Cargando…

iNOS aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtDNA-mediated cGAS-STING pathway

Background: Sterile inflammation contributes to the pathogenesis of cardiac dysfunction caused by various conditions including pressure overload in hypertension. Mitochondrial DNA (mtDNA) released from damaged mitochondria has been implicated in cardiac inflammation. However, the upstream mechanisms...

Descripción completa

Detalles Bibliográficos
Autores principales: Guo, Yongzheng, You, Yuehua, Shang, Fei-Fei, Wang, Xiaowen, Huang, Bi, Zhao, Boying, Lv, Dingyi, Yang, Shenglan, Xie, Ming, Kong, Lingwen, Du, Dingyuan, Luo, Suxin, Tian, Xin, Xia, Yong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10405855/
https://www.ncbi.nlm.nih.gov/pubmed/37554263
http://dx.doi.org/10.7150/thno.84049
_version_ 1785085625846726656
author Guo, Yongzheng
You, Yuehua
Shang, Fei-Fei
Wang, Xiaowen
Huang, Bi
Zhao, Boying
Lv, Dingyi
Yang, Shenglan
Xie, Ming
Kong, Lingwen
Du, Dingyuan
Luo, Suxin
Tian, Xin
Xia, Yong
author_facet Guo, Yongzheng
You, Yuehua
Shang, Fei-Fei
Wang, Xiaowen
Huang, Bi
Zhao, Boying
Lv, Dingyi
Yang, Shenglan
Xie, Ming
Kong, Lingwen
Du, Dingyuan
Luo, Suxin
Tian, Xin
Xia, Yong
author_sort Guo, Yongzheng
collection PubMed
description Background: Sterile inflammation contributes to the pathogenesis of cardiac dysfunction caused by various conditions including pressure overload in hypertension. Mitochondrial DNA (mtDNA) released from damaged mitochondria has been implicated in cardiac inflammation. However, the upstream mechanisms governing mtDNA release and how mtDNA activates sterile inflammation in pressure-overloaded hearts remain largely unknown. Here, we investigated the role of inducible NO synthase (iNOS) on pressure overload-induced cytosolic accumulation of mtDNA and whether mtDNA activated inflammation through the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. Methods: To investigate whether the cGAS-STING cascade was involved in sterile inflammation and cardiac dysfunction upon pressure overload, cardiomyocyte-specific STING depletion mice and mice injected with adeno-associated virus-9 (AAV-9) to suppress the cGAS-STING cascade in the heart were subjected to transverse aortic constriction (TAC). iNOS null mice were used to determine the role of iNOS in cGAS-STING pathway activation in pressure-stressed hearts. Results: iNOS knockout abrogated mtDNA release and alleviated cardiac sterile inflammation resulting in improved cardiac function. Conversely, activating the cGAS-STING pathway blunted the protective effects of iNOS knockout. Moreover, iNOS activated the cGAS-STING pathway in isolated myocytes and this was prevented by depleting cytosolic mtDNA. In addition, disruption of the cGAS-STING pathway suppressed inflammatory cytokine transcription and modulated M1/M2 macrophage polarization, and thus mitigated cardiac remodeling and improved heart function. Finally, increased iNOS expression along with cytosolic mtDNA accumulation and cGAS-STING activation were also seen in human hypertensive hearts. Conclusion: Our findings demonstrate that mtDNA is released into the cytosol and triggers sterile inflammation through the cGAS-STING pathway leading to cardiac dysfunction after pressure overload. iNOS controls mtDNA release and subsequent cGAS activation in pressure-stressed hearts.
format Online
Article
Text
id pubmed-10405855
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-104058552023-08-08 iNOS aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtDNA-mediated cGAS-STING pathway Guo, Yongzheng You, Yuehua Shang, Fei-Fei Wang, Xiaowen Huang, Bi Zhao, Boying Lv, Dingyi Yang, Shenglan Xie, Ming Kong, Lingwen Du, Dingyuan Luo, Suxin Tian, Xin Xia, Yong Theranostics Research Paper Background: Sterile inflammation contributes to the pathogenesis of cardiac dysfunction caused by various conditions including pressure overload in hypertension. Mitochondrial DNA (mtDNA) released from damaged mitochondria has been implicated in cardiac inflammation. However, the upstream mechanisms governing mtDNA release and how mtDNA activates sterile inflammation in pressure-overloaded hearts remain largely unknown. Here, we investigated the role of inducible NO synthase (iNOS) on pressure overload-induced cytosolic accumulation of mtDNA and whether mtDNA activated inflammation through the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway. Methods: To investigate whether the cGAS-STING cascade was involved in sterile inflammation and cardiac dysfunction upon pressure overload, cardiomyocyte-specific STING depletion mice and mice injected with adeno-associated virus-9 (AAV-9) to suppress the cGAS-STING cascade in the heart were subjected to transverse aortic constriction (TAC). iNOS null mice were used to determine the role of iNOS in cGAS-STING pathway activation in pressure-stressed hearts. Results: iNOS knockout abrogated mtDNA release and alleviated cardiac sterile inflammation resulting in improved cardiac function. Conversely, activating the cGAS-STING pathway blunted the protective effects of iNOS knockout. Moreover, iNOS activated the cGAS-STING pathway in isolated myocytes and this was prevented by depleting cytosolic mtDNA. In addition, disruption of the cGAS-STING pathway suppressed inflammatory cytokine transcription and modulated M1/M2 macrophage polarization, and thus mitigated cardiac remodeling and improved heart function. Finally, increased iNOS expression along with cytosolic mtDNA accumulation and cGAS-STING activation were also seen in human hypertensive hearts. Conclusion: Our findings demonstrate that mtDNA is released into the cytosol and triggers sterile inflammation through the cGAS-STING pathway leading to cardiac dysfunction after pressure overload. iNOS controls mtDNA release and subsequent cGAS activation in pressure-stressed hearts. Ivyspring International Publisher 2023-07-24 /pmc/articles/PMC10405855/ /pubmed/37554263 http://dx.doi.org/10.7150/thno.84049 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Guo, Yongzheng
You, Yuehua
Shang, Fei-Fei
Wang, Xiaowen
Huang, Bi
Zhao, Boying
Lv, Dingyi
Yang, Shenglan
Xie, Ming
Kong, Lingwen
Du, Dingyuan
Luo, Suxin
Tian, Xin
Xia, Yong
iNOS aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtDNA-mediated cGAS-STING pathway
title iNOS aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtDNA-mediated cGAS-STING pathway
title_full iNOS aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtDNA-mediated cGAS-STING pathway
title_fullStr iNOS aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtDNA-mediated cGAS-STING pathway
title_full_unstemmed iNOS aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtDNA-mediated cGAS-STING pathway
title_short iNOS aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtDNA-mediated cGAS-STING pathway
title_sort inos aggravates pressure overload-induced cardiac dysfunction via activation of the cytosolic-mtdna-mediated cgas-sting pathway
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10405855/
https://www.ncbi.nlm.nih.gov/pubmed/37554263
http://dx.doi.org/10.7150/thno.84049
work_keys_str_mv AT guoyongzheng inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT youyuehua inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT shangfeifei inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT wangxiaowen inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT huangbi inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT zhaoboying inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT lvdingyi inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT yangshenglan inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT xieming inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT konglingwen inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT dudingyuan inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT luosuxin inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT tianxin inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway
AT xiayong inosaggravatespressureoverloadinducedcardiacdysfunctionviaactivationofthecytosolicmtdnamediatedcgasstingpathway