Hyperglycemia Induces Myocardial Dysfunction via Epigenetic Regulation of JunD

Circ Res. 2020 Oct 23;127(10):1261-1273. doi: 10.1161/CIRCRESAHA.120.317132. Epub 2020 Aug 20.

Abstract

Rationale: Hyperglycemia -induced reactive oxygen species are key mediators of cardiac dysfunction. JunD (Jund proto-oncogene subunit), a member of the AP-1 (activator protein-1) family of transcription factors, is emerging as a major gatekeeper against oxidative stress. However, its contribution to redox state and inflammation in the diabetic heart remains to be elucidated.

Objective: The present study investigates the role of JunD in hyperglycemia-induced and reactive oxygen species-driven myocardial dysfunction.

Methods and results: JunD mRNA and protein expression were reduced in the myocardium of mice with streptozotocin-induced diabetes mellitus as compared to controls. JunD downregulation was associated with oxidative stress and left ventricular dysfunction assessed by electron spin resonance spectroscopy as well as conventional and 2-dimensional speckle-tracking echocardiography. Furthermore, myocardial expression of free radical scavenger superoxide dismutase 1 and aldehyde dehydrogenase 2 was reduced, whereas the NOX2 (NADPH [nicotinamide adenine dinucleotide phosphatase] oxidase subunit 2) and NOX4 (NADPH [nicotinamide adenine dinucleotide phosphatase] oxidase subunit 4) were upregulated. The redox changes were associated with increased NF-κB (nuclear factor kappa B) binding activity and expression of inflammatory mediators. Interestingly, mice with cardiac-specific overexpression of JunD via the α MHC (α- myosin heavy chain) promoter (α MHC JunDtg) were protected against hyperglycemia-induced cardiac dysfunction. We also showed that JunD was epigenetically regulated by promoter hypermethylation, post-translational modification of histone marks, and translational repression by miRNA (microRNA)-673/menin. Reduced JunD mRNA and protein expression were confirmed in left ventricular specimens obtained from patients with type 2 diabetes mellitus as compared to nondiabetic subjects.

Conclusions: Here, we show that a complex epigenetic machinery involving DNA methylation, histone modifications, and microRNAs mediates hyperglycemia-induced JunD downregulation and myocardial dysfunction in experimental and human diabetes mellitus. Our results pave the way for tissue-specific therapeutic modulation of JunD to prevent diabetic cardiomyopathy.

Keywords: diabetes mellitus; heart failure; microRNA; reactive oxygen species; transcription factors.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • DNA Methylation
  • Diabetic Cardiomyopathies / etiology
  • Diabetic Cardiomyopathies / genetics*
  • Diabetic Cardiomyopathies / metabolism
  • Epigenesis, Genetic*
  • Histone Code
  • Humans
  • Hyperglycemia / complications*
  • Male
  • Mice
  • Mice, Inbred C57BL
  • MicroRNAs / metabolism
  • Myocardium / metabolism
  • NADPH Oxidase 2 / genetics
  • NADPH Oxidase 2 / metabolism
  • NADPH Oxidase 4 / genetics
  • NADPH Oxidase 4 / metabolism
  • NF-kappa B / genetics
  • NF-kappa B / metabolism
  • Proto-Oncogene Mas
  • Proto-Oncogene Proteins / genetics
  • Proto-Oncogene Proteins / metabolism
  • Proto-Oncogene Proteins c-jun / genetics*
  • Proto-Oncogene Proteins c-jun / metabolism
  • Reactive Oxygen Species / metabolism
  • Superoxide Dismutase-1 / genetics
  • Superoxide Dismutase-1 / metabolism

Substances

  • MAS1 protein, human
  • Men1 protein, mouse
  • MicroRNAs
  • NF-kappa B
  • Proto-Oncogene Mas
  • Proto-Oncogene Proteins
  • Proto-Oncogene Proteins c-jun
  • Reactive Oxygen Species
  • junD protein, mouse
  • Sod1 protein, mouse
  • Superoxide Dismutase-1
  • Cybb protein, mouse
  • NADPH Oxidase 2
  • NADPH Oxidase 4
  • Nox4 protein, mouse