Regulation of Cigarette Smoke (CS)-Induced Autophagy by Nrf2.

Cigarette smoke (CS) has been reported to induce autophagy in airway epithelial cells. The subsequent autophagic cell death has been proposed to play an important pathogenic role in chronic obstructive pulmonary disease (COPD); however, the underlying molecular mechanism is not entirely clear. Using...

Full description

Bibliographic Details
Main Authors: Lingxiang Zhu, Erika C Barrett, Yuxue Xu, Zuguo Liu, Aditya Manoharan, Yin Chen
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2013-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC3621864?pdf=render
id doaj-e8951ded51a34ec4844631a144c625f7
record_format Article
spelling doaj-e8951ded51a34ec4844631a144c625f72020-11-25T00:47:14ZengPublic Library of Science (PLoS)PLoS ONE1932-62032013-01-0184e5569510.1371/journal.pone.0055695Regulation of Cigarette Smoke (CS)-Induced Autophagy by Nrf2.Lingxiang ZhuErika C BarrettYuxue XuZuguo LiuAditya ManoharanYin ChenCigarette smoke (CS) has been reported to induce autophagy in airway epithelial cells. The subsequent autophagic cell death has been proposed to play an important pathogenic role in chronic obstructive pulmonary disease (COPD); however, the underlying molecular mechanism is not entirely clear. Using CS extract (CSE) as a surrogate for CS, we found that it markedly increased the expressions of both LC3B-I and LC3B-II as well as autophagosomes in airway epithelial cells. This is in contrast to the common autophagy inducer (i.e., starvation) that increases LC3B-II but reduces LC3B-I. Further studies indicate that CSE regulated LC3B at transcriptional and post-translational levels. In addition, CSE, but not starvation, activated Nrf2-mediated adaptive response. Increase of cellular Nrf2 by either Nrf2 overexpression or the knockdown of Keap1 (an Nrf2 inhibitor) significantly repressed CSE-induced LC3B-I and II as well as autophagosomes. Supplement of NAC (a GSH precursor) or GSH recapitulated the effect of Nrf2, suggesting the increase of cellular GSH level is responsible for Nrf2 effect on LC3B and autophagosome. Interestingly, neither Nrf2 activation nor GSH supplement could restore the repressed activities of mTOR or its downstream effctor-S6K. Thus, the Nrf2-dependent autophagy-suppression was not due to the re-activation of mTOR-the master repressor of autophagy. To search for the downstream effector of Nrf2 on LC3B and autophagosome, we tested Nrf2-dependent genes (i.e., NQO1 and P62) that are also increased by CSE treatment. We found that P62, but not NQO1, could mimic the effect of Nrf2 activation by repressing LC3B expression. Thus, Nrf2->P62 appears to play an important role in the regulation of CSE-induced LC3B and autophagosome.http://europepmc.org/articles/PMC3621864?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Lingxiang Zhu
Erika C Barrett
Yuxue Xu
Zuguo Liu
Aditya Manoharan
Yin Chen
spellingShingle Lingxiang Zhu
Erika C Barrett
Yuxue Xu
Zuguo Liu
Aditya Manoharan
Yin Chen
Regulation of Cigarette Smoke (CS)-Induced Autophagy by Nrf2.
PLoS ONE
author_facet Lingxiang Zhu
Erika C Barrett
Yuxue Xu
Zuguo Liu
Aditya Manoharan
Yin Chen
author_sort Lingxiang Zhu
title Regulation of Cigarette Smoke (CS)-Induced Autophagy by Nrf2.
title_short Regulation of Cigarette Smoke (CS)-Induced Autophagy by Nrf2.
title_full Regulation of Cigarette Smoke (CS)-Induced Autophagy by Nrf2.
title_fullStr Regulation of Cigarette Smoke (CS)-Induced Autophagy by Nrf2.
title_full_unstemmed Regulation of Cigarette Smoke (CS)-Induced Autophagy by Nrf2.
title_sort regulation of cigarette smoke (cs)-induced autophagy by nrf2.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2013-01-01
description Cigarette smoke (CS) has been reported to induce autophagy in airway epithelial cells. The subsequent autophagic cell death has been proposed to play an important pathogenic role in chronic obstructive pulmonary disease (COPD); however, the underlying molecular mechanism is not entirely clear. Using CS extract (CSE) as a surrogate for CS, we found that it markedly increased the expressions of both LC3B-I and LC3B-II as well as autophagosomes in airway epithelial cells. This is in contrast to the common autophagy inducer (i.e., starvation) that increases LC3B-II but reduces LC3B-I. Further studies indicate that CSE regulated LC3B at transcriptional and post-translational levels. In addition, CSE, but not starvation, activated Nrf2-mediated adaptive response. Increase of cellular Nrf2 by either Nrf2 overexpression or the knockdown of Keap1 (an Nrf2 inhibitor) significantly repressed CSE-induced LC3B-I and II as well as autophagosomes. Supplement of NAC (a GSH precursor) or GSH recapitulated the effect of Nrf2, suggesting the increase of cellular GSH level is responsible for Nrf2 effect on LC3B and autophagosome. Interestingly, neither Nrf2 activation nor GSH supplement could restore the repressed activities of mTOR or its downstream effctor-S6K. Thus, the Nrf2-dependent autophagy-suppression was not due to the re-activation of mTOR-the master repressor of autophagy. To search for the downstream effector of Nrf2 on LC3B and autophagosome, we tested Nrf2-dependent genes (i.e., NQO1 and P62) that are also increased by CSE treatment. We found that P62, but not NQO1, could mimic the effect of Nrf2 activation by repressing LC3B expression. Thus, Nrf2->P62 appears to play an important role in the regulation of CSE-induced LC3B and autophagosome.
url http://europepmc.org/articles/PMC3621864?pdf=render
work_keys_str_mv AT lingxiangzhu regulationofcigarettesmokecsinducedautophagybynrf2
AT erikacbarrett regulationofcigarettesmokecsinducedautophagybynrf2
AT yuxuexu regulationofcigarettesmokecsinducedautophagybynrf2
AT zuguoliu regulationofcigarettesmokecsinducedautophagybynrf2
AT adityamanoharan regulationofcigarettesmokecsinducedautophagybynrf2
AT yinchen regulationofcigarettesmokecsinducedautophagybynrf2
_version_ 1725261038401617920