4.5 Article

Activating transcription factor 3 inhibits angiotensin II-induced cardiomyocyte viability and fibrosis by activating the transcription of cysteine-rich angiogenic protein 61

期刊

MOLECULAR MEDICINE REPORTS
卷 26, 期 5, 页码 -

出版社

SPANDIDOS PUBL LTD
DOI: 10.3892/mmr.2022.12852

关键词

activating transcription factor 3; cardiac hypertrophy; angiotensin II; proliferation; fibrosis; cysteine-rich angiogenic protein 61

资金

  1. Scientific Research and Cultivation Project of Meizhou People's Hospital, China [PY-C20210026]

向作者/读者索取更多资源

This study reveals that ATF3 inhibits fibrosis and viability of cardiac fibroblasts by activating the transcription of Cyr61 in stress overload-induced hearts. This finding enhances our understanding of the role of ATF3 in the development of cardiac diseases and provides a new target for the treatment of heart diseases.
Depletion of activating transcription factor 3 (ATF3) expression has previously been reported to promote hypertrophy, dysfunction and fibrosis in stress overload-induced hearts; however, the mechanism involved remains poorly understood. In the present study, the mechanism underlying the activation of cysteine-rich angiogenic protein 61 (Cyr61) by ATF3 in hyperproliferative and fibrotic human cardiac fibroblasts (HCFs), induced by angiotensin II (Ang II), was evaluated. The mRNA and protein expression levels of ATF3 and Cyr61 were assessed using reverse transcription-quantitative PCR and western blotting, respectively. The Cell Counting Kit-8 assay was used to assess cell viability. Cell migration was assessed using the wound healing assay and western blotting, whereas the extent of cell fibrosis was evaluated using immunofluorescence staining and western blotting. The binding site of ATF3 to the Cyr61 promoter was predicted using the JASPAR database, and verified using luciferase reporter and chromatin immunoprecipitation assays. The results demonstrated that the mRNA and protein expression levels of ATF3 were significantly upregulated in Ang II-induced HCFs. Overexpression of ATF3 significantly inhibited the Ang II-induced viability, migration and fibrosis of HCFs, whereas ATF3 knockdown mediated significant opposing effects. Mechanistically, ATF3 was demonstrated to transcriptionally activate Cyr61. Cyr61 silencing was subsequently revealed to reverse the effects of ATF3 overexpression on HCFs potentially via regulation of the TGF-beta /Smad signaling pathway. The results of the present study suggested that ATF3 could suppress HCF viability and fibrosis via the TGF-beta /Smad signaling pathway by activating the transcription of Cyr61.

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

4.5
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