Dynamic changes in chromatin accessibility, altered adipogenic gene expression, and total versus de novo fatty acid synthesis in subcutaneous adipose stem cells of normal-weight polycystic ovary syndrome (PCOS) women during adipogenesis: evidence of cellular programming

Clin Epigenetics. 2020 Nov 23;12(1):181. doi: 10.1186/s13148-020-00970-x.

Abstract

Background: Normal-weight polycystic ovary syndrome (PCOS) women exhibit adipose resistance in vivo accompanied by enhanced subcutaneous (SC) abdominal adipose stem cell (ASC) development to adipocytes with accelerated lipid accumulation per cell in vitro. The present study examines chromatin accessibility, RNA expression and fatty acid (FA) synthesis during SC abdominal ASC differentiation into adipocytes in vitro of normal-weight PCOS versus age- and body mass index-matched normoandrogenic ovulatory (control) women to study epigenetic/genetic characteristics as well as functional alterations of PCOS and control ASCs during adipogenesis.

Results: SC abdominal ASCs from PCOS women versus controls exhibited dynamic chromatin accessibility during adipogenesis, from significantly less chromatin accessibility at day 0 to greater chromatin accessibility by day 12, with enrichment of binding motifs for transcription factors (TFs) of the AP-1 subfamily at days 0, 3, and 12. In PCOS versus control cells, expression of genes governing adipocyte differentiation (PPARγ, CEBPα, AGPAT2) and function (ADIPOQ, FABP4, LPL, PLIN1, SLC2A4) was increased two-sixfold at days 3, 7, and 12, while that involving Wnt signaling (FZD1, SFRP1, and WNT10B) was decreased. Differential gene expression in PCOS cells at these time points involved triacylglycerol synthesis, lipid oxidation, free fatty acid beta-oxidation, and oxidative phosphorylation of the TCA cycle, with TGFB1 as a significant upstream regulator. There was a broad correspondence between increased chromatin accessibility and increased RNA expression of those 12 genes involved in adipocyte differentiation and function, Wnt signaling, as well as genes involved in the triacylglycerol synthesis functional group at day 12 of adipogenesis. Total content and de novo synthesis of myristic (C14:0), palmitic (C16:0), palmitoleic (C16:1), and oleic (C18:1) acid increased from day 7 to day 12 in all cells, with total content and de novo synthesis of FAs significantly greater in PCOS than controls cells at day 12.

Conclusions: In normal-weight PCOS women, dynamic chromatin remodeling of SC abdominal ASCs during adipogenesis may enhance adipogenic gene expression as a programmed mechanism to promote greater fat storage.

Keywords: Adipogenesis; Adipose stem cells; Cellular programming; Chromatin accessibility; De novo synthesis of fatty acids; Fat storage; Gene expression; Polycystic ovary syndrome; Total content; Transcriptional factors.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adipocytes / metabolism
  • Adipogenesis / genetics*
  • Adult
  • Body Mass Index
  • Case-Control Studies
  • Cell Differentiation / genetics
  • Chromatin / genetics*
  • Epigenomics / methods
  • Fatty Acids / metabolism*
  • Female
  • Gene Expression
  • Humans
  • Lipid Metabolism / genetics
  • Polycystic Ovary Syndrome / diagnosis
  • Polycystic Ovary Syndrome / genetics*
  • Polycystic Ovary Syndrome / pathology
  • RNA / genetics*
  • RNA / isolation & purification
  • Stem Cells / metabolism
  • Subcutaneous Fat / cytology
  • Subcutaneous Fat / growth & development
  • Subcutaneous Fat / metabolism
  • Transforming Growth Factor beta1 / metabolism
  • Wnt Signaling Pathway / genetics

Substances

  • Chromatin
  • Fatty Acids
  • TGFB1 protein, human
  • Transforming Growth Factor beta1
  • RNA