Cargando…

Cell sheets of co-cultured BMP-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro

Bone tissue engineering provides a substitute for bone transplantation to address various bone defects. However, bone regeneration involves a large number of cellular events. In addition, obtaining sufficient source material for autogenous bone or alloplastic bone substitutes remains an unsolved iss...

Descripción completa

Detalles Bibliográficos
Autores principales: He, Jia, Han, Xuesong, Wang, Songmei, Zhang, Ying, Dai, Xiaoming, Liu, Boyan, Liu, Liu, Zhao, Xian
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6777308/
https://www.ncbi.nlm.nih.gov/pubmed/31602206
http://dx.doi.org/10.3892/etm.2019.7982
_version_ 1783456604273770496
author He, Jia
Han, Xuesong
Wang, Songmei
Zhang, Ying
Dai, Xiaoming
Liu, Boyan
Liu, Liu
Zhao, Xian
author_facet He, Jia
Han, Xuesong
Wang, Songmei
Zhang, Ying
Dai, Xiaoming
Liu, Boyan
Liu, Liu
Zhao, Xian
author_sort He, Jia
collection PubMed
description Bone tissue engineering provides a substitute for bone transplantation to address various bone defects. However, bone regeneration involves a large number of cellular events. In addition, obtaining sufficient source material for autogenous bone or alloplastic bone substitutes remains an unsolved issue. In previous studies, it was confirmed that bone marrow stromal cells (BMSCs) and endothelial progenitor cells (EPCs) had the capacity to promote bone regeneration. Additionally, bone morphogenetic protein-2 (BMP-2) has been demonstrated to be an active inducer of osteoblast differentiation. Therefore, the aim of the present study was to produce an effective integration system, including a scaffold, reparative cells and growth factors, that may enhance bone regeneration. Firstly, bone marrow-derived BMSCs and EPCs were isolated and identified by flow cytometry. Cell proliferation ability, secreted BMP-2 levels and alkaline phosphatase (ALP) activity were highest in the cell sheets containing BMP-2-modified BMSCs and EPCs. In addition, the expression levels of osteogenesis-associated genes, including runt related transcription factor 2 (Runx2), distal-less homeobox 5 (Dlx5), ALP and integrin-binding sialoprotein (Ibsp), and osteogenesis-associated proteins, including Runx2, Dlx, ALP, Ibsp, vascular endothelial growth factor, osteonectin, osteopontin and type I collagen, gradually increased during the co-culture of ad-BMP-2-BMSCs/EPCs. The levels of these genes and proteins were increased compared with those observed in the BMSC, EPC and BMP-2-modified BMSC groups. Finally, scanning electron microscopy observation also demonstrated that the BMP2-modified BMSCs were able to combine well with EPCs to construct a cell sheet for bone formation. Collectively, these results describe an adenovirus (ad)-BMP2-BMSCs/EPCs co-culture system that may significantly accelerate bone regeneration compared with a BMSCs/EPCs co-culture system or ad-BMP2-BMSCs alone.
format Online
Article
Text
id pubmed-6777308
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-67773082019-10-10 Cell sheets of co-cultured BMP-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro He, Jia Han, Xuesong Wang, Songmei Zhang, Ying Dai, Xiaoming Liu, Boyan Liu, Liu Zhao, Xian Exp Ther Med Articles Bone tissue engineering provides a substitute for bone transplantation to address various bone defects. However, bone regeneration involves a large number of cellular events. In addition, obtaining sufficient source material for autogenous bone or alloplastic bone substitutes remains an unsolved issue. In previous studies, it was confirmed that bone marrow stromal cells (BMSCs) and endothelial progenitor cells (EPCs) had the capacity to promote bone regeneration. Additionally, bone morphogenetic protein-2 (BMP-2) has been demonstrated to be an active inducer of osteoblast differentiation. Therefore, the aim of the present study was to produce an effective integration system, including a scaffold, reparative cells and growth factors, that may enhance bone regeneration. Firstly, bone marrow-derived BMSCs and EPCs were isolated and identified by flow cytometry. Cell proliferation ability, secreted BMP-2 levels and alkaline phosphatase (ALP) activity were highest in the cell sheets containing BMP-2-modified BMSCs and EPCs. In addition, the expression levels of osteogenesis-associated genes, including runt related transcription factor 2 (Runx2), distal-less homeobox 5 (Dlx5), ALP and integrin-binding sialoprotein (Ibsp), and osteogenesis-associated proteins, including Runx2, Dlx, ALP, Ibsp, vascular endothelial growth factor, osteonectin, osteopontin and type I collagen, gradually increased during the co-culture of ad-BMP-2-BMSCs/EPCs. The levels of these genes and proteins were increased compared with those observed in the BMSC, EPC and BMP-2-modified BMSC groups. Finally, scanning electron microscopy observation also demonstrated that the BMP2-modified BMSCs were able to combine well with EPCs to construct a cell sheet for bone formation. Collectively, these results describe an adenovirus (ad)-BMP2-BMSCs/EPCs co-culture system that may significantly accelerate bone regeneration compared with a BMSCs/EPCs co-culture system or ad-BMP2-BMSCs alone. D.A. Spandidos 2019-11 2019-09-06 /pmc/articles/PMC6777308/ /pubmed/31602206 http://dx.doi.org/10.3892/etm.2019.7982 Text en Copyright: © He et al. This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
He, Jia
Han, Xuesong
Wang, Songmei
Zhang, Ying
Dai, Xiaoming
Liu, Boyan
Liu, Liu
Zhao, Xian
Cell sheets of co-cultured BMP-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro
title Cell sheets of co-cultured BMP-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro
title_full Cell sheets of co-cultured BMP-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro
title_fullStr Cell sheets of co-cultured BMP-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro
title_full_unstemmed Cell sheets of co-cultured BMP-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro
title_short Cell sheets of co-cultured BMP-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro
title_sort cell sheets of co-cultured bmp-2-modified bone marrow stromal cells and endothelial progenitor cells accelerate bone regeneration in vitro
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6777308/
https://www.ncbi.nlm.nih.gov/pubmed/31602206
http://dx.doi.org/10.3892/etm.2019.7982
work_keys_str_mv AT hejia cellsheetsofcoculturedbmp2modifiedbonemarrowstromalcellsandendothelialprogenitorcellsaccelerateboneregenerationinvitro
AT hanxuesong cellsheetsofcoculturedbmp2modifiedbonemarrowstromalcellsandendothelialprogenitorcellsaccelerateboneregenerationinvitro
AT wangsongmei cellsheetsofcoculturedbmp2modifiedbonemarrowstromalcellsandendothelialprogenitorcellsaccelerateboneregenerationinvitro
AT zhangying cellsheetsofcoculturedbmp2modifiedbonemarrowstromalcellsandendothelialprogenitorcellsaccelerateboneregenerationinvitro
AT daixiaoming cellsheetsofcoculturedbmp2modifiedbonemarrowstromalcellsandendothelialprogenitorcellsaccelerateboneregenerationinvitro
AT liuboyan cellsheetsofcoculturedbmp2modifiedbonemarrowstromalcellsandendothelialprogenitorcellsaccelerateboneregenerationinvitro
AT liuliu cellsheetsofcoculturedbmp2modifiedbonemarrowstromalcellsandendothelialprogenitorcellsaccelerateboneregenerationinvitro
AT zhaoxian cellsheetsofcoculturedbmp2modifiedbonemarrowstromalcellsandendothelialprogenitorcellsaccelerateboneregenerationinvitro