Cargando…

Glibenclamide ameliorates the disrupted blood–brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome

BACKGROUND: Glibenclamide is a widely used sulfonylurea drug prescribed to treat type II diabetes mellitus. Previous studies have demonstrated that glibenclamide has neuroprotective effects in central nervous system injury. However, the exact mechanism by which glibenclamide acts on the blood–brain...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Fulin, Shen, Gang, Su, Zuopeng, He, Zijian, Yuan, Lutao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6456786/
https://www.ncbi.nlm.nih.gov/pubmed/30859754
http://dx.doi.org/10.1002/brb3.1254
_version_ 1783409810103861248
author Xu, Fulin
Shen, Gang
Su, Zuopeng
He, Zijian
Yuan, Lutao
author_facet Xu, Fulin
Shen, Gang
Su, Zuopeng
He, Zijian
Yuan, Lutao
author_sort Xu, Fulin
collection PubMed
description BACKGROUND: Glibenclamide is a widely used sulfonylurea drug prescribed to treat type II diabetes mellitus. Previous studies have demonstrated that glibenclamide has neuroprotective effects in central nervous system injury. However, the exact mechanism by which glibenclamide acts on the blood–brain barrier (BBB) after intracerebral hemorrhage (ICH) remains unclear. The purpose of this study was to validate the neuroprotective effects of glibenclamide on ICH and to explore the mechanisms underlying these effects. METHODS: We investigated the effects of glibenclamide on experimental ICH using the autologous blood infusion model. Glibenclamide was administrated either immediately or 2 hr after ICH. Brain edema was quantified using the wet–dry method 3 days after injury. BBB integrity was evaluated by Evans Blue extravasation and degradation of the tight junction protein zona occludens‐1 (ZO‐1). mRNA levels of inflammatory cytokines were determined by quantitative polymerase chain reaction. Activation of the nucleotide‐binding oligomerization domain‐like receptor with a pyrin domain 3 (NLRP3) inflammasome and cell viability were also measured in cerebral microvascular endothelial b.End3 cells exposed to hemin. Neurological changes were evaluated by the Garcia score and rotarod test. RESULTS: After ICH, the brain water content, Evans Blue extravasation, and inflammatory cytokines decreased significantly in the ipsilateral hemisphere of the experimental compared to the vehicle group. Glibenclamide treatment and NLRP3 knockdown significantly reduced hemin‐induced activation of the NLRP3 inflammasome, release of extracellular lactate dehydrogenase, apoptosis, and loss of ZO‐1 in b.End3 cells. However, NLRP3 knockdown abolished the protective effect of glibenclamide. CONCLUSION: Glibenclamide maintained BBB integrity in experimental ICH by inhibiting the activation of the NLRP3 inflammasome in microvessel endothelial cells. Our findings will contribute to elucidating the pharmacological mechanism of action of glibenclamide and to developing a novel therapy for clinical ICH.
format Online
Article
Text
id pubmed-6456786
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-64567862019-04-19 Glibenclamide ameliorates the disrupted blood–brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome Xu, Fulin Shen, Gang Su, Zuopeng He, Zijian Yuan, Lutao Brain Behav Original Research BACKGROUND: Glibenclamide is a widely used sulfonylurea drug prescribed to treat type II diabetes mellitus. Previous studies have demonstrated that glibenclamide has neuroprotective effects in central nervous system injury. However, the exact mechanism by which glibenclamide acts on the blood–brain barrier (BBB) after intracerebral hemorrhage (ICH) remains unclear. The purpose of this study was to validate the neuroprotective effects of glibenclamide on ICH and to explore the mechanisms underlying these effects. METHODS: We investigated the effects of glibenclamide on experimental ICH using the autologous blood infusion model. Glibenclamide was administrated either immediately or 2 hr after ICH. Brain edema was quantified using the wet–dry method 3 days after injury. BBB integrity was evaluated by Evans Blue extravasation and degradation of the tight junction protein zona occludens‐1 (ZO‐1). mRNA levels of inflammatory cytokines were determined by quantitative polymerase chain reaction. Activation of the nucleotide‐binding oligomerization domain‐like receptor with a pyrin domain 3 (NLRP3) inflammasome and cell viability were also measured in cerebral microvascular endothelial b.End3 cells exposed to hemin. Neurological changes were evaluated by the Garcia score and rotarod test. RESULTS: After ICH, the brain water content, Evans Blue extravasation, and inflammatory cytokines decreased significantly in the ipsilateral hemisphere of the experimental compared to the vehicle group. Glibenclamide treatment and NLRP3 knockdown significantly reduced hemin‐induced activation of the NLRP3 inflammasome, release of extracellular lactate dehydrogenase, apoptosis, and loss of ZO‐1 in b.End3 cells. However, NLRP3 knockdown abolished the protective effect of glibenclamide. CONCLUSION: Glibenclamide maintained BBB integrity in experimental ICH by inhibiting the activation of the NLRP3 inflammasome in microvessel endothelial cells. Our findings will contribute to elucidating the pharmacological mechanism of action of glibenclamide and to developing a novel therapy for clinical ICH. John Wiley and Sons Inc. 2019-03-11 /pmc/articles/PMC6456786/ /pubmed/30859754 http://dx.doi.org/10.1002/brb3.1254 Text en © 2019 The Authors. Brain and Behavior published by Wiley Periodicals, Inc. This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Research
Xu, Fulin
Shen, Gang
Su, Zuopeng
He, Zijian
Yuan, Lutao
Glibenclamide ameliorates the disrupted blood–brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome
title Glibenclamide ameliorates the disrupted blood–brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome
title_full Glibenclamide ameliorates the disrupted blood–brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome
title_fullStr Glibenclamide ameliorates the disrupted blood–brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome
title_full_unstemmed Glibenclamide ameliorates the disrupted blood–brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome
title_short Glibenclamide ameliorates the disrupted blood–brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of NLRP3 inflammasome
title_sort glibenclamide ameliorates the disrupted blood–brain barrier in experimental intracerebral hemorrhage by inhibiting the activation of nlrp3 inflammasome
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6456786/
https://www.ncbi.nlm.nih.gov/pubmed/30859754
http://dx.doi.org/10.1002/brb3.1254
work_keys_str_mv AT xufulin glibenclamideamelioratesthedisruptedbloodbrainbarrierinexperimentalintracerebralhemorrhagebyinhibitingtheactivationofnlrp3inflammasome
AT shengang glibenclamideamelioratesthedisruptedbloodbrainbarrierinexperimentalintracerebralhemorrhagebyinhibitingtheactivationofnlrp3inflammasome
AT suzuopeng glibenclamideamelioratesthedisruptedbloodbrainbarrierinexperimentalintracerebralhemorrhagebyinhibitingtheactivationofnlrp3inflammasome
AT hezijian glibenclamideamelioratesthedisruptedbloodbrainbarrierinexperimentalintracerebralhemorrhagebyinhibitingtheactivationofnlrp3inflammasome
AT yuanlutao glibenclamideamelioratesthedisruptedbloodbrainbarrierinexperimentalintracerebralhemorrhagebyinhibitingtheactivationofnlrp3inflammasome