Cargando…

Activating cGAS–STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis

BACKGROUND: Neuroinflammation-induced injury is intimately associated with poor prognosis in patients with cerebral venous sinus thrombosis (CVST). The cyclic GMP-AMP synthase–stimulator of interferon gene (cGAS–STING) axis is a cytoplasmic double-stranded DNA (dsDNA) sensing pathway has recently em...

Descripción completa

Detalles Bibliográficos
Autores principales: Ding, Rui, Li, Haiyan, Liu, Yaqi, Ou, Weiyang, Zhang, Xifang, Chai, Huihui, Huang, Xiaofei, Yang, Weijie, Wang, Qiujing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9188164/
https://www.ncbi.nlm.nih.gov/pubmed/35689216
http://dx.doi.org/10.1186/s12974-022-02511-0
_version_ 1784725314687991808
author Ding, Rui
Li, Haiyan
Liu, Yaqi
Ou, Weiyang
Zhang, Xifang
Chai, Huihui
Huang, Xiaofei
Yang, Weijie
Wang, Qiujing
author_facet Ding, Rui
Li, Haiyan
Liu, Yaqi
Ou, Weiyang
Zhang, Xifang
Chai, Huihui
Huang, Xiaofei
Yang, Weijie
Wang, Qiujing
author_sort Ding, Rui
collection PubMed
description BACKGROUND: Neuroinflammation-induced injury is intimately associated with poor prognosis in patients with cerebral venous sinus thrombosis (CVST). The cyclic GMP-AMP synthase–stimulator of interferon gene (cGAS–STING) axis is a cytoplasmic double-stranded DNA (dsDNA) sensing pathway has recently emerged as a crucial mediator of neuroinflammation in ischemic stroke. However, the role of the cGAS–STING pathway in modulating post-CVST inflammation and the underlying mechanisms involved remain unclear. METHODS: A CVST model was induced by ferric chloride in male C57BL/6J mice. The selective cGAS inhibitor RU.521, STING agonist 2′3′-cGAMP, and STING siRNA were delivered by intranasal administration or intraventricular injection. Post-CVST assessments included rotarod test, TUNEL staining, Fluoro-Jade C staining, dihydroethidium staining, western blotting, qPCR, immunofluorescence, immunohistochemistry, ELISA and flow cytometry. RESULTS: cGAS, STING, NLRP3 and GSDMD were significantly upregulated after CVST and mostly in the microglia of the mouse brain. CVST triggered the release of dsDNA into the cytoplasm and elicited an inflammatory response via activating the cGAS–STING axis. RU.521 decreased the levels of 2′3′-cGAMP, STING and downstream inflammatory cytokines, and suppressed the expressions of NLRP3 inflammasome and pyroptosis-pertinent components containing cleaved caspase-1, GSDMD, GSDMD-C, pro- and cleaved IL-1β, and cleaved IL-1β/pro-IL-1β. Besides, RU.521 treatment also reduced oxidative stress, lessened the numbers of microglia and neutrophils, and ameliorated neuronal apoptosis, degeneration along with neurological deficits post-CVST. 2′3'-cGAMP delivery enhanced the expressions of STING and related inflammatory mediators, NLRP3 inflammasome and pyroptosis-relevant proteins, whereas these alterations were significantly abrogated by the silencing of STING by siRNA. CONCLUSIONS: Our data demonstrate that repression of the cGAS–STING pathway diminishes the neuroinflammatory burden of CVST and highlight this approach as a potential therapeutic tactic in CVST-mediated pathologies. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02511-0.
format Online
Article
Text
id pubmed-9188164
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-91881642022-06-12 Activating cGAS–STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis Ding, Rui Li, Haiyan Liu, Yaqi Ou, Weiyang Zhang, Xifang Chai, Huihui Huang, Xiaofei Yang, Weijie Wang, Qiujing J Neuroinflammation Research BACKGROUND: Neuroinflammation-induced injury is intimately associated with poor prognosis in patients with cerebral venous sinus thrombosis (CVST). The cyclic GMP-AMP synthase–stimulator of interferon gene (cGAS–STING) axis is a cytoplasmic double-stranded DNA (dsDNA) sensing pathway has recently emerged as a crucial mediator of neuroinflammation in ischemic stroke. However, the role of the cGAS–STING pathway in modulating post-CVST inflammation and the underlying mechanisms involved remain unclear. METHODS: A CVST model was induced by ferric chloride in male C57BL/6J mice. The selective cGAS inhibitor RU.521, STING agonist 2′3′-cGAMP, and STING siRNA were delivered by intranasal administration or intraventricular injection. Post-CVST assessments included rotarod test, TUNEL staining, Fluoro-Jade C staining, dihydroethidium staining, western blotting, qPCR, immunofluorescence, immunohistochemistry, ELISA and flow cytometry. RESULTS: cGAS, STING, NLRP3 and GSDMD were significantly upregulated after CVST and mostly in the microglia of the mouse brain. CVST triggered the release of dsDNA into the cytoplasm and elicited an inflammatory response via activating the cGAS–STING axis. RU.521 decreased the levels of 2′3′-cGAMP, STING and downstream inflammatory cytokines, and suppressed the expressions of NLRP3 inflammasome and pyroptosis-pertinent components containing cleaved caspase-1, GSDMD, GSDMD-C, pro- and cleaved IL-1β, and cleaved IL-1β/pro-IL-1β. Besides, RU.521 treatment also reduced oxidative stress, lessened the numbers of microglia and neutrophils, and ameliorated neuronal apoptosis, degeneration along with neurological deficits post-CVST. 2′3'-cGAMP delivery enhanced the expressions of STING and related inflammatory mediators, NLRP3 inflammasome and pyroptosis-relevant proteins, whereas these alterations were significantly abrogated by the silencing of STING by siRNA. CONCLUSIONS: Our data demonstrate that repression of the cGAS–STING pathway diminishes the neuroinflammatory burden of CVST and highlight this approach as a potential therapeutic tactic in CVST-mediated pathologies. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12974-022-02511-0. BioMed Central 2022-06-10 /pmc/articles/PMC9188164/ /pubmed/35689216 http://dx.doi.org/10.1186/s12974-022-02511-0 Text en © The Author(s) 2022 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
Ding, Rui
Li, Haiyan
Liu, Yaqi
Ou, Weiyang
Zhang, Xifang
Chai, Huihui
Huang, Xiaofei
Yang, Weijie
Wang, Qiujing
Activating cGAS–STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis
title Activating cGAS–STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis
title_full Activating cGAS–STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis
title_fullStr Activating cGAS–STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis
title_full_unstemmed Activating cGAS–STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis
title_short Activating cGAS–STING axis contributes to neuroinflammation in CVST mouse model and induces inflammasome activation and microglia pyroptosis
title_sort activating cgas–sting axis contributes to neuroinflammation in cvst mouse model and induces inflammasome activation and microglia pyroptosis
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9188164/
https://www.ncbi.nlm.nih.gov/pubmed/35689216
http://dx.doi.org/10.1186/s12974-022-02511-0
work_keys_str_mv AT dingrui activatingcgasstingaxiscontributestoneuroinflammationincvstmousemodelandinducesinflammasomeactivationandmicrogliapyroptosis
AT lihaiyan activatingcgasstingaxiscontributestoneuroinflammationincvstmousemodelandinducesinflammasomeactivationandmicrogliapyroptosis
AT liuyaqi activatingcgasstingaxiscontributestoneuroinflammationincvstmousemodelandinducesinflammasomeactivationandmicrogliapyroptosis
AT ouweiyang activatingcgasstingaxiscontributestoneuroinflammationincvstmousemodelandinducesinflammasomeactivationandmicrogliapyroptosis
AT zhangxifang activatingcgasstingaxiscontributestoneuroinflammationincvstmousemodelandinducesinflammasomeactivationandmicrogliapyroptosis
AT chaihuihui activatingcgasstingaxiscontributestoneuroinflammationincvstmousemodelandinducesinflammasomeactivationandmicrogliapyroptosis
AT huangxiaofei activatingcgasstingaxiscontributestoneuroinflammationincvstmousemodelandinducesinflammasomeactivationandmicrogliapyroptosis
AT yangweijie activatingcgasstingaxiscontributestoneuroinflammationincvstmousemodelandinducesinflammasomeactivationandmicrogliapyroptosis
AT wangqiujing activatingcgasstingaxiscontributestoneuroinflammationincvstmousemodelandinducesinflammasomeactivationandmicrogliapyroptosis