Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Brain Inj ; 30(11): 1293-1301, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27712117

RESUMEN

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.


Asunto(s)
Biomarcadores/metabolismo , Lesiones Encefálicas , Modelos Animales de Enfermedad , Percusión , Animales , Síndrome de Behçet/etiología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/diagnóstico , Lesiones Encefálicas/etiología , Lesiones Encefálicas/terapia , Humanos , Morbilidad , Percusión/efectos adversos
2.
J Neurosurg ; 116(6): 1368-78, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22462511

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/genética , Lesiones Encefálicas/fisiopatología , Corteza Cerebral/lesiones , Corteza Cerebral/fisiopatología , Reparación del ADN/genética , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Destreza Motora/fisiología , Regeneración Nerviosa/genética , Equilibrio Postural/fisiología , Reflejo de Enderezamiento/fisiología , Proteína de la Xerodermia Pigmentosa del Grupo A/genética , Animales , Lesiones Encefálicas/patología , Muerte Celular/genética , Muerte Celular/fisiología , Corteza Cerebral/patología , Genotipo , Hipocampo/patología , Hipocampo/fisiopatología , Ratones , Ratones Noqueados , Ratones Transgénicos , Tálamo/patología , Tálamo/fisiopatología
3.
J Neurosci Res ; 88(15): 3414-23, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20890990

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Actividad Motora/fisiología , Neuronas/patología , Proteína p53 Supresora de Tumor/genética , Animales , Ratones , Ratones Noqueados , Recuperación de la Función
4.
Brain ; 133(11): 3232-42, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20736189

RESUMEN

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.


Asunto(s)
Conmoción Encefálica/diagnóstico , Conmoción Encefálica/metabolismo , Encefalopatías Metabólicas/diagnóstico , Encefalopatías Metabólicas/metabolismo , Lesiones Encefálicas/metabolismo , Espectroscopía de Resonancia Magnética , Recuperación de la Función/fisiología , Adolescente , Adulto , Conmoción Encefálica/fisiopatología , Encefalopatías Metabólicas/fisiopatología , Lesiones Encefálicas/diagnóstico , Lesiones Encefálicas/fisiopatología , Estudios de Cohortes , Femenino , Humanos , Masculino , Protones , Factores de Tiempo , Adulto Joven
5.
J Neurotrauma ; 27(7): 1297-309, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20486800

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/genética , Lesiones Encefálicas/metabolismo , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/genética , Eliminación de Gen , Proteínas de la Mielina/antagonistas & inhibidores , Proteínas de la Mielina/genética , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/genética , Animales , Lesiones Encefálicas/fisiopatología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Femenino , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/fisiología , Inyecciones Intraventriculares , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de la Mielina/fisiología , Receptor Nogo 1 , Receptores de Superficie Celular/fisiología
6.
Surg Neurol ; 71(5): 527-31; discussion 531, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18789503

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/fisiopatología , Eritropoyetina/farmacología , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/fisiopatología , Fármacos Neuroprotectores/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Lesiones Encefálicas/metabolismo , Citoprotección/efectos de los fármacos , Citoprotección/fisiología , Modelos Animales de Enfermedad , Eritropoyetina/uso terapéutico , Humanos , Degeneración Nerviosa/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/uso terapéutico
7.
Crit Care Med ; 37(2): 659-65, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19114897

RESUMEN

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.


Asunto(s)
Conducta Animal/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Lesiones Encefálicas/prevención & control , Proteína Inhibidora del Complemento C1/uso terapéutico , Animales , Lesiones Encefálicas/psicología , Proteína Inhibidora del Complemento C1/farmacología , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Estudios Prospectivos
8.
Restor Neurol Neurosci ; 26(1): 45-56, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18431005

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/complicaciones , Trastornos del Conocimiento , Hipocampo/patología , Neuronas/fisiología , Receptor trkB/fisiología , Transducción Genética/métodos , Análisis de Varianza , Animales , Lesiones Encefálicas/patología , Lesiones Encefálicas/terapia , Recuento de Células/métodos , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Trastornos del Conocimiento/terapia , Dependovirus/fisiología , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/biosíntesis , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Tiempo de Reacción , Receptor trkB/genética , Factores de Tiempo
9.
Eur J Trauma Emerg Surg ; 34(4): 397-409, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26815818

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-17937233

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Proteína GAP-43/metabolismo , Hipocampo/metabolismo , Proteínas de la Mielina/antagonistas & inhibidores , Animales , Conducta Animal , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Corteza Cerebral/lesiones , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Cognición , Regulación hacia Abajo/fisiología , Inmunoglobulina G/farmacología , Masculino , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/patología , Regeneración Nerviosa/efectos de los fármacos , Proteínas Nogo , Ratas , Ratas Sprague-Dawley
11.
Restor Neurol Neurosci ; 25(2): 77-90, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17726266

RESUMEN

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.


Asunto(s)
Anticonvulsivantes/farmacología , Conducta Animal/efectos de los fármacos , Edema Encefálico/patología , Lesiones Encefálicas/fisiopatología , Lesiones Encefálicas/psicología , Carbamatos/farmacología , Sistema Nervioso/fisiopatología , Animales , Edema Encefálico/etiología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/mortalidad , Muerte Celular/efectos de los fármacos , Cognición/efectos de los fármacos , Masculino , Aprendizaje por Laberinto , Actividad Motora/efectos de los fármacos , Sistema Nervioso/efectos de los fármacos , Desempeño Psicomotor , Ratas , Ratas Sprague-Dawley , Natación
12.
Eur J Neurosci ; 24(11): 3063-72, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17156367

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Enfermedades Desmielinizantes/tratamiento farmacológico , Glicoproteína Asociada a Mielina/antagonistas & inhibidores , Recuperación de la Función/efectos de los fármacos , Degeneración Walleriana/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Axones/efectos de los fármacos , Axones/inmunología , Axones/patología , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/fisiopatología , Lesiones Encefálicas/inmunología , Lesiones Encefálicas/fisiopatología , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Glicoproteína Asociada a Mielina/inmunología , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/inmunología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/inmunología , Resultado del Tratamiento , Degeneración Walleriana/inmunología , Degeneración Walleriana/fisiopatología
13.
J Neuropathol Exp Neurol ; 65(5): 478-88, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16772871

RESUMEN

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.


Asunto(s)
Traumatismos Cerrados de la Cabeza/patología , Núcleos Talámicos/patología , Adolescente , Adulto , Análisis de Varianza , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Recuento de Células , Distribución de Chi-Cuadrado , Estudios Transversales , Evaluación de la Discapacidad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Traumatismos Cerrados de la Cabeza/clasificación , Traumatismos Cerrados de la Cabeza/metabolismo , Humanos , Inmunohistoquímica/métodos , Antígeno de Macrófago-1/metabolismo , Masculino , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/patología , Núcleos Talámicos/metabolismo
14.
J Neurotrauma ; 23(6): 976-84, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16774481

RESUMEN

Traumatic brain injury (TBI) has been associated with intravascular coagulation, which may be a result of thromboplastin released following brain injury. Clots thus formed are lysed by plasmin, which is activated by tissue-type and urokinase-type plasminogen activators (uPA). To evaluate the association between traumatic intravascular coagulation and post-traumatic outcome, uPA knockout (uPA-/-) transgenic mice (n=12) or wild-type littermates (WT; n=12) were anesthetized and subjected to controlled cortical impact (CCI) brain injury. A second group of uPA-/- (n=12) and WT mice (n=12) were subjected to sham injury. Motor function was assessed over 2 weeks using the composite neuroscore test and cognition (learning) was assessed with the Morris Water Maze (MWM) at 2 weeks post-injury, whereupon the animals were sacrificed for cortical lesion volume analysis. Motor function was significantly worse in the brain-injured uPA-/- mice when compared to brain-injured WT mice at 48 h (p<0.05) and one week post-injury (p<0.05). These differences resolved by 2 weeks post-injury. There was no significant difference in post-injury cognitive function between uPA-/- mice and WT mice. However, at 2 weeks post-injury, the brain-injured uPA-/- had a significantly larger volume of cortical tissue loss than their WT counterparts (p<0.05). These results demonstrate that the absence of uPA in mice aggravates acute motor deficit and exacerbates cortical tissue loss following CCI brain injury, and suggests a neuroprotective role of the fibrinolytic process following TBI.


Asunto(s)
Lesiones Encefálicas/patología , Fibrinólisis/fisiología , Animales , Lesiones Encefálicas/psicología , Cognición/fisiología , Femenino , Lateralidad Funcional/fisiología , Inmunohistoquímica , Trombosis Intracraneal/patología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Activador de Plasminógeno de Tipo Uroquinasa/genética
15.
Restor Neurol Neurosci ; 24(2): 109-14, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16720946

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/fisiopatología , Cognición/fisiología , Aprendizaje por Laberinto/fisiología , Animales , Conducta Animal , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
16.
Curr Pharm Des ; 12(13): 1645-80, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16729876

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Animales , Antiinflamatorios/uso terapéutico , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Bloqueadores de los Canales de Calcio/uso terapéutico , Calpaína/antagonistas & inhibidores , Inhibidores de Caspasas , Antagonistas de Aminoácidos Excitadores/uso terapéutico , Humanos , Factores de Crecimiento Nervioso/uso terapéutico , Óxido Nítrico Sintasa/antagonistas & inhibidores , Especies de Nitrógeno Reactivo , Especies Reactivas de Oxígeno , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Transmisión Sináptica
17.
Eur J Neurosci ; 23(8): 2119-34, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16630059

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/cirugía , Diferenciación Celular/fisiología , Trasplante de Células/métodos , Trastornos del Conocimiento/cirugía , Factor Neurotrófico Derivado de la Línea Celular Glial/uso terapéutico , Neuronas/fisiología , Células Madre/fisiología , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/fisiopatología , Supervivencia Celular/fisiología , Células Cultivadas , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Factor Neurotrófico Derivado de la Línea Celular Glial/biosíntesis , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica/métodos , Masculino , Aprendizaje por Laberinto/fisiología , Actividad Motora/fisiología , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-ret/metabolismo , Desempeño Psicomotor/fisiología , Ratas , Tiempo de Reacción/fisiología , Prueba de Desempeño de Rotación con Aceleración Constante/métodos , Factores de Tiempo , Transducción Genética/métodos
18.
J Neurotrauma ; 23(2): 241-61, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16503807

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/complicaciones , Modelos Animales de Enfermedad , Epilepsia/etiología , Animales , Lesiones Encefálicas/fisiopatología , Epilepsia/fisiopatología , Humanos , Ratas
19.
Exp Neurol ; 198(2): 361-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16448651

RESUMEN

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.


Asunto(s)
Lesiones Encefálicas/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Convulsiones/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Anticonvulsivantes/farmacología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Diazepam/farmacología , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Activación Enzimática/fisiología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Ácido Kaínico , Masculino , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente
20.
J Neurotrauma ; 23(1): 86-96, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16430375

RESUMEN

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.


Asunto(s)
Acetilcisteína/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Corteza Cerebral/efectos de los fármacos , Proteínas del Tejido Nervioso/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Acetilcisteína/uso terapéutico , Proteínas Adaptadoras del Transporte Vesicular , Animales , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/fisiopatología , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Modelos Animales de Enfermedad , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/uso terapéutico , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatología , Masculino , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Proteínas del Tejido Nervioso/metabolismo , Neurotoxinas/metabolismo , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Proteínas SNARE/efectos de los fármacos , Proteínas SNARE/metabolismo , Membranas Sinápticas/efectos de los fármacos , Membranas Sinápticas/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...