Cargando…

Decreased MFN2 activates the cGAS-STING pathway in diabetic myocardial ischaemia–reperfusion by triggering the release of mitochondrial DNA

BACKGROUND: The cause of aggravation of diabetic myocardial damage is yet to be elucidated; damage to mitochondrial function has been a longstanding focus of research. During diabetic myocardial ischaemia–reperfusion (MI/R), it remains unclear whether reduced mitochondrial fusion exacerbates myocard...

Descripción completa

Detalles Bibliográficos
Autores principales: Xiong, Yonghong, Leng, Yan, Tian, Hao, Deng, Xinqi, Li, Wenyuan, Li, Wei, Xia, Zhongyuan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10398939/
https://www.ncbi.nlm.nih.gov/pubmed/37537600
http://dx.doi.org/10.1186/s12964-023-01216-y
_version_ 1785084140626903040
author Xiong, Yonghong
Leng, Yan
Tian, Hao
Deng, Xinqi
Li, Wenyuan
Li, Wei
Xia, Zhongyuan
author_facet Xiong, Yonghong
Leng, Yan
Tian, Hao
Deng, Xinqi
Li, Wenyuan
Li, Wei
Xia, Zhongyuan
author_sort Xiong, Yonghong
collection PubMed
description BACKGROUND: The cause of aggravation of diabetic myocardial damage is yet to be elucidated; damage to mitochondrial function has been a longstanding focus of research. During diabetic myocardial ischaemia–reperfusion (MI/R), it remains unclear whether reduced mitochondrial fusion exacerbates myocardial injury by generating free damaged mitochondrial DNA (mitoDNA) and activating the cGAS-STING pathway. METHODS: In this study, a mouse model of diabetes was established (by feeding mice a high-fat diet (HFD) plus a low dose of streptozotocin (STZ)), a MI/R model was established by cardiac ischaemia for 2 h and reperfusion for 30 min, and a cellular model of glycolipid toxicity induced by high glucose (HG) and palmitic acid (PA) was established in H9C2 cells. RESULTS: We observed that altered mitochondrial dynamics during diabetic MI/R led to increased mitoDNA in the cytosol, activation of the cGAS-STING pathway, and phosphorylation of the downstream targets TBK1 and IRF3. In the cellular model we found that cytosolic mitoDNA was the result of reduced mitochondrial fusion induced by HG and PA, which also resulted in cGAS-STING signalling and activation of downstream targets. Moreover, inhibition of STING by H-151 significantly ameliorated myocardial injury induced by MFN2 knockdown in both the cell and mouse models. The use of a fat-soluble antioxidant CoQ10 improved cardiac function in the mouse models. CONCLUSIONS: Our study elucidated the critical role of cGAS-STING activation, triggered by increased cytosolic mitoDNA due to decreased mitochondrial fusion, in the pathogenesis of diabetic MI/R injury. This provides preclinical insights for the treatment of diabetic MI/R injury. GRAPHICAL ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12964-023-01216-y.
format Online
Article
Text
id pubmed-10398939
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-103989392023-08-04 Decreased MFN2 activates the cGAS-STING pathway in diabetic myocardial ischaemia–reperfusion by triggering the release of mitochondrial DNA Xiong, Yonghong Leng, Yan Tian, Hao Deng, Xinqi Li, Wenyuan Li, Wei Xia, Zhongyuan Cell Commun Signal Research BACKGROUND: The cause of aggravation of diabetic myocardial damage is yet to be elucidated; damage to mitochondrial function has been a longstanding focus of research. During diabetic myocardial ischaemia–reperfusion (MI/R), it remains unclear whether reduced mitochondrial fusion exacerbates myocardial injury by generating free damaged mitochondrial DNA (mitoDNA) and activating the cGAS-STING pathway. METHODS: In this study, a mouse model of diabetes was established (by feeding mice a high-fat diet (HFD) plus a low dose of streptozotocin (STZ)), a MI/R model was established by cardiac ischaemia for 2 h and reperfusion for 30 min, and a cellular model of glycolipid toxicity induced by high glucose (HG) and palmitic acid (PA) was established in H9C2 cells. RESULTS: We observed that altered mitochondrial dynamics during diabetic MI/R led to increased mitoDNA in the cytosol, activation of the cGAS-STING pathway, and phosphorylation of the downstream targets TBK1 and IRF3. In the cellular model we found that cytosolic mitoDNA was the result of reduced mitochondrial fusion induced by HG and PA, which also resulted in cGAS-STING signalling and activation of downstream targets. Moreover, inhibition of STING by H-151 significantly ameliorated myocardial injury induced by MFN2 knockdown in both the cell and mouse models. The use of a fat-soluble antioxidant CoQ10 improved cardiac function in the mouse models. CONCLUSIONS: Our study elucidated the critical role of cGAS-STING activation, triggered by increased cytosolic mitoDNA due to decreased mitochondrial fusion, in the pathogenesis of diabetic MI/R injury. This provides preclinical insights for the treatment of diabetic MI/R injury. GRAPHICAL ABSTRACT: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12964-023-01216-y. BioMed Central 2023-08-03 /pmc/articles/PMC10398939/ /pubmed/37537600 http://dx.doi.org/10.1186/s12964-023-01216-y Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This 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
Xiong, Yonghong
Leng, Yan
Tian, Hao
Deng, Xinqi
Li, Wenyuan
Li, Wei
Xia, Zhongyuan
Decreased MFN2 activates the cGAS-STING pathway in diabetic myocardial ischaemia–reperfusion by triggering the release of mitochondrial DNA
title Decreased MFN2 activates the cGAS-STING pathway in diabetic myocardial ischaemia–reperfusion by triggering the release of mitochondrial DNA
title_full Decreased MFN2 activates the cGAS-STING pathway in diabetic myocardial ischaemia–reperfusion by triggering the release of mitochondrial DNA
title_fullStr Decreased MFN2 activates the cGAS-STING pathway in diabetic myocardial ischaemia–reperfusion by triggering the release of mitochondrial DNA
title_full_unstemmed Decreased MFN2 activates the cGAS-STING pathway in diabetic myocardial ischaemia–reperfusion by triggering the release of mitochondrial DNA
title_short Decreased MFN2 activates the cGAS-STING pathway in diabetic myocardial ischaemia–reperfusion by triggering the release of mitochondrial DNA
title_sort decreased mfn2 activates the cgas-sting pathway in diabetic myocardial ischaemia–reperfusion by triggering the release of mitochondrial dna
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10398939/
https://www.ncbi.nlm.nih.gov/pubmed/37537600
http://dx.doi.org/10.1186/s12964-023-01216-y
work_keys_str_mv AT xiongyonghong decreasedmfn2activatesthecgasstingpathwayindiabeticmyocardialischaemiareperfusionbytriggeringthereleaseofmitochondrialdna
AT lengyan decreasedmfn2activatesthecgasstingpathwayindiabeticmyocardialischaemiareperfusionbytriggeringthereleaseofmitochondrialdna
AT tianhao decreasedmfn2activatesthecgasstingpathwayindiabeticmyocardialischaemiareperfusionbytriggeringthereleaseofmitochondrialdna
AT dengxinqi decreasedmfn2activatesthecgasstingpathwayindiabeticmyocardialischaemiareperfusionbytriggeringthereleaseofmitochondrialdna
AT liwenyuan decreasedmfn2activatesthecgasstingpathwayindiabeticmyocardialischaemiareperfusionbytriggeringthereleaseofmitochondrialdna
AT liwei decreasedmfn2activatesthecgasstingpathwayindiabeticmyocardialischaemiareperfusionbytriggeringthereleaseofmitochondrialdna
AT xiazhongyuan decreasedmfn2activatesthecgasstingpathwayindiabeticmyocardialischaemiareperfusionbytriggeringthereleaseofmitochondrialdna