Cargando…

IRAK-M Deficiency Exacerbates Ischemic Neurovascular Injuries in Experimental Stroke Mice

Background: Innate immune response to neuronal death is one of the key events of the pathogenesis of ischemic brain injury. Interleukin-1 receptor-associated kinase (IRAK)-M, encoded by gene Irak3, negatively regulates toll-like receptor signaling by interacting with the MyD88–IRAK-4–IRAK-1 complex...

Descripción completa

Detalles Bibliográficos
Autores principales: Lyu, Chenfei, Zhang, Yongfang, Gu, Minhua, Huang, Yusheng, Liu, Guanghui, Wang, Chen, Li, Miaodan, Chen, Shumin, Pan, Suyue, Gu, Yong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6308305/
https://www.ncbi.nlm.nih.gov/pubmed/30622459
http://dx.doi.org/10.3389/fncel.2018.00504
_version_ 1783383167433965568
author Lyu, Chenfei
Zhang, Yongfang
Gu, Minhua
Huang, Yusheng
Liu, Guanghui
Wang, Chen
Li, Miaodan
Chen, Shumin
Pan, Suyue
Gu, Yong
author_facet Lyu, Chenfei
Zhang, Yongfang
Gu, Minhua
Huang, Yusheng
Liu, Guanghui
Wang, Chen
Li, Miaodan
Chen, Shumin
Pan, Suyue
Gu, Yong
author_sort Lyu, Chenfei
collection PubMed
description Background: Innate immune response to neuronal death is one of the key events of the pathogenesis of ischemic brain injury. Interleukin-1 receptor-associated kinase (IRAK)-M, encoded by gene Irak3, negatively regulates toll-like receptor signaling by interacting with the MyD88–IRAK-4–IRAK-1 complex and blocking the phosphorylation and dissociation of IRAK-1. Its function in the ischemic stroke is unknown. Objective: This study aims to investigate whether IRAK-M deficiency could exacerbate neuroinflammation and neurovascular injuries during cerebral ischemia and reperfusion. Methods: Male C57BL/6 mice and Irak3 knockout mice were subjected to 45 min of middle cerebral artery occlusion and 4 or 24 h of reperfusion. Transcription of Irak3 gene was evaluated by quantitative real-time PCR (qRT-PCR). Then, infarct volume, neurological score, brain water content, and Evans blue leakage were compared between knock-out and wild-type mice after reperfusion. Through the observation of gross brain specimen after cerebral ischemia, the incidence of hemorrhage transformation was compared between KO and WT mice. To explore underlying signaling pathways involved in IRAK-M deficiency, major proinflammatory cytokines and NF-κB signaling were measured by qRT-PCR and Western blot. Results: The expression of IRAK-M peaked at 1 h after reperfusion, and then gradually decreased within the first 24 h, which was abolished by blocking the expression of hypoxia induced factor 1α. IRAK-M deficiency increased infarct volume, brain edema, the incidence of hemorrhage transformation, and the permeability of blood–brain barrier. In addition, the NF-κB-mediated expressions of proinflammatory cytokines and the activation of microglia in the ipsilateral brain from knock-out mice were much higher than those in wild-type littermates. Conclusion: IRAK-M deletion exacerbates neurovascular damages which are related to the pronounced activation of NF-κB signaling and neuroinflammatory responses during cerebral ischemia-reperfusion in mice. Our study indicates that IRAK-M has neuroprotective effect and has potential to facilitate the development of new pharmaceuticals that reduce neurovascular complications.
format Online
Article
Text
id pubmed-6308305
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-63083052019-01-08 IRAK-M Deficiency Exacerbates Ischemic Neurovascular Injuries in Experimental Stroke Mice Lyu, Chenfei Zhang, Yongfang Gu, Minhua Huang, Yusheng Liu, Guanghui Wang, Chen Li, Miaodan Chen, Shumin Pan, Suyue Gu, Yong Front Cell Neurosci Neuroscience Background: Innate immune response to neuronal death is one of the key events of the pathogenesis of ischemic brain injury. Interleukin-1 receptor-associated kinase (IRAK)-M, encoded by gene Irak3, negatively regulates toll-like receptor signaling by interacting with the MyD88–IRAK-4–IRAK-1 complex and blocking the phosphorylation and dissociation of IRAK-1. Its function in the ischemic stroke is unknown. Objective: This study aims to investigate whether IRAK-M deficiency could exacerbate neuroinflammation and neurovascular injuries during cerebral ischemia and reperfusion. Methods: Male C57BL/6 mice and Irak3 knockout mice were subjected to 45 min of middle cerebral artery occlusion and 4 or 24 h of reperfusion. Transcription of Irak3 gene was evaluated by quantitative real-time PCR (qRT-PCR). Then, infarct volume, neurological score, brain water content, and Evans blue leakage were compared between knock-out and wild-type mice after reperfusion. Through the observation of gross brain specimen after cerebral ischemia, the incidence of hemorrhage transformation was compared between KO and WT mice. To explore underlying signaling pathways involved in IRAK-M deficiency, major proinflammatory cytokines and NF-κB signaling were measured by qRT-PCR and Western blot. Results: The expression of IRAK-M peaked at 1 h after reperfusion, and then gradually decreased within the first 24 h, which was abolished by blocking the expression of hypoxia induced factor 1α. IRAK-M deficiency increased infarct volume, brain edema, the incidence of hemorrhage transformation, and the permeability of blood–brain barrier. In addition, the NF-κB-mediated expressions of proinflammatory cytokines and the activation of microglia in the ipsilateral brain from knock-out mice were much higher than those in wild-type littermates. Conclusion: IRAK-M deletion exacerbates neurovascular damages which are related to the pronounced activation of NF-κB signaling and neuroinflammatory responses during cerebral ischemia-reperfusion in mice. Our study indicates that IRAK-M has neuroprotective effect and has potential to facilitate the development of new pharmaceuticals that reduce neurovascular complications. Frontiers Media S.A. 2018-12-21 /pmc/articles/PMC6308305/ /pubmed/30622459 http://dx.doi.org/10.3389/fncel.2018.00504 Text en Copyright © 2018 Lyu, Zhang, Gu, Huang, Liu, Wang, Li, Chen, Pan and Gu. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Neuroscience
Lyu, Chenfei
Zhang, Yongfang
Gu, Minhua
Huang, Yusheng
Liu, Guanghui
Wang, Chen
Li, Miaodan
Chen, Shumin
Pan, Suyue
Gu, Yong
IRAK-M Deficiency Exacerbates Ischemic Neurovascular Injuries in Experimental Stroke Mice
title IRAK-M Deficiency Exacerbates Ischemic Neurovascular Injuries in Experimental Stroke Mice
title_full IRAK-M Deficiency Exacerbates Ischemic Neurovascular Injuries in Experimental Stroke Mice
title_fullStr IRAK-M Deficiency Exacerbates Ischemic Neurovascular Injuries in Experimental Stroke Mice
title_full_unstemmed IRAK-M Deficiency Exacerbates Ischemic Neurovascular Injuries in Experimental Stroke Mice
title_short IRAK-M Deficiency Exacerbates Ischemic Neurovascular Injuries in Experimental Stroke Mice
title_sort irak-m deficiency exacerbates ischemic neurovascular injuries in experimental stroke mice
topic Neuroscience
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6308305/
https://www.ncbi.nlm.nih.gov/pubmed/30622459
http://dx.doi.org/10.3389/fncel.2018.00504
work_keys_str_mv AT lyuchenfei irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice
AT zhangyongfang irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice
AT guminhua irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice
AT huangyusheng irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice
AT liuguanghui irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice
AT wangchen irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice
AT limiaodan irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice
AT chenshumin irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice
AT pansuyue irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice
AT guyong irakmdeficiencyexacerbatesischemicneurovascularinjuriesinexperimentalstrokemice