Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Brain Inj ; 30(11): 1293-1301, 2016.
Article in English | MEDLINE | ID: mdl-27712117

ABSTRACT

BACKGROUND: After 30 years of characterisation and implementation, fluid percussion injury (FPI) is firmly recognised as one of the best-characterised reproducible and clinically relevant models of TBI, encompassing concussion through diffuse axonal injury (DAI). Depending on the specific injury parameters (e.g. injury site, mechanical force), FPI can model diffuse TBI with or without a focal component and may be designated as mild-to-severe according to the chosen mechanical forces and resulting acute neurological responses. Among FPI models, midline FPI may best represent clinical diffuse TBI, because of the acute behavioural deficits, the transition to late-onset behavioural morbidities and the absence of gross histopathology. REVIEW: The goal here was to review acute and chronic physiological and behavioural deficits and morbidities associated with diffuse TBI induced by midline FPI. In the absence of neurodegenerative sequelae associated with focal injury, there is a need for biomarkers in the diagnostic, prognostic, predictive and therapeutic approaches to evaluate outcomes from TBI. CONCLUSIONS: The current literature suggests that midline FPI offers a clinically-relevant, validated model of diffuse TBI to investigators wishing to evaluate novel therapeutic strategies in the treatment of TBI and the utility of biomarkers in the delivery of healthcare to patients with brain injury.


Subject(s)
Biomarkers/metabolism , Brain Injuries , Disease Models, Animal , Percussion , Animals , Behcet Syndrome/etiology , Brain Injuries/complications , Brain Injuries/diagnosis , Brain Injuries/etiology , Brain Injuries/therapy , Humans , Morbidity , Percussion/adverse effects
2.
J Neurosurg ; 116(6): 1368-78, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22462511

ABSTRACT

OBJECT: This study investigates the outcome after traumatic brain injury (TBI) in mice lacking the essential DNA repair gene xeroderma pigmentosum group A (XPA). As damage to DNA has been implicated in neuronal cell death in various models, the authors sought to elucidate whether the absence of an essential DNA repair factor would affect the outcome of TBI in an experimental setting. METHODS: Thirty-seven adult mice of either wild-type (n = 18) or XPA-deficient ("knock-out" [n = 19]) genotype were subjected to controlled cortical impact experimental brain trauma, which produced a focal brain injury. Sham-injured mice of both genotypes were used as controls (9 in each group). The mice were subjected to neurobehavoral tests evaluating learning/acquisition (Morris water maze) and motor dysfunction (Rotarod and composite neuroscore test), pre- and postinjury up to 4 weeks. The mice were killed after 1 or 4 weeks, and cortical lesion volume, as well as hippocampal and thalamic cell loss, was evaluated. Hippocampal staining with doublecortin antibody was used to evaluate neurogenesis after the insult. RESULTS: Brain-injured XPA(-/-) mice exhibited delayed recovery from impairment in neurological motor function, as well as pronounced cognitive dysfunction in a spatial learning task (Morris water maze), compared with injured XPA(+/+) mice (p < 0.05). No differences in cortical lesion volume, hippocampal damage, or thalamic cell loss were detected between XPA(+/+) and XPA(-/-) mice after brain injury. Also, no difference in the number of cells stained with doublecortin in the hippocampus was detected. CONCLUSIONS: The authors' results suggest that lack of the DNA repair factor XPA may delay neurobehavioral recovery after TBI, although they do not support the notion that this DNA repair deficiency results in increased cell or tissue death in the posttraumatic brain.


Subject(s)
Brain Injuries/genetics , Brain Injuries/physiopathology , Cerebral Cortex/injuries , Cerebral Cortex/physiopathology , DNA Repair/genetics , Maze Learning/physiology , Memory/physiology , Motor Skills/physiology , Nerve Regeneration/genetics , Postural Balance/physiology , Reflex, Righting/physiology , Xeroderma Pigmentosum Group A Protein/genetics , Animals , Brain Injuries/pathology , Cell Death/genetics , Cell Death/physiology , Cerebral Cortex/pathology , Genotype , Hippocampus/pathology , Hippocampus/physiopathology , Mice , Mice, Knockout , Mice, Transgenic , Thalamus/pathology , Thalamus/physiopathology
3.
J Neurosci Res ; 88(15): 3414-23, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20890990

ABSTRACT

Deletion of the tumor suppressor gene p53 has been shown to improve the outcome in experimental models of focal cerebral ischemia and kainate-induced seizures. To evaluate the potential role of p53 in traumatic brain injury, genetically modified mice lacking a functional p53 gene (p53(-/-), n = 9) and their wild-type littermates (p53(+/+), n = 9) were anesthetized and subjected to controlled cortical impact (CCI) experimental brain trauma. After brain injury, neuromotor function was assessed by using composite neuroscore and rotarod tests. By 7 days posttrauma, p53(-/-) mice exhibited significantly improved neuromotor function, in the composite neuroscore (P = 0.002) as well as in two of three individual tests, when compared with brain-injured p53(+/+) animals. CCI resulted in the formation of a cortical cavity (mean volume = 6.1 mm(3)) 7 days postinjury in p53(+/+) as well as p53(-/-) mice. No difference in lesion volume was detected between the two genotypes (P = 0.95). Although significant cell loss was detected in the ipsilateral hippocampus and thalamus of brain-injured animals, no differences between p53(+/+) and p53(-/-) mice were detected. Although our results suggest that lack of the p53 gene results in augmented recovery of neuromotor function following experimental brain trauma, they do not support a role for p53 acting as a mediator of neuronal death in this context, underscoring the complexity of its role in the injured brain.


Subject(s)
Brain Injuries/genetics , Brain Injuries/pathology , Motor Activity/physiology , Neurons/pathology , Tumor Suppressor Protein p53/genetics , Animals , Mice , Mice, Knockout , Recovery of Function
4.
Brain ; 133(11): 3232-42, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20736189

ABSTRACT

Concussive head injury opens a temporary window of brain vulnerability due to the impairment of cellular energetic metabolism. As experimentally demonstrated, a second mild injury occurring during this period can lead to severe brain damage, a condition clinically described as the second impact syndrome. To corroborate the validity of proton magnetic resonance spectroscopy in monitoring cerebral metabolic changes following mild traumatic brain injury, apart from the magnetic field strength (1.5 or 3.0 T) and mode of acquisition, we undertook a multicentre prospective study in which a cohort of 40 athletes suffering from concussion and a group of 30 control healthy subjects were admitted. Athletes (aged 16-35 years) were recruited and examined at three different institutions between September 2007 and June 2009. They underwent assessment of brain metabolism at 3, 15, 22 and 30 days post-injury through proton magnetic resonance spectroscopy for the determination of N-acetylaspartate, creatine and choline-containing compounds. Values of these representative brain metabolites were compared with those observed in the group of non-injured controls. Comparison of spectroscopic data, obtained in controls using different field strength and/or mode of acquisition, did not show any difference in the brain metabolite ratios. Athletes with concussion exhibited the most significant alteration of metabolite ratios at Day 3 post-injury (N-acetylaspartate/creatine: -17.6%, N-acetylaspartate/choline: -21.4%; P < 0.001 with respect to controls). On average, metabolic disturbance gradually recovered, initially in a slow fashion and, following Day 15, more rapidly. At 30 days post-injury, all athletes showed complete recovery, having metabolite ratios returned to values detected in controls. Athletes self-declared symptom clearance between 3 and 15 days after concussion. Results indicate that N-acetylaspartate determination by proton magnetic resonance spectroscopy represents a non-invasive tool to accurately measure changes in cerebral energy metabolism occurring in mild traumatic brain injury. In particular, this metabolic evaluation may significantly improve, along with other clinical assessments, the management of athletes suffering from concussion. Further studies to verify the effects of a second concussive event occurring at different time points of the recovery curve of brain metabolism are needed.


Subject(s)
Brain Concussion/diagnosis , Brain Concussion/metabolism , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/metabolism , Brain Injuries/metabolism , Magnetic Resonance Spectroscopy , Recovery of Function/physiology , Adolescent , Adult , Brain Concussion/physiopathology , Brain Diseases, Metabolic/physiopathology , Brain Injuries/diagnosis , Brain Injuries/physiopathology , Cohort Studies , Female , Humans , Male , Protons , Time Factors , Young Adult
5.
J Neurotrauma ; 27(7): 1297-309, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20486800

ABSTRACT

Functional recovery is markedly restricted following traumatic brain injury (TBI), partly due to myelin-associated inhibitors including Nogo-A, myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMgp), that all bind to the Nogo-66 receptor-1 (NgR1). In previous studies, pharmacological neutralization of both Nogo-A and MAG improved outcome following TBI in the rat, and neutralization of NgR1 improved outcome following spinal cord injury and stroke in rodent models. However, the behavioral and histological effects of NgR1 inhibition have not previously been evaluated in TBI. We hypothesized that NgR1 negatively influences behavioral recovery following TBI, and evaluated NgR1(-/-) mice (NgR1(-/-) study) and, in a separate study, soluble NgR1 infused intracerebroventricularly immediately post-injury to neutralize NgR1 (sNgR1 study) following TBI in mice using a controlled cortical impact (CCI) injury model. In both studies, motor function, TBI-induced loss of tissue, and hippocampal beta-amyloid immunohistochemistry were not altered up to 5 weeks post-injury. Surprisingly, cognitive function (as evaluated with the Morris water maze at 4 weeks post-injury) was significantly impaired both in NgR1(-/-) mice and in mice treated with soluble NgR1. In the sNgR1 study, we evaluated hippocampal mossy fiber sprouting using the Timm stain and found it to be increased at 5 weeks following TBI. Neutralization of NgR1 significantly increased mossy fiber sprouting in sham-injured animals, but not in brain-injured animals. Our data suggest a complex role for myelin-associated inhibitors in the behavioral recovery process following TBI, and urge caution when inhibiting NgR1 in the early post-injury period.


Subject(s)
Brain Injuries/genetics , Brain Injuries/metabolism , Cognition Disorders/chemically induced , Cognition Disorders/genetics , Gene Deletion , Myelin Proteins/antagonists & inhibitors , Myelin Proteins/genetics , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Animals , Brain Injuries/physiopathology , Cognition Disorders/physiopathology , Disease Models, Animal , Female , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/genetics , GPI-Linked Proteins/physiology , Injections, Intraventricular , Male , Maze Learning , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin Proteins/physiology , Nogo Receptor 1 , Receptors, Cell Surface/physiology
6.
Surg Neurol ; 71(5): 527-31; discussion 531, 2009 May.
Article in English | MEDLINE | ID: mdl-18789503

ABSTRACT

BACKGROUND: In the United States, TBI remains a major cause of morbidity and mortality in children and young adults. A total of 1.5 million Americans experience head trauma every year, and the yearly economic cost of this exceeds $56 billion. The magnitude of this problem has generated a great deal of interest in elucidating the complex molecular mechanism underlying cell death and dysfunction after TBI and in the development of neuroprotective agents that will reduce morbidity and mortality. METHODS: A review of recent literature on EPO, TBI, and apoptosis is conducted with analysis of pathophysiologic mechanisms of TBI. In addition, animal experiments and clinical trials pertaining to mechanisms of cell death in TBI and EPO as a neuroprotective agent are reviewed. CONCLUSION: The literature and evidence for EPO as a potent inhibitor of apoptosis and promising therapeutic agent in a variety of neurological insults, including trauma, are mounting. With the recent interest in clinical trials of EPO in human stroke, it is both timely and prudent to consider the use of this pharmaceutical avenue in TBI in man.


Subject(s)
Brain Injuries/drug therapy , Brain Injuries/physiopathology , Erythropoietin/pharmacology , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Neuroprotective Agents/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Brain Injuries/metabolism , Cytoprotection/drug effects , Cytoprotection/physiology , Disease Models, Animal , Erythropoietin/therapeutic use , Humans , Nerve Degeneration/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/therapeutic use
7.
Crit Care Med ; 37(2): 659-65, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19114897

ABSTRACT

OBJECTIVE: The aim of the study was to evaluate the effects of C1-inhibitor (C1-INH), an endogenous inhibitor of complement and kinin systems, on neurobehavioral and histological outcome following controlled cortical impact brain injury. DESIGN: Experimental prospective randomized study in mice. SETTING: Experimental laboratory. SUBJECTS: Male C57Bl/6 mice (n = 81). INTERVENTIONS: Mice were subjected to controlled cortical impact brain injury followed by an intravenous bolus of either C1-INH (15 U either at 10 minutes or 1 hour postinjury) or saline (equal volume, 150 microl at 10 minutes postinjury). Sham-operated mice received identical surgery and saline injection without brain injury. Neurological motor function was evaluated weekly for 4 weeks using the Composite Neuroscore. Cognitive function was evaluated at 4 weeks postinjury using the Morris Water Maze. Histological outcome was performed by measuring the contusion volume at 1 week and 4 weeks postinjury. MEASUREMENTS AND MAIN RESULTS: Brain-injured mice receiving C1-INH at 10 minutes postinjury showed attenuated motor deficits, cognitive dysfunction and reduced contusion volume compared to brain-injured mice receiving saline. Mice receiving C1-INH at 1 hour postinjury showed reduced motor deficits compared to brain-injured mice receiving saline, but no significantly different cognitive and histological outcome. Immunohistochemical analysis showed that 20 minutes after infusion, C1-INH was localised on endothelial cells and in brain tissue surrounding brain capillaries of the injured hemisphere. CONCLUSION: Our results show that post-traumatic administration of C1-INH attenuates neuro-behavioral deficits and histological damage associated with traumatic brain injury.


Subject(s)
Behavior, Animal/drug effects , Brain Injuries/drug therapy , Brain Injuries/prevention & control , Complement C1 Inhibitor Protein/therapeutic use , Animals , Brain Injuries/psychology , Complement C1 Inhibitor Protein/pharmacology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Prospective Studies
8.
Restor Neurol Neurosci ; 26(1): 45-56, 2008.
Article in English | MEDLINE | ID: mdl-18431005

ABSTRACT

PURPOSE: The ability of brain-derived neurotrophic factor (BDNF) to attenuate secondary damage and influence behavioral outcome after experimental traumatic brain injury (TBI) remains controversial. Because TBI can result in decreased expression of the trkB receptor, thereby preventing BDNF from exerting potential neuroprotective effects, the contribution of both BDNF and its receptor trkB to hippocampal neuronal loss and cognitive dysfunction were evaluated. METHODS: Full-length trkB was overexpressed in the left hippocampus of adult C57Bl/6 mice using recombinant adeno-associated virus serotype 2/5 (rAAV 2/5). EGFP (enhanced green fluorescent protein) expression was present at two weeks after AAV-EGFP injection and remained sustained up to four weeks after the injection. At 2 weeks following gene transduction, mice were subjected to parasagittal controlled cortical impact (CCI) brain injury, followed by either BDNF or PBS infusion into the hippocampus. RESULTS: No differences were observed in learning ability at two weeks post-injury or in motor function from 48 hours to two weeks among treatment groups. The number of surviving pyramidal neurons in the CA2-CA3 region of the hippocampus was also not different among treatment groups. CONCLUSIONS: These data suggest that neither overexpression of trkB, BNDF infusion or their combination affects neuronal survival or behavioral outcome following experimental TBI in mice.


Subject(s)
Brain Injuries/complications , Cognition Disorders , Hippocampus/pathology , Neurons/physiology , Receptor, trkB/physiology , Transduction, Genetic/methods , Analysis of Variance , Animals , Brain Injuries/pathology , Brain Injuries/therapy , Cell Count/methods , Cognition Disorders/etiology , Cognition Disorders/pathology , Cognition Disorders/therapy , Dependovirus/physiology , Disease Models, Animal , Green Fluorescent Proteins/biosynthesis , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Motor Activity/physiology , Reaction Time , Receptor, trkB/genetics , Time Factors
9.
Eur J Trauma Emerg Surg ; 34(4): 397-409, 2008 Aug.
Article in English | MEDLINE | ID: mdl-26815818

ABSTRACT

Effective methods for treating cerebral edema have recently become a matter of both extensive research and significant debate within the neurosurgery and trauma surgery communities. The pathophysiologic progression and outcome of different forms of cerebral edema associated with traumatic brain injury have yet to be fully elucidated. There are heterogeneous factors influencing the onset and progress of post-traumatic cerebral edema, including the magnitude and type of head injury, age, co-morbid conditions of the patient, the critical window for therapeutic intervention and the presence of secondary insults including hypoxia, hypotension, hypo/hyperthermia, degree of raised intracranial pressure (ICP), and disruption of blood brain barrier (BBB) integrity. Although numerous studies have been designed to improve our understanding of the etiology of post-traumatic cerebral edema, therapeutic interventions have traditionally been focused on minimizing secondary insults especially raised ICP and improving cerebral perfusion pressure. More recently, fluid resuscitation strategies using hyperosmolar agents such as pentastarch and hypertonic saline (HS) have achieved some success. HS treatment is of particular interest due to its apparent advantageous action over other types of hyper-osmotic solutions in both clinical and laboratory studies. In this review, we provide a summary of recent literature concerning the pathogenesis and mechanisms involved in the various types of cerebral edema, and the possible mechanisms of action of HS for the treatment cerebral edema.

10.
J Neurosurg ; 107(4): 844-53, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17937233

ABSTRACT

OBJECT: Central nervous system axons regenerate poorly after traumatic brain injury (TBI), partly due to inhibitors such as the protein Nogo-A present in myelin. The authors evaluated the efficacy of anti-Nogo-A monoclonal antibody (mAb) 7B12 administration on the neurobehavioral and cognitive outcome of rats following lateral fluid-percussion brain injury, characterized the penetration of the 7B12 or control antibodies into target brain regions, and evaluated the effects of Nogo-A inhibition on hemispheric tissue loss and sprouting of uninjured motor tracts in the cervical cord. To elucidate a potential molecular response to Nogo-A inhibition, we evaluated the effects of 7B12 on hippocampal GAP-43 expression. METHODS: Beginning 24 hours after lateral fluid-percussion brain injury or sham injury in rats, the mAb 7B12 or control antibody was infused intracerebroventricularly over 14 days, and behavior was assessed over 4 weeks. RESULTS: Immunoreactivity for 7B12 or immunoglobulin G was detected in widespread brain regions at 1 and 3 weeks postinjury. The brain-injured animals treated with 7B12 showed improvement in cognitive function (p < 0.05) at 4 weeks but no improvement in neurological motor function from 1 to 4 weeks postinjury compared with brain-injured, vehicle-treated controls. The enhanced cognitive function following inhibition of Nogo-A was correlated with an attenuated postinjury downregulation of hippocampal GAP-43 expression (p < 0.05). CONCLUSIONS: Increased GAP-43 expression may be a novel molecular mechanism of the enhanced cognitive recovery mediated by Nogo-A inhibition after TBI in rats.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Injuries/drug therapy , GAP-43 Protein/metabolism , Hippocampus/metabolism , Myelin Proteins/antagonists & inhibitors , Animals , Behavior, Animal , Brain Injuries/metabolism , Brain Injuries/pathology , Cerebral Cortex/injuries , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cognition , Down-Regulation/physiology , Immunoglobulin G/pharmacology , Male , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Nerve Regeneration/drug effects , Nogo Proteins , Rats , Rats, Sprague-Dawley
11.
Restor Neurol Neurosci ; 25(2): 77-90, 2007.
Article in English | MEDLINE | ID: mdl-17726266

ABSTRACT

PURPOSE: To evaluate the therapeutic efficacy of two antiepileptic compounds, RWJ-333369 and RWJ-333369-A in a well-established experimental model of lateral fluid percussion (FP) traumatic brain injury (TBI) in the rat. METHODS: Anethestized Male Sprague-Dawley rats (n=227) were subjected to lateral FP brain injury or sham-injury. Animals were randomized to receive treatment with RWJ-333369 (60 mg/kg, p.o.) or its analog RWJ-333369-A (60 mg/kg, p.o.), or vehicle (equal volume) at 15 minutes, 4, 8, and 24 hours post-injury. In Study I, animals were assessed at 48 hours for acute motor and cognitive function and then sacrificed to evaluate regional cerebral edema. In Study II, animals were evaluated post-injury for motor function at 48 hours and weekly thereafter from 1 to 4 weeks. Post-traumatic learning ability was assessed 4 weeks post-injury, followed by evaluation of hemispheric tissue loss. RESULTS: In Study I, no improvement in acute memory or motor function was observed following administration of either RWJ-333369 or RWJ-333369-A in brain-injured animals compared to vehicle-treated, brain-injured animals. However, brain-injured animals receiving treatment with RWJ-333369-A had a significant reduction in post-traumatic cerebral edema in both injured and contralateral hippocampus compared to brain-injured, vehicle-treated controls (p<0.05). In Study II, treatment with either compound did not result in any improvement of neuromotor function, learning ability or change in lesion volume following brain injury. CONCLUSION: These results indicate that the novel antiepileptic compound RWJ-333369-A reduces post-traumatic hippocampal edema without affecting neurobehavioral or histological outcome. It remains unclear whether this small effect on hippocampal edema ie related to the ability of this compound to attenuate seizure activity.


Subject(s)
Anticonvulsants/pharmacology , Behavior, Animal/drug effects , Brain Edema/pathology , Brain Injuries/physiopathology , Brain Injuries/psychology , Carbamates/pharmacology , Nervous System/physiopathology , Animals , Brain Edema/etiology , Brain Injuries/complications , Brain Injuries/mortality , Cell Death/drug effects , Cognition/drug effects , Male , Maze Learning , Motor Activity/drug effects , Nervous System/drug effects , Psychomotor Performance , Rats , Rats, Sprague-Dawley , Swimming
12.
Eur J Neurosci ; 24(11): 3063-72, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17156367

ABSTRACT

Axonal injury is a hallmark of traumatic brain injury (TBI) and is associated with a poor clinical outcome. Following central nervous system injury, axons regenerate poorly, in part due to the presence of molecules associated with myelin that inhibit axonal outgrowth, including myelin-associated glycoprotein (MAG). The involvement of MAG in neurobehavioral deficits and tissue loss following experimental TBI remains unexplored and was evaluated in the current study using an MAG-specific monoclonal antibody (mAb). Anesthetized rats (n=102) were subjected to either lateral fluid percussion brain injury (n=59) or sham injury (n=43). In surviving animals, beginning at 1 h post-injury, 8.64 microg anti-MAG mAb (n=33 injured, n=21 sham) or control IgG (n=26 injured, n=22 sham) was infused intracerebroventricularly for 72 h. One group of these rats (n=14 sham, n=11 injured) was killed at 72 h post-injury for verification of drug diffusion and MAG immunohistochemistry. All other animals were evaluated up to 8 weeks post-injury using tests for neurologic motor, sensory and cognitive function. Hemispheric tissue loss was also evaluated at 8 weeks post-injury. At 72 h post-injury, increased immunoreactivity for MAG was seen in the ipsilateral cortex, thalamus and hippocampus of brain-injured animals, and anti-MAG mAb was detectable in the hippocampus, fimbria and ventricles. Brain-injured animals receiving anti-MAG mAb showed significantly improved recovery of sensorimotor function at 6 and 8 weeks (P<0.01) post-injury when compared with brain-injured IgG-treated animals. Additionally, at 8 weeks post-injury, the anti-MAG mAb-treated brain-injured animals demonstrated significantly improved cognitive function and reduced hemispheric tissue loss (P<0.05) when compared with their brain-injured controls. These results indicate that MAG may contribute to the pathophysiology of experimental TBI and treatment strategies that target MAG may be suitable for further evaluation.


Subject(s)
Antibodies, Monoclonal/pharmacology , Brain Injuries/drug therapy , Demyelinating Diseases/drug therapy , Myelin-Associated Glycoprotein/antagonists & inhibitors , Recovery of Function/drug effects , Wallerian Degeneration/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Axons/drug effects , Axons/immunology , Axons/pathology , Brain/drug effects , Brain/immunology , Brain/physiopathology , Brain Injuries/immunology , Brain Injuries/physiopathology , Demyelinating Diseases/immunology , Demyelinating Diseases/physiopathology , Disease Models, Animal , Immunohistochemistry , Male , Myelin-Associated Glycoprotein/immunology , Nerve Regeneration/drug effects , Nerve Regeneration/immunology , Rats , Rats, Sprague-Dawley , Recovery of Function/immunology , Treatment Outcome , Wallerian Degeneration/immunology , Wallerian Degeneration/physiopathology
13.
J Neuropathol Exp Neurol ; 65(5): 478-88, 2006 May.
Article in English | MEDLINE | ID: mdl-16772871

ABSTRACT

Paraffin-embedded blocks from the thalamus of 9 control patients, 9 moderately disabled, 12 severely disabled, and 10 vegetative head-injured patients assessed using the Glasgow Outcome Scale and identified from the Department of Neuropathology archive. Neurons, astrocytes, macrophages, and activated microglia were differentiated by Luxol fast blue/cresyl violet, GFAP, CD68, and CR3/43 staining and stereological techniques used to estimate cell number in a 28-microm-thick coronal section. Counts were made in subnuclei of the mediodorsal, lateral posterior, and ventral posterior nuclei, the intralaminar nuclei, and the related internal lamina. Neuronal loss occurred from mediodorsal parvocellularis, rostral center medial, central lateral and paracentral nuclei in moderately disabled patients; and from mediodorsal magnocellularis, caudal center medial, rhomboid, and parafascicular nuclei in severely disabled patients; and all of the above and the centre median nucleus in vegetative patients. Neuronal loss occurred primarily from cognitive and executive function nuclei, a lesser loss from somatosensory nuclei and the least loss from limbic motor nuclei. There was an increase in the number of reactive astrocytes, activated microglia, and macrophages with increasing severity of injury. The study provides novel quantitative evidence for differential neuronal loss, with survival after human head injury, from thalamic nuclei associated with different aspects of cortical activation.


Subject(s)
Head Injuries, Closed/pathology , Thalamic Nuclei/pathology , Adolescent , Adult , Analysis of Variance , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Count , Chi-Square Distribution , Cross-Sectional Studies , Disability Evaluation , Female , Glial Fibrillary Acidic Protein/metabolism , Head Injuries, Closed/classification , Head Injuries, Closed/metabolism , Humans , Immunohistochemistry/methods , Macrophage-1 Antigen/metabolism , Male , Middle Aged , Neurons/metabolism , Neurons/pathology , Thalamic Nuclei/metabolism
14.
Curr Pharm Des ; 12(13): 1645-80, 2006.
Article in English | MEDLINE | ID: mdl-16729876

ABSTRACT

Traumatic brain injury (TBI) is a devastating disease, predominately affecting young people. Although the prognosis for TBI victims has improved in recent years, many survivors of TBI suffer from emotional, cognitive and motor disturbances and a decreased quality of life. In recent years, there has been a rapid increase in the number of pharmacological targets evaluated in clinically-relevant experimental TBI models, showing improved cognitive and motor outcome and decreased loss of brain tissue. Despite the completion of several recent clinical trials using compounds showing neuroprotection in preclinical studies, pharmaceutical treatment strategies with proven clinical benefit are still lacking. This paper reviews the preclinical pharmacological treatment studies evaluated to date in experimental models of TBI. Although human TBI is a complex and multifaceted disease, these studies provide encouraging translational data suggesting that pharmacological compounds, delivered in a clinically-relevant time window, may improve the outcome of TBI patients.


Subject(s)
Brain Injuries/drug therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Brain Injuries/metabolism , Brain Injuries/physiopathology , Calcium Channel Blockers/therapeutic use , Calpain/antagonists & inhibitors , Caspase Inhibitors , Excitatory Amino Acid Antagonists/therapeutic use , Humans , Nerve Growth Factors/therapeutic use , Nitric Oxide Synthase/antagonists & inhibitors , Reactive Nitrogen Species , Reactive Oxygen Species , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Synaptic Transmission
15.
Restor Neurol Neurosci ; 24(2): 109-14, 2006.
Article in English | MEDLINE | ID: mdl-16720946

ABSTRACT

PURPOSE: As deficits in memory and cognition are commonly observed in survivors of traumatic brain injury (TBI), causing reduced quality of life for the patient, a major goal in experimental TBI research is to identify and evaluate cognitive dysfunction. The present study assessed the applicability of the serial Morris water maze (MWM) test to determine cognitive function following experimental TBI in the same group of rats which is particularly important for long-term studies and increasingly valuable for the evaluation of novel treatment strategies. METHODS: Male Sprague-Dawley rats (n = 27) were anesthetized and subjected to either sham injury (n = 9) or lateral fluid percussion (FP) brain injury of moderate severity (n = 18). At 4 weeks post-injury, animals were trained in a water maze over 3 days (acquisition/learning phase) to find a submerged platform. At 8 weeks post-injury the hidden platform was then moved to the opposite quadrant, and animals were trained to find the new position of the platform over 3 days. Forty-eight hours later, animals were tested for memory retention in a probe trial in which the platform was not present. RESULTS: Brain-injured animals had significant learning impairment (p < 0.0001), shifted-learning impairment (p < 0.001) and memory retention deficits (p < 0.01) in comparison to their sham-injured counterparts over the 8 week testing period. Swim speed and distance were not significantly altered by brain injury at any time point. CONCLUSION: The validation of this testing paradigm using a clinically relevant experimental brain injury model is an important addition to behavioral outcome testing.


Subject(s)
Brain Injuries/physiopathology , Cognition/physiology , Maze Learning/physiology , Animals , Behavior, Animal , Disease Models, Animal , Male , Rats , Rats, Sprague-Dawley , Time Factors
16.
Eur J Neurosci ; 23(8): 2119-34, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16630059

ABSTRACT

We sought to evaluate the potential of C17.2 neural progenitor cells (NPCs) engineered to secrete glial cell line-derived neurotrophic factor (GDNF) to survive, differentiate and promote functional recovery following engraftment into the brains of adult male Sprague-Dawley rats subjected to lateral fluid percussion brain injury. First, we demonstrated continued cortical expression of GDNF receptor components (GFRalpha-1, c-Ret), suggesting that GDNF could have a physiological effect in the immediate post-traumatic period. Second, we demonstrated that GDNF over-expression reduced apoptotic NPC death in vitro. Finally, we demonstrated that GDNF over-expression improved survival, promoted neuronal differentiation of GDNF-NPCs at 6 weeks, as compared with untransduced (MT) C17.2 cells, following transplantation into the perilesional cortex of rats at 24 h post-injury, and that brain-injured animals receiving GDNF-C17.2 transplants showed improved learning compared with those receiving vehicle or MT-C17.2 cells. Our results suggest that transplantation of GDNF-expressing NPCs in the acute post-traumatic period promotes graft survival, migration, neuronal differentiation and improves cognitive outcome following traumatic brain injury.


Subject(s)
Brain Injuries/surgery , Cell Differentiation/physiology , Cell Transplantation/methods , Cognition Disorders/surgery , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Neurons/physiology , Stem Cells/physiology , Animals , Brain Injuries/complications , Brain Injuries/physiopathology , Cell Survival/physiology , Cells, Cultured , Cognition Disorders/etiology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , Male , Maze Learning/physiology , Motor Activity/physiology , Neurons/drug effects , Proto-Oncogene Proteins c-ret/metabolism , Psychomotor Performance/physiology , Rats , Reaction Time/physiology , Rotarod Performance Test/methods , Time Factors , Transduction, Genetic/methods
17.
J Neurotrauma ; 23(2): 241-61, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16503807

ABSTRACT

Epilepsy is a major unfavorable long-term consequence of traumatic brain injury (TBI). Moreover, TBI is one of the most important predisposing factors for the development of epilepsy, particularly in young adults. Understanding the molecular and cellular cascades that lead to the development of post-traumatic epilepsy (PTE) is key for preventing its development or modifying the disease process in such a way that epilepsy, if it develops, is milder and easier-to-treat. Tissue from TBI patients undergoing epileptogenesis is not available for such studies, which underscores the importance of developing clinically relevant animal models of PTE. The goal of this review is to (1) provide a description of PTE in humans, which is critical for the development of clinically relevant models of PTE, (2) review the characteristics of currently available PTE models, and (3) provide suggestions for the development of future models of PTE based on our current understanding of the mechanisms of TBI and epilepsy. The development of clinically relevant models of PTE is critical to advance our understanding of the mechanisms of post-traumatic epileptogenesis and epilepsy, as well as for producing breakthroughs in the development and testing of novel antiepileptogenic treatments.


Subject(s)
Brain Injuries/complications , Disease Models, Animal , Epilepsy/etiology , Animals , Brain Injuries/physiopathology , Epilepsy/physiopathology , Humans , Rats
18.
Exp Neurol ; 198(2): 361-9, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16448651

ABSTRACT

Traumatic brain injury (TBI) is characterized by a progressive cell loss and a lack of axonal regeneration. In the central nervous system (CNS), the Rho signaling pathway regulates the neuronal response to growth inhibitory proteins and regeneration of damaged axons, and Rho activation is also correlated with an increased susceptibility to apoptosis. To evaluate whether traumatic brain injury (TBI) results in changes in Rho activation in vulnerable regions of the brain, GTP-RhoA pull down assays were performed on rat cortical and hippocampal tissue homogenates obtained from 24 h to 3 days following lateral fluid percussion brain injury (FPI). Following FPI, a significantly increased RhoA activation was observed from 24 h to 3 days post-injury in the cortex and by 3 days in the hippocampus ipsilateral to the injury. We also detected activated RhoA in the cortex and hippocampus contralateral to the injury, without concomitant changes in total RhoA levels. To determine if immediate post-traumatic events such as seizures may activate Rho, we examined RhoA activation in the brains of rats with kainic acid-induced seizures. Severe seizures resulted in bilateral RhoA activation in the cortex and hippocampus. Together, these results indicate that RhoA is activated in vulnerable brain regions following traumatic and epileptic insults to the CNS.


Subject(s)
Brain Injuries/metabolism , Gene Expression Regulation, Enzymologic/physiology , Seizures/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Anticonvulsants/pharmacology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Diazepam/pharmacology , Disease Models, Animal , Drug Interactions , Enzyme Activation/physiology , Gene Expression Regulation, Enzymologic/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Kainic Acid , Male , Rats , Rats, Sprague-Dawley , Seizures/chemically induced
19.
J Neurotrauma ; 23(1): 86-96, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16430375

ABSTRACT

Alteration of excitatory neurotransmission is a key feature of traumatic brain injury (TBI) in which extracellular glutamate levels rise. Although increased synaptic release of glutamate occurs at the injury site, the precise mechanism is unclear. Complexin I and complexin II constitute a family of cytosolic proteins involved in the regulation of neurotransmitter release, competing with the chaperone protein alpha-SNAP (soluble N-ethylmaleimide-sensitive factor-attachment protein) for binding to the synaptic vesicle protein synaptobrevin as well as the synaptic membrane proteins SNAP-25 and syntaxin, which together form the SNAP receptor (SNARE) complex. Complexin I is predominantly a marker of axosomatic (inhibitory) synapses, whereas complexin II mainly labels axodendritic and axospinous synapses, the majority of which are excitatory. In order to examine the role of these proteins in TBI, we have studied levels of both complexins in the injured hemisphere by immunoblotting over a time period ranging from 6 h to 7 days following lateral fluid-percussion brain injury in the rat. Transient increases in the levels of complexin I and complexin II proteins were detected in the injured cerebral cortex 6 h following TBI. This increase was followed by a decrease of complexin I in the injured cortex and hippocampus, and a decrease in both complexins in the injured thalamus region at day 3 and day 7 post-injury. The early, transient increase in the injured cortex was completely blocked by N-acetylcysteine (NAC) administered 5 min following trauma, suggesting an involvement of oxidative stress. Neuronal loss was also reduced in the injured hemisphere with post-TBI NAC treatment. Our findings suggest a dysregulation of both inhibitory and excitatory neurotransmission following traumatic injury that is responsive to antioxidant treatment. These alterations in complexin levels may also play an important role in neuronal cell loss following TBI, and thus contribute to the pathophysiology of cerebral damage following brain injury.


Subject(s)
Acetylcysteine/pharmacology , Brain Injuries/drug therapy , Cerebral Cortex/drug effects , Nerve Tissue Proteins/drug effects , Oxidative Stress/drug effects , Acetylcysteine/therapeutic use , Adaptor Proteins, Vesicular Transport , Animals , Brain Injuries/metabolism , Brain Injuries/physiopathology , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Disease Models, Animal , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/metabolism , Neurotoxins/metabolism , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , SNARE Proteins/drug effects , SNARE Proteins/metabolism , Synaptic Membranes/drug effects , Synaptic Membranes/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Time Factors , Up-Regulation/drug effects , Up-Regulation/physiology
20.
Crit Care Med ; 34(2): 492-501, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16424733

ABSTRACT

OBJECTIVES: Posttraumatic hypotension is believed to increase morbidity and mortality in traumatically brain-injured patients. Using a clinically relevant model of combined traumatic brain injury with superimposed hemorrhagic hypotension in rats, the present study evaluated whether a reduction in mean arterial blood pressure aggravates regional brain edema formation, regional cell death, and neurologic motor/cognitive deficits associated with traumatic brain injury. DESIGN: Experimental prospective, randomized study in rodents. SETTING: Experimental laboratory at a university hospital. SUBJECTS: One hundred nineteen male Sprague-Dawley rats weighing 350-385 g. INTERVENTIONS: Experimental traumatic brain injury of mild to moderate severity was induced using the lateral fluid percussion brain injury model in anesthetized rats (n = 89). Following traumatic brain injury, in surviving animals one group of animals was subjected to pressure-controlled hemorrhagic hypotension, maintaining the mean arterial blood pressure at 50-60 mm Hg for 30 mins (n = 47). The animals were subsequently either resuscitated with lactated Ringer's solution (three times shed blood volume, n = 18) or left uncompensated (n = 29). Other groups of animals included those with isolated traumatic brain injury (n = 34), those with isolated hemorrhagic hypotension (n = 8), and sham-injured control animals receiving anesthesia and surgery alone (n = 22). MEASUREMENTS AND MAIN RESULTS: The withdrawal of 6-7 mL of arterial blood significantly reduced mean arterial blood pressure by 50% without decreasing arterial oxygen saturation or Pao2. Brain injury induced significant cerebral edema (p < .001) in vulnerable brain regions and cortical tissue loss (p < .01) compared with sham-injured animals. Neither regional brain edema formation at 24 hrs postinjury nor the extent of cortical tissue loss assessed at 7 days postinjury was significantly aggravated by superimposed hemorrhagic hypotension. Brain injury-induced neurologic deficits persisted up to 20 wks after injury and were also not aggravated by the hemorrhagic hypotension. Cognitive dysfunction persisted for up to 16 wks postinjury. The superimposition of hemorrhagic hypotension significantly delayed the time course of cognitive recovery. CONCLUSIONS: A single, acute hypotensive event lasting 30 mins did not aggravate the short- and long-term structural and motor deficits but delayed the speed of recovery of cognitive function associated with experimental traumatic brain injury.


Subject(s)
Brain Injuries/complications , Cognition Disorders/etiology , Fluid Therapy , Hypotension/complications , Shock/complications , Animals , Blood Gas Analysis , Blood Pressure , Hypotension/therapy , Male , Psychomotor Performance , Rats , Rats, Sprague-Dawley , Shock/therapy , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL