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

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Biol Chem ; 288(19): 13831-41, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23553633

RESUMO

BACKGROUND: FMR1 CGG expansion repeats in the premutation range have not been linked to astrocyte pathophysiology. RESULTS: Premutation cortical astrocytes display decreased Glu transporter expression/activity and enhanced asynchronous Ca(2+) oscillations. CONCLUSION: Glu transport and Ca(2+) signaling defects in premutation astrocytes could contribute to FXTAS neuropathology. SIGNIFICANCE: Premutation astrocytes may have an etiological role in FXTAS neuropathology. Premutation CGG repeat expansions (55-200 CGG repeats; preCGG) within the fragile X mental retardation 1 (FMR1) gene can cause fragile X-associated tremor/ataxia syndrome. Defects in early neuronal migration and morphology, electrophysiological activity, and mitochondria trafficking have been described in a premutation mouse model, but whether preCGG mutations also affect astrocyte function remains unknown. PreCGG cortical astrocytes (∼170 CGG repeats) displayed 3-fold higher Fmr1 mRNA and 30% lower FMR1 protein (FMRP) when compared with WT. PreCGG astrocytes showed modest reductions in expression of glutamate (Glu) transporters GLT-1 and GLAST and attenuated Glu uptake (p < 0.01). Consistent with astrocyte cultures in vitro, aged preCGG mice cerebral cortex also displayed reduced GLAST and GLT-1 expression. Approximately 65% of the WT and preCGG cortical astrocytes displayed spontaneous asynchronous Ca(2+) oscillations. PreCGG astrocytes exhibited nearly 50% higher frequency of asynchronous Ca(2+) oscillations (p < 0.01) than WT, a difference mimicked by chronic exposure of WT astrocytes to l-trans-pyrrolidine-2,4-dicarboxylic acid (l-trans-PDC) or by partial suppression of GLAST using siRNA interference. Acute challenge with Glu augmented the frequency of Ca(2+) oscillations in both genotypes. Additionally, 10 µm Glu elicited a sustained intracellular Ca(2+) rise in a higher portion of preCGG astrocytes when compared with WT. Pharmacological studies showed that mGluR5, but not NMDA receptor, contributed to Glu hypersensitivity in preCGG astrocytes. These functional defects in preCGG astrocytes, especially in Glu signaling, may contribute to fragile X-associated tremor/ataxia syndrome neuropathology.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio , Proteína do X Frágil da Deficiência Intelectual/genética , Ácido Glutâmico/metabolismo , Expansão das Repetições de Trinucleotídeos , Animais , Astrócitos/efeitos dos fármacos , Ataxia/genética , Transporte Biológico , Células Cultivadas , Córtex Cerebral/patologia , Ácidos Dicarboxílicos/farmacologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Transportador 1 de Aminoácido Excitatório/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Feminino , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Ácido Glutâmico/fisiologia , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , N-Metilaspartato/farmacologia , Inibidores da Captação de Neurotransmissores/farmacologia , Pirrolidinas/farmacologia , Receptores de Glutamato/metabolismo , Receptores de Ácido Caínico/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Tremor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA