Cargando…

PINK1 contained in huMSC-derived exosomes prevents cardiomyocyte mitochondrial calcium overload in sepsis via recovery of mitochondrial Ca(2+) efflux

BACKGROUND: Sepsis is a systemic inflammatory response to a local severe infection that may lead to multiple organ failure and death. Previous studies have shown that 40–50% of patients with sepsis have diverse myocardial injuries and 70 to 90% mortality rates compared to 20% mortality in patients w...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhou, Qin, Xie, Min, Zhu, Jing, Yi, Qin, Tan, Bin, Li, Yasha, Ye, Liang, Zhang, Xinyuan, Zhang, Ying, Tian, Jie, Xu, Hao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8101124/
https://www.ncbi.nlm.nih.gov/pubmed/33957982
http://dx.doi.org/10.1186/s13287-021-02325-6
_version_ 1783688908196806656
author Zhou, Qin
Xie, Min
Zhu, Jing
Yi, Qin
Tan, Bin
Li, Yasha
Ye, Liang
Zhang, Xinyuan
Zhang, Ying
Tian, Jie
Xu, Hao
author_facet Zhou, Qin
Xie, Min
Zhu, Jing
Yi, Qin
Tan, Bin
Li, Yasha
Ye, Liang
Zhang, Xinyuan
Zhang, Ying
Tian, Jie
Xu, Hao
author_sort Zhou, Qin
collection PubMed
description BACKGROUND: Sepsis is a systemic inflammatory response to a local severe infection that may lead to multiple organ failure and death. Previous studies have shown that 40–50% of patients with sepsis have diverse myocardial injuries and 70 to 90% mortality rates compared to 20% mortality in patients with sepsis without myocardial injury. Therefore, uncovering the mechanism of sepsis-induced myocardial injury and finding a target-based treatment are immensely important. OBJECTIVE: The present study elucidated the mechanism of sepsis-induced myocardial injury and examined the value of human umbilical cord mesenchymal stem cells (huMSCs) for protecting cardiac function in sepsis. METHODS: We used cecal ligation and puncture (CLP) to induce sepsis in mice and detect myocardial injury and cardiac function using serological markers and echocardiography. Cardiomyocyte apoptosis and heart tissue ultrastructure were detected using TdT-mediated dUTP Nick-End Labeling (TUNEL) and transmission electron microscopy (TEM), respectively. Fura-2 AM was used to monitor Ca(2+) uptake and efflux in mitochondria. FQ-PCR and Western blotting detected expression of mitochondrial Ca(2+) distribution regulators and PTEN-induced putative kinase 1 (PINK1). JC-1 was used to detect the mitochondrial membrane potential (Δψm) of cardiomyocytes. RESULTS: We found that expression of PINK1 decreased in mouse hearts during sepsis, which caused cardiomyocyte mitochondrial Ca(2+) efflux disorder, mitochondrial calcium overload, and cardiomyocyte injury. In contrast, we found that exosomes isolated from huMSCs (huMSC-exo) carried Pink1 mRNA, which could be transferred to recipient cardiomyocytes to increase PINK1 expression. The reduction in cardiomyocyte mitochondrial calcium efflux was reversed, and cardiomyocytes recovered from injury. We confirmed the effect of the PINK1-PKA-NCLX axis on mitochondrial calcium homeostasis in cardiomyocytes during sepsis. CONCLUSION: The PINK1-PKA-NCLX axis plays an important role in mitochondrial calcium efflux in cardiomyocytes. Therefore, PINK1 may be a therapeutic target to protect cardiomyocyte mitochondria, and the application of huMSC-exo is a promising strategy against sepsis-induced heart dysfunction. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13287-021-02325-6.
format Online
Article
Text
id pubmed-8101124
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-81011242021-05-06 PINK1 contained in huMSC-derived exosomes prevents cardiomyocyte mitochondrial calcium overload in sepsis via recovery of mitochondrial Ca(2+) efflux Zhou, Qin Xie, Min Zhu, Jing Yi, Qin Tan, Bin Li, Yasha Ye, Liang Zhang, Xinyuan Zhang, Ying Tian, Jie Xu, Hao Stem Cell Res Ther Research BACKGROUND: Sepsis is a systemic inflammatory response to a local severe infection that may lead to multiple organ failure and death. Previous studies have shown that 40–50% of patients with sepsis have diverse myocardial injuries and 70 to 90% mortality rates compared to 20% mortality in patients with sepsis without myocardial injury. Therefore, uncovering the mechanism of sepsis-induced myocardial injury and finding a target-based treatment are immensely important. OBJECTIVE: The present study elucidated the mechanism of sepsis-induced myocardial injury and examined the value of human umbilical cord mesenchymal stem cells (huMSCs) for protecting cardiac function in sepsis. METHODS: We used cecal ligation and puncture (CLP) to induce sepsis in mice and detect myocardial injury and cardiac function using serological markers and echocardiography. Cardiomyocyte apoptosis and heart tissue ultrastructure were detected using TdT-mediated dUTP Nick-End Labeling (TUNEL) and transmission electron microscopy (TEM), respectively. Fura-2 AM was used to monitor Ca(2+) uptake and efflux in mitochondria. FQ-PCR and Western blotting detected expression of mitochondrial Ca(2+) distribution regulators and PTEN-induced putative kinase 1 (PINK1). JC-1 was used to detect the mitochondrial membrane potential (Δψm) of cardiomyocytes. RESULTS: We found that expression of PINK1 decreased in mouse hearts during sepsis, which caused cardiomyocyte mitochondrial Ca(2+) efflux disorder, mitochondrial calcium overload, and cardiomyocyte injury. In contrast, we found that exosomes isolated from huMSCs (huMSC-exo) carried Pink1 mRNA, which could be transferred to recipient cardiomyocytes to increase PINK1 expression. The reduction in cardiomyocyte mitochondrial calcium efflux was reversed, and cardiomyocytes recovered from injury. We confirmed the effect of the PINK1-PKA-NCLX axis on mitochondrial calcium homeostasis in cardiomyocytes during sepsis. CONCLUSION: The PINK1-PKA-NCLX axis plays an important role in mitochondrial calcium efflux in cardiomyocytes. Therefore, PINK1 may be a therapeutic target to protect cardiomyocyte mitochondria, and the application of huMSC-exo is a promising strategy against sepsis-induced heart dysfunction. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13287-021-02325-6. BioMed Central 2021-05-06 /pmc/articles/PMC8101124/ /pubmed/33957982 http://dx.doi.org/10.1186/s13287-021-02325-6 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
Zhou, Qin
Xie, Min
Zhu, Jing
Yi, Qin
Tan, Bin
Li, Yasha
Ye, Liang
Zhang, Xinyuan
Zhang, Ying
Tian, Jie
Xu, Hao
PINK1 contained in huMSC-derived exosomes prevents cardiomyocyte mitochondrial calcium overload in sepsis via recovery of mitochondrial Ca(2+) efflux
title PINK1 contained in huMSC-derived exosomes prevents cardiomyocyte mitochondrial calcium overload in sepsis via recovery of mitochondrial Ca(2+) efflux
title_full PINK1 contained in huMSC-derived exosomes prevents cardiomyocyte mitochondrial calcium overload in sepsis via recovery of mitochondrial Ca(2+) efflux
title_fullStr PINK1 contained in huMSC-derived exosomes prevents cardiomyocyte mitochondrial calcium overload in sepsis via recovery of mitochondrial Ca(2+) efflux
title_full_unstemmed PINK1 contained in huMSC-derived exosomes prevents cardiomyocyte mitochondrial calcium overload in sepsis via recovery of mitochondrial Ca(2+) efflux
title_short PINK1 contained in huMSC-derived exosomes prevents cardiomyocyte mitochondrial calcium overload in sepsis via recovery of mitochondrial Ca(2+) efflux
title_sort pink1 contained in humsc-derived exosomes prevents cardiomyocyte mitochondrial calcium overload in sepsis via recovery of mitochondrial ca(2+) efflux
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8101124/
https://www.ncbi.nlm.nih.gov/pubmed/33957982
http://dx.doi.org/10.1186/s13287-021-02325-6
work_keys_str_mv AT zhouqin pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT xiemin pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT zhujing pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT yiqin pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT tanbin pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT liyasha pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT yeliang pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT zhangxinyuan pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT zhangying pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT tianjie pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux
AT xuhao pink1containedinhumscderivedexosomespreventscardiomyocytemitochondrialcalciumoverloadinsepsisviarecoveryofmitochondrialca2efflux