the nucleus ambiguus. Note that flushing is likely the result of
inhibition of skin vasoconstrictor preganglionic neurons above the lesion by the baroreceptor reflex, and not due to the activation of
sympathetic preganglionic neurons below the lesion by the dysreflexia-inducing stimulus.
272

been modified as a result of the injury. As a result, References


following acute thoracic spinal cord transection,
multiple neuronal networks in the thoracolumbar Al-Chaer, E.D., Feng, Y. and Willis, W.D. (1999) Comparative
study of viscerosomatic input onto postsynaptic dorsal
cord generate sympathetic activity upon activation
column and spinothalamic tract neurons in the primate. J.
from as yet unidentified neurons in the L6/S1 seg- Neurophysiol., 82: 1876–1882.
ments of the cord. The ascending terminals of Al-Chaer, E.D., Lawand, N.B., Westlund, K.N. and Willis,
propriospinal neurons may also undergo morpho- W.D. (1996) Pelvic visceral input into the nucleus gracilis is
logic changes including the aberrant convergence largely mediated by the postsynaptic dorsal column pathway.
and formation of synapses with sympathetically J. Neurophysiol., 76: 2675–2690.
Al-Chaer, E.D., Westlund, K.N. and Willis, W.D. (1997) Sen-
correlated neurons or sympathetic preganglionic sitization of postsynaptic dorsal column neuronal responses
neurons in the intermediolateral cell column by colon inflammation. Neuroreport, 8: 3267–3273.
(Weaver et al., 1997; Llewellyn-Smith and Anderson, C.R. and Edwards, S.L. (1994) Intraperitoneal in-
Weaver, 2001). In various ways, the pelvic jections of Fluorogold reliably labels all sympathetic pre-
visceral signal may be conveyed to the ultimate ganglionic neurons in the rat. J. Neurosci. Meth., 53:
137–141.
vasoconstrictor post-ganglionic sympathetic Bahr, R., Bartel, B., Blumberg, H. and Janig, W. (1986)
neurons to induce dysreflexia (Fig. 5). Functional characterization of preganglionic neurons
projecting in the lumbar splanchnic nerves: vasoconstrictor
Conclusion neurons. J. Auton. Nerv. Syst., 15: 131–140.
Baron, R., Janig, W. and McLachlan, E.M. (1985) The afferent
and sympathetic components of the lumbar spinal outflow to
Further studies with gene delivery, and perhaps the colon and pelvic organs in the cat. III. The colonic nerves,
pharmacological agents that mimic plasticity-al- incorporating an analysis of all components of the lumbar
tering agents, are needed to clarify the relative prevertebral outflow. J. Comp. Neurol., 238: 158–168.
contribution of changes in primary afferent versus Birder, L.A., Roppolo, J.R., Iadarola, M.J. and de Groat, W.C.
(1991) Electrical stimulation of visceral afferent pathways in
interneuronal/propriospinal systems to autonomic the pelvic nerve increases c-fos in the rat lumbosacral spinal
dysreflexia. Such approaches may also address in- cord. Neurosci. Lett., 129: 193–196.
traspinal plasticity in other forms of dysfunction Brown, A., Ricci, M.J. and Weaver, L.C. (2004) NGF message
and/or recovery after spinal cord injury, such as and protein distribution in the injured rat spinal cord. Exp.
chronic pain and locomotion. These results are the Neurol., 188: 115–127.
Cameron, A.A., Smith, G.M., Randall, D.C., Brown, D.R. and
first demonstration that the remodeling of endog- Rabchevsky, A.G. (2004) Genetic manipulation of afferent
enous circuitry, which may underlie the develop- fiber sprouting following spinal cord injury modulates the
ment of autonomic dysreflexia, can be altered by severity of autonomic dysreflexia. J. Neurotrauma, 21: 1271.
genetic manipulation of axon guidance and inhib- Chau, D., Johns, D.G. and Schramm, L.P. (2000) Ongoing and
itory molecules to modulate plasticity-induced au- stimulus-evoked activity of sympathetically correlated neu-
rons in the intermediate zone and dorsal horn of acutely
tonomic pathophysiology after complete spinal spinalized rats. J. Neurophysiol., 83: 2699–2707.
cord injury. de Groat, W.C., Kawatani, M., Hisamitsu, T., Cheng, C.L.,
Ma, C.P., Thor, K., Steers, W. and Roppolo, J.R. (1990)
Acknowledgments Mechanisms underlying the recovery of urinary bladder
function following spinal cord injury. J. Auton. Nervous
The author thanks Dr. Adrian A. Cameron for System, 30(Suppl): S71–S77.
Finestone, H.M. and Teasell, R.W. (1993) Autonomic
contributions to the text and Drs. David C. Randall dysreflexia after brainstem tumor resection. A case report.
and George M. Smith for critical review. Addi- Am. J. Phys. Med. Rehabil., 72: 395–397.
tionally, Kainath Durre, Johnna Shipp, Igor Harati, Y. (1997) Autonomic disorders associated with spinal
Voskresensky, and Leslie Schwindel are appreci- cord injury. In: Low P.A. (Ed.), Clinical Autonomic Disor-
ders (2nd ed). Lippincott-Raven, Philadelphia, pp. 455–461.
ated for expert technical assistance. AGR is sup-
Hirshberg, R.M., Al-Chaer, E.D., Lawand, N.B., Westlund,
ported by grants from the International Spinal K.N. and Willis, W.D. (1996) Is there a pathway in the pos-
Research Trust, the Kentucky Spinal Cord and terior funiculus that signals visceral pain? Pain, 67: 291–305.
Head Injury Research Trust, and the NIH/NINDS Karlsson, A.K. (1999) Autonomic dysreflexia. Spinal Cord, 37:
(R01 NS-049901-01). 383–391.
273

Keast, J.R. and De Groat, W.C. (1992) Segmental distribution clip-compression model of spinal cord injury. J. Neurotrau-
and peptide content of primary afferent neurons innervating ma, 19: 1531–1541.
the urogenital organs and colon of male rats. J. Comp. Martinez, V., Wang, L., Mayer, E. and Tache, Y. (1998)
Neurol., 319: 615–623. Proximal colon distention increases Fos expression in the
Klimaschewski, L. (2001) Increased innervation of rat pre- lumbosacral spinal cord and activates sacral parasympathetic
ganglionic sympathetic neurons by substance P containing NADPHd-positive neurons in rats. J. Comp. Neurol., 390:
nerve fibers in response to spinal cord injury. Neurosci. Lett., 311–321.
307: 73–76. Matsushita, M. (1998) Ascending propriospinal afferents to
Krassioukov, A.V., Johns, D.G. and Schramm, L.P. (2002) area X (substantia grisea centralis) of the spinal cord in the
Sensitivity of sympathetically correlated spinal interneurons, rat. Exp. Brain Res., 119: 356–366.
renal sympathetic nerve activity, and arterial pressure to McCarthy, P.W. and Lawson, S.N. (1990) Cell type and
somatic and visceral stimuli after chronic spinal injury. conduction velocity of rat primary sensory neurons with
J. Neurotrauma, 19: 1521–1529. calcitonin gene-related peptide-like immunoreactivity.
Krassioukov, A.V. and Weaver, L.C. (1995) Episodic hyper- Neuroscience, 34: 623–632.
tension due to autonomic dysreflexia in acute and chronic Michael, G.J., Kaya, E., Averill, S., Rattray, M., Clary, D.O.
spinal cord-injured rats. Am. J. Physiol., 268: H2077–H2083. and Priestley, J.V. (1997) TrkA immunoreactive neurones in
Krenz, N.R., Meakin, S.O., Krassioukov, A.V. and Weaver, the rat spinal cord. J. Comp. Neurol., 385: 441–455.
L.C. (1999) Neutralizing intraspinal nerve growth factor Ness, T.J. and Gebhart, G.F. (1987) Characterization of neu-
blocks autonomic dysreflexia caused by spinal cord injury. J. ronal responses to noxious visceral and somatic stimuli in the
Neurosci., 19: 7405–7414. medial lumbosacral spinal cord of the rat. J. Neurophysiol.,
Krenz, N.R. and Weaver, L.C. (1998a) Sprouting of primary 57: 1867–1892.
afferent fibers after spinal cord transection in the rat. Ness, T.J. and Gebhart, G.F. (1988) Characterization of neu-
Neuroscience, 85: 443–458. rons responsive to noxious colorectal distension in the T13-
Krenz, N.R. and Weaver, L.C. (1998b) Changes in the mor- L2 spinal cord of the rat. J. Neurophysiol., 60: 1419–1438.
phology of sympathetic preganglionic neurons parallel the Ness, T.J. and Gebhart, G.F. (1989) Characterization of
development of autonomic dysreflexia after spinal cord injury superficial T13-L2 dorsal horn neurons encoding for
in rats. Neurosci. Lett., 243: 61–64. colorectal distension in the rat: comparison with neurons in
Landrum, L.M., Jones, S.L. and Blair, R.W. (2002) The ex- deep laminae. Brain Res., 46: 301–309.
pression of Fos-labeled spinal neurons in response to colo- Ondarza, A.B., Ye, Z. and Hulsebosch, C.E. (2003) Direct ev-
rectal distension is enhanced after chronic spinal cord idence of primary afferent sprouting in distant segments fol-
transection in the rat. Neuroscience, 110: 569–578. lowing spinal cord injury in the rat: colocalization of GAP-43
Lawson, S.N., McCarthy, P.W. and Prabhakar, E. (1996) and CGRP. Exp. Neurol., 184: 373–380.
Electrophysiological properties of neurones with CGRP- Petkó, M. and Antal, M. (2000) Propriospinal afferent and
like immunoreactivity in rat dorsal root ganglia. J. Comp. efferent connections of the lateral and medial areas of the
Neurol., 365: 355–366. dorsal horn (laminae I–IV) in the rat lumbar spinal cord.
Lawson, S.N., Perry, M.J., Prabhakar, E. and McCarthy, J. Comp. Neurol., 422: 312–325.
P.W. (1993) Primary sensory neurones: neurofilament, Romero, M.I., Rangappa, N., Garry, M.G. and Smith, G.M.
neuropeptides, and conduction velocity. Brain Res. Bull., (2001) Functional regeneration of chronically injured sensory
30: 239–243. afferents into adult spinal cord after neurotrophin gene ther-
Lindan, R., Joiner, E., Freehafer, A.A. and Hazel, C. (1980) apy. J. Neurosci., 21: 8408–8416.
Incidence and clinical features of autonomic dysreflexia in Seki, S., Sasaki, K., Fraser, M.O., Igawa, Y., Nishizawa, O.,
patients with spinal cord injury. Paraplegia, 18: 285–292. Chancellor, M.B., de Groat, W.C. and Yoshimura, N. (2002)
Llewellyn-Smith, I.J. and Weaver, L.C. (2001) Changes in Immunoneutralization of nerve growth factor in lumbosacral
synaptic inputs to sympathetic preganglionic neurons after spinal cord reduces bladder hyperreflexia in spinal cord
spinal cord injury. J. Comp. Neurol., 435: 226–240. injured rats. J. Urol., 168: 2269–2274.
Maiorov, D.N., Fehlings, M.G. and Krassioukov, A.V. (1998) Snow, J.C., Sideropoulos, H.P., Kripke, B.J., Freed, M.M.,
Relationship between severity of spinal cord injury and ab- Shah, N.K. and Schlesinger, R.M. (1978) Autonomic hype-
normalities in neurogenic cardiovascular control in conscious rreflexia during cystoscopy in patients with high spinal cord
rats. J. Neurotrauma, 15: 365–374. injuries. Paraplegia, 15: 327–332.
Marsh, D.R. and Weaver, L.C. (2004) Autonomic dysreflexia, Tang, X., Neckel, N.D. and Schramm, L.P. (2003) Locations
induced by noxious or innocuous stimulation, does not de- and morphologies of sympathetically correlated neurons
pend on changes in dorsal horn substance p. J. Neurotrauma, in the T(10) spinal segment of the rat. Brain Res., 976:
21: 817–828. 185–193.
Marsh, D.R., Wong, S.T., Meakin, S.O., MacDonald, J.I., Tang, X.Q., Tanelian, D.L. and Smith, G.M. (2004)
Hamilton, E.F. and Weaver, L.C. (2002) Neutralizing Semaphorin3A inhibits nerve growth factor-induced sprout-
intraspinal nerve growth factor with a trkA-IgG fusion pro- ing of nociceptive afferents in adult rat spinal cord.
tein blocks the development of autonomic dysreflexia in a J. Neurosci., 24: 819–827.
274

Vizzard, M.A. (2000) Increased expression of spinal cord for growth associated protein-43 suggest reorganization of
Fos protein induced by bladder stimulation after spinal cord synapses on spinal sympathetic neurons after cord transec-
injury. Am. J. Physiol. Regul. Integr. Comp. Physiol., 279: tion. Neuroscience, 81: 535–551.
R295–R305. Weaver, L.C., Verghese, P., Bruce, J.C., Fehlings, M.G., Krenz,
Vizzard, M.A., Brisson, M. and de Groat, W.C. (2000) N.R. and Marsh, D.R. (2001) Autonomic dysreflexia and
Transneuronal labeling of neurons in the adult rat central primary afferent sprouting after clip-compression injury of
nervous system following inoculation of pseudorabies virus the rat spinal cord. J. Neurotrauma, 18: 1107–1119.
into the colon. Cell Tissue Res., 299: 9–26. Willis, W.D., Al-Chaer, E.D., Quast, M.J. and Westlund, K.N.
Wang, C.C., Willis, W.D. and Westlund, K.N. (1999) Ascend- (1999) A visceral pain pathway in the dorsal column of the
ing projections from the area around the spinal cord central spinal cord. Proc. Natl. Acad. Sci. USA, 96: 7675–7679.
canal: a Phaseolus vulgaris leucoagglutinin study in rats. J. Zagon, A. and Smith, A.D. (1993) Monosynaptic projections
Comp. Neurol., 415: 341–367. from the rostral ventrolateral medulla oblongata to identified
Weaver, L.C., Cassam, A.K., Krassioukov, A.V. and sympathetic preganglionic neurons. Neuroscience, 54:
Llewellyn-Smith, I.J. (1997) Changes in immunoreactivity 729–743.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 18

Spinal cord injury alters cardiac electrophysiology


and increases the susceptibility to
ventricular arrhythmias

Heidi L. Collins1, David W. Rodenbaugh2 and Stephen E. DiCarlo1,

1
Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
2
Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA

Abstract: The autonomic nervous system modulates cardiac electrophysiology and abnormalities of au-
tonomic function are known to increase the risk of ventricular arrhythmias. The abnormal and unstable
autonomic control of the cardiovascular system following spinal cord injury also is well known. For
example, individuals with mid-thoracic spinal cord injury have elevated resting heart rates, increased blood
pressure variability, episodic bouts of life-threatening hypertension as part of a condition termed auto-
nomic dysreflexia, and elevated sympathetic activity above the level of the lesion. Furthermore, cardio-
vascular morbidity and mortality are high in individuals with spinal cord injuries due to a relatively
sedentary lifestyle and higher prevalence of other cardiovascular risk factors, including obesity and di-
abetes. Therefore, spinal cord injury may alter cardiac electrophysiology and increase the risk for ven-
tricular arrhythmias. In this chapter, we discuss how the autonomic changes associated with cord injury can
influence cardiac electrophysiology and the susceptibility to ventricular arrhythmias.

Individuals with spinal cord injury in the cervical new injuries are reported each year in the United
and upper thoracic (T) segments (T1–T6) have States alone. Spinal cord injuries cost the United
unstable arterial pressure and heart rate. In addi- States at least $9.7 billion per year for medical
tion to other factors, the unstable arterial pressure care, equipment and disability support (DeVivo,
and heart rate after spinal cord injury enhances the 1997; Berkowitz et al., 1998). With the advances in
risk for blood pressure-related cardiovascular dis- acute care and rehabilitation, the life expectancy of
ease above levels seen in hypertensive subjects individuals with spinal cord injury has increased to
(Rodenbaugh et al., 2003a). Thus, it is not sur- near that for able-bodied individuals. However,
prising that cardiovascular disease is a leading cardiovascular disease is now a leading cause of
cause of morbidity and mortality for individuals death and morbidity for individuals with spinal
with spinal cord injury. In this chapter, we will cord injury (Le and Price, 1982; Wicks et al.,
discuss the impact of spinal cord injury on the 1983; Cardiovascular–Cardiopulmonary Second-
autonomic control of the circulation and its rela- ary Disabilities, 1991; DeVivo et al., 1992, 1993;
tionship to increased risk for cardiac arrhythmias. Whiteneck et al., 1992; Frankel et al., 1998; Soden
An estimated 2 million individuals worldwide et al., 2000). Cardiovascular morbidity and mor-
live with spinal cord injuries. On average, 11,000 tality are high for individuals with spinal cord in-
juries, presumably due to a relatively sedentary
Corresponding author. Tel.: +(313) 577-1557; lifestyle and higher prevalence of other cardiovas-
Fax: +(313) 577-5494; E-mail: sdicarlo@med.wayne.edu cular risk factors, including obesity and diabetes

DOI: 10.1016/S0079-6123(05)52018-1 275


276

(Duckworth et al., 1980; Levi et al., 1995; Rodenbaugh et al., 2003a, b, c). This phenomenon
Karlsson, 1999). In fact, individuals with spinal is illustrated in Fig. 1 that shows the heart rate for
cord injury are at the lowest end of the human intact and cord-injured rats, recorded by radio-
fitness spectrum (Dearwater et al., 1986; Washburn telemetry, averaged over each week for 7 weeks
and Figoni, 1998). Similarly, cord-injured rats have (panel A) or for the entire 7 weeks of study (panel
lower levels of physical activity than healthy intact B). Animals with cord transection at T5 had
rats (Rodenbaugh et al., 2003a). Additionally, in- increased heart rates each week during the entire 7
dividuals with cord injury have blood lipid profiles weeks of study (panel A). Specifically, the average
characterized by elevated total cholesterol and heart rate was 19% higher in cord-injured rats
low-density lipoprotein cholesterol, and depressed during the entire 7 weeks of study (panel B).
high-density lipoprotein cholesterol, a lipid profile Numerous studies have documented that an
normally associated with, if not a direct result of, elevated heart rate is a strong risk factor for the
the sedentary lifestyle (Bauman et al., 1992, 1999; development of cardiovascular disease (Palatini
Cardus et al., 1992). There is a strong association and Julius, 1997, 1999; Julius et al., 1998). The
between low physical activity, increased sympa- elevated heart rates observed in individuals with
thetic nervous system activity, and cardiovascular mid-thoracic cord injury may be mediated by a
disease risk factors (Zimmet et al., 1991; Collins reflex compensatory mechanism to offset the de-
et al., 2000). Importantly, increased physical ac- creased stroke volume (Hjeltnes, 1977; Hopman
tivity lowers sympathetic nerve activity (DiCarlo et al., 1993a). The lower stroke volume has been
et al., 1997) and cardiovascular disease risk factors
(Jennings et al., 1984; Collins et al., 2000; DiCarlo
et al., 2002). Therefore, exercise with the arms is
often recommended for individuals with cord in-
jury, based on studies demonstrating improve-
ments in aerobic capacity and lipoprotein profiles
(DiCarlo, 1982, 1988; DiCarlo et al., 1983; Glaser,
1985; Hoffman, 1986; Hooker and Wells, 1989).
In fact, the Center for Disease Control has rec-
ommended further research to evaluate the effica-
cy of exercise to prevent the development of
cardiovascular disease in individuals with cord
injury (Cardiovascular–Cardiopulmonary Second-
ary Disabilities, 1991). It is interesting to speculate
that one mechanism contributing to the improved
cardiopulmonary status after exercise in indi-
viduals with spinal cord injury (DiCarlo, 1982;
DiCarlo et al., 1983) may be a reduced incidence
and/or severity of cardiac arrhythmias.
Cord-injured individuals have distinct hemody-
namic responses to a variety of activities of daily
living. For example, it is well established that Fig. 1. Average heart rate for intact and cord-injured rats, re-
individuals with mid-thoracic cord injury have corded by radio-telemetry, each week for 7 weeks (panel A) and
elevated heart rates and lower stroke volumes at the heart rates averaged during the entire 7 weeks of the study
rest and during activity than able-bodied indi- (panel B). Heart rate was higher in the cord-injured rats at every
time point during the 7 weeks of the study. Specifically, heart
viduals (Jacobs et al., 2002). Similarly, rats with
rate was 19% higher in cord-injured rats during the entire 7
cord injury at the 5th thoracic segment (T5) have weeks of the study (panel B). In this and subsequent figures,
elevated heart rates (Krassioukov and Weaver, variability is indicated by the standard error of the mean
1995; Maiorov et al., 1997; Mayorov et al., 2001; 
Pp0:05:
277

attributed to reduced venous return from the in- activity whereas a decreased heart rate is mediated
active regions below the level of the injury due to by an increase in parasympathetic activity and a
loss of supraspinal control of sympathetically me- concomitant decrease in sympathetic activity.
diated veno- and vasoconstriction (Kinzer and Overactivity, or dominant activity in the cardiac
Convertino, 1989; Hopman et al., 1993b). The loss parasympathetic nerves can produce or contribute
of supraspinal control of sympathetically mediated to a variety of brady-arrhythmias whereas over-
veno- and vasoconstriction impairs the ability to activity or dominant activity of the cardiac sym-
effectively distribute blood throughout the vascu- pathetic nerves can produce or contribute to a
lar system and results in venous pooling. Preven- variety of tachy-arrhythmias (Katz, 1992). In rest-
tion of venous pooling with an external ‘‘muscle ing able-bodied individuals parasympathetic activ-
pump’’ using transcutaneous electrical stimulation ity dominates. Thus, when both divisions of the
markedly enhances stroke volume in cord-injured autonomic nervous system are blocked (by drugs
individuals (Davis et al., 1990). Cardiac output, such as atropine for parasympathetic and prop-
the volume of blood pumped by the heart each ranolol for sympathetic) in resting able-bodied
minute, is the product of heart rate and stroke individuals, the heart rate increases to approxi-
volume and is lower in cord-injured people (Jacobs mately 100 beats per minute (Jose, 1966; Katona
et al., 2002). et al., 1982). This increase in heart rate following
In able-bodied individuals the average heart rate complete autonomic nervous system blockade doc-
at rest is 60–80 beats per minute. As noted above, uments the dominance of the parasympathetic sys-
heart rate is significantly higher in individuals with tem on resting heart rate. The prevailing heart rate
mid-thoracic cord injury. During emotional ex- after block of both sympathetic and parasympa-
citement or muscular activity, heart rate increases thetic cardiac influences is called the intrinsic heart
reaching values as high as 220 beats per minute rate. Importantly, because the cardiac parasym-
minus the age of the individual. The maximum pathetic fibers never pass through the spinal cord,
heart rate is not altered in individuals with spinal cord injury does not interrupt cardiac para-
mid-thoracic cord injury (Jacobs et al., 2002). The sympathetic activity. Cardiac parasympathetic
increase in heart rate is mediated, mainly, by chang- fibers originate in the dorsal motor nucleus of
es in the autonomic nervous system. The process the vagus and the nucleus ambiguus in the medulla
of excitation of the heart originates in the sino- oblongata (Loewy and Spyer, 1990) and travel in
atrial node, the intrinsic pacemaker of the heart, the vagus nerve to the heart. Subsequently, these
located in the right atrium. These cells depolarize fibers synapse with postganglionic cells on the
spontaneously and generate action potentials at an epicardial surface or within the walls of the heart
intrinsic rate of about 100 per minute (Jose, 1966; near the sinoatrial node and atrioventricular node.
Katona et al., 1982). The sinoatrial node is under In contrast, spinal cord injury above the 1st
the direct influence of the sympathetic and para- thoracic segment (T1) disrupts central control of
sympathetic divisions of the autonomic nervous preganglionic cardiac sympathetic activity. Specif-
system. In general, the sympathetic division of the ically, preganglionic cardiac sympathetic fibers
autonomic nervous system increases heart rate by originate in the intermediolateral cell column of
increasing the rate at which the sinoatrial node the upper five or six thoracic segments of the spinal
generates action potentials, whereas the parasym- cord (Bonica, 1968). These fibers exit the spinal
pathetic division of the autonomic nervous system cord through the white communicating rami and
decreases heart rate by decreasing the rate of ac- enter the paravertebral chains of ganglia. The
tion potential generation by the sinoatrial node. cardiac preganglionic and postganglionic neurons
Usually, changes in heart rate involve the recipro- synapse in the middle cervical and upper thoracic
cal actions of the two divisions of the autonomic paravertebral ganglia, especially the stellate
nervous system. Specifically, an increased heart ganglion (Bonica, 1968). Cervical and upper tho-
rate is mediated by a reduction in parasympathetic racic spinal cord injury disrupts the interaction
activity and a concomitant increase in sympathetic of the cardiac sympathetic and parasympathetic
278

innervation. Disruption of this balance may have a increase the risk for the development of ventricu-
profound influence on cardiac rate, performance lar arrhythmias and sudden cardiac death
and rhythm. For example, the higher heart rates (Schwartz and Stone, 1980; Schwartz et al.,
observed in individuals and animals with mid- 1993). Therefore, the elevated heart rates and in-
thoracic cord injury suggest higher cardiac sym- creased cardiac sympathetic activity in individuals
pathetic activity or lower cardiac parasympathetic with mid-thoracic cord injury (Karlsson et al.,
activity. To test this hypothesis we measured heart 1998; Rodenbaugh et al., 2003b) may result in an
rate in intact rats and rats with cord injury increased mortality associated with changes in
at T5 under normal resting conditions and cardiac electrophysiology and the incidence of
during ganglionic blockade achieved by intrave- arrhythmias. In fact, it has recently been docu-
nous administration of hexamethonium chloride. mented that rats with cord transection at the 5th
Hexamethonium chloride abolishes postganglionic thoracic segment have a lower electrical stimula-
sympathetic and parasympathetic nervous system tion threshold to induce cardiac arrhythmias
activity and reduces arterial pressure and heart (Rodenbaugh et al., 2003b, c).
rate slightly in intact rats and more substantially in Arterial baroreceptors, located in the aortic arch
cord-injured rats. Resting heart rate was signifi- and carotid sinuses, have a profound influence on
cantly higher in injured rats and decreased signif- cardiac rate, performance, and rhythm by reflexly
icantly more in response to ganglionic blockade. altering cardiac sympathetic and parasympathetic
Figure 2 presents heart rate before and after activity. The arterial baroreceptors respond to a
ganglionic blockade in intact and cord-injured reduction in arterial pressure by decreasing activity
rats. Ganglionic blockade reduced heart rate more of the parasympathetic nerves and increasing
in injured rats compared with intact rats (36% vs. activity of the sympathetic nerves. This imbalance
9%, respectively). These findings are consistent of the autonomic nervous system in favor of sym-
with the idea that an increase in sympathetic ac- pathetic dominance can lead to tachy-arrhythmias.
tivity above the level of the lesion in cord-injured In contrast, the arterial baroreceptors respond to
rats contributes to a greater tonic sympathetic an increase in arterial pressure by increasing
support of heart rate. activity of the parasympathetic nerves and
Elevations in cardiac sympathetic activity or decreasing activity of the sympathetic nerves. This
reductions in cardiac parasympathetic activity imbalance favoring parasympathetic dominance
can lead to brady-arrhythmias. Arterial pressure is
usually lower and unstable after cervical and upper
thoracic spinal cord injury. Hypotension occurs
immediately after the injury because of loss of
tonic supraspinal excitatory drive to spinal
sympathetic neurons (Calaresu and Yardley,
1988). Subsequently, resting arterial pressure
returns toward normal. However, episodic hyper-
tension often develops as part of the condition
termed autonomic dysreflexia (Naftchi, 1990;
Mathias and Frankel, 1992). In addition, activi-
ties of daily living produce large variations of
blood pressure in individuals with cord injury
(Stiens et al., 1995; Faghri et al., 2001). This is due,
in part, to the fact that arterial baroreflex control
Fig. 2. Heart rate before (control) and after ganglionic block-
of the sympathetic nervous system is lost below the
ade (Gag-X) in intact and cord-injured rats. Ganglionic block-
ade (equivalent to elimination of all neural input to the heart) level of the lesion, resulting in reduced buffering of
reduced heart rate more in cord-injured rats than in intact rats changes in arterial pressure. Thus, individuals and
(36% vs. 9%, respectively). animals with spinal cord injury have increased
279

blood pressure variability after mid-thoracic cord entire duration of the study (panel C). Mid-
injury (Rodenbaugh et al., 2003a). Increased blood thoracic cord injury significantly increased SBP-
pressure variability and heart rate significantly in- SD each week for the entire 7 weeks of the study.
crease the risk of cardiovascular diseases (Rizzoni Specifically, SBP-SD was 22% higher in the in-
et al., 1992; Stevenson et al., 1997; Julius and jured versus intact rats over the entire 7 weeks
Valentini, 1998). These risk factors are highly cor- (panel C). Similarly, the average diastolic blood
related with end organ damage, as well as vascular pressure standard deviation (DBP-SD) for intact
structure changes and an increased incidence of and cord-injured rats was examined (panels B and
myocardial infarction, stroke, and cardiac ar- D) and cord injury increased DBP-SD during the
rhythmias (Frattola et al., 1993; Stamler et al., entire 7 weeks of the study. Specifically, DBP-SD
1993; Palatini and Julius, 1997, 1999). The in- was 13% higher in the injured versus intact rats
creased blood pressure variability mediates chang- during the entire 7 weeks (panel D). Taken to-
es in arterial baroreceptor activity that reflexly gether, these data suggest that the loss of arterial
alters cardiac sympathetic and parasympathetic baroreflex control of the autonomic nervous sys-
activity and profoundly affects cardiac rate, per- tem below the level of the lesion increases arterial
formance, and rhythm. Importantly, cord-injured blood pressure variability.
rats have increased blood pressure-related cardio- As noted above, cardiac tachy-arrhythmias are
vascular disease risk factors (Rodenbaugh et al., associated with elevations of cardiac sympathetic
2003a). For example, Fig. 3 presents the average efferent activity (Schwartz and Stone, 1980, 1982).
systolic blood pressure standard deviation (SBP- Cardiac a- and b-adrenergic receptors mediate the
SD) for intact and cord-injured rats each week for response to cardiac sympathetic stimulation. b-
7 weeks (panel A) and SBP-SD averaged over the Adrenergic receptor blockade, which reduces the

Fig. 3. Systolic blood pressure standard deviation (SBP-SD) and diastolic blood pressure deviation (DBP-SD) for intact and cord-
injured rats, averaged each week for 7 weeks (panels A, B) and averaged over the entire duration of the study (panels C, D). Spinal cord
injury significantly increased SBP-SD and DBP-SD. Specifically, SBP-SD was 22% higher in the injured versus intact rats over the
entire 7 weeks (panel C) and DBP-SD was 13% higher in the injured vs. intact rats over the entire 7 weeks (panel D)  Pp0:05:
280

effect of cardiac sympathetic efferent activity, has adrenergic signals alter cardiac electrophysiology
been shown to reduce cardiovascular mortality over the time course of hours to days by altering
(Frishman, 1992). The sympathetic nervous system transcription of genes encoding ion channels and
can affect the generation and conduction of action calcium-regulating proteins. For example, suscep-
potentials in the heart by activating mainly b-ad- tibility to cardiac arrhythmias may increase by al-
renergic receptors (Berne and Levy, 2001). b-Ad- tering the expression of specific genes encoding
renergic receptor stimulation, which increases proteins critical to myocyte calcium homeostasis
intracellular cAMP, increases heart rate, atrioven- [the sarco(endo)plasmic reticulum Ca2+ ATPase
tricular nodal conduction and contractile force, (SERCA), the Na+/Ca2+ exchanger encoded by
and shortens atrial and ventricular refractoriness the NCX1 gene, and the L-type calcium channel]
(Murray and Roden, 1996). In addition, it en- in a manner that will favor increasing intracellular
hances the plateau phase of the action potential by Ca2+ (Rodenbaugh et al., 2003c). The ability of
increasing current through L-type Ca2+ channels cardiac myocytes to maintain cytosolic Ca2+ with-
while repolarization is accelerated due to an in- in a tightly controlled range is important to pre-
crease in both the delayed rectifier current and the vent cardiac electrical disturbances. Disturbances
chloride current (Murray and Roden, 1996). Thus, in Ca2+ homeostasis are closely related to cardiac
b-adrenergic receptor stimulation may shorten or electrophysiological events. The Na+/Ca2+ ex-
prolong action potential duration depending on changer and the L-type calcium channel provide
which of the currents predominate. b-Adrenergic the primary calcium efflux and influx pathways,
receptor stimulation also causes more rapid pace- respectively. SERCA is the predominant pathway
maker activity in the sinus node by its action on through which cytosolic calcium is sequestered.
diastolic currents (Murray and Roden, 1996). a- Signaling through a- and b-adrenergic receptors
Adrenergic receptor stimulation enhances cardiac modulates calcium homeostasis by altering the ex-
contractility due to Ca2+ influx (Berne and Levy, pression of these calcium regulatory proteins. For
2001). Furthermore, adrenergic stimulation en- example, signaling through a- and b-adrenergic
hances the development of after-depolarizations receptors coordinately regulates the expression of
that lead to electrocardiogram (ECG) complexes the a 1C-subunit of the L-type calcium channel
termed triggered beats (Katz, 1992). In this situ- and the Na+/Ca2+ exchanger by activating pro-
ation, multiple ionic mechanisms are involved, and tein kinase A and protein kinase C pathways
elevated intracellular calcium is a common feature. (Golden et al., 2000, 2002). Specifically, b-ad-
Adrenergic stimulation results in a reduction of the renergic receptor activation in vitro increases the
electrical stimulus threshold to induce ventricular expression of the L-type Ca2+ channel and NCX1
fibrillation as well as an increase in the likelihood genes, whereas a-adrenergic signaling reduces their
of spontaneous ventricular arrhythmias. b-Ad- mRNA levels (Golden et al., 2000, 2001, 2002).
renergic receptor blockade and enhanced para- Several facts have been well documented. The
sympathetic tone inhibit these effects and are autonomic nervous system modulates cardiac
known to be protective against ventricular tachy- electrophysiology and abnormalities of autonom-
arrhythmias and sudden death (Engel, 1978; Zipes, ic function can increase the risk of cardiac
1991; Wharton et al., 1992; Schwartz et al., 1993). arrhythmias. Furthermore, autonomic control of
Thus, interventions that reduce sympathetic activ- the cardiovascular system is abnormal and unsta-
ity or enhance parasympathetic activity may be ble following spinal cord injury. For example, in-
useful in preventing deadly tachy-arrhythmias. dividuals with spinal cord injury have elevated
Spinal cord injury, depending on the specific site heart rates, increased blood pressure variability,
of the lesion, may affect sympathetic and para- elevated sympathetic activity above the level of the
sympathetic activity in a manner that promotes lesion, and episodic bouts of life-threatening hy-
arrhythmias. pertension as part of a condition termed auto-
In addition to transmembrane signals that alter nomic dysreflexia. Autonomic dysreflexia occurs in
ionic currents over the time course of seconds, as many as 85% of individuals with spinal cord
281

injuries above the 6th thoracic segment and is


characterized by severe hypertension (Lee et al.,
1995). If not prevented or treated promptly, the
hypertension may produce cerebral and sub-
arachnoid hemorrhage, seizures, renal failure, car-
diac arrhythmias, and may lead to death (McGuire
and Kumar, 1986). An example of autonomic
dysreflexia in rats is presented in Fig. 4, illustrating
an analog recording of arterial pressure, ECG, and
heart rate responses to colon distension in a quad-
riplegic rat (Collins and DiCarlo, 2002). Colon
distension produced an increase in arterial pres-
sure, bradycardia, and arrhythmias. This observa-
tion demonstrates that abnormal control of the
cardiovascular system after spinal cord injury can
increase the susceptibility to cardiac arrhythmias.
Autonomic dysreflexia is the second most com-
mon long-term secondary medical complication
associated with cord injury and thus is a major
health concern (McKinley et al., 1999). In fact,
autonomic dysreflexia is the most prominent life-
threatening situation for individuals with spinal
cord injury (Comarr and Eltorai, 1997). The long-
term consequence of repeated episodes of severe
hypertension has yet to be determined, however, it
is well documented that increased blood pressure
variability is a significant cardiovascular disease
risk factor (Frattola et al., 1993; Collins et al.,
2000). Thus, the incidence of cardiac arrhythmias
may be increased in individuals with spinal cord
injury due to the unstable arterial blood pressure.
In support of this concept, there are a large
number of case reports documenting cardiac
arrhythmias in individuals with spinal cord injury
(Guttmann and Whitteridge, 1947; Frankel et al.,
1975; Colachis and Clinchot, 1997). For example,
as early as 1947, Guttmann and Whitteridge re-
ported premature atrial and ventricular contrac-
tions as well as changes in the amplitude of the
T-waves in individuals with spinal cord injuries Fig. 4. Analog recording of arterial pressure, ECG, and heart
during urodynamic testing. Arrhythmias have also rate response to colon distension (50 mmHg for 1 min) in one
quadriplegic rat. Dashed line indicates the onset of colon dis-
been observed in a woman with a spinal lesion at
tension. Colon distension produced pressor and bradycardic
T3 during labor (Guttmann et al., 1965). Prema- responses. Of particular interest are the arrhythmias produced
ture ventricular contractions and atrio-ventricular by colon distension (insert). Reprinted with permission from
dissociation are common when individuals with spi- Collins and DiCarlo (2002).
nal cord injuries have distended bladders (Guttmann
and Whitteridge, 1947) and asystole as well as
other arrhythmias have been reported during
282

tracheal suctioning (Frankel et al., 1975) and individuals and individuals with spinal cord
autonomic dysreflexia. Profound bradycardia is a injury. These investigators examined the ECG of
common complication in the early post-traumatic 26,734 able-bodied male veterans with a mean age
period following cervical spinal cord damage. It is of 56 years and compared the data with the ECG
thought to be due to temporary inactivity of the of 654 individuals with SCI with a mean age of 50
sympathetic nervous system after separation from years. Similarly, Leaf et al. (1993) studied 47 in-
supraspinal control, coupled with unopposed dividuals with chronic SCI (35–3605 days post-in-
parasympathetic dominance because of parasym- jury). Twenty-five individuals were classified as
pathetic, vagus nerve sparing (Mathias, 1976; paraplegic (mean age 39 years). No differences
Piepmeier et al., 1985; Lehmann et al., 1987; Bravo were recorded in the incidence of abnormalities
et al., 2004). Hypoxia, hypo-ventilation, and trache- between the individuals with paraplegia and quad-
al suctioning appear to intensify the bradycardia. In riplegia. Thus, the data regarding ECG abnormal-
animals, acute spinal cord transection produces a ities associated with SCI are equivocal.
variety of arrhythmias (Greenhoot et al., 1972). Two reported studies tested the effect of spinal
Several human studies have documented ECG cord injury at T4 on cardiac electrophysiology and
characteristics of individuals with spinal cord in- the susceptibility to ventricular arrhythmias
jury. Blocker et al. (1983) analyzed the resting (Rodenbaugh et al., 2003a, c). In the first study,
ECG of 98 individuals with chronic cord injury conscious female hypertensive cord-injured rats
and compared their findings with results from two had a significantly lower electrical stimulation
studies of healthy able-bodied individuals. The in- threshold to induce ventricular arrhythmias com-
vestigators reported that ECG abnormalities were pared to intact rats. The protocol used to induce
more prevalent in the individuals with cord injury. arrhythmias is presented in Fig. 5. Ventricular
The mean age of the individuals with cord injury arrhythmia was defined as sustained ventricular
was 47 years and more than half of the individuals tachycardia resulting in a reduction in arterial
had sustained their injury before the age of 40. The pressure. The intensity of current required to
most common ECG abnormalities were ST seg- cause a ventricular arrhythmia was 62% lower in
ment depression and T-wave inversion. Axis devi- cord-injured rats compared with intact rats. Cord-
ation, ventricular conduction delays, frequent injured rats also had a 35% lower effective refrac-
premature ventricular contractions, and low QRS tory period compared to intact rats. Associated
amplitude were also recorded. As might be ex- with the increased susceptibility to ventricular
pected, ECG abnormalities were most prevalent in arrhythmias was a significantly higher resting
the 50–59 age group. The sample contained an heart rate and cardiac sympathetic tone (as deter-
equal number of subjects with cervical and tho- mined by the effect of propranolol on heart rate
racic cord injury. ECG abnormalities were most after the administration of atropine (Chen and
often recorded when the injury was cervical. The DiCarlo, 1997). Triggered beats occur more fre-
incidence of ECG abnormalities did not relate to quently in the presence of increased heart rate
the time interval after the cord injury. Individuals (Rosen and Reder, 1981). The enhanced cardiac
with lumbar spinal cord injury had no ECG ab- sympathetic activity in the cord-injured rat may be
normalities. Lehmann et al. (1989) also analyzed the cause of increased susceptibility to ventricular
the ECG of individuals with spinal cord injury. arrhythmias. In this first study, female spontane-
These investigators reported altered ventricular ously hypertensive rats were studied because they
depolarization in individuals with cervical spinal have elevated cardiac sympathetic activity
cord injury. These results were confirmed recently (Chandler and DiCarlo, 1998) and are susceptible
(Marcus et al., 2002). The ECG abnormalities to cardiac arrhythmias. In the second study
consisted of an upwardly concave ST-segment. (Rodenbaugh et al., 2003c), changes in cardiac
In contrast to the reports cited above, Prakash calcium regulatory proteins (Fig. 6), cardiac elect-
et al. (2002) documented that the prevalence of rophysiology parameters, and the susceptibility to
ECG abnormalities was the same for able-bodied ventricular arrhythmias were examined in intact
283

Fig. 5. Analog recording of arterial pressure and the ECG during step increases in current delivered to the heart in a conscious cord-
injured hypertensive rat. A MacLab programmable stimulator delivered 10 s trains of pulses (frequency 50 Hz and duration of 10 ms).
The current was increased every 10 s in 10 mA increments. Ventricular arrhythmia was identified by both the ECG as rapid, wide QRS
complexes and a decrease in arterial pressure to 40 mmHg (inset). In this animal, the threshold current required to induce the
arrhythmia was 100 mA. Normal sinus rhythm reappears upon termination of the stimulation (not shown). Reprinted with permission
from Rodenbaugh et al. (2003b).

and cord-injured rats (Fig. 7). Mid-thoracic cord spontaneous calcium release, thereby leading to
injury was associated with alterations in the ectopic activity. Importantly, these molecular
abundance of cardiac calcium regulatory proteins. changes were associated with enhanced cardiac
For example, cord injury increased the relative electrophysiology parameters and a reduced elec-
protein expression of SERCA2 (45%) and the trical stimulation threshold to induce ventricular
Na+/Ca2+ exchanger (40%), whereas relative arrhythmias. The cord-injured rats had a reduced
protein expression of phospholamban was signif- electrical stimulation threshold to induce ventricu-
icantly decreased (28%, Fig. 6). It is well doc- lar arrhythmias (48%, Fig. 7) as well as shorter
umented that reductions in phospholamban and/ atrial-ventricular interval, sinus node recovery
or increases in SERCA protein abundance result time and Wenckebach cycle length. In this study,
in an increased sarco(endo)plasmic reticulum mean arterial pressure was not significantly differ-
calcium load (Ji et al., 2000). The sarco(endo)- ent between the intact and cord-injured rats.
plasmic reticulum calcium overload may produce However, injured rats had significantly higher
284

Fig. 7. Electrical stimulation threshold to induce ventricular


arrhythmias in intact (n ¼ 10) and cord-injured rats (n ¼ 6).
Spinal cord injury significantly reduced the stimulation thresh-
old by 48%. The stimulation threshold was not significantly
different in the rats that served as time controls.  Pp0:05; in-
tact vs. injured rats. Reprinted with permission from Rodenb-
augh et al. (2003c).

a similar injury (Davis and Shephard, 1988; Kinzer


and Convertino, 1989; Jacobs et al., 2002).
Intravenous epinephrine in humans mimicked the
effects of mid-thoracic spinal cord injury on heart
rate, effective refractory period and atrioventricu-
lar conduction (Morady et al., 1988). A b-ad-
renergic receptor antagonist blocked these effects
of epinephrine. Conversely, perturbations that
Fig. 6. Western blots for sarco(endo)plasmic reticulum Ca2+
lower sympathetic activity and/or raise parasym-
ATPase (SERCA; A), phospholamban (PLM; B), and sodium pathetic activity slow the conductive properties
calcium exchanger (NCX; C) using the hearts of two intact and and intrinsic excitability of the heart (Stein et al.,
two cord-injured male Wistar rats. Quantified relative abun- 2002; Such et al., 2002). Taken together, these re-
dance for each protein of interest in intact (n ¼ 8) and cord- sults suggest that the increased susceptibility to
injured (n ¼ 8) rat hearts. Spinal cord injury increased the rel-
ative abundance of SERCA (by 45%) and Na/Ca exchanger (by
ventricular arrhythmias in cord-injured rats may
40%) with a concomitant decrease in phospholamban (28%); be due, in part, to increased cardiac sympathetic

Pp0:05; intact vs. injured rats. Reprinted with permission activity. The increased cardiac sympathetic activ-
from Rodenbaugh, et al. (2003c). ity, higher heart rates, and changes in calcium
regulatory proteins in the cord-injured rats favor
heart rates (Mayorov et al., 2001; Jacobs et al., conditions of calcium overload that increases the
2002; Rodenbaugh et al., 2003b, c). likelihood for ventricular arrhythmias.
These results were consistent with clinical
reports suggesting an increased susceptibility to
cardiac arrhythmias (Guttmann and Whitteridge, Conclusion
1947; Frankel et al., 1975; Colachis and Clinchot,
1997) as well as alterations in the ECG of indi- Individuals with spinal cord injury have unstable
viduals with spinal cord injuries (Lehmann et al., arterial pressure and heart rate due to profound
1989; Marcus et al., 2002). The results were also changes in the autonomic nervous system. Fur-
consistent with a cardiac sympatho-excitation in thermore, the autonomic nervous system modu-
rats with mid-thoracic cord injury (Maiorov et al., lates cardiac electrophysiology and abnormalities
1997; Rodenbaugh et al., 2003b) and humans with of autonomic function can increase the risk of
285

ventricular arrhythmias. Recent data suggest an Cardus, D., Ribas-Cardus, F. and McTaggart, W.G. (1992)
increased susceptibility to ventricular arrhythmias Lipid profiles in spinal cord injury. Paraplegia, 30: 775–782.
Chandler, M.P. and DiCarlo, S.E. (1998) Acute exercise and
with concomitant changes in cardiac electrophys-
gender alter cardiac autonomic tonus differently in hyper-
iology parameters and the abundance of calcium tensive and normotensive rats. Am. J. Physiol. Reg. Integ.
regulatory proteins in a conscious chronic model Comp. Physiol., 274: R510–R516.
of mid-thoracic spinal cord injury. These effects Chen, C.-Y. and DiCarlo, S.E. (1997) Endurance exercise
may be mediated by an increased cardiac sympa- training-induced resting bradycardia: a brief review. Sport.
thetic activity. It is interesting to speculate that Med. Train. Rehabil., 8: 37–77.
Colachis III, S.C. and Clinchot, D.M. (1997) Autonomic hype-
these cardiac changes contribute, in part, to the rreflexia associated with recurrent cardiac arrest: case report.
fact that cardiovascular disease is a leading cause Spinal Cord, 35: 256–257.
of death for individuals with chronic spinal Collins, H.L. and DiCarlo, S.E. (2002) TENS attenuates re-
cord injury. sponse to colon distension in paraplegic and quadriplegic rats.
Am. J. Physiol. Heart Circ. Physiol., 283: H1734–H1739.
Collins, H.L., Rodenbaugh, D.W. and DiCarlo, S.E. (2000)
Acknowledgments Daily exercise attenuates the development of arterial blood
pressure related cardiovascular risk factors in hypertensive
rats. Clin. Exp. Hypertens., 22: 193–202.
We gratefully acknowledge the expert technical
Comarr, A.E. and Eltorai, I. (1997) Autonomic dysreflexia/
assistance of Dustin G. Nowacek. This work was hyperreflexia. J. Spinal Cord Med., 20: 345–354.
supported, in part, by the National Heart Lung Davis, G.M., Servedio, F.J., Glaser, R.M., Gupta, S.C. and
and Blood Institute Grant HL-74122. Suryaprasad, A.G. (1990) Cardiovascular responses to arm
cranking and FNS-induced leg exercise in paraplegics.
J. Appl. Physiol., 69: 671–677.
References Davis, G.M. and Shephard, R.J. (1988) Cardiorespiratory fit-
ness in highly active versus inactive paraplegics. Med. Sci.
Bauman, W.A., Adkins, R.H., Spungen, A.M., Herbert, R., Sports Exerc., 20: 463–468.
Schechter, C., Smith, D., Kemp, B.J., Gambino, R., Dearwater, S.R., LaPorte, R.E., Robertson, R.J., Brenes, G.,
Maloney, P. and Waters, R.L. (1999) Is immobilization as- Adams, L.L. and Becker, D. (1986) Activity in the spinal
sociated with an abnormal lipoprotein profile? Observations cord-injured patient: an epidemiologic analysis of metabolic
from a diverse cohort. Spinal Cord, 37: 485–493. parameters. Med. Sci. Sports Exerc., 18: 541–544.
Bauman, W.A., Spungen, A.M., Zhong, Y.G., Rothstein, J.L., DeVivo, M.J. (1997) Causes and costs of spinal cord injury in
Petry, C. and Gordon, S.K. (1992) Depressed serum high the United States. Spinal Cord, 35: 809–813.
density lipoprotein cholesterol levels in veterans with spinal DeVivo, M.J., Black, K.J. and Stover, S.L. (1993) Causes of
cord injury. Paraplegia, 30: 697–703. death during the first 12 years after spinal cord injury. Arch.
Berkowitz, M., O’Leary, P.K., Kruse, D.L. and Harvey, C. Phys. Med. Rehabil., 74: 248–254.
(1998) Spinal Cord Injury: An Analysis of Medical and So- DeVivo, M.J., Shewchuk, R.M., Stover, S.L., Black, K.J. and
cial Costs. Demos Medical Publishing, Inc., New York. Go, B.K. (1992) A cross-sectional study of the relationship
Berne, R. and Levy, M. (2001) Cardiovascular Physiology. 8th between age and current health status for persons with spinal
Mosby-Year Book Inc., St. Loius, MO. cord injuries. Paraplegia, 30: 820–827.
Blocker, W.P., Merrill, J.M., Krebs, M.A., Cardus, D.P. and DiCarlo, S.E. (1982) Improved cardiopulmonary status after a
Ostermann, H.J. (1983) An electrocardiographic survey of two-month program of graded arm exercise in a patient with
patients with chronic spinal cord injury. Am. Correct Ther. C6 quadriplegia: a case report. Phys. Ther., 62: 456–459.
J., 37: 101–104. DiCarlo, S.E. (1988) Effect of arm ergometry training on
Bonica, J.J. (1968) Autonomic innervation of the viscera in re- wheelchair propulsion endurance of individuals with quad-
lation to nerve block. Anesthesiology, 29: 793–813. riplegia. Phys. Ther., 68: 40–44.
Bravo, G., Guizar-Sahagun, G., Ibarra, A., Centurion, D. and DiCarlo, S.E., Collins, H.L., Rodenbaugh, D.W., Smitha,
Villalon, C.M. (2004) Cardiovascular alterations after spinal M.R., Berger, R.D. and Yeragani, V.K. (2002) Daily exercise
cord injury: an overview. Curr. Med. Chem. Cardiovasc. reduces measures of heart rate and blood pressure variability
Hematol. Agents, 2: 133–148. in hypertensive rats. Clin. Exp. Hypertens., 24: 221–234.
Calaresu, F.R. and Yardley, C.P. (1988) Medullary basal sym- DiCarlo, S.E., Stahl, L.K. and Bishop, V.S. (1997) Daily ex-
pathetic tone. Ann. Rev. Physiol., 50: 511–524. ercise attenuates the sympathetic nerve response to exercise
Cardiovascular–Cardiopulmonary Secondary Disabilities. by enhancing cardiac afferents. Am. J. Physiol., 273:
(1991) First colloquium on preventing secondary disabilities H1606–H1610.
among people with spinal cord injuries. Center for Disease DiCarlo, S.E., Supp, M.D. and Taylor, H.C. (1983) Effect
Control Atlanta, pp. 47–54. of arm ergometry training on physical work capacity of
286

individuals with spinal cord injuries. Phys. Ther., 63: Hopman, M.T., Pistorius, M., Kamerbeek, I.C. and Binkhorst,
1104–1107. R.A. (1993a) Cardiac output in paraplegic subjects at high
Duckworth, W.C., Solomon, S.S., Jallepalli, P., Heckemeyer, exercise intensities. Eur. J. Appl. Physiol. Occup. Physiol., 66:
C., Finnern, J. and Powers, A. (1980) Glucose intolerance 531–535.
due to insulin resistance in patients with spinal cord injuries. Hopman, M.T., Verheijen, P.H. and Binkhorst, R.A. (1993b)
Diabetes, 29: 906–910. Volume changes in the legs of paraplegic subjects during arm
Engel, G.L. (1978) Psychologic stress, vasodepressor (vasova- exercise. J. Appl. Physiol., 75: 2079–2083.
gal) syncope, and sudden death. Ann. Intern. Med., 89: Jacobs, P.L., Mahoney, E.T., Robbins, A. and Nash, M. (2002)
403–412. Hypokinetic circulation in persons with paraplegia. Med. Sci.
Faghri, P.D., Yount, J.P., Pesce, W.J., Seetharama, S. and Sports Exerc., 34: 1401–1407.
Votto, J.J. (2001) Circulatory hypokinesis and functional Jennings, G.L., Nelson, L., Esler, M.D., Leonard, P. and
electric stimulation during standing in persons with spinal Korner, P.I. (1984) Effects of changes in physical activity on
cord injury. Arch. Phys. Med. Rehabil., 82: 1587–1595. blood pressure and sympathetic tone. J. Hypertens. Suppl., 2:
Frankel, H.L., Coll, J.R., Charlifue, S.W., Whiteneck, G.G., S139–S141.
Gardner, B.P., Jamous, M.A., Krishnan, K.R., Nuseibeh, I., Ji, Y., Lalli, M.J., Babu, G.J., Xu, Y., Kirkpatrick, D.L.,
Savic, G. and Sett, P. (1998) Long-term survival in spinal Liu, L.H., Chiamvimonvat, N., Walsh, R.A., Shull, G.E. and
cord injury: a fifty year investigation. Spinal Cord, 36: Periasamy, M. (2000) Disruption of a single copy of the SE-
266–274. RCA2 gene results in altered Ca2+ homeostasis and card-
Frankel, H.L., Mathias, C.J. and Spalding, J.M. (1975) Mech- iomyocyte function. J. Biol. Chem., 275: 38073–38080.
anisms of reflex cardiac arrest in tetraplegic patients. Lancet, Jose, A.D. (1966) Effect of combined sympathetic and para-
13: 1183–1185. sympathetic blockade on heart rate and cardiac function in
Frattola, A., Parati, G., Cuspidi, C., Albini, F. and Mancia, G. man. Am. J. Cardiol., 18: 476–478.
(1993) Prognostic value of 24-hour blood pressure variability. Julius, S., Palantini, P. and Nesbitt, S.D. (1998) Tachycardia:
J. Hypertens., 11: 1133–1137. an important determinant of coronary risk in hypertension.
Frishman, W.H. (1992) Beta-adrenegic blockers as cardiopro- J. Hypertens. Suppl., 16: S9–S15.
tective agents. Am. J. Cardiol., 70: 2I–6I. Julius, S. and Valentini, M. (1998) Consequences of the in-
Glaser, R.M. (1985) Exercise and locomotion for the spinal creased autonomic nervous drive in hypertension, heart fail-
cord injured. Exerc. Sport. Sci. Rev., 13: 263–303. ure and diabetes. Blood Pressure., 3(Suppl): 5–13.
Golden, K.L., Fan, Q.I., Chen, B., Ren, J., O’Connor, J. and Karlsson, A.K. (1999) Insulin resistance and sympathetic func-
Marsh, J.D. (2000) Adrenergic stimulation regulates Na(+)/ tion in high spinal cord injury. Spinal Cord, 37: 494–500.
Ca(2+) exchanger expression in rat cardiac myocytes. Karlsson, A.K., Friberg, P., Lonnroth, P., Sullivan, L. and
J. Mol. Cell. Cardiol., 32: 611–620. Elam, M. (1998) Regional sympathetic function in high spi-
Golden, K.L., Ren, J., Dean, A. and Marsh, J.D. (2002) Nor- nal cord injury during mental stress and autonomic dysre-
epinephrine regulates the in vivo expression of the L-type flexia. Brain, 121: 1711–1719.
calcium channel. Mol. Cell. Biochem., 236: 107–114. Katona, P.G., McLean, M., Dighton, D.H. and Guz, A. (1982)
Golden, K.L., Ren, J., O’Connor, J., Dean, A., DiCarlo, S.E. Sympathetic and parasympathetic cardiac control in athletes
and Marsh, J.D. (2001) In vivo regulation of Na/Ca ex- and nonathletes at rest. J. Appl. Physiol., 52: 1652–1657.
changer expression by adrenergic effectors. Am. J. Physiol. Katz, A.M. (1992) Physiology of the Heart. Raven Press,
Heart Circ. Physiol., 280: H1376–H1382. New York.
Greenhoot, J.H., Shiel, F.O. and Mauck, H.P.J. (1972) Exper- Kinzer, S.M. and Convertino, V.A. (1989) Role of leg
imental spinal cord injury. Electrocardiographic abnormali- vasculature in the cardiovascular response to arm work in
ties and fuchsinophilic myocardial degeneration. Arch. wheelchair-dependent populations. Clin. Physiol., 9:
Neurol., 26: 524–529. 525–533.
Guttmann, L., Frankel, H.L. and Paeslack, V. (1965) Cardiac Krassioukov, A.V. and Weaver, L.C. (1995) Episodic hyper-
irregularities during labour in paraplegic women. Paraplegia, tension due to autonomic dysreflexia in acute and chronic
3: 144–151. spinal cord-injured rats. Am. J. Physiol. Heart Circ. Physiol.,
Guttmann, L. and Whitteridge, D. (1947) Effects of bladder 268: H2077–H2083.
distension on autonomic mechanisms after spinal cord inju- Le, C.T. and Price, M. (1982) Survival from spinal cord injury.
ries. Brain, 70: 361–404. J. Chronic Dis., 35: 487–492.
Hjeltnes, N. (1977) oxygen uptake and cardiac output in graded Leaf, D.A., Bahl, R.A. and Adkins, R.H. (1993) Risk of cardiac
arm exercise in paraplegics with low level spinal lesions. dysrhythmias in chronic spinal cord injury patients. Paraple-
Scand. J. Rehabil. Med., 9: 107–113. gia, 31: 571–575.
Hoffman, M.D. (1986) Cardiorespiratory fitness and training in Lee, B.Y., Karmakar, M.G., Herz, B.L. and Sturgill, R.A.
quadriplegics and paraplegics. Sports Med., 3: 312–330. (1995) Autonomic dysreflexia revisited. J. Spinal Cord Med.,
Hooker, S.P. and Wells, C.L. (1989) Effects of low- and mod- 18: 75–87.
erate-intensity training in spinal cord-injured persons. Med. Lehmann, K.G., Lane, J.G., Piepmeier, J.M. and Batsford,
Sci. Sports Exerc., 21: 18–22. W.P. (1987) Cardiovascular abnormalities accompanying
287

acute spinal cord injury in humans: incidence, time course Piepmeier, J.M., Lehmann, K.B. and Lane, J.G. (1985) Cardi-
and severity. J. Am. Coll. Cardiol., 10: 46–52. ovascular instability following acute cervical spinal cord
Lehmann, K.G., Shandling, A.H., Yusi, A.U. and Froelicher, trauma. Cent. Nerv. Syst. Trauma, 2: 153–160.
V.F. (1989) Altered ventricular repolarization in central sym- Prakash, M., Raxwal, V., Froelicher, V.F., Kalisetti, D., Vieira,
pathetic dysfunction associated with spinal cord injury. Am. A., O’Mara, G., Marcus, R., Myers, J., Kiratli, J. and
J. Cardiol., 63: 1498–1504. Perkash, I. (2002) Electrocardiographic findings in patients
Levi, R., Hultling, C. and Seiger, A. (1995) The Stockholm with chronic spinal cord injury. Am. J. Phys. Med. Rehabil.,
spinal cord injury study. 3. Health-related issues of the 81: 601–608.
Swedish annual level-of-living survey in SCI subjects and Rizzoni, D., Muiesan, M.L., Montani, G., Zulli, R., Calebich,
controls. Paraplegia, 33: 726–730. S. and Agabiti-Rosei, E. (1992) Relationship between initial
Loewy, A. and Spyer, K. (1990) Vagal preganglionic neurons. cardiovascular structural changes and daytime and nighttime
Central Regulation of Autonomic Functions. Oxford Uni- blood pressure monitoring. Am. J. Hypertens., 5: 180–186.
versity Press, New York, pp. 68–87. Rodenbaugh, D.W., Collins, H.L. and DiCarlo, S.E. (2003a)
Maiorov, D.N., Weaver, L.C. and Krassioukov, A.V. (1997) Paraplegia differentially increases arterial blood pressure re-
Relationship between sympathetic activity and arterial pres- lated cardiovascular disease risk factors in normotensive and
sure in conscious spinal rats. Am. J. Physiol. Heart Circ. hypertensive rats. Brain Res., 980: 242–248.
Physiol., 272: H625–H631. Rodenbaugh, D.W., Collins, H.L. and DiCarlo, S.E. (2003b)
Marcus, R.R., Kalisetti, D., Raxwal, V., Kiratli, B.J., Myers, J., Increased susceptibility to ventricular arrhythmias in hyper-
Perkash, I. and Froelicher, V.F. (2002) Early repolari- tensive paraplegic rats. Clin. Exp. Hypertens., 25: 349–358.
zation in patients with spinal cord injury: prevalence and Rodenbaugh, D.W., Collins, H.L., Nowacek, D.G. and
clinical significance. J. Spinal Cord Med., 25: 33–38 discus- DiCarlo, S.E. (2003c) Increased susceptibility to ventricular
sion 39. arrhythmias is associated with changes in Ca2+ regulatory
Mathias, C.J. (1976) Bradycardia and cardiac arrest during proteins in paraplegic rats. Am. J. Physiol. Heart Circ.
tracheal suction — mechanisms in tetraplegic patients. Eur. J. Physiol., 285: H2605–H2613.
Intensive Care Med., 2: 147–156. Rosen, M.R. and Reder, R.F. (1981) Does triggered activity
Mathias, C.J. and Frankel, H.L. (1992) The cardiovascular have a role in the genesis of cardiac arrhythmias? Ann. In-
system in tetraplegia and paraplegia. In: Frankel H.L. (Ed.), tern. Med., 94: 794–801.
Handbook of Clinical Neurology. Elsevier Science Publish- Schwartz, P.J., Priori, S.G. and Napolitano, C. (1993) Role of
ers, Amsterdam, pp. 435–456. the Autonomic Nervous System in Sudden Cardiac Death.
Mayorov, D.N., Adams, M.A. and Krassioukov, A.V. (2001) Sudden Cardiac Death. Blackwell Scientific Publications,
Telemetric blood pressure monitoring in conscious rats Cambridge, MA, pp. 16–37.
before and after compression injury of spinal cord. Schwartz, P.J. and Stone, H.L. (1980) Left stellectomy in the
J. Neurotrauma, 18: 727–736. prevention of ventricular fibrillation caused by acute myo-
McGuire, T.J. and Kumar, V.N. (1986) Autonomic dysreflexia cardial ischemia in conscious dogs with anterior myocardial
in the spinal cord injured: what the physician should know infarction. Circulation, 62: 1256–1265.
about this medical emergency. Postgrad. Med., 80: 81–89. Schwartz, P.J. and Stone, H.L. (1982) The role of the auto-
McKinley, W.O., Jackson, A.B., Cardenas, D.D. and DeVivo, nomic nervous system in sudden coronary death. Ann. NY
M.J. (1999) Long-term medical complications after traumatic Acad. Sci., 382: 162–180.
spinal cord injury: a regional model systems analysis. Arch. Soden, R.J., Walsh, J., Middleton, J.W., Craven, M.L.,
Phys. Med. Rehabil., 80: 1402–1410. Rutkowski, S.B. and Yeo, J.D. (2000) Causes of death after
Morady, F., Nelson, S.D., Kou, W.H., Pratley, R., Schmaltz, spinal cord injury. Spinal Cord, 38: 604–610.
S., De Buitleir, M. and Halter, J.B. (1988) Electrophysiologic Stamler, J., Stamler, R. and Neaton, J.D. (1993) Blood pres-
effects of epinephrine in humans. J. Am. Coll. Cardiol., 11: sure, systolic and diastolic, and cardiovascular risks. Arch.
1235–1244. Intern. Med., 153: 598–615.
Murray, K.T. and Roden, D.M. (1996) Cardiac arrhythmias Stein, R., Medeiros, C.M., Rosito, G.A., Zimerman, L.I. and
and sudden death. In: Robertson D., Low P.A. and Polinsky Ribeiro, J.P. (2002) Intrinsic sinus and atrioventricular node
R.J. (Eds.), Primer on the Autonomic Nervous System. Ac- electrophysiologic adaptations in endurance athletes. J. Am.
ademic Press, San Diego, CA, pp. 149–152. Coll. Cardiol., 39: 1033–1038.
Naftchi, N.E. (1990) Mechanism of autonomic dysreflexia: Stevenson, E.T., Davy, K.P., Jones, P.P., Desouza, C.A. and
contributions of catecholamine and peptide neurotransmit- Seals, D.R. (1997) Blood pressure risk factors in healthy
ters. Ann. NY Acad. Sci., 579: 133–148. postmenopausal women: physical activity and hormone re-
Palatini, P. and Julius, S. (1997) Association of tachycardia placement. J. Appl. Physiol., 82: 652–660.
with morbidity and mortality: pathophysiological consider- Stiens, S., Johnson, M. and Lyman, P. (1995) Cardiac rehabil-
ations. J. Hum. Hypertens., 11(Suppl 1): S19–S27. itation in patients with spinal cord injury. Phys. Med. Re-
Palatini, P. and Julius, S. (1999) Relevance of heart rate as a habil. Clin. N. Am., 6: 263–295.
risk factor in hypertension. Curr. Hypertens. Rep., 1: Such, L., Rodriguez, A., Alberola, A., Lopez, L., Ruiz, R.,
219–224. Artal, L., Pons, I., Pons, M.L., Garcia, C. and Chorro, F.J.
288

(2002) Intrinsic changes on automatism, conduction, and re- persons spinal cord injured more than 20 years ago. Para-
fractoriness by exercise in isolated rabbit heart. J. Appl. plegia, 30: 617–630.
Physiol., 92: 225–229. Wicks, J.R., Oldridge, N.B., Cameron, N.B. and Jones, N.L.
Washburn, R.A. and Figoni, S.F. (1998) Physical activity and (1983) Arm cranking and wheelchair ergometry in elite
chronic cardiovascular disease prevention in spinal cord in- spinal cord injured athletes. Med. Sci. Sports Exerc., 15:
jury: a comprehensive literature review. Top. Spinal Cord 224–231.
Injury Rehabil., 3: 16–32. Zimmet, P.Z., Collins, V.R., Dowse, G.K., Alberti, K.G.,
Wharton, J.M., Coleman, R.E. and Strauss, H.C. (1992) The Tuomilehto, J., Gareeboo, H. and Chitson, P. (1991) The
role of the autonomic nervous system in sudden cardiac relation of physical activity to cardiovascular disease risk
death. Trends Cardiovas. Med., 2: 65–71. factors in Mauritians. Mauritius Noncommunicable Disease
Whiteneck, G.G., Charlifue, S.W., Frankel, H.L., Fraser, Study Group. Am. J. Epidemiol., 134: 862–875.
M.H., Gardner, B.P., Gerhart, K.A., Krishnan, K.R., Zipes, D.P. (1991) The long QT interval syndrome. A Rosetta
Menter, R.R., Nuseibeh, I., Short, D.J. and Silver, J.R. stone for sympathetic related ventricular tachyarrhythmias
(1992) Mortality, morbidity, and psychosocial outcomes of [comment]. Circulation, 84: 1414–1419.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 19

Adaptations of peripheral vasoconstrictor pathways


after spinal cord injury

Elspeth M. McLachlan and James A. Brock

Spinal Injuries Research Centre, Prince of Wales Medical Research Institute, Gate 1, Barker Street, Randwick,
NSW 2031, Australia

Abstract: The consequences of spinal cord injury on the function of sympathetic pathways in the periphery
have generally been ignored. We discuss two types of plasticity that follow disruption of sympathetic
pathways in rats. The first relates to the partial denervation of sympathetic ganglia that would follow the
loss of some preganglionic neurones. Sprouting of residual connections rapidly reinnervates many post-
ganglionic neurones, restoring functional transmission within a few weeks, but other neurones may be
permanently decentralized. Some of the new functional connections may generate inappropriate pathways
leading to abnormal reflexes. The second type of plasticity concerns the markedly enhanced and prolonged
contractile responses to nerve activity in arterial vessels to which ongoing sympathetic activity has been
reduced or silenced following spinal cord transection or ganglion decentralization. In a cutaneous artery
(the rat tail artery), the mechanisms underlying this arterial hyperreactivity differ from those in the
splanchnic arteries (the rat mesenteric artery). In the former, hyperreactivity is mainly postjunctional but
independent of changes in a1 -adrenoceptor sensitivity, whereas the increased responsiveness in the latter
vessels can be attributed to a greater responsiveness to a1 -adrenoceptor activation. There are enough data
from humans to suggest that both of these novel findings in experimental animals are likely to apply after
spinal cord injury and contribute to autonomic dysreflexia .

The disconnection of sympathetic vasoconstrictor baroreceptor control makes it difficult for people
pathways from brainstem control that can follow with spinal injury to tolerate sitting up quickly
severe spinal cord injury leads initially to a pro- from a supine position, particularly if most of the
found fall in resting arterial blood pressure that sympathetic preganglionic outflow is uncontrolled,
gradually recovers. The long-term adjustment is i.e. when the lesion is above the sixth thoracic
thought to occur via endocrine mechanisms, such segment (T6). It has generally been found that the
as the renin–angiotensin system and anti-diuretic involvement of the splanchnic vascular bed is crit-
hormone. The fall in blood pressure presumably ical, such that reflex recovery of blood pressure is
reflects a marked decrease in sympathetic activity adequate provided the resistance of the splanchnic
in vasoconstrictor axons (Wallin and Stjernberg, component can be regulated through the bar-
1984; Stjernberg et al., 1986). In the absence of oreceptors. The innervation of the splanchnic bed
sympathetic vasoconstrictor tone, mean resting largely arises below T6.
arterial pressure is 70 mmHg when the subject A secondary consequence of severe spinal inju-
is upright (Lee et al., 1995). The loss of reflex ries above T6 is the development of autonomic
dysreflexia , an inappropriate and massive sympa-
Corresponding author. Tel.: +61 2 9399 1031; Fax: thetic response to activation of nociceptive and
+61 2 9399 1034; E-mail: e.mclachlan@unsw.edu.au non-nociceptive afferents from viscera and skin.

DOI: 10.1016/S0079-6123(05)52019-3 289


290

When there are large bladder or colon distensions, consideration. Spinal cord injury not only dener-
undetected injuries or pressure sores, the arterial vates preganglionic neurones below the injury but,
pressure can rise excessively because reflex com- if the lesion is in the thoracic or upper lumbar seg-
pensation by vasodilation in the upper body and ments, is also likely to destroy preganglionic neu-
bradycardia is limited. Systolic blood pressure can rones that are located at the site of the injury or in
peak at over 300 mmHg (Lee et al., 1995). Reflex the surrounding area involved in secondary degen-
sweating may also occur that is not present in in- eration. While the loss of descending excitatory in-
tact individuals. Although removal of the stimulus put to preganglionic neurones below the level of the
will alleviate the dangerous hypertension , mean lesion silences the tonic activity of postganglionic
blood pressure remains above resting levels for
many minutes. This condition can be life-threaten-
ing and normally requires hospitalization; death by
stroke is not unusual when this cannot be achieved.
While it is widely believed that the basis for au-
tonomic dysreflexia is the loss of baroreflex com-
pensation, changes in the injured spinal cord are
now also thought to contribute. Peptidergic noc-
iceptor afferents (expressing calcitonin gene-relat-
ed peptide) have been reported to sprout within
the dorsal horn of thoracic segments below a se-
vere spinal injury at T4–T5 in rats that exhibit
autonomic dysreflexia (Krenz and Weaver, 1998;
Weaver et al., 2001). Both the sprouting and the
hypertensive response to colon distension are at-
tentuated by intrathecal administration of anti-
body to nerve growth factor (Krenz et al., 1999).
Rearrangement of synaptic connections below the
lesion, particularly an increase in peptidergic
synapses, might contribute to enhanced reflex
sympathetic excitation by noxious visceral disten-
sion. More recently, substantial reductions in the
abnormal hypertensive reflexes in rats were
achieved by administering antibodies to the
CD11d subunit of the CD11d/CD18 integrin (Gris
et al., 2004). This treatment blocks the infiltration
of neutrophils and haematogenous monocytes/ma-
crophages at the site of the injury during the first 2 Fig. 1. Diagrammatic representation of the connections in the
days and reduces free radical formation and the lower lumbar sympathetic paravertebral ganglia in the guinea
extent of secondary damage, resulting in improved pig. In the control situation, preganglionic axons arising in
several lumbar segments (grey) project to the ganglia associated
motor, sensory and autonomic function.
with segments below the lowest white ramus (at L4). In L5
paravertebral ganglion , each ganglion cell (black) receives in-
Changes in ganglionic transmission after loss of puts from several of these segments. When the paravertebral
preganglionic neurones chain was transected above L4 paravertebral ganglion (L4 on-
ly), only the preganglionic axons arising in L4 remained, leaving
many ganglion cells denervated (white). After 4–6 weeks, the L5
The possible role of changes in the peripheral
ganglion cells became reinnervated by L4 collaterals. As a re-
pathways from the spinal cord as a contribut- sult, signals from L4 preganglionic neurones would be trans-
ing factor in the generation of heightened and mitted to the targets of neurones previously not innervated by
abnormal autonomic responses has received little this spinal segment.
291

vasoconstrictor neurones, the destruction of pre-


ganglionic neurones will partially denervate the pe-
ripheral ganglia in which they synapse.
The consequences of partial denervation can be
predicted from experiments in which the removal
of some preganglionic inputs led to sprouting and
reinnervation within sympathetic paravertebral
ganglia (Murray and Thompson, 1957; Maehlen
and Nja, 1984; Liestol et al., 1987). In a recent
electrophysiological study of synaptic transmission
in lower lumbar sympathetic ganglia of guinea
pigs, the changes in connectivity were analyzed
following transection of the lumbar trunk just
proximal to the last white ramus at the fourth
lumbar segment (L4) (Ireland, 1999). This lesion
removes all preganglionic axons (inputs) arising
from the cell bodies in T13 to L3 spinal segments
that project to the L5 ganglion , leaving only a Fig. 2. Frequency distributions of the number of preganglionic
small component (10%) of the original pregangl- inputs received by ganglion cells in L5 paravertebral ganglia
ionic input (Fig. 1, middle panel). The responses of under the three conditions illustrated in Fig. 1. The number of
‘‘weak’’ or subthreshold (hatched columns) and ‘‘strong’’ or sup-
the postganglionic neurones in L5 ganglion to
rathreshold (black columns) inputs is shown, together with the
stimulation of all preganglionic axons (in the lum- frequency that these inputs were not detected (white columns). In
bar sympathetic trunk immediately rostral to the the control situation (upper panels), usually one strong and sev-
ganglion ) were recorded with intracellular micro- eral weak inputs are received by the ganglion cells, arising from
electrodes. These responses were recorded at 2–3 several spinal segments. Only the strong synapses transmit sig-
nals to the peripheral target. Inputs that arise from L4 only
days after cutting the chain, when the terminals of
(middle panels) are mainly weak and many ganglion cells ini-
the preganglionic axons originating proximally to tially lack any inputs after the paravertebral chain is transected.
the L4 segment would have degenerated, and at Surprisingly, when the ganglion cells became reinnervated by L4
4–5 weeks after cutting the chain when the re- collaterals after sprouting of the residual L4 preganglionic axons
maining L4 inputs would have sprouted within the (lower panels), most of the neurones received a strong input,
indicating that these were formed preferentially. Figure modified
ganglion. These synaptic responses were compared
from Fig. 3 in Ireland (1999).
to the responses in control ganglia.
Under control conditions (Fig. 1, upper panel),
each neurone in the L5 paravertebral ganglia re- 10% of L5 postganglionic neurones, one of the
ceives, on average, 4 preganglionic cholinergic inputs from the pool of preganglionic neurones in
inputs of which at least one is usually suprathresh- L4 was strong whereas 40% of the neurones re-
old or ‘‘strong’’ (Fig. 2, upper panels). The number ceived only weak inputs from L4, and there was no
of quanta of acetylcholine released from this L4 input at all to the remaining 50% of neurones
strong preganglionic axon is large enough to en- (Fig. 2, middle panels). The summed synaptic con-
sure that ganglionic transmission (i.e. initiation of ductance change produced by stimulating all L4
the postganglionic action potential) occurs with a inputs was only 26% of the total received from the
high safety factor, as at the skeletal neuromuscular entire preganglionic outflow.
junction. The other, subthreshold or ‘‘weak,’’ in-
puts generate post-synaptic potentials of only a Sprouting of residual preganglionic axons rapidly
few mV in amplitude that do not activate the cell. reinnervates many postganglionic neurones
That is, ganglionic transmission normally occurs
only by direct relay of the signal carried by the One month after transecting the paravertebral
strong input. Two to three days after section, in chain between L3 and L4 ganglia, the responses
292

recorded in the L5 postganglionic neurones had come reinnervated would depend on the extent to
substantially recovered, reaching 80% of the which individual preganglionic neurones are re-
original summed synaptic conductance. This was stricted in the number of additional synapses they
achieved both by the formation of new synapses can support, i.e. whether the size of the sympa-
and by the enlargement of existing ones. Indeed, thetic ‘‘neural unit’’ (Purves and Wigston, 1983) is
only 22% of neurones remained denervated where- limited. The neural unit is the equivalent of a mo-
as 60% of neurones had a strong input of normal tor unit, being the number of postganglionic neu-
magnitude (Fig. 2, lower panel). The probability rones with which a single preganglionic neurone
was 3.5 times higher that the collateral branch of synapses. Thus, the remaining L4 preganglionic
an L4 axon formed a strong, rather than a weak, neurones might not have the capacity to take over
input. This implies that the postganglionic neu- control of all denervated postganglionic neurones.
rones send a powerful signal to whichever pre- To summarize, the evidence in animal experi-
ganglionic terminals make contact so that one of ments indicates that rapid sprouting of remaining
them develops into a strong synapse. From these sympathetic preganglionic terminals in ganglia re-
data, it is evident that, when postganglionic neu- stores transmission to many postganglionic neu-
rones lose some inputs after spinal injury, sprout- rones that have lost their strong inputs when
ing of intact preganglionic inputs caudal to the preganglionic neurones are damaged. Thus, post-
injury can quickly restore ganglionic transmission. ganglionic neurones denervated by spinal cord dam-
age may soon be reconnected with the cord below
the lesion and can participate at least in spinal re-
New connections in ganglia may be inappropriate flexes. The potential for functionally inappropriate
reconnections may explain some of the abnormal
Because the preganglionic inputs that sprouted to reflex responses observed in cord-injured people.
reinnervate the partially denervated ganglion arose
only from L4 and supplied neurones normally not
innervated from this segment, it is likely that some Changes in neurovascular transmission after spinal
of the strong (i.e. effective) synapses that devel- transection
oped in the first few weeks after the injury were
formed on functionally incorrect postganglionic Another contributing factor to autonomic dysre-
neurones. Preganglionic axons from segments con- flexia might reside in the properties of the arterial
taining the original source of innervation will dis- resistance vessels after spinal cord injury. Subjects
place the erroneous sprouts if they are able to with cervical spinal lesions have increased pressor
reenter the ganglion (Murray and Thompson, responses to intravenously infused noradrenaline
1957; Liestol et al., 1987) although, even after dis- (Mathias et al., 1976; Krum et al., 1992). While
placement of the sprouted connections, the degree this change is likely to reflect, in part, loss of bar-
of reinnervation is poor compared with that fol- oreceptor reflexes (Mathias et al., 1976), it has
lowing reinnervation after complete denervation been suggested that increased reactivity of the vas-
(Maehlen and Nja, 1984). However, if the original cular smooth muscle to a-adrenoceptor activation
preganglionic neurones have been destroyed, the contributes to the augmented pressor responses to
potential for correction of inappropriate novel noradrenaline (Krum et al., 1992). Consistent with
connections is lost. this idea, the foot veins of quadriplegic patients
It is not clear whether some postganglionic neu- with recurrent bouts of autonomic dysreflexia have
rones may have remained permanently decentral- a markedly increased sensitivity to locally infused
ized if the survival period in the above experiments noradrenaline (Arnold et al., 1995). This interven-
had been longer, but clearly, in humans, this could tion produced marked local venoconstriction in
follow spinal injuries that destroyed part of the the absence of baroreflex involvement.
thoracolumbar cord. The likelihood that all the Because the responses to exogenous agonists
denervated postganglionic neurones would be- are often unlike the responses to nerve-released
293

transmitter (Hirst et al., 1992), it seems more rel- outflow to the tail artery (T13–L2) (Rathner and
evant to identify whether responses to nerve ac- McAllen, 1998). Segments of tail artery about 1 cm
tivity are enhanced. The responses of cutaneous long were dissected from the animals and studied
vessels to brief bursts of vasoconstrictor nerve ac- in vitro using intracellular microelectrodes inserted
tivity elicited by increasing bladder pressure, or by in the vascular smooth muscle.
electrical stimulation of the skin of the contralat- In control tail arteries, stimulation of the peri-
eral leg, led to vasoconstrictions lasting over 30 s in vascular nerves evokes an excitatory junction po-
subjects with cervical or high thoracic spinal inju- tential in the vascular smooth muscle cells (Fig. 3a,
ries. In intact individuals responses to similar upper record). Following a train of five such ex-
bursts lasted only 10 s (Wallin and Stjernberg, citatory junction potentials evoked at 1 Hz, a slow
1984). These observations suggest that spinal dam- depolarization develops, which peaks at 15 s and
age modifies the response of cutaneous vessels to lasts for 1 min (Fig. 3a, lower record). The exci-
sympathetic activity in such a way that neurogenic tatory junction potential is blocked by purinergic
vasoconstriction is prolonged. receptor (P2X) antagonists such as suramin and so
is likely to be due to the release of adenosine 50 -
triphosphate. The slow depolarization is blocked
Transmitter release may be increased in tail arteries by a2 -adrenoceptor antagonists such as idazoxan,
after spinal cord transection and reflects the release of noradrenaline .
When these signals were recorded in arteries
By examining the electrophysiological responses in from animals at 2 months after spinal cord tran-
the smooth muscle of the isolated rat -tail artery to section, the amplitude of both the excitatory junc-
stimulation of the perivascular nerves , we have tion potentials and the slow depolarizations
obtained direct evidence that sympathetic neuro- evoked by supramaximal electrical stimuli were
vascular transmission is enhanced after spinal cord about double those recorded in vessels from age-
injury. The spinal cord of female Wistar rat was matched control animals (Fig. 3b). In addition,
transected at T7–T8, well above the preganglionic the frequency of spontaneous excitatory junction

Fig. 3. Spinal transection at T7–T8 increases the amplitude of intracellularly recorded excitatory junction potentials (EJP) and the
slow noradrenaline -induced depolarization (NAD) in a proximal segment of the rat -tail artery. Tissues were studied 2 months post-
operatively. (a) Traces showing the averaged response to 5 pulses at 1 Hz. The upper trace is the first part of the lower record shown on
an expanded time base. (b) EJP and NAD amplitude for arteries isolated from sham-operated rats (white columns, n ¼ 11) and rats
after spinal cord transection (hatched columns, n ¼ 13) rats , *Po0:01;**Po0:001; unpaired t-test.
294

potentials (thought to reflect the release of indi- cord-injured subjects, the contractile responses to
vidual quanta of adenosine triphosphate) was in- nerve stimulation were prolonged in time course
creased in recordings from many preparations relative to control (Fig. 4a). Taken together, these
after spinal cord transection. The simplest expla- data show that the rat -tail artery quite rapidly
nation of these findings is that transmitter release becomes more reactive to nerve activity after spi-
from the perivascular terminals is increased. nal transection and that this hyperreactivity is
maintained.
The degree of block of nerve-evoked responses
Nerve-evoked contractions are enhanced in tail produced by supramaximal concentrations of a-
arteries from rats with spinal cord transection antagonists was less (88%) than in controls
(95%) (Yeoh et al., 2004a). This suggests a greater
The more dramatic change observed in the tail ar- role for co-transmitters, such as adenosine tripho-
tery of rats after spinal cord transection was the sphate, after spinal transection. However, in com-
increase in the contractile responses evoked by parison with control arteries, the effectiveness of
nerve stimulation. Contractile responses to brief partially blocking concentrations of the competi-
trains of perivascular stimuli were recorded from tive a1 - and a2 -adrenoceptors antagonists, prazo-
isolated ring segments of tail artery mounted in a sin and idazoxan, was markedly lower in arteries
myograph (Yeoh et al., 2004a). Two weeks and 2 from cord-injured animals. This finding may be
months following spinal cord transection, brief explained by an increase in the amount of nor-
trains of supramaximal electrical stimuli at adrenaline released per impulse from the perivas-
0.1–10 Hz applied transmurally led to contractions cular axons following spinal transection.
that were up to 25-fold larger after spinal cord Much of the enhancement of the responses to
transection than in control arteries (Fig. 4a). The nerve stimulation occurred postjunctionally, be-
degree of enhancement was more marked during cause the amplitude of contractions to depolari-
trains at lower stimulation frequencies (0.1 Hz). zation evoked by applying saline containing 60
Further, consistent with Wallin and Stjernberg’s mM K+ almost doubled (Yeoh et al., 2004a). Like
observations in the cutaneous vascular beds of the responses to nerve stimulation, the duration of

Fig. 4. Spinal transection markedly increases nerve-evoked contractions of both (a) rat -tail artery and (b) rat mesenteric artery. The
spinal cord was transected at T7–T8 for the tail artery experiments and at T4 for the mesenteric artery experiments. Tissues were
studied 2 months post-operatively. Both panels show traces for arteries isolated from a sham-operated rat (upper) and a cord-
transected rat (lower). In (a), the tail artery was stimulated with 25 pulses at 0.1, 0.3 and 0.5 Hz and 100 pulses at 1 Hz. In (b), the
mesenteric artery was stimulated with 100 pulses at 1, 2, 3 and 5 Hz.
295

the contraction to increased [K+] was also pro- co-transmitters in the decentralized arteries, as the
longed. The arteries also showed increased sensi- blockade of the nerve-evoked contraction by sup-
tivity to the a2 -adrenoceptor agonist, clonidine, ramaximal concentrations of a-adrenoceptor an-
and a transient increase in sensitivity to the a1 - tagonists was similar to control (i.e. 95%
adrenoceptor agonist, phenylephrine. After 2 blockade).
months, however, the marked increase in the
nerve-evoked response was maintained but was
not associated with enhanced a1 -adrenoceptor sen- Nerve-evoked responses in mesenteric arteries are
sitivity. Interestingly, as contractions to both high also potentiated after spinal transection
[K+] and clonidine are dependent on extracellular
Ca2+ (Abe et al., 1987; Chen and Rembold, 1995), More recent experiments have confirmed that re-
the augmentation of contraction to both these sponses of second-order mesenteric arteries from
agents may indicate that the contractile mecha- rats with spinal cord transection at T4 are en-
nism is selectively sensitized to Ca2+ entering the hanced in a manner similar to the responses of tail
cell or that there in an increase in stimulus-induced arteries from rats with spinal cord transection
Ca2+ entry. (Fig. 4b). Mesenteric vessels are normally control-
led by preganglionic neurones that project from
T4 to T13 (Anderson et al., 1989; Taylor and
Decentralization mimics spinal cord transection in Weaver, 1992). In the rat mesenteric artery, the
enhancing vascular reactivity contractile responses to supramaximal electrical
stimulation of the sympathetic axons are very
The underlying mechanism for the hyperreactivity small so that the lowest frequency of stimulation
after spinal transection might be related to the re- that can be evaluated in vitro is 1 Hz.The potent-
duced activity of sympathetic postganglionic ax- iation of mesenteric artery contraction at 1 Hz was
ons. We tested this idea by silencing the relatively greater (8-fold) than that of the tail ar-
sympathetic outflow to the tail by cutting the lum- tery (1.5-fold) (Fig. 4). Unlike for the tail artery,
bar sympathetic chain below L3, i.e. decentralizing the change in the nerve-evoked responses of the
the postganglionic neurones supplying the tail ar- mesenteric arteries was closely correlated with an
tery that are located in the sacral sympathetic increased sensitivity to a1 -adrenoceptor agonists.
chain (Sittiracha et al., 1987). In the tail arteries Further, the responses of the mesenteric artery to
from these animals, the responses to stimulation of depolarization with increased extracellular [K+]
the perivascular nerves were enhanced and pro- were not larger. Thus, the enhancement of neuro-
longed, mimicking the changes after spinal tran- vascular transmission in the mesenteric artery ap-
section (Yeoh et al., 2004b). In this case, enhanced pears to be due primarily to an increased
responses to nerve stimulation were observed 2 responsiveness to a1 -adrenoceptor activation.
days following decentralization, they had in- These findings indicate that the mechanisms
creased after 2 weeks and were maintained for at underlying the augmented responses differ bet-
least 7 weeks. ween the cutaneous and splanchnic vascular beds.
The data indicate that the vascular changes after These differences probably reflect the equally
decentralization and spinal cord transection are distinctive mechanisms by which sympathetic
not identical (Yeoh et al., 2004a, b). At two weeks, vasoconstrictor nerves normally activate smooth
the augmentation of nerve-evoked responses was muscle of these two vascular beds. In the tail
consistently larger after decentralization than it artery, both a1 - and a2 -adrenoceptors contribute
was after cord transection. This difference may to the nerve-evoked contraction in vitro and
reflect residual vasoconstrictor nerve activity to co-transmitters play only a minor role (Bao
arteries from cord-injured rats , evoked reflexly et al., 1993; Brock et al., 1997; Yeoh et al.,
from afferent input below the lesion. In addition, 2004b). In contrast, in the mesenteric vascular bed,
there was no evidence for an increased role of nerve-released noradrenaline produces contraction
296

through activation of a1 -adrenoceptors , while tivity, which is debated (Silver, 2000), the changes
both adenosine triphosphate and neuropeptide Y in the vascular muscle produced by abolishing
appear to contribute a significant component (Do- normal ongoing nerve activity by spinal cord tran-
noso et al., 1997; Han et al., 1998). In other ar- section or decentralization are evident within a few
terial vessels, the mechanisms underlying neurally days. Damage to preganglionic neurones that par-
evoked contraction differ from those in either the tially denervates vasoconstrictor neurones might
tail or the mesenteric artery. For example, con- silence them and also rapidly result in hyperreac-
tractions of guinea-pig submucosal arterioles are tivity of the vasculature. Whether or not this
entirely due to neurally released adenosine tripho- would be reversed when the postganglionic neu-
sphate (Evans and Surprenant, 1992). Further- rones become reinnervated in the ensuing weeks is
more, in arteries like the mesenteric artery that are unclear, but it is likely that some vasoconstrictor
innervated by a perivascular plexus of afferent ax- innervation remains chronically inactive.
ons, it is not clear whether ongoing activity in There is no doubt that changed responsiveness
these peptidergic terminals has an effect on the of the vasculature is likely to contribute to the
behaviour of the vascular smooth muscle. There- prolonged vasoconstriction that occurs when sym-
fore, the effects of silencing ongoing sympathetic pathetic activity is evoked below a spinal injury
nerve activity on hyperreactivity are likely to differ (Wallin and Stjernberg, 1984; Teasell et al., 2000).
between vascular beds. Similar increases in vascular reactivity may well
occur in most types of blood vessel below a spinal
injury, although the underlying mechanisms may
Conclusions differ. What is less clear is whether vascular hyper-
reactivity contributes to other problems in patients
The result of our experiments clearly show signif- such as inadequate temperature regulation and the
icant changes in the peripheral components of the poor healing of pressure sores. Our data imply that
sympathetic pathways involved in autonomic sympathetic nerve activity modulates the process
dysreflexia . of neurovascular transmission and the behaviour
(i) The experiments in guinea pigs imply that of the vascular smooth muscle itself. Until more is
any loss of preganglionic neurones that occurs as a known about these processes, both in control sit-
direct or indirect result of spinal damage will den- uations and in conditions when nerve activity is
ervate postganglionic neurones and lead to cessa- modified, it will not be easy to predict which ther-
tion of their activity. This state will not persist for apeutic approaches are the most appropriate to
long, as sprouting of residual intact terminals ameliorate the functional disorders that appear
within the ganglia will soon restore strong (effec- after injury. Another question is whether non-vas-
tive) synaptic transmission to many of the dener- cular target organs in the periphery, whose pat-
vated neurones. However, this might lead to terns of autonomic nerve activity are modified
erroneous connections that could account for in- after spinal cord injury have as yet undiscovered
appropriate functional reflexes such as sweating in changes in neuroeffector function.
response to noxious stimuli.
(ii) The experiments in rats show that the re-
sponsiveness of both the tail artery (which supplies Acknowledgments
a major thermoregulatory cutaneous bed) and the
small mesenteric arteries is dramatically enhanced This work was supported by grants from the Na-
after spinal cord injury. tional Health & Medical Research Council of
These changes appear to result from the reduc- Australia (970852, 209632) and the Christopher
tion in sympathetic traffic as similar hyperreactiv- Reeve Paralysis Foundation (BAC1-0101-1,
ity follows surgical decentralization. BAC1-0101-2). We thank David Ireland and Me-
Irrespective of whether or not spinal shock re- lanie Yeoh for their major contributions to the
moves spinal reflex -evoked sympathetic nerve ac- conduct of the experiments.
297

References Krum, H., Louis, W.J., Brown, D.J. and Howes, L.G. (1992)
Pressor dose responses and baroreflex sensitivity in quadri-
Abe, K., Matsuki, N. and Kasuya, Y. (1987) Pharmacological plegic spinal cord injury patients. J. Hypertens., 10:
and electrophysiological discrimination of contractile re- 245–250.
sponses to selective a1- and a2-adrenoceptor agonists in rat Lee, B.Y., Karmakar, M.G., Herz, B.L. and Sturgill, R.A.
tail artery. Jpn. J. Pharmacol., 45: 249–261. (1995) Autonomic dysreflexia revisited. J. Spinal Cord Med.,
Anderson, C.R., McLachlan, E.M. and Srb-Christie, O. (1989) 18: 75–87.
Distribution of sympathetic preganglionic neurons and mon- Liestol, K., Maehlen, J. and Nja, A. (1987) Two types of
oaminergic nerve terminals in the spinal cord of the rat. J. synaptic selectivity and their interrelation during sprouting in
Comp. Neurol., 283: 269–284. the guinea-pig superior cervical ganglion. J. Physiol., 384:
Arnold, J.M., Feng, Q.P., Delaney, G.A. and Teasell, R.W. 233–245.
(1995) Autonomic dysreflexia in tetraplegic patients: evidence Maehlen, J. and Nja, A. (1984) Rearrangement of synapses on
for a-adrenoceptor hyper-responsiveness. Clin. Auton. Res., guinea-pig sympathetic ganglion cells after partial interrup-
5: 267–270. tion of the preganglionic nerve. J. Physiol., 348: 43–56.
Bao, J.X., Gonon, F. and Stjarne, L. (1993) Frequency- and Mathias, C.J., Frankel, H.L., Christensen, N.J. and Spalding,
train length-dependent variation in the roles of postjunctio- J.M. (1976) Enhanced pressor response to noradrenaline in
nal a1- and a2-adrenoceptors for the field stimulation-in- patients with cervical spinal cord transection. Brain, 99:
duced neurogenic contraction of rat tail artery. Naunyn 757–770.
Schmiedebergs Arch. Pharmacol., 347: 601–616. Murray, J.G. and Thompson, J.W. (1957) The occurrence and
Brock, J.A., McLachlan, E.M. and Rayner, S.E. (1997) Con- function of collateral sprouting in the sympathetic nervous
tribution of a-adrenoceptors to depolarization and contrac- system of the cat. J. Physiol., 135: 133–162.
tion evoked by continuous asynchronous sympathetic nerve Purves, D. and Wigston, D.J. (1983) Neural units in the supe-
activity in rat tail artery. Br. J. Pharmacol., 120: 1513–1521. rior cervical ganglion of the guinea-pig. J. Physiol., 334:
Chen, X.L. and Rembold, C.M. (1995) Phenylephrine contracts 169–178.
rat tail artery by one electromechanical and three pharmaco- Rathner, J.A. and McAllen, R.M. (1998) The lumbar pre-
mechanical mechanisms. Am. J. Physiol., 268: H74–H81. ganglionic sympathetic supply to rat tail and hindpaw. J.
Donoso, M.V., Steiner, M. and Huidobro-Toro, J.P. (1997) Auton. Nervous System, 69: 127–131.
BIBP 3226, suramin and prazosin identify neuropeptide Y, Silver, J.R. (2000) Early autonomic dysreflexia. Spinal Cord,
adenosine 50 -triphosphate and noradrenaline as sympathetic 38: 229–233.
cotransmitters in the rat arterial mesenteric bed. J. Pharma- Sittiracha, T., McLachlan, E.M. and Bell, C. (1987) The in-
col. Exp. Ther., 282: 691–698. nervation of the caudal artery of the rat. Neuroscience, 21:
Evans, R.J. and Surprenant, A. (1992) Vasoconstriction of 647–659.
guinea-pig submucosal arterioles following sympathetic nerve Stjernberg, L., Blumberg, H. and Wallin, B.G. (1986) Sympa-
stimulation is mediated by the release of ATP. Br. J. Pharma- thetic activity in man after spinal cord injury. Outflow to
col., 106: 242–249. muscle below the lesion. Brain, 109: 695–715.
Gris, D., Marsh, D.R., Oatway, M.A., Chen, Y., Hamilton, Taylor, R.B. and Weaver, L.C. (1992) Spinal stimulation to
E.F., Dekaban, G.A. and Weaver, L.C. (2004) Transient locate preganglionic neurons controlling the kidney, spleen,
blockade of the CD11d/CD18 integrin reduces secondary or intestine. Am. J. Physiol., 263: H1026–H1033.
damage after spinal cord injury, improving sensory, auto- Teasell, R.W., Arnold, J.M., Krassioukov, A. and Delaney,
nomic, and motor function. J. Neurosci., 24: 4043–4051. G.A. (2000) Cardiovascular consequences of loss of supra-
Han, S., Yang, C.L., Chen, X., Naes, L., Cox, B.F. and West- spinal control of the sympathetic nervous system after spinal
fall, T. (1998) Direct evidence for the role of neuropeptide Y cord injury. Arch. Phys. Med. Rehabil., 81: 506–516.
in sympathetic nerve stimulation-induced vasoconstriction. Wallin, B.G. and Stjernberg, L. (1984) Sympathetic activity in
Am. J. Physiol., 274: H290–H294. man after spinal cord injury. Outflow to skin below the le-
Hirst, G.D., Bramich, N.J., Edwards, F.R. and Klemm, M. sion. Brain, 107: 183–198.
(1992) Transmission at autonomic neuroeffector junctions. Weaver, L.C., Verghese, P., Bruce, J.C., Fehlings, M.G., Krenz,
Trends Neurosci., 15: 40–46. N.R. and Marsh, D.R. (2001) Autonomic dysreflexia and
Ireland, D.R. (1999) Preferential formation of strong synapses primary afferent sprouting after clip-compression injury of
during re-innervation of guinea-pig sympathetic ganglia. J. the rat spinal cord. J. Neurotrauma, 18: 1107–1109.
Physiol., 520(Part 3): 827–837. Yeoh, M., McLachlan, E.M. and Brock, J.A. (2004a) Tail ar-
Krenz, N.R., Meakin, S.O., Krassioukov, A.V. and Weaver, teries from chronically spinalized rats have potentiated re-
L.C. (1999) Neutralizing intraspinal nerve growth factor sponses to nerve stimulation in vitro. J. Physiol., 556:
blocks autonomic dysreflexia caused by spinal cord injury. J. 545–555.
Neurosci., 19: 7405–7414. Yeoh, M., McLachlan, E.M. and Brock, J.A. (2004b) Chronic
Krenz, N.R. and Weaver, L.C. (1998) Sprouting of primary decentralization potentiates neurovascular transmission in
afferent fibers after spinal cord transection in the rat. Ne- the isolated rat tail artery, mimicking the effects of spinal
uroscience, 85: 443–458. transection. J. Physiol., 561: 583–596.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 20

Genetic approaches to autonomic dysreflexia

A. Brown and J.E. Jacob

Biotherapeutics Research Group, The Spinal Cord Injury Team, Robarts Research Institute and The Graduate Program in
Neuroscience, The University of Western Ontario, P.O. Box 5015, 100 Perth Drive, London, ON N6A 5K8, Canada

Abstract: Autonomic dysreflexia is a potentially life-threatening condition in which episodic hypertension


occurs after injuries above the mid-thoracic segments of the spinal cord. Despite the seriousness of this
condition, little is known of the molecular mechanisms that lead to its development. The completed
sequencing of the mouse genome, its dense genetic map, and the large repository of engineered and
spontaneous mouse mutants, make the mouse an ideal model organism in which to study the molecular
mechanisms underlying autonomic dysreflexia. We subjected two wild-type strains of mice, 129Sv and
C57BL/6, and one spontaneous mouse mutant, Wallerian degeneration slow (Wlds), to spinal cord tran-
section and clip-compression injury. We found that the incidence of autonomic dysreflexia is greatly
reduced, compared to spinal cord-transected wild-type mice, in Wlds mice after both injury paradigms and
in 129Sv and C57BL/6 that have undergone the clip-compression injury. We also found that the amplitude
of the dysreflexic response was greater in cord-compressed 129Sv than in C57BL/6 mice. These results
implicate axonal degeneration as an important source of signals that trigger the development of autonomic
dysreflexia and are discussed in the context of mouse genetics, interstrain differences and possible molecular
mechanisms underlying autonomic dysreflexia after spinal cord injury.

Introduction Yardley, 1988). However, after spinal cord injury,


spinal reflexes that increase sympathetic outflow
Autonomic dysreflexia is a condition that often from sympathetic preganglionic neurons dominate
develops after midthoracic or higher spinal cord the regulation of arterial pressure. The activity of
injuries and is characterized by episodes of hyper- the spinal reflexes, unchecked by supraspinal inhi-
tension and other signs of sympathetic hyperac- bition, leads to autonomic dysreflexia. These ex-
tivity that are triggered by sensory input entering aggerated reflexes begin within weeks of cord
the spinal cord below the level of the lesion. Au- injury and can be caused by stimulation of the
tonomic dysreflexia develops in 50–90% of people skin, pressure sores, distension or inflammation of
with tetraplegia or high paraplegia (Corbett et al., the urinary bladder or gastrointestinal tract, and
1975; Lindan et al., 1980; Mathias and Frankel, also by muscle spasms that often develop after
1993; Lee et al., 1995; Giannantoni et al., 1998). In spinal cord injury (Corbett et al., 1975; Mathias
uninjured animals and people, blood pressure con- and Frankel, 1993). Autonomic dysreflexia may be
trol depends upon supraspinal regulation of sym- mild, characterized only by sweating, piloerection,
pathetic preganglionic neurons (Calaresu and and small increases in arterial pressure or it may be
severe and lead to debilitating headaches, seizures,
Corresponding author. Tel.: +519 663 5777 ext. 34308; strokes, and death. Even in rehabilitated tetra-
Fax: +519 663 3789; E-mail: abrown@robarts.ca plegics and paraplegics, this condition can become

DOI: 10.1016/S0079-6123(05)52020-X 299


300

uncontrolled, leading to life-threatening hyperten- demonstrate autonomic dysreflexia in mice. Specif-


sion (Naftchi, 1990; Mathias and Frankel, 1993; ically, we found that we could elicit a mean arterial
Lee et al., 1995; Giannantoni et al., 1998). blood pressure elevation of approximately 35 mmHg
As is the case for any biological phenomenon, in response to colon distension and cutaneous
autonomic dysreflexia can be studied using a va- stimulation below the level of the lesion in spinal
riety of techniques. Unfortunately, few if any lab- cord-transected mice (Fig. 1) (Jacob et al., 2001).
oratories are undertaking a genetic approach to
this disorder. We will describe our own experience
in using a spontaneous mouse mutant to uncover A definition of autonomic dysreflexia
some of the molecular underpinnings of autonom-
ic dysreflexia. We will also describe some of the Once we began to use spinal cord-injured mice to
strain differences between C57BL/6 and 129Sv address mechanisms in the development of auto-
mice that we found with respect to their expression nomic dysreflexia, it became obvious that this
of autonomic dysreflexia and their development of could not be done without a proper definition of
afferent arbor plasticity after spinal cord injury. autonomic dysreflexia. The importance of this def-
Finally, we will briefly discuss and evaluate genetic inition was evident by the observation that even
and genomic approaches to the study of autonom- uninjured mice could be shown to display eleva-
ic dysreflexia. tions in mean arterial blood pressure in response
to colon distension (Fig. 1). To formulate a def-
inition of autonomic dysreflexia, we compared the
A mouse model of autonomic dysreflexia pressor responses of uninjured and spinal cord-
transected mice to colon distension. We considered
We have been interested in developing a mouse cord-transected animals to be obligately dysre-
model to study autonomic dysreflexia because of flexic since their sympathetic reflexes cannot be
the many advantages of the mouse over other affected by supraspinal inputs. We considered un-
model systems. First, with the exception of man, injured mice to be obligately eureflexic because of
we know much more about the genetics of mice their intact neuraxis. Uninjured mice demonstrat-
than of any other mammal (Copeland and Jenkins, ed an erratic blood pressure response to colon dis-
1991; Rinchik, 1991; Copeland et al., 1993; tention with readings falling above and below
Dietrich et al., 1994, 1998; Collins et al., 1998; baseline values during the stimulus. Upon the
Marra et al., 1999; Nusbaum et al., 1999). Second, withdrawal of the stimulus the blood pressures of
there are a large number of spontaneous mouse uninjured mice returned, within a 10 s interval, to
mutants that can be studied with respect to spinal baseline values (the longest time taken for any
cord injury (Green and Witham, 1991; Roths unoperated animal’s blood pressure to return to
et al., 1999). Third, large genetic screens have been baseline was 10.2 s). The blood pressure response
undertaken in mice (Schimenti and Bucan, 1998). of cord-transected mice to colon distension usually
Fourth, embryonic stem cell technology provides increased above baseline for the duration of the
the ability to design mice with mutations in any stimulus and remained increased well after the
nonlethal gene of interest to test its potential role stimulus ceased. Thus we consider a mouse to be
in autonomic dysreflexia (Roths et al., 1999). dysreflexic if (1) its blood pressure is consistently
Although autonomic dysreflexia had been well increased above baseline in response to colon dis-
documented in humans and rats (Osborn et al., tension and, (2) its blood pressure returns to base-
1990; Krassioukov and Weaver, 1995; Maiorov line with a delay greater than 10 s after colon
et al., 1997a, b; Bravo et al., 2004), there were no distension is terminated. It is interesting to note
previous reports characterizing autonomic dysre- that spinal cord-injured mice show a bimodal dis-
flexia in the spinal cord-injured mouse. Using 129Sv tribution with respect to the amount of time it
mice and a simple transection injury at the second takes blood pressure to return to baseline after
thoracic spinal segment (T2) we were able to colo-rectal distension. Their blood pressures either
301

Fig. 1. Recordings of arterial blood pressure changes in response to colon distension in uninjured and spinal cord-transected mice. An
uninjured C57BL/6 mouse (A) and a C57BL/6 spinal cord-transected mouse (B) were subjected to a 0.3 ml colon distension using a
balloon-tipped catheter. On the blood pressure tracings the time points marked ‘‘start’’ indicate the point at which the balloons were
fully inflated and the time points marked ‘‘stop’’ indicate the time at which the balloons were fully deflated and removed. The dashed
lines indicate the baseline blood pressure before balloon inflation. Note that while this stimulus produces a pressor response in
uninjured mice, the blood pressure elevation is erratic and the blood pressure returns to baseline or below baseline before the stimulus
ceased. In the spinal cord-transected mice there is a rapid increase in blood pressure that remains stable above baseline for the duration
of the stimulus and for a prolonged period afterward. Modified from Jacob et al. (2003). With permission from Elsevier Science B.V.

returned to baseline within 10 s of terminating the (Mathias and Frankel, 1993). Indeed, the modu-
stimulus (a non-dysreflexic response) or their lation of spinal reflex excitation of sympathetic
blood pressures remained elevated for minutes af- preganglionic neurons by baroreceptors and other
ter the stimulus was terminated. Once a mouse is supraspinal inhibitory systems may either be lost
judged dysreflexic, its degree of dysreflexia can be following complete spinal cord transection or re-
determined by measuring the increase in its blood duced following partial injuries. This lack of inhi-
pressure above baseline in response to a particular bition likely has a role in determining the
stimulus. magnitude and duration of the hypertensive epi-
sodes. However, the loss of inhibition develops al-
Proposed mechanisms for the development of most instantaneously after the injury, whereas
autonomic dysreflexia autonomic dysreflexia in humans and experimental
animals take weeks or months to develop (Mathias
Both peripheral and central mechanisms have been and Frankel, 1992; Krassioukov and Weaver,
proposed to explain the development of autonom- 1995; Maiorov et al., 1997a). Thus the loss of
ic dysreflexia. Peripheral mechanisms for the de- supraspinal inputs alone cannot account for the
velopment of autonomic dysreflexia include development of autonomic dysreflexia.
increased expression and/or responsiveness of vas- We and others have suggested that the time-
cular catecholamine receptors and increased neu- dependent, progressive development of autonomic
ral release of catecholamines (Naftchi, 1990; Lee dysreflexia implies that in addition to spinal dys-
et al., 1995; Karlsson et al., 1998; Karlsson, 1999; inhibition, synaptic plasticity leading to the grad-
Teasell et al., 2000). An attempt to quantify the ual remodeling of the spinal reflexes that control
contribution of peripheral mechanisms to the de- sympathetic preganglionic neuron output may
velopment of autonomic dysreflexia in the rat sug- underlie the development of this disorder (Mathias
gests that about half of the dysreflexic response and Frankel, 1992; Krassioukov and Weaver,
may be attributed to changes in vascular respon- 1996; Cassam et al., 1997; Weaver et al., 1997;
siveness after spinal cord injury (Collins and Jacob et al., 2001). Synaptic plasticity may be due
Dicarlo, 2002). Central mechanisms include to the unmasking of ‘‘silent’’ synapses (Guth,
loss of the baroreceptor reflex and loss of tonic 1976; Goshgarian et al., 1989), adjustments to
bulbospinal inhibitory input to spinal neurons synaptic strength (Kang and Schuman, 1995;
302

Scanziani et al., 1996), or to axonal sprouting that using a modified aneurysm clip calibrated to a 24 g
culminates in reinnervation of denervated neurons weight, which produces a severe injury in the
(Steward, 1989). Many of these synaptic altera- mouse (Joshi and Fehlings, 2002a). This model of
tions are triggered by axonal degeneration (Grob- spinal cord injury closely replicates the key patho-
stein and Chow, 1987). We therefore set out to physiological features of human injury by produc-
evaluate the importance of axonal degeneration to ing a prolonged, rapidly applied, extradural
the development of autonomic dysreflexia by: (1) compression. This model produces mechanical in-
assessing autonomic dysreflexia in a strain of mice, jury (primary injury) and secondary damage by a
designated Wallerian degeneration slow (Wlds) variety of well-characterized mechanisms includ-
mice, that undergo delayed Wallerian degenera- ing microvasculature disruption, hemorrhage, is-
tion, and (2) comparing autonomic dysreflexia in chemia, increases in intracellular calcium, calpain
cord-transected mice and -compressed mice in activation, progressive axonal loss, and glutamate
which maximal axonal degeneration would be ex- toxicity (Agrawal and Fehlings, 1996, 1997a, b;
pected to be delayed for sometime after the initial Agrawal et al., 1998; Schumacher et al., 1999;
injury (Jacob et al., 2003). Weaver et al., 2001, 2002). As an alternative to the
use of Wlds mice to evaluate the role of axonal
degeneration in the development of autonomic
Wlds mice, a genetic model to study the effects of
dysreflexia, we compared mice after spinal
delayed axonal degeneration
cord transection when all axons should undergo
Wallerian degeneration starting at the time of in-
The Wlds mutation arose in a substrain of C57BL/
jury to mice after clip-compression injury when one
6 mice being bred in England. The distinguishing
would expect a great deal of Wallerian degeneration
feature of Wlds mutants is that while the distal
to begin days after the initial insult due to the pro-
segment of an injured nerve normally degenerates
gressive nature of the secondary injury (Dumont
within 48 h, their axons survive for a long time
et al., 2001). The progressive loss of neurons due to
after being severed and separated from their cell
secondary events triggered by optic nerve crush has
bodies (Perry et al., 1990a, b, 1991). In fact, the
been well demonstrated (Yoles and Schwartz, 1998).
distal segments of severed Wlds axons not only
appear intact but may also conduct electrical im-
pulses up to 2 weeks after injury. While the genetic
Histological assessments of spinal cord transection
lesion accounting for the Wlds mutation has been
and clip-compression injury in wild-type and
identified as a fusion of the N-terminal portion of
Wlds mice
the ubiquitination factor E4B (Ube4b) to the ni-
cotinamide mononucleotide adenylyltransferase
We began our analysis by evaluating the lesion
(Nmnat) gene, the mechanism through which this
sites after spinal cord transection and clip-
mutation results in delayed Wallerian degenera-
compression injury in two strains of wild-type
tion is unclear (Mack et al., 2001). Wlds mice have
mice, 129Sv and C57BL/6, and in Wlds mice. The
previously been utilized to demonstrate the im-
three strains of mice demonstrated very similar
portance of Wallerian degeneration as a trigger for
histopathological changes after each injury para-
the plasticity involved in locomotor recovery after
digm as demonstrated by staining with hem-
spinal cord injury in mice (Zhang et al., 1998).
atoxylin and eosin. Two weeks after spinal cord
transection and clip-compression injury, lesion ep-
The clip-compression injury, a nongenetic model to icenters were largely filled by fibrous material, ma-
study the effects of delayed axonal degeneration crophages, and fibroblasts. In all cases small
cavities were present around the lesion, however,
The clip-compression injury model in the mouse large central cavities such as are found in lesioned
has been previously described (Joshi and Fehlings, rat spinal cords were not found (Fig. 2). These
2002a, b). Clip-compression injury is carried out findings corroborate other reports that C57BL/6
Fig. 2. Histology of the lesion epicenter in the mouse and rat after spinal cord injury. Two weeks after spinal cord transection (SCT, A), and clip-compression injury
(CCI, B and C) the spinal cords of C57BL/6 mice and rats were sectioned longitudinally on a cryostat and stained with hematoxylin and eosin. The lesion epicenters in
spinal cord-transected mice (A) were characterized by well-demarcated scars that were fibrous compact, whereas the lesion epicenters in mice with clip-compression injury
(B) were less fibrous and more diffuse and cellular. Unlike spinal cord-injured rats, that demonstrate large central cavities after spinal cord injury (C), mouse spinal cords
after either injury paradigm demonstrate multiple small cavities confined to within 1–2 mm of the lesion epicenter. Cav, cavity; FS, fibrous scar. Scale bars ¼ 0.5 mm. Rat
spinal cord illustration is a photomontage. Modified from Jacob et al. (2003). With permission from Elsevier Science B.V.

303
304

and Wlds spinal cords undergo very little cavita- part, on signals released by degenerating axons is
tion after spinal cord injury compared to the pro- to evaluate autonomic dysreflexia in an injury
gressive necrosis and cavitation seen in rats (Kuhn paradigm in which axonal degeneration is delayed
and Wrathall, 1998; Steward et al., 1999; Jakeman compared to transection. We have argued above
et al., 2000; Ma et al., 2001; Joshi and Fehlings, that clip-compression injury is an injury paradigm
2002a, b). Other groups have performed more de- in which axonal degeneration increases over time
tailed analysis comparing the cellular reactions of and is therefore somewhat delayed compared to
C57BL/6 and Wlds mice to spinal cord injury spinal cord transection. Blood pressure responses
(Fujiki et al., 1996; Zhang et al., 1996). These after spinal cord transection and clip-compression
studies have shown that in Wlds mice there is a injury were measured at 2 weeks post-injury in re-
delay in wound healing that becomes apparent af- sponse to colon distension and cutaneous pinch
ter 2 weeks post-injury and a delay in macrophage caudal to the injury. While all spinal cord-
and astrocyte activation that is obvious at 1 week transected C57BL/6 mice developed autonomic
post-injury. While we were unable to demonstrate dysreflexia (7/7), only approximately 50% did so
any histological differences between the strains for after clip-compression injury (5/9). Another wild-
either injury paradigm, we noted an obvious differ- type strain being studied in our laboratory, 129Sv,
ence between the mice that had undergone spinal also consistently demonstrated half the incidence
cord transection versus those that had undergone of autonomic dysreflexia after clip-compression
clip-compression injury. The lesion epicenters in injury (5/9) compared to after spinal cord transec-
mice that had undergone spinal cord transection tion (6/6). Thus we suggest that the incidence of
were thinner, more fibrous, and better circumscribed autonomic dysreflexia after clip-compression inju-
than the lesions after clip-compression injury. ry is lower than the incidence after spinal cord
transection because some of the Wallerian degen-
Incidence of autonomic dysreflexia in Wlds mice eration that is immediate after spinal cord tran-
section is delayed after clip-compression injury.
Since injured axons in Wlds mice undergo delayed We speculate that at sometime point greater than 2
Wallerian degeneration, we used this mutant to weeks post-injury, when Wallerian degeneration
assess the role of axon degeneration in the devel- has peaked in cord-compressed mice and in cord-
opment of autonomic dysreflexia. We reasoned transected Wlds mice, all these mice would develop
that if autonomic dysreflexia develops as a result autonomic dysreflexia (assuming that they eventu-
of events triggered by axonal degeneration, such as ally show the same amount of Wallerian degener-
changes in synaptic strength, axonal sprouting, or ation as in spinal cord transection wild-type mice).
reactive synaptogenesis, then the development of This work using genetic models and injury para-
autonomic dysreflexia should be delayed in Wlds digms that delay axonal degeneration suggests that
mice. In our studies the incidence of autonomic the development of autonomic dysreflexia does
dysreflexia at 2 weeks post-spinal cord transection depend on signals from degenerating axons. This
was reduced by half (4/8) in Wlds mice compared finding has profound importance to the treatment
to its wild-type parental strain, C57BL/6 (7/7). of spinal cord injury, suggesting that neuroprotec-
This supports the hypothesis that the development tive measures that delay Wallerian degeneration
of autonomic dysreflexia depends in part, on and support axonal survival may be able to pre-
synaptic plasticity that is triggered by signals elab- vent the synaptic plasticity associated with auto-
orated by degenerating axons. nomic dysreflexia.

Incidence of autonomic dysreflexia after spinal cord Strain differences in the amplitude of
transection and clip-compression injury autonomic dysreflexia

A nongenetic way to test the hypothesis that the During the analysis of autonomic dysreflexia in
development of autonomic dysreflexia depends in C57BL/6 and Wlds mice, identical experiments
305

were ongoing in our laboratory to evaluate the (Christensen and Hulsebosch, 1997a). Indeed ther-
same responses to spinal cord transection and clip- apeutic strategies designed to stop the sprouting of
compression injury in another wild-type strain, primary afferent fibers have been used to decrease
129Sv. A comparison of our results revealed in- autonomic dysreflexia and hyperalgesia in animal
teresting similarities and potentially important in- models following spinal cord injury (Christensen
ter-strain differences in response to spinal cord and Hulsebosch, 1997b; Krenz et al., 1999). In-
injury. Both 129Sv and C57BL/6 mice had a sim- creases in size of the small diameter primary af-
ilar reduction in the incidence of autonomic ferent arbor after spinal cord injury may be
dysreflexia after clip-compression injury compared detected by an increased area of Calcitonin gene-
to after spinal cord transection (see above). Fur- related peptide-immunoreactivity (CGRP-Ir) in
thermore, both strains showed approximately the laminae III–V of the dorsal horn. In our studies,
same magnitude of the blood pressure responses to 129Sv, C57BL/6, and Wlds mice sham and spinal
colon distension and cutaneous stimulation after cord-injured animals were perfused with fixative 2
spinal cord transection. However among the mice weeks post-injury and their spinal cords were sec-
that underwent clip-compression injury and were tioned and processed immunohistochemically for
judged to be dysreflexic (5/9 for both 129Sv and CGRP-Ir. Sections were analyzed and an area val-
C57BL/6) the magnitude of the blood pressure re- ue (mm2) for CGRP-immunoreactive fibers was
sponses during episodes of dysreflexia were signif- determined within laminae III–V (Fig. 4). Where-
icantly greater in 129Sv than C57BL/6 mice. as, the area of CGRP-Ir was increased at all spinal
Inspection of the data shows that 129Sv and segments in 129Sv mice after both spinal cord
C57BL/6 mice experienced a mean arterial blood transection and clip-compression injury, in both
pressure increase of approximately 35 mmHg after C57BL/6 and Wlds mice, CGRP-Ir was not sig-
spinal cord transection in response to colon dis- nificantly increased by spinal cord injury despite
tension or cutaneous pinch. 129Sv mice demon- the fact that all mice analyzed for CGRP-Ir were
strated the same degree of dysreflexia after clip- dysreflexic. We therefore concluded that sprouting
compression injury, whereas C57BL/6 mice only of small diameter primary afferent fibers after spi-
showed a mean arterial blood pressure increase of nal cord injury is not mandatory for the develop-
approximately 20 mmHg after clip-compression ment of autonomic dysreflexia, at least in C57BL/6
injury (Fig. 3). and Wlds mice. However, from these experiments,
we cannot determine the potential importance of
an increased small diameter primary afferent arbor
Increases in the size of the small diameter primary to the development of autonomic dysreflexia in
afferent arbor as a mechanism for the development animals that demonstrate this phenomenon.
of autonomic dysreflexia

It has been proposed that sprouting of the small Strain differences in CGRP-Ir in the dorsal horn
diameter primary afferent arbor may lead to au-
tonomic dysreflexia by increasing sensory input Interesting strain differences were also observed in
onto interneurons in the dorsal horn that control the size of the small diameter primary afferent ar-
sympathetic preganglionic neuron output. In sup- bor measured in control and injured spinal cords.
port of this hypothesis, increases in the arbor of First, we found that the size of the small diameter
small diameter afferent fibers in laminae III–V of primary afferent arbor, as measured by CGRP-Ir,
the dorsal horn have been demonstrated to corre- was significantly greater in control C57BL/6 and
late with the development of autonomic dysreflex- Wlds mice than in control 129Sv mice. Second, we
ia (Krenz and Weaver, 1998; Krenz et al., 1999). also found that, regardless of which injury was
Pain, another complication arising from spinal used, spinal cord transection or clip-compression
cord injury, has also been shown to be associated injury, an increase in the size of the primary af-
with sprouting of small diameter afferent fibers ferent arbor depended solely on the strain of
306

Fig. 3. Blood pressure changes in dysreflexic 129Sv (gray bars) and C57BL/6 (black bars) mice 2 weeks after clip-compression injury.
The change in blood pressure was measured by subtracting the baseline blood pressure from the maximal blood pressure reached
during stimulation. In each group, the blood pressure changes were significantly different from baseline, *po0:05: In addition, the
blood pressure changes were significantly greater in 129Sv than C57BL/6 dysreflexic mice; +po0:05: From Jacob et al. (2003) and
partially reproduced with permission from Elsevier Science B.V.

mouse under consideration. Thus, whether as- same group also showed that after spinal cord
sessed after spinal cord transection or clip-com- crush injury, 129/SvEMS mice developed a signif-
pression injury, 129Sv mice inevitably icantly greater lesion size than C57BL/6 mice at
demonstrated an increased afferent arbor while late (21 and 56 days post-injury) but not at early
C57BL/6 mice (and their mutant derivative Wlds) (7 and 14 days post-injury) time points (Inman
did not. et al., 2002). Other previously reported strain
Other groups also have documented strain dif- differences point to a possible explanation for the
ferences between C57BL/6 and 129Sv mice. For 129Sv/C57BL/6 strain differences observed after
example, it has been shown that kainic acid pro- spinal cord injury. For example, the recruitment
duces excitotoxic cell death in 129/SvEMS mice, as of neutrophils and macrophages is defective in
described in the rat, but that C57BL/6 are highly 129X1/SvJ compared to C57BL/6 mice in response
resistant to this neurotoxin (Schauwecker and to chemical inflammation (White et al., 2002).
Steward, 1997). Interestingly, both strains devel- Similarly, it has also been shown that the cellular
op seizures after the same kainic acid regime. The response after spinal cord injury is different in
307

129X1/SvJ compared to C57BL/6 mice (Ma et al.,


2004). In particular this group has shown that the
lesion site in C57BL/6 mice is occupied by more
macrophages than the lesion sites in 129X1/SvJ
mice and that the macrophages are distributed
uniformly throughout the lesion in C57BL/6,
whereas the macrophages in 129X1/SvJ mice are
distributed in patches separated by tissue matrix.
They also demonstrated more astrocytic processes
and axon profiles in the lesions of 129X1/SvJ
compared to C57BL/6 mice. Thus this study sug-
gests that the inflammatory response generated by
129X1/SvJ mice in response to spinal cord injury
results in a lesion that encourages more plasticity,
as evidenced by axonal sprouting into the lesion,
than that seen in C57BL/6 mice.
The strain differences summarized above sug-
gest that, after an equivalent injury, 129X1/SvJ
mice may elicit greater plasticity at the lesion ep-
icenter than C57BL/6. Can this help us to under-
stand strain differences in autonomic dysreflexia
that are more likely to be affected by plasticity
caudal to the injury where sympathetic outflow
controlling the major splanchnic vascular beds
originates? We have shown that 129Sv mice have a
greater degree of plasticity below the level of the
lesion, as measured by greater changes in CGRP-
Ir than C57BL/6. Assuming that the slightly dif-
ferent substrain of 129Sv mice, 129S3Svimj, used
in our studies, demonstrate the same muted in-
flammatory response to spinal cord injury as
shown for 129X1/SvJ mice, it is reasonable to
speculate that the mechanisms generating in-
creased plasticity at the lesion epicenter and at
more caudal segments may be the same. In this
regard there are two possibilities. First, since the
inflammatory response after spinal cord injury
adds to the destruction of spinal cord tissue sur-
Fig. 4. Area of CGRP-Ir fibers (mm2) in the dorsal horn of
rounding the lesion epicenter (Bethea, 2000), we
129Sv, C57Bl, and Wlds mice 2 weeks after a sham operation, propose that a less robust inflammatory reaction
spinal cord transection or clip-compression injury. The area of allows for greater sparing in the injured cord that
CGRP-Ir was measured within an area of interest in laminae may provide more substrate for the development
III–V of the dorsal horn as described in the text. The areas of of synaptic plasticity even at segmental levels cau-
CGRP-Ir were increased at all spinal segments in the 129Sv
cord-transected and clip-compressed mice (A) but not in C57BL
dal to the primary injury. Thus, the less efficient
(B) or Wlds (C) mice after either injury. *Significantly different recruitment of inflammatory cells to the injury site
from sham-operated group of the same segment, po0:05: From in 129Sv mice after clip-compression injury, might
Jacob et al. (2003) and reproduced with permission from lead to greater plasticity and therefore greater
Elsevier Science B.V. blood pressure responses to sensory stimulation
308

than cord-compressed C57BL/6 dysreflexic mice. sympathetic preganglionic neurons to suppress au-
Second, there may be a greater proclivity for plas- tonomic dysreflexia, whereas axonal growth might
ticity in the injured spinal cord in 129Sv mice reflect a spinal cord environment that encourages
compared to C57BL/6 mice for genetic reasons enhanced plasticity and promotes the development
unrelated to inflammatory response. Genetic map- of autonomic dysreflexia. Thus we stress the im-
ping studies that might allow these important portance of adding autonomic dysreflexia to the
genes to be identified will be discussed below. We outcome measures used by those studying spinal
speculate that we do not see differences in the de- cord injury in mouse mutants as it is a sensitive
gree of dysreflexia between these strains after spi- measure of both axonal sparing and of deleterious
nal cord transection because the total disruption of plasticity (whereas regenerative growth allowing
descending inputs to sympathetic preganglionic for improved locomotor function might be con-
neurons promotes maximal dysreflexia regardless sidered ameliorative plasticity). These types of
of other modulating effects. studies may then have the potential to delineate
the genetic pathways that lead to productive as
opposed to harmful plasticity. Our study and the
Conclusions work of Zhang et al. (1996, 1998), suggest that the
trigger to deleterious and beneficial plasticity may
The study of C57BL/6 and Wlds mice was a useful be the same, namely axonal degeneration. The
attempt to use mouse genetics to address the mo- identification of molecules that are unique to the
lecular mechanisms responsible for autonomic development of one form of plasticity over the
dysreflexia and supports the importance of axon- other will be critical to therapeutic advances.
al degeneration as a trigger to the cascade of In contrast to the study of mutant mice in which
events that leads to autonomic dysfunction. How- the genetic lesion has been identified, studying
ever, because of the nature of the mutation (a fu- phenotypic differences between strains of mice is
sion of the N-terminus of Ube4b to the Nmnat somewhat more resistant to genetic dissection. The
gene) it is difficult to speculate on the molecular different responses to spinal cord injury observed
signals that might be affected. Others have used between C57BL/6 and 129Sv must be due to ge-
engineered mouse mutants (knockouts and trans- netic differences between these two strains of mice.
genics) to ask the roles of particular genes in var- Inter strain genetic differences can be identified
ious aspects of recovery from spinal cord injury with proper analysis and have yielded dividends in
(Kim et al., 2003; Simonen et al., 2003; Wells et al., a number of fields including obesity, atherosclero-
2003; Zheng et al., 2003; Kerr and Patterson, 2004; sis, and cancer research (Devereux and Kaplan,
Song et al., 2004). Unfortunately, none of these 1998; Diament et al., 2003; Smith, 2003). For ex-
studies have evaluated autonomic dysreflexia in ample, we have found that after clip-compression
these mouse mutants and, while one might assume injury, 129Sv mice have nearly a two-fold greater
that better locomotor recovery will translate into blood pressure response to tail pinch or colon dis-
less autonomic dysreflexia, caution is warranted. tention than C57BL/6 mice. These two mouse
For example, whereas analysis of spinal cord in- strains could be crossed and the resulting F1 gen-
jury in a mouse mutant that demonstrates im- eration sibmated or backcrossed to one of the pa-
proved neuroprotection and tissue sparing might rental strains. By phenotyping (autonomic testing)
be safely expected to have a concomitant reduction the F2 or backcross generation and then genotyp-
in autonomic dysreflexia, it is also possible that ing them using molecular markers (microsatellite
mutants that show enhanced axonal growth, sequences, restriction fragment length polymorph-
sprouting or regeneration and therefore improved isms, or single nucleotide polymorphisms) an as-
locomotor outcomes might actually have more se- sociation between one or more genetic loci and an
vere autonomic dysreflexia. This is because neuro- autonomic dysreflexia phenotype could be
protection and tissue sparing might allow for achieved. These types of genetic mapping studies
enough descending inhibitory inputs into thoracic can narrow down the genomic region of interest to
309

about 1 cM (Diament et al., 2003). Through the profiling of multiple conditions that can be corre-
judicious use of congenic and recombinant inbred lated to a degree of autonomic dysfunction. While
strains, the genomic area of interest may be further several groups have conducted gene expression
narrowed and allow one to evaluate candidate studies at various time points after spinal cord in-
genes from that locus (Mullerova and Hozak, jury in the rat using microarrays (Carmel et al.,
2004). This genetic approach may require patience 2001; Song et al., 2001; Nesic et al., 2002; Tachi-
to identify genes responsible for autonomic dysre- bana et al., 2002; Pan et al., 2004; Resnick et al.,
flexia but it offers the following two very powerful 2004; Zhang et al., 2004), none have tried to cor-
advantages. First, because the analysis begins with relate these expression patterns to autonomic
an interesting autonomic phenotype (a mouse dysreflexia. However, based on the premise that
more or less vulnerable to develop autonomic sparing of descending inputs may protect against
dysreflexia after spinal cord injury) one can be autonomic dysreflexia, a related approach may be
fairly certain that the genes being sought do have a to profile gene expression after experimental treat-
role to play in autonomic physiology. This stands ments that promote neuroprotection and tissue
in stark contrast to a candidate gene approach that sparing. For example one group has profiled gene
begins with a gene that may or may not be in- expression using microarrays after treating spinal
volved in the phenomenon under study. Second, cord explants with a variety of anti-inflammatory
this approach offers the potential to identify genes compounds (Pan et al., 2004). This analysis re-
with an important role to play in autonomic vealed a neuroprotective gene cluster that was in-
dysreflexia that may not have been proposed based duced by a COX-2 inhibitor. In a similar fashion,
on the literature. This unbiased approach to iden- we are analyzing changes in gene expression asso-
tify genes involved in autonomic dysreflexia is not ciated with the administration of an early anti-in-
limited by our imagination to conjecture a priori flammatory strategy after spinal cord injury in the
what that gene may be. Thus this type of research rat. This anti-integrin strategy has been shown to
may possibly open up new avenues of research into be neuroprotective and to reduce greatly auto-
causes of autonomic dysreflexia. nomic dysreflexia in spinal cord-injured rats (Gris
Another unbiased strategy to identify genes in- et al., 2004). The identification of a gene or gene
volved in autonomic dysreflexia would be to em- cluster induced or suppressed by this treatment
ploy genomic technologies. In contrast to the may be a step toward identification of genes in-
analysis of mouse mutants or gene mapping stud- volved in autonomic dysreflexia.
ies that allow one to consider the role of one or a Autonomic dysreflexia is a complex disorder
few genes in a particular process, genomics allows that is likely the product of trauma and many ge-
one to analyze tens-of-thousands of genes at once. netic interactions. Strategies to identify genes im-
As genomic analysis rests on gene expression pro- portant to its development include the analysis of
files that correlate gene expression with a physio- mouse mutants and the use of genetics and gen-
logical process, a gene or group of genes may be omics. Our own genetic studies have helped to
implicated with a particular process but their in- uncover the role of axonal degeneration in trig-
volvement in that process must be proven by func- gering the development of autonomic dysreflexia.
tional studies. The advantage of this approach lies Our studies also reveal interesting strain differenc-
in: (1) the ability to interrogate the entire tran- es between 129Sv and C57BL/6 mice in their
scriptome for correlations to autonomic dysreflex- dysreflexic responses and in their expression of
ia, (2) the unbiased nature of this approach, and plasticity after spinal cord injury as expressed by
(3) the speed with which one can generate candi- changes in their primary afferent arbor. We
date genes responsible for autonomic dysreflexia strongly urge those carrying out genetic and gen-
compared to mapping studies. The biggest disad- omic studies of spinal cord injury in rodents to use
vantage to this approach is the problem of han- autonomic function as an outcome measure as it is
dling and mining the volumes of data. A genomics as important to identify the genes involved in
approach to autonomic dysreflexia requires gene harmful plasticity, that may lead to autonomic
310

dysfunction, as it is to identify the genes involved Copeland, N.G. and Jenkins, N.A. (1991) Development and
in regenerative plasticity that may lead to im- applications of a molecular genetic linkage map of the mouse
proved locomotor function. Analysis of strain dif- genome. Trends Genet., 7: 113–118.
Copeland, N.G., Jenkins, N.A., Gilbert, D.J., Eppig, J.T.,
ferences, new studies employing genetically Maltais, L.J., Miller, J.C., Dietrich, W.F., Weaver, A.,
modified mice, and genomic analysis of cord-in- Lincoln, S.E., Steen, R.G., Stein, L.D., Nadeau, J.H. and
jured rodents under conditions that lead to varying Lander, E.S. (1993) A genetic linkage map of the mouse:
degrees of dysreflexia will undoubtedly lead to the current applications and future prospects. Sci. New Series,
identification of genes responsible for autonomic No. 5130, Genome Issue, 262: 57–66.
Corbett, J.L., Debarge, O., Frankel, H.L. and Mathias, C.
dysreflexia and point the way to new innovative (1975) Cardiovascular responses in tetraplegic man to muscle
approaches to its treatment. spasm bladder percussion and head-up tilt Clin. Exp.
Pharmacol. Physiol., 2: 189–193.
Devereux, T.R. and Kaplan, N.L. (1998) Use of quantitative
References trait loci to map murine lung tumor susceptibility genes. Exp.
Lung Res., 24: 407–417.
Agrawal, S.K. and Fehlings, M.G. (1996) Mechanisms of sec- Diament, A.L., Fisler, J.S. and Warden, C.H. (2003) Studies of
ondary injury to spinal cord axons in vitro: role of Na+ natural allele effects in mice can be used to identify genes
Na(+)-K(+)-ATPase the Na(+)-H(+) exchanger and the causing common human obesity. Obes. Rev., 4: 249–255.
Na(+)-Ca2+ exchanger. J. Neurosci., 16: 545–552. Dietrich, W., Katz, H., Lincoln, S., Shin, H., Friedman, J.,
Agrawal, S.K. and Fehlings, M.G. (1997a) The effect of the Dracopoli, N. and Lander, E. (1998) A genetic map of the
sodium channel blocker QX-314 on recovery after acute spi- mouse suitable for typing intraspecific crosses. Genetics, 131:
nal cord injury. J. Neurotrauma, 14: 81–88. 423–447.
Agrawal, S.K. and Fehlings, M.G. (1997b) Role of NMDA and Dietrich, W., Miller, J., Steen, R., Merchant, M., Damron, D.,
non-NMDA ionotropic glutamate receptors in traumatic NAhf, R., Gross, A., Joyce, D., Wessel, M., Dredge, R.,
spinal cord axonal injury. J. Neurosci., 17: 1055–1063. Marquis, A., Stein, L., Goodman, N., Page, D. and Lander,
Agrawal, S.K., Theriault, E. and Fehlings, M.G. (1998) Role of E. (1994) A genetic map of the mouse with 4,006 simple
group I metabotropic glutamate receptors in traumatic spinal sequence length polymorphisms. Nat. Genet., 7: 220–245.
cord white matter injury. J. Neurotrauma, 15: 929–941. Dumont, R.J., Okonkwo, D.O., Verma, S., Hurlbert, R.J.,
Bethea, J.R. (2000) Spinal cord injury-induced inflammation: a Boulos, P.T., Ellegala, D.B. and Dumont, A.S. (2001) Acute
dual-edged sword. Prog. Brain Res., 128: 33–42. spinal cord injury, part I: pathophysiologic mechanisms.
Bravo, G., Guizar-Sahagun, G., Ibarra, A., Centurion, D. and Clin. Neuropharmacol., 24: 254–264.
Villalon, C.M. (2004) Cardiovascular alterations after spinal Fujiki, M., Zhang, Z., Guth, L. and Steward, O. (1996) Genetic
cord injury: an overview. Curr. Med. Chem. Cardiovasc. influences on cellular reactions to spinal cord injury: activa-
Hematol. Agents, 2: 133–148. tion of macrophages/microglia and astrocytes is delayed in
Calaresu, F.R. and Yardley, C.P. (1988) Medullary basal sym- mice carrying a mutation (WldS) that causes delayed Wall-
pathetic tone. Annu. Rev. Physiol., 50: 511–524. erian degeneration. J .Comp. Neurol., 371: 469–484.
Carmel, J.B., Galante, A., Soteropoulos, P., Tolias, P., Recce, Giannantoni, A., Di Stasi, S.M., Scivoletto, G., Mollo, A.,
M., Young, W. and Hart, R.P. (2001) Gene expression pro- Silecchia, A., Fuoco, U. and Vespasiani, G. (1998) Auto-
filing of acute spinal cord injury reveals spreading inflamma- nomic dysreflexia during urodynamics [see comments]. Spinal
tory signals and neuron loss. Physiol. Genomics, 7: 201–213. Cord, 36: 756–760.
Cassam, A., Llewellyn-Smith, I. and Weaver, L. (1997) Cat- Goshgarian, H.G., Yu, X.J. and Rafols, J.A. (1989) Neuronal
echolamine enzymes and neuropeptides are expressed in fi- and glial changes in the rat phrenic nucleus occurring within
bres and somata in the intermediate gray matter in chronic hours after spinal cord injury. J. Comp. Neurol., 284: 519–533.
spinal rats. Neuroscience, 78: 829–841. Green, M.C. and Witham, B.A. (1991) Handbook on Genet-
Christensen, M. and Hulsebosch, C. (1997a) Chronic central ically Standardized Jax Mice. Jackson Laboratory, Bar Har-
pain after spinal cord injury. J. Neurotrauma, 14: 517–537. bor, Maine.
Christensen, M.D. and Hulsebosch, C.E. (1997b) Spinal cord Gris, D., Marsh, D.R., Oatway, M.A., Chen, Y., Hamilton,
injury and anti-NGF treatment results in changes in CGRP E.F., Dekaban, G.A. and Weaver, L.C. (2004) Transient
density and distribution in the dorsal horn in the rat. Exp. blockade of the CD11d/CD18 integrin reduces secondary
Neurol., 147: 463–475. damage after spinal cord injury, improving sensory auto-
Collins, F.S., Patrinos, A., Jordan, E., Chakravarti, A., Gest- nomic, and motor function. J. Neurosci., 24: 4043–4051.
eland, R. and Walters, L. (1998) New goals for the U.S. Grobstein, P. and Chow, K.L. (1987) Visual system develop-
Human Genome Project: 1998–2003. Science, 282: 682–689. ment plasticity. In: Adelman G. (Ed.), Encyclopedia of
Collins, H.L. and Dicarlo, S.E. (2002) Acute exercise reduces Neuroscience. Birkhauser, Boston, pp. 1294–1297.
the response to colon distension in T(5) spinal rats. Am. J. Guth, L. (1976) Functional plasticity in the respiratory pathway
Physiol. Heart Circ. Physiol., 282: H1566–H1570. of the mammalian spinal cord. Exp. Neurol., 51: 414–420.
311

Inman, D., Guth, L. and Steward, O. (2002) Genetic influences Lee, B.Y., Karmakar, M.G., Herz, B.L. and Sturgill, R.A.
on secondary degeneration and wound healing following (1995) Autonomic dysreflexia revisited. J. Spinal Cord Med.,
spinal cord injury in various strains of mice. J. Comp. 18: 75–87.
Neurol., 451: 225–235. Lindan, R., Joiner, E., Freehafer, A.A. and Hazel, C. (1980)
Jacob, J.E., Gris, P., Fehlings, M.G., Weaver, L.C. and Brown, Incidence and clinical features of autonomic dysreflexia in
A. (2003) Autonomic dysreflexia after spinal cord transection patients with spinal cord injury. Paraplegia, 18: 285–292.
or compression in 129Sv, C57BL, and Wallerian degenera- Ma, M., Basso, D.M., Walters, P., Stokes, B.T. and Jakeman,
tion slow mutant mice. Exp. Neurol., 183: 136–146. L.B. (2001) Behavioral and histological outcomes following
Jacob, J.E., Pniak, A., Weaver, L.C. and Brown, A. (2001) graded spinal cord contusion injury in the C57Bl/6 mouse.
Autonomic dysreflexia in a mouse model of spinal cord in- Exp. Neurol., 169: 239–254.
jury. Neuroscience, 108: 687–693. Ma, M., Wei, P., Wei, T., Ransohoff, R.M. and Jakeman, L.B.
Jakeman, L.B., Guan, Z., Wei, P., Ponnappan, R., Dzwonczyk, (2004) Enhanced axonal growth into a spinal cord contusion
R., Popovich, P.G. and Stokes, B.T. (2000) Traumatic spinal injury site in a strain of mouse (129X1/SvJ) with a diminished
cord injury produced by controlled contusion in mouse. inflammatory response. J. Comp. Neurol., 474: 469–486.
J. Neurotrauma, 17: 299–319. Mack, T.G., Reiner, M., Beirowski, B., Mi, W., Emanuelli, M.,
Joshi, M. and Fehlings, M.G. (2002a) Development and char- Wagner, D., Thomson, D., Gillingwater, T., Court, F.,
acterization of a novel, graded model of clip compressive Conforti, L., Fernando, F.S., Tarlton, A., Andressen, C.,
spinal cord injury in the mouse: part 1. Clip design, behavi- Addicks, K., Magni, G., Ribchester, R.R., Perry, V.H. and
oral outcomes, and histopathology. J. Neurotrauma, 19: Coleman, M.P. (2001) Wallerian degeneration of injured ax-
175–190. ons and synapses is delayed by a Ube4b/Nmnat chimeric
Joshi, M. and Fehlings, M.G. (2002b) Development and char- gene. Nat. Neurosci., 4: 1199–1206.
acterization of a novel, graded model of clip compressive Maiorov, D.N., Krenz, N.R., Krassioukov, A.V. and Weaver,
spinal cord injury in the mouse: part 2. Quantitative neuro- L.C. (1997a) Role of spinal NMDA and AMPA receptors in
anatomical assessment and analysis of the relationships be- episodic hypertension in conscious spinal rats. Am. J.
tween axonal tracts, residual tissue, and locomotor recovery. Physiol., 273: H1266–H1274.
J. Neurotrauma, 19: 191–203. Maiorov, D.N., Weaver, L.C. and Krassioukov, A.V. (1997b)
Kang, H. and Schuman, E.M. (1995) Long-lasting neurotro- Relationship between sympathetic activity and arterial pres-
phin-induced enhancement of synaptic transmission in the sure in conscious spinal rats. Am. J. Physiol., 272:
adult hippocampus. Science, 267: 1658–1662. H625–H631.
Karlsson, A.K. (1999) Autonomic dysreflexia [see comments]. Marra, M., Hillier, L.D., Kucaba, T., Allen, M., Barstead, R.,
Spinal Cord, 37: 383–391. Beck, C., Blistain, A., Bonaldo, M., Bowers, Y., Bowles, L.,
Karlsson, A.K., Friberg, P., Lonnroth, P., Sullivan, L. and Cardenas, M., Chamberlain, A., Chappell, J., Clifton, S.,
Elam, M. (1998) Regional sympathetic function in high spi- Favello, A., Geisel, S., Gibbons, M., Harvey, N., Hill, F.,
nal cord injury during mental stress and autonomic dysre- Jackson, Y., Kohn, S., Lennon, G., Mardis, E., Martin, J.,
flexia. Brain, 121: 1711–1719. Mila, L.A., McCann, R., Morales, R., Pape, D., Person, B.,
Kerr, B.J. and Patterson, P.H. (2004) Potent pro-inflammatory Prange, C., Ritter, E., Soares, M., Schurk, R., Shin, T.,
actions of leukemia inhibitory factor in the spinal cord of the Steptoe, M., Swaller, T., Theising, B., Underwood, K.,
adult mouse. Exp. Neurol., 188: 391–407. Wylie, T., Yount, T., Wilson, R. and Waterston, R. (1999)
Kim, J.E., Li, S., GrandPre, T., Qiu, D. and Strittmatter, S.M. An encyclopedia of mouse genes. Nat. Genet., 21: 191–194.
(2003) Axon regeneration in young adult mice lacking Nogo- Mathias, C.J. and Frankel, H.L. (1992) The cardiovascular
A/B. Neuron, 38: 187–199. system in tetraplegia and paraplegia. In: Frankel H.L. (Ed.),
Krassioukov, A.V. and Weaver, L.C. (1995) Episodic hyper- Handbook of Clinical Neurology. Elsevier Science Publish-
tension due to autonomic dysreflexia in acute and chronic ers, B.V., pp. 435–456.
spinal cord-injured rats. Am. J. Physiol., 268: H2077–H2083. Mathias, C.J. and Frankel, H.L. (1993) Autonomic disturbanc-
Krassioukov, A.V. and Weaver, L.C. (1996) Morphological es in spinal cord lesions. In: Bannister R. and Mathias C.J.
changes in sympathetic preganglionic neurons after spinal (Eds.), Autonomic Failure. A Textbook of Clinical Disorders
cord injury in rats. Neuroscience, 70: 211–225. of the Autonomic Nervous System. Oxford Medical Publi-
Krenz, N. and Weaver, L. (1998) Changes in the morphology of cations, Oxford.
sympathetic preganglionic neurons parallel the development Mullerova, J. and Hozak, P. (2004) Use of recombinant con-
of autonomic dysreflexia after spinal cord injury in rats. genic strains in mapping disease-modifying genes. News.
Neurosci. Lett., 243: 61–64. Physiol. Sci., 1: 105–109.
Krenz, N.R., Meakin, S.O., Krassioukov, A.V. and Weaver, Naftchi, N.E. (1990) Mechanism of autonomic dysreflexia.
L.C. (1999) Neutralizing intraspinal nerve growth factor Contributions of catecholamine and peptide neurotransmit-
blocks autonomic dysreflexia caused by spinal cord injury. ters. Ann. NY. Acad. Sci., 579: 133–148.
J. Neurosci., 19: 7405–7414. Nesic, O., Svrakic, N.M., Xu, G.Y., McAdoo, D., Westlund,
Kuhn, P. and Wrathall, J. (1998) A mouse model of graded K.N., Hulsebosch, C.E., Ye, Z., Galante, A., Soteropoulos,
contusive spinal cord injury. J. Neurotrauma, 15: 125–140. P., Tolias, P., Young, W., Hart, R.P. and Perez-Polo, J.R.
312

(2002) DNA microarray analysis of the contused spinal cord: loss of dephosphorylated NF200, following traumatic spinal
effect of NMDA receptor inhibition. J. Neurosci. Res., 68: cord injury. Neuroscience, 91: 733–744.
406–423. Simonen, M., Pedersen, V., Weinmann, O., Schnell, L., Buss,
Nusbaum, C., Slonim, D.K., Harris, K.L., Birren, B.W., Steen, A., Ledermann, B., Christ, F., Sansig, G., van der Putten, H.
R.G., Stein, L.D., Miller, J., Dietrich, W.F., Nahf, R., Wang, and Schwab, M.E. (2003) Systemic deletion of the myelin-
V., Merport, O., Castle, A.B., Husain, Z., Farino, G., Gray, associated outgrowth inhibitor Nogo-A improves regenera-
D., Anderson, M.O., Devine, R., Horton Jr., L.T., Ye, W., tive and plastic responses after spinal cord injury. Neuron,
Wu, X., Vardouhie, K., Zemsteva, I.S., Wu, Y., Collymore, 38: 201–211.
A.J., Courtney, D.F, Tam, J., Cadman, M., Haynes, A.R., Smith, J. (2003) Quantitative trait locus mapping for athero-
Heuston, C., Marsland, T., Southwell, A., Trickett, P., Striv- sclerosis susceptibility. Curr. Opin. Lipidol., 14: 499–504.
ens, M.A., Ross, M.T., Makalowski, W., Xu, Y., Boguski, Song, G., Cechvala, C., Resnick, D.K., Dempsey, R.J. and
M.S., Carter, N.P., Denny, P., Brown, S.D.M., Hudson, T.J. Rao, V.L. (2001) GeneChip analysis after acute spinal cord
and Lander, E.S. (1999) A YAC-based physical map of the injury in rat. J. Neurochem., 79: 804–815.
mouse genome. Nat. Genet., 22: 388–393. Song, X.Y., Zhong, J.H., Wang, X. and Zhou, X.F. (2004)
Osborn, J.W., Taylor, R.F. and Schramm, L.P. (1990) Chronic Suppression of p75NTR does not promote regeneration of
cervical spinal cord injury and autonomic hyperreflexia in injured spinal cord in mice. J. Neurosci., 24: 542–546.
rats. Am. J. Physiol., 258: R169–R174. Steward, O. (1989) Reorganization of neuronal connections
Pan, J.Z., Jornsten, R. and Hart, R.P. (2004) Screening anti- following CNS trauma: principles and experimental para-
inflammatory compounds in injured spinal cord with mic- digms. J. Neurotrauma, 6: 99–152.
roarrays: a comparison of bioinformatics analysis approach- Steward, O., Schauwecker, P.E., Guth, L., Zhang, Z., Fujiki,
es. Physiol. Genomics, 17: 201–214. M., Inman, D., Wrathall, J., Kempermann, G., Gage, F.H.,
Perry, V., Brown, M. and Lunn, E. (1991) Very slow retrograde Saatman, K.E., Raghupathi, R. and McIntosh, T. (1999)
and Wallerian degeneration in the CNS of C57BL6/Ola mice. Genetic approaches to neurotrauma research: opportunities
Eur. J. Neurosci., 3: 102–105. and potential pitfalls of murine models. Exp. Neurol., 157:
Perry, V., Brown, M., Lunn, E., Tree, P. and Gordon, S. 19–42.
(1990a) Evidence that very slow Wallerian degeneration in Tachibana, T., Noguchi, K. and Ruda, M.A. (2002) Analysis of
C57BL/Ola mice is an intrinsic property of the peripheral gene expression following spinal cord injury in rat using
nerve. Eur. J. Neurosci., 2: 802–808. complementary DNA microarray. Neurosci. Lett., 327:
Perry, V., Lunn, E., Brown, M., Cahusac, S. and Gordon, S. 133–137.
(1990b) Evidence that the rate of Wallerian degeneration is Teasell, R.W., Arnold, J.M., Krassioukov, A. and Delaney,
controlled by a single autosomal dominant gene. Eur. J. G.A. (2000) Cardiovascular consequences of loss of supra-
Neurosci., 2: 408–413. spinal control of the sympathetic nervous system after spinal
Resnick, D.K., Schmitt, C., Miranpuri, G.S., Dhodda, V.K., cord injury. Arch. Phys. Med. Rehabil., 81: 506–516.
Isaacson, J. and Vemuganti, R. (2004) Molecular evidence of Weaver, L., Cassam, A., Krassioukov, A. and Llewellyn-Smith,
repair and plasticity following spinal cord injury. Neurore- I. (1997) Changes in immunoreactivity for growth associated
port, 15: 837–839. protein-43 suggest reorganization of synapses on spinal sym-
Rinchik, E.M. (1991) Chemical mutagenesis and fine-structure pathetic neurons after cord transection. Neuroscience, 81:
functional analysis of the mouse genome. Trends Genet., 7: 535–551.
15–21. Weaver, L., Verghese, P., Bruce, J., Fehlings, M., Krenz, N.R.
Roths, J.B., Foxworth, W.B., McArthur, M.J., Montgomery, and Marsh, D. (2001) Autonomic dysreflexia and primary
C.A. and Kier, A.B. (1999) Spontaneous and engineered afferent sprouting after clip compression injury of the rat
mutant mice as models for experimental and comparative spinal cord. J. Neurotrauma, 18: 1107–1119.
pathology: history comparison and developmental technolo- Weaver, L.C., Marsh, D.R., Gris, D., Meakin, S.O. and
gy. Lab. Anim. Sci., 49: 12–34. Dekaban, G.A. (2002) Central mechanisms for autonomic
Scanziani, M., Malenka, R.C. and Nicoll, R.A. (1996) Role of dysreflexia after spinal cord injury. Prog. Brain Res., 137:
intercellular interactions in heterosynaptic long-term depres- 83–95.
sion. Nature, 380: 446–450. Wells, J.E., Rice, T.K., Nuttall, R.K., Edwards, D.R., Zekki,
Schauwecker, P.E. and Steward, O. (1997) Genetic determi- H., Rivest, S. and Yong, V.W. (2003) An adverse role for
nants of susceptibility to excitotoxic cell death: implications matrix metalloproteinase 12 after spinal cord injury in mice.
for gene targeting approaches. Proc. Natl. Acad. Sci. USA., J. Neurosci., 23: 10107–10115.
94: 4103–4108. White, P., Liebhaber, S.A. and Cooke, N.E. (2002) 129X1/SvJ
Schimenti, J. and Bucan, M. (1998) Functional genomics in the Mouse strain has a novel defect in inflammatory cell recruit-
mouse: phenotype-based mutagenesis screens. Genome Res., ment. J. Immunol., 168: 869–874.
8: 698–710. Yoles, E. and Schwartz, M. (1998) Degeneration of spared ax-
Schumacher, P.A., Eubanks, J.H. and Fehlings, M.G. (1999) ons following partial white matter lesion: implications for
Increased calpain I-mediated proteolysis, and preferential optic nerve neuropathies. Exp. Neurol., 153: 1–7.
313

Zhang, K.H., Xiao, H.S., Lu, P.H., Shi, J., Li, G.D., Wang, Zhang, Z., Guth, L. and Steward, O. (1998) Mechanisms of
Y.T., Han, S., Zhang, F.X., Lu, Y.J., Zhang, X. and Xu, motor recovery after subtotal spinal cord injury insights
X.M. (2004) Differential gene expression after complete spi- from the study of mice carrying a mutation (Wlds)
nal cord transection in adult rats: an analysis focused on a that delays cellular responses to injury. Exp. Neurol., 149:
subchronic post-injury stage. Neuroscience, 128: 375–388. 221–229.
Zhang, Z., Fujiki, M., Guth, L. and Steward, O. (1996) Genetic Zheng, B., Ho, C., Li, S., Keirstead, H., Steward, O. and
influences on cellular reactions to spinal cord injury: a Tessier-Lavigne, M. (2003) Lack of enhanced spinal regen-
wound-healing response present in normal mice is impaired eration in Nogo-deficient mice. Neuron, 38: 213–224.
in mice carrying a mutation (WldS) that causes delayed
Wallerian degeneration. J. Comp. Neurol., 371: 485–495.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 21

Gastrointestinal symptoms related to autonomic


dysfunction following spinal cord injury

Eric A.L. Chung and Anton V. Emmanuel

St Mark’s Hospital, Northwick Park, Watford Road, Harrow, Middlesex, HA1 3UJ, UK

Abstract: The impact of spinal cord injury on an individual’s gastrointestinal tract function is often poorly
understood by the general public and also by those involved with persons with spinal cord injury. This
chapter reviews the anatomy, physiology and function of the gastrointestinal tract, with particular em-
phasis on neurological control mechanisms. In turn, it relates the effect that spinal cord injury has on the
neurological control of the gastrointestinal tract. The symptoms that are encountered by patients in the
acute phase following injury, and by individuals in the months/years after injury, with particular reference
to the effect of altered autonomic nervous system control of the gastrointestinal tract, are discussed.
Together with a following summary of current bowel management regimens and techniques, this chapter
aims to provide an overall view of the effect that autonomic dysfunction due to spinal cord injury has on
gastrointestinal function.

Introduction published work has been dedicated to the urolog-


ical sequelae of spinal cord injury, compared to the
Among the general population, the perceived im- effects on the gastrointestinal system. However, in
pact that spinal cord injury has on people is often recent years this has begun to be researched. This
limited to the noticeable effects of impaired mo- has been fuelled by work that has shown that a
bility. Less well appreciated by the general popu- considerable proportion of cord-injured people
lation is the impact that spinal cord injury has on rate bowel dysfunction as a greater source of
pelvic function, resulting in bladder, bowel and distress than bladder and sexual problems (Stone
sexual dysfunction. et al., 1990a; Glickman and Kamm, 1996; De Looze
Control of the gastrointestinal system involves et al., 1998; Han et al., 1998).
complex interactions between autonomic and so- In both the acute and long-term phases of spinal
matic innervation acting ultimately at the level of cord injury, patients report high levels of gastroin-
the intrinsic enteric nervous system. Following testinal morbidity (Cosman et al., 1991; Krogh
spinal cord injury, this fine control mechanism is et al., 1997; De Looze et al., 1998; Miller et al.,
interrupted to varying degrees, dependent upon 2001). Gastrointestinal problems are a large cause of
the level and extent of the spinal cord injury. The rehospitalization among people with spinal cord in-
result is a spectrum of possible gastrointestinal jury, accounting for 11% of readmissions in a recent
symptoms. To date, the effects of spinal cord in- Australian study (Middleton et al., 2004). This mor-
jury on bowel function and management have bidity has significant cost implications to healthcare
been poorly understood. A much larger volume of systems in the acute hospital and community setting
(Harvey et al., 1992; Johnson et al., 1996).
Corresponding author. Tel.: +020-8235-4084; In the acute spinal cord injury setting, symp-
Fax: +020-8235-4162; E-mail: a.emmanuel@imperial.ac.uk toms can affect any region of the gastrointestinal

DOI: 10.1016/S0079-6123(05)52021-1 317


318

tract. At the upper end of the gut, problems in- a compliant tubular sac, approximately 1.5 m in
clude gastric dilatation, ileus, superior mesenteric length (Sinnatamby, 1999), which can be divided
artery syndrome, peptic ulceration and pan- anatomically into five parts; appendix, cecum,
creatitis. Chronic gastrointestinal symptoms en- colon, rectum and anus (Fig. 2). Embryologically,
countered by people with spinal cord injury the large bowel develops from two separate sourc-
include poorly localized abdominal pain, bloating, es: the proximal colon up to the transverse colon
upper gastrointestinal symptoms such as nausea arising from the mid-gut, and the colon distal to
and vomiting, incontinence and constipation. This the mid-transverse colon, arising from the embry-
list of symptoms which is partially attributable to onic hindgut. Proximally it commences at the
autonomic dysfunction, exists alongside the po- ileocecal valve and distally it ends with the anal
tential for any other acute abdominal pathology, sphincter. The former is of little functional signif-
the diagnosis and treatment of which may often be icance, whereas the latter has obvious major phys-
complicated by the reduction of visceral sensitivity iological importance. The colon follows the
(Bar-On and Ohry, 1995; Miller et al., 2001). The general structure of the gastrointestinal tract, with
symptoms of lower gastrointestinal tract dysfunc- an inner circular smooth muscle layer and a thin
tion following spinal cord injury are more appar- outer longitudinal muscle layer that is gathered up
ent to clinicians, typically presenting later with into thickened cords forming the taenia coli. At
constipation and fecal incontinence. the distal rectum is the anal canal formed from
This chapter aims to give an overview of neuro- anal mucosa overlying two layers of muscle, the
logical control within the gastrointestinal tract and internal and external anal sphincters. The internal
also to review the current understanding of the anal sphincter is formed from a condensation of
symptoms and pathology behind the effect that spi- the inner circular smooth muscle and hence is not
nal cord injury has on the gastrointestinal system. under voluntary control. The external anal sphinc-
ter is made up of a circumferential ‘voluntary’
Background striated muscle band, which is continuous with the
pelvic floor. These sphincters work in conjunction
Bowel anatomy and innervation with the puborectalis muscle, which forms a sling
around the distal rectum and is tethered to the
The gastrointestinal tract from the oesophagus to pubic symphysis to maintain the puborectal angle,
the rectum follows a similar structural pattern of a a minor contributor to the maintenance of fecal
tube whose lumen is formed by concentric layers continence. Tonic contraction of the internal anal
of mucosa, submucosa, circular and longitudinal sphincter provides 80% of resting anal pressure
muscle layers, and an outer serosal covering layer (Schweiger, 1979). When the urge to defecate
(Fig. 1). Between the muscular layers and beneath occurs with rectal distension and puborectalis
the mucosa are collections of nerve cells that form stretch, contraction of the external anal sphincter
plexuses (the submucosal Meissner’s and muscular and puborectalis helps to maintain continence un-
Auerbach’s plexuses) that participate in the con- til there is a suitable moment to void. In addition
trol of gut peristalsis and secretion. Dependent to the voluntary control of external anal sphincter
upon the position in the gastrointestinal tract these function, there is a reflex component, which can be
layers vary in thickness and complexity. This pat- experimentally triggered by a cough or Valsalva
tern is consistent between the gastrointestinal manoeuvre, and which serves physiologically to
tracts of most vertebrates. maintain continence during episodes of raised
The term ‘neurogenic bowel’ relates to colonic intra-abdominal pressure.
dysfunction (constipation, fecal incontinence and
disordered defecation) following disruption of The enteric nervous system
normal control, and is the largest contributor to
gastrointestinal symptoms following spinal cord The enteric nervous system controls the gastroin-
injury. The adult human large intestine consists of testinal tract, via a network of sensory neurones
319

Fig. 1. The layers of the gastrointestinal tract. From Feldman’s GastroAtlas Online, with permission.

Fig. 2. Colon anatomy. From Feldman’s GastroAtlas Online, with permission.


320

relaying information from the gut which in turn (GABA)] and smaller molecules (e.g., nitric oxide)
communicates with a network of interneurones (Olsson and Holmgren, 2001). The role of each
and effector neurones to produce an effect on gut transmitter varies, in each region of the gut, de-
secretion, blood flow and motor function. It has pendent upon the interaction with other local
been estimated that the enteric nervous system transmitters and receptor density on target cells
contains 80–100 million neurones (Furness and (Schemann and Neunlist, 2004). A functional prin-
Costa, 1987) a similar number to that in the spinal ciple states that neuropeptides often act as
cord itself (Goyal and Hirano, 1996). The enteric neuromodulators, as opposed to direct neuro-
nervous system can function independently of the transmitters, in the gut. Enteric nervous system
central nervous system but the central nervous neurones can be broadly classed into intrinsic af-
system plays a large role in coordinating gut func- ferents, interneurones and motor neurones (Lynch
tion. This has led to the concept of the brain gut et al., 2001). The intrinsic afferents form the sen-
axis, with the ‘larger brain’ in the cranium and the sory limb of motor and secretory reflexes, project-
‘mini brain’ in the abdomen. The central nervous ing into the interneurones of both plexuses.
system exerts its effect on the bowel via afferent Excitatory motor and secretory neurones project
and efferent sympathetic, parasympathetic and so- to circular muscle locally or rostrally, whereas in-
matic innervation (Sarna, 1991), which interacts hibitory neurones project caudally. This pattern of
with the intrinsic nervous system. An analogy proximal relaxation with local and distal contrac-
would be of the enteric nervous system being like tion helps coordinate churning and peristaltic gut
constant traffic running through a town, while the contractions. The afferent and motor neurones are
central nervous system is the system of traffic linked by interneurones, forming multi-synaptic
lights and roundabouts that controls the smooth pathways that fine-tune gut secretion and gut mo-
flow of that traffic. tility. While the enteric nervous system coordinates
Nerve cell bodies in the enteric nervous system segmental motility and some peristaltic movement,
are grouped into small ganglia that are connected global colonic movements are triggered by spinal
to each other by nerve processes, producing two cord-mediated reflexes, acting via pelvic nerves.
main plexuses that constitute this intrinsic nervous
system. The myenteric (Auerbach’s) plexus is well
developed, made up of unmyelinated fibres and Parasympathetic innervation
postganglionic parasympathetic cell bodies and
lies between the longitudinal and circular muscles Parasympathetic autonomic innervation from the
of the gut and coordinates peristalsis. It supplies vagus (10th cranial nerve), originating from the
the mucosa with secretomotor innervation and has brainstem, supplies the gastrointestinal tract from
connections with the sympathetic ganglia (Fig. 3). the esophagus up to the colonic splenic flexure
The submucosal (Meissner’s) plexus lies on the (Devroede and Lamarche, 1974) (Fig. 4). Para-
luminal side of the circular muscle in the submu- sympathetic innervation to the splenic flexure, de-
cosa together with connective tissue, glands and scending colon and rectum arises from the sacral
small vessels. It conveys local sensory and motor (S) spinal roots S2–S4 that form the pelvic plexus
responses to Auerbach’s plexus and to the central and give rise to the nervi erigentes. In man, the
nervous system (Stiens et al., 1997). It also has a precise point at which the vagal innervation to the
role in the control of secretions, endocrine cells bowel stops and pelvic innervation starts is a
and the submucosal vasculature. The exact signal- source of controversy with some authors describ-
ling controls between gut neurones have yet to be ing vagal innervation down to the rectum, and
fully elucidated. Neurones have been shown to others reporting pelvic nerve branches travelling
contain the classical adrenergic and cholinergic proximally to innervate the entire colon (Stiens
neurotransmitters, together with putative trans- et al., 1997). The parasympathetic supply to the
mitters such as peptides (e.g., substance P), amino internal anal sphincter is derived from the sacral
acids [e.g., glutamate, g amino-butyric acid spinal cord and joins the pelvic nerves. It relaxes
321

Fig. 3. Layers of the submucosal and myenteric plexuses. From Feldman’s GastroAtlas Online, with permission.

the sphincter by the effect of pre-ganglionic postganglionic fibres innervate the rectum and
cholinergic neurones exciting nicotinic and mu- anus. The internal anal sphincter has sympathetic
scarinic receptors. supply from the inferior mesenteric ganglion via
the hypogastric nerves. Sympathetic tone is exci-
tatory to the internal sphincter musculature and
Sympathetic innervation helps to maintain continence.

Preganglionic axons from thoracic (T) nerve roots


T6–T12 pass via rami communicantes to the sym- Colonic function, reflexes and control
pathetic chain and travel via thoracic splanchnic
nerves, synapsing at the celiac and superior mes- The colon serves several functions: stool storage;
enteric plexus, supplying small bowel and the as- stool propulsion when socially appropriate; the
cending colon. Sympathetic innervation distal to provision of an environment for symbiotic bacte-
the splenic flexure to the upper rectum arises from rial growth; and even absorption of amino acids,
lumbar (L) nerve roots L1–L3. The nerves travel short chain fatty acids and fluid. Following the
to the sympathetic chain and via the lumbar disruption of supraspinal control systems, the
splanchnic nerves, synapse at the inferior mesen- dominant autonomic tone is inhibitory to colonic
teric ganglia. The supply then follows the arterial propulsion, which contributes towards constipa-
blood supply to the left colon. The lower rectum tion. The understanding of these neural mecha-
and anal canal sympathetic supply is derived from nisms and their aberrant behaviour after spinal
the aortic and lumbar splanchnics which unite to cord injury may provide a basis to treat symptoms.
form the hypogastric plexus, giving off the pre- Normal patterns of colonic contraction can be
sacral branches to form the sacral plexus, whose classified into three groups: (i) individual phasic
322

Fig. 4. Autonomic innervation of the colon. From Feldman’s GastroAtlas Online, with permission.

contractions (of long or short duration), which mechanisms controlling it are inadequately under-
have the effect of kneading and mixing stool; (ii) stood. Enteric reflexes, with serotonin as the
organized groups (migratory and non-migratory neurotransmitter, stimulate peristalsis (Hansen,
motor complexes), which are propulsive in small 2003), as demonstrated by its continuation after
regions of colon; (iii) giant migratory contractions, the gut is removed from the body. These enteric
which produce movements of content and expel reflexes also contribute to the colonic slow wave
stool during defecation (Christensen, 1991; Sarna, activity (Olsson and Holmgren, 2001). Recent
1993). In addition, distension of the wall of the studies have suggested that interstitial cells of
colon causes proximal muscle contraction and Cajal, found in the submucosa, intra- and inter-
distal relaxation, resulting in caudal propagation. muscle layers of the gut, especially in the right co-
The colon has intrinsic rhythmic slow wave lon, generate spontaneously active pacemaker
activity that is thought to be important in encour- currents (Horowitz et al., 1999; Takaki, 2003).
aging fluid reabsorption from the colonic mucosa. These ‘pacemaker’ cells in the colon are likely to
The origin of this activity varies and the have profound effects on colonic smooth muscles.
323

One of the specialized aspects of gastrointestinal properties of these two reflexes are exploited in
motility is the gastrocolic reflex. This reflex causes bowel management, to aid defecation, as long as
increased small bowel and colonic propulsive mo- the spinal cord lesion is above the level of the conus.
tility, and is mediated by neural and endocrine Spinal cord injury results in disruption of the
mechanisms. The neural substrates comprise cho- interaction that normally occurs between the in-
linergic motor neurones which are activated by the trinsic and extrinsic nervous system. Studies look-
ingestion of a meal (Connell et al., 1963). The ing at the effect of spinal cord and peripheral nerve
stomach is not the source of the stimulus as the lesions on the enteric nervous system have shown
response is triggered if food bypasses the stomach ganglion cell loss and secondary Schwann
(by a feeding tube) and enters the duodenum di- cell proliferation in the colon (Devroede and
rectly (Snape, Jr. et al., 1979; Christensen, 1991) Lamarche, 1974; Devroede et al., 1979). Once
and even by the psychological anticipation or established, recovery of this disruption is restrict-
smell of food. The proposed mechanisms for the ed. However, recent work with enteric glia cells has
reflex include a role for central vagal mediation, shown that they have the potential to aid axonal
possibly long enteric reflexes via the enteric nerv- growth (Jiang et al., 2003) and may be a source of
ous system and humoral components related to future regenerative therapies.
release of cholecystokinin, gastrin and motilin
(Christensen, 1991; Saltzstein et al., 1995). Studies
in spinal cord-injured subjects have demonstrated Gastrointestinal dysfunction with acute spinal
differing recorded responses of the reflex, either cord injury
showing it to be intact or absent (Glick et al.,
1984). The reasons for this discrepancy may simply In the acute phase of spinal cord injury, tonic ex-
be methodological, and in the clinical setting at citatory input to ganglionic and enteric neurones is
least the reflex is frequently used as a management lost and the neurones are less excitable, resulting in
tool for treating constipation in cord-injured peo- overall lack of neural input to the gut. Commonly
ple. By ingesting food or a calorific drink approx- encountered complications in this acute period of
imately 30 min before bowel management is ‘spinal shock’ include ileus, gastric dilatation, pep-
planned, reflex colonic contractions can aid stool tic ulcer disease, pancreatitis and superior mesen-
emptying (Longo et al., 1989). Fatty foods tend to teric artery syndrome (Tibbs et al., 1979; Gore
have larger and longer action on the reflex com- et al., 1981; Berlly and Wilmot, 1984). The latter
pared to protein or carbohydrate-dominant foods complication, which is compression of the third
(Spiller, 2000). part of the duodenum by the superior mesenteric
Pelvic sacral reflexes are excitatory, with the re- artery, tends to occur due to alterations of vascular
flex arc conveyed from the sacral spinal cord seg- tone to the viscera. This spectrum of ‘spinal shock’
ments in the conus, to and from the colon via pelvic abnormalities tends to settle over a varying period
nerves. Parasympathetic stimulation of splanchnic of days to weeks after the initial injury (Ditunno
nerves leads to a significant propulsive colonic re- et al., 2004). Peptic ulceration in the acute setting
sponse. From the colon, enteric nerves trigger this is thought to occur as a result of unopposed para-
reflex in response to stretch or dilation, reinforcing sympathetic activity from the vagus and transient
inherent colonic enteric-mediated peristalsis. The loss of sympathetic innervation, resulting in raised
rectocolic reflex is another pelvic reflex that is trig- gastrin levels and a reduced pH (Pollock and
gered by mechanical or chemical stimulation in the Finkelman, 1954; Bowen et al., 1974; Tanaka
rectum or anus. It also produces colonic peristalsis, et al., 1979). Analgesic and corticosteroid admin-
which brings stool down to the rectum. Stool istration following spinal injury may exacerbate
entering the rectum can then trigger the recto-anal the condition. These drugs have also been impli-
inhibitory reflex, a reflex relaxation of the internal cated in the prevalence of pancreatitis in acute spi-
anal sphincter in response to rectal distension nal cord injury. Other causes of pancreatitis which
allowing expulsion of stool from the rectum. The have been described include autonomic imbalance
324

causing over stimulation of the sphincter of Oddi, same lines as in able-bodied patients. The focus is
hypercalcaemia due to immobilization and thick- on antacids (alginates), acid suppressants (hista-
ened pancreatic secretions (Hyman et al., 1972; mine (H)2-receptor antagonists and proton pump
Carey et al., 1977; Maynard and Imai, 1977). inhibitors) and motility stimulating agents (dom-
With regard to lower gut function, animal peridone, metoclopramide).
studies have shown decreased colonic motility Nausea and vomiting related to gastric dilata-
immediately after thoracic cord transection tion and ileus are common symptoms in the acute
(Meshkinpour et al., 1985). Inhibitory reflexes spinal cord injury setting. Both tend to improve as
below the lesion are lost and there is a loss of spinal shock resolves. Nausea, however, can also
facilitation from above. This loss of supra-lesional be a persistent and troublesome symptom in the
input to the bowel in large part explains the longer term (Stone et al., 1990a; Glickman and
reduced transit through the bowel that is found. Kamm, 1996). There are a number of possible
Ileus occurs almost immediately in patients with causes for these symptoms: gastric stasis (second-
thoracolumbar cord injury but can be delayed in ary to denervation), gallstone disease (which is
high thoracic and cervical nerve injuries. However, more prevalent in spinal cord injury patients) and
it is most commonly seen in patients with higher constipation (Camilleri, 1990; Pfeifer et al., 1996;
lesions when cord injury occurs at or above the Tola et al., 2000; Cubeddu, 2003).
level of visceral innervation, namely T5. Gastric emptying is delayed after spinal cord
injury (Lu et al., 1998; Kao et al., 1999). Vagal
Gastrointestinal dysfunction with chronic spinal parasympathetic innervation to the upper gastro-
cord injury intestinal tract originates from the brainstem and
its control tends to be preserved in spinal cord in-
Upper gastrointestinal symptoms jury. However, the sympathetic outflow arises
from the thoracic lumbar cord (T5–T12) and its
Little work has been published on the extent and loss with a lesion about this level results in exces-
mechanisms of upper gastrointestinal dysfunction sive splanchnic sympathetic activity from the tho-
affecting cord-injured people, and much of it is racic cord, and hence gastroparesis (decreased
contradictory. Mild upper gastrointestinal symp- gastric emptying). After lower level injuries, there
toms have been reported to affect a third of cord- is no loss of supra-spinal influence and the auto-
injured people (Lu et al., 1998). Heartburn and nomic hyper-reflexia and delayed gastric emptying
dysphagia have been reported in 61 and 30%, is not seen (Fealey et al., 1984; Nino-Murcia and
respectively, of injured individuals, which is of Friedland, 1992). There is some evidence to sug-
greater prevalence than in matched controls. This gest that gastric emptying tends to return towards
symptom-burden is associated with high levels normal over time as a degree of autonomic nerv-
of endoscopic and histological evidence of es- ous system-mediated homeostasis and regulation
ophagitis (Stinneford et al., 1993). Oesophageal returns in long-term spinal cord-injured individu-
motility studies also show abnormal slow wave als (Segal et al., 1987).
peristaltic propagation, the equivalent of the Orocecal transit times are delayed after spinal
slowed motor abnormalities seen further down cord injury (Chen et al., 2004). Using a non-
the gastrointestinal tract. The cause and relevance invasive hydrogen breath test method, these au-
of these findings remains unknown. thors have shown that cord-injured subjects have
A high prevalence of hiatus hernia is found after overall mean orocecal transit times of 180 min com-
spinal cord injury, which appears to be related to a pared to 98 min in controls. The net result of this
reduction of diaphragmatic motion, muscle atro- prolonged small bowel transit time, is to predispose
phy and weakening of fibrous tissue at the gastro- to disturbance of digestion and bacterial over-
oesophageal junction due to chronically raised growth which can exacerbate nausea. Nausea has
intra-abdominal pressures. Treatment of hiatus also been attributed, in part, to the higher incidence
hernia and other reflux type symptoms is along the of gallstone disease found in cord-injured patients
325

(Ketover et al., 1996). Studies looking at the gall- symptoms in these patients. Constipation, fecal
bladder following spinal cord injury, have shown incontinence and incoordinated defecation are the
reduced contractility and this has been postulated most frequently reported symptoms. The incidence
as possible reason for the increase in incidence of constipation reported in the literature ranges
(Fong et al., 2003). Gallstones have been implicated from 20 to 58%. This discrepancy in the reported
in causing non-specific symptoms such as nausea figures can be attributed to a disparity between
and bloating in cord-injured people (Moonka et al., definitions of constipation and bowel management
1999). However, given the prevalence of gallstones practices used between spinal injury units. Incon-
after spinal cord injury, the presence of vague ab- tinence to feces and flatus is reported to affect up
dominal symptoms that occur should not be put to 75% of the spinal cord injury population al-
down to the presence of gallstones alone (Moonka though the percentage of cord-injured people in
et al., 2000). Gastric or colonic pathology should be whom this occurs on a regular basis (more than
considered in the diagnostic workup. monthly) is only approximately 15% (Krogh et al.,
1997). However, the threat of episodes of fecal in-
continence causes psychological stresses to cord-
Pain
injured people and their carers and can result in
social isolation.
Abdominal pain experienced by people after spinal
Colonic diverticulae are found more frequently
cord injury needs to be carefully investigated to
and at younger ages in the spinal cord injury pop-
exclude common abdominal pathology (such as
ulation compared with controls (Gore et al., 1981).
neoplasm, peptic ulceration and ischemia), as di-
This may be due to the contribution of the high
agnosis in this population can be fraught with dif-
pressures that uncoordinated segmental peristalsis
ficulty (Ingersoll, 1985; Bar-On and Ohry, 1995).
can produce and of chronic intraluminal disten-
Chronic neurological visceral pain does affect
sion (Gore et al., 1981). Hemorrhoids are also
cord-injured people but the extent of this problem
common with up to three-quarters of cord-
is poorly documented and understood partly as a
injured people having the problem (Stone et al.,
result of poor classification (Beric, 2003). Studies
1990a).
quote the prevalence of chronic visceral pain as
between 3 and 10% (Cardenas et al., 2002). Al-
Upper versus lower motor neurone lesions: effect on
though not as common as musculoskeletal or ne-
the bowel
uropathic pain, visceral pain is perceived as being
of higher intensity (severe/excruciating) compared
When describing symptoms attributable to bowel
to musculoskeletal pain. Visceral pain tends to de-
dysfunction after spinal cord injury, an under-
velop months or years after injury, compared with
standing of the effect that the level of injury has on
other pain types that more likely have an early
the bowel is necessary, as this determines the pat-
onset. This probably results following the devel-
tern of colonic motility. Upper motor neurone
opment of visceral organ problems associated with
spinal cord injury lesions occur above the level of
spinal cord injury such as constipation, bladder
the conus medullaris, which in adults lies at the
infection and renal calculi. Visceral pain may be
lumbar (L)1,2 level. The colon in these cases is
due to normal afferent sensation via the sympa-
described as ‘spastic’ with increased colonic wall
thetic and vagus nerves in paraplegics, or vagal
and striated external anal sphincter muscle tone.
innervation alone in tetraplegics (Richards, 1992;
Baseline colonic activity is higher in this group
Siddall and Loeser, 2001).
compared to controls (Aaronson et al., 1985).
Rectal tone is high (Krogh et al., 2002), resulting
Lower gastrointestinal symptoms and pathology in a reduced capacity to hold stool and therefore
increasing the risk of fecal incontinence episodes.
Colorectal dysfunction following spinal cord This gives rise to poorly coordinated peristalsis,
injury is the major source of gastrointestinal with excessive segmental and reduced propulsive
326

peristalsis. The resulting slow whole gut transit recto-anal inhibitory reflex described above is
results in constipation that is often exacerbated by present after spinal cord injury but differs from
changes in puborectalis muscle function. Evacua- controls in that it can be triggered with lower vol-
tion of feces is achieved by triggering reflex def- umes compared to controls. It has been hypoth-
ecation either by mechanical (digitation) or esized that the cause of this could be decreased
chemical means (suppositories or enemas). rectal compliance (a less distensible rectum) re-
Lower motor neurone lesions occur with injuries sulting in lower threshold for stimulating the reflex
at the level of the conus, cauda equina or pelvic (Meshkinpour et al., 1983; Glick et al., 1984). This
nerves resulting in the disruption of parasympa- combination of a less distensible rectum and reflex
thetic innervation to the bowel. Loss of parasym- anal relaxation contributes towards triggering ep-
pathetic control results in a flaccid bowel and low isodes of fecal incontinence in spinal cord injury.
internal anal sphincter tone. There is also an ab-
sence of spinal cord-mediated reflex peristalsis, and Constipation and incontinence
hence stool propulsion occurs with intrinsic my-
enteric plexus-triggered segmental peristalsis. With Constipation is common after spinal cord injury
the loss of external anal sphincter control and the (Glickman and Kamm, 1996). The frequency of
absence of internal anal sphincter parasympathetic constipation is affected by the level of injury, with
supply, the anal sphincter complex resting tone is up to three-quarters of quadriplegics being affect-
low. This low pressure makes cord-injured people ed, falling to a third in paraplegics with lesions
susceptible to passive fecal leakage. This tendency between the T10 and L2 cord segments (De Looze
is exacerbated by the associated loss of rectal tone, et al., 1998). This is due to delayed colonic transit,
resulting in a capacious rectum full of stool. Man- disordered evacuation and changes in visceral sen-
ual removal of stool, aided by increases in intra- sitivity. Investigating and researching constipation
abdominal pressure (such as with a Valsalva) is the is difficult since imaging and measuring bowel
mainstays of management of such people. motility is not straightforward. However there are
techniques available: radionucleotide and radio-
Incoordinate anal sphincter function opaque marker studies (van der Sijp et al., 1993);
solid state pressure catheters (Fajardo et al., 2003);
There are a number of reports in the literature of balloon distension and Barostat recorders
abnormal anorectal physiology in spinal cord- (Bruninga and Camilleri, 1997). Using these tech-
injured people. Resting sphincter tone (mainly a niques, it is possible to understand the patient’s
reflection of internal anal sphincter function) is bowel motility pattern and allow therapeutic in-
reduced in cord-injured people compared to con- terventions to be directed appropriately. Most
trols, and is maintained mainly by internal anal studies show overall colonic transit times to be
sphincter activity possibly due to tonic excitatory prolonged following spinal cord injury (Menardo
sympathetic discharge (Lynch et al., 2000). The et al., 1987). Some studies have suggested that this
Valsalva manoeuvre causes a rise in intra-abdom- delay in colonic transit is segmental, most mark-
inal pressure that is thought to stimulate pelvic edly in the distal colon and rectum (Menardo
floor tension receptors into triggering reflex exter- et al., 1987; Beuret-Blanquart et al., 1990). By
nal anal sphincter contraction (MacDonagh et al., contrast, others show a pan-colonic increase in
1992). In able-bodied individuals, a cortically me- transit times (Keshavarzian et al., 1995). In the
diated pathway relaxes the external anal sphincter clinical setting, groups of cord-injured patients
during straining to defecate. With upper motor benefit from oral laxatives in addition to rectal
neurone lesions where the reflex pathway is intact medications and reflex stimulation, indicating a
but the supra-lesional input is absent, Valsalva reduction in whole colon motility. Poor coordina-
manoeuvres for bowel emptying may actually tion of the anal sphincter complex described pre-
worsen attempts to evacuate stool, as the external viously, leading to outlet obstruction, can also
anal sphincter tone increases on straining. The contribute towards constipation. The importance
327

of a good bowel care programme, to prevent con- phase of spinal shock when peristalsis is reduced,
stipation, is underlined by some studies that show digital or manual evacuation of stool is required
high levels (73%) of megacolon (dilated colon sec- (Halm, 1990). When bowel function stabilizes, a
ondary to constipation) in people with spinal cord regular bowel care program can be initiated. Cur-
injury (Harari and Minaker, 2000). rent programs vary between institutions, where
Incontinence is a threat to cord-injured people management is often empirical, given the lack of
due to a combination of factors — lack of aware- well-designed controlled trials (Wiesel et al., 2001).
ness of rectal fullness, overflow following poor There have been published proposed bowel pro-
evacuation management, and weak sphincter func- grams (Correa and Rotter, 2000) but none have
tion as described above (particularly in lower mo- been universally adopted and for many cord-in-
tor neurone lesion patients). Poor control of flatus jured people, bowel management regimens are far
and fecal leakage can lead to physical, psycholog- from ideal. One series reported 41% of cord-in-
ical, sexual and social problems (DeLisa and jured individuals spending more than 1 h on bowel
Kirshblum, 1997). Effective bowel management evacuation (Harari et al., 1997) and some people
strategies to prevent these symptoms are impor- report having to spend 3 h or more a day on their
tant for the well being of cord-injured people. bowel care. It has been quoted that the ideal bowel
Regular evacuation and preventing loose stool management regime should be self controlled and
formation by ensuring an adequate fibre intake, spontaneous, with or without oral medication,
preventing gastrointestinal infections and regulat- performed at least once every 2 days, and com-
ing diet are core techniques that will be discussed pleted within 30 min to result in effective evacua-
further later in this chapter. tion without complication and this was achieved
by only 32% of subjects in one study (Han et al.,
Therapies that exacerbate symptoms 1998). In the clinical setting, it is accepted that
there have been improvements in bowel care over
Spinal cord injuries cause dysfunction to many the last two decades although hard evidence to
organ systems and the treatment of these can result back this up is scarce.
in adverse effects on the bowel. Side effects from The foundation of good bowel management
prescribed medications are very common. Anti- program involves the implementation of a regular
cholinergics used in the treatment of bladder routine, which addresses the specific issues such as
dyssynergia, opiates and anti-spasmodics slow constipation, incontinence and functional mobility,
bowel transit and dry the stool thereby exacerbat- using the appropriate interventions. A bowel care
ing the constipation. Broad-spectrum antibiotics routine should be timed to coincide with colonic
can cause diarrhea by altering the balance of com- giant migratory contractions, to take advantage of
mensal enteric flora in the gut. Additionally, ther- any stool propulsion. Giant migratory contrac-
apies used in bowel management can exacerbate tions tend to occur after meals and in the morning,
symptoms. Anal digitation, evacuation and rectal on waking. This regime should take into account
medication administration can cause local trauma the person’s social, sexual, cultural and vocational
potentially irritating hemorrhoids and predispos- beliefs. Also, the question of functional mobility
ing towards anal fissure and solitary rectal ulcer needs to be addressed, ensuring carers and appro-
formation. All treatments and therapies should priate equipment such as commode chairs are
therefore be evaluated for side effects, and vigi- available. Logistical issues such as access to toilet
lance observed for their onset. facilities, are fundamental but often overlooked.

Bowel management Diet

Early implementation of a regulated-controlled Simple dietary measures can benefit bowel man-
bowel management program is held to be the best agement. Adequate fluid intake aids gut transit by
practice for patients after their injuries. During the softening stool. In people with spinal cord injury,
328

fibre does not increase colonic transit time but acts fecal incontinence. Senna may be used as an oc-
to absorb excess water and to keep the stool soft casional night-time dose to ‘prime’ the bowel for a
and formed, thereby reducing the problems of in- morning evacuation. Combinations of the differ-
continence (Banwell et al., 1993). ent classes of laxatives often deliver the desired
results. For example, a regular bulking agent with
Laxatives a stimulant laxative can lead to the regular evac-
uation of soft, formed stools.
There are several groups of medications that can
be taken orally to aid bowel movements or to re-
Suppositories and enemas
lieve constipation. Lubricants and stool softeners
(e.g., docusate sodium, liquid paraffin and mineral
Glycerine suppositories are used to stimulate rectal
oils) ‘grease’ the stool and make passage through
contraction due to its irritant and hyperosmotic
the bowel easier. Bulking agents (e.g., isogel gran-
action and result in bowel movements in
ules, ispaghula husk, methylcellulose, psyllium)
15–30 min. Bisacodyl can be administered in sup-
are indicated if dietary fibre cannot be adequately
pository form and it acts on sensory afferent
ingested. They act by absorbing water in the gut
nerves of the mucosa producing a parasympathet-
thereby softening and bulking stool. Fluid intake
ically mediated reflex peristaltic contraction of the
must be adequate when on these treatments, al-
entire colon (Stiens et al., 1998), which aids bowel
though this can be difficult to achieve in people
emptying and reduces time spent on bowel man-
with bladder management difficulties. Bloating
agement (Frisbie, 1997). Saline, water or docusate
and flatulence can be problematic but tend to set-
sodium enemas can be used. These work by trig-
tle if people can persevere with the treatment.
gering reflex colonic peristalsis, lubricating and in
Indigestible carbohydrates (lactitol, lactulose,
the case of docusate softening the stool. Auto-
polyethylene glycols) and salts [e.g., magnesium
nomic dysreflexia is the condition of abrupt onset,
(Epsom) salts] act osmotically to draw fluid into
potentially lethal hypertension in people with spi-
the colon. Stimulant laxatives (bisacodyl, da-
nal cord injury above the level of T6 (see chapters
nthron, senna) induce and augment peristaltic
addressing cardiovascular dysfunction, this vol-
movement of the bowel, thus aiding stool progres-
ume). It is caused by uncontrolled sympathetic
sion and reducing the time allowed for water and
discharge triggered by any noxious stimulus and
electrolyte resorption. Senna is broken down in
many innocuous stimuli below the level of the le-
absorbable anthraquinones which directly stimu-
sion. Common triggers of this condition relate to
late the myenteric plexus. Oral stimulant laxatives
bladder and bowel distension or irritation (Adsit
can all cause the side effects of cramps, diarrhea
and Bishop, 1995). The maneuvers involved with
and dehydration. Their chronic use can lead to
bowel management such as digitation, use of en-
colonic mucosal staining due to macrophage
emas and evacuation can trigger autonomic
phagocytosis of pigments derived from laxatives
dysreflexia particularly if there is local pathology
(melanosis coli) (Menter et al., 1997). There is no
such as anal fissure or rectal ulceration. Use of
evidence in cord-injured people that their already
local anesthetic agents, such as lidocaine gel can
very slow transit is further compromised by reg-
reduce the incidence of attacks of autonomic
ular use of these agents. The delay of onset for
dysreflexia during bowel management.
these laxatives is 1–2 days, except for magnesium
salts that have a faster onset of action of about 4 h
(Frisbie, 1997; Amir et al., 1998). The attraction of Prokinetic agents
osmotic agents is their speed of action and their
ability to be titrated, done according to stool con- Metoclopramide and domperidone are dopamine
sistency. Care must be taken however to avoid too antagonists that increase the rate of gastric emp-
loose a stool, especially in people with sphincter tying and of small gut transit. They have no effect
compromise, as they may lead frequent episodes of on colonic peristalsis and are most commonly used
329

in the acute setting of spinal cord injury when try- (Chait et al., 1997) or endoscopic placement (De
ing to overcome the gastric dilatation and ileus that Peppo et al., 1999). These irrigation methods have
accompanies spinal shock (Miller and Fenzl, 1981; been most studied in pediatric practice, especially
Segal et al., 1987). Erythromycin is a macrolide in children with myelomeningocoele. While often
antibiotic with prokinetic effects which is also used efficient in the short term, infective and mechanical
in the acute setting to enhance transit through the complications around the tube entry site can be
upper gut (Clanton and Bender, 1999). problematic. Furthermore, there is evidence that
The parasympathomimetic drugs neostigmine, with time, the irrigation method may become less
bethanechol, distigmine and pyridostigmine, all efficient, and indeed the antegrade continence en-
enhance parasympathetic effects on the gut to in- ema openings in the abdominal wall frequently
crease motility but are rarely used in the clinical stenose (McAndrew and Malone, 2002).
setting for this purpose, due to their side effects. Severe refractory constipation, prolonged bowel
They may have a role in treating the rare situation care time, fecal incontinence and chronic peri-anal
of acute pseudo-obstruction of the gut (Ogilvie’s ulcers are reasons for cord-injured people to con-
Syndrome) which is seen in some cases after acute sider stoma formation for their bowel care
injury, related to sudden loss of autonomic tone to (Deshmukh et al., 1996; Pfeifer et al., 1996). How-
the viscera (Delgado-Aros and Camilleri, 2003). ever, the decision to opt for surgery should not be
Cisapride was used for its prokinetic properties on taken lightly as there are issues of assessment of
the upper and lower gut and increased colonic the current bowel care program, body image, life-
transit speed (Binnie et al., 1988; Geders et al., style, and required nursing assistance to be taken
1995; Longo et al., 1995) but has been withdrawn into account together with the high risks of surgery
from clinical use because of an association with in this group of patients. That said, the formation
fatal cardiac arrhythmias (Prescrire Int, 2000; of a stoma (ileostomy or colostomy) has been
Cubeddu, 2003). shown in several studies to improve the quality of
life for people who opt for this treatment option as
it can simplify bowel management, reduce incon-
Mechanical devices and surgical interventions tinence and bloating and increase independence
(Stone et al., 1990b; Randell et al., 2001; Branagan
Anal plug devices can be utilized to prevent leak- et al., 2003). Choice of type and position of stoma
age of flatus and feces (Kim et al., 2001) for cord- should be assessed according to the person, de-
injured people with lower motor neurone lesions, pendent upon their colonic transit characteristics
who often have an atonic anal sphincter. They are and their mobility (Safadi et al., 2003).
best tolerated by people who have no preservation Sacral anterior root stimulators have been
of anal sensation, but tend to be inefficient if large implanted since 1977 (Brindley et al., 1986), orig-
volumes of stool are being lost. Pulsed irrigation inally being used for functional electrical stimula-
enemas have been used, in which a catheter with tion to control bladder emptying (Brindley and
an inflatable retention cuff is passed into the rec- Rushton, 1990; Binnie et al., 1991; Brindley, 1994).
tum followed by a program of tap water pulses The implant consists of a subcutaneous radio-re-
(Puet et al., 1997). This loosens and suspends stool ceiver connected to S2, S3 and S4 nerve roots via
that is removed via a conduit drain running tunnelled wires. Trains of high-frequency stimula-
through the centre of the catheter. Antegrade con- tion trigger complex, high pressure, phasic colonic
tinence enemas require appendicocecostomies to and rectal contractions resembling peristaltic
be surgically fashioned to deliver washout fluid movements, which act to bring stool down distal-
into the proximal colon to allow controlled daily ly. Following stimulation, defecation may occur
emptying. Water or saline is infused into the (Varma et al., 1986; MacDonagh et al., 1990;
cecum and passes through to produce bowel evac- Varma, 1992) and indeed this procedure was found
uation minutes later (Malone, 2004). The tech- to improve bowel management in addition to
nique has been modified to allow radiological bladder care in some patients.
330

Conclusion Binnie, N.R., Creasey, G.H., Edmond, P. and Smith, A.N.


(1988) The action of cisapride on the chronic constipation of
Gastrointestinal symptoms and the required pro- paraplegia. Paraplegia, 26: 151–158.
Binnie, N.R., Smith, A.N., Creasey, G.H. and Edmond, P.
cedures for management of the bowel in people (1991) Constipation associated with chronic spinal cord in-
with spinal cord injury are very problematic and jury: the effect of pelvic parasympathetic stimulation by the
distressing. Unlike the advances in limb and blad- Brindley stimulator. Paraplegia, 29: 463–469.
der dysfunction, the understanding of the effects of Bowen, J.C., Fleming, W.H. and Thompson, J.C. (1974) In-
spinal cord injury on the bowel is still very poor. creased gastrin release following penetrating central nervous
system injury. Surgery, 75: 720–724.
Improvement in the quality of life for this group of Branagan, G., Tromans, A. and Finnis, D. (2003) Effect of
people requires on-going basic clinical research in stoma formation on bowel care and quality of life in patients
this field. Greater understanding of the influence with spinal cord injury. Spinal Cord, 41: 680–683.
of neural disconnection on the residual function of Brindley, G.S. (1994) The first 500 patients with sacral anterior
the gut (via the enteric nervous system) is needed. root stimulator implants: general description. Paraplegia, 32:
795–805.
Specifically, the understanding of the role of pelvic
Brindley, G.S., Polkey, C.E., Rushton, D.N. and Cardozo, L.
reflexes in controlling evacuation and continence is (1986) Sacral anterior root stimulators for bladder control in
required to offer the prospect of possible future paraplegia: the first 50 cases. J. Neurol. Neurosurg. Psychiat.,
neuromodulation of these reflexes. Finally, under- 49: 1104–1114.
standing of the quality of life implications of bowel Brindley, G.S. and Rushton, D.N. (1990) Long-term follow-up
of patients with sacral anterior root stimulator implants.
dysfunction and developments of simple means to
Paraplegia, 28: 469–475.
remedy the socially isolating problems is required. Bruninga, K. and Camilleri, M. (1997) Colonic motility and
The above necessitates a combination of labora- tone after spinal cord and cauda equina injury. Am. J.
tory and clinical research, in addition to advances Gastroenterol., 92: 891–894.
in nursing care. All of this requires a greater pri- Camilleri, M. (1990) Disorders of gastrointestinal motility in
neurologic diseases. Mayo Clin. Proc., 65: 825–846.
ority to be placed on the research and the clinical
Cardenas, D.D., Turner, J.A., Warms, C.A. and Marshall, H.M.
agenda for spinally injured people. (2002) Classification of chronic pain associated with spinal
cord injuries. Arch. Phys. Med. Rehabil., 83: 1708–1714.
Carey, M.E., Nance, F.C., Kirgis, H.D., Young, H.F., Megison
References Jr., L.C. and Kline, D.G. (1977) Pancreatitis following spinal
cord injury. J. Neurosurg., 47: 917–922.
Aaronson, M.J., Freed, M.M. and Burakoff, R. (1985) Colonic Chait, P.G., Shandling, B., Richards, H.M. and Connolly, B.L.
myoelectric activity in persons with spinal cord injury. Dig. (1997) Fecal incontinence in children: treatment with percu-
Dis. Sci., 30: 295–300. taneous cecostomy tube placement — a prospective study.
Adsit, P.A. and Bishop, C. (1995) Autonomic dysreflexia — Radiology, 203: 621–624.
don’t let it be a surprise. Orthop. Nurs., 14: 17–20. Chen, C.Y., Chuang, T.Y., Tsai, Y.A., Tai, H.C., Lu, C.L., Kang,
Amir, I., Sharma, R., Bauman, W.A. and Korsten, M.A. (1998) L.J., Lu, R.H., Chang, F.Y. and Lee, S.D. (2004) Loss of sym-
Bowel care for individuals with spinal cord injury: compar- pathetic coordination appears to delay gastrointestinal transit
ison of four approaches. J. Spinal Cord Med., 21: 21–24. in patients with spinal cord injury. Dig. Dis. Sci., 49: 738–743.
Banwell, J.G., Creasey, G.H., Aggarwal, A.M. and Mortimer, Christensen, J. (1991) The motor function of the colon. In:
J.T. (1993) Management of the neurogenic bowel in patients Yamada T. (Ed.), Textbook of Gastroenterology. Lippincott,
with spinal cord injury. Urol. Clin. North Am., 20: 517–526. Philadelphia, pp. 180–196.
Bar-On, Z. and Ohry, A. (1995) The acute abdomen in spinal Clanton Jr., L.J. and Bender, J. (1999) Refractory spinal cord
cord injury individuals. Paraplegia, 33: 704–706. injury induced gastroparesis: resolution with erythromycin
Beric, A. (2003) Spinal cord injury pain. Eur. J. Pain, 7: lactobionate, a case report. J. Spinal Cord Med., 22: 236–238.
335–338. Connell, A.M., Frankel, H. and Guttman, L. (1963) The
Berlly, M.H. and Wilmot, C.B. (1984) Acute abdominal emer- motility of the pelvic colon following complete lesions of the
gencies during the first four weeks after spinal cord injury. spinal cord. Paraplegia, 104: 98–115.
Arch. Phys. Med. Rehabil., 65: 687–690. Correa, G.I. and Rotter, K.P. (2000) Clinical evaluation and
Beuret-Blanquart, F., Weber, J., Gouverneur, J.P., management of neurogenic bowel after spinal cord injury.
Demangeon, S. and Denis, P. (1990) Colonic transit time Spinal Cord, 38: 301–308.
and anorectal manometric anomalies in 19 patients with Cosman, B.C., Stone, J.M. and Perkash, I. (1991) Gastrointes-
complete transection of the spinal cord. J. Auton. Nerv. tinal complications of chronic spinal cord injury. J. Am.
Syst., 30: 199–207. Paraplegia Soc., 14: 175–181.
331

Cubeddu, L.X. (2003) QT prolongation and fatal arrhythmias: Gore, R.M., Mintzer, R.A. and Calenoff, L. (1981) Gastroin-
a review of clinical implications and effects of drugs. Am. J. testinal complications of spinal cord injury. Spine, 6:
Ther., 10: 452–457. 538–544.
De Looze, D., Van Laere, M., De Muynck, M., Beke, R. and Goyal, R.K. and Hirano, I. (1996) The enteric nervous system.
Elewaut, A. (1998) Constipation and other chronic gastro- N. Engl. J. Med., 334: 1106–1115.
intestinal problems in spinal cord injury patients. Spinal Halm, M.A. (1990) Elimination concerns with acute spinal cord
Cord, 36: 63–66. trauma. Assessment and nursing interventions. Crit. Care
De Peppo, F., Iacobelli, B.D., De Gennaro, M., Colajacomo, Nurs. Clin. North Am., 2: 385–398.
M. and Rivosecchi, M. (1999) Percutaneous endoscopic Han, T.R., Kim, J.H. and Kwon, B.S. (1998) Chronic gastro-
cecostomy for antegrade colonic irrigation in fecally incon- intestinal problems and bowel dysfunction in patients with
tinent children. Endoscopy, 31: 501–503. spinal cord injury. Spinal Cord, 36: 485–490.
Delgado-Aros, S. and Camilleri, M. (2003) Clinical manage- Hansen, M.B. (2003) Neurohumoral control of gastrointestinal
ment of acute colonic pseudo-obstruction in patients: a sys- motility. Physiol. Res., 52: 1–30.
tematic review of the literature. Gastroenterol. Hepatol., 26: Harari, D. and Minaker, K.L. (2000) Megacolon in patients
646–655. with chronic spinal cord injury. Spinal Cord, 38: 331–339.
DeLisa, J.A. and Kirshblum, S. (1997) A review: frustrations Harari, D., Sarkarati, M., Gurwitz, J.H., McGlinchey-Berroth,
and needs in clinical care of spinal cord injury patients. G. and Minaker, K.L. (1997) Constipation-related symptoms
J. Spinal. Cord Med., 20: 384–390. and bowel program concerning individuals with spinal cord
Deshmukh, G.R., Barkel, D.C., Sevo, D. and Hergenroeder, P. injury. Spinal Cord, 35: 394–401.
(1996) Use or misuse of colostomy to heal pressure ulcers. Harvey, C., Wilson, S.E., Greene, C.G., Berkowitz, M. and
Dis. Colon Rectum, 39: 737–738. Stripling, T.E. (1992) New estimates of the direct costs of
Devroede, G., Arhan, P., Duguay, C., Tetreault, L., Akoury, H. traumatic spinal cord injuries: results of a nationwide survey.
and Perey, B. (1979) Traumatic constipation. Gastroenterol- Paraplegia, 30: 834–850.
ogy, 77: 1258–1267. Horowitz, B., Ward, S.M. and Sanders, K.M. (1999) Cellular
Devroede, G. and Lamarche, J. (1974) Functional importance and molecular basis for electrical rhythmicity in gastrointes-
of extrinsic parasympathetic innervation to the distal colon tinal muscles. Annu. Rev. Physiol., 61: 19–43.
and rectum in man. Gastroenterology, 66: 273–280. Hyman, L.R., Boner, G., Thomas, J.C. and Segar, W.E. (1972)
Ditunno, J.F., Little, J.W., Tessler, A. and Burns, A.S. (2004) Immobilization hypercalcemia. Am. J. Dis. Child, 124: 723–727.
Spinal shock revisited: a four-phase model. Spinal Cord, 42: Ingersoll, G.L. (1985) Abdominal pathology in spinal cord in-
383–395. jured persons. J. Neurosurg. Nurs., 17: 343–348.
Fajardo, N.R., Pasiliao, R.V., Modeste-Duncan, R., Creasey, Jiang, S., Wang, J., Khan, M.I., Middlemiss, P.J., Salgado-
G., Bauman, W.A. and Korsten, M.A. (2003) Decreased co- Ceballos, H., Werstiuk, E.S., Wickson, R. and Rathbone,
lonic motility in persons with chronic spinal cord injury. Am. M.P. (2003) Enteric glia promote regeneration of transected
J. Gastroenterol., 98: 128–134. dorsal root axons into spinal cord of adult rats. Exp. Neurol.,
Fealey, R.D., Szurszewski, J.H., Merritt, J.L. and DiMagno, 181: 79–83.
E.P. (1984) Effect of traumatic spinal cord transection on Johnson, R.L., Brooks, C.A. and Whiteneck, G.G. (1996) Cost
human upper gastrointestinal motility and gastric emptying. of traumatic spinal cord injury in a population-based regis-
Gastroenterology, 87: 69–75. try. Spinal Cord, 34: 470–480.
Fong, Y.C., Hsu, H.C., Sun, S.S., Kao, A., Lin, C.C. and Lee, Kao, C.H., Ho, Y.J., Changlai, S.P. and Ding, H.J. (1999)
C.C. (2003) Impaired gallbladder function in spinal cord in- Gastric emptying in spinal cord injury patients. Dig. Dis. Sci.,
jury on quantitative Tc-99m DISIDA cholescintigraphy. Ab- 44: 1512–1515.
dom. Imaging, 28: 87–91. Keshavarzian, A., Barnes, W.E., Bruninga, K., Nemchausky,
Frisbie, J.H. (1997) Improved bowel care with a polyethylene B., Mermall, H. and Bushnell, D. (1995) Delayed colonic
glycol based bisacadyl suppository. J. Spinal Cord Med., 20: transit in spinal cord-injured patients measured by indium-
227–229. 111 Amberlite scintigraphy. Am. J. Gastroenterol., 90:
Furness, J.B. and Costa, M. (1987) The Enteric Nervous Sys- 1295–1300.
tem. Churchill Livingstone, Edinburgh. Ketover, S.R., Ansel, H.J., Goldish, G., Roche, B. and Geb-
Geders, J.M., Gaing, A., Bauman, W.A. and Korsten, M.A. hard, R.L. (1996) Gallstones in chronic spinal cord injury: is
(1995) The effect of cisapride on segmental colonic transit impaired gallbladder emptying a risk factor? Arch. Phys.
time in patients with spinal cord injury. Am. J. Gas- Med. Rehabil., 77: 1136–1138.
troenterol., 90: 285–289. Kim, J., Shim, M.C., Choi, B.Y., Ahn, S.H., Jang, S.H. and
Glick, M.E., Meshkinpour, H., Haldeman, S., Hoehler, F., Shin, H.J. (2001) Clinical application of continent anal plug
Downey, N. and Bradley, W.E. (1984) Colonic dysfunction in in bedridden patients with intractable diarrhea. Dis. Colon
patients with thoracic spinal cord injury. Gastroenterology, Rectum, 44: 1162–1167.
86: 287–294. Krogh, K., Mosdal, C., Gregersen, H. and Laurberg, S. (2002)
Glickman, S. and Kamm, M.A. (1996) Bowel dysfunction in Rectal wall properties in patients with acute and chronic
spinal-cord-injury patients. Lancet, 347: 1651–1653. spinal cord lesions. Dis. Colon Rectum, 45: 641–649.
332

Krogh, K., Nielsen, J., Djurhuus, J.C., Mosdal, C., Sabroe, S. Moonka, R., Stiens, S.A., Eubank, W.B. and Stelzner, M.
and Laurberg, S. (1997) Colorectal function in patients with (1999) The presentation of gallstones and results of biliary
spinal cord lesions. Dis. Colon Rectum, 40: 1233–1239. surgery in a spinal cord injured population. Am. J. Surg.,
Longo, W.E., Ballantyne, G.H. and Modlin, I.M. (1989) The 178: 246–250.
colon, anorectum and spinal cord patient. A review of the Moonka, R., Stiens, S.A. and Stelzner, M. (2000) Atypical
functional alterations of the denervated hindgut. Dis. Colon gastrointestinal symptoms are not associated with gallstones
Rectum, 32: 261–267. in patients with spinal cord injury. Arch. Phys. Med. Re-
Longo, W.E., Woolsey, R.M., Vernava, A.M., Virgo, K.S., habil., 81: 1085–1089.
McKirgan, L. and Johnson, F.E. (1995) Cisapride for con- Nino-Murcia, M. and Friedland, G.W. (1992) Functional ab-
stipation in spinal cord injured patients: a preliminary report. normalities of the gastrointestinal tract in patients with spinal
J. Spinal Cord Med., 18: 240–244. cord injuries: evaluation with imaging procedures. Am. J.
Lu, C.L., Montgomery, P., Zou, X., Orr, W.C. and Chen, J.D. Roentgenol., 158: 279–281.
(1998) Gastric myoelectrical activity in patients with cervical Olsson, C. and Holmgren, S. (2001) The control of gut motility.
spinal cord injury. Am. J. Gastroenterol., 93: 2391–2396. Comp. Biochem. Physiol. A Mol. Integr. Physiol., 128: 481–503.
Lynch, A.C., Anthony, A., Dobbs, B.R. and Frizelle, F.A. Pfeifer, J., Agachan, F. and Wexner, S.D. (1996) Surgery for
(2000) Anorectal physiology following spinal cord injury. constipation: a review. Dis. Colon Rectum, 39: 444–460.
Spinal Cord, 38: 573–580. Pollock, L.J. and Finkelman, I. (1954) The digestive apparatus
Lynch, A.C., Antony, A., Dobbs, B.R. and Frizelle, F.A. (2001) in injuries to the spinal cord and cauda equina. Surg. Clin.
Bowel dysfunction following spinal cord injury. Spinal Cord, North Am., 98: 259–268.
39: 193–203. Prescrire, Int. (2000) Severe cardiac arrythmia on cisapride.
MacDonagh, R., Sun, W.M., Thomas, D.G., Smallwood, R. Prescrire Int., 9: 144–145.
and Read, N.W. (1992) Anorectal function in patients with Puet, T.A., Jackson, H. and Amy, S. (1997) Use of pulsed ir-
complete supraconal spinal cord lesions. Gut, 33: 1532–1538. rigation evacuation in the management of the neuropathic
MacDonagh, R.P., Sun, W.M., Smallwood, R., Forster, D. and bowel. Spinal Cord, 35: 694–699.
Read, N.W. (1990) Control of defecation in patients with Randell, N., Lynch, A.C., Anthony, A., Dobbs, B.R., Roake,
spinal injuries by stimulation of sacral anterior nerve roots. J.A. and Frizelle, F.A. (2001) Does a colostomy alter quality
BMJ, 300: 1494–1497. of life in patients with spinal cord injury? A controlled study.
Malone, P.S. (2004) The antegrade continence enema proce- Spinal Cord, 39: 279–282.
dure. BJU Int., 93: 248–249. Richards, J.S. (1992) Chronic pain and spinal cord injury: re-
Maynard, F.M. and Imai, K. (1977) Immobilization hype- view and comment. Clin. J. Pain, 8: 119–122.
rcalcemia in spinal cord injury. Arch. Phys. Med. Rehabil., Safadi, B.Y., Rosito, O., Nino-Murcia, M., Wolfe, V.A. and
58: 16–24. Perkash, I. (2003) Which stoma works better for colonic
McAndrew, H.F. and Malone, P.S. (2002) Continent catheter- dysmotility in the spinal cord injured patient? Am. J. Surg.,
izable conduits: which stoma, which conduit and which res- 186: 437–442.
ervoir? BJU Int., 89: 86–89. Saltzstein, R.J., Mustin, E. and Koch, T.R. (1995) Gut hor-
Menardo, G., Bausano, G., Corazziari, E., Fazio, A., mone release in patients after spinal cord injury. Am. J. Phys.
Marangi, A., Genta, V. and Marenco, G. (1987) Large-bowel Med. Rehabil., 74: 339–344.
transit in paraplegic patients. Dis. Colon Rectum, 30: 924–928. Sarna, S.K. (1991) Physiology and pathophysiology of colonic
Menter, R., Weitzenkamp, D., Cooper, D., Bingley, J., motor activity (2). Dig. Dis. Sci., 36: 998–1018.
Charlifue, S. and Whiteneck, G. (1997) Bowel management Sarna, S.K. (1993) Colonic motor activity. Surg. Clin. North
outcomes in individuals with long-term spinal cord injuries. Am., 73: 1201–1223.
Spinal Cord, 35: 608–612. Schemann, M. and Neunlist, M. (2004) The human enteric nerv-
Meshkinpour, H., Harmon, D., Thompson, R. and Yu, J. ous system. Neurogastroenterol. Motil., 16(Suppl. 1): 55–59.
(1985) Effects of thoracic spinal cord transection on colonic Schweiger, M. (1979) Method for determining individual con-
motor activity in rats. Paraplegia, 23: 272–276. tributions of voluntary and involuntary anal sphincters to
Meshkinpour, H., Nowroozi, F. and Glick, M.E. (1983) resting tone. Dis. Colon Rectum, 22: 415–416.
Colonic compliance in patients with spinal cord injury. Arch. Segal, J.L., Milne, N., Brunnemann, S.R. and Lyons, K.P.
Phys. Med. Rehabil., 64: 111–112. (1987) Metoclopramide-induced normalization of impaired
Middleton, J.W., Lim, K., Taylor, L., Soden, R. and Rutkowski, gastric emptying in spinal cord injury. Am. J. Gastroenterol.,
S. (2004) Patterns of morbidity and rehospitalisation follow- 82: 1143–1148.
ing spinal cord injury. Spinal Cord, 42: 359–367. Siddall, P.J. and Loeser, J.D. (2001) Pain following spinal cord
Miller, B.J., Geraghty, T.J., Wong, C.H., Hall, D.F. and injury. Spinal Cord, 39: 63–73.
Cohen, J.R. (2001) Outcome of the acute abdomen in patients Sinnatamby, C.S. (1999) Last’s Anatomy, Regional and Ap-
with previous spinal cord injury. ANZ J. Surg., 71: 407–411. plied (10th ed.). Churchill Livingstone, Edinburgh.
Miller, F. and Fenzl, T.C. (1981) Prolonged ileus with acute Snape Jr., W.J., Wright, S.H., Battle, W.M. and Cohen, S.
spinal cord injury responding to metaclopramide. Paraplegia, (1979) The gastrocolic response: evidence for a neural mech-
19: 43–45. anism. Gastroenterology, 77: 1235–1240.
333

Spiller, R.C. (2000) Haute cuisine and the colon. Gut, 46: Tanaka, M., Uchiyama, M. and Kitano, M. (1979) Gastrodu-
150–151. odenal disease in chronic spinal cord injuries. An endoscopic
Stiens, S.A., Bergman, S.B. and Goetz, L.L. (1997) Neurogenic study. Arch. Surg., 114: 185–187.
bowel dysfunction after spinal cord injury: clinical evaluation Tibbs, P.A., Bivins, B.A. and Young, A.B. (1979) The problem
and rehabilitative management. Arch. Phys. Med. Rehabil., of acute abdominal disease during spinal shock. Am. Surg.,
S86–S102. 45: 366–368.
Stiens, S.A., Luttrel, W. and Binard, J.E. (1998) Polyethylene Tola, V.B., Chamberlain, S., Kostyk, S.K. and Soybel, D.I.
glycol versus vegetable oil based bisacodyl suppositories to (2000) Symptomatic gallstones in patients with spinal cord
initiate side-lying bowel care: a clinical trial in persons with injury. J. Gastrointest. Surg., 4: 642–647.
spinal cord injury. Spinal Cord, 36: 777–781. van der Sijp, J.R., Kamm, M.A., Nightingale, J.M., Britton,
Stinneford, J.G., Keshavarzian, A., Nemchausky, B.A., Doria, K.E., Mather, S.J., Morris, G.P., Akkermans, L.M. and
M.I. and Durkin, M. (1993) Esophagitis and esophageal Lennard-Jones, J.E. (1993) Radioisotope determination of
motor abnormalities in patients with chronic spinal cord in- regional colonic transit in severe constipation: comparison
juries. Paraplegia, 31: 384–392. with radio opaque markers. Gut, 34: 402–408.
Stone, J.M., Nino-Murcia, M., Wolfe, V.A. and Perkash, I. Varma, J.S. (1992) Autonomic influences on colorectal motility
(1990a) Chronic gastrointestinal problems in spinal cord in- and pelvic surgery. World J. Surg., 16: 811–819.
jury patients: a prospective analysis. Am. J. Gastroenterol., Varma, J.S., Binnie, N., Smith, A.N., Creasey, G.H. and
85: 1114–1119. Edmond, P. (1986) Differential effects of sacral anterior root
Stone, J.M., Wolfe, V.A., Nino-Murcia, M. and Perkash, I. stimulation on anal sphincter and colorectal motility in spin-
(1990b) Colostomy as treatment for complications of spinal ally injured man. Br. J. Surg., 73: 478–482.
cord injury. Arch. Phys. Med. Rehabil., 71: 514–518. Wiesel, P.H., Norton, C. and Brazzelli, M. (2001) Management
Takaki, M. (2003) Gut pacemaker cells: the interstitial cells of of faecal incontinence and constipation in adults with central
Cajal (ICC). J. Smooth Muscle Res., 39: 137–161. neurological diseases. Cochrane Database. Syst. Rev., 4:
CD002115.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 22

Colorectal motility and defecation after spinal cord


injury in humans

A.C. Lynch and F.A. Frizelle

Colorectal Unit, Department of Surgery, Christchurch Hospital and Burwood Spinal Unit, Christchurch, New Zealand

Abstract: Following spinal cord injury, colorectal problems are a significant cause of morbidity, and
chronic gastrointestinal problems remain common with increasing time after injury. Although many cord-
injured patients achieve an adequate bowel frequency with drugs and manual stimulation, the risk and
occurrence of fecal incontinence, difficulties with evacuation, and need for assistance remain significant
problems. The underlying physiology of colorectal motility and defecation is reviewed, and consequences of
spinal cord injury on defecation are reported. A discussion of present management techniques is under-
taken and new directions in management and research are suggested. There is need for more intervention in
regard to bowel function that could improve quality of life, but there is also a need for more research in this
area.

Introduction with increasing time after injury (Stone et al.,


1990a). The inability to defecate normally means
Bowel dysfunction is a problem following spinal that bowel care often occupies a significant part of
cord injury the day, and, although many cord-injured people
achieve an adequate bowel frequency with drugs
Bowel dysfunction following spinal cord injury is and manual stimulation, the risk and occurrence
increasingly recognized as an area of major phys- of fecal incontinence, difficulties with evacuation,
ical and psychological difficulty. Surveys of spinal and need for assistance remain significant life-lim-
cord-injured people show that bowel function is as iting problems (Stone et al., 1990a, b; Levi et al.,
much of a problem as loss of mobility or sexual 1995; Glickman and Kamm, 1996; Han et al.,
function. The problem is twofold as, not only does 1998; Lynch et al., 2001).
spinal cord injury result in changes to bowel mo- Long-term gastrointestinal complications can
tility and sphincter control, but also the concur- develop in cord-injured people. Fecal impaction is
rent loss of mobility and gross motor dexterity common. Diverticular disease and volvulus are
makes bowel management a major life-limiting more frequent and are perhaps related to higher
problem. intracolonic pressures in those with upper motor
Immediately after spinal cord injury, colorectal neuron lesions. The occurrence of hemorrhoids
problems are a significant cause of morbidity, and and mucosal prolapse was also identified by Lynch
chronic gastrointestinal problems remain common et al. (2000c) as occurring more frequently after
spinal cord injury by an incidence of hemorrhoi-
Corresponding author. Tel.: +64-3-3640-640; dectomy of 9% compared with a control group
Fax: +64-3-3640-352; E-mail: frank.frizelle@chmeds.ac.nz incidence of 1.5% (po0.001). This may be

DOI: 10.1016/S0079-6123(05)52022-3 335


336

multifactorial due to altered anorectal tone or frequency similar to a control population. Chang-
trauma with manual evacuation, and is a frequent es to the extrinsic autonomic innervation of the
source of bleeding or autonomic dysreflexia. bowel are presumed to decrease the normal post-
prandial increase in motility and to decrease
colonic compliance. Laxative use among the
Impact on lifestyle cord-injured population is common as a means
of regulating bowel habit. Two-thirds of those
Spinal cord-injured people rate difficulties with with high injuries report using laxatives either
bowel management as similar to problems associ- occasionally or regularly, compared to the 4% in a
ated with loss of mobility and sexual function. control group drawn from the general population
Hanson and Franklin (1976) reported that 80% of (po0.0001, Fisher’s exact test, Lynch et al.,
male paraplegics and 46% of male tetraplegics 2000c). However, the diarrhea produced by laxa-
would rank bladder and bowel as their greatest tives and the resultant risk of incontinence may
functional loss after loss of mobility. It was inter- limit their use for some.
esting that when they asked the same question to Patterns of gut dysmotility have been described
spinal unit staff, only 39% ranked bladder and for different levels and degrees of spinal cord in-
bowel problems as high. jury with the level of the spinal cord lesion deter-
mining the effect on colonic motility. Marker
Toileting transit studies show that lesions above the first
thoracic segment (T1) result in delayed mouth-to-
Survey data shows that 61% of cord-injured peo- caecum time, but lesions below this level result in
ple would spend more than 15 min per day toile- normal transit times to the caecum. Beyond the
ting, compared with only 9% of controls (Lynch ileocaecal valve, transit times are markedly de-
et al., 2001). Those doing manual evacuations layed (Menardo et al., 1987). For people with an
spend the longest time. Half of all cord-injured upper motor neuron lesion, transit studies and
people need assistance with toileting. The need for scintigraphy have demonstrated variable changes
assistance with toileting relates very closely with in colonic transit. If the spinal cord lesion is above
level of injury and has implications for provision the lumbar region, transit is slowed throughout the
of carers and dependence on family members. It is whole colon. The velocity of the median position
recognised that having family members perform of bowel contents throughout the colon was sig-
such intimate tasks can be emotionally charged nificantly slower in cord-injured people (0.637
and negatively affect family interrelationships. 0.33 cm/h in cord-injured; 2.5871.2 cm/h in con-
This was significantly associated with the report- trols, po0.001). One study by Nino-Murcia et al.
ed perception that bowel function was a source of (1990), involving 28 cord-injured subjects, also
distress. demonstrated distal small bowel dilatation in 10
people, all of whom had abdominal symptoms and
9 of whom had a spinal cord lesion above T5. A
Colonic function following spinal cord injury lower motor neuron injury from a lesion affecting
the conus, cauda equina or pelvic nerves results in
Questionnaires exploring bowel function in spinal interruption of the parasympathetic supply to the
injured people have found that over half of those colon and reduced spinal cord-mediated reflex
with an injury above the second lumbar segment peristalsis. Stool propulsion is by segmental co-
(L2) suffer from constipation (DeLooze et al., lonic peristalsis only.
1998). People with higher injuries defecate less The mechanism for colonic dysmotility follow-
frequently compared to those with lower injuries ing a spinal cord injury may be a loss of descend-
and the general population. It is apparent that ing inhibitory modulation from the sympathetic
even with medications and other methods, spinal nervous system. This theory is supported by
cord-injured people do not achieve a bowel motion studies in the cat by Gillis et al. (1987) in which
337

a2-adrenergic receptor activation resulted in pro- to accelerate the transit of a charcoal meal in rats.
found inhibition of colonic motility, and section- It increases intraluminal pressure mainly by circu-
ing of splanchnic nerves (containing preganglionic lar muscle contraction by direct action on the
sympathetic innervation of the intestine) produced muscle as well as by simultaneous activation of
an increase in colonic contraction. excitatory cholinergic pathways and of inhibitory
In cord-injured people, colonic transit delays are vasoactive intestinal polypeptide-independent, ni-
more profound in higher injuries, where the sym- tric oxide-regulated pathways.
pathetic injury should be more pronounced. The Substance P is reduced in the colonic mucosa of
delay may in part be due to loss of colonic com- patients with chronic constipation, and mucosal
pliance. With a spinal cord lesion above L1, the substance P levels correlate significantly with dis-
left colon has an abnormal response to increasing ease state (Goldin et al., 1989). A similar scenario
volume. Distension with water produces a pres- exists with diabetic constipation where substance P
sure–volume curve (colometrogram) showing a in the rectal mucosa of diabetics with constipation
steep increase in intracolonic pressure with in- is significantly lower than in diabetics with normal
creasing volume. This is similar to the hype- bowel function (Lysy et al., 1993). The fact that
rreflexic response described by Meshkinpour mucosal substance P levels are associated with two
et al. (1983) during bladder cystometry for inju- disorders of colonic transit suggests a role in the
ries at a similar level. For injuries above T5, the pathogenesis of intestinal transit disorders.
right colon is also affected. The lack of compliance Whereas mucosal substance P may be decreased
leads to functional obstruction, increased transit with chronic constipation, concentrations in the
times, abdominal distension, bloating and discom- muscle layers may be increased. Sjolund et al.
fort. It suggests that the central nervous system (1997) examined tissue from the colon of 18 sub-
(CNS) is necessary to modulate colonic motility. jects with slow-transit constipation. Tissue con-
Colonic myoelectric activity has been recorded in a centrations of vasoactive intestinal polypeptide
group of spinal injury subjects with injuries at and substance P were measured by radioimmuno-
varying levels and controls. This demonstrated a assay. Significantly increased concentrations of
significantly higher level of basal colonic activity in both peptides were found in the ascending colon,
cord-injured subjects (12.6 vs. 3.3 spikes per and in the descending colon, substance P was in-
10 min), and no demonstrable gastrocolic reflex. creased in the myenteric plexus.
This would support the assumption that the CNS Recovery of bladder and bowel function follow-
exerts a tonic inhibitory influence on basal colonic ing traumatic spinal cord injury is dependent on
activity and is consistent with the hypertonicity reorganisation of reflex pathways in the periphery
seen on colometrograms. and CNS. Part of this reorganisation may be in-
fluenced by spinal cord–target organ interactions
mediated by neurotrophic factors released by the
Colonic neurotransmitters following spinal peripheral organs. Interrupting the descending
cord injury modulation from the CNS may lead to changes
in the autonomic and somatic outflow reaching
The intramural distribution of regulatory neuro- target organs from the spinal cord caudal to the
peptides within the bowel wall is distinct. Sub- lesion and alter target organ function. In rats, spi-
stance P is exclusively localized in nerves (Ferri nal cord injury leads to hypertrophy of the bladder
et al., 1983). Large numbers of vasoactive intestinal as well as electrophysiological and morphological
polypeptide- and substance P-containing enteric changes in bladder afferent neurons (de Groat
nerves supply the ganglionated plexuses and are et al., 1993; Yoshimura et al., 1993). In the colon,
especially numerous in the circular muscle layer. however, the intrinsic enteric nervous system ap-
They have a role in colon motility while those pears intact. Nerve fibres containing the intrinsic
supplying the mucosa are involved with electrolyte neurotransmitters substance P and vasoactive
and fluid transport. Substance P has been shown intestinal polypeptide appear to be present in
338

approximately similar amounts in specimens from also continues to show tonic activity, but again
cord-injured and control subjects (Lynch et al., generates a lower than normal pressure. Mano-
2000b). The colon may therefore continue to metric studies on cord-injured subjects show a
function independently of CNS modulation after maximal mean basal sphincter pressure (which
spinal cord injury. probably reflects external anal sphincter pressure)
significantly lower than control group pressures.
Spinal cord-injured subjects can produce a small
Anorectal function rise in sphincter pressure with voluntary squeeze
(p40.05). This is, however, much less than the in-
Continence crease in pressure generated by control subjects
performing a similar maneuver who can generate a
The incidence of fecal incontinence in people with four-fold increase in external anal sphincter pres-
spinal cord injury is more common than in the sure. People with incomplete injuries can produce
general population. When compared to matched a greater increase in sphincter pressure with a
controls by using standardized scoring systems, Valsalva maneuver than those with complete inju-
the mean fecal incontinence score was higher for ries. The Valsalva maneuver is expiration against a
cord-injured people than controls (po0.0001), and closed glottis. This may reflect the greater increase
for complete spinal cord injury compared with in- in intra-abdominal pressure, as measured by rectal
complete injury ðp ¼ 0:0023Þ: Having fecal incon- pressure, that people with incomplete or low inju-
tinence also impacts on the quality of life of those ries are able to generate due to incomplete paral-
with a spinal cord injury more frequently than of ysis of their abdominal musculature. For those
neurologically intact persons [62% of cord-injured with a complete injury, attempts at squeezing re-
people report that fecal incontinence impacts upon sult in a straining response rather than a true
their everyday life, compared to 8% of control squeeze. People with lesions above T5 will be un-
subjects, po0.0001 (Lynch et al., 2000c)]. able to use abdominal muscles and rely on inter-
Fecal continence requires the ability to maintain costal and diaphragmatic muscle contraction to
internal anal sphincter resting tone and to contract increase intra-abdominal pressure. Those with cer-
the external anal sphincter in response to increased vical injuries can only use the diaphragm. These
intra-abdominal pressure, rectal distension and observations fit the concept that external anal
rectal contraction. These are spinal reflexes that sphincter contraction is mediated by a spinal re-
are intact following spinal cord injury, but no flex, triggered by tension receptors in the pelvic
longer modulated by cortical input. Basal sphinc- floor that respond to an increase in intra-abdom-
ter tone is mainly an activity of the internal anal inal pressure. This is supported by another study
sphincter, the maintenance of which seems to be by MacDonagh et al. (1992) that found the rise in
due to a tonic excitatory sympathetic discharge. sphincter pressure with the Valsalva maneuver to
Frenckner and Ihre (1976) observed that anorectal be directly proportional to the rise in intra-
manometry performed on cord injury subjects abdominal pressure.
shows a persistent anal tone that is reduced com-
pared to control subjects. They described changes
in anal tone in eight healthy subjects following Rectal sensation
spinal anesthesia. High spinal anesthesia resulted
in a significantly lower resting anal pressure than All normal subjects experiencing rectal distension
either low spinal or pudendal block. Of note, peo- as part of anorectal manometry studies report a
ple with lumbosacral injuries, who have external range of sensation starting at a rectal volume of
anal sphincter paralysis but persistent internal anal about 10 ml, and ranging from sensations of ‘wind’
sphincter activity, still appear to maintain a degree to pain. This is compared to 78% of cord-injured
of anorectal tone, higher than rectal pressure, but subjects with complete injuries, and 43% of those
lower than normal. The external anal sphincter with incomplete injuries, who report no sensation
339

on rectal distension (Lynch et al., 2000a). Those injury. It becomes even more significant as a qual-
that did report sensation described non-specific ity of life issue when reduced mobility and poor
abdominal sensation that did not prevent further hand dexterity are compounded by the difficulties
rectal distension. A previous study by MacDonagh associated with finding a wheelchair-able toilet.
et al. (1992), examining similar sensations, pro- Fecal urgency can be assessed by asking respond-
posed that sympathetic nerves entering the tho- ents how long defecation can be delayed. Overall
racic spinal cord above the level of the injury 81% of controls can delay defecation, compared
conveyed this dull pelvic sensation. However, such with only 41% of cord-injured people. There is
sensations have also been identified in five people also an approximately ten-fold increase in the
with complete cervical injuries, making the origin proportion of cord-injured people, compared to
of this sensation unclear. controls, who have to defecate immediately. Of
note, many people with complete injuries have no
sensation, and thus never sense the need to defe-
Rectal compliance
cate. The incidence of fecal incontinence is often
higher for cord-injured people who are unable to
A normally compliant rectum accommodates an
delay defecation.
increase in volume with little change in pressure.
As rectal volume increases, a normal sphincter re-
sponse is the relaxation of the internal anal sphinc- Defecation
ter with continence being maintained by continued
external anal sphincter contraction. The ability of Many defecatory problems seen after spinal cord
the rectum to distend to store bowel volume is an injury are the result of altered anorectal function.
important component of normal bowel function, Defecation requires the complex integration of re-
as it means defecation can be delayed until an ap- flex and voluntary muscular control. Cortical in-
propriate time. hibition of the external anal sphincter occurs in a
People with complete cervical injuries can have coordinated fashion on straining as the rectal
increased rectal tone with low compliance, i.e., a smooth muscle contracts. This modulation of the
sharp rise in rectal pressure occurs with rectal dis- intrinsic nervous system by the extrinsic system is
tension, as the rectum does not expand to accom- disrupted following a complete supra-conal spinal
modate the increase in volume. Sphincter tone can cord injury. This means that straining by increas-
also increase with rectal distension, because inter- ing intra-abdominal pressure does not improve
nal anal sphincter contraction is an enteric reflex, evacuation by the usual external anal sphincter
normally suppressed by descending inhibitory relaxation in response to the rectal and intra-ab-
pathways. The loss of inhibitive sympathetic tone dominal pressure increase. Rather, external anal
has also been proposed as a mechanism for the sphincter tone increases. Thus, coordinated reflex
absent rectal relaxation and linear pressure/vol- defecation is difficult for cord-injured people. It is
ume relationship during rectal distension (Mac- often inefficient and incomplete, resulting in in-
Donagh et al., 1992). Most people with low continence and/or constipation. Therefore, defe-
lumbosacral injuries have an areflexic rectum with cation is planned on regular basis to avoid
an attenuated sphincter response to rectal disten- constipation or an increased chance of fecal in-
sion (Shafik, 1995). The rectum is flaccid and ca- continence.
pacious producing no rise in rectal pressure with
increasing volume.
High spinal cord injuries

Urgency The difficulties with defecation following high


spinal cord injury result from discoordinate anal
Fecal urgency, or an inability to delay defecation, sphincter function. The normal synergistic activity
is more often a problem following spinal cord of colonic smooth muscle and pelvic striated muscle
340

is lost. There is a loss of conscious sphincter con- to changes in intra-abdominal pressure. Thus, a
trol and, due to abdominal muscle paralysis, an Valsalva maneuver can result in fecal leakage, and
inability to significantly increase intra-abdominal the rectum has to be kept empty to avoid fecal
pressure. Loss of rectal sensation and a spastic incontinence. Reflex-mediated defecation does not
external anal sphincter require defecation to be occur with spinal cord lesions below the conus, so
anticipated. stool has to be removed digitally, assisted by a
The conus-mediated increase in external anal Valsalva maneuver and abdominal massage.
sphincter tone with increasing intra-abdominal
pressure acts against straining to defecate. How-
Change in bowel function with time from injury
ever, reflex relaxation of the internal and external
anal sphincters by mucosal stimulation, either dig-
Studies do not demonstrate a change in fecal in-
itally or with a suppository, can be exploited in
continence with either duration of injury or in-
order to defecate. Insertion of a gloved finger into
creasing age for cord-injured people. One survey
the rectum with gentle sustained pressure towards
of bowel dysfunction in cord-injured people by
the sacrum relaxes the spastic external anal sphinc-
Stone et al. (1990a) found that chronic gastroin-
ter and pelvic muscles. Rapid or excessive stretch-
testinal problems were rare in the first 5 years fol-
ing can precipitate sphincter spasm. Rotation of
lowing injury, but problems with defecation
the finger continues the stimulation until a reflex
became more common with increasing time. The
peristaltic wave is generated in the rectum, flatus is
incidence of abdominal pain and distension was
passed and stool comes down. The recto-anal in-
increased in long-term cord-injured people (more
hibitory reflex is initiated, the internal anal sphinc-
than 18 years since injury) whose bowel regimen
ter relaxes, and the recto-colic reflex stimulates
was less frequent than once a day (Stone et al.,
pelvic nerve-mediated peristalsis. If there is no re-
1990a). Another study of chronic gastrointestinal
flex relaxation of the external anal sphincter com-
problems in cord-injured subjects by Han et al.
plex, evacuation will not occur or be incomplete.
(1998) concluded that bowel habit appeared to
Manual evacuation or enemata are often required
settle by about 6 months after injury, and that
in this situation.
subsequent bowel dysfunction was not related to
This reflex relaxation can also lead to fecal in-
age, duration or level of injury.
continence by two means. The anal sphincter may
relax at relatively low rectal volume in response to
a small increase in intra-abdominal pressure, or as Current management strategies
people with high-level injuries often have no sen-
sation of rectal fullness, reflex defecation in re- The approach to bowel management after spinal
sponse to a full rectum can result in fecal cord injury should address specific issues such as
incontinence that is unpredictable and episodic. fecal incontinence, constipation and functional mo-
bility. This must be within the context of the patient
as a whole person and consider his/her cultural,
Low spinal cord injuries social, sexual and vocational roles. A bowel care
regimen needs to be generated that fits the person’s
Complete or partial injuries to the cauda equina long-term routine. The aim should be effective
result in a lower motor neuron pattern of injury. A colonic evacuation without fecal incontinence or
person with a lower motor neuron lesion following other complications. Regularity of evacuation pre-
spinal cord injury will have absent external anal vents excessive build-up of feces and impaction.
sphincter tone, flaccid pelvic muscles and decreased Appropriate equipment, such as commode chairs
reflex peristalsis. Rectal compliance is increased in and wheelchair-able toilets, needs to be supplied for
response to rectal distension. The loss of parasym- an adequate long-term bowel program.
pathetic control of the internal anal sphincter means Dietary manipulation is important. Adequate
that resting anal tone is low and unresponsive water intake promotes transit by keeping the stool
341

soft. Fibre is promoted to give the stool bulk and Spinal cord-injured people with upper motor
plasticity. This is thought to assist colonic transit neuron lesions can exploit the recto-colic reflex to
in neurologically intact patients, probably by pro- effect defecation. Digital stimulation can result in
moting propulsive activity secondary to increased a reflex wave of conus-mediated rectal peristalsis.
colonic wall distension. The effects of increased The recto-anal inhibitory reflex is intact, so this
fibre on colonic function after spinal cord injury causes internal anal sphincter relaxation and def-
are not yet fully understood, and may be counter- ecation. Rectal sensation is reduced, however, so
productive. A study carried out by Cameron et al. defecation has to be anticipated on a regular basis.
(1996) at the Spinal Injury Unit, Austin Hospital, These people require a bowel management pro-
Heidelberg, Australia demonstrated that signifi- gram that keeps the rectum empty to reduce the
cantly increasing dietary fibre in a group of cord- incidence of incontinence. If people with upper
injured subjects with a range of injuries resulted in motor neuron lesions are unable to defecate using
an increase in mean colonic transit time from 28.2 the recto-colic reflex, then a management plan
to 42.2 h (po0.05), and in mean recto-sigmoid needs to minimize anorectal trauma but still allow
transit time from 7.9 to 23.3 h (po0.02). However, adequate rectal evacuation to avoid constipation.
an increase in stool bulk may mean more time Cord-injured people with lower motor neuron
spent with bowel care. lesions have a rectum that is areflexic, reduced
Stool softeners other than fibre, such as docu- sphincter tone and an attenuated sphincter re-
sate sodium increase the amount of water in stool sponse to rectal distension or a Valsalva mane-
without increasing volume and have no effect on uver. These dysfunctions lead to an increased risk
bowel motility. They can also affect the intestinal of incontinence, especially with liquid stool. The
absorption of other drugs, resulting in higher plas- aim, therefore, is to keep stool consistency firm.
ma levels. The stool is more likely to be liquid, so Local anorectal reflexes are often insufficient to
continence will not be improved. They are most result in defecation, and a compliant rectum acts
useful when fecal incontinence is not a risk and as a large reservoir, so stool is digitally removed.
straining is to be avoided, such as for patients with Arnold et al. (1986) noted that a Brindley sacral
hemorrhoids or autonomic dysreflexia. anterior nerve root stimulator (S2–S4) can be used
Stimulant laxatives act by increasing intestinal for electromicturition to achieve regular, complete
motility, resulting in less time for water reabsorp- bladder emptying. Often deafferentiation of the
tion. Senna has a direct stimulant effect on the sacral posterior nerve roots is performed before
myenteric plexus and also increases intraluminal the stimulator is implanted to produce detrusor
fluid. Bisacodyl has a similar mode of action and is areflexia and interim urinary continence. The deaf-
often used as a suppository to initiate bowel evac- ferentiation also results in loss of the sacral reflexes
uation. Dose-dependent side effects can occur. necessary for defecation. The stimulator can then
These include abdominal cramping, diarrhea and be used to initiate defecation. This does not occur
electrolyte imbalance. Chronic use of stimulant during stimulation due to the simultaneous rectal
laxatives, especially senna, can result in a progres- and sphincter contraction, but when stimulation
sive unresponsiveness. Osmotic laxatives such as stops the external anal sphincter relaxes instanta-
lactulose draw fluid into the colon. They can result neously and the rectum relaxes slowly, resulting in
in more liquid stool and cause cramping. Proki- spontaneous defecation. This method has been
netic agents such as cisapride have been employed shown to result in quicker, more controllable def-
to reduce constipation in cord-injured people. ecation than the reflex method.
Transit times are improved, but cardiac arrhyth-
mias have been noted with long-term use. Enemata
are often employed when suppositories or digital Does a colostomy improve bowel function?
stimulation fail. Long-term use can result in enema
dependences and side effects such as rectal trauma Colostomy has been reported to result in improved
and autonomic dysreflexia can occur. quality of life for people after spinal cord injury
342

(Frisbie et al., 1986; Saltzstein and Romano, 1990; general bowel function and identify problems such
Stone et al., 1990b; Randell et al., 2001). It can not as constipation, fecal impaction, anal fissures and
only reduce fecal incontinence, but also simplify hemorrhoids. Simple tests of anorectal function
bowel care in those for whom bowel evacuation is are available that can be performed on all cord-
difficult, reducing the amount of time spent on injured people in the same manner that bladder
bowel care from 99 to 18 min per day (Stone et al., dysfunction is investigated. Anorectal manometry
1990b). The only long-term management problem will identify those with dyssynergic sphincter func-
reported has been occasional appliance leakage tion. For those with abdominal bloating and con-
and mucoid discharge per rectum. All subjects in- stipation, abdominal X-ray and colonic motility
volved in a study by Craven and Etchells (1998) studies can be helpful.
found the stoma had impacted significantly on Although improving colonic motility and appro-
their lifestyle with increased feelings of independ- priate bowel management may help, some cord-
ence, freedom and raised self-esteem. injured people will have ongoing bowel problems.
Sacral pressure ulcers are a common reason for Colostomy formation has been used to provide the
stoma formation in people who had no bowel patient with relief from constipation and anorectal
function problems previously. Deshmukh et al. dysfunction and with an independent means of
(1996) examined the use of colostomy as an adjunct managing their own bowel function. Further re-
measure in the healing of pressure sores in cord- search needs to be done to examine the differences
injured people. Their findings similarly showed that in quality of life and bowel function following co-
a colostomy improved bowel management and was lostomy formation in cord-injured people.
well accepted. However, they noted that the pri-
mary goal of pressure sore healing was accom-
plished in only 6 out of 27 cord-injured people, and References
stoma formation resulted in significant morbidity
with stomal prolapse and difficulties with wound Arnold, E.P., Gowland, S.P., MacFarlane, M.R., Bean, A.R.
and Utley, W.L.F. (1986) Sacral anterior root stimulation of
healing. Of the 19 subjects reviewed in this study, the bladder in paraplegics. Aust. N.Z. J. Surg., 56: 319–324.
none wanted their stoma reversed, and all felt that Cameron, K.J., Nyulasi, I.B., Collier, G.R. and Brown, D.J.
their quality of life was improved. (1996) Assessment of the effect of increased dietary fibre in-
A colostomy can improve bowel management take on bowel function in patients with spinal cord injury.
for some people after cord injury. Dependence on Spinal Cord, 34(5): 277–283.
Craven, M.L. and Etchells, J. (1998) A review of the outcome of
assistance with toileting is decreased as even tet- stoma surgery on spinal cord injured patients. J. Adv. Nurs.,
raplegics with limited dexterity are able to manage 27: 922–926.
suitable stoma appliances. Enthusiasm for bowel de Groat, W.C. (1993) Anatomy and physiology of the lower
diversion procedures should be tempered with an urinary tract. Urol. Clin. North Am., 20(3): 383–401.
DeLooze, D., Van Laere, M., De Muynck, M., Beke, R. and
acknowledgment that the risks associated with any
Elewault, A. (1998) Constipation and other chronic gastro-
surgical procedure may not make it acceptable to intestinal problems in spinal cord injury patients. Spinal
all cord-injured people. Cord, 36: 63–66.
Deshmukh, G.R., Barkel, D.C., Sevo, D. and Hergenroeder, P.
(1996) Use or misuse of colostomy to heal pressure ulcers.
Future objectives for the investigation and Dis. Colon Rectum, 39: 737–738.
Ferri, G.-L., Adrian, T.E., Ghatei, M.A., O’Shaughnessy, D.J.,
management of bowel dysfunction Probert, L., Lee, Y.C., Buchan, A.M., Polak, J.M. and
Bloom, S.R. (1983) Tissue localisation and relative distribu-
Bowel dysfunction has a major impact on the tion of regulatory peptides in separated layers from the hu-
quality of life for many people with spinal cord man bowel. Gastroenterology, 84: 777–786.
Frenckner, B. and Ihre, T. (1976) Influence of autonomic nerves
injury. This difficulty has been shown to improve
on the internal anal sphincter in man. Gut, 17: 306–312.
with appropriate early identification and manage- Frisbie, J.H., Tun, C.G. and Nguyen, C.H. (1986) Effect of
ment of their problems. Interview and clinical ex- enterostomy on quality of life in spinal cord injury patients.
amination can generate an impression of their J. Am. Paraplegia Soc., 9(1–2): 3–5.
343

Gillis, R.A., Dias Souza, J., Hicks, K.A., Mangel, A.W., MacDonagh, R., Sun, W.M., Thomas, D.G., Smallwood, R.
Pagani, F.D., Hamilton, B.L., Garvey III, T.Q., Pace, and Read, N.W. (1992) Anorectal function in patients
D.G., Browne, R.K. and Norman, W.P. (1987) Inhibitory with complete supraconal spinal cord lesions. Gut, 33:
control of proximal colonic motility by the sympathetic 1532–1538.
nervous system. Am. J. Physiol., 253(4, Pt 1): G531–G539. Menardo, G., Bausano, G., Corazziari, E., Fazio, A., Marangi,
Glickman, S. and Kamm, M.A. (1996) Bowel dysfunction in A., Genta, V. and Marenco, G.L. (1987) Large bowel transit
spinal-cord-injury patients. Lancet, 347: 1651–1653. in paraplegic patients. Dis. Colon Rectum, 30(12): 924–928.
Goldin, E., Karmeli, F., Selinger, Z. and Rachmilewitz, D. Meshkinpour, H., Nowroozi, F. and Glick, M.E. (1983) Co-
(1989) Colonic substance P levels are increased in ulcerative lonic compliance in patients with spinal cord injury. Arch.
colitis and decreased in chronic severe constipation. Dig. Dis. Phys. Med. Rehabil., 64: 111–112.
Sci., 34(5): 754–757. Nino-Murcia, M., Stone, J.M., Chang, P.J. and Perkash, I.
Han, T.R., Kim, J.H. and Kwon, B.S. (1998) Chronic gastro- (1990) Colonic transit in spinal cord-injured patients. Invest.
intestinal problems and bowel dysfunction in patients with Radiol., 25(2): 109–112.
spinal cord injury. Spinal Cord, 36: 485–490. Randell, N., Lynch, A., Anthony, A., Dobbs, B., Roake, J. and
Hanson, R.W. and Franklin, M.R. (1976) Sexual loss in rela- Frizelle, F. (2001) Does a colostomy alter quality of life in
tion to other functional losses for spinal cord injured males. patients with spinal cord injury? A controlled study. Spinal
Arch. Phys. Med. Rehabil., 57: 291–293. Cord, 39(5): 279–282.
Levi, R., Hultling, C., Nash, M.S. and Seiger, Å. (1995) The Saltzstein, R.J. and Romano, J. (1990) The efficacy of colos-
Stockholm spinal cord injury study: 1. medical problems in a tomy as a bowel management alternative is selected spinal
regional SCI population. Paraplegia, 33: 308–315. cord injury patients. J. Am. Paraplegia Soc., 13(2): 9–13.
Lynch, A., Anthony, A., Dobbs, B. and Frizelle, F. (2000a) Shafik, A. (1995) Electrorectogram study of the neuropathic
Anorectal physiology following spinal cord injury. Spinal rectum. Paraplegia, 33: 346–349.
Cord, 38: 73–80. Sjolund, K., Fasth, S., Ekman, R., Hulten, L., Jiborn, H.,
Lynch, A., Anthony, A., Dobbs, B. and Frizelle, F. (2000b) Nordgren, S. and Sundler, F. (1997) Neuropeptides in
Colonic neurotransmitters following spinal cord injury. Tech. idiopathic chronic constipation (slow transit constipation).
Coloproct., 4: 93–97. Neurogastroenterol. Motil., 9(3): 143–150.
Lynch, A., Anthony, A., Dobbs, B. and Frizelle, F. (2001) Stone, J.M., Nino-Murcia, M., Wolfe, V.A. and Perkash, I.
Bowel dysfunction following spinal cord injury; a review. (1990a) Chronic gastrointestinal problems in spinal cord in-
Spinal Cord, 39: 193–203. jury patients: a prospective analysis. Am. J. Gastroenterol.,
Lynch, A., Wong, C., Anthony, A., Dobbs, B. and Frizelle, F. 85(9): 1114–1119.
(2000c) Bowel dysfunction following spinal cord injury: Stone, J.M., Wolfe, V.A., Nino-Murcia, M. and Perkash, I.
a description of bowel function in a spinal cord-injured pop- (1990b) Colostomy as treatment for complications of spinal
ulation and comparison with age and gender matched con- cord injury. Arch. Phys. Med. Rehabil., 71: 514–518.
trols. Spinal Cord, 38: 717–723. Yoshimura, N. and de Groat, W.C. (1993) Changes in elect-
Lysy, J., Karmeli, F. and Goldin, E. (1993) Substance P levels in rophysiological and pharmacological properties of rat blad-
the rectal mucosa of diabetic patients with normal bowel func- der afferent neurons following spinal cord injury. J. Urol.,
tion and constipation. Scand. J. Gastroenterology, 28: 49–52. 149: 340A.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 23

Mechanisms controlling normal defecation and the


potential effects of spinal cord injury

A.F. Brading1, and T. Ramalingam2

1
Oxford Continence Group, University Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK
2
Specialist Registrar in Colorectal Surgery, Headquarters Army Medical Directorate, Former Army Staff College,
Camberley, Surrey GU16 4NP, UK

Abstract: Spinal cord injury frequently leads to bowel dysfunction with the result that emptying the bowel
can occupy a significant part of the day and reduce the quality of life. This chapter contains an overview of
the function and morphology of the normal distal gut in the human, and of gut behaviour in normal
defecation. In humans, this can be monitored and is described, but knowledge of the mechanisms con-
trolling it is limited. Work on animals has shown that the intrinsic activity of the smooth muscles and their
interactions with the enteric nervous system can program the activity that is necessary to expel waste
material, but the external anal sphincter is controlled through somatic nerves. The gut however also
receives input from the central nervous system through autonomic nerves, and a spinal reflex centre exists.
Voluntary effort to induce defecation can influence all the control mechanisms, but the precise importance
of each is not understood.
The behaviour and properties of the individual muscles in the normal human rectum and anal canal are
described, including their responses to intrinsic nerve stimulation and adrenergic and cholinergic agonists.
The effects of established spinal cord injury are then considered. For convenience, supraconal and conal/
cauda equina lesions are considered as two categories. Prolongation of transit times and disordered def-
ecation are common problems. Supraconal lesions result in reduced resting anal pressures and increased
risk of fecal incontinence. The acute effects of spinal cord injury are described, with injury causing ileus
(prolonged total gastrointestinal transit times), constipation (prolonged colonic transit times) and fecal
incontinence (passive leakage).

Introduction cause of morbidity and remain a problem long


after the time of injury. Constipation and dis-
Problems related to the control of defecation are a turbed defecation means that this routine bodily
source of physical, psychological and social dis- function can occupy a significant part of the day in
tress to people with spinal cord injuries (SCI). those with spinal cord injuries. Successful bowel
Approximately 30% of these people consider bow- management often requires considerable control of
el dysfunction to be a greater concern than either dietary intake, strict adherence to a regime, man-
bladder or sexual dysfunction (Krogh et al., 1997). ual stimulation and the use of a concoction of
Chronic gastrointestinal problems are a significant drugs to aid the process. Those with higher lesions
and poor arm and hand control may have to rely
Corresponding author. Tel.: +44 1865 271875; on others for assistance with the accompanying
loss of dignity, making bowel management a ma-
Fax: +44 1865 281120;
E-mail: alison.brading@pharm.ox.ac.uk jor life-limiting problem. Some also experience

DOI: 10.1016/S0079-6123(05)52023-5 345


346

symptoms of fecal incontinence, with its inevitable striated muscle, the external anal sphincter. The
consequences on their quality of life (Glickman internal anal sphincter is considered to be the con-
and Kamm, 1996). tinuation of the circular muscle while the longitu-
Our current understanding of the control of dinal muscle layer of the rectum splits to envelope
normal defecation is still somewhat sketchy, and the internal sphincter ultimately inserting into the
thus the precise mechanisms through which spinal subcutaneous tissue (see Fig. 1).
cord injury affects the process are also not entirely
clear. In this chapter, the mechanisms controlling
normal defecation will be discussed. We will in- Overview of defecation
clude colonic morphology in relation to motility
and transit, and the behaviour and role of the In the gut, waste material, usually either solid or
anorectum during evacuation, since effective def- semi-solid feces or flatus (gas), can be stored in the
ecation requires both these components to func- rectum and distal colon. The anal sphincter pro-
tion properly. We will go on to describe the vides continence and is equivalent to the external
current appreciation of the potential effects of SCI urethral sphincter. Functional differences between
in each of these component areas. the distal gut and the urinary tract are due to the
fact that fecal material can be returned to the co-
lon from the rectum, and that a detection mech-
Normal gut anism is present in the anal canal which allows
assessment of the rectal content, so that flatus can
Outline of colorectal function and morphology be voided at times when voiding feces would not
be appropriate.
The large intestine extends from the ileocaecal The sequence of events that occurs in storage
valve of the terminal ileum to the anus. It com- and defecation is as follows. The rectum remains
prises of the caecum, ascending colon, transverse empty for most of the time, but will fill as fecal
colon, descending colon, sigmoid colon, rectum matter accumulates in the descending and sigmoid
and the anus (Bannister, 1995). Its main functions colon, and is pushed forward through the occa-
are the absorption of water and salts, the degra- sional peristaltic waves (mass movements) that
dation of short-chain fatty acids and the transport occur. As fecal matter enters the rectum, the walls
and storage of luminal contents to the anorectum relax, and filling can occur with little increase in
until it is deemed convenient for defecation. rectal pressure. Distension of the rectum triggers a
The colon contains two layers of smooth mus- recto-anal inhibitory reflex that lowers the pressure
cle, an inner, circular layer and an outer longitu- in the anal canal, allowing the rectal contents to
dinal layer. The anatomy of the longitudinal layer enter and contact the mucosa. Periodic relaxations
is not consistent throughout the large intestine.
From the caecum to the sigmoid colon, it is con-
centrated in three bands that are symmetrically
placed: the taenia coli.
Bundles of smooth muscle cells within these lay-
ers are connected by gap junctions at intermittent
points, enabling them to function as a syncitium.
The anorectum forms the distal end of the
colon. At the rectum, the taenia coli coalesce to
form the longitudinal smooth muscle layer. The
anal sphincter consists of an inner ring of smooth
muscle, the internal anal sphincter, surrounded by a
thin outer longitudinal smooth muscle, the conjoint
longitudinal coat, and a thicker outer ring of Fig. 1. Diagram of the human rectum and anal canal.
347

of the internal anal sphincter allow anorectal sam-


pling of the contents (Miller et al., 1988a, b) using
the rich sensory innervation of the anal canal
which allows us to discriminate between gas, liquid
and solid. During these phases, the external stri-
ated sphincter is contracted to maintain conti-
nence. As filling continues, sensory information
ascending to the brain leads to the sensation of
rectal fullness. If defecation is deemed appropriate,
voluntary relaxation of the external sphincter oc-
curs and peristalsis in the colon and rectum is in-
itiated, usually by abdominal straining resulting in
relaxation of the internal sphincter and expulsion
of the rectal contents. If defecation is not appro-
priate, the rectal contents may return to the colon.

Overview of neural control

Unlike the situation in the urinary tract, however,


the gut wall contains all of the machinery (intrinsic
pacemakers and neural networks) to programme
the activity of the smooth muscle that is necessary
to expel the waste material (relaxation of the in-
ternal anal sphincter, initiation and co-ordination
of peristalsis). The intrinsic or enteric nervous sys-
tem consists of two major plexuses of intercon-
necting ganglia, namely the submucous
Fig. 2. Innervation of the distal gut. CP, celiac plexus; HGN,
(Meissner’s) plexus and the myenteric (Auer-
hypogastric nerve; PEL, pelvic nerve; PP, pelvic plexus; PUD,
bach’s) plexus (Furness and Costa, 1987) whose pudendal nerve; S2–4, sacral roots; SC, sympathetic chain; SHP,
complex circuitry is the focus of considerable ex- superior hypogastric plexus; SN, sacral nerve.
perimental interest, and which can choreograph
most of the activity of the gut. It is only the ex- filling, and to help switch on the peristaltic activity
ternal striated sphincter that absolutely requires necessary to produce defecation, as well as to in-
extrinsic innervation to contract, through activa- itiate the correct pattern of activity in the somatic
tion of the somatic motor neurons whose axons nerves to the extrinsic sphincter. The voluntary
run in the pudendal nerves. The distal gut does, input to defecation is through control of the stri-
however, receive extrinsic innervation through the ated muscles of the anal sphincter and the abdom-
autonomic nervous system, with parasympathetic inal muscles.
input to the proximal colon through the vagus,
and to the whole colon and anorectum via the
sacral roots (S2–S4) through the pelvic nerves, and Underlying smooth muscle properties
with sympathetic input from the lumbar cord run-
ning with the superior and inferior mesenteric Basic rhythmic activity
blood vessels and passing through the mesenteric Smooth muscles of the non-sphincteric elements of
and pelvic plexuses (Figs. 2 and 3). A spinal reflex the gut show basic rhythmic changes in electrical
centre exists which uses this autonomic extrinsic activity, called slow waves. These slow waves,
pathway to maintain rectal compliance during if large enough, trigger action potentials through
348

Fig. 3. Diagram of the autonomic innervation of the gut wall. CMS, circular smooth muscle; LSM, longitudinal smooth muscle; Epi,
epithelium. Green: preganglionic parasympathetic nerves. Blue: pre- and post-ganglionic sympathetic nerves, brown: sensory nerves,
orange and yellow: interneurones. Note that this diagram does not include interstitial cells of Cajal.

activation of L-type Ca channels, and resulting using immunohistochemical labelling with anti-
smooth muscle contraction (Tomita, 1981). bodies to the Kit receptor. Interstitial cells are ar-
Rhythmic contractions of the longitudinal mus- ranged in distinct ways in different parts of the
cles may generate pendular activity in the gut wall, gut. In the longitudinal elements there is an ex-
and similar contractions in the circular muscle tensive plexus running with the neurons in the
generate segmentation. These types of activity may myenteric plexus, and often another plexus at the
occur in the absence of neural input. The rhythms submucosal surface of the circular muscle. In
and sizes of the underlying slow waves can be these, the interstitial cells are linked to each other,
modulated by neuronal input and by local gut and to the adjacent smooth muscle cells through
hormones. gap-junctions. Interstitial cells also run along the
outside of smooth muscle bundles within the mus-
cle layers. The interstitial cells associated with the
Pacemaking myenteric plexus and submucosal plexus are
This spontaneous, or more accurately, non- thought to be pacemakers in the human colon
neurogenic contractile activity in gastrointestinal and rectum. The interstitial cells along the muscle
smooth muscles is now known not to be generated bundles may be involved in helping conduct the
by the smooth muscles themselves, but by activity within the muscle layers. Isolated intersti-
another cell type, the interstitial cells of Cajal. tial cells undergo rhythmic large depolarizations at
These cells are named after Santiago Ramon frequencies similar to the phasic contractions seen
y Cajal, a Spanish histologist who described them in intact smooth muscles, and when held under
at the end of the 19th century (Cajal, 1893). Recent voltage clamp conditions, will generate depolariz-
studies of these cells have been facilitated by the ing currents with the same rhythm. The underlying
discovery that they express on their surface mechanism has not been completely resolved, and
membrane a receptor tyrosine kinase that is the may vary in different parts of the gut. Integrated
gene product of c-kit, a proto-oncogene (for Ca2+ handling by the endoplasmic reticulum and
reviews, see Sanders et al., 1999, 2002; Ward and the mitochondria seem to be involved (Ward et al.,
Sanders, 2001). These cells can now be identified 2000), and it is thought that changes in [Ca2+]i
349

determined by phasic release of Ca2+ from endo- our knowledge of defecation in humans comes
plasmic reticulum through an IP3-dependent from less direct methodologies such as imaging the
mechanism and also uptake of Ca2+ by mi- gut (e.g., X-rays and ultrasound) behavioural
tochondria, lead to activation of Ca2+-dependent studies, recordings of pressure in the gastrointes-
conductances (possibly Cl channels or non-selec- tinal tract, and of electrical activity using needle or
tive cation channels) that can generate depolariz- surface electrodes. Information about the control
ing (inward) currents. When linked together in of the individual muscles can be obtained using
networks and activated synchronously, the inter- human tissue resected during surgical procedures.
stitial cells can inject sufficient current into adja- In the next section, examination of the gross and
cent smooth muscle cells to produce the more detailed properties of the various compo-
characteristic slow waves of depolarization seen nents will be made. Much of the work on the hu-
in gastrointestinal smooth muscles. man anorectum and its intrinsic innervation
described in this chapter is from research using
tissue retrieved from patients at surgery by clini-
Underlying mechanisms in defecation cians working for higher degrees in the Oxford
Continence Group, using a superfusion organ bath
Experimental approaches set-up (Brading and Sibley, 1982).
Our understanding of the basic mechanisms comes
from work on animals. Initially much work was
carried out on the overall organisation (central Colorectal motility and transport
and peripheral) controlling defecation using anaes- In health, the proximal colon (ascending and
thetised cats or dogs (Martner, 1975; de Groat and transverse segments) acts as a reservoir while the
Krier, 1978; Mackel, 1979; de Groat et al., 1981, descending colon serves as a conduit (Proano
1982; Fukuda et al., 1981; Fukuda and Fukai, et al., 1990). The movement of colonic contents is
1986; Takaki et al., 1987), but the current climate a discontinuous process, such that residue may be
with respect to animal research has limited the use retained for prolonged periods in the ascending
of these species. Studies of the properties of iso- colon and mass movements can deliver its contents
lated segments of the whole gut have been useful to the sigmoid colon in seconds. Colonic contrac-
for studying reflexes intrinsic to the enteric nerv- tions can either be phasic or tonic. The former are
ous system, but to keep these preparations alive the result of spike potentials and have definite be-
and functioning in vitro, it is necessary to use in- ginnings and endings, causing elevations in intra-
testine from small animals to enable proper oxy- luminal pressure (Garcia et al., 1991; Gregersen
genation of the tissue and survival of the intrinsic and Ehrlein, 1996). Ambulatory studies have
nerves; thus more recent experimental work has shown there to be several patterns of phasic co-
concentrated on rodents. Many of the detailed in- lonic contractions: single non-propagating con-
vestigations into the properties of the enteric nerv- tractions, antegrade pressure waves, retrograde
ous system have been carried out on guinea-pig gut pressure waves and periodic colonic motor activ-
(e.g., see Furness and Costa, 1987), including el- ity. Of note, high-amplitude propagating contrac-
egant demonstrations of the morphological, tions or giant migrating/mass contractions, the
immunohistochemical and functional properties of result of antegrade pressure waves, occur as iso-
the individual neurons in the plexuses. The motor lated events or in bursts (Sarna, 1991). They may
control of the smooth muscles of the lower gut can or may not result in propagation of colonic con-
be examined in the organ bath by dissecting strips tents. Mass contractions are mostly generated in
of smooth muscle and recording their spontaneous the daytime, especially after awakening or after
contractile behaviour and their evoked responses to meals (known as the gastro-colonic response).
electrical field stimulation of the intrinsic nerves. In relation to the gastro-colonic response, some
Many of the experimental techniques used on investigators report the response to be most
animals cannot be applied to normal humans, and intense in the sigmoid and descending colon
350

segments (Quigley, 2002). Localized phasic contractions thus arise from a zero baseline. In
contractions function to mix and move colonic contrast, the longitudinal smooth muscle may de-
contents over short distances (Cook et al., 2000; velop a basal tension of some 0.2 g/mg tissue, and
Rao et al., 2001). Tonic colonic contractions are the phasic contractions rise from this. These spon-
less well defined than phasic contractions. They taneous contractions are unaffected by tetrodo-
are long lasting and may or may not be associated toxin, and are probably evoked by slow waves
with increased intraluminal pressure (Gregersen generated in the interstitial cells of Cajal (see
and Ehrlein, 1996). above). Both muscle layers respond to activation
Three types of phasic rectal contractions have of their muscarinic receptors by contracting.
been described: isolated ones, short clusters of low Catecholamines abolish the spontaneous contrac-
amplitude contractions (frequency 5–6/min) and tions in both muscle layers and reduce the baseline
powerful contractions termed rectal phasic motor tension in the longitudinal smooth muscle (Fig. 4).
activity, every 60–120 min (Akervall et al., 1989). Activation of either a- or b-adrenoceptors can
The phasic activity is most common during the induce relaxation, exemplified by the fact that
night and is usually associated with simultaneous phenylephrine and isoproterenol produce the
colonic contractions and contractions of the anal same relaxant responses as norepinephrine. a-
sphincter, thus preventing defecation. adrenoceptors are known to induce relaxation of
Rectal tone appears to be influenced by the rate the longitudinal elements of the gut in many mam-
of volume change. Rapid volume waves lasting less malian species through opening of Ca-activated K
than 2 min have been frequently associated with channels (Kuriyama et al., 1998).
increase in luminal pressure, while slow volume The enteric nervous system is well developed
waves of greater than 2 min duration have not. in the rectum (O’Kelly et al., 1994), as shown in
Although the significance of these volume waves is Fig. 5. Extrapolation from detailed studies in other
uncertain, changes in the rectal tone may act to species (e.g., Gabella, 2001) suggests that the den-
alter its function between a capacious reservoir sity of innervation directly to the smooth muscle
and a conduit (Akervall et al., 1989). cells is fairly sparse, and that the interstitial cells of
The transit time for colonic contents is the cul- Cajal may be the main targets of the intrinsic
mination of anatomical and motility effects and is nerves (Sanders, 1996). Transmural stimulation of
relatively easily quantifiable (Abrahamsson et al.,
1988). Normally, the average mouth to caecum
transit time for ingested food is about 6 h and re-
gional transit times, measured using radio-opaque
marker method, are about 2 h each in the ascend-
ing, descending and sigmoid colon segments.
Although the mean colonic transit time is 36 h,
the range is wide since many factors can affect this
such as diet, drugs, etc. (Metcalf et al., 1987).

Properties of the rectal smooth muscles


The rectum is surrounded by a complete layer of
outer longitudinal and inner circular smooth mus-
cle. The properties of the two layers are probably
rather similar to those in other longitudinal ele-
ments of the gut. In humans, both the circular and
longitudinal smooth muscle layers generate phasic
contractions at a frequency of about 3–4/min
(Stebbing, 1998). In the circular smooth muscle, Fig. 4. Contractile responses of small strips of rectal smooth
there is little if any intrinsic tone, and these muscle to agonist application.
351

a-adrenoceptor antagonist, suggesting a tonic ex-


citatory sympathetic discharge. There does not
appear to be a tonic parasympathetic input to the
internal sphincter in vivo.
In addition, superimposed on the tonic state of
the internal sphincter are two intermittent wave-
form activities, namely, slow and ultraslow, ac-
cording to their respective frequencies (Sun et al.,
1990). The relevance of these is not yet clear but it
has been suggested that they may act either to op-
pose rectal pressure waves (Sorensen et al., 1989)
or to keep the anal canal closed to prevent desen-
sitization of the anoderm (Zbar et al., 2000).
The intrinsic innervation of the anal canal
has been examined in sections, (Holmes, 1961;
Aldridge and Campbell, 1968; Weinberg, 1970;
Baumgarten et al., 1971) and also in whole mounts
where it can be shown that the myenteric plexus
penetrates as far as the middle third of the anal
Fig. 5. Preparations of the human myenteric plexus stained canal, although the number of ganglia seems to be
with NADPH diaphorase to indicate nerves containing nitric less than in the rectum. Nitric oxide-producing
oxide synthase. (A) Myenteric plexus from the main rectum. (B) nerves divide and ramify into the internal anal
Enlarged view of ganglion cells in rectal myenteric plexus sphincter, as shown in Fig. 5 (O’Kelly et al., 1994).
showing pronounced staining. (C) Myenteric plexus from the
Functionally, the internal anal sphincter has
lower rectum. (D) Myenteric plexus from the anal canal. Scale
bars in (A), (B) and (D): 1 mm; in (C): 100 mm. From O’Kelly a robust inhibitory innervation. Burleigh and
et al. (1994), with permission. colleagues showed that electric field stimulation
caused a relaxation, which is blocked by tetrodo-
strips of rectal smooth muscle produce responses toxin but not by atropine or guanethidine (Burleigh
that are normally quite small and varied — where et al., 1979). This suggested that the relaxation
there is initial tone, the normal response is a tran- is nerve mediated, but not by cholinergic or ad-
sient small relaxation followed by a small contrac- renergic pathways, paving the way for a possible
tion. If tone is initiated by application of an non-adrenergic non-cholinergic mediator. The prop-
agonist such as histamine, field stimulation initiates erties of the human sphincter including the inhibitory
a robust relaxant response that is attenuated by innervation were reviewed by Penninckx et al. (1992).
nitric oxide synthase inhibition (Stebbing, 1998). O’Kelly and colleagues extended this work on
the human sphincter, and showed that strips of the
Properties of the internal anal sphincter sphincter relaxed to transmural stimulation of
The internal anal sphincter is in a continuous state their intrinsic nerves, and also relaxed in response
of contraction as a result of its intrinsic myogenic to nitric oxide donors. Nerve-mediated relaxation
properties (O’Kelly et al., 1993a, b) and extrinsic was inhibited in a concentration-dependent man-
innervation by the autonomic nerves (Gutierrez ner by the nitric oxide synthase blocker, L-N-9-
and Shah, 1975; Frenckner and Ihre, 1976). It nitroarginine, which competes with the normal
contributes up to 85% of the overall resting anal substrate for nitric oxide synthesis, L-arginine. The
pressure. Using in vivo experiments, investigators antagonistic effect was countered by the addition
have shown that reduction in sphincter tone can be of excess L-arginine (O’Kelly et al., 1993a),
achieved following sympathetic blockade by either providing good evidence for a nitrergic inhibitory
high spinal (T6–T12) anaesthesia (Frenckner and innervation (see Fig. 6). O’Kelly also showed that
Ihre, 1976) or by infusion of phentolamine, an the sphincter relaxes in response to muscarinic
352

activity (O’Kelly et al., 1993b). They respond to


activation of both a-adrenoceptors and muscarinic
receptors by contracting (Fig. 8).
Table 1 compares the basic properties of the
four smooth muscles of the anal canal and rectum,
emphasising their heterogeneity. The responsive-
ness of these smooth muscles to stimulation of
their adrenergic receptors reflects their overall
response to circulating epinephrine and activation
of sympathetic nerves, which will ensure that in
Fig. 6. Responses of a strip of human internal anal sphincter to ‘flight and fight’ conditions, activity will be re-
transmural stimulation (10 V, 0.5 ms, 8 Hz, 1 s train) at the dots. duced in the longitudinal parts of the gut, while the
The responses were abolished by application of the nitric oxide smooth muscle sphincters will be closed, ensuring
synthase blocker L-NOARG (10 5 M), were not affected by continence whilst reducing overall energy expend-
additional application of the inactive D isomer of arginine, but
restored by application of the natural substrate L-arginine
iture. However, under extreme conditions of fear,
which competes with L-NOARG for the enzyme. if the circulating levels of epinephrine become too
high, the relaxant responses to b-adrenoceptor
stimulation on the sphincteric smooth muscle may
underlie the anecdotal accounts of involuntary loss
of feces and urine.

Properties of the external anal sphincter


Since the inherent automaticity of the internal anal
sphincter confers the sphincter complex with a
resting tone, which in turn helps maintain our
subconscious control of continence, there has been
a greater emphasis on research looking into the
physiology and pharmacology of this compared to
the external sphincter. The striated muscle of the
external sphincter has a somatic nerve supply with
Fig. 7. Responses of strips of human internal anal sphincter to acetylcholine as the excitatory transmitter at the
10 s applications of 5  10 5 M carbachol (arrows) and trans- neuromuscular junction working via nicotinic re-
mural field stimulation (10 V, 0.5 ms, 8 Hz, 1 sec train) at the ceptors. The cell bodies of the motor neurons lie in
dots. The relaxant responses to both stimuli were abolished by the ventral horn of the second and third sacral
L-NOARG, and restored by additional application of L-argi-
segments of the spinal cord (Schroder, 1981).
nine.
The external sphincter can contribute up to 50%
of the anal pressure during periods of rectal dis-
receptor stimulation, contracts in response to
tension (Frenckner and Euler, 1975). When
activation of a-adrenoceptors, and relaxes in re-
required, the voluntary component can be called
sponse to stimulation of b-adrenoceptors (O’Kelly
upon to momentarily raise the anal pressure in
et al., 1993b). He postulated that muscarinic
order to defer defecation.
receptors were present on nitrergic neurons since
inhibitors of nitric oxide synthase attenuated the
relaxant response of the sphincter to carbachol, an Main areas of ignorance
acetylcholine analogue (Fig. 7). In contrast to the
internal anal sphincter, strips from the conjoined The above account perhaps illustrates that we
longitudinal coat only generate a small amount of can describe overall events, and have increasing
basic tone but show little spontaneous phasic knowledge of the properties of the individual
353

Fig. 8. A comparison of the effects of carbachol and norepinephrine (5  10 5 M, 10 sec) on the strips of smooth muscle dissected
from the conjoined longitudinal coat and the internal anal sphincter. Hexamethoium (10 6 M) was present to prevent any stimulation
of nicotinic acetylcholine receptors.

Table 1. Properties of smooth muscle from human ano-rectum

Tone Response to muscarinic Response to


agonist noradrenaline

Rectal circular m k
Rectal longitudinal + m k
Internal anal sphincter +++ k m
Conjoined longitudinal + m m

m contraction; k relaxation

muscles involved in defecation. However, we are considerable significance in patients with spinal
extremely ignorant about exactly how the process injury, since injuries at different levels of the cord,
of defecation is controlled. Although evidence and the likelihood that there may also be addi-
strongly suggests that there is a spinal defecation tional damage to autonomic nerves means that the
centre, the relative roles of the enteric nervous spectra of disruption to defecation must be ex-
system, the spinal centre and voluntary control is tremely varied. Exact knowledge of the extent of
hard to determine. Voluntary control via increas- the damage would be required to predict the dis-
ing intra-abdominal pressure can trigger increased ruption to defecation that is likely to occur for a
activity in the enteric nervous system through ac- particular patient, as well as the relative impor-
tivation of pressure sensitive nerves, and these may tance of the potential levels of control. More re-
enhance activity at any of the several sites — lo- search, in particular on whole animal models, may
cally in the enteric nervous system, through influ- help throw light onto the underlying processes.
encing autonomic ganglia en route to the CNS, at
the level of the spinal reflex centre and at higher Spinal cord injury
centres. Sensory nerve activity may enter the CNS
along with the parasympathetic, sympathetic, or Introduction
somatic nerves, and the CNS can alter activity in
the enteric nervous system through the parasym- Traditionally, lesions of motor neurons are classi-
pathetic and sympathetic nerves. The relative fied into supranuclear (supraconal or upper motor
importance of these different systems will be of neuron) or infranuclear (conal/cauda equina or
354

lower motor neuron) lesions. In striated muscles left (descending) colon transit times to be pro-
and the smooth muscle of the urinary bladder, longed (Beuret-Blanquart et al., 1990).
upper motor neuron lesions result in hyperactive In their relatively large study that examined the
or spastic paresis while lower motor neuron lesions changes in segmental colonic transit times among
lead to hyporeactive or flaccid paresis. 32 people with spinal cord injuries, Krogh and
However, the effects of spinal cord injury on the colleagues observed that chronic supraconal injury
colon, rectum and sphincter complex are less well significantly prolonged transit times at the trans-
known. This is in part due to our relatively poor verse and descending colon segments but not at the
understanding of the complex regulation of bowel rectosigmoid (Krogh et al., 1997). Chronic conal
function and, in particular, defecation. This is or cauda equina lesions were shown to prolong
compounded by the variable clinical effects ob- transit times significantly, not only at the trans-
served in spinal cord injury with 60% of those af- verse and descending colonic segments, but also at
flicted having incomplete lesions and 59% of these the rectosigmoid region. They also showed that
having significant recovery of function (Buckle there was no statistically significant difference in
et al., 1999). the total gastrointestinal transit times or segmental
We will therefore describe the potential effects colonic transit times between people with lesions
of spinal cord injury in relation to the clinical affecting the sympathetic outflow and those with
manifestation of disturbed colorectal transport/ lesions beyond this level, reinforcing the percep-
motility, and impaired defecation. As a matter of tion that the parasympathetic input is more im-
convenience, we have two categories: people with portant in colorectal transport.
supraconal or conal/cauda equina lesions. We will A subsequent study by the same group of in-
also briefly describe colorectal dysfunction after vestigators, albeit using a new scintigraphic tech-
acute injury. nique for quantitative assessment of segmental
colorectal transport, showed impaired emptying of
the rectosigmoid segment in people with sacral le-
sions when compared with a healthy volunteer
Effects on colorectal motility and transport group (Krogh et al., 2003). The gastro-colonic
response in people with supraconal lesions is also
In a study of the effects of spinal cord injuries on generally reduced or absent (Bruninga and
the total gastrointestinal transit time and colonic Camilleri, 1997).
transit times, Krogh and colleagues showed that
these parameters were significantly increased
chronically (Krogh et al., 2000). The regions in Disordered defecation
the colon and rectum responsible for the delays are
unclear, however. Devroede and colleagues found In a questionnaire survey of 424 members of the
prolonged transit times of the entire colon and Danish Paraplegia Association, constipation and
rectum in four subjects with incomplete lumbosa- disturbed defecation were the most common colo-
cral lesions (Devroede et al., 1979). In two other rectal concern affecting 81% of those with spinal
studies, colorectal transit times were also shown to cord injuries (Krogh et al., 1997). Symptoms as-
be prolonged after cervical and thoracic lesions. sociated with autonomic dysreflexia, a response
Nino-Murcia, who looked at transit times after usually generated by distended bladder or bowel,
cervical or thoracic lesions, found the total and left were noted in 25% before and during defecation.
(descending) colon transit times to be prolonged Studies have suggested rectal tone to be in-
but not the right (ascending) colon transit times creased after supraconal lesions (Sun et al., 1995)
(Nino-Murcia et al., 1990). In contrast, Beuret- and decreased after conal or cauda equina lesions
Blanquat and colleagues, in a study of 19 people (Krogh et al., 2002), similar to the effects seen in
with complete thoracic, lumbar or sacral lesions, the urinary bladder (Koldewijn et al., 1994). There
found neither the mean right (ascending) colon nor also appears to be a relative increase in giant rectal
355

contractions in people with supraconal injuries any contractile activity in a subject who had sus-
that may be important in their ability to induce tained supraconal cord injury days earlier (Denny-
defecation by means of mechanical (digital) stim- Brown and Robertson, 1935). Reflex defecation is
ulation of the rectum. It is postulated that this however, thought to return between 4 and 8 weeks
action may trigger both giant rectal contractions of the injury (Kuhn, 1947; Krogh et al., 2002).
and the recto-anal inhibitory reflex simultaneously Finally, the anal resting pressures were not shown
(Krogh et al., 2003). to be significantly lowered in an acute setting with
The effects of spinal cord injury on the recto- either supraconal or conal lesions (Krogh et al.,
anal inhibitory reflex are unclear. The reflex has 2002) but the ability to voluntarily contract the
been shown to be both increased (Sun et al., external sphincter was reduced or absent (Frenck-
1990b) and not (Frenckner, 1975) in those with ner, 1975; Sun et al., 1990a).
chronic supraconal injury. It has also been shown In the acute setting, therefore, spinal cord injury
to be increased (Devroede et al., 1979) or normal causes ileus (prolonged total gastrointestinal tran-
(Sun et al., 1990) in people with conal or cauda sit times), constipation (prolonged colonic transit
equina lesions. times) and fecal incontinence (passive leakage).
The anal sphincter resting pressure has been
shown to be lowered in subjects with supraconal
lesions but not in those with conal or caudal le- Conclusion
sions (Krogh et al., 2002). Fecal incontinence can
therefore be a common problem and is strongly The number of people with traumatic spinal cord
associated with complete lesions (Krogh et al., injury in most countries can only be estimated,
1997). Inevitably a component of fecal inconti- although the overall incidence is thought to be in
nence can be the result of using laxatives to over- the region of 12–40 per million annually (Illis,
come constipation. Finally, although we are 2004). With improved medical awareness and care,
increasingly aware of the dynamic role of the pel- the life expectancy for paraplegics surviving the
vic floor musculature in defecation, the extent of acute phase is now approaching the life expectancy
its likely dysfunction in people with spinal cord of the general population (Hartkopp et al., 1997).
injury is unclear. Consequently, activities that affect quality of life
such as effective bowel management are deemed
by patients to be important, more so even than
Colorectal transport and defecation in acute spinal bladder or sexual functions.
cord injury The inability to defecate normally means that
this ‘normal’ bodily function not only occupies a
Acutely (within 3 weeks of injury) colorectal tran- significant part of daily living but also acts as a
sit times (and total gastrointestinal transit times) in drain on resources e.g., the use of laxatives, toi-
those with both supraconal and conal injuries have letries, dependence on physical assistance (Glick-
been found to be at least twice as long as those of man and Kamm, 1996; Krogh et al., 1997). There
a healthy control group (Krogh et al., 2002). are also potential long-term effects of poor colo-
Rectosigmoid transit times were also significantly rectal motility and function in these people who
prolonged after conal/caudal lesions. No doubt may go on to develop diverticular disease, volvulus
these prolonged transit times may also, however, and haemorrhoids (Steins et al., 1997). As a result,
be the consequence of other factors encountered some have advocated early colostomies (Branagan
by the acutely ill spinal cord injury patients such et al., 2003).
as immobilisation, usage of opioid analgesics, The mechanisms and control of defecation both
reduced enteral diet intake and the unpredictable in healthy and spinal cord injured individuals are
effects of spinal shock and trauma generally. still poorly understood. The recent development
With regard to the anorectum, Denny-Brown of a pig model of defecation (Ramalingam,
and Robertson showed that the rectal wall lacked personal communication), should enable research
356

into defecation and its control in a species with de Groat, W.C., Booth, A.M., Milne, R.J. and Roppolo, J.R.
similar ano-rectal function to the human. Areas of (1982) Parasympathetic preganglionic neurons in the sacral
research using imaging techniques, such as dy- spinal cord. J. Auton. Nerv. Syst., 5: 23–43.
de Groat, W.C. and Krier, J. (1978) The sacral parasympathetic
namic magnetic resonance imaging, therapeutic reflex pathway regulating colonic motility and defaecation in
techniques, such as sacral nerve stimulation, and the cat. J. Physiol., 276: 481–500.
genetic manipulation, such as stem cell therapy, de Groat, W.C., Nadelhaft, I., Milne, R.J., Booth, A.M.,
will also no doubt help further unfold the Morgan, C. and Thor, K. (1981) Organization of the sacral
physiology of defecation. parasympathetic reflex pathways to the urinary bladder and
large intestine. J. Auton. Nerv. Syst., 3: 135–160.
Denny-Brown, D. and Robertson, E.G. (1935) An investigation
of the nervous control of defecation. Brain, 58: 256–310.
References Devroede, G., Arhan, P., Duguay, C., Tetreault, L., Akoury, H.
and Perey, B. (1979) Traumatic constipation. Gastroenterol-
Abrahamsson, H., Antov, S. and Bosaeus, I. (1988) Gastroin- ogy, 77: 1258–1267.
testinal and colonic segmental transit time evaluated by a Frenckner, B. (1975) Function of the anal sphincters in spinal
single abdominal X-ray in healthy subjects and constipated man. Gut, 16: 638–644.
patients. Scand. J. Gastroenterol,, 152(Suppl.): 72–80. Frenckner, B. and Euler, C.V. (1975) Influence of pudendal
Akervall, S., Fasth, S., Nordgren, S., Oresland, T. and Hulten, block on the function of the anal sphincters. Gut, 16: 482–489.
L. (1989) Rectal reservoir and sensory function studied by Frenckner, B. and Ihre, T. (1976) Influence of autonomic nerves
graded isobaric distension in normal man. Gut, 30: 496–502. on the internal and sphincter in man. Gut, 17: 306–312.
Aldridge, R.T. and Campbell, P.E. (1968) Ganglion cell distri- Fukuda, H. and Fukai, K. (1986) Postural change and straining
bution in the normal rectum and anal canal. A basis for the induced by distension of the rectum, vagina and urinary
diagnosis of Hirschsprung’s disease by anorectal biopsy. bladder of decerebrate dogs. Brain Res., 380: 276–286.
J. Pediatr. Surg., 3: 475–490. Fukuda, H., Fukai, K., Yamane, M. and Okada, H. (1981)
Bannister, L.H. (1995) Alimentary system. In: Williams P.L. Pontine reticular unit responses to pelvic nerve and colonic
(Ed.), Gray’s Anatomy (38th ed.). Churchill-Livingstone, mechanical stimulation in the dog. Brain Res., 207: 59–71.
London, pp. 1774–1786. Furness, J.B. and Costa, M. (1987) The Enteric Nervous Sys-
Baumgarten, H.G., Holstein, A.F. and Stelzner, F. (1971) Dif- tem. Churchill-Livingstone, New York.
ferences in the innervation of the large intestine and the in- Gabella, G. (2001) Development and ageing of intestinal mus-
ternal sphincter of the anus in mammals and humans. Verh. culature and nerves: the guinea-pig taenia coli. J. Ne-
Anat. Ges., 66: 43–47. urocytol., 30: 733–766.
Beuret-Blanquart, F., Weber, J., Gouverneur, J.P., De- Garcia, D., Hita, G., Mompean, B., Hernandez, A., Pellicer, E.,
mangeon, S. and Denis, P. (1990) Colonic transit time and Morales, G. and Parrilla, P. (1991) Colonic motility: electric
anorectal manometric anomalies in 19 patients with complete and manometric description of mass movement. Dis. Colon
transection of the spinal cord. J. Auton. Nerv. Syst., 30: Rectum,, 34: 577–584.
199–207. Glickman, S. and Kamm, M.A. (1996) Bowel dysfunction in
Brading, A.F. and Sibley, G.N. (1982) A superfusion apparatus spinal-cord-injury patients. Lancet, 347: 1651–1653.
to study field stimulation of smooth muscle from mammalian Gregersen, H. and Ehrlein, H.-J. (1996) Motility studies in lab-
urinary bladder. J. Physiol., 334: 11–12. oratory animals. In: Jensen S.L., Grgersen H., Amiri S. and
Branagan, G., Tromans, A. and Finnis, D. (2003) Effect of Moody F. (Eds.), Essentials of Experimental Surgery. Gastro-
stoma formation on bowel care and quality of life in patients enterology. Harwood Academic Publishers, New York,
with spinal cord injury. Spinal Cord, 41: 680–683. pp. 17/11–17/39.
Bruninga, K. and Camilleri, M. (1997) Colonic motility and Gutierrez, J.G. and Shah, A.N. (1975) Autonomic control of
tone after spinal cord and cauda equina injury. Am. J. Gas- the internal anal sphincter in man. In: Trappen V. (Ed.),
troenterol., 92: 891–894. Fifth International Symposium on Gastrointestinal Motility.
Buckle, D.A., McKenna, J. and Guanci, M. (1999) Spinal cord Typoff Press, Leuven, pp. 363–373.
trauma. Nurs. Clin. North Am., 34: 661–687. Hartkopp, A., Bronnum-Hansen, H., Seidenschnur, A.M. and
Burleigh, D.E., D’Mello, A. and Parks, A.G. (1979) Responses Biering-Sorensen, F. (1997) Survival and cause of death after
of isolated human internal anal sphincter to drugs and elec- traumatic spinal cord injury. A long-term epidemiological
trical field stimulation. Gastroenterology, 77: 484–490. survey from Denmark. Spinal Cord, 35: 76–85.
Cajal, S.R. (1893) Sur les ganglions et plexus nerveux de l’in- Holmes, A.M. (1961) Observations on the intrinsic innervation
testin. C.R. Soc. Biol., 45: 217–223. of the rectum and anal canal. J. Anat., 95: 416–422.
Cook, I.J., Furukawa, Y., Panagopoulos, V., Collins, P.J. and Illis, L.S. (2004) The case for specialist units. Spinal Cord,
Dent, J. (2000) Relationships between spatial patterns of co- 42: 443–446.
lonic pressure and individual movements of content. Am. J. Koldewijn, E.L., Van Kerrebroeck, P.E., Rosier, P.F.,
Physiol. Gastrointest. Liver Physiol., 278: G329–G341. Wijkstra, H. and Debruyne, F.M. (1994) Bladder compliance
357

after posterior sacral root rhizotomies and anterior sacral Proano, M., Camilleri, M., Phillips, S.F., Brown, M.L. and
root stimulation. J. Urol., 151: 955–960. Thomforde, G.M. (1990) Transit of solids through the hu-
Krogh, K., Mosdal, C., Gregersen, H. and Laurberg, S. (2002) man colon: regional quantification in the unprepared bowel.
Rectal wall properties in patients with acute and chronic Am. J. Physiol., 258: G856–G862.
spinal cord lesions. Dis. Colon Rectum, 45: 641–649. Quigley, E.M. (2002) Colonic motility and colonic function. In:
Krogh, K., Mosdal, C. and Laurberg, S. (2000) Gastrointestinal Pemberton J., Swash M. and Henry M. (Eds.), The Pelvic
and segmental colonic transit times in patients with acute and Floor: Its Function and Disorders. W.B Saunders, London,
chronic spinal cord lesions. Spinal Cord, 38: 615–621. pp. 84–93.
Krogh, K., Nielsen, J., Djurhuus, J.C., Mosdal, C., Sabroe, S. Rao, S.S., Sadeghi, P., Beaty, J., Kavlock, R. and Ackerson, K.
and Laurberg, S. (1997) Colorectal function in patients with (2001) Ambulatory 24-h colonic manometry in healthy hu-
spinal cord lesions. Dis. Colon Rectum,, 40: 1233–1239. mans. Am. J. Physiol. Gastrointest. Liver Physiol., 280:
Krogh, K., Olsen, N., Christensen, P., Madsen, J.L. and G629–G639.
Laurberg, S. (2003) Colorectal transport during defecation in Sanders, K.M. (1996) A case for interstitial cells of Cajal as
patients with lesions of the sacral spinal cord. Ne- pacemakers and mediators of neurotransmission in the gas-
urogastroenterol. Motil., 15: 25–31. trointestinal tract. Gastroenterology, 111: 492–515.
Kuhn, W.G. (1947) The care and rehabilitation of patients with Sanders, K.M., Ordog, T., Koh, S.D., Torihashi, S. and Ward,
injuries of the spinal cord and cauda equina. J. Neurosurg., 4: S.M. (1999) Development and plasticity of interstitial cells of
40–68. Cajal. Neurogastroenterol. Motil., 11: 311–338.
Kuriyama, H., Kitamura, K., Itoh, T. and Inoue, R. (1998) Sanders, K.M., Ordog, T. and Ward, S.M. (2002) Physiology
Physiological features of visceral smooth muscle cells, with and pathophysiology of the interstitial cells of Cajal: from
special reference to receptors and ion channels. Physiol. Rev.,
bench to bedside. IV. Genetic and animal models of GI mo-
78: 811–920.
tility disorders caused by loss of interstitial cells of Cajal. Am.
Mackel, R. (1979) Segmental and descending control of the
J. Physiol. Gastrointest. Liver Physiol., 282: G747–G756.
external urethral and anal sphincters in the cat. J. Physiol.,
Sarna, S.K. (1991) Physiology and pathophysiology of colonic
294: 105–122.
motor activity (1). Dig. Dis. Sci., 36: 827–862.
Martner, J. (1975) Influences on the defecation and micturition
Schroder, H.D. (1981) Onuf’s nucleus X: a morphological study
reflexes by the cerebellar fastigial nucleus. Acta Physiol.
of a human spinal nucleus. Anat. Embryol. (Berl.), 162:
Scand., 94: 95–104.
443–453.
Metcalf, A.M., Phillips, S.F., Zinsmeister, A.R., MacCarty, R.L.,
Sorensen, S.M., Gregersen, H., Sorensen, S. and Djurhuus, J.C.
Beart, R.W. and Wolff, B.G. (1987) Simplified assessment of
(1989) Spontaneous anorectal pressure activity. Evidence of
segmental colonic transit. Gastroenterology, 92: 40–47.
Miller, R., Bartolo, D.C., Cervero, F. and Mortensen, N.J. internal anal sphincter contractions in response to rectal
(1988a) Anorectal sampling: a comparison of normal and pressure waves. Scand. J. Gastroenterol., 24: 115–200.
incontinent patients. Br. J. Surg., 75: 44–47. Stebbing, J.F. (1998) Nitric oxide synthase neurones and ne-
Miller, R., Lewis, G.T., Bartolo, D.C., Cervero, F. and Mor- uromuscular behaviour of the anorectum. Ann. R. Coll.
tensen, N.J. (1988b) Sensory discrimination and dynamic Surg. Engl., 80: 137–145.
activity in the anorectum: evidence using a new ambulatory Steins, S.A. and Bergman. S.B. and Goetz, L.L. (1997) Ne-
technique. Br. J. Surg., 75: 1003–1007. urogenic bowel dysfunction after spinal cord injury: clinical
Nino-Murcia, M., Stone, J.M., Chang, P.J. and Perkash, I. evaluation and rehabilitative management. Arch. Phys. Med.
(1990) Colonic transit in spinal cord-injured patients. Invest. Rehabil., 78: S86–S102.
Radiol., 25: 109–112. Sun, W.M., MacDonagh, R., Forster, D., Thomas, D.G.,
O’Kelly, T., Brading, A. and Mortensen, N. (1993a) Nerve Smallwood, R. and Read, N.W. (1995) Anorectal function in
mediated relaxation of the human internal anal sphincter: the patients with complete spinal transection before and after
role of nitric oxide. Gut, 34: 689–693. sacral posterior rhizotomy. Gastroenterology, 108: 990–998.
O’Kelly, T.J., Brading, A. and Mortensen, N.J. (1993b) In vitro Sun, W.M., Read, N.W. and Donnelly, T.C. (1990a) Anorectal
response of the human anal canal longitudinal muscle layer function in incontinent patients with cerebrospinal disease.
to cholinergic and adrenergic stimulation: evidence of sphinc- Gastroenterology, 99: 1372–1379.
ter specialization. Br. J. Surg., 80: 1337–1341. Sun, W.M., Read, N.W., Miner, P.B., Kerrigan, D.D. and
O’Kelly, T.J., Davies, J.R., Brading, A.F. and Mortensen, N.J. Donnelly, T.C. (1990b) The role of transient internal sphinc-
(1994) Distribution of nitric oxide synthase containing neu- ter relaxation in faecal incontinence? Int. J. Colorectal Dis.,
rons in the rectal myenteric plexus and anal canal. Morpho- 5: 31–36.
logic evidence that nitric oxide mediates the rectoanal Takaki, M., Neya, T. and Nakayama, S. (1987) Functional role
inhibitory reflex. Dis. Colon Rectum,, 37: 350–357. of lumbar sympathetic nerves and supraspinal mechanism in
Penninckx, F., Lestar, B. and Kerremans, R. (1992) The inter- the defecation reflex of the cat. Acta. Med. Okayama,, 41:
nal anal sphincter: mechanisms of control and its role in 249–257.
maintaining anal continence. Baillieres Clin. Gastroenterol., Tomita, T. (1981) Electrical activity (spikes and slow waves) in
6: 193–214. gastrointestinal smooth muscle. In: Bulbring E., Brading
358

A.F., Jones A.W. and Tomita T. (Eds.), Smooth Muscle: An tial cells of Cajal networks. Am. J. Physiol. Gastrointest.
assessment of Current Knowledge. E. Arnold, London. Liver Physiol., 281: G602–G611.
Ward, S.M., Ordog, T., Koh, S.D., Baker, S.A., Jun, J.Y., Weinberg, A.G. (1970) The anorectal myenteric plexus: its re-
Amberg, G., Monaghan, K. and Sanders, K.M. (2000) Pace- lation to hypoganglionosis of the colon. Am. J. Clin. Pathol.,
making in interstitial cells of Cajal depends upon calcium 54: 637–642.
handling by endoplasmic reticulum and mitochondria. J. Zbar, A.P., Jayne, D.G., Mathur, D., Ambrose, N.S. and
Physiol., 525(Part 2): 355–361. Guillou, P.J. (2000) The importance of the internal anal
Ward, S.M. and Sanders, K.M. (2001) Physiology and patho- sphincter (IAS) in maintaining continence: anatomical, phys-
physiology of the interstitial cell of Cajal: from bench to iological and pharmacological considerations. Colorectal
bedside. I. Functional development and plasticity of intersti- Dis., 2: 193–202.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 24

Alterations in eliminative and sexual reflexes after


spinal cord injury: defecatory function and
development of spasticity in pelvic floor musculature

Yvette S. Nout1, Gail M. Leedy2, Michael S. Beattie1 and Jacqueline C. Bresnahan1,

1
Department of Neuroscience, Laboratory of CNS Repair and Spinal Trauma and Repair Laboratories, The Ohio State
University College of Medicine and Public Health, Columbus, OH, USA
2
Division of Social Work, University of Wyoming, Laramie, WY, USA

Abstract: Spinal cord injury often results in loss of normal eliminative and sexual functions. This chapter is
focused on defecatory function, although aspects of micturition and erectile function will be covered as well
due to the overlap in anatomical organization and response to injury. These systems have both autonomic
and somatic components, and are organized in the thoracolumbar (sympathetic), lumbosacral (somatic),
and sacral (parasympathetic) spinal cord. Loss of supraspinal descending control and plasticity-mediated
alterations at the level of the spinal cord, result in loss of voluntary control and in abnormal functioning of
these systems including the development of dyssynergies and spasticity. There are several useful models of
spinal cord injury in rodents that exhibit many of the autonomic dysfunctions observed after spinal cord
injury in humans. Numerous studies involving these animal models have demonstrated development of
abnormalities in bladder, external anal sphincter, and erectile function, such as detrusor–sphinc-
ter–dyssynergia and external anal sphincter hyperreflexia. Here we review many of these studies and
show some of the anatomical alterations that develop within the spinal cord during the development of
these hyperreflexias. Furthermore, we show that spasticity develops in other pelvic floor musculature as
well, such as the bulbospongiosus muscle, which results in increased duration and magnitude of pressures
developed during erectile events and increased duration of micturition. Advances and continued improve-
ment in the use of current animal models of spinal cord injury should encourage and increase the laboratory
work devoted to this relatively neglected area of experimental spinal cord injury.

Introduction and somatic motor pools in the caudal lumbar/


upper sacral cord. Although the basic afferent and
Both eliminative and sexual functions are com- efferent limbs of the reflex components are seg-
promised following spinal cord injury (Comarr, mentally organized, the loss of brainstem and
1970; Frenckner, 1975; Pedersen, 1983). These sys- forebrain descending input results in loss of vol-
tems have both autonomic and somatic compo- untary control and in abnormal functioning of
nents, and are organized by sympathetic neuronal these systems including the development of dyssy-
pools in the caudal thoracic/upper lumbar cord, nergies and spasticity. Urinary function has been
parasympathetic neuronal pools in the sacral cord, studied most in animal models, and the complete
loss of descending input initially causes an absence
Corresponding author. Tel.: +614-292-2206; of bladder emptying which is gradually replaced
E-mail: bresnahan.1@osu.edu by spinal reflex circuit activity that is initiated by a

DOI: 10.1016/S0079-6123(05)52024-7 359


360

different population of afferents (e.g., c-fiber by thickening of circular smooth muscle that lies
afferents) than in the normal condition (De Groat immediately inside the anus. Similar to the rest of
et al., 1990, 1998). While some recovery of this the gastrointestinal smooth muscle, the internal
function occurs, residual deficits remain in the co- anal sphincter contains a myenteric plexus within
ordination of the autonomic efferents to the the tunica muscularis. Furthermore, the internal
detrusor muscle and the somatic efferents to the sphincter muscle receives autonomic innervation
external urethral sphincter muscle (so-called via the pelvic plexus. The external anal sphincter is
bladder-sphincter dyssynergia) producing inade- composed of striated muscle that surrounds the
quate bladder emptying. Defecatory function has internal anal sphincter and extends distal to it. The
been less well studied but exhibits similar respons- external sphincter is controlled by nerve fibers in
es to the loss of descending control causing inad- the pudendal nerve, which in the rat has motor,
equate voiding, fecal impaction, and chronic sensory, and autonomic components (Rexed, 1954;
constipation in man (Frenckner 1975; Cosman Katagiri et al., 1986; Holstege and Tan, 1987;
et al., 1991) due, perhaps, to dyssynergia and Paxinos, 1995). While the internal anal sphincter is
external anal sphincter spasticity (Holmes et al., the main mechanism for continence, accounting for
1998, 2005). Sexual reflexes are similarly disrupted; the majority of the resting pressure, subconsciously
in humans, loss of descending input results in the external sphincter is usually kept constricted
facilitated spinally mediated erectile activity but unless conscious signals inhibit constriction
disruption of ejaculatory function and seminal flu- (Gonella et al., 1987). Motor units of the external
id composition (Higgins, 1979; Basu et al., 2004). anal sphincter have been shown to be tonically
The focus of the present chapter will be on def- active (Krier, 1985; Gonella et al., 1987), whereas
ecation as other eliminative and sexual functions the other pudendal motor units are effectively qui-
will be covered in other chapters (for urinary blad- escent (Dubrovsky et al., 1985; Holmes et al., 1994).
der and sexual function, see other chapters in this The rectum and anal canal are supported by the
volume). However, we will discuss these other sys- pelvic diaphragmatic musculature, which partici-
tems to some extent here as well, as their anatom- pates in the functions of fecal continence and def-
ical organization is co-extensive, their functions ecation. Paired rectococcygeal muscles originate
overlap, and response to injury is similar. We will on each side of the rectum and attach dorsally to
first describe the anatomy of the distal gastrointes- the base of the tail. These smooth muscles are in-
tinal tract, its innervation including the central and nervated by autonomic fibers from the pelvic plex-
peripheral components; this will be followed by a us and shorten the rectum during defecation to
discussion of normal eliminative function and then assist evacuation of feces. Laterally the illeocauda-
of dysfunctions resulting from spinal cord injury. lis and pubocaudalis and the coccygeus muscles
surround the rectum. These striated muscles are
innervated by the 6th lumbar (L6) to 1st sacral
Neuroanatomy (S1) nerves in the rat (Bremer et al., 2003) and help
to compress the rectum during defecation.
Peripheral components

The most distal portion of the gastrointestinal Pelvic plexus


tract is formed by the rectum and anus. Innerva-
tion of the rectum is similar to that of the colon, The pelvic plexus is a crossroad of autonomic
but innervation of the anus is more complex (for a nerves and ganglia supplying the rectum, the lower
comprehensive review, see Jänig and McLachlan, urinary tract, and the genital tract (Paxinos, 1995).
1987). The anus controls passage of fecal matter In the male rat, the main components of this plex-
by the degree of constriction of the internal and us are the right and left pelvic ganglia, also referred
external anal sphincters (Schuster, 1968; Gonella to as the hypogastric ganglia, or the major pelvic
et al., 1987). The internal anal sphincter is formed ganglia. These ganglia are positioned on either side
361

of the prostate, ventral to the rectum, and caudal obturator, ventral and dorsal bulbospongiosus
to the ureter and vas deferens, and are unusual (also termed bulbocavernosus), ischiocavernosus,
among autonomic ganglia, since they contain both external urethral and anal sphincter muscles, and
parasympathetic and sympathetic post-ganglionic afferent fibers from the penis, prepuce, scrotum,
neurons. The main afferents to the pelvic ganglia and ventral-proximal tail (McKenna and Nadelhaft,
are the hypogastric nerve and the pelvic nerve 1986). The sacral plexus is the complex formed by
(Paxinos, 1995). the bridge-like structure connecting the pudendal
The majority of fibers comprising the hypogastric nerve with the lumbosacral trunk, and two nerve
nerve originate in the intermediolateral cell column branches emerging from it, one innervating the
of the thoracolumbar cord [thoracic (T)11–L2] proximal half of the scrotal skin, and the other,
(Baron and Jänig, 1991). These preganglionic sym- known as the motor branch, innervating the mus-
pathetic neurons send their axons to sympathetic cles at the base of the penis (Pacheco et al., 1997).
ganglion cells of the bladder in the sympathetic These branches are only considered as a part of the
chain (T12–L6), pelvic ganglia, and to the inferior sacral plexus because they integrate axons from
mesenteric ganglia (Vera and Nadelhaft, 1992). The both the lumbosacral trunk and pudendal nerve.
caudal continuation of the inferior mesenteric gan- In rats, retrograde tracer studies of the pudendal
glion forms the hypogastric nerve, which carries the nerve have demonstrated the presence of two mo-
majority of the sympathetic input to the pelvic tor nuclei in the ventral horn gray matter of the
plexus. A small number of hypogastric fibers have L5–L6 spinal cord segments, the dorsomedial, and
their cells of origin located in either the sympathetic dorsolateral cell columns (Fig. 1; Breedlove and
chain or the inferior mesenteric ganglion. Arnold, 1980; McKenna and Nadelhaft, 1986;
The pelvic nerve carries the parasympathetic in- Vera and Nadelhaft, 1992). These nuclei are the
put and originates from the last lumbar (L6) and homologs to the nucleus of Onuf, which is found
first sacral (S1) spinal nerves in the rat (Purinton in other species, and contains motor neurons in-
et al., 1973). The pelvic nerve also carries a small nervating the anal and urethral sphincters, bulb-
portion of sympathetic fibers to the pelvic viscera. ospongiosus, and ischiocavernosus muscles, all
Numerous small efferents arise from the pelvic located in one cell group. McKenna and Nadelhaft
ganglion and supply the rectum, the ureter, the (1986) also demonstrated that in the female rat,
vas deferens, the seminal vesicles, the prostate, the urethral sphincter motor neurons accounted for
bladder, and the urethra. In the female rat almost all the dorsolateral nucleus motor neurons,
the pelvic ganglion, which is also referred to as and anal sphincter motor neurons accounted for
the paracervical or Frankenhauser ganglion, is almost all the dorsomedial motor neurons; the is-
smaller than that in males and lies against the lat- chiocavernosus and bulbospongiosus muscles are
eral wall of the uterine cervix. Afferent neurons vestigial in the female rat. In the male, neurons
with fibers in the pelvic nerve are located in the innervating the external anal sphincter and bulb-
L6–S1 dorsal root ganglia in the rat and carry ospongiosus muscles are found intermingled in the
sensory information from the descending colon, dorsomedial nucleus. In contrast, in the dorsolat-
bladder, urethra, and sex organs (Nadelhaft and eral nucleus, the urethral sphincter neurons are
Booth, 1984). These fibers enter the tract of located in the lateral portion of the nucleus and the
Lissauer, encircle the dorsal horn with a medial ischiocavernosus neurons are located in the medial
component terminating in the dorsal commissural portion (McKenna and Nadelhaft, 1986; Collins
gray and the lateral component terminating in the et al., 1992; Hermann et al., 1998). There are sig-
sacral parasympathetic nucleus. nificantly more neurons in both nuclei in the male
than in the female and the neurons are larger in the
Pudendal nerve male. However, perineal muscle tracer injections
have established that there is no difference be-
The pudendal nerve arises from L5–L6 in the rat tween males and females in the number of motor
and carries efferent fibers to the coccygeus, internal neurons innervating the external anal or urethral
362

Fig. 1. The caudal lumbar and rostral sacral spinal cord contains the neuronal circuitry that organizes the parasympathetic com-
ponents of bladder, bowel, and sexual function. DCG: dorsal commissural gray; SPN: sacral parasympathetic nucleus; DL: dorso-
lateral nucleus; DM: dorsomedial nucleus; DF: dorsal funiculus; DH: dorsal horn; CC: central canal; VM: ventromedial nucleus; RDL:
retrodorsolateral nucleus. (A) The cord at this level contains the SPN and the associated somatic motor columns innervating the pelvic
floor musculature, the DM, and DL nuclei. (B, C) The L6 segment contains the DM and DL nuclei that are characterized by a lack of
myelinated fibers; this is most easily seen in the DL. The boxed area in (B) is shown at higher magnification in (C). In this particular
section, bulbospongiosus motor neuron cell bodies are retrogradely labeled with horseradish peroxidase (HRP) and can be easily
identified in the higher power image shown in (C). This is a plastic embedded section containing the labeled bulbospongiosus motor
neurons cell bodies; diaminobenzidine was used to visualize the horseradish peroxidase in the neurons. After processing the tissue for
HRP, it was stained en bloc with osmium and uranyl acetate and embedded in plastic for subsequent sectioning for electron mi-
croscopy. Myelinated fibers appear black (note the darker staining of the ventral white commissure just above the DM nuclei and in the
white matter of the dorsal lateral and ventral funiculi). The retrogradely labeled neurons also appear dark in this section (arrow in C).
(D) This micrograph shows a 1.0 mm thick, toluidine blue stained section of the DM area enclosed by the circle in (C). Two motor
neuron cell bodies are shown in cross section; the cell on the left has a clear nucleus, clumps of rough endoplasmic reticulum in the
cytoplasm, and a large primary dendrite extending dorsally (toward the letter D). The other cell body has clumps of rough endoplasmic
reticulum and short primary dendrites that extend mostly out of the plane of section. There is a high density of cross-cut dendrites that
appear as small white circles; these are rostrocaudally oriented bundles of dendrites. Black myelinated fibers can be seen at the top of
the micrograph and a couple of fibers can be seen to traverse the nucleus between the two cell bodies. The magnification is too low to
identify the HRP-stained elements in the cytoplasm. (E, F). These low power dark field micrographs of adjacent sections show the
extensive dendritic arborizations of bulbospongiosus motor neurons retrogradely labeled with horseradish peroxidase from muscle
injections (tetramethyl benzidine processing). The bundles extend into the DCG region that receives pudendal and pelvic nerve afferent
input, into the DL nucleus that contains motor neurons of the ischiocavernosus and external urethral sphincter muscles, and over to
the contralateral DM nucleus.
363

sphincters (McKenna and Nadelhaft, 1986). These 1990). For example, the external anal sphincter
nuclei have a number of special anatomic features motor neurons project dendrites into the dorsal
(see Fig. 1) similar to the phrenic nucleus, a motor band of the sacral parasympathetic nucleus where
nucleus in the cervical spinal cord that innervates the colonic preganglionics are located, and the ex-
the diaphragm. These nuclei lack myelinated fibers ternal urethral sphincter motor neurons into its
making them appear translucent, similar to the lateral band where the bladder preganglionics re-
substantia gelatinosa. The motor neurons exhibit side. Similarly, the bulbospongiosus motor neurons
extensive dendritic bundling (Roney et al., 1979; in the rat extend profuse dendrites into the dorsal
Schroder, 1980; McKenna and Nadelhaft, 1986; commissural region (Fig. 1E) where interneurons
Sasaki, 1994); the rostrocaudally oriented dendri- important in sexual reflex function are located
tic bundles can be seen in Fig. 1D. Dense bundles (Peshori et al., 1995; Nadelhaft and Vera, 2001).
of dendrites from bulbospongiosus motor neurons Pudendal nerve afferent neurons are located in
can be seen in Figs. 1E and F (arrows), intercon- the L6 and S1 dorsal root ganglia (Nadelhaft and
necting the dorsomedial nuclei on the two sides as Booth, 1984). In both sexes, pudendal afferent
well as the dorsomedial and dorsolateral nuclei on fibers in the spinal cord are located in the dorsal
the same side. A dorsally extending group projects columns, the medial half of Lissauer’s tract, the
into the dorsal commissural gray (Figs. 1E and F). extreme medial edge of the dorsal horn, both ipsi-
Peshori et al. (1995) demonstrated that motor laterally and contralaterally, and in a large termi-
neurons in the contralateral dorsomedial nucleus nal field in the dorsal gray commissure. No
could be transneuronally labeled with wheatgerm- afferents have been detected in the intermediate
agglutinin horseradish peroxidase after a unilater- or ventral gray (McKenna and Nadelhaft, 1986).
al muscle injection and dye-coupling has been
demonstrated between bulbocavernosus motor
neurons by Matsumoto et al. (1988). At the ultra- Defecation
structural level, the dendrites are connected with
tight junctions or puncta adherentia (Ramirez- Defecation reflexes
Leon and Ulfhake, 1993; see Figs. 2A and B) as
well as through presynaptic dendrites (PSD; see In normal animals and humans, defecation is in-
Figs. 2A–C, and D). The synaptic arrangements in itiated by defecation reflexes (Gonella, 1987;
this region are also somewhat unique, in that axon Guyton and Hall, 2000). One of these reflexes is
terminals are commonly contacting more than one an intrinsic reflex mediated by the local enteric
dendrite (see, e.g., AT1 and AT2 in Fig. 2; nervous system in the rectal wall. Distention of the
Ramirez-Leon and Ulfhake, 1993). The dense in- rectal wall initiates afferent signals that spread
nervation of this region by g-amino butyric acid through the myenteric plexus to initiate peristaltic
(GABA)-containing elements is also of note waves in the descending colon, sigmoid, and rec-
(Ramirez-Leon and Ulfhake, 1993; Li et al., 1995). tum, forcing feces toward the anus. As the per-
It is likely that these anatomical arrangements can istaltic wave approaches the anus, the internal anal
provide for a more synchronized activation of sphincter is relaxed by inhibitory signals from the
motor neurons controlling pelvic floor musculature myenteric plexus. A rectoanal inhibition reflex oc-
(e.g., the sphincters and erectile musculature). curs in which as the rectum fills and the internal
The pudendal motor neuron dendrites occupy anal sphincter relaxes, the external anal sphincter
most of the ventral horn and reach dorsally be- contracts involuntarily and defecation only occurs
yond the central canal (Sasaki, 1994; Peshori et al., following further rectal distension inducing con-
1995; Hermann et al., 1998). In the cat, the sphinc- comitant external anal sphincter relaxation
ter motor neuron dendritic arbors have been (Gonella et al., 1987). This local intrinsic defeca-
shown to extend into the regions where function- tion reflex, however, is by itself relatively weak and
ally associated preganglionic neurons in the sacral usually must be fortified by another type of def-
parasympathetic nucleus are located (Beattie et al., ecation reflex, a parasympathetic defecation reflex
364

Fig. 2. Electron micrographs showing a variety of interactions between dendritic elements in Onuf’s nucleus in the cat, the homolog of
the rat dorsomedial (DM), and dorsolateral (DL) nuclei. Two dendrites (D1 and D2) of retrogradely identified pudendal motoneurons
receive synaptic input from a variety of axon terminals, some of which contact both dendrites (AT1 and AT2). The dendrites also are
closely apposed and have regions of direct appositions with puncta adherentia (small arrows in B). An adjacent dendrite (PSD) gives
rise to an element that appears to be presynaptic to D1 (synaptic specializations on D1 are indicated by the small arrows on the bottom
of the higher power figure shown in D). This dendritic excrescence is also post-synaptic to two adjacent axon terminals, one on the right
and one on the left, both of which are presynaptic to D1. In the higher power figure shown in (D), synaptic vesicles in the right terminal
are clustered at the membrane and a clear post-synaptic density is visible between the small arrows. The dendrite (D1) also has a region
containing vesicles (VES) and may be presynaptic to a small dendrite (shown in C at higher magnification). There is a synaptic
specialization between the small arrows at the interface with D1 as well as with a synaptic terminal surrounding this dendritic element
(small arrows on the left).

that involves the sacral segments of the spinal sphincter, and thus convert the intrinsic myenter-
cord. When nerve endings in the rectum are stim- ic defecation reflex from a weak effort into a pow-
ulated by distension, signals are transmitted first erful process of defecation.
into the spinal cord and then reflexively back to The afferent defecation signals entering the spi-
the descending colon, rectum, and anus by way of nal cord initiate other effects such as closure of the
parasympathetic nerve fibers in the pelvic nerves. glottis and contracture of the abdominal wall
These parasympathetic signals greatly intensify the muscles to force fecal contents of the colon down-
peristaltic waves and relax the internal anal ward and at the same time cause the pelvic floor to
365

relax downward and pull outward on the anal ring motor neurons and terminals of nucleus raphe
to evaginate feces. obscurus projections in the lumbosacral spinal
cord have been shown to co-localize with se-
rotonin, thyrotropin-releasing hormone, and sub-
Supraspinal control mechanisms stance P (Ramirez-Leon et al., 1994).
In fact, the nucleus raphe obscurus is thought to
Although in the rat reflex defecation is mainly or- be an overall regulator of autonomic functions
ganized at the spinal level, there is evidence for controlled by both cranial and spinal autonomic
supraspinal centers that may modify the functional efferents. Rostral portions of the nucleus raphe
response (Maggi et al., 1988). These supraspinal obscurus project to the dorsal vagal complex,
inputs strongly appear to influence the magnitude which controls proximal digestive functions
and coordination of these reflexes, and generally (McCann et al., 1989), whereas caudal portions
this descending supraspinal input is inhibitory to of the nucleus raphe obscurus project to the caudal
the lumbosacral reflexes (described above as the intermediolateral cell column, sacral parasympa-
parasympathetic defecation reflex). Retrograde thetic nucleus, and related somatic motor neurons,
tracer injections into the ventromedial gray of which control distal digestive and other pelvic au-
the lumbar spinal cord labeled supraspinal input tonomic functions. Also, the nucleus raphe ob-
primarily from the vestibular nuclei, the giganto- scurus maintains direct projections to both
cellular reticular nuclei, the medullary raphe nu- autonomic and somatic regions and to regions of
clei, and the hypothalamic paraventricular nuclei the intermediate gray, which contain putative in-
(Marson and McKenna, 1990; Shen et al., 1990; terneurons that contribute to the organization of
Monaghan and Breedlove, 1991; Marson et al., pelvic floor reflexes. The nucleus raphe obscurus is
1992). The ventrolateral gray of the lumbar spinal thus in a position to modulate autonomic pre-
cord has supraspinal input from the dorsolateral ganglionic and functionally related skeletal motor
pontine tegmental region, also referred to as Bar- neuron activity (Hermann et al., 1998).
rington’s nucleus or the pontine micturition center Electrical stimulation of the nucleus raphe
(Ding et al., 1995). Furthermore, transneuronal obscurus results in reduction of spontaneous ano-
tracing studies using pseudorabies virus injections rectal activity, providing evidence for a direct
directly into the muscles involved in pelvic floor brainstem inhibitory circuit (Holmes et al., 1997a).
reflexes have consistently confirmed these same Also, nucleus raphe obscurus lesions elicit tran-
regions to be the source of supraspinal input sient increases in anorectal reflex activity (Beattie
(Marson and McKenna, 1996; Tang et al., 1999; et al., 1996; Holmes et al., 2002) and disrupt male
Vizzard et al., 2000). Some studies have focused on copulatory behavior in rats (Yamanouchi and
supraspinal input originating from the nucleus Kakeyama, 1992). However, nucleus raphe ob-
raphe obscurus in the brainstem and the region in scurus lesions do not have an effect on measures of
the ventrolateral medulla referred to as the gig- ex copula penile reflexes (Holmes et al., 2002). In
antocellular–lateral paragigantocellular complex. contrast, lesions of the gigantocellular–lateral
Projections from this latter area are widespread paragigantocellular complex did not affect meas-
and include fibers in close apposition to external ures of external anal sphincter activity but altered
anal sphincter and bulbospongiosus motor neu- penile reflexes significantly (Holmes et al., 2002). It
rons, and although targets from the nucleus raphe has been shown that the bulbospongiosus muscle
obscurus are much more restricted, they also in- displays electromyographic activity during the
clude contacts between nucleus raphe obscurus passage of urine (Sachs and Leipheimer, 1988;
terminals and bulbospongiosus and external anal Schmidt et al., 1995), and it is suggested that the
sphincter motor neurons (Hermann et al., 1998, nucleus raphe obscurus projections may be specific
2003). More specifically, fibers from the nucleus to pudendal eliminative reflexes, while other
raphe obscurus ramify in close apposition to both descending brainstem projections are specific to
bulbospongiosus and external anal sphincter sexual reflexes (Holmes et al., 2002).
366

Projections from the gigantocellular–lateral gastric secretion and motility, intestinal transport,
paragigantocellular complex are diffuse, descend- and is thought to play a role in stress-induced in-
ing to all levels of the spinal cord affecting sensory, creases in defecation (Miyata et al., 1992).
motor, and autonomic control circuits beyond Although thyrotropin-releasing hormone appears
those for pudendal reflexes. The gigantocellu- to have an excitatory effect on external anal sphinc-
lar–lateral paragigantocellular complex may glo- ter motor neurons, intrathecally applied thyrotro-
bally modulate multiple behaviors with somatic pin-releasing hormone has an inhibitory effect on
and autonomic components such as reproductive penile erections, suggesting that although external
reflexes, acoustic startle, and nociception as pro- anal sphincter and bulbospongiosus motor neurons
posed by Hermann et al. (2003). are co-mingled within the same spinal nucleus, they
Neurotransmitters implicated in the descending are discretely and differentially regulated by sepa-
supraspinal control of pelvic and pudendal motor rate neural circuits (Holmes et al., 1997b).
neurons include serotonin, thyrotropin-releasing
hormone, and GABA (Arvidsson et al., 1990).
Medullary thyrotropin-releasing hormone neurons Eliminative functions following spinal cord injury
diffusely project to the ventral horn of the spinal
cord and sympathetic neurons within the inter- Following acute injury to the spinal cord an initial
mediolateral cell column (Appel et al., 1987; period of ‘‘spinal shock’’ occurs, which is charac-
Hirsch and Helke, 1988). When applied intrathec- terized by areflexia and generally lasts for approx-
ally at the level of the L5–L6 spinal cord, thy- imately 24 h in humans (Ditunno et al., 2004).
rotropin-releasing hormone resulted in contraction Disruption of spinal cord tracts proximal to the
of the internal anal sphincter, predominantly lumbosacral cord leads to loss of supraspinal con-
through activation of the pelvic nerve. Although trol over normal eliminative and reproductive be-
sectioning of the hypogastric nerve had no effect haviors. These functions, which reflect coordinated
on thyrotropin-releasing hormone-induced inter- activity of both somatic and autonomic compo-
nal anal sphincter activity, a role for thyrotropin- nents of the nervous system, are important ther-
releasing hormone activation of sympathetic apeutic targets as they represent critical problems
preganglionic neurons should not be excluded for the spinal cord injured population, and have
since the pelvic nerve has been shown to contain been less well studied than locomotion in models
both sympathetic and parasympathetic fibers of spinal cord injury (Anderson, 2004). After the
(Hulsebosch and Coggeshall, 1982). It is suggest- initial post-injury period of spinal shock, the basic
ed that the relative roles of the pelvic and spinal reflexes control these functions independent
hypogastric nerves in internal anal sphincter from supraspinal input. During the recovery phase
contractility differ with changes in activity of rec- from spinal shock (1 day to 12 months), a gradual
tal afferents (Holmes et al., 1995). Also, pudendal return of reflexes may take place as well as the
motor neurons are under spinal descending development of hyperreflexia and spasticity
inhibitory control. Similar to the effect thyrotro- (Ditunno et al., 2004). Interestingly in the rat,
pin-releasing hormone has on the internal anal the initial loss and subsequent recovery of func-
sphincter, high doses of thyrotropin-releasing hor- tions that have autonomic components show a
mone applied to the lumbosacral spinal cord re- similar pattern of recovery as locomotor function
sulted in an increase in firing rate of the external (Holmes et al., 2005), suggesting commonality of
anal sphincter (Holmes et al., 1997b). Thus, thy- underlying recovery mechanisms.
rotropin-releasing hormone appears to be involved In human patients with spinal cord injury,
in maintaining fecal continence. Other studies have coordination of autonomic and somatic defeca-
shown that thyrotropin-releasing hormone plays tion reflexes is lost secondary to disruption of
a critical role in modulation of gastrointestinal supraspinal control pathways. Compressive de-
function (McCann et al., 1989). Thyrotropin- struction of the conus medullaris of the spinal cord
releasing hormone has been shown to increase can destroy the distal segments of the cord where,
367

in humans, the cord defecation reflex is integrated, of micturition, however, is unknown. Recovery
and this almost paralyzes defecation. More fre- of micturition function has been observed to oc-
quently the spinal cord is injured more proximal, cur through amelioration of detrusor–sphinc-
between the conus medullaris and the brain, in ter–dyssynergia without noticeable change of de-
which case the voluntary portion of the defecation trusor hyperreflexia (Mitsui et al., 2003).
act is blocked while the basic spinal cord reflex for We suggest that in addition to these muscles,
defecation is still intact. other pelvic floor muscles, and specifically the
In rat models, in addition to loss of control of bulbospongiosus musculature develops hype-
eliminative and sexual function, the lumbosacral rreflexia and spasticity following spinal cord inju-
reflexes develop hyperreactivity, including in- ry. This change in reflex activity is reminiscent
creased external anal sphincter contractions after of hyperreflexia observed in other segmentally
distention and hyperreflexia of erections in re- mediated reflexes (Bose et al., 2002). Interestingly,
sponse to slight tactile stimuli (Holmes et al., 1998, McKenna et al. (1991) observed simultaneous
2005). This is consistent with the finding in contraction of all pelvic floor musculature (bulb-
humans that removal of supraspinal control leads ospongiosus, ischiocavernosus, external anal
to spasticity and dyssynergia of urethral and anal sphincter, and external urethral sphincter) during
sphincters. In addition to hyperreflexia of the the urethrogenital reflex after spinal cord transec-
external anal sphincter, it is well recognized that tion. This reflex is induced by stimulation of the
hyperreflexia of the detrusor muscle occurs after urethra and these authors suggest that the motor
spinal cord injury, as well as detrusor–sphinc- program is controlled by a spinal pattern genera-
ter–dyssynergia. Detrusor-sphincter-dyssynergia is tor in the absence of descending input and that the
thought to be the main reason for the observed spinal circuitry is responsible for the rhythmic,
increased voiding pressures and decreased voiding coordinated bursting activity of all these muscles.
efficiency, and has been documented in rats (Kruse Certainly, the extensive co-mingling of the dend-
et al., 1993; Pikov et al., 1998; Yoshiyama et al., rites of these motor neurons in the spinal cord
2000; Cheng and de Groat, 2004). Although ex- (as well as their electrotonic and synaptic cou-
ternal urethral sphincter bursting activity can be pling) could be an anatomical substrate for this
mediated by spinal reflex mechanisms, the bursting and other coordinated activities (Coolen et al.,
activity that is seen after spinal cord injury is ab- 2004; Giuliano and Rampin, 2004).
normal, leading to shorter urethral opening times A closer examination of the bulbospongiosus
and this presumably contributes to the inefficient muscle activity after contusion injury of the spinal
voiding and increased voiding pressures (Cheng cord (Nout et al., 2005) in awake behaving rats,
and de Groat, 2004). Also, the amplitude from shows that the loss of descending input produces
external urethral sphincter electromyographic re- an exaggerated contraction pattern in this muscle
cordings has been shown to increase following similar to that seen in the external anal sphincter.
spinal cord injury (Pikov and Wrathall, 2001). This activity was observed during erectile events as
Suppression of external urethral sphincter activity well as during micturition. Using pressure record-
results in improvement of all voiding parameters ings from the corpus spongiosum penis it is pos-
(Kruse et al., 1993; Yoshiyama et al., 2000). In sible to reliably assess both micturition and erectile
addition to dyssynergia, detrusor hyperreflexia has events (Schmidt et al., 2004, 2005). Following
been well characterized in rats following spinal spinal cord damage, increased corpus spongiosum
cord injury (Mitsui et al., 2003) and is thought penis mean pressures and increased duration
to be due to a lack of supraspinal inhibition of micturition as well as of erectile events occur
with or without an increase of afferent signaling. (Figs. 3 and 4). Increased corpus spongiosum penis
Moreover, enlargement of the bladder may result pressures indicate bulbospongiosus muscle spasti-
in plasticity of afferents, further contributing to city, since pressure within the bulb of the corpus
this hyperreflexia (De Groat et al., 1998). The spongiosum penis is directly dependent on the
significance of this syndrome in the recovery activity in the surrounding bulbospongiosus
368

Fig. 3. Pressures recorded from the corpus spongiosum penis in


conscious awake rats during micturition. A moderate spinal
cord contusion [12.5 g cm weight drop Multicenter Animal
Spinal Cord Injury Study (MASCIS) injury] results in a signif-
icant trend toward the development of slightly higher pressures
Fig. 4. Pressures recorded from the corpus spongiosum penis in
and prolonged duration of these events. This is likely due to
conscious awake rats during erectile events. A moderate spinal
development of bulbospongiosus muscle spasticity (time scale: a
cord contusion (12.5 g cm MASCIS injury) results in the de-
25 s interval is shown). (Adapted from Nout et al., 2005.)
velopment of slightly higher pressures and prolonged duration
of the peaks occurring during these events. This is likely due to
development of bulbospongiosus muscle spasticity (time scale: a
musculature (Schmidt et al., 1995). In addition to
25 sc interval is shown). (Adapted from Nout et al., 2005.)
changes in corpus spongiosum penis pressures, the
total number of erectile events per 24-h period was
decreased after injury. We hypothesize that the erections. In addition, during ex copula reflex test-
number of partial erectile events decreased due to ing in these same preparations, there was a short-
disinhibition of supraspinal input allowing almost ened latency to induction of erections consistent
all initiated erectile events to develop into full with previous reports (Hart, 1968; Schmidt et al.,
369

1999; Hubscher and Johnson, 2000; Holmes et al., calcitonin gene-related peptide immunoreactive
2001, 2005). terminals appears to increase (Beattie et al.,
One of the possible explanations of our finding 2000), suggesting that the effect of such input
of a reduced number of erectile events following might be even more functionally significant after
spinal cord injury is the assumption that erections spinal cord injury. Interestingly, the synaptic in-
occur secondary to genital grooming, which is re- puts to Onuf’s nucleus in the same studies showed
duced markedly following spinal cord injury. This similar synaptic rearrangements, i.e., reduced size
could then account for a reduction of the number of terminals apposed to Onuf’s motor neurons af-
of erectile events following spinal cord injury. Also, ter spinal cord injury (Beattie et al., 1993), and an
ascending sensory pathways from the male genita- increase in their terminal coverage by GABA-
lia, that are bilaterally located within the dorsal immunoreactive terminals (Beattie et al., 2000).
quadrant at the midthoracic level of the spinal The plasticity that is seen in this area of the cord
cord, are damaged in this model of spinal cord in- forms an important target for potential therapeu-
jury (Hubscher and Johnson, 1999). Another pos- tic interference. For example, it has been shown
sible explanation for a reduction of the number of that interference in the development of the prima-
erectile events following spinal cord injury, would ry afferent plasticity affected the development of
be through disruption of descending excitatory autonomic dysreflexia (Weaver et al., 2001) and
mechanisms in addition to disruption of inhibitory detrusor–sphincter–dyssynergia (Seki et al., 2004).
tracts. Although most studies have concluded that Recently, Cameron et al. (2004) presented a strat-
the descending tracts modulating sexual reflexes egy for modulation of post-traumatic spinal plas-
are inhibitory in nature, recent studies have dem- ticity in both sacral afferents and propriospinal
onstrated the significance of descending excitatory projection neurons via targeted gene therapy in a
tracts particularly from the paraventricular nucleus model of autonomic dysreflexia.
of the hypothalamus (Giuliano and Rampin, 2000).
In addition to loss of descending inhibitory con-
trol of spinal reflex pathways, plasticity within the Conclusions
spinal cord is thought to contribute to recovery
but also to development of exaggerated spinal re- Defecation, urination, and male sexual function are
flex responses. For example, Weaver and her col- compromised after spinal cord injury. The resulting
leagues have shown that sprouting of dorsal root dysfunction represents a combination of recovery
afferents containing calcitonin gene-related pep- of reflexes and plasticity that produce hyperactive
tide may contribute to the development of auto- reflexes. Treatments aimed at ameliorating second-
nomic dysreflexia after severe spinal cord injury ary injury and enhancing sprouting or regeneration
(Weaver et al., 2001). Similarly, calcitonin gene- may have positive effects on lumbosacral auto-
related peptide fibers contributing to the parasym- nomic function, but more information regarding
pathetic components of bladder, bowel, and sexual the normal and pathophysiological eliminative and
reflex function have also been demonstrated to sexual functions is needed to plan treatment strat-
sprout after spinal cord injury in cats (Beattie egies. The strategies should include pharmacolog-
et al., 2000). Spinal cord injury, in this species, ical therapies guided by experimental studies. The
produces a chronic (measured at 6 weeks) dener- availability of several useful models of spinal cord
vation of identified parasympathetic preganglionic injury in rodents that mimic many of the auto-
neurons (Beattie et al., 1993); the proportion of nomic functions of human spinal cord injury
the somatic and proximal dendritic membrane of should encourage an increase in the laboratory
these efferent neurons contacted by synaptic ter- work devoted to this relatively neglected area of
minals is significantly reduced, the size of the ter- experimental spinal cord injury. This should be
minals is smaller, and glial coverage is reciprocally especially true since spinal cord-injured people
increased. In additional studies, an increase in the consider recovery of these functions to be of high-
proportion of the membrane area contacted by est priority (Anderson, 2004).
370

Acknowledgments Bremer, R.E., Barber, M.D., Coates, K.W., Dolber, P.C. and
Thor, K.B. (2003) Innervation of the levator ani and co-
We would like to thank Dr. Gregory M. Holmes ccygeus muscles of the female rat. Anat. Rec., 275: 1031–1041.
Cameron, A.A., Smith, G.M., Randall, D.C. and Rabchevsky,
for his helpful comments on this manuscript, and A.G. (2004) Genetic manipulation of afferent fiber sprouting
Mr. John Komon for preparing the figures. Sup- following spinal cord injury modulates the severity of auto-
ported by NIH grant NS-31193. nomic dysreflexia. Soc. Neurosci. Abstr., Program No. 459.15.
Cheng, C.L. and de Groat, W.C. (2004) The role of capsaicin-
sensitive afferent fibers in the lower urinary tract dysfunction
induced by chronic spinal cord injury in rats. Exp. Neurol.,
187: 445–454.
References Collins, W.F.I., Alspaugh, W.H. and Erichsen, J.T. (1992)
Morphology of external urethral sphincter motoneurons and
Anderson, K.D. (2004) Targeting recovery: priorities of the localization of putative sphincter interneurons in the rat spi-
spinal cord injured population. J. Neurotrauma, 21: nal cord. Soc. Neurosci. Abstr., 18: 1050.
1371–1383. Comarr, A.E. (1970) Sexual function among patients with spi-
Appel, N.M., Wessendorf, W. and Elde, R.P. (1987) Thryrotro- nal cord injury. Urol. Int., 25: 134–168.
pin-releasing hormone in spinal cord: coexistence with se- Coolen, L.M., Allard, J., Truitt, W.A. and McKenna, K.E. (2004)
rotonin and with substance P in fibers and terminals Central regulation of ejaculation. Physiol. Beh., 83: 203–215.
apposing identified preganglionic sympathetic neurons. Brain Cosman, B.C., Stone, J.M. and Perkash, I. (1991) Gastrointes-
Res., 415: 137–143. tinal complications of chronic spinal cord injury. J. Am.
Arvidsson, U., Cullheim, S., Ulfhake, B., Bennett, G.W., Fone, Paraplegia Soc., 14: 175–181.
K.C., Cuello, A.C., Verhofstad, A.A.J., Visser, T.J. and De Groat, W.C., Araki, I., Vizzard, M.A., Yoshiyama, M.,
Hokfelt, T. (1990) 5-Hydroxytryptamine, substance P, and Yoshimura, M., Sugaya, K., Tai, C. and Roppolo, J.R.
thyrotropin releasing hormone in the adult cat spinal cord (1998) Developmental and injury induced plasticity in the
segment L7: immunohistochemical and chemical studies. micturition reflex pathway. Behav. Brain Res., 92: 127–140.
Synapse, 6: 237–270. De Groat, W.C., Kawatani, M., Hisamitsu, T., Cheng, C.L.,
Baron, R. and Janig, W. (1991) Afferent and sympathetic neu- Ma, C.P., Thor, K., Steers, W. and Roppolo, J.R. (1990)
rons projecting into lumbar visceral nerves of the male rat. J. Mechanisms underlying the recovery of urinary bladder
Comp. Neurol., 314: 429–436. function following spinal cord injury. J. Autonom. Nerv.
Basu, S., Aballa, T.C., Ferrell, S.M., Lynne, C.M. and Brack- Sys., 30(Suppl): S71–S77.
ett, N.L. (2004) Inflammatory cytokine concentrations are Ding, Y.Q., Takada, M., Tokuno, H. and Mizuno, N. (1995)
elevated in seminal plasma of men with spinal cord injuries. J. Direct projections from the dorsolateral pontine tegmentum
Androl., 25: 250–254. to pudendal motoneurons innervating the external urethral
Beattie, M.S., Holmes, G.M., Hermann, G.E., Rogers, R.C. and sphincter muscle in the rat. J. Comp. Neurol., 357: 318–330.
Bresnahan, J.C. (1996) Pudendal motoneuron function in in- Ditunno, J.F., Little, J.W., Tessler, A. and Burns, A.S. (2004)
tact and CNS injured rats: alterations following brainstem le- Spinal shock revisited: a four-phase model. Spinal Cord, 42:
sions and spinal transection. Soc. Neurosci. Abstr., 22: 722.4. 383–395.
Beattie, M.S., Leedy, M.G. and Bresnahan, J.C. (1993) Evi- Dubrovsky, B., Martinez-Gomez, M. and Pacheco, P. (1985) Spi-
dence for alterations of synaptic inputs to sacral spinal reflex nal control of pelvic floor muscles. Exp. Neurol., 88: 277–287.
circuits after spinal cord transection in the rat. Exp. Neurol., Frenckner, B. (1975) Function of the anal sphincters in spinal
123: 35–50. man. Gut, 16: 638–644.
Beattie, M.S., Li, Q. and Bresnahan, J.C. (2000) Cell death and Giuliano, F. and Rampin, O. (2000) Central neural regulation
plasticity after experimental spinal cord injury. In: Seil F.J. of penile erection. Neurosci. Biobehav. Rev., 24: 517–533.
(Ed.) Progress in Brain Research, Vol. 128. Elsevier, Am- Giuliano, F. and Rampin, O. (2004) Neural control of erection.
sterdam, pp. 9–21. Physiol. Behav., 83: 189–201.
Beattie, M.S., Li, Q., Leedy, M.G. and Bresnahan, J.C. (1990) Gonella, J., Bouvier, M. and Blanquet, F. (1987) Extrinsic
Motoneurons innervating the external anal and urethral nervous control of motility of small and large intestined and
sphincters of the female cat have different patterns of den- related sphicters. Physiol. Rev., 67: 902–961.
dritic arborization. Neurosci. Lett., 111: 69–74. Guyton, A.C. and Hall, J.E. (2000) Textbook of Medical Phys-
Bose, P., Parmer, R. and Thompson, F.J. (2002) Velocity- iology (10th ed.). W.B. Saunders Co., Philadelphia, PA.
dependent ankle torque in rats after contusion injury of the Hart, B.L. (1968) Sexual reflexes and mating behavior in the
midthoracic spinal cord: time course. J. Neurotrauma, 19: male rat. J. Comp. Physiol. Psychol., 65: 453–460.
1231–1249. Hermann, G.E., Bresnahan, J.C., Holmes, G.M., Rogers, R.C.
Breedlove, S.M. and Arnold, A.P. (1980) Hormone accumula- and Beattie, M.S. (1998) Descending projections from the
tion in a sexually dimorphic motor nucleus of the rat spinal nucleus raphe obscurus to pudendal motoneurons in the male
cord. Science, 210: 564–566. rat. J. Comp. Neurol., 397: 458–474.
371

Hermann, G.E., Holmes, G.M., Rogers, R.C., Beattie, M.S. Katagiri, T., Gibson, S.J., Su, H.C. and Polak, J.M. (1986) Com-
and Bresnahan, J.C. (2003) Descending spinal projections position and central projections of the pudendal nerve in the rat
from the rostral gigantocellular reticular nuclei complex. J. investigated by combined peptide immunocytochemistry and
Comp. Neurol., 455: 210–221. retrograde fluorescent labelling. Brain Res., 372: 313–322.
Higgins Jr, G.E. (1979) Sexual responses in spinal cord injured Krier, J. (1985) Discharge patterns of pudendal efferent fibers
adults: a review of the literature. Arch. Sex. Behav., 8: 173–196. innervating the external anal sphincter of the cat. J. Physiol.,
Hirsch, M.D. and Helke, C.J. (1988) Bulbospinal thryrotrophin 368: 471–480.
releasing hormone projections to the intermediolateral cell Kruse, M.N., Belton, A.L. and De Groat, W.C. (1993) Changes
column: a double fluorescence immunohistochemical retro- in bladder and external urethral sphincter function after spi-
grade tracing study in the rat. Neuroscience, 25: 625–637. nal cord injury in the rat. Am. J. Physiol., 264: R1157–R1163.
Holmes, G.M., Bresnahan, J.C., Rogers, R.C., Hermann, G.E. Li, Q., Beattie, M.S. and Bresnahan, J.C. (1995) Onuf’s nucleus
and Beattie, M.S. (1994) Differential characteristics of rat (ON) motoneurons (MNs) have more GABA-ergic synapses
dorsomedial motoneurons. Soc. Neurosci. Abstr., 20: 791. than other somatic MNs: a quantitative immunocytochem-
Holmes, G.M., Hermann, G.E., Rogers, R.C., Bresnahan, J.C. ical study in the cat. Soc. Neurosci. Abstr., 21: 1201.
and Beattie, M.S. (2002) Dissociation of the effects of nucleus Maggi, C.A., Giuliani, S., Santicioli, P., Patacchini, R. and Meli,
raphe obscurus or rostral ventrolateral medulla lesions A. (1988) Neural pathways and pharmacological modulation
on eliminatory and sexual reflexes. Physiol. Behav., 75: 49–55. of defecation reflex in rats. Gen. Pharmacol., 19: 517–523.
Holmes, G.M., Martau, J.M., Hermann, G.E., Rogers, R.C., Marson, L., List, M.S. and McKenna, K.E. (1992) Lesions of
Bresnahan, J.C. and Beattie, M.S. (1997a) Nucleus raphe the nucleus paragigantocellularis alter ex copula penile re-
obscurus (nRO) regulation of anorectal motility in rats. Brain flexes. Brain Res., 592: 187–192.
Res., 759: 197–204. Marson, L. and McKenna, K.E. (1990) The identification of a
Holmes, G.M., Rogers, R.C., Bresnahan, J.C. and Beattie, brainstem site controlling spinal sexual reflexes in male rats.
M.S. (1995) Thyrotropin-releasing hormone (TRH) and CNS Brain Res., 515: 303–308.
regulation of anorectal motility in the rat. J. Auton. Nerv. Marson, L. and McKenna, K.E. (1996) CNS cell groups in-
Syst., 56: 8–14. volved in the control of the ischiocavernosus and bulb-
Holmes, G.M., Rogers, R.C., Bresnahan, J.C. and Beattie, ospongiosus muscles: a transneuronal tracing study using
M.S. (1997b) Differential effects of intrathecal thyrotropin- pseudorabies virus. J. Comp. Neurol., 374: 161–179.
releasing hormone (TRH) on perineal reflexes in male rats. Matsumoto, A., Arnold, A.P., Zampighi, G.A. and Micevych,
Physiol. Behav., 61: 57–63. P.E. (1988) Androgenic regulation of gap junctions between
Holmes, G.M., Rogers, R.C., Bresnahan, J.C. and Beattie, motoneurons in the male rat. J. Neurosci., 8: 4177–4183.
M.S. (1998) External anal sphincter hyperreflexia following McCann, M.J., Hermann, G.E. and Rogers, R.C. (1989) Nu-
spinal transection in the rat. J. Neurotrauma, 15: 451–457. cleus raphe obscurus (nRO) influences vagal control of gas-
Holmes, G.M., Stephens, R.L., Bresnahan, J.C. and Beattie, M.S. tric motility in rats. Brain Res., 486: 181–184.
(2001) Intrathecal methiothepin and thyrotropin-releasing hor- McKenna, K.E. and Nadelhaft, I. (1986) The organization of
mone effects on penile erection. Physiol. Behav., 73: 59–64. the pudendal nerve in the male and female rat. J. Comp.
Holmes, G.M., Van Meter, M.T., Beattie, M.S. and Bresnahan, Neurol., 248: 532–549.
J.C. (2005) Serotonergic fiber sprouting to external anal McKenna, K.E., Sookja, K.C. and McVary, K.T. (1991) A
sphincter motoneurons after spinal cord contusion. Exp. Ne- model for the study of sexual function in anesthetized male
urol., 193: 29–42. and female rats. Am. J. Physiol., 261(Regul. Integr. Comp.
Holstege, G. and Tan, J. (1987) Supraspinal control of moto- Physiol. 30): R1276–R1285.
neurons innervating the striated muscles of the pelvic floor Mitsui, T., Kakizaki, H., Tanaka, H., Shibata, T., Matsuoka, I.
including urethral and anal sphincters in the cat. Brain, and Koyanagi, T. (2003) Immortalized neural stem cells
110(Part 5): 1323–1344. transplanted into the injured spinal cord promote recovery of
Hubscher, C.H. and Johnson, R.D. (1999) Effects of acute and voiding function in the rat. J. Urol., 170: 1421–1425.
chronic midthoracic spinal cord injury on neural circuits for Miyata, K., Kamato, T., Nishida, A., Ito, H., Yuki, H.,
male sexual function. I. Ascending pathways. J. Neuro- Yamano, M., Tsutsumi, R., Katsuyama, Y. and Honda, K.
physiol., 82: 1381–1389. (1992) Role of the serotonin3 receptor in stress-induced def-
Hubscher, C.H. and Johnson, R.D. (2000) Effects of acute and ecation. J. Pharmacol. Exp. Ther., 261: 297–303.
chronic midthoracic spinal cord injury on neural circuits for Monaghan, E.P. and Breedlove, S.M. (1991) Brain sites pro-
male sexual function. II. Descending pathways. J. Neuro- jecting to the spinal nucleus of the bulbocavernosus. J.
physiol., 83: 2508–2518. Comp. Neurol., 307: 370–374.
Hulsebosch, C.E. and Coggeshall, R.E. (1982) An analysis of Nadelhaft, I. and Booth, A.M. (1984) The location and mor-
the axon populations in the nerves to the pelvic viscera in the phology of preganglionic neurons and the distribution of
rat. J. Comp. Neurol., 211: 1–10. visceral afferents from the rat pelvic nerve: a horseradish
Jänig, W. and McLachlan, E.M. (1987) Organization of lumbar peroxidase study. J. Comp. Neurol., 226: 238–245.
spinal outflow to distal colon and pelvic organs. Physiol. Nadelhaft, I. and Vera, P.L. (2001) Separate urinary bladder
Rev., 67: 1332–1404. and external urethral sphincter neurons in the central nervous
372

system of the rat: simultaneous labeling with wo immunohisto- spongiosum penis (CSP) pressure. Society for Neuroscience,
chemically distinguishable pseudorabies viruses. Brain Res., Online., Program No. 922.16. Abstract Viewer/Itinerary
903: 33–44. Planner. Washington, DC.
Nout, Y.S., Schmidt, M.H., Tovar, C.A., Culp, E., Beattie, Schmidt, M.H., Nout, Y.S., Culp, E., Tovar, C.A., Beattie,
M.S. and Bresnahan, J.C. (2005) Telemetric monitoring of M.S. and Bresnahan, J.C. (2005) A new technique for the
corpus spongiosum penis pressures in conscious rats for as- simultaneous recording of micturition and penile erections in
sessment of micturition and sexual function following spinal freely behaving rats: telemetric monitoring of corpus
cord contusion injury. J. Neurotrauma, 22(4): 429–444. spongiosum penis (CSP) pressure, in preparation.
Pacheco, P., Camacho, M.A., Garcia, L.I., Hernandez, M.E., Schmidt, M.H., Sakai, K., Valatx, J.L. and Jouvet, M. (1999)
Carrillo, P. and Manzo, J. (1997) Electrophysiological evi- The effects of spinal or mesencephalic transections on sleep-
dence for the nomenclature of the pudendal nerve and sacral related erections and ex-copula penile reflexes in the rat.
plexus in the male rat. Brain Res., 763: 202–208. Sleep, 22: 409–418.
Paxinos, G. (1995) The Rat Nervous System (2nd ed.). Aca- Schmidt, M.H., Valatx, J.L., Sakai, K., Debilly, G. and Jouvet,
demic Press Inc., San Diego, CA. M. (1995) Corpus spongiosum penis pressure and perineal
Pedersen, E. (1983) Regulation of bladder and colon-rectum in muscle activity during reflexive erections in the rat. Am. J.
patients with spinal lesions. J. Auton. Nerv. Syst., 7: 329–338. Physiol., 269: R904–R913.
Peshori, K.R., Erichsen, J.T. and Collins 3rd, W.F. (1995) Dif- Schroder, H.D. (1980) Organization of the motor neurons in-
ferences in the connectivity of rat pudendal motor nuclei as nervating the pelvic muscles of the male rat. J. Comp. Ne-
revealed by retrograde transneuronal transport of wheat urol., 192: 567–587.
germ agglutinin. J. Comp. Neurol., 353: 119–128. Schuster, M.M. (1968) Motor action of rectum and anal
Pikov, V., Gillis, R.A., Jasmin, L. and Wrathall, J.R. (1998) sphincters in continence and defecation. In: Code C.F. (Ed.)
Assessment of lower urinary tract functional deficit in rats Handbook of Physiology, Alimentary Canal Motility, Sec-
with contusive spinal cord injury. J. Neurotrauma, 15: tion 6, Vol. IV. Chapter 103. American Physiological Society,
375–386. Washington, DC, pp. 2121–2146.
Pikov, V. and Wrathall, J.R. (2001) Coordination of the blad- Seki, S., Sasaki, K., Igawa, Y., Nishizawa, O., Chancellor,
der detrusor and the external urethral sphincter in a rat M.B., DeGroat, W.C. and Yoshimura, N. (2004) Suppres-
model of spinal cord injury: effect of injury severity. J. Ne- sion of detrusor-sphincter dyssynergia by immunoneutrali-
urosci., 21: 559–569. zation of nerve growth factor in lumbosacral spinal cord in
Purinton, P.T., Fletcher, T.F. and Bradley, W.E. (1973) Gross spinal cord injured rats. J. Urol., 171: 478–482.
and light microscopic features of the pelvic plexus in the rat. Shen, P., Arnold, A.P. and Micevych, P.E. (1990) Supraspinal
Anat. Rec., 175: 697–705. projections to the ventromedial lumbar spinal cord in adult
Ramirez-Leon, V. and Ulfake, B. (1993) GABA-like immuno- male rats. J. Comp. Neurol., 300: 263–272.
reactive innervation and dendro-dendritic eontacts in the Tang, Y., Rampin, O., Giuliano, F. and Ugolini, G. (1999)
ventrolateral dendritic bundle in the cat S1 spinal segment: Spinal and brain circuits to motoneurons of the bulb-
and electron microscopic study. Exp. Brain Res., 97: 1–12. ospongiosus muscle: retrograde transneuronal tracing with
Ramirez-Leon, V., Ulfake, B., Arvidsson, U., Verhofstad, rabies virus. J. Comp. Neurol., 414: 167–192.
A.A., Visser, T.J. and Hokfelt, T. (1994) Serotonergic, Vera, P.L. and Nadelhaft, I. (1992) Afferent and sympathetic
peptidergic, and GABAergic innervation of the ventrolater- innervation of the dome and the base of the urinary bladder
al and dorsolateral motor nuclei in the cat S1/S2 segments: of the female rat. Brain Res. Bull., 29: 651–658.
an immunofluorescence study. J. Chem. Neuroanat., 7: Vizzard, M.A., Brisson, M. and De Groat, W.C. (2000)
87–103. Transneuronal labeling of neurons in the adult rat central
Rexed, B. (1954) A cytoarchitectonic atlas of the spinal cord in nervous system following inoculation of pseudorabies virus
the cat. J. Comp. Neurol., 100: 297–379. into the colon. Cell Tissue Res., 299: 9–26.
Roney, K.J., Scheibel, A.B. and Shaw, G.L. (1979) Dendritic Weaver, L.C., Verghese, P., Bruce, J.C., Fehlings, M.G., Krenz,
bundles: survey of anatomical experiments and physiological N.R. and Marsh, C.R. (2001) Autonomic dysreflexia and
theories. Brain Res. Rev., 1: 225–271. primary afferent sprouting after clip-compression injury of
Sachs, B.D. and Leipheimer, R.E. (1988) Rapid effect of testos- the rats spinal cord. J. Neurotrauma, 18: 1107–1119.
terone on striated muscle activity in rats. Neuroendocrino- Yamanouchi, K. and Kakeyama, M. (1992) Effect of medullary
logy, 48: 453–458. raphe lesions on sexual behavior in male rats with or without
Sasaki, M. (1994) Morphological analysis of external urethral treatments of p-chlorophenylalanine. Physiol. Behav., 51:
and external anal sphincter motoneurones of cat. J. Comp. 575–579.
Neurol., 349: 269–287. Yoshiyama, M., De Groat, W.C. and Fraser, M.O. (2000) In-
Schmidt, M.H., Nout, Y.S., Culp, E., Tovar, C.A., Beattie, fluences of external urethral sphincter relaxation induced by
M.S. and Bresnahan, J.C. (2004) A new technique for the alpha-bungarotoxin, a neuromuscular junction blocking
simultaneous recording of micturition and penile erections in agent, on voiding dysfunction in the rat with spinal cord
freely behaving rats: telemetric monitoring of corpus injury. Urology, 55: 956–960.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 25

Upper and lower gastrointestinal motor and sensory


dysfunction after human spinal cord injury

Paul Enck1,, Irmgard Greving2, Sibylle Klosterhalfen3 and Beate Wietek4

1
Department of Psychosomatic Medicine, University Hospitals Tuingen, Schaffhausenstr 113, 72072 Tubingen, Germany
2
Department of Internal Medicine, Elisabeth Hospital, Gelsenkirchen, Germany
3
Institute of Medical Psychology, University Hospitals Dusseldorf, Germany
4
Department of Radiology, University Hospitals Tubingen, Germany

Abstract: This chapter describes the results of investigations of the upper and lower gastrointestinal tract in
subjects with complete and incomplete spinal cord injury. In one study, gastric emptying was investigated
and found delayed. The delay was tentatively attributed to a colo-gastric inhibitory reflex triggered by
inappropriate colonic emptying. In another study, anorectal motor and sensory functions were measured.
Decreased tone of the internal anal sphincter, exaggerated recto-anal reflexes following rectal distension
and spontaneous high-amplitude rectal contractions at low distension volumes were among the findings of
the study. Some of the subjects, classified as having a complete injury according to usual clinical criteria
(American Spinal Injury Association, ASIA), reported sensation of distension of the rectum. This raises the
issue of the need for better methods for the clinical assessment of sensory transmission in the spinal cord.
Promising results obtained with functional magnetic resonance imaging of the brain during rectal stim-
ulation in a small group of paraplegics, with complete injuries by ASIA criteria, showed evidence of
activation of several brain regions.

Introduction innervation is unknown. The purpose of this


chapter is to review data from functional investi-
People with traumatic spinal cord lesions often gations of the upper and lower gastrointestinal
suffer from persistent upper and lower gastroin- tract in spinal cord-injured people with complete
testinal complaints such as dyspepsia, nausea, or incomplete injury. Both motor and sensory
slowed defecation, reflex and uncontrolled rectal dysfunctions are addressed.
evacuation, and chronic abdominal pain (Glickman
and Kamm, 1996; Kirk et al., 1997; Menter et al.,
1997; De Looze et al., 1998a, b; Han et al., 1998).
Gastric emptying
Symptoms tend to increase with time and peak at
around 5 years post-trauma (Stone et al., 1990),
People with incomplete or complete spinal cord
but the reasons for these long-term changes are
injury often complain of upper gastrointestinal
unclear. The effect of disconnection of the spinal
symptoms such as heartburn, bloating, and post-
cord from supraspinal centers on autonomic (para-
prandial fullness (De Vault et al., 1996; Kao et al.,
sympathetic/sympathetic) and intrinsic (enteric)
1999; Singh and Triadafilopoulos, 2000; Chen
et al., 2004), but these symptoms have rarely been
Corresponding author. Tel.: +49 7071 9387374; investigated in a systematic fashion. Clinically,
Fax.: +49 7071 9387379; E-mail: paul.enck@uni-tuebingen.de they are often regarded as secondary to immobility

DOI: 10.1016/S0079-6123(05)52025-9 373


374

and bed rest, to inadequate eating habits, or as a Delayed gastric emptying seems to be quite
consequence of inappropriate colonic emptying common in people with complete spinal cord in-
and constipation, i.e. secondary to a colo-gastric jury and may explain the occurrence of upper gas-
inhibitory reflex (Youle and Read, 1984). While trointestinal symptoms in these people in everyday
this may be the case with lower spinal cord inju- life (Fealey et al., 1984). However, the data are
ries, a higher cord lesion may also directly affect conflicting, since some authors found no delay in
visceral motor functions such as gastric emptying, gastric emptying in people with spinal cord injury
via reflex spinal pathways. or concluded that, if a delay occurs early after spi-
The following study of gastric emptying was done nal trauma, it diminishes over time (Mollen
using 15 subjects with complete injury of the spinal et al., 1999). It may be argued that the delays are
cord (according to ASIA criteria) (Ditunno et al., secondary to immobility and bed rest, or to inad-
1994), 5 of whom had lesions in the cervical (C) equate eating habits; more likely, however, they are
segments C1 to C7 and 10 who had lesions in the a consequence of inappropriate colonic emptying
thoracic (TH) and lumbar (L) regions (TH1 to L5). and constipation, i.e. are secondary to a colo-gas-
The ratio of males to females investigated was 14:1. tric inhibitory reflex. Delayed gastric emptying has
Studies were undertaken between 3 months and 28 been shown to occur following experimental rectal
years following trauma. All subjects completed a balloon distension in healthy volunteers (Youle
questionnaire related to gastrointestinal symptoms. and Read, 1984; Mollen et al., 1999), and also in
Gastric emptying was measured by means of the 13C patients with chronic constipation (Coremans
(carbon 13) breath test: in short, a standardized et al., 2004). Final proof of direct or indirect
mixed test meal labeled with [13C]octanoic acid for effects of spinal cord injury on upper gastrointes-
measurement of emptying of solids (Zahn et al., tinal transit remains to be obtained in the future.
2003), and a standard test drink, labeled with
[13C]sodium acetate for emptying of liquids (Braden Anorectal motor and sensory function
et al., 1995), were provided on two separate days in
randomized sequence. Breath samples were collect- People with complete or incomplete spinal cord
ed at baseline, and every 10 min over 2 h for liquid lesions due to trauma often also suffer from def-
emptying and every 15 min for 4 h for emptying of ecation disorders (Longo et al., 1989; Enck et al.,
solids, and analyzed by infrared spectrometry (IRIS, 1991; Bruninga and Camilleri, 1997; De Looze
Wagner Analysentechnik, Bremen, Germany) for et al., 1998a). As early as 1948 (Gaston, 1948), this
the 12C:13C ratio. The time series of each data set problem was attributed to loss of motor and/or
permits the relevant parameters of gastric emptying sensory control of the anorectum and/or pelvic
to be calculated, i.e. half-time emptying (T/2) and floor. In studies of anorectal motor function after
the lag-phase (t-lag) that measures the time between traumatic complete (MacDonagh et al., 1992) or
the end of the meal and the first bolus leaving the incomplete (Sun et al., 1990) spinal injury, various
stomach (Chew et al., 2003). continence mechanisms have been shown to be
All subjects reported relevant upper gastrointes- disturbed. Patients with spinal tumors (May, 1991)
tinal symptoms such as heartburn, post-prandial and other causes of denervation (Wheatley et al.,
fullness, nausea, and bloating. Compared to 1977) also have defecation disorders, resulting in
the normal values of the test (T/2o75 min, either constipation or incontinence or both. This
t-lago50 min for solid emptying; T/2o60 min, problem may relate to motor and sensory deficits
t-lago20 min for liquid emptying), subjects with in the somatic and visceral compartments of the
high spinal cord lesions had significantly delayed nervous system. Colonic transit time is usually de-
gastric emptying for solids (T/2 ¼ 313 min, layed in people with spinal cord injury (Devroede
t-lag ¼ 167 min) but not for liquids, whereas sub- et al., 1979; Glick et al., 1984; Beuret-Blanquart
jects with lower spinal cord lesions had significant et al., 1990; Keshavarzian et al., 1995), but since
delay of both solid (T/2 ¼ 153, t-lag ¼ 109) and transit in upper gastrointestinal compartments of-
liquid emptying (T/2 ¼ 58, t-lag ¼ 36). ten is also abnormal (Fealey et al., 1984; see also
375

the preceding section), constipation may be either Sun, 1992; Rao et al., 2002) was routinely per-
of the slow transit-type or of the ‘‘outlet obstruc- formed in left-lateral position after a conventional
tion’’-type. Incontinence, if present, is mostly at- bowel preparation (saline or sorbid enema). The
tributable to the loss of visceral sensation on the following parameters were assessed (Tables 2 and
one hand, and to the loss of voluntary control over 3) — anal resting pressure, representing the func-
the external anal sphincter on the other (Sun et al., tion of the smooth muscle of the internal anal
1995; De Looze et al., 1998b). In the case of a low sphincter; squeeze pressure, the increment above
spinal cord lesion, the combination of intact sen- resting pressure due to voluntary contraction of
sation and voluntary control of the external anal the external anal sphincter; reflex relaxation of the
sphincter suggests a lesion of the conus medullaris internal anal sphincter (%) during balloon disten-
or cauda equina (MacDonagh et al., 1992). In sion of the rectum 10 cm above the anal canal
people with high spinal cord lesions, loss of vol- (called the recto-anal inhibitory reflex, RAIR);
untary contraction of the external anal sphincter, reflex contraction of the external anal sphincter
blunted or absent rectal sensation and exaggerated during a sudden abdominal pressure increase
reflex activity of the sphincter, without inconti- produced by coughing, as an indicator of stress
nence, are often seen. In these people, the residual continence (both anal and abdominal pressures are
pressure, i.e. the remaining pressure during sphinc- presented); and paradoxical contraction of pelvic
ter relaxation induced by rectal distension, is sig- floor muscles, especially of the external anal
nificantly higher than in people with a low spinal sphincter, with attempts to strain for defecation
cord injury (Read and Sun, 1992). No data are (% occurrence). If spontaneous expulsion (% oc-
available on the short-term recovery of anorectal currence) of the distending balloon was observed,
functions following trauma, except after spinal the respective balloon volume (ml) was noted. If
surgery (Sun et al., 1995). Furthermore, the rela- the subject reported sensation of balloon disten-
tionship between clinical data and anorectal func- sion, the pressure at which the sensation appeared
tions has not been determined. was noted (sensation threshold) and the procedure
We conducted studies to establish a set of criteria was repeated at least once to ensure reproducibil-
that would best predict short-term recovery of ity of the sensation. If, as the rectal balloon was
anorectal functions, permitting selection of patients inflated, the subject experienced pain, the pressure
for therapy programs such as biofeedback training. at which this happened was noted (pain threshold).
Thirty-two subjects with spinal cord injury were Data from healthy subjects served as controls for
recruited from the Spinal Trauma Unit of the the manometry measurements.
Department of Surgery at the University Hospital Most anorectal parameters analyzed in the cord-
‘‘Bergmannsheil’’, Bochum, Germany. A mini- injured subjects differed significantly from those of
mum of 4 days elapsed between trauma and the the control subjects (Table 2). In addition, spon-
clinical functional investigations to allow recovery taneous defecation of the rectal balloon, never ob-
from traumatic inflammation (Table 1). Subjects served in the healthy subjects, was initially present
underwent assessment of completeness of spinal in nine subjects with complete, and one subject
injury by clinical signs (ASIA criteria; Ditunno with incomplete lesions, usually preceded by giant
et al., 1994), of clinical symptoms of incontinence rectal contractions. These responses were inde-
(urinary, fecal), of constipation or diarrhea, and of pendent of the spinal level of the lesions. Signif-
the necessity for medication, compared to pre- icant changes in anorectal function, as assessed by
trauma history. Fifteen of the subjects had com- manometry, were never observed in the cord-in-
plete injury and the remaining 17 had incomplete jured subjects investigated two or three times dur-
injury. The subjects had anorectal manometry that ing the course of disease (Table 3), and no patient
included visceral (rectal) sensitivity testing (see reported a significant clinical change of symptoms,
below) and this testing was repeated after an either with incontinence or with constipation.
average of 64 days (n ¼ 29) and again after 140 People with traumatic spinal cord lesions
days (n ¼ 20). Anorectal manometry (Read and usually have disturbances of voluntary anorectal
376

Table 1. Patient characteristics and clinical investigations performed

No Init Age Sex C/IC Level Invest Interv Neuro UI FI

1 RV 20 1 IC T5 2 5 Y N N
2 MW 27 1 IC L3 3 33 Y Y Y
3 RS 23 2 IC T12 3 6 N Y Y
4 HDS 41 2 C T11 3 14 N Y Y
5 JS 38 2 IC C6 3 6 N Y Y
6 PP 25 2 IC L2 1 17 N Y Y
7 NM 17 1 C T7 3 8 Y Y Y
8 HJL 48 2 C C4 3 46 N Y Y
9 JH 32 1 IC T12 2 17 Y Y Y
10 EH 65 1 IC C6 2 22 Y Y Y
11 WE 37 2 IC L1 2 67 N Y Y
12 MBO 21 2 C C6 3 25 N Y Y
13 OB 18 2 C C4 2 32 N Y Y
14 RC 31 2 IC T6 1 42 N Y Y
15 RG 53 1 IC L1 3 9 Y Y Y
16 BP 16 2 IC C5 3 23 N Y Y
17 CT 24 2 C T8 3 12 N Y Y
18 WK 58 2 IC T4 3 74 N N N
19 MH 20 2 C C7 3 29 N Y Y
20 MB 33 2 C C4 3 51 N Y Y
21 HI 49 2 IC T11 3 51 N Y Y
22 HJS 58 2 IC C4 2 20 Y Y Y
23 DH 19 2 IC L1 2 22 Y N N
24 AL 40 2 C L3 2 301 Y N N
25 FT 18 2 IC T10 3 38 Y Y Y
26 CW 21 2 C T12 2 4 Y N N
27 MN 19 2 C C6 3 11 Y Y Y
28 JB 21 2 C C4 3 17 Y Y Y
29 IW 47 2 C C6 3 34 N Y Y
30 HE 29 2 C T6 3 26 Y Y Y
31 MM 24 1 IC T7 3 10 Y Y N
32 HN 17 2 C C6 1 13 Y Y Y

Note: C/IC, complete, incomplete; Level, spinal level of lesion; Invest, investigated 1, 2, or 3 times; Interv, interval (days) between trauma and 1st
investigation; Neuro, neurological investigation performed (Yes, No); UI, urinary incontinence (Yes, No); FI, fecal incontinence (Yes, No); Sex,
1 ¼ male, 2 ¼ female.

motor function (MacDonagh et al., 1992), and the rectum. It is of interest and clinical relevance
these problems originate from various kinds of that anorectal functions assessed early post-trau-
dysfunctions (Sun et al., 1990, 1995; MacDonagh ma, within the first few weeks, do not change
et al., 1992). Our data are in accordance with these significantly during the subsequent period, irre-
previous reports. People with complete lesions ex- spective of completeness or incompleteness of cord
hibit lower resting anal pressures, i.e. decreased injury, despite the fact that this time usually in-
tone of the internal anal sphincter muscle, some- cludes intense rehabilitation of the other motor
times exaggerated recto-anal reflexes following functions. However, since these programs do not
rectal distension, and some abnormalities not include pelvic floor training for regaining conti-
found in normal controls or in patients with in- nence control, one cannot dismiss the possibility
continence of other origins. For example, some that patients, at least those with incomplete le-
patients have spontaneous high-amplitude rectal sions, would benefit from programs such as bio-
contractions, at rather low distension volumes that feedback training. This hypothesis should be
uncontrollably expel the distending balloon from tested in the future.
377

Table 2. Manometric study of cord-injured subjects and controls (1st investigation)

Patients Controls Statistics

N 32 15
Age 32714 2674 po0.05
Sex (male/female) 25:7 11:4 n.s.
Resting pressureb 65725 78711 po0.02
Squeeze pressureb 66740 206740 po0.01
RAIRa (%) 75715 73713 n.s.
Sensation thresholdb 53743 (n ¼ 20) 45732 n.s.
Urge to defecate 95774 (n ¼ 19) 94745 n.s.
Pain threshold 154767 (n ¼ 14) 167770 n.s.
Cough reflex (anal)b 60732 (n ¼ 20) 176752 po0.01
Cough reflex (abdom)b 40742 (n ¼ 20) 104732 po0.01
Paradoxical contractiond 18 0 po0.01
Spontaneous defecationd 10 0 po0.01
Volumec 154765 (n ¼ 10)
a
Recto-anal inhibitory reflex. b All pressures are reported in mmHg. c Balloon volume (ml) initiating spontaneous defecation. d Number
of subjects with response.

Table 3. Manometric study of the course of disease

Investigation number

First Second Third

N 32 29 20
Age 32714 33714 31714
Sex (male/female) 25:7 22:7 16:4
Resting pressureb 65725 65721 74727
Squeeze pressureb 66740 76765 67738
RAIRa (%) 75715 (n ¼ 32) 74715 (n ¼ 29) 68711 (n ¼ 20)
Sensation thresholdb 53743 (n ¼ 20) 66742 (n ¼ 20) 57738 (n ¼ 11)
Urge to defecateb 95774 (n ¼ 19) 62732 (n ¼ 16) 82740 (n ¼ 11)
Pain thresholdb 154767 (n ¼ 14) 147778 (n ¼ 13) 190788 (n ¼ 7)
Cough reflex (anal)b 60732 (n ¼ 20) 65740 (n ¼ 18) 59735 (n ¼ 15)
Cough reflex (abdom)b 40742 (n ¼ 20) 43730 (n ¼ 15) 45722 (n ¼ 8)
Paradoxical contractiond 18 13 10
Spontaneous defecationd 10 11 8
Volumec 154765 (n ¼ 10) 156.3786 (n ¼ 11) 180778 (n ¼ 8)
a
Recto-anal inhibitory reflex. b All pressures are reported in mmHg. c Balloon volume (ml) initiating spontaneous defecation. d Number
of subjects with response.

Completeness of spinal injury is assessed clini- floor or lower limb often are used to confirm the
cally by the absence of sensory perception and the diagnosis (Neill and Swash, 1980; Herdmann et al.,
lack of motor control of the lower limbs. Trans- 1991; Enck et al., 1992). Occasionally, people with
cranial recordings of somatosensory and motor complete spinal cord injury report diffuse sensa-
cortical potentials, evoked by stimulation of the tions from the viscera, e.g., the feeling of rectal
pelvic floor or lower limbs, and recordings of the fullness preceding involuntary colonic mass move-
electrical activity, evoked by transcranial stimula- ments and defecation. These reports challenge the
tion of the motor cortex, in muscles of the pelvic reliability of the ASIA criteria (Ditunno et al.,
378

1994) for clinical assessment of cases of complete


spinal cord injury. However, these reports have
usually been attributed to re-interpretation of
bowel sounds arising from the abdominal cavity.
In the above study of anorectal motor function,
we also assessed anorectal sensory functions by
testing the subject’s sensitivity to rectal balloon
distension. During anorectal manometry, as de-
scribed above, the thresholds for minimal sensa-
tion, urge to defecate and pain following stepwise
rectal balloon distension (ml) were determined. If a
sensation of the distension (which was blinded for
the patients) was noted, it was repeated at least
once to assure reproducibility.
When subjects were grouped according to com-
plete or incomplete spinal cord injury, by clinical
assessment, 5 of 15 cases of complete injury had
some sensory function and 3 of 15 even reported
pain during rectal distension. In one subject, pain
thresholds were identical when retested and in two
subjects, minimal perception thresholds to disten-
sion were almost identical on repetition. Three
subjects (No. 17, 20, and 27, Table 1) who were
tested three times consistently reported perception
of the distension on all three occasions. One of
them had been evaluated neurologically (No. 27).
Conversely, in one subject judged as incomplete,
no remaining motor or sensory function could be
detected by anorectal manometry (No. 3, Table 1).
Patients with remaining visceral sensation had re-
producible perception thresholds and experienced
pain with rectal distension volumes up to 200 ml.
Persistence of visceral sensation, despite the clas- Fig. 1. Level of injury in subjects with complete (left) and in-
sification of ‘‘complete’’ injury, occurred with complete (right) spinal injuries. Numbers refer to the individ-
lesions at all spinal levels (Fig. 1). uals in Table 1. Bold (outer) numbers refer to those subjects
investigated neurophysiologically. See Table 1 for clinical data
As described above, 5 of 15 cases diagnosed as
and Table 3 for cases with residual sensation. Reproduced by
complete spinal cord injury by clinical assessment permission of Blackwell from Greving et al., 1998.
had some anorectal sensory function. Because of
this discrepancy between the anorectal manometry
data and the clinical assessment, electrophysiolog- area of the cortex. In the same two groups, con-
ical techniques were used to provide another indi- centric needle recording electrodes were placed
cation of completeness of injury. In 15 subjects in the external anal sphincter (Herdmann et al.,
diagnosed as complete injury and in 17 others di- 1991) while transcranial stimulation of the motor
agnosed as having incomplete injury by ASIA cri- cortex was performed with a magneto-electric coil.
teria (Ditunno et al., 1994), electrical stimulation The subjects of the incomplete group showed
of the anal mucosa (Enck et al., 1992) was per- sensory- and motor-evoked potentials with late-
formed while recording electrodes were placed bi- ncies of 53.8723.5 and 27.875.1 ms, respectively.
laterally on the scalp overlying the sensory-motor No evoked potentials were recorded in the
379

complete group. The fact that the 5 cases with nociceptive information to the thalamus (Al-Chaer
rectal sensation (out of 15 cases), diagnosed as et al., 1996a, b). However, since evoked potential
complete by ASIA criteria, showed neither senso- recordings are thought to represent predominantly
ry- nor motor-evoked potentials suggests that dorsal column transmission of information, other
electrophysiological testing is not more ‘‘sensitive’’ pathways must process the remaining sensation in
than the routine neurological examination. On the our subjects with ‘‘complete’’ lesions.
other hand, anorectal manometry provides infor- Visceral afferent information is also carried in
mation on sensory function that seems more useful the spinoreticular, spinomesencephalic and spino-
than either electrophysiology or routine neurolog- solitary tracts that project to the thalamus via re-
ical examination for the diagnosis of completeness lays in the brainstem (e.g. the nucleus of the
of spinal cord injury. tractus solitarius) and in the midbrain (see Aziz
These findings about visceral sensation in sub- and Thompson, 1998). These pathways mediate
jects with spinal cord injury also raise the question the integration of somatic and visceral input from
of the pathways by which these sensations are wide areas of the body, and also allow afferent
processed. The pelvic nerve innervates the distal information encoded within vagal afferent projec-
parts of the colon, rectum, and the urogenital tions to modulate afferent information encoded
organs. Although afferent nerves from different within spinal afferent nerves (Randich and Gebhart,
intra-abdominal nerve trunks enter multiple seg- 1992; Mayer and Gebhart, 1994). Whereas spinal
ments of the cord, each nerve shows peak projec- afferent nerves are usually only considered as the
tions to one or two adjacent segments. Thus, pathway for transmission of nociceptive informa-
innervation of different visceral organs has con- tion to the CNS, the majority of afferent projections
siderable segmental overlap, which probably have stimulus response functions that cover both
explains the poor viscerotopic localization of physiological and nociceptive ranges of stimulation
sensation in the gastrointestinal tract. Additional (Aziz and Thompson, 1998). This idea is supported
convergence of somatic afferent projections onto by the fact that, in our study, five cord-injured sub-
the same spinal segments is thought to be the basis jects reported non-painful and three subjects re-
for the referral of visceral sensation to somatic ported painful sensation following rectal distension.
structures (Aziz and Thompson, 1998). Due to the complex innervation of the viscera,
Visceral afferent information is transmitted pathways that remain partially intact following
along the spinal cord via a number of tracts, of trauma may still be able to transmit sensory in-
which the spinothalamic tracts and the dorsal col- formation from the rectum, and may even be able
umns are the most important. The lateral and me- to take over processing of visceral information to
dial subdivisions of the spinothalamic tract project and from the brain. Whereas plasticity has been
to the ventral, ventral posterior lateral, medial, and shown to be a feature of reorganization of the in-
intralaminar nuclei of the thalamus, respectively. jured spinal cord (Schnell et al., 1994; Schwab and
The lateral spinothalamic neurons mediate the Bartholdi, 1996), the application of this to visceral
sensory-discriminative aspects of pain whereas the functions remains to be shown in the future.
medial spinothalamic neurons mediate the motiva-
tional-affective aspects of pain. In contrast to con- Cortical representation of sensory functions from
ventional wisdom that the dorsal columns do not the anorectum
mediate visceral afferent information, recent evi-
dence from human studies now suggests that they Anecdotal evidence suggests that residual anorec-
do, since posterior midline myelotomy that inter- tal sensation is present in some cord-injured people
rupts the dorsal columns alleviates pelvic visceral with clinically classified complete injury of the
pain in patients with colon cancer (Gildenberg and spinal cord. This has also been shown with inves-
Hirshberg, 1984). Recent evidence from animal tigations of anorectal functions in these cord-
studies also confirms that the dorsal column is injured subjects (Greving et al., 1998, and previous
an important pathway for transmitting visceral section). We examined this phenomenon with
380

brain activation imaging. Cortical reorganization healthy controls minus patients or post-treatment
following traumatic spinal cord injury is a com- minus pre-treatment). Parameter estimates of in-
mon phenomenon, but is investigated predomi- terest of each subject were computed, and mean
nantly for motor functions of the upper and lower images across all four were used to contrast the
extremities. In people with complete spinal cord responses of the cord-injured subject and those of
injury, cortical areas adjacent to the denervated a healthy volunteer group (Lotze et al., 2001). In
one may eventually take over its functions, as comparison with a healthy group, contrast anal-
shown by cortical functional representations, and yses revealed only the activation pattern of the
this phenomenon is accompanied by clinical im- healthy subjects (Fig. 3). This implies that subjects
provements (Lotze et al., 1999). with residual visceral perception exhibit cortical
Ten paraplegic subjects with complete traumatic activation in areas similar to that found in healthy
spinal cord injury (according to ASIA criteria) volunteers, but this activation is less extensive and
(Ditunno et al., 1994) at different levels (TH3–L3) less well coordinated. We recently were able to re-
were investigated during non-painful stimulation investigate one of these four patients with residual
of the distal rectum and anal canal by means of subjective perception and cortical activation fol-
pneumatic dilatation with a balloon probe. Func- lowing anorectal stimulation. In addition to the
tional magnetic resonance imaging data were ac- stimulation, putative activation was recorded dur-
quired across the whole brain with a commercial ing anticipation of stimuli that were not delivered
1.5 Tesla tomograph (Siemens, Vision) using Echo (sham stimulation). In the initial investigation in
Planar Imaging (Lotze et al., 2001). this patient, relevant activation was revealed in
Although complete impairment was clinically prefrontal (rectal stimulation) and the second so-
diagnosed in all 10 cord-injured subjects, four of matosensory cortex (anal stimulation) only. After
the subjects experienced reproducible sensations one year, significant activation was detected bilat-
during anal and/or rectal stimulation. In the re- erally in the cerebellum for both stimulation sites,
maining six subjects who had no sensations, inju- as well as in the second somatosensory cortex for
ries of the dura or an empty spinal canal had been rectal stimulation and in the first somatosensory
reported from a surgical procedure. For the four cortex for anal stimulation. This was accompanied
patients with sensation, data analysis for each was by further improvement of subjective awareness of
undertaken using SPM99 (Welcome Department anorectal sensations. Sham stimulation did not re-
of Cognitive Neurology, London) to identify the veal any cortical activation at all.
cortical areas that show significant activation in an These data indicate that the diagnosis of com-
event-related stimulus design. Figure 2 shows im- plete spinal cord injury by ASIA criteria (Ditunno
ages from one individual during rectal stimulation et al., 1994) alone may be insufficient, as has been
in sagittal, coronal, and transverse projection speculated previously after interoception testing in
planes. Individual data analysis in these four pa- these patients (Greving et al., 1998). Cord-injured
tients revealed predominant activation in the right people with residual visceral sensation exhibit cor-
secondary somatosensory cortex, the posterior tical activation in areas similar to those found in
cingulate gyrus, and the left posterior cerebellar healthy volunteers but this activation is less ex-
lobe. In addition, left orbitofrontal cortex activa- tensive and less well coordinated. It exhibits, how-
tion was observed in one of the patients. ever, significant plasticity in the course of the
Significant activation was found with individual disease and may improve over time.
data analysis but not for the whole group, most Several questions remain that cannot be an-
likely due to differences between subjects in injury swered by results from the small number of pa-
patterns and in the levels of the spinal cord lesions. tients that we have investigated. Do sensations
With a less strict statistical criterion, group anal- arising from other gastrointestinal segments acti-
ysis was possible. For group comparison it is con- vate similar brain responses? What is the time
ventional to compute a contrast between two course of the cortical activations? Is the cortical
conditions (condition A minus condition B, e.g. network activated sufficient to explain clinical
Fig. 2. Functional magnetic resonance imaging of the brain of a cord-injured subject during non-painful stimulation of the distal rectum or of the anal canal with a
balloon. The injury was complete in the basis of ASIA criteria but the subject reported anal/rectal sensation. The brain is shown in three projections (sagittal, coronal, and
transverse). Significant cortical activation is present (po0.05 corrected) in different regions during rectal (top) and anal (bottom) stimulation. Top: Second somatosensory
cortex bilaterally, right posterior cingulate cortex, and left cerebellum. Bottom: right first and bilateral second somatosensory cortex, posterior cingulate cortex, and left
cerebellum.

381
382

Fig. 3. Group analysis (contrasts: healthy minus paraplegic subjects) following rectal stimulation revealed significant activation in the
right second somatosensory cortex and left cerebellar cortex during rectal stimulation (see text for details).

symptoms? Does it respond to therapeutic inter- pairment, ranging from disruption of sympathetic
ventions with plasticity and change? Can thera- and parasympathetic (extrinsic) pathways that af-
peutic approaches and patient management be fect autonomic motor and sensory functions to
built upon this phenomenon, e.g. biofeedback changes secondary to altered individual mobility
therapy? This warrants future investigations. and behaviors. Further research is needed to in-
vestigate these possibilities, and to develop man-
Summary and conclusions agement strategies for the bowel problems in those
with spinal cord injury.
The research discussed above demonstrates that
both sensory and motor functions of the gastro- Acknowledgment
intestinal system can be significantly impaired in
people with complete or incomplete spinal cord Supported by grants from Deutsche Forschungs-
injury. Multiple mechanisms may underlie this im- gemeinschaft, DFG En 50/18 and 50/21.
383

References Enck, P., Bielefeldt, K., Rathmann, W., Purrmann, J., Tschöpe,
D. and Erckenbrecht, J.F. (1991) Epidemiology of faecal in-
Al-Chaer, E.D., Lawand, N.B., Westlund, K.N. and Willis, continence in selected patient groups. Int. J. Colorect. Dis., 6:
W.D. (1996a) Visceral nociceptive input into the ventral 143–146.
posterolateral nucleus of the thalamus. A new function for Enck, P., Herdmann, J., Börgermann, K., Theisen, U., Zacchi,
the dorsal column pathway. J. Neurophysiol., 76: 2661–2674. P. and Lübke, H.J. (1992) Up and down the spinal cord:
Al-Chaer, E.D., Lawand, N.B., Westlund, K.N. and Willis, afferent and efferent innervation of the human external anal
W.D. (1996b) Pelvic visceral input into the nucleus gracilis is sphincter. J. Gastrointest. Motil., 4: 271–277.
largely mediated by the postsynaptic dorsal column pathway. Fealey, R.D., Szurszewski, J.H., Merrit, J.L. and DiMagno,
J. Neurophysiol., 76: 2675–2690. E.P. (1984) Effect of traumatic spinal cord transection on
Aziz, Q. and Thompson, D.G. (1998) Brain-gut axis in health human upper gastrointestinal motility and gastric emptying.
and disease. Gastroenterology, 114: 559–578. Gastroenterology, 87: 69–75.
Beuret-Blanquart, F., Weber, J., Gouverneurm, J.P., Demangeon, Gaston, E.A. (1948) The physiology of fecal continence. Surg.
S. and Denis, P. (1990) Colonic transit time and anorectal Gynec. Obstr., 87: 280–290.
manometric anomalies in 19 patients with complete transection Gildenberg, P.L. and Hirshberg, R.M. (1984) Limited myelo-
of the spinal cord. J. Auton. Nerv. Syst., 30: 199–208. tomy for the treatment of intractable cancer pain. J. Neurol.
Braden, B., Adams, S., Duan, L.P., Orthm, K.H., Maul, F.D., Neurosurg. Psychiatry,, 47: 94–96.
Lembcke, B., Hor, G. and Caspary, W.F. (1995) The [13C]ac- Glick, M.E., Meshkinpour, H., Haldeman, S., Hoehler, F.,
etate breath test accurately reflects gastric emptying of liquids Downey, N. and Bradley, W.E. (1984) Colonic dysfunction in
in both liquid and semisolid test meals. Gastroenterology, patients with thoracic spinal cord injury. Gastroenterology,
108: 1048–1055. 86: 287–294.
Bruninga, K. and Camilleri, M. (1997) Colonic motility and tone Glickman, S. and Kamm, M.A. (1996) Bowel dysfunction in
after spinal cord injury. Am. J. Gastroenterol., 92: 891–894. spinal cord injury patients. Lancet, 347: 1651–1653.
Chen, C.Y., Chuang, T.Y., Tsai, Y.A., Tai, H.C., Lu, C.L., Greving, I., Nedjat, S., Orth, G., Tegenthoff, M., Bötel, U.,
Kang, L.J., Lu, R.H., Chang, F.Y. and Lee, S.D. (2004) Loss Micklefieldt, G., May, B. and Enck, P. (1998) Anorectal
of sympathetic coordination appears to delay gastrointestinal functions in patients with spinal cord injury. Ne-
transit in patients with spinal cord injury. Dig. Dis. Sci., 49: urogastroenterol. Motil., 10: 509–515.
738–743. Han, T.R., Kim, J.H. and Kwon, B.S. (1998) Chronic gastro-
Chew, C.G., Bartholomeusz, F.D., Bellon, M. and Chatterton, intestinal problems and bowel dysfunction in patients with
B.E. (2003) Simultaneous 13C/14C dual isotope breath test spinal cord injury. Spinal Cord, 36: 485–490.
measurement of gastric emptying of solid and liquid in nor- Herdmann, J., Bielefeldt, K. and Enck, P. (1991) Quantification
mal subjects and patients: comparison with scintigraphy. of motor pathways to the pelvic floor in humans. Am. J.
Nucl. Med. Rev. Cent. East Eur., 6: 29–33. Physiol., 260: G720–723.
Coremans, G., Geypens, B., Vos, R., Tack, J., Margaritis, V., Kao, C.H., Ho, Y.J., Changlai, S.P. and Ding, H.J. (1999)
Ghoos, Y. and Janssens, J. (2004) Influence of continuous Gastric emptying in spinal cord injury patients. Dig. Dis. Sci.,
isobaric rectal distension on gastric emptying and small bow- 44: 1512–1515.
el transit in young healthy women. Neurogastroenterol. Keshavarzian, A., Barnes, W.E., Bruninga, K., Nemchausky, B.,
Motil., 16: 107–111. Mermall, H. and Bushnell, D. (1995) Delayed colonic transit
De Looze, D.A., De Muynck, M.C., Van Laere, M., De Vo, in spinal cord-injured patients measured by Indium-111
M.M. and Elewaut, A.G. (1998b) Pelvic floor function in amberlite scintigraphy. Am. J. Gastroenterol., 90: 1295–1300.
patients with clinically complete spinal cord injury and its Kirk, P.M., King, R.B., Temple, R., Bourjaily, J. and Thomas,
relation to constipation. Dis. Colon Rectum,, 41: 778–786. P. (1997) Long-term follow-up of bowel management after
De Looze, D.A., Van Laere, M., De Muynck, M.C., Beke, R. spinal cord injury. SCI Nurs., 14: 56–63.
and Elewaut, A.G. (1998a) Constipation and other chronic Longo, W.E., Ballantyne, G.H. and Modlin, I.M. (1989) The
gastrointestinal problems in spinal cord injury patients. Spi- colon, anorectum, and spinal cord patient. A review of the
nal Cord, 36: 63–66. functional alterations of the denervated hindgut. Dis. Colon
DeVault, K.R., Beacham, S., Castell, D.O., Streletz, L.J. and Rectum, 32: 261–267.
Ditunno, J.F. (1996) Esophageal sensation in spinal cord- Lotze, M., Laubis-Herrmann, U., Erb, M., Topka, H. and
injured patients: balloon distension and cerebral evoked po- Grodd, W. (1999) Reorganization in the primary motor cor-
tential recording. Am. J. Physiol., 271: G937–941. tex after spinal cord injury — a functional magnetic reso-
Devroede, G., Arhan, P., Duguay, C., Tétreault, L., Akoury, H. nance study. Restor. Neurol.Neurosci., 14: 183–187.
and Perey, B. (1979) Traumatic constipation. Gastroenterol- Lotze, M., Wietek, B., Birbaumer, N., Ehrhardt, J., Grodd, W.
ogy, 77: 1258–1267. and Enck, P. (2001) Cerebral activation during anal and rec-
Ditunno, J.F., Young, W., Donovan, W.H. and Creasey, G. tal stimulation. NeuroImage, 14: 1027–1034.
(1994) The international standard booklet for neurological MacDonagh, R., Sun, W.M., Thomas, D.G., Smallwood, R.
and functional classification of spinal cord injury. Paraplegia, and Read, N.W. (1992) Anorectal function in patients with
32: 70–80. complete supraconal spinal cord lesions. Gut, 33: 1532–1538.
384

May, R.J. (1991) Fecal incontinence in a 39-year-old man. Schwab, M.E. and Bartholdi, D. (1996) Degeneration and
Gastroenterology, 101: 1120–1126. regeneration of axons in the lesioned spinal cord. Physiol.
Mayer, E.A. and Gebhart, G.F. (1994) Basic and clinical aspects Rev., 76: 319–370.
of visceral hyperalgesia. Gastroenterology, 107: 271–293. Singh, G. and Triadafilopoulos, G. (2000) Gastroesophageal
Menter, R., Weitzenkamp, D., Cooper, D., Bingley, J., Charlifue, reflux disease in patients with spinal cord injury. J. Spinal
S. and Whiteneck, G. (1997) Bowel management outcomes in Cord Med., 23: 23–27.
individuals with long-term spinal cord injuries. Spinal Cord, Stone, J.M., Nino-Murcia, M. and Wolfe, V.A. (1990) Gastro-
35: 608–612. intestinal problems in spinal cord injury patients: a prospec-
Mollen, R.M., Hopman, W.P., Kuijpers, H.H. and Jansen, J.B. tive analysis. Am. J. Gastroenterol., 85: 1114–1119.
(1999) Abnormalities of upper gut motility in patients with Sun, W.M., MacDonagh, R., Forster, D., Thomas, D.G.,
slow-transit constipation. Eur. J. Gastroenterol. Hepatol., 11: Smallwood, R. and Read, N.W. (1995) Anorectal function
701–708. in patients with complete spinal transection before and
Neill, M.E. and Swash, M. (1980) Increased motor unit fibre after sacral posterior rhizotomy. Gastroenterology, 109:
density in the external anal sphincter muscle in ano-rectal 990–998.
incontinence: a single fibre EMG study. J. Neurol. Ne- Sun, W.M., Read, N.W. and Donnelly, T.C. (1990) Anorectal
urosurg. Pychiatry,, 43: 343–347. function in incontinent patients with cerebrospinal disease.
Randich, A. and Gebhart, G.F. (1992) Vagal afferent modu- Gastroenterology, 99: 1372–1379.
lation of nociception. Brain Res. Rev., 17: 77–99. Wheatley, I.C., Hardy, K.J. and Dent, J. (1977) Anal pressure
Rao, S.S.R., Azpiroz, F., Diamant, N., Enck, P., Tougas, G. studies in spinal patients. Gut, 18: 488–490.
and Wald, A. (2002) Minimum standards of anorectal man- Youle, M.S. and Read, N.W. (1984) Effect of painless rectal
ometry. Neurogastroenterol. Motil., 14: 553–559. distension on gastrointestinal transit of solid meal. Dig. Dis.
Read, N.W. and Sun, W.M. (1992) Anorectal manometry. In: Sci., 29: 902–906.
Henry M.M. and Swash M. (Eds.), Coloproctology and the Zahn, A., Langhans, C.D., Hoffner, S., Haberkorn, U., Rating,
Pelvic Floor (2nd ed). Butterworth, London, pp. 119–145. D., Haass, M., Enck, P., Stremmel, W. and Ruhl, A. (2003)
Schnell, L., Schneider, R., Kolbeck, R., Barde, Y.A. and Measurement of gastric emptying by 13C-octanoic acid
Schwab, M.E. (1994) Neurotrophin-3 enhances sprouting of breath test versus scintigraphy in diabetics. Z. Gastroenterol.,
corticospinal tract during development and after adult spinal 41: 383–390.
cord lesions. Nature, 367: 170–173.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 26

Problems of sexual function after spinal cord injury

Stacy L. Elliott1,2,3,4,

1
Departments of Psychiatry and Urology, University of British Columbia, BC, Canada
2
British Columbia Center for Sexual Medicine, Echelon-5, 855 West 12th Avenue, Vancouver, BC, V5Z 1M9, Canada
3
Vancouver Sperm Retrieval Clinic, Vancouver Hospital, Vancouver, BC, Canada
4
G.F. Strong Rehabilitation Centre, Vancouver, BC, Canada

Abstract: Sex is a legitimate and fundamental need in humans. Substantial changes to both the autonomic
and somatic nervous system occur after spinal cord injury, and result in altered sexual function and fertility
potential. This chapter provides a clinical overview of the main sexual and reproductive concerns and
priorities men and women face after spinal cord injury. Besides genital functioning, other autonomic
functions affect sexuality, such as bladder and bowel function, cardiovascular control and temperature
regulation. These interlinked autonomic functions are presented in their impact on sexuality. The mind-
body interaction and spinal feedback loops are discussed. It is proposed that human sexuality after spinal
cord injury can be a model for investigating integrated autonomic function. Recent research on the meas-
urement of cardiovascular parameters during vibrostimulation and ejaculation demonstrates the discord-
ance between objective and subjective signs of autonomic dysreflexia. It is hoped that health care
professionals and researchers will become motivated to attend to the unmet sexual health care needs of this
population.

What is sex? Sex is a highly individualized experience. The


drive to be sexual is a combination of biological
Sex is a legitimate and fundamental need in human drive, the degree of success in presenting (and ac-
beings. Sexual functioning is recognized by the cepting) oneself as a sexual person to the world,
health care profession as an area of joy for many positive sexual experiences and harmonic integra-
people, but it can also be an area of great mental tion with cultural and spiritual perspectives. Ulti-
and physical suffering. Medicine is mandated to mately, the priority any one individual places on
relieve suffering. After spinal cord injury, in gen- sex depends on the ‘‘pay-off’’ that comes with be-
eral, sexual satisfaction decreases (Berkman et al., ing sexual: does it result in a pleasurable physical
1983; Alexander et al., 1993; Reitz et al., 2004). experience one wishes to repeat, a more intimate
However, sexuality after spinal cord injury remains relationship with a partner, an improved mood
a central motivating factor in life (Reitz et al., and attitude about life, or better societal accept-
2004). Both health care professionals treating ance in some way?
patients and clients and researchers in the area of Physiologically, for sex to unfold in a natural
somatic and autonomic function must address this and rewarding way, people have to trust their
significant area. bodies. The sexual ‘‘pay-off’’ has to be worth it, or
there will be less motivation to repeat the be-
havior. Thus, an erection should occur with pre-
Corresponding author. Tel.: +604-875-8273; dictability and not be a source of embarrassment
Fax: +604-875-8249; E-mail: stacy.elliott@vch.ca or disappointment, and vaginal lubrication from

DOI: 10.1016/S0079-6123(05)52026-0 387


388

adequate sexual arousal should assist with com- examined the psychological, emotional and rela-
fortable sexual intercourse. Although orgasm is tionship aspects of sexuality in the lives of 15
not a prerequisite to a rewarding sexual experi- women who sustained a complete spinal cord in-
ence, reaching orgasm with reliability after spinal jury between the levels of the thoracic (T)6 and
cord injury is a bonus. lumbar (L)2 segments. These researchers were in-
When a person sustains a spinal cord injury, terested in the ‘‘real-life experience’’. The theme of
there is a new body to learn, to adapt to and to readiness was addressed; even though the study
trust. Not only do the sensory and motor changes subjects had regained their sense of identity as
affect the ability to get on with the tasks of daily people in the world, their sense of sexual identity
living, they severely affect sexual capacities as well. lagged years behind their overall self-esteem re-
Without, for example, sensation in one’s genitalia, covery. With time, experience with a partner and
how can one interpret genital touch by a partner as changes in self and relationships, these women
sexual? With interruption of sexual reflexes, not eventually set sexual exploration as a higher pri-
only are automaticity and reliability of sexual re- ority. A set of continuous themes was identified:
sponses diminished or gone, they are sometimes cognitive genital dissociation (purposefully ‘‘shut-
cross-wired with other pelvic automatic responses ting out’’ sexuality) followed by sexual disenfran-
such as perspiration, bladder and bowel function. chisement (feeling ‘‘shut out’’ and having poor
What a daunting experience to relearn how to sexual self-esteem) and eventually sexual rediscov-
make one’s sexual body parts be functional again ery. The latter was often triggered by a turning
and to use therapies to accomplish this! Even more point (e.g., new partner, orgasmic experience, etc.).
complicated is the integration of this discovered The subjects identified a relationship with a part-
function with the learned subtleties of post-injury ner to be the most significant event affecting their
sexual arousal and gratification that come with sexual recovery. Similarly, in another study (Ek-
experimentation, experience (positive and nega- land and Lawrie, 2004), women with spinal cord
tive) and patience. Some men and women with injury underwent three distinct phases while redis-
cord injury claim their injury has spurred them covering sexuality after injury: avoidance, increas-
onto a level of sexual depth and understanding ing comfort and exploration. Our clinical
they would not have otherwise known. experience with men following spinal cord injury
Readiness to address sexuality will vary from suggests that men move faster in their sexual re-
queries during the acute management (‘‘Will I ever habilitation than women. Regaining sexual func-
be able to have sex again? Can I still have kids?’’) tion appears to be an earlier priority for them.
to queries several years after injury (‘‘How do I However, this has not been substantiated by
have intercourse with my new boyfriend with this ‘‘lived-experience’’ studies similar to those done
indwelling catheter?’’). It takes time and shaping in women.
experiences to become open to sexual experimen- These findings emphasize that sexual recovery is
tation, to learn a ‘‘map’’ of sexual potential on part of post-spinal cord injury rehabilitation, and
sensate areas not traditionally thought to be sex- that sex is important in the lives of men and wom-
ually rewarding and to appreciate the sexual ca- en after injury. Past researchers have not always
pacity that high mental arousal alone can provide. felt that way. Cole et al. (1973) fought against the
Clinicians in the field of sexual medicine are ac- dogma that sexual rehabilitation rated low against
quainted with the tenacity and positiveness that other rehabilitation variables, by noting that in the
many men and women with spinal cord injury lay literature many persons with cord injury
bring to the table when sexuality is finally a focus mourned the loss of sexual function more than
in their lives. other deficits such as loss of walking. For the next
The majority of studies of sex following spinal 30 years many well-meaning rehabilitation profes-
cord injury focus on physical changes, with far sionals did not recognize that sex was a priority for
fewer studies on sexual satisfaction and prediction their patients: their focus was survival, then inde-
of positive sexual outcomes. Tepper et al. (2001) pendence. Sexual rehabilitation was a luxury, and
389

an uncomfortable one at that to discuss or treat. In vicarious? He may need assistance to maximize the
the 1970s and early 1980s, not many attractive great sexual potential that remains and to pursue
medical therapies were available; sexual counseling higher arousal and even orgasmic release through
and adoption of children rather than procreation more non-conventional avenues (e.g., Tantric sex
were the mainstays. experimentation discussed by Tepper: www.sexu-
In her 2004 survey of priorities for over 600 alhealth.com). Therefore the issues of past sexual
persons with spinal cord injury, Anderson con- experiences, willingness to experiment and readi-
firmed what health care professionals dealing with ness are of paramount importance. These are
sexuality have always known: sexuality is very im- espoused in a set of sexual rehabilitation principles
portant to the majority of persons after spinal cord (Elliott, 2003). These principles include the max-
injury (Anderson, 2004). The survey found regain- imization of the underlying physiology, followed
ing sexual function to be either the first or second by adaptation to the remaining limitations with
highest priority of 28.3% of quadriplegics and the use of medical enhancement and, finally, being
45.5% of paraplegics, compared with other prior- open to potentially new therapies and attitudes
ities such as recovering motor function and blad- previously not part of pre-injury sexual practice.
der and bowel issues. Sex is no longer to be
disregarded as relatively unimportant for the ma-
jority of persons with spinal cord injury. Impact on sexual function of the autonomic,
In this era of early scientific progress in sexuality motor and sensory changes associated with spinal
following cord injury, it is critical to provide use- cord injury
ful, practical assistance to persons with spinal cord
injury as soon as they are ready. What a difference Despite the recent advances in the knowledge of
proper sexual rehabilitation can make: addressing male sexual function and in therapies for male
the mind-body connection together versus focus- sexual dysfunction, sexual neurophysiology, as a
ing only on the genitalia, can make all the differ- science, is a complicated subject with many un-
ence to a successful outcome. Misinformation or knowns. The highly complex role of central auto-
unrealistic expectations can lead to months or nomic and somatic control, the interconnection
years of non-productive sexual experience accom- between spinal reflex pathways, and the continual
panied by negative psychological overlay. For ex- local and generalized moment-to-moment cerebral
ample, even though the reflex component of and intraspinal feedback that happens during sex-
orgasm for women may need to be relearned and ual activity are very hard to systematize accurate-
reinforced with practice, starting too early after ly, especially in human basic science research.
injury to experiment with recovering reflexes may Human spinal cord injury has been a paramount
undermine a woman’s sexual confidence and make model in our understanding of sexual neurophys-
her give up hope of a future sex life. Comarr and iology. The role of the autonomic nervous system
Vigue (1978) suggested that it might take at least 6 is crucial in sexual functioning. Human sexuality
months to know the level of erectile recovery, es- after spinal cord injury is a model for investigating
pecially among men with incomplete lesions. For integrated autonomic function. To learn, or rather
patients with lower motor neuron or cauda equina to relearn, a sexual sense of self in a new body
injury, it may take a year since there is more pos- requires the brain’s interpretation, not just of sen-
sibility of nerve regeneration as compared with sory and motor, but also of autonomic, compo-
injury located within the spinal cord. Providing a nents. In the simplest of terms, the physiology
pharmacological rigid erection for a young injured associated with sexual arousal is primarily para-
man may provide tremendous relief and opportu- sympathetic. As sexual arousal increases, a critical
nity to regain that part of his life soon after injury. neurological threshold (often termed the ‘‘orgas-
But when he is unable to ejaculate or experience mic’’ or ‘‘ejaculatory’’ threshold) is reached, and
orgasm for years he may feel discouraged. What if the sympathetic nervous system predominates.
his only source of pleasure with a partner is When the autonomic nervous system is altered
390

below the level of spinal cord injury, sexual arous- sadness and anger at being robbed of sexual
al can generate altered cardiovascular responses sensations, frustration at altered motor abil-
and disordered sexual, bladder and bowel proc- ity and autonomic coordination and all the
esses. More stimuli may be required to trigger influences mentioned below make the mind
what remains of sexual reflexes, and extragenital the ultimate arbitrator of sexual pursuit.
stimuli (i.e., around the hypersensitive level of in- 2. Genital functioning: The spinal sexual reflexes
jury, or the face, neck and ears) may take on are under tonic inhibitory control by supra-
a new, amplified and critical importance in the spinal neurons (Chuang and Steers, 1999). If
genesis of sexual arousal. this control is removed by spinal cord injury,
The use of animal models to research sexual the spinal reflexes are free to be triggered
functioning, while critical to our current knowl- without conscious control (i.e., reflex erec-
edge base, is limited, as subjective data about sex- tions). However, the intactness of the genital
ual experiences is not available. If we listen and reflexes required for sexual functioning
observe carefully, the men and women with spinal (arousal, orgasm, etc.) is not necessarily re-
cord injury are the best teachers of sexuality after lated to the intactness of sensory or motor
injury. They dictate what needs to be researched at function. Remaining reflexes can be thera-
a human trial level. What have we, as health care peutically enhanced: alternate learning is re-
professionals and researchers, learned in terms of quired for sexual rehabilitation.
autonomic responses? 3. Bladder functioning: Bladder functioning de-
Sexual feelings and responses in the spinal cord pends on remaining autonomic pathways.
injured population (and in the able-bodied for that Factors such as bladder continence dictate
matter) constitute a head-to-toe approach. After freedom from leakage or accidents during
spinal cord injury, the changes instigated by the sexual activity. Neurogenic bladders increase
altered autonomic system affecting sexual function the risk for urinary tract and kidney infec-
and manifestations of sexuality are as equally, if tions. Urine odors and various external urine
not more, important as those from motor and collection apparatuses are usually associated
sensory alterations. This suggested head-to-toe negatively with sexuality.
‘‘autonomic’’ framework of sexual changes fol- 4. Bowel function: Bowel-emptying practices are
lowing spinal cord injury is meant to elucidate the often time-consuming. Bowel evacuation pat-
interconnection between autonomic functions that terns can affect general wellness and energy.
either interfere with sexuality, or are a conse- Flatulence or stool incontinence can be one
quence of sexual activity. With real-life spinal cord of the most socially embarrassing and anti-
injury experiences, altered genital functioning may sexual events that a person with a spinal cord
or may not be viewed, by the injured person, as the injury can experience.
factor most affecting his or her sexuality. This is 5. Cardiovascular and respiratory function: Al-
important for researchers and clinicians to appre- tered cardiovascular responses during sexual
ciate. Research and clinical efforts need to be di- arousal and orgasm/ejaculation occur after
rected at the most important sexual rehabilitation cord injury. Altered heart rates and rhythms,
priorities identified by men and women with spinal symptomatic hypotension in quadriplegics or
cord injury. To date, such preference studies are the hypertension of autonomic dysreflexia
few. can alter the safety and willingness to be sex-
An altered nervous system can affect sexuality ual. The same effect can result from respira-
from head to toe. tory problems like loss of vital capacity due
to paralysis of the intercostal muscles, in-
1. Cerebral functioning: The mind is the ruler of crease in airway resistance due to the decrease
sexual desire, sexual self-image and mood al- in bronchodilator sympathetic neuronal ac-
terations following spinal cord injury. De- tivity and loss or attenuation of the cough
pression from overwhelming life changes, reflex. In most cases, autonomic dysreflexia is
391

a deterrent to safe or enjoyable sexual prac- sexual intercourse if her bowel reflexes are trig-
tices and can interfere with sperm retrieval gered by penile penetration (Szasz, 1983). Bladder
for fertility purposes in men with spinal cord or bowel fullness can impede or facilitate the trig-
injury. gering of ejaculation by vibrostimulation for
6. Altered thermal regulation and skin integrity: sperm retrieval. Autonomic dysreflexia can result
Unnatural appearance or texture of the skin in avoidance of sexual activity by both sexes. Al-
can affect sexual self-esteem. Skin integrity is though these examples demonstrate the negative
altered and extended pressure leads to skin consequences of sexual arousal and activity, there
breakdown, affecting sexual positioning. are positive consequences also. Obviously, the
Lack of sweating and vasodilatory responses pursuit of sexual activity after injury is a result of
below the level of lesion or unattractive ab- weighing the pros and cons, and fortunately for
normal flushing and sweating above the level most, the pros prevail. For example, some men
of lesion can impede the motivation to be and women with spinal cord injury who experience
sexual. Inability to tolerate nakedness due to mild autonomic dysreflexia learn, with time, to in-
chilling can be restrictive for both partners. terpret those altered cardiovascular processes as
Cold extremities are not uncommon after sexually enhancing.
spinal cord injury, and are often avoided by a With spinal cord injury, disturbances in the sen-
sexual partner. sory input to the brain, descending autonomic
7. Sensory deficits: These severely affect the control of reflex centers in the spinal cord and al-
neural pathways reinforced from previous tered efferent reflex abilities result in various com-
sexual learning. The highly motivated person plete, incomplete or even aborted sexual responses.
with spinal cord injury can reprogram senso- Along with the changes seen in physiological sex-
ry afferents from previously ‘‘non-sexual’’ in- ual responses following spinal cord injury, the
puts on the body to have sexual flavoring. person with this injury must learn and adapt to his
This may be brain plasticity or other relearn- or her sexual new body. Psychological feedback
ing phenomenon (Bach-y-Rita, 1999), a vir- feeds into recovery progression. This mind-body
tually unexplored area. interaction leads to, or deters, sexual curiosity, sex-
8. Motor deficits: Loss of abdominal tone (i.e., seeking behaviors and motivation to express inti-
‘‘quad belly’’) affects sexual self-view. De- macy in a sexual manner.
creased muscle tone and bulk affects sexual Sexual activity in people with spinal cord injury
self-esteem and pride, especially if this was a may have positive consequences, such as relief
source of sexual confidence before the injury. from spasms following ejaculation (Courtois et al.,
Loss of motor coordination or strength can 2004). This anti-spastic effect has been reported
preclude even holding or caressing a partner. following ejaculation either by vibrostimulation
Inability to balance or support the upper (Szasz and Carpenter, 1989; Elliott, 2003; Laessoe
trunk affects sexual positioning options and et al., 2004) or electroejaculation (Halstead et al.,
ability to thrust with the pelvis. Muscle 1993) and can last for several hours.
spasms and clonus can affect sexual enjoy-
ment and may preclude sexual positioning
options. Changes in sexual responses after spinal cord injury

Medications and other pre-existing medical con- Masters and Johnston (1966) outlined the complex
ditions can also influence sexual function and re- genital and cardiovascular changes associated with
ceptivity. arousal, orgasm and ejaculation through the de-
There are many clinical examples of interplay of velopment of a sexual response cycle. They defined
various autonomic functions during sexual activ- four phases of a ‘‘sexual mountain’’, including ex-
ity. For example, a woman with spinal cord injury citement, plateau, orgasm and resolution. Not
may be at risk for bowel incontinence during much has been written about how the sexual
392

response cycle changes after cord injury. For ex- Approximately 50% of women with spinal cord
ample, Cole (1975) noted that with sexual arousal, injury (including women with complete injury)
the labia in women with spinal cord injury only who participated in a study by Sipski et al. (1995)
swell, whereas they swell and open in non-injured were able to achieve orgasm (see also Sipski and
women. In men with spinal cord injury, ejaculation Arenas, this volume). In the Kinsey survey (Dono-
is almost always absent. When erection occurs, it hue and Gebhard, 1995), women with incomplete
may not be as reliable as experienced before injury lesions attained orgasm more frequently than
and may detumesce with pressure or the penis may women with complete lesions, regardless of the
have an altered filling capacity (i.e., the glans may level of injury. For women with spinal cord injury,
not fill with high arousal). Although typically with the incidence of orgasm generated by self-stimula-
sexual arousal and orgasm, muscle tone increases, tion decreased, and the time required to attain or-
and heart rate, respiratory rate and blood pressure gasm increased (Donohue and Gebhard, 1995;
increase (Masters and Johnson, 1966), cardiovas- Sipski et al., 1995; Ferrerio-Velasco et al., 2005).
cular responses with arousal in spinal cord injured Breast stimulation and mechanical genital stimu-
people may be overexaggerated (i.e., autonomic lation were often employed. Many compensated
dysreflexia). Resolution of cardiovascular changes for the loss of genital sensitivity by stimulating a
may take longer than pre-injury, especially if there sensate part of their body. Greater sexual knowl-
is atypical autonomic dysreflexia (Elliott and edge and higher sex drive were two variables in
Krassioukov, 2005). women that could predict a higher chance of
reaching orgasm after spinal cord injury (Donohue
and Gebhard, 1995).
Clinical issues of female sexual function after spinal After spinal cord injury, women have many sex-
cord injury ual concerns other than orgasmic difficulties. They
include altered arousal capacity (i.e., vaginal lu-
The literature on sexuality after spinal cord injury brication and accommodation), impaired motor
is dominated by men. Men are approximately 75% ability, decreased sexual satisfaction and concerns
of the spinal cord injury population and, therefore, about attractiveness and partnerships. In their
too few women are available to warrant statisti- survey of women with spinal cord injury, Jackson
cally significantly sized samples in many studies and Wadley (1999) noted an increased concern
(Siosteen et al., 1990). Like children, women have about sexually transmitted diseases. They also
traditionally been excluded from many medical noted that, whereas fertility is not affected, preg-
studies. Surveys of women with spinal cord injury nancy is associated with risks and complications
show definite loss of sexual functioning and sexual (increased urinary tract infections, changes in
satisfaction post injury. While rates of sexual in- bladder management, increased risk for skin
tercourse and orgasmic success both drop after breakdown, difficult transfers, increased risk for
injury, they both increase in frequency over time, deep vein thrombosis and delayed bowel empty-
although not to pre-injury levels (Jackson and ing). Labor and delivery must be monitored since,
Wadley, 1999). In one large survey by the Kinsey depending on the level of lesion, labor may not be
Institute (Donohue and Gebhard, 1995), about felt. Women with spinal cord injury also give birth
60% of the women with spinal cord injury mas- to lower birth weight infants (Jackson and Wad-
turbated after injury, with half experiencing vag- ley, 1999), the etiology of which is not clearly un-
inal lubrication and only a third experiencing derstood.
orgasm. In another survey (White et al., 1993), the As noted previously, many surveys have shown
researchers noted less intercourse but relatively that the greatest sexual concerns for women with
good sexual satisfaction among women with spinal spinal cord injury are problems associated with
cord injury. They also found that injury before the urinary and bowel accidents and with autonomic
age of 18 years implied a greater risk of not having dysreflexia (Jackson and Wadley, 1999). While
an active sex life. dysreflexia usually presents in women with injury
393

higher than neurological level T6, this survey also muscles (McKenna, 1999). The result is an increase
noted reports of autonomic dysreflexia in a small in penile intracavernosal pressure, leading to pe-
number of women with lumbar/sacral injury. nile rigidity (Schmidt and Schmidt, 1993).
Menopausal changes mirror able-bodied coun- As will be discussed by Brown, Hill and Baker
terparts (Dannels and Charlifue, 2004). The peri- (this volume), basically two distinct control mech-
menopause presents a unique challenge for women anisms induce penile erection: reflexogenic and
with spinal cord injury since the symptoms may psychogenic. To understand why this distinction is
mimic or mask conditions associated with spinal clinically relevant on a daily basis for the man with
cord injury, such as autonomic dysreflexia, infec- spinal cord injury, a brief description of the auto-
tions, impaired temperature regulation and spinal nomic involvement is presented. The tactile-de-
cord cyst development. Autonomic dysreflexia pendent reflexogenic erection is mediated by a
during pregnancy and labor must be distinguished reflex arc that is complete at the sacral spinal level.
from pre-eclampsia, another hypertensive condi- The afferent limb is composed of the dorsal nerve
tion associated with pregnancy that requires dif- of the penis/pudendal nerve, and the efferent limb
ferent treatment (Pope et al., 2001; Yarkony and consists of preganglionic axons traveling in the
Chen, 1995). There are reports of cerebral intra- pelvic nerve to the pelvic plexus, where ganglion
ventricular hemorrhage (McGregor and Meeuw- cells send axons to the penis via the cavernous
sen, 1985) with resultant neurological deficits and nerve (Chuang and Steers, 1999). Spinal cord in-
death (Abouleish, 1980) associated with unrecog- jury above the sacral level will not only preserve
nized autonomic dysreflexia during labor or deliv- the reflex but can even enhance it, especially if the
ery. Methods to prevent autonomic dysreflexia in lesion is complete (Chuang and Steers, 1999), due
these circumstances include the use of epidural to loss of tonic inhibitory control. This loss results
anesthesia to block the reflex arc or prompt deliv- in a decrease in the sensory threshold and latency
ery by caesarian section (Yarkony and Chen, 1995; of erection (McKenna, 1999). Clinically, men with
Pereira, 2003). cervical injuries come to expect reflex erections to
non-sexual touch stimulation associated with sit-
uations such as catheterization, chafing from
Clinical issues of male sexual function after spinal clothing, or bumping on rough road in their
cord injury wheelchair. Such reflex erections are labeled
‘‘spontaneous’’; they are reflex in origin and are
Central to male sexual functioning after spinal produced by the same touch mechanism that pro-
cord injury is the presence of an erection and the duces sexual erections.
option of sexual intercourse. While it is true that Alternately, the ability to have reflex erections is
non-coital activities can be highly satisfying sexu- lost if the sacral spinal cord is injured or if the
ally, most men with spinal cord injury need to pudendal nerve or pelvic nerve is destroyed
know whether an erection adequate for sexual in- (Chuang and Steers, 1999). The bulbocavernosus
tercourse is possible, either naturally or assisted reflex, a polysynaptic response elicited by low
(Dr. Claus Hultling, personal communication). threshold pudendal sensory fibers, activates pu-
Erection requires participation of sacral parasym- dendal motorneurons to contract the striated peri-
pathetic (pelvic), thoracolumbar sympathetic neal muscles (McKenna, 1999). Since tactile
(hypogastric and lumbar sympathetic chain) and stimulation, especially to the glans penis, activates
somatic (pudendal) nerves (Chuang and Steers, the bulbocavernosus reflex, Szasz (1986) suggests
1999). Vasodilator preganglionic neurons (prima- that the clinical test of the bulbocavernosus reflex
rily parasympathetic) become activated, the activ- can determine the potential for reflex erections by
ity of the vasoconstrictor preganglionic neurons demonstrating an intact sacral reflex.
(mainly sympathetic) become suppressed and pe- Psychogenic erections occur in men with an in-
nile tumescence occurs. Activation of the somatic tact nervous system in response to various stimuli
motoneurons causes contraction of the pelvic floor (Chuang and Steers, 1999). The sensory inputs
394

(or afferent limb) are processed in the higher centers results in frustration and feeling of worthlessness
(imaginative in the limbic system, olfactory in the (Szasz, 1983).
rhinencephalon, visual in the occipital regions and Reflexogenic and psychogenic mechanisms
tactile in the thalamus inputs) and are integrated in probably act synergistically to determine the erec-
the medial pre-optic and anterior hypothalamic tile response via a final common pathway involv-
regions and paraventricular nucleus. The brain also ing a sacral parasympathetic route (Chuang and
receives sensory input from the penis via ascending Steers, 1999). Courtois et al. (1993) state that neu-
spinal pathways. The efferent limb of this reflex rologically, a spinal cord lesion between the two
pathway from the brain is through the lumbar erection centers should maintain both the reflex
sympathetic and sacral parasympathetic outflow to and psychogenic erection potential. However, in
the penis. Men with injuries to their sacral cord my clinical experience and that of others (Szasz,
or lower are often dependent on these intact 1983), this specific lesion can result in the impor-
psychogenic pathways to elicit an erection; they tant loss of cord communication between the two
are also dependent on maintaining their mental centers, resulting in a poorer erection than expec-
sexual arousal, unlike the men with reflex erections, ted. McKenna (1999) suggests that a strict division
in order to obtain and maintain their erection. between psychogenic and reflexogenic erections
Oral phosphodiesterase-5 inhibitor (PDE5 in- may not be possible, since even erections generated
hibitors) medications such as Viagras (sildenafil), by higher neural activity may be facilitated by
Levitras (vardenafil), or Cialiss (tadalafil) are sensory stimuli elicited by peripheral sexual arous-
used with efficacy in the spinal cord injured pop- al, and that loss of this positive feedback system
ulation. PDE5 inhibitors may make spontaneous following spinal cord injury may explain why
or reflex erections more frequent when the drug is erections are often not sustained and why ejacu-
still active in the body. Clinically, men with sacral, lation often requires very strong stimuli.
cauda equina or conus medullaris lesions do not Ejaculation and orgasm pose an even bigger
seem to respond as well to the PDE5 inhibitors. problem. In a large-scale study done at the Kinsey
Although not studied, this may be due to less ne- Institute, only 12–15% of men with all levels of
urogenic nitric oxide release (a neurotransmitter spinal cord injury could ejaculate (Donohue and
essential for penile smooth muscle relaxation) with Gebhard, 1995). The ejaculatory reflex, consisting
poorly mediated psychogenic erections in men with of seminal emission (sympathetic) and ejaculation
flaccid paralysis as compared with the more robust (parasympathetic and somatic) (Elliott, 2003) can
reflexogenic erections in men with spastic paralysis. be disrupted by spinal cord injury, but the most
Granting the oversimplification, while the para- common problem is that of absence of ejaculation
sympathetic nervous system is primarily responsi- or of triggering the reflex. Unlike reflex erections,
ble for vasodilatation of the penile vasculature and the triggering of ejaculation in a man with com-
erection, and the sympathetic nervous system is plete upper motor neuron injury requires a specific
responsible for detumescence (loss of erection), it stimulus, and usually a sexual stimulus alone is not
appears that the sympathetic nervous system also adequate. Penile vibrostimulation increases the
maintains erections after injury to parasympathet- chance of ejaculation if the thoracolumbar reflexes
ic pathways (Chuang and Steers, 1999). This com- are intact, but again, the vibrator needs to be spe-
pensatory mechanism brings with it another cifically calibrated for the response to occur
potential clinical problem for the man dependent (Sonksen et al., 1994). Fertility in men is there-
on psychogenic erections; while he struggles to fore compromised due to erection and ejaculatory
maintain his erection through activation of problems. Fertility is also affected due to changes
sexually arousing mental thought, he is activating in semen quality after spinal cord injury (Lin-
the fibers of the sympathetic chain, and such ad- senmeyer and Perkash, 1991; Brackett et al., 1997;
renergic stimulation will bring about the first stage Elliott, 2003).
of ejaculation, seminal emission. This flow of Ejaculation is not necessarily associated with
seminal emission results in detumescence. This erection, and erection itself may be variable
395

throughout the ejaculatory procedure in men with the pudendal nerve activated during phases of
spinal cord injury (Szasz and Carpenter, 1989; emission and ejaculation (deGroat and Booth,
Sonksen et al., 1994; Elliott, 2003). In sperm re- 1980). It is associated with reversal of the physi-
trieval procedures, for example, the loss of erection ological changes occurring with the buildup of
is not an indication to stop vibrostimulation, nor is sexual excitement, or the release of this sexual
the presence of a strong erection a reliable predic- tension (Klein, 1988). However, clinical examples
tor of ejaculation (Szasz and Carpenter, 1989; have shown that orgasm appears to be far more
Elliott, 2003). This dissociation of erection and complicated. For example, orgasm can occur in
ejaculation is also seen in other neurogenic condi- men with spinal cord injury who are not capable of
tions, such as post-radical prostatectomy and mul- ejaculation, and altered capacities for orgasm seem
tiple sclerosis. Despite difficulties with ejaculation, possible in some spinal cord injured men and
up to 42% of men with various levels and com- women through stimulation of non-genital or oth-
pleteness of spinal cord injury have reported or- er sensory inputs (Elliott, 2003). Orgasm could be
gasm (Alexander et al., 1993). Other researchers a genitally based, learned reflex as Sipski suggests
(Talbot, 1949; Cole and Cole, 1981; Szasz, 1983) from her research (see Sipski and Arenas, this vol-
have stated that their subjects were able to expe- ume), an ‘‘efferent’’ cerebral interpretation of ei-
rience what they considered to be highly pleasur- ther centrally generated activity or of sensory
able orgasms by focusing on a sensate part of their input arriving from a different neural system, such
body and intensifying the sensation to be sexual, as the vagus nerve (Komisaruk and Whipple,
or reassigning the sensation to their genitals. In the 1991) or other typical genital or non-genital sourc-
Kinsey study (Donohue and Gebhard, 1995), al- es (Bach-y-Rita, 1999). Orgasm is likely a combi-
though almost all males prior to spinal cord injury nation of a genitally, reflex-based component (that
could attain orgasm with masturbation, just under improves with reinforcement) and non-genital or
50% could attain orgasm after injury, and not non-pudendal nerve-dependent components that
with reliability. It appeared that those with para- may play a larger, compensatory role when the
plegia and those with incomplete injuries had a reflex component of orgasm is impaired.
better chance of experiencing orgasm than those Therefore, although an erection can be provided
with quadriplegia and those with complete inju- through safe and effective erection enhancement
ries. Orgasm attainment also required more time methods – a big breakthrough – not much can be
(Donohue and Gebhard, 1995). Tepper (Tepper done to provide feelings to the penis in order for
dissertation, website) discussed lived experiences the man to fully experience touch, intercourse and
that could either impede or facilitate orgasm for genital orgasm. Nor can his urine loss with genital
both men and women with spinal cord injury. The stimulation and ejaculation be eliminated without
role of the PDE5 inhibitor Levitras (vardenafil), drastic measures, or his risk of autonomic dysre-
used successfully for erection enhancement in men flexia with ejaculation be removed. Fertility op-
with spinal cord injury, was recently noted to tions for men with spinal cord injury have had the
double the ejaculation rate in men with spinal cord biggest improvement over the last ten years with
injury (19% with vardenafil vs. 10% for placebo) effective sperm retrieval methods and the refine-
(Giuliano, 2004). This was not noted in the orig- ment of in vitro fertilization and intracytoplasmic
inal studies with Viagras in men with cord injury sperm injection. However, we cannot reverse his
(Derry et al., 1998; Maytom et al., 1999). ejaculatory dysfunction.
From an autonomic point of view, the neuro-
physiology of orgasm is unclear. In men, although Sexual consequences: the example of autonomic
both orgasm and ejaculation occur almost simul- dysreflexia
taneously, they are two distinct entities (Elliott,
2002) that can be separated by medical conditions. Autonomic dysreflexia is a response to a noxious
Orgasm is traditionally described in urology texts or non-noxious stimulus to the body below the
as the cerebral processing of afferent stimuli via level of the injury, resulting in symptoms such as
396

headache, nausea, sweating above the level of in- occur during such ejaculatory procedures (Elliott
jury and severe hypertension that could lead to et al., 2005).
sequelae such as stroke and death (Teasell et al., Clinically, various adjustments to the severity
2000). By definition, autonomic dysreflexia is only and/or personal interpretation of autonomic dys-
recognized in the neurologically disabled popula- reflexia symptoms can occur as time progresses.
tion, such as those after spinal cord injury or with Some persons have ceased sexual activity because
multiple sclerosis (Bateman and Goldish, 2002). of it, others have adapted to the discomfort, and
Sexual arousal, a phenomenon of vasoconges- some have incorporated it as a positive aspect of
tion and neuromuscular tension (Masters and sexual activity. Others have noted a drop in sub-
Johnson, 1966) resulting in penile erection in jective awareness of dysreflexia with continued
males or vaginal lubrication and accommodation sexual activity (primarily with repeated ejacula-
in females, has a cardiovascular component. Blood tion), even to the point of dysreflexia becoming
pressure increases with arousal and tends to peak symptomatically ‘‘silent’’. Silent autonomic dysre-
with the advent of genital orgasm. In one study of flexia is a recognized phenomenon in voiding and
able-bodied healthy males (Nemec et al., 1976), bowel procedures (Linsenmeyer and Perkash,
mean blood pressure at orgasm was in the range of 1991; Kirshbium et al., 2002) and during sperm
161/77 mmHg–163/81 mmHg depending on the retrieval procedures. Sexual activity has also pro-
sexual position. The risk of cardiac complications voked atypical dysreflexia termed ‘‘malignant’’
during sex is minimal in the able-bodied popula- (Elliott and Krassioukov, 2005) lasting for days
tion (Drory, 2002), due to the briefness of the car- beyond the sexual experience.
diovascular stress and the intact compensatory Descriptions of autonomic dysreflexia and sex-
mechanisms for blood pressure control. Whereas ual activity from patients with cord injury have
autonomic dysreflexia during sexual activity or included both positive and negative consequences.
sperm retrieval procedures is usually short-lived For some, the rising feeling of headache, facial
due to the causative stimuli being removed, occa- warmth and discomfort or nausea is anti-sexual. If
sionally a worsening and or prolongation of the there is no accompanying orgasm or other sexual
dysreflexia induced by penile vibrostimulation has payoff, a pounding headache from dysreflexia
been observed after this stimulation is removed. leads to avoidance of high arousal or ejaculation,
For example, at the Vancouver Sperm Retrieval except when medically required (i.e., fertility pro-
Clinic, one incomplete quadriplegic (ASIA C) who cedures). For example, one woman who had a very
is often symptomatically hypotensive (blood pres- active and rewarding sexual life pre-injury stated
sure approximately 60/40 mmHg) states that, at she felt a tremendous loss at not being able to at-
ejaculation induced by vibrostimulation, he feels tain high arousal without autonomic dysreflexia
an intense adrenaline rush leading to an out- making her sweaty and nauseated, and turning her
of-body experience, almost like ‘‘death’’. His and her partner off. Despite this, she was persistent
blood pressure during ejaculation on several oc- in being sexual, determined to regain some sem-
casions has been consistently recorded at greater blance of her past; sex remained important to her.
than 200 mmHg systolic with a diastolic pressure Other patients have stated that they have learned
of over 100 mmHg. Over the next 10 min after to use their particular experience of autonomic
ejaculation, his blood pressure fluctuates around dysreflexia to enhance or extend the sexual feelings
three times his resting value, until it settles at they were already experiencing. What was formerly
140–160 mmHg systolic within 20 min post-ejacu- distressing was consciously enveloped into the over-
lation. Understandably, this atypically prolonged all sexual arousal, and eventually interpreted as pos-
autonomic dysreflexia makes him highly anxious itive and sexually heightening. These anecdotal
about repeating the process. On the other hand, remarks have come primarily from men with in-
silent dysreflexia, a condition in which systolic complete spinal cord injury who can ejaculate. The
blood pressure increases by at least 20 mmHg in severity of subjective symptoms of autonomic
the absence of any subjective symptoms, can also dysreflexia (headache, facial warmth and discomfort
397

or nausea) has also been noted to decrease with re- major psychological, social and economic reper-
peated ejaculations in some men at our clinic. These cussions. Knowing that autonomic function lies at
men who are long-term patients are now able to the basis of sexuality and sexual ability should
ejaculate with very little symptoms of dysreflexia. hopefully spur research in this area and add to the
For example, symptoms of headache are either ab- very important downstream effects that are fun-
sent or are substantially reduced in duration. damental to human happiness.
Our current research in ejaculation-induced au-
tonomic dysreflexia demonstrates that subjective
symptoms do not predict or reflect objective blood Acknowledgments
pressure readings, and that cardiac arrhythmias
can occur at this time (Claydon et al., 2005; Sheel International Collaboration of Repair Discoveries
et al., 2005). We have confirmed the established (ICORD), and its Director, John Steeves, PhD, for
knowledge that men with cervical injuries are more his research support and encouragement, The Rick
likely to experience autonomic dysreflexia during Hansen Institute for their funding of research in
vibrostimulation than are men with thoracic inju- sexuality over the last ten years, collaborator And-
ries. However, an important clinical point is that rei Krassioukov, MD, PhD, research colleagues
symptoms are not predictive of severity of blood (in particular Marci Ekland RN, BSN, CRRN and
pressure rise, and so cannot be used to estimate Kate McBride RN, BSN, CRRN), clinical co-di-
safety of sexual practices or clinical procedures. rector Mark Nigro MD, FRCP(C) and mentor,
Professor Emeritus George Szasz, MD.

Conclusions
References
In conclusion, just as human spinal cord injury is
the model for the neurophysiology of sexual spinal Abouleish, E. (1980) Hypertension in a paraplegic parturient.
Anesthesiology, 53: 348–349.
reflexes, human sexuality after spinal cord injury Alexander, C.J., Sipski, M.L. and Findley, T.W. (1993) Sexual
can be the model for integrated autonomic func- activities, desire and satisfaction in males pre- and post-spinal
tion. Within the framework of a head-to-toe au- cord injury. Arch. Sex. Behav., 22(3): 217–228.
tonomic approach to sexuality, multiple Anderson, K.D. (2004) Targeting recovery: Priorities of the spinal
autonomic systems could be investigated. Ejacu- cord-injured population. J. Neurotrauma, 21(10): 1371–1383.
Bach-y-Rita, P. (1999) Theoretical aspects of sensory substitu-
lation also provides a good predictive model for tion and of neurotransmission-related reorganization in spi-
observable autonomic dysreflexia and how it nal cord injury. Spinal Cord, 37: 465–474.
changes over time with repeated challenges. Over- Bateman, A.M. and Goldish, G.D. (2002) Autonomic dysre-
all, the uniqueness of sexual rehabilitation lies in flexia in multiple sclerosis. J. Spinal Cord Med., 25(1): 40–42.
Berkman, A.H., Weissman, R. and Friedlich, M.H. (1983) Sex-
its potential to grow even when the motor and
ual adjustment of spinal cord injured veterans living in the
sensory recovery has met its limits. community. Arch. Phys. Med. Rehabil., 64: 47–52.
It is unfortunate that the time devoted to re- Brackett, N.L., Padron, O.F. and Lynne, C.M. (1997) Semen
search or clinical rehabilitation in the sexual and quality of spinal cord injured men is better when obtained by
fertility area is not reflective of the need, since sex vibratory stimulation versus electroejaculation. J. Urol., 157:
is clearly a priority to men and women following 151.
Chuang, A.T. and Steers, W.D. (1999) Neurophysiology of pe-
spinal cord injury. Researchers and clinicians in- nile erection. In: Carson C., Kirby R. and Goldstein I. (Eds.),
terested in sexual and fertility rehabilitation can Textbook of Erection Dysfunction. ISIS Medical Media Inc.,
find an area in autonomic dysfunction where they Oxford, UK, pp. 59–72.
feel they could make a difference. To paraphrase Claydon, V., Elliott, S., Sheel, A.W. and Krassioukov, A.V.
(2005) Incidence of cardiac dysrhythmias during sperm re-
Anderson (2004), basic science researchers trained
trieval in men with SCI. Proceedings of American Spine In-
in physiological function need to appreciate that jury Association. The ASIA abstracts were published in
any improvement their work leads to in quality of Journal of Spinal Cord Medicine, American Paraplegic
life for persons with spinal cord injury, results in Association (United States).
398

Cole, T. and Cole, S. (1981) Sexual attitude reassessment Ferrerio-Velasco, M.E., Barca-Buyo, A., Salvador DeLaBarr-
programs for spinal cord injured adults. Their partners and era, S., Monto, M., Arques, A., Miguens Vazquez, X. and
health care professionals. In: Sha’ked A. (Ed.), Human Rodriguez-Sotillo, A. (2005) Sexual issues in a sample of
Sexual and Rehabilitative Medicine. Sexual functioning after women with spinal cord injury. Spinal Cord, Advance on
Spinal Cord Injury. Williams and Wilkins, Baltimore, pp. Line Publication. Spinal Cord, 43(1): 51–55.
80–90. Giuliano, F. (2004) Vardenafil in challenging – to - treat pa-
Cole, T.M. (1975) Sexuality and the spinal cord injured. In: tients: The RESPITE study Presented symposium 11th
Green R. (Ed.), Human Sexuality: A Health Practitioner’s World Congress of the International Society for Sexual and
Text. Williams and Wilkins, Baltimore, pp. 243–263. Impotence Research. Buenos Aires, Argentina.
Cole, T.M., Chilgrem, R. and Rosenberg, P. (1973) A new Halstead, L.S., Seager, S.W., Houston, J.M., Whitesell, K.,
programme of sex education and counseling for spinal cord Dennis, M. and Nance, P.W. (1993) Relief of spasticity in
injured adults and health care professionals. Paraplegia, 11: men and woman using rectal probe stimulation. Paraplegia,
111–124. 31: 715–721.
Comarr, A.E. and Vigue, M. (1978) Sexual counseling among Jackson, A.B. and Wadley, V. (1999) A multicenter study of
male and female patients with spinal cord injury and/or women’s self-reported reproductive health after spinal cord
cauda equina injury. Am. J. Phys. Med., 57(Part 1): 107–122. injury. Arch. Phys. Med. Rehabil., 80: 1420–1428.
Courtois, F.J., Charvier, K.F., Leriche, A. and Raymond, D.P. Kirshbium, S.C., House, J.G. and O’Connor, K.C. (2002) Silent
(1993) Sexual function in spinal injury men I. Assessing sex- autonomic dysreflexia during a routine bowel program in
ual capacity. Paraplegia, 31: 771–784. persons with traumatic spinal cord injury: A preliminary
Courtois, F.J., Geoffrion, R., Landry, E. and Belanger, M.H. study. Arch. Phys. Med. Rehabil., 83: 1774–1776.
(2004) Reflex and physiologic measures of ejaculation in men Klein, E.A. (1988) The anatomy and physiology of normal male
with spinal cord injury. Arch. Phys. Med. Rehab., 85: sexual function. In: Montague D. (Ed.), Disorders of Male
910–918. Sexual Function. Year Book Medical Publishers, Inc.,
Dannels, A. and Charlifue, S. (2004) The perimenopause expe- Chicago, USA, pp. 2–19.
rience for women with spinal cord injury. SCI Nurs., 21(1): Komisaruk, B.R. and Whipple, B. (1991) Physiological and
9–13. perceptual correlates of orgasm produced by genital or non-
deGroat, W.C. and Booth, A.M. (1980) Physiology of male genital stimulation. In: Kothari P. (Ed.), The Proceedings of
sexual function. Ann. Intern. Med., 92(Pt 2): 329–331. the First International Conference on Orgasm, India. VRP
Derry, F.A., Dinsmore, W.W., Fraser, M., Gardner, B.P., Publishers, Bombay, pp. 69–73.
Glass, G.A., Maytom, M.C. and Smith, M.D. (1998) Efficacy Laessoe, L., Nielsen, J.B., Biering-Sorensen, F. and Sonksen, J.
and safety of oral sildenafil (viagra) in men with erectile dys- (2004) Antispastic effect of penile vibration in men with spi-
function caused by spinal cord injury. Neurology, 51: nal cord injury. Arch. Phys. Med. Rehabil., 85: 919–924.
129–163. Linsenmeyer, T.A. and Perkash, I. (1991) Infertility in men
Donohue, J. and Gebhard, P. (1995) The Kinsey Institute/In- with spinal cord injury. Arch. Phys. Med. Rehab., 72:
diana University report on sexual and spinal cord injury. Sex 747–754.
Disabil., 13(1): 7–84. Masters, W.H. and Johnson, V.E. (1966) Human Sexual
Drory, Y. (2002) Sexual activity and cardiovascular risk. Eur. Response. Little Brown and Co, Boston, pp. 174–176.
Heart J., 4(Suppl. H): 413–418. Maytom, M.C., Derry, F.A., Dinsmore, W.W., Glass, C.A.,
Ekland, M. and Lawrie, B. (2004) How a woman’s sexual ad- Smith, M.D., Orr, M. and Osterloh, I.H. (1999) A two-part
justment after sustaining a spinal cord injury impacts sexual pilot study of sildenafil (Viagra) in men with erectile dys-
health interventions. SCI Nurs., 21(1): 14–19. function caused by spinal cord injury. Spinal Cord, 37:
Elliott, S. (2002) Ejaculation and orgasm: Sexuality in men with 110–116.
SCI. Top Spinal Cord Inj. Rehabil., 8(1): 1–15. McGregor, J.A. and Meeuwsen, J. (1985) Autonomic hype-
Elliott, S. (2003) Sexual dysfunction and infertility in men with rreflexia: A mortal danger for spinal cord-damaged women in
spinal cord disorders. In: Lin V. (Ed.), Spinal Cord Medicine: labor. Am. J. Obstet. Gynecol., 151(3): 330–333.
Principles and Practice. Demos Medical Publishing, New McKenna, K. (1999) The brain is the master organ in sexual
York, pp. 350–365. function: Central nervous system control of male and female
Elliott, S.L. and Krassioukov, A. (2005) Malignant autonomic sexual function. Int. J. Impot. Res., 11(Suppl. 1): S48–S55.
dysreflexia following ejaculation in spinal cord injured men. Nemec, E.D., Mansfield, L. and Kennedy, J.W. (1976) Heart
Spinal Cord, Stockholm Press, England. rate and blood pressure responses during sexual activity in
Elliott, S.L., McBride, K., Ekland, M. and Krassioukov, A. normal males. Am. Heart J., 92(3): 274–277.
(2005) Characteristics of autonomic dysreflexia in men with Pereira, L. (2003) Obstetric management of the patient with
SCI undergoing 296 procedures of vibrostimulation for spinal cord injury. Obstet. Gynecol. Surv., 58(10): 678–687.
sperm retrieval. Proceedings of American Spine Injury As- Pope, C.S., Markenson, G.R., Bayer-Zwirello, L.A. and
sociation. The ASIA abstracts were published in Journal of Maissel, G.S. (2001) Pregnancy complicated by chronic
Spinal Cord Medicine, American Paraplegic Association spinal cord injury and history of autonomic hyperreflexia.
(United States). Obstet. Gynecol., 97(5): 802–803.
399

Reitz, A., Tobe, V., Knapp, P.A. and Schurch, B. (2004) Szasz, G. (1986) Sexual function in the spinal cord injured. In:
Impact of spinal cord injury on sexual health and quality of Bloch R.F. and Basbaum M. (Eds.), Management of Spinal
life. Int. J. Impotence Res., 16: 167–174. Cord Injuries. Williams and Wilkins, Baltimore.
Schmidt, M.H. and Schmidt, H.S. (1993) The ischiocavernosus Szasz, G. and Carpenter, C. (1989) Clinical observations in vi-
and bulbospongiosus muscles in mammalian penile rigidity. bratory stimulation of the penis in men with spinal cord in-
Sleep, 16: 171–183. jury. Arch. Sex. Behav., 18: 461–474.
Sheel, A.W., Krassioukov, A.V., Inglis, J.T. and Elliott, S.L. Talbot, H. (1949) A report on sexual function in paraplegics.
(2005) Autonomic dysreflexia during sperm retrieval in spinal J. Urol., 61: 265–270.
cord Injury: Influence of lesion level and sildenafil citrate. J. Teasell, R.W., Arnold, J.M. and Krassioukov, A. (2000)
Appl. Physiol., 99(July): 53–58. Cardiovascular consequences of loss of supraspinal control
Siosteen, A., Lundqvist, C., Blomstrand, C., Sullivan, L. and of the sympathetic nervous system after spinal cord injury.
Sullivan, M. (1990) Sexual ability, activity, attitudes and Arch. Phys. Med. Rehabil., 81(4): 506–516.
satisfaction as art of adjustment in spinal cord injured Tepper, M.S., Whipple, B., Richard, E. and Komisaruk, B.R.
subjects. Paraplegia, 28: 285–295. (2001) Women with complete spinal cord injury: A phen-
Sipski, M.L., Alexander, C.J. and Rosen, R.C. (1995) Orgasm omenological study of sexual experiences. J. Sex Marital
in women with spinal cord injuries: A laboratory based Ther., 27: 615–623.
assessment. Arch. Phys. Med. Rehab., 76: 811–818. White, M.J., Rintala, D.H., Hart, K.A. and Fuhrer, M.J. (1993)
Sonksen, J., Biering-Sorensen, F. and Kristensen, J.K. (1994) Sexual activities, concerns and interests of women with spinal
Ejaculation by penile vibratory stimulation in men with spi- cord injury living in the Community. Am. J. Phys. Med.
nal cord injured. Paraplegia, 32: 651–660. Rehabil., 72(6): 372–378.
Szasz, G. (1983) Sexual health care. In: Zejdlik C. (Ed.), Yarkony, G.M. and Chen, D. (1995) Sexuality in patients with
Management of the Spinal Cord Injured, Wadsworth Health spinal cord injury. Phys. Med. Rehab., 9(2): 325–343.
Sciences Division. Monterey, California, pp. 125–152.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Published by Elsevier B.V.

CHAPTER 27

Ascending spinal pathways from sexual organs:


effects of chronic spinal lesions

Charles H. Hubscher

Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville,
KY 40292, USA

Abstract: A recent survey of paraplegics indicates that regaining sexual function is of the highest priority
for both males and females (Anderson, K.D. (2004) Targeting recovery: priorities of the spinal cord-injured
population J. Newrotrauma, 21: 1371–1383). Our understanding of the neural pathways and mechanisms
underlying sexual behavior and function is limited at the present time. More studies are obviously needed to
direct experiments geared toward developing effective therapeutic interventions. In this chapter, a review of
studies on the processing of sensory inputs from the male and female reproductive organs is presented with
a review of what is known about the location of ascending spinal pathways conveying this information. The
effect of spinal cord injury on sexual function and the problems that ensue are discussed.

Afferent innervation of the male and female Peripheral structures of primary importance for
reproductive organs reproduction include pelvic visceral organs, the
external genitalia and somatic elements of the
Compared to the voluminous literature on the hindquarters. The internal pelvic visceral organs
neural pathways involved in the sensation and involved in reproduction include the ovaries,
motor control of the limbs, there have been oviduct, uterus, cervix and vagina in females and
relatively few studies on the male and female the testis, epididymis, vas deferens, seminal vesicle,
reproductive organs. This may partly be due to the ejaculatory duct and prostate in males. These
complexity of the neural pathways involved in organs are innervated by visceral afferent fibers,
sexual behavior and functions, which include which consist primarily of unmyelinated and some
somatic, sympathetic, and parasympathetic nerves thinly myelinated fibers that convey information
integrated with two interconnected portions of the centrally mainly via the hypogastric and pelvic
spinal cord, which in turn make connections with nerves. Organ-specific and organ-characteristic
the brainstem and cerebral cortex (de Groat and information is conveyed in a rostro-caudal topo-
Booth, 1980, 1993; de Groat et al., 1981; deGroat graphic array to the caudal spinal cord (Berkley
and Steers, 1988; Berkley and Hubscher, 1995b; and Hubscher, 1995b). The external genitalia
Giuliano et al., 1995; McKenna, 2000). The main (clitoris/penis), skin and muscles of the perineum
focus of this review is on experiments that use are innervated by somatic myelinated and un-
rodents, since the majority of studies on sexual myelinated afferent fibers of all sizes. These fibers
function use rats to model humans. convey information to the lower lumbar and upper
sacral segments of the spinal cord mainly via the
pudendal nerve, but also via branches of the pelvic,
Corresponding author. Tel.: +(502)852-3058; genitofemoral, ilioinguinal and anococcygeal
Fax: +(502)852-6228; E-mail: chhubs01@louisville.edu nerves (Peters et al., 1987; Bonica, 1990).

DOI: 10.1016/S0079-6123(05)52027-2 401


402

Hypogastric nerve necessary to evoke a response in the hypogastric


nerve (Berkley et al., 1987, 1993c). Behavioral
The hypogastric nerve contains both afferent and studies also show that rats will engage in an escape
efferent fibers. Removal of one axonal component behavior only at noxious levels of uterine disten-
(e.g. dorsal root ganglionectomy from the 12th sion, and that this behavior is eliminated after
thoracic (T12) to 2nd sacral (S2) ganglia to remove bilateral hypogastric neurectomy (Berkley et al.,
sensory fibers) demonstrated that most of the 1995b; Temple et al., 1999). The noxious level
axonal population is efferent (92%) (Hulsebosch is equivalent to the distension volume at which
and Coggeshall, 1982). Cell bodies of hypogastric ischemia of the uterine vessels is produced. Blood
nerve afferents have been identified (using the vessels in the uterus of the non-pregnant rat have
transganglionic transport of horseradish per- been shown to be highly innervated (Garfield,
oxidase) from T10 to the 4th lumbar (L4) 1986; Haase et al., 1997). Uterine distension late in
ganglion (Neuhuber, 1982). In females, a variety pregnancy leads to hormonal changes, which ac-
of tracing techniques and electrophysiological count for the initiation of delivery and onset of
recordings have been used to demonstrate that maternal behavior (Graber and Kristal, 1977).
the T13–L3 (predominantly) dorsal root ganglion, Whether or not this nerve serves a similar role in
via the hypogastric nerve (Nance et al., 1988), males (re-nociception) is not known.
provide afferent innervation to the cervix, uterus
and broad ligament (Peters et al., 1987; Berkley Pelvic nerve
et al., 1988, 1993a; Nance et al., 1988). Sensory
axons in the hypogastric nerve of male and female The pelvic nerve in the male rat contains both
adult rats (with cell bodies concentrated in the L1 afferent (34%) and efferent (66%) fibers with the
and L2 dorsal root ganglia) are similar in number, majority of the myelinated axons and slightly less
110 and 148, respectively. This is not the case than half of the unmyelinated axons being sensory
for efferent hypogastric fibers. In male rats, an (Hulsebosch and Coggeshall, 1982). Cell bodies of
average of 415 sympathetic preganglionic neurons pelvic nerve afferents (95%) are located in the
were found, while in females, only 110 were found L6 and S1 dorsal root ganglia (Nadelhaft and
(Nadelhaft and McKenna, 1987). Booth, 1984). In the female rat, the pelvic nerve
The reproductive structures innervated by the was shown to bifurcate into a viscero-cutaneous
hypogastric nerve are involved in contraction and branch and a muscular branch shortly after
movement of substances. In the male, stimulation separating from the lumbosacral trunk (Pacheco
of the hypogastric nerve produces contractions of et al., 1989), with both branches conveying sensory
the distal vas deferens and seminal vesicle but and motor information. In females, afferent
not erection (Quinlan et al., 1989). Thus, the innervation of the cervical portion of the uterus,
hypogastric nerve is believed to play a role in vagina, base of the bladder and rectum via the
sperm motility (ejaculation). Bilateral sectioning of pelvic nerve was demonstrated with HRP injec-
the hypogastric nerve has no effect on mating be- tions of the L6 and S1 dorsal root ganglia (Nance
havior (Larsson and Swedin, 1971). These findings et al., 1988). Electrophysiological studies showed
are consistent with those of Bacq (1930) who that pelvic nerve fibers have receptive fields
showed that sympathetic denervation affects sem- extending from the uterine cervix to the distal
inal discharge but not mounts and intromissions. end of the vagina (Berkley et al., 1990). Sensory
In the female, stimulation of the hypogastric nerve input from the midline perineal skin and viscera
produces contractions of the uterus (Sato et al., travel through the viscerocutaneous branch and
1989). Hypogastric afferents in the female rat from the iliococcygeus and pubococcygeus muscles
convey information about intense stimulation through the muscular branch (Pacheco et al.,
(mechanical and chemical) to the central nervous 1989).
system (Berkley et al., 1993c). Intense mechanical Stimulation of the pelvic nerve produces penile
or chemical stimulation of the uterus or cervix is erection and contractions of the distal vas deferens
403

and seminal vesicle in the male (Quinlan et al., pelvic nerve has also been shown to be important
1989) and produces displacement of the vaginal for the display of paced mating behaviors (Erskine,
wall (Pacheco et al., 1989) and contractions of the 1992). Mating pattern (position, contact) is affected
uterus in the female (Sato et al., 1989). Although in cervically denervated females (Diakow, 1970).
there is a substantial amount of information
available regarding the function of the pelvic nerve Pudendal nerve
in the female rat, less experimentation has been
done in the male. In females, pelvic afferents serve The pudendal nerve in the male and female rat,
important informative, affective and behavioral which shares its origin with the pelvic nerve, runs
functions during mating, conception and parturi- from the L6–S1 trunk to the sacral plexus and
tion. The pelvic nerve, which has both internal and contains fibers that arise from the L6–S1 trunk as
external sensory fields, conveys information that is well as the lumbosacral trunk (Reiner et al., 1981;
considerably different from the hypogastric nerve. McKenna and Nadelhaft, 1986; Pacheco et al.,
The external sensory field is involved in triggering 1997). Revised nomenclature of the pudendal
lordosis and may play a role in positioning for nerve in the male rat includes three components:
intromission, whereas the internal fields are a sensory branch (dorsal nerve of the penis
sensitive to vaginal stimulation, especially at the (Calaresu, 1970; Nunez et al., 1986)) that supplies
cervical end of the vagina (Peters et al., 1987). the penis and prepuce, a motor branch that supplies
Responses to noxious pinch stimulation can the perineal muscles (McKenna and Nadelhaft,
be attenuated by probing the vaginal cervix 1986) and cutaneous branches that supply the
(Komisaruk and Wallman, 1977). It is hypothe- scrotum (Pacheco et al., 1997). The glans penis has
sized that this attenuation may be especially sig- an abundance of free nerve endings, and the pri-
nificant during parturition, because it may reduce mary afferent population in the dorsal nerve of the
stress, which would otherwise interfere with ma- penis contains both slowly and rapidly adapting
ternal behavior. Bilateral sectioning of the pelvic fibers (Kitchell et al., 1982; Johnson and Halata,
nerve renders the entire intravaginal mucosa com- 1991). In the female, afferents arising from the
pletely insensitive to gentle and intense mechanical clitoris and perigenital skin send information to
stimulation, and prevents pseudopregnancy (arrest the L6 and S1 dorsal root ganglia through the pu-
of estrus) upon intense mechanical stimulation of dendal nerve sensory branch (McKenna and
the cervix (Kollar, 1953; Carlson and De Feo, Nadelhaft, 1986). A few of the dorsal root gangli-
1965; Ross et al., 1979). Prolongation of the on cells at L6 respond to electrical stimulation of
process of parturition is seen in bilaterally pelvic myelinated fibers in both the pudendal and sciatic
neurectomized rats due to the elimination of the nerves (Pierau et al., 1982; Taylor et al., 1982),
fetus-expulsion reflex (contraction of abdominal demonstrating prespinal convergence of sensory
muscles and diaphragm) in response to mechanical nerve fibers. The pudendal nerve in male and
stimulation of the upper vagina and cervix female rats varies greatly, however, in both size
(Higuchi et al., 1987). Only the sensory branch and number of afferent and efferent neurons. For
of the pelvic nerve is required for normal vaginal example, there are twice as many dorsal root
delivery in the rat (Burden et al., 1990). Pelvic ganglion neurons from the pudendal nerve sensory
neurectomy also eliminates the lordosis response branch in the male rat than in the female
(Carlson and De Feo, 1965). Komisaruk and (McKenna and Nadelhaft, 1986) and in cross sec-
Wallman (1977) hypothesize that immobilization tion, the sensory branch is three times larger in males
during lordosis could facilitate sperm transport (Moore and White, 1996). One well-known differ-
and consequently pregnancy. The lordosis reflex ence between the male and female rat, for example,
has been studied in the male, but not in the context involves the spinal nucleus of the bulbocavernosus
of neural control [for example, effects of cholecys- muscle (Breedlove and Arnold, 1980). Adult
tokinin (Bloch et al., 1988); modulation by the main female rats lack both this spinal nucleus and its
olfactory system (Chateau and Aron, 1990)]. The target muscles (perineal bulbocavernosus). The
404

presence of the nucleus is dependent on the action the ovaries, oviduct, the entire uterine horns and
of androgens at an early stage of life (Breedlove cervix (Burden et al., 1983; Ortega-Villalobos
and Arnold, 1980; Breedlove, 1985). et al., 1990; Collins et al., 1999). Abdominal vago-
In males, bilateral transection of the pudendal tomy disrupts the rat’s estrous cycle (Burden et al.,
nerves, affecting the dorsal nerve of the penis, 1981). Whether or not the vagus innervates
prevents erection, intromission, ejaculation, sexual portions of the male urogenital tract is unknown.
motivation and some (mounts and intromissions),
but not all (time-outs and mount-bout periods),
Central processing of inputs from the male and
mating behaviors (Larsson and Sodersten, 1973;
female reproductive organs
Lodder and Zeilmaker, 1976). Stimulation of the
pudendal nerve does not, however, produce penile
Sensory information conveyed centrally from the
erection (Quinlan et al., 1989). Desensitization of
internal reproductive organs has important conse-
the penis by local anesthesia also causes an
quences for both reproduction and for sensation
impairment of reflexogenic erection, intromission
(from pleasure to pain). In females for example,
and ejaculation (Hart and Leedy, 1985). In fe-
cervix stimulation in both rats and humans pro-
males, application of the local anesthetic lidocaine
duces effects that are of considerable importance
to the clitoris (sensory innervation by pudendal
for reproductive behaviors such as during mating
nerve) prevents the occurrence of intromission
and parturition (Komisaruk and Whipple, 1988),
patterns (Baum et al., 1974). When the pudendal
and for pain, such as is evident in women during
nerve is sectioned, there is a significant decrease in
invasive gynecological procedures or for many,
lordosis, although this decrease can be overridden
with deep penile penetration during vaginal inter-
by elevated estrogen levels (Kow and Pfaff, 1973;
course (dyspareunia — affecting 10–15% of sex-
Kow, 1976).
ually active women) (Meana et al., 1997).
Ovarian/testicular nerves
Spinal processing
Sensory fibers in the ovarian plexus nerves and the
superior ovarian nerves innervate the ovaries, ovi- In males, the lumbosacral segments (L5–S1 in the
duct and rostral pole of the uterine horns (Burden rat) contain a center for erection and the expulsive
and Lawrence, 1978; Baljet and Drukker, 1979, part of ejaculation mediated by preganglionic
1980; Marchetti et al., 1987; Klein et al., 1989; parasympathetic motor axons in the pelvic nerve
Berkley and Hubscher, 1995b; Serghini et al., and somatic motor axons in the pudendal nerve
1997). Dorsal root ganglion neurons from T10 to supplying the striated perineal muscles of the
L2 are labeled unilaterally following injection of pelvic floor. Stimulation of the dorsal nerve of
HRP into the ipsilateral ovary (Burden et al., 1983; the penis induces Fos labeling in the dorsal horn,
Nance et al., 1988). In males, the testis is supplied dorsal gray commissure and sacral parasympa-
by the superior spermatic nerve (Kumazawa, thetic nucleus (Rampin et al., 1997). Electrophys-
1986). Most of the testicular afferents are poly- iological techniques have been used to investigate
modal, responding to mechanical, chemical and single spinal cord interneurons in the dorsal horn
thermal (noxious heat) stimulation (Kumazawa and intermediate zone of primarily L6–S1 that re-
et al., 1995). In dogs, HRP injection labels dorsal ceive input from dorsal nerve of the penis afferents
root ganglia between T10 and L4, with most cells (Johnson, 1989). All of the penile interneurons
labeled in L1 and L2 (Kumazawa et al., 1995). exhibit receptive fields on the penis that are sig-
nificantly larger than the receptive fields for single
Vagus nerve primary afferent neurons, thereby demonstrating
a central convergence of penile sensory input.
In female rats, the innervation of the reproductive Almost all of the penile interneurons have recep-
tract by abdominal branches of the vagus includes tive fields on both sides of the body and their
405

response characteristics strongly suggest a mo- gigantocellularis, nucleus reticularis gigantocellularis


nosynaptic input from both ipsilateral and cont- pars alpha, lateral paragigantocellular reticular
ralateral dorsal nerve of the penis fibers. There is nucleus, raphe pallidus, raphe magnus, A5 nor-
also an extensive representation from the distal adrenergic cell groups, parapyramidal area, lateral
glans (cup) region in these spinal cord interneu- vestibular nucleus, Barrington’s nucleus, nucleus
rons. Afferent fibers in the dorsal nerve of the locus coeruleus, nucleus subcoeruleus, caudal pon-
penis produce bilateral (crossed and uncrossed) tine reticular nucleus, periaqueductal gray, intra-
reflex facilitation of pudendal motoneurons locat- laminar thalamic nuclei, ventroposterolateral
ed in L5–L6. A lumbar (L3–L4) reflex region for thalamic nucleus, ventrolateral thalamic nucleus,
ejaculation has also been proposed (Truitt and ventromedial thalamic nucleus, anterior thalamic
Coolen, 2002) that depends on input from afferent nuclei, nucleus submedius, subparafascicular nu-
systems releasing Substance P. cleus, medial preoptic area, lateral septum, bed
In females, information from the reproductive nucleus of the stria terminalis, ventromedial hypo-
organs is conveyed in a rostro-caudal topographic thalamus, hypothalamic paraventricular nucleus,
array to the caudal spinal cord (Berkley et al., medial amygdala, globus pallidus, mesencephalic
1993c; Berkley and Hubscher, 1995b). Electro- central gray and cortex (Allen et al., 1981;
physiological and anatomical studies indicate that Haldeman et al., 1982; Haskins and Moss, 1983;
there is an extensive system of neurons in thoracic, Akaishi et al., 1988; Shen et al., 1990; Baum and
lumbar and sacral spinal cord that receive repro- Everitt, 1992; Berkley et al., 1993a, 1995a; Marson
ductive organ input (Berkley et al., 1993b; Lee and et al., 1993; Tetel et al., 1993; Hubscher and
Erskine, 1996, 2000). The cervix, which is inner- Berkley, 1994; Berkley and Hubscher, 1995b;
vated by both the hypogastric and pelvic nerves, Hubscher and Johnson, 1996, 2003; Pfaus et al.,
has inputs to dorsal horn neurons at all three lev- 1996; Bradley et al., 1998; Papka et al., 1998;
els, although the neurons are concentrated vent- Van der Horst and Holstege, 1998; Ding et al.,
rally in the dorsal horn at T13–L1 and throughout 1999; Lee and Erskine, 2000). Areas receiving
the dorsal horn at L4–L5 and L6–S1 segments reproductive organ input such as those targeted by
(Berkley et al., 1993b). Cervix-responsive dorsal Hubscher and colleagues, for example, which in-
horn neurons in both regions receive convergent clude the nucleus reticularis gigantocellularis and
inputs from other pelvic organs as well as cutane- surrounding nuclei within the medullary reticular
ous regions, although the receptive fields tend to formation, the nucleus gracilis and solitarius, and
be larger at T13–L1 and more confined to the various subregions of the thalamus have also been
perineum for the neurons at L6-S2 that are located shown to process and relay a vast array of con-
in the dorsal part of the dorsal horn (Berkley et al., vergent somatic and pelvic visceral sensory inputs
1993b). Many cervix-responsive neurons at L6–S2 (Hubscher and Berkley, 1994; Berkley and
respond to uterine distension by being inhibited. Hubscher, 1995a; Hubscher and Johnson, 1996,
This uterine input originates from distant roots 2001, 2003). Our electrophysiological data from
(uterus innervated by the hypogastric nerve), as extracellular single unit recordings in the rostral
shown in experiments, where T13–L2 roots were ventromedial medulla in male rats has demon-
sectioned bilaterally (Wall et al., 1993). How these strated a significant degree of convergence from
interactions sculpt the actions of these neurons for the reproductive organs (dorsal nerve of penis;
various aspects of reproduction is still unclear. pelvic nerve) and rostral skin areas (including the
ears and forepaws) (Hubscher and Johnson, 1996).
Supraspinal processing Examples from recordings obtained in a male and
female rat are provided in Fig. 1. The majority of
Many regions throughout the brain receive input these neuronal responses were to noxious levels of
from the reproductive organs. These regions in- stimulation. Some neurons required windup (re-
clude (but are not limited to) the nucleus tractus petitive stimuli) to respond to bilateral dorsal nerve
solitarius, nucleus gracilis, nucleus reticularis of the penis (or clitoris) stimulation. In males,
406

it is hypothesized that these neurons may be firing likely involved in mating, some likely play a role in
near the ejaculatory threshold (Hubscher and nociceptive processing as well (Bowsher, 1976;
Johnson, 1996), which depends on a gradual Peschanski and Besson, 1984; Zhuo and Gebhart,
build-up of activity produced by multiple intr- 1991; Berkley et al., 1993a; Al-Chaer et al., 1996b;
omissions (Sachs and Meisel, 1988). Large lesions Yang et al., 1998; Mason, 2001).
that include the gigantocellular (ventral and pars
alpha) and lateral paragigantocellular nuclei have Ascending pathways
been shown to affect ejaculatory bursts in perineal
muscles (Marson and McKenna, 1990). Limited information is available on the location of
In female rats, neurons in the nucleus reticularis ascending spinal pathways that convey information
gigantocellularis and surrounding regions respond- originating from the internal reproductive organs
ing to probing of the vaginal canal (Hornby and and external genitalia. The results of several stud-
Rose, 1976; Hubscher and Johnson, 2001) have ies, when taken together, indicate that there are
been shown to be involved in female circuitry likely many spinal pathways that convey this input
responsible for lordosis behavior (Modianos and to the brain, and these projections are likely all
Pfaff, 1979; Schwartz-Giblin et al., 1996; Daniels bilateral. These pathways include most, if not
et al., 1999). Although many of these neurons are all, of the following: dorsal column, post-synaptic

Fig. 1. Example on the left shows excitatory responses in a male rat of a single neuron located in right n. reticularis gigantocellularis
pars alpha to uni- and bi-lateral stimulation of the pelvic nerve and dorsal nerve of the penis as well as stimulation of the abdominal
branch of the vagus, distention of the colon and mechanical stimulation of the penis. This neuron had excitatory responses to pinching
of the entire body, including the face, dorsal trunk, forepaw and ears (responses shown are only to pinch of the glans penis). Note that
the only low threshold responses obtained were from stroking of the penis (rest of body responded to noxious pinch). Example on the
right shows excitatory responses in a female rat of a single neuron located in left nucleus reticularis gigantocellularis to bilateral (b)
stimulation of the dorsal nerve of the clitoris (DNC) and pelvic nerve (PN), pressure on the cervix, distention of the left uterine horn
and distal colon. The same neuron also had excitatory responses to distention of the right uterine horn and bladder, and to bilateral
stimulation (pinching) of the perineum, anus, trunk, ears, toes of the hindpaw and forepaw (not shown). Note that this particular
neuron did not respond to distention of the vaginal canal (not shown); the vagus was not tested. For both neurons shown, note the
stimulus artifacts at the onset of the electrical nerve stimulation train (see arrows, which indicate the onset of the electrical stimulation
train). The horizontal bar indicates the duration of a natural stimulus. The two large dots with smaller dots in between indicate the
onset and end, respectively, of maximal balloon stimulus intensity (by vol.). Adapted from Hubscher et al. (2004b).
407

dorsal column, spinoreticular, spinothalamic, spino- At the present time, it is unclear whether these
solitary, spinoparabrachial, spinohypothalamic, projections reach the medullary reticular forma-
spinoamygdalar, spinomesencephalic and spinocer- tion directly or indirectly via one or more synaptic
ebellar pathways (Menetrey and de Pommery, 1991; contacts in other regions of the brain. There is
Berkley and Hubscher, 1995b). little evidence in the literature that would strongly
In males, the most extensive data exist for support or refute either of the two possibilities.
central projections of input from the penis. The For example, the location of direct spinoreticular
location of projections originating from the dorsal projections ascending in the white matter of the rat
nerve of the penis to the medullary reticular spinal cord is unknown, with the exception of one
formation neurons has been examined (Hubscher anatomical study that used large medullary injec-
and Johnson, 1999, 2004). The results from these tions of horseradish peroxidase to demonstrate the
studies indicate that there are at least two central loss of retrogradely labeled L4–L6 cells around the
projections originating from the male genitalia central canal following a ventrolateral quadrant
located in the dorsal quadrant at the mid-thoracic lesion at T12 (Nahin et al., 1986). The evidence
spinal level (conveyed rostrally from lumbosacral from that study relative to Hubscher and Johnson
dorsal horn cells to the medullary reticular forma- (1999) is inconclusive, since in Nahin et al. (i) the
tion). All responses to bilateral electrical stimula- lesions appeared to encroach upon the ventral
tion of the dorsal nerve of the penis were lost portion of the dorsolateral quadrant, (ii) the
following a complete dorsal hemisection. Specifi- lesions were at T12 (T7/T8 in Hubscher and
cally, responses to gentle stimulation of the penis Johnson) and there is evidence for a dorsal shift
were lost following either an acute or chronic of axons as they ascend rostrally (Willis and
mid-thoracic dorsal column lesion (bilateral). Coggeshall, 1991), and (iii) the study focused on
Responses to pinching of the penis were not lost cells adjacent to the central canal, which are some
following a complete dorsal column lesion or of many dorsal horn neurons to (a) project directly
following a subsequent unilateral lesion of the upon neurons in the medullary reticular formation
dorsolateral quadrant, but were lost after both (Menetrey et al., 1980; Chaouch et al., 1983) and
dorsolateral quadrant’s had been lesioned. It (b) respond to bilateral dorsal nerve of the penis
is important to note that, although a chronic stimulation (Johnson, 1989). There is also evidence
mid-thoracic spinal cord dorsal hemisection for the existence of several different ascending
eliminates medullary reticular formation neuronal pathways within the dorsolateral quadrant of the
responses to bilateral dorsal nerve of the penis rat, any of which could provide indirect projec-
stimulation (Hubscher and Johnson, 1999), this tions to medullary reticular formation and thus,
electrical search stimulus activates dorsal nerve may convey pelvic and visceral information. Such
of the penis afferents in the A-b and A-d range pathways include a spinomesencephalic pathway
only. Therefore, additional pathways likely exist and a spinohypothalamic pathway (Zemlan et al.,
that convey, to the medullary reticular formation, 1978; Menetrey et al., 1980; McMahon and Wall,
information originating from the large population 1983; Cliffer et al., 1991; Burstein et al., 1996;
of C-fibers that are contained within the Kostarczyk et al., 1997).
dorsal nerve of the penis (Johnson and Halata, In addition, the medullary reticular formation
1991). Candidates include spinothalamic or spin- has been shown in the rat to be interconnected
oreticulothalamic pathways in the ventrolateral with both the gracilis nucleus and the solitary nu-
quadrant (Giesler et al., 1981; Peschanski and cleus in the caudal brainstem (Tomasulo and
Besson, 1984). Specific thalamic subregions relay Emmers, 1972; Odutola, 1977; Jean, 1991; Mtui
extensive inputs from the dorsal nerve of the et al., 1995). In females, neurons in the gracilis nu-
penis (Truitt and Coolen, 2002; Hubscher and cleus have been shown to receive input (albeit in-
Johnson, 2003). Other thalamic subregions may be direct) from female reproductive organs (Hubscher,
associated with ejaculation (Truitt and Coolen, 1994; Berkley and Hubscher, 1995a). The dorsal
2002). column-medial lemniscal pathway has therefore
408

been implicated as a source of input about lesion. In male patients with clinically complete
innocuous and noxious events from pelvic viscera spinal cord injuries cranial to T9–10, for example,
(uterus, cervix, vagina and colon) and skin to the spinal reflex arcs for erection and ejaculation
lateral thalamus, suggesting that both the dorsal are intact, although supraspinal input has been
column-medial lemniscal and spinothalamic removed (see Sachs and Bitran, 1990). In females,
pathways are involved in pain (Hubscher, 1994; having a spinal cord injury has important conse-
Berkley and Hubscher, 1995a). The dorsal quences for pregnancy (Cross et al., 1992), such as
columns have also been shown to convey visceral frequent urinary tract infections, and the location
nociceptive inputs from the colon (Al-Chaer et al., of the spinal lesion has important consequences
1996a, b; Willis et al., 1999) and pancreas for childbirth (Sauer and Harvey, 1993), since
(Houghton et al., 1997). Ascending projections damage above T5 can result in the life-threatening
from neurons in the area around the central canal, complication of autonomic dysreflexia (Crosby
an area known to receive primary afferent input et al., 1992).
from somatic and pelvic visceral structures in the
cat (Honda, 1985), has been implicated in the
transmission of second order afferent information Sensation
for visceral nociception to the gracilis nucleus
(Wang et al., 1999). Most male spinal cord-injured patients with intact
The solitary nucleus has also been shown to reflex arcs demonstrate reflexogenic erections of
relay input (potentially noxious) from pelvic varying degrees in response to very slight mechan-
visceral sources centrally (from the cervix, vagina ical stimulation of the penis (Sarkarati et al., 1987;
and uterus), via both a spino-solitary and Ver Voort, 1987). Although these erections are
vagal-solitary pathway (Hubscher and Berkley, easily initiated, they are not easily sustained or
1994, 1995; Komisaruk et al., 1996). Different firm enough for vaginal penetration (Bodner et al.,
pathways were identified for different portions of 1987), which has been proposed to be a result of
the reproductive tract, which involved electrophys- altered penile sensitivity (Goldstein, 1988). Studies
iological recording of responses to pelvic organ show that central pathway impairment and not
stimulation in the solitary nucleus, pre- and post- peripheral pathways may play a role in erectile
combinations of acute spinal transection and bi- dysfunction in diabetic patients (Sartucci et al.,
lateral vagotomy (Hubscher and Berkley, 1995). 1999). In cases where the spinal reflex arc is dam-
The results demonstrating a vagal-solitary projec- aged (such as for conus medullaris or cauda equina
tion from the uterus are consistent with anatom- injury), sensations are completely absent or signif-
ical tracing and other types of experiments icantly diminished (Pavlakis et al., 1983).
(Ortega-Villalobos et al., 1990; Collins et al., Among women with spinal cord injuries, only
1999; Guevara-Guzman et al., 2001). In addition, some cannot achieve orgasm, although most of
although bilateral vagotomy had an effect on the those with lower motor neuron dysfunction cannot
responsiveness of neurons in the solitary nucleus to achieve orgasm (Sipski et al., 2001). For those
cervix/vaginal stimulation (suggestive of an ana- spinal cord-injured women who can achieve or-
tomical connection), the responses were only elim- gasm, time to orgasm significantly increases versus
inated after a subsequent acute spinal transection able-bodied controls (Sipski et al., 2001). A survey
(Hubscher, 1994; Hubscher and Berkley, 1995). shows that following injury, the majority of wom-
en are satisfied with their sexual experiences
(Charlifue et al., 1992). Women with spinal cord
Clinical implications of damage to these spinal injuries can also perceive menstrual cramps (Axel,
pathways: males versus females 1982). There is growing evidence of a nociceptive
vagal-solitary pathway from the vaginocervix
In man, the degree of sexual dysfunction caused region in humans with clinically complete spinal
by spinal cord injury depends on the level of the cord injuries (Komisaruk et al., 1997; Komisaruk
409

and Sansone, 2003; Komisaruk et al., 2004). Such et al., 1992; Westgren and Levi, 1994). A delay in
a pathway is consistent with some of the basic the reproductive cycle has also been found exper-
scientific studies that have been done using rats imentally in adult rats (44%) following contusion
(Ortega-Villalobos et al., 1990; Hubscher and injury at the mid-thoracic spinal level (Hubscher
Berkley, 1994, 1995; Komisaruk et al., 1996; Collins et al., 2004a). Only a minority of spinal cord-in-
et al., 1999). However, in pregnancy, perception of jured women report heavier, longer and irregular
fetal movement and uterine contractions does not periods post injury, although many report changes
occur in women with injuries above T8 (Sauer and in frequency and amount of vaginal discharge
Harvey, 1993). (Charlifue et al., 1992). Thus, fertility is less of an
issue for females than males with spinal cord in-
jury. Although there are significant risks associat-
Fertility ed with pregnancy following spinal cord injury,
most women have full-term healthy babies (Cross
Infertility in males relates to impairments in ejac- et al., 1991, 1992; Burns and Jackson, 2001) and
ulation and poor quality of semen once ejaculation succeed in parenting despite their physical limita-
is achieved (Beretta et al., 1989; Linsenmeyer et al., tions (Westgren and Levi, 1994; Alexander et al.,
1994; Sedor and Hirsch, 1995). In 95% of male 2002).
spinal patients with lesions cranial to T10, ejacu-
lation is severely impaired or impossible (Ver
Voort, 1987; Seftel et al., 1991). Despite the intact References
spinal reflex arc, the segmental ejaculatory reflex
circuitry may be dependent on (1) supraspinal Akaishi, T., Robbins, A., Sakuma, Y. and Sato, Y. (1988)
facilitation or disinhibition and/or (2) the integrity Neural inputs from the uterus to the paraventricular magno-
of a spino–bulbo–spinal loop for the coordination cellular neurons in the rat. Neurosci. Lett., 84: 57–62.
of perineal muscle activities. Thus, any decrease in Al-Chaer, E.D., Lawand, N.B., Westlund, K.N. and Willis,
W.D. (1996a) Pelvic visceral input into the nucleus gracilis is
penile sensation resulting from damage to ascend- largely mediated by the postsynaptic dorsal column pathway.
ing pathways would affect this ejaculatory circuit- J. Neurophysiol., 76: 2675–2690.
ry (mechanisms discussed in Chapter 28 by Al-Chaer, E.D., Lawand, N.B., Westlund, K.N. and Willis,
Johnson in this book). There are a number of W.D. (1996b) Visceral nociceptive input into the ventral
techniques used to assist spinal cord-injured males posterolateral nucleus of the thalamus: a new function for the
dorsal column pathway. J. Neurophysiol., 76: 2661–2674.
with fertility (Beretta et al., 1989; Chung et al., Alexander, C.J., Hwang, K. and Sipski, M.L. (2002) Mothers
1995; Dahlberg et al., 1995; Nehra et al., 1996). with spinal cord injuries: impact on marital, family, and
Many spinal cord-injured patients who do not re- children’s adjustment. Arch. Phys. Med. Rehabil., 83: 24–30.
spond to normal tactile stimulation of the penis Allen, T.O., Adler, N.T., Greenberg, J.H. and Reivich, M.
(1981) Vaginocervical stimulation selectively increases meta-
may ejaculate to very intense vibratory stimulation
bolic activity in the rat brain. Science, 211: 1070–1072.
of the ventral penile midline (Sonksen and Ohl, Axel, S.J. (1982) Spinal cord injured women’s concerns: men-
2002) suggesting that massive recruitment of all struation and pregnancy. Rehabil. Nurs., 7: 10–15.
low and high threshold penile mechanoreceptive Bacq, Z.M. (1930) Impotence of the male rodent after sympa-
afferent neurons can provide enough input to the thetic denervation of the genital organs. In: From the Lab-
spinal ejaculatory circuit. Quality of semen has oratories of Physiology in the Harvard Medical School,
pp. 321–330.
been shown to vary with different assisted ejacu- Baljet, B. and Drukker, J. (1979) The extrinsic innervation of
lation procedures, with vibratory stimulation the abdominal organs in the female rat. Acta Anat., 104:
being better than electroejaculation (Brackett 243–267.
et al., 1997). Baljet, B. and Drukker, J. (1980) The extrinsic innervation of
the pelvic organs in the female rat. Acta Anat., 107: 241–267.
In females, although menses is initially altered
Baum, M.J. and Everitt, B.J. (1992) Increased expression of c-
(on average 5 months in 60% of patients) follow- fos in the medial preoptic area after mating in male rats: role
ing spinal cord injury, cycling resumes to pre- of afferent inputs from the medial amygdala and midbrain
injury status (Comarr, 1966; Axel, 1982; Charlifue central tegmental field. J. Neurosci., 50: 627–646.
410

Baum, M.J., Sodersten, P. and Vreeburg, J.T. (1974) Mounting Bradley, W.E., Farrell, D.F. and Ojemann, G.A. (1998) Human
and receptive behavior in the ovariectomized female rat: in- cerebrocortical potentials evoked by stimulation of the dorsal
fluence of estradiol, dihydrotestosterone, and genital an- nerve of the penis. Somatosens Mot. Res., 15: 118–127.
esthetization. Horm. Behav., 5: 175–190. Breedlove, S.M. (1985) Hormonal control of the anatomical
Beretta, G., Chelo, E. and Zanollo, A. (1989) Reproductive specificity of motoneuron-to-muscle innervation in rats. Sci-
aspects in spinal cord injured males. Paraplegia, 27: 113–118. ence, 227: 1357–1359.
Berkley, K.J., Benoist, J.M., Gautron, M. and Guilbaud, G. Breedlove, S.M. and Arnold, A.P. (1980) Hormone accumula-
(1995a) Responses of neurons in the caudal intralaminar tion in a sexually dimorphic motor nucleus of the rat spinal
thalamic complex of the rat to stimulation of the uterus, va- cord. Science, 210: 564–566.
gina, cervix, colon and skin. Brain Res., 695: 92–95. Burden, H.W. and Lawrence Jr., I.E. (1978) Experimental
Berkley, K.J., Guilbaud, G., Benoist, J.M. and Gautron, M. studies on the acetylcholinesterase-positive nerves in the ova-
(1993a) Responses of neurons in and near the thalamic vent- ry of the rat. Anat. Rec., 190: 233–241.
robasal complex of the rat to stimulation of uterus, cervix, Burden, H.W., Lawrence Jr., I.E., Louis, T.M. and Hodson,
vagina, colon and skin. J. Neurophysiol., 69: 557–568. C.A. (1981) Effect of abdominal vagotomy on the estrous
Berkley, K.J., Hotta, H., Robbins, A. and Sato, Y. (1990) cycle of the rat and the induction of pseudopregnancy. Ne-
Functional properties of afferent fibers supplying reproduc- uroendocrinology, 33: 218–222.
tive and other pelvic organs in pelvic nerve of female rat. J. Burden, H.W., Leonard, M., Smith, C.P. and Lawrence Jr., I.E.
Neurophysiol., 63: 256–272. (1983) The sensory innervation of the ovary: a horseradish
Berkley, K.J. and Hubscher, C.H. (1995a) Are there separate peroxidase study in the rat. Anat. Rec., 207: 623–627.
central nervous system pathways for touch and pain? Nat. Burden, H.W., Price, G.T., Renegar, R.H. and Hodson, C.A.
Med., 1: 766–773. (1990) Effects of peripheral nerve lesions during pregnancy
Berkley, K.J. and Hubscher, C.H. (1995b) Visceral and somatic on parturition in rats. Anat. Embryol., 182: 499–501.
sensory tracks through the neuraxis and their relation to pain: Burns, A.S. and Jackson, A.B. (2001) Gynecologic and repro-
lessons from the rat female reproductive system. In: Gebhart ductive issues in women with spinal cord injury. Phys. Med.
G.F. (Ed.), Visceral Pain. IASP Press, Seattle, pp. 195–216. Rehabil. Clin. N. Am., 12: 183–199.
Berkley, K.J., Hubscher, C.H. and Wall, P.D. (1993b) Neu- Burstein, R., Falkowsky, O., Borsook, D. and Strassman, A.
ronal responses to stimulation of the cervix, uterus, colon and (1996) Distinct lateral and medial projections of the spin-
skin in the rat spinal cord. J. Neurophysiol., 69: 545–556. ohypothalamic tract of the rat. J. Comp. Neurol., 373:
Berkley, K.J., Robbins, A. and Sato, Y. (1987) Uterine afferent 549–574.
fibers in the rat. In: Schmidt R.F., Schaible H.-G. and Vahle- Calaresu, F.R. (1970) Experimental studies on the dorsal nerve
Hinz C. (Eds.), Fine Afferent Nerve Fibers and Pain. VCH, of the penis of the rat. Am. J. Anat., 127: 415–422.
Weinheim, pp. 129–136. Carlson, R.R. and De Feo, V.J. (1965) Role of the pelvic nerve
Berkley, K.J., Robbins, A. and Sato, Y. (1988) Afferent fibers vs. the abdominal sympathetic nerves in the reproductive
supplying the uterus in the rat. J. Neurophysiol., 59: 142–163. function of the female rat. Endocrinology, 77: 1014–1022.
Berkley, K.J., Robbins, A. and Sato, Y. (1993c) Functional Chaouch, A., Menetrey, D., Binder, D. and Besson, J.M. (1983)
differences between afferent fibers in the hypogastric and Neurons at the origin of the medial component of the bulb-
pelvic nerves innervating female reproductive organs in the opontine spinoreticular tract in the rat: an anatomical study
rat. J. Neurophysiol., 69: 533–544. using horseradish peroxidase retrograde transport. J. Comp.
Berkley, K.J., Wood, E., Scofield, S.L. and Little, M. (1995b) Neurol., 214: 309–320.
Behavioral responses to uterine or vaginal distension in the Charlifue, S.W., Gerhart, K.A., Menter, R.R., Whiteneck,
rat. Pain, 61: 121–131. G.G. and Manley, M.S. (1992) Sexual issues of women with
Bloch, G.J., Babcock, A.M., Gorski, R.A. and Micevych, P.E. spinal cord injuries. Paraplegia, 30: 192–199.
(1988) Effects of cholecystokinin on male copulatory behavior Chateau, D. and Aron, C. (1990) Peripheral anosmia and
and lordosis behavior in male rats. Physiol. Behav., 43: 351–357. display of lordosis behaviour in the male rat. Behav. Proc., 22:
Bodner, D.R., Lindan, R., Leffler, E., Kursh, E.D. and 33–40.
Resnick, M.I. (1987) The application of intracavernous Chung, P.H., Yeko, T.R., Mayer, J.C., Sanford, E.J. and
injection of vasoactive medications for erection in men with Maroulis, G.B. (1995) Assisted fertility using electroejaculation
spinal cord injury. J. Urol., 138: 310–311. in men with spinal cord injury—a review of literature. Fertil.
Bonica, J.J. (1990) General considerations of pain in the pelvis Steril., 64: 1–9.
and perineum. In: Bonica J.J. (Ed.), The Management of Pain Cliffer, K.D., Burstein, R. and Giesler Jr., G.J. (1991)
(2nd ed). Lea & Febiger, Philadelphia, PA, pp. 1283–1312. Distributions of spinothalamic, spinohypothalamic, and
Bowsher, D. (1976) Role of the reticular formation in responses spinotelencephalic fibers revealed by anterograde transport
to noxious stimulation. Pain, 2: 361–378. of PHA-L in rats. J. Neurosci., 11: 852–868.
Brackett, N.L., Padron, O.F. and Lynne, C.M. (1997) Semen Collins, J.J., Lin, C.E., Berthoud, H.R. and Papka, R.E.
quality of spinal cord injured men is better when obtained by (1999) Vagal afferents from the uterus and cervix provide
vibratory stimulation versus electroejaculation. J. Urol., 157: direct connections to the brainstem. Cell Tissue Res., 295:
151–157. 43–54.
411

Comarr, A.E. (1966) Observations on menstruation and preg- and nitric oxide in the nucleus of the solitary tract varies as
nancy among female spinal cord injury patients. Paraplegia, a function of the oestrus cycle. Brain Res., 898: 303–313.
3: 263–272. Haase, E.B., Buchman, J., Tietz, A.E. and Schramm, L.P.
Crosby, E., St-Jean, B., Reid, D. and Elliott, R.D. (1992) Ob- (1997) Pregnancy-induced uterine neuronal degeneration in
stetrical anaesthesia and analgesia in chronic spinal cord-in- the rat. Cell Tissue Res., 288: 293–306.
jured women. Can. J. Anaesth., 39: 487–494. Haldeman, S., Bradley, W.E., Bhatia, N.N. and Johnson, B.K.
Cross, L.L., Meythaler, J.M., Tuel, S.M. and Cross, A.L. (1982) Pudendal evoked responses. Arch. Neurol., 39:
(1991) Pregnancy following spinal cord injury. West J. Med., 280–283.
154: 607–611. Haskins, J.T. and Moss, R.L. (1983) Action of estrogen and
Cross, L.L., Meythaler, J.M., Tuel, S.M. and Cross, A.L. mechanical vaginocervical stimulation on the membrane
(1992) Pregnancy, labor and delivery post spinal cord injury. excitability of hypothalamic and midbrain neurons. Brain
Paraplegia, 30: 890–902. Res. Bull., 10: 489–496.
Dahlberg, A., Ruutu, M. and Hovatta, O. (1995) Pregnancy Higuchi, T., Uchide, K., Honda, K. and Negoro, H. (1987)
results from a vibrator application, electroejaculation, and a Pelvic neurectomy abolishes the fetus-expulsion reflex and
vas aspiration programme in spinal-cord injured men. Hum. induces dystocia in the rat. Exp. Neurol., 96: 443–455.
Reprod., 10: 2305–2307. Honda, C.N. (1985) Visceral and somatic afferent convergence
Daniels, D., Miselis, R.R. and Flanagan-Cato, L.M. (1999) onto neurons near the central canal in the sacral spinal cord
Central neuronal circuit innervating the lordosis-producing of the cat. J. Neurophysiol., 53: 1059–1078.
muscles defined by transneuronal transport of pseudorabies Hornby, J.B. and Rose, J.D. (1976) Responses of caudal brain
virus. J. Neurosci., 19: 2823–2833. stem neurons to vaginal and somatosensory stimulation in
de Groat, W.C. and Booth, A.M. (1980) Physiology of male the rat and evidence of genital-nociceptive interactions. Exp.
sexual function. Ann. Intern. Med., 92: 329–331. Neurol., 51: 363–376.
de Groat, W.C. and Booth, A.M. (1993) Neural control of pe- Houghton, A.K., Kadura, S. and Westlund, K.N. (1997)
nile erection. In: Maggi C.A. (Ed.), The Autonomic Nervous Dorsal column lesions reverse the reduction of homecage
System. Harwood Academic, London, pp. 465–516. activity in rats with pancreatitis. Neuroreport, 8: 3795–3800.
de Groat, W.C., Nadelhaft, I., Milne, R.J., Booth, A.M., Mor- Hubscher, C.H. (1994) Sensory input from pelvic reproductive
gan, C. and Thor, K. (1981) Organization of the sacral para- organs to the gracile and solitary nuclei in the female rat.
sympathetic reflex pathways to the urinary bladder and large Department of Psychology: Florida State University,
intestine. J. Auton. Nerv. Syst., 3: 135–160. Tallahassee, p. 80.
deGroat, W.C. and Steers, W.D. (1988) Neuroanatomy and ne- Hubscher, C.H. and Berkley, K.J. (1994) Responses of neurons
urophysiology of penile erection. In: Tanagho E.A., Lue T.F. in caudal solitary nucleus of female rats to stimulation of
and McClure R.D. (Eds.), Contemporary Management Of vagina, cervix, uterine horn and colon. Brain Res., 664: 1–8.
Impotence and Infertility. Williams and Wilkins, Baltimore, Hubscher, C.H. and Berkley, K.J. (1995) Spinal and vagal in-
pp. 3–27. fluences on the responses of rat solitary nucleus neurons to
Diakow, C. (1970) Effects of genital desensitization on the stimulation of uterus, cervix and vagina. Brain Res., 702:
mating pattern of female rats as determined by motion pic- 251–254.
ture analysis. Am. Zool., 10: 486. Hubscher, C.H., Brooks, D.L. and Johnson, J.R. (2004a)
Ding, Y.Q., Shi, J., Wang, D.S., Xu, J.Q., Li, J.L. and Ju, G. Effects of spinal cord injury on the rat estrous cycle. Program
(1999) Primary afferent fibers of the pelvic nerve terminate in No. 458.15. In: 2004 Abstract Viewer/Itinerary Planner.
the gracile nucleus of the rat. Neurosci. Lett., 272: 211–214. Washington, DC: Society for Neuroscience.
Erskine, M.S. (1992) Pelvic and pudendal nerves influence the Hubscher, C.H. and Johnson, R.D. (1996) Responses of med-
display of paced mating behavior in response to estrogen and ullary reticular formation neurons to input from the male
progesterone in the female rat. Behav. Neurosci., 106: 690–697. genitalia. J. Neurophysiol., 76: 2474–2482.
Garfield, R.E. (1986) Structural studies of innervation on non- Hubscher, C.H. and Johnson, R.D. (1999) Effects of acute and
pregnant rat uterus. Am. J. Physiol., 251: C41–C54. chronic midthoracic spinal cord injury on neural circuits
Giesler Jr., G.J., Spiel, H.R. and Willis, W.D. (1981) Organ- for male sexual function. I. Ascending pathways. J.
ization of spinothalamic tract axons within the rat spinal Neurophysiol., 82: 1381–1389.
cord. J. Comp. Neurol., 195: 243–252. Hubscher, C.H. and Johnson, R.D. (2001) Supraspinal
Giuliano, F.A., Rampin, O., Benoit, G. and Jardin, A. (1995) processing of inputs from the cervix: effects of hormonal
Neural control of penile erection. Urol. Clin. North Am., 22: status. Soc. Neurosci. Abstr., 27: Program No. 840–841.
747–766. Hubscher, C.H. and Johnson, R.D. (2003) Responses of
Graber, G.C. and Kristal, M.B. (1977) Uterine distention fa- thalamic neurons to input from the male genitalia. J. Ne-
cilitates the onset of maternal behavior in pseudopregnant urophysiol., 89: 2–11.
but not in cycling rats. Physiol. Behav., 19: 133–137. Hubscher, C.H. and Johnson, R.D. (2004) Effects of chronic
Guevara-Guzman, R., Buzo, E., Larrazolo, A., de la Riva, C., dorsal column lesions on pelvic viscerosomatic convergent
Da Costa, A.P. and Kendrick, K.M. (2001) Vaginocervical medullary reticular formation neurons. J. Neurophysiol., 92:
stimulation-induced release of classical neurotransmitters 3596–3600.
412

Hubscher, C.H., Kaddumi, E.G. and Johnson, R.D. (2004b) Kow, L.M. and Pfaff, D.W. (1973) Effects of estrogen treat-
Brain stem convergence of pelvic viscerosomatic inputs via ment on the size of receptive field and response threshold of
spinal and vagal afferents. Neuroreport, 15: 1299–1302. pudendal nerve in the female rat. Neuroendocrinology, 13:
Hulsebosch, C.E. and Coggeshall, R.E. (1982) An analysis of 299–313.
the axon populations in the nerves to the pelvic viscera in the Kumazawa, T. (1986) Sensory innervation of reproductive or-
rat. J. Comp. Neurol., 211: 1–10. gans. Prog. Brain Res., 67: 115–131.
Jean, A. (1991) The nucleus tractus solitarius: neuroanatomic, Kumazawa, T., Mizumura, K., Koda, H., Tamura, R. and
neurochemical and functional aspects. Arch. Int. Physiol. Sato, J. (1995) Mechanisms of chemical modulation of tes-
Biochem. Biophys., 99: A3–A52. ticular afferents. In: Gebhart G.F. (Ed.), Visceral Pain
Johnson, R.D. (1989) Physiology of single spinal cord sensory Progress in Pain Research and Management. IASP Press,
neurons responding to penile stimulation in the rat. Soc. Seattle.
Neurosci. Abstr., 15: 756. Larsson, K. and Sodersten, P. (1973) Mating in male rats after
Johnson, R.D. and Halata, Z. (1991) Topography and ultra- section of the dorsal penile nerve. Physiol. Behav., 10:
structure of sensory nerve endings in the glans penis of the 567–571.
rat. J. Comp. Neurol., 312: 299–310. Larsson, K. and Swedin, G. (1971) The sexual behavior of male
Kitchell, R.L., Gilanpour, H. and Johnson, R.D. (1982) Elect- rats after bilateral section of the hypogastric nerve and re-
rophysiologic studies of penile mechanoreceptors in the rats. moval of the accessory genital glands. Physiol. Behav., 6:
Exp. Neurol., 75: 229–244. 251–253.
Klein, C.M., Ray, R.H. and Burden, H.W. (1989) Direct elec- Lee, J.W. and Erskine, M.S. (1996) Vaginocervical stimulation
trical stimulation of the superior ovarian nerve in rats causes suppresses the expression of c-fos induced by mating in tho-
an increase in neuronal activity in the ipsilateral ovarian racic, lumbar and sacral segments of the female rat. Neuro-
plexus nerve. Brain Res., 479: 194–200. science, 74: 237–249.
Kollar, E.J. (1953) Reproduction in the female rat after pelvic Lee, J.W. and Erskine, M.S. (2000) Pseudorabies virus tracing
nerve neurectomy. Anat. Rec., 115: 641–658. of neural pathways between the uterine cervix and CNS: ef-
Komisaruk, B.R., Bianca, R., Sansone, G., Gomez, L.E., fects of survival time, estrogen treatment, rhizotomy, and
Cueva-Rolon, R., Beyer, C. and Whipple, B. (1996) Brain- pelvic nerve transection. J. Comp. Neurol., 418: 484–503.
mediated responses to vaginocervical stimulation in spinal Linsenmeyer, T.A., Pogach, L.M., Ottenweller, J.E. and
cord-transected rats: role of the vagus nerves. Brain Res., Huang, H.F. (1994) Spermatogenesis and the pituitary-tes-
708: 128–134. ticular hormone axis in rats during the acute phase of spinal
Komisaruk, B.R., Gerdes, C.A. and Whipple, B. (1997) ‘Com- cord injury. J. Urol., 152: 1302–1307.
plete’ spinal cord injury does not block perceptual responses Lodder, J. and Zeilmaker, G.H. (1976) Effects of pelvic nerve
to genital self-stimulation in women. Arch. Neurol., 54: and pudendal nerve transection on mating behavior in the
1513–1520. male rat. Physiol. Behav., 16: 745–751.
Komisaruk, B.R. and Sansone, G. (2003) Neural pathways Marchetti, B., Cioni, M., Badr, M., Follea, N. and Pelletier, G.
mediating vaginal function: the vagus nerves and spinal cord (1987) Ovarian adrenergic nerves directly participate in the
oxytocin. Scand. J. Psychol., 44: 241–250. control of luteinizing hormone-releasing hormone and beta-
Komisaruk, B.R. and Wallman, J. (1977) Antinociceptive ef- adrenergic receptors during puberty: a biochemical and au-
fects of vaginal stimulation in rats: neurophysiological and toradiographic study. Endocrinology, 121: 219–226.
behavioral studies. Brain Res., 137: 85–107. Marson, L. and McKenna, K.E. (1990) The identification of a
Komisaruk, B.R. and Whipple, B. (1988) The role of vaginal brainstem site controlling spinal sexual reflexes in male rats.
stimulation-produced analgesia in reproductive processes. In: Brain Res., 515: 303–308.
Genazzani A.R., Nappi G., Facchinetti F. and Martignoni E. Marson, L., Platt, K.B. and McKenna, K.E. (1993) Central
(Eds.), Pain and Reproduction. Parthenon Publishing nervous system innervation of the penis as revealed by the
Group, New Jersey, pp. 125–140. transneuronal transport of pseudorabies virus. Neuroscience,
Komisaruk, B.R., Whipple, B., Crawford, A., Liu, W.C., 55: 263–280.
Kalnin, A. and Mosier, K. (2004) Brain activation during Mason, P. (2001) Contributions of the medullary raphe and
vaginocervical self-stimulation and orgasm in women with ventromedial reticular region to pain modulation and other
complete spinal cord injury: fMRI evidence of mediation by homeostatic functions. Ann. Rev. Neurosci., 24: 737–777.
the vagus nerves. Brain Res., 1024: 77–88. McKenna, K.E. (2000) The neural control of female sexual
Kostarczyk, E., Zhang, X. and Giesler Jr., G.J. (1997) Spin- function. NeuroRehabilitation, 15: 133–143.
ohypothalamic tract neurons in the cervical enlargement of McKenna, K.E. and Nadelhaft, I. (1986) The organization of
rats: locations of antidromically identified ascending axons the pudendal nerve in the male and female rat. J. Comp.
and their collateral branches in the contralateral brain. J. Neurol., 248: 532–549.
Neurophysiol., 77: 435–451. McMahon, S.B. and Wall, P.D. (1983) A system of rat spinal
Kow, L.M. (1976) Sensory requirements for the lordosis reflex cord lamina 1 cells projecting through the contralateral
in female rats. Brain Res., 101: 47–66. dorsolateral funiculus. J. Comp. Neurol., 214: 217–223.
413

Meana, M., Binik, Y.M., Khalife, S., Bergeron, S., Pagidas, K. the rat uterus: an electrophysiological and HRP study. Brain
and Berkley, K.J. (1997) Dyspareunia: more than bad sex. Res. Bull., 25: 365–371.
Pain, 71: 211–212. Pacheco, P., Camacho, M.A., Garcia, L.I., Hernandez, M.E.,
Menetrey, D., Chaouch, A. and Besson, J.M. (1980) Location Carrillo, P. and Manzo, J. (1997) Electrophysiological evi-
and properties of dorsal horn neurons at origin of spin- dence for the nomenclature of the pudendal nerve and sacral
oreticular tract in lumbar enlargement of the rat. J. Neuro- plexus in the male rat. Brain Res., 763: 202–208.
physiol., 44: 862–877. Pacheco, P., Martinez-Gomez, M., Whipple, B., Beyer, C. and
Menetrey, D. and de Pommery, J. (1991) Origins of spinal as- Komisaruk, B.R. (1989) Somato-motor components of the
cending pathways that reach central areas involved in vis- pelvic and pudendal nerves of the female rat. Brain Res., 490:
ceroception and visceronociception in the rat. Eur. J. 85–94.
Neurosci., 3: 249–259. Papka, R.E., Williams, S., Miller, K.E., Copelin, T. and Puri, P.
Modianos, D. and Pfaff, D. (1979) Medullary reticular forma- (1998) CNS location of uterine-related neurons revealed by
tion lesions and lordosis reflex in female rats. Brain Res., 171: trans-synaptic tracing with pseudorabies virus and their re-
334–338. lation to estrogen receptor-immunoreactive neurons. Neuro-
Moore, C.L. and White, R.H. (1996) Sex differences in sensory science, 84: 935–952.
and motor branches of the pudendal nerve of the rat. Horm. Pavlakis, A.J., Siroky, M.B., Goldstein, I. and Krane, R.J.
Behav., 30: 590–599. (1983) Neurourologic findings in conus medullaris and cauda
Mtui, E.P., Anwar, M., Reis, D.J. and Ruggiero, D.A. (1995) equina injury. Arch. Neurol., 40: 570–573.
Medullary visceral reflex circuits: local afferents to nucleus Peschanski, M. and Besson, J.M. (1984) A spino-reticulo-
tractus solitarii synthesize catecholamines and project to thalamic pathway in the rat: an anatomical study with ref-
thoracic spinal cord. J. Comp. Neurol., 351: 5–26. erence to pain transmission. Neuroscience, 12: 165–178.
Nadelhaft, I. and Booth, A.M. (1984) The location and mor- Peters, L.C., Kristal, M.B. and Komisaruk, B.R. (1987) Sen-
phology of preganglionic neurons and the distribution of sory innervation of the external and internal genitalia of the
visceral afferents from the rat pelvic nerve: a horseradish female rat. Brain Res., 408: 199–204.
peroxidase study. J. Comp. Neurol., 226: 238–245. Pfaus, J.G., Marcangione, C., Smith, W.J., Manitt, C. and
Nadelhaft, I. and McKenna, K.E. (1987) Sexual dimorphism in Abillamaa, H. (1996) Differential induction of Fos in the
sympathetic preganglionic neurons of the rat hypogastric female rat brain following different amounts of vaginocervi-
nerve. J. Comp. Neurol., 256: 308–315. cal stimulation: modulation by steroid hormones. Brain Res.,
Nahin, R.L., Madsen, A.M. and Giesler Jr., G.J. (1986) Funic- 741: 314–330.
ular location of the ascending axons of neurons adjacent to the Pierau, F.K., Taylor, D.C., Abel, W. and Friedrich, B. (1982)
spinal cord central canal in the rat. Brain Res., 384: 367–372. Dichotomizing peripheral fibres revealed by intracellular re-
Nance, D.M., Burns, J., Klein, C.M. and Burden, H.W. (1988) cording from rat sensory neurones. Neurosci. Lett., 31:
Afferent fibers in the reproductive system and pelvic viscera 123–128.
of female rats: anterograde tracing and immunocytochemical Quinlan, D.M., Nelson, R.J., Partin, A.W., Mostwin, J.L. and
studies. Brain Res. Bull., 21: 701–709. Walsh, P.C. (1989) The rat as a model for the study of penile
Nehra, A., Werner, M.A., Bastuba, M., Title, C. and Oates, erection. J. Urol., 141: 656–661.
R.D. (1996) Vibratory stimulation and rectal probe electro- Rampin, O., Gougis, S., Giuliano, F. and Rousseau, J.P. (1997)
ejaculation as therapy for patients with spinal cord injury: Spinal Fos labeling and penile erection elicited by stimulation
semen parameters and pregnancy rates. J. Urol., 155: of dorsal nerve of the rat penis. Am. J. Physiol., 272:
554–559. R1425–R1431.
Neuhuber, W. (1982) The central projections of visceral pri- Reiner, P., Woolsey, J., Adler, N. and Morrison, A. (1981) A
mary afferent neurons of the inferior mesenteric plexus and gross anatomical study of the peripheral nerves associated
hypogastric nerve and the location of the related sensory and with reproductive function in the female albino rat. In: Adler
preganglionic sympathetic cell bodies in the rat. Anat. Em- N.T. (Ed.), Neuroendocrinology of Reproduction. Plenum
bryol., 164: 413–425. Press, New York, pp. 545–549.
Nunez, R., Gross, G.H. and Sachs, B.D. (1986) Origin and Ross, E.L., Komisaruk, B.R. and O’Donnell, D. (1979) Evi-
central projections of rat dorsal penile nerve: possible direct dence that probing the vaginal cervix is analgesic in rats,
projection to autonomic and somatic neurons by primary using an operant paradigm. J. Comp. Physiol. Psychol., 93:
afferents of nonmuscle origin. J. Comp. Neurol., 247: 330–336.
417–429. Sachs, B.D. and Meisel, R.L. (1988) The physiology of male
Odutola, A.B. (1977) On the location of reticular neurons pro- sexual behavior. In: Knobil E. and Neill J.D. (Eds.), The
jecting to the cuneogracile nuclei in the rat. Exp. Neurol., 54: Physiology of Reproduction. Raven, New York,
54–59. pp. 1393–1485.
Ortega-Villalobos, M., Garcia-Bazan, M., Solano-Flores, L.P., Sarkarati, M., Rossier, A.B. and Fam, B.A. (1987) Experience
Ninomiya-Alarcon, J.G., Guevara-Guzman, R. and Wayner, in vibratory and electro-ejaculation techniques in spinal cord
M.J. (1990) Vagus nerve afferent and efferent innervation of injury patients: a preliminary report. J. Urol., 138: 59–62.
414

Sartucci, F., Piaggesi, A., Logi, F., Bonfiglio, L., Bongioanni, Tetel, M.J., Getzinger, M.J. and Blaustein, J.D. (1993) Fos ex-
P., Pellegrinetti, A., Baccetti, F., Navalesi, R. and Murri, L. pression in the rat brain following vaginal-cervical stimula-
(1999) Impaired ascendant central pathways conduction in tion by mating and manual probing. J. Neuroendocrinol., 5:
impotent diabetic subjects. Acta Neurol. Scand., 99: 381–386. 397–404.
Sato, S., Hayashi, R.H. and Garfield, R.E. (1989) Mechanical Tomasulo, K.C. and Emmers, R. (1972) Activation of neurons
responses of the rat uterus, cervix, and bladder to stimulation in the gracile nucleus by two afferent pathways in the rat.
of hypogastric and pelvic nerves in vivo. Biol. Reprod., 40: Exp. Neurol., 36: 197–206.
209–219. Truitt, W.A. and Coolen, L.M. (2002) Identification of a po-
Sauer, P.M. and Harvey, C.J. (1993) Spinal cord injury and tential ejaculation generator in the spinal cord. Science, 297:
pregnancy. J. Perinat. Neonatal. Nurs., 7: 22–34. 1566–1569.
Schwartz-Giblin, S., McCarthy, M.M. and Robbins, A. (1996) Van der Horst, V.G. and Holstege, G. (1998) Sensory and mo-
The medullary reticular formation is a site of muscle relaxant tor components of reproductive behavior: pathways and
action of diazepam on deep back and neck muscles in the plasticity. Behav. Brain Res., 92: 157–167.
female rat. Brain Res., 710: 178–188. Ver Voort, S. (1987) Infertility in spinal cord injured male.
Sedor, J.F. and Hirsch, I.H. (1995) Evaluation of sperm mor- Urology, XXIX(29): 157–165.
phology of electroejaculates of spinal cord-injured men by Wall, P.D., Hubscher, C.H. and Berkley, K.J. (1993) Intraspinal
strict criteria. Fertil. Steril., 63: 1125–1127. modulation of neuronal responses to uterine and cervix stim-
Seftel, A.D., Oates, R.D. and Krane, R.J. (1991) Disturbed ulation in rat L1 and L6 dorsal horn. Brain Res., 622: 71–78.
sexual function in patients with spinal cord disease. Neurol. Wang, C.C., Willis, W.D. and Westlund, K.N. (1999) Ascend-
Clin., 9: 757–778. ing projections from the area around the spinal cord central
Serghini, R., Prud’homme, M.J., Vaudry, H. and Rousseau, canal: a Phaseolus vulgaris leucoagglutinin study in rats. J.
J.P. (1997) Involvement of the pelvic plexus and the supra- Comp. Neurol., 415: 341–367.
renal ganglia in the neuropeptide Y (NPY) innervation of the Westgren, N. and Levi, R. (1994) Motherhood after traumatic
cervix and the uterus of the rat. J. Auton. Nerv. Syst., 67: spinal cord injury. Paraplegia, 32: 517–523.
38–50. Willis, W.D., Al-Chaer, E.D., Quast, M.J. and Westlund, K.N.
Shen, P., Arnold, A.P. and Micevych, P.E. (1990) Supraspinal (1999) A visceral pain pathway in the dorsal column of the
projections to the ventromedial lumbar spinal cord in adult spinal cord. Proc. Natl. Acad. Sci. USA, 96: 7675–7679.
male rats. J. Comp. Neurol., 300: 263–272. Willis, W.D. and Coggeshall, R.E. (1991) Sensory Mechanisms
Sipski, M.L., Alexander, C.J. and Rosen, R. (2001) Sexual of the Spinal Cord (2nd ed). Plenum Press, New York.
arousal and orgasm in women: effects of spinal cord injury. Yang, S.W., Follett, K.A., Piper, J.G. and Ness, T.J. (1998) The
Ann. Neurol., 49: 35–44. effect of morphine on responses of mediodorsal thalamic
Sonksen, J. and Ohl, D.A. (2002) Penile vibratory stimulation nuclei and nucleus submedius neurons to colorectal disten-
and electroejaculation in the treatment of ejaculatory dys- sion in the rat. Brain Res., 779: 41–52.
function. Int. J. Androl., 25: 324–332. Zemlan, F.P., Leonard, C.M., Kow, L.M. and Pfaff, D.W.
Taylor, D.C., Korf, H.W. and Pierau, F.K. (1982) Distribution (1978) Ascending tracts of the lateral columns of the rat spi-
of sensory neurons of the pudendal nerve in the dorsal root nal cord: a study using the silver impregnation and horse-
ganglia and their projection to the spinal cord. Horseradish- radish peroxidase techniques. Exp. Neurol., 62: 298–334.
peroxidase studies in the rat. Cell Tissue Res., 226: 555–564. Zhuo, M. and Gebhart, G.F. (1991) Spinal serotonin receptors
Temple, J.L., Bradshaw, H.B., Wood, E. and Berkley, K.J. mediate descending facilitation of a nociceptive reflex from
(1999) Effects of hypogastric neurectomy on escape responses the nuclei reticularis gigantocellularis and gigantocellularis
to uterine distention in the rat. Pain, 6(Suppl.): S13–S20. pars alpha in the rat. Brain Res., 550: 35–48.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Published by Elsevier B.V.

CHAPTER 28

Descending pathways modulating the spinal circuitry


for ejaculation: effects of chronic spinal cord injury

Richard D. Johnson

Department of Physiological Sciences, College of Veterinary Medicine and the McKnight Brain Institute, University of
Florida, Gainesville, FL 32610-0144, USA

Abstract: Sexual dysfunction is a common complication in men with chronic spinal cord injury. In par-
ticular, ejaculation is severely compromised or absent and the resulting infertility issues are important to
this group of predominantly young men. To investigate the neural circuits and descending spinal pathways
involved in ejaculation, animal models have been developed in normal and spinal cord-injured prepara-
tions. Primarily through studies in rats, spinal ejaculatory circuits have been described including (i) au-
tonomic circuits at the thoracolumbar and lumbosacral levels mediating the emission phase of ejaculation,
(ii) somatic circuits at the lumbosacral level controlling the expulsion phase of ejaculation through se-
quential and rhythmic contraction of perineal striated muscles (e.g. bulbospongiosus), and (iii) a proposed
ejaculatory pattern generator in the lumbar cord. Midthoracic incomplete chronic spinal cord injury has
revealed the dependency of spinal ejaculatory circuits on bilateral spinal pathways from the brainstem via
modulation of pudendal motor neuron reflexes and pudendal nerve autonomic fibers. Accordingly, sensory
input from the dorsal nerve of the penis, required to trigger the ejaculatory response in animals and
humans, is no longer inhibited from the lateral paragigantocellularis nucleus in the ventrolateral medulla.
This inhibitory effect, likely presynaptic through a serotonergic pathway, is thought to be necessary to
provide the rhythmic, bursting, and sequential contractions of the perineal muscles during ejaculation.
Chronic lateral hemisection injury, which severs half of the descending lateral funiculus-located pathways,
results in new functional connections of the pudendal reflex inhibitory and pudendal sympathetic activation
pathways across the midline, above and below the lesion, respectively. Clinical correlations in spinal cord-
injured men have demonstrated the validity of the rodent animal for the study of ejaculatory dysfunction
after chronic injury.

Introduction recent survey of men with spinal cord injury by


Anderson (2004) has demonstrated that among
Male sexual dysfunction is a common complica- paraplegics, regaining normal sexual function is
tion resulting from chronic spinal cord injury. The the most important goal, even surpassing locomo-
human spinal cord-injured population is predom- tion. Moreover, the ability to ejaculate is severely
inantly male and of this group, 85% are men compromised making infertility a critical issue.
between the ages of 18 and 45 (Ohl et al., 1989). A Clinical and animal studies over the last 25 years
have investigated the integrity and degree of
Corresponding author. Tel.: +352-392-4700 ext 3834; recovery of male sexual function following spinal
Fax: +352-392-5145; E-mail: johnson@ufbi.ufl.edu injury in order to (i) document the neural pathways

DOI: 10.1016/S0079-6123(05)52028-4 415


416

affected, (ii) evaluate the effects of different de- intraspinal connections that span several spinal
grees of chronic injury, and (iii) devise treatment segments [in the rat; thoracic (T)10–sacral (S)1].
strategies to improve functional outcome. Studies in animals and humans, detailed below,
Of paramount importance has been the devel- have shown that descending control from supra-
opment of animal models for the study of male spinal sites are required for coordination of the
sexual function following acute and chronic spinal visceral and somatic motor events mediating the
cord injury. Although the investigation of normal forceful, rhythmic expulsion of semen in ejacula-
sexual neural mechanisms have included impor- tion and moving the blood into the glans penis in
tant studies in the cat and dog, the rat has emerged rigid erection. These two propulsive-like sexual
as the most appropriate animal model particularly events are dependent on a coordinated contraction
with regard to studying the chronic injury. Several sequence of smooth and striated muscles similar to
advantages to using the rat model are (i) the ex- other eliminative viscerosomatic responses like
istence of a substantial behavioral database on defecation and micturition.
normal sexual function, (ii) a variety of spinal le- The ejaculation response consists of two phases.
sions can be performed with few complications, The initial emission phase involves accessory gland
(iii) the accessibility of single neurons or nerve/ secretions and smooth muscle-mediated movement
muscle ensembles for recording in anesthetized of sperm and of the resultant mixture of sperm
preparations, and (iv) combined electrophysiolog- with glandular secretions (semen) through the in-
ical, neuroanatomical, pharmacological, and be- ternal segments of the reproductive tract into the
havioral studies can be carried out in the same pelvic urethra. The emission phase is mediated by
animal. the sympathetic and parasympathetic divisions of
In this chapter, a consideration of the studies the autonomic nervous system. Sympathetic pre-
using animal models will concentrate on the chronic ganglionic emission neurons are located in the in-
spinal cord injury-induced alteration of ejaculato- termediolateral cell column and central gray of the
ry circuitry and provide a structural template on T12 to lumbar (L)2 spinal cord and send axons
which future studies can be designed. When ap- into the hypogastric nerve and sympathetic trunk
propriate, data from human clinical studies will be (Nadelhaft and McKenna, 1987). Parasympathetic
correlated with data from animal models. preganglionic emission neurons reside in the sacral
portion of the intermediolateral cell column (also
known as the sacral parasympathetic nucleus) and
Spinal reflex circuitry for ejaculation project peripherally through the pelvic nerve. Fol-
lowing emission, the expulsion phase of ejacula-
Much of the neural circuitry for erection and ejac- tion consists of a rapid, forceful, and rhythmic
ulation is contained within specific regions of the propulsion of semen through the penile urethra
spinal cord (reviewed by McKenna, 2000; Steers, and out through the external urethral orifice. Ex-
2000; Coolen et al., 2004; Giuliano and Rampin, pulsion of semen is produced by the coordinated
2004). Spinal cord-injured humans and animals, and rhythmic contraction of the striated perineal
however, lose portions of these sexual responses muscles; bulbospongiosus, ischiocavernosus, and
when the lesion severs ascending and descending external urethral sphincter. In the rat, the motor
pathways to the brainstem and higher centers, neurons innervating these muscles are located in
suggesting the segmental circuits require a long- the dorsomedial (bulbospongiosus) and dorsolat-
loop connection to maintain optimum function. eral (ischiocavernosus, external urethral sphincter)
The spinal circuits are very complex primarily be- motor nuclei in the ventral horn of L5–L6
cause they involve the integration of both divisions (McKenna and Nadelhaft, 1986; Collins et al.,
of the autonomic motor system (parasympathetic 1991). In humans and certain other species (primate,
and sympathetic), the somatic motor system, and cat, dog), these motoneuronal pools are combined
all three divisions of general sensory afferent fibers in Onuf’s nucleus (Schroder, 1985; Beattie et al.,
(visceral, somatic, mucocutaneous) in a network of 1993) located in the sacral segments. The perineal
417

motor neurons send their axons peripherally via may connect with a proposed spinal pattern
the pudendal nerve (via the motor branch in rats). generator for ejaculation, located in the L3–L4
The perineal muscle that is primarily involved in central gray with connections to the autonomic
ejaculatory expulsion and in the firm erection of and somatic spinal circuits for sexual function
the glans penis (Johnson, 1988; Holmes et al., (Truitt and Coolen, 2002; Carro-Juarez et al.,
1991; Schmidt and Schmidt, 1993; Yang and 2003; Coolen et al., 2004).
Bradley, 1999) is the bulbospongiosus (also known The primary sensory input to the pudendal seg-
as the bulbocavernosus), a striated muscle that mental reflex circuit and probably to the proposed
surrounds the bulb of the penis and its internal ejaculatory spinal pattern generator, is the dorsal
corpus spongiosum. nerve of the penis, the most distal portion of the
The segmental reflex circuit for perineal muscle pudendal nerve innervating sensory endings in the
contraction resides in the L5–S1 spinal cord of the penis and prepuce (Johnson, 1988; Johnson and
rat. The bulbospongiosus and ischiocavernosus Halata, 1991). Penile mechanoreceptors have been
motor neurons have many properties that are dif- shown to (i) respond to vibratory and tangential
ferent from typical somatic motor neurons inner- surface tactile stimulation (Johnson and Murray,
vating skeletal muscles and have many similarities 1992), (ii) spontaneously respond to blood flow
to autonomic motor neurons. For example, they engorging the erectile tissue in the absence of skin
are characterized by lack of a monosynaptic reflex stimulation, an internal response to stretch (Johnson,
arc from the homonymous muscle spindle recep- 1988), and (iii) increase their tactile sensitivity
tors (Bowens et al., 1984; McKenna and Nadelhaft, during erection possibly through autonomic fiber
1989; Collins et al., 1991), resistance to paradox- modulation of the sensory ending (Johnson, 1988;
ical sleep-triggered inhibition (Mann et al., 2003) Johnson and Halata, 1991). Behavioral and func-
and to damage from amyotrophic lateral sclerosis tional studies in animals and humans have dem-
(Carvalho et al., 1995) but exhibit susceptibility to onstrated that the integrity of the penile sensory
autonomic motor neuron disorders (Sung et al., axons is essential for triggering ejaculation (Hart
1979; Dubrovsky and Filipini, 1990). In addition, and Leedy, 1985; Meisel and Sachs, 1994; Wieder
these neurons are trophically dependent on the et al., 2000). Visceral afferents innervating the
presence of testosterone (Breedlove and Arnold, urethral mucosa (McKenna and Nadelhaft, 1989)
1980), are each bilaterally organized for simulta- and erectile tissue in the corpus cavernosum/
neous contraction, and can have dendritic arbors spongiosum (Johnson, 1988; Johnson and Halata,
that cross the midline forming gap junction-medi- 1991) along with the mucocutaneous afferents in-
ated electrical communication with their contra- nervating the glans penis and external urethral or-
lateral counterpart (Collins et al., 1991; Coleman ifice (Johnson and Halata, 1991) are all present
and Sengelaub, 2002). The polysynaptic segmental within the dorsal nerve of the penis and pudendal
reflex activation of these pudendal motoneurons nerve. Electrical, tactile, or chemical activation of
requires the participation of many spinal interneu- the pudendal primary afferents triggers a polysy-
rons that have been identified, via transsynaptic naptic reflex discharge of pudendal motoneurons
chemical and viral retrograde tracers (Collins (McKenna and Nadelhaft, 1989; Johnson, 1995;
et al., 1991; Marson and McKenna, 1996), bilat- Johnson and Hubscher, 1998), produces ejacula-
erally in the central and intermediate gray of the tion-like motor patterns (McKenna et al., 1991;
lumbosacral cord. Spinal interneurons receiving Carro-Juarez et al., 2003) and drives the bulbo-
bilateral synaptic input from dorsal nerve of the cavernosus reflex in men (Yang and Bradley, 1999).
penis afferent terminations (Nunez et al., 1986) To investigate the electrophysiological charac-
have been recorded from throughout the T13–S1 teristics of the pudendal reflex circuit in male rats
spinal cord (R.D. Johnson, unpublished obser- with and without chronic spinal cord injury, our
vations). In addition to connecting pudendal laboratory at the University of Florida developed
motoneurons synaptically with segmental primary an in vivo animal model. By unilaterally and
afferent terminals (see below), the interneurons bilaterally recording and stimulating the pudendal
418

reflex elements simultaneously with activation of (PET)-scan studies in men (Holstege and Georgiadis,
descending modulatory pathways from the medul- 2004). However, as these areas do not project to
la (Fig. 1), the effects of different spinal lesions and the spinal cord, any modulatory effect they might
postlesion intervals could be determined. In unin- have would likely be relayed through the medul-
jured anesthetized animals, stimulation of penile lary spinal pathways.
afferents produced polysynaptic bilateral reflex The descending spinal pathways from the vent-
discharges that were subject to rate depression and rolateral medulla and medullary reticular forma-
presynaptic inhibition (Johnson, 1995; Johnson tion modulate many viscerosomatic functions in
and Hubscher, 1998). In contrast, stimulation of the spinal cord including nociceptive reflexes, how-
pelvic nerve afferents elicited bilateral reflex dis- ever, they have been proposed to organize and
charges that were resistant to rate depression and temporally sequence complex sensorimotor activ-
presynaptic inhibition. These results suggested that ities which require both somatic and visceral
the spinal interneurons activated by these two dif- (autonomic) motor neurons such as gagging, vom-
ferent inputs are separate and may be modulated by iting, defecation, and ejaculation (Holstege, 1991;
different descending pathways from the brainstem. Mason, 2001). Electrical stimulation of the lateral
paragigantocellularis nucleus has been shown to
produce field potentials in the lumbosacral spinal
Descending brainstem control of ejaculation cord near the pudendal motor nuclei (Tanaka and
Arnold, 1993). Large lesions that included the
Several supraspinal regions have been shown to reticularis gigantocellularis nuclear complex
exert various degrees of facilitatory or inhibitory eliminated ejaculatory bursts in perineal muscles
control on the spinal centers for ejaculation (re- triggered by urethral stimulation (urethrogenital
viewed by Coolen et al., 2004). Excitatory influ- reflex; Marson and McKenna, 1990). Smaller
ences have been proposed from several regions of lesions of the nucleus raphe obscurus or lateral
the hypothalamus, the medial preoptic area (Pehek paragigantocellularis nucleus had significant effects
et al., 1989; Markowski et al., 1994), the paraven- on external anal sphincter and perineal muscle
tricular nucleus (Marson and McKenna, 1994), reflexes, respectively (Holmes et al., 2002) and
and the lateral hypothalamus (Kippin et al., 2004). reduced ejaculatory behavior (Yells et al., 1992).
Strong inhibitory influences descend to the spinal Using bilateral electrical stimulation of the
cord from the lateral portion of the nucleus par- dorsal nerve of the penis as the search stimulus,
agigantocellularis in the medullary reticular for- we found penile-responsive neurons in the gig-
mation (Marson and McKenna, 1992). Numerous antocellularis complex in anesthetized, uninjured
neuroanatomical studies have shown robust axon- male rats (Hubscher and Johnson, 1996; see
al projections to the perineal motoneuronal pools Hubscher, this volume). The majority of these
from the lateral paragigantocellularis (Marson and neurons exhibited an excitatory response to me-
McKenna, 1996; Hermann et al., 2003), the adja- chanical stimulation of receptive fields on either
cent nucleus raphe obscurus (Hermann et al., side of the glans penis and many exhibited a wind-
1998; Holmes et al., 2002), and the paraventricular up of firing during repeated stimulation as would
nucleus in the thalamus (Wagner and Clemens, occur during copulation. This suggested that a
1991). Although the medial preoptic area does not spino–bulbo–spinal loop for coordination and
project axons to the spinal cord, it likely has an control of the lumbosacral ejaculatory reflex cir-
influence through connections to the lateral par- cuitry involved the gigantocellularis nuclear com-
agigantocellularis via the periaqueductal gray plex. Unilateral microstimulation of neurons in and
(Murphy and Hoffman, 2001). Other supraspinal adjacent to the lateral paragigantocellularis nucleus
regions above the brainstem that may be active (Fig. 1; left panel) produced a profound bilateral
prior to, during, or after ejaculation have been inhibition (decrease in amplitude and increase in
identified in neuroanatomical studies in rats (Coolen latency) of the short latency pudendal motoneuron
et al., 2004) and positron emission tomography reflex discharges elicited by stimulation of the
Fig. 1. Schematic representation of two descending pathways that modulate activity of somatic and autonomic motor neurons in the
pudendal nerve, a nerve innervating the perineal muscles involved in the expulsive phase of ejaculation. Data from Johnson and
Hubscher (1998, 2000) and Hubscher and Johnson (1999, 2000) were obtained from anesthetized spinal cord-injured and -uninjured
male rats in which electrodes were placed bilaterally around the dorsal nerve of the penis and pelvic nerve for the stimulation (S) of
segmental sensory afferents and the motor branch of the pudendal nerve for the recording (R) of elicited polysynaptic pudendal motor
neuron reflex discharges and the firing of postganglionic sympathetic fibers. The most robust medullary site (gray regions) for
inhibitory modulation of pudendal motor neuron reflexes (left panel) and activation of pudendal sympathetic fibers (right panel) were
obtained by electrical microstimulation of the lateral paragigantocellularis (LPGi) nuclear region located ventrolateral to the gig-
antocellularis (Gi, GiA) nuclear complex, dorsolateral to the pyramidal (py) tracts and lateral to the raphe magnus (RMg). The
location of each descending pathway in the lateral funiculus of T8 was determined by documenting the effect of microstimulating the
ipsilateral or contralateral medulla after a variety of acute and chronic midthoracic spinal lesions. Bilateral chronic lesions of the dorsal
3/5 of T8 were necessary to eliminate the effects of both pathways. Chronic (30 day), but not acute (several hours) lateral hemisection
lesions revealed evidence of reorganization and/or plasticity. The pudendal reflex inhibitory pathway, normally ipsilateral and un-
crossed above T8 but exerting a bilateral inhibition on pudendal segmental circuits, exhibited crossed inhibition after chronic lateral
hemisection lesions suggesting the development of novel crossed connections above the lesion (dashed lines). In addition, the pudendal
sympathetic activation pathway, normally unilateral and uncrossed below T8, exhibited bilateral excitation of sympathetic post-
ganglionics suggesting the development of novel crossed connections below the lesion. Pudendal motor neurons are located in the
dorsomedial (DM) and dorsolateral (DL) nuclei of the L5–L6 ventral horn. Pudendal sympathetic neurons are located in the in-
termediate zone of the T13–L2 spinal cord. Templates of the medullary and spinal sections modified from Paxinos and Watson (1998).
420

dorsal nerve of the penis (Johnson and Hubscher, the lateral paragigantocellularis nucleus (Fig. 1,
1998). In marked contrast, no inhibitory effects right panel; Johnson and Hubscher, 1998). As was
could be seen on pudendal reflex discharges elic- true for the pudendal reflex depression sites, si-
ited by pelvic nerve afferents. Bilateral microstim- multaneous microstimulation of both left and
ulation was always more effective in depression of right sympathetic premotor axons, as opposed to
penile afferent-elicited reflexes than unilateral mi- unilateral stimulation, produced the maximum
crostimulation for motoneuron pools on either level of the long latency sympathetic postgangl-
side of the cord. Microstimulation (conditioning ionic axon firing, again demonstrating a decussat-
stimulus alone) never produced direct firing of pu- ion across the midline. Terminations of lateral
dendal motoneurons and the latency of effect paragigantocellularis axons have been found in the
strongly suggests the lumbosacral site of action intermediate zone of the thoracic cord using an-
involves presynaptic inhibition of dorsal nerve of terograde (Hermann et al., 2003) and viral tracers
the penis afferents. The function of the descending (Stornetta et al., 2004). The unmyelinated sympa-
inhibitory pathway may be to coordinate, through thetic postganglionic axons in the motor branch of
the periodic presynaptic inhibition of excitatory the pudendal nerve are synaptically connected to
penile afferent input, the proper sequence of pro- T13–L1 preganglionic neurons in the intermedio-
pulsive contraction bursts interfaced with closure lateral cell column as shown by pseudorabies viral
of the bladder neck and emission of semen. The tracing from the perineal muscles (Marson and
50–75 ms period of penile afferent inhibition elic- McKenna, 1996; Marson and Carson, 1999) and
ited from lateral paragigantocellularis microstim- travel through the caudal sympathetic trunk, the
ulation corresponds to the period of quiescence L5 spinal nerve, but not the pelvic nerve (Johnson
between sequential bursts of contractions of the and Hubscher, 1998). Although the peripheral tar-
bulbospongiosus during ejaculation in the behav- get of the activated sympathetic axons is still un-
ing rat (Meisel and Sachs, 1994). Neurons in the known, they likely innervate blood vessels, glands
lateral paragigantocellularis nucleus contain se- in the penile bulb, and/or structures in the prox-
rotonin (5-HT; Marson and McKenna, 1992), a imal portions of the corpus cavernosum erectile
neurotransmitter known to produce presynaptic tissue (Galindo et al., 1997).
inhibition on primary afferent terminals in the
spinal cord (Peng et al., 2001; Schwartz et al.,
2005), and have been shown to project axons Effects of chronic spinal cord injury on
through the spinal cord to terminate in and adja- sexual function
cent to the pudendal motor nuclei (Marson and
McKenna, 1992; Hermann et al., 2003). The lack Development of an animal model for the study of
of inhibitory effect on pelvic nerve-elicited puden- sexual function following chronic spinal cord in-
dal reflexes argues against a direct postsynaptic jury must carefully take into account the level of
inhibition of pudendal motoneurons from these injury. Since the spinal cord segments containing
microstimulation sites. Presynaptic inhibition of erectile and ejaculatory circuits span the T10–S1
pudendal and urethral afferents of cats has been levels in the rat, studies designed to investigate the
shown in the spinal micturition circuit (reviewed effects of removing long ascending or descending
by Shefchyk, 2002) and recent data has shown pathways need to avoid injuries within this span
5-HT receptors on primary afferent terminals that would disrupt intrinsic spinal segmental cir-
(Maxwell et al., 2003). cuits and their putative interconnections as well as
In addition to the descending inhibition of pe- produce the equivalent of lower motor neuron
nile afferent-elicited pudendal motoneuron reflex- signs. Historically, studies of the effects of chronic
es, sympathetic postganglionic neurons in the (at least 30 day) spinal cord transection in rats
motor branch of the pudendal nerve are strongly have used injuries to the midthoracic (T6–T9) spi-
activated by ipsilateral or contralateral microstim- nal cord, rostral to the autonomic centers for
ulation of a slightly more caudolateral region of erection and emission, and caudal to the T1–T5
421

levels within which severe injuries could produce the visceral afferent inputs originating in the pelvic
complications from autonomic dysreflexia (re- nerve. This reorganization of inputs on the pu-
viewed by Weaver, this volume). It is also impor- dendal motor neuron circuits likely takes time as
tant to use postlesion intervals of at least 30 days shown by the lack of immediate effect of an acute
(chronic lesion), since studies on acute injuries transection (4–6 h). Alternatively, injury discharg-
(hours to a few days) are subject to lingering ef- es and spinal shock may have contributed to some
fects of spinal shock and active plasticity, show of the acute–chronic injury differences.
vastly different results when compared to the Utilizing the electrophysiological model for in-
chronic lesion, and do not provide the degree of vestigating the spino–bulbo–spinal loop (Fig. 1),
clinical correlation to chronic injuries in humans. various degrees of chronic (30 day) incomplete le-
Early behavioral studies on rats with spinal cord sions of the midthoracic cord were used to deter-
transections at the midthoracic level for approxi- mine the bilateral nature of the descending
mately 30 days (Sachs and Garinello, 1979; Hart pudendal inhibitory and sympathetic facilitatory
and Odell, 1981; Mas et al., 1987) documented pathways, the midthoracic white matter pathway
enhanced erectile and depressed ejaculatory reflex- location, and the behavioral effect on sexual re-
es due to loss of supraspinal influences. Dorsal flexes (Hubscher and Johnson, 2000). As expected,
nerve of the penis afferents that are required for lateral midthoracic hemisections did not eliminate
ejaculation and reflexogenic erection likely play a the effects of microstimulating either descending
role in the reorganization of spinal circuits follow- pathway from the medulla, corroborating the bi-
ing chronic injury. In normal unoperated animals lateral terminal architecture in electrophysiological
and in those with an acute 4–6 h T8 transection, (Johnson and Hubscher, 1998, 2000) and neuro-
the magnitude of the pudendal reflex evoked by anatomical (Hermann et al., 2003) studies, and
supramaximal stimulus strength of the penile had a negligible effect on sexual or bladder reflex-
afferent nerves was significantly greater than es. However, chronic lateral hemisections revealed
that evoked by pelvic nerve afferent stimulation a reorganization of both descending pathways that
(Johnson, 1995). This ratio changed over the may have contributed to the functional recovery.
course of 30–60 days after transection such that The pudendal reflex inhibitory pathway, normally
penile afferent stimulation became increasingly less ipsilateral and uncrossed above T8 but exerting a
effective than pelvic afferent stimulation in eliciting bilateral inhibition on pudendal segmental circuits,
a reflex discharge. The decrease in synaptic efficacy exhibited crossed inhibition after chronic lateral
of penile afferents was particularly pronounced on hemisection lesions suggesting the development of
the contralateral (crossed) activation of pudendal novel crossed connections above the lesion (Fig. 1;
motoneurons. Although the pelvic nerve afferent broken lines in left panel). In addition, the puden-
synaptic efficacy did not decrease and may have dal sympathetic activation pathway, normally uni-
increased slightly, the contralateral changes were lateral and uncrossed below T8, exhibited bilateral
not seen with this input circuit. excitation of sympathetic postganglionics suggest-
At 30 and 60 days after spinal cord transection, ing the development of novel crossed connections
a reduction in synaptic efficacy of dorsal nerve of below the lesion (Fig. 1; broken lines in right pan-
the penis afferents was coupled with the relative el). These novel synaptic connections were not seen
strengthening of the pelvic nerve afferent inputs in acute (4–6 h) lateral hemisections (Hubscher
suggesting that some descending pathway(s), likely and Johnson, 2000). The new connections revealed
from the brainstem, normally exerts some type of across the thoracic spinal cord below the level of a
selective facilitation of pudendal afferent input chronic lesion may be due a reorganization of seg-
onto pudendal motoneuron circuitry. Elimination mental autonomic circuits as described by Weaver
of that facilitatory influence after transection et al. (1997, 2001).
injury may have left those circuits approachable In contrast to the spared and novel bilateral
by other segmental systems, as demonstrated by connections following chronic lateral hemisections,
Beattie et al. (1993) in the cat, including possibly severe contusion or dorsal hemisection injuries
422

(dorsal 3/5 of the cord) eliminated the effects of feelings (e.g. arousal, libido) cannot be obtained in
both descending pathways and produced a behavi- animal models, the clinical picture can greatly
oral status similar to spinal transection; reduction expand the interpretation of neuronal mechanisms.
of erectile reflex initiation latency, and the devel- The great majority of men with severe lesions
opment of bladder-sphincter dyssynergia (de cranial to the T10 spinal segment (i) can obtain
Groat, 1995). It is important to note that func- reflexogenic (parasympathetically mediated) erec-
tional behavioral measures of mating-triggered tions but usually lack the desired penile rigidity
ejaculation cannot be performed because of the and/or duration even under high states of arousal
inability of the severely injured rat to physically (Szasz and Carpenter, 1989), (ii) cannot ejaculate
mount a receptive female. By reconstructing and or sense genital-based orgasm (Donohue and
overlapping the lesion extent of the variable acute Gebhard, 1995), and (iii) often have difficulty in
and chronic injuries (Hubscher and Johnson, controlling urethral and anal sphincters during
2000), the approximate lateral white matter loca- sexual activity (Elliott, 2003). All these scenarios
tion of these two pathways was determined (Fig. 1) point to inappropriate, weak, or uncoordinated
in general agreement with neuroanatomical tracing perineal muscle contractions likely due to the loss
studies on normal animals (Martin et al., 1985; of the spinal–bulbo–spinal or segmental coordina-
Hermann et al., 2003). In support of this finding, tion pathways described above.
pseudorabies virus injections into the perineal mus- By activating the afferent fibers in the dorsal
cles failed to infect neurons in the gigantocellularis nerve of the penis with high-intensity penile vibra-
complex 30 days after lesions of the dorsal 3/5 of tory stimulation (PVS), an ejaculation sufficient
the T8 spinal cord (Chadha et al., 2004). for sperm collection can often be elicited (Sønksen
Chronic spinal cord injury compromises normal and Ohl, 2002; Brackett et al., 1997), provided the
sexual function and the disruption of the descend- T11–S4 spinal cord, the dorsal nerve of the penis,
ing pathways modulating the pudendal moto- and the bulbocavernosus reflex are intact (Szasz
neuron reflex circuit likely contributes to the and Carpenter, 1989; Wieder et al., 2000; Bird
incoordination of perineal muscle contractions. et al., 2001). Thus it would seem that the penile
The inappropriate tonic contractions of these mus- afferent input to the pudendal reflex circuitry and
cles following severe chronic injury likely interfere the spinal ejaculation centers inclusive to the T11
with the timing of phasic events mediating elim- segment, are crucial for the application of this
inative functions such as ejaculation, defecation, therapy. The high magnitude and long duration of
and micturition. The differential descending inhi- PVS needed for optimum effects (2.5 mm displace-
bition of penile nerve but not pelvic nerve inputs to ment; Sønksen et al., 1994) are beyond the stim-
segmental reflex circuits may correlate with the ulation intensity needed for reflexogenic erection
finding that the segmental inputs from the penile and normal sensation in able-bodied men, sug-
nerve but not the pelvic nerve become progres- gesting that the central synaptic efficacy of penile
sively ineffective following long-term transection afferents gradually diminishes after injury, similar
injury, possibly resulting from a reorganization of to the data obtained in rat studies.
input-specific interneurons. Another therapeutic strategy, electroejaculation,
involves the stimulation of the pelvic nerve affer-
ents through the wall of the rectum (Halstead
Clinical correlations in men with spinal injury et al., 1987). When successful, this procedure trig-
gers emission but not the expulsive phase of ejac-
Studies of sexual function in men with spinal cord ulation and often produces retrograde emission
injury (reviewed by Elliot, this volume) have into the bladder (Biering-Sorensen and Sønksen,
documented complications, signs, and treatment 2001). Therefore, as in the spinal cord-injured rat,
strategies that correlate with data obtained in ani- an increased synaptic efficacy of pelvic nerve af-
mal studies. In some instances, in which the verbal ferents on segmental ejaculatory circuits may oc-
descriptions of sensations (e.g. orgasm) or emotional cur in chronic injury. However, because of
423

unpleasant sensations, electroejaculation can only afferent terminals (Peng et al., 2001; Schwartz
be applied under anesthesia if any residual as- et al., 2005) possibly through the 5-HT3 receptor
cending sensory pathways are retained (Sønksen found on primary afferent terminals (Maxwell
and Ohl, 2002), presumably in the ventral half of et al., 2003). In addition to serotonin, studies by
the spinal cord. PVS and electroejaculation utilize Holmes et al. in normal and spinal-injured rats
the dorsal nerve of the penis and pelvic nerve sen- (2001) describe a medullary-spinal pathway from
sory afferents, respectively, to activate autonomic the nucleus raphe obscurus which releases thyrotro-
circuits in the thoracolumbar spinal cord including phin releasing hormone on pudendal motor neurons
the proposed spinal ejaculation center (at L3–L4 in and acts synergistically with serotonin to specifically
the rat; Truitt and Coolen, 2002). If the lesion is inhibit sexual and anal sphincter reflexes.
cranial to T7, however, autonomic dysreflexia may The inhibitory neurotransmitter gamma amino-
be produced by either procedure (Frankel and butyric acid (GABA), found in some of the
Mathias, 1980; Szasz and Carpenter, 1989) or by spinally projecting neurons of the lateral par-
stimulation of the dorsal nerve of the penis (Reitz agigantocellularis nucleus (Jones et al., 1991) as
et al., 2003). A recent study has demonstrated well as in spinal interneurons, likely plays a role in
positive sympathetic skin (sudomotor) responses ejaculatory dysfunction after chronic spinal cord
in the hand following stimulation of the urethra in injury. The GABAB receptor is found on primary
men with incomplete spinal cord injuries caudal to afferent terminals, mediates presynaptic inhibi-
the upper thoracic levels (Schmid et al., 2004). tion, and is the site of action of baclofen
Recording a sympathetic skin response in the hand (Li et al., 2004). In rats and humans, baclofen
following stimulation of the penile or pelvic nerves administration inhibits ejaculation and erection of
may become useful in determining whether upper the glans penis (Leipheimer and Sachs, 1988;
thoracic sympathetic centers can be activated by Vaidyanathan et al., 2004) possibly through the
ascending intraspinal pathways through the lesion. tonic presynaptic inhibition of penile afferents.

Pharmacological considerations Summary

The neurotransmitters, neuromodulators, and re- The ejaculatory dysfunction experienced by the
ceptor subtypes identified in the neurons subserv- vast majority of spinal cord-injured men is likely
ing erection and ejaculation are numerous (see caused by the removal of supraspinal modulatory
Giuliano and Rampin, 2004). Some of these are pathways that normally act to coordinate the ac-
likely important for the mechanisms described tivity in the ejaculatory spinal circuits and spinal
above. Several animal studies have demonstrated pattern generator. Continued development of an-
that serotonin (5-HT), released by several descend- imal models for the investigation of electrophys-
ing brainstem pathways, is involved through acti- iological, neuroanatomical, pharmacological, and
vation of certain receptor subtypes primarily as an behavioral characteristics of male sexual responses
inhibitory neurotransmitter (see McKenna, 2000; following chronic spinal cord injury will provide
Marson and Gravitt, 2004). The 5-HT1A receptor, valuable information about a complex visceroso-
however, facilitates ejaculation in rats (Carro- matic system dependent on the rhythmic and co-
Juarez et al., 2003; Truitt et al., 2003) and has been ordinated contraction of smooth and striated
proposed to be hypersensitive in men with prema- muscles. Injury-induced alteration of segmental
ture ejaculation (Waldinger, 2004). The 5-HT2C and descending control of the spinal circuits sub-
receptor has been found on perineal motor serving somatic and autonomic elements of the
neurons (Bancila et al., 1999) along with the 5- perineal muscles, including the development of nov-
HT5A receptor (Doly et al., 2004) and may inhibit el connections, involves spinal pathways from the
ejaculation (Waldinger, 2004). Serotonin release lateral paragigantocellularis nucleus in the medulla
from brainstem pathways has been shown to pro- acting on reflex inputs from the dorsal nerve of the
duce presynaptic inhibition of spinal primary penis and associated autonomic interneurons.
424

Acknowledgments Collins III, W.F., Erichsen, J.T. and Rose, R.D. (1991) Puden-
dal motor and premotor neurons in the male rat: A WGA
Supported by grants from the National Institutes transneuronal study. J. Comp. Neurol., 308: 28–41.
Coolen, L.M., Allard, J., Truitt, W.A. and McKenna, K.E. (2004)
of Health, the Christopher Reeve Paralysis Asso- Central regulation of ejaculation. Physiol. Behav., 83: 203–215.
ciation, the Paralyzed Veterans of America, and de Groat, W.C. (1995) Mechanisms underlying the recovery of
the Brain and Spinal Cord Injury Trust Fund of lower urinary tract function following spinal cord injury.
Florida. Paraplegia, 33: 493–505.
Doly, S., Fischer, J., Brisorgueil, M.J., Verge, D. and Conrath,
M. (2004) 5-HT5A receptor localization in the rat spinal cord
suggests a role in nociception and control of pelvic floor
musculature. J. Comp. Neurol., 476: 316–329.
References Donohue, J. and Gebhard, P. (1995) The Kinsey Institute/In-
diana University report on sexual and spinal cord injury. Sex.
Anderson, K.D. (2004) Targeting recovery: priorities of the Disabil., 13(1): 7–84.
spinal cord-injured population. J. Neurotrauma, 21: Dubrovsky, B. and Filipini, D. (1990) Neurobiological aspects
1371–1383. of the pelvic floor muscles involved in defecation. Neurosci.
Bancila, M., Verge, D., Rampin, O., Backstrom, J.R., Biobehav. Rev., 14: 157–168.
Sanders-Bush, E., McKenna, K.E., Marson, L., Calas, A. Elliott, S. (2003) Sexual dysfunction and infertility in men with
and Giuliano, F. (1999) 5-Hydroxytryptamine2C receptors spinal cord disorders. In: Lin V. (Ed.), Spinal Cord Medicine:
on spinal neurons controlling penile erection in the rat. Principles and Practice (Chapter 26). Demos Medical Pub-
Neuroscience, 92: 1523–1537. lishing, New York, pp. 350–365.
Beattie, M.S., Leedy, M.G. and Bresnahan, J.C. (1993) Frankel, H.L. and Mathias, C.J. (1980) Severe hypertension in
Evidence for alterations of synaptic inputs to sacral spinal patients with high spinal cord lesions undergoing electro-
reflex circuits after spinal cord transection in the cat. Exp. ejaculation–management with prostaglandin E2. Paraplegia,
Neurol., 123: 35–50. 18: 293–299.
Biering-Sorensen, F. and Sønksen, J. (2001) Sexual function in Galindo, R., Barba, V. and Dail, W.G. (1997) The sensory
spinal cord lesioned men. Spinal Cord, 39: 455–470. branch of the pudendal nerve is the major route for adrenergic
Bird, V.G., Brackett, N.L., Lynne, C.M., Aballa, T.C. and innervation of the penis in the rat. Anat. Rec., 247: 479–485.
Ferrell, S.M. (2001) Reflexes and somatic responses as Giuliano, F. and Rampin, O. (2004) Neural control of erection.
predictors of ejaculation by penile vibratory stimulation in Physiol. Behav., 83(2): 189–201.
men with spinal cord injury. Spinal Cord, 39: 514–519. Halstead, L.S., VerVoort, S. and Seager, S.W. (1987) Rectal
Bowens, J.M., Hughes, B.J. and Bradley, W.E. (1984) probe electrostimulation in the treatment of anejaculatory
Anatomic, physiologic, and pharmacologic properties of the spinal cord injured men. Paraplegia, 25: 120–129.
feline bulbospongiosus muscle. Am. J. Vet. Res., 45: 308–313. Hart, B.L. and Leedy, M.G. (1985) Neurological bases of male
Brackett, N.L., Padron, O.F. and Lynne, C.M. (1997) Semen sexual behavior: a comparative analysis. In: Adler N., Pfaff
quality of spinal cord injured men is better when obtained by D. and Goy R.W. (Eds.) Handbook of Behavioral Neuro-
vibratory stimulation versus electroejaculation. J. Urol., 157: biology, Vol. 7. Reproduction. Plenum Press, New York,
151. pp. 373–422.
Breedlove, S.M. and Arnold, A.P. (1980) Hormone accumula- Hart, B.L. and Odell, V. (1981) Elicitation of ejaculation and
tion in a sexually dimorphic motor nucleus of the rat spinal penile reflexes in spinal male rats by peripheral electric shock.
cord. Science, 210: 564–566. Physiol. Behav., 26: 623–626.
Carro-Juarez, M., Cruz, S.L. and Rodriguez-Manzo, G. (2003) Hermann, G.E., Bresnahan, J.C., Holmes, G.M., Rogers, R.C.
Evidence for the involvement of a spinal pattern generator in and Beattie, M.S. (1998) Descending projections from the
the control of the genital motor pattern of ejaculation. Brain nucleus raphe obscurus to pudendal motoneurons in the male
Res., 975: 222–228. rat. J. Comp. Neurol., 397: 458–474.
Carvalho, M., Schwartz, M.S. and Swash, M. (1995) Involve- Hermann, G.E., Holmes, G.M., Rogers, R.C., Beattie, M.S.
ment of the external anal sphincter in amyotrophic lateral and Bresnahan, J.C. (2003) Descending spinal projections
sclerosis. Muscle Nerve, 18: 848–853. from the rostral gigantocellular reticular nuclei complex.
Chadha, H.K., Johnson, R.D. and Hubscher, C.H. (2004) J. Comp. Neurol., 455: 210–221.
Effects of chronic partial mid-thoracic spinal cord injury on Holmes, G.M., Bresnahan, J.C. and Beattie, M.S. (2001) Inhi-
the transneuronal tracing of pseudorabies virus from the bition of pudendal reflexes in spinal rats. Reassessing the role
perineal musculature to the brainstem. Soc. Neurosci. Abstr., of serotonin. Physiol. Behav., 74: 57–64.
30: 458.16. Holmes, G.M., Chapple, W.D., Leipheimer, R.E. and Sachs,
Coleman, A.M. and Sengelaub, D.R. (2002) Patterns of dye B.D. (1991) Electromyographic analysis of male rat perineal
coupling in lumbar motor nuclei of the rat. J. Comp. Neurol., muscles during copulation and reflexive erections. Physiol.
454: 34–41. Behav., 49: 1235–1246.
425

Holmes, G.M., Hermann, G.E., Rogers, R.C., Bresnahan, J.C. Mann, K., Pankok, J., Connemann, B. and Roschke, J. (2003)
and Beattie, M.S. (2002) Dissociation of the effects of Temporal relationship between nocturnal erections and rapid
nucleus raphe obscurus or rostral ventrolateral medulla eye movement episodes in healthy men. Neuropsychobiol.,
lesions on eliminatory and sexual reflexes. Physiol. Behav., 47: 109–114.
75: 49–55. Markowski, V.P., Eaton, R.C., Lumley, L.A., Moses, J. and
Holstege, G. (1991) Descending motor pathways and the spinal Hull, E.M. (1994) A D1 agonist in the MPOA facilitates
motor system: limbic and non-limbic components. Prog. copulation in male rats. Pharmacol. Biochem. Behav., 47:
Brain Res., 87: 307–421. 483–486.
Holstege, G. and Georgiadis, J.R. (2004) The emotional brain: Marson, L. and Carson III, C.C. (1999) Central nervous system
neural correlates of cat sexual behavior and human male innervation of the penis, prostate, and perineal muscles:
ejaculation. Prog. Brain Res., 143: 39–45. a transneuronal tracing study. Mol. Urol., 3: 43–50.
Hubscher, C.H. and Johnson, R.D. (1996) Responses of med- Marson, L. and Gravitt, K. (2004) Spinal neurons activated
ullary reticular formation neurons to input from the male with the urethrogenital reflex in the male rat. Brain Res.,
genitalia. J. Neurophysiol., 76: 2474–2482. 1026: 108–115.
Hubscher, C.H. and Johnson, R.D. (1999) Effects of acute and Marson, L. and McKenna, K.E. (1990) The identification of a
chronic midthoracic spinal cord injury on neural circuits for brainstem site controlling spinal sexual reflexes in male rats.
male sexual function. I. Ascending pathways. J. Neuro- Brain Res., 515: 303–308.
physiol., 82: 1381–1389. Marson, L. and McKenna, K.E. (1992) A role for 5-hydroxy-
Hubscher, C.H. and Johnson, R.D. (2000) Effects of acute and tryptamine in descending inhibition of spinal sexual reflexes.
chronic midthoracic spinal cord injury on neural circuits for Exp. Brain Res., 88: 313–320.
male sexual function. II. Descending pathways. J. Neuro- Marson, L. and McKenna, K.E. (1994) Stimulation of the
physiol., 83: 2508–2518. hypothalamus initiates the urethrogenital reflex in male rats.
Johnson, R.D. (1988) Efferent modulation of penile me- Brain Res., 638: 103–108.
chanoreceptor activity. In: Iggo A. and Hamann W. (Eds.) Marson, L. and McKenna, K.E. (1996) CNS cell groups
Progress in Brain Research, Vol. 73. Transduction and Cel- involved in the control of ischiocavernosus and bulb-
lular Mechanisms in Sensory Receptors. Elsevier, Amster- ospongiosus muscles: a transneuronal tracing study using
dam, pp. 319–324. pseudorabies virus. J. Comp. Neurol., 374: 161–179.
Johnson, R.D. (1995) Effects of chronic spinal cord injury on Martin, G.F., Vertes, R.P. and Waltzer, R. (1985) Spinal pro-
segmental circuits controlling eliminative and sexual func- jections of the gigantocellular reticular formation in the rat.
tions. J. Neurotrauma, 12: 359. Evidence for projections from different areas to laminae
Johnson, R.D. and Halata, Z. (1991) Topography and ultra- I and II and lamina IX. Exp. Brain Res., 58: 154–162.
structure of sensory nerve endings in the glans penis of the Mas, M., del Castillo, A.R., Guerra, M., Davidson, J.M. and
rat. J. Comp. Neurol., 312: 299–310. Battaner, E. (1987) Neurochemical correlates of male sexual
Johnson, R.D. and Hubscher, C.H. (1998) Brainstem micro- behavior. Physiol. Behav., 41: 341–345.
stimulation differentially inhibits pudendal motoneuron Mason, P. (2001) Contributions of the medullary raphe and
reflex inputs. Neuroreport, 9: 341–345. ventromedial reticular region to pain modulation and other
Johnson, R.D. and Hubscher, C.H. (2000) Brainstem micro- homeostatic functions. Annu. Rev. Neurosci., 24: 737–777.
stimulation activates sympathetic fibers in pudendal nerve Maxwell, D.J., Kerr, R., Rashid, S. and Anderson, E. (2003)
motor branch. Neuroreport, 11: 1–4. Characterisation of axon terminals in the rat dorsal horn that
Johnson, R.D. and Murray, F.T. (1992) Reduced sensitivity of are immunoreactive for serotonin 5-HT3A receptor subunits.
penile mechanoreceptors in aging rats with sexual dysfunc- Exp. Brain Res., 149: 114–124.
tion. Brain Res. Bull., 28: 61–64. McKenna, K.E. (2000) Some proposals regarding the organi-
Jones, B.E., Holmes, C.J., Rodriguez-Viega, E. and Mainville, zation of the central nervous system control of penile erec-
L. (1991) GABA-synthesizing neurons in the medulla: their tion. Neurosci. Biobehav. Rev., 24: 535–540.
relationship to serotonin-containing and spinally projecting McKenna, K.E., Chung, S.K. and McVary, K.T. (1991) A
neurons in the rat. J. Comp. Neurol., 312: 1–19. model for the study of sexual reflexes in anesthetized male
Kippin, T.E., Sotiropoulos, V., Badih, J. and Pfaus, J.G. (2004) and female rats. Am. J. Physiol., 261: R1276–R1285.
Opposing roles of the nucleus accumbens and anterior lateral McKenna, K.E. and Nadelhaft, I. (1986) The organization of
hypothalamic area in the control of sexual behaviour in the the pudendal nerve in the male and female rat. J. Comp.
male rat. Eur. J. Neurosci., 19: 698–704. Neurol., 248: 532–549.
Leipheimer, R.E. and Sachs, B.D. (1988) GABAergic regula- McKenna, K.E. and Nadelhaft, I. (1989) The pudendo-puden-
tion of penile reflexes and copulation in rats. Physiol. Behav., dal reflex in male and female rats. J. Autonom. Nerv. Syst.,
42: 351–357. 27: 67–77.
Li, Y., Li, X., Harvey, P.J. and Bennett, D.J. (2004) Effects of Meisel, R.L. and Sachs, B.D. (1994) The physiology of male
baclofen on spinal reflexes and persistent inward currents in sexual behavior. In: Knobil E. and Neill J.D.D. (Eds.) The
motoneurons of chronic spinal rats with spasticity. J. Neuro- Physiology of Reproduction, Vol. 2. Raven Press, New York,
physiol., 92: 2694–2703. pp. 3–105.
426

Murphy, A.Z. and Hoffman, G.E. (2001) Distribution of go- Steers, W.D. (2000) Neural pathways and central sites involved
nadal steroid receptor-containing neurons in the preoptic- in penile erection: neuroanatomy and clinical implications.
periaqueductal gray-brainstem pathway: a potential circuit Neurosci. Biobehav. Rev., 24: 507–516.
for the initiation of male sexual behavior. J. Comp. Neurol., Stornetta, R.L., McQuiston, T.J. and Guyenet, P.G. (2004)
438: 191–212. GABAergic and glycinergic presympathetic neurons of rat
Nadelhaft, I. and McKenna, K.E. (1987) Sexual dimorphism in medulla oblongata identified by retrograde transport of
sympathetic preganglionic neurons of the rat hypogastric pseudorabies virus and in situ hybridization. J. Comp.
nerve. J. Comp. Neurol., 256: 308–315. Neurol., 479: 257–270.
Nunez, R., Gross, G.H. and Sachs, B.D. (1986) Origin and Sung, J.H., Mastri, A.R. and Segal, E. (1979) Pathology of
central projections of rat dorsal penile nerve: possible direct Shy-Drager syndrome. J. Neuropathol. Exp. Neurol., 38:
projection to autonomic and somatic neurons by primary af- 353–368.
ferents of nonmuscle origin. J. Comp. Neurol., 247: 417–429. Szasz, G. and Carpenter, C. (1989) Clinical observations in vi-
Ohl, D.A., Bennett, C.J., McCabe, M., Menge, A.C. and bratory stimulation of the penis in men with spinal cord in-
McGuire, E.J. (1989) Predictors of success in electroejacula- jury. Arch. Sexual Behav., 18: 461–474.
tion of spinal cord injured men. J. Urol., 142: 1483–1486. Tanaka, J. and Arnold, A.P. (1993) An electrophysiological
Paxinos, G. and Watson, C. (1998) The Rat Brain In Stereo- study of descending projections to the lumbar spinal cord in
taxic Coordinates (4th ed.). Academic Press, San Diego. adult male rats. Exp. Brain Res., 96: 117–124.
Pehek, E.A., Thompson, J.T. and Hull, E.M. (1989) The effects Truitt, W.A. and Coolen, L.M. (2002) Identification of a
of intracranial administration of the dopamine agonist apo- potential ejaculation generator in the spinal cord. Science,
morphine on penile reflexes and seminal emission in the rat. 297: 1566–1569.
Brain Res., 500: 325–332. Truitt, W.A., Shipley, M.T., Veening, J.G. and Coolen, L.M.
Peng, Y.B., Wu, J., Willis, W.D. and Kenshalo, D.R. (2001) (2003) Activation of a subset of lumbar spinothalamic neu-
GABA(A) and 5-HT(3) receptors are involved in dorsal root rons after copulatory behavior in male but not female rats.
reflexes: possible role in periaqueductal gray descending in- J. neurosci., 23: 325–331.
hibition. J. Neurophysiol., 86: 49–58. Vaidyanathan, S., Watt, J.W., Singh, G., Hughes, P.L.,
Reitz, A., Schmid, D.M., Curt, A., Knapp, P.A. and Schurch, Selmi, F., Oo, T., Soni, B.M. and Sett, P. (2004) Man-
B. (2003) Autonomic dysreflexia in response to pudendal agement of recurrent priapism in a cervical spinal cord
nerve stimulation. Spinal Cord, 41: 539–542. injury patient with oral baclofen therapy. Spinal Cord,
Sachs, B.D. and Garinello, L.D. (1979) Spinal pacemaker con- 42: 134–135.
trolling sexual reflexes in male rats. Brain Res., 171: 152–156. Wagner, C.K. and Clemens, L.G. (1991) Projections of the
Schmid, D.M., Reitz, A., Curt, A., Hauri, D. and Schurch, B. paraventricular nucleus of the hypothalamus to the sexually
(2004) Urethral evoked sympathetic skin responses and vis- dimorphic lumbosacral region of the spinal cord. Brain Res.,
cerosensory evoked potentials as diagnostic tools to evaluate 539: 254–262.
urogenital autonomic afferent innervation in spinal cord in- Waldinger, M.D. (2004) Lifelong premature ejaculation: from
jured patients. J. Urol., 171: 1156–1160. authority-based to evidence-based medicine. BJU Int., 93:
Schmidt, M.H. and Schmidt, H.S. (1993) The ischiocavernosus 201–207.
and bulbospongiosus muscles in mammalian penile rigidity. Weaver, L.C., Cassam, A.K., Krassioukov, A.V. and
Sleep, 16: 171–183. Llewellyn-Smith, I.J. (1997) Changes in immunoreactivity
Schroder, H.D. (1985) Anatomical and pathoanatomical studies for growth associated protein-43 suggest reorganization of
on the spinal efferent systems innervating pelvic structures. synapses on spinal sympathetic neurons after cord transec-
1. Organization of spinal nuclei in animals. 2. The nucleus tion. Neuroscience, 81: 535–551.
X-pelvic motor system in man. J. Auton. Nerv. Syst., 14: 23–48. Weaver, L.C., Verghese, P., Bruce, J.C., Fehlings, M.G., Krenz,
Schwartz, E.J., Gerachshenko, T. and Alford, S. (2005) 5-HT N.R. and Marsh, D.R. (2001) Autonomic dysreflexia and
prolongs ventral root bursting via presynaptic inhibition primary afferent sprouting after clip-compression injury of
of synaptic activity during fictive locomotion in lamprey. the rat spinal cord. J. Neurotrauma, 18: 1107–1119.
J. Neurophysiol., 93: 980–988. Wieder, J.A., Brackett, N.L., Lynne, C.M., Green, J.T. and
Shefchyk, S.J. (2002) Spinal cord neural organization control- Aballa, T.C. (2000) Anesthetic block of the dorsal penile
ling the urinary bladder and striated sphincter. Prog. Brain nerve inhibits vibratory-induced ejaculation in men with spi-
Res., 137: 71–82. nal cord injuries. Urology, 55: 915–917.
Sønksen, J., Biering-Sorensen, F. and Kristensen, J.K. (1994) Yang, C.C. and Bradley, W.E. (1999) Somatic innervation of
Ejaculation by penile vibratory stimulation in men with the human bulbocavernosus muscle. Clin. Neurophysiol.,
spinal cord injured. Paraplegia, 32: 651–660. 110: 412–418.
Sønksen, J. and Ohl, D.A. (2002) Penile vibratory stimulation Yells, D.P., Hendricks, S.E. and Prendergast, M.A. (1992)
and electroejaculation in the treatment of ejaculatory dys- Lesions of the nucleus paragigantocellularis: effects on
function. Int. J. Androl., 25: 324–332. mating behavior in male rats. Brain Res., 596: 73–79.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 29

Male fertility and sexual function after spinal


cord injury

D.J. Brown1,, S.T. Hill1,2 and H.W.G. Baker2,3

1
Victorian Spinal Cord Service, Austin Health, Heidelberg, Vic., Australia
2
Melbourne IVF Reproductive Services, Royal Women’s Hospital, Melbourne, Vic., Australia
3
University of Melbourne Department of Obstetrics and Gynaecology, Carlton, Vic. 3058, Australia

Abstract: Spinal cord injury has an enormous impact upon the sexual relationship of a man and his
partner. Erection may be partial or absent, orgasm altered or impossible, and fertility severely impaired.
New understanding of the physiology of sexual function and improved treatment can enable most cord-
injured men to achieve erections suitable for sexual satisfaction. Modern methods of sperm collection and
fertility treatment mean that many can also be fathers. The best results are obtained by a team approach
involving rehabilitation and reproductive medicine clinicians, nurses, spinal cord injury specialists and
counselors with the cord-injured man and his partner. Erections can be achieved by drugs, such as si-
ldenafil, that block phosphodiesterase 5, prolonging the action of nitric oxide with resultant smooth muscle
relaxation. Intracavernosal prostaglandin E1 and mechanical systems, such as vacuum pumps and con-
striction rings, are also effective. Sexual gratification can be promoted in the context of an understanding
relationship in which the cord-injured person can gain pleasure from pleasing his partner and also from his
partner’s exploration of erotogenic areas not affected by the spinal cord injury. An emphasis on the broader
view of sexuality in relationships allows for a continuance and strengthening of bonds between the couple.
Vibration ejaculation or electroejaculation can be used to collect semen. For a limited period in the acute
phase, usually for about 6–12 days after injury, normal semen can be obtained by electroejaculation from
some cord-injured men. With chronic spinal cord injury the semen is of variable quality. Some patients have
necrospermia, which may be improved by regular ejaculation. Others have poor quality semen or sperm-
atogenic disorders and, in this situation, in vitro fertilization techniques must be used to achieve parent-
hood. Trials of assisted ejaculation help individualize cost-effective management of the infertility.

Introduction significant impact upon the man and his partner.


In younger people in particular, the bigger picture
‘‘Sexual disturbances, which inevitably follow a se- of sexuality and personal relationship is lost as
vere injury of the spinal cord, regardless of whether they focus on sexual performance and hopes of
the lesion is complete or incomplete, constitute a parenthood. To the cord-injured man and his
complex problem in the rehabilitation of paraplegics partner, the closely integrated facets of erection,
and even more in tetraplegics’’ (Guttmann, 1973). ejaculation and orgasm become separated and,
The profound changes in sexual function and fer- with this, the prospects of sexual pleasure and
tility in men after spinal cord injury have a very parenthood seem like lost dreams. The loss of
erections suitable for intercourse is a major blow
Corresponding author. Tel.: +94963030; Fax: +9496 3626; to the cord-injured man’s self-esteem and self-con-
E-mail: DouglasJ.Brown@austin.org.au fidence and can lead to a belief that no woman

DOI: 10.1016/S0079-6123(05)52029-6 427


428

would want him. The prospect of infertility in the acquired necrospermia suggests that genital
a partner also affects the woman’s hopes and tract stasis is the main cause but a number of
expectations. The more important coitus and hav- unresolved questions remain (Mallidis et al., 2000).
ing children are to a cord-injured person and his This work has also aided understanding the devel-
partner, the more profound the impact on their opment of sperm defects, particularly necrospermia,
relationship. in able-bodied men. In this chapter we review the
And so it was. But now the scene has changed pathophysiology of autonomic dysfunction of the
almost beyond recognition due to improved un- male reproductive tract after spinal cord injury,
derstanding of the physiology and pathophysiolo- previous studies of the associated testicular and
gy of sexual function. The pioneering work of sperm defects, studies on necrospermia after chron-
people like Giles Brindley in the treatment of im- ic spinal cord injury, a possible mechanism of the
potence and the development of in vitro fertiliza- development of the common sperm defects present
tion programs, in particular intracytoplasmic after cord injury and clinical approaches for the
sperm injection, have given hope of an improved management of the male infertility and sexual dys-
sexual life and of parenthood (Brindley, 1981a, b, function after spinal cord injury.
1982, 1983a, 1983b, 1984, 1988; Brindley et al.,
1982; Schatte et al., 2000). The introduction of
Pathophysiology of autonomic dysfunction affecting
sexual counseling about these developments dur-
male fertility and sexual function
ing rehabilitation has given new hope to cord-in-
jured individuals and their partners.
The names of Bors and Comarr are associated
Modern management of sexual dysfunction fol-
with the early studies quantifying sexual dysfunc-
lowing spinal cord injury is based upon better un-
tion following spinal cord injury. They found, in
derstanding of the physiology of erection, the
their classic work, that the higher the spinal cord
development of sophisticated techniques for re-
injury, the more likely the man was to have reflex
trieval of sperm and the achievement of fertiliza-
but no psychogenic erections, whereas lesions in
tion in spite of highly adverse circumstances.
the sacral region were likely to be associated with
However, our understanding of the mechanisms
psychogenic, rather than reflex, erections (Bors
whereby disruption of the nervous system causes
and Comarr, 1960). Ejaculation in men with com-
such profound sexual disability remains poor. We
plete upper motor neuron lesions was rare. Men
still do not know clearly how erection, ejaculation
with lower motor neuron lesions could rarely have
and orgasm are normally integrated, nor do we
erections or ejaculate. The less complete the lesion,
know the cause of male infertility after spinal cord
the more likely was the man to have reflex and
injury. Treatments are therefore incomplete.
psychogenic erections and ejaculate. However,
Male fertility and sexual function are dramati-
some men suffer pain and spasm with sexual ac-
cally impaired by spinal cord injury with less than
tivity (Slot et al., 1989).
5% of men able to procreate without medical in-
tervention (Talbot, 1955; Thomas, 1983). Modern
vibroejaculation and electroejaculation equipment Erection
allows sperm to be collected from the majority of
cord-injured men (Lim et al., 1994; Brackett, Penile erection occurs in 95% of men with com-
1999). However, with chronic spinal cord injury, plete spinal cord injury and upper motor neuron
the semen quality is variable and generally poor; lesions, although the erection is often of poor
particularly, there is necrospermia (low sperm mo- quality (insufficient rigidity) or poorly sustained.
tility and viability, Mallidis et al., 2000). The fact Why is this so, particularly when the reflex path-
that collected sperm is normal in the acute phase ways appear to be intact? Contact stimulation of-
of spinal cord injury indicates that this defect takes ten does not lead to erections as good as those
some time to develop (Mallidis et al., 1994). which occurred before the injury. Before exploring
Research aimed at discovering the mechanism of the causes for this dysfunction, the innervation
429

and neurotransmission key to the erectile response polypeptide by the same axons. This peptide is a
must be reviewed (see also Burns et al., 2001). The powerful smooth muscle relaxant. In addition,
male sex organs are innervated by three sets of acetylcholine triggers the release of nitric oxide,
nerves: the pelvic nerves (nervi erigentes, para- another powerful smooth muscle relaxant, by end-
sympathetic), the hypogastric nerves (sympathetic) othelial cells and other cells of the erectile tissue.
and the pudendal nerves (somatic). The pelvic The nitric oxide binds to smooth muscle receptors
nerves contain the axons of sacral parasympathetic of the corpus cavernosum to increase cyclic gua-
preganglionic neurons situated in the intermedio- nosine monophosphate (cGMP) levels and thus
lateral column of sacral (S) spinal cord segments relax smooth muscles. Reversal of this process by
S2–S4. These axons leave the spinal cord in the phosphodiesterase 5, converting cGMP into GMP,
ventral roots S2–S4. Electrical stimulation of these can be inhibited to promote and prolong erection.
ventral roots causes erection (Brindley et al., This is the basis of the use of sildenafil and other
1982). The hypogastric nerves contain the axons phosphodiesterase 5 inhibitors, such as vardenafil
of the sympathetic preganglionic neurons situated and taladafil, in the treatment of impotence (Derry
in the intermediolateral column of the 11th and et al., 2002).
12th thoracic (T) and 1st and 2nd lumbar (L) spi- While acetylcholinesterase positive axons have
nal segments. The pudendal nerves contain the been described in human cavernous tissue, and
axons of somatic motoneurons situated in the acetylcholine synthesis and release has been dem-
ventral horn of sacral cord segments S2–S4. Sym- onstrated in the same tissue (Blanco et al., 1988),
pathetic and parasympathetic preganglionic axons Shirai et al. (1973) could find no evidence of ace-
in the hypogastric and pelvic nerve, respectively, tylcholinesterase in cavernous tissue from able-
synapse in the pelvic plexus with ganglionic neu- bodied men or from men with a variety of diseases
rons. The postganglionic axons form the cavern- causing impotence. The mechanism of action of
ous nerves that innervate the smooth muscle of the parasympathetic nerves in producing erections
blood vessels, erectile tissue and nonerectile tissue must still be considered open to question (Lopez
(e.g., vas deferens) and glandular tissue. Some of and Koller, 2000).
these axons are cholinergic, some are adrenergic The sympathetic nerves of the pelvic plexus can
and the rest are noncholinergic, nonadrenergic. also cause erection when stimulated (Brindley,
Somatic motor axons in the pudendal nerves con- 1988). However, it is known that infusion of
trol the bulbo-cavernous and ischio-cavernous alpha2-adrenergic antagonists cause erections,
muscles that are striated muscles situated at the showing that noradrenalin is, in fact, the main
base of the penis. Sensory axons from the male sex anti-erectile neurotransmitter (Levin and Wein,
organs run mainly in the pudendal nerves. Erec- 1980). In a study of corpora cavernosa noradren-
tion is produced by dilatation of the arterioles that aline and acetylcholine content, Melman et al.
supply the erectile tissue of the corpora cavernosa. (1980) found normal levels of noradrenalin in men
As the erectile tissue fills with blood, the veins that with spinal cord injury. Thus the sympathetic sys-
drain the erectile tissue are compressed against the tem seems to have both erectile and anti-erectile
stiff fibrous envelope of the corpora cavernosa and actions, particularly the latter. How these oppos-
venous outflow decreases. Erection is likely to in- ing actions are regulated and integrated with para-
volve inhibition of the activity of noradrenergic sympathetic activity is not known.
axons innervating the corpora cavernosa, since Intracavernosal injection therapy aims at in-
noradrenaline causes constriction of the arterioles hibiting sympathetic tone or at relaxing smooth
that supply the erectile tissue by acting on alpha- muscle by a direct action, thereby causing an
adrenergic receptors. Erection is also likely to erection. Brindley (1983b) showed that the alpha-
involve increased activity of cholinergic axons. adrenergic receptor blocker, phenoxybenzamine,
Acetylcholine is thought to act by inhibiting the was effective. The drugs most commonly used to-
release of noradrenaline by noradrenergic axons day are papaverine, phentolamine and prostaglan-
and to facilitate the release of vasoactive intestinal din E1. Papaverine increases cyclic adenosine
430

monophosphate, promotes calcium efflux from semen to the proximal urethra. The delivery is due
cells and thereby induces smooth muscle relaxa- to contraction of the smooth muscle of the
tion. Phentolamine blocks alpha1- and alpha2-ad- epididymis, vas deferens, prostate and seminal
renergic receptors and has a direct relaxant effect vesicles (innervated by sympathetic axons). Spinal
on smooth muscles. Given alone it does not pro- cord injury causes loss of coordination of these
duce an erection. It is commonly used in conjunc- reflexes and loss of emission and ejaculation.
tion with papaverine. Prostaglandin E1, the most Psychogenic and genital stimulation usually are
widely used drug for intracavernosal injection, unsuccessful in producing ejaculation in complete
causes vasodilatation, smooth muscle relaxation injuries above T10. Injuries below that level may
and therefore penile erection. Men with spinal permit psychogenic ejaculation (Thomas, 1983).
cord injury are much more sensitive to prostaglan-
din E1 than men with other forms of impotence
and may respond to as little as 1–2 mg. Testicular disorders associated with spinal
A vacuum pump and ring can be used to cause an cord injury
erection suitable for intercourse (Denil et al., 1996).
Penile prostheses are used rarely now that less Many factors could affect testicular function in
destructive treatments for impotence are available. men with spinal cord injury. Hypogonadism and
Objective measurement of sexual function in the gynecomastia were described to be common in
clinical setting can be difficult as it largely relies on paraplegic men from the time of the Second World
self-reporting. The International Index of Erectile War. It is possible that the testicular failure was
Function (IIEF) has been developed to measure, in related to a severe catabolic state following the
a standardized way, five areas of sexual function – spinal cord injury and, with recovery in the sur-
erectile function, intercourse satisfaction, orgasm vivors, there was a period of relative estrogen
function, sexual desire and overall satisfaction. excess causing the development of gynecomastia
This tool has been used for measuring the efficacy [re-feeding gynecomastia (Baker, 2001)]. This
of phosphodiesterase inhibitors and comparing the pattern is not seen today because of better
relative benefits of newer agents such as tadalafil nutrition and rehabilitation techniques. Other
and vardenafil to the more established agent coincidental injuries could cause gonadotropin
sildenafil (Del Popolo, 2004). deficiency, direct testicular damage or disruption
of the male genital tract. Drugs used in the
management of spasticity and other complications
Ejaculation of chronic spinal cord injury could also cause
hyperprolactinemia or gonadotropin suppression.
Ejaculation is a complex process involving coor- While hormonal abnormalities are still reported in
dination of erection, propulsion of semen and some men with chronic spinal cord injury (Naderi
prevention of retrograde seminal flow. A review by and Safarinejad, 2003), most investigators do not
Thomas (1983) provides a clear, concise account of consistently find abnormalities (Baker, 2001).
these events. Ejaculation may be defined as the Autonomic dysfunction has been shown in
expulsion of semen (sperm plus secretions of pros- animals with experimental spinal cord damage to
tate, seminal vesicles and other glands) from the affect spermatogenesis (Ohl et al., 2001; Huang
distal urethra. It is produced by rhythmic contrac- et al., 2004). Specific patterns of disrupted sperm
tions of the urethral smooth muscle (innervated by production have been claimed to occur with spinal
sympathetic axons) and of two striated muscles cord lesions at particular levels in men; however,
at the base of the penis, the ischio- and bulbo- this has not been confirmed (Bors and Comarr,
cavernosus muscle (innervated by somatic motor 1963; Chapelle et al., 1993; Elliott et al., 2000).
axons). The external bladder sphincter (striated Nonspecific defects of spermatogenesis such as
muscle innervated by somatic motor axons) is hypospermatogenesis and germ cell arrest are
closed. Emission may be defined as the delivery of found in some men with chronic spinal cord
431

injury but they may be coincidental (Elliott et al., seminal vesicles and distal vas deferens accounting
2000; Mallidis et al., 2000). Urinary infections for the azoospermic samples obtained during
spreading to the vasa deferentia, epididymides and spinal shock. There is also a requirement for func-
testes may also temporarily or permanently impair tional perivasal nerves for stimulation of contrac-
sperm production or genital tract patency. tion of the cauda epididymis to deliver the stored
Urinary bladder catheterization may cause genital sperm, which is possible once the spinal shock
tract obstruction. Obstruction may result in the abates. Failure to stimulate the pudendal nerves is
formation of sperm antibodies (Hirsch et al., probably the reason that emission, rather than
1992). Fever from sepsis or disturbed temperature ejaculation, occurs.
regulation could also impair spermatogenesis or For electroejaculation we use a rechargeable,
epididymal function (see below). However, these battery-powered, electrical stimulator, the ‘‘CGS
conditions are not generally present in all men Electrojector’’ (Ratek Industries P/L 60 Wadhurst
with spinal cord injury and the most frequent Drive, Boronia, Vic., 3155, Australia, Lim et al.,
dysfunction is defective ejaculation that occurs in 1994; Mallidis et al., 1994). It provides a sine wave
about 95% of cord-injured men (Talbot, 1955). current at 20 Hz of progressively increasing
amplitude to a maximum of 500 mA. The stimula-
tor is fitted with a blunt-end Delrin probe which
Alterations of the semen associated with spinal houses three anteriorly placed, stainless steel,
cord injury longitudinal, bar electrodes. A thermocouple in
the middle electrode monitors temperature at the
Methods of sperm collection electrode-mucosal interface. The temperature is
read on a screen built into the stimulator casing.
Guttmann (1973) developed assisted ejaculation In 9–41% men with complete spinal cord injury
with intrathecal neostigmine for humans with spi- undergoing electroejaculation, there is partial or
nal cord injury in the 1940s. Brindley (1981b) complete retrograde ejaculation, that is, retrograde
pioneered the technique of electroejaculation, flow of semen into the bladder (Brindley, 1984;
using small electrode-containing probes embedded Siosteen et al., 1990; Lim et al., 1994). This is rec-
in a plastic sheath that fit over the index finger. ognized by failure of antegrade flow of semen and
After insertion into the rectum the electrodes were the presence of sperm in the urine collected from
placed in different positions until an emission was the bladder after the procedure. To prevent retro-
obtained upon stimulation. Probes with electrodes grade ejaculation, we use a nontoxic, all-silicone
with larger area have become standard as they Foley catheter inserted into the bladder. Lubricat-
seem more effective and safer than the original ing gels are avoided, as they are known to be toxic
probes (Halstead et al., 1987). The large area elec- to sperm (Lim et al., 1994). Once the catheter is in
trode equipment has been further improved. The situ, the balloon is inflated with 10 ml of saline.
probe is placed in the rectum with the electrodes Urine is drained into a collection bag. An assistant
anterior and the electrical stimulation is presumed applies constant, gentle traction to the catheter to
to stimulate directly the accessory sex organs and tamponade the bladder neck (Lim et al., 1994).
nerves responsible for emission. By performing After proctoscopy to detect any pre-existing
repeated electroejaculation in patients in the acute lesions, the rectal probe is inserted and the
phase of spinal cord injury, we found that during stimulator output progressively increased toward
spinal shock semen with few or no sperm was the maximum output or until emission occurs. The
obtained. Thereafter, good quality semen was rectal temperature is kept below 401C. Antegrade
often produced for a few days until about 2 weeks emission occurs through the urethra around the
after the injury when sperm numbers or motility catheter. The semen is collected in a warm, sterile,
became low (Mallidis et al., 1994). We believe this is plastic jar. The proctoscope is reintroduced to
consistent with a direct effect of electroejaculation check the condition of the rectal mucosa and the
on the autonomic nerve supply of the prostate, catheter removed.
432

The technique of vibration ejaculation (vibro- may be used if assisted ejaculation is not available
ejaculation) has also improved recently (Brindley, or fails, intracytoplasmic sperm injection is
1981a; Brackett, 1999). The specifications of the required as usually insufficient mature sperm are
machinery vary. The key is adequate vibration fre- obtained for artificial insemination.
quency (80 Hz) and amplitude (2.5 mm; Brindley,
1981a; Brackett, 1999). The technique relies on an Semen abnormalities
intact spinal reflex arc, including sacral segments
and thoracolumbar segments up to T10, and y it seems that non-drainage is one factor in the
works by an augmentation of the normal ejacula- poor quality of paraplegic semen. Almost certainly
tory reflex (Wieder et al., 2000). The vibrator is chronic infection in the genital tract is another, yA
applied to the ventral surface of the glans penis to third potentially remediable factor is raised scrotal
initiate the ejaculatory reflex. Brackett (1999) temperaturey (Brindley, 1983a)
reported a 50% success rate of vibroejaculation Analysis of semen collected by assisted ejacula-
in 211 cord-injured men using a 2.5 mm amplitude tion gives variable results from person to person
vibrator. The success rate was higher than when a after cord injury. In some subjects there are no
standard 1.6 mm vibrator was used. The success sperm; occasionally the semen is normal; usually
rate with the 2.5 mm vibrator was related to level there is a normal to high sperm concentration with
of spinal cord injury. Lesions at C3–7, T1–5, extremely low sperm motility and viability. The
T6–10, and T11–L3 were associated with success latter semen pattern is called necrospermia and it
rates of 66%, 54%, 41% and 36%, respectively. It also occurs, rarely, in able-bodied infertile men
has been noted that, when successful, vibroejacu- who can ejaculate (see below; Wilton et al., 1988;
lation produces sperm of better quality than does Mallidis et al., 2000). A number of abnormalities
electroejaculation (Brackett et al., 1997, 1998). of the semen collected from men with chronic
Assisted ejaculation may induce autonomic spinal cord injury have been described, including:
dysreflexia in dysreflexia-prone men, i.e., those increased leukocyte numbers (from very low num-
with spinal cord injury above T6 (Guttmann, 1973; bers, o1 million/ml), increased levels of inflam-
Brackett, 1999; Scheutzow and Bockenek, 2000). matory cytokines (normally found in semen),
Careful monitoring during trials of vibroejaculation increased presence of reactive oxygen species (from
or electroejaculation and the prophylactic use very low values normally) and changes in prostate-
of the calcium channel blocker nifedipine are specific antigen (De Lamirande et al., 1995; Aird
essential (Brackett, 1999). Electroejaculation et al., 1999; Lynne et al., 1999; Trabulsi et al.,
requires general anesthesia in men with incom- 2002; Basu et al., 2004). The increased numbers of
plete lesions as pain is induced in those with leukocyte and levels of cytokines may occur
sensory awareness in their sacral dermatomes. because of inflammation and they further impair
Rectal trauma may be induced by the procedure sperm quality but probably are not the only, or
for electroejaculation. Other methods of stimulat- main, cause of semen abnormality. Some have
ing emission of semen with oral or intrathecal reported that the seminal plasma will impair the
drugs (e.g., the catecholamine uptake blocker, motility of normal test sperm (Brackett et al.,
imipramine, or the cholinesterase inhibitor, 1996; Monga et al., 2001). While some investiga-
neostigmine) are rarely effective and are potential- tors believe abnormalities of the secretions of the
ly dangerous (Guttmann, 1973). accessory sex organs are responsible for the defec-
Sperm may also be obtained from the genital tive sperm motility, at this stage it remains unclear
tract by prostatic massage or surgically by hemi- whether these changes cause the sperm defect or
section of the vas deferens, epididymal aspiration are merely associations.
or needle aspiration or open testicular biopsy It is important to consider the possibility that
(Guttmann, 1973; Hovatta and von Smitten, 1993; the assisted ejaculation technique itself may
Buch, 1994; Watkins et al., 1996; Marina et al., contribute to the poor semen analysis result
1999; Brackett et al., 2000). While these approaches through, for example, exposure of the sperm to
433

toxic material in rubber or plastic, urine contam- random assisted ejaculation in men with chronic
ination or failure to stimulate emission from the spinal cord injury may come from the seminal
epididymal sperm store (Lim et al., 1994). The vesicles (Ohl et al., 1999).
person’s bladder management also relates to the
results of assisted ejaculation (Ohl et al., 1992; Mechanisms of semen abnormalities
Rutkowski et al., 1995; Brackett et al., 1998).
There are several reports that sperm obtained Testicular temperature
from the vas deferens of men with chronic spinal
cord injury have better motility than sperm in the Brindley (1982, 1983a) demonstrated that intra-
semen collected by assisted ejaculation (Hovatta scrotal temperatures were elevated in 29 men with
and von Smitten, 1993; Buch, 1994; Brackett et al., chronic spinal cord injury sitting in wheel chairs
2000). Brackett et al. (2000) examined sperm from with temperatures about 11C higher (36.21C) than
the vas deferens aspirates and from semen of men in clothed able-bodied controls sitting in wheel
with chronic spinal cord injury and from controls chairs for 20 min (35.31C). He found no difference
undergoing vasectomy. The mean sperm motility in scrotal temperatures of cord-injured men and
and viability (expressed as a percentage of the controls lying in bed. In a smaller data set he
sperm) in the cord-injured men’s semen were 14% found suggestive evidence for poorer results of
and 26% and in vas deferens aspirates were 54% electroejaculation in that only two of nine with
and 74%, respectively. In the control men, semen scrotal temperatures above 36.41C had motile
values had 74% motility and 85% viability and vas sperm compared with eight of nine with lower
deferens aspirates had 78% motility and 89% vi- scrotal temperatures. Old data also suggested
ability. While the sperm motility is better in the vas a relationship between impaired spermatogenesis,
deferens of the cord-injured men than in their semen, level of lesion and impairment of sweating
it is still lower than in the controls. Interestingly they (Guttmann, 1973). However, others have not been
also found, in two cord-injured men, abnormalities able to confirm a clear relationship between
of the vas deferens described as edema or fibrosis. testicular temperature and results of assisted ejac-
It is known that the cauda epididymis is the ulation (Brackett et al., 1994). Bedford (1991,
storage organ of sperm and that the seminal 1994) has reviewed the effects of elevated temper-
vesicles do not normally contain sperm. Jarow ature on cauda epididymal function in rats, rabbits
(1996) performed bilateral needle aspirations of and hamsters and suggests there may be a parallel
the seminal vesicles under transrectal ultrasound in humans. In animals, the size of the tail of the
guidance and confirmed that the seminal vesicles epididymis was reduced and the rate of transit of
of fertile men ejaculating regularly do not contain sperm through the epididymis was increased.
significant numbers of sperm. However, with 5 Sperm in the tail of the epididymis were reduced
days of abstinence from ejaculation the sperm in number, motility and viability in the rat. Sperm
concentration in seminal vesicle aspirates was quality improved with return of normal temperature
found to range up to 10  106/ml in one-third of control or with frequent ejaculation (Bedford, 1994).
the subjects. No sperm aspirated from the seminal We suspect genital tract stasis and disturbed
vesicles were actively motile. The suggestion was thermoregulation impairing epididymal sperm
that the number of sperm present might be directly storage are probably the main causes of the com-
related to the duration of the abstinence. If mon pattern of low sperm motility and viability in
neuropathic anejaculation behaved as a ‘‘function- the semen of men with chronic spinal cord injury.
al obstruction’’ causing a sludging of sperm in the
reproductive tract there may be reflux of sperm into Stasis
the seminal vesicles. This has been confirmed by
Ohl et al. (1999). By performing electroejaculation We have shown that the poor quality of semen
after seminal vesicle aspiration, these workers commonly obtained by assisted ejaculation from
estimated that up to half the sperm collected by men with chronic spinal cord injury is consistent
434

with a rare cause of infertility in able-bodied men with impaired cauda epididymal sperm storage
known as necrospermia (Mallidis et al., 2000). The accounting for the low sperm motility in the vasa
reproductive tract stasis in spinal cord injury and the rapid drop off in sperm count with re-
caused by the neuropathic anejaculation is similar peated ejaculation. The disturbed genital tract
to a functional obstruction. Recently a similar sit- motility results in accumulation and sludging of
uation has been found in men with adult polycystic sperm in the ampullae of the vasa and seminal
kidney disease and necrospermia (Fang and Baker, vesicles. This produces the characteristic result
2003). These men appear to have stasis of genital with the first electroejaculation: often discolored
tract contents in cysts of the ejaculatory ducts, semen with high sperm concentration but very low
prostate or seminal vesicles. This remarkable motility. Once this material is cleared, sperm with
association suggests that stasis from any cause: better motility can be obtained over the next 1 to 2
neuropathic, partial distal genital tract obstruction days by successive daily electroejaculations. The
or cystic dilatation will cause necrospermia fact that sperm cannot continue to be collected in
(Mallidis et al., 2000; Fang and Baker, 2003). the majority of cord-injured men beyond 3–4 days
suggests that reduced sperm production, not ap-
parent on routine testicular histology, may also be
Possible evolution of the sperm defects with spinal a common problem.
cord injury
Management of male infertility with spinal
While multiple factors could affect sperm produc-
cord injury
tion in chronic spinal cord injury and certain cord-
injured men may have spermatogenic failure,
The team approach
accessory sex organ inflammation, genital tract
obstruction or sperm autoimmunity as the pre-
While failure of ejaculation is the major contrib-
dominant cause, the majority of individuals do not
utor to the infertility of men with spinal cord in-
have such problems. Our studies suggest the fol-
jury, sperm quality is often impaired and
lowing possible sequence in the majority of men
intracytoplasmic sperm injection may be required.
with spinal cord injury (Lim et al., 1994; Mallidis
However, as some cord-injured men have normal
et al., 1994, 2000). In the acute phase, spermato-
semen, a thorough evaluation is required to pro-
genesis is likely to be suppressed because of an
vide cost-effective management. This has been
acute shut down of gonadotropin production as
achieved by close co-operation between the Victo-
response to critical illness including the stresses of
rian Spinal Cord Service at Austin Health and
anesthesia, surgery, nutritional deprivation and
Melbourne IVF Reproductive Services at the Roy-
drug administration. Once spinal shock abates
al Women’s Hospital. This partnership has also
sperm already in the epididymis can be recovered
facilitated clinical research. Similar team ap-
by electroejaculation. Subsequently sperm collec-
proaches have been reported from around the
tion depends on the resumption of spermatogen-
world (Linsenmeyer, 2000; Biering-Sorensen and
esis and this may take several months in some
Sonksen, 2001; Heruti et al., 2001; Shieh et al.,
men. Those men with spinal cord lesions that al-
2003).
low coordinated autonomic function may have
normal genital tract motility and thermoregulation
and good quality antegrade semen may be collect- Clinical evaluation
ed by either vibroejaculation or electroejaculation.
Rarely some men in this group may be able to The cord-injured men undergo andrologic assess-
ejaculate without assistance. Many cord-injured ment, including particularly a history of factors
men however have abnormal genital tract motility that might affect testicular function, physical
so they do not respond well to vibroejaculation. examination of secondary sex characteristics and
There is also defective scrotal thermoregulation thorough scrotal examination, serum levels of
435

follicle-stimulating hormone and antisperm anti- Both vibration ejaculation and electroejacula-
bodies and other hormonal assays and testicular tion can be performed with Foley catheter bladder
biopsy as indicated by signs of testicular failure. neck tamponade if retrograde ejaculation is
The female partner is evaluated by a infertility expected. Both procedures are likely to precipitate
specialist gynecologist, has pre-pregnancy screens autonomic dysreflexia in those who suffer lesions
for general health, rubella immunity, infections, above T6. Close monitoring of the blood pressure
genetic conditions as indicated and is advised during the procedure is mandatory. Prophylactic
about preventative strategies such as taking folate anti-hypertensive medication can be given. If
and avoiding smoking and excessive alcohol autonomic dysreflexia does develop cessation of
consumption. A plan of management is devised the procedure is usually adequate management.
depending on the success of assisted ejaculation With persistent blood pressure elevation, the rapid
trials and on the sperm quality (Table 1). administration of hypotensive agents sublingually
or, in rare instances, intravenously is effective.
Testicular aspiration is utilized when vibration and
electroejaculation procedures have failed.
Assisted ejaculation The semen collected is analyzed by standard
methods. Sperm may be cryopreserved if not im-
The management approach is from the least inva- mediately required for artificial insemination or
sive and costly intervention that may be successful intracytoplasmic sperm injection.
toward the more technologically complex and
costly (Table 1). In determining the method of
assisted ejaculation, vibration stimulus is prefer- Austin Health/Royal Women’s Hospital fertility
entially chosen for men who have an intact reflex program results
arc for ejaculation. Vibration, if successful, may
allow home insemination, particularly when auto- Acute collection of semen is not offered routinely.
nomic dysreflexia is not a risk. It is offered to cord-injured men who were actively
Electroejaculation is tried when vibration fails attempting to father children at the time of their
and in men without an intact reflex ejaculation arc. injury and to men for whom this was a major
Cord-injured men with sacral spinal segment immediate issue psychosocially. Near normal
preservation will be offered the procedure under semen was collected in 50% of cases in which acute
general anesthetic. acquisition was attempted (Mallidis et al., 1994).

Table 1. Clinical features of management in the chronic spinal cord injury patient

Clinical evaluation including


General condition stable, no recent febrile illness or urinary infection
Neurologic assessment level (upper or lower motor neuron), completeness
Andrologic assessment clinical examination, follicle stimulating hormone and sperm antibody assay, possibly testicular biopsy
Trial of assisted ejaculation vibroejaculation or electroejaculation, need for general anesthesia, autonomic dysreflexia risk
Female fertility evaluation prepregnancy checkup
Management plan artifical insemination or intracytoplasmic sperm injection
Infertility counselor information and psychological support
Management options
Home insemination if vibration ejaculation successful and safe
Repeated ejaculation for necrospermia (daily for 2–3 days) timed with ovulation for fresh intrauterine insemination
Semen cryopreservation for artificial insemination (concentration 420  106/ml, post thaw motility 420%) or intracytoplasmic sperm
injection
Fine-needle tissue aspiration testicular biopsy to obtain sperm for intracytoplasmic sperm injection if assisted ejaculation fails or
contraindicated.
Donor insemination if no sperm obtainable
436

In the chronic spinal cord injury group fifty- Halstead et al., 1987; Dahlberg et al., 1995; Brack-
eight men have entered the program over a 12-year ett, 1999; Pryor et al., 2001).
period. Thirty-eight men were or are engaged in ART including in vitro fertilization or intracy-
active treatment. Sixteen men have fathered twen- toplasmic sperm injection can be used if there is
ty live births. Three live births were achieved from coexisting female infertility, artificial insemination
sperm acquired by testicular aspiration, twelve fails after a reasonable number of attempts, if the
from sperm acquired by assisted ejaculation and semen quality is inadequate for artificial insemi-
six from home-based sperm acquisition and arti- nation or if the sperm are obtained from the gen-
ficial insemination. No live sperm was retrievable ital tract. Standard in vitro fertilization may be
in six men. The drop out rate was approximately possible if the semen is adequate. The results of
50% after one or two treatments in couples en- assisted reproductive technology seem no different
gaged in hospital-based assisted reproductive tech- from those of other patients although a lower
nology (ART) treatments. This is a similar figure pregnancy rate has been reported (Schatte et al.,
to the able-bodied engaged in assisted reproduc- 2000).
tive technology. Ten cord-injured men are pres-
ently engaged in active treatment with the partners
Summary
of two of them having pregnancies of 3 months
duration.
Sexual function
Although some groups have reported that re-
peated ejaculation does not improve semen quality
Treatment of sexual dysfunction in males with
with chronic spinal cord injury, this is not the
spinal cord injury is often focused on the erection
general experience (Brindley, 1983a; Brackett,
disorder. Modern approaches emphasize the need
1999; Sonksen et al., 1999; Mallidis et al., 2000;
for sexual health counseling where broader issues
Heruti et al., 2001). We find sperm motility can be
of intimacy, relationship and communication are
improved with frequent ejaculation (daily for up
addressed. Without attention to these issues the
to 2–3 days) in most cord-injured men with
success of erection-promoting agents may be com-
necrospermia (Mallidis et al., 2000). This has
promised. At this time all have a place clinically
clinical significance for vibroejaculation in the
but further comparative research may elucidate
home setting. The men are advised to have fre-
which groups of cord-injured men may be more
quent ejaculation (two or more times a week) and
suited to some agents than others. While the trend
intravaginal artificial insemination of the semen
is toward the evolution of further oral agents, es-
daily for 1–3 days around the time of ovulation
tablished options such as intracavernosal therapy,
determined by symptoms or detection of the lute-
vacuum tumescence pump, sacral anterior root
inizing hormone surge in urine. Also with electro-
electrical stimulation and even penile prostheses
ejaculation, if semen is not immediately suitable
continue to have a place in clinical care.
for artificial insemination, then repeating the pro-
cedure and collection of semen on the second or
third day may allow artificial insemination to be Male infertility
timed with ovulation or cryopreservation of semen
adequate for subsequent artificial insemination. An inability to ejaculate in the first few days after
Generally the cryopreserved semen is used to pre- acute spinal cord injury, before return of visceral
pare a motile sperm suspension by density gradient reflexes, is probably due to neural refractoriness
centrifugation for intrauterine insemination at the associated with spinal shock. Except in those with
time of ovulation (Bourne et al., 2004). Although systemic illness or multi-trauma, electroejaculation
there are many reports of low pregnancy rates with performed 6–12 days after injury will enable col-
artificial insemination of sperm obtained by lection of sperm of normal or near normal quality.
assisted ejaculation, the results of adequately Thereafter sperm quality deteriorates. The main
timed inseminations can be good (Brindley, 1984; reason for this is not clear and may be related to
437

elevated scrotal temperatures, infrequent ejacula- Basu, S., Aballa, T.C., Ferrell, S.M., Lynne, C.M. and Brack-
tion (functional obstruction) or other factors. The ett, N.L. (2004) Inflammatory cytokine concentrations are
fact that it occurs at approximately 2 weeks after elevated in seminal plasma of men with spinal cord injuries.
J. Androl., 25: 250–254.
injury suggests that there is a neurogenic factor Bedford, M.J. (1991) Effects of elevated temperature on the
and this would therefore appear to be related to loss epididymis and testis: experimental studies. In: Zorgniotti
of input from the brain stem. However the relevant A.W. (Ed.), Temperature and Environmental Effects on the
neurogenic factor is not understood at this stage. Testis. Plenum Press, New York, pp. 19–32.
We have shown that most men with chronic Bedford, M.J. (1994) The status and the state of the human
epididymis. Hum. Reprod., 11: 2187–2199.
spinal cord injury have adequate spermatogenesis Biering-Sorensen, F. and Sonksen, J. (2001) Sexual function in
but many have a defect of sperm motility that can spinal cord lesioned men. Spinal Cord, 39: 455–470.
be improved by frequent ejaculation. This is Blanco, R., Saenz De Tejada, I., Goldstein, I., Krane, R.J.,
similar to the epididymal necrospermia seen in Wotiz, H.H. and Cohen, R.A. (1988) Cholinergic neuro-
able-bodied men. We postulate that neuropathic transmission in human corpus cavernosum II Acetylcholine
synthesis. Am. J. Physiol., 254: H468–H472.
anejaculation behaves as a functional ejaculatory
Bors, E. and Comarr, A.E. (1960) Neurological disturbances of
duct obstruction and that the distal genital tract sexual function with special reference to 529 patients with
fills with degenerating sperm. Repeated ejaculation spinal cord injury. Urol. Surv., 10: 191–222.
clears these sperm allowing collection of sperm Bourne, H., Edgar, D.H. and Baker, H.W.G. (2004) Sperm
with better motility. Although spermatogenesis is preparation techniques. In: Gardner D.K., Weissman A.,
Howles C.M. and Shoham Z. (Eds.), Textbook of
not severely impaired on conventional histology, it
Assisted Reproductive Techniques Laboratory and Clinical
is possible that sperm production is abnormal as Perspectives (2nd ed.). Taylor and Francis, London,
suggested by animal studies, because we have gen- pp. 79–92.
erally been unable to continue to collect semen Brackett, N.L. (1999) Semen retrieval by penile vibratory stim-
daily beyond 2–4 days. Although not all studies ulation in men with spinal cord injury. Hum. Reprod.,
show significant relationships between scrotal tem- Update, 5: 216–222.
Brackett, N.L., Bloch, W.E. and Lynne, C.M. (1998) Predictors
perature and semen quality, we suspect elevated of necrospermia in men with spinal cord injury. J. Urol., 159:
temperature does affect spermatogenesis and par- 844–847.
ticularly epididymal sperm storage. Further re- Brackett, N.L., Davi, R.C., Padron, O.F. and Lynne, C.M.
search to examine in detail the testes and genital (1996) Seminal plasma of spinal cord injured men inhibits
tract of men with chronic spinal cord injury is sperm motility of normal men. J. Urol., 155: 1632–1635.
Brackett, N.L., Lynne, C.M., Aballa, T.C. and Ferrell, S.M.
needed to confirm our hypotheses. However, the (2000) Sperm motility from the vas deferens of spinal cord
findings that semen quality improves with repeated injured men is higher than from the ejaculate. J. Urol., 164:
ejaculation has obvious therapeutic implications as 712–715.
described above. We anticipate further clinical Brackett, N.L., Lynne, C.M., Weizman, M.S., Bloch, W.E. and
research will enhance our approach to male infer- Padron, O.F. (1994) Scrotal and oral temperatures are not
related to semen quality of serum gonadotropin levels in spi-
tility associated with spinal cord injury and also
nal cord-injured men. J Androl., 15: 614–619.
benefit the able-bodied male population with Brackett, N.L., Padron, O.F. and Lynne, C.M. (1997) Semen
infertility through better understanding of these quality of spinal cord injured men is better when obtained by
interesting acquired sperm defects. vibratory stimulation versus electroejaculation. J. Urol., 157:
151–157.
Brindley, G.S. (1981a) Reflex ejaculation under vibratory stim-
ulation in paraplegic men. Paraplegia, 19: 299–302.
References Brindley, G.S. (1981b) Electroejaculation: its technique, neu-
rological implication and uses. J. Neurol. Neurosurg. Psych.,
Aird, I.A., Vince, G.S., Bates, M.D., Hohnson, P.M. and 44: 9–18.
Lewis-Jones, I.D. (1999) Leukocytes in semen from men with Brindley, G.S. (1982) Deep scrotal temperature and the effect
spinal cord injuries. Fertil. Steril., 72: 97–103. on it of clothing, air temperature, activity, posture and par-
Baker, H.W.G. (2001) Testicular function in systemic disease. aplegia. Br. J. Urol., 54: 49–55.
In: Becker K.L. (Ed.), Principles and Practice of Endocrino- Brindley, G.S. (1983a) Physiology of erection and management
logy and Metabolism (3rd ed.), Chapter 116. Lippincott of paraplegic infertility. In: Hargreave T.B. (Ed.), Male In-
Williams and Wilkins, Philadelphia, PA, pp. 1150–1158. fertility. Springer, Berlin, pp. 261–279.
438

Brindley, G.S. (1983b) Cavernosal alpha-blockade: a new tech- Hirsch, I.H., Sedor, J., Callahan, H.J. and Staas, W.E. (1992)
nique for investigation and treating erectile impotence. Br. J. Antisperm antibodies in seminal plasma of spinal cord-in-
Psych., 143: 332–337. jured men. Urol., 39: 243–247.
Brindley, G.S. (1984) The fertility of men with spinal cord in- Hovatta, O. and von Smitten, K. (1993) Sperm aspiration from
juries. Paraplegia, 22: 337–348. vas deferens and in-vitro fertilization in cases of non-treat-
Brindley, G.S. (1988) The actions of parasympathetic and sym- able anejaculation. Hum. Reprod., 8: 1691–1698.
pathetic nerves in human micturition, erection and seminal Huang, H.F., Wang, S., Molina, C.A. and Ottenweller, J.E.
emission, and their restoration in parasympathetic patients (2004) Preservation of spermatogenesis in spinal cord injured
by implanted electrical stimulation. Ferrier Lecture 1986. rats with exogenous testosterone. Relationship with serum
Proc. R. Soc. Lond. [Biol.], 235: 111–120. testosterone levels and cellular localization of camp respon-
Brindley, G.S., Polkey, C.E. and Rushdon, D.N. (1982) Sacral sive element modulator. J. Androl., 25: 95–103.
anterior root stimulators for bladder control in paraplegia. Jarow, J.P. (1996) Seminal vesicle aspiration of fertile men.
J. Paraplegia, 20: 365–381. J. Urol., 156: 1005–1007.
Buch, J.P. (1994) Greatly improved sperm motility from vas Levin, R.M. and Wein, A.J. (1980) Adrenergic alpha-receptors
deferens sperm retrieval: a case for accessory gland related outnumber beta-receptors in human penile corpus caverno-
subfertility in spinal cord injured men. Case report. Paraple- sum. Invest. Urol., 18: 225–226.
gia, 32: 501–504. Lim, T.C., Mallidis, C., Hill, S.T., Skinner, D.J., Carter, P.D.,
Burns, A.S., Rivas, D.A. and Ditunno, J.F. (2001) The man- Brown, D.J. and Baker, H.W.G. (1994) A simple technique
agement of the neurogenic bladder and sexual dysfunction to prevent retrograde ejaculation during assisted ejaculation.
after spinal cord injury. Spine, 26(Suppl. 24): 129–136. Paraplegia, 32: 142–149.
Chapelle, P.A., Roby-Brami, A., Jondet, M., Piechaud, T. and Linsenmeyer, T.A. (2000) Sexual function and infertility fol-
Bussel, B. (1993) Trophic effects on testes in paraplegics. lowing spinal cord injury. Phys. Med. Rehabil. Clin. N. Am.,
Paraplegia, 31: 576–583. 11: 141–156.
Dahlberg, A., Ruutu, M. and Hovatta, O. (1995) Pregnancy Lopez, G.J. and Kollar, W.C. (2000) Sexual function, dysfunc-
results from a vibrator application, electroejaculation, and a tion, orientation and the autonomic nervous system. In:
vas aspiration programme in spinal-cord injured men. Hum. Appenzeller O. (Ed.), Handbook of Clinical Neurology, Vol.
Reprod., 10: 2305–2307. 75 (31). The Autonomic Nervous System, Part ii Dysfunc-
De Lamirande, E., Hassouna, M., Leduc, B.E., Gagnmon, C. tions, Chapter 3, Elsevier, Amsterdam, pp. 85–103.
and Iwasaki, A. (1995) Increased Reactive Oxygen Species Lynne, C.M., Aballa, T.C., Wang, T.J., Rittenhouse, H.G.,
Formation in Semen of Patients with Spinal Cord Injury. Ferrell, S.M. and Brackett, N.L. (1999) Serum and semen
Fertil. Steril., 53: 637–642. prostate specific antigen concentrations are different in
Del Popolo, G., Li Marzi, V., Mondaini, N. and Lombardi, G. young spinal cord injured men compared to normal con-
(2004) Time/duration effectiveness of sildenafil versus ta- trols. J. Urol., 162: 89–91.
dalafil in the treatment of erectile dysfunction in male spinal Mallidis, C., Lim, T.C., Hill, S.T., Skinner, D.J., Brown, D.J.,
cord-injured patients. Spinal Cord, 42: 643–648. Johnston, W.I.H. and Baker, H.W.G. (1994) Collection of
Denil, J., Ohl, D.A. and Smythe, C. (1996) Vacuum erection semen from men in acute phase of spinal cord injury. Lancet,
device in spinal cord injured men: patient and partner sat- 343: 1072–1073.
isfaction. Arch. Phys. Med. Rehabil., 77: 750–753. Mallidis, C., Lim, T.C., Hill, S.T., Skinner, D.J., Brown, D.J.,
Derry, F., Hultling, C., Seftel, A.D. and Sipski, M.L. (2002) Johnston, W.I.H. and Baker, H.W.G. (2000) Necrospermia
Efficacy and safety of sildenafil citrate (Viagra) in men with and chronic spinal cord injury. Fertil. Steril., 74: 221–227.
erectile dysfunction and spinal cord injury: a review. Urol- Marina, S., Marina, F., Alcolea, R., Nadal, J., Pons, M.C.,
ogy, 60: 49–57. Grossman, M., Esposito, R. and Vidal, J. (1999) Triplet
Elliott, S.P., Orejuela, F., Hirsch, I.H., Lipshultz, L.I., Lamb, pregnancy achieved through intracytoplasmic sperm injection
D.J. and Kim, E.D. (2000) Testis biopsy findings in the spinal with spermatozoa obtained by prostatic massage of a para-
cord injured patient. J. Urol., 163: 792–795. plegic patient. Hum. Reprod., 14: 1546–1548.
Fang, S. and Baker, H.W.G. (2003) Male infertility and adult Melman, A., Henry, D.P., Felten, D.L. and O’Connor, B.L.
polycystic kidney disease are associated with necrospermia. (1980) Alteration of the penile corpora in patients with erec-
Fertil. Steril., 79: 643–644. tile impotence. Invest. Urol., 17: 474–477.
Guttmann, L. (1973) Spinal Cord Injuries Comprehensive Monga, M., Dunn, K. and Rajasekaran, M. (2001) Character-
Management and Research. Blackwell, Oxford, pp. 446–477. ization of ultrastructural and metabolic abnormalities in se-
Halstead, L.S., VerVoort, S.M. and Seager, S.W. (1987) Rectal men from men with spinal cord injury. J. Spinal Cord Med.,
probe electrostimulation in the treatment of anejaculatory 24: 41–46.
spinal cord injured men. Paraplegia, 25: 120–129. Naderi, A.R. and Safarinejad, M.R. (2003) Endocrine profiles
Heruti, R.J., Katz, H., Menashe, Y., Weissenberg, R., Raviv, and semen quality in spinal cord injured men. Clin. End-
G., Madjar, I. and Ohry, A. (2001) Treatment of male in- ocrinol. (Oxf.),, 58: 177–184.
fertility due to spinal cord injury using rectal probe electro- Ohl, D.A., Denil, J., Fitzgerald-Shelton, K., McCabe, M.,
ejaculation: the Israeli experience. Spinal Cord, 39: 168–175. McGuire, E.J., Menge, A.C. and Randolph, J.F. (1992)
439

Fertility of spinal cord injured males: Effect of genitourinary Shirai, M., Sasaki, K. and Rikimaru, A. (1973) A histochemical
infection and bladder management on results of electroejac- investigation of the distribution of adrenergic and cholinergic
ulation. J. Am. Paraplegia Soc., 15: 5359. nerves in the human male genital organs. Tohoku J. Exp.
Ohl, D.A., Menge, A.C. and Jarow, J.P. (1999) Seminal vesicle Med., 111: 281–291.
aspiration in spinal cord injured men: Insight into poor Siosteen, A., Forssman, L., Steen, Y., Sullivan, L. and Wick-
sperm quality. J. Urol., 162: 2048–2051. strom, I. (1990) Quality of semen after repeated ejaculation
Ohl, D.A., Sonksen, J., Wedemeyer, G., Zaborniak, M.C., treatment in spinal cord injury men. Paraplegia, 28: 96–104.
Dam, T.N., Menge, A.C., Putzi, M.J. and Papadopoulos, Slot, O., Drewes, A., Andreasen, A. and Olsson, A. (1989)
S.M. (2001) Canine model of infertility after spinal cord in- Erectile and ejaculatory function of males with spinal cord
jury: Time course of acute changes in semen quality and injury. Int. Disabil. Stud., 11: 75–77.
spermatogenesis. J. Urol., 166: 1181–1184. Sonksen, J., Ohl, D.A., Giwercman, A., Biering-Sorensen, F.,
Pryor, J.L., Kuneck, P.H., Blatz, S.M., Thorp, C., Cornwell, Skakkebaek, N.E. and Kristensen, J.K. (1999) Effect of re-
C.E. and Carrell, D.T. (2001) Delayed timing of intrauterine peated ejaculation on semen quality in spinal cord injured
insemination results in a significantly improved pregnancy men. J. Urol., 161: 1163–1165.
rate in female partners of quadriplegic men. Fertil. Steril., 76: Talbot, H.S. (1955) The sexual function in paraplegia. J. Urol.,
1130–1135. 77: 157–168.
Rutkowski, S.B., Middleton, J.W., Truman, G., Hagen, D.L. Thomas, A.J. (1983) Ejaculatory dysfunction. Fertil. Steril., 39:
and Ryan, J.P. (1995) The influence of bladder management 445–454.
on fertility in spinal cord injured males. Paraplegia, 33: Trabulsi, E.J., Shupp-Byrne, D., Sedor, J. and Hirsch, I.H.
236–263. (2002) Leukocyte subtypes in electroejaculates of spinal cord
Schatte, E.C., Orejuela, F.J., Lipshultz, L.I., Kim, E.D. and injured men. Arch. Phys. Med. Rehabil., 83: 31–34.
Lamb, D.J. (2000) Treatment of infertility due to anejacu- Watkins, W., Lim, T., Bourne, H., Baker, H.W.G. and
lation in the male with electroejaculation and intracytoplas- Wutthiphan, B. (1996) Testicular aspiration of sperm for in-
mic sperm injection. J. Urol., 163: 1717–1720. tracytoplasmic sperm injection: an alternative treatment to
Scheutzow, M.H. and Bockenek, W.L. (2000) An unusual electro-emission: case report. Spinal Cord, 34: 696–698.
complication during electroejaculation in an individual with Wieder, J.A., Brackett, N.L., Lynne, C.M., Green, J.T. and
tetraplegia. J. Spinal Cord Med., 23: 28–30. Aballa, T.C. (2000) Anesthetic block of the dorsal penile
Shieh, J.Y., Chen, S.U., Wang, Y.H., Chang, H.C., Ho, H.N. nerve inhibits vibratory-induced ejaculation in men with spi-
and Yang, Y.S. (2003) A protocol of electroejaculation and nal cord injuries. Urol., 55: 915–917.
systematic assisted reproductive technology achieved high Wilton, L.J., Temple-Smith, P.D., Baker, H.W.G. and de
efficiency and efficacy for pregnancy for anejaculatory men Kretser, D.M. (1988) Human male infertility caused by de-
with spinal cord injury. Arch. Phys. Med. Rehabil., 84: generation and death of sperm in the epididymis. Fertil.
535–540. Steril., 49: 1052–1058.
L.C. Weaver and C. Polosa (Eds.)
Progress in Brain Research, Vol. 152
ISSN 0079-6123
Copyright r 2006 Elsevier B.V. All rights reserved

CHAPTER 30

Female sexual function after spinal cord injury

Marca L. Sipski1,2, and Adriana Arenas2

1
Veterans Administration Rehabilitation Research and Development, Center of Excellence in Functional Recovery and
Spinal Cord Injury, Miami, FL 33101, USA
2
Department of Rehabilitation Medicine, University of Miami School of Medicine, P.O. Box 016960 (D-461), Miami,
FL 33101, USA

Abstract: Over the past 10 years, studies of the impact of spinal cord injuries on female sexuality have
expanded from questionnaire studies in small populations with unknown levels and degrees of injury to
laboratory-based analyses of women with known injury patterns. These studies have provided detailed
information on how specific injury patterns affect specific aspects of the female sexual response. Research
findings have supported the hypothesis that the sympathetic nervous system is regulatory for psychogenic
genital vasocongestion and that orgasm is a reflex response of the autonomic nervous system. Based on
these results, a new system for the classification of sexual function in women with spinal cord injury (SCI) is
proposed. Moreover, studies related to the treatment of sexual dysfunction in women with cord injury are
reviewed.

Our understanding of the impact of spinal cord sexuality (Charlifue et al., 1992; Sipski and
injury (SCI) on the female sexual response and Alexander, 1993; Jackson and Wadley, 1999;
sexuality is probably greater than that of any other Fisher et al., 2002). These studies, although lack-
neurologic disorder. Evolving from a stage in ing appropriate controls, definition of the injury
which only questionnaire studies were available to and validation of the questionnaires, showed
address the effects of cord injury on female sex- that the frequency of sexual activity and sexual
uality, there is now a relatively large body of lit- satisfaction is diminished in women after SCI
erature that documents the impact of various types (Charlifue et al., 1992; Sipski and Alexander, 1993;
of cord injuries on the female sexual response. Fisher et al., 2002). Sexual activities engaged in by
Additionally, methods of treatment have begun to women after cord injury (Sipski and Alexander,
be explored. The goal of this chapter is to review 1993) are unaltered; however, a surprisingly low
the literature pertaining to the impact of SCI on percentage of women with cord injury masturbate.
the female sexual response, to discuss the issue of
diagnosis of sexual dysfunction in the population
Effect of SCI on female sexual arousal
of women with cord injury and to discuss potential
treatment methods currently being evaluated.
The effect of cord injury on female sexual arousal
has been evaluated via multiple laboratory-based
The impact of SCI on female sexuality trials (Sipski et al., 1995, 1996, 1997, 2001). Two
separate pathways control female genital sexual
Prior to the 1990s, a number of questionnaire arousal: a psychogenic pathway and a reflex path-
studies documented the effects of SCI on women’s way. Thus, the impact of SCI on sexual response
depends on which of these pathways is altered by
Corresponding author.; E-mail: m.sipski@worldnet.att.net the neurologic injury. Women with complete cord

DOI: 10.1016/S0079-6123(05)52030-2 441


442

injury at or above the level of the 6th thoracic increases in their level of genital arousal with
segment (T6) were evaluated in the laboratory to manual stimulation, regardless of whether they
see the effects of psychogenic, and psychogenic had concomitant increases in level of subjective
combined with manual genital stimulation on arousal (Sipski et al., 1997). This was interpreted
heart rate, respiratory rate, blood pressure, vagi- as evidence for the maintenance of reflex genital
nal pulse amplitude (Lann and Everaerd, 1998) arousal in women with upper motor neuron inju-
(as a measure of genital arousal) and subjective ries affecting their sacral spinal segments. Further
sexual arousal. A 78 min protocol was used that research compared the effect of the addition of
consisted of 6 min baseline periods alternating with manual to psychogenic stimulation in women with
two 12 min periods of audiovisual erotic stimula- upper versus lower motor neuron injuries affecting
tion and two 12 min periods of audiovisual erotic their sacral segments (Sipski et al., 2001). The
combined with manual genital stimulation. Women subjects with upper versus lower motor neuron
with complete upper motor neuron injuries affect- injury did not differ in vaginal pulse amplitude,
ing their sacral spinal cord had significant increas- but vaginal pulse amplitude tended to increase
es in subjective sexual arousal with audiovisual with upper motor neuron injuries, as would be
erotic stimulation alone, but no increase in vaginal expected in the presence of reflex genital vasocon-
pulse amplitude. Adding manual genital stimula- gestion. Despite the lack of significance in these
tion to the audiovisual stimulation resulted in results, the overall psychophysiologic data tend to
augmentation in vaginal pulse amplitude without validate the hypothesis that reflex lubrication is
continuation of the increased subjective sexual maintained in women with SCI and upper motor
arousal. These results were interpreted as a dem- neuron injuries affecting their sacral segments, and
onstration of the presence of reflex genital vaso- that women with cord injury and lower motor
congestion in women with complete SCI. neuron incomplete injuries should still have partial
The neurologic control of psychogenic arousal preservation of reflex lubrication. It follows that
was also studied in the laboratory in women with a the only subset of women who should not have
wide range of levels of SCI (Sipski et al., 2001). the potential for reflex lubrication should be
Using the identical 78 min research protocol de- those women with complete lower motor neuron
scribed above, the ability to achieve psychogenic injuries.
genital arousal was shown to be related to the In another laboratory-based study (Komisaruk
preservation of the combined ability to perceive et al., 1997) of the effects of SCI on sexual re-
pinprick and light touch sensation in the T11–L2 sponse, the authors hypothesized that vaginal and/
dermatomes (Sipski et al., 2001). Women with or cervical self-stimulation will not produce per-
combined scores of 24–32 in these dermatomes as ceptual responses in women with complete SCI (as
determined by the ASIA standards (Sipski et al., by the ASIA standards) at or above the highest
2001), were significantly more likely to demon- level of entry of the hypogastric nerves (T10–T12),
strate psychogenic genital vasocongestion than but will produce perceptual responses if the injury
women with scores of 9–23. In turn, women with is below T10. The authors studied 6 women with
scores of 9–23 were significantly more likely to complete SCI (T10 and/or above), 10 women with
achieve psychogenic genital vasocongestion than complete lower cord injury (below T10) and 5 un-
women with scores of 0–8. This information was injured women as a control group. Perceptual re-
interpreted to be evidence for a role of the sym- sponse to vaginal and/or cervical self-stimulation
pathetic preganglionic neurons (with cell bodies at was quantified as the magnitude of analgesia pro-
the T11–L2 level) in the control of psychogenic duced by a calibrated finger compressive force ap-
genital arousal. plied to the vagina or cervix for a period of 12 min
The control of reflex arousal was also further interspersed with non-stimulation control periods.
studied in the laboratory. Women with incomplete Significant analgesia was noted in all the groups
SCI who were first subjected to psychogenic, then including the lower cord injury group. The authors
to manual genital stimulation showed further interpreted the finding of analgesia in the group
443

with lower injury as support for their original hy- laboratory, 44% of cord-injured subjects were or-
pothesis. The unexpected finding of analgesia in gasmic compared to 100% of able-bodied con-
the group with injury above T10 was proposed as trols. The characteristics of orgasm in able-bodied
evidence for a genital afferent pathway that by- versus cord-injured subjects were also analyzed.
passes the spinal cord, such as the vagus nerve. Despite previous reports that non-genital stimula-
However, women with upper cord injury may also tion is often used as a means to achieve orgasm,
experience menstrual discomfort, awareness of only one woman in this study (Sipski et al., 2001)
vaginal and/or cervical stimulation and orgasms. chose non-genital stimulation in combination with
Thus, one must realize that the definition of injury genital stimulation. The average latency to orgasm
as ‘‘complete’’ is based solely on preservation of was significantly greater in cord-injured versus
voluntary rectal contraction and/or preservation able-bodied subjects (26 min versus 16 min). The
of anal sensation and does not provide any other heart rate, systolic blood pressure and respiratory
information with respect to remaining autonomic rate at orgasm compared to baseline were signif-
function. Moreover, the results of this study must icantly greater for both cord-injured and able-
be considered in view of the fact that the protocol bodied subjects; however, there were no significant
in which subjects were asked to participate re- differences between the two groups at any time
quired them to stimulate their cervix at a specific period. Diastolic blood pressure was similar in
intensity with a device constructed as a diaphragm both groups of subjects at orgasm and baseline;
with a handle on it. In addition, the subjects were additionally there was no significant increase in
not alone during this test, but had an investigator diastolic blood pressure at orgasm versus baseline.
in the room with them administering both tactile When two investigators were blinded to subjects’
and pain testing. Under these circumstances, it is descriptions of orgasms, they were unable to de-
possible that the alterations in sensory perception termine whether the women had complete or in-
found in these subjects were related to a distrac- complete SCI or were able-bodied control subjects.
tion effect of the study methodology. The women’s ability to achieve orgasms were
also compared between various groups of women
with spinal cord injuries. No statistically signifi-
cant differences were observed in orgasmic ability
Effects of SCI on orgasm based upon grouping of subjects according to re-
maining sensation at the T11–L2 or S2–S5 derma-
The ability to achieve orgasm in women with SCI tomes, completeness of injury, or upper or lower
has also been assessed via questionnaire studies motor neuron injury affecting their sacral cord
and in the laboratory (Sipski et al., 1995; Whipple segments. Subjects with complete lower motor
et al., 1996). In the largest laboratory-based series neuron injuries affecting S2–S5 ðn ¼ 6Þ were sig-
to date (Sipski et al., 2001), the ability of women nificantly less likely to report the historical ability
with traumatic SCI to achieve orgasm was assessed to achieve orgasm as compared to those with all
both historically and in the laboratory. Sixty-two other patterns and degrees of SCI (n ¼ 56; 17%
women with SCI and 21 able-bodied control sub- lower motor neuron complete, 59% all other cord
jects participated. They were set up with monitors injuries combined; w2 ¼ 3:91; p ¼ 0:048). In the
for heart rate, respiratory rate and blood pressure, laboratory, however, there were no significant dif-
brought into the laboratory, provided with an ferences in the ability of these women to achieve
erotic video and asked to stimulate themselves to orgasm (17% lower motor neuron complete; 46%
orgasm any way they would like. Women with all all other cord injuries combined; w2 ¼ 1:96;
levels and degrees of SCI were significantly less p ¼ 0:16). Based on the belief that the historical
likely than able-bodied control subjects to achieve reporting of orgasm is more accurate than the
orgasm. Fifty-five percent of women with SCI re- women’s performance in the laboratory, these re-
ported the ability to achieve orgasm compared to sults were taken as evidence that the occurrence of
100% of able-bodied control subjects. In the orgasm depends on the presence of an intact sacral
444

reflex arc. These authors hypothesized that orgasm the development of treatment methods to remedy
is a reflex response of the autonomic nervous sys- orgasmic dysfunction.
tem that can be either facilitated or inhibited by Another group of investigators also performed
cerebral input. laboratory studies of the effects of SCI on orgasm.
Recent research points to the fact that orgasm Sixteen women with complete cord injury at or
may be associated with a pattern generator in the below the level T6 were studied during self-stim-
spinal cord. The suggestion has been made that an ulation of the anterior vaginal wall and cervix with
ejaculation generator is present in the spinal cord a device constructed as a diaphragm with a handle
(Truitt and Coolen, 2002) of male rats. These re- on it, designed for laboratory studies (mentioned
searchers documented activation of a subset of above) (Whipple et al., 1996; Komisaruk et al.,
lumbar spinothalamic neurons after copulatory 1997). Nine-minute control periods alternated with
behavior in male but not in female rats (Truitt 12 min stimulation periods. The stimulation was at
et al., 2003). This finding in male rats is similar to a specific intensity that was monitored by the sub-
that of the urogenital reflex that is found in anest- jects. Three of the subjects with SCI reported or-
hetized animals with spinal cord transections gasms from cervical stimulation and one subject
above T9. The reflex consists of rhythmic firing also reported orgasm from vaginal stimulation.
of the hypogastric, pelvic and pudendal motor Moreover, two of the three subjects had multiple
nerves in response to self-stimulation (McKenna orgasms during the experimental sessions. These
et al., 1991; Chung et al., 1988). The peripheral authors hypothesized that the neural pathway ac-
activity displayed during the urogenital reflex counting for these subjects’ ability to achieve or-
strongly resembles that seen during human or- gasm was the vagus nerve, citing laboratory
gasm (Bohlen et al., 1982). Vaginal, uterine and studies of animals as the reason for this hypoth-
anal sphincter rhythmic contractions are present in esis (Ortega-Villalobos et al., 1990; Cueva-Rolon
both the urogenital reflex and orgasm, and both et al., 1996; Komisaruk et al., 1996; Komisaruk
are relatively insensitive to gonadal hormones. et al., 2004;). In one of these animal studies, an-
Thus, a pattern of neural activity similar to the algesia and pupil dilatation in response to genital
urogenital reflex may underlie the response ob- stimulation of rats persisted after genital deaff-
served in women with SCI. erentation by bilateral pelvic, hypogastric and pu-
The authors hypothesized that if the neurologic dendal neurectomy (Cueva-Rolon et al., 1996).
potential to achieve orgasm exists in approximate- These two residual responses were abolished after
ly 50% of women with all levels of SCI (except bilateral vagotomy; thus, the authors concluded
those with complete lower motor neuron injury of that the pelvic, hypogastric and pudendal nerves
S2–S5), then those women who did not achieve are not the only vaginocervical afferent pathways
orgasm must have some intervening variable pre- and that the vagus pathway could remain intact
cluding them from achieving orgasm. Lack of ed- and functional after SCI. The authors further sur-
ucation and interfering psychologic issues were mised that this pathway could account, at least in
mentioned as possible problems in addition to the part, for the reports of perceptual responses to
overall negative viewpoint in the medical literature vaginal or cervical self-stimulation in women with
that has previously existed (Money, 1960; Fitting complete SCI.
et al., 1978). Other possible contributing factors This last hypothesis was recently tested in a pilot
include medications that are commonly taken by study by Komisaruk (Komisaruk et al., 2004) us-
women with SCI such as antidepressant and anti- ing functional magnetic resonance imaging to as-
spasticity medications that could diminish sexual certain whether the region of the brainstem to
responsiveness. Based upon research findings, the which the sensory component of the vagus nerves
authors recommended that women with SCI project, the nucleus tracti solitarii in the medulla
should be educated that longer and potentially oblongata, is activated by vaginal/cervical self-
more intense genital stimulation is necessary to stimulation in women with complete SCI at or
achieve orgasm. Furthermore, they recommended above T10. They found an overall increase in
445

activation at orgasm of multiple brain regions in- presence or absence of sexual dysfunction in wom-
cluding the nucleus tractus solitarius and the en with SCI.
hypothalamus. While this observation is sugges-
tive, it does not constitute proof. Proof would re-
quire the reversible loss of activation of the Improving sexual responsiveness
nucleus tractus solitarius by reversible block of
the vagus nerve. A number of studies have begun to test therapies
to improve sexual responsiveness in women with
SCI. The majority of these studies have used treat-
Documentation of sexual dysfunction in women ments previously used in able-bodied women, ex-
with SCI cept one drug study that tested the efficacy of
medications utilized in men (Sipski et al., 2000a).
The above documentation of the impact of SCI on The first series of therapies can be described as
sexual response provides a framework for under- cognitive. False positive feedback was used in a
standing how the injury affects sexual response. laboratory-based study (Sipski et al., 2000b) to
But it does not give us any information about increase the level of sexual arousal in a sample of
whether a woman with a cord injury has sexual women with SCI. It was unknown whether or not
dysfunction. According to the International Con- these women complained of sexual dysfunction.
sensus Development Conference on Female Sexual False positive feedback was shown to increase
Dysfunction (Basson et al., 2000), sexual dysfunc- psychogenic arousal in women with both complete
tion implies personal distress. Therefore a woman or incomplete SCI; however, genital arousal was
with a cord injury who has alterations in her sexual only increased in women with incomplete injuries
response related to her injury, but does not com- who had preservation of sensory function in the
plain of distress, does not have sexual dysfunction. T11–L2 dermatomes. This study concluded that
Conversely, a woman with a SCI who has no in- cognitively based therapies might be useful to im-
jury-related alterations in her sexual response but prove function in this subset of women with SCI.
complains of sexual distress, suffers from sexual In another study, the same authors studied the
dysfunction. In order to remedy this lack of a impact of an anxiety-provoking video on sexual
means of documentation, the Female Spinal Sex- arousal (Sipski et al., 2004). Subjects viewed two
ual Function Classification (FSSFC) was proposed erotic videos, one of which was preceded by a
(Sipski et al., 2002). This classification system re- neutral video and another that was preceded by an
lies on previous research to define four categories anxiety-provoking video. In subjects with impaired
of sexual function after SCI, document their pres- genital responsiveness to psychogenic erotic stim-
ence and associated characteristics and determine ulation (T11–L2 combined ASIA scores less than
which aspects of the neurologic examination 23), anxiety pre-exposure resulted in a small in-
should be used to determine the likely capacity crease in genital responsiveness to erotic stimula-
for sexual response. Based upon the performance tion compared to neutral pre-exposure. In subjects
of the neurologic examination and detailed histo- who had mostly intact genital responsiveness (cord
ry, one should be able to document the expected injury subjects with T11–L2 combined ASIA
effects of the injury on specific components of scores 24–32) and able-bodied subjects, anxiety
sexual response and also document whether the pre-exposure resulted in decreased genital respon-
subject reports any sexual dysfunction. This latter siveness compared to neutral pre-exposure. On the
issue becomes especially important when the issue assumption that anxiety is associated with sympa-
of clinical trials for remedying sexual dysfunction thetic activation, the authors concluded that there
after SCI is addressed. The FSSFC is currently is a therapeutic benefit in manipulating the sym-
being utilized in a study of women with SCI and pathetic nervous system in cord-injured subjects
multiple sclerosis, to assess its utility for docu- with impaired, but not absent, ability to achieve
mentation of the remaining sexual function and psychogenic genital vasocongestion.
446

Only one study has been published on the effects could be useful to treat sexual dysfunction in
of medications on sexual responsiveness after SCI women with SCI.
in females. In a laboratory-based, double-blind Compared to other neurologic injuries, knowl-
crossover design study (Sipski et al., 2000a) the edge of the impact of SCI on the female sexual
effects of sildenafil 50 mg versus placebo were response is relatively advanced. A study of women
compared on vaginal pulse amplitude, subjective with predominantly spinal multiple sclerosis has
arousal and autonomic function. Participants un- recently been initiated to determine if the impact
derwent a 78 min protocol, in which they were of spinal multiple sclerosis lesions on sexual re-
subject to audiovisual erotic stimulation and au- sponse in women will be similar to that of trau-
diovisual erotic combined with self-applied man- matic SCI. It is hoped that knowledge about the
ual stimulation alternating with baseline periods. impact of SCI on the female sexual response can
A statistically significant increase in subjective serve as a model to study not only multiple scle-
arousal was noted with the use of the medication. rosis but also other neurologic disabilities.
A borderline significant effect of drug administra-
tion was noted on vaginal pulse amplitude. In both
Acknowledgments
treatment conditions, sildenafil and placebo, both
visual and visual plus manual stimulation resulted
This work was supported in part by funds from
in small increases in blood pressure (3–5 mmHg)
NIH R01 HD 30149
above baseline. Regardless of the type of stimula-
tion, however, the mean blood pressure of subjects
on sildenafil was 4–5 mmHg lower compared to References
placebo. Similarly, regardless of the type of stim-
ulation, there was a slight increase in baseline Basson, R., Berman, J., Burnett, A., Derogatis, L., Ferguson,
heart rate (4–5 beats/min) with sildenafil compared D., Fourcroy, J., Goldstein, I., Graziottin, A., Heiman, J.,
to placebo. Laan, E., Leiblum, S., Padma-Nathan, H., Rosen, R., Se-
graves, K., Segraves, R.T., Shabsigh, R., Sipski, M., Wagner,
Although the above studies begin to outline G. and Whipple, B. (2000) Report of the international con-
possible methods of treatment for sexual arousal sensus development conference on female sexual dysfunction:
dysfunction for women with SCI, data are still definitions and classification. J. Urol., 163: 888–893.
preliminary and have yet to be translated into Billups, K.L., Berman, L., Berman, J., Metz, M.E., Glennon,
clinical utility. Other studies currently in progress M.E. and Goldstein, I. (2001) A new non-pharmacological
vacuum therapy for female sexual dysfunction. J. Sex Marital
are examining the effect of the use of medications Ther., 27: 1435–1441.
to improve sexual response. As new medications, Bohlen, J.G., Held, J.P., Sanderson, M.O. and Anderson, M.O.
such as the testosterone patch, become available, (1982) The female orgasm: pelvic contractions. Arch. Sex
their effects on sexual desire and arousal dysfunc- Behav., 11: 367–386.
Charlifue, S.W., Gerhart, K.A., Menter, R.R., Whiteneck,
tion after SCI will be assessed.
G.G. and Manley, M.S. (1992) Sexual issues of women with
No controlled clinical trials have been designed spinal cord injuries. Paraplegia, 30: 192–199.
to treat orgasmic dysfunction after SCI. Based Chung, S.K., Mc Vary, K.T. and Mc Kenna, K.E. (1988) Sex-
upon the assumption that orgasm is a reflex re- ual reflexes in male and female rats. Neurosci. Lett., 94:
sponse of the autonomic nervous system and that 343–348.
a spinal pattern generator exists, it may be possible Cueva-Rolon, R., Sane, G., Bianca, R., Gomez, L., Beyer, C.,
Whipple, B. and Komisaruk, B.R. (1996) Vagotomy blocks
to train the pattern generator. This concept is responses to Vaginocervical stimulation after genitospinal
similar to work being done with ambulation train- neurectomy in rats. Physio. Behav., 60: 19–24.
ing (Wernig et al., 1995, 1998) and is the founda- Fisher, T.L., Laud, P.W., Byfield, M.G., Brown, T.T., Hayat,
tion for a study to test the efficacy of EROS M.J. and Fiedler, I.G. (2002) Sexual health after spinal cord
injury: a longitudinal study. Arch. Phys. Med. Rehabil., 83:
(clitorical vacuum stimulation procedure) therapy
1043–1051.
(Billups et al., 2001) versus vibratory stimulation Fitting, M.D., Salisbury, S., Davies, N.H. and Mayclin, D.K.
that is currently underway. Both of these therapies (1978) Self-concept and sexuality of spinal injured women.
are designed to stimulate a reflex response and Arch. Sex Behav., 7: 143–156.
447

Jackson, A.B. and Wadley, V. (1999) A multicenter study of Sipski, M.L., Alexander, C.J. and Rosen, R.C. (1995) Physio-
women’s self-reported reproductive health after spinal cord logic parameters associated with psychogenic sexual arousal
injury. Arch. Phys. Med. Rehabil., 80: 1420–1428. in women with complete spinal cord injuries. Arch. Phys.
Komisaruk, B.R., Bianca, R., Sansone, G., Gomez, L.E., Med. Rehabil., 76: 811–818.
Cueva-Rolon, R., Beyer, C. and Whipple, V. (1996) Brain- Sipski, M.L., Alexander, C.J. and Rosen, R.C. (1996) Physio-
mediated responses to vaginocervical stimulation in spinal logic parameters associated with the performance of a dis-
cord-transacted rats: role of the vagus nerves. Brain Res., tracting task and genital self-stimulation in women with
708: 128–134. complete spinal cord injuries. Arch. Phys. Med. Rehabil., 77:
Komisaruk, B.R., Gerdes, C.A. and Whipple, B. (1997) ‘‘Com- 419–424.
plete’’ spinal cord injury does not block percepetual respons- Sipski, M.L., Alexander, C.J. and Rosen, R.C. (1997) Physio-
es to genital self-stimulation in women. Arch. Neurol., 544: logic parameters associated with psychogenic sexual arousal
1513–1520. in women with incomplete spinal cord injuries. Arch. Phys.
Komisaruk, B.R., Whipple, B., Crawford, A., Liu, W.C., Med. Rehabil., 78: 305–313.
Kalnin, A. and Mosier, K. (2004) Brain activation during Sipski, M.L., Alexander, C.J. and Rosen, R.C. (2001) The ne-
Vaginocervical self-stimulation and orgasm in women with urologic basis of sexual arousal and orgasm in women: effects
complete spinal cord injury: FMRI evidence of mediation by of spinal cord injury. Ann. Neurol., 49: 35–44.
the vagus nerves. Brain Res., 1024: 1513–1520. Sipski, M.L., Rosen, R.C., Alexander, C.J. and Hamer, R.M.
Lann, E. and Everaerd, W. (1998) Physiological measures of (2000a) Sildenafil effects on sexual and cardiovascular re-
vaginal vasocongestion. Int. J. Impot. Res., 10: 107–110. sponses in women with spinal cord injury. Urology, 55:
McKenna, K.E., Chung, S.K. and McVary, K.T. (1991) 812–815.
A model for the study of sexual function in anesthetized Sipski, M.L., Rosen, R., Alexander, C.J. and Hamer, R.
male and female rats. Am. J. Physiol., 30: R1276–R1285. (2000b) A controlled trial of positive feedback to increase
Money, J. (1960) Orgasm in the dreams of paraplegic men and sexual arousal in women with spinal cord injuries. Neuro.
women. Arch. Gen. Psychiatry., 3: 373–382. Rehabil., 15: 145–153.
Ortega-Villalobos, M., Garcia-Bazan, M., Solano-Flores, L., Truitt, W.A. and Coolen, L.M. (2002) Identification of a po-
Ninomiya-Alarcon, J., Guvara-Guzman, R. and Wayne, M. tential ejaculation generator in the spinal cord. Science,
(1990) Vagus nerve afferent and efferent innervation of the 297(5586): 1566–1569.
rat uterus: an electrophysiological and HRP study. Brain Truitt, W.A., Shipley, M.T., Veening, J.G. and Coolen, L.M.
Res. Bull., 25: 365–371. (2003) Activation of a subset of lumbar spinothalamic neu-
Sipsk, M.L., Rosen, R.C., Alexander, C.J. and Gomez-Marin, rons after copulator behavior in male but not female rats.
O. (2004) Sexual responsiveness in women with spinal cord J. Neurosci., 23: 325–331.
injuries: differential effects of anxiety-eliciting stimulation. Wernig, A., Muller, S., Nanassy, A. and Cagol, E. (1995) La-
Arch. Sex Behav., 33(3): 295–302. ufband therapy based on ‘‘Rules of spinal locomotion’’ is
Sipski, M.L. and Alexander, C.J. (1993) Sexual activities, re- effective in spinal cord injured persons. Eur. J. Neurosci., 7:
sponse and satisfaction in women pre- and post-spinal cord 823–829.
injury. Arch. Phys. Med. Rehabil., 74: 1025–1029. Wernig, A., Nanassy, A. and Muller, S. (1998) Maintenance of
Sipski, M.L. and Alexander, C.J. (2002) Documentation of the locomotor abilities following Laufband (treadmill) therapy in
impact of spinal cord injury on female sexual function: the para- and tetraplegic persons: follow-up studies. Spinal Cord,
female spinal sexual function classification. Top Spinal Cord 36: 744–749.
Inj. Rehabil., 8(1): 63–73. Whipple, B., Gerdes, C.A. and Komisaruk, B.R. (1996) Sexual
Sipski, M.L., Alexander, C.J. and Rosen, R.C. (1995) Orgasm response to self-stimulation in women with complete spinal
in women with spinal cord injuries: a laboratory-based as- cord injury. J. Sex Res., 33: 231–240.
sessment. Arch. Phys. Med. Rehabil., 76: 1097–1102.
Subject Index

adrenergic 52–54, 61–63, 65 bladder detrusor 118, 121, 126, 139, 195, 206–207
adrenergic antagonists 429 bladder dyssynergia 147, 327
adrenoceptor 62, 238–239, 241, 289, 292–296, 350, 352 bladder guarding reflex 213
afferent 53, 249, 266 bladder hyperactivity 158
afferent arbor 249, 251–252, 255, 259, 300, 305–306, 309 bladder hyperreactivity 143
afferent neurons 59, 63, 75–76, 100–101, 361, 363, 404, bladder hyperreflexia 97, 107, 158, 205
409 bladder hyporeflexia 136
anal canal 207–209, 321, 345–346, 351–352, 360, 375, bladder incontinence 155, 184
380–381 bladder inflammation 142
anatomy 27–28, 52, 60, 65, 164, 317, 346, 360 bladder interneurons 154
anejaculation 433–434, 437 bladder preganglionic 182, 363
anorectal 326, 336, 338–339, 341–342, 347, 365, 373–376, bladder primary afferents 147, 149–150, 155, 158
378–380 bladder reflex 74, 77, 97, 99–101, 107, 121, 126,
anorectum 346–347, 349, 355 200, 213
anti-spasticity 444 bladder urothelium 137, 143
anticholinergic 53, 164, 171, 178, 327 bladder–external urethral sphincter 119, 124–126, 130,
areflexia 73, 119, 341 132
areflexic rectum 339 bladder–sphincter 97, 99
areflexive bladder 164 bladder–sphincter dyssynergia 85, 97, 107, 130, 154, 422
arousal 388–392, 394, 396, 422, 441–442, 445–446 bladder, urethra 69
arrhythmias 256, 275–285, 329, 341, 397 blood pressure 11, 41–42, 45, 172, 223–227, 231–241,
arterial pressure 39–40, 42, 226, 231, 245–246, 269–270, 246, 267, 275, 278–281, 289–290, 299–301, 304–308,
275, 278, 281, 283–284, 289, 299 392, 396, 435, 443, 446
autonomic 42, 73, 237, 241, 265–266, 272, 281–282, 292, bowel 19, 53, 172, 188, 195, 200–201, 205–211, 213, 215,
296, 392, 396 217, 225–226, 234, 241, 246, 265–266, 317–318,
autonomic dysreflexia 11, 13, 27, 35, 39–42, 74, 76, 320–321, 323–330, 335–337, 339–342, 345, 354–355,
147–148, 166, 173, 178, 223–226, 233, 235, 237–241, 362, 369, 375, 378, 382, 387–392, 396
245–249, 251–252, 255–256, 259, 265–266, 268–272, bowel dysfunction 315, 317, 335, 340, 342, 345
275, 278, 280–281, 289–290, 299–302, 304–305, bowel guarding reflex 213
307–310, 328, 336, 341, 354, 369, 387, 390–391, bowel incontinence 391, see also fecal incontinence
393, 395–397, 408, 421, 423, 432, 435 bulbospongiosus 359, 361–363, 365–368, 415–417, 420

barrier function 54, 137, 140 cardiovascular 11, 17, 27, 39–45, 59, 104, 221, 223–225,
bladder 51–56, 59–65, 68–78, 85–86, 88–90, 92, 97–104, 227–228, 231, 235, 238, 245, 275–276, 279–281, 285,
106–109, 117–124, 126–127, 130–132, 135–137, 328, 387, 390–392, 396
139–143, 147–156, 158, 163–188, 195, 200–201, cell replacement 201–202
205–211, 213, 215, 217, 225–226, 236, 238–239, cholinergic 11, 17, 53–54, 60, 62, 65, 78, 92, 107, 211,
241, 246, 251, 259, 265–267, 290, 293, 317, 325, 271, 291, 320–321, 323, 337, 345, 351, 429
328–330, 336–337, 341–342, 345, 354–355, 359–362, colon 206, 208, 210, 246–247, 251, 256, 259, 267, 281,
367, 387–390, 392, 402, 406, 420–422, 430–431, 433, 290, 300–301, 304–305, 308, 318, 320–323, 325–329,
435 337, 341, 346–350, 354, 360, 363–364, 379, 406, 408
bladder afferent neurons 59, 63–64, 75–76, 103, 108, 111, colonic 266, 268, 271, 318, 320–329, 337, 339–342, 346,
155, 337 349–350, 354–355
bladder cholinergic 143 colonic transit times 345, 354, see also bowel transit
bladder control 51, 119, 165, 176, 184, 186–188 times

449
450

constipation 210, 318, 321, 323–329, 336–337, 339–342, fecal incontinence 205, 318, 325–326, 328–329, 335,
345, 354–355, 360, 374–375 338–342, 345–346, 355, 376, see also bowel
incontinence
defecation 174, 177, 206, 209, 234, 318, 322–323, female infertility 436
325–326, 329, 335, 339–341, 345–347, 349–350, female sexual function 392, 441
352–356, 360, 363–364, 366–367, 369, 373–375, flaccid bladder 52, 166
377, 416, 418, 422 functional magnetic resonance imaging 373, 381, 444
detrusor 51–54, 60, 65, 73–74, 76, 78, 98, 107, 119, 122, gallbladder 325
126–128, 135, 139, 143, 148, 155, 164, 168, 170–176, ganglion 20, 31, 33, 60–61, 63, 75, 102, 104, 106, 108,
178–179, 184–188, 206, 208, 210–211, 213, 360, 112, 150, 152, 271, 277, 289–292, 323, 351, 361, 393,
367 402–404
detrusor areflexia 74, 341 ganglionectomy 402
detrusor hyperactivity 52, 142 ganglionic 137, 164, 207, 235, 239, 241, 248, 272, 278,
detrusor hyperreflexia 53, 73–74, 76–78, 135, 164, 166, 290–292, 323, 348, 361, 429
170–171, 176, 211, 213, 215, 367 gastric emptying 324, 328, 373–374
detrusor–external urethral sphincter 119, 123, 125, 127, gastrointestinal 102, 104, 235, 239, 246, 299, 317–320,
130 323–325, 327, 330, 335, 340, 345, 348–349, 354–355,
detrusor–sphincter 53, 73, 74, 76 360, 366, 373–374, 379–380, 382
detrusor–sphincter dyssynergia 74, 76, 112, 155, 148, gastrointestinal transit times 354–355, see also colonic
164, 166–167, 170, 178–179, 185, 188, 354, 359, transit times
367 gene therapy 369
dorsolateral funiculus 12, 45, 67, 91, 104, 149, 199 genitalia 369, 388–389, 401, 406–407
dysreflexia 13, 39, 42, 45, 73, 237, 241, 265–266, 272,
glutamic acid 13
281–282, 292, 296, 301, 392, 396, 432
guarding reflex 205, 208–210, 211, 213, 215
dyssynergia 52–54, 73–74, 76, 85, 90, 92, 99, 360,
367
human spinal cord 45, 373, 389, 397, 415
dyssynergy 52, 54
humoral 323
hyperactivity 51, 53, 148
ejaculation 239, 387, 391–392, 394–397, 402,
404–405, 407–409, 415–423, 427–428, hyperreactivity 289, 294–296, 367
430–437, 444 hyperreflexia 11, 53, 59, 76, 97, 103, 112, 135, 164, 173,
ejaculation vibroejaculation 435 210–211, 213, 238, 359, 366–367
electrical stimulation 90, 119, 163, 168, 170–171, hyperreflexic bladder detrusor 119
173–174, 176–178, 181–183, 185–188, 213, 277, hyperreflexive bladder 74, 171–173, 176, 178, 180, 183,
284, 295, 365, 378, 418, 429, 431, 436 186
electroejaculation 391, 409, 422–423, 427–428, 431–436 hypertension 11, 39, 42, 54, 73, 223, 224–225, 231–232,
electromicturition 341 235, 237–239, 241, 245–246, 265–271, 275, 278,
electrophysiology 36, 275, 278, 280, 282, 284–285, 379 280–281, 290, 299–300, 328, 390, 396
enkephalin 17, 21, 72, 100, 111 hypogastric 52, 60–62, 97, 207–208, 241, 267, 270, 321,
enteric 206, 208, 317–318, 320, 323, 327, 330, 337, 345, 347, 360–361, 366, 401–403, 405, 416, 429, 442, 444
347, 349–350, 353, 363, 373 hyporeflexive bladders 167
enteric reflexes 322–323, 339 hypotension 223
erection 12, 174, 177, 235, 366–369, 387, 389–390,
392–396, 402, 404, 408, 416–417, 420–423, immunocytochemistry 252
427–430, 436 implantation 154–155, 166, 170, 173–178, 181, 183, 185,
eureflexic 300 187, 199–200
external sphincter 52 incontinence 11, 51, 53, 55, 117, 130, 143, 164, 166,
external urethral sphincter 61–62, 67–68, 71, 77, 85, 88, 171–172, 175, 177, 184–185, 205, 209, 211, 325–329,
99–100, 117–123, 125–128, 130, 132, 148, 155, 164, 336, 339, 341, 345, 374–376, 390
169, 174, 176–177, 182, 184, 362, 367, 416 inflammation 103, 107, 111, 135, 137, 140, 152, 155, 246,
external urethral sphincter motoneurons 85, 87 254–255, 259, 299, 306, 375, 432, 434
451

intermediolateral cell column 12–14, 17, 20–21, 40, neurogenic bladder 51, 54–55, 72, 163, 165, 168,
258–259, 267, 269, 272, 365–366, 416 170–171, 175, 211, 390
interneuron 11–12, 17, 21–22, 28–34, 77, 86–87, 89–92, neurogenic detrusor 73, 164, 172
101, 118, 148–149, 153–154, 181–182, 209, 223, 249, neurons 249, 265–266, 299, 402
266, 268–271, 305, 363, 365, 404–405, 417–418, 423 neuropeptide 13, 15, 17, 21, 63, 72, 100, 111, 143, 237,
interstitial cells 322, 348–350 296, 320, 337
intraurethral 60, 163, 167, 169, 179–180, 182, 185 neurophysiology 27–30, 34, 36, 168, 205–207, 209,
215–217, 378, 389, 395, 397
lateral funiculus 34, 39–40, 42–43, 45, 67, 69, 415, 419 neuroprosthesis 171–172, 176, 183, 186, 188
lower urinary tract 51, 59–60, 62, 65–67, 69, 72, 74, 78, neuroprosthetic 174, 187
90, 92, 97–99, 101–103, 107–108, 111, 112, 117–122, neuroprotection 195, 199, 202, 308–309
125, 126–127, 130, 142–143, 155, 163–165, 168–169, neurotrophic factors 76, 108, 111, 112, 150, 152, 155,
175, 184–186, 188, 195, 200–202, 206–207, 211, 232 157, 200, 259, 337
lumbosacral 59–60, 63, 73, 97–101, 103–104, 107, 109, neurotrophin 108, 136, 152, 154, 156, 199
117, 126, 128, 147, 152–154, 158, 195–198, 200, 202, neurovascular 295–296
205, 251, 265–271, 338–339, 354, 359, 361, 365–367, neurovascular transmission 292
369, 402–404, 407, 415, 417–418, 420, 423 non-cholinergic 60, 429
lumbosacral preganglionic 103 noradrenaline 14, 19, 87–88, 232–233, 235–238, 247,
lumbosacral propriospinal 268–269, 271 292–295, 353, 429
lumbosacral propriospinal projections 265 noradrenergic 69
lumbosacral sympathetic 207
lumbosacral ventral root avulsion 198 orgasm 388–389, 391–392, 394–396, 408, 422, 427–428,
430, 441, 443–446
orgasm/ejaculation 390
male infertility 428, 434, 436–437
orgasmic 388–389, 392, 443–444, 446
male sexual function 369, 389, 393, 415
orthostatic hypotension 223, 225–227, 231–236
mechanical devices 163, 165, 168, 185, 329
medulla 266
parasympathetic 52, 60–62, 64–65, 68–70, 76, 78,
megacolon 327
97–102, 104–105, 108, 118, 125–128, 142, 148, 151,
mesenteric 318, 321, 323, 347
154, 164, 173, 177–183, 197, 200–201, 206–208,
mesenteric ganglion 321 210, 215, 226, 231–232, 268, 277–280, 282, 284,
microstimulation 181–182, 187, 418–420 320, 323–324, 326, 329, 336, 340, 347, 351,
microstimulation, urethral 163 353–354, 359, 361–366, 369, 382, 389, 393–394,
micturition 59–60, 62, 65, 69–74, 76, 85–86, 88–92, 401, 404, 416, 429
97–100, 103, 111, 117–118, 122, 124, 128, 130–131, parasympathetic defecation reflex 363, 365
137, 147–149, 152–156, 158, 164, 168–170, 172, 174, parasympathetic ganglia 62
176–178, 181–183, 195, 200, 206–208, 213, 234, 359, parasympathetic micturition 70
365, 367–368, 416, 420, 422 parasympathetic postganglionic 78, 101
micturition reflex 52, 59, 70, 72, 74–77, 90, 97–100, 103, parasympathetic preganglionic 21–22, 66–67, 69–70, 77,
107–108, 111–112, 119, 130, 147–149, 152–153, 154, 101, 148–149, 173–174, 180, 184, 198, 369, 416, 429
155, 158 parasympathetic reflex 65
motoneurons 28, 31–32, 65, 67, 70, 73, 77, 85–92, 99, paroxysmal hypertension 231, 235, 241, 266
101, 112, 126, 128–130, 164, 170, 177, 182, 196–201, pelvic 22, 52, 59–63, 69–70, 73, 86, 97–98, 101–102, 140,
208–209, 393, 405, 416, 420–421, 429 148, 154–155, 163–164, 168–169, 172–173, 177, 181,
motoneurons, sympathetic preganglionic neurons 27 184–185, 197, 200–201, 205–211, 215–217, 252, 265,
mouse genetics 299, 308 267, 318, 320, 323, 326, 336, 338–340, 347, 360–362,
myenteric 101–102, 320–321, 326, 328, 337, 341, 364–367, 374–377, 379, 388, 393, 401–408, 416,
347–348, 351 418–423, 429, 444
pelvic floor 52, 169, 170–171, 355, 359, 363, 367, 393
necrospermia 427–428, 432, 434–437 pelvic ganglion 21–22, 196, 198, 361
nerve growth factor 76, 108–112, 140, 150–152, 155–156, pelvic reflex 213, 323
158, 199, 265–271, 290 pelvic somato-visceral reflexes 205
452

pelvic sphincters 215 rectum 206–208, 210, 215, 235, 239, 318, 320–321, 323,
pelvic sympathetic 33 326, 329, 339–342, 345–348, 350–355, 360–361,
pelvic urethra 416 363–364, 373, 375–376, 379–381, 402, 422, 431
pelvic viscera 11, 21–22, 86, 101, 206–207, 235, 239, 265, reflex 51–52, 55, 60, 64–65, 69, 74, 76, 78, 85, 89, 91–92,
267, 270–272, 361, 379, 401 97, 101, 103, 111, 119, 121, 127, 130, 135, 137, 143,
penis 100, 171, 361, 367–368, 393–395, 401, 403–409, 148–149, 153, 168, 173, 195, 205, 207–211, 215, 224,
415, 417–423, 429–430, 432 233, 235–236, 239, 241, 245–246, 248–249, 252, 256,
perineal 86–87, 89–90, 148, 154, 174, 178, 195, 361, 393, 268, 276, 289–290, 292, 296, 301, 318, 323, 326, 328,
402–404, 406, 409, 415–420, 422–423 336–341, 346–347, 353, 355, 359, 363, 367–369,
373–375, 377, 393–395, 403, 405, 408–409, 416–419,
periurethral 60
421–423, 428, 432, 435, 441–442, 444, 446
perivascular nerves 293, 295
reflex arousal 442
piloerection 246
reflex bladder 74, 76, 97, 117, 120–121, 128, 131
postganglionic 61, 78, 98, 277–278, 289–292, 295–296, reflex colonic 323, 328
321, 420, 429 reflex defecation 174, 178, 340, 355
postganglionic parasympathetic 320 reflex detrusor 55, 73
postganglionic sympathetic 28, 419 reflex ejaculation 435
preganglionic 18, 40, 52, 60–61, 67, 69–72, 87, 100–101, reflex erections 174, 178, 393–394
103, 149, 154, 164, 197, 200–201, 207, 231, 236, reflex micturition 148, 178
248–249, 251, 258–259, 265–266, 271, 277, 289–293, reflex sympathetic 290
295–296, 299, 321, 337, 363, 365, 393, 402, 404, 420 reflex vasoconstrictor 226
preganglionic neurons 18, 28, 31–32, 118, 266, 271, 308, reflexes 27, 52, 59, 65, 69, 76–77, 88, 91, 98–100,
321 118–119, 164, 183, 195, 205–206, 208–211, 213, 215,
preganglionic parasympathetic 118, 172, 196–201, 348 224–225, 231, 233, 235–236, 245–246, 248–249,
preganglionic sympathetic 269, 361 289–290, 292, 296, 299, 301, 320–321, 323–324,
preproneuropeptide 17 330, 338, 341, 349, 359–360, 363, 365–367, 369, 373,
presynaptic 423 376, 397, 430, 436
presynaptic inhibition 89–90, 92, 418, 420, 423 reflexive bladder 122, 164, 171, 173, 178
primary afferents 27–28, 35–36, 90, 92, 149–152, 155, reflexogenic 394, 404, 408
196, 248, 267–268 reflexogenic erection 393–394
proenkephalin 155 reinnervation 13, 195, 200–202, 291–292, 302
propriospinal 33–35, 265, 268, 270–272, 369 renal sympathetic 28–30, 33–36, 248
reproductive 366, 387, 401–402, 404–409, 416, 436
propriospinal interneurons 268
psychogenic 394
secondary injury 302, 369
pudendal 52, 54, 60, 62, 64, 68, 86–90, 97, 164, 169, 171,
semaphorin 3A 265, 267–268, 270
181–184, 207–210, 213, 215, 338, 347, 360–363,
sensory 51, 136
365–366, 393, 395, 401, 403–405, 415, 417, 419–421,
serotonergic 69
423, 429, 431, 444
serotonin 14–15, 87, 88, 117–118, 125, 130, 258, 322,
pudendal motoneuron 67, 364, 418, 420–422
365–366, 420, 423
pudendal motor nuclei 418 sexual dysfunction 195, 201, 259, 317, 345, 389, 408, 415,
pudendal primary afferents 417 428, 436, 441, 445–446
pudendal reflex 210, 213, 415, 417–422 sexual rehabilitation 388–390, 397
pudendal sympathetic 415, 419, 421 sexual response 388, 391–392, 416, 423, 441–442, 445–446
sexuality 387–390, 392, 397, 427, 441
rat 20–21, 33–36, 40, 60, 62–63, 66–72, 74–78, 88–89, 92, somato-visceral 207, 210, 213, 216
98, 100–103, 107, 117–120, 122, 124, 127, 131, somatosympathetic 98
151–154, 158, 196–200, 238, 246–253, 256–259, spasticity 52–53, 170, 177, 225, 359–360, 366–368, 430
265–271, 276, 278–279, 281–284, 289–290, sphincter 11, 51–55, 59, 64, 67–68, 70, 73–74, 77–78,
293–296, 301–303, 306, 309, 360–361, 363–368, 85–86, 88–92, 97–99, 121–122, 135, 147, 154, 178,
416–417, 420, 422–423, 433 197, 205–209, 211, 213, 215, 241, 318, 320–321,
rectal sensation 338, 340–341, 375, 379, 381 323–329, 335, 338–342, 345–347, 350–355, 359–361,
453

363–367, 373, 375–376, 378, 416, 418, 422–423, 430, transducer function 136
444 transganglionic 100, 402
sphincter dyssynergia 74, 85, 90–91, 155, 205, transit times 324, 326, 336–337, 341, 345, 350, 354–355
211, 213 transmission 295–296
sphincter guarding reflexes 211 transurethral 177
sphincter hyperreflexia 359 tyrosine hydroxylase 14
sphincter motoneuron 67, 70, 85–92
sphincter reflex 77, 85, 92, 205, 215 ultrastructure 137, 139–140
sphincter spasticity 52, 360 urethra 51–52, 55, 59–65, 68–72, 74, 76, 78, 85–86,
sphincterotomy 54, 148 88–90, 92, 97, 100, 103, 121–122, 135, 150, 154,
spinal sympathetic interneurons 27–28, 30 163–167, 174–175, 177, 180–183, 185, 187, 207–209,
spinal voiding reflexes 99 211, 239, 361, 416–418, 420, 422–423, 430–431
sprouting 35, 76, 99, 128, 147–148, 150–151, 153–155, urethra sphincter motoneurons 88
157–158, 213, 224, 245, 248, 251–252, 258–259, urethral sphincter 62, 65, 68, 70, 73, 78, 85–86, 97, 99,
265–271, 289–292, 296, 305, 307–308, 369 118,135, 346
sprouting primary afferents 147 urethral sphincter reflex 86
stimulation 278, 282–283 urinary 51–55, 135–136
stoma 329, 342 urinary bladder 19, 21, 51–52, 55, 59–60, 64–65, 67–68,
substance P 17, 20, 77, 87, 100, 111, 136, 149, 266–267, 70, 73, 75, 76, 97–100, 102–104, 107–108, 111, 112,
320, 337, 405 118,135, 136, 140–141, 143, 163, 207–208, 235, 237,
sudden cardiac death 278 239, 241, 246, 299, 354, 360, 431
sympathetic 17, 19, 21–22, 27–30, 32–33, 36, 39–40, 45,
urinary bladder areflexia 99
52, 60–62, 69, 73, 97–99, 101, 118, 164, 206–208,
urinary bladder detrusor 99
223–226, 231–236, 238–239, 246, 248–249, 258,
urinary bladder hyperreflexia 101
265–267, 270–272, 275–280, 282, 284–285, 289–293,
urinary bladder urothelium 142
295–296, 299, 308, 320–321, 323–326, 328, 336–339,
urinary tract 62, 68
347, 351–354, 359, 361, 366, 382, 389–390, 393–394,
urothelium 54, 63–64, 135–137, 140–143, 166
401–402, 416, 420–421, 423, 429–430, 441, 445
sympathetic activation 445
sympathetic adrenergic 54 vasoconstrictor 73, 233, 237, 272, 289, 291, 293, 295–296,
sympathetic ganglia 239, 289, 291, 361 393
sympathetic interneurons 27–36 vasoconstrictor preganglionic 271
sympathetic neurons 278 vasomotor 45
sympathetic pathways 61 vasomotor pathways 39, 42, 44–45, 225
sympathetic postganglionic 31, 419–421 ventral root avulsion 197–200
sympathetic preganglionic 18–22, 27–36, 39–40, 66, 101, ventrolateral medulla 13–14, 266, 365, 415
223–226, 246, 248, 265, 267–268, 270–272, 299, 301, vesicosympathetic reflex 69
305, 308, 366, 416, 429, 442 vesicoureteric 166
sympathetic reflex 27, 33, 36, 69, 246, 266, 300 vibroejaculation 428, 432, 434, 436
sympathoadrenal preganglionic neurons 21 viscera 140, 265, 408
synaptic plasticity 301, 304, 307 viscero-sympathetic reflex 265

tachy-arrhythmias 280 wallerian degeneration 299, 302, 304


testicular function 430, 434 wlds 299, 302, 304–308

Potrebbero piacerti anche