Cargando…

Anti-inflammatory effect of Yu-Ping-Feng-San via TGF-β1 signaling suppression in rat model of COPD

OBJECTIVE(S): Yu-Ping-Feng-San (YPFS) is a classical traditional Chinese medicine that is widely used for treatment of the diseases in respiratory systems, including chronic obstructive pulmonary disease (COPD) recognized as chronic inflammatory disease. However, the molecular mechanism remains uncl...

Descripción completa

Detalles Bibliográficos
Autores principales: Yang, Zhong-Shan, Yan, Jin-Yuan, Han, Ni-Ping, Zhou, Wei, Cheng, Yu, Zhang, Xiao-Mei, Li, Ning, Yuan, Jia-Li
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Mashhad University of Medical Sciences 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5080430/
https://www.ncbi.nlm.nih.gov/pubmed/27803787
_version_ 1782462712227299328
author Yang, Zhong-Shan
Yan, Jin-Yuan
Han, Ni-Ping
Zhou, Wei
Cheng, Yu
Zhang, Xiao-Mei
Li, Ning
Yuan, Jia-Li
author_facet Yang, Zhong-Shan
Yan, Jin-Yuan
Han, Ni-Ping
Zhou, Wei
Cheng, Yu
Zhang, Xiao-Mei
Li, Ning
Yuan, Jia-Li
author_sort Yang, Zhong-Shan
collection PubMed
description OBJECTIVE(S): Yu-Ping-Feng-San (YPFS) is a classical traditional Chinese medicine that is widely used for treatment of the diseases in respiratory systems, including chronic obstructive pulmonary disease (COPD) recognized as chronic inflammatory disease. However, the molecular mechanism remains unclear. Here we detected the factors involved in transforming growth factor beta 1 (TGF-β1)/Smad2 signaling pathway and inflammatory cytokines, to clarify whether YPFS could attenuate inflammatory response dependent on TGF-β1/Smad2 signaling in COPD rats or cigarette smoke extract (CSE)-treated human bronchial epithelial (Beas-2B) cells. MATERIALS AND METHODS: The COPD rat model was established by exposure to cigarette smoke and intratracheal instillation of lipopolysaccharide, YPFS was administered to the animals. The efficacy of YPFS was evaluated by comparing the severity of pulmonary pathological damage, pro-inflammation cytokines, collagen related genes and the activation of TGF-β1/Smad2 signaling pathway. Furthermore, CSE-treated cells were employed to confirm whether the effect of YPFS was dependent on the TGF-β1/Smad2 signaling via knockdown Smad2 (Si-RNA), or pretreatment with the inhibitor of TGF-β1. RESULTS: Administration of YPFS effectively alleviated injury of lung, suppressed releasing of pro-inflammatory cytokines and collagen deposition in COPD animals (P<0.05), whereas exogenous TGF-β1 promoted releasing of IL-1β, IL-6, TNFα (P<0.05). Administration YPFS reduced inflammatory response significantly, also down-regulated TGF-β1/Smad2 signaling in vivo and in vitro. Unexpectedly, knockdown Smad2 or inhibition of TGF-β1 abolished anti-inflammatory effect of YPFS in CSE-treated cells. CONCLUSION: YPFS accomplished anti-inflammatory effects mainly by suppressing phosphorylation of Smad2, TGF-β1/Smad2 signaling pathway was required for YPFS-mediated anti-inflammation in COPD rats or CSE-treated Beas-2B cells.
format Online
Article
Text
id pubmed-5080430
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Mashhad University of Medical Sciences
record_format MEDLINE/PubMed
spelling pubmed-50804302016-11-01 Anti-inflammatory effect of Yu-Ping-Feng-San via TGF-β1 signaling suppression in rat model of COPD Yang, Zhong-Shan Yan, Jin-Yuan Han, Ni-Ping Zhou, Wei Cheng, Yu Zhang, Xiao-Mei Li, Ning Yuan, Jia-Li Iran J Basic Med Sci Original Article OBJECTIVE(S): Yu-Ping-Feng-San (YPFS) is a classical traditional Chinese medicine that is widely used for treatment of the diseases in respiratory systems, including chronic obstructive pulmonary disease (COPD) recognized as chronic inflammatory disease. However, the molecular mechanism remains unclear. Here we detected the factors involved in transforming growth factor beta 1 (TGF-β1)/Smad2 signaling pathway and inflammatory cytokines, to clarify whether YPFS could attenuate inflammatory response dependent on TGF-β1/Smad2 signaling in COPD rats or cigarette smoke extract (CSE)-treated human bronchial epithelial (Beas-2B) cells. MATERIALS AND METHODS: The COPD rat model was established by exposure to cigarette smoke and intratracheal instillation of lipopolysaccharide, YPFS was administered to the animals. The efficacy of YPFS was evaluated by comparing the severity of pulmonary pathological damage, pro-inflammation cytokines, collagen related genes and the activation of TGF-β1/Smad2 signaling pathway. Furthermore, CSE-treated cells were employed to confirm whether the effect of YPFS was dependent on the TGF-β1/Smad2 signaling via knockdown Smad2 (Si-RNA), or pretreatment with the inhibitor of TGF-β1. RESULTS: Administration of YPFS effectively alleviated injury of lung, suppressed releasing of pro-inflammatory cytokines and collagen deposition in COPD animals (P<0.05), whereas exogenous TGF-β1 promoted releasing of IL-1β, IL-6, TNFα (P<0.05). Administration YPFS reduced inflammatory response significantly, also down-regulated TGF-β1/Smad2 signaling in vivo and in vitro. Unexpectedly, knockdown Smad2 or inhibition of TGF-β1 abolished anti-inflammatory effect of YPFS in CSE-treated cells. CONCLUSION: YPFS accomplished anti-inflammatory effects mainly by suppressing phosphorylation of Smad2, TGF-β1/Smad2 signaling pathway was required for YPFS-mediated anti-inflammation in COPD rats or CSE-treated Beas-2B cells. Mashhad University of Medical Sciences 2016-09 /pmc/articles/PMC5080430/ /pubmed/27803787 Text en Copyright: © Iranian Journal of Basic Medical Sciences http://creativecommons.org/licenses/by-nc-sa/3.0 This is an open-access article distributed under the terms of the Creative Commons Attribution-Noncommercial-Share Alike 3.0 Unported, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Article
Yang, Zhong-Shan
Yan, Jin-Yuan
Han, Ni-Ping
Zhou, Wei
Cheng, Yu
Zhang, Xiao-Mei
Li, Ning
Yuan, Jia-Li
Anti-inflammatory effect of Yu-Ping-Feng-San via TGF-β1 signaling suppression in rat model of COPD
title Anti-inflammatory effect of Yu-Ping-Feng-San via TGF-β1 signaling suppression in rat model of COPD
title_full Anti-inflammatory effect of Yu-Ping-Feng-San via TGF-β1 signaling suppression in rat model of COPD
title_fullStr Anti-inflammatory effect of Yu-Ping-Feng-San via TGF-β1 signaling suppression in rat model of COPD
title_full_unstemmed Anti-inflammatory effect of Yu-Ping-Feng-San via TGF-β1 signaling suppression in rat model of COPD
title_short Anti-inflammatory effect of Yu-Ping-Feng-San via TGF-β1 signaling suppression in rat model of COPD
title_sort anti-inflammatory effect of yu-ping-feng-san via tgf-β1 signaling suppression in rat model of copd
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5080430/
https://www.ncbi.nlm.nih.gov/pubmed/27803787
work_keys_str_mv AT yangzhongshan antiinflammatoryeffectofyupingfengsanviatgfb1signalingsuppressioninratmodelofcopd
AT yanjinyuan antiinflammatoryeffectofyupingfengsanviatgfb1signalingsuppressioninratmodelofcopd
AT hanniping antiinflammatoryeffectofyupingfengsanviatgfb1signalingsuppressioninratmodelofcopd
AT zhouwei antiinflammatoryeffectofyupingfengsanviatgfb1signalingsuppressioninratmodelofcopd
AT chengyu antiinflammatoryeffectofyupingfengsanviatgfb1signalingsuppressioninratmodelofcopd
AT zhangxiaomei antiinflammatoryeffectofyupingfengsanviatgfb1signalingsuppressioninratmodelofcopd
AT lining antiinflammatoryeffectofyupingfengsanviatgfb1signalingsuppressioninratmodelofcopd
AT yuanjiali antiinflammatoryeffectofyupingfengsanviatgfb1signalingsuppressioninratmodelofcopd