[HTML][HTML] Mitophagy-dependent necroptosis contributes to the pathogenesis of COPD

K Mizumura, SM Cloonan, K Nakahira… - The Journal of …, 2014 - Am Soc Clin Investig
K Mizumura, SM Cloonan, K Nakahira, AR Bhashyam, M Cervo, T Kitada, K Glass, CA Owen
The Journal of clinical investigation, 2014Am Soc Clin Investig
The pathogenesis of chronic obstructive pulmonary disease (COPD) remains unclear, but
involves loss of alveolar surface area (emphysema) and airway inflammation (bronchitis) as
the consequence of cigarette smoke (CS) exposure. Previously, we demonstrated that
autophagy proteins promote lung epithelial cell death, airway dysfunction, and emphysema
in response to CS; however, the underlying mechanisms have yet to be elucidated. Here,
using cultured pulmonary epithelial cells and murine models, we demonstrated that CS …
The pathogenesis of chronic obstructive pulmonary disease (COPD) remains unclear, but involves loss of alveolar surface area (emphysema) and airway inflammation (bronchitis) as the consequence of cigarette smoke (CS) exposure. Previously, we demonstrated that autophagy proteins promote lung epithelial cell death, airway dysfunction, and emphysema in response to CS; however, the underlying mechanisms have yet to be elucidated. Here, using cultured pulmonary epithelial cells and murine models, we demonstrated that CS causes mitochondrial dysfunction that is associated with a reduction of mitochondrial membrane potential. CS induced mitophagy, the autophagy-dependent elimination of mitochondria, through stabilization of the mitophagy regulator PINK1. CS caused cell death, which was reduced by administration of necrosis or necroptosis inhibitors. Genetic deficiency of PINK1 and the mitochondrial division/mitophagy inhibitor Mdivi-1 protected against CS-induced cell death and mitochondrial dysfunction in vitro and reduced the phosphorylation of MLKL, a substrate for RIP3 in the necroptosis pathway. Moreover, Pink1–/– mice were protected against mitochondrial dysfunction, airspace enlargement, and mucociliary clearance (MCC) disruption during CS exposure. Mdivi-1 treatment also ameliorated CS-induced MCC disruption in CS-exposed mice. In human COPD, lung epithelial cells displayed increased expression of PINK1 and RIP3. These findings implicate mitophagy-dependent necroptosis in lung emphysematous changes in response to CS exposure, suggesting that this pathway is a therapeutic target for COPD.
The Journal of Clinical Investigation