The PPARγ ligand rosiglitazone attenuates hypoxia-induced endothelin signaling in vitro and in vivo

BY Kang, JM Kleinhenz… - American Journal of …, 2011 - journals.physiology.org
BY Kang, JM Kleinhenz, TC Murphy, CM Hart
American Journal of Physiology-Lung Cellular and Molecular …, 2011journals.physiology.org
Peroxisome proliferator-activated receptor (PPAR) γ activation attenuates hypoxia-induced
pulmonary hypertension (PH) in mice. The current study examined the hypothesis that
PPARγ attenuates hypoxia-induced endothelin-1 (ET-1) signaling to mediate these
therapeutic effects. To test this hypothesis, human pulmonary artery endothelial cells
(HPAECs) were exposed to normoxia or hypoxia (1% O2) for 72 h and treated with or
without the PPARγ ligand rosiglitazone (RSG, 10 μM) during the final 24 h of exposure …
Peroxisome proliferator-activated receptor (PPAR) γ activation attenuates hypoxia-induced pulmonary hypertension (PH) in mice. The current study examined the hypothesis that PPARγ attenuates hypoxia-induced endothelin-1 (ET-1) signaling to mediate these therapeutic effects. To test this hypothesis, human pulmonary artery endothelial cells (HPAECs) were exposed to normoxia or hypoxia (1% O2) for 72 h and treated with or without the PPARγ ligand rosiglitazone (RSG, 10 μM) during the final 24 h of exposure. HPAEC proliferation was measured with MTT assays or cell counting, and mRNA and protein levels of ET-1 signaling components were determined. To explore the role of hypoxia-activated transcription factors, selected HPAECs were treated with inhibitors of hypoxia-inducible factor (HIF)-1α (chetomin) or nuclear factor (NF)-κB (caffeic acid phenethyl ester, CAPE). In parallel studies, male C57BL/6 mice were exposed to normoxia (21% O2) or hypoxia (10% O2) for 3 wk with or without gavage with RSG (10 mg·kg−1·day−1) for the final 10 days of exposure. Hypoxia increased ET-1, endothelin-converting enzyme-1, and endothelin receptor A and B levels in mouse lung and in HPAECs and increased HPAEC proliferation. Treatment with RSG attenuated hypoxia-induced activation of HIF-1α, NF-κB activation, and ET-1 signaling pathway components. Similarly, treatment with chetomin or CAPE prevented hypoxia-induced increases in HPAEC ET-1 mRNA and protein levels. These findings indicate that PPARγ activation attenuates a program of hypoxia-induced ET-1 signaling by inhibiting activation of hypoxia-responsive transcription factors. Targeting PPARγ represents a novel therapeutic strategy to inhibit enhanced ET-1 signaling in PH pathogenesis.
American Physiological Society