Hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2

Abstract Background Delivery of endothelial cells into the ischemic tissue is emerging as an alternative approach in revascularization of injured tissues by means of angiogenesis to restore organ function. Adipose-derived stem cells (ASCs) are a readily accessible source of the mesenchymal stem cell...

Full description

Bibliographic Details
Main Authors: Ting Shang, Shuaijun Li, Yun Zhang, Laiya Lu, Lei Cui, Fang Fang Guo
Format: Article
Language:English
Published: BMC 2019-05-01
Series:Stem Cell Research & Therapy
Subjects:
Online Access:http://link.springer.com/article/10.1186/s13287-019-1233-x
id doaj-23ea664ae5f549ce9f2c5eeb70a1a596
record_format Article
spelling doaj-23ea664ae5f549ce9f2c5eeb70a1a5962020-11-25T03:14:57ZengBMCStem Cell Research & Therapy1757-65122019-05-0110111210.1186/s13287-019-1233-xHypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2Ting Shang0Shuaijun Li1Yun Zhang2Laiya Lu3Lei Cui4Fang Fang Guo5Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical UniversityDepartment of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical UniversityDepartment of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical UniversityDepartment of Orthopedics, Shanghai Tongji Hospital, Tongji University School of MedicineDepartment of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical UniversityDepartment of Plastic and Reconstructive surgery, Zhongda Hospital, Southeast UniversityAbstract Background Delivery of endothelial cells into the ischemic tissue is emerging as an alternative approach in revascularization of injured tissues by means of angiogenesis to restore organ function. Adipose-derived stem cells (ASCs) are a readily accessible source of the mesenchymal stem cell with rapid expansion and multidifferentiation potential. The view has emerged that endothelial cells (ECs) differentiated from ASCs is a step forward for adult vascular repair in regenerative medicine and construction of the blood vessel by tissue engineering approach. Methods In this study, differentiation of human ASCs (hASCs) into vascular EC lineage was induced by combined treatment of vascular endothelial growth factor (VEGF) and bone morphogenetic protein-4 (BMP4) under hypoxia condition. The expression of CD31, VEGF-R2, and VE-cadherin was determined by immunofluorescent staining, real-time PCR, and western blot analysis. These differentiated cells acquired functional characteristics of mature ECs as determined by their tube formation ability, DiI-ac-LDL uptake, and nitric oxide secretion in vitro. The methylation status in the proximal promoter CpGs was determined by bisulfite sequencing. Results hASCs expressed endothelial cell markers including CD31, VEGF-R2, and VE-cadherin by combined treatment of VEGF and BMP4 under hypoxia condition. These differentiated cells exhibited the angiogenesis potential in vitro, and injection of these differentiated cells enhanced angiogenesis in the ischemic hindlimb of diabetic mice. Furthermore, it was found that hypoxia increased significantly EphrinB2 expression EC differentiation, which is greatly downregulated with EphrinB2 blockage. The methylation status in the proximal promoter CpG results showed that methylation of EphrinB2 promoter decreased in hASCs with exposure to hypoxia. Conclusion Our data demonstrate that hASCs can be efficiently induced to differentiate into vascular EC lineages which are mediated by demethylation of ephrinB2 under hypoxia condition.http://link.springer.com/article/10.1186/s13287-019-1233-xHypoxiaAdipose-derived stem cellsVascular endothelial cellDemethylationephrinB2
collection DOAJ
language English
format Article
sources DOAJ
author Ting Shang
Shuaijun Li
Yun Zhang
Laiya Lu
Lei Cui
Fang Fang Guo
spellingShingle Ting Shang
Shuaijun Li
Yun Zhang
Laiya Lu
Lei Cui
Fang Fang Guo
Hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2
Stem Cell Research & Therapy
Hypoxia
Adipose-derived stem cells
Vascular endothelial cell
Demethylation
ephrinB2
author_facet Ting Shang
Shuaijun Li
Yun Zhang
Laiya Lu
Lei Cui
Fang Fang Guo
author_sort Ting Shang
title Hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2
title_short Hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2
title_full Hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2
title_fullStr Hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2
title_full_unstemmed Hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2
title_sort hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinb2
publisher BMC
series Stem Cell Research & Therapy
issn 1757-6512
publishDate 2019-05-01
description Abstract Background Delivery of endothelial cells into the ischemic tissue is emerging as an alternative approach in revascularization of injured tissues by means of angiogenesis to restore organ function. Adipose-derived stem cells (ASCs) are a readily accessible source of the mesenchymal stem cell with rapid expansion and multidifferentiation potential. The view has emerged that endothelial cells (ECs) differentiated from ASCs is a step forward for adult vascular repair in regenerative medicine and construction of the blood vessel by tissue engineering approach. Methods In this study, differentiation of human ASCs (hASCs) into vascular EC lineage was induced by combined treatment of vascular endothelial growth factor (VEGF) and bone morphogenetic protein-4 (BMP4) under hypoxia condition. The expression of CD31, VEGF-R2, and VE-cadherin was determined by immunofluorescent staining, real-time PCR, and western blot analysis. These differentiated cells acquired functional characteristics of mature ECs as determined by their tube formation ability, DiI-ac-LDL uptake, and nitric oxide secretion in vitro. The methylation status in the proximal promoter CpGs was determined by bisulfite sequencing. Results hASCs expressed endothelial cell markers including CD31, VEGF-R2, and VE-cadherin by combined treatment of VEGF and BMP4 under hypoxia condition. These differentiated cells exhibited the angiogenesis potential in vitro, and injection of these differentiated cells enhanced angiogenesis in the ischemic hindlimb of diabetic mice. Furthermore, it was found that hypoxia increased significantly EphrinB2 expression EC differentiation, which is greatly downregulated with EphrinB2 blockage. The methylation status in the proximal promoter CpG results showed that methylation of EphrinB2 promoter decreased in hASCs with exposure to hypoxia. Conclusion Our data demonstrate that hASCs can be efficiently induced to differentiate into vascular EC lineages which are mediated by demethylation of ephrinB2 under hypoxia condition.
topic Hypoxia
Adipose-derived stem cells
Vascular endothelial cell
Demethylation
ephrinB2
url http://link.springer.com/article/10.1186/s13287-019-1233-x
work_keys_str_mv AT tingshang hypoxiapromotesdifferentiationofadiposederivedstemcellsintoendothelialcellsthroughdemethylationofephrinb2
AT shuaijunli hypoxiapromotesdifferentiationofadiposederivedstemcellsintoendothelialcellsthroughdemethylationofephrinb2
AT yunzhang hypoxiapromotesdifferentiationofadiposederivedstemcellsintoendothelialcellsthroughdemethylationofephrinb2
AT laiyalu hypoxiapromotesdifferentiationofadiposederivedstemcellsintoendothelialcellsthroughdemethylationofephrinb2
AT leicui hypoxiapromotesdifferentiationofadiposederivedstemcellsintoendothelialcellsthroughdemethylationofephrinb2
AT fangfangguo hypoxiapromotesdifferentiationofadiposederivedstemcellsintoendothelialcellsthroughdemethylationofephrinb2
_version_ 1724641410921005056