Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
BMC Neurosci ; 25(1): 16, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38468222

ABSTRACT

BACKGROUND: Myelin oligodendrocyte glycoprotein-associated disorders (MOGAD) is an autoimmune central nervous system disease. Antigen-specific immune tolerance using nanoparticles such as Polylactic-co-glycolic acid (PLGA) have recently been used as a new therapeutic tolerization approach for CNS autoimmune diseases. We examined whether MOG1-125 conjugated with PLGA could induce MOG-specific immune tolerance in an experimental autoimmune encephalitis (EAE) mouse model. EAE was induced in sixty C57BL/6 J wild-type mice using MOG1-125 peptide with complete Freund's Adjuvant. The mice were divided into 12 groups (n = 5 each) to test the ability of MOG1-125 conjugated PLGA intervention to mitigate the severity or improve the outcomes from EAE with and without rapamycin compared to antigen alone or PLGA alone. EAE score and serum MOG-IgG titers were compared among the interventions.Kindly check and confirm the processed Affiliation “4” is appropriate.I confirmed the Aff 4.Affiliation: Corresponding author information have been changed to present affiliation. Kindly check and confirm.I checked and confirmed the Corresponding author's information. RESULTS: Mice with EAE that were injected intraperitoneally with MOG1-125 conjugated PLGA + rapamycin complex showed dose-dependent mitigation of EAE score. Intraperitoneal and intravenous administration resulted in similar clinical outcomes, whereas 80% of mice treated with subcutaneous injection had a recurrence of clinical score worsening after approximately 1 week. Although there was no significant difference in EAE scores between unconjugated-PLGA and MOG-conjugated PLGA, serum MOG-IgG tended to decrease in the MOG-conjugated PLGA group compared to controls. CONCLUSION: Intraperitoneal administration of PLGA resulted in dose-dependent and longer-lasting immune tolerance than subcutaneous administration. The induction of immune tolerance using PLGA may represent a future therapeutic option for patients with MOGAD.


Subject(s)
Encephalitis , Encephalomyelitis, Autoimmune, Experimental , Hashimoto Disease , Polyesters , Humans , Mice , Animals , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Mice, Inbred C57BL , Glycols/adverse effects , Sirolimus/pharmacology , Immunoglobulin G/adverse effects
2.
Br J Pharmacol ; 180(23): 2989-3007, 2023 12.
Article in English | MEDLINE | ID: mdl-37400950

ABSTRACT

BACKGROUND AND PURPOSE: Fibroblast growth factors and receptors (FGFR) have been shown to modulate inflammation and neurodegeneration in multiple sclerosis (MS). The selective FGFR inhibitor infigratinib has been shown to be effective in cancer models. Here, we investigate the effects of infigratinib on prevention and suppression of first clinical episodes of myelin oligodendrocyte glycoprotein (MOG)35-55 -induced experimental autoimmune encephalomyelitis (EAE) in mice. EXPERIMENTAL APPROACH: The FGFR inhibitor infigratinib was given over 10 days from the time of experimental autoimmune encephalomyelitis induction or the onset of symptoms. The effects of infigratinib on proliferation, cytotoxicity and FGFR signalling proteins were studied in lymphocyte cell lines and microglial cells. KEY RESULTS: Administration of infigratinib prevented by 40% and inhibited by 65% first clinical episodes of the induced experimental autoimmune encephalomyelitis. In the spinal cord, infiltration of lymphocytes and macrophages/microglia, destruction of myelin and axons were reduced by infigratinib. Infigratinib enhanced the maturation of oligodendrocytes and increased remyelination. In addition, infigratinib resulted in an increase of myelin proteins and a decrease in remyelination inhibitors. Further, lipids associated with neurodegeneration such as lysophosphatidylcholine and ceramide were decreased as were proliferation of T cells and microglial cells. CONCLUSION AND IMPLICATIONS: This proof of concept study demonstrates the therapeutic potential of targeting FGFRs in a disease model of multiple sclerosis. Application of oral infigratinib resulted in anti-inflammatory and remyelinating effects. Thus, infigratinib may have the potential to slow disease progression or even to improve the disabling symptoms of multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Remyelination , Mice , Animals , Multiple Sclerosis/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Spinal Cord/metabolism , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein/metabolism , Anti-Inflammatory Agents/pharmacology , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Fibroblast Growth Factor/therapeutic use , Mice, Inbred C57BL
3.
Fundam Clin Pharmacol ; 37(2): 215-225, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36300567

ABSTRACT

Macrophage M2 (MP2)-based cell therapy is a novel medicinal treatment for animals with Experimental Autoimmune Encephalomyelitis (EAE) as an experimental model of multiple sclerosis (MS). This systematic review and meta-analysis study was designed to assess the overall therapeutic effects of MP2 cell therapy on Clinical Score and motor impairment in EAE-induced animals. All experiments on MP2 cell therapy in animals with EAE were gathered (by October 2, 2022) from English (PubMed, Scopus, WoS, Science Direct, and ISC) and Persian (MagIran and SID) databases. The searching strategy was designed using "Experimental Autoimmune Encephalomyelitis," "Multiple Sclerosis," and "Macrophage M2" keywords. Following primary and secondary screenings, eligible papers were selected based on the PRISMA 2020 guideline, and the study quality was assessed using the Animal Research: Reporting of In Vivo Experiments (ARRIVE) checklist. The difference in means of Clinical Score (score 0-5) as the effect size (ES) was analyzed based on the random effect model (CMA software, v.2). Subgrouping (EAE phases of Onset, Peak, and Recovery) was applied, and I2 index was used to assess the heterogeneity index. Publication bias and sensitivity indices were also evaluated. P < 0.05 was considered significant, and the confidence interval (CI) was determined 95%. Among 22 gathered papers, medium to high quality studies were selected for meta-analysis. Difference in means, P value, and I2 for Onset, Peak, and Recovery phases were 0.082 (CI95%: -0.323-0.159, P value: 0.504, I2 : 67.961%), -0.606 (CI95%: -1.518 to -0.305, P value: 0.192, I2 : 96.070%), and -1.103 (CI95%: -1.390 to -0.816, P value: 0.000, I2 : 30.880%), respectively and Overall Effect was found -0.509 (CI95%: -0.689 to -0.328, P value < 0.001). Also, P value (two-tailed) indices for publication bias were 0.366 and 0.583 for Egger's regression intercept and Begg rank correlation, respectively. The P value for sensitivity was detected 0.003. Cell therapy procedure using MP2 can potentially alleviate the Clinical Scores Index and correct the motor defects in Recovery phase of EAE animals. In healthy mice, the brain and myelin surrounding neurons are in a healthy and physiological state (1). To evaluate MS in humans, it is necessary to model this type of disease in animals using EAE procedure through subcutaneous injection of CFA, MOG35-55 , MT, and Pert. Thus, inflammation and autoimmunity occur, which finally lead to myelin destruction and motor symptoms (2). By aspiration of progenitor cells available in bone marrow, the MP2 can be isolated and cultured. By activation of these types of cells, a rich collection of MP2 can be prepared for the cell-therapy process (3). After injection through the tail vein or intra-peritoneal procedure, these cells can be located in CNS through crossing from the BBB. They begin their anti-inflammatory activities and help repair the damaged myelin (4). Eventually, the clinical symptoms can be modified considerably, and the animal motor function improves (5). CFA, complete Freund's adjuvant; MOG35-55 , myelin oligodendrocyte glycoprotein; MT, Mycobacterium tuberculosis; Pert, pertussis; EAE, Experimental Autoimmune Encephalomyelitis; BM, bone marrow; MP2, macrophage M2; and BBB, blood brain barrier.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Humans , Mice , Animals , Encephalomyelitis, Autoimmune, Experimental/therapy , Multiple Sclerosis/therapy , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Anti-Inflammatory Agents , Macrophages/metabolism , Mice, Inbred C57BL , Peptide Fragments/adverse effects
4.
Front Immunol ; 12: 615898, 2021.
Article in English | MEDLINE | ID: mdl-33776998

ABSTRACT

Interleukin-19 (IL-19) acts as a negative-feedback regulator to limit proinflammatory response of macrophages and microglia in autocrine/paracrine manners in various inflammatory diseases. Multiple sclerosis (MS) is a major neuroinflammatory disease in the central nervous system (CNS), but it remains uncertain how IL-19 contributes to MS pathogenesis. Here, we demonstrate that IL-19 deficiency aggravates experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, by promoting IL-17-producing helper T cell (Th17 cell) infiltration into the CNS. In addition, IL-19-deficient splenic macrophages expressed elevated levels of major histocompatibility complex (MHC) class II, co-stimulatory molecules, and Th17 cell differentiation-associated cytokines such as IL-1ß, IL-6, IL-23, TGF-ß1, and TNF-α. These observations indicated that IL-19 plays a critical role in suppression of MS pathogenesis by inhibiting macrophage antigen presentation, Th17 cell expansion, and subsequent inflammatory responses. Furthermore, treatment with IL-19 significantly abrogated EAE. Our data suggest that IL-19 could provide significant therapeutic benefits in patients with MS.


Subject(s)
Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Interleukins/metabolism , Animals , Biomarkers , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Immunohistochemistry , Immunophenotyping , Interleukins/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
5.
Cells ; 9(1)2019 12 19.
Article in English | MEDLINE | ID: mdl-31861635

ABSTRACT

Multiple sclerosis (MS) is a demyelinating autoimmune disease of the central nervous system (CNS) mediated by autoreactive lymphocytes. The role of autoreactive lymphocytes in the CNS demyelination is well described, whereas very little is known about their role in remyelination during MS remission. In this study, we identified a new subpopulation of myelin-specific CD49d+CD154+ lymphocytes presented in the peripheral blood of MS patients during remission, that proliferated in vitro in response to myelin peptides. These lymphocytes possessed the unique ability to migrate towards maturing oligodendrocyte precursor cells (OPCs) and synthetize proinflammatory chemokines/cytokines. The co-culture of maturing OPCs with myelin-specific CD49d+CD154+ lymphocytes was characterized by the increase in proinflammatory chemokine/cytokine secretion that was not only a result of their cumulative effect of what OPCs and CD49d+CD154+ lymphocytes produced alone. Moreover, maturing OPCs exposed to exogenous myelin peptides managed to induce CD40-CD154-dependent CD49d+CD154+ lymphocyte proliferation. We confirmed, in vivo, the presence of CD49d+CD154+ cells close to maturating OPCs and remyelinating plaque during disease remission in the MS mouse model (C57Bl/6 mice immunized with MOG35-55) by immunohistochemistry. Three weeks after an acute phase of experimental autoimmune encephalomyelitis, CD49d+/CD154+ cells were found to be co-localized with O4+ cells (oligodendrocyte progenitors) in the areas of remyelination identified by myelin basic protein (MBP) labelling. These data suggested that myelin-specific CD49d+CD154+ lymphocytes present in the brain can interfere with remyelination mediated by oligodendrocytes probably as a result of establishing proinflammatory environment.


Subject(s)
CD40 Ligand/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Integrin alpha4/metabolism , Multiple Sclerosis/immunology , Myelin Sheath/metabolism , Adult , Animals , Case-Control Studies , Cell Proliferation , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Lymphocytes/cytology , Lymphocytes/immunology , Male , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/immunology , Peptide Fragments/adverse effects , Remyelination
6.
Cells ; 8(11)2019 11 14.
Article in English | MEDLINE | ID: mdl-31739589

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is the most commonly used multiple sclerosis animal model. EAE mice typically develop motor deficits in a caudal-to-rostral pattern when inflammatory lesions have already developed. However, to monitor more subtle behavioral deficits during lesion development (i.e., pre-clinical phase), more sophisticated methods are needed. Here, we investigated whether high speed ventral plane videography can be applied to monitor early motor deficits during 'pre-clinical' EAE. For this purpose, EAE was induced in C57BL/6 mice and gait abnormalities were quantified using the DigiGait™ apparatus. Gait deficits were related to histopathological changes. 10 out of 10 control (100%), and 14 out of 18 (77.8%) pre-clinical EAE mice could be evaluated using DigiGait™. EAE severity was not influenced by DigiGait™-related mice handlings. Most gait parameters recorded from day 6 post-immunization until the end of the experiment were found to be stable in control mice. During the pre-clinical phase, when conventional EAE scorings failed to detect any functional impairment, EAE mice showed an increased Swing Time, increased %Swing Stride, decreased %Stance Stride, decreased Stance/Swing, and an increased Absolute Paw Angle. In summary, DigiGait™ is more sensitive than conventional scoring approaches to study motor deficits during the EAE pre-clinical phase.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Gait Analysis/instrumentation , Animals , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Mice , Mice, Inbred C57BL , Motor Skills , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Rotarod Performance Test , Severity of Illness Index , Video Recording
7.
J Exp Med ; 216(10): 2427-2447, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31324740

ABSTRACT

Signal strength controls the outcome of αß T cell selection in the thymus, resulting in death if the affinity of the rearranged TCR is below the threshold for positive selection, or if the affinity of the TCR is above the threshold for negative selection. Here we show that deletion of the GTPase RRAS2 results in exacerbated negative selection and above-normal expression of positive selection markers. Furthermore, Rras2-/- mice are resistant to autoimmunity both in a model of inflammatory bowel disease (IBD) and in a model of myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). We show that MOG-specific T cells in Rras2-/- mice have reduced affinity for MOG/I-Ab tetramers, suggesting that enhanced negative selection leads to selection of TCRs with lower affinity for the self-MOG peptide. An analysis of the TCR repertoire shows alterations that mostly affect the TCRα variable (TRAV) locus with specific VJ combinations and CDR3α sequences that are absent in Rras2-/- mice, suggesting their involvement in autoimmunity.


Subject(s)
Clonal Selection, Antigen-Mediated , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Rearrangement, alpha-Chain T-Cell Antigen Receptor/immunology , Membrane Proteins/immunology , Monomeric GTP-Binding Proteins/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor/immunology , Membrane Proteins/genetics , Mice , Mice, Knockout , Monomeric GTP-Binding Proteins/genetics , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein/pharmacology
8.
Front Immunol ; 9: 944, 2018.
Article in English | MEDLINE | ID: mdl-29867938

ABSTRACT

Multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE) have been proved to be T cell-mediated autoimmune diseases. Recent researches indicate that humoral immunity is also involved in the pathogenesis of these disorders. T follicular helper (Tfh) cells are critical for B cell differentiation and antibody production. However, the role of Tfh cells in MS and EAE remains unclear. Here, we found elevated frequencies of CD4+CXCR5+PD-1+ Tfh-like cells in both MS patients and EAE. In EAE mice, Tfh-like cells, together with B cells, were found in the ectopic lymphoid structures in spinal cords. Moreover, Tfh-like cells promoted the antibody production via IL-21/IL-21R and CD40 ligand/CD40 interaction and the synergy effect of STAT3 and non-canonical NF-κB signaling pathway inside B cells. Moreover, adoptive transfer of Tfh-like cells could increase the severity and delay the remission of EAE. In conclusion, our data indicate that Tfh-like cells contribute to the pathogenesis of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Adult , Animals , Autoantibodies/immunology , Autoimmunity , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Brain/immunology , Brain/metabolism , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/pathology , Humans , Lymphocyte Activation , Lymphocyte Count , Male , Mice , Middle Aged , Multiple Sclerosis, Relapsing-Remitting , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Peptide Fragments/adverse effects , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , Young Adult
9.
J Hand Surg Am ; 43(9): 873.e1-873.e4, 2018 09.
Article in English | MEDLINE | ID: mdl-29526530

ABSTRACT

Accidental needlestick injuries are common in laboratory and health care workers. Injection of atypical pathogens, such as those encountered in the animal laboratory setting, may pose considerable problems at the site of inoculation. We present the case of an otherwise healthy laboratory worker who accidentally self-injected Freund complete adjuvant with heat-killed Mycobacterium tuberculosis into her hand, requiring multiple debridement operations over a prolonged treatment course.


Subject(s)
Freund's Adjuvant/administration & dosage , Hand Injuries/therapy , Mycobacterium tuberculosis , Needlestick Injuries/therapy , Accidents, Occupational , Adult , Debridement , Female , Freund's Adjuvant/adverse effects , Glucocorticoids/therapeutic use , Granuloma/etiology , Granuloma/surgery , Humans , Laboratory Personnel , Methylprednisolone/therapeutic use , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Needlestick Injuries/complications , Peptide Fragments/administration & dosage , Peptide Fragments/adverse effects , Triamcinolone Acetonide/therapeutic use
10.
Int Immunol ; 30(2): 69-78, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29528433

ABSTRACT

Schlafen-8 (Slfn8) is a member of the Schlafen family of proteins, which harbor helicase domains and are induced by LPS and interferons. It has been reported that the Schlafen family are involved in various cellular functions, including proliferation, differentiation and regulation of virus replication. Slfn8 has been implicated in T-cell differentiation in the thymus. However, the roles of Slfn8 in the immune system remains unclear. In this study, we generated Slfn8 knockout mice (Slfn8-/-) and investigated the immunological role of Slfn8 using the T-cell-mediated autoimmune model experimental autoimmune encephalomyelitis (EAE). We found that the clinical score was reduced in Slfn8-/- mice. IL-6 and IL-17A cytokine production, which are associated with EAE onset and progression, were decreased in the lymph nodes of Slfn8-/- mice. Immune cell populations in Slfn8-/- mice, including macrophages, neutrophils, T cells and B cells, did not reveal significant differences compared with wild-type mice. In vitro activation of Slfn8-/- T cells in response to TCR stimulation also did not reveal significant differences. To confirm the involvement of non-hematopoietic cells, we isolated CD45- CD31+ endothelial cells and CD45-CD31- gp38+ fibroblastic reticular cells by FACS sorting. We showed that the levels of IL-6 and Slfn8 mRNA in CD45- CD31+ endothelial cells were increased after EAE induction. In contrast, the level of IL-6 mRNA after EAE induction was markedly decreased in CD31+ endothelial cells from Slfn8-/- mice. These results indicate that Slfn8 may play a role in EAE by regulating inflammation in endothelial cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelial Cells/metabolism , Animals , Biomarkers , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunity, Innate , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Peptide Fragments/adverse effects , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
J Korean Med Sci ; 32(9): 1502-1507, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28776347

ABSTRACT

Multiple sclerosis (MS) is a T-lymphocyte-mediated autoimmune disease that is characterized by inflammation in the central nervous system (CNS). Although many disease-modifying therapies (DMTs) are presumed effective in patients with MS, studies on the efficacy and safety of DMTs for preventing MS relapse are limited. Therefore, we tested the immunosuppressive anti-inflammatory effects of oral-formulated tacrolimus (FK506) on MS in a mouse model of experimental autoimmune encephalomyelitis (EAE). The mice were randomly divided into 3 experimental groups: an untreated EAE group, a low-dose tacrolimus-treated EAE group, and a high-dose tacrolimus-treated EAE group. After autoimmunization of the EAE mice with myelin oligodendrocyte glycoprotein, symptom severity scores, immunohistochemistry of the myelination of the spinal cord, and western blotting were used to evaluate the EAE mice. After the autoimmunization, the symptom scores of each EAE group significantly differed at times. The group treated with the larger tacrolimus dose had the lowest symptom scores. The tacrolimus-treated EAE groups exhibited less demyelination and inflammation and weak immunoreactivity for all of the immunization biomarkers. Our results revealed that oral-formulated tacrolimus inhibited the autoimmunization in MS pathogenesis by inactivating inflammatory cells.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Tacrolimus/therapeutic use , Administration, Oral , Animals , Anti-Inflammatory Agents/chemistry , Biomarkers/metabolism , CD4 Antigens/metabolism , Calcium-Binding Proteins/metabolism , Drug Compounding , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Severity of Illness Index , Spinal Cord/pathology , Tacrolimus/chemistry
12.
Glia ; 64(7): 1124-37, 2016 07.
Article in English | MEDLINE | ID: mdl-27120265

ABSTRACT

The Na(+) /Ca(2+) exchanger NCX3, recently identified as a myelin membrane component, is involved in the regulation of [Ca(2+) ]i during oligodendrocyte maturation. Here NCX3 involvement was studied in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. Western blotting and quantitative colocalization studies performed in wild-type ncx3(+/+) mice at different stages of EAE disease showed that NCX3 protein was intensely upregulated during the chronic stage, where it was intensely coexpressed with the oligodendrocyte precursor cells (OPC) marker NG2 and the premyelinating marker CNPase. Moreover, MOG35-55 -immunized mice lacking the ncx3 gene displayed not only a reduced diameter of axons and an intact myelin ring number but also a dramatic decrease in OPC and pre-myelinating cells in the white matter of the spinal cord when compared with ncx3(+/+) . Accordingly, ncx3(-/-) and ncx3(+/-) mutants developed early onset of EAE and more severe clinical symptoms. Interestingly, cytofluorimetric analysis revealed that during the peak stage of the disease, the number of immune T-cell subsets in ncx3(-/-) mice, was not statistically different from that measured in ncx3(+/+) . Our findings demonstrate that knocking-out NCX3 impairs oligodendrocyte response and worsens clinical symptoms in EAE without altering the immune T-cell population. GLIA 2016;64:1124-1137.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Oligodendrocyte Precursor Cells/metabolism , Oligodendrocyte Precursor Cells/pathology , Sodium-Calcium Exchanger/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Antigens/metabolism , Axons/metabolism , Axons/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Nerve Tissue Proteins/metabolism , Proteoglycans/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Sodium-Calcium Exchanger/genetics , Spinal Cord/metabolism , Spinal Cord/pathology , Spleen/metabolism , Spleen/pathology , Up-Regulation/genetics , Up-Regulation/immunology
13.
J Neuroimmunol ; 292: 58-67, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26943960

ABSTRACT

Immoderate immunoreaction of antigen-specific Th17 and Treg cell dysfunction play critical roles in the pathogenesis of multiple sclerosis. We examined Th17/Treg immune responses and the underlying mechanisms in response to metformin in C57BL/6 mice with experimental autoimmune encephalomyelitis (EAE). Metformin reduced Th17 and increased Treg cell percentages along with the levels of associated cytokines. Molecules involved in cellular metabolism were altered in mice with EAE. Suppressed activation of mTOR and its downstream target, HIF-1α, likely mediated the protective effects of metformin. Our findings demonstrate that regulation of T cell metabolism represents a new therapeutic target for CNS autoimmune disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Analysis of Variance , Animals , Cytokines/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Hypoglycemic Agents/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit , Metformin/pharmacology , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/adverse effects , Peptide Fragments/immunology , RNA, Messenger/metabolism
14.
Daru ; 23: 48, 2015 Oct 21.
Article in English | MEDLINE | ID: mdl-26489494

ABSTRACT

BACKGROUND: The present study was designed to investigate the efficacy of a new formulation of alone, purified cannabidiol (CBD) (>98 %), the main non-psychotropic cannabinoid of Cannabis sativa, as a topical treatment in an experimental model of autoimmune encephalomyelitis (EAE), the most commonly used model for multiple sclerosis (MS). Particularly, we evaluated whether administration of a topical 1 % CBD-cream, given at the time of symptomatic disease onset, could affect the EAE progression and if this treatment could also recover paralysis of hind limbs, qualifying topical-CBD for the symptomatic treatment of MS. METHODS: In order to have a preparation of 1 % of CBD-cream, pure CBD have been solubilized in propylene glycoland basic dense cream O/A. EAE was induced by immunization with myelin oligodendroglial glycoprotein peptide (MOG35-55) in C57BL/6 mice. After EAE onset, mice were allocated into several experimental groups (Naïve, EAE, EAE-1 % CBD-cream, EAE-vehicle cream, CTRL-1 % CBD-cream, CTRL-vehicle cream). Mice were observed daily for signs of EAE and weight loss. At the sacrifice of the animals, which occurred at the 28(th) day from EAE-induction, spinal cord and spleen tissues were collected in order to perform histological evaluation, immunohistochemistry and western blotting analysis. RESULTS: Achieved results surprisingly show that daily treatment with topical 1 % CBD-cream may exert neuroprotective effects against EAE, diminishing clinical disease score (mean of 5.0 in EAE mice vs 1.5 in EAE + CBD-cream), by recovering of paralysis of hind limbs and by ameliorating histological score typical of disease (lymphocytic infiltration and demyelination) in spinal cord tissues. Also, 1 % CBD-cream is able to counteract the EAE-induced damage reducing release of CD4 and CD8α T cells (spleen tissue localization was quantified about 10,69 % and 35,96 % of positive staining respectively in EAE mice) and expression of the main pro-inflammatory cytokines as well as several other direct or indirect markers of inflammation (p-selectin, IL-10, GFAP, Foxp3, TGF-ß, IFN-γ), oxidative injury (Nitrotyrosine, iNOS, PARP) and apoptosis (Cleaved caspase 3). CONCLUSION: All these data suggest an interesting new profile of CBD that could lead to its introduction in the clinical management of MS and its associated symptoms at least in association with current conventional therapy.


Subject(s)
Cannabidiol/administration & dosage , Cannabidiol/isolation & purification , Cytokines/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Administration, Topical , Animals , Cannabidiol/pharmacology , Chemistry, Pharmaceutical , Cytokines/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Male , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Skin Cream , Weight Loss/drug effects
15.
J Neuroinflammation ; 12: 169, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26377397

ABSTRACT

BACKGROUND: Experimental autoimmune encephalomyelitis (EAE) in the common marmoset monkey (Callithrix jacchus) is a relevant preclinical model for translational research into immunopathogenic mechanisms operating in multiple sclerosis (MS). Prior studies showed a core pathogenic role of T and B cells specific for myelin oligodendrocyte glycoprotein (MOG). However, in those studies, the quality of the response against MOG epitopes was strongly biased by bacterial antigens in the complete Freund's adjuvant (CFA), in which the immunizing recombinant human (rh) MOG protein had been formulated. In response to the need of a more refined EAE model, we have tested whether disease could also be induced with rhMOG in incomplete Freund's adjuvant (IFA). METHOD: Marmosets were immunized with rhMOG emulsified in IFA in the dorsal skin. Monkeys that did not develop neurological deficit were given booster immunizations at 28-day interval with the same antigen preparation. In a second experiment, three marmoset twin pairs were sensitized against MOG peptides in IFA to study a possibility for suppressive activity towards pathogenic T cells directed against the encephalitogenic epitope MOG40-48. RESULTS: Despite the absence of strong danger signals in the rhMOG/IFA inoculum, all monkeys developed clinically evident EAE symptoms. Moreover, in all monkeys, demyelinated lesions were present in the white matter and in two cases also in the cortical grey matter. Immune profiling at height of the disease showed a dominant T cell response against the overlapping peptides 14-36 and 24-46, but reactivity against the pathogenically most relevant peptide 34-56 was conspicuously absent. In the second experiment, there was an indication for a possible suppressive mechanism. CONCLUSIONS: Immunization of marmoset monkeys with rhMOG in IFA elicits clinical EAE in all animals. Moreover, rhMOG contains pathogenic and regulatory epitopes, but the pathogenic hierarchy of rhMOG epitopes is strongly influenced by the adjuvant in which the protein is formulated.


Subject(s)
Callithrix , Encephalomyelitis, Autoimmune, Experimental/immunology , Freund's Adjuvant/immunology , Lipids/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Animals , Antibodies/blood , Brain/metabolism , Brain/pathology , Cell Proliferation/drug effects , Cytokines/blood , Cytokines/genetics , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Freund's Adjuvant/adverse effects , Humans , Immunization/adverse effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Lipids/adverse effects , Lymphocyte Activation/immunology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Peptides/adverse effects , Peptides/immunology , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , T-Lymphocytes/immunology
16.
Clin Exp Immunol ; 180(3): 419-31, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25619397

ABSTRACT

Recently, parasite infections or parasite-derived products have been suggested as a therapeutic strategy with suppression of immunopathology, which involves the induction of regulatory T cells or/and T helper type 2 (Th2) responses. In a recent study, researchers reported that constructed recombinant galectin (rTl-gal) isolated from an adult worm of the gastrointestinal nematode parasite Toxascaris leonina attenuated clinical symptoms of inflammatory bowel disease in mice treated with dextran sulphate sodium. Noting the role of rTl-gal in inflammatory disease, we attempted to investigate the effect of the parasite via its rTl-gal on neuronal autoimmune disease using experimental autoimmune encephalomyelitis (EAE), a mouse inflammatory and demyelinating autoimmune disease model of human multiple sclerosis. In this model, rTl-gal-treated experimental autoimmune encephalomyelitis (EAE) mice failed to recover after the peak of the disease, leading to persistent central nervous system (CNS) damage, such as demyelination, gliosis and axonal damage. Further, rTl-gal-treated EAE mice markedly increased the number of CD45R/B220(+) B cells in both infiltrated inflammation and the periphery, along with the increased production of autoantibody [anti-myelin oligodendrocyte glycoprotein (MOG)35-55 ] in serum at chronic stage. Upon antigen restimulation, rTl-gal treatment affected the release of overall cytokines, especially interferon (IFN)-γ and tumour necrosis factor (TNF)-α. Our results suggest that galectin isolated from a gastrointestinal parasite can deliver a harmful effect to EAE contrary to its beneficial effect on inflammatory bowel disease.


Subject(s)
Autoantibodies/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Galectins/immunology , Immunomodulation/drug effects , Parasites/chemistry , Animals , Autoantibodies/blood , Axons/immunology , Axons/metabolism , Axons/pathology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Cytokines/biosynthesis , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Female , Galectins/adverse effects , Galectins/isolation & purification , Gliosis/immunology , Gliosis/metabolism , Gliosis/pathology , Leukocyte Common Antigens/metabolism , Mice , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/adverse effects , Peptide Fragments/immunology , Severity of Illness Index , Spinal Cord/immunology , Spinal Cord/pathology
17.
J Immunol ; 193(9): 4429-38, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25267971

ABSTRACT

In this study, we investigate the basis of T cell recognition of myelin that governs the progression from acute symptoms into disease remission, relapse, and chronic progression in a secondary progressive model of demyelinating disease. Until now, the frequency and affinity of myelin-reactive CD4 T cells that elicit relapsing-remitting disease have not been quantified. The micropipette adhesion frequency assay was used to obtain a sensitive and physiologically relevant two-dimensional measurement of frequency and TCR affinity for myelin, as the inherent low affinity does not allow the use of specific peptide:MHC-II tetramers for this purpose. We found the highest affinity and frequency of polyclonal myelin oligodendrocyte glycoprotein-reactive cells infiltrate the CNS during acute disease, whereas affinities during remission, relapse, and chronic disease are not significantly different from each other. Frequency analysis revealed that the vast majority of CNS-infiltrating CD4 T cells are myelin oligodendrocyte glycoprotein reactive at all time points, demonstrating epitope spread is not a predominant factor for disease progression. Furthermore, time points at which mice were symptomatic were characterized by an infiltration of Th17 cells in the CNS, whereas symptom remission showed an enrichment of cells producing IFN-γ. Also, the ratio of regulatory T cells to Foxp3(-) CD4 T cells was significantly higher in the CNS at remission than during acute disease. The results of this study indicate that a high frequency of T cells specific for a single myelin Ag, rather than increased TCR affinity or epitope spread, governs the transition from acute symptoms through remission, relapse, and chronic disease states.


Subject(s)
Demyelinating Autoimmune Diseases, CNS/immunology , Demyelinating Autoimmune Diseases, CNS/metabolism , Epitopes/immunology , Receptors, Antigen, T-Cell/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Demyelinating Autoimmune Diseases, CNS/pathology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental , Female , Interleukin-17/biosynthesis , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred NOD , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/adverse effects , Peptide Fragments/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
18.
PLoS One ; 9(8): e104430, 2014.
Article in English | MEDLINE | ID: mdl-25116424

ABSTRACT

Recent studies have demonstrated that atypical antipsychotic agents, which are known to antagonize dopamine D2 and serotonin 5-HT2a receptors, have immunomodulatory properties. Given the potential of these drugs to modulate the immune system both peripherally and within the central nervous system, we investigated the ability of the atypical anti-psychotic agent, risperidone, to modify disease in the animal model of multiple sclerosis (MS)4, experimental autoimune encephalomyelitis (EAE). We found that chronic oral administration of risperidone dose-dependently reduced the severity of disease and decreased both the size and number of spinal cord lesions. Furthermore, risperidone treatment substantially reduced antigen-specific interleukin (IL)-17a, IL-2, and IL-4 but not interferon (IFN)-γ production by splenocytes at peak disease and using an in vitro model, we show that treatment of macrophages with risperidone alters their ability to bias naïve T cells. Another atypical antipsychotic agent, clozapine, showed a similar ability to modify macrophages in vitro and to reduce disease in the EAE model but this effect was not due to antagonism of the type 1 or type 2 dopamine receptors alone. Finally, we found that while risperidone treatment had little effect on the in vivo activation of splenic macrophages during EAE, it significantly reduced the activation of microglia and macrophages in the central nervous system. Together these studies indicate that atypical antipsychotic agents like risperidone are effective immunomodulatory agents with the potential to treat immune-mediated diseases such as MS.


Subject(s)
Antipsychotic Agents/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Risperidone/pharmacology , Animals , Antigens/immunology , Antipsychotic Agents/administration & dosage , Cell Survival , Cytokines/biosynthesis , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/diagnosis , Epitopes, T-Lymphocyte/immunology , Female , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Nitric Oxide/metabolism , Peptide Fragments/adverse effects , Risperidone/administration & dosage , Severity of Illness Index , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
J Immunol ; 192(8): 3518-3529, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24639351

ABSTRACT

Tumor progression locus 2 (TPL-2) expression is required for efficient polarization of naive T cells to Th1 effector cells in vitro, as well as for Th1-mediated immune responses. In the present study, we investigated the potential role of TPL-2 in Th17 cells. TPL-2 was found to be dispensable for Th17 cell differentiation in vitro, and for the initial priming of Th17 cells in experimental autoimmune encephalomyelitis (EAE), a Th17 cell-mediated disease model for multiple sclerosis. Nevertheless, TPL-2-deficient mice were protected from EAE, which correlated with reduced immune cell infiltration, demyelination, and axonal damage in the CNS. Adoptive transfer experiments demonstrated that there was no T cell-intrinsic function for TPL-2 in EAE, and that TPL-2 signaling was not required in radiation-sensitive hematopoietic cells. Rather, TPL-2 signaling in radiation-resistant stromal cells promoted the effector phase of the disease. Importantly, using a newly generated mouse strain expressing a kinase-inactive form of TPL-2, we demonstrated that stimulation of EAE was dependent on the catalytic activity of TPL-2 and not its adaptor function to stabilize the associated ubiquitin-binding protein ABIN-2. Our data therefore raise the possibility that small molecule inhibitors of TPL-2 may be beneficial in multiple sclerosis therapy.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression Regulation , MAP Kinase Kinase Kinases/genetics , Proto-Oncogene Proteins/genetics , Animals , Astrocytes/immunology , Astrocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/immunology , Enzyme Activation , Lymphocyte Activation/immunology , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Peptide Fragments/adverse effects , Proto-Oncogene Proteins/metabolism , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
20.
PLoS One ; 9(1): e86643, 2014.
Article in English | MEDLINE | ID: mdl-24475162

ABSTRACT

BACKGROUND: Multiple microarray analyses of multiple sclerosis (MS) and its experimental models have been published in the last years. OBJECTIVE: Meta-analyses integrate the information from multiple studies and are suggested to be a powerful approach in detecting highly relevant and commonly affected pathways. DATA SOURCES: ArrayExpress, Gene Expression Omnibus and PubMed databases were screened for microarray gene expression profiling studies of MS and its experimental animal models. STUDY ELIGIBILITY CRITERIA: Studies comparing central nervous system (CNS) samples of diseased versus healthy individuals with n >1 per group and publically available raw data were selected. MATERIAL AND METHODS: Included conditions for re-analysis of differentially expressed genes (DEGs) were MS, myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE) in rats, proteolipid protein-induced EAE in mice, Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD), and a transgenic tumor necrosis factor-overexpressing mouse model (TNFtg). Since solely a single MS raw data set fulfilled the inclusion criteria, a merged list containing the DEGs from two MS-studies was additionally included. Cross-study analysis was performed employing list comparisons of DEGs and alternatively Gene Set Enrichment Analysis (GSEA). RESULTS: The intersection of DEGs in MS, EAE, TMEV-IDD, and TNFtg contained 12 genes related to macrophage functions. The intersection of EAE, TMEV-IDD and TNFtg comprised 40 DEGs, functionally related to positive regulation of immune response. Over and above, GSEA identified substantially more differentially regulated pathways including coagulation and JAK/STAT-signaling. CONCLUSION: A meta-analysis based on a simple comparison of DEGs is over-conservative. In contrast, the more experimental GSEA approach identified both, a priori anticipated as well as promising new candidate pathways.


Subject(s)
Central Nervous System/metabolism , Demyelinating Diseases/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression Profiling , Multiple Sclerosis/metabolism , Animals , Demyelinating Diseases/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Humans , Mice , Microarray Analysis , Multiple Sclerosis/genetics , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Proteolipids/adverse effects , Rats , Species Specificity , Theilovirus , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL