IL-1R1 signaling in TBI: assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed-head injury

Abstract Neuroinflammation contributes to secondary injury cascades following traumatic brain injury (TBI), with alternating waves of inflammation and resolution. Interleukin-1 (IL-1), a critical neuroinflammatory mediator originating from brain endothelial cells, microglia, astrocytes, and peripher...

Full description

Bibliographic Details
Main Authors: Jonathan C. Vincent, Colleen N. Garnett, James B. Watson, Emma K. Higgins, Teresa Macheda, Lydia Sanders, Kelly N. Roberts, Ryan K. Shahidehpour, Eric M. Blalock, Ning Quan, Adam D. Bachstetter
Format: Article
Language:English
Published: BMC 2023-10-01
Series:Journal of Neuroinflammation
Subjects:
Online Access:https://doi.org/10.1186/s12974-023-02934-3
_version_ 1797414431988121600
author Jonathan C. Vincent
Colleen N. Garnett
James B. Watson
Emma K. Higgins
Teresa Macheda
Lydia Sanders
Kelly N. Roberts
Ryan K. Shahidehpour
Eric M. Blalock
Ning Quan
Adam D. Bachstetter
author_facet Jonathan C. Vincent
Colleen N. Garnett
James B. Watson
Emma K. Higgins
Teresa Macheda
Lydia Sanders
Kelly N. Roberts
Ryan K. Shahidehpour
Eric M. Blalock
Ning Quan
Adam D. Bachstetter
author_sort Jonathan C. Vincent
collection DOAJ
description Abstract Neuroinflammation contributes to secondary injury cascades following traumatic brain injury (TBI), with alternating waves of inflammation and resolution. Interleukin-1 (IL-1), a critical neuroinflammatory mediator originating from brain endothelial cells, microglia, astrocytes, and peripheral immune cells, is acutely overexpressed after TBI, propagating secondary injury and tissue damage. IL-1 affects blood–brain barrier permeability, immune cell activation, and neural plasticity. Despite the complexity of cytokine signaling post-TBI, we hypothesize that IL-1 signaling specifically regulates neuroinflammatory response components. Using a closed-head injury (CHI) TBI model, we investigated IL-1's role in the neuroinflammatory cascade with a new global knock-out (gKO) mouse model of the IL-1 receptor (IL-1R1), which efficiently eliminates all IL-1 signaling. We found that IL-1R1 gKO attenuated behavioral impairments 14 weeks post-injury and reduced reactive microglia and astrocyte staining in the neocortex, corpus callosum, and hippocampus. We then examined whether IL-1R1 loss altered acute neuroinflammatory dynamics, measuring gene expression changes in the neocortex at 3, 9, 24, and 72 h post-CHI using the NanoString Neuroinflammatory panel. Of 757 analyzed genes, IL-1R1 signaling showed temporal specificity in neuroinflammatory gene regulation, with major effects at 9 h post-CHI. IL-1R1 signaling specifically affected astrocyte-related genes, selectively upregulating chemokines like Ccl2, Ccl3, and Ccl4, while having limited impact on cytokine regulation, such as Tnfα. This study provides further insight into IL-1R1 function in amplifying the neuroinflammatory cascade following CHI in mice and demonstrates that suppression of IL-1R1 signaling offers long-term protective effects on brain health.
first_indexed 2024-03-09T05:31:58Z
format Article
id doaj.art-1afd2662b1b249409d8005a54850aaee
institution Directory Open Access Journal
issn 1742-2094
language English
last_indexed 2024-03-09T05:31:58Z
publishDate 2023-10-01
publisher BMC
record_format Article
series Journal of Neuroinflammation
spelling doaj.art-1afd2662b1b249409d8005a54850aaee2023-12-03T12:31:11ZengBMCJournal of Neuroinflammation1742-20942023-10-0120111710.1186/s12974-023-02934-3IL-1R1 signaling in TBI: assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed-head injuryJonathan C. Vincent0Colleen N. Garnett1James B. Watson2Emma K. Higgins3Teresa Macheda4Lydia Sanders5Kelly N. Roberts6Ryan K. Shahidehpour7Eric M. Blalock8Ning Quan9Adam D. Bachstetter10Department of Neuroscience, University of KentuckyDepartment of Neuroscience, University of KentuckyDepartment of Neuroscience, University of KentuckyDepartment of Neuroscience, University of KentuckyDepartment of Neuroscience, University of KentuckyDepartment of Neuroscience, University of KentuckyDepartment of Neuroscience, University of KentuckyDepartment of Neuroscience, University of KentuckyDepartment of Pharmacology and Nutritional Sciences, University of KentuckyDepartment of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic UniversityDepartment of Neuroscience, University of KentuckyAbstract Neuroinflammation contributes to secondary injury cascades following traumatic brain injury (TBI), with alternating waves of inflammation and resolution. Interleukin-1 (IL-1), a critical neuroinflammatory mediator originating from brain endothelial cells, microglia, astrocytes, and peripheral immune cells, is acutely overexpressed after TBI, propagating secondary injury and tissue damage. IL-1 affects blood–brain barrier permeability, immune cell activation, and neural plasticity. Despite the complexity of cytokine signaling post-TBI, we hypothesize that IL-1 signaling specifically regulates neuroinflammatory response components. Using a closed-head injury (CHI) TBI model, we investigated IL-1's role in the neuroinflammatory cascade with a new global knock-out (gKO) mouse model of the IL-1 receptor (IL-1R1), which efficiently eliminates all IL-1 signaling. We found that IL-1R1 gKO attenuated behavioral impairments 14 weeks post-injury and reduced reactive microglia and astrocyte staining in the neocortex, corpus callosum, and hippocampus. We then examined whether IL-1R1 loss altered acute neuroinflammatory dynamics, measuring gene expression changes in the neocortex at 3, 9, 24, and 72 h post-CHI using the NanoString Neuroinflammatory panel. Of 757 analyzed genes, IL-1R1 signaling showed temporal specificity in neuroinflammatory gene regulation, with major effects at 9 h post-CHI. IL-1R1 signaling specifically affected astrocyte-related genes, selectively upregulating chemokines like Ccl2, Ccl3, and Ccl4, while having limited impact on cytokine regulation, such as Tnfα. This study provides further insight into IL-1R1 function in amplifying the neuroinflammatory cascade following CHI in mice and demonstrates that suppression of IL-1R1 signaling offers long-term protective effects on brain health.https://doi.org/10.1186/s12974-023-02934-3NeuroinflammationInterleukin-1Interleukin-1 receptor-1AstrocyteMicrogliaTraumatic brain injury
spellingShingle Jonathan C. Vincent
Colleen N. Garnett
James B. Watson
Emma K. Higgins
Teresa Macheda
Lydia Sanders
Kelly N. Roberts
Ryan K. Shahidehpour
Eric M. Blalock
Ning Quan
Adam D. Bachstetter
IL-1R1 signaling in TBI: assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed-head injury
Journal of Neuroinflammation
Neuroinflammation
Interleukin-1
Interleukin-1 receptor-1
Astrocyte
Microglia
Traumatic brain injury
title IL-1R1 signaling in TBI: assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed-head injury
title_full IL-1R1 signaling in TBI: assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed-head injury
title_fullStr IL-1R1 signaling in TBI: assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed-head injury
title_full_unstemmed IL-1R1 signaling in TBI: assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed-head injury
title_short IL-1R1 signaling in TBI: assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed-head injury
title_sort il 1r1 signaling in tbi assessing chronic impacts and neuroinflammatory dynamics in a mouse model of mild closed head injury
topic Neuroinflammation
Interleukin-1
Interleukin-1 receptor-1
Astrocyte
Microglia
Traumatic brain injury
url https://doi.org/10.1186/s12974-023-02934-3
work_keys_str_mv AT jonathancvincent il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT colleenngarnett il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT jamesbwatson il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT emmakhiggins il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT teresamacheda il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT lydiasanders il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT kellynroberts il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT ryankshahidehpour il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT ericmblalock il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT ningquan il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury
AT adamdbachstetter il1r1signalingintbiassessingchronicimpactsandneuroinflammatorydynamicsinamousemodelofmildclosedheadinjury