4.3 Article

Long noncoding RNA MIAT inhibits the progression of diabetic nephropathy and the activation of NF-κB pathway in high glucose-treated renal tubular epithelial cells by the miR-182-5p/GPRC5A axis

期刊

OPEN MEDICINE
卷 16, 期 1, 页码 1336-1349

出版社

DE GRUYTER POLAND SP Z O O
DOI: 10.1515/med-2021-0328

关键词

MIAT; DN; miR-182-5p; GPRC5A; NF-kappa B pathway

资金

  1. Shaanxi Science and Technology Research and Development Project Fund [2017SF-263]

向作者/读者索取更多资源

This study demonstrates that MIAT acts as an inhibitory factor in the pathogenesis of DN by regulating the miR-182-5p/GPRC5A axis, thereby preventing cell damage induced by high glucose and inactivating the NF-kappa B pathway.
Background - Diabetic nephropathy (DN) is a common diabetic complication. Long noncoding RNAs (lncRNAs) have been identified as essential regulators in DN progression. This study is devoted to the research of lncRNA-myocardial infarction-associated transcript (MIAT) in DN. Methods - DN cell model was established by high glucose (HG) treatment for human renal tubular epithelial cells (HK-2). Cell viability and colonizing capacity were analyzed by Cell Counting Kit-8 (CCK-8) and colony formation assay. Apoptosis was assessed via caspase-3 detection and flow cytometry. Enzyme-linked immunosorbent assay (ELISA) was used for evaluating inflammation. The protein determination was completed using western blot. MIAT, microRNA-182-5p (miR-182-5p), and G protein-coupled receptor class C group 5 member A (GPRC5A) levels were all examined via reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Intergenic binding was verified using dual-luciferase reporter, RNA immunoprecipitation (RIP), and RNA pull-down assays. Results - HG induced the inhibition of cell growth, but accelerated apoptosis and inflammation as well as the activation of nuclear factor kappa B (NF-kappa B) pathway. MIAT reestablishment prevented the HG-induced cell damages and NF-kappa B signal activation. Mechanistically, MIAT was proved as a miR-182-5p sponge and regulated the expression of GPRC5A that was a miR-182-5p target. The rescued experiments demonstrated that MIAT downregulation or miR-182-5p upregulation aggravated the HG-induced cell damages and activated the NF-kappa B pathway via the respective regulation of miR-182-5p or GPRC5A. Conclusion - Taken together, MIAT functioned as an inhibitory factor in the pathogenesis to impede the development of DN and inactivate the NF-kappa B pathway via regulating the miR-182-5p/GPRC5A axis.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.3
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据