Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Review

J Vet Intern Med 1999;13:399–407

Current Concepts in the Management of Acute Spinal Cord Injury


Natasha Olby

Acute injury to the spinal cord initiates a sequence of vascular, biochemical, and inflammatory events that result in the development
of secondary tissue damage. Experimental studies and clinical trials in humans have demonstrated that the extent of this secondary
tissue damage can be limited by pharmacologic intervention at appropriate intervals after injury. High doses of methylprednisolone
sodium succinate (MPSS) improve the outcome of acute spinal cord injury in humans if treatment is initiated within 8 hours of
injury. Starting therapy more than 8 hours after injury is detrimental to outcome. The clinical efficacy of methylprednisolone in
improving the outcome of canine spinal cord injuries has not yet been demonstrated and its use by veterinarians is controversial.
Many dogs are not seen by a veterinarian within the 8-hour window after injury, and these dogs frequently are treated with
nonsteroidal anti-inflammatory drugs or large doses of dexamethasone or prednisone before methylprednisolone treatment can be
initiated, thus increasing the risk of severe adverse effects. Other drugs that may provide safer and more effective alternatives to
methylprednisolone include thyrotropin-releasing hormone (TRH), the 21-aminosteroids, and kappa opioid agonists. Recent studies
suggest that modulation of the influx of inflammatory cells and activation of endogenous microglial cells may provide another
means of improving the outcome of acute spinal cord injuries. Many drugs being developed to treat acute spinal cord injury have
shown promising results when evaluated experimentally. However, any proposed therapeutic strategy should be evaluated in pro-
spective blinded clinical trials before being adopted in clinics.
Key words: Disk disease; Methylprednisolone; Secondary injury; Spinal cord trauma; Thyrotropin-releasing hormone.

A cute spinal cord injuries form an important part of the


case load of veterinarians in general and specialty
practice. Such injuries are most commonly the result of
perimental drug trials. A useful experimental model of spi-
nal cord injury should fulfill several criteria including pro-
duction of reproducible, quantifiable pathologic changes
explosive protrusion of an intervertebral disc causing both comparable to clinical lesions and production of measurable
concussion and compression of the spinal cord. Other caus- neurologic deficits. Models of spinal cord injury have been
es include fractures, dislocation, or subluxation of the ver- developed to mimic concussion of the cord,7 compression
tebral column potentially causing laceration, compression, of the cord,8 and spinal cord ischemia.9 Most of these mod-
or concussion of the spinal cord; infarction (usually the els fulfill 1 or more of the above criteria, but a common
result of a fibrocartilagenous embolus); and gun shot problem is lack of lesion reproducibility.10 The resultant
wounds causing laceration of the spinal cord. Although re- decrease in the statistical power of the model necessitates
covery from incomplete spinal cord injuries is good, with the use of large numbers of animals in experimental groups
the majority of animals regaining motor function,1,2 animals to establish significant differences. Conflicting conclusions
with more severe injuries have a much poorer prognosis.3 drawn by different groups testing the same treatment par-
Treatment of spinal cord injuries by veterinarians has in adigm often can be ascribed to inadequate group size as
the past been aimed at surgical decompression of the spinal well as to the use of different models and different methods
cord where indicated, stabilization of the vertebral column, of assessing the effect of a treatment. When interpreting the
and strict rest.4 Corticosteroids have been used historically results of such an experiment, it is necessary to assess in-
with the aim of decreasing spinal cord swelling. More re- tragroup variability, the parameters that have been mea-
cently, high doses of corticosteroids with free radical scav- sured, and the relevance of the model to the clinical situ-
enging properties have been recommended after reports of ation in which its findings may be used.
successful clinical trials in humans.5,6 However, the current With the exception of injuries that cause lacerations of
interest in treating central nervous system (CNS) injuries the spinal cord, the primary tissue damage that occurs at
has led to a large number of often contradictory drug trials the time of injury is minor when compared to the secondary
that have been published over the last 2 decades. As a re- damage that develops over the next 48 hours. The period
sult, considerable controversy exists over the most appro-
after an injury can be divided into 3 different therapeutic
priate way to manage acute spinal cord injury. Much of this
windows: biochemical and vascular events, starting at the
controversy is a result of the difficulty of performing ex-
time of injury and continuing for up to 48 hours; the influ-
ence of inflammatory cells, starting within hours of injury
From the College of Veterinary Medicine, North Carolina State Uni- and peaking at 4 days after injury; and axonal regeneration
versity, Raleigh, NC. and lesion repair (CNS plasticity) from 1 week after injury.
Reprint requests: Natasha Olby, Vet MB, PhD, Department of Com- This paper has 3 goals: first, to review the pathophysi-
panion Animals and Special Species, College of Veterinary Medicine, ology of the development of secondary tissue damage fo-
North Carolina State University, 4700 Hillsborough Street, Raleigh, cusing on vascular, biochemical, and inflammmatory
NC 27606; e-mail: natashapolby@csu.edu.
events; second, to review experimental and clinical thera-
Received March 23, 1998; Revised September 1, 1998 and Decem-
ber 22, 1998; Accepted February 16, 1999. peutic trials targeted at the developing lesion and the in-
Copyright q 1999 by the American College of Veterinary Internal flammatory response; and third, to highlight those treatment
Medicine strategies that have been shown to have a beneficial effect
0891-6640/99/1305-0002/$3.00/0 clinically. Research developments in transplantation and the
400 Olby

use of growth factors aimed at promoting axonal regener- though 1 study has demonstrated a neuroprotective effect
ation and CNS plasticity have been reviewed elsewhere.11–14 of halothane anesthesia (postulated to be the result of a
direct vasodilatory effect),40 this effect was only apparent
Vascular and Biochemical Events During when halothane anesthesia was administered at the time of
Lesion Development: Pathophysiology and injury, a delay of even 10 minutes abolished any benefits.
Therapeutic Strategies Several different classes of drugs have been demonstrat-
ed to improve spinal cord blood flow after injury as a result
Vascular Abnormalities of local effects on the vasculature. These include calcium
Acute injury to the spinal cord causes both systemic and channel antagonists,28,29 nonsteroidal anti-inflammatory
local vascular abnormalities, which ultimately result in a agents,29 free radical scavengers,29 and opioid antagonists
progressive decrease in perfusion and necrosis of the in- such as naloxone.41,42 These drugs will be discussed further
jured section of the spinal cord.15–19 The development of in the appropriate sections below.
spinal cord ischemia has been proposed to be one of the
fundamental causes of secondary tissue damage.20 The de- Biochemical Events
crease in spinal cord perfusion develops over several hours,
giving clinicians the opportunity for therapeutic interven- The complex sequence of biochemical events initiated by
tion. The abnormalities that occur have been reviewed in trauma to the spinal cord has been reviewed elsewhere.15–19
detail,20 and can be summarized as follows Separate events are summarized below.
Initial Systemic Hypertension. Initial systemic hyperten- Increases in Intracellular Calcium. After acute trau-
sion is followed within 10 minutes by sympathetic paralysis matic spinal cord injury, a rapid increase in intracellular
and hypotension that is maintained for several hours.21–25 calcium concentrations occurs.43,44 The routes of calcium
These changes occur in the rat,25 cat,23 dog,21 and primate.22 entry include direct membrane damage, voltage-gated cal-
The severity of the changes is dependent on the degree of cium channels triggered by membrane depolarization, and
injury. activation of N-methyl D-aspartate (NMDA) receptors by
Progressive Decrease in Spinal Cord Blood Flow. This the excitatory neurotransmitter glutamate. The increase in
change is the result of loss of autoregulation (see below), calcium results in activation of intracellular proteases and
destruction of microvasculature,20 thrombus formation,26,27 phospholipase A2, impairment of mitochondrial function,
and vasospasm induced by increased intracellular calcium spasm of vascular smooth muscle, and binding of phos-
concentrations28 and release of vasoactive chemicals such phates by calcium, thus depleting the cell of energy sourc-
as prostanoids.29 These events affect gray matter before es.45
white matter and in severe injuries have been maintained The calcium channel antagonist nimodipine has been
for up to 7 days after injury.30,31 In less severe injuries, shown to improve posttraumatic spinal cord blood flow and
reactive hyperemia of the damaged cord may occur.30 axonal function in the rat when used at a dosage of 0.02
Loss of Autoregulation. The spinal cord can normally mg/kg for 1 hour in combination with dextran to support
maintain constant blood flow in the face of changing sys- systemic blood pressure.28 Nimodipine also supported spi-
temic blood pressure, but this property is lost after inju- nal cord blood flow when used in baboons with compres-
ry.23,32,33 sive spinal cord lesions at a dosage of 0.02 mg/kg/hour for
Spinal cord blood flow and injury severity are inversely 2 hours and then 0.04 mg/kg/hour for 1 week.46 Neither of
related,28 and many treatments therefore are aimed at sup- these studies evaluated functional recovery. Other groups,
porting spinal cord blood flow. Decreasing systemic blood using similar injury models and comparable drug regimes,
pressure results in a decrease in spinal cord perfusion be- have been unable to demonstrate a beneficial effect of ni-
cause of loss of autoregulation,34 and therefore supporting modipine in spinal cord injury, bringing the efficacy of ni-
systemic blood pressure has been proposed to support spi- modipine into question.47–49 Other calcium channel blockers
nal cord blood flow.20 Maintaining systemic blood pressure also have been evaluated. Pretreatment with diltiazem (150
with pressors, whole blood transfusions, or dextran will im- mg/kg given as a bolus IV 30 minutes before injury fol-
prove spinal cord perfusion postinjury,35,36 but only 1 study lowed by 5 mg/kg/minute as a constant-rate IV infusion for
has demonstrated an improvement in spinal cord function.37 4 hours) and nifedipine (10 mg/kg given as a bolus IV 30
Moreover, hypertension does not result in an increase in minutes before injury followed by 1 mg/kg/minute as a con-
spinal cord blood flow, and indeed may increase chances stant rate IV infusion for 4 hours) in a weight drop model
of hemorrhage and edema.38,39 The importance of maintain- of spinal cord injury in the cat resulted in an improvement
ing systemic blood pressure has become more apparent in in spinal cord blood flow.29 Verapamil did not produce this
studies testing other drugs. For example the calcium chan- effect. In the same study, animals treated with calcium
nel blocker nimodipine has been demonstrated to improve channel antagonists did not improve to the same extent as
axonal function and spinal cord blood flow after injury those treated with the antioxidants vitamin E and selenium.
when used in combination with dextran to maintain system- Flunarizine has been evaluated in a model of acute spinal
ic blood pressure.28 In view of the importance of systemic cord injury in the cat, and proved more effective than meth-
blood pressure, the recommended procedure when manag- ylprednisolone. Unfortunately, this study only demonstrated
ing acute spinal cord injury is to maintain normotension.20 a greater restoration of somatosensory evoked potential am-
This is important to bear in mind as many veterinary pa- plitudes at 4 hours after injury. Flunarizine had no effect
tients undergo prolonged general anesthesia for myelogra- on spinal cord blood flow, and a full functional study was
phy and surgery soon after severe spinal cord injuries. Al- not performed.45 Drug treatment in these studies was insti-
Acute Spinal Cord Injury 401

tuted either before or immediately after injury, and the ef- able free radical-induced damage is occurring. A more re-
fects of delaying treatment were not examined. cent clinical trial in humans demonstrated that if treatment
Calcium channel antagonists in combination with a pres- with MPSS is initiated within 3 hours of injury, a regime
sor may provide a useful clinical treatment for spinal cord that continues a maintenance infusion of the drug for 24
injury in the future.50 However, experimental studies eval- hours should be used.65 If treatment is initiated between 3
uated indirect markers of recovery, with few studies eval- and 8 hours after injury, the infusion should be continued
uating functional recovery. This class of drug has not been for 48 hours.65 The same study demonstrated increased oc-
tested in clinical trials on spinal cord injury, and the clinical currence of severe pneumonia and sepsis when MPSS treat-
efficacy of calcium channel blockers remains unclear at ment was continued for 48 hours, but this finding was not
present. associated with increased mortality. In another clinical
Free Radical Production. Demopoulos et al.51 suggested study, high doses of MPSS in acute spinal cord injury were
that free radical-induced lipid peroxidative damage to mem- associated with prolonged hospitalization as a result of ste-
branes was a major factor causing secondary tissue destruc- roid-related adverse effects. This study questioned the ben-
tion after spinal cord injury. Increased intracellular calcium efits of MPSS use.66 Despite the positive reports on the use
concentrations, ischemic conditions, reperfusion, and the of MPSS in spinal cord injury, use of this drug remains
presence of iron and copper complexes in petechial hem- controversial.
orrhages all contribute to the production of free radicals.52,53 In veterinary neurology, a similar controversy exists con-
When the intrinsic protective free radical scavenging sys- cerning the beneficial effects of MPSS on the injured spinal
tems are overwhelmed, free radicals start to react with cell cord. A study by Coates et al67 failed to demonstrate a pro-
enzymes and unsaturated fatty acids in cell membranes, tective effect of MPSS in a model of spinal cord injury in
causing lipid peroxidation. The result is direct lipid per- dogs. However, only 4 animals were used to test MPSS and
oxidative damage of neuronal and glial cell membranes. In such a small number would only demonstrate a difference
addition, peroxidation of endothelial cell membranes trig- if the model were extremely reproducible. In contrast, an-
gers platelet aggregation, which results in intensification other study found a beneficial effect of MPSS in dogs un-
and extension of the area of posttraumatic spinal cord is- dergoing spinal surgery, but no controls were included, and
chemia.28,51,53 the relevance of these findings was unclear.68 As a result, it
Pretreatment of cats for 5 days with a combination of the is still uncertain whether the use of MPSS is helpful in dogs
free radical scavengers vitamin E (1,000 IU/day) and se- with spinal cord injury. Most experimental and clinical ev-
lenium (50 mg/day) has been shown to preserve spinal cord idence in other species indicates that MPSS is beneficial,
perfusion28 and improve neurologic outcome after spinal but only when given within a certain time period after in-
cord injury.54 The length of time required for these anti- jury and following a particular dosage regimen. The regi-
oxidants to reach therapeutic concentrations in the CNS men recommended for cats is to start with an initial dosage
precludes their use after acute spinal cord injury, and de- of 30 mg/kg IV, followed at 2 and 6 hours with dosages of
velopment of other drugs for this purpose has been neces- 15 mg/kg and then to continue with an IV infusion of 2.5
sary. The most notable of these drugs is methylprednisolone mg/kg/hour for 42 hours.53 The regimen adopted by Sie-
sodium succinate (MPSS). MPSS is a glucocorticoid that mering and Vomering68 for dogs before performing spinal
has free radical scavenging properties when used at very surgery was to give an initial IV bolus of 30 mg/kg fol-
high dosages.52,53 Many experimental studies using models lowed by a 24-hour continuous IV infusion of 5.4 mg/kg/
of both acute spinal cord concussion and compression have hour as in the clinical trials in humans. However, little in-
demonstrated that MPSS has a neuroprotective effect when formation is available on the optimal dosage regimen in
given at the time of or within minutes after spinal cord dogs.
injury.55–59 Despite variation in reported dosage regimes, the High doses of MPSS are associated with several prob-
common aim is to maintain therapeutic concentrations of lems in veterinary patients. A recent study of dogs under-
MPSS in the spinal cord during the 24–48 hours of lesion going spinal surgery that received a single bolus of 30 mg/
development. kg of MPSS followed by a second half to full dose 2–4
Multicenter clinical trials have tested the use of MPSS hours later reported that 90% of the dogs developed occult
in humans after acute spinal cord injury and have demon- gastrointestinal hemorrhage.69 Unfortunately, many patients
strated a positive outcome when MPSS is given within 8 with spinal cord injuries that are seen at referral institutions
hours of injury.5,6 Patients received an initial IV bolus of already have been treated with large doses of prednisone,
30 mg/kg of MPSS followed by an IV infusion of 5.4 mg/ dexamethasone, or a nonsteroidal anti-inflammatory agent.
kg/hour of MPSS for 24 hours. The clinically detectable How such treatment would influence the effect of subse-
benefits are small but significant and involve both long tract quent treatment with MPSS is not known, but such com-
and segmental function.60 These trials also demonstrated binations of drugs may predispose the patient to the adverse
that initiation of MPSS treatment in those patients with in- effects of high doses of MPSS. Another important consid-
complete injuries more than 8 hours after injury resulted in eration is the time interval after injury. In referred patients,
a worse outcome.6,60 The proposed mechanism for this find- this interval is often greater than 8 hours, and experience
ing is that glucocorticoids interfere with normal regenera- in clinical trials involving humans suggests that the use of
tion61,62 and will enhance neurodegeneration by inhibiting MPSS is contraindicated in such patients.60 As a result of
neuronal glucose uptake in the face of ischemia and glu- the potential adverse effects of MPSS use and the limited
tamate-induced damage.63,64 MPSS should therefore be giv- efficacy of this drug, care should be exercised before using
en within 8 hours after spinal cord injury, when consider- it. MPSS should be reserved for patients with severe spinal
402 Olby

cord injuries that have occurred within 8 hours of presen- flow.78 Concern also exists about other toxic effects such
tation and that have not been pretreated with other steroids. as fever and renal toxicity.18,50
Some veterinary institutions pretreat dogs with MPSS Increases in Excitatory Amino Acids. Extracellular con-
when performing potentially traumatic procedures such as centrations of the excitatory amino acids (EAAs) glutamate
myelography and spinal surgery. Although seemingly log- and aspartate are increased after trauma because of depo-
ical, the benefits of such MPSS use are unclear. The main larization-induced release, release from damaged neurons,
concern is that the animal has already suffered an acute and decreased uptake by ischemic astrocytes.79 These amino
spinal cord injury more than 8 hours previously, and further acids interact with NMDA and non-NMDA receptors caus-
doses of MPSS may be detrimental. Five days of pretreat- ing a rapid increase in intracellular sodium concentration
ment with vitamin E and selenium may be considered and cellular swelling, and a slower increase in intracellular
whenever an elective procedure that may result in trauma calcium concentration.79 The toxicity of excess EAAs has
to the spinal cord is to be performed. This approach has been demonstrated conclusively both in vitro and in
the advantage of affording a similar degree of neuroprotec- vivo.79,80 Moreover, after acute thoracolumbar intervertebral
tion as MPSS without the potential detrimental effects.28 disc herniation in dogs, the concentration of glutamate in
The adverse effects associated with high doses of MPSS lumbar cerebrospinal fluid is increased in proportion to the
have led to controversy over their use66 and have resulted severity of the injury.81 NMDA and non-NMDA receptor
in development of the 21-aminosteroids. These drugs are antagonists such as MK801, thienylphencyclidine, and
similar in structure to MPSS but lack the glucocorticoid NBQX have been tested in ischemic, concussive, and com-
receptor-mediated effects. These drugs have been effective pressive models of spinal cord injury in the rat.82–85 These
in experimental studies in cats70,71 and rats57,72 but not in studies all demonstrated improvement in functional recov-
dogs.67 One of these drugs (tirilazad mesylate or U74006F) ery, but unfortunately these drugs can cause adverse effects
recently was assessed in clinical trials in humans (NASCIS including sedation and ataxia, motor stimulation, cardio-
3)65 in which a 48-hour dosage regimen was shown to have vascular changes, and vacuolation of certain neuronal pop-
efficacy similar to that of MPSS if initiated within 3 hours ulations.86 Therefore, the search for drugs that counteract
of injury. However, tirilazad was shown to be inferior to the toxic effects of EAAs with minimal adverse effects is
MPSS when treatment was initiated more than 3 hours after continuing.
injury.65 A drawback of this study was that all patients re- Endorphin-Associated Ischemia. The importance of en-
cieved an initial bolus of 30 mg/kg of MPSS (as a result dorphins in mediating local ischemia in the injured spinal
of the demonstrated benefits of MPSS) and objective eval- cord is well recognized.42,87 Naloxone, a nonspecific opioid
uation of the true efficacy of tirilazad is difficult. In addi- antagonist, supports systemic blood pressure and spinal
tion, an optimal dosage regimen for tirilazad has yet to be cord blood flow in models of spinal cord injury in the rat
determined. Further clinical trials on the use of tirilazad to and cat, resulting in an improvement in functional out-
treat acute spinal cord injury are planned. come.42,74,87,88 A clinical trial of the efficacy of naloxone in
The use of large doses of dexamethasone by veterinarians humans, using a 5.4 mg/kg IV bolus, followed by 4 mg/
with the aim of reducing posttraumatic spinal cord swelling kg/hour constant-rate infusion for 23 hours, revealed im-
is widespread. However, experimental trials examining the provement in descending motor function in patients with
efficacy of dexamethasone have failed to show a beneficial incomplete spinal cord injuries if administered within 8
effect.73,74 Moreover, evidence exists that glucocorticoids hours of injury.60 Because naloxone is both more expensive
can be detrimental61–64 and the use of high doses of dexa- and less effective than MPSS it is not used routinely.60
methasone in the treatment of acute spinal cord injuries Research associated with manipulation of the endoge-
should be avoided. Anti-inflammatory doses of prednisone nous opioids is concentrated on k-opioid agonists (eg, U-
commonly are used for dogs with thoracolumbar and cer- 50488H) and antagonists (eg, nalmefene). Paradoxically,
vical spinal pain. However, such dogs are at increased risk both of these drugs improve the functional outcome of
of gastrointestinal hemorrhage, and care should be taken to acute spinal cord injury in rats.89–91 A good explanation for
ensure that the animal is adequately confined while on pred- these results has not been provided, but it is believed to be
nisone. due either to nonopioid actions of the drugs, or to action at
The ability of the free radical scavenger dimethyl sulf- distinct k-receptor subtypes.90 The k-opioid agonist CI-977
oxide (DMSO) to decrease the severity of spinal cord injury exerts a neuroprotective effect on experimental models of
has been investigated in rats,75 cats,73 and dogs.76,77 How- cerebral ischemia by its ability to decrease glutamate re-
ever, results have been conflicting, varying from providing lease.92 However, in a model of spinal cord ischemia, CI-
good neuroprotection81 to no apparent effect.73,76 The stud- 977 had a detrimental effect believed to be the result of a
ies used similar dosage regimens for DMSO, and why such decrease in systemic blood pressure.93 The development of
discrepancy exists in the results is not clear. However, all both k-opioid agonists and antagonists is continuing, but as
of these studies were performed at least 15 years ago, when yet no drug has emerged that is superior to MPSS.
physiologic parameters such as systemic blood pressure and
end tidal carbon dioxide concentrations were not controlled. The Role of Thyrotropin-Releasing Hormone
A study of the effect of DMSO on the circulation of the (TRH)
dog showed that a dosage of 2 g/kg (ie, the standard initial
bolus used in the treatment of experimental spinal cord TRH has a wide variety of physiologic actions in addi-
trauma) caused significant hemolysis with a decrease in he- tion to its well-known effect on production and release of
matocrit and a significant decrease in spinal cord blood thyroxine. These include the ability to block the autonomic
Acute Spinal Cord Injury 403

effects of opioids41 and antagonism of the effects of both roprotection in rats after spinal cord injury by administering
vasoactive agents94,95 and EAAs.96 Treatment with TRH (0.2 lipopolysaccharide to increase the release of growth-pro-
mg/kg/hour IV) after experimental spinal cord injury in the moting cytokines, in combination with a nonsteroidal anti-
cat improves functional outcome,74,97 even when adminis- inflammatory agent and steroid, to suppress the effects of
tration is delayed for up to 1 week after injury.98 This per- cytotoxic cytokines.117 However, this finding has not been
haps is a result of TRH’s neurotrophic effects, influences reproduced by other groups.
on plasticity, and facilitation of motor neuron firing.57 Sub- The neuroprotective effect of MPSS may be due in part
sequent studies in cats and rats also have demonstrated that to glucocorticoid receptor-mediated inhibition of phospho-
the use of TRH and its analog in spinal cord injury is ben- lipase A2.58 However, MPSS has no effect on postinjury
eficial and superior to naloxone, MPSS, U-50488H, and concentrations of the products of phospholipase A2 activa-
nalmefene.74,75,90 Only 1 study failed to demonstrate a pos- tion, supporting the hypothesis that MPSS’s neuroprotective
itive effect from the use of TRH in cats, but this study action is mediated by free radical scavenging rather than
suffered from having only 4 animals per treatment group.99 anti-inflammatory actions.59
TRH has passed the 1st phase of clinical trials in humans
(at a dosage of 0.2 mg/kg initial IV bolus followed by a 6- Miscellaneous Therapeutic Strategies
hour IV infusion of 0.2 mg/kg/hour initiated within 12
hours of injury).100 TRH has disadvantages, including its Gangliosides
analeptic, endocrine, and autonomic effects, but a new gen-
eration of TRH analogs has been developed that have the Gangliosides are lipid components of the outer bilayer of
protective effects of TRH without its adverse effects.101 plasma membranes that play vital roles in the normal func-
These drugs could be of use to veterinarians both because tion of the nervous system.118 Several studies have shown
of their beneficial effect even when given at intervals of up the ganglioside GM1 to attenuate the effects of EAAs, be
to 1 week after injury, and also because of their apparent neuroprotective in acute CNS injury, and to potentiate the
superiority to other drugs. effect of neurotrophic factors.118 The ganglioside GM1 was
tested for efficacy in improving the outcome of acute spinal
The Inflammatory Response: Pathophysiology cord injury in a small-scale clinical trial in humans and was
found to be beneficial when given as late as 48–72 hours
and Therapeutic Strategies after injury at a dosage of 100 mg IV per day for 3–4
The inflammatory response to CNS damage results in the weeks.119 A subsequent experimental study failed to dem-
production of a variety of cytotoxic and protective onstrate a beneficial effect of GM1 when used to treat spi-
agents.102–106 Therefore, it is not surprising that the role nal cord trauma in the rat, and in addition suggested that
played by the inflammatory response in the development of GM1 antagonized the effect of MPSS.58 Larger-scale clin-
secondary spinal cord damage is controversial. Recruitment ical trials using GM1 in combination with an initial IV bo-
of inflammatory cells into spinal cord lesions lags behind lus of MPSS have just been completed and suggest that
the development of acute secondary cell death,107 but co- GM1 has a small beneficial effect when used in this way.120
incides with demyelination of surviving axons and chronic
neuronal death.108–110 Several studies have reported that in- Hypothermia
hibiting the infiltration of mononuclear cells after spinal
cord injury is beneficial.111–114 The delayed pathologic effect A decrease in brain temperature is remarkably neuropro-
of macrophages infiltrating the damaged spinal cord of the tective after an ischemic insult.121 The neuroprotective
guinea pig is thought to be a consequence of their produc- mechanisms of hypothermia include decreasing the rate of
tion of the NMDA agonist quinolinic acid. This hypothesis depletion of high-energy phosphates, decreasing the extra-
is supported by the observation that antagonists to quino- cellular concentrations of glutamate and other neurotrans-
linate reduce the severity of delayed injury.106 However, not mitters, and decreasing activation of enzymes such as pro-
all species produce significant amounts of quinolinic acid tein kinase II.121 The effect of hypothermia on acute spinal
and the clinical importance of this finding remains to be cord injury also has been evaluated. The original trials per-
assessed in the dog and cat. formed in the 1970s involved prolonged perfusion (2.5
The situation is further complicated when the microglial hours) of previously injured spinal cord with isotonic saline
cell is considered. Microglial cells are the endogenous (58C) to achieve a spinal cord temperature of approximately
phagocytic cells of the CNS, and can release potentially 108C. These studies demonstrated neuroprotection even
cytotoxic chemicals such as hydrogen peroxide, nitric ox- when hypothermia was initiated 3–6 hours after injury.122
ide, proteinases, and the cytokines interleukin-1 (IL-1) and This finding led to trials in humans that failed because of
tumour necrosis factor-a within minutes of injury.102,104 Ad- a high mortality rate (apparently due to respiratory com-
ministration of an IL-1 receptor antagonist at 30 minutes plications in cervical injuries) and the difficulty of a tech-
before and 10 minutes after inducing cerebral ischemia in nique that required early surgical intervention to perform a
the rat significantly decreased infarct volume and this effect multilevel laminectomy to expose the spinal cord.122 More
was thought to be due to antagonism of IL-1 produced by recently, very modest decreases in brain temperature (1–
microglial cells.115 The presence of such cytokines in the 38C) were found to be neuroprotective121 and interest in the
injured spinal cord has been described,116 but their role in use of modest hypothermia as a treatment of spinal cord
the production of secondary tissue damage has yet to be injury has been renewed. A 1–38C drop in spinal cord tem-
investigated in detail. One study demonstrated marked neu- perature may be achieved by decreasing rectal temperature
404 Olby

to 31–328C and studies are underway to assess the efficacy References


of this approach.122
1. Davies JV, Sharp NJH. A comparison of conservative treatment
and fenestration for thoracolumbar disc disease in the dog. J Small
Cerebrospinal Fluid (CSF) Drainage Anim Pract 1983;24:721–729.
2. Butterworth SJ, Denny HR. Follow-up of 100 cases with thora-
Recently CSF drainage has been adopted as a means of
columbar disc protrusions treated by lateral fenestration. J Small Anim
maintaining cerebral perfusion pressure following acute Pract 1991;32:443–447.
brain injury.123 The efficacy of CSF drainage at improving 3. Duval J, Dewey C, Roberts R, Aron D. Spinal cord swelling as
the outcome of ischemic spinal cord injury has now been a myelographic indicator of prognosis: A retrospective study in dogs
evaluated.124 An increase in CSF pressure has been dem- with interverterbral disc disease and loss of deep pain perception. Vet
onstrated to decrease spinal cord blood flow.125 The aim of Surg 1996;25:6–12.
CSF drainage is to maintain perfusion pressure within the 4. Wheeler SJ, Sharp NJH. Small Animal Spinal Disorders. Diag-
spinal cord and to prevent the decrease in spinal cord blood nosis and Surgery. London: Mosby-Wolfe;1994:171–191.
flow that occurs after acute injury. The cited study found 5. Bracken MB, Shepard MJ, Collins WF, et al. A randomized, con-
trolled trial of methyl-prednisolone or naloxone in the treatment of
that a combination of CSF drainage and the 21-aminoster-
acute spinal-cord injury. N Engl J Med 1990;322:1405–1411.
oid tirilazad was most effective at reducing the severity of
6. Bracken MB, Shepard MJ, Collins WF, et al. Methylprednisolone
spinal cord injury after acute spinal cord ischemia in the or naloxone treatment after acute spinal cord injury: 1-year follow-up
rabbit.124 data. J Neurosurg 1992;76:23–31.
7. Wrathall JR, Pettegrew RK, Harvey F. Spinal cord contusion in
Conclusions the rat: Production of graded, reproducible, injury groups. Exp Neurol
1985;88:108–122.
The experimental and clinical findings to date suggest 8. Rivlin AS, Tator CH. Objective clinical assessment of motor
that the functional outcome of acute spinal cord injury in function after experimental spinal cord injury in the rat. J Neurosurg
humans, rats, and cats can be influenced by management 1977;47:577–581.
9. Watson BD, Prado R, Dietrich WD, et al. Photochemically in-
of systemic blood pressure and by the use of MPSS within
duced spinal cord injury in the rat. Brain Res 1986;367:296–300.
8 hours of injury. At present, whether use of MPSS in this 10. Khan M, Griebel R. Acute spinal cord injury in the rat: Com-
way is beneficial to dogs with acute spinal cord injuries is parison of three experimental techniques. Can J Neurol Sci 1983;10:
not clear, but the efficacy of MPSS in other species is com- 161–165.
pelling. Unfortunately, the clinical experience in humans 11. Cheng H, Cao Y, Olson L. Spinal cord repair in adult paraplegic
indicates that MPSS should be used only if it can be ad- rats: Partial restoration of hind limb function. Science 1996;273:510–
ministered within 8 hours of injury. In view of the time that 513.
often elapses between injury and presentation of animals to 12. Bregman BS, Kunkel-Bagden E, Reier PJ, et al. Recovery of
the veterinarian, treatment paradigms that can be instituted function after spinal cord injury: Mechanisms underlying transplant-
after a longer delay from the time of injury (eg, TRH an- mediated recovery of function differ after spinal cord injury in new-
born and adult rats. Exp Neurol 1993;123:3–16.
alogs, GM1) and drugs that have not as yet been shown to
13. Li Y, Field PM, Raisman G. Repair of adult rat corticospinal
have the same detrimental effects as MPSS (eg, 21-ami- tract by transplantation of olfactory ensheathing cells. Science 1997;
nosteroids) should be studied. Little support exists for the 277:2000–2002.
use of dexamethasone to treat acute spinal cord injuries, 14. Zompa EA, Cain LD, Everhart AW, et al. Transplant therapy:
and this practice is to be discouraged in light of the high Recovery of function after spinal cord injury. J Neurotrauma 1997;14:
frequency of adverse gastrointestinal effects associated with 479–506.
this drug and its potential detrimental effects on the recov- 15. Janssens LAA. Mechanical and pathophysiological aspects of
ering nervous system. For those animals that are to undergo acute spinal cord trauma. J Small Anim Pract 1991;32:527–579.
elective procedures during which acute spinal cord injury 16. Coughlan AR. Secondary injury mechanisms in acute spinal
could occur, experimental evidence exists that prophylactic cord trauma. J Small Anim Pract 1993;34:117–122.
17. Braund KG, Shores A, Brawner WR. The etiology, pathology,
use of a combination of vitamin E and selenium could be
and pathophysiology of acute spinal cord trauma. Vet Med 1990;85:
beneficial. 684–691.
New experimental treatment paradigms for acute spinal 18. Kraus KH. The pathophysiology of spinal cord injury and its
cord injury are emerging, and a critical evaluation of each clinical implications. Semin Vet Med Surg 1996;11:201–207.
study should be made in order to identify those that are 19. Jeffrey NH. Pathophysiology of spinal cord trauma. Chapter 3.
worth pursuing further. Double-blinded clinical trials form In: Handbook of Small Animal Spinal Surgery. London: WB Saun-
the standard for adoption of treatment protocols in human ders; 1995:64–84.
medicine, but few such trials have been performed on drugs 20. Tator CH, Fehlings MG. Review of the secondary injury theory
used in veterinary neurology. In order to establish the most of acute spinal cord trauma with emphasis on vascular mechanisms. J
appropriate way of managing acute spinal cord injury in Neurosurg 1991;75:15–26.
21. Griffiths IR. Vasogenic edema following acute and chronic spi-
small animals, large-scale prospective clinical trials should
nal cord compression in the dog. J Neurosurg 1975;42:155–165.
be performed.
22. Sandler AN, Tator CH. Effect of acute spinal cord compression
injury on regional spinal cord blood flow in primates. J Neurosurg
Acknowledgments 1976;45:660–676.
23. Senter HJ, Venes JL. Altered blood flow and secondary injury
I would like to thank Dr Karen Muñana and Dr Nick in experimental spinal cord trauma. J Neurosurg 1978;49:569–578.
Sharp for their comments on this manuscript. 24. Rawe SE, Perot PL. Pressor response resulting from experi-
Acute Spinal Cord Injury 405

mental contusion injury to the spinal cord. J Neurosurg 1979;50:58– channel antagonist nimodipine in experimental spinal cord ischemia.
63. J Neurosurg 1984;60:796–799.
25. Guha A, Tator CH. Acute cardiovascular effects of experimental 48. Ford RW, Malm DN. Failure of nimodipine to reverse acute
spinal cord injury. J Trauma 1988;28:481–490. spinal cord injury. Cent Nerv Syst Trauma 1985;2:9–17.
26. Balentine JD. Pathology of experimental spinal cord trauma I. 49. Haghighi SS, Stiens T, Oro JJ, Madsen R. Evaluation of calcium
The necrotic lesion as a function of vascular injury. Lab Invest 1978; channel antagonist nimodipine after experimental spinal cord injury.
39:236–253. Surg Neurol 1993;39:403–408.
27. Dow-Edwards D, DeCrescito V, Tomasula JJ, Flamm ES. Effect 50. Meintjes E, Hosgood G, Daniloff J. Pharmaceutic treatment of
of aminophylline and isoproterenol on spinal cord blood flow after acute spinal cord trauma. Compend Cont Educ 1996;18:625–635.
impact injury. J Neurosurg 1980;53:385–390. 51. Demopoulos HB, Flamm ES, Pietronigro DD. The free radical
28. Fehlings MG, Tator CH, Linden RD. The effect of nimodipine pathology and the microcirculation in the major central nervous system
and dextran on axonal function and blood flow following experimental disorders. Acta Physiol Scand 1980;492:91–119.
spinal cord injury. J Neurosurg 1989;71:403–416. 52. Hall ED. The neuroprotective pharmacology of methylprednis-
29. Hall ED, Wolf DL. A pharmacological analysis of the patho- olone. J Neurosurg 1992;76:13–22.
physiological mechanisms of posttraumatic spinal cord ischemia. J 53. Brown SA, Hall ED. Role of oxygen-derived free radicals in
Neurosurg 1986;64:951–961. the pathogenesis of shock and trauma, with focus on central nervous
30. Ducker TB, Salcman M, Perot PL, Ballantine D. Experimental system injuries. J Am Vet Med Assoc 1992;200:1849–1859.
spinal cord trauma, I: Correlation of blood flow, tissue oxygen and 54. Anderson DK, Waters TR, Means ED. Pretreatment with alpha
neurologic status in the dog. Surg Neurol 1978;10:60–63. tocopherol enhances neurologic recovery after experimental spinal
31. Ducker TB, Salcman M, Lucas JT, et al. Experimental spinal cord compression. J Neurotrauma 1988;5:61–67.
cord trauma, II: Blood flow, tissue oxygen, evoked potentials in paretic 55. Means ED, Anderson DK, Waters TR, Kalaf L. Effect of meth-
and plegic monkeys. Surg Neurol 1978;10:64–70. ylprednisolone in compression trauma to the feline spinal cord. J Neu-
32. Kobrine AI, Doule TF, Rizzoli HV. Spinal cord blood flow as rosurg 1981;55:200–208.
affected by changes in systemic arterial pressure. J Neurosurg 1976; 56. Braughler JM, Hall ED, Means ED, et al. Evaluation of an
44:12–15. intensive methylprednisolone sodium succinate regimen in experimen-
33. Young W, DeCrescito V, Tomasula JJ. Effect of sympathectomy tal spinal cord injury. J Neurosurg 1987;67:102–105.
on spinal cord blood flow autoregulation and posttraumatic ischaemia. 57. Behrmann DL, Bresnahan JC, Beattie MS. Modelling of acute
J Neurosurg 1982;56:706–710. spinal cord injury in the rat: Neuroprotection and enhanced recovery
34. Guha A, Tator CH, Rochon J. Spinal cord blood flow and sys- with methylprednisolone, U-74006F and YM-14673. Exp Neurol
temic blood pressure after experimental spinal cord injury in rats. 1994;126:1–75.
Stroke 1989;20:372–377. 58. Constantini S, Young W. The effects of methylprednisolone and
35. Dolan EJ, Tator CH. The effect of blood transfusion, dopamine the ganglioside GM1 on acute spinal cord injury in rats. J Neurosurg
and gamma hydroxybutyrate on posttraumatic ischemia of the spinal 1994;80:97–111.
cord. J Neurosurg 1982;56:350–358. 59. Hall ED, Yonkers PA, Taylor BM, Sun FF. Lack of effect of
36. Guha A, Tator CH, Smith CR, Piper I. Improvement in post- postinjury treatment with methylprednisolone or tirilazad mesylate on
traumatic spinal cord blood flow with a combination of a calcium the increase in eicosanoid levels in the acutely injured cat spinal cord.
channel blocker and a vasopressor. J Trauma 1989;29:1440–1447. J Neurotrauma 1995;12:245–256.
37. Young WF, Rosenwasser RH, Vasthare US, Tuma RF. Preser- 60. Bracken MB, Holford TR. Effects of timing of methylprednis-
vation of post-compression spinal cord function by infusion of hyper- olone or naloxone administration on recovery of segmental and long-
tonic saline. J Neurosurg Anesthesiol 1994;6:122–127. tract neurological function in NASCIS 2. J Neurosurg 1993;79:500–
38. Rawe SE, Lee WA, Perot PL. The histopathology of experi- 507.
mental spinal cord trauma. The effect of systemic blood pressure. J 61. Scheff SW, Hoff SF, Anderson KJ. Altered regulation of lesion-
Neurosurg 1978;48:1002–1007. induced synaptogenesis by adrenalectomy and corticosterone in young
39. Alderman JL, Osterholm JL, D’Amore BR, et al. Influences of adult rats. Exp Neurol 1986;93:456–470.
arterial blood pressure upon central hemorrhagic necrosis after severe 62. Jap Tjoen San ERA, Schmidt-Michels M, Oestreicher AB, et
spinal cord injury. Neurosurgery 1979;4:53–55. al. Dexamethasone-induced effects on B-50/GAP-43 expression and
40. Salzman SK, Lee WA, Sabuto S, et al. Halothane anesthesia is neurite outgrowth in PC12 cells. J Mol Neurosci 1992;3:189–195.
neuroprotective in experimental spinal cord injury: Early hemodynam- 63. Sapolsky RM. Glucocorticoids, stress and exacerbation of ex-
ic mechanisms of action. Res Commun Chem Pathol Pharmacol 1993; citotoxic neuron death. Semin Neurosci 1994;6:323–331.
80:59–81. 64. Smith-Swintosky VL, Pettegrew LC, Sapolsky RM, et al. Me-
41. Faden AI. Role of thyrotropin-releasing hormone and opiate tyrapone, an inhibitor of glucocorticoid production, reduces brain in-
receptor antagonists in limiting central nervous system injury. Adv jury induced by focal and global ischemia and seizures. J Cereb Blood
Neurol 1988;47:531–546. Flow Metab 1996;16:585–598.
42. Young W, Flamm ES, Demopoulos HB, et al. Effect of nalox- 65. Bracken MB, Shepard MJ, Holford TR, et al. Administration
one on posttraumatic ischemia in experimental spinal contusion. J of methylprednisolone for 24 or 48 hours or tirilazad mesylate for 48
Neurosurg 1981;55:209–219. hours in the treatment of acute spinal cord injury. JAMA 1997;277:
43. Stokes BT, Fox P, Hollinden G. Extracellular calcium activity 1597–1604.
in the injured spinal cord. Exp Neurol 1983;80:561–572. 66. Galandiuk S, Raque G, Appel S, Polk HC Jr. The two-edged
44. Young W, Koreh I. Potassium and calcium changes in injured sword of large-dose steroids for spinal cord trauma. Ann Surg 1993;
spinal cords. Brain Res 1986;365:42–53. 218:419–425.
45. De Lay G, Laybaert L. Effect of flunarizine and methylpred- 67. Coates JR, Sorjonen DC, Simpson ST, et al. Clinicopathologic
nisolone on functional recovery after experimental spinal injury. J effects of a 21-aminosteroid compound (U74389G) and high-dose
Neurotrauma 1993;10:25–35. methylprednisolone on spinal cord function after simulated spinal cord
46. Pointillart V, Gense D, Gross C, et al. Effects of nimodipine on trauma. Vet Surg 1995;24:128–139.
posttraumatic spinal cord ischemia in baboons. J Neurotrauma 1993; 68. Siemering GB, Vomering ML. High dose methylprednisolone
10:201–213. sodium succinate: An adjunct to surgery for canine intervertebral disc
47. Faden AI, Jacobs TP, Smith MT. Evaluation of the calcium herniation. Vet Surg 1992;21:406, (abstract).
406 Olby

69. Hanson SM, Bostwick DR, Twedt DC, Smith MO. Clinical 91. Hall ED, Wolf DL, Althaus JS, Von Voigt-Lander PF. Beneficial
evaluation of cimetidine, sucralfate and misoprostol for prevention of effects of the kappa opioid receptor agonist U-50488H in experimental
gastrointestinal tract bleeding in dogs undergoing spinal surgery. Am acute brain and spinal cord injury. Brain Res 1987;435:174–180.
J Vet Res 1997;58:1320–1323. 92. Hayward NJ, McKnight AT, Woodruff GN. Neuroprotective ef-
70. Hall ED. Effects of the 21-aminosteroid U74006F on posttrau- fect of the k-agonist enadoline (CI-977) in rat models of focal cerebral
matic spinal cord ischemia in cats. J Neurosurg 1988;68:462–465. ischaemia. Eur J Neurosci 1993;5:961–967.
71. Anderson DK, Braughler JM, Hall ED, et al. Effects of treat- 93. Olby NJ, Jeffrey ND, Blakemore WF. Trial of the efficacy of
ment with U-74006F on neurological outcome following experimental the kappa opioid agonist, enadoline, at limiting the extent of tissue
spinal cord injury. J Neurosurg 1988;69:562–567. damage that develops following photochemically-induced injury. Exp
72. Holtz A, Gerdin B. Blocking weight-induced spinal cord injury Neurol 1999. Submitted.
in rats: Therapeutic effect of the 21-aminosteroid U74006F. J Neuro- 94. Lux WE, Feuerstein G, Faden AI. Alteration of leukotriene D4
trauma 1991;8:239–245. hypotension by thyrotropin-releasing hormone. Nature 1983;302:822–824.
73. Hoerlein BF, Redding RW, Hoff EJ, McGuire JA. Evaluation of 95. Lux WE, Feuerstein G, Snyder F, Faden AI. Effect of thyrotro-
dexamethasone, DMSO, mannitol, and solcoseryl in acute spinal cord pin-releasing hormone on hypotension produced by platelet-activating
trauma. J Am Anim Hosp Assoc 1983;19:216–225. factor. Circ Shock 1983;10:262.
74. Faden AI, Jacobs TP, Smith MT, Holaday JW. Comparison of 96. Renaud LP, Blumne HW, Pittman QJ, et al. Thyrotropin-releas-
thyrotropin-releasing hormone (TRH), naloxone, and dexamethasone ing hormone selectively depresses glutamate excitation of cerebral cor-
treatments in experimental spinal injury. Neurology 1983;33:673–678. tical neurons. Science 1979;205:1275–1276.
75. Zileli M, Ovul I, Dalbasti T. Effects of methyl prednisolone, 97. Faden AI, Jacobs TP, Smith MT. Thyrotropin-releasing hormone
dimethyl sulfoxide and naloxone in experimental spinal cord injury in in experimental spinal cord injury. Dose response and late treatment.
rats. Neurol Res 1988;10:232–235. Neurology 1984;34:1280–1284.
76. Parker AJ, Smith CW. Lack of functional recovery from spinal 98. Hashimoto T, Fukada N. Effect of thyrotropin-releasing hor-
cord trauma following dimethylsulfoxide and epsilon amino caproic mone on the neurologic impairment in rats with spinal cord injury:
acid therapy in dogs. Res Vet Sci 1979;27:253–255. Treatment starting 24 hours and 7 days after injury. Eur J Pharmacol
77. Kajihara K, Kawanga H, de la Torre J, Mullan S. Dimethyl 1991;23:25–32.
sulfoxide in the treatment of experimental spinal cord injury. Surg 99. Hoerlein BF, Redding RW, Hoff EJ, McGuire JA. Evaluation of
Neurol 1973;1:16–22. naloxone, crocetin, thyrotropin-releasing hormone, methylpredniso-
78. Kassell NF, Sprowell JA, Boarini DJ, Olin JJ. Effect of dimethyl lone, partial myelotomy and hemilaminectomy in the treatment of
sulfoxide on the cerebral and systemic circulations of the dog. Neu- acute spinal cord trauma. J Am Anim Hosp Assoc 1985;2:67–77.
rosurgery 1983;12:24–28. 100. Pitts LH, Ross A, Chase GA, Faden AI. Treatment with thy-
79. Choi DW. Calcium-mediated neurotoxicity: Relationship to spe- rotropin-releasing hormone (TRH) in patients with traumatic spinal
cific channel types and role in ischemic damage. Trends Neurosci cord injuries. J Neurotrauma 1995;12:235–243.
1988;11:465–469. 101. Fox GB, Faden AI. A novel cyclic dipeptide analog of a TRH
80. Choi DW, Maulucci-Gedde MA, Kriegstein AR. Glutamate neu- derivative exhibits neuroprotective and neurotropic properties follow-
rotoxicity in cortical cell culture. J Neurosci 1987;7:357–368. ing traumatic brain injury in the rodent. Soc Neurosci Abstr 1998;24:
81. Olby NJ, Sharp NDH, Munana KR, Papich MG. Chronic and 1728.
acute compressive spinal cord lesions in dogs are associated with in- 102. Rothwell NJ, Relton JK. Involvement of cytokines in acute
creased lumbar CSF glutamate levels. J Vet Intern Med 1999;13:240 neurodegeneration in the CNS. Neurosci Behav Rev 1993;17:217–227.
(abstract). 103. Perry VH, Andersson P-B, Gordon S. Macrophages and in-
82. Gomez-Pinella F, Tram H, Cotman CW, Nieto-Sampedro M. flammation in the central nervous system. Trends Neurosci 1993;16:
Neuroprotective effect of MK-801 and U-50488H after contusive spi- 268–272.
nal cord injury. Exp Neurol 1989;104:118–124. 104. Giulian D, Vaca K. Inflammatory glia mediate delayed neu-
83. Faden AI, Lemke M, Simon RP, Noble LJ. N-methyl-D-aspar- ronal damage after ischemia in the central nervous system. Stroke
tate antagonist MK801 improves outcome following traumatic spinal 1993;24:84–90.
cord injury in rats: Behavioural, anatomic, and neurochemical studies. 105. Gehrmann J, Matsumoto Y, Kreutzberg GW. Microglia: Intrin-
J Neurotrauma 1988;5:33–45. sic immunoeffector cell of the brain. Brain Res Rev 1995;20:269–287.
84. Pencalet P, Ohanna F, Poulat P, et al. Thienyl-phencyclidine pro- 106. Blight AR, Cohen TI, Saito K, Heyes MP. Quinolinic acid
tection for the spinal cord of adult rats against extension of lesions accumulation and functional deficits following experimental spinal
secondary to a photochemical injury. J Neurosurg 1993;78:603–609. cord injury. Brain 1995;118:735–752.
85. Wrathall JR, Choiniere D, Dong Teng Y. Dose-dependent re- 107. Dusart I, Schwab ME. Secondary cell death and the inflam-
duction of tissue loss and functional impairment after spinal cord trau- matory reaction after dorsal hemisection of the rat spinal cord. Eur J
ma with the AMPA/kainate antagonist NBQX. J Neurosci 1994;14: Neurosci 1994;6:712–724.
6598–6607. 108. Blight AR. Morphometric analysis of a model of spinal cord
86. Olney JW, Labruyere J, Price MT. Pathological changes induced injury in guinea pigs, with behavioural evidence of delayed secondary
in cerebrocortical neurons by phencyclidine and related drugs. Science pathology. J Neurol Sci 1991;103:156–171.
1989;244:1360–1362. 109. Olby NJ, Blakemore WF. Primary demyelination and regen-
87. Faden AI, Jacobs TP, Mougey E, Holaday JW. Endorphins in eration of ascending axons in the dorsal funiculus of the rat spinal
experimental spinal injury: Therapeutic effect of naloxone. Ann cord following photochemically-induced injury. J Neurocytol 1996;25:
Neurol 1981;10;326–332. 465–480.
88. Faden AI, Jacobs TP, Holaday JW. Opiate antagonist improves 110. Crowe MJ, Bresnahan JC, Shuman SL, et al. Apoptosis and
neurologic recovery after spinal injury. Science 1981;211:493–494. delayed degeneration after spinal cord injury in rats and monkeys. Nat
89. Faden AI. Opioid and nonopioid mechanisms may contribute to Med 1997;3:73–76.
dynorphin’s pathophysiological actions in spinal cord injury. Ann 111. Blight AR. Effects of silica on the outcome from experimental
Neurol 1990;27:67–74. spinal cord injury: Implication of macrophages in secondary tissue
90. Behrmann DL, Bresnahan JC, Beattie MS. A comparison of damage. Neuroscience 1994;60:263–273.
YM-14673, U-50488H, and nalmefene after spinal cord injury in the 112. Giulian D, Robertson C. Inhibition of mononuclear phagocytes
rat. Exp Neurol 1993;119:258–267. reduces ischaemic injury in the spinal cord. Ann Neurol 1990;27:33–42.
Acute Spinal Cord Injury 407

113. Taoka Y, Okajima K, Uchiba M, et al. Gabexate mesilate, a 119. Geisler FH, Dorsey FC, Coleman WP. Recovery of motor
synthetic protease inhibitor, prevents compression-induced spinal cord function after spinal-cord injury—A randomized, placebo-controlled
injury by inhibiting activation of leukocytes in rats. Crit Care Med trial with GM-1 ganglioside. N Engl J Med 1991;324:1829–1838.
1997;25:874–879. 120. Geisler FH, Dorsey FC, Paternello F, et al. Sygen (GM1-gan-
114. Taoka Y, Okajima K, Uchiba M, et al. Reduction of spinal glioside) acute spinal cord injury study. Congress of Neurosurgical
cord injury by administration of iloprost, a stable prostacyclin ana- Surgeons, Seattle, WA, October 8, 1998.
logue. J Neurosurg 1997;86:1007–1011. 121. Ginsberg MD, Sternau L, Globus MY-T, et al. Therapeutic
115. Relton JK, Rothwell NJ. Interleukin-1 receptor antagonist in- modulation of brain temperature: Relevance to ischemic brain injury.
hibits neuronal damage induced by cerebral ischaemia or NMDA-re- Cereb Blood Flow Metab 1992;4:189–225.
ceptor activation in the rat. Brain Res Bull 1992;585:135–160. 122. Martinez-Arizala A, Green B. Hypothermia in spinal cord in-
116. Bartholdi D, Schwab ME. Expression of pro-inflammatory cy- jury. J Neurotrauma 1992;9(Suppl 2):497–505.
tokine and chemokine mRNA upon experimental spinal cord injury in 123. Bullock R, Chestnut RM, Clifton G, et al. In Guidelines for
mouse: An in situ hybridization study. Eur J Neurosci 1997;9:1422– the Management of Severe Head Injury. New York, NY: Brain Trauma
1438. Foundation; 1997:5.
117. Guth L, Zhang Z, Roberts E. Key role for pregnolone in com- 124. Francel PC, Long BA, Malik JM, et al. Limiting ischemic
bination therapy that promotes recovery after spinal cord injury. Proc spinal cord injury using a free radical scavenger 21-aminosteroid and/
Natl Acad Sci USA 1994;91:12308–12312. or cerebrospinal fluid drainage. J Neurosurg 79:742–751.
118. Skaper SD, Leon A. Monosialogangliosides, neuroprotection 125. Griffiths IR, Pitts LH, Crawford RA, Trench JG. Spinal cord
and neuronal repair processes. J Neurotrauma 1992;9(Suppl 2):507– compression and blood flow. I. The effects of raised cerebrospinal fluid
516. pressure on spinal cord blood flow. Neurology 1978;28:1145–1151.

You might also like