Cargando…

Insulin impedes osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence via the TGF-β1 pathway

The dysfunction of bone marrow stromal cells (BMSCs) may be a core factor in Type 2 diabetes mellitus (T2DM) associated osteoporosis. However, the underlying mechanism is not well understood. Here, we delineated the critical role of insulin impeding osteogenesis of BMSCs in T2DM. Compared with BMSCs...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Ping, Zhang, Hengguo, Lin, Jialin, Xiao, Tao, Xu, Rongyao, Fu, Yu, Zhang, Yuchao, Du, Yifei, Cheng, Jie, Jiang, Hongbing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Impact Journals 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7041775/
https://www.ncbi.nlm.nih.gov/pubmed/32017705
http://dx.doi.org/10.18632/aging.102723
_version_ 1783501207766040576
author Zhang, Ping
Zhang, Hengguo
Lin, Jialin
Xiao, Tao
Xu, Rongyao
Fu, Yu
Zhang, Yuchao
Du, Yifei
Cheng, Jie
Jiang, Hongbing
author_facet Zhang, Ping
Zhang, Hengguo
Lin, Jialin
Xiao, Tao
Xu, Rongyao
Fu, Yu
Zhang, Yuchao
Du, Yifei
Cheng, Jie
Jiang, Hongbing
author_sort Zhang, Ping
collection PubMed
description The dysfunction of bone marrow stromal cells (BMSCs) may be a core factor in Type 2 diabetes mellitus (T2DM) associated osteoporosis. However, the underlying mechanism is not well understood. Here, we delineated the critical role of insulin impeding osteogenesis of BMSCs in T2DM. Compared with BMSCs from healthy people (H-BMSCs), BMSCs from T2DM patient (DM-BMSCs) showed decreased osteogenic differentiation and autophagy level, and increased senescent phenotype. H-BMSCs incubated in hyperglycemic and hyperinsulinemic conditions similarly showed these phenotypes of DM-BMSCs. Notably, enhanced TGF-β1 expression was detected not only in DM-BMSCs and high-glucose and insulin-treated H-BMSCs, but also in bone callus of streptozocin-induced diabetic rats. Moreover, inhibiting TGF-β1 signaling not only enhanced osteogenic differentiation and autophagy level of DM-BMSCs, but also delayed senescence of DM-BMSCs, as well as promoted mandible defect healing of diabetic rats. Finally, we further verified that it was TGF-β receptor II (TβRII), not TβRI, markedly increased in both DM-BMSCs and insulin-treated H-BMSCs. Our data revealed that insulin impeded osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence, which it should be responsible for T2DM-induced bone loss, at least in part. These findings suggest that inhibiting TGF-β1 pathway may be a potential therapeutic target for T2DM associated bone disorders.
format Online
Article
Text
id pubmed-7041775
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Impact Journals
record_format MEDLINE/PubMed
spelling pubmed-70417752020-03-04 Insulin impedes osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence via the TGF-β1 pathway Zhang, Ping Zhang, Hengguo Lin, Jialin Xiao, Tao Xu, Rongyao Fu, Yu Zhang, Yuchao Du, Yifei Cheng, Jie Jiang, Hongbing Aging (Albany NY) Research Paper The dysfunction of bone marrow stromal cells (BMSCs) may be a core factor in Type 2 diabetes mellitus (T2DM) associated osteoporosis. However, the underlying mechanism is not well understood. Here, we delineated the critical role of insulin impeding osteogenesis of BMSCs in T2DM. Compared with BMSCs from healthy people (H-BMSCs), BMSCs from T2DM patient (DM-BMSCs) showed decreased osteogenic differentiation and autophagy level, and increased senescent phenotype. H-BMSCs incubated in hyperglycemic and hyperinsulinemic conditions similarly showed these phenotypes of DM-BMSCs. Notably, enhanced TGF-β1 expression was detected not only in DM-BMSCs and high-glucose and insulin-treated H-BMSCs, but also in bone callus of streptozocin-induced diabetic rats. Moreover, inhibiting TGF-β1 signaling not only enhanced osteogenic differentiation and autophagy level of DM-BMSCs, but also delayed senescence of DM-BMSCs, as well as promoted mandible defect healing of diabetic rats. Finally, we further verified that it was TGF-β receptor II (TβRII), not TβRI, markedly increased in both DM-BMSCs and insulin-treated H-BMSCs. Our data revealed that insulin impeded osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence, which it should be responsible for T2DM-induced bone loss, at least in part. These findings suggest that inhibiting TGF-β1 pathway may be a potential therapeutic target for T2DM associated bone disorders. Impact Journals 2020-02-03 /pmc/articles/PMC7041775/ /pubmed/32017705 http://dx.doi.org/10.18632/aging.102723 Text en Copyright © 2020 Zhang et al. http://creativecommons.org/licenses/by/3.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Paper
Zhang, Ping
Zhang, Hengguo
Lin, Jialin
Xiao, Tao
Xu, Rongyao
Fu, Yu
Zhang, Yuchao
Du, Yifei
Cheng, Jie
Jiang, Hongbing
Insulin impedes osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence via the TGF-β1 pathway
title Insulin impedes osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence via the TGF-β1 pathway
title_full Insulin impedes osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence via the TGF-β1 pathway
title_fullStr Insulin impedes osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence via the TGF-β1 pathway
title_full_unstemmed Insulin impedes osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence via the TGF-β1 pathway
title_short Insulin impedes osteogenesis of BMSCs by inhibiting autophagy and promoting premature senescence via the TGF-β1 pathway
title_sort insulin impedes osteogenesis of bmscs by inhibiting autophagy and promoting premature senescence via the tgf-β1 pathway
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7041775/
https://www.ncbi.nlm.nih.gov/pubmed/32017705
http://dx.doi.org/10.18632/aging.102723
work_keys_str_mv AT zhangping insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway
AT zhanghengguo insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway
AT linjialin insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway
AT xiaotao insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway
AT xurongyao insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway
AT fuyu insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway
AT zhangyuchao insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway
AT duyifei insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway
AT chengjie insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway
AT jianghongbing insulinimpedesosteogenesisofbmscsbyinhibitingautophagyandpromotingprematuresenescenceviathetgfb1pathway