Cargando…

Platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro

BACKGROUND: Adipose-derived mesenchymal stem cells (ADSC)-based therapy is an outstanding treatment strategy for ischaemic disease. However, the therapeutic efficacy of this strategy is not ideal due to the poor paracrine function and low survival rate of ADSCs in target regions. Platelet extracellu...

Descripción completa

Detalles Bibliográficos
Autores principales: Tang, Yanan, Li, Jiayan, Wang, Weiyi, Chen, Bingyi, Chen, Jinxing, Shen, Zekun, Hou, Jiaxuan, Mei, Yifan, Liu, Shuang, Zhang, Liwei, Li, Zongjin, Lu, Shaoying
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8427862/
https://www.ncbi.nlm.nih.gov/pubmed/34503551
http://dx.doi.org/10.1186/s13287-021-02561-w
_version_ 1783750260734033920
author Tang, Yanan
Li, Jiayan
Wang, Weiyi
Chen, Bingyi
Chen, Jinxing
Shen, Zekun
Hou, Jiaxuan
Mei, Yifan
Liu, Shuang
Zhang, Liwei
Li, Zongjin
Lu, Shaoying
author_facet Tang, Yanan
Li, Jiayan
Wang, Weiyi
Chen, Bingyi
Chen, Jinxing
Shen, Zekun
Hou, Jiaxuan
Mei, Yifan
Liu, Shuang
Zhang, Liwei
Li, Zongjin
Lu, Shaoying
author_sort Tang, Yanan
collection PubMed
description BACKGROUND: Adipose-derived mesenchymal stem cells (ADSC)-based therapy is an outstanding treatment strategy for ischaemic disease. However, the therapeutic efficacy of this strategy is not ideal due to the poor paracrine function and low survival rate of ADSCs in target regions. Platelet extracellular vesicles (PEVs) are nanoparticles derived from activated platelets that can participate in communication between cells. Accumulating evidence indicates that PEVs can regulate the biological functions of several cell lines. In the present study, we aimed to investigate whether PEVs can modulate the proangiogenic potential of ADSCs in vitro and in vivo. METHODS: PEVs were identified using scanning electron microscope (SEM), flow cytometry (FCM) and nanoparticle tracking analysis (NTA). The CCK8 assay was performed to detect proliferation of cells. Transwell and wound healing assays were performed to verify migration capacity of cells. AnnexinV-FITC/PI apoptosis kit and live/dead assay were performed to assess ADSCs apoptosis under Cocl(2)-induced hypoxia condition. The underlying mechanisms by which PEVs affected ADSCs were explored using real time-PCR(RT-PCR) and Western blot. In addition, matrigel plug assays were conducted and mouse hindlimb ischaemic models were established to investigate the proangiogenic potential of PEV-treated ADSCs in vivo. RESULTS: We demonstrated that ADSC could internalize PEVs, which lead to a series of biological reactions. In vitro, dose-dependent effects of PEVs on ADSC proliferation, migration and antiapoptotic capacity were observed. Western blotting results suggested that multiple proteins such as ERK, AKT, FAK, Src and PLCγ1 kinase may contribute to these changes. Furthermore, PEVs induced upregulation of several growth factors expression in ADSCs and amplified the proliferation, migration and tube formation of HUVECs induced by ADSC conditioned medium (CM). In in vivo experiments, compared with control ADSCs, the injection of PEV-treated ADSCs resulted in more vascularization in matrigel plugs, attenuated tissue degeneration and increased blood flow and capillary density in ischaemic hindlimb tissues. CONCLUSION: Our data demonstrated that PEVs could enhance the proangiogenic potential of ADSCs in mouse hindlimb ischaemia. The major mechanisms of this effect included the promotion of ADSC proliferation, migration, anti-apoptosis ability and paracrine secretion.
format Online
Article
Text
id pubmed-8427862
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-84278622021-09-10 Platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro Tang, Yanan Li, Jiayan Wang, Weiyi Chen, Bingyi Chen, Jinxing Shen, Zekun Hou, Jiaxuan Mei, Yifan Liu, Shuang Zhang, Liwei Li, Zongjin Lu, Shaoying Stem Cell Res Ther Research BACKGROUND: Adipose-derived mesenchymal stem cells (ADSC)-based therapy is an outstanding treatment strategy for ischaemic disease. However, the therapeutic efficacy of this strategy is not ideal due to the poor paracrine function and low survival rate of ADSCs in target regions. Platelet extracellular vesicles (PEVs) are nanoparticles derived from activated platelets that can participate in communication between cells. Accumulating evidence indicates that PEVs can regulate the biological functions of several cell lines. In the present study, we aimed to investigate whether PEVs can modulate the proangiogenic potential of ADSCs in vitro and in vivo. METHODS: PEVs were identified using scanning electron microscope (SEM), flow cytometry (FCM) and nanoparticle tracking analysis (NTA). The CCK8 assay was performed to detect proliferation of cells. Transwell and wound healing assays were performed to verify migration capacity of cells. AnnexinV-FITC/PI apoptosis kit and live/dead assay were performed to assess ADSCs apoptosis under Cocl(2)-induced hypoxia condition. The underlying mechanisms by which PEVs affected ADSCs were explored using real time-PCR(RT-PCR) and Western blot. In addition, matrigel plug assays were conducted and mouse hindlimb ischaemic models were established to investigate the proangiogenic potential of PEV-treated ADSCs in vivo. RESULTS: We demonstrated that ADSC could internalize PEVs, which lead to a series of biological reactions. In vitro, dose-dependent effects of PEVs on ADSC proliferation, migration and antiapoptotic capacity were observed. Western blotting results suggested that multiple proteins such as ERK, AKT, FAK, Src and PLCγ1 kinase may contribute to these changes. Furthermore, PEVs induced upregulation of several growth factors expression in ADSCs and amplified the proliferation, migration and tube formation of HUVECs induced by ADSC conditioned medium (CM). In in vivo experiments, compared with control ADSCs, the injection of PEV-treated ADSCs resulted in more vascularization in matrigel plugs, attenuated tissue degeneration and increased blood flow and capillary density in ischaemic hindlimb tissues. CONCLUSION: Our data demonstrated that PEVs could enhance the proangiogenic potential of ADSCs in mouse hindlimb ischaemia. The major mechanisms of this effect included the promotion of ADSC proliferation, migration, anti-apoptosis ability and paracrine secretion. BioMed Central 2021-09-09 /pmc/articles/PMC8427862/ /pubmed/34503551 http://dx.doi.org/10.1186/s13287-021-02561-w Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Tang, Yanan
Li, Jiayan
Wang, Weiyi
Chen, Bingyi
Chen, Jinxing
Shen, Zekun
Hou, Jiaxuan
Mei, Yifan
Liu, Shuang
Zhang, Liwei
Li, Zongjin
Lu, Shaoying
Platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro
title Platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro
title_full Platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro
title_fullStr Platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro
title_full_unstemmed Platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro
title_short Platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro
title_sort platelet extracellular vesicles enhance the proangiogenic potential of adipose-derived stem cells in vivo and in vitro
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8427862/
https://www.ncbi.nlm.nih.gov/pubmed/34503551
http://dx.doi.org/10.1186/s13287-021-02561-w
work_keys_str_mv AT tangyanan plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT lijiayan plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT wangweiyi plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT chenbingyi plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT chenjinxing plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT shenzekun plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT houjiaxuan plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT meiyifan plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT liushuang plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT zhangliwei plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT lizongjin plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro
AT lushaoying plateletextracellularvesiclesenhancetheproangiogenicpotentialofadiposederivedstemcellsinvivoandinvitro