Loss of tet methyl cytosine dioxygenase 3 (TET3) enhances cardiac fibrosis via modulating the DNA damage repair response.

IF 4.8 2区 医学 Q1 GENETICS & HEREDITY
Sandip Kumar Rath, Gunsmaa Nyamsuren, Björn Tampe, David Sung-Wen Yu, Melanie S Hulshoff, Denise Schlösser, Sabine Maamari, Michael Zeisberg, Elisabeth M Zeisberg
{"title":"Loss of tet methyl cytosine dioxygenase 3 (TET3) enhances cardiac fibrosis via modulating the DNA damage repair response.","authors":"Sandip Kumar Rath, Gunsmaa Nyamsuren, Björn Tampe, David Sung-Wen Yu, Melanie S Hulshoff, Denise Schlösser, Sabine Maamari, Michael Zeisberg, Elisabeth M Zeisberg","doi":"10.1186/s13148-024-01719-6","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Cardiac fibrosis is the hallmark of all forms of chronic heart disease. Activation and proliferation of cardiac fibroblasts are the prime mediators of cardiac fibrosis. Existing studies show that ROS and inflammatory cytokines produced during fibrosis not only signal proliferative stimuli but also contribute to DNA damage. Therefore, as a prerequisite to maintain sustained proliferation in fibroblasts, activation of distinct DNA repair mechanism is essential.</p><p><strong>Result: </strong>In this study, we report that TET3, a DNA demethylating enzyme, which has been shown to be reduced in cardiac fibrosis and to exert antifibrotic effects does so not only through its demethylating activity but also through maintaining genomic integrity by facilitating error-free homologous recombination (HR) repair of DNA damage. Using both in vitro and in vivo models of cardiac fibrosis as well as data from human heart tissue, we demonstrate that the loss of TET3 in cardiac fibroblasts leads to spontaneous DNA damage and in the presence of TGF-β to a shift from HR to the fast but more error-prone non-homologous end joining repair pathway. This shift contributes to increased fibroblast proliferation in a fibrotic environment. In vitro experiments showed TET3's recruitment to H2O2-induced DNA double-strand breaks (DSBs) in mouse cardiac fibroblasts, promoting HR repair. Overexpressing TET3 counteracted TGF-β-induced fibroblast proliferation and restored HR repair efficiency. Extending these findings to human cardiac fibrosis, we confirmed TET3 expression loss in fibrotic hearts and identified a negative correlation between TET3 levels, fibrosis markers, and DNA repair pathway alteration.</p><p><strong>Conclusion: </strong>Collectively, our findings demonstrate TET3's pivotal role in modulating DDR and fibroblast proliferation in cardiac fibrosis and further highlight TET3 as a potential therapeutic target.</p>","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":4.8000,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11350970/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Epigenetics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1186/s13148-024-01719-6","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"GENETICS & HEREDITY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Cardiac fibrosis is the hallmark of all forms of chronic heart disease. Activation and proliferation of cardiac fibroblasts are the prime mediators of cardiac fibrosis. Existing studies show that ROS and inflammatory cytokines produced during fibrosis not only signal proliferative stimuli but also contribute to DNA damage. Therefore, as a prerequisite to maintain sustained proliferation in fibroblasts, activation of distinct DNA repair mechanism is essential.

Result: In this study, we report that TET3, a DNA demethylating enzyme, which has been shown to be reduced in cardiac fibrosis and to exert antifibrotic effects does so not only through its demethylating activity but also through maintaining genomic integrity by facilitating error-free homologous recombination (HR) repair of DNA damage. Using both in vitro and in vivo models of cardiac fibrosis as well as data from human heart tissue, we demonstrate that the loss of TET3 in cardiac fibroblasts leads to spontaneous DNA damage and in the presence of TGF-β to a shift from HR to the fast but more error-prone non-homologous end joining repair pathway. This shift contributes to increased fibroblast proliferation in a fibrotic environment. In vitro experiments showed TET3's recruitment to H2O2-induced DNA double-strand breaks (DSBs) in mouse cardiac fibroblasts, promoting HR repair. Overexpressing TET3 counteracted TGF-β-induced fibroblast proliferation and restored HR repair efficiency. Extending these findings to human cardiac fibrosis, we confirmed TET3 expression loss in fibrotic hearts and identified a negative correlation between TET3 levels, fibrosis markers, and DNA repair pathway alteration.

Conclusion: Collectively, our findings demonstrate TET3's pivotal role in modulating DDR and fibroblast proliferation in cardiac fibrosis and further highlight TET3 as a potential therapeutic target.

tet 甲基胞嘧啶二氧酶 3 (TET3) 的缺失会通过调节 DNA 损伤修复反应增强心脏纤维化。
背景:心脏纤维化是各种慢性心脏病的标志。心脏成纤维细胞的活化和增殖是心脏纤维化的主要介质。现有研究表明,纤维化过程中产生的 ROS 和炎性细胞因子不仅是增殖刺激的信号,也会导致 DNA 损伤。因此,作为维持成纤维细胞持续增殖的先决条件,激活独特的 DNA 修复机制至关重要:在这项研究中,我们报告了一种 DNA 去甲基化酶 TET3,这种酶在心脏纤维化中被证明会减少并发挥抗纤维化作用,它不仅通过其去甲基化活性,还通过促进 DNA 损伤的无差错同源重组(HR)修复来维持基因组的完整性。我们利用体外和体内心脏纤维化模型以及来自人类心脏组织的数据证明,心脏成纤维细胞中 TET3 的缺失会导致自发性 DNA 损伤,并在 TGF-β 的存在下导致从 HR 转向快速但更容易出错的非同源末端连接修复途径。这种转变导致成纤维细胞在纤维化环境中增殖增加。体外实验显示,在小鼠心脏成纤维细胞中,TET3 被招募到 H2O2 诱导的 DNA 双链断裂(DSB)处,促进 HR 修复。过表达 TET3 可抵消 TGF-β 诱导的成纤维细胞增殖,并恢复 HR 修复效率。将这些发现扩展到人类心脏纤维化,我们证实了纤维化心脏中 TET3 的表达缺失,并确定了 TET3 水平、纤维化标志物和 DNA 修复途径改变之间的负相关:总之,我们的研究结果证明了 TET3 在心脏纤维化中调节 DDR 和成纤维细胞增殖的关键作用,并进一步突出了 TET3 作为潜在治疗靶点的作用。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 求助全文
来源期刊
自引率
5.30%
发文量
150
期刊介绍: Clinical Epigenetics, the official journal of the Clinical Epigenetics Society, is an open access, peer-reviewed journal that encompasses all aspects of epigenetic principles and mechanisms in relation to human disease, diagnosis and therapy. Clinical trials and research in disease model organisms are particularly welcome.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信