author_facet Huang, Huaping
Chen, Mingjing
Liu, Feng
Wu, Haifeng
Wang, Jie
Chen, Jialiang
Liu, Meihua
Li, Xi
Huang, Huaping
Chen, Mingjing
Liu, Feng
Wu, Haifeng
Wang, Jie
Chen, Jialiang
Liu, Meihua
Li, Xi
author Huang, Huaping
Chen, Mingjing
Liu, Feng
Wu, Haifeng
Wang, Jie
Chen, Jialiang
Liu, Meihua
Li, Xi
spellingShingle Huang, Huaping
Chen, Mingjing
Liu, Feng
Wu, Haifeng
Wang, Jie
Chen, Jialiang
Liu, Meihua
Li, Xi
Bioscience Reports
N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
Cell Biology
Molecular Biology
Biochemistry
Biophysics
author_sort huang, huaping
spelling Huang, Huaping Chen, Mingjing Liu, Feng Wu, Haifeng Wang, Jie Chen, Jialiang Liu, Meihua Li, Xi 0144-8463 1573-4935 Portland Press Ltd. Cell Biology Molecular Biology Biochemistry Biophysics http://dx.doi.org/10.1042/bsr20190681 <jats:title>Abstract</jats:title> <jats:p>Silicosis is a lethal pneumoconiosis disease characterized by chronic lung inflammation and fibrosis. The present study was to explore the effect of against crystalline silica (CS)-induced pulmonary fibrosis. A total of 138 wild-type C57BL/6J mice were divided into control and experimental groups, and killed on month 0, 1, 2, 3, 4, and 5. Different doses of N-acetylcysteine (NAC) were gavaged to the mice after CS instillation to observe the effect of NAC on CS induced pulmonary fibrosis and inflammation. The pulmonary injury was evaluated with Hematoxylin and eosin/Masson staining. Reactive oxygen species level was analyzed by DCFH-DA labeling. Commercial ELISA kits were used to determine antioxidant activity (T-AOC, glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) and cytokines (TNF-α, IL-1β, IL-4, and IL-6). The expression of oxidising enzymes (NOX2, iNOS, SOD2, and XO) were detected by real time PCR. Immunohistochemistry (IHC) staining was performed to examine epithelial-mesenchymal transition-related markers. The mice treated with NAC presented markedly reduced CS-induced pulmonary injury and ameliorated CS-induced pulmonary fibrosis and inflammation. The level of malondialdehyde was reduced, while the activities of GSH-PX, SOD, and T-AOC were markedly enhanced by NAC. We also found the down-regulation of oxidising enzymes (NOX2, iNOS, SOD2, and XO) after NAC treatment. Moreover, E-cadherin expression was increased while vimentin and Cytochrome C expressions were decreased by NAC. These encouraging findings suggest that NAC exerts pulmonary protective effects in CS-induced pulmonary fibrosis and might be considered as a promising agent for the treatment of silicosis.</jats:p> N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis Bioscience Reports
doi_str_mv 10.1042/bsr20190681
facet_avail Online
Free
finc_class_facet Biologie
Chemie und Pharmazie
Physik
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTA0Mi9ic3IyMDE5MDY4MQ
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTA0Mi9ic3IyMDE5MDY4MQ
institution DE-Gla1
DE-Zi4
DE-15
DE-Rs1
DE-Pl11
DE-105
DE-14
DE-Ch1
DE-L229
DE-D275
DE-Bn3
DE-Brt1
DE-Zwi2
DE-D161
imprint Portland Press Ltd., 2019
imprint_str_mv Portland Press Ltd., 2019
issn 0144-8463
1573-4935
issn_str_mv 0144-8463
1573-4935
language English
mega_collection Portland Press Ltd. (CrossRef)
match_str huang2019nacetylcysteinetiherapeuticallyprotectsagainstpulmonaryfibrosisinamousemodelofsilicosis
publishDateSort 2019
publisher Portland Press Ltd.
recordtype ai
record_format ai
series Bioscience Reports
source_id 49
title N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_unstemmed N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_full N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_fullStr N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_full_unstemmed N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_short N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_sort n-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
topic Cell Biology
Molecular Biology
Biochemistry
Biophysics
url http://dx.doi.org/10.1042/bsr20190681
publishDate 2019
physical
description <jats:title>Abstract</jats:title> <jats:p>Silicosis is a lethal pneumoconiosis disease characterized by chronic lung inflammation and fibrosis. The present study was to explore the effect of against crystalline silica (CS)-induced pulmonary fibrosis. A total of 138 wild-type C57BL/6J mice were divided into control and experimental groups, and killed on month 0, 1, 2, 3, 4, and 5. Different doses of N-acetylcysteine (NAC) were gavaged to the mice after CS instillation to observe the effect of NAC on CS induced pulmonary fibrosis and inflammation. The pulmonary injury was evaluated with Hematoxylin and eosin/Masson staining. Reactive oxygen species level was analyzed by DCFH-DA labeling. Commercial ELISA kits were used to determine antioxidant activity (T-AOC, glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) and cytokines (TNF-α, IL-1β, IL-4, and IL-6). The expression of oxidising enzymes (NOX2, iNOS, SOD2, and XO) were detected by real time PCR. Immunohistochemistry (IHC) staining was performed to examine epithelial-mesenchymal transition-related markers. The mice treated with NAC presented markedly reduced CS-induced pulmonary injury and ameliorated CS-induced pulmonary fibrosis and inflammation. The level of malondialdehyde was reduced, while the activities of GSH-PX, SOD, and T-AOC were markedly enhanced by NAC. We also found the down-regulation of oxidising enzymes (NOX2, iNOS, SOD2, and XO) after NAC treatment. Moreover, E-cadherin expression was increased while vimentin and Cytochrome C expressions were decreased by NAC. These encouraging findings suggest that NAC exerts pulmonary protective effects in CS-induced pulmonary fibrosis and might be considered as a promising agent for the treatment of silicosis.</jats:p>
container_issue 7
container_start_page 0
container_title Bioscience Reports
container_volume 39
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
_version_ 1792344669081829380
geogr_code not assigned
last_indexed 2024-03-01T17:11:16.404Z
geogr_code_person not assigned
openURL url_ver=Z39.88-2004&ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fvufind.svn.sourceforge.net%3Agenerator&rft.title=N-acetylcysteine+tiherapeutically+protects+against+pulmonary+fibrosis+in+a+mouse+model+of+silicosis&rft.date=2019-07-31&genre=article&issn=1573-4935&volume=39&issue=7&jtitle=Bioscience+Reports&atitle=N-acetylcysteine+tiherapeutically+protects+against+pulmonary+fibrosis+in+a+mouse+model+of+silicosis&aulast=Li&aufirst=Xi&rft_id=info%3Adoi%2F10.1042%2Fbsr20190681&rft.language%5B0%5D=eng
SOLR
_version_ 1792344669081829380
author Huang, Huaping, Chen, Mingjing, Liu, Feng, Wu, Haifeng, Wang, Jie, Chen, Jialiang, Liu, Meihua, Li, Xi
author_facet Huang, Huaping, Chen, Mingjing, Liu, Feng, Wu, Haifeng, Wang, Jie, Chen, Jialiang, Liu, Meihua, Li, Xi, Huang, Huaping, Chen, Mingjing, Liu, Feng, Wu, Haifeng, Wang, Jie, Chen, Jialiang, Liu, Meihua, Li, Xi
author_sort huang, huaping
container_issue 7
container_start_page 0
container_title Bioscience Reports
container_volume 39
description <jats:title>Abstract</jats:title> <jats:p>Silicosis is a lethal pneumoconiosis disease characterized by chronic lung inflammation and fibrosis. The present study was to explore the effect of against crystalline silica (CS)-induced pulmonary fibrosis. A total of 138 wild-type C57BL/6J mice were divided into control and experimental groups, and killed on month 0, 1, 2, 3, 4, and 5. Different doses of N-acetylcysteine (NAC) were gavaged to the mice after CS instillation to observe the effect of NAC on CS induced pulmonary fibrosis and inflammation. The pulmonary injury was evaluated with Hematoxylin and eosin/Masson staining. Reactive oxygen species level was analyzed by DCFH-DA labeling. Commercial ELISA kits were used to determine antioxidant activity (T-AOC, glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) and cytokines (TNF-α, IL-1β, IL-4, and IL-6). The expression of oxidising enzymes (NOX2, iNOS, SOD2, and XO) were detected by real time PCR. Immunohistochemistry (IHC) staining was performed to examine epithelial-mesenchymal transition-related markers. The mice treated with NAC presented markedly reduced CS-induced pulmonary injury and ameliorated CS-induced pulmonary fibrosis and inflammation. The level of malondialdehyde was reduced, while the activities of GSH-PX, SOD, and T-AOC were markedly enhanced by NAC. We also found the down-regulation of oxidising enzymes (NOX2, iNOS, SOD2, and XO) after NAC treatment. Moreover, E-cadherin expression was increased while vimentin and Cytochrome C expressions were decreased by NAC. These encouraging findings suggest that NAC exerts pulmonary protective effects in CS-induced pulmonary fibrosis and might be considered as a promising agent for the treatment of silicosis.</jats:p>
doi_str_mv 10.1042/bsr20190681
facet_avail Online, Free
finc_class_facet Biologie, Chemie und Pharmazie, Physik
format ElectronicArticle
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
geogr_code not assigned
geogr_code_person not assigned
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTA0Mi9ic3IyMDE5MDY4MQ
imprint Portland Press Ltd., 2019
imprint_str_mv Portland Press Ltd., 2019
institution DE-Gla1, DE-Zi4, DE-15, DE-Rs1, DE-Pl11, DE-105, DE-14, DE-Ch1, DE-L229, DE-D275, DE-Bn3, DE-Brt1, DE-Zwi2, DE-D161
issn 0144-8463, 1573-4935
issn_str_mv 0144-8463, 1573-4935
language English
last_indexed 2024-03-01T17:11:16.404Z
match_str huang2019nacetylcysteinetiherapeuticallyprotectsagainstpulmonaryfibrosisinamousemodelofsilicosis
mega_collection Portland Press Ltd. (CrossRef)
physical
publishDate 2019
publishDateSort 2019
publisher Portland Press Ltd.
record_format ai
recordtype ai
series Bioscience Reports
source_id 49
spelling Huang, Huaping Chen, Mingjing Liu, Feng Wu, Haifeng Wang, Jie Chen, Jialiang Liu, Meihua Li, Xi 0144-8463 1573-4935 Portland Press Ltd. Cell Biology Molecular Biology Biochemistry Biophysics http://dx.doi.org/10.1042/bsr20190681 <jats:title>Abstract</jats:title> <jats:p>Silicosis is a lethal pneumoconiosis disease characterized by chronic lung inflammation and fibrosis. The present study was to explore the effect of against crystalline silica (CS)-induced pulmonary fibrosis. A total of 138 wild-type C57BL/6J mice were divided into control and experimental groups, and killed on month 0, 1, 2, 3, 4, and 5. Different doses of N-acetylcysteine (NAC) were gavaged to the mice after CS instillation to observe the effect of NAC on CS induced pulmonary fibrosis and inflammation. The pulmonary injury was evaluated with Hematoxylin and eosin/Masson staining. Reactive oxygen species level was analyzed by DCFH-DA labeling. Commercial ELISA kits were used to determine antioxidant activity (T-AOC, glutathione peroxidase (GSH-Px), and superoxide dismutase (SOD) and cytokines (TNF-α, IL-1β, IL-4, and IL-6). The expression of oxidising enzymes (NOX2, iNOS, SOD2, and XO) were detected by real time PCR. Immunohistochemistry (IHC) staining was performed to examine epithelial-mesenchymal transition-related markers. The mice treated with NAC presented markedly reduced CS-induced pulmonary injury and ameliorated CS-induced pulmonary fibrosis and inflammation. The level of malondialdehyde was reduced, while the activities of GSH-PX, SOD, and T-AOC were markedly enhanced by NAC. We also found the down-regulation of oxidising enzymes (NOX2, iNOS, SOD2, and XO) after NAC treatment. Moreover, E-cadherin expression was increased while vimentin and Cytochrome C expressions were decreased by NAC. These encouraging findings suggest that NAC exerts pulmonary protective effects in CS-induced pulmonary fibrosis and might be considered as a promising agent for the treatment of silicosis.</jats:p> N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis Bioscience Reports
spellingShingle Huang, Huaping, Chen, Mingjing, Liu, Feng, Wu, Haifeng, Wang, Jie, Chen, Jialiang, Liu, Meihua, Li, Xi, Bioscience Reports, N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis, Cell Biology, Molecular Biology, Biochemistry, Biophysics
title N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_full N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_fullStr N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_full_unstemmed N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_short N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_sort n-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
title_unstemmed N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis
topic Cell Biology, Molecular Biology, Biochemistry, Biophysics
url http://dx.doi.org/10.1042/bsr20190681