[HTML][HTML] Hypoxia mediates mutual repression between microRNA-27a and PPARγ in the pulmonary vasculature

BY Kang, KK Park, DE Green, KM Bijli, CD Searles… - PloS one, 2013 - journals.plos.org
BY Kang, KK Park, DE Green, KM Bijli, CD Searles, RL Sutliff, CM Hart
PloS one, 2013journals.plos.org
Pulmonary hypertension (PH) is a serious disorder that causes significant morbidity and
mortality. The pathogenesis of PH involves complex derangements in multiple pathways
including reductions in peroxisome proliferator-activated receptor gamma (PPARγ).
Hypoxia, a common PH stimulus, reduces PPARγ in experimental models. In contrast,
activating PPARγ attenuates hypoxia-induced PH and endothelin 1 (ET-1) expression. To
further explore mechanisms of hypoxia-induced PH and reductions in PPARγ, we examined …
Pulmonary hypertension (PH) is a serious disorder that causes significant morbidity and mortality. The pathogenesis of PH involves complex derangements in multiple pathways including reductions in peroxisome proliferator-activated receptor gamma (PPARγ). Hypoxia, a common PH stimulus, reduces PPARγ in experimental models. In contrast, activating PPARγ attenuates hypoxia-induced PH and endothelin 1 (ET-1) expression. To further explore mechanisms of hypoxia-induced PH and reductions in PPARγ, we examined the effects of hypoxia on selected microRNA (miRNA or miR) levels that might reduce PPARγ expression leading to increased ET-1 expression and PH. Our results demonstrate that exposure to hypoxia (10% O2) for 3-weeks increased levels of miR-27a and ET-1 in the lungs of C57BL/6 mice and reduced PPARγ levels. Hypoxia-induced increases in miR-27a were attenuated in mice treated with the PPARγ ligand, rosiglitazone (RSG, 10 mg/kg/d) by gavage for the final 10 d of exposure. In parallel studies, human pulmonary artery endothelial cells (HPAECs) were exposed to control (21% O2) or hypoxic (1% O2) conditions for 72 h. Hypoxia increased HPAEC proliferation, miR-27a and ET-1 expression, and reduced PPARγ expression. These alterations were attenuated by treatment with RSG (10 µM) during the last 24 h of hypoxia exposure. Overexpression of miR-27a or PPARγ knockdown increased HPAEC proliferation and ET-1 expression and decreased PPARγ levels, whereas these effects were reversed by miR-27a inhibition. Further, compared to lungs from littermate control mice, miR-27a levels were upregulated in lungs from endothelial-targeted PPARγ knockout (ePPARγ KO) mice. Knockdown of either SP1 or EGR1 was sufficient to significantly attenuate miR-27a expression in HPAECs. Collectively, these studies provide novel evidence that miR-27a and PPARγ mediate mutually repressive actions in hypoxic pulmonary vasculature and that targeting PPARγ may represent a novel therapeutic approach in PH to attenuate proliferative mediators that stimulate proliferation of pulmonary vascular cells.
PLOS