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.
Acta Neurobiol Exp (Wars) ; 82(3): 358-372, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36214718

RESUMO

Hypoxia inducible factor 1α (HIF­1α) has been reported to play a key role in protecting neurons from ischaemic injury. However, the exact molecular mechanisms remain largely unclear. PC12 cells were exposed to oxygen glucose deprivation/reoxygenation (OGD/R) conditions to mimic ischaemic injury in vitro. The expression of the HIF­1α mRNA, miR­20a­5p, and kinesin family member 5A (KIF5A) mRNA was tested using qRT-PCR. Levels of the HIF­1α, LC3I/II, P62, LAMP2, cathepsin B (CTSB) and KIF5A proteins were determined using western blotting. The CCK­8 assay was conducted to assess PC12 cell viability. DQ­Red­BSA and LysoSensor Green DND­189 dyes were employed to measure the proteolytic activity and pH of lysosomes, respectively. The interaction between miR­20a­5p and HIF­1α or KIF5A was verified by performing chromatin immunoprecipitation (ChIP) and/or dual­luciferase reporter assays. TUNEL staining was adopted to assess PC12 cell death. GFP­LC3 and RFP­GFP­LC3 probes were used to examine the autophagy status and autophagy flux of PC12 cells. A rat middle cerebral artery occlusion­reperfusion (MCAO/R) model was established to investigate the role of the HIF­1α/miR­20a­5p/KIF5A axis in ischaemic stroke in vivo. OGD/R exposure initiated PC12 cell autophagy and injury. HIF­1α expression was substantially increased in PC12 cells after OGD/R exposure. Overexpression of HIF­1α reversed the effects of OGD/R on reducing cell viability, blocking autophagy flux and inducing lysosome dysfunction. These rescue effects of HIF­1α depended on KIF5A. HIF­1α negatively regulated miR­20a­5p expression by targeting its promoter region, and miR­20a­5p directly targeted and negatively regulated the KIF5A mRNA. Overexpression of miR­20a­5p abolished the effects of HIF­1α on rescuing OGD/R­induced injury in PC12 cells. The effects of the HIF­1α/miR­20a­5p/KIF5A axis were verified in MCAO/R rats. HIF­1α protects PC12 cells from OGD/R­induced cell injury by regulating autophagy flux through the miR­20a­5p/KIF5A axis.


Assuntos
Isquemia Encefálica , Subunidade alfa do Fator 1 Induzível por Hipóxia , Cinesinas , MicroRNAs , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Animais , Apoptose , Autofagia , Catepsina B , Sobrevivência Celular , Glucose/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Cinesinas/genética , MicroRNAs/genética , Oxigênio , Células PC12 , RNA Mensageiro , Ratos , Traumatismo por Reperfusão/metabolismo
2.
J Neurochem ; 163(6): 500-516, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35997641

RESUMO

Ischemic stroke is a major global health issue. Ischemia and subsequent reperfusion results in stroke-related brain injury. Previous studies have demonstrated that nuclear-enriched abundant transcript 1 (NEATa and early growth response 1 (EGR1) are involved in ischemia reperfusion (IR) injury). In this study, we aimed to explore the roles of NEAT1/EGR1 axis as well as its downstream effector RNA binding motif protein 25 (RBM25) in cerebral IR injury. Oxygen-glucose deprivation/reperfusion (OGD/R) and middle cerebral artery occlusion (MCAO) were used to establish in vitro and in vivo models of cerebral IR injury, respectively. According to our data, NEAT1, EGR1, and RBM25 levels were elevated in OGD/R-exposed SK-N-SH and SH-SY5Y cells and cerebral cortex of MCAO mice. NEAT1, EGR1, or RBM25 knockdown effectively reduced infarct volumes and apoptosis, and improved neurological function. Mechanistically, NEAT1 directly interacted with EGR1, which restrained WW domain containing E3 ubiquitin protein ligase 1 (WWP1)-mediated ubiquitination of EGR1 and subsequently caused EGR1 accumulation. EGR1 bound to RBM25 promoter and transcriptionally activated RBM25. Rescue experiments indicated that RBM25 overexpression abolished the therapeutic effects of NEAT1 knockdown. In conclusion, this work identified a novel NEAT1/EGR1/RBM25 axis in potentiating brain injury after IR insults, suggesting a potential therapeutic target for ischemic stroke.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , AVC Isquêmico , MicroRNAs , Neuroblastoma , RNA Longo não Codificante , Traumatismo por Reperfusão , Humanos , Camundongos , Animais , RNA Longo não Codificante/genética , Traumatismo por Reperfusão/metabolismo , Infarto da Artéria Cerebral Média , Oxigênio/metabolismo , Apoptose/genética , Glucose/metabolismo , Motivos de Ligação ao RNA , Isquemia Encefálica/metabolismo , MicroRNAs/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
3.
Brain Res ; 1785: 147884, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35304105

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a major neurodegenerative disorder. The functions of lncRNA RMRP have been characterized mainly in various human cancers. However, the functional network of RMRP in AD progression remains unknown. METHODS: Human serum samples, AD transgenic (Tg) mice as well as SH-SY5Y cells were used in this study. The RNA expression patterns of RMRP, miR-3142 and TRIB3 were assessed by quantitative real-time PCR (qRT-PCR). Levels of apoptosis- or autophagy-associated biomarkers and TRIB3 level were evaluated using immunohistochemistry (IHC), western blotting or immunofluorescence assays, respectively. Bioinformatics methods and luciferase assays were used to predict and validate the interactions among RMRP, miR-3142, and TRIB3. Flow cytometry, TUNEL staining and EdU assays were used to examine the apoptosis and proliferation of neurons, respectively. RESULTS: The elevated RMRP and TRIB3 expressions and activation of autophagy were observed in AD. Knockdown of RMRP restrained neuronal apoptosis and autophagy activation in vitro and in vivo. Interestingly, TRIB3 overexpression reversed the biological effects of RMRP silencing on Aß1-42-induced cell apoptosis and autophagy. Further mechanistic analysis showed RMRP acted as a sponge of miR-3142 to elevate TRIB3 level. CONCLUSION: These data illustrated that knockdown of RMRP inhibited autophagy and apoptosis via regulating miR-3142/TRIB3 axis in AD, suggesting that inhibition of RMRP maybe a therapeutic strategy for AD.


Assuntos
Doença de Alzheimer , MicroRNAs , RNA Longo não Codificante , Doença de Alzheimer/genética , Animais , Apoptose , Autofagia , Linhagem Celular Tumoral , Camundongos , MicroRNAs/metabolismo , Neurônios/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA