Cargando…

Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway

Oxidative stress caused by chronic intermittent hypoxia (CIH) is the hallmark of obstructive sleep apnea (OSA). Among the first line of defense against oxidative stress is the dismutation of superoxide radicals, which in the mitochondria is carried out by manganese superoxide dismutase (SOD2). In th...

Descripción completa

Detalles Bibliográficos
Autores principales: Song, Jie-qiong, Jiang, Li-yan, Fu, Cui-ping, Wu, Xu, Liu, Zi-long, Xie, Liang, Wu, Xiao-dan, Hao, Sheng-yu, Li, Shan-qun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer Singapore 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7608479/
https://www.ncbi.nlm.nih.gov/pubmed/32066884
http://dx.doi.org/10.1038/s41401-019-0349-y
_version_ 1783604849161535488
author Song, Jie-qiong
Jiang, Li-yan
Fu, Cui-ping
Wu, Xu
Liu, Zi-long
Xie, Liang
Wu, Xiao-dan
Hao, Sheng-yu
Li, Shan-qun
author_facet Song, Jie-qiong
Jiang, Li-yan
Fu, Cui-ping
Wu, Xu
Liu, Zi-long
Xie, Liang
Wu, Xiao-dan
Hao, Sheng-yu
Li, Shan-qun
author_sort Song, Jie-qiong
collection PubMed
description Oxidative stress caused by chronic intermittent hypoxia (CIH) is the hallmark of obstructive sleep apnea (OSA). Among the first line of defense against oxidative stress is the dismutation of superoxide radicals, which in the mitochondria is carried out by manganese superoxide dismutase (SOD2). In this study, wild-type (WT) and SOD2-heterozygous knockout (SOD2(+/−)) mice were exposed to CIH or normoxic (Nor) conditions. After 4 weeks, pulmonary artery pressure was measured, and the mice were processed to harvest either serum for cytokine assays or lungs for flow cytometry and histopathological studies. Herein, we showed that heterozygous deletion of SOD2 markedly deteriorated pulmonary remodeling and increased the oxidative stress, especially promoted the infiltration of macrophages in the lungs of CIH mouse. Moreover, in the intermittent hypoxia (IH)-treated RAW264.7 cells, SOD2 knockdown increased the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome activation accompanied with the IL-1β elevation and caspase-1 activity. Additionally, mitochondrial ROS (mtROS) scavenger mito-TEMPO abolished NLRP3 inflammasome activation in IH-treated RAW264.7 cells. Collectively, our results supported that SOD2 contributed to the pathogenesis of CIH-induced lung remodeling. Meanwhile, SOD2 knockdown exacerbates oxidative damage through assembly and activation of NLRP3 inflammasome in macrophages. SOD2 may be a novel therapeutic target for CIH-induced pulmonary inflammation and arteriole remodeling.
format Online
Article
Text
id pubmed-7608479
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Springer Singapore
record_format MEDLINE/PubMed
spelling pubmed-76084792020-11-05 Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway Song, Jie-qiong Jiang, Li-yan Fu, Cui-ping Wu, Xu Liu, Zi-long Xie, Liang Wu, Xiao-dan Hao, Sheng-yu Li, Shan-qun Acta Pharmacol Sin Article Oxidative stress caused by chronic intermittent hypoxia (CIH) is the hallmark of obstructive sleep apnea (OSA). Among the first line of defense against oxidative stress is the dismutation of superoxide radicals, which in the mitochondria is carried out by manganese superoxide dismutase (SOD2). In this study, wild-type (WT) and SOD2-heterozygous knockout (SOD2(+/−)) mice were exposed to CIH or normoxic (Nor) conditions. After 4 weeks, pulmonary artery pressure was measured, and the mice were processed to harvest either serum for cytokine assays or lungs for flow cytometry and histopathological studies. Herein, we showed that heterozygous deletion of SOD2 markedly deteriorated pulmonary remodeling and increased the oxidative stress, especially promoted the infiltration of macrophages in the lungs of CIH mouse. Moreover, in the intermittent hypoxia (IH)-treated RAW264.7 cells, SOD2 knockdown increased the nucleotide-binding domain-like receptor protein 3 (NLRP3) inflammasome activation accompanied with the IL-1β elevation and caspase-1 activity. Additionally, mitochondrial ROS (mtROS) scavenger mito-TEMPO abolished NLRP3 inflammasome activation in IH-treated RAW264.7 cells. Collectively, our results supported that SOD2 contributed to the pathogenesis of CIH-induced lung remodeling. Meanwhile, SOD2 knockdown exacerbates oxidative damage through assembly and activation of NLRP3 inflammasome in macrophages. SOD2 may be a novel therapeutic target for CIH-induced pulmonary inflammation and arteriole remodeling. Springer Singapore 2020-02-17 2020-09 /pmc/articles/PMC7608479/ /pubmed/32066884 http://dx.doi.org/10.1038/s41401-019-0349-y Text en © CPS and SIMM 2020
spellingShingle Article
Song, Jie-qiong
Jiang, Li-yan
Fu, Cui-ping
Wu, Xu
Liu, Zi-long
Xie, Liang
Wu, Xiao-dan
Hao, Sheng-yu
Li, Shan-qun
Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway
title Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway
title_full Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway
title_fullStr Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway
title_full_unstemmed Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway
title_short Heterozygous SOD2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtROS-NLRP3 signaling pathway
title_sort heterozygous sod2 deletion deteriorated chronic intermittent hypoxia-induced lung inflammation and vascular remodeling through mtros-nlrp3 signaling pathway
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7608479/
https://www.ncbi.nlm.nih.gov/pubmed/32066884
http://dx.doi.org/10.1038/s41401-019-0349-y
work_keys_str_mv AT songjieqiong heterozygoussod2deletiondeterioratedchronicintermittenthypoxiainducedlunginflammationandvascularremodelingthroughmtrosnlrp3signalingpathway
AT jiangliyan heterozygoussod2deletiondeterioratedchronicintermittenthypoxiainducedlunginflammationandvascularremodelingthroughmtrosnlrp3signalingpathway
AT fucuiping heterozygoussod2deletiondeterioratedchronicintermittenthypoxiainducedlunginflammationandvascularremodelingthroughmtrosnlrp3signalingpathway
AT wuxu heterozygoussod2deletiondeterioratedchronicintermittenthypoxiainducedlunginflammationandvascularremodelingthroughmtrosnlrp3signalingpathway
AT liuzilong heterozygoussod2deletiondeterioratedchronicintermittenthypoxiainducedlunginflammationandvascularremodelingthroughmtrosnlrp3signalingpathway
AT xieliang heterozygoussod2deletiondeterioratedchronicintermittenthypoxiainducedlunginflammationandvascularremodelingthroughmtrosnlrp3signalingpathway
AT wuxiaodan heterozygoussod2deletiondeterioratedchronicintermittenthypoxiainducedlunginflammationandvascularremodelingthroughmtrosnlrp3signalingpathway
AT haoshengyu heterozygoussod2deletiondeterioratedchronicintermittenthypoxiainducedlunginflammationandvascularremodelingthroughmtrosnlrp3signalingpathway
AT lishanqun heterozygoussod2deletiondeterioratedchronicintermittenthypoxiainducedlunginflammationandvascularremodelingthroughmtrosnlrp3signalingpathway