Cargando…

PINK1-PARK2-mediated mitophagy in COPD and IPF pathogeneses

Mitochondria regulate not only cell functions through energy generation but also aging-associated cell phenotypes. Impaired mitochondrial structural and functional integrity accompanied by excessive mitochondrial reactive oxygen species (mtROS) production is associated with enhanced programmed cell...

Descripción completa

Detalles Bibliográficos
Autores principales: Tsubouchi, Kazuya, Araya, Jun, Kuwano, Kazuyoshi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6199723/
https://www.ncbi.nlm.nih.gov/pubmed/30386443
http://dx.doi.org/10.1186/s41232-018-0077-6
_version_ 1783365185230077952
author Tsubouchi, Kazuya
Araya, Jun
Kuwano, Kazuyoshi
author_facet Tsubouchi, Kazuya
Araya, Jun
Kuwano, Kazuyoshi
author_sort Tsubouchi, Kazuya
collection PubMed
description Mitochondria regulate not only cell functions through energy generation but also aging-associated cell phenotypes. Impaired mitochondrial structural and functional integrity accompanied by excessive mitochondrial reactive oxygen species (mtROS) production is associated with enhanced programmed cell death (PCD) and cellular senescence. Dysregulation of mechanisms for mitochondrial integrity, including mitophagy, induces accumulation of mitochondrial damage. Mitophagy is a highly conserved mechanism of selectively delivering damaged mitochondria for lysosomal degradation and is mainly governed by phosphatase and tensin homolog (PTEN)-induced putative protein kinase 1 (PINK1) and PARK2. Accumulating evidence suggests that PINK1-PARK2-mediated mitophagy has an important role in the pathogenesis of aging-associated pulmonary disorders, represented by chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). COPD characterized by progressive airflow limitation is mainly caused by cigarette smoke (CS) exposure, and accumulation of damaged mitochondria in bronchial epithelial cells (BEC) has been demonstrated. Intriguingly, both enhanced and impaired mitophagy have been implicated in COPD pathogenesis. Enhanced mitophagy induced by increased PINK1 expression has been associated with programmed necrosis, necroptosis. On the other hand, reduced PARK2 levels were linked to insufficient mitophagy, resulting in accelerated cellular senescence in BEC. Although dominant involvement of PCD and cellular senescence remains unclear, PINK1-PARK2-mediated mitophagy regulates mitochondrial ROS and cell fate during COPD pathogenesis. Involvement of insufficient mitophagy has been proposed in lung fibrosis development during IPF pathogenesis. Accumulation of dysmorphic mitochondria and increased ROS production linked to decrease in PINK1 expression were demonstrated in type II alveolar epithelial cells (AECIIs) in IPF lungs, which can be associated with enhanced apoptosis and cellular senescence. Furthermore, reduced PARK2 expression levels have been shown in myofibroblasts in IPF lungs. Insufficient mitophagy caused by PARK2 deficiency induced mtROS production with concomitantly activated platelet-derived growth factor receptor (PDGFR)/mammalian target of rapamycin (mTOR) signaling, resulting in increased myofibroblast differentiation and proliferation. Inappropriate PINK1-PARK2-mediated mitophagy appears to be mainly responsible for regulating cell fate, including PCD, cellular senescence, and myofibroblast differentiation during COPD and IPF pathogeneses. Modalities to achieve specific and appropriate levels of PINK1-PARK2-mediated mitophagy activation may be a promising therapeutic option to regulate the aging-associated pathology, COPD, and IPF.
format Online
Article
Text
id pubmed-6199723
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-61997232018-10-31 PINK1-PARK2-mediated mitophagy in COPD and IPF pathogeneses Tsubouchi, Kazuya Araya, Jun Kuwano, Kazuyoshi Inflamm Regen Review Mitochondria regulate not only cell functions through energy generation but also aging-associated cell phenotypes. Impaired mitochondrial structural and functional integrity accompanied by excessive mitochondrial reactive oxygen species (mtROS) production is associated with enhanced programmed cell death (PCD) and cellular senescence. Dysregulation of mechanisms for mitochondrial integrity, including mitophagy, induces accumulation of mitochondrial damage. Mitophagy is a highly conserved mechanism of selectively delivering damaged mitochondria for lysosomal degradation and is mainly governed by phosphatase and tensin homolog (PTEN)-induced putative protein kinase 1 (PINK1) and PARK2. Accumulating evidence suggests that PINK1-PARK2-mediated mitophagy has an important role in the pathogenesis of aging-associated pulmonary disorders, represented by chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). COPD characterized by progressive airflow limitation is mainly caused by cigarette smoke (CS) exposure, and accumulation of damaged mitochondria in bronchial epithelial cells (BEC) has been demonstrated. Intriguingly, both enhanced and impaired mitophagy have been implicated in COPD pathogenesis. Enhanced mitophagy induced by increased PINK1 expression has been associated with programmed necrosis, necroptosis. On the other hand, reduced PARK2 levels were linked to insufficient mitophagy, resulting in accelerated cellular senescence in BEC. Although dominant involvement of PCD and cellular senescence remains unclear, PINK1-PARK2-mediated mitophagy regulates mitochondrial ROS and cell fate during COPD pathogenesis. Involvement of insufficient mitophagy has been proposed in lung fibrosis development during IPF pathogenesis. Accumulation of dysmorphic mitochondria and increased ROS production linked to decrease in PINK1 expression were demonstrated in type II alveolar epithelial cells (AECIIs) in IPF lungs, which can be associated with enhanced apoptosis and cellular senescence. Furthermore, reduced PARK2 expression levels have been shown in myofibroblasts in IPF lungs. Insufficient mitophagy caused by PARK2 deficiency induced mtROS production with concomitantly activated platelet-derived growth factor receptor (PDGFR)/mammalian target of rapamycin (mTOR) signaling, resulting in increased myofibroblast differentiation and proliferation. Inappropriate PINK1-PARK2-mediated mitophagy appears to be mainly responsible for regulating cell fate, including PCD, cellular senescence, and myofibroblast differentiation during COPD and IPF pathogeneses. Modalities to achieve specific and appropriate levels of PINK1-PARK2-mediated mitophagy activation may be a promising therapeutic option to regulate the aging-associated pathology, COPD, and IPF. BioMed Central 2018-10-24 /pmc/articles/PMC6199723/ /pubmed/30386443 http://dx.doi.org/10.1186/s41232-018-0077-6 Text en © The Author(s) 2018 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Review
Tsubouchi, Kazuya
Araya, Jun
Kuwano, Kazuyoshi
PINK1-PARK2-mediated mitophagy in COPD and IPF pathogeneses
title PINK1-PARK2-mediated mitophagy in COPD and IPF pathogeneses
title_full PINK1-PARK2-mediated mitophagy in COPD and IPF pathogeneses
title_fullStr PINK1-PARK2-mediated mitophagy in COPD and IPF pathogeneses
title_full_unstemmed PINK1-PARK2-mediated mitophagy in COPD and IPF pathogeneses
title_short PINK1-PARK2-mediated mitophagy in COPD and IPF pathogeneses
title_sort pink1-park2-mediated mitophagy in copd and ipf pathogeneses
topic Review
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6199723/
https://www.ncbi.nlm.nih.gov/pubmed/30386443
http://dx.doi.org/10.1186/s41232-018-0077-6
work_keys_str_mv AT tsubouchikazuya pink1park2mediatedmitophagyincopdandipfpathogeneses
AT arayajun pink1park2mediatedmitophagyincopdandipfpathogeneses
AT kuwanokazuyoshi pink1park2mediatedmitophagyincopdandipfpathogeneses