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

Circ Res. 2017 Aug 4;121(4):392-410. doi: 10.1161/CIRCRESAHA.117.311143. Epub 2017 Jun 21.

Abstract

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, TNFα (tumor necrosis factor α) can, under certain circumstances, promote improvements in mitochondrial integrity and function, phenomena that can be ascribed to the existence of TNFR2 (TNFα receptor 2).

Objective: The present study aimed to investigate whether and how TNFR2 activation mediates the effects of TNFα 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.

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

MeSH terms

  • Animals
  • Animals, Newborn
  • Aortic Diseases / genetics
  • Aortic Diseases / metabolism
  • Cells, Cultured
  • GTP Phosphohydrolases / biosynthesis*
  • GTP Phosphohydrolases / chemistry
  • GTP Phosphohydrolases / genetics
  • Gene Expression
  • HEK293 Cells
  • Humans
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Mitochondrial Dynamics / physiology*
  • Myocytes, Cardiac / metabolism
  • NF-kappa B / chemistry
  • NF-kappa B / metabolism*
  • Protein Structure, Secondary
  • Protein Structure, Tertiary
  • Receptors, Tumor Necrosis Factor, Type II / chemistry
  • Receptors, Tumor Necrosis Factor, Type II / metabolism*
  • STAT3 Transcription Factor / chemistry
  • STAT3 Transcription Factor / metabolism*

Substances

  • NF-kappa B
  • Receptors, Tumor Necrosis Factor, Type II
  • STAT3 Transcription Factor
  • Stat3 protein, mouse
  • GTP Phosphohydrolases
  • Opa1 protein, mouse