Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neurosci Lett ; 738: 135324, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32860887

RESUMEN

BACKGROUND: The hyperpolarizing activity of γ-aminobutyric acid A (GABAA) receptors depends on the intracellular chloride gradient that is developmentally regulated by the activity of the chloride extruder potassium (K) chloride (Cl) cotransporter 2 (KCC2). In humans and rodents, KCC2 expression can be detected at birth. In rodents, KCC2 expression progressively increases and reaches adult-like levels by the second postnatal week of life. Several studies report changes in KCC2 expression levels in response to early-life injuries. However, the functional contribution of KCC2 in maintaining the excitation-inhibition balance in the neonatal brain is not clear. In the current study, we examined the effect of KCC2 antagonism on the neonatal brain activity under hyperexcitable conditions ex vivo and in vivo. METHODS: Ex vivo electrophysiology experiments were performed on hippocampal slices prepared from 7 to 9 days-old (P7-P9) male rats. Excitability of CA1 pyramidal neurons bathed in zero-Mg2+ buffer was measured using single-unit extracellular (loose) or cell-attach protocol before and after application of VU0463271, a specific antagonist of KCC2. To examine the functional role of KCC2 in vivo, the effect of VU0463271 on hypoxia-ischemia (HI)-induced ictal (seizures and brief runs of epileptiform discharges - BREDs), and inter-ictal spike and sharp-wave activity was measured in P7 male rats. A highly sensitive LC-MS/MS method was used to determine the distribution and the concentration of VU0463271 in the brain. RESULTS: Ex vivo blockade of KCC2 by VU0463271 significantly increased the frequency of zero-Mg2+-triggered spiking in CA1 pyramidal neurons. Similarly, in vivo administration of VU0463271 significantly increased the number of ictal events, BREDs duration, and spike and sharp-wave activity in HI rats. LC-MS/MS data revealed that following systemic administration, VU0463271 rapidly reached brain tissues and distributed well among different brain regions. CONCLUSION: The results suggest that KCC2 plays a critical functional role in maintaining the balance of excitation-inhibition in the neonatal brain, and thus it can be used as a therapeutic target to ameliorate injury associated with hyperexcitability in newborns.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Hipocampo/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Simportadores/antagonistas & inhibidores , Potenciales de Acción/fisiología , Animales , Electroencefalografía , Hipocampo/fisiopatología , Masculino , Células Piramidales/fisiología , Ratas , Ratas Sprague-Dawley , Convulsiones/fisiopatología , Cotransportadores de K Cl
2.
Pediatr Res ; 88(2): 202-208, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31896131

RESUMEN

BACKGROUND: Hypoxia-ischemia (HI) is the most common cause of brain injury in newborns and the survivors often develop cognitive and sensorimotor disabilities that undermine the quality of life. In the current study, we examined the effectiveness of flupirtine, a potassium channel opener, shown previously in an animal model to have strong anti-neonatal-seizure efficacy, to provide neuroprotection and alleviate later-life disabilities caused by neonatal hypoxic-ischemic injury. METHODS: The rats were treated with a single dose of flupirtine for 4 days following HI induction in 7-day-old rats. The first dose of flupirtine was given after the induction of HI and during the reperfusion period. The effect of treatment was examined on acute and chronic brain injury, motor functions, and cognitive abilities. RESULTS: Flupirtine treatment significantly reduced HI-induced hippocampal and cortical tissue loss at acute time point. Furthermore, at chronic time point, flupirtine reduced contralateral hippocampal volume loss and partially reversed learning and memory impairments but failed to improve motor deficits. CONCLUSION: The flupirtine treatment regimen used in the current study significantly reduced brain injury at acute time point in an animal model of neonatal hypoxic-ischemic encephalopathy. However, these neuroprotective effects were not persistent and only modest improvement in functional outcomes were observed at chronic time points.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Canales de Potasio/metabolismo , Aminopiridinas/uso terapéutico , Animales , Animales Recién Nacidos , Anticonvulsivantes/uso terapéutico , Encéfalo/efectos de los fármacos , Lesiones Encefálicas/metabolismo , Arterias Carótidas/patología , Cognición , Modelos Animales de Enfermedad , Fuerza de la Mano , Hipoxia , Masculino , Aprendizaje por Laberinto , Destreza Motora , Enfermedades del Sistema Nervioso/metabolismo , Neuroprotección , Fármacos Neuroprotectores/uso terapéutico , Calidad de Vida , Ratas , Convulsiones/tratamiento farmacológico
3.
Neurobiol Dis ; 124: 141-151, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30423475

RESUMEN

Epilepsy is a brain disorder characterized by a predisposition to suffer epileptic seizures. Acquired epilepsy might be the result of brain insults like head trauma, stroke, brain infection, or status epilepticus (SE) when one of these triggering injuries starts a transformative process known as epileptogenesis. There is some data to suggest that, during epileptogenesis, seizures themselves damage the brain but there is no conclusive evidence to demonstrate that spontaneous recurrent seizures themselves injure the brain. Our recent evidence indicates that calpain overactivation might be relevant for epileptogenesis. Here, we investigated if spontaneous recurrent seizures that occur during an early period of epileptogenesis show any correlation with the levels of calpain activation and/or expression. In addition, we also investigated a possible association between the occurrence of spontaneous seizures and increased levels of cell death, gliosis and inflammation (typical markers associated with epileptogenesis). We found that the number of spontaneous seizures detected prior to sample collection was correlated with altered calpain activity and expression. Moreover, the levels of hippocampal neurodegeneration were also correlated with seizure occurrence. Our findings suggest that, at least during early epileptogenesis, there is a correlation between seizure occurrence, calpain activity and neurodegeneration. Thus, this study opens the possibility that aberrant calpain reactivation by spontaneous seizures might contribute to the manifestation of future spontaneous seizures.


Asunto(s)
Calpaína/metabolismo , Epilepsia/metabolismo , Hipocampo/metabolismo , Neuronas/metabolismo , Convulsiones/metabolismo , Animales , Muerte Celular , Encefalitis/etiología , Encefalitis/metabolismo , Epilepsia/complicaciones , Epilepsia/patología , Gliosis , Hipocampo/patología , Masculino , Microglía/metabolismo , Ratas Sprague-Dawley , Convulsiones/complicaciones , Convulsiones/patología
4.
Neurobiol Dis ; 102: 1-10, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28237317

RESUMEN

In this study, we used the pilocarpine model of epilepsy to evaluate the involvement of calpain dysregulation on epileptogenesis. Detection of spectrin breakdown products (SBDPs, a hallmark of calpain activation) after induction of pilocarpine-induced status epilepticus (SE) and before appearance of spontaneous seizure suggested the existence of sustained calpain activation during epileptogenesis. Acute treatment with a cell permeable inhibitor of calpain, MDL-28170, resulted in a partial but significant reduction on seizure burden. The reduction on seizure burden was associated with a limited reduction on the generation of SBDPs but was correlated with a reduction in astrocytosis, microglia activation and cell sprouting. Together, these observations provide evidence for the role of calpain in epileptogenesis. In addition, provide proof-of-principle for the use of calpain inhibitors as a novel strategy to prevent epileptic seizures and its associated pathologies.


Asunto(s)
Anticonvulsivantes/farmacología , Dipéptidos/farmacología , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Glicoproteínas/farmacología , Animales , Calpaína/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/patología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Masculino , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Pilocarpina , Distribución Aleatoria , Ratas Sprague-Dawley , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Convulsiones/patología
5.
eNeuro ; 3(1)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27057559

RESUMEN

Brain-derived neurotrophic factor (BDNF) levels are elevated after status epilepticus (SE), leading to activation of multiple signaling pathways, including the janus kinase/signal transducer and activator of transcription pathway that mediates a decrease in GABAA receptor α1 subunits in the hippocampus (Lund et al., 2008). While BDNF can signal via its pro or mature form, the relative contribution of these forms to signaling after SE is not fully known. In the current study, we investigate changes in proBDNF levels acutely after SE in C57BL/6J mice. In contrast to previous reports (Unsain et al., 2008; Volosin et al., 2008; VonDran et al., 2014), our studies found that levels of proBDNF in the hippocampus are markedly elevated as early as 3 h after SE onset and remain elevated for 7 d. Immunohistochemistry studies indicate that seizure-induced BDNF localizes to all hippocampal subfields, predominantly in principal neurons and also in astrocytes. Analysis of the proteolytic machinery that cleaves proBDNF to produce mature BDNF demonstrates that acutely after SE there is a decrease in tissue plasminogen activator and an increase in plasminogen activator inhibitor-1 (PAI-1), an inhibitor of extracellular and intracellular cleavage, which normalizes over the first week after SE. In vitro treatment of hippocampal slices from animals 24 h after SE with a PAI-1 inhibitor reduces proBDNF levels. These findings suggest that rapid proBDNF increases following SE are due in part to reduced cleavage, and that proBDNF may be part of the initial neurotrophin response driving intracellular signaling during the acute phase of epileptogenesis.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Precursores de Proteínas/metabolismo , Procesamiento Proteico-Postraduccional , Estado Epiléptico/metabolismo , Animales , Astrocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Pilocarpina , Estado Epiléptico/inducido químicamente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA