Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 233
Filter
1.
Proc Natl Acad Sci U S A ; 121(39): e2408324121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39288173

ABSTRACT

Myasthenia gravis (MG) is a chronic and severe disease of the skeletal neuromuscular junction (NMJ) in which the effects of neurotransmitters are attenuated, leading to muscle weakness. In the most common forms of autoimmune MG, antibodies attack components of the postsynaptic membrane, including the acetylcholine receptor (AChR) or muscle-specific kinase (MuSK). MuSK, a master regulator of NMJ development, associates with the low-density lipoprotein-related receptor 4 (Lrp4) to form the signaling receptor for neuronal Agrin, a nerve-derived synaptic organizer. Pathogenic antibodies to MuSK interfere with binding between MuSK and Lrp4, inhibiting the differentiation and maintenance of the NMJ. MuSK MG can be debilitating and refractory to treatments that are effective for AChR MG. We show here that recombinant antibodies, derived from MuSK MG patients, cause severe neuromuscular disease in mice. The disease can be prevented by a MuSK agonist antibody, presented either prophylactically or after disease onset. These findings suggest a therapeutic alternative to generalized immunosuppression for treating MuSK MG by selectively and directly targeting the disease mechanism.


Subject(s)
Myasthenia Gravis , Neuromuscular Junction , Receptor Protein-Tyrosine Kinases , Receptors, Cholinergic , Animals , Receptor Protein-Tyrosine Kinases/immunology , Receptor Protein-Tyrosine Kinases/metabolism , Mice , Neuromuscular Junction/drug effects , Neuromuscular Junction/immunology , Receptors, Cholinergic/immunology , Receptors, Cholinergic/metabolism , Myasthenia Gravis/immunology , Myasthenia Gravis/drug therapy , Humans , LDL-Receptor Related Proteins/immunology , Autoantibodies/immunology , Female , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Antibodies/immunology , Antibodies/pharmacology , Disease Models, Animal , Fatty Acids, Monounsaturated
2.
Allergol Immunopathol (Madr) ; 52(5): 59-64, 2024.
Article in English | MEDLINE | ID: mdl-39278852

ABSTRACT

Acquired myasthenia (AM), a debilitating autoimmune disease, is typically characterized by skeletal muscle fatigue and weakness. Despite advances in myasthenia gravis treatment, current approaches remain unsatisfactory and many result in unexpected side effects. Traditional Chinese medicine has shown great potential in the treatment of myasthenia gravis, including relieving myasthenic symptoms, improving patients' quality of life, and reducing Western medicine side effects. This study investigates the protective effects and mechanism of BZYQD in mice with acquired myasthenia. BZYQD alleviates the reduced grip strength and increased expression of MAFbx and MuRF-1 in mice with acquired myasthenia. It also reduces levels of pro-inflammatory factors IL-1ß, IL-6, and TNF-α in the mouse serum. In addition, BZYQD reduces ROS accumulation and the mitochondrial ROS production rate, while increasing ATP levels and mitochondrial membrane potential in mice with acquired myasthenia. Moreover, BZYQD decreases the expression of p-JAK2, p-STAT3, and p-AKT in the skeletal muscle of mice with acquired myasthenia. In summary, BZYQD reduces inflammation, enhances mitochondrial function, and regulates the JAK2/STAT3/AKT signaling pathway to treat acquired myasthenia.


Subject(s)
Drugs, Chinese Herbal , Janus Kinase 2 , Mitochondria , Proto-Oncogene Proteins c-akt , STAT3 Transcription Factor , Signal Transduction , Animals , Janus Kinase 2/metabolism , STAT3 Transcription Factor/metabolism , Drugs, Chinese Herbal/pharmacology , Mice , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Myasthenia Gravis/drug therapy , Myasthenia Gravis/immunology , Female , Inflammation/drug therapy , Inflammation/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Myasthenia Gravis, Autoimmune, Experimental/immunology , SKP Cullin F-Box Protein Ligases/metabolism , Tripartite Motif Proteins/metabolism , Reactive Oxygen Species/metabolism , Muscle Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
3.
J Neuroinflammation ; 18(1): 244, 2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34702288

ABSTRACT

BACKGROUND: Diabetes mellitus (DM) is a common concomitant disease of late-onset myasthenia gravis (MG). However, the impacts of DM on the progression of late-onset MG were unclear. METHODS: In this study, we examined the immune response in experimental autoimmune myasthenia gravis (EAMG) rats with DM or not. The phenotype and function of the spleen and lymph nodes were determined by flow cytometry. The serum antibodies, Tfh cells, and germinal center B cells were determined by ELISA and flow cytometry. The roles of advanced glycation end products (AGEs) in regulating Tfh cells were further explored in vitro by co-culture assays. RESULTS: Our results indicated clinical scores of EAMG rats were worse in diabetes rats compared to control, which was due to the increased production of anti-R97-116 antibody and antibody-secreting cells. Furthermore, diabetes induced a significant upregulation of Tfh cells and the subtypes of Tfh1 and Tfh17 cells to provide assistance for antibody production. The total percentages of B cells were increased with an activated statue of improved expression of costimulatory molecules CD80 and CD86. We found CD4+ T-cell differentiation was shifted from Treg cells towards Th1/Th17 in the DM+EAMG group compared to the EAMG group. In addition, in innate immunity, diabetic EAMG rats displayed more CXCR5 expression on NK cells. However, the expression of CXCR5 on NKT cells was down-regulated with the increased percentages of NKT cells in the DM+EAMG group. Ex vivo studies further indicated that Tfh cells were upregulated by AGEs instead of hyperglycemia. The upregulation was mediated by the existence of B cells, the mechanism of which might be attributed the elevated molecule CD40 on B cells. CONCLUSIONS: Diabetes promoted both adaptive and innate immunity and exacerbated clinical symptoms in EAMG rats. Considering the effect of diabetes, therapy in reducing blood glucose levels in MG patients might improve clinical efficacy through suppressing the both innate and adaptive immune responses. Additional studies are needed to confirm the effect of glucose or AGEs reduction to seek treatment for MG.


Subject(s)
Adaptive Immunity/physiology , Diabetes Mellitus, Experimental/immunology , Immunity, Innate/physiology , Inflammation Mediators/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Coculture Techniques , Diabetes Mellitus, Experimental/metabolism , Female , Inflammation Mediators/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Rats , Rats, Inbred Lew , Th17 Cells/immunology , Th17 Cells/metabolism
4.
Immunol Invest ; 50(6): 671-684, 2021 Aug.
Article in English | MEDLINE | ID: mdl-32597289

ABSTRACT

Antigen-specific immune responses are crucially involved in both multiple sclerosis (MS) and myasthenia gravis (MG). Teriflunomide is an immunomodulatory agent approved for treatment of MS through inhibition of lymphocyte proliferation. MG associated with muscle-specific tyrosine kinase (MuSK) antibodies often manifests with a severe disease course, prompting development of effective treatment methods. To evaluate whether teriflunomide treatment may ameliorate MuSK-autoimmunity, experimental autoimmune MG (EAMG) was induced by immunizing C57BL/6 (B6) mice three times with MuSK in complete Freund's adjuvant (CFA) (n = 17). MuSK-immunized mice were treated daily with teriflunomide (n = 8) or PBS (n = 9) starting from the third immunization (week 8) to termination (week 14). Clinical severity of EAMG was monitored. Immunological alterations were evaluated by measurement of anti-MuSK IgG, neuromuscular junction deposits, and flow cytometric analysis of lymph node cells. In MS patients under teriflunomide treatment, the peripheral blood B cell subset profile was analyzed. B6 mice treated with teriflunomide displayed relatively preserved body weight, lower EAMG prevalence, reduced average clinical grades, higher inverted screen scores, diminished anti-MuSK antibody and NMJ deposit levels. Amelioration of EAMG findings was associated with reduced memory B cell ratios in the lymph nodes. Similarly, MS patients under teriflunomide treatment showed reduced memory B cell, plasma cell, and plasmablast ratios. Teriflunomide treatment has effectively ameliorated MuSK-autoimmunity and thus may putatively be used in long-term management of MuSK-MG as an auxiliary treatment method. Teriflunomide appears to exert beneficial effects through inhibition of effector B cells.


Subject(s)
B-Lymphocyte Subsets/drug effects , Crotonates/administration & dosage , Hydroxybutyrates/administration & dosage , Multiple Sclerosis/drug therapy , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Nitriles/administration & dosage , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Cholinergic/immunology , Toluidines/administration & dosage , Adult , Animals , B-Lymphocyte Subsets/immunology , Female , Humans , Male , Mice , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/diagnosis , Multiple Sclerosis/immunology , Myasthenia Gravis, Autoimmune, Experimental/blood , Myasthenia Gravis, Autoimmune, Experimental/diagnosis , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptor Protein-Tyrosine Kinases/administration & dosage , Receptors, Cholinergic/administration & dosage , Treatment Outcome
5.
J Neuroinflammation ; 17(1): 294, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33032631

ABSTRACT

BACKGROUND: Myasthenia gravis (MG) is a rare autoimmune disease mainly mediated by autoantibodies against the acetylcholine receptor (AChR) at the neuromuscular junction. The thymus is the effector organ, and its removal alleviates the symptoms of the disease. In the early-onset form of MG, the thymus displays functional and morphological abnormalities such as B cell infiltration leading to follicular hyperplasia, and the production of AChR antibodies. Type-I interferon (IFN-I), especially IFN-ß, is the orchestrator of thymic changes observed in MG. As Dicer and miR-29 subtypes play a role in modulating the IFN-I signalization in mouse thymus, we investigated their expression in MG thymus. METHODS: The expression of DICER and miR-29 subtypes were thoroughly investigated by RT-PCR in human control and MG thymuses, and in thymic epithelial cells (TECs). Using miR-29a/b-1-deficient mice, with lower miR-29a/b-1 expression, we investigated their susceptibility to experimental autoimmune MG (EAMG) as compared to wild-type mice. RESULTS: DICER mRNA and all miR-29 subtypes were down-regulated in the thymus of MG patients and DICER expression was correlated with the lower expression of miR-29a-3p. A decreased expression of miR-29 subtypes was similarly observed in MG TECs; a decrease also induced in TECs upon IFN-ß treatment. We demonstrated that miR-29a/b-1-deficient mice were more susceptible to EAMG without higher levels of anti-AChR IgG subtypes. In the thymus, if no B cell infiltration was observed, an increased expression of Ifn-ß associated with Baff expression and the differentiation of Th17 cells associated with increased expression of Il-6, Il-17a and Il-21 and decreased Tgf-ß1 mRNA were demonstrated in miR-29a/b-1-deficient EAMG mice. CONCLUSIONS: It is not clear if the decreased expression of miR-29 subtypes in human MG is a consequence or a causative factor of thymic inflammation. However, our results from the EAMG mouse model indicated that a reduction in miR-29a/b1 may contribute to the pathophysiological process involved in MG by favoring the increased expression of IFN-ß and the emergence of pro-inflammatory Th17 cells.


Subject(s)
MicroRNAs/biosynthesis , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Myasthenia Gravis/metabolism , Adolescent , Adult , Animals , Autoantibodies/immunology , Autoantibodies/metabolism , Cells, Cultured , Female , Gene Expression , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Myasthenia Gravis/genetics , Myasthenia Gravis/immunology , Myasthenia Gravis, Autoimmune, Experimental/genetics , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptors, Cholinergic/immunology , Receptors, Cholinergic/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Young Adult
6.
Med Sci Monit ; 26: e924393, 2020 Sep 13.
Article in English | MEDLINE | ID: mdl-32920588

ABSTRACT

BACKGROUND Myasthenia gravis (MG) is an autoimmune neurological disorder of neuromuscular junctions. In this study we established experimental autoimmune myasthenia gravis (EAMG) rat models to investigate the effects of AEB-071 (AEB), which is a specific inhibitor of protein kinase C that prevents T lymphocyte activation. MATERIAL AND METHODS We utilized animals divided into 4 groups: (1) control rats, (2) EAMG, (3) AEB-071+EAMG, and (4) AZP+EAMG. Drug treatment was continued for 10 days. Ten weeks after immunization we measured body weights, assessed mortality rates, and used Lennon scores to evaluate EAMG grades. We also assessed the proportions of Treg, Th1, Th2, Th17, and lymphocytes using flow cytometry. RESULTS In the absence of drug treatment, we found a significant decline in body weights in the EAMG group in comparison to control rats, and EAMG group rats also had higher Lennon scores (P<0.05). Interestingly, we found that AEB-071 restored the body weight of EAMG rats and the decreased mortality rate compared to AZP treatment. Although a decrease in the number of Treg cells was observed, the proportion of Th lymphocytes was significantly increased in the EAMG group, and AEB-071 treatment decreased the proportion of Th lymphocytes. CONCLUSIONS We concluded that AEB-071 treatment imparts beneficial effects in EAMG rat models by reducing mortality rate and restoring Th lymphocyte balance, and thus may be an attractive candidate for use in MG treatment.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental/immunology , Pyrroles/pharmacology , Quinazolines/pharmacology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Animals , Female , Lymphocyte Activation/drug effects , Muscle Weakness/etiology , Rats , Rats, Inbred Lew
7.
Med Sci Monit ; 26: e919150, 2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32052794

ABSTRACT

BACKGROUND Myasthenia gravis (MG) is a progressive autoimmune disorder caused by the production of antibodies directed against acetylcholine receptors (AChRs), resulting in muscle weakness and fatigue. This study aimed to explore the effect and mechanism of grilled nux vomica (GNV) in experimental autoimmune myasthenia gravis (EAMG) rats. MATERIAL AND METHODS Rat 97-116 peptides were used to mediate disease in the EAMG model in SPF female Lewis rats. The treatment groups received grilled nux vomica (75 mg/kg, 150 mg/kg, and 225 mg/kg). The autoantibody and inflammatory cytokines levels were measured by enzyme-linked immunosorbent assay (ELISA). RNA profiling was performed on high-dose and model group rats. Profiling results and TLR-4/NF-kappaB signaling were validated by q-PCR and Western blot analysis. RESULTS The results showed that GNV could attenuate the symptoms of EAMG rats. There was a decreased level of AChR-ab, IFN-γ, TNF-alpha, IL-2, IL-4, and IL-17 levels, and an increased level of TGF-ß1. In total, 235 differentially expressed genes (DEGs), consisting of 175 upregulated DEGs and 60 downregulated DEGs, were identified. Functional annotation demonstrated that DEGs were largely associated with leukocyte cell-cell adhesion, NF-kappa B signaling pathway, muscle contraction, and cardiac muscle contraction pathway. Rac2, Itgb2, Lcp2, Myl3, and Tnni1 were considered as hub genes with a higher degree value in the protein-protein interaction (PPI) network. The q-PCR and Western blot results of hub genes were consistent with RNA profiles. GNV treatment also significantly reduced the TLR-4 and NF-kappaB p65 protein expression in EAMG rats. CONCLUSIONS These results indicate that grilled nux vomica ameliorates EAMG by depressing the TLR-4/NF-kappaB signaling pathway, and hub genes may serve as potential targets for MG treatment.


Subject(s)
Drugs, Chinese Herbal/administration & dosage , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , RNA, Messenger/metabolism , Signal Transduction/drug effects , Strychnos nux-vomica/chemistry , Animals , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/pathology , NF-kappa B/metabolism , RNA-Seq , Rats , Rats, Inbred Lew , Signal Transduction/genetics , Signal Transduction/immunology , Specific Pathogen-Free Organisms , Toll-Like Receptor 4/metabolism
8.
Eur J Immunol ; 48(3): 498-508, 2018 03.
Article in English | MEDLINE | ID: mdl-29205338

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease characterized by muscle weakness and fatigue in the presence of circulating antibodies against components of the neuromuscular junction. Most patients have a good prognosis, but some are refractory to standard-of-care immunosuppressive treatment and suffer from recurrent myasthenic crises. Functional sphingosine-1-phosphate (S1P) antagonists like fingolimod and siponimod (BAF312) are successfully used for the treatment of multiple sclerosis, and fingolimod was shown to prevent the development of myasthenic symptoms in experimental autoimmune myasthenia gravis (EAMG), the standard model of MG. Here, we investigated whether fingolimod or siponimod improves outcome in EAMG mice when administered after disease onset, modeling the clinical setting in human MG. Both S1P antagonists inhibited lymphocyte egress, resulting in peripheral lymphopenia. After stimulation, there were differences in T-cell responses, but no change in either antibody titers or total or antigen-specific plasma cell populations after treatment. Most importantly, disease incidence and severity were not influenced by fingolimod or siponimod therapy. Although fingolimod and siponimod did lead to subtle changes in T-cell responses, they had no significant effect on antibody titers and disease severity. In conclusion, our data show no evidence of a therapeutic potential for S1P receptor antagonists in MG treatment.


Subject(s)
Azetidines/pharmacology , Benzyl Compounds/pharmacology , Fingolimod Hydrochloride/pharmacology , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Receptors, Lysosphingolipid/antagonists & inhibitors , Animals , Antibody Formation/drug effects , Cytokines/biosynthesis , Female , Humans , Immunosuppressive Agents/pharmacology , Lymphopenia/chemically induced , Male , Mice , Mice, Inbred C57BL , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Plasma Cells/drug effects , Receptors, Lysosphingolipid/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
9.
J Neuroinflammation ; 16(1): 282, 2019 Dec 29.
Article in English | MEDLINE | ID: mdl-31884963

ABSTRACT

BACKGROUND: Recent studies have demonstrated that natural killer (NK) cells can modulate other immune components and are involved in the development or progression of several autoimmune diseases. However, the roles and mechanisms of NK cells in regulating experimental autoimmune myasthenia gravis (EAMG) remained to be illustrated. METHODS: To address the function of NK cells in experimental autoimmune myasthenia gravis in vivo, EAMG rats were adoptively transferred with splenic NK cells. The serum antibodies, and splenic follicular helper T (Tfh) cells and germinal center B cells were determined by ELISA and flow cytometry. The roles of NK cells in regulating Tfh cells were further verified in vitro by co-culturing splenocytes or isolated T cells with NK cells. Moreover, the phenotype, localization, and function differences between different NK cell subtypes were determined by flow cytometry, immunofluorescence, and ex vivo co-culturation. RESULTS: In this study, we found that adoptive transfer of NK cells ameliorated EAMG symptoms by suppressing Tfh cells and germinal center B cells. Ex vivo studies indicated NK cells inhibited CD4+ T cells and Tfh cells by inducing the apoptosis of T cells. More importantly, NK cells could be divided into CXCR5- and CXCR5+ NK subtypes according to the expression of CXCR5 molecular. Compared with CXCR5- NK cells, which were mainly localized outside B cell zone, CXCR5+ NK were concentrated in the B cell zone and exhibited higher expression levels of IL-17 and ICOS, and lower expression level of CD27. Ex vivo studies indicated it was CXCR5- NK cells not CXCR5+ NK cells that suppressed CD4+ T cells and Tfh cells. Further analysis revealed that, compared with CXCR5- NK cells, CXCR5+ NK cells enhanced the ICOS expression of Tfh cells. CONCLUSIONS: These findings highlight the different roles of CXCR5- NK cells and CXCR5+ NK cells. It was CXCR5- NK cells but not CXCR5+ NK cells that suppressed Tfh cells and inhibited the autoimmune response in EAMG models.


Subject(s)
Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptors, CXCR5/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Female , Mice , Rats , Rats, Inbred Lew
10.
J Neuroinflammation ; 16(1): 202, 2019 Nov 03.
Article in English | MEDLINE | ID: mdl-31679515

ABSTRACT

BACKGROUND: The thymus plays an essential role in the pathogenesis of myasthenia gravis (MG). In patients with MG, natural regulatory T cells (nTreg), a subpopulation of T cells that maintain tolerance to self-antigens, are severely impaired in the thymuses. In our previous study, upregulated nTreg cells were observed in the thymuses of rats in experimental autoimmune myasthenia gravis after treatment with exosomes derived from statin-modified dendritic cells (statin-Dex). METHODS: We evaluated the effects of exosomes on surface co-stimulation markers and Aire expression of different kinds of thymic stromal cells, including cTEC, mTEC, and tDCs, in EAMG rats. The isolated exosomes were examined by western blot and DLS. Immunofluorescence was used to track the exosomes in the thymus. Flow cytometry and western blot were used to analyze the expression of co-stimulatory molecules and Aire in vivo and in vitro. RESULTS: We confirmed the effects of statin-Dex in inducing Foxp3+ nTreg cells and found that both statin-Dex and DMSO-Dex could upregulate CD40 but only statin-Dex increased Aire expression in thymic stromal cells in vivo. Furthermore, we found that the role of statin-Dex and DMSO-Dex in the induction of Foxp3+ nTreg cells was dependent on epithelial cells in vitro. CONCLUSIONS: We demonstrated that statin-Dex increased expression of Aire in the thymus, which may further promote the Foxp3 expression in the thymus. These findings may provide a new strategy for the treatment of myasthenia gravis.


Subject(s)
Exosomes/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myasthenia Gravis, Autoimmune, Experimental/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Atorvastatin/pharmacology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/metabolism , Female , Rats , Rats, Inbred Lew , T-Lymphocytes, Regulatory/cytology , Thymus Gland , Transcription Factors/metabolism , AIRE Protein
11.
J Immunol ; 199(9): 3051-3062, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28972088

ABSTRACT

The transcription factor NFAT1 plays a pivotal role in the homeostasis of T lymphocytes. However, its functional importance in non-CD4+ T cells, especially in systemic immune disorders, is largely unknown. In this study, we report that NFAT1 regulates dendritic cell (DC) tolerance and suppresses systemic autoimmunity using the experimental autoimmune myasthenia gravis (EAMG) as a model. Myasthenia gravis and EAMG are T cell-dependent, Ab-mediated autoimmune disorders in which the acetylcholine receptor is the major autoantigen. NFAT1-knockout mice showed higher susceptibility to EAMG development with enhanced Th1/Th17 cell responses. NFAT1 deficiency led to a phenotypic alteration of DCs that show hyperactivation of NF-κB-mediated signaling pathways and enhanced binding of NF-κB (p50) to the promoters of IL-6 and IL-12. As a result, NFAT1-knockout DCs produced much higher levels of proinflammatory cytokines such as IL-1ß, IL-6, IL-12, and TNF-α, which preferentially induce Th1/Th17 cell differentiation. Our data suggest that NFAT1 may limit the hyperactivation of the NF-κB-mediated proinflammatory response in DCs and suppress autoimmunity by serving as a key regulator of DC tolerance.


Subject(s)
Dendritic Cells/immunology , Lymphocyte Activation , Myasthenia Gravis, Autoimmune, Experimental/immunology , NFATC Transcription Factors/immunology , Signal Transduction/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Immune Tolerance/genetics , Mice , Mice, Transgenic , Myasthenia Gravis, Autoimmune, Experimental/genetics , Myasthenia Gravis, Autoimmune, Experimental/pathology , NF-kappa B/genetics , NF-kappa B/immunology , NFATC Transcription Factors/genetics , Signal Transduction/genetics , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
12.
Immunol Invest ; 47(7): 712-724, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29944018

ABSTRACT

Interleukin-9 (IL-9) is a multifunctional cytokine involved in protective immunity or immunopathology depending on the microenvironment and specific disease settings. Our early study determined that IL-9 and Th9 cells participate in and promote the progression of experimental autoimmune myasthenia gravis (EAMG). The data from this study showed that exogenous recombinant rat IL-9 (rrIL-9) acted as an IL-9 receptor antagonist, reduced the incidence of EAMG in rats, alleviated the severity of the disease, and reduced the anti-acetylcholine receptor (AChR) IgG antibody levels by altering the Th-subset distribution. These data suggest that administration of rrIL-9 may provide a novel therapeutic strategy against MG or related autoimmune diseases. Abbreviations: 2-Mercaptoethanol (2-ME); antibodies (Abs); ?-bungarotoxin (?-BTX); acetylcholine receptor (AChR); airway hyper-reactivity (AHR); allophycocyanin-conjugated (APC); antigen presenting cells (APCs); complete Freund's adjuvant (CFA); Cyanine dye 3 (Cy3); dendritic cells (DCs); experimental autoimmune encephalomyelitis (EAE); experimental autoimmune myasthenia gravis (EAMG); flow cytometry (FACS); fetal bovine serum (FBS); fetal calf serum (FCS); Fluorescein isothiocyanate (FITC); gamma chain (?c); intraperitoneally (i.p.); Incomplete Freund's adjuvant (IFA); interferon (IFN); immunoglobulin (Ig); Interleukin (IL); Janus kinase (JAK); myasthenia gravis (MG); Mononuclear cells (MNC); neuromuscular junctions (NMJ); optical density (OD); ovalbumin (OVA); phosphate-buffered saline (PBS); phycoerythrin (PE); Peridinin chlorophyll protein complex (Percp); Rat AChR ? subunit (R-AChR97-116); Recombinant Rat (rr); room temperature (RT); signal transducer and activator of transcription (STAT); T helper cells (Th).


Subject(s)
Immunotherapy/methods , Interleukin-9/immunology , Myasthenia Gravis, Autoimmune, Experimental/therapy , Myasthenia Gravis/therapy , Recombinant Proteins/immunology , Animals , Autoantibodies/blood , Autoantigens/immunology , Female , Humans , Interleukin-9/therapeutic use , Myasthenia Gravis/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Peptides/immunology , Rats , Rats, Inbred Lew , Receptors, Cholinergic/immunology , Receptors, Interleukin-9/antagonists & inhibitors , Recombinant Proteins/therapeutic use
13.
J Biol Regul Homeost Agents ; 32(5): 1217-1223, 2018.
Article in English | MEDLINE | ID: mdl-30334416

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease characterized by fatigue and muscle weakness. Artemisinin and its derivatives were reported to be experimentally used to treat autoimmune diseases, such as systemic lupus erythematosus (SLE) and experimental allergic encephalomyelitis (EAE). Here, we tested the effects of artemisinin on experimental autoimmune myasthenia gravis (EAMG). Our data confirmed that artemisinin markedly ameliorated the symptoms of EAMG rats. There was a decreased level of tumor necrosis factor-α (TNF-α) and IL-17+ cells in mononuclear cells (MNCs), and an increased level of transforming growth factor-ß1 (TGF-ß1) and Treg cells in MNCs. These findings indicate that artemisinin may be a new choice for MG treatment.


Subject(s)
Artemisinins/therapeutic use , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Myasthenia Gravis, Autoimmune, Experimental/immunology , T-Lymphocytes, Regulatory/drug effects , Th1 Cells/drug effects , Th17 Cells/drug effects , Th17 Cells/immunology , Animals , Artemisinins/pharmacology , Rats , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology
14.
Pharmazie ; 73(9): 526-532, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30223936

ABSTRACT

Artesunate is a semi-synthetic derivative of a Chinese herb named Artemisia annua L. that is commonly used as an antimalarial agent in the history of traditional Chinese medicine. Many studies have reported artesunate possesses anti-inflammatory and immunoregulation properties. The present study was conducted to explore whether artesunate was effective in experimental autoimmune myasthenia gravis (EAMG) in Lewis rats. Our data showed that artesunate could improve the clinical symptoms and suppress the development of EAMG. Artesunate exerted its immunomodulatory effects by inhibiting lymphocyte proliferation and the expression of costimulatory molecules CD86, modulating Th1/Th2 cytokine expression levels, and enhancing the level of Treg cells. The final result of administration of artesunate was the decreased synthesis of anti-R97-116 IgG, IgG2a, and IgG2b antibodies. The treatment effect of artesunate was more obvious at dose of 10 mg/kg. These date suggest that artesunate might be a potential drug for the treatment of human myasthenia gravis (MG).


Subject(s)
Artesunate/pharmacology , Immunologic Factors/pharmacology , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , T-Lymphocytes, Regulatory/immunology , Animals , Antimalarials/administration & dosage , Antimalarials/pharmacology , Artesunate/administration & dosage , Cell Proliferation/drug effects , Cytokines/immunology , Dose-Response Relationship, Drug , Female , Immunologic Factors/administration & dosage , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/physiopathology , Rats , Rats, Inbred Lew , Th1 Cells/immunology , Th2 Cells/immunology , Up-Regulation
15.
Clin Immunol ; 183: 121-131, 2017 10.
Article in English | MEDLINE | ID: mdl-28822831

ABSTRACT

Abnormal CD4+T cell activation is known to play roles in the pathogenesis of myasthenia gravis (MG). However, little is known about the mechanisms underlying the roles of lncRNAs in regulating CD4+ T cell. In this study, we discovered that the lncRNA IFNG-AS1 is abnormally expressed in MG patients associated with quantitative myasthenia gravis (QMG) and the positive anti-AchR Ab levels patients. IFNG-AS1 influenced Th1/Treg cell proliferation and regulated the expression levels of their transcription factors in an experimental autoimmune myasthenia gravis (EAMG)model. IFNG-AS1 could reduce the expression of HLA-DRB and HLA-DOB and they had a negative correlation in MG. Furthermore IFNG-AS1 influenced the expression levels of CD40L and CD4+ T cells activation in MG patient partly depend on effecting the HLA-DRB1 expression. It suggests that IFNG-AS1 may be involved in CD4+T cell-mediated immune responses in MG.


Subject(s)
Cell Proliferation , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis/immunology , RNA, Long Noncoding/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Adolescent , Adult , Animals , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/immunology , Case-Control Studies , Female , HLA-D Antigens/immunology , HLA-DRB1 Chains/immunology , Humans , Male , Mice , Middle Aged , RNA, Long Noncoding/pharmacology , Young Adult
16.
J Neuroinflammation ; 14(1): 117, 2017 06 09.
Article in English | MEDLINE | ID: mdl-28599652

ABSTRACT

Multiple sclerosis (MS) is a chronic debilitating disease of the central nervous system primarily mediated by T lymphocytes with specificity to neuronal antigens in genetically susceptible individuals. On the other hand, myasthenia gravis (MG) primarily involves destruction of the neuromuscular junction by antibodies specific to the acetylcholine receptor. Both autoimmune diseases are thought to result from loss of self-tolerance, which allows for the development and function of autoreactive lymphocytes. Although the mechanisms underlying compromised self-tolerance in these and other autoimmune diseases have not been fully elucidated, one possibility is numerical, functional, and/or migratory deficits in T regulatory cells (Tregs). Tregs are thought to play a critical role in the maintenance of peripheral immune tolerance. It is believed that Tregs function by suppressing the effector CD4+ T cell subsets that mediate autoimmune responses. Dysregulation of suppressive and migratory markers on Tregs have been linked to the pathogenesis of both MS and MG. For example, genetic abnormalities have been found in Treg suppressive markers CTLA-4 and CD25, while others have shown a decreased expression of FoxP3 and IL-10. Furthermore, elevated levels of pro-inflammatory cytokines such as IL-6, IL-17, and IFN-γ secreted by T effectors have been noted in MS and MG patients. This review provides several strategies of treatment which have been shown to be effective or are proposed as potential therapies to restore the function of various Treg subsets including Tr1, iTr35, nTregs, and iTregs. Strategies focusing on enhancing the Treg function find importance in cytokines TGF-ß, IDO, interleukins 10, 27, and 35, and ligands Jagged-1 and OX40L. Likewise, strategies which affect Treg migration involve chemokines CCL17 and CXCL11. In pre-clinical animal models of experimental autoimmune encephalomyelitis (EAE) and experimental autoimmune myasthenia gravis (EAMG), several strategies have been shown to ameliorate the disease and thus appear promising for treating patients with MS or MG.


Subject(s)
Multiple Sclerosis/immunology , Multiple Sclerosis/therapy , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/therapy , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Immune Tolerance/drug effects , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunotherapy/methods , Immunotherapy/trends , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Multiple Sclerosis/metabolism , Myasthenia Gravis, Autoimmune, Experimental/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
17.
Am J Pathol ; 186(10): 2559-68, 2016 10.
Article in English | MEDLINE | ID: mdl-27658713

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease mediated by autoantibodies that target proteins at the neuromuscular junction, primarily the acetylcholine receptor (AChR) and the muscle-specific kinase. Because downstream of kinase 7 (Dok-7) is essential for the full activation of muscle-specific kinase and consequently for dense clustering of AChRs, we hypothesized that reduced levels of Dok-7 increase the susceptibility to passive transfer MG. To test this hypothesis, Dok-7 expression was reduced by transfecting shRNA-coding plasmids into the tibialis anterior muscle of adult rats by in vivo electroporation. Subclinical MG was subsequently induced with a low dose of anti-AChR monoclonal antibody 35. Neuromuscular transmission was significantly impaired in Dok-7-siRNA-electroporated legs compared with the contralateral control legs, which correlated with a reduction of AChR protein levels at the neuromuscular junction (approximately 25%) in Dok-7-siRNA-electroporated muscles, compared with contralateral control muscles. These results suggest that a reduced expression of Dok-7 may play a role in the susceptibility to passive transfer MG, by rendering AChR clusters less resistant to the autoantibody attack.


Subject(s)
Autoantibodies/immunology , Muscle Proteins/genetics , Myasthenia Gravis, Autoimmune, Experimental/genetics , Animals , Disease Models, Animal , Disease Susceptibility , Down-Regulation , Female , Gene Silencing , Genes, Reporter , HEK293 Cells , Humans , Muscle Proteins/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/physiopathology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/physiopathology , Neuromuscular Junction/immunology , Neuromuscular Junction/physiopathology , Rats , Rats, Inbred Lew , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Cholinergic/genetics , Receptors, Cholinergic/metabolism , Synaptic Transmission
18.
Cytokine ; 96: 279-285, 2017 08.
Article in English | MEDLINE | ID: mdl-28599246

ABSTRACT

An array of cytokines influences the pathogenesis of early onset myasthenia gravis (MG) and its animal model, experimental autoimmune myasthenia gravis (EAMG). Patients with MG, in particular those with more severe weakness, have elevations of the pro-inflammatory cytokine IL-17 in the blood. We assessed the role of IL-17A in autoimmunity by inducing EAMG in mice with knockout of IL-17 and found a reduction of EAMG severity, but not a complete ablation of disease. The IL-17ko mice had no evidence of weakness, low levels of acetylcholine receptor antibodies, and retention of acetylcholine receptor at the neuromuscular junction. Splenic germinal center size was reduced in EAMG IL-17ko mice along with elevations of Foxp3 and BCL-6 gene expression, suggesting a shift away from pro-inflammatory signals. The results emphasize the importance of IL-17 in EAMG development and that IL-17 independent pathways drive the autoimmune reaction.


Subject(s)
Autoimmunity/immunology , Interleukin-17/genetics , Interleukin-17/physiology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/physiopathology , Animals , Autoantibodies , Cytokines/immunology , Disease Models, Animal , Forkhead Transcription Factors/genetics , Humans , Immunoglobulin G/blood , Interleukin-17/deficiency , Mice , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6/genetics , Real-Time Polymerase Chain Reaction , Severity of Illness Index , T-Lymphocytes, Regulatory
19.
Immunol Invest ; 46(5): 490-499, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28375749

ABSTRACT

BACKGROUND: Myasthenia gravis (MG) is an autoimmune disease of the neuromuscular junction (NMJ), characterized with muscle weakness. While MG develops due to acetylcholine receptor (AChR) antibodies in most patients, antibodies to muscle-specific receptor tyrosine kinase (MuSK) or low-density lipoprotein receptor-related protein 4 (LRP4) may also be identified. Experimental autoimmune myasthenia gravis (EAMG) has been previously induced by both LRP4 immunization and passive transfer of LRP4 antibodies. OBJECTIVE: Our aim was to confirm previous results and to test the pathogenic effects of LRP4 immunization in a commonly used mouse strain C57BL/6 (B6) using a recombinantly expressed human LRP4 protein. METHODS: B6 mice were immunized with human LRP4 in CFA, Torpedo Californica AChR in CFA or only CFA. Clinical and pathogenic aspects of EAMG were compared among groups. RESULTS: LRP4- and AChR-immunized mice showed comparable EAMG clinical severity. LRP4-immunized mice displayed serum antibodies to LRP4 and NMJ IgG and complement factor C3 deposits. IgG2 was the dominant anti-LRP4 isotype. Cultured lymph node cells of LRP4- and AChR-immunized mice gave identical pro-inflammatory cytokine (IL-6, IFN-γ and IL-17) responses to LRP4 and AChR stimulation, respectively. CONCLUSION: Our results confirm the EAMG-inducing action of LRP4 immunization and identify B6 as a LRP4-EAMG-susceptible mouse strain. Demonstration of complement fixing anti-LRP4 antibodies in sera and complement/IgG deposits at the NMJ of LRP4-immunized mice indicates complement activation as a putative pathogenic mechanism. We have thus developed a practical LRP4-induced EAMG model using a non-conformational protein and a widely available mouse strain for future investigation of LRP4-related MG.


Subject(s)
Complement Activation/drug effects , Immunization/methods , Immunoglobulin G/biosynthesis , LDL-Receptor Related Proteins/administration & dosage , Myasthenia Gravis, Autoimmune, Experimental/immunology , Receptors, Cholinergic/administration & dosage , Animals , Complement C3/metabolism , Freund's Adjuvant/administration & dosage , Humans , Immunoglobulin Isotypes/biosynthesis , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , LDL-Receptor Related Proteins/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/pathology , Male , Mice , Mice, Inbred C57BL , Myasthenia Gravis, Autoimmune, Experimental/chemically induced , Myasthenia Gravis, Autoimmune, Experimental/pathology , Primary Cell Culture , Receptors, Cholinergic/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Severity of Illness Index , Torpedo/metabolism
20.
Eur J Immunol ; 45(5): 1339-47, 2015 May.
Article in English | MEDLINE | ID: mdl-25676041

ABSTRACT

The role of Th17 cells in the pathogenesis of autoantibody-mediated diseases is unclear. Here, we assessed the contribution of Th17 cells to the pathogenesis of experimental autoimmune myasthenia gravis (EAMG), which is induced by repetitive immunizations with Torpedo californica acetylcholine receptor (tAChR). We show that a significant fraction of tAChR-specific CD4(+) T cells is producing IL-17. IL-17(ko) mice developed fewer or no EAMG symptoms, although the frequencies of tAChR-specific CD4(+) T cells secreting IL-2, IFN-γ, or IL-21, and the percentage of FoxP3(+) Treg cells were similar to WT mice. Even though the total anti-tAChR antibody levels were equal, the complement fixating IgG2b subtype was reduced in IL-17(ko) as compared to WT mice. Most importantly, pathogenic anti-murine AChR antibodies were significantly lower in IL-17(ko) mice. Furthermore, we confirmed the role of Th17 cells in EAMG pathogenesis by the reconstitution of TCR ß/δ(ko) mice with WT or IL-17(ko) CD4(+) T cells. In conclusion, we show that the level of IgG2b and the loss of B-cell tolerance, which results in pathogenic anti-murine AChR-specific antibodies, are dependent on IL-17 production by CD4(+) T cells. Thus, we describe here for the first time how Th17 cells are involved in the induction of classical antibody-mediated autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Interleukin-17/biosynthesis , Myasthenia Gravis, Autoimmune, Experimental/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , Antigens/administration & dosage , Immune Tolerance , Immunoglobulin G/biosynthesis , Interleukin-17/deficiency , Interleukin-17/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myasthenia Gravis, Autoimmune, Experimental/etiology , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/deficiency , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Cholinergic/immunology , Torpedo/immunology
SELECTION OF CITATIONS
SEARCH DETAIL