321 results on '"Myasthenia Gravis, Autoimmune, Experimental"'
Search Results
2. Effect of Core Strength Training on Wushu C-level Difficulty Movements ,12 Weeks of Core Strength Training
- Author
-
Li Long, Principal Investigator
- Published
- 2021
3. Metformin inhibits the pathogenic functions of AChR-specific B and Th17 cells by targeting miR-146a
- Author
-
Yue Hao, Wei Zhao, Lulu Chang, Xingfan Chen, Chonghui Liu, Yang Liu, Lixuan Hou, Yinchun Su, Hao Xu, Yu Guo, Qixu Sun, Lili Mu, Jinghua Wang, Hulun Li, Junwei Han, and Qingfei Kong
- Subjects
B-Lymphocytes ,MicroRNAs ,Immunology ,Animals ,Th17 Cells ,Immunology and Allergy ,Receptors, Cholinergic ,Biomarkers ,Metformin ,Autoantibodies ,Myasthenia Gravis, Autoimmune, Experimental ,Rats - Abstract
Myasthenia gravis (MG) is characterized by fatigable skeletal muscle weakness with a fluctuating and unpredictable disease course and is caused by circulating autoantibodies and pathological T helper cells. Regulation of B-cell function and the T-cell network may be a potential therapeutic strategy for MG. MicroRNAs (miRNAs) have emerged as potential biomarkers in immune disorders due to their critical roles in various immune cells and multiple inflammatory diseases. Aberrant miR-146a signal activation has been reported in autoimmune diseases, but a detailed exploration of the relationship between miR-146a and MG is still necessary. Using an experimental autoimmune myasthenia gravis (EAMG) rat model, we observed that miR-146a was highly expressed in the spleen but expressed at low levels in the thymus and lymph nodes in EAMG rats. Additionally, miR-146a expression in T and B cells was also quite different. EAMG-specific Th17 and Treg cells had lower miR-146a levels, while EAMG-specific B cells had higher miR-146a levels, indicating that targeted intervention against miR-146a might have diametrically opposite effects. Metformin, a drug that was recently demonstrated to alleviate EAMG, may rescue the functions of both Th17 cells and B cells by reversing the expression of miR-146a. We also investigated the downstream target genes of miR-146a in both T and B cells using bioinformatics screening and qPCR. Taken together, our study identifies a complex role of miR-146a in the EAMG rat model, suggesting that more caution should be paid in targeting miR-146a for the treatment of MG.
- Published
- 2022
- Full Text
- View/download PDF
4. Silencing of FCRLB by shRNA ameliorates MuSK-induced EAMG in mice.
- Author
-
Koral G, Ulusoy C, Cossins J, Lazaridis K, Türkoğlu R, Dong YY, Tüzün E, and Yılmaz V
- Subjects
- Humans, Mice, Animals, Receptor Protein-Tyrosine Kinases, Neuromuscular Junction, Immunization methods, Autoantibodies, Immunoglobulin G, Myasthenia Gravis, Autoimmune, Experimental
- Abstract
Introduction: Muscle specific kinase (MuSK) antibody positive myasthenia gravis (MG) often presents with a severe disease course and resistance to treatment. Treatment-refractory patients may respond to B cell depleting treatment methods. Our aim was to investigate whether inhibition of Fc receptor-like B (FCRLB) could effectively suppress autoimmunity without diminishing B cell counts in animal model of MG, a classical antibody-mediated autoimmune disease., Methods: Experimental autoimmune MG was induced in Balb/C mice with two s.c. immunizations with recombinant human MuSK in complete Freund's adjuvant. FCRLB was silenced with a lentiviral particle transported shRNA in myasthenic mice with a single i.p. injection during second MuSK-immunization. Control immunized mice received scrambled shRNA or saline. Mice were observed for clinical parameters for 28 days and at termination, anti-MuSK IgG, neuromuscular junction (NMJ) deposits, muscle AChR expression and lymph node B and T cell ratios were assessed by ELISA, immunofluorescence, immunoblotting and flow cytometry, respectively., Results: FCRLB shRNA-treated mice showed no muscle weakness or weight loss at termination. Also, they exhibited higher grip strength and muscle AChR levels, lower anti-MuSK IgG and NMJ IgG/C3 levels than control mice. Flow cytometry analysis showed that ratios of major effector lymph node B and T cell populations were not altered by FCRLB silencing. However, regulatory T and CD19 + CD5+ B cell ratios were decreased in FCRLB shRNA-group., Conclusion: Our results provide evidence regarding involvement and therapeutic value of FCRLB in MuSK-MG. Silencing of FCRLB appears to substantially inhibit antibody production without interfering with survival of major lymphocyte populations., Competing Interests: Declaration of Competing Interest The authors declare no disclosures of any financial interest., (Copyright © 2023. Published by Elsevier B.V.)
- Published
- 2023
- Full Text
- View/download PDF
5. Discovery of functionally distinct anti-C7 monoclonal antibodies and stratification of anti-nicotinic AChR positive Myasthenia Gravis patients
- Author
-
Eleonora, Lekova, Wioleta M, Zelek, David, Gower, Claus, Spitzfaden, Isabelle H, Osuch, Elen, John-Morris, Lasse, Stach, Darren, Gormley, Andrew, Sanderson, Angela, Bridges, Elizabeth R, Wear, Sebastien, Petit-Frere, Michael N, Burden, Richard, Priest, Trevor, Wattam, Semra J, Kitchen, Maria, Feeney, Susannah, Davis, B Paul, Morgan, and Eva-Maria, Nichols
- Subjects
Nicotine ,Immunology ,Antibodies, Monoclonal ,Complement System Proteins ,Receptors, Nicotinic ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Epitopes ,Macaca fascicularis ,Antineoplastic Agents, Immunological ,Animals ,Humans ,Immunology and Allergy ,Receptors, Cholinergic ,Autoantibodies - Abstract
Myasthenia Gravis (MG) is mediated by autoantibodies against acetylcholine receptors that cause loss of the receptors in the neuromuscular junction. Eculizumab, a C5-inhibitor, is the only approved treatment for MG that mechanistically addresses complement-mediated loss of nicotinic acetylcholine receptors. It is an expensive drug and was approved despite missing the primary efficacy endpoint in the Phase 3 REGAIN study. There are two observations to highlight. Firstly, further C5 inhibitors are in clinical development, but other terminal pathway proteins, such as C7, have been relatively understudied as therapeutic targets, despite the potential for lower and less frequent dosing. Secondly, given the known heterogenous mechanisms of action of autoantibodies in MG, effective patient stratification in the REGAIN trial may have provided more favorable efficacy readouts. We investigated C7 as a target and assessed thein vitrofunction, binding epitopes and mechanism of action of three mAbs against C7. We found the mAbs were human, cynomolgus monkey and/or rat cross-reactive and each had a distinct, novel mechanism of C7 inhibition. TPP1820 was effective in preventing experimental MG in rats in both prophylactic and therapeutic dosing regimens. To enable identification of MG patients that are likely to respond to C7 inhibition, we developed a patient stratification assay and showed in a small cohort of MG patients (n=19) that 63% had significant complement activation and C7-dependent loss of AChRs in thisin vitroset up. This study provides validation of C7 as a target for treatment of MG and provides a means of identifying patients likely to respond to anti-C7 therapy based on complement-activating properties of patient autoantibodies.
- Published
- 2022
- Full Text
- View/download PDF
6. High mobility group box 1 is involved in the pathogenesis of passive transfer myasthenia gravis model
- Author
-
Tetsuya Kanai, Yuta Kojima, Satoshi Kuwabara, Bibinu Maimaitiming, Manato Yasuda, Akiyuki Uzawa, and Yukiko Ozawa
- Subjects
0301 basic medicine ,medicine.medical_specialty ,chemical and pharmacologic phenomena ,Inflammation ,HMGB1 ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,HMGB1 Protein ,Muscle, Skeletal ,Acetylcholine receptor ,Autoimmune disease ,biology ,business.industry ,General Neuroscience ,Autoantibody ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,030104 developmental biology ,Endocrinology ,biology.protein ,Biomarker (medicine) ,Female ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Myasthenia gravis (MG) is an autoimmune disease with autoantibodies against the mainly nicotinic acetylcholine receptor (AChR). High mobility group box1 (HMGB1) acts as a danger signal and drives the pathogenesis of autoimmune-mediated diseases. However, the role of HMGB1 in the pathogenesis of MG is not fully understood. Therefore, in this study, we analyzed serum levels of HMGB1 and immunohistochemical HMGB1 staining of muscle tissues in the passive transfer MG model to investigate the role of HMGB1 in MG. As a result, serum HMGB1 levels tended to be higher and the quantitative score of muscle pathology showed greater HMGB1 deposition (P = 0.02) along with sparser AChR staining and more severe inflammation in the passive transfer MG rats (n = 6) than those in control rats (n = 6). These findings indicate that HMGB1 is an important mediator and biomarker for inflammation in the pathogenesis of MG and can be a therapeutic target in MG.
- Published
- 2021
- Full Text
- View/download PDF
7. CD59 Expression in Skeletal Muscles and Its Role in Myasthenia Gravis
- Author
-
Kazuo Iwasa, Yutaka Furukawa, Hiroaki Yoshikawa, Masahito Yamada, and Kenjiro Ono
- Subjects
Neurology ,Activities of Daily Living ,Animals ,Humans ,CD59 Antigens ,Neurology (clinical) ,Complement System Proteins ,Thymus Neoplasms ,RNA, Messenger ,Muscle, Skeletal ,Myasthenia Gravis, Autoimmune, Experimental - Abstract
Background and ObjectivesComplement regulatory proteins at the neuromuscular junction (NMJ) could offer protection against complement-mediated damage in myasthenia gravis (MG). However, there is limited information on their expression at the human NMJ. Thus, this study aimed at investigating the expression of the cluster of differentiation 59 (CD59) at the NMJ of human muscle specimens and demonstrating the overexpression ofCD59mRNA and protein in the muscles of patients with MG.MethodsIn this observational study, muscle specimens from 16 patients with MG (9 and 7 patients with and without thymoma, respectively) and 6 nonmyopathy control patients were examined. Immunohistochemical stains, Western blot analysis, and quantitative real-time reverse transcription PCR were used to evaluate the CD59 expression.ResultsA strong localized expression of CD59 was observed at the NMJ in both patients with and without MG. Moreover, the CD59/glyceraldehyde-3-phosphate dehydrogenase protein ratio in patients with MG was significantly higher than that in the nonmyopathy controls (MG; n = 16, median 0.16, interquartile range (IQR) 0.08–0.26 and nonmyopathy controls; n = 6, median 0.03, IQR 0.02–0.11,p= 0.01). The proportion ofCD59mRNA expression relative toAChRmRNA expression (ΔCtCD59/AChR) was associated with the quantitative MG score, MG activities of daily living score, and MG of Foundation of America Clinical Classification (r= 0.663,p= 0.01;r= 0.638,p= 0.014; andr= 0.715,p= 0.003, respectively).DiscussionCD59, which acts as a complement regulator, may protect the NMJ from complement attack. Our findings could provide a basis for further research that investigates the underlying pathogenesis in MG and the immunomodulating interactions of the muscle cells.
- Published
- 2022
8. Effects of Teriflunomide on B Cell Subsets in MuSK-Induced Experimental Autoimmune Myasthenia Gravis and Multiple Sclerosis
- Author
-
Konstantinos Lazaridis, Erdem Tüzün, Vuslat Yilmaz, Sabastian Hajtovic, Murat Kürtüncü, Canan Ulusoy, John Tzartos, and Recai Turkoglu
- Subjects
Adult ,Male ,0301 basic medicine ,Multiple Sclerosis ,Toluidines ,Immunology ,B-Lymphocyte Subsets ,Hydroxybutyrates ,medicine.disease_cause ,Autoimmunity ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Nitriles ,Teriflunomide ,Muscle-Specific Kinase ,medicine ,Animals ,Humans ,Receptors, Cholinergic ,Immunomodulatory Agent ,B cell ,business.industry ,Multiple sclerosis ,Receptor Protein-Tyrosine Kinases ,General Medicine ,Middle Aged ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Crotonates ,030220 oncology & carcinogenesis ,Female ,business - 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.
- Published
- 2020
- Full Text
- View/download PDF
9. miR-1933-3p is upregulated in skeletal muscles of MuSK+ EAMG mice and affects Impa1 and Mrpl27
- Author
-
Tanel Punga, Anna Rostedt Punga, Evgenii Bogatikov, and Ida Lindblad
- Subjects
0301 basic medicine ,Muscle tissue ,medicine.medical_specialty ,Cell Culture Techniques ,Mitochondrial Ribosomes ,Myoblasts ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Atrophy ,Downregulation and upregulation ,Internal medicine ,medicine ,Extracellular ,Animals ,Muscle, Skeletal ,Chemistry ,General Neuroscience ,Receptor Protein-Tyrosine Kinases ,General Medicine ,medicine.disease ,Phosphoric Monoester Hydrolases ,Muscle atrophy ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Mice, Inbred C57BL ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Female ,medicine.symptom ,C2C12 ,030217 neurology & neurosurgery ,Intracellular - Abstract
MuSK antibody seropositive (MuSK+) Myasthenia Gravis (MG) typically affects skeletal muscles of the bulbar area, including the omohyoid muscle, causing focal fatigue, weakness and atrophy. The profile of circulating extracellular microRNA (miRNA) is changed in MuSK + MG, but the intracellular miRNA profile in skeletal muscles of MuSK + MG and MuSK + experimental autoimmune MG (EAMG) remains unknown. This study elucidated the intracellular miRNA profile in the omohyoid muscle of mice with MuSK + EAMG. The levels of eleven mouse miRNAs were elevated and two mouse miRNAs were reduced in muscles of MuSK + EAMG mice. Transient expression of miR-1933-3p and miR-1930-5p in mouse muscle (C2C12) cells revealed several downregulated genes, out of which five had predicted binding sites for miR-1933-3p. The mRNA expression of mitochondrial ribosomal protein L27 (Mrpl27) and Inositol monophosphatase I (Impa1) was reduced in miR-1933-3p transfected C2C12 cells compared to control cells (p = 0.032 versus p = 0.020). Further, transient expression of miR-1933-3p reduced Impa1 protein accumulation in C2C12 cells. These findings provide novel insights of dysregulated miRNAs and their intracellular pathways in muscle tissue afflicted with MuSK + EAMG, providing a possible link to mitochondrial dysfunction and muscle atrophy observed in MuSK + MG.
- Published
- 2020
- Full Text
- View/download PDF
10. The effect of bone marrow stem cells on the treatment of experimental autoimmune myasthenia gravis rat
- Author
-
Yu FU, Yin⁃yan TENG, Chao⁃wei XU, and Xu ZHANG
- Subjects
Myasthenia gravis, autoimmune, experimental ,Mesenchymal stem cell transplantation ,Blotting, western ,Bone marrow transplantation ,Flow cytometry ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Objective To investigate the effect of bone marrow stem cells (BMSCs) on the treatment of experimental autoimmune myasthenia gravis (EAMG) and the possible pathogenesis. Methods BMSCs were obtained from Lewis rat bone marrow selected by cell attachment, cultured and proliferated in vitro. The expression of surface molecules of CD34 and CD44 was analyzed by flow cytometry. The Lewis rats were divided randomly into group A and group B. The rats in group A were transplanted with the third passage BMSCs, and the rats in group B were injected with physiological saline. Seven days after injection, the rat model of EAMG was established by injecting mAb35 to rats in both groups. Clinical signs and symptoms were assessed with Lennon score. Western blotting method was used to determine the expression of acetylcholine receptor (AChR) protein. The CD4+CD25+ Treg cell ratio was analyzed by flow cytometry. Plasma TGF ⁃ β, IFN ⁃ γ and IL ⁃ 4 expressions were detected by ELISA. Results The clinical scores of rats treated with BMSCs were significantly lower than those untreated (t = 8.062, P = 0.000). In comparison with group B, the expression levels of AChR protein (t = 8.092, P = 0.000) and TGF⁃β (t = 9.859, P = 0.000) in group A were all higher, while IFN⁃γ (t = 9.040, P = 0.000) and IL⁃4 (t = 3.320, P = 0.004) were all lower in group A. Conclusion Transplantation with BMSCs can relieve the clinical syndrome of EAMG and reduce the lost of AChR by promoting the level of TGF⁃β, up regulating the expression of CD4+CD25+ Treg cell ratio, and suppressing IFN⁃γ and IL⁃4. DOI:10.3969/j.issn.1672⁃6731.2012.02.014
- Published
- 2012
11. A Recombinant Acetylcholine Receptor α1 Subunit Extracellular Domain Is a Promising New Drug Candidate for Treatment Of Myasthenia Gravis
- Author
-
Konstantinos Lazaridis, Maria Fernandez-Santoscoy, Vasiliki Baltatzidou, Jan-Olof Andersson, Richard Christison, John Grünberg, Socrates Tzartos, Björn Löwenadler, and Charlotte Fribert
- Subjects
T-Lymphocytes ,Immunology ,Immunology and Allergy ,Animals ,Epitopes, B-Lymphocyte ,Receptors, Cholinergic ,Receptors, Nicotinic ,Myasthenia Gravis, Autoimmune, Experimental ,Rats - Abstract
Background and AimsMyasthenia gravis (MG) is a T-cell dependent antibody-mediated autoimmune disease in which the nicotinic acetylcholine receptor (AChR) is the major autoantigen, comprising several T and B cell auto-epitopes. We hypothesized that an efficacious drug candidate for antigen-specific therapy in MG should comprise a broad range of these auto-epitopes and be administered in a noninflammatory and tolerogenic context.MethodsWe used a soluble mutated form of the extracellular domain of the α1 chain of the AChR (α1-ECDm), which represents the major portion of auto-epitopes involved in MG, and investigated, in a well-characterized rat model of experimental autoimmune myasthenia gravis (EAMG) whether its intravenous administration could safely and efficiently treat the autoimmune disease.ResultsWe demonstrated that intravenous administration of α1-ECDm abrogates established EAMG, in a dose and time dependent manner, as assessed by clinical symptoms, body weight, and compound muscle action potential (CMAP) decrement. Importantly, the effect was more pronounced compared to drugs representing current standard of care for MG. The protein had a short plasma half-life, most of what could be recovered was sequestered in the liver, kidneys and spleen. Further, we did not observe any signs of toxicity or intolerability in animals treated with α1-ECDm.ConclusionWe conclude that intravenous treatment with α1-ECDm is safe and effective in suppressing EAMG. α1-ECDm is in preclinical development as a promising new drug candidate for MG.
- Published
- 2021
12. Diabetes mellitus exacerbates experimental autoimmune myasthenia gravis via modulating both adaptive and innate immunity
- Author
-
Tong Du, Heng Li, Ru-Tao Liu, Peng Zhang, Cong-Cong Wang, Meng-Ru Ge, Chun-Lin Yang, Ying-Chun Dou, Rui-Sheng Duan, and Yu-Dong Liu
- Subjects
medicine.medical_specialty ,Neurology ,Immunology ,Tfh ,Disease ,Adaptive Immunity ,Diabetes Mellitus, Experimental ,Cellular and Molecular Neuroscience ,Diabetes mellitus ,medicine ,Animals ,RC346-429 ,B cell ,Antibody ,Innate immunity ,B-Lymphocytes ,Innate immune system ,biology ,business.industry ,General Neuroscience ,Research ,Correction ,medicine.disease ,Myasthenia gravis ,Coculture Techniques ,Immunity, Innate ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Killer Cells, Natural ,medicine.anatomical_structure ,Rats, Inbred Lew ,Concomitant ,Myasthenia gravis (MG) ,biology.protein ,Th17 Cells ,Female ,Neurology. Diseases of the nervous system ,Inflammation Mediators ,business ,Diabetes mellitus (DM) - 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.
- Published
- 2021
13. A Targeted Complement Inhibitor CRIg/FH Protects Against Experimental Autoimmune Myasthenia Gravis in Rats
- Author
-
Jie, Song, Rui, Zhao, Chong, Yan, Sushan, Luo, Jianying, Xi, Peipei, Ding, Ling, Li, Weiguo, Hu, and Chongbo, Zhao
- Subjects
Recombinant Fusion Proteins ,Autoimmunity ,Cell Differentiation ,Lymphocyte Activation ,Severity of Illness Index ,T-Lymphocytes, Regulatory ,Cell Line ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Immunomodulation ,Disease Models, Animal ,Complement Inactivating Agents ,Immunoglobulin G ,Animals ,Cytokines ,Humans ,Disease Susceptibility ,Inflammation Mediators ,Complement Activation ,Autoantibodies - Abstract
Antibody-induced complement activation may cause injury of the neuromuscular junction (NMJ) and is thus considered as a primary pathogenic factor in human myasthenia gravis (MG) and animal models of experimental autoimmune myasthenia gravis (EAMG). In this study, we tested whether CRIg/FH, a targeted complement inhibitor, could attenuate NMJ injury in rat MG models. We first demonstrated that CRIg/FH could inhibit complement-dependent cytotoxicity on human rhabdomyosarcoma TE671 cells induced by MG patient-derived IgG
- Published
- 2021
14. Takotsubo Cardiomyopathy Complicated with Left Ventricular Thrombus in Myasthenic Crisis: A Case Report
- Author
-
Natnicha Pongbangli, Sasivimon Jae-aue, Arintaya Phrommintikul, and Wanwarang Wongcharoen
- Subjects
medicine.medical_specialty ,Cardiomyopathy ,030204 cardiovascular system & hematology ,QT interval ,Embolism and Thrombosis ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,law ,Takotsubo Cardiomyopathy ,Internal medicine ,Myasthenia Gravis ,medicine ,Humans ,cardiovascular diseases ,Aged ,business.industry ,Coronary Thrombosis ,General Medicine ,Articles ,Left ventricular thrombus ,medicine.disease ,Intensive care unit ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,030220 oncology & carcinogenesis ,Concomitant ,Heart failure ,Cardiology ,cardiovascular system ,Female ,medicine.symptom ,business ,Hyperkinesia - Abstract
Patient: Female, 67 Final Diagnosis: Takotsubo cardiomyopathy complicating with left ventricular thrombus Symptoms: Acute respiratory failure Medication: — Clinical Procedure: Echocardiogram Specialty: Cardiology Objective: Unusual clinical course Background: Myasthenia gravis can precipitate severe stress particularly during a myasthenic crisis episode. Takotsubo cardiomyopathy has been demonstrated in several conditions associated with emotional or physical stress. As a result, Takotsubo cardiomyopathy is not uncommon in patients with MG. The severe complications of Takotsubo cardiomyopathy include heart failure and left ventricular thrombus associated with thromboembolic risk. The concomitant myasthenic crisis and Takotsubo cardiomyopathy with apical left ventricular thrombus has never been reported. Case Report: A 67-year- old Thai female diagnosed with myasthenia gravis was admitted to the intensive care unit due to the myasthenic crisis. The 12-lead electrocardiogram showed marked QT interval prolongation and diffuse large T-wave inversion. Echocardiogram demonstrated basal hyperkinesia and apical akinesia with apical ballooning. Hyperechoic mass was noted in akinetic left ventricular apex. Takotsubo cardiomyopathy with apical left ventricular thrombus was diagnosed. Both conditions were successfully treated in this patient without any complications. Conclusions: The electrocardiogram surveillance in patients with myasthenic crisis is essential to detect the occurrence of Takotsubo cardiomyopathy and its complications. Early diagnosis and treatments may decrease mortality and morbidity related with this condition.
- Published
- 2019
15. Enhancement of T Follicular Helper Cell-Mediated Humoral Immunity Reponses During Development of Experimental Autoimmune Myasthenia Gravis
- Author
-
Siying Qu, Ying-Zhe Cui, Bo Sun, Tongshuai Zhang, Guangyou Wang, Jiarui Zhao, Qingfei Kong, Lili Mu, Hulun Li, and Lulu Chang
- Subjects
0301 basic medicine ,Physiology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Animals ,Medicine ,Receptors, Cholinergic ,Lymph node ,Autoimmune disease ,B-Lymphocytes ,biology ,business.industry ,General Neuroscience ,Germinal center ,Receptor Cross-Talk ,T-Lymphocytes, Helper-Inducer ,General Medicine ,medicine.disease ,Myasthenia gravis ,Immunity, Humoral ,Myasthenia Gravis, Autoimmune, Experimental ,Disease Models, Animal ,Protein Subunits ,030104 developmental biology ,medicine.anatomical_structure ,Immunization ,Rats, Inbred Lew ,Humoral immunity ,Immunology ,Proto-Oncogene Proteins c-bcl-6 ,biology.protein ,Female ,Original Article ,Lymph Nodes ,Antibody ,business ,030217 neurology & neurosurgery - Abstract
Myasthenia gravis (MG) is a prototypical antibody-mediated neurological autoimmune disease with the involvement of humoral immune responses in its pathogenesis. T follicular helper (Tfh) cells have been implicated in many autoimmune diseases. However, whether and how Tfh cells are involved in MG remain unclear. Here, we established and studied a widely-used and approved animal model of human MG, the rat model with acetylcholine receptor alpha (AChRα) subunit (R-AChR(97–116))-induced experimental autoimmune myasthenia gravis (EAMG). This model presented mild body-weight loss 10 days after the first immunization (representing the early stage of disease) and more obvious clinical manifestations and body-weight loss 7 days after the second immunization (representing the late stage of disease). AChR-specific pre-Tfh cells and mature Tfh cells were detected in these two stages, respectively. In co-cultures of Tfh cells and B cells, the number of IgG2b-secreting B cells and the level of anti-AChR antibodies in the supernatant were higher in the cultures containing EAMG-derived Tfh cells. In immunohistochemistry and immunofluorescence assays, a substantial number of CD4(+)/Bcl-6(+) T cells and a greater number of larger germinal centers were observed in lymph node tissues resected from EAMG rats. Based on these results, we hypothesize that an AChR-specific Tfh cell-mediated humoral immune response contributes to the development of EAMG.
- Published
- 2019
- Full Text
- View/download PDF
16. Berberine attenuates experimental autoimmune myasthenia gravis via rebalancing the T cell subsets
- Author
-
Jie Song, Jie Yang, Sisi Jing, Chong Yan, Xiao Huan, Sheng Chen, Huahua Zhong, Jun Lu, Jianying Xi, Lijun Luo, Xi Chen, Ziyuan Wang, Chongbo Zhao, Ming Chu, and Sushan Luo
- Subjects
Neurology ,Berberine ,Rats, Inbred Lew ,T-Lymphocyte Subsets ,Immunology ,Immunology and Allergy ,Animals ,Female ,Neurology (clinical) ,Myasthenia Gravis, Autoimmune, Experimental ,Rats - Abstract
Myasthenia Gravis (MG) is a T cell-driven, autoantibody-mediated disease. Here we show that oral Berberine (BBR) ameliorated clinical symptoms of experimental autoimmune myasthenia gravis(EAMG) rat model via decreasing the frequencies of Th1, Th17, Th1/17 cell subsets. JAK-STAT pathway was highlighted by transcriptomic analysis with EAMG mononuclear cells (MNCs). Surface plasmon resonance identified ligand binding interaction between BBR and JAK2, and electrostatic interaction was proposed by molecular dynamic simulation. Reduced phosphorylated JAK1/2/3 and STAT1/3 in MNCs from BBR-fed EAMG rats were demonstrated. These results suggest that BBR might improve EAMG by rebalancing T cell subsets through targeting JAK-STAT pathway.
- Published
- 2021
17. B cell-specific mAb-siRNA conjugates improve experimental myasthenia.
- Author
-
Ibtehaj N, Bahauddin A, Ivannikov M, Rytting E, Jamaluddin M, Liang Y, Sun J, Haller SL, Wu X, and Huda R
- Subjects
- Mice, Animals, RNA, Small Interfering, Receptors, Cholinergic, Antibodies, Monoclonal, Autoantibodies, Myasthenia Gravis, Autoimmune, Experimental
- Abstract
Myasthenia gravis (MG) is a debilitating autoimmune disease characterized by muscle fatigue and weakness caused by autoantibody- and complement-mediated damage to the neuromuscular junction. This study sought to compare the efficacy of unique sets of monoclonal antibody-siRNA conjugates, individually (mono) or in combination (duo), against the crucial receptors predominantly or solely expressed on two subsets of B cells-plasma B cells and their precursor (transitional mature B) cells in a mouse model of MG. At the optimized doses, the conjugates, likely due to the combined activities of mAb and siRNA, substantially decreased the expression levels of CD268 (B cell-activating factor receptor) in mature B cells and CD269 (B-cell maturation antigen) in plasma cells concomitantly with reducing the levels of acetylcholine receptor (AChR)-specific autoantibodies. PEGylation, but not pretreatment with an antibody against type 1 interferon receptor, further improved duoconjugate-induced reduction in the autoantibody levels. Our results show that the duoconjugate treatment significantly improved the clinical symptoms of MG, consistent with the preservation of bungarotoxin-bound functional AChRs. In the future, developing similar target-specific combination molecules can potentially turn into a new and effective therapeutic approach for MG., Competing Interests: Declaration of competing interest The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The invention disclosed here has been filed for a patent., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Blocking interleukin-23 ameliorates neuromuscular and thymic defects in myasthenia gravis.
- Author
-
Villegas JA, Van Wassenhove J, Merrheim J, Matta K, Hamadache S, Flaugère C, Pothin P, Truffault F, Hascoët S, Santelmo N, Alifano M, Berrih-Aknin S, le Panse R, and Dragin N
- Subjects
- Mice, Humans, Animals, Interleukin-23 Subunit p19, Receptors, Cholinergic, Neuromuscular Junction pathology, Autoantibodies, Interleukin-23, Myasthenia Gravis, Autoimmune, Experimental
- Abstract
Acetylcholine receptor (AChR) myasthenia gravis (MG) is a chronic autoimmune disease characterized by muscle weakness. The AChR
+ autoantibodies are produced by B-cells located in thymic ectopic germinal centers (eGC). No therapeutic approach is curative. The inflammatory IL-23/Th17 pathway is activated in the thymus as well as in the blood and the muscle, contributing to the MG pathogenic events. We aimed to study a potential new therapeutic approach that targets IL-23p19 (IL-23) in the two complementary preclinical MG models: the classical experimental MG mouse model (EAMG) based on active immunization and the humanized mouse model featuring human MG thymuses engrafted in NSG mice (NSG-MG). In both preclinical models, the anti-IL-23 treatment ameliorated MG clinical symptoms. In the EAMG, the treatment reduced IL-17 related inflammation, anti-AChR IgG2b antibody production, activated transduction pathway involved in muscle regeneration and ameliorated the signal transduction at the neuromuscular junction. In the NSG-MG model, the treatment reduced pathogenic Th17 cell population and expression of genes involved in eGC stabilization and B-cell development in human MG thymus biopsies. Altogether, these data suggest that a therapy targeting IL-23p19 may promote significant clinical ameliorations in AChR+ MG disease due to concomitant beneficial effects on the thymus and skeletal muscle defects., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
19. [Rapamycin alleviates the symptoms of experimental autoimmune myasthenia gravis rats by down-regulating Th17 cell/regulatory T cell ratio]
- Author
-
Xinlei, Gao, Yujun, Wen, Zhao, Wang, Guowei, Wang, Jiayu, Guo, Liming, Yu, and Zhenhai, Wang
- Subjects
Sirolimus ,Mice ,Rats, Inbred Lew ,Animals ,Th17 Cells ,T-Lymphocytes, Regulatory ,Myasthenia Gravis, Autoimmune, Experimental ,Rats - Abstract
Objective To study the therapeutic effect of rapamycin (RAPA) on experimental autoimmune myasthenia gravis (EAMG) rats and to explore the related immune mechanisms. Methods The mouse-derived acetylcholine receptor alpha subunit 97-116 peptide (R97-116) was used to immunize Lewis rats to establish an EAMG rat model. The rats were randomly divided into three groups: complete Freund's adjuvant control group (CFA group), EAMG model control group, and RAPA treatment group [1 mg/(kg.d)]. The Lennon muscle strength scoring scale was used to evaluate rats' clinical symptoms in each group once every two days, and their body mass was recorded. ELISA was performed to detect the level of anti-R97-116 antibodies in the peripheral blood of rats. Flow cytometry was used to detect the numbers of Th17 cells and regulatory T cells (Tregs) in rat splenocytes. Splenocytes were stimulated with 5 μg/mL concanavalin A (ConA), 10 μg/mL R97-116 and RPMI1640 medium, and the proliferation activity of rat splenocytes was tested by CCK-8 assay. Results RAPA treatment significantly improved the body mass and clinical scores in EAMG rats. Compared with the CFA group, the number of Th17 cells in the spleen of the EAMG group increased, and the number of Tregs decreased. Compared with the EAMG group, the number of Th17 cells in the spleen of RAPA-treated rats significantly dropped, the number of Tregs went up, and the level of anti-R97-116 antibodies in the serum went down. RAPA treatment inhibited the proliferation of lymphocytes induced by RPMI1640 medium, R97-116, and ConA stimulation. Conclusion RAPA may alleviate the clinical symptoms of EAMG rats by down-regulating the ratio of Th17 cells/Tregs.
- Published
- 2021
20. Leflunomide ameliorates experimental autoimmune myasthenia gravis by regulating humoral and cellular immune responses
- Author
-
Huan Huang, Weibin Liu, Yaru Lu, Xiaoxi Liu, Hao Ran, Changyi Ou, Lu Yu, Zhongqiang Lin, Li Qiu, and Wenhao Yang
- Subjects
0301 basic medicine ,STAT3 Transcription Factor ,Immunology ,Adaptive Immunity ,03 medical and health sciences ,Psoriatic arthritis ,0302 clinical medicine ,Immune system ,Immunology and Allergy ,Medicine ,Animals ,Humans ,Receptor ,Th1-Th2 Balance ,Cells, Cultured ,Leflunomide ,Pharmacology ,biology ,business.industry ,Multiple sclerosis ,Interleukins ,T-Lymphocytes, Helper-Inducer ,medicine.disease ,Germinal Center ,Myasthenia gravis ,Immunity, Humoral ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,030104 developmental biology ,Rats, Inbred Lew ,030220 oncology & carcinogenesis ,Rheumatoid arthritis ,biology.protein ,Female ,Antibody ,business ,Immunosuppressive Agents ,medicine.drug ,Signal Transduction - Abstract
Leflunomide, an immunosuppressive disease-modifying anti-rheumatic drug (DMARD), is widely used in the treatment of rheumatoid arthritis (RA), psoriatic arthritis (PA) as well as multiple sclerosis. However, its role in myasthenia gravis (MG) has not yet been clearly explored. Here, we investigated the effect of leflunomide on experimental autoimmune myasthenia gravis (EAMG) in vivo and in vitro. The results demonstrated that leflunomide alleviated the severity of EAMG associated with reduced serum total anti-acetylcholine receptor (AChR) IgG levels. During the development of EAMG, the increase of follicular helper T cells (Tfh) 1, Tfh 17 cells and decrease of follicular regulatory T cells (Tfr) were reversely altered after leflunomide administration. Our work further found that leflunomide might inhibit Tfh cells through the IL-21/STAT3 pathway to reduce the secretion of antibodies by B cells. In addition, leflunomide rebuilt the balance of Th1/Th2/Th17/Treg subsets. These results suggested that leflunomide ameliorated EAMG severity by regulating humoral immune responses and Th cell profiles thereby providing a novel effective treatment strategy for MG.
- Published
- 2021
21. Prophylactic administration of fingolimod (FTY720) ameliorated experimental autoimmune myasthenia gravis by reducing the number of dendritic cells, follicular T helper cells and antibody-secreting cells
- Author
-
Rui-Sheng Duan, Yu-Dong Liu, Ying Liu, Bing Yang, Meng-Ru Ge, Ying Lian, Heng Li, Xiao-Li Li, Peng Zhang, Tong Du, and Chun-Lin Yang
- Subjects
0301 basic medicine ,T cell ,Immunology ,medicine.disease_cause ,Autoantigens ,Autoimmunity ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Myasthenia Gravis ,Immunology and Allergy ,Medicine ,Animals ,Humans ,Receptors, Cholinergic ,Lymphocyte homing receptor ,Antibody-Producing Cells ,Pharmacology ,biology ,business.industry ,Fingolimod Hydrochloride ,T helper cell ,Dendritic Cells ,T-Lymphocytes, Helper-Inducer ,medicine.disease ,Myasthenia gravis ,Immunity, Humoral ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Thymocyte ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Rats, Inbred Lew ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Antibody ,business ,Peptides ,Immunosuppressive Agents - Abstract
Fingolimod (FTY720), a sphingosine 1-phosphate (S1P) receptor antagonist, possesses potent immunomodulatory activity via lymphocyte homing. The effects of FTY720 have been widely studied in various T-cell-mediated autoimmune diseases, while the immunomodulatory effects on experimental autoimmune myasthenia gravis (EAMG), a typical disease model for antibody-mediated autoimmunity, remain elusive. In the present study, FTY720 was administered to EAMG rats as prophylaxis. The clinical scores were recorded every other day, and serum antibodies at different time points were measured by enzyme-linked immunosorbent assay (ELISA). The immune cell subsets in the spleen, bone marrow, circulation, and thymus were determined by flow cytometry. The prophylactic administration alleviated EAMG symptoms by reducing the level of serum antibodies IgG and its isotype IgG2b on days 30 and 46 post immunization, as well as IgG and Ig kappa antibody-secreting cells in the spleen and bone marrow. The mitigated humoral immune response can be attributed to the decreased dendritic cells, follicular T help cells (Tfh) and Tfh subsets (Tfh1, Tfh2, and Tfh17), and T helper cell subsets (Th1, Th2, and Th17) in the spleen. The promotion of lymphocyte homing and inhibition of thymocyte egress contribute to the effects of FTY720 on these effector T cell subsets. Overall, the prophylactic administration of FTY720 ameliorated EAMG partially by regulating humoral immune response,suggesting that FTY720 could be part of a pharmacological strategy for managing myasthenia gravis.
- Published
- 2020
22. AEB-071 Ameliorates Muscle Weakness by Altering Helper T Lymphocytes in an Experimental Autoimmune Myasthenia Gravis Rat Model
- Author
-
Qian Ma, Sheng-Jie Xu, Wei Huang, Yu-Xiu Guan, Feng Jing, Chang-Jin Wu, and Bing Chen
- Subjects
medicine.medical_specialty ,Helper T lymphocyte ,Lymphocyte ,Rat model ,Neurological disorder ,030204 cardiovascular system & hematology ,Lymphocyte Activation ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Pyrroles ,Protein kinase C ,Muscle Weakness ,medicine.diagnostic_test ,business.industry ,Muscle weakness ,T-Lymphocytes, Helper-Inducer ,General Medicine ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,medicine.anatomical_structure ,Endocrinology ,Rats, Inbred Lew ,030220 oncology & carcinogenesis ,Quinazolines ,Female ,medicine.symptom ,business - 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
- Published
- 2020
- Full Text
- View/download PDF
23. Qiangji Jianli Decoction promotes mitochondrial biogenesis in skeletal muscle of myasthenia gravis rats via AMPK/PGC-1α signaling pathway
- Author
-
Jingwei Song, Qing Li, Lanqi Li, Wei Jiao, Fangyu Hu, Lingling Ke, Jian Liang, Yafang Song, Jinqiu Li, and Zhiwei Chen
- Subjects
0301 basic medicine ,AMPK ,ATPase ,PGC-1α ,RM1-950 ,Pharmacology ,AMP-Activated Protein Kinases ,03 medical and health sciences ,Gastrocnemius muscle ,0302 clinical medicine ,Mitochondrial biogenesis ,medicine ,Animals ,Receptors, Cholinergic ,Muscle, Skeletal ,Qiangji Jianli Decoction ,Myasthenia gravis ,Organelle Biogenesis ,biology ,Chemistry ,Skeletal muscle ,General Medicine ,TFAM ,medicine.disease ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Peptide Fragments ,Mitochondria, Muscle ,Myasthenia Gravis, Autoimmune, Experimental ,030104 developmental biology ,medicine.anatomical_structure ,mitochondrial fusion ,Gene Expression Regulation ,Rats, Inbred Lew ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Therapeutics. Pharmacology ,Drugs, Chinese Herbal ,Signal Transduction - Abstract
The Qiangji Jianli Decoction (QJJLD) is an effective Chinese medicine formula for treating Myasthenia gravis (MG) in the clinic. QJJLD has been proven to regulate mitochondrial fusion and fission of skeletal muscle in myasthenia gravis. In this study, we investigated whether QJJLD plays a therapeutic role in regulating mitochondrial biogenesis in MG and explored the underlying mechanism. Rats were experimentally induced to establish autoimmune myasthenia gravis (EAMG) by subcutaneous immunization with R97-116 peptides. The treatment groups were administered three different dosages of QJJLD respectively. After the intervention of QJJLD, the pathological changes of gastrocnemius muscle in MG rats were significantly improved; SOD, GSH-Px, Na+-K+ ATPase and Ca2+-Mg2+ ATPase activities were increased; and MDA content was decreased in the gastrocnemius muscle. Moreover, AMPK, p38MAPK, PGC-1α, NRF-1, Tfam and COX IV mRNA and protein expression levels were also reversed by QJJLD. These results implied that QJJLD may provide a potential therapeutic strategy through promoting mitochondrial biogenesis to alleviate MG via activating the AMPK/PGC-1α signaling pathway.
- Published
- 2020
24. MuSK EAMG: Immunological Characterization and Suppression by Induction of Oral Tolerance
- Author
-
Revital Aricha, Debby Reuveni, Sara Fuchs, and Miriam C. Souroujon
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,medicine.medical_treatment ,T regulatory cells ,Immunology ,MuSK protein ,Administration, Oral ,Receptor tyrosine kinase ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Immune Tolerance ,Immunology and Allergy ,Medicine ,Animals ,Original Research ,myasthenia gravis ,neuromuscular junction (NMJ) ,biology ,business.industry ,oral tolerance ,Antibody titer ,Receptor Protein-Tyrosine Kinases ,Immunosuppression ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,030104 developmental biology ,biology.protein ,Nasal administration ,Female ,Antibody ,business ,lcsh:RC581-607 ,muscle-specific receptor tyrosine kinase (MuSK) ,030215 immunology - Abstract
Myasthenia gravis (MG) with antibodies to the muscle-specific receptor tyrosine kinase (MuSK) is a distinct sub-group of MG, affecting 5-8% of all MG patients. MuSK, a receptor tyrosine kinase, is expressed at the neuromuscular junctions (NMJs) from the earliest stages of synaptogenesis and plays a crucial role in the development and maintenance of the NMJ. MuSK-MG patients are more severely affected and more refractory to treatments currently used for MG. Most patients require long-term immunosuppression, stressing the need for improved treatments. Ideally, preferred treatments should specifically delete the antigen-specific autoimmune response, without affecting the entire immune system. Mucosal tolerance, induced by oral or nasal administration of an auto-antigen through the mucosal system, resulting in an antigen-specific immunological systemic hyporesponsiveness, might be considered as a treatment of choice for MuSK-MG. In the present study we have characterized several immunological parameters of murine MuSK-EAMG and have employed induction of oral tolerance in mouse MuSK-EAMG, by feeding with a recombinant MuSK protein one week before disease induction. Such a treatment has been shown to attenuate MuSK-EAMG. Both induction and progression of disease were ameliorated following oral treatment with the recombinant MuSK fragment, as indicated by lower clinical scores and lower anti-MuSK antibody titers.
- Published
- 2020
- Full Text
- View/download PDF
25. Sorting nexin 17 increases low-density lipoprotein receptor-related protein 4 membrane expression: A novel mechanism of acetylcholine receptor aggregation in myasthenia gravis.
- Author
-
He X, Zhou S, Ji Y, Zhang Y, Lv J, Quan S, Zhang J, Zhao X, Cui W, Li W, Liu P, Zhang L, Shen T, Fang H, Yang J, Zhang Y, Cui X, Zhang Q, and Gao F
- Subjects
- Animals, Mice, Acetylcholine, LDL-Receptor Related Proteins, Lipoproteins, LDL, Receptor Protein-Tyrosine Kinases, Sorting Nexins genetics, Myasthenia Gravis, Autoimmune, Experimental, Receptors, Cholinergic
- Abstract
Myasthenia gravis (MG) is characterized by autoimmune damage to the postsynaptic membrane of the neuromuscular junction (NMJ) with impaired postsynaptic acetylcholine receptor (AChR) aggregation. Low-density lipoprotein receptor-related protein 4 (LRP4) plays an important role in AChR aggregation at endplate membranes via the Agrin-LRP4-muscle-specific receptor tyrosine kinase (MuSK) cascade. Sorting nexin 17 (SNX17) regulates the degradation and recycling of various internalized membrane proteins. However, whether SNX17 regulates LRP4 remains unclear. Therefore, we examined the regulatory effects of SNX17 on LRP4 and its influence on AChR aggregation in MG. We selected C2C12 myotubes and induced LRP4 internalization via stimulation with anti-LRP4 antibody and confirmed intracellular interaction between SNX17 and LRP4. SNX17 knockdown and overexpression confirmed that SNX17 promoted MuSK phosphorylation and AChR aggregation by increasing cell surface LRP4 expression. By establishing experimental autoimmune MG (EAMG) mouse models, we identified that SNX17 upregulation improved fragmentation of the AChR structure at the NMJ and alleviated leg weakness in EAMG mice. Thus, these results reveal that SNX17 may be a novel target for future MG therapy., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 He, Zhou, Ji, Zhang, Lv, Quan, Zhang, Zhao, Cui, Li, Liu, Zhang, Shen, Fang, Yang, Zhang, Cui, Zhang and Gao.)
- Published
- 2022
- Full Text
- View/download PDF
26. Heterogeneity of auto-antibodies against nAChR in myasthenic serum and their pathogenic roles in experimental autoimmune myasthenia gravis
- Author
-
Jyoji Yamate, Tomohiro Makino, Ryuichi Nakamura, Hirokazu Shiraishi, Maki Terakawa, Masakatsu Motomura, Takeshi Hanada, and Kazuhiro Nagahira
- Subjects
0301 basic medicine ,medicine.drug_class ,Immunology ,Receptors, Nicotinic ,Monoclonal antibody ,Autoantigens ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Immunology and Allergy ,Autoantibodies ,biology ,Chemistry ,Autoantibody ,medicine.disease ,Phenotype ,Myasthenia gravis ,In vitro ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Nicotinic acetylcholine receptor ,030104 developmental biology ,Neurology ,Polyclonal antibodies ,biology.protein ,Female ,Neurology (clinical) ,Antibody ,030217 neurology & neurosurgery - Abstract
Many myasthenia gravis (MG) patients have auto-antibodies against the nicotinic acetylcholine receptor (nAChR), and monoclonal antibodies against the main immunogenic region (MIR) of nAChR can induce experimental autoimmune MG (EAMG). We investigated whether Fab fragment of MIR antibody (Fab35) could block the pathogenicity of polyclonal antibodies. Fab35 partially inhibited nAChR downmodulation, blocked EAMG serum-induced binding of polyclonal antibodies and complement deposition in vitro. Moreover, Fab35 did not ameliorate the EAMG serum-induced EAMG phenotype in rats. These results suggested that the EAMG serum possessed several different pathogenic antibodies that might be sufficient to induce the EAMG phenotype.
- Published
- 2018
- Full Text
- View/download PDF
27. Specific inhibition of acetylcholinesterase as an approach to decrease muscarinic side effects during myasthenia gravis treatment
- Author
-
Oksana A. Lenina, Alexandra D. Kharlamova, Marina E. Sitdykova, Ayrat R. Nurtdinov, Victor I. Ilyin, Irina V. Zueva, Evgeny E. Nikolsky, and Konstantin A. Petrov
- Subjects
0301 basic medicine ,Urinary Bladder ,lcsh:Medicine ,Pharmacology ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Muscarinic acetylcholine receptor ,Medicine ,Animals ,Humans ,Uracil ,lcsh:Science ,Butyrylcholinesterase ,Multidisciplinary ,Urinary bladder ,business.industry ,lcsh:R ,Muscle weakness ,Muscle, Smooth ,medicine.disease ,Acetylcholinesterase ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Intestines ,Quaternary Ammonium Compounds ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Pyridostigmine ,Pyridostigmine Bromide ,lcsh:Q ,Cholinesterase Inhibitors ,medicine.symptom ,business ,medicine.drug ,Muscle Contraction - Abstract
Non-selective inhibitors of cholinesterases (ChEs) are clinically used for treatment of myasthenia gravis (MG). While being generally safe, they cause numerous adverse effects including induction of hyperactivity of urinary bladder and intestines affecting quality of patients life. In this study we have compared two ChEs inhibitors, a newly synthesized compound C547 and clinically used pyridostigmine bromide, by their efficiency to reduce muscle weakness symptoms and ability to activate contractions of urinary bladder in a rat model of autoimmune MG. We found that at dose effectively reducing MG symptoms, C547 did not affect activity of rat urinary bladder. In contrast, at equipotent dose, pyridostigmine caused a significant increase in tonus and force of spontaneous contractions of bladder wall. We also found that this profile of ChEs inhibitors translates into the preparation of human urinary bladder. The difference in action observed for C547 and pyridostigmine we attribute to a high level of pharmacological selectivity of C547 in inhibiting acetylcholinesterase as compared to butyrylcholinesterase. These results raise reasonable hope that selective acetylcholinesterase inhibitors should show efficacy in treating MG in human patients with a significant reduction in adverse effects related to hyperactivation of smooth muscles.
- Published
- 2018
- Full Text
- View/download PDF
28. Discovery of functionally distinct anti-C7 monoclonal antibodies and stratification of anti-nicotinic AChR positive Myasthenia Gravis patients.
- Author
-
Lekova E, Zelek WM, Gower D, Spitzfaden C, Osuch IH, John-Morris E, Stach L, Gormley D, Sanderson A, Bridges A, Wear ER, Petit-Frere S, Burden MN, Priest R, Wattam T, Kitchen SJ, Feeney M, Davis S, Morgan BP, and Nichols EM
- Subjects
- Animals, Antibodies, Monoclonal metabolism, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Autoantibodies metabolism, Complement System Proteins metabolism, Epitopes, Humans, Macaca fascicularis, Nicotine, Rats, Receptors, Cholinergic, Antineoplastic Agents, Immunological, Myasthenia Gravis, Autoimmune, Experimental, Receptors, Nicotinic
- Abstract
Myasthenia Gravis (MG) is mediated by autoantibodies against acetylcholine receptors that cause loss of the receptors in the neuromuscular junction. Eculizumab, a C5-inhibitor, is the only approved treatment for MG that mechanistically addresses complement-mediated loss of nicotinic acetylcholine receptors. It is an expensive drug and was approved despite missing the primary efficacy endpoint in the Phase 3 REGAIN study. There are two observations to highlight. Firstly, further C5 inhibitors are in clinical development, but other terminal pathway proteins, such as C7, have been relatively understudied as therapeutic targets, despite the potential for lower and less frequent dosing. Secondly, given the known heterogenous mechanisms of action of autoantibodies in MG, effective patient stratification in the REGAIN trial may have provided more favorable efficacy readouts. We investigated C7 as a target and assessed the in vitro function, binding epitopes and mechanism of action of three mAbs against C7. We found the mAbs were human, cynomolgus monkey and/or rat cross-reactive and each had a distinct, novel mechanism of C7 inhibition. TPP1820 was effective in preventing experimental MG in rats in both prophylactic and therapeutic dosing regimens. To enable identification of MG patients that are likely to respond to C7 inhibition, we developed a patient stratification assay and showed in a small cohort of MG patients (n=19) that 63% had significant complement activation and C7-dependent loss of AChRs in this in vitro set up. This study provides validation of C7 as a target for treatment of MG and provides a means of identifying patients likely to respond to anti-C7 therapy based on complement-activating properties of patient autoantibodies., Competing Interests: EL, DGow, CS, IO, LS, DGorm, AS, AB, EW, MB, TW, RP, SK, MF, SD, E-MN are employees and shareholders of GSK. SP-F is an employee of GSK. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Lekova, Zelek, Gower, Spitzfaden, Osuch, John-Morris, Stach, Gormley, Sanderson, Bridges, Wear, Petit-Frere, Burden, Priest, Wattam, Kitchen, Feeney, Davis, Morgan and Nichols.)
- Published
- 2022
- Full Text
- View/download PDF
29. NFAT1 Regulates Systemic Autoimmunity through the Modulation of a Dendritic Cell Property
- Author
-
Chang-Suk Chae, Eun Sil Park, Sin-Hyeog Im, Yung Joon Yoo, Choong-Gu Lee, Gi-Cheon Kim, Ravi Verma, Hagg-Lim Cho, and Chang-Duk Jun
- Subjects
0301 basic medicine ,Cellular differentiation ,Immunology ,Mice, Transgenic ,Biology ,Lymphocyte Activation ,medicine.disease_cause ,Proinflammatory cytokine ,Autoimmunity ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Immune Tolerance ,medicine ,Animals ,Immunology and Allergy ,NFATC Transcription Factors ,NF-kappa B ,Dendritic Cells ,Dendritic cell ,Th1 Cells ,NFKB1 ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,030104 developmental biology ,Cytokines ,Th17 Cells ,Signal transduction ,Signal Transduction ,030215 immunology - 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-1β, 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.
- Published
- 2017
- Full Text
- View/download PDF
30. Transcriptional repressor Blimp1 regulates follicular regulatory T-cell homeostasis and function
- Author
-
Jue Hu, Dandan Zheng, Guancheng Li, Xiaosu Yang, Guang Yang, Liqun Xu, Huan Yang, Jing Li, Anjiao Peng, Erdem Tüzün, Junmei Zhang, Baifeng Yang, and Wenbin Zhou
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Adoptive cell transfer ,Regulatory T cell ,Immunology ,Lymphocyte Activation ,T-Lymphocytes, Regulatory ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,T-Lymphocyte Subsets ,Internal medicine ,medicine ,Animals ,Homeostasis ,Immunology and Allergy ,Neuroinflammation ,B-Lymphocytes ,biology ,Germinal center ,Original Articles ,Germinal Center ,Myasthenia Gravis, Autoimmune, Experimental ,Cell biology ,030104 developmental biology ,Neuroimmunology ,medicine.anatomical_structure ,Endocrinology ,Gene Expression Regulation ,biology.protein ,Female ,Positive Regulatory Domain I-Binding Factor 1 ,Antibody ,030217 neurology & neurosurgery - Abstract
The B-lymphocyte-induced maturation protein 1 (Blimp1) regulates T-cell homeostasis and function. Loss of Blimp1 could double the proportion of follicular regulatory T (Tfr) cells. However, the effects that Blimp1 may have on the function of Tfr cells remain unknown. Here we document the function for Blimp1 in Tfr cells in vitro and in vivo. Data presented in this study demonstrate that Tfr cells indirectly inhibit the activation and differentiation of B cells by negatively regulating follicular helper T cells, so lowering the secretion of antibody. Lack of Blimp1 makes the immune suppression function of Tfr cells impaired in vitro. In the in vivo study, adoptive transfer of Tfr cells could reduce immune responses in germinal centres and relieve the muscle weakness symptoms of mice with experimental autoimmune myasthenia gravis. Blimp1 deficiency resulted in reduced suppressive ability of Tfr cells. This study identifies that Tfr cells are potent suppressors of immunity and are controlled by Blimp1.
- Published
- 2017
- Full Text
- View/download PDF
31. Decreased bone mineral density in experimental myasthenia gravis in C57BL/6 mice
- Author
-
Minako Oshima, Takahiro Maruta, M. Zouhair Atassi, Philip R. Deitiker, and Akiko Iida-Klein
- Subjects
Fish Proteins ,Male ,0301 basic medicine ,medicine.medical_specialty ,Time Factors ,Bone density ,Freund's Adjuvant ,Immunology ,Osteoporosis ,Torpedo ,Severity of Illness Index ,Mice ,03 medical and health sciences ,Absorptiometry, Photon ,0302 clinical medicine ,Immune system ,Bone Density ,Weight loss ,Internal medicine ,medicine ,Animals ,Immunology and Allergy ,Receptors, Cholinergic ,Femur ,Tibia ,Bone Resorption ,Lumbar Vertebrae ,Muscle Weakness ,business.industry ,Age Factors ,Muscle weakness ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Experimental autoimmune myasthenia gravis (EAMG), an animal model of myasthenia gravis (MG), can be induced in C57BL/6 (B6, H-2 b) mice by 2-3 injections with Torpedo californica AChR (tAChR) in complete Freund's adjuvant. Some EAMG mice exhibit weight loss with muscle weakness. The loss in body weight, which is closely associated with bone structure, is particularly evident in EAMG mice with severe muscle weakness. However, the relationship between muscle weakness and bone loss in EAMG has not been studied before. Recent investigations on bone have shed light on association of bone health and immunological states. It is possible that muscle weakness in EAMG developed by anti-tAChR immune responses might accompany bone loss. We determined whether reduced muscle strength associates with decreased bone mineral density (BMD) in EAMG mice. EAMG was induced by two injections at 4-week interval of tAChR and adjuvants in two different age groups. The first tAChR injection was either at age 8 weeks or at 15 weeks. We measured BMD at three skeletal sites, including femur, tibia, and lumbar vertebrae, using dual energy X-ray absorptiometry. Among these bone areas, femur of EAMG mice in both age groups showed a significant decrease in BMD compared to control adjuvant-injected and to non-immunized mice. Reduction in BMD in induced EAMG at a later-age appears to parallel the severity of the disease. The results indicate that anti-tAChR autoimmune response alone can reduce bone density in EAMG mice. BMD reduction was also observed in adjuvant-injected mice in comparison to normal un-injected mice, suggesting that BMD decrease can occur even when muscle activity is normal. Decreased BMD observed in both tAChR-injected and adjuvant-injected mice groups were discussed in relation to innate immunity and bone-related immunology involving activated T cells and tumour necrosis factor-related cytokines that trigger osteoclastogenesis and bone loss.
- Published
- 2017
- Full Text
- View/download PDF
32. Ablation of IL-17 expression moderates experimental autoimmune myasthenia gravis disease severity
- Author
-
Henry J. Kaminski, Jarrod Sheehan, Yanchen Xie, Gabriela Aguilo-Seara, and Linda L. Kusner
- Subjects
Weakness ,medicine.medical_specialty ,medicine.medical_treatment ,Immunology ,Autoimmunity ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Severity of Illness Index ,T-Lymphocytes, Regulatory ,Biochemistry ,Neuromuscular junction ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Humans ,Immunology and Allergy ,Molecular Biology ,Autoantibodies ,Acetylcholine receptor ,Mice, Knockout ,business.industry ,Interleukin-17 ,FOXP3 ,Forkhead Transcription Factors ,Hematology ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,Cytokine ,Immunoglobulin G ,Proto-Oncogene Proteins c-bcl-6 ,Cytokines ,Interleukin 17 ,medicine.symptom ,business ,030217 neurology & neurosurgery ,030215 immunology - 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.
- Published
- 2017
- Full Text
- View/download PDF
33. Therapeutic Effect of Bifidobacterium Administration on Experimental Autoimmune Myasthenia Gravis in Lewis Rats
- Author
-
Elena Rinaldi, Lorenzo Morelli, Alessandra Fontana, Chiara Cordiglieri, Alessandra Consonni, Fulvio Baggi, Grazia Sacco, Marina Elli, Renato Mantegazza, and Camilla Crasà
- Subjects
lcsh:Immunologic diseases. Allergy ,immunoregulation ,Immunology ,Spleen ,Autoimmunity ,Biology ,Gut flora ,EAMG ,law.invention ,Probiotic ,Immune system ,law ,Cell Movement ,RNA, Ribosomal, 16S ,medicine ,Immunology and Allergy ,Animals ,Bifidobacterium ,Original Research ,Lachnospiraceae ,MG ,FOXP3 ,High-Throughput Nucleotide Sequencing ,Akkermansia ,therapeutic treatment ,Dendritic Cells ,biology.organism_classification ,Gastrointestinal Microbiome ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Disease Models, Animal ,medicine.anatomical_structure ,probiotics ,Rats, Inbred Lew ,Settore AGR/16 - MICROBIOLOGIA AGRARIA ,Metagenome ,Female ,Metagenomics ,lcsh:RC581-607 - Abstract
Beneficial effects of probiotics on gut microbiota homeostasis and inflammatory immune responses suggested the investigation of their potential clinical efficacy in experimental models of autoimmune diseases. Indeed, administration of two bifidobacteria and lactobacilli probiotic strains prevented disease manifestations in the Lewis rat model of Myasthenia Gravis (EAMG). Here, we demonstrate the clinical efficacy of therapeutic administration of vital bifidobacteria (i.e., from EAMG onset). The mechanisms involved in immunomodulation were investigated with ex vivo and in vitro experiments. Improvement of EAMG symptoms was associated to decreased anti-rat AChR antibody levels, and differential expression of TGFβ and FoxP3 immunoregulatory transcripts in draining lymph nodes and spleen of treated-EAMG rats. Exposure of rat bone marrow-derived dendritic cells to bifidobacteria or lactobacilli strains upregulated toll-like receptor 2 mRNA expression, a key molecule involved in bacterium recognition via lipotheicoic acid. Live imaging experiments of AChR-specific effector T cells, co-cultured with BMDCs pre-exposed to bifidobacteria, demonstrated increased percentages of motile effector T cells, suggesting a hindered formation of TCR-peptide-MHC complex. Composition of gut microbiota was studied by 16S rRNA gene sequencing, and α and β diversity were determined in probiotic treated EAMG rats, with altered ratios between Tenericutes and Verrucomicrobia (phylum level), and Ruminococcaceae and Lachnospiraceae (family level). Moreover, the relative abundance of Akkermansia genus was found increased compared to healthy and probiotic treated EAMG rats. In conclusion, our findings confirms that the administration of vital bifidobacteria at EAMG onset has beneficial effects on disease progression; this study further supports preclinical research in human MG to evaluate probiotic efficacy as supplementary therapy in MG.
- Published
- 2019
- Full Text
- View/download PDF
34. Exosomes derived from statin-modified bone marrow dendritic cells increase thymus-derived natural regulatory T cells in experimental autoimmune myasthenia gravis
- Author
-
Rui-Sheng Duan, Yu-Dong Liu, Min Zhang, Chun-Lin Yang, Tong Du, Ying-Chun Dou, Ru-Tao Liu, Peng Zhang, Heng Li, Xiao-Li Li, and Meng-Ru Ge
- Subjects
0301 basic medicine ,Stromal cell ,Immunology ,Bone Marrow Cells ,Thymus Gland ,Exosomes ,T-Lymphocytes, Regulatory ,lcsh:RC346-429 ,Flow cytometry ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,polycyclic compounds ,Atorvastatin ,medicine ,Animals ,lcsh:Neurology. Diseases of the nervous system ,CD40 ,medicine.diagnostic_test ,biology ,Research ,Experimental autoimmune myasthenia gravis (EAMG) ,General Neuroscience ,FOXP3 ,Cell Differentiation ,Dendritic Cells ,Foxp3+ natural regulatory T cells ,medicine.disease ,Microvesicles ,In vitro ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Thymus ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Rats, Inbred Lew ,Aire ,Cancer research ,biology.protein ,Female ,Bone marrow ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,hormones, hormone substitutes, and hormone antagonists ,030217 neurology & neurosurgery ,Transcription Factors - 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.
- Published
- 2019
- Full Text
- View/download PDF
35. Antigen-specific immunoadsorption of MuSK autoantibodies as a treatment of MuSK-induced experimental autoimmune myasthenia gravis
- Author
-
Konstantinos Lazaridis, Vasiliki Baltatzidou, Socrates J. Tzartos, and Nikolaos Tektonidis
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Neuromuscular junction ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Animals ,Humans ,Receptors, Cholinergic ,Immunoadsorption ,Autoantibodies ,Autoimmune disease ,biology ,business.industry ,Autoantibody ,Histocompatibility Antigens Class II ,Receptor Protein-Tyrosine Kinases ,Plasmapheresis ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Titer ,030104 developmental biology ,medicine.anatomical_structure ,Treatment Outcome ,Neurology ,Rats, Inbred Lew ,biology.protein ,Female ,Immunization ,Neurology (clinical) ,Antibody ,business ,030217 neurology & neurosurgery - Abstract
Myasthenia gravis (MG) is an autoimmune disease affecting the neuromuscular junction. Approximately 9% of MG patients have autoantibodies targeting the muscle specific kinase (MuSK), and are challenging therapeutically, since they often present with more severe symptoms. A useful therapy is plasmapheresis, but it is highly non-specific. Antigen-specific immunoadsorption would only remove the pathogenic autoantibodies, minimizing the possible side effects and maximizing the benefit. We used rats with human MuSK-induced experimental autoimmune MG to perform antigen-specific immunoadsorptions, and found it very effective, resulting in a dramatic autoantibody titer decrease, while immunoadsorbed, but not mock-treated, animals showed an significant improvement of their clinical symptoms. Overall, the procedure was efficient, supporting its application for MG treatment.
- Published
- 2019
36. Efgartigimod improves muscle weakness in a mouse model for muscle-specific kinase myasthenia gravis
- Author
-
Inge E. van Es, Jan J.G.M. Verschuuren, Hans de Haard, Maartje G. Huijbers, Samar Kamar-Al Majidi, Jaap J. Plomp, Peter Ulrichts, Silvère M. van der Maarel, Yvonne E. Fillié-Grijpma, and Erik G. Hofman
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Neuroimmunology ,Action Potentials ,Neuromuscular junction ,Electromyography ,Mice, SCID ,Receptors, Fc ,In Vitro Techniques ,Motor Endplate ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neonatal Fc receptor ,Developmental Neuroscience ,In vivo ,Mice, Inbred NOD ,Internal medicine ,medicine ,Animals ,Humans ,Myasthenia gravis ,Acetylcholine receptor ,MuSK ,Muscle Weakness ,medicine.diagnostic_test ,business.industry ,Facial weakness ,Muscle weakness ,Receptor Protein-Tyrosine Kinases ,medicine.disease ,Immunoglobulin Fc Fragments ,Myasthenia Gravis, Autoimmune, Experimental ,Neuromuscular diseases ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Neurology ,Immunoglobulin G ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Muscle Contraction - Abstract
Myasthenia gravis is hallmarked by fatigable muscle weakness resulting from neuromuscular synapse dysfunction caused by IgG autoantibodies. The variant with muscle-specific kinase (MuSK) autoantibodies is characterized by prominent cranial and bulbar weakness and a high frequency of respiratory crises. The majority of MuSK MG patients requires long-term immunosuppressive treatment, but the result of these treatments is considered less satisfactory than in MG with acetylcholine receptor antibodies. Emergency treatments are more frequently needed, and many patients develop permanent facial weakness and nasal speech. Therefore, new treatment options would be welcome. The neonatal Fc receptor protects IgG from lysosomal breakdown, thus prolonging IgG serum half-life. Neonatal Fc receptor antagonism lowers serum IgG levels and thus may act therapeutically in autoantibody-mediated disorders. In MuSK MG, IgG4 anti-MuSK titres closely correlate with disease severity. We therefore tested efgartigimod (ARGX-113), a new neonatal Fc receptor blocker, in a mouse model for MuSK myasthenia gravis. This model involves 11 daily injections of purified IgG4 from MuSK myasthenia gravis patients, resulting in overt myasthenic muscle weakness and, consequently, body weight loss. Daily treatment with 0.5 mg efgartigimod, starting at the fifth passive transfer day, reduced the human IgG4 titres about 8-fold, despite continued daily injection. In muscle strength and fatigability tests, efgartigimod-treated myasthenic mice outperformed control myasthenic mice. Electromyography in calf muscles at endpoint demonstrated less myasthenic decrement of compound muscle action potentials in efgartigimod-treated mice. These substantial in vivo improvements of efgartigimod-treated MuSK MG mice following a limited drug exposure period were paralleled by a tendency of recovery at neuromuscular synaptic level (in various muscles), as demonstrated by ex vivo functional studies. These synaptic improvements may well become more explicit upon longer drug exposure. In conclusion, our study shows that efgartigimod has clear therapeutic potential in MuSK myasthenia gravis and forms an exciting candidate drug for many autoantibody-mediated neurological and other disorders.
- Published
- 2019
37. Correction to: Exosomes derived from atorvastatin-modified bone marrow dendritic cells ameliorate experimental autoimmune myasthenia gravis by up-regulated levels of IDO/Treg and partly dependent on FasL/Fas pathway
- Author
-
Xiao-Li Li, Heng Li, Min Zhang, Hua Xu, Long-Tao Yue, Xin-Xin Zhang, Shan Wang, Cong-Cong Wang, Yan-Bin Li, Ying-Chun Dou, and Rui-Sheng Duan
- Subjects
Fas Ligand Protein ,Immunology ,Exosomes ,T-Lymphocytes, Regulatory ,lcsh:RC346-429 ,Cellular and Molecular Neuroscience ,Bone Marrow ,Animals ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,fas Receptor ,lcsh:Neurology. Diseases of the nervous system ,General Neuroscience ,Correction ,Forkhead Transcription Factors ,Dendritic Cells ,Flow Cytometry ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Up-Regulation ,Disease Models, Animal ,Microscopy, Electron ,Neurology ,Rats, Inbred Lew ,Cytokines ,Female ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Signal Transduction - Abstract
Previously, we have demonstrated that spleen-derived dendritic cells (DCs) modified with atorvastatin suppressed immune responses of experimental autoimmune myasthenia gravis (EAMG). However, the effects of exosomes derived from atorvastatin-modified bone marrow DCs (BMDCs) (statin-Dex) on EAMG are still unknown.Immunophenotypical characterization of exosomes from atorvastatin- and dimethylsulfoxide (DMSO)-modified BMDCs was performed by electron microscopy, flow cytometry, and western blotting. In order to investigate whether statin-DCs-derived exosomes (Dex) could induce immune tolerance in EAMG, we administrated statin-Dex, control-Dex, or phosphate-buffered saline (PBS) into EAMG rats via tail vein injection. The tracking of injected Dex and the effect of statin-Dex injection on endogenous DCs were performed by immunofluorescence and flow cytometry, respectively. The number of Foxp3(+) cells in thymuses was examined using immunocytochemistry. Treg cells, cytokine secretion, lymphocyte proliferation, cell viability and apoptosis, and the levels of autoantibody were also carried out to evaluate the effect of statin-Dex on EAMG rats. To further investigate the involvement of FasL/Fas in statin-Dex-induced apoptosis, the underlying mechanisms were studied by FasL neutralization assays.Our data showed that the systemic injection of statin-Dex suppressed the clinical symptoms of EAMG rats. These statin-Dex had immune regulation functions in immune organs, such as the spleen, thymus, and popliteal and inguinal lymph nodes. Furthermore, statin-Dex exerted their immunomodulatory effects in vivo by decreasing the expression of CD80, CD86, and MHC class II on endogenous DCs. Importantly, the therapeutic effects of statin-Dex on EAMG rats were associated with up-regulated levels of indoleamine 2,3-dioxygenase (IDO)/Treg and partly dependent on FasL/Fas pathway, which finally resulted in decreased synthesis of anti-R97-116 IgG, IgG2a, and IgG2b antibodies.Our data suggest that atorvastatin-induced immature BMDCs are able to secrete tolerogenic Dex, which are involved in the suppression of immune responses in EAMG rats. Importantly, our study provides a novel cell-free approach for the treatment of autoimmune diseases.
- Published
- 2019
38. Profiling of patient-specific myocytes identifies altered gene expression in the ophthalmoplegic subphenotype of myasthenia gravis
- Author
-
Sharon Prince, Jeannine M. Heckmann, and Melissa Nel
- Subjects
0301 basic medicine ,Adult ,Male ,medicine.medical_specialty ,Adolescent ,Subphenotype ,lcsh:Medicine ,030105 genetics & heredity ,Biology ,Receptors, Nicotinic ,Extraocular muscles ,MyoD ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,Gene expression ,Exome Sequencing ,medicine ,Myocyte ,Humans ,Pharmacology (medical) ,Gene ,Myasthenia gravis ,Genetics (clinical) ,Exome sequencing ,Cells, Cultured ,Acetylcholine receptor ,Skin ,Muscle Cells ,Transdifferentiation ,Ophthalmoplegia ,Research ,lcsh:R ,General Medicine ,Fibroblasts ,medicine.disease ,Myasthenia Gravis, Autoimmune, Experimental ,medicine.anatomical_structure ,Endocrinology ,Gene Expression Regulation ,Female ,Myotranscriptome ,030217 neurology & neurosurgery - Abstract
Background: While extraocular muscles are affected early in myasthenia gravis (MG), but respond to treatment, we observe a high incidence of treatment-resistant ophthalmoplegia (OP-MG) among MG subjects with African genetic ancestry. Previously, using whole exome sequencing, we reported potentially functional variants which associated with OP-MG. The aim of this study was to profile the expression of genes harbouring the OP-MG associated variants using patient-derived subphenotype-specific ‘myocyte’ cultures. Methods From well-characterised MG patients we developed the ‘myocyte’ culture models by transdifferentiating dermal fibroblasts using an adenovirus expressing MyoD. These myocyte cultures were treated with homologous acetylcholine receptor antibody-positive myasthenic sera to induce muscle transcripts in response to an MG stimulus. Gene expression in myocytes derived from OP-MG (n = 10) and control MG subjects (MG without ophthalmoplegia; n = 6) was quantified using a custom qPCR array profiling 93 potentially relevant genes which included the putative OP-MG susceptibility genes and other previously reported genes of interest in MG and experimental autoimmune myasthenia gravis (EAMG). Results OP-MG myocytes compared to control MG myocytes showed altered expression of four OP-MG susceptibility genes (PPP6R2, CANX, FAM136A and FAM69A) as well as several MG and EAMG genes (p 0.78, p
- Published
- 2019
39. Ginsenoside Rb1: The new treatment measure of myasthenia gravis
- Author
-
Yan-Jun Wang, Shen Liu, Qing-Fang Meng, Yan-Bin Li, Heng Li, Chun-hong Li, Xiao-Li Li, Wei Chen, Jun-Kang Sui, and Cong-Cong Wang
- Subjects
0301 basic medicine ,Neuromuscular disease ,Ginsenosides ,Immunology ,T-Lymphocytes, Regulatory ,03 medical and health sciences ,chemistry.chemical_compound ,Ginseng ,Th2 Cells ,0302 clinical medicine ,Immune system ,Animals ,Immunologic Factors ,Immunology and Allergy ,Medicine ,Pharmacology ,biology ,business.industry ,Muscle weakness ,medicine.disease ,eye diseases ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Interleukin 10 ,030104 developmental biology ,chemistry ,Rats, Inbred Lew ,Ginsenoside ,Immunoglobulin G ,Leukocytes, Mononuclear ,biology.protein ,Cytokines ,Th17 Cells ,Female ,Lymph Nodes ,Antibody ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Myasthenia gravis (MG) is an autoimmune neuromuscular disease characterized by fatigue and muscle weakness. Ginseng is used in the treatment of MG. Ginsenoside Rb1 (G-Rb1), the most abundant ginsenoside in ginseng root, has been proved to be immune regulatory in various diseases. In this study, we investigated the effects and mechanisms of G-Rb1 in treatment for MG in a rat model. Our data showed that G-Rb1 treatment markedly ameliorated the symptoms of experimental autoimmune myasthenia gravis (EAMG) rats, decreased the percentage of Th17 cells in mononuclear cells (MNCs), and increased the number of Treg and Th2 cells in MNCs. We also found that G-Rb1 treatment decreased the serum level of anti-R97-116 peptides IgG1 and IgG2a antibodies. Our findings provide strong evidence that G-Rb1 treatment has immune regulatory effects in EAMG rats, which indicate that G-Rb1 may be employed as a therapeutic medication for MG.
- Published
- 2016
- Full Text
- View/download PDF
40. Curcumin ameliorates experimental autoimmune myasthenia gravis by diverse immune cells
- Author
-
Heng Li, Cong-Cong Wang, Ying Liu, Min Zhang, Long-Tao Yue, Shan Wang, Peng Zhang, and Rui-Sheng Duan
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Curcumin ,medicine.medical_treatment ,T cell ,Genes, MHC Class II ,Natural killer cell ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Lymphocytes ,CD86 ,Chemistry ,General Neuroscience ,Natural killer T cell ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Killer Cells, Natural ,030104 developmental biology ,medicine.anatomical_structure ,Cytokine ,Rats, Inbred Lew ,Immunology ,B7-1 Antigen ,Leukocytes, Mononuclear ,Cytokines ,Natural Killer T-Cells ,Female ,B7-2 Antigen ,Inflammation Mediators ,CD80 ,030215 immunology - Abstract
Curcumin is a traditional Asian medicine with diverse immunomodulatory properties used therapeutically in the treatment of many autoimmune diseases. However, the effects of curcumin on myasthenia gravis (MG) remain undefined. Here we investigated the effects and potential mechanisms of curcumin in experimental autoimmune myasthenia gravis (EAMG). Our results demonstrated that curcumin ameliorated the clinical scores of EAMG, suppressed the expression of T cell co-stimulatory molecules (CD80 and CD86) and MHC class II, down-regulated the levels of pro-inflammatory cytokines (IL-17, IFN-γ and TNF-α) and up-regulated the levels of the anti-inflammatory cytokine IL-10, shifted the balance from Th1/Th17 toward Th2/Treg, and increased the numbers of NKR-P1(+) cells (natural killer cell receptor protein 1 positive cells, including NK and NKT cells). Moreover, the administration of curcumin promoted the differentiation of B cells into a subset of B10 cells, increased the anti-R97-166 peptide IgG1 levels and decreased the relative affinity indexes of anti-R97-116 peptide IgG. In summary, curcumin effectively ameliorate EAMG, indicating that curcumin may be a potential candidate therapeutic agent for MG.
- Published
- 2016
- Full Text
- View/download PDF
41. ROCK inhibitor abolishes the antibody response in experimental autoimmune myasthenia gravis
- Author
-
Shuai Miao, Peng Zhang, Long-Tao Yue, Yan-Bin Li, Min Zhang, Rui-Sheng Duan, Ying-Chun Dou, Cong-Cong Wang, Heng Li, and Xiao-Li Li
- Subjects
0301 basic medicine ,Autoimmunity ,Biology ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Immune system ,1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine ,medicine ,Animals ,Protein Kinase Inhibitors ,Molecular Biology ,Rho-associated protein kinase ,B-Lymphocytes ,rho-Associated Kinases ,Fasudil ,Germinal center ,T-Lymphocytes, Helper-Inducer ,Cell Biology ,Germinal Center ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,030104 developmental biology ,Rats, Inbred Lew ,Rho kinase inhibitor ,Immunology ,biology.protein ,Female ,Antibody ,CD80 - Abstract
The Rho/Rho kinase (ROCK) pathway serves as molecular switches in many biological processes including the immune response. ROCK inhibitors lead to amelioration of some autoimmune diseases. The present study was designed to define whether a selective ROCK inhibitor, fasudil, was effective in experimental autoimmune myasthenia gravis (EAMG) and investigate the underlying mechanisms. Here we found fasudil effectively attenuated the development of ongoing EAMG. Fasudil abolished the antibody production and function by decreasing follicular helper T cells and CD19(+) B cells, especially germinal center B cells. Moreover, fasudil reduced the expression of CD80 on lymph node mononuclear cells. These findings suggest the inhibition of ROCK might be a potential therapeutic strategy for antibody-mediated autoimmune diseases.
- Published
- 2016
- Full Text
- View/download PDF
42. Review on Toll-Like Receptor Activation in Myasthenia Gravis: Application to the Development of New Experimental Models
- Author
-
Rozen Le Panse, Marieke Robinet, Mélanie A. Cron, Sonia Berrih-Aknin, Solène Maillard, Centre de recherche en myologie, Université Pierre et Marie Curie - Paris 6 (UPMC)-Association française contre les myopathies (AFM-Téléthon)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut de Myologie, Université Pierre et Marie Curie - Paris 6 (UPMC)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Association française contre les myopathies (AFM-Téléthon)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), HAL-UPMC, Gestionnaire, Centre de recherche en Myologie – U974 SU-INSERM, and Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU)
- Subjects
0301 basic medicine ,Autoimmune diseases ,Mouse experimental models ,[SDV.IMM.II]Life Sciences [q-bio]/Immunology/Innate immunity ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Interferon ,Myasthenia Gravis ,Animals ,Humans ,Immunology and Allergy ,Medicine ,[SDV.IMM.II] Life Sciences [q-bio]/Immunology/Innate immunity ,Sensitization ,Inflammation ,Innate immunity ,Toll-like receptor ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,business.industry ,Toll-Like Receptors ,Autoantibody ,Germinal center ,General Medicine ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,Complete Freund's adjuvant ,TLR4 ,Interferon type-I ,business ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,030217 neurology & neurosurgery ,medicine.drug - Abstract
International audience; Abnormal toll-like receptor (TLR) activation and uncontrolled resolution of inflammation are suspected to play a key role in the development of autoimmune diseases. Acquired myasthenia gravis (MG) is an invalidating neuromuscular disease leading to muscle weaknesses. MG is mainly mediated by anti-acetylcholine receptor (AChR) autoantibodies, and thymic hyperplasia characterized by ectopic germinal centers is a common feature in MG. An abnormal expression of certain TLRs is observed in the thymus of MG patients associated with the overexpression of interferon (IFN)-β, the orchestrator of thymic changes in MG. Experimental models have been developed for numerous autoimmune diseases. These models are induced by animal immunization with a purified antigen solubilized in complete Freund’s adjuvant (CFA) containing heat-inactivated mycobacterium tuberculosis (MTB). Sensitization against the antigen is mainly due to the activation of TLR signaling pathways by the pathogen motifs displayed by MTB, and attempts have been made to substitute the use of CFA by TLR agonists. AChR emulsified in CFA is used to induce the classical experimental autoimmune MG model (EAMG). However, the TLR4 activator lipopolysaccharide (LPS) has proved to be efficient to replace MTB and induce a sensitization against purified AChR. Poly(I:C), the well-known TLR3 agonist, is also able by itself to induce MG symptoms in mice associated with early thymic changes as observed in human MG. In this review, we discuss the abnormal expression of TLRs in MG patients and we describe the use of TLR agonists to induce EAMG in comparison with other autoimmune experimental models.
- Published
- 2016
- Full Text
- View/download PDF
43. Suppression of experimental autoimmune myasthenia gravis by autologous T regulatory cells
- Author
-
Revital Aricha, Miriam C. Souroujon, Sara Fuchs, and Debby Reuveni
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Lymphoid Tissue ,Immunology ,Population ,CD11c ,chemical and pharmacologic phenomena ,Cell Communication ,Receptors, Nicotinic ,T-Lymphocytes, Regulatory ,Immunophenotyping ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,T-Lymphocyte Subsets ,Splenocyte ,medicine ,Animals ,Immunology and Allergy ,education ,Immunosuppression Therapy ,education.field_of_study ,business.industry ,FOXP3 ,hemic and immune systems ,Dendritic Cells ,medicine.disease ,Adoptive Transfer ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Disease Models, Animal ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,Female ,Bone marrow ,business ,CD8 ,030215 immunology - Abstract
Adoptive transfer of regulatory T (Treg) cells have been employed effectively for suppression of several animal models for autoimmune diseases. In order to employ Treg cell therapy in patients, it is necessary to generate Treg cells from the patient's own cells (autologous) that would be able to suppress effectively the disease in vivo, upon their reintroduction to the patient. Towards this objective, we report in the present study on a protocol for a successful immune-regulation of experimental autoimmune myasthenia gravis (EAMG) by ex vivo--generated autologous Treg cells. For this protocol bone marrow (BM) cells, are first cultured in the presence of GM-CSF, giving rise to a population of CD11c(+)MHCII(+)CD45RA(+)CD8(-) DCs (BMDCs). Splenic CD4(+) T cells are then co-cultured with the differentiated BM cells and expand to 90% of Foxp3(+) Treg cells. In vitro assay exhibits a similar dose dependent manner in the suppression of T effector cells proliferation between Treg cells obtained from either healthy or sick donors. In addition, both Treg cells inhibit similarly the secretion of IFN-γ from activated splenocytes. Administration of 1 × 10(6) ex-vivo generated Treg cells, I.V, to EAMG rats, modulates the disease following a single treatment, given 3 days or 3 weeks after disease induction. Similar disease inhibition was achieved when CD4 cells were taken from either healthy or sick donors. The disease suppression was accompanied by reduced levels of total AChR specific antibodies in the serum. Moreover, due to the polyclonality of the described Treg cell, we have examined whether this treatment approach could be also employed for the treatment of other autoimmune diseases involving Treg cells. Indeed, we demonstrated that the ex-vivo generated autologous Treg cells suppress Adjuvant Arthritis (AA) in rats. This study opens the way for the application of induced autologous Treg cell therapy for myasthenia gravis, as well as for other human autoimmune diseases involving Treg cells.
- Published
- 2016
- Full Text
- View/download PDF
44. Remission of Severe Myasthenia Gravis After Massive-Dose Vitamin D Treatment
- Author
-
Flávio Adsuara Cadegiani
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Specialty ,MEDLINE ,Severity of Illness Index ,03 medical and health sciences ,Remission induction ,0302 clinical medicine ,Internal medicine ,Severity of illness ,Myasthenia Gravis ,Vitamin D and neurology ,Medicine ,Humans ,Vitamin D ,Dose-Response Relationship, Drug ,business.industry ,Remission Induction ,Muscle weakness ,General Medicine ,Articles ,Vitamins ,Middle Aged ,medicine.disease ,Myasthenia gravis ,030104 developmental biology ,Immunology ,Myasthenia gravis, Autoimmune, Experimental ,Female ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Patient: Female, 49 Final Diagnosis: Myasthenia Gravis Symptoms: Muscle weakness • shortness of breath Medication: — Clinical Procedure: Vitamin D Specialty: Endocrinology and Metabolic Objective: Unusual or unexpected effect of treatment Background: Vitamin D has been shown to be related to autoimmune diseases, such as multiple sclerosis and psoriasis. Correlations have been reported between vitamin D levels and prevalence and severity of other autoimmune disorders, and also between vitamin D therapy and disease improvement and remission. Case Report: This is a case report of a patient with severe and refractory myasthenia gravis (MG) who followed a “high-dose vitamin D treatment”, a massive-dose treatment (80 000 to 120 000 IU/day) promoted by a medical center in Brazil (but still not proven), and she had her first complete remission after this type of treatment with increased vitamin D serum levels (400 to 700 ng/mL). Conclusions: This case report may reinforce the reported correlation between vitamin D level and disease severity and introduces a possible new use for vitamin D as a potential target for treating autoimmune diseases. We recommend large, double-blind, placebo-controlled, randomized studies using high-dose vitamin D treatment for refractory autoimmune diseases to reliably assess this pharmacotherapy target for these diseases.
- Published
- 2016
45. Effect of Grilled Nux Vomica on Differential RNA Expression Profile of Gastrocnemius Muscle and Toll-Like Receptor 4 (TLR-4)/Nuclear Factor kappa B (NF-κB) Signaling in Experimental Autoimmune Myasthenia Gravis Rats
- Author
-
Xu Hong Jiang, Di Yi Zhou, Hui Qiu, Yi Chen, Yang Yang Ding, and Chang Lin Qiu
- Subjects
030204 cardiovascular system & hematology ,Pharmacology ,Proinflammatory cytokine ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Western blot ,medicine ,Animals ,Humans ,RNA, Messenger ,RNA-Seq ,Muscle, Skeletal ,Acetylcholine receptor ,medicine.diagnostic_test ,Chemistry ,Animal Study ,Gene Expression Profiling ,Autoantibody ,NF-kappa B ,Muscle weakness ,Strychnos nux-vomica ,General Medicine ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Specific Pathogen-Free Organisms ,Toll-Like Receptor 4 ,MYL3 ,Gene Expression Regulation ,Rats, Inbred Lew ,030220 oncology & carcinogenesis ,Female ,medicine.symptom ,Drugs, Chinese Herbal ,Signal Transduction - 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-s1. 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.
- Published
- 2020
46. A Natural Variant of the Signaling Molecule Vav1 Enhances Susceptibility to Myasthenia Gravis and Influences the T Cell Receptor Repertoire
- Author
-
Bernard, Isabelle, Sacquin, Antoine, Kassem, Sahar, Benamar, Mehdi, Colacios, Céline, Gador, Mylène, Pérals, Corine, Fazilleau, Nicolas, Saoudi, Abdelhadi, Centre de Physiopathologie Toulouse Purpan ex IFR 30 et IFR 150 (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre de Physiopathologie Toulouse Purpan (CPTP), This work was supported by INSERM, Association Française contre les Myopathies, Fight-MG (FP7-Health-2009-242210), the Agence Nationale de la Recherche and the Aninfimip, an EquipEx program (Equipement d’Excellence) supported by the French government through the Investments for the Future program (ANR-11-EQPX-0003). AS is supported by Centre National de la Recherche Scientifique., Centre de Physiopathologie Toulouse Purpan (CPTP - U1043 INSERM - UMR5282 CNRS - UT3), and Fazilleau, Nicolas
- Subjects
CD4-Positive T-Lymphocytes ,lcsh:Immunologic diseases. Allergy ,[SDV.IMM] Life Sciences [q-bio]/Immunology ,T-Lymphocytes ,[SDV]Life Sciences [q-bio] ,Immunology ,Receptors, Antigen, T-Cell ,T cells ,T-Cell Antigen Receptor Specificity ,Receptors, Nicotinic ,Mice ,Immunology and Allergy ,Animals ,Proto-Oncogene Proteins c-vav ,ComputingMilieux_MISCELLANEOUS ,Original Research ,myasthenia gravis ,Genetic Variation ,animal models ,Myasthenia Gravis, Autoimmune, Experimental ,Phenotype ,[SDV.IMM.IA]Life Sciences [q-bio]/Immunology/Adaptive immunology ,[SDV.IMM.IA] Life Sciences [q-bio]/Immunology/Adaptive immunology ,Vav1 ,Cytokines ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Disease Susceptibility ,lcsh:RC581-607 ,T cell repertoire ,Signal Transduction - Abstract
International audience; The guanine nucleotide exchange factor Vav1 is essential for transducing T cell receptor (TCR) signals and plays an important role in T cell development and activation. Previous genetic studies identified a natural variant of Vav1 characterized by the substitution of an arginine (R) residue by a tryptophane (W) at position 63 (Vav1R63W). This variant impacts Vav1 adaptor functions and controls susceptibility to T cell-mediated neuroinflammation. To assess the implication of this Vav1 variant on the susceptibility to antibody-mediated diseases, we used the animal model of myasthenia gravis, experimental autoimmune myasthenia gravis (EAMG). To this end, we generated a knock-in (KI) mouse model bearing a R to W substitution in the Vav1 gene (Vav1R63W) and immunized it with either torpedo acetylcholine receptor (tAChR) or the α146-162 immunodominant peptide. We observed that the Vav1R63W conferred increased susceptibility to EAMG, revealed by a higher AChR loss together with an increased production of effector cytokines (IFN-γ, IL-17A, GM-CSF) by antigen-specific CD4+ T cells, as well as an increased frequency of antigen-specific CD4+ T cells. This correlated with the emergence of a dominant antigen-specific T cell clone in KI mice that was not present in wild-type mice, suggesting an impact on thymic selection and/or a different clonal selection threshold following antigen encounter. Our results highlight the key role of Vav1 in the pathophysiology of EAMG and this was associated with an impact on the TCR repertoire of AChR reactive T lymphocytes.
- Published
- 2018
- Full Text
- View/download PDF
47. Artemisinin ameliorates the symptoms of experimental autoimmune myasthenia gravis by regulating the balance of TH1 cells, TH17 cells and Treg cells
- Author
-
W, Chen, F F, Li, C, Li, J K, Sui, Q F, Meng, X L, Li, H, Li, C H, Li, and Y B, Li
- Subjects
Animals ,Th17 Cells ,Th1 Cells ,T-Lymphocytes, Regulatory ,Artemisinins ,Myasthenia Gravis, Autoimmune, Experimental ,Rats - 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-β1 (TGF-β1) and Treg cells in MNCs. These findings indicate that artemisinin may be a new choice for MG treatment.
- Published
- 2018
48. Therapeutic potential of artesunate in experimental autoimmune myasthenia gravis by upregulated T regulatory cells and regulation of Th1/Th2 cytokines
- Author
-
Qing-Fang, Meng, Xin-Xin, Zhang, Zheng, Zhang, Wei, Chen, Xiao-Li, Li, Yan-Jun, Wang, Fei-Fei, Li, and Yan-Bin, Li
- Subjects
Dose-Response Relationship, Drug ,Artesunate ,Th1 Cells ,T-Lymphocytes, Regulatory ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,Up-Regulation ,Antimalarials ,Th2 Cells ,Rats, Inbred Lew ,Animals ,Cytokines ,Immunologic Factors ,Female ,Cell Proliferation - 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).
- Published
- 2018
49. Exogenous IL-9 Ameliorates Experimental Autoimmune Myasthenia Gravis Symptoms in Rats
- Author
-
Xiuhua Yao, Lixia Xu, Hulun Li, Lili Mu, Jinghua Wang, Qingfei Kong, Bo Sun, Xiaoli Xie, and Jiarui Zhao
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Autoantigens ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Interferon ,Myasthenia Gravis ,medicine ,Animals ,Humans ,Receptors, Cholinergic ,Fluorescein isothiocyanate ,Antigen-presenting cell ,Autoantibodies ,Receptors, Interleukin-9 ,biology ,Chemistry ,Experimental autoimmune encephalomyelitis ,Interleukin-9 ,General Medicine ,medicine.disease ,Myasthenia gravis ,Recombinant Proteins ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,030104 developmental biology ,Cytokine ,Rats, Inbred Lew ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Immunotherapy ,Antibody ,Peptides ,Fetal bovine serum ,medicine.drug - 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).
- Published
- 2018
50. Treatment of experimental autoimmune myasthenia gravis rats with FTY720 and its effect on Th1/Th2 cells
- Author
-
Ting Zhang, Jian-Kang Huang, Yuwu Zhao, and Hongmei Wang
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_specialty ,Biochemistry ,Neuromuscular junction ,03 medical and health sciences ,Th2 Cells ,Immune system ,Interferon ,hemic and lymphatic diseases ,Internal medicine ,Genetics ,Animals ,Medicine ,Molecular Biology ,Acetylcholine receptor ,biology ,Fingolimod Hydrochloride ,business.industry ,Interleukin ,Th1 Cells ,medicine.disease ,Myasthenia gravis ,Myasthenia Gravis, Autoimmune, Experimental ,Rats ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Oncology ,Rats, Inbred Lew ,biology.protein ,Molecular Medicine ,Experimental pathology ,Female ,Antibody ,business ,Immunosuppressive Agents ,medicine.drug - Abstract
Myasthenia gravis (MG) is an autoimmune neurological disease that is characterized by the expression of anti-acetylcholine receptor (AChR) antibodies. The immune response at AChRs of neuromuscular junction is disrupted in patients with MG, which manifests as skeletal muscle fatigue and is aggravated following periods of activity and alleviated following rest. Although a novel immune suppressant FTY720 drug, which exhibits strong immune suppression efficacy and minor adverse effects, is available, its role and mechanism in MG have not been elucidated. The aim of this study was to investigate the role of FTY720 in MG. A total of 60 healthy female Lewis rats were randomly assigned into 4 groups: Control group, Model group of experimental autoimmune myasthenia gravis (EAMG), 0.5 mg/kg FTY720-treatment EAMG group and 1.0 mg/kg FTY720‑treatment EAMG group. Body weight and symptoms were examined; Lennon score was used to evaluate improvement of clinical symptoms. Reverse transcription‑quantitative polymerase chain reaction and ELISA were used to test the mRNA and protein expression levels, respectively, of the helper T (Th)1 and Th2 cell cytokines, including interleukin (IL)‑2, interferon (IFN)‑γ, IL‑4 and IL‑6 in thymus tissue and serum. FTY720 treatment improved rat MG symptoms, increased body weight and decreased Lennon score. FTY720 treatments also reduced tissue and serum levels of IL‑2, IFN‑γ and IL‑6, but not IL‑4 expression levels. FTY720 suppressed the inflammatory response and improved EAMG symptoms by inhibiting the secretion of inflammatory factors.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.