Cargando…

Pretreatment of cardiac progenitor cells with bradykinin attenuates H(2)O(2)-induced cell apoptosis and improves cardiac function in rats by regulating autophagy

BACKGROUND: Previous studies have demonstrated that human cardiac c-Kit(+) progenitor cells (hCPCs) can effectively improve ischemic heart disease. However, the major challenge in applying hCPCs to clinical therapy is the low survival rate of graft hCPCs in the host heart, which limited the benefit...

Descripción completa

Detalles Bibliográficos
Autores principales: Wu, Chan, Zhou, Xiao-Xia, Li, Jing-Zhou, Qiang, Hai-Feng, Wang, Yan, Li, Gang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8340370/
https://www.ncbi.nlm.nih.gov/pubmed/34353364
http://dx.doi.org/10.1186/s13287-021-02503-6
_version_ 1783733757022306304
author Wu, Chan
Zhou, Xiao-Xia
Li, Jing-Zhou
Qiang, Hai-Feng
Wang, Yan
Li, Gang
author_facet Wu, Chan
Zhou, Xiao-Xia
Li, Jing-Zhou
Qiang, Hai-Feng
Wang, Yan
Li, Gang
author_sort Wu, Chan
collection PubMed
description BACKGROUND: Previous studies have demonstrated that human cardiac c-Kit(+) progenitor cells (hCPCs) can effectively improve ischemic heart disease. However, the major challenge in applying hCPCs to clinical therapy is the low survival rate of graft hCPCs in the host heart, which limited the benefit of transplanted hCPCs. Bradykinin (BK) is a principal active agent of the tissue kinin-kallikrein system. Our previous studies have highlighted that BK mediated the growth and migration of CPCs by regulating Ca(2+) influx. However, the protective effect of BK on CPCs, improvement in the survival rate of BK-pretreated hCPCs in the infarcted heart, and the related mechanism remain elusive. METHODS: HCPCs were treated with H(2)O(2) to induce cell apoptosis and autophagy, and different concentration of BK was applied to rescue the H(2)O(2)-induced injury detected by MTT assay, TUNEL staining, flow cytometry, western blotting, and mitoSOX assays. The role of autophagy in the anti-apoptotic effect of BK was chemically activated or inhibited using the autophagy inducer, rapamycin, or the inhibitor, 3-methyladenine (3-MA). To explore the protective effect of BK on hCPCs, 3-MA or BK-pretreated hCPCs were transplanted into the myocardial infarcted rats. An echocardiogram was used to determine cardiac function, H&E and Masson staining were employed to assess pathological characteristics, HLA gene expression was quantified by qRT-PCR, and immunostaining was applied to examine neovascularization using confocal microscopy. RESULTS: The in vitro results showed that BK suppressed H(2)O(2)-induced hCPCs apoptosis and ROS production in a concentration-dependent manner by promoting pAkt and Bcl-2 expression and reducing cleaved caspase 3 and Bax expression. Moreover, BK restrained the H(2)O(2)-induced cell autophagy by decreasing LC3II/I, Beclin1, and ATG5 expression and increasing P62 expression. In the in vivo experiment, the transplanted BK- or 3-MA-treated hCPCs were found to be more effectively improved cardiac function by decreasing cardiomyocyte apoptosis, inflammatory infiltration, and myocardial fibrosis, and promoting neovascularization in the infarcted heart, compared to untreated-hCPCs or c-kit(-) cardiomyocytes (CPC(-) cells). CONCLUSIONS: Our present study established a new method to rescue transplanted hCPCs in the infarcted cardiac area via regulating cell apoptosis and autophagy of hCPCs by pretreatment with BK, providing a new therapeutic option for heart failure. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13287-021-02503-6.
format Online
Article
Text
id pubmed-8340370
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-83403702021-08-06 Pretreatment of cardiac progenitor cells with bradykinin attenuates H(2)O(2)-induced cell apoptosis and improves cardiac function in rats by regulating autophagy Wu, Chan Zhou, Xiao-Xia Li, Jing-Zhou Qiang, Hai-Feng Wang, Yan Li, Gang Stem Cell Res Ther Research BACKGROUND: Previous studies have demonstrated that human cardiac c-Kit(+) progenitor cells (hCPCs) can effectively improve ischemic heart disease. However, the major challenge in applying hCPCs to clinical therapy is the low survival rate of graft hCPCs in the host heart, which limited the benefit of transplanted hCPCs. Bradykinin (BK) is a principal active agent of the tissue kinin-kallikrein system. Our previous studies have highlighted that BK mediated the growth and migration of CPCs by regulating Ca(2+) influx. However, the protective effect of BK on CPCs, improvement in the survival rate of BK-pretreated hCPCs in the infarcted heart, and the related mechanism remain elusive. METHODS: HCPCs were treated with H(2)O(2) to induce cell apoptosis and autophagy, and different concentration of BK was applied to rescue the H(2)O(2)-induced injury detected by MTT assay, TUNEL staining, flow cytometry, western blotting, and mitoSOX assays. The role of autophagy in the anti-apoptotic effect of BK was chemically activated or inhibited using the autophagy inducer, rapamycin, or the inhibitor, 3-methyladenine (3-MA). To explore the protective effect of BK on hCPCs, 3-MA or BK-pretreated hCPCs were transplanted into the myocardial infarcted rats. An echocardiogram was used to determine cardiac function, H&E and Masson staining were employed to assess pathological characteristics, HLA gene expression was quantified by qRT-PCR, and immunostaining was applied to examine neovascularization using confocal microscopy. RESULTS: The in vitro results showed that BK suppressed H(2)O(2)-induced hCPCs apoptosis and ROS production in a concentration-dependent manner by promoting pAkt and Bcl-2 expression and reducing cleaved caspase 3 and Bax expression. Moreover, BK restrained the H(2)O(2)-induced cell autophagy by decreasing LC3II/I, Beclin1, and ATG5 expression and increasing P62 expression. In the in vivo experiment, the transplanted BK- or 3-MA-treated hCPCs were found to be more effectively improved cardiac function by decreasing cardiomyocyte apoptosis, inflammatory infiltration, and myocardial fibrosis, and promoting neovascularization in the infarcted heart, compared to untreated-hCPCs or c-kit(-) cardiomyocytes (CPC(-) cells). CONCLUSIONS: Our present study established a new method to rescue transplanted hCPCs in the infarcted cardiac area via regulating cell apoptosis and autophagy of hCPCs by pretreatment with BK, providing a new therapeutic option for heart failure. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13287-021-02503-6. BioMed Central 2021-08-05 /pmc/articles/PMC8340370/ /pubmed/34353364 http://dx.doi.org/10.1186/s13287-021-02503-6 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
Wu, Chan
Zhou, Xiao-Xia
Li, Jing-Zhou
Qiang, Hai-Feng
Wang, Yan
Li, Gang
Pretreatment of cardiac progenitor cells with bradykinin attenuates H(2)O(2)-induced cell apoptosis and improves cardiac function in rats by regulating autophagy
title Pretreatment of cardiac progenitor cells with bradykinin attenuates H(2)O(2)-induced cell apoptosis and improves cardiac function in rats by regulating autophagy
title_full Pretreatment of cardiac progenitor cells with bradykinin attenuates H(2)O(2)-induced cell apoptosis and improves cardiac function in rats by regulating autophagy
title_fullStr Pretreatment of cardiac progenitor cells with bradykinin attenuates H(2)O(2)-induced cell apoptosis and improves cardiac function in rats by regulating autophagy
title_full_unstemmed Pretreatment of cardiac progenitor cells with bradykinin attenuates H(2)O(2)-induced cell apoptosis and improves cardiac function in rats by regulating autophagy
title_short Pretreatment of cardiac progenitor cells with bradykinin attenuates H(2)O(2)-induced cell apoptosis and improves cardiac function in rats by regulating autophagy
title_sort pretreatment of cardiac progenitor cells with bradykinin attenuates h(2)o(2)-induced cell apoptosis and improves cardiac function in rats by regulating autophagy
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8340370/
https://www.ncbi.nlm.nih.gov/pubmed/34353364
http://dx.doi.org/10.1186/s13287-021-02503-6
work_keys_str_mv AT wuchan pretreatmentofcardiacprogenitorcellswithbradykininattenuatesh2o2inducedcellapoptosisandimprovescardiacfunctioninratsbyregulatingautophagy
AT zhouxiaoxia pretreatmentofcardiacprogenitorcellswithbradykininattenuatesh2o2inducedcellapoptosisandimprovescardiacfunctioninratsbyregulatingautophagy
AT lijingzhou pretreatmentofcardiacprogenitorcellswithbradykininattenuatesh2o2inducedcellapoptosisandimprovescardiacfunctioninratsbyregulatingautophagy
AT qianghaifeng pretreatmentofcardiacprogenitorcellswithbradykininattenuatesh2o2inducedcellapoptosisandimprovescardiacfunctioninratsbyregulatingautophagy
AT wangyan pretreatmentofcardiacprogenitorcellswithbradykininattenuatesh2o2inducedcellapoptosisandimprovescardiacfunctioninratsbyregulatingautophagy
AT ligang pretreatmentofcardiacprogenitorcellswithbradykininattenuatesh2o2inducedcellapoptosisandimprovescardiacfunctioninratsbyregulatingautophagy