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.
PLoS Biol ; 21(6): e3002142, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37289842

RESUMO

Rab26 is known to regulate multiple membrane trafficking events, but its role in insulin secretion in pancreatic ß cells remains unclear despite it was first identified in the pancreas. In this study, we generated Rab26-/- mice through CRISPR/Cas9 technique. Surprisingly, insulin levels in the blood of the Rab26-/- mice do not decrease upon glucose stimulation but conversely increase. Deficiency of Rab26 promotes insulin secretion, which was independently verified by Rab26 knockdown in pancreatic insulinoma cells. Conversely, overexpression of Rab26 suppresses insulin secretion in both insulinoma cell lines and isolated mouse islets. Islets overexpressing Rab26, upon transplantation, also failed to restore glucose homeostasis in type 1 diabetic mice. Immunofluorescence microscopy revealed that overexpression of Rab26 results in clustering of insulin granules. GST-pulldown experiments reveal that Rab26 interacts with synaptotagmin-1 (Syt1) through directly binding to its C2A domain, which interfering with the interaction between Syt1 and SNAP25, and consequently inhibiting the exocytosis of newcomer insulin granules revealed by TIRF microscopy. Our results suggest that Rab26 serves as a negative regulator of insulin secretion, via suppressing insulin granule fusion with plasma membrane through sequestering Syt1.


Assuntos
Diabetes Mellitus Experimental , Células Secretoras de Insulina , Insulinoma , Ilhotas Pancreáticas , Neoplasias Pancreáticas , Animais , Camundongos , Diabetes Mellitus Experimental/metabolismo , Exocitose/fisiologia , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Ilhotas Pancreáticas/metabolismo , Neoplasias Pancreáticas/metabolismo
2.
Cell Death Dis ; 12(4): 284, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33731709

RESUMO

Rab proteins play crucial roles in membrane trafficking. Some Rab proteins are implicated in cancer development through regulating protein sorting or degradation. In this study, we found that the expression of Rab26 is suppressed in the aggressive breast cancer cells as compared to the levels in non-invasive breast cancer cells. Over-expression of Rab26 inhibits cell migration and invasion, while Rab26 knockdown significantly promotes the migration and invasion of breast cancer cells. Rab26 reduces focal adhesion association of Src kinase and induces endosomal translocation of Src. Further experiments revealed that Rab26 mediates the autophagic degradation of phosphorylated Src through interacting with ATG16L1, consequently, resulting in the suppression of the migration and invasion ability of breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Proteínas rab de Ligação ao GTP/metabolismo , Quinases da Família src/metabolismo , Autofagia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Humanos , Fosforilação
3.
Biochem Biophys Res Commun ; 534: 240-247, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33272569

RESUMO

Mild hypothermia is a well-established technique for alleviating neurological injuries in clinical surgery. RNA-binding protein motif 3 (RBM3) has been identified as a crucial factor in mediating hypothermic neuroprotection, providing its induction as a promising strategy for mimicking therapeutic hypothermia. However, little is known about molecular control of RBM3 and signaling pathways affected by hypothermia. In the present study, human SH-SY5Y neuroblastoma cells were used as a neural cell model. Screening of signaling pathways showed that cold exposure led to inactivation of ERK and AMPK pathways, and activation of FAK and PLCγ pathways, with activities of p38, JNK and AKT pathways moderately changed. Next, various small molecule inhibitors specific to these signaling pathways were applied. Interestingly, only FAK-specific inhibitor exhibited a significant inhibitory effect on hypothermia-induced RBM3 gene transcription and protein expression. Likewise, FAK silencing using siRNA technique significantly abrogated the induction of RBM3 by hypothermia. Moreover, FAK inhibition accounted for an inactivation of Src, a known kinase downstream of FAK. Next, either the silencing of Src by siRNA or its inactivation by a chemical inhibitor, strongly blocked the induction of RBM3 by cooling. Notably, in HEK293 and PC12 cells, FAK/Src activation was also shown to be indispensable for hypothermia-stimulated RBM3 expression. Lastly, the CCK8 and Western blot assays showed that both FAK/Src inacitivation and their knockdown substantially abrogate the neuroprotective effects of mild hypothermia against rotenone in SH-SY5Y cells. These data suggest that FAK/Src signaling axis regulates the transcription of Rbm3 gene and mediates neuroprotective effects of mild hypothermia.


Assuntos
Temperatura Baixa , Quinase 1 de Adesão Focal/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Proteínas de Ligação a RNA/biossíntese , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Células HEK293 , Humanos , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Neurônios/enzimologia , Proteínas de Ligação a RNA/genética , Ratos , Rotenona/toxicidade , Transcrição Gênica
4.
J Cell Mol Med ; 23(10): 7010-7020, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31436914

RESUMO

Mild hypothermia and its key product, cold-inducible protein RBM3, possess robust neuroprotective effects against various neurotoxins. However, we previously showed that mild hypothermia fails to attenuate the neurotoxicity from MPP+ , one of typical neurotoxins related to the increasing risk of Parkinson disease (PD). To better understand the role of mild hypothermia and RBM3 in PD progression, another known PD-related neurotoxin, rotenone (ROT) was utilized in this study. Using immunoblotting, cell viability assays and TUNEL staining, we revealed that mild hypothermia (32°C) significantly reduced the apoptosis induced by ROT in human neuroblastoma SH-SY5Y cells, when compared to normothermia (37°C). Meanwhile, the overexpression of RBM3 in SH-SY5Y cells mimicked the neuroprotective effects of mild hypothermia on ROT-induced cytotoxicity. Upon ROT stimulation, MAPK signalling like p38, JNK and ERK, and AMPK and GSK-3ß signalling were activated. When RBM3 was overexpressed, only the activation of p38, JNK and ERK signalling was inhibited, leaving AMPK and GSK-3ß signalling unaffected. Similarly, mild hypothermia also inhibited the activation of MAPKs induced by ROT. Lastly, it was demonstrated that the MAPK (especially p38 and ERK) inhibition by their individual inhibitors significantly decreased the neurotoxicity of ROT in SH-SY5Y cells. In conclusion, these data demonstrate that RBM3 mediates mild hypothermia-related neuroprotection against ROT by inhibiting the MAPK signalling of p38, JNK and ERK.


Assuntos
Temperatura Baixa , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Neurotoxinas/toxicidade , Proteínas de Ligação a RNA/metabolismo , Rotenona/toxicidade , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Citoproteção/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Hipotermia Induzida
5.
BMJ Open ; 9(5): e023724, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-31101692

RESUMO

INTRODUCTION: Left ventricular ejection fraction (LVEF) ≤35%, as current significant implantable cardioverter-defibrillator (ICD) indication for primary prevention of sudden cardiac death (SCD) in heart failure (HF) patients, has been widely recognised to be inefficient. Improvement of patient selection for low LVEF (≤35%) is needed to optimise deployment of ICD. Most of the existing prediction models are not appropriate to identify ICD candidates at high risk of SCD in HF patients with low LVEF. Compared with traditional statistical analysis, machine learning (ML) can employ computer algorithms to identify patterns in large datasets, analyse rules automatically and build both linear and non-linear models in order to make data-driven predictions. This study is aimed to develop and validate new models using ML to improve the prediction of SCD in HF patients with low LVEF. METHODS AND ANALYSIS: We will conduct a retroprospective, multicentre, observational registry of Chinese HF patients with low LVEF. The HF patients with LVEF ≤35% after optimised medication at least 3 months will be enrolled in this study. The primary endpoints are all-cause death and SCD. The secondary endpoints are malignant arrhythmia, sudden cardiac arrest, cardiopulmonary resuscitation and rehospitalisation due to HF. The baseline demographic, clinical, biological, electrophysiological, social and psychological variables will be collected. Both ML and traditional multivariable Cox proportional hazards regression models will be developed and compared in the prediction of SCD. Moreover, the ML model will be validated in a prospective study. ETHICS AND DISSEMINATION: The study protocol has been approved by the Ethics Committee of the First Affiliated Hospital of Nanjing Medical University (2017-SR-06). All results of this study will be published in international peer-reviewed journals and presented at relevant conferences. TRIAL REGISTRATION NUMBER: ChiCTR-POC-17011842; Pre-results.


Assuntos
Morte Súbita Cardíaca/etiologia , Insuficiência Cardíaca/complicações , Aprendizado de Máquina , Sistema de Registros , Projetos de Pesquisa , China , Insuficiência Cardíaca/fisiopatologia , Humanos , Estudos Multicêntricos como Assunto , Estudos Prospectivos , Estudos Retrospectivos , Volume Sistólico , Função Ventricular Esquerda
6.
Med Sci Monit ; 24: 8870-8877, 2018 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-30531686

RESUMO

BACKGROUND Angiogenesis plays a crucial role in myocardial infarction (MI) treatment by ameliorating myocardial remodeling, thus improving cardiac function and preventing heart failure. Muscone has been reported to have beneficial effects on cardiac remodeling in MI mice. However, the effects of muscone on angiogenesis in MI mice and its underlying mechanisms remain unknown. MATERIAL AND METHODS Mice were randomly divided into sham, MI, and MI+muscone groups. The MI mouse model was established by ligating the left anterior descending coronary artery. Mice in the sham group received the same procedure except for ligation. Mice were administered muscone or an equivalent volume of saline for 4 consecutive weeks. Cardiac function was evaluated by echocardiograph after MI for 2 and 4 weeks. Four weeks later, all mice were sacrificed and Masson's trichrome staining was used to assess myocardial fibrosis. Isolectin B4 staining was applied to evaluate the angiogenesis in mouse hearts. Immunohistochemistry, Western blot analysis, and quantitative real-time polymerase chain reaction (qPCR) were performed to analyze expression levels of HIF-1a and its downstream genes. RESULTS Compared with the MI group, muscone treatment significantly improved cardiac function and reduced myocardial fibrosis. Moreover, muscone enhanced angiogenesis in the peri-infarct region and p-VEGFR2 expression in the vascular endothelial cells. Western blot analysis and qPCR showed that muscone upregulated expression levels of HIF-1a and VEGFA. CONCLUSIONS Muscone improved cardiac function in MI mice through augmented angiogenesis. The potential mechanism of muscone treatment in regulating angiogenesis of MI mice was upregulating expression levels of HIF-1α and VEGFA.


Assuntos
Cicloparafinas/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Indutores da Angiogênese , Animais , Modelos Animais de Doenças , Ecocardiografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Dados Preliminares , Função Ventricular Esquerda , Remodelação Ventricular/fisiologia
7.
J Mol Neurosci ; 63(2): 142-151, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28831692

RESUMO

Induced by hypothermia, cold-inducible protein RBM3 (RNA-binding protein motif 3), has been implicated in neuroprotection against various toxic insults such as hypoxia and ischemia. However, whether mild hypothermia and RBM3 prevent neural cells from UV irradiation-elicited apoptosis is unclear. In the present study, human neuroblastoma cell line SH-SY5Y was used as a cell model for neural cell death, and it was demonstrated that mild hypothermia protects SH-SY5Y cells from UV irradiation-induced apoptosis. However, the protective effect of mild hypothermia was abrogated when RBM3 was silenced. Conversely, the overexpression of RBM3 rescued SH-SY5Y cells from UV-induced apoptosis, as indicated by the decreased levels of cleaved caspase-3 and PARP, and increased cell survival. The analysis on the mechanism underlying RBM3-mediated neuroprotection against UV insult showed that RBM3 could substantially block the activation of p38 and JNK signaling pathways. In addition, the overexpression of RBM3 reduced the expression of pro-apoptotic proteins Bax and Bad, leaving the pro-survival protein Bcl-2 unaffected. In conclusion, RBM3 is the key mediator of mild hypothermia-related protection against UV in neuroblastoma cells, and the neuroprotective effect might be exerted through interfering with pro-apoptotic signaling pathways p38 and JNK and regulating pro-apoptotic proteins Bax and Bad.


Assuntos
Apoptose , Sistema de Sinalização das MAP Quinases , Neuroblastoma/metabolismo , Proteínas de Ligação a RNA/metabolismo , Linhagem Celular Tumoral , Humanos , MAP Quinase Quinase 4/metabolismo , Neurônios/metabolismo , Neurônios/efeitos da radiação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas de Ligação a RNA/genética , Raios Ultravioleta , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Sci Rep ; 7: 41738, 2017 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-28134320

RESUMO

Nitric oxide (NO)-induced apoptosis in neurons is an important cause of neurodegenerative disease in humans. The cold-inducible protein RBM3 mediates the protective effects of cooling on apoptosis induced by various insults. However, whether RBM3 protects neural cells from NO-induced apoptosis is unclear. This study aimed to investigate the neuroprotective effect of RBM3 on NO-induced apoptosis in human SH-SY5Y neuroblastoma cells. Firstly, we demonstrated that mild hypothermia (32 °C) induces RBM3 expression and confers a potent neuroprotective effect on NO-induced apoptosis, which was substantially diminished when RBM3 was silenced by siRNA. Moreover, overexpression of RBM3 exhibited a strong protective effect against NO-induced apoptosis. Signaling pathway screening demonstrated that only p38 inhibition by RBM3 provided neuroprotective effect, although RBM3 overexpression could affect the activation of p38, JNK, ERK, and AKT signaling in response to NO stimuli. Notably, RBM3 overexpression also blocked the activation of p38 signaling induced by transforming growth factor-ß1. Furthermore, both RBM3 overexpression and mild hypothermia abolished the induction of miR-143 by NO, which was shown to mediate the cytotoxicity of NO in a p38-dependent way. These findings suggest that RBM3 protects neuroblastoma cells from NO-induced apoptosis by suppressing p38 signaling, which mediates apoptosis through miR-143 induction.


Assuntos
Apoptose , Sistema de Sinalização das MAP Quinases , MicroRNAs/genética , Neuroblastoma/genética , Neuroblastoma/metabolismo , Óxido Nítrico/metabolismo , Proteínas de Ligação a RNA/metabolismo , Apoptose/genética , Linhagem Celular Tumoral , Expressão Gênica , Inativação Gênica , Humanos , Proteínas de Ligação a RNA/genética , Temperatura , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA