Cargando…

Induced Pluripotent Stem Cells Inhibit Bleomycin-Induced Pulmonary Fibrosis in Mice through Suppressing TGF-β1/Smad-Mediated Epithelial to Mesenchymal Transition

Pulmonary fibrosis is a progressive and irreversible fibrotic lung disorder with high mortality and few treatment options. Recently, induced pluripotent stem (iPS) cells have been considered as an ideal resource for stem cell-based therapy. Although, an earlier study demonstrated the therapeutic eff...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhou, Yan, He, Zhong, Gao, Yuan, Zheng, Rui, Zhang, Xiaoye, Zhao, Li, Tan, Mingqi
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5108931/
https://www.ncbi.nlm.nih.gov/pubmed/27895584
http://dx.doi.org/10.3389/fphar.2016.00430
_version_ 1782467442200543232
author Zhou, Yan
He, Zhong
Gao, Yuan
Zheng, Rui
Zhang, Xiaoye
Zhao, Li
Tan, Mingqi
author_facet Zhou, Yan
He, Zhong
Gao, Yuan
Zheng, Rui
Zhang, Xiaoye
Zhao, Li
Tan, Mingqi
author_sort Zhou, Yan
collection PubMed
description Pulmonary fibrosis is a progressive and irreversible fibrotic lung disorder with high mortality and few treatment options. Recently, induced pluripotent stem (iPS) cells have been considered as an ideal resource for stem cell-based therapy. Although, an earlier study demonstrated the therapeutic effect of iPS cells on pulmonary fibrosis, the exact mechanisms remain obscure. The present study investigated the effects of iPS cells on inflammatory responses, transforming growth factor (TGF)-β1 signaling pathway, and epithelial to mesenchymal transition (EMT) during bleomycin (BLM)-induced lung fibrosis. A single intratracheal instillation of BLM (5 mg/kg) was performed to induce pulmonary fibrosis in C57BL/6 mice. Then, iPS cells (c-Myc-free) were administrated intravenously at 24 h following BLM instillation. Three weeks after BLM administration, pulmonary fibrosis was evaluated. As expected, treatment with iPS cells significantly limited the pathological changes, edema, and collagen deposition in lung tissues of BLM-induced mice. Mechanically, treatment with iPS cells obviously repressed the expression ratios of matrix metalloproteinase-2 (MMP-2) to its tissue inhibitor -2 (TIMP-2) and MMP-9/TIMP-1 in BLM-induced pulmonary tissues. In addition, iPS cell administration remarkably suppressed BLM-induced up-regulation of pulmonary inflammatory mediators, including tumor necrosis factor-α, interleukin (IL)-1β, IL-6, inducible nitric oxide synthase, nitric oxide, cyclooxygenase-2 and prostaglandin E(2). We further demonstrated that transplantation of iPS cells markedly inhibited BLM-mediated activation of TGF-β1/Mothers against decapentaplegic homolog 2/3 (Smad2/3) and EMT in lung tissues through up-regulating epithelial marker E-cadherin and down-regulating mesenchymal markers including fibronectin, vimentin and α-smooth muscle actin. Moreover, in vitro, iPS cell-conditioned medium (iPSC-CM) profoundly inhibited TGF-β1-induced EMT signaling pathway in mouse alveolar epithelial type II cells (AECII). Collectively, our results suggest that transplantation of iPS cells could suppress inflammatory responses, TGF-β1/Smad2/3 pathway and EMT during the progression of BLM-induced pulmonary fibrosis, providing new useful clues regarding the mechanisms of iPS cells in the treatment for this disease.
format Online
Article
Text
id pubmed-5108931
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-51089312016-11-28 Induced Pluripotent Stem Cells Inhibit Bleomycin-Induced Pulmonary Fibrosis in Mice through Suppressing TGF-β1/Smad-Mediated Epithelial to Mesenchymal Transition Zhou, Yan He, Zhong Gao, Yuan Zheng, Rui Zhang, Xiaoye Zhao, Li Tan, Mingqi Front Pharmacol Pharmacology Pulmonary fibrosis is a progressive and irreversible fibrotic lung disorder with high mortality and few treatment options. Recently, induced pluripotent stem (iPS) cells have been considered as an ideal resource for stem cell-based therapy. Although, an earlier study demonstrated the therapeutic effect of iPS cells on pulmonary fibrosis, the exact mechanisms remain obscure. The present study investigated the effects of iPS cells on inflammatory responses, transforming growth factor (TGF)-β1 signaling pathway, and epithelial to mesenchymal transition (EMT) during bleomycin (BLM)-induced lung fibrosis. A single intratracheal instillation of BLM (5 mg/kg) was performed to induce pulmonary fibrosis in C57BL/6 mice. Then, iPS cells (c-Myc-free) were administrated intravenously at 24 h following BLM instillation. Three weeks after BLM administration, pulmonary fibrosis was evaluated. As expected, treatment with iPS cells significantly limited the pathological changes, edema, and collagen deposition in lung tissues of BLM-induced mice. Mechanically, treatment with iPS cells obviously repressed the expression ratios of matrix metalloproteinase-2 (MMP-2) to its tissue inhibitor -2 (TIMP-2) and MMP-9/TIMP-1 in BLM-induced pulmonary tissues. In addition, iPS cell administration remarkably suppressed BLM-induced up-regulation of pulmonary inflammatory mediators, including tumor necrosis factor-α, interleukin (IL)-1β, IL-6, inducible nitric oxide synthase, nitric oxide, cyclooxygenase-2 and prostaglandin E(2). We further demonstrated that transplantation of iPS cells markedly inhibited BLM-mediated activation of TGF-β1/Mothers against decapentaplegic homolog 2/3 (Smad2/3) and EMT in lung tissues through up-regulating epithelial marker E-cadherin and down-regulating mesenchymal markers including fibronectin, vimentin and α-smooth muscle actin. Moreover, in vitro, iPS cell-conditioned medium (iPSC-CM) profoundly inhibited TGF-β1-induced EMT signaling pathway in mouse alveolar epithelial type II cells (AECII). Collectively, our results suggest that transplantation of iPS cells could suppress inflammatory responses, TGF-β1/Smad2/3 pathway and EMT during the progression of BLM-induced pulmonary fibrosis, providing new useful clues regarding the mechanisms of iPS cells in the treatment for this disease. Frontiers Media S.A. 2016-11-15 /pmc/articles/PMC5108931/ /pubmed/27895584 http://dx.doi.org/10.3389/fphar.2016.00430 Text en Copyright © 2016 Zhou, He, Gao, Zheng, Zhang, Zhao and Tan. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Pharmacology
Zhou, Yan
He, Zhong
Gao, Yuan
Zheng, Rui
Zhang, Xiaoye
Zhao, Li
Tan, Mingqi
Induced Pluripotent Stem Cells Inhibit Bleomycin-Induced Pulmonary Fibrosis in Mice through Suppressing TGF-β1/Smad-Mediated Epithelial to Mesenchymal Transition
title Induced Pluripotent Stem Cells Inhibit Bleomycin-Induced Pulmonary Fibrosis in Mice through Suppressing TGF-β1/Smad-Mediated Epithelial to Mesenchymal Transition
title_full Induced Pluripotent Stem Cells Inhibit Bleomycin-Induced Pulmonary Fibrosis in Mice through Suppressing TGF-β1/Smad-Mediated Epithelial to Mesenchymal Transition
title_fullStr Induced Pluripotent Stem Cells Inhibit Bleomycin-Induced Pulmonary Fibrosis in Mice through Suppressing TGF-β1/Smad-Mediated Epithelial to Mesenchymal Transition
title_full_unstemmed Induced Pluripotent Stem Cells Inhibit Bleomycin-Induced Pulmonary Fibrosis in Mice through Suppressing TGF-β1/Smad-Mediated Epithelial to Mesenchymal Transition
title_short Induced Pluripotent Stem Cells Inhibit Bleomycin-Induced Pulmonary Fibrosis in Mice through Suppressing TGF-β1/Smad-Mediated Epithelial to Mesenchymal Transition
title_sort induced pluripotent stem cells inhibit bleomycin-induced pulmonary fibrosis in mice through suppressing tgf-β1/smad-mediated epithelial to mesenchymal transition
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5108931/
https://www.ncbi.nlm.nih.gov/pubmed/27895584
http://dx.doi.org/10.3389/fphar.2016.00430
work_keys_str_mv AT zhouyan inducedpluripotentstemcellsinhibitbleomycininducedpulmonaryfibrosisinmicethroughsuppressingtgfb1smadmediatedepithelialtomesenchymaltransition
AT hezhong inducedpluripotentstemcellsinhibitbleomycininducedpulmonaryfibrosisinmicethroughsuppressingtgfb1smadmediatedepithelialtomesenchymaltransition
AT gaoyuan inducedpluripotentstemcellsinhibitbleomycininducedpulmonaryfibrosisinmicethroughsuppressingtgfb1smadmediatedepithelialtomesenchymaltransition
AT zhengrui inducedpluripotentstemcellsinhibitbleomycininducedpulmonaryfibrosisinmicethroughsuppressingtgfb1smadmediatedepithelialtomesenchymaltransition
AT zhangxiaoye inducedpluripotentstemcellsinhibitbleomycininducedpulmonaryfibrosisinmicethroughsuppressingtgfb1smadmediatedepithelialtomesenchymaltransition
AT zhaoli inducedpluripotentstemcellsinhibitbleomycininducedpulmonaryfibrosisinmicethroughsuppressingtgfb1smadmediatedepithelialtomesenchymaltransition
AT tanmingqi inducedpluripotentstemcellsinhibitbleomycininducedpulmonaryfibrosisinmicethroughsuppressingtgfb1smadmediatedepithelialtomesenchymaltransition