Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Neurotrauma ; 37(2): 236-247, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31530242

RESUMEN

Traumatic brain injury (TBI) carries a risk of developing post-traumatic epilepsy (PTE). Currently, animal models that replicate clinical PTE (delayed spontaneous and recurrent seizures) are limited, which hinders pre-clinical research. In this study, we used two rat models of penetrating ballistic-like brain injury (PBBI) and closed-head injury (CHI) to induce spontaneous seizures and also measure changes in seizure susceptibility. In the PBBI model, two trajectories (frontal and lateral) and two injury severities for each trajectory, were evaluated. In the CHI model, a single projectile impact to the dorsal/lateral region of the head was tested. Continuous video-electroencephalographic (EEG) recordings were collected for 10 days at 1 or 6 month(s) post-injury. After EEG recording, all rats were given a sub-convulsant dose of pentylenetetrazole (PTZ) to challenge the seizure susceptibility. The video-EEG recording did not detect PTE following the PBBI. Only one CHI rat demonstrated persistent and recurrent non-convulsive seizures detected at 6 months post-injury. However, after PTZ challenge, 50-100% of the animals across different TBI groups experienced seizures. Seizure susceptibility increased over time from 1 to 6 months post-injury across the majority of TBI groups. Injury severity effects were not apparent within the PBBI model, but were evident between PBBI and CHI models. These results demonstrated the difficulties in detecting delayed spontaneous post-traumatic seizures even in a high-risk model of penetrating brain injury. The PTZ-induced increase in seizure susceptibility indicated the existence of vulnerable risk of epileptogenesis following TBI, which may be considered as an alternative research tool for pre-clinical studies of PTE.


Asunto(s)
Lesiones Traumáticas del Encéfalo/etiología , Modelos Animales de Enfermedad , Epilepsia Postraumática/etiología , Traumatismos Cerrados de la Cabeza/complicaciones , Traumatismos Penetrantes de la Cabeza/complicaciones , Animales , Masculino , Ratas , Ratas Sprague-Dawley , Convulsiones/etiología
2.
J Trauma Acute Care Surg ; 83(1 Suppl 1): S25-S34, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28452872

RESUMEN

BACKGROUND: Posttraumatic seizures are a medical problem affecting patients with traumatic brain injury. Yet effective treatment is lacking owing to the limitations of antiepileptic drugs (AEDs) applicable to these patients. METHODS: In this study, we evaluated the dose-response efficacy of levetiracetam (12.5-100.0 mg/kg) and gabapentin (1.25-25.0 mg/kg) administered either individually or in pairs at fixed-dose ratios as a combination in mitigating posttraumatic nonconvulsive seizures induced by severe penetrating ballistic-like brain injury (PBBI) in rats. Seizures were detected by continuous electroencephalogram (EEG) monitoring for 72 hours postinjury. Animals were treated twice per day for 3 days by intravenous injections. RESULTS: Both levetiracetam (25-100 mg/kg) and gabapentin (6.25-25 mg/kg) significantly reduced PBBI-induced seizure frequency by 44% to 73% and 61% to 69%, and seizure duration by 45% to 64% and 70% to 78%, respectively. However, the two drugs manifested different dose-response profiles. Levetiracetam attenuated seizure activity in a dose-dependent fashion, whereas the beneficial effects of gabapentin plateaued across the three highest doses tested. Combined administration of levetiracetam and gabapentin mirrored the more classic dose-response profile of levetiracetam monotherapy. However, no additional benefit was derived from the addition of gabapentin. Furthermore, isobolographic analysis of the combination dose-response profile of levetiracetam and gabapentin failed to reach the expected level of additivity, suggesting an unlikelihood of favorable interactions between these two drugs against spontaneously occurring posttraumatic seizure activities at the particular set of dose ratios tested. CONCLUSION: This study was the first attempt to apply isobolographic approach to studying AED combination therapy in the context of spontaneously occurring posttraumatic seizures. Despite the failure to achieve additivity from levetiracetam and gabapentin combination, it is important to recognize the objectivity of the isobolographic approach in the evaluation of AED combination therapy against seizures directly associated with brain injuries.


Asunto(s)
Aminas/farmacología , Ácidos Ciclohexanocarboxílicos/farmacología , Traumatismos Penetrantes de la Cabeza/complicaciones , Piracetam/análogos & derivados , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Ácido gamma-Aminobutírico/farmacología , Animales , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Electroencefalografía , Gabapentina , Levetiracetam , Masculino , Piracetam/farmacología , Ratas , Ratas Sprague-Dawley
3.
Restor Neurol Neurosci ; 34(2): 257-70, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26890099

RESUMEN

PURPOSE: We assessed the therapeutic efficacy of FDA-approved anti-epileptic drug Levetiracetam (LEV) to reduce post-traumatic nonconvulsive seizure (NCS) activity and promote neurobehavioral recovery following 10% frontal penetrating ballistic-like brain injury (PBBI) in male Sprague-Dawley rats. METHODS: Experiment 1 anti-seizure study: 50 mg/kg LEV (25 mg/kg maintenance doses) was given twice daily for 3 days (LEV3D) following PBBI; outcome measures included seizures incidence, frequency, duration, and onset. Experiment 2 neuroprotection studies: 50 mg/kg LEV was given twice daily for either 3 (LEV3D) or 10 days (LEV10D) post-injury; outcome measures include motor (rotarod) and cognitive (water maze) functions. RESULTS: LEV3D treatment attenuated seizure activity with significant reductions in NCS incidence (54%), frequency, duration, and delayed latency to seizure onset compared to vehicle treatment. LEV3D treatment failed to improve cognitive or motor performance; however extending the dosing regimen through 10 days post-injury afforded significant neuroprotective benefit. Animals treated with the extended LEV10D dosing regimen showed a twofold improvement in rotarod task latency to fall as well as significantly improved spatial learning performance (24%) in the MWM task. CONCLUSIONS: These findings support the dual anti- seizure and neuroprotective role of LEV, but more importantly identify the importance of an extended dosing protocol which was specific to the therapeutic targets studied.


Asunto(s)
Traumatismos Penetrantes de la Cabeza/complicaciones , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Piracetam/análogos & derivados , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Trastornos Neurológicos de la Marcha/tratamiento farmacológico , Trastornos Neurológicos de la Marcha/etiología , Regulación de la Expresión Génica/efectos de los fármacos , Levetiracetam , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Piracetam/farmacología , Piracetam/uso terapéutico , Ratas , Ratas Sprague-Dawley , Tiempo de Reacción/efectos de los fármacos , Estadísticas no Paramétricas , Factores de Tiempo , Índices de Gravedad del Trauma , Resultado del Tratamiento
4.
J Neurotrauma ; 31(5): 505-14, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24138024

RESUMEN

Abstract Blood-brain barrier (BBB) disruption is a pathological hallmark of severe traumatic brain injury (TBI) and is associated with neuroinflammatory events contributing to brain edema and cell death. The goal of this study was to elucidate the profile of BBB disruption after penetrating ballistic-like brain injury (PBBI) in conjunction with changes in neuroinflammatory markers. Brain uptake of biotin-dextran amine (BDA; 3 kDa) and horseradish peroxidase (HRP; 44 kDa) was evaluated in rats at 4 h, 24 h, 48 h, 72 h, and 7 days post-PBBI and compared with the histopathologic and molecular profiles for inflammatory markers. BDA and HRP both displayed a uniphasic profile of extravasation, greatest at 24 h post-injury and which remained evident out to 48 h for HRP and 7 days for BDA. This profile was most closely associated with markers for adhesion (mRNA for intercellular adhesion molecule-1) and infiltration of peripheral granulocytes (mRNA for matrix metalloproteinase-9 [MMP-9] and myeloperoxidase staining). Improvement of BBB dysfunction coincided with increased expression of markers implicated in tissue remodeling and repair. The results of this study reveal a uniphasic and gradient opening of the BBB after PBBI and suggest MMP-9 and resident inflammatory cell activation as candidates for future neurotherapeutic intervention after PBBI.


Asunto(s)
Barrera Hematoencefálica/lesiones , Edema Encefálico/fisiopatología , Lesiones Encefálicas/fisiopatología , Traumatismos Penetrantes de la Cabeza/fisiopatología , Inflamación/fisiopatología , Animales , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Edema Encefálico/patología , Lesiones Encefálicas/patología , Traumatismos Penetrantes de la Cabeza/patología , Inflamación/patología , Masculino , Modelos Animales , Ratas , Ratas Sprague-Dawley
5.
J Neurotrauma ; 30(23): 1973-82, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23822888

RESUMEN

Acute seizures frequently occur following severe traumatic brain injury (TBI) and have been associated with poor patient prognosis. Silent or nonconvulsive seizures (NCS) manifest in the absence of motor convulsion, can only be detected via continuous electroencephalographic (EEG) recordings, and are often unidentified and untreated. Identification of effective anti-epileptic drugs (AED) against post-traumatic NCS remains crucial to improve neurological outcome. Here, we assessed the anti-seizure profile of ethosuximide (ETX, 12.5-187.5 mg/kg) and phenytoin (PHT, 5-30 mg/kg) in a spontaneously occurring NCS model associated with penetrating ballistic-like brain injury (PBBI). Rats were divided between two drug cohorts, PHT or ETX, and randomly assigned to one of four doses or vehicle within each cohort. Following PBBI, NCS were detected by continuous EEG monitoring for 72 h post-injury. Drug efficacy was evaluated on NCS parameters of incidence, frequency, episode duration, total duration, and onset latency. Both PHT and ETX attenuated NCS in a dose-dependent manner. In vehicle-treated animals, 69-73% experienced NCS (averaging 9-10 episodes/rat) with average onset of NCS occurring at 30 h post-injury. Compared with control treatment, the two highest PHT and ETX doses significantly reduced NCS incidence to 13-40%, reduced NCS frequency (1.8-6.2 episodes/rat), and delayed seizure onset: <20% of treated animals exhibited NCS within the first 48 h. NCS durations were also dose-dependently mitigated. For the first time, we demonstrate that ETX and PHT are effective against spontaneously occurring NCS following PBBI, and suggest that these AEDs may be effective at treating post-traumatic NCS.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Lesiones Encefálicas/complicaciones , Etosuximida/uso terapéutico , Fenitoína/uso terapéutico , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Animales , Anticonvulsivantes/farmacocinética , Encéfalo/patología , Lesiones Encefálicas/patología , Relación Dosis-Respuesta a Droga , Electroencefalografía/efectos de los fármacos , Etosuximida/farmacocinética , Traumatismos Penetrantes de la Cabeza/complicaciones , Traumatismos Penetrantes de la Cabeza/patología , Masculino , Fenitoína/farmacocinética , Ratas , Ratas Sprague-Dawley
6.
Ther Hypothermia Temp Manag ; 1(1): 33-42, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-24716886

RESUMEN

Induced hypothermia has been reported to provide neuroprotection against traumatic brain injury. We recently developed a novel method of selective brain cooling (SBC) and demonstrated its safety and neuroprotection efficacy in a rat model of ischemic brain injury. The primary focus of the current study was to evaluate the potential neuroprotective efficacy of SBC in a rat model of penetrating ballistic-like brain injury (PBBI) with a particular focus on the acute cerebral pathophysiology, neurofunction, and cognition. SBC (34°C) was induced immediately after PBBI, and maintained for 2 hours, followed by a spontaneous re-warming. Intracranial pressure (ICP) and regional cerebral blood flow were monitored continuously for 3 hours, and the ICP was measured again at 24 hours postinjury. Brain swelling, blood-brain barrier permeability, intracerebral hemorrhage, lesion size, and neurological status were assessed at 24 hours postinjury. Cognitive abilities were evaluated in a Morris water maze task at 12-16 days postinjury. Results showed that SBC significantly attenuated PBBI-induced elevation of ICP (PBBI = 33.2 ± 10.4; PBBI + SBC = 18.8 ± 6.7 mmHg) and reduced brain swelling, blood-brain barrier leakage, intracerebral hemorrhage, and lesion volume by 40%-45% for each matrix, and significantly improved neurologic function. However, these acute neuroprotective benefits of SBC did not translate into improved cognitive performance in the Morris water maze task. These results indicate that 34°C SBC is effective in protecting against acute brain damage and related neurological dysfunction. Further studies are required to establish the optimal treatment conditions (i.e., duration of cooling and/or combined therapeutic approaches) needed to achieve significant neurocognitive benefits.

7.
J Neurotrauma ; 27(9): 1635-41, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20568960

RESUMEN

Penetrating ballistic brain injury involves a leading shockwave producing a temporary cavity causing substantial secondary injury. In response to the prevalence of this type of brain trauma in the military, a rat model of penetrating ballistic-like brain injury (PBBI) was established. This study focuses on cerebral physiological responses resulting from a PBBI, specifically the immediate and delayed changes in intracranial pressure (ICP) and cerebral perfusion pressure (CPP). ICP/CPP was measured continuously in rats subjected to PBBI, probe insertion alone, or sham injury. Immediately following the PBBI, a transient (<0.1 sec) and dramatic elevation of ICP reaching 280.0 ± 86.0 mm Hg occurred, accompanied by a profound decrease in CPP to -180.2 ± 90.1 mm Hg. This emergent ICP/CPP response resolved spontaneously within seconds, but was followed by a slowly-developing and sustained secondary phase, which peaked at 24 h post-injury, reaching 37.2 ± 10.4 mm Hg, and remained elevated until 72 h post-injury. The measured decrease in CPP reached 85.3 ± 17.2 mm Hg at 3 h post-injury. By comparison, probe insertion alone did not produce the immediate ICP crisis (28.6 ± 9.1 mm Hg), and only a mild and sustained increase in ICP (13.5 ± 2.1 mm Hg) was observed in the following 3 h post-injury. Injury severity, as measured by lesion volume, brain swelling, and neurological deficits at 1, 3, and 7 days post-injury, also reflected the distinctive differences between the dynamics of the PBBI versus controls. These results not only reinforced the severe nature of this model in mimicking the ballistic effect of PBBI, but also established cerebral pathophysiological targets for neuroprotective therapies.


Asunto(s)
Modelos Animales de Enfermedad , Traumatismos Penetrantes de la Cabeza/patología , Traumatismos Penetrantes de la Cabeza/fisiopatología , Presión Intracraneal/fisiología , Animales , Análisis de los Gases de la Sangre/métodos , Edema Encefálico/etiología , Edema Encefálico/patología , Edema Encefálico/fisiopatología , Traumatismos Penetrantes de la Cabeza/complicaciones , Masculino , Ratas , Ratas Sprague-Dawley
8.
J Neurosci Methods ; 189(1): 84-7, 2010 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-20346980

RESUMEN

A rat model of penetrating ballistic-like brain injury (PBBI) was recently established to study military-relevant severe traumatic brain injury (TBI). The purpose of this study was to conduct a side-by-side evaluation of two well-established cognitive testing paradigms: the novel object recognition (NOR) task and the Morris water maze (MWM) task. Accordingly, male Sprague-Dawley rats were subjected to PBBI and their cognitive abilities were assessed at 7 and 21 days post-PBBI. Although PBBI animals had more difficulty completing both tasks compared to sham animals, their performance on the NOR task was confounded by a high degree of within-group variability that was likely due to attention deficits produced by the injury. In contrast, PBBI produced consistent, significant spatial learning deficits in the MWM task. Overall, these results suggest that the MWM task provides a more appropriate cognitive test for the PBBI model that would be useful for testing promising neuroprotective therapeutics.


Asunto(s)
Lesiones Encefálicas/diagnóstico , Trastornos del Conocimiento/diagnóstico , Cognición/fisiología , Evaluación de la Discapacidad , Pruebas Neuropsicológicas/normas , Animales , Atención/fisiología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/fisiopatología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/fisiopatología , Modelos Animales de Enfermedad , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/diagnóstico , Trastornos de la Memoria/etiología , Trastornos de la Memoria/fisiopatología , Variaciones Dependientes del Observador , Trastornos de la Percepción/diagnóstico , Trastornos de la Percepción/etiología , Trastornos de la Percepción/fisiopatología , Valor Predictivo de las Pruebas , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/fisiología , Sensibilidad y Especificidad
9.
J Neuroinflammation ; 6: 19, 2009 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-19656406

RESUMEN

BACKGROUND: Inflammatory cytokines play a crucial role in the pathophysiology of traumatic brain injury (TBI), exerting either deleterious effects on the progression of tissue damage or beneficial roles during recovery and repair. NNZ-2566, a synthetic analogue of the neuroprotective tripeptide Glypromate, has been shown to be neuroprotective in animal models of brain injury. The goal of this study was to determine the effects of NNZ-2566 on inflammatory cytokine expression and neuroinflammation induced by penetrating ballistic-like brain injury (PBBI) in rats. METHODS: NNZ-2566 or vehicle (saline) was administered intravenously as a bolus injection (10 mg/kg) at 30 min post-injury, immediately followed by a continuous infusion of NNZ-2566 (3 mg/kg/h), or equal volume of vehicle, for various durations. Inflammatory cytokine gene expression from the brain tissue of rats exposed to PBBI was evaluated using microarray, quantitative real time PCR (QRT-PCR), and enzyme-linked immunosorbent assay (ELISA) array. Histopathology of the injured brains was examined using hematoxylin and eosin (H&E) and immunocytochemistry of inflammatory cytokine IL-1beta. RESULTS: NNZ-2566 treatment significantly reduced injury-mediated up-regulation of IL-1beta, TNF-alpha, E-selectin and IL-6 mRNA during the acute injury phase. ELISA cytokine array showed that NZ-2566 treatment significantly reduced levels of the pro-inflammatory cytokines IL-1beta, TNF-alpha and IFN-gamma in the injured brain, but did not affect anti-inflammatory cytokine IL-6 levels. CONCLUSION: Collectively, these results suggest that the neuroprotective effects of NNZ-2566 may, in part, be functionally attributed to the compound's ability to modulate expression of multiple neuroinflammatory mediators in the injured brain.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Citocinas/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Oligopéptidos/farmacología , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/fisiopatología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Encefalitis/etiología , Encefalitis/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Traumatismos Penetrantes de la Cabeza/complicaciones , Traumatismos Penetrantes de la Cabeza/fisiopatología , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Masculino , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
10.
J Cereb Blood Flow Metab ; 29(12): 1924-32, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19638995

RESUMEN

Ischemic and traumatic brain injuries often induce non-convulsive seizures (NCSs), which likely contribute to the worsening of neurological outcomes. Here, we evaluated the effect of glycyl-L-methylprolyl-L-glutamic acid (NNZ-2566) to lessen the severity of NCSs caused by permanent middle cerebral artery occlusion (pMCAo). Continuous electroencephalographic recordings were performed in rats during pMCAo. Glycyl-L-methylprolyl-L-glutamic acid (3, 10, or 100 mg/kg bolus followed by an infusion of a fixed dose of 3 mg/kg per hour for 12 h) was delivered at 20 mins after pMCAo (before the first NCS event) or delayed until immediately after the first NCS event occurred. Control rats received pMCAo and saline treatment. The results revealed that 91% of the saline-treated animals had NCSs (23 episodes per rat and 1238 secs per rat) with an onset latency of 35 mins after injury. When NNZ-2566 was administered before the NCS events, it dose-dependently reduced the NCS incidence to 36%-80%, decreased NCS frequency to 5-16 episodes per rat, and shortened the total duration of NCS to 251-706 secs per rat. The two high doses significantly reduced the infarct volume by 28%-30%. Delayed treatment also attenuated NCS duration but had no effect on the infarct volume. Results indicate that NNZ-2566 possesses a unique therapeutic potential as a safe prophylactic agent that synergistically provides neuroprotection and reduces injury-induced seizures.


Asunto(s)
Isquemia Encefálica/complicaciones , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , Convulsiones/tratamiento farmacológico , Animales , Infarto Encefálico/tratamiento farmacológico , Electroencefalografía , Masculino , Oligopéptidos/química , Ratas , Ratas Sprague-Dawley , Convulsiones/etiología
11.
Pharmacol Biochem Behav ; 94(1): 56-62, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19619574

RESUMEN

Dextromethorphan (DM) has been well-characterized as a neuroprotective agent in experimental models of CNS injury. The goal of this study was to determine the neuroprotective profile of DM in a military-relevant model of penetrating ballistic-like brain injury (PBBI). In an acute (3 day) dose-response study, anesthetized male Sprague-Dawley rats were exposed to a unilateral frontal PBBI with DM (0.156-10 mg/kg) or vehicle delivered as an i.v. bolus from 30 min to 48 h post-injury. In a follow-up (7 day) experiment, the 10-mg/kg bolus injections of DM were administered in conjunction with a 6-h infusion (5 mg/kg/h). DM bolus injections alone produced a dose-dependent improvement in motor recovery on a balance beam task at 3 days post-injury. However, more rapid recovery (24 h) was observed on this task when the bolus injections were combined with the 6-h infusion. Moreover, the DM bolus/infusion treatment regimen resulted in a significant (76%) improvement in cognitive performance in a novel object recognition (NOR) task at 7 days post-injury. Although post-injury administration of DM (all doses) failed to reduce core lesion size, the maximum dose of DM (10 mg/kg) was effective in reducing silver-stained axonal fiber degeneration in the cortical regions adjacent to the injury.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Dextrometorfano/uso terapéutico , Lóbulo Frontal/lesiones , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Lesiones Encefálicas/patología , Lesiones Encefálicas/cirugía , Cognición/efectos de los fármacos , Dextrometorfano/administración & dosificación , Lesión Axonal Difusa/patología , Relación Dosis-Respuesta a Droga , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/cirugía , Traumatismos Penetrantes de la Cabeza/patología , Traumatismos Penetrantes de la Cabeza/cirugía , Procesamiento de Imagen Asistido por Computador , Masculino , Actividad Motora/efectos de los fármacos , Degeneración Nerviosa/patología , Fármacos Neuroprotectores/administración & dosificación , Ratas , Ratas Sprague-Dawley , Reconocimiento en Psicología/efectos de los fármacos , Factores de Tiempo
12.
J Neurotrauma ; 25(5): 549-59, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18435596

RESUMEN

Systemic cooling to achieve brain hypothermia has been investigated as a neuroprotective therapy but can present serious adverse effects. Here we describe a novel method to selectively cool the rat brain and investigate its neuroprotective effects following transient middle cerebral artery occlusion (MCAo). The novelty of our method of selective brain cooling (SBC) was that the extraluminal cooling of the carotid arterial blood was achieved by using a cooling cuff wrapped around each common carotid artery (CCA). Within 20 min of CCA cooling, brain temperature could be lowered by 2-5 degrees C below the baseline and maintained stable for approximately 2 h while maintaining body temperature at 37 degrees C. No adverse effects of SBC were observed on systemic physiology, regional cerebral blood flow (rCBF), bleeding time, or tissue histology in normal animals. In rats having sustained 2-h MCAo, intra-ischemic SBC for 90 min, initiated 30 min following the onset of ischemia, significantly reduced infarction measured at 24 h post-injury (normothermic rats=312+/-51 mm3, SBC rats=139+/-83 mm3). In subgroup experiments, the incidence of peri-infarct depolarization (PID) was assessed during the MCAo and cooling period. Compared to normothermic but ischemic rats, SBC significantly reduced the number of PID events from 6.2+/-2.5 to 2.0+/-2.5, and reduced infarct volumes from 323+/-79 to 139+/-102 mm3. In conclusion, this extralumimal cooling method of SBC provides a safe and efficient approach to rapidly and safely achieve hypothermic neuroprotection.


Asunto(s)
Encéfalo/irrigación sanguínea , Arterias Carótidas , Hipertermia Inducida/métodos , Infarto de la Arteria Cerebral Media/terapia , Degeneración Nerviosa/prevención & control , Animales , Encéfalo/patología , Circulación Cerebrovascular/fisiología , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratas , Ratas Sprague-Dawley
13.
J Neurotrauma ; 23(12): 1828-46, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17184192

RESUMEN

Acute and delayed cerebral injury was assessed in a recently developed rat model of a penetrating ballistic-like brain injury (PBBI). A unilateral right frontal PBBI trajectory was used to induce survivable injuries to the frontal cortex and striatum. Three distinct phases of injury progression were observed. Phase I (primary injury, 0-6 h) began with immediate (<5 min) intracerebral hemorrhage (ICH) that reached maximal volumetric size at 6 h (27.0 +/- 2.9 mm(3)). During Phase II (secondary injury, 6-72 h), a core lesion of degenerate neurons surrounding the injury track expanded into peri-lesional areas to reach a maximal volume of 69.9 +/- 6.1 mm(3) at 24 h. The core lesion consisted of predominately necrotic cell death and included marked infiltration of both neutrophils (24 h) and macrophages (72 h). Phase III (delayed degeneration, 3-7 days) involved the degeneration of neurons and fiber tracts remote from the core lesion including the thalamus, internal capsule, external capsule, and cerebral peduncle. Overall, different time courses of hemorrhage, lesion evolution, and inflammation were consistent with complementary roles in injury development and repair, providing key information about these mediators of primary, secondary, and delayed brain injury development. The similarities/differences of PBBI to other focal brain injury models are discussed.


Asunto(s)
Lesiones Encefálicas/patología , Hemorragia Cerebral Traumática/etiología , Encefalitis/etiología , Degeneración Nerviosa/etiología , Heridas por Arma de Fuego/patología , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/inmunología , Hemorragia Cerebral Traumática/patología , Modelos Animales de Enfermedad , Encefalitis/patología , Masculino , Necrosis/etiología , Necrosis/patología , Degeneración Nerviosa/patología , Infiltración Neutrófila , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Heridas por Arma de Fuego/complicaciones , Heridas por Arma de Fuego/inmunología
14.
J Pharmacol Exp Ther ; 318(3): 947-55, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16728590

RESUMEN

Acute seizures following brain injury have been associated with a worsening of patient outcome, but they are often undiagnosed and untreated when they occur without motor convulsions. Here, we sought to compare the antiseizure profile of ethosuximide (EXM; 125-312.5 mg/kg i.v.) and gabapentin (GBP; 0.3-50 mg/kg. i.v.) in a rat model of nonconvulsive seizures (NCS) induced by brain ischemia. Seizures were detected by continuous electroencephalographic monitoring for 24 h following permanent middle cerebral artery occlusion (MCAo). Both "preseizure" and "postseizure" treatment effects were evaluated. Control rats experienced a 91% incidence of NCS (averaging 10-11 NCS/rat), which was significantly reduced following preseizure treatment (delivered 20 min post-MCAo) with either EXM (ED(50) = 161 mg/kg) or GBP (ED(50) = 10.5 mg/kg). In contrast to preseizure treatment effects, only GBP reduced NCS when given after the first seizure event. A further, albeit nonsignificant, 20% reduction in NCS incidence was measured when given in combination postseizure. Drug treatment also reduced infarct volume, which was positively correlated to the number of NCS events (r = 0.475; P < 0.001). EXM and GBP treatment of cultured neurons exposed to neurotoxic or ischemic insults showed no neuroprotective effects, suggesting that in vivo neuroprotection can be attributed to anti-seizure effects. We conclude that EXM and GBP significantly attenuate NCS in a dose-related manner and may help to improve patient outcome from brain ischemia-induced seizure activity.


Asunto(s)
Aminas/uso terapéutico , Isquemia Encefálica/complicaciones , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Etosuximida/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Convulsiones/tratamiento farmacológico , Ácido gamma-Aminobutírico/uso terapéutico , Enfermedad Aguda , Animales , Relación Dosis-Respuesta a Droga , Electroencefalografía , Gabapentina , Masculino , Ratas , Ratas Sprague-Dawley , Convulsiones/fisiopatología
15.
Pharmacol Biochem Behav ; 81(1): 182-9, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15894077

RESUMEN

In the present study, we evaluated delayed treatment effects of the proteasome inhibitor and anti-inflammatory agent MLN519 (initiated 10 h post-injury) to improve recovery following ischemic brain injury in rodents. Male rats were exposed to 2 h of middle cerebral artery occlusion (MCAo) and treated with MLN519 (1.0 mg/kg, i.v. @ 10, 24, and 48 h post-occlusion) or vehicle. By 2 weeks post-injury, 60% (6/10) of vehicle animals survived, which was improved (although non-significantly) to 78% (7/9) following MLN519 treatment. The percent loss of tissue in the ipsilateral brain hemisphere (at 2 weeks) was significantly reduced from 27+/-4% (vehicle) to 15+/-4% (MLN519). MLN519 treated animals also lost significantly less body weight (39%) and showed significant improvement in overall neurological function across the 2-week recovery period. However, no significant treatment effects were observed to reduce foot-fault deficits (balance beam) or improve recovery of operant performance (active avoidance test). Overall, delayed treatment with MLN519 provided significant improvement in 3 of 6 test metrics (histopathology, body weight, and neurological dysfunction) supporting improved outcome for brain-injured subjects.


Asunto(s)
Acetilcisteína/análogos & derivados , Isquemia Encefálica/tratamiento farmacológico , Recuperación de la Función/efectos de los fármacos , Acetilcisteína/administración & dosificación , Animales , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Inyecciones Intraventriculares , Masculino , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/fisiología , Factores de Tiempo
16.
J Med Chem ; 48(7): 2319-24, 2005 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-15801825

RESUMEN

Two representative glutamate carboxypeptidase II (GCP II) inhibitors, 2-(hydroxypentafluorophenylmethyl-phosphinoylmethyl)pentanedioic acid 2 and 2-(3-mercaptopropyl)pentanedioic acid 3, were synthesized in high optical purities (>97%ee). The two enantiomers of 2 were prepared from previously reported chiral intermediates, (R)- and (S)-2-(hydroxyphosphinoylmethyl)pentanedioic acid benzyl esters 8. The synthesis of (R)- and (S)-3 involves the hydrolysis of (R)- and (S)-3-(2-oxo-tetrahydro-thiopyran-3-yl)propionic acids, (R)- and (S)-11, the corresponding optically pure thiolactones delivered by chiral chromatographic separation of the racemic 11. GCP II inhibitory assay revealed that (S)-2 is 40-fold more potent than (R)-2. In contrast, both enantiomers of 3 inhibited GCP II with nearly equal potency. The efficacy observed in subsequent animal studies with these enantiomers correlated well with the inhibitory potency in a GCP II assay.


Asunto(s)
Glutamato Carboxipeptidasa II/antagonistas & inhibidores , Glutaratos/síntesis química , Ácidos Fosfínicos/síntesis química , Compuestos de Sulfhidrilo/síntesis química , Analgésicos/síntesis química , Analgésicos/química , Analgésicos/farmacología , Animales , Isquemia Encefálica/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Constricción Patológica/complicaciones , Cristalografía por Rayos X , Glutamato Carboxipeptidasa II/química , Glutaratos/química , Glutaratos/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , L-Lactato Deshidrogenasa/metabolismo , Estructura Molecular , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Fármacos Neuroprotectores/farmacología , Dolor/tratamiento farmacológico , Dolor/etiología , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/etiología , Ácidos Fosfínicos/química , Ácidos Fosfínicos/farmacología , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/farmacología , Técnicas de Cultivo de Tejidos
17.
Brain Res ; 978(1-2): 99-103, 2003 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-12834903

RESUMEN

Poly(ADP-ribose) is synthesized from nicotinamide adenine dinucleotide (NAD(+)) by poly(ADP-ribose) polymerase (PARP) and degraded by poly(ADP-ribose) glycohydrolase (PARG). Overactivation of the poly(ADP-ribose) pathway increases nicotinamide and decreases cellular NAD(+)/ATP, which leads to cell death. Blocking poly(ADP-ribose) metabolism by inactivating PARP has been shown to reduce ischemia injury. We investigated whether disrupting the poly(ADP-ribose) cycle by PARG inhibition could achieve similar protection. We demonstrate that either pre- or post-ischemia treatment with 40 mg/kg of N-bis-(3-phenyl-propyl)9-oxo-fluorene-2,7-diamide, a novel PARG inhibitor, significantly reduces brain infarct volumes by 40-53% in a rat model of focal cerebral ischemia. Our result provides the first evidence that PARG inhibitors can ameliorate ischemic brain damage in vivo, in support of PARG as a new therapeutic target for treating ischemia injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Infarto Cerebral/tratamiento farmacológico , Diamida/análogos & derivados , Diamida/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Glicósido Hidrolasas/antagonistas & inhibidores , Animales , Encéfalo/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Infarto Cerebral/metabolismo , Infarto Cerebral/patología , Colorantes/farmacocinética , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratas , Ratas Sprague-Dawley , Reperfusión/métodos , Sales de Tetrazolio/farmacocinética , Factores de Tiempo
18.
J Neurochem ; 86(4): 896-906, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12887688

RESUMEN

The sodium-vitamin C co-transporter SVCT2 is primarily responsible for the accumulation of the important antioxidant ascorbate into brain cells. In vitro studies have demonstrated strong expression of this transporter in cultured astrocytes, whereas in situ hybridization analysis has so far detected SVCT2 only in neurons. In the present study, we examined the response of SVCT2 mRNA expression in the brain to focal ischemia induced for 2 h by unilateral middle cerebral artery occlusion. The mRNA expression patterns of SVCT2 and the glutamate-activated immediate early gene Arc were investigated at 2 and 22 h after ischemia. SVCT2 and Arc mRNA expression was lost in the ischemic core at both time points. In areas outside the core, Arc was strongly up-regulated, primarily at 2 h, whereas SVCT2 showed an increase at 2 and 22 h. SVCT2 expression was increased in neurons as well as in astrocytes, providing the first evidence for SVCT2 expression in astrocytes in situ. These findings underscore the importance of ascorbate as a neuroprotective agent and may have implications for therapeutic strategies. In addition, the increase of SVCT2 in astrocytes after ischemia suggests that cultured astrocytes are exposed to chronic oxidative stress.


Asunto(s)
Encéfalo/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Proteínas del Tejido Nervioso , Transportadores de Anión Orgánico Sodio-Dependiente/genética , ARN Mensajero/metabolismo , Simportadores/genética , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encéfalo/irrigación sanguínea , Encéfalo/patología , Proteínas del Citoesqueleto , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Hibridación in Situ , Infarto de la Arteria Cerebral Media/patología , Masculino , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Transportadores de Sodio Acoplados a la Vitamina C
19.
Fundam Clin Pharmacol ; 17(5): 581-93, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14703719

RESUMEN

The spatiotemporal electroencephalogram (EEG) pathology associated with brain injury was studied using high-resolution, 10-electrode cortical EEG mapping in a rat model of middle cerebral artery occlusion (MCAo). Using this model we evaluated the ability of the novel sodium channel blocker and neuroprotective agent RS100642 to resolve injury-induced EEG abnormalities as a measure of neurophysiological recovery from brain injury. The middle cerebral artery (MCA) was occluded for 1 h during which a dramatic loss of EEG power was measured over the injured cortex with near complete recovery upon reperfusion of blood to the MCA region in all rats. The resultant progression of the MCAo/reperfusion injury (6-72 h) included the appearance of diffuse polymorphic delta activity (PDA), as visually indicated by the presence of high-amplitude slow-waves recorded from both brain hemispheres. PDA was associated with large increases in EEG power, particularly evident in outer 'peri-infarct' regions of the ipsilateral parietal cortex as visualized using topographic EEG mapping. Post-injury treatment with RS100642 (1.0 mg/kg, i.v.) significantly reduced the PDA activity and attenuated the increase in EEG power throughout the course of the injury. These effects were associated with a reduction in brain infarct volume and improved neurological function. These methods of EEG analysis may be helpful tools to evaluate the physiological recovery of the brain from injury in humans following treatment with an experimental neuroprotective compound.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Ritmo Delta/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Bloqueadores de los Canales de Sodio/uso terapéutico , Canales de Sodio/fisiología , Animales , Isquemia Encefálica/fisiopatología , Mapeo Encefálico/métodos , Ritmo Delta/métodos , Electroencefalografía/efectos de los fármacos , Electroencefalografía/métodos , Masculino , Fármacos Neuroprotectores/farmacología , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA