Cargando…

Restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization

Macrophage-mediated inflammation compromises bone repair in diabetic patients. Electrical signaling cues are known to regulate macrophage functions. However, the biological effects of electrical microenvironment from charged biomaterials on the immune response for regulating osteogenesis under diabe...

Descripción completa

Detalles Bibliográficos
Autores principales: Dai, Xiaohan, Heng, Boon Chin, Bai, Yunyang, You, Fuping, Sun, Xiaowen, Li, Yiping, Tang, Zhangui, Xu, Mingming, Zhang, Xuehui, Deng, Xuliang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: KeAi Publishing 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7787955/
https://www.ncbi.nlm.nih.gov/pubmed/33474514
http://dx.doi.org/10.1016/j.bioactmat.2020.12.020
_version_ 1783632935034814464
author Dai, Xiaohan
Heng, Boon Chin
Bai, Yunyang
You, Fuping
Sun, Xiaowen
Li, Yiping
Tang, Zhangui
Xu, Mingming
Zhang, Xuehui
Deng, Xuliang
author_facet Dai, Xiaohan
Heng, Boon Chin
Bai, Yunyang
You, Fuping
Sun, Xiaowen
Li, Yiping
Tang, Zhangui
Xu, Mingming
Zhang, Xuehui
Deng, Xuliang
author_sort Dai, Xiaohan
collection PubMed
description Macrophage-mediated inflammation compromises bone repair in diabetic patients. Electrical signaling cues are known to regulate macrophage functions. However, the biological effects of electrical microenvironment from charged biomaterials on the immune response for regulating osteogenesis under diabetic conditions remain to be elucidated. Herein the endogeneous electrical microenvironment of native bone tissue was recapitulated by fabricating a ferroelectric BaTiO(3)/poly (vinylidene fluoridetrifluoroethylene) (BTO/P(VDF-TrFE)) nanocomposite membrane. In vitro, the polarized BaTiO(3)/poly (vinylidene fluoridetrifluoroethylene) (BTO/P(VDF-TrFE)) nanocomposite membranes inhibited high glucose-induced M1-type inflammation, by effecting changes in cell morphology, M1 marker expression and pro-inflammatory cytokine secretion in macrophages. This led to enhanced osteogenic differentiation of human bone marrow mesenchymal stem cells (BM-MSCs). In vivo, the biomimetic electrical microenvironment recapitulated by the polarized nanocomposite membranes switched macrophage phenotype from the pro-inflammatory (M1) into the pro-healing (M2) phenotype, which in turn enhanced bone regeneration in rats with type 2 diabetes mellitus. Mechanistic studies revealed that the biomimetic electrical microenvironment attenuated pro-inflammatory M1 macrophage polarization under hyperglycemic conditions by suppressing expression of AKT2 and IRF5 within the PI3K-AKT signaling pathway, thereby inducing favorable osteo-immunomodulatory effects. Our study thus provides fundamental insights into the biological effects of restoring the electrical microenvironment conducive for osteogenesis under DM conditions, and offers an effective strategy to design functionalized biomaterials for bone regeneration therapy in diabetic patients.
format Online
Article
Text
id pubmed-7787955
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher KeAi Publishing
record_format MEDLINE/PubMed
spelling pubmed-77879552021-01-19 Restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization Dai, Xiaohan Heng, Boon Chin Bai, Yunyang You, Fuping Sun, Xiaowen Li, Yiping Tang, Zhangui Xu, Mingming Zhang, Xuehui Deng, Xuliang Bioact Mater Article Macrophage-mediated inflammation compromises bone repair in diabetic patients. Electrical signaling cues are known to regulate macrophage functions. However, the biological effects of electrical microenvironment from charged biomaterials on the immune response for regulating osteogenesis under diabetic conditions remain to be elucidated. Herein the endogeneous electrical microenvironment of native bone tissue was recapitulated by fabricating a ferroelectric BaTiO(3)/poly (vinylidene fluoridetrifluoroethylene) (BTO/P(VDF-TrFE)) nanocomposite membrane. In vitro, the polarized BaTiO(3)/poly (vinylidene fluoridetrifluoroethylene) (BTO/P(VDF-TrFE)) nanocomposite membranes inhibited high glucose-induced M1-type inflammation, by effecting changes in cell morphology, M1 marker expression and pro-inflammatory cytokine secretion in macrophages. This led to enhanced osteogenic differentiation of human bone marrow mesenchymal stem cells (BM-MSCs). In vivo, the biomimetic electrical microenvironment recapitulated by the polarized nanocomposite membranes switched macrophage phenotype from the pro-inflammatory (M1) into the pro-healing (M2) phenotype, which in turn enhanced bone regeneration in rats with type 2 diabetes mellitus. Mechanistic studies revealed that the biomimetic electrical microenvironment attenuated pro-inflammatory M1 macrophage polarization under hyperglycemic conditions by suppressing expression of AKT2 and IRF5 within the PI3K-AKT signaling pathway, thereby inducing favorable osteo-immunomodulatory effects. Our study thus provides fundamental insights into the biological effects of restoring the electrical microenvironment conducive for osteogenesis under DM conditions, and offers an effective strategy to design functionalized biomaterials for bone regeneration therapy in diabetic patients. KeAi Publishing 2020-12-31 /pmc/articles/PMC7787955/ /pubmed/33474514 http://dx.doi.org/10.1016/j.bioactmat.2020.12.020 Text en © 2020 The Authors. Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd. http://creativecommons.org/licenses/by-nc-nd/4.0/ This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Article
Dai, Xiaohan
Heng, Boon Chin
Bai, Yunyang
You, Fuping
Sun, Xiaowen
Li, Yiping
Tang, Zhangui
Xu, Mingming
Zhang, Xuehui
Deng, Xuliang
Restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization
title Restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization
title_full Restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization
title_fullStr Restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization
title_full_unstemmed Restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization
title_short Restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization
title_sort restoration of electrical microenvironment enhances bone regeneration under diabetic conditions by modulating macrophage polarization
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7787955/
https://www.ncbi.nlm.nih.gov/pubmed/33474514
http://dx.doi.org/10.1016/j.bioactmat.2020.12.020
work_keys_str_mv AT daixiaohan restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization
AT hengboonchin restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization
AT baiyunyang restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization
AT youfuping restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization
AT sunxiaowen restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization
AT liyiping restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization
AT tangzhangui restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization
AT xumingming restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization
AT zhangxuehui restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization
AT dengxuliang restorationofelectricalmicroenvironmentenhancesboneregenerationunderdiabeticconditionsbymodulatingmacrophagepolarization