Increased prostaglandin-D2 in male STAT3-deficient hearts shifts cardiac progenitor cells from endothelial to white adipocyte differentiation

PLoS Biol. 2020 Dec 28;18(12):e3000739. doi: 10.1371/journal.pbio.3000739. eCollection 2020 Dec.

Abstract

Cardiac levels of the signal transducer and activator of transcription factor-3 (STAT3) decline with age, and male but not female mice with a cardiomyocyte-specific STAT3 deficiency conditional knockout (CKO) display premature age-related heart failure associated with reduced cardiac capillary density. In the present study, isolated male and female CKO-cardiomyocytes exhibit increased prostaglandin (PG)-generating cyclooxygenase-2 (COX-2) expression. The PG-degrading hydroxyprostaglandin-dehydrogenase-15 (HPGD) expression is only reduced in male cardiomyocytes, which is associated with increased prostaglandin D2 (PGD2) secretion from isolated male but not female CKO-cardiomyocytes. Reduced HPGD expression in male cardiomyocytes derive from impaired androgen receptor (AR)-signaling due to loss of its cofactor STAT3. Elevated PGD2 secretion in males is associated with increased white adipocyte accumulation in aged male but not female hearts. Adipocyte differentiation is enhanced in isolated stem cell antigen-1 (SCA-1)+ cardiac progenitor cells (CPC) from young male CKO-mice compared with the adipocyte differentiation of male wild-type (WT)-CPC and CPC isolated from female mice. Epigenetic analysis in freshly isolated male CKO-CPC display hypermethylation in pro-angiogenic genes (Fgfr2, Epas1) and hypomethylation in the white adipocyte differentiation gene Zfp423 associated with up-regulated ZFP423 expression and a shift from endothelial to white adipocyte differentiation compared with WT-CPC. The expression of the histone-methyltransferase EZH2 is reduced in male CKO-CPC compared with male WT-CPC, whereas no differences in the EZH2 expression in female CPC were observed. Clonally expanded CPC can differentiate into endothelial cells or into adipocytes depending on the differentiation conditions. ZFP423 overexpression is sufficient to induce white adipocyte differentiation of clonal CPC. In isolated WT-CPC, PGD2 stimulation reduces the expression of EZH2, thereby up-regulating ZFP423 expression and promoting white adipocyte differentiation. The treatment of young male CKO mice with the COX inhibitor Ibuprofen or the PGD2 receptor (DP)2 receptor antagonist BAY-u 3405 in vivo increased EZH2 expression and reduced ZFP423 expression and adipocyte differentiation in CKO-CPC. Thus, cardiomyocyte STAT3 deficiency leads to age-related and sex-specific cardiac remodeling and failure in part due to sex-specific alterations in PGD2 secretion and subsequent epigenetic impairment of the differentiation potential of CPC. Causally involved is the impaired AR signaling in absence of STAT3, which reduces the expression of the PG-degrading enzyme HPGD.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adipocytes, White / metabolism
  • Animals
  • Cell Differentiation / genetics
  • Cells, Cultured
  • Cyclooxygenase 2 / metabolism
  • Endothelial Cells / metabolism
  • Female
  • Heart Failure / genetics
  • Humans
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Multipotent Stem Cells / metabolism
  • Myocytes, Cardiac / metabolism*
  • Prostaglandin D2 / metabolism*
  • Prostaglandin D2 / physiology
  • STAT3 Transcription Factor / genetics
  • STAT3 Transcription Factor / metabolism*
  • Signal Transduction / genetics
  • Stem Cells / metabolism

Substances

  • STAT3 Transcription Factor
  • Ptgs2 protein, mouse
  • Cyclooxygenase 2
  • Prostaglandin D2

Grants and funding

This work was supported with financial funding by the German Research Foundation (DFG, HI 842/3-2 to D.H.-K.), by the DFG Clinical Research Group (DFG KFO311, HI 842/10-1 to D.H.-K., HI 842/10-2 to D.H.-K., RI 2531/2-1 to M.R.-H., RI 2531/2-2 to M.R.-H.), by the State of Lower Saxony and the Volkswagen Foundation (VWZN3009 to D.H.-K. and M.R.-H.), Hannover, Germany and by REBIRTH I/II as well as the BMBF project de.STAIR (031L0106D to S.H.). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. The authors received no specific funding for this work.