Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Shock ; 60(2): 248-254, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37267223

RESUMEN

ABSTRACT: Several studies have demonstrated the clinical utility of tranexamic acid (TXA) for use in trauma patients presenting with significant hemorrhage. Tranexamic acid is an antifibrinolytic that inhibits plasminogen activation, and plasmin activity has been shown to mitigate blood loss and reduce all-cause mortality in the absence of adverse vascular occlusive events. Recent clinical developments indicate TXA is safe to use in patients with concomitant traumatic brain injury (TBI); however, the prehospital effects are not well understood. Importantly, TXA has been associated with seizure activity. Therefore, this study sought to evaluate the effects of early administration of TXA on neurological recovery and electroencephalogram (EEG) abnormalities following penetrating TBI with concomitant hypoxemia and hemorrhagic shock. We hypothesized that early administration of TXA will provide hemodynamic stabilization and reduce intracerebral hemorrhage, which will result in improved neurological function. To test this hypothesis, Sprague-Dawley rats received a unilateral, frontal penetrating ballistic-like brain injury by inserting a probe into the frontal cortex of the anesthetized rat. Five minutes following brain injury, animals underwent 30 min of respiratory distress and 30 min of hemorrhage. Upon completion of the hemorrhage phase, animals received the initial dose of drug intravenously over 10 min after which the prehospital phase was initiated. During the prehospital phase, animals received autologous shed whole blood as needed to maintain a MAP of 65 mm Hg. After 90 min, "in-hospital" resuscitation was performed by administering the remaining shed whole blood providing 100% oxygen for 15 min. Upon recovery from surgery, animals were administered their second dose of vehicle or TXA intravenously over 8 h. Tranexamic acid induced an early improvement in neurologic deficit, which was statistically significant compared with vehicle at 24, 48, and 72 h at three doses tested. Analysis of cerebral hemoglobin content and intracerebral lesion progression revealed 100 mg/kg provided the optimal effects for improvement of neuropathology and was continued for determination of adverse treatment effects. We observed no exacerbation of cerebral thrombosis, but TXA treatment caused an increased risk of EEG abnormalities. These results suggest that TXA following polytrauma with concomitant brain injury may provide mild neuroprotective effects by preventing lesion progression, but this may be associated with an increased risk of abnormal EEG patterns. This risk may be associated with TXA inhibition of glycine receptors and may warrant additional considerations during the use of TXA in patients with severe TBI.


Asunto(s)
Antifibrinolíticos , Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Traumatismos Penetrantes de la Cabeza , Traumatismo Múltiple , Ácido Tranexámico , Animales , Ratas , Ácido Tranexámico/uso terapéutico , Ratas Sprague-Dawley , Hemorragia/tratamiento farmacológico , Hemorragia/etiología , Antifibrinolíticos/uso terapéutico , Traumatismo Múltiple/complicaciones , Traumatismo Múltiple/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Encefálicas/tratamiento farmacológico , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Electroencefalografía/efectos adversos , Fibrina
2.
Mil Med ; 184(Suppl 1): 291-300, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30901408

RESUMEN

This study assessed the effect of caffeine on neurobehavioral recovery in the WRAIR penetrating ballistic-like brain injury (PBBI) model. Unilateral frontal PBBI was produced in the right hemisphere of anesthetized rats at moderate (7%-PBBI) or severe (10%-PBBI) injury levels. Animals were randomly assigned to pretreatment groups: acute caffeine (25 mg/kg CAF gavage, 1 h prior to PBBI), or chronic caffeine (0.25 g/L CAF drinking water, 30 days prior to PBBI). Motor function was evaluated on the rotarod at fixed-speed increments of 10, 15, and 20 RPM. Cognitive performance was evaluated on the Morris water maze. Acute caffeine showed no significant treatment effect on motor or cognitive outcome. Acute caffeine exposure prior to 10%-PBBI resulted in a significantly higher thigmotaxic response compared to vehicle-PBBI groups, which may indicate caffeine exacerbates post-injury anxiety/attention decrements. Results of the chronic caffeine study revealed a significant improvement in motor outcome at 7 and 10 days post-injury in the 7%-PBBI group. However, chronic caffeine exposure significantly increased the latency to locate the platform in the Morris water maze task at all injury levels. Results indicate that chronic caffeine consumption prior to a penetrating TBI may provide moderate beneficial effects to motor recovery, but may worsen the neurocognitive outcome.


Asunto(s)
Cafeína/farmacología , Cognición/efectos de los fármacos , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Actividad Motora/efectos de los fármacos , Análisis de Varianza , Animales , Cafeína/uso terapéutico , Estimulantes del Sistema Nervioso Central/farmacología , Estimulantes del Sistema Nervioso Central/uso terapéutico , Cognición/fisiología , Modelos Animales de Enfermedad , Traumatismos Penetrantes de la Cabeza/fisiopatología , Masculino , Actividad Motora/fisiología , Ratas , Ratas Sprague-Dawley/lesiones , Prueba de Desempeño de Rotación con Aceleración Constante , Resultado del Tratamiento
3.
J Neurosci Res ; 96(4): 545-555, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28480980

RESUMEN

The use of progesterone following brain injury has a controversial history. On one hand, some lab-based models have showed progesterone as being neuroprotective, but on the other, clinical trials have showed quite the opposite. One of many complaints that arose from this discrepancy was the lack of a diverse pool of animal models and paradigms employed during the preclinical phase. However, over the past decade, the zebra finch has emerged as an optimal organism for the study of steroid-mediated neuroprotection. Following an injury, steroid hormones and receptors are upregulated, serving to decrease neuroinflammation and overall damage to the brain. As compared to other vertebrate models, zebra finches can upregulate expression of both estrogens and androgens at a faster and more robust response, suggesting that vertebrates differ in their neuroprotective mechanisms and timing following injury. Therefore, to expand the types organisms studied in pre-clinical trials, we chose to use zebra finches. While the majority of work in the zebra finch brain has focused on estrogens and androgens, we sought to clarify the role of progesterone following injury. Adult male zebra finches were given daily injections of progesterone following a penetrating injury and then were assessed for the size of injury and expression of various genes associated with neuroinflammation and cell survival. Treatment with progesterone decreased the injury size in zebra finches over controls and increased expression of various genes associated with cell survival and neuroinflammation. These data suggest that progesterone does mediate neuroprotection, most likely through the alteration of neuroinflammatory and cell survival pathways.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Traumatismos Penetrantes de la Cabeza/metabolismo , Progesterona/farmacología , Animales , Lesiones Traumáticas del Encéfalo/patología , Pinzones/metabolismo , Traumatismos Penetrantes de la Cabeza/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Modelos Animales , Neuroprotección , Proteínas Serina-Treonina Quinasas/metabolismo , ARN/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
4.
Biomaterials ; 87: 157-169, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26923363

RESUMEN

Intracortical neural probes enable researchers to measure electrical and chemical signals in the brain. However, penetration injury from probe insertion into living brain tissue leads to an inflammatory tissue response. In turn, microglia are activated, which leads to encapsulation of the probe and release of pro-inflammatory cytokines. This inflammatory tissue response alters the electrical and chemical microenvironment surrounding the implanted probe, which may in turn interfere with signal acquisition. Dexamethasone (Dex), a potent anti-inflammatory steroid, can be used to prevent and diminish tissue disruptions caused by probe implantation. Herein, we report retrodialysis administration of dexamethasone while using in vivo two-photon microscopy to observe real-time microglial reaction to the implanted probe. Microdialysis probes under artificial cerebrospinal fluid (aCSF) perfusion with or without Dex were implanted into the cortex of transgenic mice that express GFP in microglia under the CX3CR1 promoter and imaged for 6 h. Acute morphological changes in microglia were evident around the microdialysis probe. The radius of microglia activation was 177.1 µm with aCSF control compared to 93.0 µm with Dex perfusion. T-stage morphology and microglia directionality indices were also used to quantify the microglial response to implanted probes as a function of distance. Dexamethasone had a profound effect on the microglia morphology and reduced the acute activation of these cells.


Asunto(s)
Antiinflamatorios/uso terapéutico , Dexametasona/uso terapéutico , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Microdiálisis/instrumentación , Microglía/efectos de los fármacos , Animales , Antiinflamatorios/administración & dosificación , Encéfalo/efectos de los fármacos , Dexametasona/administración & dosificación , Traumatismos Penetrantes de la Cabeza/complicaciones , Traumatismos Penetrantes de la Cabeza/patología , Inflamación/complicaciones , Inflamación/patología , Ratones Transgénicos , Microglía/patología , Prótesis e Implantes
5.
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
6.
J Neurotrauma ; 32(20): 1621-32, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25794265

RESUMEN

Post-traumatic seizures can exacerbate injurious outcomes of severe brain trauma, yet effective treatments are limited owing to the complexity of the pathology underlying the concomitant occurrence of both events. In this study, we tested C-10068, a novel deuterium-containing analog of (+)-N-methyl-3-ethoxymorphinan, in a rat model of penetrating ballistic-like brain injury (PBBI) and evaluated the effects of C-10068 on PBBI-induced nonconvulsive seizures (NCS), acute neuroinflammation, and neurofunctional outcomes. NCS were detected by electroencephalographic monitoring. Neuroinflammation was evaluated by immunohistochemical markers, for example, glial fibrillary acidic protein and major histocompatibility complex class I, for activation of astrocytes and microglia, respectively. Neurofunction was tested using rotarod and Morris water maze tasks. Three infusion doses of C-10068 (1.0, 2.5, and 5.0 mg/kg/h × 72 h) were tested in the antiseizure study. Neuroinflammation and neurofunction were evaluated in animals treated with 5.0 mg/kg/h × 72 h C-10068. Compared to vehicle treatment, C-10068 dose dependently reduced PBBI-induced NCS incidence (40-50%), frequency (20-70%), and duration (30-82%). The most effective antiseizure dose of C-10068 (5.0 mg/kg/h × 72 h) also significantly attenuated hippocampal astrocyte activation and perilesional microglial reactivity post-PBBI. Within C-10068-treated animals, a positive correlation was observed in reduction in NCS frequency and reduction in hippocampal astrocyte activation. Further, C-10068 treatment significantly attenuated astrocyte activation in seizure-free animals. However, C-10068 failed to improve PBBI-induced motor and cognitive functions with the dosing regimen used in this study. Overall, the results indicating that C-10068 exerts both potent antiseizure and antiinflammatory effects are promising and warrant further investigation.


Asunto(s)
Antiinflamatorios , Anticonvulsivantes , Dextrometorfano , Antagonistas de Aminoácidos Excitadores , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/farmacología , Astrocitos/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Dextrometorfano/administración & dosificación , Dextrometorfano/análogos & derivados , Dextrometorfano/farmacología , Modelos Animales de Enfermedad , Electroencefalografía , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Antagonistas de Aminoácidos Excitadores/farmacología , Traumatismos Penetrantes de la Cabeza/complicaciones , Traumatismos Penetrantes de la Cabeza/inmunología , Hipocampo/efectos de los fármacos , Inflamación/etiología , Masculino , Microglía/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Convulsiones/etiología
7.
Restor Neurol Neurosci ; 33(2): 189-203, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25588460

RESUMEN

PURPOSE: The present work compared the behavioral outcomes of ACCS therapy delivered either intravenously (i.v.) or intracerebroventricularly (i.c.v.) after penetrating ballistic-like brain injury (PBBI). Histological markers for neuroinflammation and neurodegeneration were employed to investigate the potential therapeutic mechanism of ACCS. METHODS: Experiment-1, ACCS was administered either i.v. or i.c.v. for 1 week post-PBBI. Outcome metrics included behavioral (rotarod and Morris water maze) and gross morphological assessments. Experiment-2, rats received ACCS i.c.v for either 1 or 2 weeks post-PBBI. The inflammatory response was determined by immunohistochemistry for neutrophils and microglia reactivity. Neurodegeneration was visualized using silver staining. RESULTS: Both i.v. and i.c.v. delivery of ACCS improved motor outcome but failed to improve cognitive outcome or tissue sparing. Importantly, only i.c.v. ACCS treatment produced persistent motor improvements at a later endpoint. The i.c.v. ACCS treatment significantly reduced PBBI-induced increase in myeloperoxidase (MPO) and ionized calcium binding adaptor molecule 1 (Iba1) expression. Concomitant reduction of both Iba1 and silver staining were detected in corpus callosum with i.c.v. ACCS treatment. CONCLUSIONS: ACCS, as a treatment for TBI, showed promise with regard to functional (motor) recovery and demonstrated strong capability to modulate neuroinflammatory responses that may underline functional recovery. However, the majority of beneficial effects appear restricted to the i.c.v. route of ACCS delivery, which warrants future studies examining delivery routes (e.g. intranasal delivery) which are more clinically viable for the treatment of TBI.


Asunto(s)
Citocinas/administración & dosificación , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Actividad Motora/efectos de los fármacos , Neuroinmunomodulación/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Amnios , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Traumatismos Penetrantes de la Cabeza/patología , Traumatismos Penetrantes de la Cabeza/fisiopatología , Inmunohistoquímica , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Proteínas de Microfilamentos/metabolismo , Actividad Motora/fisiología , Neuroinmunomodulación/fisiología , Peroxidasa/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley , Prueba de Desempeño de Rotación con Aceleración Constante , Soluciones
8.
Anal Chem ; 85(17): 8173-9, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23927692

RESUMEN

Microdialysis sampling in the brain is employed frequently in the chemical analysis of neurological function and disease, but implanting the probes, which are substantially larger than the size and spacing of brain cells and blood vessels, is injurious and triggers ischemia, gliosis, and cell death at the sampling site. The nature of the interface between the brain and the microdialysis probe is critical to the use of microdialysis as a neurochemical analysis technique. The objective of the work reported here was to investigate the potential of two compounds, dexamethasone, a glucocorticoid anti-inflammatory agent, and XJB-5-131, a mitochondrially targeted reactive oxygen species scavenger, to mitigate the penetration injury. Measurements were performed in the rat brain striatum, which is densely innervated by axons that release dopamine, an electroactive neurotransmitter. We used voltammetry to measure electrically evoked dopamine release next to microdialysis probes during the retrodialysis of dexamethasone or XJB-5-131. After the in vivo measurements, the brain tissue containing the microdialysis probe tracks was examined by fluorescence microscopy using markers for ischemia, neuronal nuclei, macrophages, and dopamine axons and terminals. Dexamethasone and XJB-5-131 each diminished the loss of evoked dopamine activity, diminished ischemia, diminished the loss of neuronal nuclei, diminished the appearance of extravasated macrophages, and diminished the loss of dopamine axons and terminals next to the probes. Our findings confirm the ability of dexamethasone and XJB-5-131 to mitigate, but not eliminate, the effects of the penetration injury caused by implanting microdialysis probes into brain tissue.


Asunto(s)
Óxidos N-Cíclicos/administración & dosificación , Dexametasona/administración & dosificación , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Microdiálisis/efectos adversos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Dopamina/análisis , Traumatismos Penetrantes de la Cabeza/etiología , Traumatismos Penetrantes de la Cabeza/patología , Microdiálisis/instrumentación , Ratas
9.
Brain Res Bull ; 98: 64-75, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23886572

RESUMEN

A penetrating brain injury produces a glial scar formed by astrocytes, oligodendrocytes, microglia and NG2 cells. Glial scar is a barrier preventing the extent of damage but it has deleterious effects in the regeneration of the axons. Estradiol and tamoxifen reduce gliosis and have neuroprotective effects in the hippocampus and the spinal cord. We evaluated the proliferation of glia and the electrocorticogram in the sensorial cortex in a brain injury model. At seven days post-injury, estradiol, tamoxifen and estradiol plus tamoxifen reduced the number of resident and proliferative NG2 and reactive astrocyte vimentin+ cells. Estradiol and tamoxifen effects on NG2 cells could be produced by the classical oestrogen receptors found in these cells. The glial scar was also reduced by tamoxifen. At thirty days post-injury, the amount of resident and proliferative astrocytes increased significantly, except in the estradiol plus tamoxifen group, whilst the oligodendrocytes proliferation in the glial scar was reduced in treated animals. Tamoxifen promotes the survival of FOX-3+ neurons in the injured area and a recovery in the amplitude of electrocorticogram waves. At thirty days, estradiol did not favour the survival of neurons but produced a greater number of reactive astrocytes. In contrast, the number of oligodendrocytes was reduced. Tamoxifen could favour brain repair promoting neuron survival and adjusting glial cell number. It seems to recover adequate neural communication.


Asunto(s)
Traumatismos Penetrantes de la Cabeza/patología , Regeneración/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/uso terapéutico , Corteza Somatosensorial/efectos de los fármacos , Tamoxifeno/uso terapéutico , Animales , Antígenos/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Ondas Encefálicas/efectos de los fármacos , Modelos Animales de Enfermedad , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Masculino , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteoglicanos/metabolismo , Ratas , Ratas Wistar , Receptores de Estrógenos/metabolismo , Corteza Somatosensorial/fisiopatología , Factores de Tiempo
10.
Neuromolecular Med ; 15(3): 504-14, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23765588

RESUMEN

The tripeptide glycine-proline-glutamate analogue NNZ-2566 (Neuren Pharmaceuticals) demonstrates neuroprotective efficacy in models of traumatic brain injury. In penetrating ballistic-like brain injury (PBBI), it significantly decreases injury-induced upregulation of inflammatory cytokines including TNF-α, IFN-γ, and IL-6. However, the mechanism by which NNZ-2566 acts has yet to be determined. The activating transcription factor-3 (ATF3) is known to repress expression of these inflammatory cytokines and was increased at the mRNA and protein level 24-h post-PBBI. This study investigated whether 12 h of NNZ-2566 treatment following PBBI alters atf3 expression. PBBI alone significantly increased atf3 mRNA levels by 13-fold at 12 h and these levels were increased by an additional fourfold with NNZ-2566 treatment. To confirm that changes in mRNA translated to changes in protein expression, ATF3 expression levels were determined in vivo in microglia/macrophages, T cells, natural killer cells (NKCs), astrocytes, and neurons. PBBI alone significantly increased ATF3 in microglia/macrophages (820%), NKCs (58%), and astrocytes (51%), but decreased levels in T cells (48%). NNZ-2566 treatment further increased ATF3 protein expression in microglia/macrophages (102%), NKCs (308%), and astrocytes (13%), while reversing ATF3 decreases in T cells. Finally, PBBI increased ATF3 levels by 55% in neurons and NNZ-2566 treatment further increased these levels an additional 33%. Since increased ATF3 may be an innate protective mechanism to limit inflammation following injury, these results demonstrating that the anti-inflammatory and neuroprotective drug NNZ-2566 increase both mRNA and protein levels of ATF3 in multiple cell types provide a cellular mechanism for NNZ-2566 modulation of neuroinflammation following PBBI.


Asunto(s)
Factor de Transcripción Activador 3/biosíntesis , Antiinflamatorios no Esteroideos/uso terapéutico , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Proteínas del Tejido Nervioso/biosíntesis , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , Factor de Transcripción Activador 3/genética , Animales , Antiinflamatorios no Esteroideos/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Traumatismos Penetrantes de la Cabeza/metabolismo , Traumatismos Penetrantes de la Cabeza/patología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Oligopéptidos/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Regulación hacia Arriba/efectos de los fármacos
12.
J Trauma Acute Care Surg ; 73(2 Suppl 1): S156-64, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22847087

RESUMEN

BACKGROUND: Previous work has shown that human amnion-derived progenitor (AMP) cell therapy is neuroprotective in a penetrating ballistic-like brain injury (PBBI) model. However, the neuroprotective capacity of AMP cells seemed to be mediated by the sustained secretion of AMP cell-derived neurotrophic factors, which are abundant in the amnion-derived cellular cytokine suspension (ACCS). To test this theory, the current study assessed the neuroprotective efficacy of long-term ACCS delivery in the PBBI model. METHODS: Experiment 1 assessed the bioactive stability and neuroprotective capacity of ACCS in an in vitro model of neurodegeneration. Experiment 2 evaluated the therapeutic effects of ACCS delivery initiated 15 minutes after PBBI and continued for 2 weeks after injury. Experiment 3 was designed to identify the therapeutic window for long-term ACCS delivery in the PBBI model. Outcome metrics included neurobehavioral assessments and neuropathologic measures of neuroinflammation and axonal/neuronal degeneration. RESULTS: Experiment 1 demonstrated that ACCS is thermally stable for 1 week at 37°C and that ACCS treatment protected neurite against staurosporine toxicity. Experiment 2 identified the optimal infusion rate of ACCS (1 µL/h) and demonstrated that long-term infusion of ACCS was capable of promoting significant protection against PBBI-induced neuropathology and motor abnormalities, but was not sufficient for reducing cognitive deficits. Finally, the results of Experiment 3 showed that ACCS is effective in promoting significant neuroprotection even when onset of treatment is delayed out to 24 hours (but not 48 hours) after PBBI. CONCLUSIONS: Collectively, our results support the hypothesis that the neuroprotective effects of AMP cells are mediated through a sustained delivery of ACCS, which implicates ACCS as a promising neuroprotection agent for clinical study.


Asunto(s)
Amnios/citología , Citocinas/uso terapéutico , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Amnios/fisiología , Animales , Técnicas In Vitro , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Destreza Motora/efectos de los fármacos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
13.
Lik Sprava ; (7): 194-7, 2012.
Artículo en Ruso | MEDLINE | ID: mdl-23350150

RESUMEN

The problem of cardiovascular disease and cancer, the effects of traumatic brain injury is now one of the major health and social problems. Every year in Ukraine registered 200 thousand cases of the victims of traumatic brain injury. Of these, 30% of people then have persistent signs of disability that results in a disability, sometimes painful existence the patient and his relatives. Therefore, in order to bring man back into society after a traumatic brain injury, to the rehabilitation phase of treatment, immediately after the stabilization of the patient.


Asunto(s)
Lesiones Encefálicas/diagnóstico , Lesiones Encefálicas/rehabilitación , Traumatismos Cerrados de la Cabeza/diagnóstico , Traumatismos Cerrados de la Cabeza/rehabilitación , Traumatismos Penetrantes de la Cabeza/diagnóstico , Traumatismos Penetrantes de la Cabeza/rehabilitación , Lesiones Encefálicas/tratamiento farmacológico , Terapia Combinada , Quimioterapia Combinada , Electroencefalografía , Traumatismos Cerrados de la Cabeza/tratamiento farmacológico , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Humanos , Imagen por Resonancia Magnética , Masculino , Pruebas Neuropsicológicas , Modalidades de Fisioterapia , Reflejoterapia/métodos , Factores de Tiempo , Resultado del Tratamiento
14.
Neurocirugia (Astur) ; 20(5): 467-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19830370

RESUMEN

Orbital penetrating injuries may cause significant harm to the optic nerves and eyeball as well as to the brain and cerebral vessels. Management of orbital foreign bodies should include prompt recognition of the extent of the injury, broad-spectrum parenteral antibiotics, tetanus prophylaxis, anticonvulsant medication and early surgical intervention under direct vision to remove the foreign body and to avoid immediate and long-term complications. We report a penetrating orbital injury caused by a bread knife that extended from the orbit to the tegmental dura mater of the temporal bone. The knife's main trajectory coursed through the temporal lobe. Adjacent cerebral structures were explored before removal of the knife.


Asunto(s)
Ceguera/etiología , Lesiones Oculares Penetrantes/cirugía , Traumatismos Penetrantes de la Cabeza/cirugía , Órbita/lesiones , Lóbulo Temporal/lesiones , Adulto , Antibacterianos/uso terapéutico , Anticonvulsivantes/uso terapéutico , Urgencias Médicas , Lesiones Oculares Penetrantes/tratamiento farmacológico , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Traumatismos Penetrantes de la Cabeza/patología , Humanos , Masculino , Órbita/cirugía , Trastornos de la Pupila/etiología , Lóbulo Temporal/cirugía , Toxoide Tetánico
15.
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
16.
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
17.
Acta Neurochir (Wien) ; 150(7): 663-8, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18473114

RESUMEN

Traumatic brain contusions may increase in size over time or may develop at a delay after injury. This may lead to neurological deterioration, long term morbidity or even death. Coagulation disorders after injury can contribute to progression of haemorrhage. Recombinant activated factor VII (rFVIIa) was used in 12 patients with a severe head injury who had no systemic coagulopathy but who were considered to be at risk of progression of their intracranial lesion. Twelve consecutive patients suffering from life-threatening acute head injuries from blunt (3 cases) and penetrating mechanisms were given with rFVIIa, either to prevent the expected development of brain contusion or to assist in bleeding control during surgery. In 11 patients, rFVIIa was given by the attending neurosurgeon. Two of the patients died of their severe penetrating injuries one of whom had severe vasospasm 2 days after administration of rFVIIa. The other 11 patients did not appear to suffer any treatment-related adverse effects. When the drug was given prophylactically to prevent brain resection (6 cases) or to limit the need for widening resection (5 cases), marked control was achieved in seven cases, and a lesser effect was observed in the other 4 cases. We conclude that, in a small and highly individually selected series of patients with severe head injury, the administration of rFVIIa did not lead to adverse effects. Although the majority of patients were considered to be at high risk of progression of their lesions, this occurred in only one. The early use of rFVIIa in head injured patients without systemic coagulopathy may reduce the occurrence of enlargement of contusions, the requirement of further operation, and adverse outcome. Prospective randomised controlled studies are required to investigate this.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Etiquetado de Medicamentos , Factor VIIa/uso terapéutico , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Heridas no Penetrantes/tratamiento farmacológico , Adolescente , Adulto , Trastornos de la Coagulación Sanguínea , Lesiones Encefálicas/cirugía , Niño , Preescolar , Progresión de la Enfermedad , Esquema de Medicación , Factor VIIa/administración & dosificación , Traumatismos Penetrantes de la Cabeza/mortalidad , Traumatismos Penetrantes de la Cabeza/cirugía , Técnicas Hemostáticas , Humanos , Persona de Mediana Edad , Procedimientos Neuroquirúrgicos , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Medición de Riesgo , Índices de Gravedad del Trauma , Heridas no Penetrantes/cirugía
18.
Niger J Clin Pract ; 10(2): 177-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17902515

RESUMEN

Cow horn eye injuries are not common but are devastating causes of uniocular blindness amongst young active population. Early and appropriate intervention can save the life of the patient depending on the severity of the injury. This uncommon cause of unilateral visual loss can be prevented if slaughtering of cows are done by trained and appropriately equipped personnel.


Asunto(s)
Ceguera/etiología , Traumatismos Penetrantes de la Cabeza/complicaciones , Cuernos , Enfermedades Orbitales/complicaciones , Cráneo/lesiones , Adolescente , Adulto , Animales , Ceguera/cirugía , Bovinos , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Traumatismos Penetrantes de la Cabeza/cirugía , Humanos , Masculino , Enfermedades Orbitales/tratamiento farmacológico , Enfermedades Orbitales/cirugía
19.
J Neurotrauma ; 21(11): 1683-91, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15684658

RESUMEN

An internal fragment of the human neuroprotective polypeptide DSEP (Diffusible Survival Evasion Peptide) was delivered at 0.4 mg/kg (subcutaneously) 20-30 min after stab wound lesions in the parietal cortex of anesthetized rats. The peptide, CHEASAAQC or CHEC-9, inhibited the inflammatory response to the lesion and the degeneration of neurons adjacent to the wound. Four days after surgery, peptide-treated animals (n = 6) had 75% fewer reactive ameboid microglia/brain macrophages in the cortical parenchyma surrounding the lesion compared to vehicle-injected control rats (n = 6, p = 0.004). The cortical laminae in area 2 adjacent to the lesion were completely obscured in controls because of the increase in inflammatory cells and frank degeneration of neurons, while there was preservation of the neurons and cytoarchitecture after peptide treatment. In parallel experiments, CHEC-9 was found to inhibit the enzymatic activity of secreted phospholipase A2 (sPLA2), including activity present in the serum of peptide-injected rats. Kinetic analysis revealed the peptide increased the average Km for serum by 318% when tested 45 min after treatment (peptide-treated, n = 6; control-treated, n = 6; p = 0.0087), suggesting the principal effect of the peptide was to lower the affinity of serum sPLA2 for substrate. The sPLA2 inhibition by this particular peptide sequence appeared to be highly specific since inversion of a single pair of amino acids eliminated the inhibitory effect. Phorbol-12-myristate-13-acetate stimulated platelet aggregation, a PLA2-regulated activity, was also inhibited by the peptide. The discovery of CHEC-9 makes it possible to study in vivo the long appreciated contribution made by PLA2-directed inflammation to both acute and chronic neurodegeneration and may be helpful in designing therapies to limit neuron death in these conditions.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/lesiones , Inhibidores Enzimáticos/farmacología , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Fosfolipasas A/antagonistas & inhibidores , Secuencia de Aminoácidos/fisiología , Animales , Corteza Cerebral/enzimología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Encefalitis/tratamiento farmacológico , Encefalitis/etiología , Encefalitis/prevención & control , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/uso terapéutico , Femenino , Fosfolipasas A2 Grupo II , Traumatismos Penetrantes de la Cabeza/enzimología , Traumatismos Penetrantes de la Cabeza/fisiopatología , Masculino , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/etiología , Degeneración Nerviosa/prevención & control , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/farmacología , Proteínas del Tejido Nervioso/uso terapéutico , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Péptidos , Péptidos Cíclicos/farmacología , Péptidos Cíclicos/uso terapéutico , Fosfolipasas A/sangre , Fosfolipasas A2 , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/fisiología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Resultado del Tratamiento
20.
Brain Res ; 991(1-2): 34-45, 2003 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-14575874

RESUMEN

The elucidation of the molecular mechanisms involved in the response of brain tissue to trauma and the recognition of substances with neuroprotective properties is a prerequisite for the development of rational therapeutic approaches. In this study, we used a model of, unilateral, penetrating stab-like brain injury and examined the possible beneficial effects of post-injury administration of insulin-like growth factor-I (IGF-I) both at the cellular level, 4 and 12 h post-injury, and on the physical condition of the animals up to 1 week following the trauma. The consequences of injury were assessed by immunohistochemically observing the expression of heat-shock protein 70 (Hsp70), which is thought to be a marker of cell stress and injury, and by staining the tissue with the TUNEL reaction, in order to detect apoptotic cell death. Injury resulted in an increase in the number of Hsp70 and TUNEL positive cells in the peritraumatic area. The physical condition of the rats was followed by measuring body weight changes, food and water intake and by estimating their "motor activity". IGF-I administration resulted in a significant decrease in the number of Hsp70 and TUNEL positive cells in the peritraumatic area. Additionally, it improved the total "motor activity" of injured rats, increased food intake and attenuated the post-injury body weight loss. IGF-I thus emerges as a factor acting both at the cellular level as a neuroprotectant and at the systemic level as an anabolic agent.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/efectos de los fármacos , Traumatismos Penetrantes de la Cabeza/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Peso Corporal/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Ingestión de Líquidos/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas HSP70 de Choque Térmico/biosíntesis , Traumatismos Penetrantes de la Cabeza/patología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Modelos Animales , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...