The transcription factor GATA6 accelerates vascular smooth muscle cell senescence-related arterial calcification by counteracting the role of anti-aging factor SIRT6 and impeding DNA damage repair

转录因子 衰老 血管平滑肌 癌症研究 钙化 下调和上调 早衰 关贸总协定6 内分泌学 细胞生物学 医学 生物 内科学 平滑肌 遗传学 生物化学 基因
作者
Xiaoxue Li,Aiting Liu,Chen Xie,Yanlian Chen,Ke Zeng,Changming Xie,Zhengzhipeng Zhang,Pei Luo,Hui Huang
出处
期刊:Kidney International [Elsevier]
卷期号:105 (1): 115-131 被引量:1
标识
DOI:10.1016/j.kint.2023.09.028
摘要

Arterial calcification is a hallmark of vascular pathology in the elderly and in individuals with chronic kidney disease (CKD). Vascular smooth muscle cells (VSMCs), after attaining a senescent phenotype, are implicated in the calcifying process. However, the underlying mechanism remains to be elucidated. Here, we reveal an aberrant upregulation of transcriptional factor GATA6 in the calcified aortas of humans, mice with CKD and mice subjected to vitamin D3 injection. Knockdown of GATA6, via recombinant adeno-associated virus carrying GATA6 shRNA, inhibited the development of arterial calcification in mice with CKD. Further gain- and loss-of function experiments in vitro verified the contribution of GATA6 in osteogenic differentiation of VSMCs. Samples of human aorta exhibited a positive relationship between age and GATA6 expression and GATA6 was also elevated in the aortas of old as compared to young mice. Calcified aortas displayed senescent features with VSMCs undergoing premature senescence, blunted by GATA6 downregulation. Notably, abnormal induction of GATA6 in senescent and calcified aortas was rescued in Sirtuin 6 (SIRT6)-transgenic mice, a well-established longevity mouse model. Suppression of GATA6 accounted for the favorable effect of SIRT6 on VSMCs senescence prevention. Mechanistically, SIRT6 inhibited the transcription of GATA6 by deacetylation and increased degradation of transcription factor Nkx2.5. Moreover, GATA6 was induced by DNA damage stress during arterial calcification and subsequently impeded the Ataxia-telangiectasia mutated (ATM)-mediated DNA damage repair process, leading to accelerated VSMCs senescence and osteogenic differentiation. Thus, GATA6 is a novel regulator in VSMCs senescence. Our findings provide novel insight in arterial calcification and a potential new target for intervention. Arterial calcification is a hallmark of vascular pathology in the elderly and in individuals with chronic kidney disease (CKD). Vascular smooth muscle cells (VSMCs), after attaining a senescent phenotype, are implicated in the calcifying process. However, the underlying mechanism remains to be elucidated. Here, we reveal an aberrant upregulation of transcriptional factor GATA6 in the calcified aortas of humans, mice with CKD and mice subjected to vitamin D3 injection. Knockdown of GATA6, via recombinant adeno-associated virus carrying GATA6 shRNA, inhibited the development of arterial calcification in mice with CKD. Further gain- and loss-of function experiments in vitro verified the contribution of GATA6 in osteogenic differentiation of VSMCs. Samples of human aorta exhibited a positive relationship between age and GATA6 expression and GATA6 was also elevated in the aortas of old as compared to young mice. Calcified aortas displayed senescent features with VSMCs undergoing premature senescence, blunted by GATA6 downregulation. Notably, abnormal induction of GATA6 in senescent and calcified aortas was rescued in Sirtuin 6 (SIRT6)-transgenic mice, a well-established longevity mouse model. Suppression of GATA6 accounted for the favorable effect of SIRT6 on VSMCs senescence prevention. Mechanistically, SIRT6 inhibited the transcription of GATA6 by deacetylation and increased degradation of transcription factor Nkx2.5. Moreover, GATA6 was induced by DNA damage stress during arterial calcification and subsequently impeded the Ataxia-telangiectasia mutated (ATM)-mediated DNA damage repair process, leading to accelerated VSMCs senescence and osteogenic differentiation. Thus, GATA6 is a novel regulator in VSMCs senescence. Our findings provide novel insight in arterial calcification and a potential new target for intervention.
最长约 10秒,即可获得该文献文件

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
1秒前
我是老大应助mkxany采纳,获得10
1秒前
2秒前
gjww应助科研通管家采纳,获得10
3秒前
gjww应助科研通管家采纳,获得10
3秒前
gjww应助科研通管家采纳,获得10
3秒前
慕青应助科研通管家采纳,获得20
3秒前
orixero应助科研通管家采纳,获得10
3秒前
Lucas应助科研通管家采纳,获得10
3秒前
Lucas应助科研通管家采纳,获得10
3秒前
李爱国应助科研通管家采纳,获得10
3秒前
星辰大海应助科研通管家采纳,获得10
3秒前
隐形曼青应助科研通管家采纳,获得10
3秒前
乐乐应助科研通管家采纳,获得10
3秒前
桐桐应助科研通管家采纳,获得10
3秒前
Owen完成签到,获得积分10
3秒前
Treasure完成签到,获得积分10
6秒前
honeyzh发布了新的文献求助10
7秒前
9秒前
10秒前
紫菜完成签到,获得积分10
11秒前
领导范儿应助负数采纳,获得10
11秒前
英俊的铭应助负数采纳,获得10
12秒前
子厘发布了新的文献求助10
14秒前
shinysparrow应助细腻曼冬采纳,获得30
14秒前
mkxany发布了新的文献求助10
16秒前
21秒前
飞翔完成签到 ,获得积分10
22秒前
廖梓康发布了新的文献求助10
25秒前
26秒前
mkxany完成签到,获得积分10
26秒前
Hsia发布了新的文献求助200
26秒前
子厘完成签到,获得积分10
27秒前
JamesPei应助丹尼采纳,获得10
29秒前
30秒前
罗布林卡应助幸运海星采纳,获得30
32秒前
32秒前
32秒前
32秒前
Forya发布了新的文献求助10
33秒前
高分求助中
Teaching Social and Emotional Learning in Physical Education 900
Plesiosaur extinction cycles; events that mark the beginning, middle and end of the Cretaceous 800
Recherches Ethnographiques sue les Yao dans la Chine du Sud 500
Two-sample Mendelian randomization analysis reveals causal relationships between blood lipids and venous thromboembolism 500
Chinese-English Translation Lexicon Version 3.0 500
Wisdom, Gods and Literature Studies in Assyriology in Honour of W. G. Lambert 400
薩提亞模式團體方案對青年情侶輔導效果之研究 400
热门求助领域 (近24小时)
化学 材料科学 医学 生物 有机化学 工程类 生物化学 纳米技术 物理 内科学 计算机科学 化学工程 复合材料 遗传学 基因 物理化学 催化作用 电极 光电子学 量子力学
热门帖子
关注 科研通微信公众号,转发送积分 2392386
求助须知:如何正确求助?哪些是违规求助? 2096953
关于积分的说明 5283278
捐赠科研通 1824520
什么是DOI,文献DOI怎么找? 909933
版权声明 559928
科研通“疑难数据库(出版商)”最低求助积分说明 486236