Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuroscience ; 289: 242-50, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25595979

RESUMEN

Neuroligin 1 (NLGN1) is a postsynaptic adhesion molecule that determines N-methyl-d-aspartate receptor (NMDAR) function and cellular localization. Our recent work showed that Nlgn1 knockout (KO) mice cannot sustain neuronal activity occurring during wakefulness for a prolonged period of time. Since NMDAR-dependent neuronal activity drives an important vascular response, we used multispectral optical imaging to determine if the hemodynamic response to neuronal stimulation is modified in Nlgn1 KO mice. We observed that Nlgn1 KO mice show a 10% lower response rate to forepaw electrical stimulation compared to wild-type (WT) and heterozygote (HET) littermates on both the contra- and ipsilateral sides of the somatosensory cortex. Moreover, Nlgn1 mutant mice showed an earlier oxyhemoglobin peak response that tended to return to baseline faster than in WT mice. Analysis of the time course of the hemodynamic response also showed that HET mice express a faster dynamics of cerebrovascular response in comparison to WT. Taken together, these data are indicative of an altered immediate response of the brain to peripheral stimulation in Nlgn1 KO mice, and suggest a role for NLGN1 in the regulation of cerebrovascular responses.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/deficiencia , Circulación Cerebrovascular/fisiología , Hemodinámica/fisiología , Corteza Somatosensorial/fisiopatología , Animales , Moléculas de Adhesión Celular Neuronal/genética , Estimulación Eléctrica , Miembro Anterior/fisiología , Masculino , Ratones Noqueados , Músculo Esquelético/fisiología , Imagen Óptica , Oxihemoglobinas/metabolismo , Propiocepción , Corteza Somatosensorial/irrigación sanguínea , Factores de Tiempo
2.
Transl Psychiatry ; 4: e347, 2014 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-24448209

RESUMEN

Sleep is critical for normal brain function and mental health. However, the molecular mechanisms mediating the impact of sleep loss on both cognition and the sleep electroencephalogram remain mostly unknown. Acute sleep loss impacts brain gene expression broadly. These data contributed to current hypotheses regarding the role for sleep in metabolism, synaptic plasticity and neuroprotection. These changes in gene expression likely underlie increased sleep intensity following sleep deprivation (SD). Here we tested the hypothesis that epigenetic mechanisms coordinate the gene expression response driven by SD. We found that SD altered the cortical genome-wide distribution of two major epigenetic marks: DNA methylation and hydroxymethylation. DNA methylation differences were enriched in gene pathways involved in neuritogenesis and synaptic plasticity, whereas large changes (>4000 sites) in hydroxymethylation where observed in genes linked to cytoskeleton, signaling and neurotransmission, which closely matches SD-dependent changes in the transcriptome. Moreover, this epigenetic remodeling applied to elements previously linked to sleep need (for example, Arc and Egr1) and synaptic partners of Neuroligin-1 (Nlgn1; for example, Dlg4, Nrxn1 and Nlgn3), which we recently identified as a regulator of sleep intensity following SD. We show here that Nlgn1 mutant mice display an enhanced slow-wave slope during non-rapid eye movement sleep following SD but this mutation does not affect SD-dependent changes in gene expression, suggesting that the Nlgn pathway acts downstream to mechanisms triggering gene expression changes in SD. These data reveal that acute SD reprograms the epigenetic landscape, providing a unique molecular route by which sleep can impact brain function and health.


Asunto(s)
Corteza Cerebral/metabolismo , Metilación de ADN/fisiología , Genoma/genética , Plasticidad Neuronal/genética , Privación de Sueño/metabolismo , Transcriptoma/genética , Animales , Moléculas de Adhesión Celular Neuronal/genética , Corteza Cerebral/fisiopatología , Metilación de ADN/genética , Electroencefalografía , Epigénesis Genética/genética , Epigénesis Genética/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Privación de Sueño/fisiopatología , Fases del Sueño/genética , Fases del Sueño/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...