myelin-oligodendrocyte-glycoprotein-(35-55) has been researched along with Chronic-Disease* in 10 studies
10 other study(ies) available for myelin-oligodendrocyte-glycoprotein-(35-55) and Chronic-Disease
Article | Year |
---|---|
The Beneficial Effect of Human Amnion Mesenchymal Cells in Inhibition of Inflammation and Induction of Neuronal Repair in EAE Mice.
Multiple sclerosis (MS) is a chronic inflammatory autoimmune disease of the central nervous system (CNS). Currently, there is still lack of curative treatment for MS. Mesenchymal stem cell- (MSC-) based therapy is recently the subject of intense interest in autoimmune diseases. Here, we investigated the therapeutic effect and potential mechanism of human amnion mesenchymal cells (hAMC) on inflammation and remyelination in experimental autoimmune encephalomyelitis (EAE) mice. C57BL/6 mice were immunized with myelin oligodendrocyte glycoprotein (MOG) 35-55 peptide. hAMC were injected intraperitoneal when EAE was successfully established. The results demonstrated that application of hAMC significantly ameliorated the disease severity and histopathological changes in EAE mice. The production of proinflammatory cytokines such as IFN- Topics: Animals; Cells, Cultured; Chronic Disease; Cytokines; Disease Models, Animal; Disease Progression; Encephalomyelitis, Autoimmune, Experimental; Female; Humans; Inflammation; Inflammation Mediators; Mesenchymal Stem Cells; Mice; Mice, Inbred C57BL; Multiple Sclerosis; Myelin-Oligodendrocyte Glycoprotein; Neurons; Peptide Fragments; Wound Healing | 2018 |
Connexin 30 Deficiency Attenuates Chronic but Not Acute Phases of Experimental Autoimmune Encephalomyelitis Through Induction of Neuroprotective Microglia.
Glial connexins (Cxs) form gap junction channels through which a pan-glial network plays key roles in maintaining homeostasis of the central nervous system (CNS). In multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), expression of astrocytic Cx43 is lost in acute lesions but upregulated in chronic plaques, while astrocytic Cx30 is very low in normal white matter and changes in its expression have not been convincingly shown. In Cx30 or Cx43 single knockout (KO) mice and even in Cx30/Cx43 double KO mice, acute EAE is unaltered. However, the effects of Cx30/Cx43 deficiency on chronic EAE remains to be elucidated. We aimed to clarify the roles of Cx30 in chronic neuroinflammation by studying EAE induced by myelin oligodendrocyte glycoprotein peptide 35-55 in Cx30 KO mice. We found that Cx30 deficiency improved the clinical symptoms and demyelination of chronic but not acute EAE without influencing CD3 Topics: Acute Disease; Animals; Arginase; Brain-Derived Neurotrophic Factor; Chronic Disease; Connexin 30; Connexin 43; Disease Models, Animal; Encephalomyelitis, Autoimmune, Experimental; Female; Humans; Mice; Mice, Inbred C57BL; Mice, Knockout; Microglia; Multiple Sclerosis; Myelin-Oligodendrocyte Glycoprotein; Neuroprotection; Peptide Fragments | 2018 |
Low dose zymosan ameliorates both chronic and relapsing experimental autoimmune encephalomyelitis.
Zymosan has previously been reported to have both pro-inflammatory and anti-inflammatory effects. Here we demonstrate that low dose zymosan prevented or reversed chronic and relapsing paralysis in EAE. In suppressing CNS autoimmune inflammation, zymosan not only regulated APC costimulator and MHC class II expression, but also promoted differentiation of regulatory T cells. Following adoptive transfer of zymosan-primed CD4(+) T cells, recipient mice were protected from EAE. In contrast, a MAPK inhibitor and a blocker of β-glucan, reversed the effects of zymosan. These results demonstrate that zymosan may be a promising beneficial agent for Multiple Sclerosis (MS). Topics: Adoptive Transfer; Animals; Anti-Inflammatory Agents, Non-Steroidal; Antigens, CD; Butadienes; CD4-Positive T-Lymphocytes; Cell Differentiation; Cell Line, Transformed; Chronic Disease; Coculture Techniques; Cytokines; Dose-Response Relationship, Drug; Encephalomyelitis, Autoimmune, Experimental; Enzyme Inhibitors; Female; Flow Cytometry; Gene Expression; Interferon-alpha; Lipopolysaccharides; Macrophages; Mice; Mice, Inbred Strains; Mice, Transgenic; Microglia; Myelin Proteolipid Protein; Myelin-Oligodendrocyte Glycoprotein; Nitriles; Peptide Fragments; Receptors, Antigen, T-Cell; Secondary Prevention; Severity of Illness Index; T-Lymphocytes, Regulatory; Time Factors; Tritium; Zymosan | 2013 |
Suppression of inflammatory responses during myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis is regulated by AKT3 signaling.
AKT3, a member of the serine/threonine kinase AKT family, is involved in a variety of biologic processes. AKT3 is expressed in immune cells and is the major AKT isoform in the CNS representing 30% of the total AKT expressed in spinal cord, and 50% in the brain. Myelin-oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE) is a mouse model in which lymphocytes and monocytes enter the CNS, resulting in inflammation, demyelination, and axonal injury. We hypothesized that during EAE, deletion of AKT3 would negatively affect the CNS of AKT3(-/-) mice, making them more susceptible to CNS damage. During acute EAE, AKT3(-/-)mice were more severely affected than wild type (WT) mice. Evaluation of spinal cords showed that during acute and chronic disease, AKT3(-/-) spinal cords had more demyelination compared with WT spinal cords. Quantitative RT-PCR determined higher levels of IL-2, IL-17, and IFN-γ mRNA in spinal cords from AKT3(-/-) mice than WT. Experiments using bone marrow chimeras demonstrated that AKT3(-/-) mice receiving AKT3-deficient bone marrow cells had elevated clinical scores relative to control WT mice reconstituted with WT cells, indicating that altered function of both CNS cells and bone marrow-derived immune cells contributed to the phenotype. Immunohistochemical analysis revealed decreased numbers of Foxp3(+) regulatory T cells in the spinal cord of AKT3(-/-) mice compared with WT mice, whereas in vitro suppression assays showed that AKT3-deficient Th cells were less susceptible to regulatory T cell-mediated suppression than their WT counterparts. These results indicate that AKT3 signaling contributes to the protection of mice against EAE. Topics: Acute Disease; Animals; Chronic Disease; Encephalomyelitis, Autoimmune, Experimental; Genetic Predisposition to Disease; Inflammation Mediators; Lumbosacral Region; Male; Mice; Mice, Inbred C57BL; Mice, Knockout; Myelin-Oligodendrocyte Glycoprotein; Peptide Fragments; Proto-Oncogene Proteins c-akt; Severity of Illness Index; Signal Transduction; Spinal Cord | 2013 |
Leptin modulates the survival of autoreactive CD4+ T cells through the nutrient/energy-sensing mammalian target of rapamycin signaling pathway.
Chronic inflammation can associate with autoreactive immune responses, including CD4(+) T cell responses to self-Ags. In this paper, we show that the adipocyte-derived proinflammatory hormone leptin can affect the survival and proliferation of autoreactive CD4(+) T cells in experimental autoimmune encephalomyelitis, an animal model of human multiple sclerosis. We found that myelin olygodendrocyte glycoprotein peptide 35-55 (MOG(35-55))-specific CD4(+) T cells from C57BL/6J wild-type mice could not transfer experimental autoimmune encephalomyelitis into leptin-deficient ob/ob mice. Such a finding was associated with a reduced proliferation of the transferred MOG(35-55)-reactive CD4(+) T cells, which had a reduced degradation of the cyclin-dependent kinase inhibitor p27(kip1) and ERK1/2 phosphorylation. The transferred cells displayed reduced Th1/Th17 responses and reduced delayed-type hypersensitivity. Moreover, MOG(35-55)-reactive CD4(+) T cells in ob/ob mice underwent apoptosis that associated with a downmodulation of Bcl-2. Similar results were observed in transgenic AND-TCR- mice carrying a TCR specific for the pigeon cytochrome c 88-104 peptide. These molecular events reveal a reduced activity of the nutrient/energy-sensing AKT/mammalian target of rapamycin pathway, which can be restored in vivo by exogenous leptin replacement. These results may help to explain a link between chronic inflammation and autoimmune T cell reactivity. Topics: Animals; Apoptosis; Cell Proliferation; Chronic Disease; Cyclin-Dependent Kinase Inhibitor p27; Encephalomyelitis, Autoimmune, Experimental; Energy Metabolism; Female; Glycoproteins; Humans; Inflammation; Leptin; Mice; Mice, Obese; Mice, Transgenic; Mitogen-Activated Protein Kinase 3; Multiple Sclerosis; Myelin-Oligodendrocyte Glycoprotein; Peptide Fragments; Phosphorylation; Proto-Oncogene Proteins c-bcl-2; Species Specificity; Th1 Cells; Th17 Cells; TOR Serine-Threonine Kinases | 2010 |
Antibodies to myelin oligodendrocyte glycoprotein are not involved in the severity of chronic non-remitting experimental autoimmune encephalomyelitis.
To elucidate the role of antibodies in development of chronic non-remitting experimental autoimmune encephalomyelitis (EAE) in C57BL/6 mice, which is a well-established Th1-mediated autoimmune disease, and the involvement of activation-induced cytidine deaminase (AID) in Th1-mediated function, we have investigated the myelin oligodendrocyte glycoprotein (MOG)-induced EAE in mice deficient of AID, which is absolutely required for class switching and somatic hypermutation. Following immunization with MOG, AID(-/-) had completely same levels of clinical and pathological severity of EAE when compared with AID(+/-) and AID(+/+), although AID(-/-) did not produce IgG and anti-MOG IgG. Similar levels of T cell proliferation and a modest increase of anti-MOG IgM synthesis were found in spleen cells of AID(-/-) stimulated with MOG. These results indicate that antibodies are not involved in development of EAE in C57BL/6 mice. Topics: Animals; Antibody Formation; Cell Proliferation; Chronic Disease; Cytidine Deaminase; Encephalomyelitis, Autoimmune, Experimental; Glycoproteins; Immunoglobulin Class Switching; Immunoglobulin G; Mice; Mice, Inbred C57BL; Mice, Knockout; Myelin Proteins; Myelin-Associated Glycoprotein; Myelin-Oligodendrocyte Glycoprotein; Peptide Fragments; Somatic Hypermutation, Immunoglobulin; T-Lymphocyte Subsets; Th1 Cells | 2009 |
Genetic deletion of Fatty Acid Amide Hydrolase results in improved long-term outcome in chronic autoimmune encephalitis.
The enzyme Fatty Acid Amide Hydrolase (FAAH) is a key regulator of the endogenous levels of a family of biologically active lipid mediators, the fatty acid amides. These include anandamide, oleoyl ethanolamide and palmitoyl ethanolamide, and their effects are mediated by a variety of downstream targets including cannabinoid receptors and peroxisome proliferator-activated receptors (PPARs). Activation of both of these may have anti-inflammatory and neuroprotective effects. Levels of all three mediators are low in normal nervous tissue, but substantially elevated in mice lacking FAAH as a result of genetic deletion. There is a long anecdotal history of cannabis use by patients suffering from multiple sclerosis, and preclinical studies have indicated beneficial effects of cannabinoid receptor stimulation on both long-term outcome and acute muscle spasm in rodent models of multiple sclerosis (experimental autoimmune encephalitis; EAE). Thus far no report has appeared on the effect of inhibition of FAAH on the progression of EAE. Using a chronic mouse EAE model, we present data indicating that mice lacking FAAH experience an initial inflammatory phase of EAE similar in severity to wild type controls, but exhibited a more substantial clinical remission compared to wild type mice. Topics: Amidohydrolases; Animals; CD3 Complex; Chronic Disease; Demyelinating Diseases; Disease Models, Animal; Encephalomyelitis, Autoimmune, Experimental; Glycoproteins; Mice; Mice, Inbred C57BL; Mice, Knockout; Myelin-Oligodendrocyte Glycoprotein; Peptide Fragments; Spinal Cord; T-Lymphocytes; Time Factors | 2008 |
Delayed administration of erythropoietin and its non-erythropoietic derivatives ameliorates chronic murine autoimmune encephalomyelitis.
Erythropoietin (EPO) mediates a wide range of neuroprotective activities, including amelioration of disease and neuroinflammation in rat models of EAE. However, optimum dosing parameters are currently unknown. In the present study, we used a chronic EAE model induced in mice by immunization with the myelin oligodendrocyte glycoprotein peptide (MOG35-55) to compare the effect of EPO given with different treatment schedules. EPO was administered intraperitoneally at 0.5, 5.0 or 50 microg/kg three times weekly starting from day 3 after immunization (preventive schedule), at the onset of clinical disease (therapeutic schedule) or 15 days after the onset of symptoms (late therapeutic schedule). The results show that EPO is effective even when given after the appearance of clinical signs of EAE, but with a reduced efficacy compared to the preventative schedule. To determine whether this effect requires the homodimeric EPO receptor (EPOR2)-mediated hematopoietic effect of EPO, we studied the effect of carbamylated EPO (CEPO) that does not bind EPOR2. CEPO, ameliorated EAE without changing the hemoglobin concentration. Another non-erythropoietic derivative, asialoEPO was also effective. Both EPO and CEPO equivalently decreased the EAE-associated production of TNF-alpha, IL-1beta and IL-1Ra in the spinal cord, and IFN-gamma by peripheral lymphocytes, indicating that their action involves targeting neuroinflammation. The lowest dosage tested appeared fully effective. The possibility to dissociate the anti-neuroinflammatory action of EPO from its hematopoietic action, which may cause undesired side effects in non-anemic patients, present new avenues to the therapy of multiple sclerosis. Topics: Analysis of Variance; Animals; Body Weight; Chronic Disease; Cytokines; Disease Models, Animal; Dose-Response Relationship, Drug; Drug Administration Schedule; Encephalomyelitis, Autoimmune, Experimental; Erythropoietin; Female; Glycoproteins; Hematocrit; Humans; Immunohistochemistry; Mice; Mice, Inbred C57BL; Myelin-Oligodendrocyte Glycoprotein; Neuroprotective Agents; Peptide Fragments; Recombinant Proteins; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Severity of Illness Index; Spinal Cord; Spleen; Statistics, Nonparametric; Time Factors; Treatment Outcome; Tumor Necrosis Factor-alpha | 2006 |
CD1-dependent regulation of chronic central nervous system inflammation in experimental autoimmune encephalomyelitis.
The existence of T cells restricted for the MHC I-like molecule CD1 is well established, but the function of these cells is still obscure; one implication is that CD1-dependent T cells regulate autoimmunity. In this study, we investigate their role in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, using CD1-deficient mice on a C57BL/6 background. We show that CD1-/- mice develop a clinically more severe and chronic EAE compared with CD1+/+ C57BL/6 mice, which was histopathologically confirmed with increased demyelination and CNS infiltration in CD1-/- mice. Autoantigen rechallenge in vitro revealed similar T cell proliferation in CD+/+ and CD1-/- mice but an amplified cytokine response in CD1-/- mice as measured by both the Th1 cytokine IFN-gamma and the Th2 cytokine IL-4. Investigation of cytokine production at the site of inflammation showed a CNS influx of TGF-beta1-producing cells early in the disease in CD1+/+ mice, which was absent in the CD1-/- mice. Passive transfer of EAE using an autoreactive T cell line induced equivalent disease in both groups, which suggested additional requirements for activation of the CD1-dependent regulatory pathway(s). When immunized with CFA before T cell transfer, the CD1-/- mice again developed an augmented EAE compared with CD1+/+ mice. We suggest that CD1 exerts its function during CFA-mediated activation, regulating development of EAE both through enhancing TGF-beta1 production and through limiting autoreactive T cell activation, but not necessarily via effects on the Th1/Th2 balance. Topics: Animals; Antigens, CD1; Brain; Cell Division; Cells, Cultured; Chronic Disease; Cytokines; Encephalomyelitis, Autoimmune, Experimental; Female; Genotype; Glycoproteins; Incidence; Lymphocyte Activation; Male; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Mice, Knockout; Myelin Sheath; Myelin-Oligodendrocyte Glycoprotein; Peptide Fragments; Severity of Illness Index; Spinal Cord; T-Lymphocytes; Transforming Growth Factor beta; Transforming Growth Factor beta1; Up-Regulation; Vaccination | 2004 |
Recombinant TCR ligand induces tolerance to myelin oligodendrocyte glycoprotein 35-55 peptide and reverses clinical and histological signs of chronic experimental autoimmune encephalomyelitis in HLA-DR2 transgenic mice.
In a previous study, we demonstrated that myelin oligodendrocyte glycoprotein (MOG)-35-55 peptide could induce severe chronic experimental autoimmune encephalomyelitis (EAE) in HLA-DR2(+) transgenic mice lacking all mouse MHC class II genes. We used this model to evaluate clinical efficacy and mechanism of action of a novel recombinant TCR ligand (RTL) comprised of the alpha(1) and beta(1) domains of DR2 (DRB1*1501) covalently linked to the encephalitogenic MOG-35-55 peptide (VG312). We found that the MOG/DR2 VG312 RTL could induce long-term tolerance to MOG-35-55 peptide and reverse clinical and histological signs of EAE in a dose- and peptide-dependent manner. Some mice treated with lower doses of VG312 relapsed after cessation of daily treatment, but the mice could be successfully re-treated with a higher dose of VG312. Treatment with VG312 strongly reduced secretion of Th1 cytokines (TNF-alpha and IFN-gamma) produced in response to MOG-35-55 peptide, and to a lesser degree purified protein derivative and Con A, but had no inhibitory effect on serum Ab levels to MOG-35-55 peptide. Abs specific for both the peptide and MHC moieties of the RTLs were also present after treatment with EAE, but these Abs had only a minor enhancing effect on T cell activation in vitro. These data demonstrate the powerful tolerance-inducing therapeutic effects of VG312 on MOG peptide-induced EAE in transgenic DR2 mice and support the potential of this approach to inhibit myelin Ag-specific responses in multiple sclerosis patients. Topics: Amino Acid Sequence; Animals; Chronic Disease; Cytokines; Dose-Response Relationship, Immunologic; Encephalomyelitis, Autoimmune, Experimental; Female; Genetic Vectors; Glycoproteins; Growth Inhibitors; HLA-DR2 Antigen; Humans; Immune Tolerance; Inflammation Mediators; Ligands; Male; Mice; Mice, Inbred C57BL; Mice, Inbred DBA; Mice, Knockout; Mice, Transgenic; Molecular Sequence Data; Myelin-Oligodendrocyte Glycoprotein; Peptide Fragments; Receptors, Antigen, T-Cell; Recombinant Proteins | 2003 |