368 442

Cited 109 times in

Carbon monoxide activates autophagy via mitochondrial reactive oxygen species formation

Authors
 Seon-Jin Lee  ;  Stefan W. Ryter  ;  Jin-Fu Xu  ;  Kiichi Nakahira  ;  Hong Pyo Kim  ;  Augustine M. K. Choi  ;  Young Sam Kim 
Citation
 AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY, Vol.45(4) : 867-873, 2011 
Journal Title
AMERICAN JOURNAL OF RESPIRATORY CELL AND MOLECULAR BIOLOGY
ISSN
 1044-1549 
Issue Date
2011
MeSH
Administration, Inhalation ; Animals ; Antioxidants/pharmacology ; Autophagy/drug effects* ; Carbon Monoxide/administration & dosage ; Carbon Monoxide/pharmacology* ; Cell Line, Tumor ; Cytoprotection ; Disease Models, Animal ; Epithelial Cells/drug effects* ; Epithelial Cells/metabolism ; Epithelial Cells/pathology ; Gases ; Humans ; Hyperoxia/metabolism ; Hyperoxia/pathology ; Hyperoxia/prevention & control* ; Mice ; Mice, Inbred C57BL ; Microscopy, Fluorescence ; Microtubule-Associated Proteins/genetics ; Microtubule-Associated Proteins/metabolism ; Mitochondria/drug effects* ; Mitochondria/metabolism ; Mitochondria/pathology ; Oxidative Stress/drug effects* ; RNA Interference ; Reactive Oxygen Species/metabolism* ; Respiratory Mucosa/drug effects* ; Respiratory Mucosa/metabolism ; Respiratory Mucosa/pathology ; Signal Transduction/drug effects ; Time Factors ; Transfection
Keywords
apoptosis ; autophagy ; carbon monoxide ; epithelial cells ; hyperoxia
Abstract
Autophagy, an autodigestive process that degrades cellular organelles and protein, plays an important role in maintaining cellular homeostasis during environmental stress. Carbon monoxide (CO), a toxic gas and candidate therapeutic molecule, confers cytoprotection in animal models of acute lung injury. The mechanisms underlying CO-dependent lung cell protection and the role of autophagy in this process remain unclear. Here, we demonstrate that CO exposure time-dependently increased the expression and activation of the autophagic protein, microtubule-associated protein-1 light chain-3B (LC3B) in mouse lung, and in cultured human alveolar (A549) or human bronchial epithelial cells. Furthermore, CO increased autophagosome formation in epithelial cells by electron microscopy and green fluorescent protein (GFP)-LC3 puncta assays. Recent studies indicate that reactive oxygen species (ROS) play an important role in the activation of autophagy. CO up-regulated mitochondria-dependent generation of ROS in epithelial cells, as assayed by MitoSOX fluorescence. Furthermore, CO-dependent induction of LC3B expression was inhibited by N-acetyl-L-cysteine and the mitochondria-targeting antioxidant, Mito-TEMPO. These data suggest that CO promotes the autophagic process through mitochondrial ROS generation. We investigated the relationships between autophagic proteins and CO-dependent cytoprotection using a model of hyperoxic stress. CO protected against hyperoxia-induced cell death, and inhibited hyperoxia-associated ROS production. The ability of CO to protect against hyperoxia-induced cell death and caspase-3 activation was compromised in epithelial cells infected with LC3B-small interfering (si)RNA, indicating a role for autophagic proteins. These studies uncover a new mechanism for the protective action of CO, in support of potential therapeutic application of this gas.
Files in This Item:
T201105159.pdf Download
DOI
10.1165/rcmb.2010-0352OC
Appears in Collections:
1. College of Medicine (의과대학) > Dept. of Internal Medicine (내과학교실) > 1. Journal Papers
Yonsei Authors
Kim, Young Sam(김영삼) ORCID logo https://orcid.org/0000-0001-9656-8482
URI
https://ir.ymlib.yonsei.ac.kr/handle/22282913/95014
사서에게 알리기
  feedback

qrcode

Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.

Browse

Links