Your browser doesn't support javascript.
loading
MicroRNA-495 inhibits the high glucose-induced inflammation, differentiation and extracellular matrix accumulation of cardiac fibroblasts through downregulation of NOD1.
Wang, Xiaowei; Jin, Haiying; Jiang, Shifeng; Xu, Yanlan.
Afiliação
  • Wang X; Department of Geriatrics, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Qingpu District, No.1158, Park East Road, Shanghai, 201707 People's Republic of China.
  • Jin H; Department of Geriatrics, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Qingpu District, No.1158, Park East Road, Shanghai, 201707 People's Republic of China.
  • Jiang S; Department of Geriatrics, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Qingpu District, No.1158, Park East Road, Shanghai, 201707 People's Republic of China.
  • Xu Y; Department of Geriatrics, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Qingpu District, No.1158, Park East Road, Shanghai, 201707 People's Republic of China.
Cell Mol Biol Lett ; 23: 23, 2018.
Article em En | MEDLINE | ID: mdl-29760746
ABSTRACT

BACKGROUND:

MicroRNAs (miRNAs) have physiological and pathophysiological functions that are involved in the regulation of cardiac fibrosis. This study aimed to investigate the effects of miR-495 on high glucose-induced cardiac fibrosis in human cardiac fibroblasts (CFs) and to establish the mechanism underlying these effects.

METHODS:

Human CFs were transfected with an miR-495 inhibitor or mimic and incubated with high glucose. The levels of NOD1 and miR-495 were then determined via quantitative RT-PCR. Pro-inflammatory cytokine levels, cell differentiation and extracellular matrix accumulation were respectively detected using ELISA, quantitative RT-PCR and western blot assays. The luciferase reporter assay, quantitative RT-PCR and western blot were used to explore whether NOD1 was a target of miR-495. The effects of miR-495 on the NF-κB and TGF-ß1/Smad signaling pathways were also detected via western blot.

RESULTS:

Our results show that high glucose can significantly increase the expression of NOD1 in a time-dependent manner. Upregulation of miR-495 significantly alleviated the high glucose-induced increases in cell differentiation and collagen accumulation of CFs. Moreover, the bioinformatics analysis predicted that NOD1 was a potential target gene for miR-495. The luciferase reporter assay showed that miR-495 can directly target NOD1. The introduction of miR-495 could significantly inhibit the high glucose-activated NF-κB and TGF-ß1/Smad signaling pathways.

CONCLUSION:

Upregulation of miR-495 ameliorates the high glucose-induced inflammatory, cell differentiation and extracellular matrix accumulation of human CFs by modulating both the NF-κB and TGF-ß1/Smad signaling pathways through downregulation of NOD1 expression. These results provide further evidence for the protective effect of miR-495 overexpression in cases of high glucose-induced cardiac fibrosis.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: MicroRNAs / Proteína Adaptadora de Sinalização NOD1 / Fibroblastos / Glucose / Inflamação Idioma: En Ano de publicação: 2018 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: MicroRNAs / Proteína Adaptadora de Sinalização NOD1 / Fibroblastos / Glucose / Inflamação Idioma: En Ano de publicação: 2018 Tipo de documento: Article