4.7 Article

TNFR2 Stimulation Promotes Mitochondrial Fusion via Stat3-and NF-kB-Dependent Activation of OPA1 Expression

期刊

CIRCULATION RESEARCH
卷 121, 期 4, 页码 392-+

出版社

LIPPINCOTT WILLIAMS & WILKINS
DOI: 10.1161/CIRCRESAHA.117.311143

关键词

acetylation; heart failure; mitochondrial dynamics; molecular dynamics simulation; tumor necrosis factor-alpha

资金

  1. National Basic Research Program of China (973 Program) [2014CB965100, 2014CB965103]
  2. National High-tech RD 863 Program [2015AA020922]
  3. National Natural Science Foundation of China [81320108003, 31371498, 81370346, 81370247, 81622006, 81670261, 81573641]
  4. Science and Technology Department of Zhejiang Province Public Welfare Projects [2013C37054]
  5. Science and Technology Department of Zhejiang Province Major Development Projects and Public Welfare Projects [2013C37054]
  6. Fundamental Research Funds for the Central Universities [2016XZZX002-03, BSF-001-00*, 2017FZA7011]
  7. National Key Research and Development Program of China [2016YFC1301204]
  8. Major Scientific and Technological Project of the Zhejiang Province Science and Technology Department [2013C03043-4]
  9. Education Department of Zhejiang Province [Y201329862]
  10. Zhejiang Provincial Natural Science Foundation [LY16H280003]

向作者/读者索取更多资源

Rationale: Mitochondria are important cellular organelles and play essential roles in maintaining cell structure and function. Emerging evidence indicates that in addition to having proinflammatory and proapoptotic effects, TNF alpha (tumor necrosis factor alpha) can, under certain circumstances, promote improvements in mitochondrial integrity and function, phenomena that can be ascribed to the existence of TNFR2 (TNF alpha receptor 2). Objective: The present study aimed to investigate whether and how TNFR2 activation mediates the effects of TNF alpha on mitochondria. Methods and Results: Freshly isolated neonatal mouse cardiac myocytes treated with shRNA targeting TNFR1 were used to study the effects of TNFR2 activation on mitochondrial function. Neonatal mouse cardiac myocytes exhibited increases in mitochondrial fusion, a change that was associated with increases in mitochondrial membrane potential, intracellular ATP levels, and oxygen consumption capacity. Importantly, TNFR2 activation-induced increases in OPA1 (optic atrophy 1) protein expression were responsible for the above enhancements, and these changes could be attenuated using siRNA targeting OPA1. Moreover, both Stat3 and RelA bound to the promoter region of OPA1 and their interactions synergistically upregulated OPA1 expression at the transcriptional level. Stat3 acetylation at lysine 370 or lysine 383 played a key role in the ability of Stat3 to form a supercomplex with RelA. Meanwhile, p300 modulated Stat3 acetylation in HEK293T (human embryonic kidney 293T) cells, and p300-mediated Stat3/RelA interactions played an indispensable role in OPA1 upregulation. Finally, TNFR2 activation exerted beneficial effects on OPA1 expression in an in vivo transverse aortic constriction model, whereby TNFR1-knockout mice exhibited better outcomes than in mice with both TNFR1 and TNFR2 knocked out. Conclusions: TNFR2 activation protects cardiac myocytes against stress by upregulating OPA1 expression. This process was facilitated by p300-mediated Stat3 acetylation and Stat3/RelA interactions, leading to improvements in mitochondrial morphology and function.

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

4.7
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