Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39000569

ABSTRACT

Regulation of neuroinflammation is critical for maintaining central nervous system (CNS) homeostasis and holds therapeutic promise in autoimmune diseases such as multiple sclerosis (MS). Previous studies have highlighted the significance of selective innate signaling in triggering anti-inflammatory mechanisms, which play a protective role in an MS-like disease, experimental autoimmune encephalomyelitis (EAE). However, the individual intra-CNS administration of specific innate receptor ligands or agonists, such as for toll-like receptor 7 (TLR7) and nucleotide-binding oligomerization-domain-containing protein 2 (NOD2), failed to elicit the desired anti-inflammatory response in EAE. In this study, we investigated the potential synergistic effect of targeting both TLR7 and NOD2 simultaneously to prevent EAE progression. Our findings demonstrate that simultaneous intrathecal administration of NOD2- and TLR7-agonists led to synergistic induction of Type I IFN (IFN I) and effectively suppressed EAE in an IFN I-dependent manner. Suppression of EAE was correlated with a significant decrease in the infiltration of monocytes, granulocytes, and natural killer cells, reduced demyelination, and downregulation of IL-1ß, CCL2, and IFNγ gene expression in the spinal cord. These results underscore the therapeutic promise of concurrently targeting the TLR7 and NOD2 pathways in alleviating neuroinflammation associated with MS, paving the way for novel and more efficacious treatment strategies.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Nod2 Signaling Adaptor Protein , Toll-Like Receptor 7 , Animals , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/agonists , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Mice , Mice, Inbred C57BL , Immunity, Innate/drug effects , Female , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/drug effects , Membrane Glycoproteins/metabolism , Interferon Type I/metabolism , Signal Transduction/drug effects
2.
Int J Mol Sci ; 23(19)2022 Sep 25.
Article in English | MEDLINE | ID: mdl-36232593

ABSTRACT

Type I interferons (IFN), including IFNß, play a protective role in multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Type I IFNs are induced by the stimulation of innate signaling, including via cytoplasmic RIG-I-like receptors. In the present study, we investigated the potential effect of a chimeric protein containing the key domain of RIG-I signaling in the production of CNS endogenous IFNß and asked whether this would exert a therapeutic effect against EAE. We intrathecally administered an adeno-associated virus vector (AAV) encoding a fusion protein comprising RIG-I 2CARD domains (C) and the first 200 amino acids of mitochondrial antiviral-signaling protein (MAVS) (M) (AAV-CM). In vivo imaging in IFNß/luciferase reporter mice revealed that a single intrathecal injection of AAV-CM resulted in dose-dependent and sustained IFNß expression within the CNS. IFNß expression was significantly increased for 7 days. Immunofluorescent staining in IFNß-YFP reporter mice revealed extraparenchymal CD45+ cells, choroid plexus, and astrocytes as sources of IFNß. Moreover, intrathecal administration of AAV-CM at the onset of EAE induced the suppression of EAE, which was IFN-I-dependent. These findings suggest that accessing the signaling pathway downstream of RIG-I represents a promising therapeutic strategy for inflammatory CNS diseases, such as MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Interferon Type I , Amino Acids , Animals , Antiviral Agents , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interferon Type I/metabolism , Interferon-beta/genetics , Interferon-beta/metabolism , Mice , Recombinant Fusion Proteins , Signal Transduction
3.
Int J Mol Sci ; 23(16)2022 Aug 11.
Article in English | MEDLINE | ID: mdl-36012246

ABSTRACT

Recently, the protective and/or pathological role of virus-specific T cells in SARS-CoV-2 infection has been the focus of many studies. We investigated the anti-spike IgG levels and SARS-CoV-2-specific T cells in 125 donors (90 vaccinated with four different vaccine platforms, 16 individuals with a previous natural infection, and 19 not vaccinated donors who did not report previous SARS-CoV-2 infections). Our data show that anti-spike IgG titers were similar between naturally infected subjects and those vaccinated with adenoviral vector vaccines. Of note, all immunized donors produced memory CD4+ and/or CD8+ T cells. A sustained polyfunctionality of SARS-CoV-2-specific T cells in all immunized donors was also demonstrated. Altogether, our data suggest that the natural infection produces an overall response like that induced by vaccination. Therefore, this detailed immunological evaluation may be relevant for other vaccine efforts especially for the monitoring of novel vaccines effective against emerging virus variants.


Subject(s)
COVID-19 , Viral Vaccines , Antibodies, Viral , CD8-Positive T-Lymphocytes , COVID-19/prevention & control , Humans , Immunoglobulin G , SARS-CoV-2 , Vaccination
4.
Glia ; 69(4): 943-953, 2021 04.
Article in English | MEDLINE | ID: mdl-33241604

ABSTRACT

Neuromyelitis optica (NMO) is an inflammatory disease of the central nervous system (CNS) most frequently mediated by serum autoantibodies against the water channel aquaporin 4, expressed on CNS astrocytes, resulting in primary astrocytopathy. There is no cure for NMO, and treatment with Type I interferon (IFNI)-IFNß is ineffective or even detrimental. We have previously shown that both NMO lesions and associated microglial activation were reduced in mice lacking the receptor for IFNß. However, the role of microglia in NMO is not well understood. In this study, we clarify the pathomechanism for IFNI dependence of and the role of microglia in experimental NMO. Transcriptome analysis showed a strong IFNI footprint in affected CNS tissue as well as in microglial subpopulations. Treatment with IFNß led to exacerbated pathology and further microglial activation as evidenced by expansion of a CD11c+ subset of microglia. Importantly, depletion of microglia led to suppression of pathology and decrease of IFNI signature genes. Our data show a pro-pathologic role for IFNI-activated microglia in NMO and open new perspectives for microglia-targeted therapies.


Subject(s)
Interferon Type I , Neuromyelitis Optica , Animals , Aquaporin 4 , Astrocytes , Mice , Microglia , Neuromyelitis Optica/drug therapy
5.
EMBO J ; 36(22): 3292-3308, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28963396

ABSTRACT

Microglia are resident macrophages of the central nervous system that contribute to homeostasis and neuroinflammation. Although known to play an important role in brain development, their exact function has not been fully described. Here, we show that in contrast to healthy adult and inflammation-activated cells, neonatal microglia show a unique myelinogenic and neurogenic phenotype. A CD11c+ microglial subset that predominates in primary myelinating areas of the developing brain expresses genes for neuronal and glial survival, migration, and differentiation. These cells are the major source of insulin-like growth factor 1, and its selective depletion from CD11c+ microglia leads to impairment of primary myelination. CD11c-targeted toxin regimens induced a selective transcriptional response in neonates, distinct from adult microglia. CD11c+ microglia are also found in clusters of repopulating microglia after experimental ablation and in neuroinflammation in adult mice, but despite some similarities, they do not recapitulate neonatal microglial characteristics. We therefore identify a unique phenotype of neonatal microglia that deliver signals necessary for myelination and neurogenesis.


Subject(s)
Brain/cytology , Brain/embryology , Microglia/metabolism , Myelin Sheath/metabolism , Neurogenesis , Aging/genetics , Animals , Animals, Newborn , Biomarkers/metabolism , Brain/ultrastructure , CD11c Antigen/metabolism , Cell Aggregation , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor I/metabolism , Mice, Inbred C57BL , Neural Plate/metabolism , Up-Regulation/genetics
6.
Scand J Immunol ; 92(5): e12963, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32851668

ABSTRACT

Myeloid cells represent the major cellular component of innate immune responses. Myeloid cells include monocytes and macrophages, granulocytes (neutrophils, basophils and eosinophils) and dendritic cells (DC). The role of myeloid cells has been broadly described both in physiological and in pathological conditions. All tissues or organs are equipped with resident myeloid cells, such as parenchymal microglia in the brain, which contribute to maintaining homeostasis. Moreover, in case of infection or tissue damage, other myeloid cells such as monocytes or granulocytes (especially neutrophils) can be recruited from the circulation, at first to promote inflammation and later to participate in repair and regeneration. This review aims to address the regulatory roles of myeloid cells in inflammatory diseases of the central nervous system (CNS), with a particular focus on recent work showing induction of suppressive function via stimulation of innate signalling in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE).


Subject(s)
Central Nervous System/immunology , Dendritic Cells/immunology , Granulocytes/immunology , Inflammation/immunology , Macrophages/immunology , Myeloid Cells/immunology , Animals , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology
7.
Mult Scler ; 26(10): 1187-1196, 2020 09.
Article in English | MEDLINE | ID: mdl-31287367

ABSTRACT

BACKGROUND: Neuromyelitis optica spectrum disorder (NMOSD) is a relapsing inflammatory central nervous system (CNS) disease for which there is no cure. Immunoglobulin G autoantibodies specific for the water channel aquaporin-4 are a serum biomarker, believed to induce complement-dependent astrocyte damage with secondary demyelination. OBJECTIVE: To investigate the effect of angiotensin AT2 receptor (AT2R) stimulation on NMOSD-like pathology and its underlying mechanism. METHODS: NMOSD-like pathology was induced in mice by intracerebral injection of immunoglobulin-G isolated from NMOSD patient serum, with complement. This mouse model produces the characteristic histological features of NMOSD. A specific AT2R agonist, Compound 21 (C21), was given intracerebrally at day 0 and by intrathecal injection at day 2. RESULTS: Loss of aquaporin-4 and glial fibrillary acidic protein was attenuated by treatment with C21. Administration of C21 induced mRNA for interleukin-10 in the brain. NMOSD-like pathology was exacerbated in interleukin-10-deficient mice, suggesting a protective role. C21 treatment did not attenuate NMOSD-like pathology in interleukin-10-deficient mice, indicating that the protective effect of AT2R stimulation was dependent on interleukin-10. CONCLUSION: Our findings identify AT2R as a novel potential therapeutic target for the treatment of NMOSD. Interleukin-10 signaling is an essential part of the protective mechanism counteracting NMOSD pathology.


Subject(s)
Neuromyelitis Optica , Animals , Aquaporin 4/genetics , Autoantibodies , Humans , Interleukin-10 , Mice , Neoplasm Recurrence, Local , Neuromyelitis Optica/drug therapy , Receptor, Angiotensin, Type 2
8.
J Neuroinflammation ; 15(1): 110, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29665816

ABSTRACT

BACKGROUND: Encounter of autoantibodies with specific antigens can lead to hypersensitivity reactions and pathology. In multiple sclerosis and neuromyelitis optica spectrum disease (NMOSD), immunoglobulin-G (IgG) deposition has been observed in pathological lesions in the central nervous system. The paradigmatic autoantibodies in NMOSD are specific for the water channel aquaporin-4, localized to astrocytic end-feet at the blood-brain barrier and ependymal cells at the cerebrospinal fluid-brain barrier. We have previously observed that IgG antibodies from NMO patients (NMO-IgG) access brain parenchyma from the cerebrospinal fluid and induce subpial and periventricular NMO-like lesions and blood-brain barrier breakdown, in a complement-dependent manner. OBJECTIVE: To investigate how IgG trafficking from cerebrospinal fluid to brain parenchyma can be influenced by injury. METHODS: IgG from healthy donors was intrathecally injected into the cerebrospinal fluid via cisterna magna at 1, 2, 4, or 7 days after a distal stereotactic sterile needle insertion to the striatum. RESULTS: Antibody deposition, detected by staining for human IgG, peaked 1 day after the intrathecal injection and was selectively seen close to the needle insertion. When NMO-IgG was intrathecally injected, we observed complement-dependent NMO-like pathology (loss of aquaporin-4 and glial fibrillary acidic protein) proximal to the insertion site, with similar kinetics. A fluorescent tracer did not show the same distribution indicating IgG-selective localization. CONCLUSION: These findings suggest that IgG from cerebrospinal fluid localize selectively in brain parenchyma at the site of injury and pathogenic NMO-IgG induce astrocyte pathology at the same location.


Subject(s)
Central Nervous System/metabolism , Immunoglobulin G/cerebrospinal fluid , Parenchymal Tissue/metabolism , Animals , Aquaporin 4/immunology , Aquaporin 4/metabolism , Central Nervous System/anatomy & histology , Cytokines/genetics , Cytokines/metabolism , Dextrans/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Immunoglobulin G/administration & dosage , Mice , Mice, Inbred C57BL , Neuromyelitis Optica/metabolism , Neuromyelitis Optica/pathology , Protein Transport/drug effects , Protein Transport/physiology , RNA, Messenger/metabolism , Time Factors
9.
J Neuroinflammation ; 14(1): 127, 2017 06 24.
Article in English | MEDLINE | ID: mdl-28646890

ABSTRACT

BACKGROUND: Antibodies with specificity for myelin oligodendrocyte glycoprotein (MOG) are implicated in multiple sclerosis and related diseases. The pathogenic importance of anti-MOG antibody in primary demyelinating pathology remains poorly characterized. OBJECTIVE: The objective of this study is to investigate whether administration of anti-MOG antibody would be sufficient for demyelination and to determine if type I interferon (IFN) signaling plays a similar role in anti-MOG antibody-mediated pathology, as has been shown for neuromyelitis optica-like pathology. METHODS: Purified IgG2a monoclonal anti-MOG antibody and mouse complement were stereotactically injected into the corpus callosum of wild-type and type I IFN receptor deficient mice (IFNAR1-KO) with and without pre-established experimental autoimmune encephalomyelitis (EAE). RESULTS: Anti-MOG induced complement-dependent demyelination in the corpus callosum of wild-type mice and did not occur in mice that received control IgG2a. Deposition of activated complement coincided with demyelination, and this was significantly reduced in IFNAR1-KO mice. Co-injection of anti-MOG and complement at onset of symptoms of EAE induced similar levels of callosal demyelination in wild-type and IFNAR1-KO mice. CONCLUSIONS: Anti-MOG antibody and complement was sufficient to induce callosal demyelination, and pathology was dependent on type I IFN. Induction of EAE in IFNAR1-KO mice overcame the dependence on type I IFN for anti-MOG and complement-mediated demyelination.


Subject(s)
Autoantibodies/metabolism , Demyelinating Diseases/metabolism , Interferon Type I/metabolism , Myelin-Oligodendrocyte Glycoprotein/metabolism , Animals , Autoantibodies/immunology , Corpus Callosum/immunology , Corpus Callosum/metabolism , Corpus Callosum/pathology , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Interferon Type I/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/immunology
10.
J Neuroinflammation ; 14(1): 204, 2017 Oct 16.
Article in English | MEDLINE | ID: mdl-29037246

ABSTRACT

BACKGROUND: Autoreactive Th1 and Th17 cells are believed to mediate the pathology of multiple sclerosis in the central nervous system (CNS). Their interaction with microglia and astrocytes in the CNS is crucial for the regulation of the neuroinflammation. Previously, we have shown that only Th1 but not Th17 effectors activate microglia. However, it is not clear which cells are targets of Th17 effectors in the CNS. METHODS: To understand the effects driven by Th17 cells in the CNS, we induced experimental autoimmune encephalomyelitis in wild-type mice and CD4+ T cell-specific integrin α4-deficient mice where trafficking of Th1 cells into the CNS was affected. We compared microglial and astrocyte response in the brain and spinal cord of these mice. We further treated astrocytes with supernatants from highly pure Th1 and Th17 cultures and assessed the messenger RNA expression of neurotrophic factors, cytokines and chemokines, using real-time PCR. Data obtained was analyzed using the Kruskal-Wallis test. RESULTS: We observed in α4-deficient mice weak microglial activation but comparable astrogliosis to that of wild-type mice in the regions of the brain populated with Th17 infiltrates, suggesting that Th17 cells target astrocytes and not microglia. In vitro, in response to supernatants from Th1 and Th17 cultures, astrocytes showed altered expression of neurotrophic factors, pro-inflammatory cytokines and chemokines. Furthermore, increased expression of chemokines in Th1- and Th17-treated astrocytes enhanced recruitment of microglia and transendothelial migration of Th17 cells in vitro. CONCLUSION: Our results demonstrate the delicate interaction between T cell subsets and glial cells and how they communicate to mediate their effects. Effectors of Th1 act on both microglia and astrocytes whereas Th17 effectors preferentially target astrocytes to promote neuroinflammation.


Subject(s)
Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gliosis/metabolism , Inflammation Mediators/metabolism , Th1 Cells/metabolism , Th17 Cells/metabolism , Animals , Astrocytes/pathology , Cell Movement/physiology , Cells, Cultured , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Gliosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Th1 Cells/pathology , Th17 Cells/pathology
11.
J Neuroinflammation ; 12: 211, 2015 Nov 18.
Article in English | MEDLINE | ID: mdl-26581581

ABSTRACT

BACKGROUND: Interferon (IFN)-ß exerts anti-inflammatory effects, coupled to remarkable neurological improvements in multiple sclerosis, a neuroinflammatory condition of the central nervous system. Analogously, it has been hypothesized that IFN-ß, by limiting inflammation, decreases neuronal death and promotes functional recovery after stroke. However, the core actions of endogenous IFN-ß signaling in stroke are unclear. METHODS: To address this question, we used two clinically relevant models of focal cerebral ischemia, transient and permanent middle cerebral artery occlusion, and two genetically modified mouse lines, lacking either IFN-ß or its receptor, the IFN-α/ß receptor. Subsets of inflammatory and immune cells isolated from the brain, blood, and spleen were studied using flow cytometry. Sensorimotor deficits were assessed by a modified composite neuroscore, the rotating pole and grip strength tests, and cerebral infarct volumes were given by lack of neuronal nuclei immunoreactivity. RESULTS: Here, we report alterations in local and systemic inflammation in IFN-ß knockout (IFN-ßKO) mice over 8 days after induction of focal cerebral ischemia. Notably, IFN-ßKO mice showed a higher number of infiltrating leukocytes in the brain 2 days after stroke. Concomitantly, in the blood of IFN-ßKO mice, we found a higher percentage of total B cells but a similar percentage of mature and activated B cells, collectively indicating a higher proliferation rate. The additional differential regulation of circulating cytokines and splenic immune cell populations in wild-type and IFN-ßKO mice further supports an important immunoregulatory function of IFN-ß in stroke. Moreover, we observed a significant weight loss 2-3 days and a reduction in grip strength 2 days after stroke in the IFN-ßKO group, while endogenous IFN-ß signaling did not affect the infarct volume. CONCLUSIONS: We conclude that endogenous IFN-ß signaling attenuates local inflammation, regulates peripheral immune cells, and, thereby, may contribute positively to stroke outcome.


Subject(s)
Brain Ischemia/pathology , Inflammation/pathology , Interferon-beta , Stroke/pathology , Animals , B-Lymphocytes/pathology , Brain/pathology , Brain Ischemia/psychology , Cytokines/blood , Hand Strength/physiology , Infarction, Middle Cerebral Artery/pathology , Interferon-beta/genetics , Ischemic Attack, Transient/pathology , Leukocytes/pathology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Postural Balance , Receptors, Interferon/genetics , Spleen/cytology , Spleen/immunology , Stroke/psychology
12.
Acta Neuropathol ; 130(1): 107-18, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25869642

ABSTRACT

The Type I interferons (IFN), beta (IFN-ß) and the alpha family (IFN-α), act through a common receptor and have anti-inflammatory effects. IFN-ß is used to treat multiple sclerosis (MS) and is effective against experimental autoimmune encephalomyelitis (EAE), an animal model for MS. Mice with EAE show elevated levels of Type I IFNs in the central nervous system (CNS), suggesting a role for endogenous Type I IFN during inflammation. However, the therapeutic benefit of Type I IFN produced in the CNS remains to be established. The aim of this study was to examine whether experimentally induced CNS-endogenous Type I IFN influences EAE. Using IFN-ß reporter mice, we showed that direct administration of polyinosinic-polycytidylic acid (poly I:C), a potent inducer of IFN-ß, into the cerebrospinal fluid induced increased leukocyte numbers and transient upregulation of IFN-ß in CD45/CD11b-positive cells located in the meninges and choroid plexus, as well as enhanced IFN-ß expression by parenchymal microglial cells. Intrathecal injection of poly I:C to mice showing first symptoms of EAE substantially increased the normal disease-associated expression of IFN-α, IFN-ß, interferon regulatory factor-7 and IL-10 in CNS, and disease worsening was prevented for as long as IFN-α/ß was expressed. In contrast, there was no therapeutic effect on EAE in poly I:C-treated IFN receptor-deficient mice. IFN-dependent microglial and astrocyte response included production of the chemokine CXCL10. These results show that Type I IFN induced within the CNS can play a protective role in EAE and highlight the role of endogenous type I IFN in mediating neuroprotection.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interferon-alpha/metabolism , Interferon-beta/metabolism , Neuroprotective Agents/pharmacology , Poly I-C/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/pathology , Brain/drug effects , Brain/immunology , Brain/pathology , Chemokine CXCL10/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interferon-alpha/genetics , Interferon-beta/genetics , Leukocytes/drug effects , Leukocytes/pathology , Leukocytes/physiology , Meninges/drug effects , Meninges/immunology , Meninges/pathology , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Microglia/pathology , Microglia/physiology , Random Allocation , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology
13.
Glia ; 62(3): 339-55, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24588027

ABSTRACT

Interferons (IFNs) are implicated as an important component of the innate immune system influencing viral infections, inflammation, and immune surveillance. We review here the complex biological activity of IFNs in the central nervous system (CNS) and associated glial­immune interactions, with focus specifically on the Type I IFNs in physiological and pathological conditions. IFN-α and IFN-ß are the predominant Type I IFNs in the CNS. They are produced in the CNS by glial cells, mostly microglia and astrocytes, as well as by neurons. A variety of mechanisms stimulate IFN production in glial cells, including innate stimuli from Toll-like and other receptors, which can recognize endogenous entities, as well as pathogens. We will review evidence that differential signaling by IFN-α versus IFN-ß through the common heterodimeric receptor IFNAR is the basis for CNS-selective Type I IFN response, and the capacity of CNS glial cells to produce and respond to Type I IFN. Differential signaling outcomes of IFN-α and IFN-ß, which have been ascribed to differential affinity for IFNAR1 and IFNAR2, determine whether Type I IFN exert pathogenic or protective roles in the CNS. These points will be discussed with reference to selected neurological diseases, and effects of Type I IFN on the integrity of the blood­brain barrier.


Subject(s)
Central Nervous System , Immune System/physiology , Interferons/metabolism , Neuroglia/physiology , Animals , Central Nervous System/cytology , Central Nervous System/immunology , Central Nervous System/metabolism , Humans , Interferons/therapeutic use , Nervous System Diseases/drug therapy , Nervous System Diseases/immunology
14.
Mult Scler ; 20(10): 1312-21, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24552747

ABSTRACT

BACKGROUND: Myelin-specific T cells are implicated in multiple sclerosis (MS) and drive experimental autoimmune encephalomyelitis (EAE). EAE is commonly induced with short peptides, whereas in MS, whole myelin proteins are available for immune response. We asked whether immunization with the immunoglobulin-like domain of myelin oligodendrocyte glycoprotein (MOG(Igd), residues 1-125) might induce distinct CD4+ T-cell response and/or a stronger CD8+ T-cell response, compared to the 21 amino acid immunodominant MHC II-associating peptide (p35-55). OBJECTIVES: Compare both EAE and T-cell responses in C57BL/6 mice immunized with MOG(Igd) and MOG p35-55. METHODS: Cytokine production, and chemokine receptor expression by CD4+ and CD8+ T cells in the mouse central nervous system (CNS), were analyzed by flow cytometry. RESULTS: MOG(Igd) triggered progression to more severe EAE than MOG p35-55, despite similar time of onset and overall incidence. EAE in MOG(Igd)-immunized mice was characterized by an increased percentage of CXCR3+ interferon-γ-producing CD4+ T cells in CNS. The CD8+ T-cell response to both immunogens was similar. CONCLUSIONS: Increased incidence of severe disease following MOG(Igd) immunization, accompanied by an increased percentage of CD4+ T cells in the CNS expressing CXCR3 and producing interferon-γ, identifies a pathogenic role for interferon-γ that is not seen when disease is induced with a single Major Histocompatibility Complex (MHC) II-associating epitope.


Subject(s)
Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/immunology , Receptors, CXCR3/immunology , Th1 Cells/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Central Nervous System/metabolism , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/metabolism , Epitopes , Female , Immunization , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Rats , Receptors, CXCR3/metabolism , Severity of Illness Index , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Time Factors
15.
J Immunol ; 185(2): 1258-64, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20562259

ABSTRACT

Innate glial response is critical for the induction of inflammatory mediators and recruitment of leukocytes to sites of the injury in the CNS. We have examined the involvement of type I IFN signaling in the mouse hippocampus following sterile injury (transection of entorhinal afferents). Type I IFNs signal through a receptor (IFNAR), which involves activation of IFN regulatory factor (IRF)9, leading to the induction of IFN-stimulated genes including IRF7, that in turn enhances the induction of type I IFN. Axonal transection induced upregulation of IRF7 and IRF9 in hippocampus. Induction of IRF7 and IRF9 mRNAs was IFNAR dependent. Double-labeling immunofluorescence showed that IRF7 selectively was induced in Mac-1/CD11b(+) macrophages/microglia in hippocampus after axonal transection. IRF7 mRNA was also detected in microglia sorted by flow cytometry. Lack of type I IFN signaling resulted in increased leukocyte infiltration into the lesion-reactive hippocampus. Axonal lesion-induced CXCL10 gene expression was abrogated, whereas matrix metalloproteinase 9 mRNA was elevated in IFNAR-deficient mice. Our findings point to a role for type I IFN signaling in regulation of CNS response to sterile injury.


Subject(s)
Central Nervous System/immunology , Inflammation/immunology , Interferon Type I/immunology , Signal Transduction/immunology , Animals , Blotting, Western , Brain Injuries/immunology , CD11b Antigen/analysis , Central Nervous System/metabolism , Central Nervous System/pathology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Female , Glial Fibrillary Acidic Protein/analysis , Hippocampus/injuries , Hippocampus/metabolism , Immunohistochemistry , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Regulatory Factor-7/metabolism , Leukocytes/immunology , Leukocytes/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptor, Interferon alpha-beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , STAT1 Transcription Factor/metabolism
16.
J Neuroinflammation ; 8: 181, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22196084

ABSTRACT

BACKGROUND: Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) with unknown etiology. Interferon-ß (IFN-ß), a member of the type I IFN family, is used as a therapeutic for MS and the IFN signaling pathway is implicated in MS susceptibility. Interferon regulatory factor 7 (IRF7) is critical for the induction and positive feedback regulation of type I IFN. To establish whether and how endogenous type I IFN signaling contributes to disease modulation and to better understand the underlying mechanism, we examined the role of IRF7 in the development of MS-like disease in mice. METHODS: The role of IRF7 in development of EAE was studied by immunizing IRF7-KO and C57BL/6 (WT) mice with myelin oligodendrocyte glycoprotein using a standard protocol for the induction of EAE. We measured leukocyte infiltration and localization in the CNS using flow cytometric analysis and immunohistochemical procedures. We determined levels of CD3 and selected chemokine and cytokine gene expression by quantitative real-time PCR. RESULTS: IRF7 gene expression increased in the CNS as disease progressed. IRF7 message was localized to microglia and infiltrating leukocytes. Furthermore, IRF7-deficient mice developed more severe disease. Flow cytometric analysis showed that the extent of leukocyte infiltration into the CNS was higher in IRF7-deficient mice with significantly higher number of infiltrating macrophages and T cells, and the distribution of infiltrates within the spinal cord was altered. Analysis of cytokine and chemokine gene expression by quantitative real-time PCR showed significantly greater increases in CCL2, CXCL10, IL-1ß and IL17 gene expression in IRF7-deficient mice compared with WT mice. CONCLUSION: Together, our findings suggest that IRF7 signaling is critical for regulation of inflammatory responses in the CNS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interferon Regulatory Factor-7/immunology , Interferon-beta/immunology , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , Chemokines/immunology , Cytokines/immunology , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Interferon Regulatory Factor-7/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Spinal Cord/pathology , Spinal Cord/physiology
17.
Front Neurosci ; 15: 682451, 2021.
Article in English | MEDLINE | ID: mdl-34149350

ABSTRACT

The pathological hallmark of multiple sclerosis (MS) is the formation of multifocal demyelinating lesions in the central nervous system (CNS). Stimulation of innate receptors has been shown to suppress experimental autoimmune encephalomyelitis (EAE), an MS-like disease in mice. Specifically, targeting Toll-like receptor 9 (TLR9) and NOD-like receptor 2 (NOD2) significantly reduced disease severity. In the present work we have developed a novel focal EAE model to further study the effect of innate signaling on demyelinating pathology. Focal lesions were induced by stereotactic needle insertion into the corpus callosum (CC) of mice previously immunized for EAE. This resulted in focal pathology characterized by infiltration and demyelination in the CC. We find that intrathecal delivery of MIS416, a TLR9 and NOD2 bispecific innate ligand, into the cerebrospinal fluid reduced focal lesions in the CC. This was associated with upregulation of type I and II interferons, interleukin-10, arginase-1, CCL-2 and CXCL-10. Analysis of draining cervical lymph nodes showed upregulation of type II interferons and interleukin 10. Moreover, intrathecal MIS416 altered the composition of early CNS infiltrates, increasing proportions of myeloid and NK cells and reducing T cells at the lesion site. This study contributes to an increased understanding of how innate immune responses can play a protective role, which in turn may lead to additional therapeutic strategies for the prevention and treatment of demyelinating pathologies.

18.
Front Neurol ; 12: 703249, 2021.
Article in English | MEDLINE | ID: mdl-34367056

ABSTRACT

Background: Optic neuritis (ON) is a common inflammatory optic neuropathy, which often occurs in neuromyelitis optica spectrum disease (NMOSD). An experimental model of NMOSD-ON may provide insight into disease mechanisms. Objective: To examine the pathogenicity of autoantibodies targeting the astrocyte water channel aquaporin-4 [aquaporin-4 (AQP4)-immunoglobulin G (AQP4-IgG)] in the optic nerve. Materials and Methods: Purified IgG from an AQP4-IgG-positive NMOSD-ON patient was together with human complement (C) given to wild-type (WT) and type I interferon (IFN) receptor-deficient mice (IFNAR1-KO) as two consecutive intrathecal injections into cerebrospinal fluid via cisterna magna. The optic nerves were isolated, embedded in paraffin, cut for histological examination, and scored semi-quantitatively in a blinded fashion. In addition, optic nerves were processed to determine selected gene expression by quantitative real-time PCR. Results: Intrathecal injection of AQP4-IgG+C induced astrocyte pathology in the optic nerve with loss of staining for AQP4 and glial fibrillary acidic protein (GFAP), deposition of C, and demyelination, as well as upregulation of gene expression for interferon regulatory factor-7 (IRF7) and CXCL10. Such pathology was not seen in IFNAR1-KO mice nor in control mice. Conclusion: We describe induction of ON in an animal model for NMOSD and show a requirement for type I IFN signaling in the disease process.

19.
J Neuroimmunol ; 358: 577657, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34315069

ABSTRACT

Astrocyte pathology is a feature of neuromyelitis optica spectrum disorder (NMOSD) pathology. Recently mitochondrial dysfunction and metabolic changes were suggested to play a role in NMOSD. To elucidate the role of mitochondrial dysfunction, astrocyte pathology was induced in C57BL/6 J female mice by intracerebral injection of aquaporin-4-immunoglobulin G from an NMOSD patient, together with complement. Etomoxir has been shown to cause mitochondrial dysfunction. Etomoxir was delivered to the central nervous system and resulted in decreased astrocyte pathology. The ameliorating effect was associated with increases in different acylcarnitines and amino acids. This suggests that mitochondria may be a therapeutic target in NMOSD.


Subject(s)
Astrocytes/immunology , Astrocytes/pathology , Autoantibodies/immunology , Epoxy Compounds/administration & dosage , Mitochondria/immunology , Animals , Astrocytes/drug effects , Female , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Neuromyelitis Optica/immunology
20.
Mult Scler Relat Disord ; 53: 103033, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34090131

ABSTRACT

BACKGROUND: Neuromyelitis optica spectrum disorder (NMOSD) is an antibody-mediated autoimmune inflammatory disease of the central nervous system (CNS), resulting in primary astrocytopathy. We have previously shown that Angiotensin AT2-receptor (AT2R) stimulation with the specific agonist Compound 21 (C21) attenuated NMOSD-like pathology. Recent studies have proposed that the mechanism behind protective effects of AT2R includes induction of brain derived neurotrophic factor (BDNF). Astrocytes are a major cellular source of BDNF. In this study we used mice with conditional BDNF deficiency in astrocytes (GfapF) to examine the involvement of astrocyte-derived BDNF in NMOSD-like pathology and in mediating the protective effect of AT2R stimulation. METHODS: Anti-aquaporin-4 IgG (AQP4-IgG) from an NMOSD patient and human complement (C) were co-injected intrastriatally to GfapF and wildtype littermate BDNFfl/fl mice (WT), together with either C21 or vehicle at day 0, followed by intrathecal injection of C21 or vehicle at day 2 and tissue collection at day 4. RESULTS: Intracerebral/intrathecal injection of C21, alone or in combination with AQP4-IgG + C, induced BDNF expression in WT mice. Injection of AQP4-IgG + C induced NMOSD-like pathology, including loss of AQP4 and GFAP. There was no difference in the severity of pathological changes between GfapF and WT mice. C21 treatment significantly and equally ameliorated NMOSD-like pathology in both WT and GfapF mice. CONCLUSION: Our findings indicate that astrocyte-derived BDNF neither reduces the severity of NMOSD-like pathology nor is it necessary for the protective effect of AT2R stimulation in NMOSD-like pathology.


Subject(s)
Neuromyelitis Optica , Angiotensins , Animals , Aquaporin 4/genetics , Astrocytes , Autoantibodies , Brain-Derived Neurotrophic Factor , Humans , Mice , Neuromyelitis Optica/drug therapy , Receptor, Angiotensin, Type 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL