IRAK-M suppresses the activation of microglial NLRP3 inflammasome and GSDMD-mediated pyroptosis through inhibiting IRAK1 phosphorylation during experimental autoimmune encephalomyelitis

Abstract The activation of the NOD-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome triggers pyroptosis proinflammatory cell death in experimental autoimmune encephalomyelitis (EAE). However, the underlying mechanisms of the inflammatory processes of microglia in EAE remai...

Full description

Bibliographic Details
Main Authors: Yuanyuan Wang, Shanshan Pei, Zhuhe Liu, Yuewen Ding, Tinglin Qian, Haixia Wen, Ssu-Wei Hsu, Zheyi Zhou, Jun Zhang, Honghao Wang
Format: Article
Language:English
Published: Nature Publishing Group 2023-02-01
Series:Cell Death and Disease
Online Access:https://doi.org/10.1038/s41419-023-05621-6
_version_ 1811165699911974912
author Yuanyuan Wang
Shanshan Pei
Zhuhe Liu
Yuewen Ding
Tinglin Qian
Haixia Wen
Ssu-Wei Hsu
Zheyi Zhou
Jun Zhang
Honghao Wang
author_facet Yuanyuan Wang
Shanshan Pei
Zhuhe Liu
Yuewen Ding
Tinglin Qian
Haixia Wen
Ssu-Wei Hsu
Zheyi Zhou
Jun Zhang
Honghao Wang
author_sort Yuanyuan Wang
collection DOAJ
description Abstract The activation of the NOD-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome triggers pyroptosis proinflammatory cell death in experimental autoimmune encephalomyelitis (EAE). However, the underlying mechanisms of the inflammatory processes of microglia in EAE remain unclear. Our previous studies suggested that interleukin-1 receptor-associated kinase (IRAK)-M down-regulates the toll-like receptor 4/interleukin-1 receptor signaling pathway. Here, we used IRAK-M knockout (IRAK-M−/−) mice and their microglia to dissect the role of IRAK-M in EAE. We found that deletion of IRAK-M increased the incidence rate and exacerbated the clinical symptoms in EAE mice. We then found that IRAK-M deficiency promoted the activation of microglia, activated NLRP3 inflammasomes, and enhanced GSDMD-mediated pyroptosis in the microglia of EAE. In contrast, over-expression of IRAK-M exerted inhibitory effects on neuroinflammation, NLRP3 activation, and pyroptosis. Moreover, IRAK-M deficiency enhanced the phosphorylation of IRAK1, while IRAK-M over-expression downregulated the level of phosphorylated IRAK1. Finally, we found upregulated binding of IRAK1 and TNF receptor-associated factor 6 (TRAF6) in IRAK-M−/− EAE mice compared to WT mice, which was blocked in AAVIRAK-M EAE mice. Our study reveals a complex signaling network of IRAK-M, which negatively regulates microglial NLRP3 inflammasomes and pyroptosis by inhibiting IRAK1 phosphorylation during EAE. These findings suggest a potential target for the novel therapeutic approaches of multiple sclerosis (MS)/EAE and NLRP3-related inflammatory diseases.
first_indexed 2024-04-10T15:40:55Z
format Article
id doaj.art-5f4c57c709304b4bae20b21773b174e4
institution Directory Open Access Journal
issn 2041-4889
language English
last_indexed 2024-04-10T15:40:55Z
publishDate 2023-02-01
publisher Nature Publishing Group
record_format Article
series Cell Death and Disease
spelling doaj.art-5f4c57c709304b4bae20b21773b174e42023-02-12T12:24:04ZengNature Publishing GroupCell Death and Disease2041-48892023-02-0114211210.1038/s41419-023-05621-6IRAK-M suppresses the activation of microglial NLRP3 inflammasome and GSDMD-mediated pyroptosis through inhibiting IRAK1 phosphorylation during experimental autoimmune encephalomyelitisYuanyuan Wang0Shanshan Pei1Zhuhe Liu2Yuewen Ding3Tinglin Qian4Haixia Wen5Ssu-Wei Hsu6Zheyi Zhou7Jun Zhang8Honghao Wang9Department of Neurology, Guangzhou First People’s Hospital, School of Medicine, South China University of TechnologyDepartment of Neurology, Nanfang Hospital, Southern Medical UniversityDepartment of Neurology, Guangzhou First People’s Hospital, School of Medicine, South China University of TechnologyDepartment of Neurology, Nanfang Hospital, Southern Medical UniversityDepartment of Neurology, Nanfang Hospital, Southern Medical UniversityDepartment of Neurology, Guangzhou First People’s Hospital, School of Medicine, South China University of TechnologyDepartment of Internal Medicine, University of California at DavisDepartment of Neurology, Hospital of Liuzhou Traditional Chinese MedicineDepartment of Internal Medicine, University of California at DavisDepartment of Neurology, Guangzhou First People’s Hospital, School of Medicine, South China University of TechnologyAbstract The activation of the NOD-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome triggers pyroptosis proinflammatory cell death in experimental autoimmune encephalomyelitis (EAE). However, the underlying mechanisms of the inflammatory processes of microglia in EAE remain unclear. Our previous studies suggested that interleukin-1 receptor-associated kinase (IRAK)-M down-regulates the toll-like receptor 4/interleukin-1 receptor signaling pathway. Here, we used IRAK-M knockout (IRAK-M−/−) mice and their microglia to dissect the role of IRAK-M in EAE. We found that deletion of IRAK-M increased the incidence rate and exacerbated the clinical symptoms in EAE mice. We then found that IRAK-M deficiency promoted the activation of microglia, activated NLRP3 inflammasomes, and enhanced GSDMD-mediated pyroptosis in the microglia of EAE. In contrast, over-expression of IRAK-M exerted inhibitory effects on neuroinflammation, NLRP3 activation, and pyroptosis. Moreover, IRAK-M deficiency enhanced the phosphorylation of IRAK1, while IRAK-M over-expression downregulated the level of phosphorylated IRAK1. Finally, we found upregulated binding of IRAK1 and TNF receptor-associated factor 6 (TRAF6) in IRAK-M−/− EAE mice compared to WT mice, which was blocked in AAVIRAK-M EAE mice. Our study reveals a complex signaling network of IRAK-M, which negatively regulates microglial NLRP3 inflammasomes and pyroptosis by inhibiting IRAK1 phosphorylation during EAE. These findings suggest a potential target for the novel therapeutic approaches of multiple sclerosis (MS)/EAE and NLRP3-related inflammatory diseases.https://doi.org/10.1038/s41419-023-05621-6
spellingShingle Yuanyuan Wang
Shanshan Pei
Zhuhe Liu
Yuewen Ding
Tinglin Qian
Haixia Wen
Ssu-Wei Hsu
Zheyi Zhou
Jun Zhang
Honghao Wang
IRAK-M suppresses the activation of microglial NLRP3 inflammasome and GSDMD-mediated pyroptosis through inhibiting IRAK1 phosphorylation during experimental autoimmune encephalomyelitis
Cell Death and Disease
title IRAK-M suppresses the activation of microglial NLRP3 inflammasome and GSDMD-mediated pyroptosis through inhibiting IRAK1 phosphorylation during experimental autoimmune encephalomyelitis
title_full IRAK-M suppresses the activation of microglial NLRP3 inflammasome and GSDMD-mediated pyroptosis through inhibiting IRAK1 phosphorylation during experimental autoimmune encephalomyelitis
title_fullStr IRAK-M suppresses the activation of microglial NLRP3 inflammasome and GSDMD-mediated pyroptosis through inhibiting IRAK1 phosphorylation during experimental autoimmune encephalomyelitis
title_full_unstemmed IRAK-M suppresses the activation of microglial NLRP3 inflammasome and GSDMD-mediated pyroptosis through inhibiting IRAK1 phosphorylation during experimental autoimmune encephalomyelitis
title_short IRAK-M suppresses the activation of microglial NLRP3 inflammasome and GSDMD-mediated pyroptosis through inhibiting IRAK1 phosphorylation during experimental autoimmune encephalomyelitis
title_sort irak m suppresses the activation of microglial nlrp3 inflammasome and gsdmd mediated pyroptosis through inhibiting irak1 phosphorylation during experimental autoimmune encephalomyelitis
url https://doi.org/10.1038/s41419-023-05621-6
work_keys_str_mv AT yuanyuanwang irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis
AT shanshanpei irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis
AT zhuheliu irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis
AT yuewending irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis
AT tinglinqian irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis
AT haixiawen irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis
AT ssuweihsu irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis
AT zheyizhou irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis
AT junzhang irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis
AT honghaowang irakmsuppressestheactivationofmicroglialnlrp3inflammasomeandgsdmdmediatedpyroptosisthroughinhibitingirak1phosphorylationduringexperimentalautoimmuneencephalomyelitis