Involvement of PARK2-mediated mitophagy in idiopathic pulmonary fibrosis pathogenesis

K Kobayashi, J Araya, S Minagawa, H Hara… - The Journal of …, 2016 - journals.aai.org
K Kobayashi, J Araya, S Minagawa, H Hara, N Saito, T Kadota, N Sato, M Yoshida…
The Journal of Immunology, 2016journals.aai.org
Fibroblastic foci, known to be the leading edge of fibrosis development in idiopathic
pulmonary fibrosis (IPF), are composed of fibrogenic myofibroblasts. Autophagy has been
implicated in the regulation of myofibroblast differentiation. Insufficient mitophagy, the
mitochondria-selective autophagy, results in increased reactive oxygen species, which may
modulate cell signaling pathways for myofibroblast differentiation. Therefore, we sought to
investigate the regulatory role of mitophagy in myofibroblast differentiation as a part of IPF …
Abstract
Fibroblastic foci, known to be the leading edge of fibrosis development in idiopathic pulmonary fibrosis (IPF), are composed of fibrogenic myofibroblasts. Autophagy has been implicated in the regulation of myofibroblast differentiation. Insufficient mitophagy, the mitochondria-selective autophagy, results in increased reactive oxygen species, which may modulate cell signaling pathways for myofibroblast differentiation. Therefore, we sought to investigate the regulatory role of mitophagy in myofibroblast differentiation as a part of IPF pathogenesis. Lung fibroblasts were used in in vitro experiments. Immunohistochemical evaluation in IPF lung tissues was performed. PARK2 was examined as a target molecule for mitophagy regulation, and a PARK2 knockout mouse was employed in a bleomycin-induced lung fibrosis model. We demonstrated that PARK2 knockdown-mediated mitophagy inhibition was involved in the mechanism for activation of the platelet-derived growth factor receptor (PDGFR)/PI3K/AKT signaling pathway accompanied by enhanced myofibroblast differentiation and proliferation, which were clearly inhibited by treatment with both antioxidants and AG1296, a PDGFR inhibitor. Mitophagy inhibition–mediated activation of PDGFR signaling was responsible for further autophagy suppression, suggesting the existence of a self-amplifying loop of mitophagy inhibition and PDGFR activation. IPF lung demonstrated reduced PARK2 with concomitantly increased PDGFR phosphorylation. Furthermore, bleomycin-induced lung fibrosis was enhanced in PARK2 knockout mice and subsequently inhibited by AG1296. These findings suggest that insufficient mitophagy-mediated PDGFR/PI3K/AKT activation, which is mainly attributed to reduced PARK2 expression, is a potent underlying mechanism for myofibroblast differentiation and proliferation in fibroblastic foci formation during IPF pathogenesis.
journals.aai.org