Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Neuropathol Commun ; 11(1): 134, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37596685

ABSTRACT

Accumulating evidence suggests that type I interferon (IFN-I) signaling is a key contributor to immune cell-mediated neuropathology in neurodegenerative diseases. Recently, we demonstrated a robust upregulation of type I interferon-stimulated genes in microglia and astrocytes following experimental traumatic brain injury (TBI). The specific molecular and cellular mechanisms by which IFN-I signaling impacts the neuroimmune response and neuropathology following TBI remains unknown. Using the lateral fluid percussion injury model (FPI) in adult male mice, we demonstrated that IFN α/ß receptor (IFNAR) deficiency resulted in selective and sustained blockade of type I interferon-stimulated genes following TBI as well as decreased microgliosis and monocyte infiltration. Molecular alteration of reactive microglia also occurred with diminished expression of genes needed for MHC class I antigen processing and presentation following TBI. This was associated with decreased accumulation of cytotoxic T cells in the brain. The IFNAR-dependent modulation of the neuroimmune response was accompanied by protection from secondary neuronal death, white matter disruption, and neurobehavioral dysfunction. These data support further efforts to leverage the IFN-I pathway for novel, targeted therapy of TBI.


Subject(s)
Brain Injuries, Traumatic , Interferon Type I , Male , Animals , Mice , Neuropathology , Brain Injuries, Traumatic/complications , Brain , Antibodies
2.
bioRxiv ; 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37333385

ABSTRACT

Accumulating evidence suggests that type I interferon (IFN-I) signaling is a key contributor to immune cell-mediated neuropathology in neurodegenerative diseases. Recently, we demonstrated a robust upregulation of type I interferon-stimulated genes in microglia and astrocytes following experimental traumatic brain injury (TBI). The specific molecular and cellular mechanisms by which IFN-I signaling impacts the neuroimmune response and neuropathology following TBI remains unknown. Using the lateral fluid percussion injury model (FPI) in adult male mice, we demonstrated that IFN α/ß receptor (IFNAR) deficiency resulted in selective and sustained blockade of type I interferon-stimulated genes following TBI as well as decreased microgliosis and monocyte infiltration. Phenotypic alteration of reactive microglia also occurred with diminished expression of molecules needed for MHC class I antigen processing and presentation following TBI. This was associated with decreased accumulation of cytotoxic T cells in the brain. The IFNAR-dependent modulation of the neuroimmune response was accompanied by protection from secondary neuronal death, white matter disruption, and neurobehavioral dysfunction. These data support further efforts to leverage the IFN-I pathway for novel, targeted therapy of TBI.

3.
J Neuroinflammation ; 18(1): 151, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34225752

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a leading cause of death and disability that lacks neuroprotective therapies. Following a TBI, secondary injury response pathways are activated and contribute to ongoing neurodegeneration. Microglia and astrocytes are critical neuroimmune modulators with early and persistent reactivity following a TBI. Although histologic glial reactivity is well established, a precise understanding of microglia and astrocyte function following trauma remains unknown. METHODS: Adult male C57BL/6J mice underwent either fluid percussion or sham injury. RNA sequencing of concurrently isolated microglia and astrocytes was conducted 7 days post-injury to evaluate cell-type-specific transcriptional responses to TBI. Dual in situ hybridization and immunofluorescence were used to validate the TBI-induced gene expression changes in microglia and astrocytes and to identify spatial orientation of cells expressing these genes. Comparative analysis was performed between our glial transcriptomes and those from prior reports in mild TBI and other neurologic diseases to determine if severe TBI induces unique states of microglial and astrocyte activation. RESULTS: Our findings revealed sustained, lineage-specific transcriptional changes in both microglia and astrocytes, with microglia showing a greater transcriptional response than astrocytes at this subacute time point. Microglia and astrocytes showed overlapping enrichment for genes related to type I interferon signaling and MHC class I antigen presentation. The microglia and astrocyte transcriptional response to severe TBI was distinct from prior reports in mild TBI and other neurodegenerative and neuroinflammatory diseases. CONCLUSION: Concurrent lineage-specific analysis revealed novel TBI-specific transcriptional changes; these findings highlight the importance of cell-type-specific analysis of glial reactivity following TBI and may assist with the identification of novel, targeted therapies.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/metabolism , Interferon Type I/biosynthesis , Microglia/metabolism , Transcriptome/physiology , Animals , Astrocytes/pathology , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/pathology , Interferon Type I/genetics , Male , Mice , Mice, Inbred C57BL , Microglia/pathology
4.
Invest Ophthalmol Vis Sci ; 61(12): 7, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33030508

ABSTRACT

Purpose: In a mouse model of blast-mediated traumatic brain injury (bTBI), interleukin-1 (IL-1)-pathway components were tested as potential therapeutic targets for bTBI-mediated retinal ganglion cell (RGC) dysfunction. Sex was also evaluated as a variable for RGC outcomes post-bTBI. Methods: Male and female mice with null mutations in genes encoding IL-1α, IL-1ß, or IL-1RI were compared to C57BL/6J wild-type (WT) mice after exposure to three 20-psi blast waves given at an interblast interval of 1 hour or to mice receiving sham injury. To determine if genetic blockade of IL-1α, IL-1ß, or IL-1RI could prevent damage to RGCs, the function and structure of these cells were evaluated by pattern electroretinogram and optical coherence tomography, respectively, 5 weeks following blast or sham exposure. RGC survival was also quantitatively assessed via immunohistochemical staining of BRN3A at the completion of the study. Results: Our results showed that male and female WT mice had a similar response to blast-induced retinal injury. Generally, constitutive deletion of IL-1α, IL-1ß, or IL-1RI did not provide full protection from the effects of bTBI on visual outcomes; however, injured WT mice had significantly worse visual outcomes compared to the injured genetic knockout mice. Conclusions: Sex does not affect RGC outcomes after bTBI. The genetic studies suggest that deletion of these IL-1 pathway components confers some protection, but global deletion from birth did not result in a complete rescue.


Subject(s)
Blast Injuries/physiopathology , Brain Injuries, Traumatic/physiopathology , Gene Expression Regulation/physiology , Interleukin-1/genetics , Retinal Ganglion Cells/physiology , Visual Acuity/physiology , Animals , Blast Injuries/metabolism , Brain Injuries, Traumatic/metabolism , Cell Survival/physiology , Electroretinography , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Sex Factors , Tomography, Optical Coherence , Transcription Factor Brn-3A/metabolism
5.
J Neurotrauma ; 37(12): 1463-1480, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32056479

ABSTRACT

The purpose of this study was to characterize acute changes in inflammatory pathways in the mouse eye after blast-mediated traumatic brain injury (bTBI) and to determine whether modulation of these pathways could protect the structure and function of retinal ganglion cells (RGC). The bTBI was induced in C57BL/6J male mice by exposure to three 20 psi blast waves directed toward the head with the body shielded, with an inter-blast interval of one hour. Acute cytokine expression in retinal tissue was measured through reverse transcription-quantitative polymerase chain reaction (RT-qPCR) four hours post-blast. Increased retinal expression of interleukin (lL)-1ß, IL-1α, IL-6, and tumor necrosis factor (TNF)α was observed in bTBI mice exposed to blast when compared with shams, which was associated with activation of microglia and macroglia reactivity, assessed via immunohistochemistry with ionized calcium binding adaptor molecule 1 and glial fibrillary acidic protein, respectively, one week post-blast. Blockade of the IL-1 pathway was accomplished using anakinra, an IL-1RI antagonist, administered intra-peritoneally for one week before injury and continuing for three weeks post-injury. Retinal function and RGC layer thickness were evaluated four weeks post-injury using pattern electroretinogram (PERG) and optical coherence tomography (OCT), respectively. After bTBI, anakinra treatment resulted in a preservation of RGC function and RGC structure when compared with saline treated bTBI mice. Optic nerve integrity analysis demonstrated a trend of decreased damage suggesting that IL-1 blockade also prevents axonal damage after blast. Blast exposure results in increased retinal inflammation including upregulation of pro-inflammatory cytokines and activation of resident microglia and macroglia. This may explain partially the RGC loss we observed in this model, as blockade of the acute inflammatory response after injury with the IL-1R1 antagonist anakinra resulted in preservation of RGC function and RGC layer thickness.


Subject(s)
Brain Injuries, Traumatic/immunology , Immunity/immunology , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Receptors, Interleukin-1/antagonists & inhibitors , Retina/immunology , Visual Perception/immunology , Animals , Blast Injuries/diagnostic imaging , Blast Injuries/drug therapy , Blast Injuries/immunology , Brain Injuries, Traumatic/diagnostic imaging , Brain Injuries, Traumatic/drug therapy , Electroretinography/methods , Immunity/drug effects , Interleukin 1 Receptor Antagonist Protein/pharmacology , Male , Mice , Mice, Inbred C57BL , Retina/diagnostic imaging , Retina/drug effects , Tomography, Optical Coherence/methods , Treatment Outcome , Visual Perception/drug effects
6.
J Neurotrauma ; 37(4): 635-646, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31621484

ABSTRACT

Traumatic brain injury (TBI) is a leading cause of death and disability that lacks targeted therapies. Successful translation of promising neuroprotective therapies will likely require more precise identification of target populations through greater study of crucial biological factors like age and sex. A growing body of work supports the impact of these factors on response to and recovery from TBI. However, age and sex are understudied in TBI animal models. The first aim of this study was to demonstrate the feasibility of lateral fluid percussion injury (FPI) in juvenile mice as a model of pediatric TBI. Subsequently, we were interested in examining the impact of young age and sex on TBI outcome. After adapting the lateral FPI model to 21-day-old male and female mice, we characterized the molecular, histological, and functional outcomes. Whereas similar tissue injury was observed in male and female juvenile mice exposed to TBI, we observed differences in neuroinflammation and neurobehavioral function. Overall, our findings revealed less acute inflammatory cytokine expression, greater subacute microglial/macrophage accumulation, and greater neurological recovery in juvenile male mice after TBI. Given that ongoing brain development may affect progression of and recovery from TBI, juvenile models are of critical importance. The sex-dependent differences we discovered after FPI support the necessity of also including this biological variable in future TBI studies. Understanding the mechanisms underlying age- and sex-dependent differences may result in the discovery of novel therapeutic targets for TBI.


Subject(s)
Brain Injuries/physiopathology , Inflammation/physiopathology , Macrophages/metabolism , Microglia/metabolism , Recovery of Function/physiology , Animals , Brain Injuries/metabolism , Cytokines/metabolism , Female , Inflammation/metabolism , Male , Mice , Models, Animal , Sex Factors
7.
J Neurogenet ; 32(4): 313-315, 2018 12.
Article in English | MEDLINE | ID: mdl-29790814

ABSTRACT

Homozygous recessive mutations in the PRICKLE1 gene were first described in three consanguineous families with myoclonic epilepsy. Subsequent studies have identified neurological abnormalities in humans and animal models with both heterozygous and homozygous mutations in PRICKLE1 orthologs. We describe a 7-year-old with a novel de novo missense mutation in PRICKLE1 associated with epilepsy, autism spectrum disorder and global developmental delay.


Subject(s)
Autism Spectrum Disorder/genetics , Epilepsies, Myoclonic/genetics , LIM Domain Proteins/genetics , Tumor Suppressor Proteins/genetics , Child , Developmental Disabilities/genetics , Humans , Intellectual Disability/genetics , Male , Mutation, Missense
8.
eNeuro ; 5(2)2018.
Article in English | MEDLINE | ID: mdl-29662944

ABSTRACT

Diffuse activation of interleukin-1 inflammatory cytokine signaling after traumatic brain injury (TBI) elicits progressive neurodegeneration and neuropsychiatric dysfunction, and thus represents a potential opportunity for therapeutic intervention. Although interleukin (IL)-1α and IL-1ß both activate the common type 1 IL-1 receptor (IL-1RI), they manifest distinct injury-specific roles in some models of neurodegeneration. Despite its potential relevance to treating patients with TBI, however, the individual contributions of IL-1α and IL-1ß to TBI-pathology have not been previously investigated. To address this need, we applied genetic and pharmacologic approaches in mice to dissect the individual contributions of IL-1α, IL-ß, and IL-1RI signaling to the pathophysiology of fluid percussion-mediated TBI, a model of mixed focal and diffuse TBI. IL-1RI ablation conferred a greater protective effect on brain cytokine expression and cognitive function after TBI than did individual IL-1α or IL-1ß ablation. This protective effect was recapitulated by treatment with the drug anakinra, a recombinant naturally occurring IL-1RI antagonist. Our data thus suggest that broad targeting of IL-1RI signaling is more likely to reduce neuroinflammation and preserve cognitive function after TBI than are approaches that individually target IL-1α or IL-1ß signaling.


Subject(s)
Brain Injuries, Traumatic , Cognitive Dysfunction/prevention & control , Inflammation/prevention & control , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Signal Transduction , Animals , Behavior, Animal/drug effects , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/metabolism , Cognitive Dysfunction/etiology , Cognitive Dysfunction/immunology , Cognitive Dysfunction/metabolism , Disease Models, Animal , Inflammation/etiology , Inflammation/immunology , Inflammation/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/deficiency , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/drug effects , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...