PPARγ regulates mitochondrial structure and function and human pulmonary artery smooth muscle cell proliferation

SM Yeligar, BY Kang, KM Bijli… - American journal of …, 2018 - atsjournals.org
SM Yeligar, BY Kang, KM Bijli, JM Kleinhenz, TC Murphy, G Torres, A San Martin, RL Sutliff…
American journal of respiratory cell and molecular biology, 2018atsjournals.org
Pulmonary hypertension (PH) is a progressive disorder that causes significant morbidity and
mortality despite existing therapies. PH pathogenesis is characterized by metabolic
derangements that increase pulmonary artery smooth muscle cell (PASMC) proliferation and
vascular remodeling. PH-associated decreases in peroxisome proliferator-activated receptor
γ (PPARγ) stimulate PASMC proliferation, and PPARγ in coordination with PPARγ
coactivator 1α (PGC1α) regulates mitochondrial gene expression and biogenesis. To further …
Pulmonary hypertension (PH) is a progressive disorder that causes significant morbidity and mortality despite existing therapies. PH pathogenesis is characterized by metabolic derangements that increase pulmonary artery smooth muscle cell (PASMC) proliferation and vascular remodeling. PH-associated decreases in peroxisome proliferator-activated receptor γ (PPARγ) stimulate PASMC proliferation, and PPARγ in coordination with PPARγ coactivator 1α (PGC1α) regulates mitochondrial gene expression and biogenesis. To further examine the impact of decreases in PPARγ expression on human PASMC (HPASMC) mitochondrial function, we hypothesized that depletion of either PPARγ or PGC1α perturbs mitochondrial structure and function to stimulate PASMC proliferation. To test this hypothesis, HPASMCs were exposed to hypoxia and treated pharmacologically with the PPARγ antagonist GW9662 or with siRNA against PPARγ or PGC1α for 72 hours. HPASMC proliferation (cell counting), target mRNA levels (qRT-PCR), target protein levels (Western blotting), mitochondria-derived H2O2 (confocal immunofluorescence), mitochondrial mass and fragmentation, and mitochondrial bioenergetic profiling were determined. Hypoxia or knockdown of either PPARγ or PGC1α increased HPASMC proliferation, enhanced mitochondria-derived H2O2, decreased mitochondrial mass, stimulated mitochondrial fragmentation, and impaired mitochondrial bioenergetics. Taken together, these findings provide novel evidence that loss of PPARγ diminishes PGC1α and stimulates derangements in mitochondrial structure and function that cause PASMC proliferation. Overexpression of PGC1α reversed hypoxia-induced HPASMC derangements. This study identifies additional mechanistic underpinnings of PH, and provides support for the notion of activating PPARγ as a novel therapeutic strategy in PH.
ATS Journals