Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Learn Mem ; 97(1): 97-104, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22023893

RESUMEN

Nitric oxide (NO) is synthesized as a result of N-methyl-d-asparate (NMDA) receptor activation, it acts as an retrograde neurotransmitter freely diffusing across cell membranes interacting with its targets in a non-synaptic manner. Consequently, NO has been described as an extension of NMDA receptor activation. The targets of NO include cellular components within the basolateral complex of the amygdala (BLA) that are necessary for the consolidation of conditioned fear as well as targets that can significantly modulate neurotransmission associated with its expression. Given that both are NMDA receptor associated processes, this implies that NO may be an important intermediary of NMDA receptor activation and both fear memory consolidation and expression. The current study sought to examine this using visual fear conditioning and fear potentiated startle. Three experiments were conducted, rats received intra-BLA microinfusions of the global nitric oxide synthase inhibitor l-NAME either prior to fear conditioning, or expression of learned fear. Furthermore, NO's ability to modulate a NMDA receptor independent fear process was assessed by microinfusing l-NAME into the BLA prior to examination of the shock sensitization of the acoustic startle affect. The results indicated that NO was, indeed, required for both the consolidation and expression of learned fear, whereas it was not required for NMDA independent shock enhanced startle responding. This study illustrates that NO plays a pivotal role in the examined NMDA associated fear processes.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Condicionamiento Clásico/fisiología , Miedo/fisiología , Óxido Nítrico/biosíntesis , Reflejo de Sobresalto/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Condicionamiento Clásico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Miedo/efectos de los fármacos , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Ratas , Ratas Wistar , Reflejo de Sobresalto/efectos de los fármacos
2.
J Neurosci ; 28(47): 12383-95, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-19020031

RESUMEN

The activity-regulated cytoskeletal-associated protein (Arc/Arg3.1) is an immediate early gene that has been widely implicated in hippocampal-dependent learning and memory and is believed to play an integral role in synapse-specific plasticity. Here, we examined the role of Arc/Arg3.1 in amygdala-dependent Pavlovian fear conditioning. We first examined the regulation of Arc/Arg3.1 mRNA and protein after fear conditioning and LTP-inducing stimulation of thalamic inputs to the lateral amygdala (LA). Quantitative real-time PCR, in situ hybridization, Western blotting and immunohistochemistry revealed a significant upregulation of Arc/Arg3.1 mRNA and protein in the LA relative to controls. In behavioral experiments, intra-LA infusion of an Arc/Arg3.1 antisense oligodeoxynucleotide (ODN) was observed to be anatomically restricted to the LA, taken up by LA cells, and to promote significant knockdown of Arc/Arg3.1 protein. Rats given intra-LA infusions of multiple doses of the Arc/Arg3.1 ODN showed an impairment of LTM (tested approximately 24 later), but no deficit in STM (tested 3 h later) relative to controls infused with scrambled ODN. Finally, to determine whether upregulation of Arc/Arg3.1 occurs downstream of ERK/MAPK activation, we examined Arc/Arg3.1 expression in rats given intra-LA infusion of the MEK inhibitor U0126. Relative to vehicle controls, infusion of U0126 impaired training-induced increases in Arc/Arg3.1 expression. These findings suggest that Arc/Arg3.1 expression in the amygdala is required for fear memory consolidation, and further suggest that Arc/Arg3.1 regulation in the LA is downstream of the ERK/MAPK signaling pathway.


Asunto(s)
Amígdala del Cerebelo/fisiología , Condicionamiento Clásico/fisiología , Proteínas del Citoesqueleto/metabolismo , Miedo , Memoria/fisiología , Proteínas del Tejido Nervioso/metabolismo , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Butadienos/farmacología , Condicionamiento Clásico/efectos de los fármacos , Proteínas del Citoesqueleto/genética , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Estimulación Eléctrica/métodos , Inhibidores Enzimáticos/farmacología , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/fisiología , Potenciación a Largo Plazo/efectos de la radiación , Masculino , Memoria/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Nitrilos/farmacología , Oligodesoxirribonucleótidos Antisentido/farmacología , ARN Mensajero , Ratas , Factores de Tiempo
3.
Artículo en Inglés | MEDLINE | ID: mdl-20161806

RESUMEN

In both invertebrate and vertebrate models of synaptic plasticity, signaling via the putative "retrograde messenger" nitric oxide (NO) has been hypothesized to serve as a critical link between functional and structural alterations at pre- and postsynaptic sites. However, while in vitro models of synaptic plasticity have consistently implicated NO signaling in linking postsynaptic induction mechanisms with accompanying presynaptic changes, a convincing role of such "retrograde signaling" in mammalian memory formation has remained elusive. Using auditory Pavlovian fear conditioning, we show that synaptic plasticity and NO signaling in the lateral nucleus of the amygdala (LA) regulate the expression of the ERK-driven immediate early gene early growth response gene I (EGR-1) in regions of the auditory thalamus that are presynaptic to the LA. Further, antisense knockdown of EGR-1 in the auditory thalamus impairs both fear memory consolidation and the training-induced elevation of two presynaptically localized proteins in the LA. These findings indicate that synaptic plasticity and NO signaling in the LA during auditory fear conditioning promote alterations in ERK-driven gene expression in auditory thalamic neurons that are required for both fear memory consolidation as well as presynaptic correlates of fear memory formation in the LA, and provide general support for a role of NO as a "retrograde signal" in mammalian memory formation.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA