NLRP3 inhibition attenuates early brain injury and delayed cerebral vasospasm after subarachnoid hemorrhage

Abstract Background The NLRP3 inflammasome is a critical mediator of several vascular diseases through positive regulation of proinflammatory pathways. In this study, we defined the role of NLRP3 in both the acute and delayed phases following subarachnoid hemorrhage (SAH). SAH is associated with dev...

Full description

Bibliographic Details
Main Authors: William S. Dodd, Imaray Noda, Melanie Martinez, Koji Hosaka, Brian L. Hoh
Format: Article
Language:English
Published: BMC 2021-07-01
Series:Journal of Neuroinflammation
Subjects:
Online Access:https://doi.org/10.1186/s12974-021-02207-x
id doaj-dc49f4eb97b14a6e89d742bf271f7299
record_format Article
spelling doaj-dc49f4eb97b14a6e89d742bf271f72992021-07-25T11:42:29ZengBMCJournal of Neuroinflammation1742-20942021-07-0118111110.1186/s12974-021-02207-xNLRP3 inhibition attenuates early brain injury and delayed cerebral vasospasm after subarachnoid hemorrhageWilliam S. Dodd0Imaray Noda1Melanie Martinez2Koji Hosaka3Brian L. Hoh4Department of Neurosurgery, College of Medicine, University of FloridaDepartment of Neurosurgery, College of Medicine, University of FloridaDepartment of Neurosurgery, College of Medicine, University of FloridaDepartment of Neurosurgery, College of Medicine, University of FloridaDepartment of Neurosurgery, College of Medicine, University of FloridaAbstract Background The NLRP3 inflammasome is a critical mediator of several vascular diseases through positive regulation of proinflammatory pathways. In this study, we defined the role of NLRP3 in both the acute and delayed phases following subarachnoid hemorrhage (SAH). SAH is associated with devastating early brain injury (EBI) in the acute phase, and those that survive remain at risk for developing delayed cerebral ischemia (DCI) due to cerebral vasospasm. Current therapies are not effective in preventing the morbidity and mortality associated with EBI and DCI. NLRP3 activation is known to drive IL-1β production and stimulate microglia reactivity, both hallmarks of SAH pathology; thus, we hypothesized that inhibition of NLRP3 could alleviate SAH-induced vascular dysfunction and functional deficits. Methods We studied NLRP3 in an anterior circulation autologous blood injection model of SAH in mice. Mice were randomized to either sham surgery + vehicle, SAH + vehicle, or SAH + MCC950 (a selective NLRP3 inhibitor). The acute phase was studied at 1 day post-SAH and delayed phase at 5 days post-SAH. Results NLRP3 inhibition improved outcomes at both 1 and 5 days post-SAH. In the acute (1 day post-SAH) phase, NLRP3 inhibition attenuated cerebral edema, tight junction disruption, microthrombosis, and microglial reactive morphology shift. Further, we observed a decrease in apoptosis of neurons in mice treated with MCC950. NLRP3 inhibition also prevented middle cerebral artery vasospasm in the delayed (5 days post-SAH) phase and blunted SAH-induced sensorimotor deficits. Conclusions We demonstrate a novel association between NLRP3-mediated neuroinflammation and cerebrovascular dysfunction in both the early and delayed phases after SAH. MCC950 and other NLRP3 inhibitors could be promising tools in the development of therapeutics for EBI and DCI.https://doi.org/10.1186/s12974-021-02207-xSubarachnoid hemorrhageMicrogliaNLRP3 inflammasomeStrokeInflammation
collection DOAJ
language English
format Article
sources DOAJ
author William S. Dodd
Imaray Noda
Melanie Martinez
Koji Hosaka
Brian L. Hoh
spellingShingle William S. Dodd
Imaray Noda
Melanie Martinez
Koji Hosaka
Brian L. Hoh
NLRP3 inhibition attenuates early brain injury and delayed cerebral vasospasm after subarachnoid hemorrhage
Journal of Neuroinflammation
Subarachnoid hemorrhage
Microglia
NLRP3 inflammasome
Stroke
Inflammation
author_facet William S. Dodd
Imaray Noda
Melanie Martinez
Koji Hosaka
Brian L. Hoh
author_sort William S. Dodd
title NLRP3 inhibition attenuates early brain injury and delayed cerebral vasospasm after subarachnoid hemorrhage
title_short NLRP3 inhibition attenuates early brain injury and delayed cerebral vasospasm after subarachnoid hemorrhage
title_full NLRP3 inhibition attenuates early brain injury and delayed cerebral vasospasm after subarachnoid hemorrhage
title_fullStr NLRP3 inhibition attenuates early brain injury and delayed cerebral vasospasm after subarachnoid hemorrhage
title_full_unstemmed NLRP3 inhibition attenuates early brain injury and delayed cerebral vasospasm after subarachnoid hemorrhage
title_sort nlrp3 inhibition attenuates early brain injury and delayed cerebral vasospasm after subarachnoid hemorrhage
publisher BMC
series Journal of Neuroinflammation
issn 1742-2094
publishDate 2021-07-01
description Abstract Background The NLRP3 inflammasome is a critical mediator of several vascular diseases through positive regulation of proinflammatory pathways. In this study, we defined the role of NLRP3 in both the acute and delayed phases following subarachnoid hemorrhage (SAH). SAH is associated with devastating early brain injury (EBI) in the acute phase, and those that survive remain at risk for developing delayed cerebral ischemia (DCI) due to cerebral vasospasm. Current therapies are not effective in preventing the morbidity and mortality associated with EBI and DCI. NLRP3 activation is known to drive IL-1β production and stimulate microglia reactivity, both hallmarks of SAH pathology; thus, we hypothesized that inhibition of NLRP3 could alleviate SAH-induced vascular dysfunction and functional deficits. Methods We studied NLRP3 in an anterior circulation autologous blood injection model of SAH in mice. Mice were randomized to either sham surgery + vehicle, SAH + vehicle, or SAH + MCC950 (a selective NLRP3 inhibitor). The acute phase was studied at 1 day post-SAH and delayed phase at 5 days post-SAH. Results NLRP3 inhibition improved outcomes at both 1 and 5 days post-SAH. In the acute (1 day post-SAH) phase, NLRP3 inhibition attenuated cerebral edema, tight junction disruption, microthrombosis, and microglial reactive morphology shift. Further, we observed a decrease in apoptosis of neurons in mice treated with MCC950. NLRP3 inhibition also prevented middle cerebral artery vasospasm in the delayed (5 days post-SAH) phase and blunted SAH-induced sensorimotor deficits. Conclusions We demonstrate a novel association between NLRP3-mediated neuroinflammation and cerebrovascular dysfunction in both the early and delayed phases after SAH. MCC950 and other NLRP3 inhibitors could be promising tools in the development of therapeutics for EBI and DCI.
topic Subarachnoid hemorrhage
Microglia
NLRP3 inflammasome
Stroke
Inflammation
url https://doi.org/10.1186/s12974-021-02207-x
work_keys_str_mv AT williamsdodd nlrp3inhibitionattenuatesearlybraininjuryanddelayedcerebralvasospasmaftersubarachnoidhemorrhage
AT imaraynoda nlrp3inhibitionattenuatesearlybraininjuryanddelayedcerebralvasospasmaftersubarachnoidhemorrhage
AT melaniemartinez nlrp3inhibitionattenuatesearlybraininjuryanddelayedcerebralvasospasmaftersubarachnoidhemorrhage
AT kojihosaka nlrp3inhibitionattenuatesearlybraininjuryanddelayedcerebralvasospasmaftersubarachnoidhemorrhage
AT brianlhoh nlrp3inhibitionattenuatesearlybraininjuryanddelayedcerebralvasospasmaftersubarachnoidhemorrhage
_version_ 1721282909321560064