Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Prog Neurobiol ; 214: 102280, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35525373

RESUMO

Alzheimer's disease (AD) is the most prevalent form of dementia in the old adult and characterized by progressive cognitive decline and neuronal damage. The mammalian Ste20-like kinase1/2 (MST1/2) is a core component in Hippo signaling, which regulates neural stem cell proliferation, neuronal death and neuroinflammation. However, whether MST1/2 is involved in the occurrence and development of AD remains unknown. In this study we reported that the activity of MST1 was increased with Aß accumulation in the hippocampus of 5xFAD mice. Overexpression of MST1 induced AD-like phenotype in normal mice and accelerated cognitive decline, synaptic plasticity damage and neuronal apoptosis in 2-month-old 5xFAD mice, but did not significantly affect Aß levels. Mechanistically, MST1 associated with p53 and promoted neuronal apoptosis by phosphorylation and activation of p53, while p53 knockout largely reversed MST1-induced AD-like cognitive deficits. Importantly, either genetic knockdown or chemical inactivation of MST1 could significantly improve cognitive deficits and neuronal apoptosis in 7-month-old 5xFAD mice. Our results support the idea that MST1-mediated neuronal apoptosis is an essential mechanism of cognitive deficits and neuronal loss for AD, and manipulating the MST1 activity as a potential strategy will shed light on clinical treatment for AD or other diseases caused by neuronal injury.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Proteínas Serina-Treonina Quinases , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Animais , Cognição , Disfunção Cognitiva/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Mamíferos/metabolismo , Camundongos , Proteína Supressora de Tumor p53/metabolismo
2.
Genes Brain Behav ; 19(6): e12678, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32468668

RESUMO

The activated mammalian Ste20-like serine/threonine kinases 1 (MST1) was found in the central nervous system diseases, such as cerebral ischemia, stroke and ALS, which were related with cognitions. The aim of this study was to examine the effect of elevated MST1 on memory functions in C57BL/6J mice. We also explored the underlying mechanism about the pattern alteration of neural oscillations, closely associated with cognitive dysfunctions, at different physiological rhythms, which were related to a wide range of basic and higher-level cognitive activities. A mouse model of the adeno-associated virus (AAV)-mediated overexpression of MST1 was established. The behavioral experiments showed that spatial memory was significantly damaged in MST1 mice. The distribution of either theta or gamma power was clearly disturbed in MST1 animals. Moreover, the synchronization in both theta and gamma rhythms, and theta-gamma cross-frequency coupling were significantly weakened in MST1 mice. In addition, the expressions of GABAA receptor, GAD67 and parvalbumin (PV) were obviously increased in MST1 mice. Meanwhile, blocking MST1 activity could inhibit the activation of FOXO3a and YAP. The above data suggest that MST1-overexpression may induce memory impairments via disturbing the patterns of neural activities, which is possibly associated with the abnormal GABAergic expression level.


Assuntos
Ritmo Gama , Proteínas Serina-Treonina Quinases/genética , Memória Espacial , Ritmo Teta , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Cognição , Proteína Forkhead Box O3/metabolismo , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Parvalbuminas/genética , Parvalbuminas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Regulação para Cima , Proteínas de Sinalização YAP
3.
Artigo em Inglês | MEDLINE | ID: mdl-30391307

RESUMO

The effect of prenatal stress (PS) on offspring's behavior was reported previously. Several studies attempted to reveal the mechanisms of PS on synaptic and molecular levels. However, the influences of PS on neural oscillations and their interaction in hippocampus are still unknown. In the present study, a PS rat model was established by using restraint stress. The local-field potentials (LFPs) were simultaneously recorded from the hippocampal CA3 and CA1 regions in young, adolescent and early-adult offspring rats. After that, LFPs were analyzed by analytic algorithms for estimating power spectrum, coherence, phase synchronization and cross-frequency coupling. The results showed that there was a significant influence of PS on power distribution from 1 to 100 Hz during different developmental stages. The identical-frequency synchronizations between CA3 and CA1 regions, including coherence and phase synchronization, were significantly reduced in PS rats compared to that in normal rats. Meanwhile, PS significantly impaired the cross-frequency coupling strength between theta and gamma rhythms. These data show that PS alters the neural oscillations and their interaction on the hippocampal CA3-CA1 pathway, which may be associated with the behavior outcomes and synaptic impairments previously reported in PS offspring rats. Moreover, the significant PS × age interactions between the effects of PS and age have been only found in the cross-frequency coupling, implying that the cross-frequency coupling more appropriately reflects the differences of the behavioral effects of PS in different postnatal ages.


Assuntos
Hipocampo/crescimento & desenvolvimento , Hipocampo/fisiopatologia , Efeitos Tardios da Exposição Pré-Natal , Estresse Psicológico , Animais , Ondas Encefálicas , Modelos Animais de Doenças , Feminino , Masculino , Vias Neurais/crescimento & desenvolvimento , Vias Neurais/fisiopatologia , Periodicidade , Gravidez , Complicações na Gravidez/fisiopatologia , Distribuição Aleatória , Ratos Wistar , Restrição Física , Estresse Psicológico/fisiopatologia
4.
J Neurogenet ; 31(4): 337-343, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29098922

RESUMO

We recently reported that presenilin-1 (PS1) induced an increase of U1 snRNA expression accompanied with the change of amyloid precursor protein expression, ß-amyloid level and cell death. In the present study, our data showed that both overexpression and knockdown of U1 snRNA could cause the loss in the function of U1 snRNA and resulted in PCPA as well as the same downstream phenomena including the expression changes of genes specific to AD, tau hyperphosphorylation on the site of Thr212, the decrease of acetylated α-tubulin, the reduction of cell viability and upregulation of RIPK1, RIPK3 and caspase8. These findings not only helped researchers better understand the functions of U1 snRNA, but also paved the way to reveal the mechanisms of AD from a different point of view and may find a new therapeutic target for the disease.


Assuntos
Doença de Alzheimer/genética , RNA Nuclear Pequeno/genética , Spliceossomos/genética , Doença de Alzheimer/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Técnicas de Silenciamento de Genes , Humanos , Fosforilação , RNA Nuclear Pequeno/metabolismo , Spliceossomos/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas tau/metabolismo
5.
Exp Neurol ; 298(Pt A): 68-78, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28882411

RESUMO

Prenatal stress (PS) brings numerous outcomes on offspring, including anxiety, depression-like behavior and other cognitive disorder. In this study, a rat model of PS was established by using restraint stress for 45min three times per day from the 15th to 21st day of pregnancy. Behavioral tests, including open field test (OPT), elevated plus-maze (EPM) and Morris water-maze (MWM), were performed in adolescent male-offspring. The bidirectional synaptic plasticity, including long-term potential (LTP) and depotentiation (DEP), from the hippocampal Schaffer collaterals to CA1 region was subsequently measured. Furthermore, Western blot assay, immunofluorescence staining and hematoxylin-eosin (HE) staining were employed. The MWM test showed that the cognitive flexibility was remarkably damaged in PS offspring. Meanwhile, PS considerably aggravated the anxiety and depression-like behavior in OPT and EPM. Both LTP and DEP were significantly inhibited by PS. Furthermore, PS considerably altered the expression of synaptic-related proteins NR2A, NR2B and PSD-95 in adolescent male-offspring. Interestingly, PS significantly elevated the autophagy level in the hippocampus of male-offspring. In order to investigate the role of autophagy on the negative impacts of PS in adolescent male-offspring, both in vitro and in vivo studies were performed. It was found that autophagy inhibitors significantly eliminated the alterations in gene expression induced by corticosterone. The results suggest that regulating autophagy may become a new targeted therapy to relieve the damage induced by PS in adolescent male-offspring.


Assuntos
Autofagia/fisiologia , Transtornos Cognitivos/fisiopatologia , Plasticidade Neuronal/fisiologia , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Estresse Psicológico/fisiopatologia , Fatores Etários , Animais , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/psicologia , Feminino , Potenciação de Longa Duração/fisiologia , Masculino , Aprendizagem em Labirinto/fisiologia , Células PC12 , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/psicologia , Distribuição Aleatória , Ratos , Ratos Wistar , Fatores Sexuais , Estresse Psicológico/complicações , Estresse Psicológico/psicologia
6.
J Mol Neurosci ; 62(3-4): 269-275, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28577205

RESUMO

U1 small nuclear RNA (snRNA) is selectively enriched in 100% of familial Alzheimer's disease (AD) resulting from presenilin1 (PS1) and amyloid precursor protein (APP) mutations. However, it remains unknown what gene or protein cause the U1 snRNA overexpression and then resulted in AD. Using SH-SY5Y cells, we discovered that PS1 induced the overexpression of U1 snRNA, which increased the production of Aß. Moreover, the U1 snRNA overexpression induced the upregulation of apoe and clu transcripts. In addition, the levels of phosphorylation of tau protein at Thr212 were significantly elevated in U1 snRNA overexpression cells. Immunofluorescence using antibodies reactive with the phosphorylation of tau at Thr212 site demonstrated the hyperphosphorylated tau localization with α-tubulin, the main component of cytoskeleton. Importantly, the increased levels of Bax, Bcl2, and Bax/Bcl2 ratio showed that the overexpression of U1 snRNA could cause cell apoptosis. Conclusively, these findings suggest that PS1 considerably increases the level of U1snRNA accompanied with the adverse change of Aß level, AD-related tau cytoskeletal pathology, and cell apoptosis.


Assuntos
Doença de Alzheimer/metabolismo , Presenilina-1/metabolismo , RNA Nuclear Pequeno/genética , Doença de Alzheimer/genética , Animais , Apoptose , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células PC12 , Fosforilação , Presenilina-1/genética , Processamento de Proteína Pós-Traducional , RNA Nuclear Pequeno/metabolismo , Ratos , Proteínas tau/metabolismo
7.
Mol Neurobiol ; 54(6): 4644-4658, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27405470

RESUMO

The aim of this study was to examine if nicotine was able to improve cognition deficits in a mouse model of chronic mild stress. Twenty-four male C57BL/6 mice were divided into three groups: control, stress, and stress with nicotine treatment. The animal model was established by combining chronic unpredictable mild stress (CUMS) and isolated feeding. Mice were exposed to CUMS continued for 28 days, while nicotine (0.2 mg/kg) was also administrated for 28 days. Weight and sucrose consumption were measured during model establishing period. The anxiety and behavioral despair were analyzed using the forced swim test (FST) and open-field test (OFT). Spatial cognition was evaluated using Morris water maze (MWM) test. Following behavioral assessment, both long-term potentiation (LTP) and depotentiation (DEP) were recorded in the hippocampal dentate gyrus (DG) region. Both synaptic and Notch1 proteins were measured by Western. Nicotine increased stressed mouse's sucrose consumption. The MWM test showed that spatial learning and reversal learning in stressed animals were remarkably affected relative to controls, whereas nicotine partially rescued cognitive functions. Additionally, nicotine considerably alleviated the level of anxiety and the degree of behavioral despair in stressed mice. It effectively mitigated the depression-induced impairment of hippocampal synaptic plasticity, in which both the LTP and DEP were significantly inhibited in stressed mice. Moreover, nicotine enhanced the expression of synaptic and Notch1 proteins in stressed animals. The results suggest that nicotine ameliorates the depression-like symptoms and improves the hippocampal synaptic plasticity closely associated with activating transmembrane ion channel receptors and Notch signaling components. Graphical Abstract ᅟ.


Assuntos
Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/etiologia , Plasticidade Neuronal , Nicotina/uso terapêutico , Estresse Psicológico/complicações , Animais , Peso Corporal/efeitos dos fármacos , Comportamento de Escolha , Doença Crônica , Transtornos Cognitivos/fisiopatologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato Descarboxilase/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal/efeitos dos fármacos , Nicotina/farmacologia , Receptores de N-Metil-D-Aspartato/metabolismo , Sacarose , Natação
8.
Oncotarget ; 8(68): 112867-112874, 2017 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-29348872

RESUMO

U1 small nuclear RNA (U1 snRNA), as one of the most abundant ncRNAs in human cells, plays an important role in splicing of pre-mRNAs. Compared to previous studies which have focused on the primary function of U1 snRNA and the neurodegenerative diseases caused by abnormalities of U1 snRNA, this study is to investigate how U1 snRNA over-expression affects the expression of mammal genes on a genome-wide scale. By comparing the gene expression profiles of U1 snRNA over-expressed cells with those of their controls using microarray experiments, 916 genes or loci were identified significantly Differentially Expressed (DE). These 595 up-regulated DE genes and 321 down-regulated DE genes were analyzed using annotations from GO categories and pathways from the KEGG database. As a result, three of 12 enriched pathways were well-known cancer pathways, while the other nine pathways were associated to cancers in previous studies. The further analysis of 73 genes involved in 12 pathways suggested that U1 snRNA could regulate cancer gene expression. The microarray data under the GEO Series accession number GSE84304 is available in the NCBI GEO database.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA