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










Base de dados
Intervalo de ano de publicação
1.
Int J Neuropsychopharmacol ; 22(6): 372-382, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31038173

RESUMO

BACKGROUND: Although depression is the leading cause of disability worldwide, its pathophysiology is poorly understood. Our previous study showed that hippocampal peroxisome proliferator-activated receptor δ (PPARδ) overexpression displays antidepressive effect and enhances hippocampal neurogenesis during chronic stress. Herein, we further extended our curiosity to investigate whether downregulating PPARδ could cause depressive-like behaviors through downregulation of neurogenesis. METHODS: Stereotaxic injection of lentiviral vector, expressing short hairpin RNA complementary to the coding exon of PPARδ, was done into the bilateral dentate gyri of the hippocampus, and the depression-like behaviors were observed in mice. Additionally, hippocampal neurogenesis, brain-derived neurotrophic factor and cAMP response element-binding protein were measured both in vivo and in vitro. RESULTS: Hippocampal PPARδ knockdown caused depressive-like behaviors and significantly decreased neurogenesis, neuronal differentiation, levels of mature brain-derived neurotrophic factor and phosphorylated cAMP response element-binding protein in the hippocampus. In vitro study further confirmed that PPARδ knockdown could inhibit proliferation and differentiation of neural stem cells. Furthermore, these effects were mimicked by repeated systemic administration of a PPARδ antagonist, GSK0660 (1 or 3 mg/kg i.p. for 21 d). CONCLUSIONS: These findings suggest that downregulation of hippocampal PPARδ is associated with depressive behaviors in mice through an inhibitory effect on cAMP response element-binding protein/brain-derived neurotrophic factor-mediated adult neurogenesis in the hippocampus, providing new insights into the pathogenesis of depression.


Assuntos
Comportamento Animal/fisiologia , Giro Denteado/metabolismo , Depressão/fisiopatologia , Neurogênese/efeitos dos fármacos , PPAR delta/fisiologia , Animais , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteína de Ligação a CREB/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Giro Denteado/efeitos dos fármacos , Depressão/induzido quimicamente , Regulação para Baixo/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Células-Tronco Neurais/efeitos dos fármacos , PPAR delta/genética , RNA Interferente Pequeno/farmacologia , Sulfonas/farmacologia , Tiofenos/farmacologia
2.
Int J Neuropsychopharmacol ; 19(1)2015 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-26362775

RESUMO

BACKGROUND: Emerging data have demonstrated that peroxisome proliferator-activated receptor δ (PPARδ) activation confers a potentially neuroprotective role in some neurodegenerative diseases. However, whether PPARδ is involved in depression is unknown. METHODS: In this study, PPARδ was firstly investigated in the chronic mild stress (CMS) and learned helplessness (LH) models of depression. The changes in depressive behaviors and hippocampal neurogenesis were investigated after PPARδ overexpression by microinfusion of the lentiviral vector, containing the coding sequence of mouse PPARδ (LV-PPARδ), into the bilateral dentate gyri of the hippocampus or PPARδ activation by repeated systemic administration of PPARδ agonist GW0742 (5 or 10mg/kg.d, i.p., for 21 d). RESULTS: We found that both CMS and LH resulted in a significant decrease in the PPARδ expression in the hippocampi of mice, and this change was reversed by treatment with the antidepressant fluoxetine. PPARδ overexpression and PPARδ activation each suppressed the CMS- and LH-induced depressive-like behavior and produced an antidepressive effect. In vivo or in vitro studies also showed that both overexpression and activation of PPARδ enhanced proliferation or differentiation of neural stem cells in the hippocampi of mice. CONCLUSIONS: These results suggest that hippocampal PPARδ upregulation represses stress-induced depressive behaviors, accompanied by enhancement of neurogenesis.


Assuntos
Depressão/complicações , Depressão/patologia , Hipocampo/metabolismo , Neurogênese/fisiologia , PPAR delta/metabolismo , Estresse Psicológico/complicações , Animais , Diferenciação Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Elevação dos Membros Posteriores , Masculino , Camundongos , Camundongos Endogâmicos ICR , Células-Tronco Neurais/fisiologia , PPAR delta/genética , Fosfopiruvato Hidratase/metabolismo , Sincalida/metabolismo , Tiazóis/farmacologia , Fatores de Tempo , Transdução Genética
3.
Neurobiol Aging ; 35(3): 590-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24269024

RESUMO

Accumulation of amyloid-ß (Aß) is thought to be associated with the progressive neuronal death observed in Alzheimer's disease, but the mechanisms underlying neurotoxicity triggered by Aß remain elusive. In the current study, we investigated the roles of cysteinyl leukotriene receptor 1 (CysLT1R) in Aß1-42-induced neurotoxicity in vitro or in vivo. In vitro exposure of mouse primary neurons to Aß1-42 caused a gradual increases in CysLT1R expression. In vivo bilateral intrahippocampal injection of Aß1-42 also elicited time-dependent increases of CysLT1R expression in the hippocampus and cortex of mice. The CysLT1R antagonist pranlukast not only reversed Aß1-42-induced upregulation of CysLT1R, but also suppressed Aß1-42-triggered neurotoxicity evidenced by enhanced nuclear factor-kappa B p65, activated caspase-3, decreased B-cell lymphoma-2 and cell viability and impaired memory. Furthermore, chronic treatment with pranlukast produced similar beneficial effects on memory behavior and hippocampal long-term potentiation to memantine or donepezil in intrahippocampal Aß1-42-injected mice. Our data indicate that CysLT1R is involved in Aß1-42-induced neurotoxicity, and that blockade of CysLT1R, such as application of CysLT1R antagonist, could be a novel and promising strategy for the treatment of Alzheimer's disease.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Córtex Cerebral/metabolismo , Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fragmentos de Peptídeos/toxicidade , Receptores de Leucotrienos/genética , Receptores de Leucotrienos/fisiologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/psicologia , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Cromonas/farmacologia , Cromonas/uso terapêutico , Hipocampo/citologia , Antagonistas de Leucotrienos/farmacologia , Antagonistas de Leucotrienos/uso terapêutico , Masculino , Memória , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Terapia de Alvo Molecular , NF-kappa B/metabolismo , Neurônios/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Leucotrienos/metabolismo , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...