Reduced estradiol-induced vasodilation and poly-(ADP-ribose) polymerase (PARP) activity in the aortas of rats with experimental polycystic ovary syndrome (PCOS)

PLoS One. 2013;8(3):e55589. doi: 10.1371/journal.pone.0055589. Epub 2013 Mar 26.

Abstract

Polycystic ovary syndrome (PCOS) is a complex endocrine disorder characterized by hyperandrogenism and insulin resistance, both of which have been connected to atherosclerosis. Indeed, an increased risk of clinical manifestations of arterial vascular diseases has been described in PCOS. On the other hand endothelial dysfunction can be detected early on, before atherosclerosis develops. Thus we assumed that vascular dysfunction is also related directly to the hormonal imbalance rather than to its metabolic consequences. To detect early functional changes, we applied a novel rodent model of PCOS: rats were either sham operated or hyperandrogenism was achieved by implanting subcutaneous pellets of dihydrotestosterone (DHT). After ten weeks, myograph measurements were performed on isolated aortic rings. Previously we described an increased contractility to norepinephrine (NE). Here we found a reduced immediate relaxation to estradiol treatment in pre-contracted aortic rings from hyperandrogenic rats. Although the administration of vitamin D3 along with DHT reduced responsiveness to NE, it did not restore relaxation to estradiol. Poly-(ADP-ribose) polymerase (PARP) activity was assessed by poly-ADP-ribose immunostaining. Increased PAR staining in ovaries and circulating leukocytes from DHT rats showed enhanced DNA damage, which was reduced by concomitant vitamin D3 treatment. Surprisingly, PAR staining was reduced in both the endothelium and vascular smooth muscle cells of the aorta rings from hyperandrogenic rats. Thus in the early phase of PCOS, vascular tone is already shifted towards vasoconstriction, characterized by reduced vasorelaxation and vascular dysfunction is concomitant with altered PARP activity. Based on our findings, PARP inhibitors might have a future perspective in restoring metabolic disorders in PCOS.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Aorta / enzymology*
  • Aorta / pathology
  • Aorta / physiopathology
  • Cholecalciferol / pharmacology
  • Endothelium, Vascular / enzymology
  • Endothelium, Vascular / pathology
  • Endothelium, Vascular / physiopathology
  • Estradiol / pharmacology*
  • Estrogens / pharmacology*
  • Female
  • Muscle, Smooth, Vascular / enzymology
  • Muscle, Smooth, Vascular / pathology
  • Muscle, Smooth, Vascular / physiopathology
  • Poly Adenosine Diphosphate Ribose / metabolism
  • Poly(ADP-ribose) Polymerases / metabolism*
  • Polycystic Ovary Syndrome / chemically induced
  • Polycystic Ovary Syndrome / metabolism*
  • Polycystic Ovary Syndrome / pathology
  • Polycystic Ovary Syndrome / physiopathology
  • Rats
  • Rats, Wistar
  • Vasoconstriction / drug effects
  • Vasodilation / drug effects*
  • Vitamins / pharmacology

Substances

  • Estrogens
  • Vitamins
  • Cholecalciferol
  • Poly Adenosine Diphosphate Ribose
  • Estradiol
  • Poly(ADP-ribose) Polymerases

Grants and funding

This study was supported by a Servier Grant from the European Foundation for the Study of Diabetes as well as by grants from the Hungarian NIH (ALAP1-01298/2009), Scientific Healthcare Council (ETT) (427/2009) and the Hungarian National Developmental Agency (NFÜ) (TÁMOP 4.2.1/B- 09/1/KMR-2010-001). This study was sponsored by a research grant of the Hungarian Society of Hypertension, and National Grants of the Hungarian Government Fund, Hungarian NIH (K81972, NF69278). Eszter M. Horvath was supported by the János Bolyai Scholarship of the Hungarian Academy of Sciences. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.