Hypoxia upregulates estrogen receptor β in pulmonary artery endothelial cells in a HIF-1α–dependent manner

AL Frump, M Selej, JA Wood, M Albrecht… - American journal of …, 2018 - atsjournals.org
AL Frump, M Selej, JA Wood, M Albrecht, B Yakubov, I Petrache, T Lahm
American journal of respiratory cell and molecular biology, 2018atsjournals.org
17β-Estradiol (E2) attenuates hypoxia-induced pulmonary hypertension (HPH) through
estrogen receptor (ER)-dependent effects, including inhibition of hypoxia-induced
endothelial cell proliferation; however, the mechanisms responsible for this remain
unknown. We hypothesized that the protective effects of E2 in HPH are mediated through
hypoxia-inducible factor 1α (HIF-1α)-dependent increases in ERβ expression. Sprague-
Dawley rats and ERα or ERβ knockout mice were exposed to hypobaric hypoxia for 2–3 …
17β-Estradiol (E2) attenuates hypoxia-induced pulmonary hypertension (HPH) through estrogen receptor (ER)-dependent effects, including inhibition of hypoxia-induced endothelial cell proliferation; however, the mechanisms responsible for this remain unknown. We hypothesized that the protective effects of E2 in HPH are mediated through hypoxia-inducible factor 1α (HIF-1α)-dependent increases in ERβ expression. Sprague-Dawley rats and ERα or ERβ knockout mice were exposed to hypobaric hypoxia for 2–3 weeks. The effects of hypoxia were also studied in primary rat or human pulmonary artery endothelial cells (PAECs). Hypoxia increased expression of ERβ, but not ERα, in lungs from HPH rats as well as in rat and human PAECs. ERβ mRNA time dependently increased in PAECs exposed to hypoxia. Normoxic HIF-1α/HIF-2α stabilization increased PAEC ERβ, whereas HIF-1α knockdown decreased ERβ abundance in hypoxic PAECs. In turn, ERβ knockdown in hypoxic PAECs increased HIF-2α expression, suggesting a hypoxia-sensitive feedback mechanism. ERβ knockdown in hypoxic PAECs also decreased expression of the HIF inhibitor prolyl hydroxylase 2 (PHD2), whereas ERβ activation increased PHD2 and decreased both HIF-1α and HIF-2α, suggesting that ERβ regulates the PHD2/HIF-1α/HIF-2α axis during hypoxia. Whereas hypoxic wild-type or ERα knockout mice treated with E2 demonstrated less pulmonary vascular remodeling and decreased HIF-1α after hypoxia compared with untreated hypoxic mice, ERβ knockout mice exhibited increased HIF-2α and an attenuated response to E2 during hypoxia. Taken together, our results demonstrate a novel and potentially therapeutically targetable mechanism whereby hypoxia, via HIF-1α, increases ERβ expression and the E2-ERβ axis targets PHD2, HIF-1α, and HIF-2α to attenuate HPH development.
ATS Journals