Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway

Abstract Background Increasing evidence has shown that mesenchymal stem cells (MSCs) yield a favorable therapeutic benefit for thermal burn skin wounds. Human amniotic MSCs (hAMSCs) derived from amniotic membrane have multilineage differentiation, immunosuppressive, and anti-inflammatory potential w...

Full description

Bibliographic Details
Main Authors: Jing-Yuan Li, Kang-Kang Ren, Wen-Jie Zhang, Ling Xiao, Han-You Wu, Qian-Yu Liu, Ting Ding, Xiang-Cheng Zhang, Wen-Jia Nie, Yu Ke, Ke-Yu Deng, Quan-Wen Liu, Hong-Bo Xin
Format: Article
Language:English
Published: BMC 2019-08-01
Series:Stem Cell Research & Therapy
Subjects:
Online Access:http://link.springer.com/article/10.1186/s13287-019-1366-y
id doaj-87b54e7beab24e0196fd51c417ff39b0
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Jing-Yuan Li
Kang-Kang Ren
Wen-Jie Zhang
Ling Xiao
Han-You Wu
Qian-Yu Liu
Ting Ding
Xiang-Cheng Zhang
Wen-Jia Nie
Yu Ke
Ke-Yu Deng
Quan-Wen Liu
Hong-Bo Xin
spellingShingle Jing-Yuan Li
Kang-Kang Ren
Wen-Jie Zhang
Ling Xiao
Han-You Wu
Qian-Yu Liu
Ting Ding
Xiang-Cheng Zhang
Wen-Jia Nie
Yu Ke
Ke-Yu Deng
Quan-Wen Liu
Hong-Bo Xin
Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway
Stem Cell Research & Therapy
Human amniotic membrane mesenchymal stem cells
Conditioned medium
Wound healing
PI3K/AKT signaling
Antibody array
author_facet Jing-Yuan Li
Kang-Kang Ren
Wen-Jie Zhang
Ling Xiao
Han-You Wu
Qian-Yu Liu
Ting Ding
Xiang-Cheng Zhang
Wen-Jia Nie
Yu Ke
Ke-Yu Deng
Quan-Wen Liu
Hong-Bo Xin
author_sort Jing-Yuan Li
title Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway
title_short Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway
title_full Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway
title_fullStr Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway
title_full_unstemmed Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway
title_sort human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating pi3k/akt signaling pathway
publisher BMC
series Stem Cell Research & Therapy
issn 1757-6512
publishDate 2019-08-01
description Abstract Background Increasing evidence has shown that mesenchymal stem cells (MSCs) yield a favorable therapeutic benefit for thermal burn skin wounds. Human amniotic MSCs (hAMSCs) derived from amniotic membrane have multilineage differentiation, immunosuppressive, and anti-inflammatory potential which makes them suitable for treating skin wounds. However, the exact effects of hAMSCs on the healing of thermal burn skin wounds and their potential mechanisms are not explored. Methods hAMSCs were isolated from amniotic membrane and characterized by RT-PCR, flow cytometry, immunofluorescence, and tumorigenicity test. We assessed the effects of hAMSCs and hAMSC conditional medium (CM) on wound healing in a deep second-degree burn injury model of mice. We then investigated the biological effects of hAMSCs and hAMSC-CM on the apoptosis and proliferation of heat stress-injured human keratinocytes HaCAT and dermal fibroblasts (DFL) both in vivo and in vitro. Next, we explored the underlying mechanisms by assessing PI3K/AKT and GSK3β/β-catenin signaling pathways in heat injured HaCAT and DFL cells after hAMSCs and hAMSC-CM treatments using PI3K inhibitor LY294002 and β-catenin inhibitor ICG001. Antibody array assay was used to identify the cytokines secreted by hAMSCs that may activate PI3K/AKT signaling pathway. Results Our results showed that hAMSCs expressed various markers of embryonic stem cells and mesenchymal stem cells and have low immunogenicity and no tumorigenicity. hAMSC and hAMSC-CM transplantation significantly promoted thermal burn wound healing by accelerating re-epithelialization with increased expression of CK19 and PCNA in vivo. hAMSCs and hAMSC-CM markedly inhibited heat stress-induced apoptosis in HaCAT and DFL cells in vitro through activation of PI3K/AKT signaling and promoted their proliferation by activating GSK3β/β-catenin signaling. Furthermore, we demonstrated that hAMSC-mediated activation of GSK3β/β-catenin signaling was dependent on PI3K/AKT signaling pathway. Antibody array assay showed that a panel of cytokines including PAI-1, C-GSF, periostin, and TIMP-1 delivered from hAMSCs may contribute to the improvement of the wound healing through activating PI3K/AKT signaling pathway. Conclusion Our results demonstrated that hAMSCs and hAMSC-CM efficiently cure heat stress-induced skin injury by inhibiting apoptosis of skin cells and promoting their proliferation through activating PI3K/AKT signaling pathway, suggesting that hAMSCs and hAMSC-CM may provide an alternative therapeutic approach for the treatment of skin injury.
topic Human amniotic membrane mesenchymal stem cells
Conditioned medium
Wound healing
PI3K/AKT signaling
Antibody array
url http://link.springer.com/article/10.1186/s13287-019-1366-y
work_keys_str_mv AT jingyuanli humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT kangkangren humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT wenjiezhang humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT lingxiao humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT hanyouwu humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT qianyuliu humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT tingding humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT xiangchengzhang humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT wenjianie humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT yuke humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT keyudeng humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT quanwenliu humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
AT hongboxin humanamnioticmesenchymalstemcellsandtheirparacrinefactorspromotewoundhealingbyinhibitingheatstressinducedskincellapoptosisandenhancingtheirproliferationthroughactivatingpi3kaktsignalingpathway
_version_ 1724570920204369920
spelling doaj-87b54e7beab24e0196fd51c417ff39b02020-11-25T03:31:54ZengBMCStem Cell Research & Therapy1757-65122019-08-0110111710.1186/s13287-019-1366-yHuman amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathwayJing-Yuan Li0Kang-Kang Ren1Wen-Jie Zhang2Ling Xiao3Han-You Wu4Qian-Yu Liu5Ting Ding6Xiang-Cheng Zhang7Wen-Jia Nie8Yu Ke9Ke-Yu Deng10Quan-Wen Liu11Hong-Bo Xin12The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityDepartment of Obstetrics and Gynecology, The First Affiliated Hospital of Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityThe National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang UniversityAbstract Background Increasing evidence has shown that mesenchymal stem cells (MSCs) yield a favorable therapeutic benefit for thermal burn skin wounds. Human amniotic MSCs (hAMSCs) derived from amniotic membrane have multilineage differentiation, immunosuppressive, and anti-inflammatory potential which makes them suitable for treating skin wounds. However, the exact effects of hAMSCs on the healing of thermal burn skin wounds and their potential mechanisms are not explored. Methods hAMSCs were isolated from amniotic membrane and characterized by RT-PCR, flow cytometry, immunofluorescence, and tumorigenicity test. We assessed the effects of hAMSCs and hAMSC conditional medium (CM) on wound healing in a deep second-degree burn injury model of mice. We then investigated the biological effects of hAMSCs and hAMSC-CM on the apoptosis and proliferation of heat stress-injured human keratinocytes HaCAT and dermal fibroblasts (DFL) both in vivo and in vitro. Next, we explored the underlying mechanisms by assessing PI3K/AKT and GSK3β/β-catenin signaling pathways in heat injured HaCAT and DFL cells after hAMSCs and hAMSC-CM treatments using PI3K inhibitor LY294002 and β-catenin inhibitor ICG001. Antibody array assay was used to identify the cytokines secreted by hAMSCs that may activate PI3K/AKT signaling pathway. Results Our results showed that hAMSCs expressed various markers of embryonic stem cells and mesenchymal stem cells and have low immunogenicity and no tumorigenicity. hAMSC and hAMSC-CM transplantation significantly promoted thermal burn wound healing by accelerating re-epithelialization with increased expression of CK19 and PCNA in vivo. hAMSCs and hAMSC-CM markedly inhibited heat stress-induced apoptosis in HaCAT and DFL cells in vitro through activation of PI3K/AKT signaling and promoted their proliferation by activating GSK3β/β-catenin signaling. Furthermore, we demonstrated that hAMSC-mediated activation of GSK3β/β-catenin signaling was dependent on PI3K/AKT signaling pathway. Antibody array assay showed that a panel of cytokines including PAI-1, C-GSF, periostin, and TIMP-1 delivered from hAMSCs may contribute to the improvement of the wound healing through activating PI3K/AKT signaling pathway. Conclusion Our results demonstrated that hAMSCs and hAMSC-CM efficiently cure heat stress-induced skin injury by inhibiting apoptosis of skin cells and promoting their proliferation through activating PI3K/AKT signaling pathway, suggesting that hAMSCs and hAMSC-CM may provide an alternative therapeutic approach for the treatment of skin injury.http://link.springer.com/article/10.1186/s13287-019-1366-yHuman amniotic membrane mesenchymal stem cellsConditioned mediumWound healingPI3K/AKT signalingAntibody array