1
|
Wang B, Zhu Y, Liu D, Hu C, Zhu R. The intricate dance of non-coding RNAs in myasthenia gravis pathogenesis and treatment. Front Immunol 2024; 15:1342213. [PMID: 38605954 PMCID: PMC11007667 DOI: 10.3389/fimmu.2024.1342213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/11/2024] [Indexed: 04/13/2024] Open
Abstract
Myasthenia gravis (MG) stands as a perplexing autoimmune disorder affecting the neuromuscular junction, driven by a multitude of antibodies targeting postsynaptic elements. However, the mystery of MG pathogenesis has yet to be completely uncovered, and its heterogeneity also challenges diagnosis and treatment. Growing evidence shows the differential expression of non-coding RNAs (ncRNAs) in MG has played an essential role in the development of MG in recent years. Remarkably, these aberrantly expressed ncRNAs exhibit distinct profiles within diverse clinical subgroups and among patients harboring various antibody types. Furthermore, they have been implicated in orchestrating the production of inflammatory cytokines, perturbing the equilibrium of T helper 1 cells (Th1), T helper 17 cells (Th17), and regulatory T cells (Tregs), and inciting B cells to generate antibodies. Studies have elucidated that certain ncRNAs mirror the clinical severity of MG, while others may hold therapeutic significance, showcasing a propensity to return to normal levels following appropriate treatments or potentially foretelling the responsiveness to immunosuppressive therapies. Notably, the intricate interplay among these ncRNAs does not follow a linear trajectory but rather assembles into a complex network, with competing endogenous RNA (ceRNA) emerging as a prominent hub in some cases. This comprehensive review consolidates the landscape of dysregulated ncRNAs in MG, briefly delineating their pivotal role in MG pathogenesis. Furthermore, it explores their promise as prospective biomarkers, aiding in the elucidation of disease subtypes, assessment of disease severity, monitoring therapeutic responses, and as novel therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | - Ruixia Zhu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
2
|
García Estévez DA, Pardo Fernández J. Myasthenia gravis. Update on diagnosis and therapy. Med Clin (Barc) 2023:S0025-7753(23)00218-X. [PMID: 37248131 DOI: 10.1016/j.medcli.2023.04.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/31/2023]
Abstract
Myasthenia gravis is an autoimmune disease caused by the presence of specific antibodies targeting different postsynaptic components of the neuromuscular junction, and is clinically characterized by the presence of fatigueable muscle weakness. In the etiopathogenesis plays a central role the thymus and the most frequently detected pathogenic autoantibodies are targeted to the acetylcholine receptor. The increase in the knowledge of the immunological components of the neuromuscular junction in the last two decades has been fundamental to identify new pathogenic antibodies, reduce the percentage of patients with seronegative myasthenia, and propose a classification of patients into subgroups with clinical-therapeutic interest. In addition, in recent years, new drugs have been developed for the treatment of patients with myasthenia gravis that are refractory to conventional immunosuppressive treatment.
Collapse
Affiliation(s)
- Daniel Apolinar García Estévez
- Servicio de Neurología, Complexo Hospitalario Universitario de Ourense, Ourense, España; Grupo de investigación Neurociencias Clínicas, Instituto de Investigaciones Sanitarias Galicia-Sur, SERGAS-UVIGO, Vigo, Pontevedra, España.
| | - Julio Pardo Fernández
- Unidad de Enfermedades Neuromusculares, Servicio de Neurología, Complexo Hospitalario Universitario de Santiago de Compostela. Facultad de Medicina, Universidad de Santiago de Compostela, Santiago de Compostela, A Coruña, España
| |
Collapse
|
3
|
Ao YQ, Jiang JH, Gao J, Wang HK, Ding JY. Recent thymic emigrants as the bridge between thymoma and autoimmune diseases. Biochim Biophys Acta Rev Cancer 2022; 1877:188730. [DOI: 10.1016/j.bbcan.2022.188730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 11/27/2022]
|
4
|
Moodley K, Bill PLA, Patel VB. A Comparison of Clinical, Electro-Diagnostic, Laboratory, and Treatment Outcome Differences in a Cohort of HIV-Infected and HIV-Uninfected Patients With Myasthenia Gravis. Front Neurol 2021; 12:738813. [PMID: 34721269 PMCID: PMC8553930 DOI: 10.3389/fneur.2021.738813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/13/2021] [Indexed: 11/13/2022] Open
Abstract
There is limited literature comparing the clinical parameters and treatment outcomes in HIV-infected and HIV-uninfected myasthenia gravis (MG) patients. The aim of the study was to investigate the clinical differences and treatment outcomes in the two categories of patients, particularly the safe use of immunosuppressive therapy in immunocompromised patients. The study was a retrospective analysis of medical records of MG patients from the neuromuscular unit at Inkosi Albert Luthuli Central Hospital in Kwa-Zulu Natal between 2003 and 2019. One hundred and seventy-eight (178) patients fulfilled the clinical criteria for MG. Twenty-four (13.4%) were HIV-infected and 154 (86.5%) were HIV-uninfected. There were 116 (65%) females, median 45 years, (IQR 40–62), 90 (50.5%) black African, 66 (37%) Indian, 20 (11.2%) white, and 2 (1.1%) of mixed ancestry. In the HIV-infected cohort, 20 (87%) had generalized MG, 12 (50%) bulbar, and 14 (60.9%) respiratory onset MG, 12 (50%) presented with MG Foundation of America (MGFA) class five diseases at diagnosis, six (25%) presented with MG crisis during the 5-year follow-up. Thirteen (54%) of the HIV-infected group required rescue therapy using (plasma exchange or IV immunoglobulin) combined with pulse cyclophosphamide compared with 17 (11%) in the HIV-uninfected cohort, respectively. At 5 years, 8 (33%) of the HIV-infected group remained refractory to treatment compared with 10 (6.5%) HIV-uninfected cohort, respectively. No adverse events were documented in HIV-infected patients receiving combination rescue therapy (PLEX or IVIG combined with IV cyclophosphamide). In conclusion HIV-infected MG patients are more likely to require combination rescue therapy with PE/IVIG and IV cyclophosphamide compared with those who were HIV-uninfected. No side effects were documented in the HIV-infected group receiving the above therapy.
Collapse
Affiliation(s)
- Kaminie Moodley
- Department of Neurology, University of KwaZulu-Natal, Durban, South Africa
| | - Pierre L A Bill
- Department of Neurology, University of KwaZulu-Natal, Durban, South Africa
| | - Vinod B Patel
- Department of Neurology, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
5
|
Liu RT, Li W, Guo D, Yang CL, Ding J, Xu JX, Duan RS. Natural killer cells promote the differentiation of follicular helper T cells instead of inducing apoptosis in myasthenia gravis. Int Immunopharmacol 2021; 98:107880. [PMID: 34174703 DOI: 10.1016/j.intimp.2021.107880] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 06/06/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023]
Abstract
Recent evidence has shown that natural killer (NK) cells have an immunoregulatory function in the pathogenesis of myasthenia gravis (MG). In this study, the phenotype and function of NK cell subsets in peripheral blood of new-onset MG (N-MG) and stable MG (S-MG) patients were explored. Circulating CD56dim and CD56bright NK cells were increased and decreased, respectively, in patients with N-MG and S-MG compared with healthy control (HC). Moreover, all circulating NK cell subsets from N-MG patients showed significantly lower expression of activating receptor NKG2D and production of Interferon (IFN) -γ than that from HC. The killing effects of NK cells on CD4+ T cells and Tfh cells were impaired in MG patients, whereas, they promoted the differentiation and activation of Tfh cells. These data indicated that the immune-regulation of NK cells on CD4+ T cells and Tfh cells in MG patients was abnormal, which may contribute to the immune-pathological mechanism of MG.
Collapse
Affiliation(s)
- Rui-Ting Liu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China; Department of Neurology, Liaocheng People's Hospital, Liaocheng 252000, Shandong Province, PR China
| | - Wei Li
- The Neurosurgical Department, Liaocheng People's Hospital, Liaocheng 252000, Shandong Province, PR China
| | - Dong Guo
- Department of Neurology, Liaocheng People's Hospital, Liaocheng 252000, Shandong Province, PR China
| | - Chun-Lin Yang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China
| | - Jie Ding
- Central Laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China
| | - Jian-Xin Xu
- Department of Neurology, Liaocheng People's Hospital, Liaocheng 252000, Shandong Province, PR China
| | - Rui-Sheng Duan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan 250014, PR China; Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, PR China; Shandong Institute of Neuroimmunology, Jinan 250014, PR China.
| |
Collapse
|
6
|
Yingchoncharoen P, Charoenngam N, Ponvilawan B, Thongpiya J, Chaikijurajai T, Ungprasert P. The Association Between Asthma and Risk of Myasthenia Gravis: A Systematic Review and Meta-analysis. Lung 2021; 199:273-280. [PMID: 33987703 DOI: 10.1007/s00408-021-00444-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 03/25/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE This study aimed to investigate the association between asthma and risk of myasthenia gravis (MG) using the method of systematic review and meta-analysis. METHODS Potentially eligible studies were identified from Medline and EMBASE databases from inception to July 2020 using search strategy that comprised terms for "Asthma" and "Myasthenia Gravis". Eligible cohort study must consist of one cohort of individuals with asthma and another cohort of individuals without asthma. Then, the study must report relative risk (RR) with 95% confidence intervals (95% CIs) of incident MG between the groups. Eligible case-control studies must include cases with MG and controls without MG. Then, the study must explore their history of asthma. Odds ratio (OR) with 95% CIs of the association between asthma status and MG must be reported. Point estimates with standard errors were retrieved from each study and were combined together using the generic inverse variance method. RESULTS A total of 6,835 articles were identified. After two rounds of independent review by five investigators, two cohort studies and three case-control studies met the eligibility criteria and were included into the meta-analysis. Pooled analysis showed that asthma was significantly associated with risk of MG with the pooled risk ratio of 1.38 (95% CI 1.02-1.86). Funnel plot was symmetric, which was not suggestive of publication bias. CONCLUSION The current study found a significant association between asthma and increased risk of MG.
Collapse
Affiliation(s)
- Pitchaporn Yingchoncharoen
- Department of Clinical Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nipith Charoenngam
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand. .,Section Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University School of Medicine, 85 E Newton St., Boston, MA, 02118, USA.
| | - Ben Ponvilawan
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jerapas Thongpiya
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thanat Chaikijurajai
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Patompong Ungprasert
- Department of Rheumatologic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
7
|
Rodríguez Cruz PM, Cossins J, Beeson D, Vincent A. The Neuromuscular Junction in Health and Disease: Molecular Mechanisms Governing Synaptic Formation and Homeostasis. Front Mol Neurosci 2020; 13:610964. [PMID: 33343299 PMCID: PMC7744297 DOI: 10.3389/fnmol.2020.610964] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/30/2020] [Indexed: 12/28/2022] Open
Abstract
The neuromuscular junction (NMJ) is a highly specialized synapse between a motor neuron nerve terminal and its muscle fiber that are responsible for converting electrical impulses generated by the motor neuron into electrical activity in the muscle fibers. On arrival of the motor nerve action potential, calcium enters the presynaptic terminal, which leads to the release of the neurotransmitter acetylcholine (ACh). ACh crosses the synaptic gap and binds to ACh receptors (AChRs) tightly clustered on the surface of the muscle fiber; this leads to the endplate potential which initiates the muscle action potential that results in muscle contraction. This is a simplified version of the events in neuromuscular transmission that take place within milliseconds, and are dependent on a tiny but highly structured NMJ. Much of this review is devoted to describing in more detail the development, maturation, maintenance and regeneration of the NMJ, but first we describe briefly the most important molecules involved and the conditions that affect their numbers and function. Most important clinically worldwide, are myasthenia gravis (MG), the Lambert-Eaton myasthenic syndrome (LEMS) and congenital myasthenic syndromes (CMS), each of which causes specific molecular defects. In addition, we mention the neurotoxins from bacteria, snakes and many other species that interfere with neuromuscular transmission and cause potentially fatal diseases, but have also provided useful probes for investigating neuromuscular transmission. There are also changes in NMJ structure and function in motor neuron disease, spinal muscle atrophy and sarcopenia that are likely to be secondary but might provide treatment targets. The NMJ is one of the best studied and most disease-prone synapses in the nervous system and it is amenable to in vivo and ex vivo investigation and to systemic therapies that can help restore normal function.
Collapse
Affiliation(s)
- Pedro M. Rodríguez Cruz
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Judith Cossins
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Angela Vincent
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Myasthenia gravis (MG) is an autoimmune neuromuscular disease that causes fluctuating weakness in ocular, bulbar, and limb muscles and can, in 15% of cases, cause myasthenic crisis, a neurologic emergency characterized by respiratory failure. Although infrequent, MG needs to be promptly recognized and treated because the potential for improvement and remission is very high. The diagnosis of MG can be challenging and delayed because of the fluctuating nature of muscle weakness and the overlap of signs and symptoms with other neuromuscular diseases.This article reviews the importance of prompt recognition of the typical signs and symptoms, best tests to confirm the diagnosis, currently available acute and chronic treatment modalities, the role of thymectomy, and the natural history of the disease. Special consideration related to the diagnosis and management in women during pregnancy and in children will also be reviewed. This article also includes an overview of congenital myasthenic syndromes. RECENT FINDINGS Recent significant efforts in standardizing and improving the care of patients with MG have occurred, as well as new momentum in developing new drugs for patients with MG who do not adequately respond to currently available treatments. The number of clinical trials and drugs in development for MG is steadily increasing. Eculizumab has been recently approved by the US Food and Drug Administration (FDA) for adult patients with generalized MG who are acetylcholine receptor-antibody positive, based on the REGAIN (Safety and Efficacy of Eculizumab in Refractory Generalized Myasthenia Gravis) study, a phase 3, randomized, double-blind, placebo-controlled, multicenter trial. An international, multicenter, randomized trial comparing thymectomy plus prednisone with prednisone alone has demonstrated that thymectomy improves clinical outcome in patients with nonthymomatous MG. Clinical care guidelines have been published, and the recommendations for clinical research standards and the Myasthenia Gravis Foundation of America MGFA clinical classification published in 2000 have become widely accepted by the clinical and research community of MG experts. SUMMARY MG is a highly treatable disease with many effective treatment modalities available and with a natural history that continues to improve thanks to better diagnostic tests and effective drugs. The diagnosis and management of patients affected by MG can be highly rewarding for any neurologist as most patients are able to live normal lives if treated appropriately. Nevertheless, future research is needed to address unresolved clinical issues, such as when and how to discontinue immunosuppressive medications in patients in remission, the role and timing of thymectomy in children, and better treatment options for refractory patients.
Collapse
|
9
|
Yan X, Gu Y, Wang C, Sun S, Wang X, Tian J, Wang M, Ji X, Duan X, Gao H, Fang Q, Dong W, Zhang X, Xue Q. Unbalanced expression of membrane-bound and soluble inducible costimulator and programmed cell death 1 in patients with myasthenia gravis. Clin Immunol 2019; 207:68-78. [PMID: 31374257 DOI: 10.1016/j.clim.2019.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 02/01/2023]
Abstract
This study aimed to investigate the possible functions and mechanisms of positive and negative costimulatory molecules in the pathological process of myasthenia gravis (MG). The expression levels of membrane-bound inducible costimulator (ICOS) and programmed cell death 1 (PD-1) in peripheral blood T cells, their corresponding ligands ICOSL and PDL-1 on B cells, and their soluble forms (sICOS, sPD-1, sICOSL, and sPDL-1) in plasma were detected in patients with untreated-stage MG (USMG) and remission-stage MG (RSMG). The results showed that the expression levels of membrane-bound ICOS and PD-1 in the peripheral blood T cells of the USMG group and their corresponding ligands ICOSL and PD-L1 on B cells were significantly increased compared to those in the RSMG group and healthy controls (HCs). The levels of sICOSL and sPD-1 were significantly upregulated in USMG patients compared to those in the RSMG and HC groups, while the levels of sICOS and sPD-L1 were not different. The expression of PD-L1 on CD19+ B cells was positively correlated with the concentrations of AchR Ab in the USMG group. The expression of ICOS and PD-1 in CD4+ T cells and the expression of ICOSL and PD-L1 on CD19+ B cells were positively correlated with the quantitative myasthenia gravis (QMG) scores in the USMG group. Also, in the USMG group, the plasma levels of sICOSL and sPD-1 were positively correlated with the QMG scores. In addition, the percentage of peripheral blood follicular helper T (Tfh) cells in the USMG group was positively correlated with ICOS and PD-1 expression on CD4+ T cells and ICOSL and PD-L1 expression on CD19+ B cells. There were positive correlations between sICOSL and sPD-1 levels and the percentage of peripheral blood Tfh cells and plasma interleukin-21 (IL-21) levels in the USMG group. The results suggest that the positive ICOS/ICOSL and negative PD-1/PD-L1 costimulatory molecule pairs participate in the pathological process of MG. Abnormal sICOSL and sPD-1 expression might interfere with the normal signal transduction of ICOS and PD-1 on Tfh cells, causing excessive activation of Tfh cells and promotion of disease progression. sICOSL and sPD-1 have potential value in monitoring MG disease states.
Collapse
Affiliation(s)
- Xiaoming Yan
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yanzheng Gu
- Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, China
| | - Caiqin Wang
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Simao Sun
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaozhu Wang
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jingluan Tian
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Mingyuan Wang
- Suzhou Red Cross Central Blood Station, Suzhou, Jiangsu 215006, China
| | - Xiaopei Ji
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiaoyu Duan
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Hanqing Gao
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Qi Fang
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, China
| | - Wanli Dong
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xueguang Zhang
- Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Qun Xue
- Neurology Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Suzhou Clinical Medical Center of Neurology, Suzhou, Jiangsu 215004, China.
| |
Collapse
|
10
|
A Neurologist's Perspective on Understanding Myasthenia Gravis: Clinical Perspectives of Etiologic Factors, Diagnosis, and Preoperative Treatment. Thorac Surg Clin 2019; 29:133-141. [PMID: 30927994 DOI: 10.1016/j.thorsurg.2018.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Myasthenia gravis (MG) is a disease of neuromuscular transmission caused by antibodies directed toward proteins concentrated at the neuromuscular junction. Mild to life-threatening weakness varies in severity over time and with level of activity. Therefore, clinical diagnosis is often challenging. MG may be categorized by autoantibody type, thymic pathologic condition, and age of onset. Treatments are tailored for each group. A key management concern is severe exacerbation of weakness resulting from infections or exposure to certain medications, including antibiotics, which may be severe enough to produce respiratory decompensation. The article reviews key diagnostic issues and treatment options.
Collapse
|
11
|
miR-1933-3p is upregulated in skeletal muscles of MuSK+ EAMG mice and affects Impa1 and Mrpl27. Neurosci Res 2019; 151:46-52. [PMID: 30763589 DOI: 10.1016/j.neures.2019.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 02/03/2019] [Accepted: 02/06/2019] [Indexed: 12/15/2022]
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.
Collapse
|
12
|
Wu DM, Wen X, Han XR, Wang S, Wang YJ, Shen M, Fan SH, Zhuang J, Zhang ZF, Shan Q, Li MQ, Hu B, Sun CH, Lu J, Zheng YL. Micro-RNA-143 inhibits proliferation and promotes apoptosis of thymocytes by targeting CXCL13 in a myasthenia gravis mouse model. Am J Physiol Cell Physiol 2019; 316:C70-C80. [PMID: 30404560 DOI: 10.1152/ajpcell.00090.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Myasthenia gravis (MG) is an autoimmune neuromuscular disorder, affecting the quality of life of millions of people worldwide. The present study aims to determine the relationship between micro-RNA-143 (miR-143) and C-X-C motif chemokine 13 (CXCL13) and whether it influences the pathogenesis of myasthenia gravis (MG). Thymus specimens were resected from patients with thymic hyperplasia combined with MG and then infused into normal mouse cavities to establish MG mouse models. Immunohistochemistry, reverse transcription-quantitative PCR, in situ hybridization detection, and Western blot analysis were employed to identify the expression of miR-143 and CXCL13 in MG and normal mice. The obtained thymocytes were cultured in vitro and transfected with a series of miR-143 mimic, miR-143 inhibitor, overexpression of CXCL13, or siRNA against CXCL13. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] and flow cytometry assays were employed to assess cell viability, cycle entry, and apoptosis of the thymocytes. Dual-luciferase reporter assay provided verification, confirming that CXCL13 was the target gene of miR-143. Low miR-143 expression in the thymus tissues of the MG mice was detected, which presented with a reciprocal relationship with the expression rate of CLCX13. Observations in relation to the interactions between miR-143 mimic or siRNA-CXCL13 exposure showed reduced cell viability, with a greater number of cells arrested at the G0/G1 phase and a greater rate of induced apoptosis. Furthermore, overexpression of CXCL13 rescued miR-143 mimic-induced apoptosis. The findings have identified the potential role of miR-143 as a MG development mediator by targeting CXCL13. The key results obtained provide a promising experimental basis for targeted intervention treatment with miR-143.
Collapse
Affiliation(s)
- Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Xin Wen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Xin-Rui Han
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Yong-Jian Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Min Shen
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Juan Zhuang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- School of Environment Science and Spatial Informatics, China University of Mining and Technology , Xuzhou , People's Republic of China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, School of Life Sciences, Huaiyin Normal University, Huai'an, People's Republic of China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Qun Shan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Chun-Hui Sun
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University , Xuzhou , People's Republic of China
- College of Health Sciences, Jiangsu Normal University , Xuzhou , People's Republic of China
| |
Collapse
|
13
|
Abstract
Acquired Myasthenia Gravis (MG) is a neuromuscular disease caused by autoantibodies against components of the neuromuscular junction. It is a prototype organ-specific autoimmune disease with well-defined antigenic targets mainly the nicotinic acetylcholine receptor (AChR). Patients suffer from fluctuating, fatigable muscle weakness that worsens with activity and improves with rest. Various therapeutic strategies have been used over the years to alleviate MG symptoms. These strategies aim at improving the transmission of the nerve impulse to muscle or at lowering the immune system with steroids or immunosuppressant drugs. Nevertheless, MG remains a chronic disease and symptoms tend to persist in many patients, some being or becoming refractory over time. In this review, based on recent experimental data on MG or based on results from clinical trials for other autoimmune diseases, we explore new potential therapeutic approaches for MG patients, going from non-specific approaches with the use of stem cells with their anti-inflammatory and immunosuppressive properties to targeted therapies using monoclonal antibodies specific for cell-surface antigens or circulating molecules.
Collapse
Affiliation(s)
- Anthony Behin
- APHP, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile-de-France, Institut de Myologie, GH Pitié-Salpêtrière, Paris, France.,AIM, Institut de Myologie, Paris, France
| | - Rozen Le Panse
- INSERM U974, Paris, France.,UPMC Sorbonne Université, Paris, France.,AIM, Institut de Myologie, Paris, France
| |
Collapse
|
14
|
Wang S, Breskovska I, Gandhy S, Punga AR, Guptill JT, Kaminski HJ. Advances in autoimmune myasthenia gravis management. Expert Rev Neurother 2018; 18:573-588. [PMID: 29932785 PMCID: PMC6289049 DOI: 10.1080/14737175.2018.1491310] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Myasthenia gravis (MG) is an autoimmune neuromuscular disorder with no cure and conventional treatments limited by significant adverse effects and variable benefit. In the last decade, therapeutic development has expanded based on improved understanding of autoimmunity and financial incentives for drug development in rare disease. Clinical subtypes exist based on age, gender, thymic pathology, autoantibody profile, and other poorly defined factors, such as genetics, complicate development of specific therapies. Areas covered: Clinical presentation and pathology vary considerably among patients with some having weakness limited to the ocular muscles and others having profound generalized weakness leading to respiratory insufficiency. MG is an antibody-mediated disorder dependent on autoreactive B cells which require T-cell support. Treatments focus on elimination of circulating autoantibodies or inhibition of effector mechanisms by a broad spectrum of approaches from plasmapheresis to B-cell elimination to complement inhibition. Expert commentary: Standard therapies and those under development are disease modifying and not curative. As a rare disease, clinical trials are challenged in patient recruitment. The great interest in development of treatments specific for MG is welcome, but decisions will need to be made to focus on those that offer significant benefits to patients.
Collapse
Affiliation(s)
- Shuhui Wang
- Department of Neurology, George Washington University, Washington DC 20008
| | - Iva Breskovska
- Department of Neurology, George Washington University, Washington DC 20008
| | - Shreya Gandhy
- Department of Neurology, George Washington University, Washington DC 20008
| | - Anna Rostedt Punga
- Department of Neuroscience, Clinical Neurophysiology, Uppsala University, Uppsala, Sweden
| | - Jeffery T. Guptill
- Department of Neurology, Duke University Medical Center, Durham, North Carolina, USA
| | - Henry J. Kaminski
- Department of Neurology, George Washington University, Washington DC 20008
| |
Collapse
|
15
|
Stöppler D, Macpherson A, Smith-Penzel S, Basse N, Lecomte F, Deboves H, Taylor RD, Norman T, Porter J, Waters LC, Westwood M, Cossins B, Cain K, White J, Griffin R, Prosser C, Kelm S, Sullivan AH, Fox D, Carr MD, Henry A, Taylor R, Meier BH, Oschkinat H, Lawson AD. Insight into small molecule binding to the neonatal Fc receptor by X-ray crystallography and 100 kHz magic-angle-spinning NMR. PLoS Biol 2018; 16:e2006192. [PMID: 29782488 PMCID: PMC5983862 DOI: 10.1371/journal.pbio.2006192] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/01/2018] [Accepted: 05/02/2018] [Indexed: 01/09/2023] Open
Abstract
Aiming at the design of an allosteric modulator of the neonatal Fc receptor (FcRn)-Immunoglobulin G (IgG) interaction, we developed a new methodology including NMR fragment screening, X-ray crystallography, and magic-angle-spinning (MAS) NMR at 100 kHz after sedimentation, exploiting very fast spinning of the nondeuterated soluble 42 kDa receptor construct to obtain resolved proton-detected 2D and 3D NMR spectra. FcRn plays a crucial role in regulation of IgG and serum albumin catabolism. It is a clinically validated drug target for the treatment of autoimmune diseases caused by pathogenic antibodies via the inhibition of its interaction with IgG. We herein present the discovery of a small molecule that binds into a conserved cavity of the heterodimeric, extracellular domain composed of an α-chain and β2-microglobulin (β2m) (FcRnECD, 373 residues). X-ray crystallography was used alongside NMR at 100 kHz MAS with sedimented soluble protein to explore possibilities for refining the compound as an allosteric modulator. Proton-detected MAS NMR experiments on fully protonated [13C,15N]-labeled FcRnECD yielded ligand-induced chemical-shift perturbations (CSPs) for residues in the binding pocket and allosteric changes close to the interface of the two receptor heterodimers present in the asymmetric unit as well as potentially in the albumin interaction site. X-ray structures with and without ligand suggest the need for an optimized ligand to displace the α-chain with respect to β2m, both of which participate in the FcRnECD-IgG interaction site. Our investigation establishes a method to characterize structurally small molecule binding to nondeuterated large proteins by NMR, even in their glycosylated form, which may prove highly valuable for structure-based drug discovery campaigns.
Collapse
Affiliation(s)
- Daniel Stöppler
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | | | | | | | | | | | | | | | - Lorna C. Waters
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom
| | | | | | | | | | | | | | | | - Amy H. Sullivan
- Beryllium Discovery, Bedford, Massachusetts, United States of America
| | - David Fox
- Beryllium Discovery, Bedford, Massachusetts, United States of America
| | - Mark D. Carr
- Leicester Institute of Structural and Chemical Biology, University of Leicester, Leicester, United Kingdom
| | | | | | - Beat H. Meier
- Laboratory of Physical Chemistry, ETH Zürich, Zürich, Switzerland
| | - Hartmut Oschkinat
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
- * E-mail: (HO); (ADL)
| | | |
Collapse
|
16
|
Misra MK, Damotte V, Hollenbach JA. The immunogenetics of neurological disease. Immunology 2018; 153:399-414. [PMID: 29159928 PMCID: PMC5838423 DOI: 10.1111/imm.12869] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 12/18/2022] Open
Abstract
Genes encoding antigen-presenting molecules within the human major histocompatibility complex (MHC) account for the highest component of genetic risk for many neurological diseases, such as multiple sclerosis, neuromyelitis optica, Parkinson's disease, Alzheimer's disease, schizophrenia, myasthenia gravis and amyotrophic lateral sclerosis. Myriad genetic, immunological and environmental factors may contribute to an individual's susceptibility to neurological disease. Here, we review and discuss the decades long research on the influence of genetic variation at the MHC locus and the role of immunogenetic killer cell immunoglobulin-like receptor (KIR) loci in neurological diseases, including multiple sclerosis, neuromyelitis optica, Parkinson's disease, Alzheimer's disease, schizophrenia, myasthenia gravis and amyotrophic lateral sclerosis. The findings of immunogenetic association studies are consistent with a polygenic model of inheritance in the heterogeneous and multifactorial nature of complex traits in various neurological diseases. Future investigation is highly recommended to evaluate both coding and non-coding variation in immunogenetic loci using high-throughput high-resolution next-generation sequencing technologies in diverse ethnic groups to fully appreciate their role in neurological diseases.
Collapse
Affiliation(s)
- Maneesh K. Misra
- Department of NeurologySan Francisco School of MedicineUniversity of CaliforniaSan FranciscoCAUSA
| | - Vincent Damotte
- Department of NeurologySan Francisco School of MedicineUniversity of CaliforniaSan FranciscoCAUSA
| | - Jill A. Hollenbach
- Department of NeurologySan Francisco School of MedicineUniversity of CaliforniaSan FranciscoCAUSA
| |
Collapse
|
17
|
Muscle satellite cells are functionally impaired in myasthenia gravis: consequences on muscle regeneration. Acta Neuropathol 2017; 134:869-888. [PMID: 28756524 DOI: 10.1007/s00401-017-1754-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 06/30/2017] [Accepted: 07/21/2017] [Indexed: 12/14/2022]
Abstract
Myasthenia gravis (MG) is a neuromuscular disease caused in most cases by anti-acetyl-choline receptor (AChR) autoantibodies that impair neuromuscular signal transmission and affect skeletal muscle homeostasis. Myogenesis is carried out by muscle stem cells called satellite cells (SCs). However, myogenesis in MG had never been explored. The aim of this study was to characterise the functional properties of myasthenic SCs as well as their abilities in muscle regeneration. SCs were isolated from muscle biopsies of MG patients and age-matched controls. We first showed that the number of Pax7+ SCs was increased in muscle sections from MG and its experimental autoimmune myasthenia gravis (EAMG) mouse model. Myoblasts isolated from MG muscles proliferate and differentiate more actively than myoblasts from control muscles. MyoD and MyoG were expressed at a higher level in MG myoblasts as well as in MG muscle biopsies compared to controls. We found that treatment of control myoblasts with MG sera or monoclonal anti-AChR antibodies increased the differentiation and MyoG mRNA expression compared to control sera. To investigate the functional ability of SCs from MG muscle to regenerate, we induced muscle regeneration using acute cardiotoxin injury in the EAMG mouse model. We observed a delay in maturation evidenced by a decrease in fibre size and MyoG mRNA expression as well as an increase in fibre number and embryonic myosin heavy-chain mRNA expression. These findings demonstrate for the first time the altered function of SCs from MG compared to control muscles. These alterations could be due to the anti-AChR antibodies via the modulation of myogenic markers resulting in muscle regeneration impairment. In conclusion, the autoimmune attack in MG appears to have unsuspected pathogenic effects on SCs and muscle regeneration, with potential consequences on myogenic signalling pathways, and subsequently on clinical outcome, especially in the case of muscle stress.
Collapse
|
18
|
Luo M, Liu X, Meng H, Xu L, Li Y, Li Z, Liu C, Luo YB, Hu B, Xue Y, Liu Y, Luo Z, Yang H. IFNA-AS1 regulates CD4 + T cell activation in myasthenia gravis though HLA-DRB1. Clin Immunol 2017; 183:121-131. [PMID: 28822831 DOI: 10.1016/j.clim.2017.08.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/04/2017] [Accepted: 08/10/2017] [Indexed: 12/17/2022]
Abstract
Abnormal CD4+T cell activation is known to play roles in the pathogenesis of myasthenia gravis (MG). However, little is known about the mechanisms underlying the roles of lncRNAs in regulating CD4+ T cell. In this study, we discovered that the lncRNA IFNG-AS1 is abnormally expressed in MG patients associated with quantitative myasthenia gravis (QMG) and the positive anti-AchR Ab levels patients. IFNG-AS1 influenced Th1/Treg cell proliferation and regulated the expression levels of their transcription factors in an experimental autoimmune myasthenia gravis (EAMG)model. IFNG-AS1 could reduce the expression of HLA-DRB and HLA-DOB and they had a negative correlation in MG. Furthermore IFNG-AS1 influenced the expression levels of CD40L and CD4+ T cells activation in MG patient partly depend on effecting the HLA-DRB1 expression. It suggests that IFNG-AS1 may be involved in CD4+T cell-mediated immune responses in MG.
Collapse
Affiliation(s)
- Mengchuan Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xiaofang Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Huanyu Meng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Liqun Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yi Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhibin Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Chang Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yue-Bei Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Bo Hu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yuanyuan Xue
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yu Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhaohui Luo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Neurology Institute of Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
19
|
Aguilo-Seara G, Xie Y, Sheehan J, Kusner LL, Kaminski HJ. Ablation of IL-17 expression moderates experimental autoimmune myasthenia gravis disease severity. Cytokine 2017; 96:279-285. [PMID: 28599246 DOI: 10.1016/j.cyto.2017.05.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/17/2017] [Accepted: 05/08/2017] [Indexed: 01/25/2023]
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.
Collapse
Affiliation(s)
| | - Yanchen Xie
- Departments of Neurology, George Washington University, Washington, DC, USA
| | - Jarrod Sheehan
- Pharmacology & Physiology, George Washington University, Washington, DC, USA
| | - Linda L Kusner
- Departments of Neurology, George Washington University, Washington, DC, USA; Pharmacology & Physiology, George Washington University, Washington, DC, USA
| | - Henry J Kaminski
- Departments of Neurology, George Washington University, Washington, DC, USA; Pharmacology & Physiology, George Washington University, Washington, DC, USA.
| |
Collapse
|
20
|
Lopomo A, Berrih-Aknin S. Autoimmune Thyroiditis and Myasthenia Gravis. Front Endocrinol (Lausanne) 2017; 8:169. [PMID: 28751878 PMCID: PMC5508005 DOI: 10.3389/fendo.2017.00169] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/30/2017] [Indexed: 12/18/2022] Open
Abstract
Autoimmune diseases (AIDs) are the result of specific immune responses directed against structures of the self. In normal conditions, the molecules recognized as "self" are tolerated by immune system, but when the self-tolerance is lost, the immune system could react against molecules from the body, causing the loss of self-tolerance, and subsequently the onset of AID that differs for organ target and etiology. Autoimmune thyroid disease (ATD) is caused by the development of autoimmunity against thyroid antigens and comprises Hashimoto's thyroiditis and Graves disease. They are frequently associated with other organ or non-organ specific AIDs, such as myasthenia gravis (MG). In fact, ATD seems to be the most associated pathology to MG. The etiology of both diseases is multifactorial and it is due to genetic and environmental factors, and each of them has specific characteristics. The two pathologies show many commonalities, such as the organ-specificity with a clear pathogenic effect of antibodies, the pathological mechanisms, such as deregulation of the immune system and the implication of the genetic predisposition. They also show some differences, such as the mode of action of the antibodies and therapies. In this review that focuses on ATD and MG, the common features and the differences between the two diseases are discussed.
Collapse
Affiliation(s)
- Angela Lopomo
- Department of Translational Research and New Technologies in Medicine and Surgery, Division of Medical Genetics, University of Pisa, Pisa, Italy
| | - Sonia Berrih-Aknin
- Sorbonne Universités, UPMC Univ Paris 06, Paris, France
- INSERM U974, Paris, France
- AIM, Institute of Myology, Paris, France
- *Correspondence: Sonia Berrih-Aknin,
| |
Collapse
|
21
|
Zhang F, Liu G, Bu Y, Ma X, Hao J. Expression profile of long noncoding RNAs and mRNAs in peripheral blood mononuclear cells from myasthenia gravis patients. J Neuroimmunol 2016; 299:124-129. [PMID: 27725110 DOI: 10.1016/j.jneuroim.2016.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/03/2016] [Accepted: 09/08/2016] [Indexed: 11/28/2022]
Abstract
For the epigenetic characterization of myasthenia gravis (MG), we determined whether long noncoding RNAs (lncRNAs) and messenger RNAs (mRNAs) are expressed differentially in subjects with and without MG. Compared with healthy control subjects, the MG patients had 1561 upregulated lncRNAs, 1034 downregulated lncRNAs, 921 upregulated mRNAs, and 806 downregulated mRNAs (fold change>2.0). Several GO terms including nucleic acid transcription factor activity, inflammatory response, regulation of leukocyte activation, lymphocyte proliferation and regulation of B cell proliferation were enriched in gene lists, suggesting a potential correlation with MG. Pathway analysis then demonstrated that cytokine-cytokine receptor interaction, intestinal immune network for lgA production, NOD-like receptor signaling pathway, NF-kappaB signaling pathway, cell adhesion molecules and TNF signaling pathway play important roles in MG. Co-expression network analysis indicated that 33 lncRNAs were predicted to have 31 cis-regulated target genes, and 65 lncRNAs appeared to regulate the patients' 45 trans target genes among differentially expressed lncRNAs. Our present study identified a subset of dysregulated lncRNAs and mRNAs in patients with MG, which may impact this disease process.
Collapse
Affiliation(s)
- Fang Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Guiyou Liu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Yali Bu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xiaofeng Ma
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Junwei Hao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
22
|
Steiner I, Goldstein L, Hellmann MA, Lotan I. Prior damage to lower motor neuron triggering myasthenia gravis. Muscle Nerve 2016; 54:167-9. [PMID: 26789735 DOI: 10.1002/mus.25026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/29/2015] [Accepted: 12/30/2015] [Indexed: 11/05/2022]
Affiliation(s)
- Israel Steiner
- Department of Neurology, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lilach Goldstein
- Department of Neurology, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mark A Hellmann
- Department of Neurology, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Itay Lotan
- Department of Neurology, Rabin Medical Center, Beilinson Campus, 49100, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
23
|
Increased expression of Toll-like receptors 7 and 9 in myasthenia gravis thymus characterized by active Epstein-Barr virus infection. Immunobiology 2015; 221:516-27. [PMID: 26723518 DOI: 10.1016/j.imbio.2015.12.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/10/2015] [Accepted: 12/10/2015] [Indexed: 11/24/2022]
Abstract
Considerable data implicate the thymus as the main site of autosensitization to the acetylcholine receptor in myasthenia gravis (MG), a B-cell-mediated autoimmune disease affecting the neuromuscular junction. We recently demonstrated an active Epstein-Barr virus (EBV) infection in the thymus of MG patients, suggesting that EBV might contribute to the onset or maintenance of the autoimmune response within MG thymus, because of its ability to activate and immortalize autoreactive B cells. EBV has been reported to elicit and modulate Toll-like receptor (TLR) 7- and TLR9-mediated innate immune responses, which are known to favor B-cell dysfunction and autoimmunity. Aim of this study was to investigate whether EBV infection is associated with altered expression of TLR7 and TLR9 in MG thymus. By real-time PCR, we found that TLR7 and TLR9 mRNA levels were significantly higher in EBV-positive MG compared to EBV-negative normal thymuses. By confocal microscopy, high expression levels of TLR7 and TLR9 proteins were observed in B cells and plasma cells of MG thymic germinal centers (GCs) and lymphoid infiltrates, where the two receptors co-localized with EBV antigens. An increased frequency of Ki67-positive proliferating B cells was found in MG thymuses, where we also detected proliferating cells expressing TLR7, TLR9 and EBV antigens, thus supporting the idea that EBV-associated TLR7/9 signaling may promote abnormal B-cell activation and proliferation. Along with B cells and plasma cells, thymic epithelium, plasmacytoid dendritic cells and macrophages exhibited enhanced TLR7 and TLR9 expression in MG thymus; TLR7 was also increased in thymic myeloid dendritic cells and its transcriptional levels positively correlated with those of interferon (IFN)-β. We suggested that TLR7/9 signaling may be involved in antiviral type I IFN production and long-term inflammation in EBV-infected MG thymuses. Our overall findings indicate that EBV-driven TLR7- and TLR9-mediated innate immune responses may participate in the intra-thymic pathogenesis of MG.
Collapse
|
24
|
Myasthenia gravis: subgroup classification and therapeutic strategies. Lancet Neurol 2015; 14:1023-36. [DOI: 10.1016/s1474-4422(15)00145-3] [Citation(s) in RCA: 563] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 06/02/2015] [Accepted: 06/19/2015] [Indexed: 12/13/2022]
|
25
|
Alahgholi-Hajibehzad M, Kasapoglu P, Jafari R, Rezaei N. The role of T regulatory cells in immunopathogenesis of myasthenia gravis: implications for therapeutics. Expert Rev Clin Immunol 2015; 11:859-70. [DOI: 10.1586/1744666x.2015.1047345] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
26
|
Systems biology of myasthenia gravis, integration of aberrant lncRNA and mRNA expression changes. BMC Med Genomics 2015; 8:13. [PMID: 25889429 PMCID: PMC4380247 DOI: 10.1186/s12920-015-0087-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 02/26/2015] [Indexed: 12/20/2022] Open
Abstract
Background A novel class of transcripts, long non-coding RNAs (lncRNAs), has recently emerged as a key player in several biological processes, and important roles for these molecules have been reported in a number of complex human diseases, such as autoimmune diseases, neurological disorders, and various cancers. However, the aberrant lncRNAs implicated in myasthenia gravis (MG) remain unknown. The aim of the present study was to explore the abnormal expression of lncRNAs in peripheral blood mononuclear cells (PBMCs) and examine mRNA regulatory relationship networks among MG patients with or without thymoma. Methods Microarray assays were performed, and the outstanding differences between lncRNAs or mRNA expression were verified through RT-PCR. The lncRNAs functions were annotated for the target genes using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) biological pathway. The potential regulatory relationships between the lncRNAs and target genes were analyzed using the ‘cis’ and ‘trans’ model. Outstanding lncRNAs were organized to generate a TF-lncRNA-gene network using Cytoscape software. Results The lncRNA and mRNA expression profile analysis revealed subsets of differentially expressed genes in MG patients with or without thymoma. A total of 12 outstanding dysregulated expression lncRNAs, such as lncRNA oebiotech_11933, were verified through real-time PCR. Several GO terms including the cellular response to interferon-γ, platelet degranulation, chemokine receptor binding and cytokine interactions were very important in MG pathogenesis. The chromosome locations of some lncRNAs and associated co-expression genes were demonstrated using ‘cis’ analysis. The results of the ‘trans’ analysis revealed that some TFs (i.e., CTCF, TAF1and MYC) regulate lncRNA and gene expression. The outstanding lncRNAs in each group were implicated in the regulation of the TF-lncRNA-target gene network. Conclusion The results of the present study provide a perspective on lncRNA expression in MG. We identify a subset of aberrant lncRNAs and mRNAs as potential biomarkers for the diagnosis of MG. The GO and KEGG pathway analysis provides an annotation to determine the functions of these lncRNAs. The results of the ‘cis’ and ‘trans’ analyses provide information concerning the modular regulation of lncRNAs. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0087-z) contains supplementary material, which is available to authorized users.
Collapse
|
27
|
Detecting key genes regulated by miRNAs in dysfunctional crosstalk pathway of myasthenia gravis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:724715. [PMID: 25705681 PMCID: PMC4331476 DOI: 10.1155/2015/724715] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/10/2014] [Indexed: 02/07/2023]
Abstract
Myasthenia gravis (MG) is a neuromuscular autoimmune disorder resulting from autoantibodies attacking components of the neuromuscular junction. Recent studies have implicated the aberrant expression of microRNAs (miRNAs) in the pathogenesis of MG; however, the underlying mechanisms remain largely unknown. This study aimed to identify key genes regulated by miRNAs in MG. Six dysregulated pathways were identified through differentially expressed miRNAs and mRNAs in MG, and significant crosstalk was detected between five of these. Notably, crosstalk between the "synaptic long-term potentiation" pathway and four others was mediated by five genes involved in the MAPK signaling pathway. Furthermore, 14 key genes regulated by miRNAs were detected, of which six-MAPK1, RAF1, PGF, PDGFRA, EP300, and PPP1CC-mediated interactions between the dysregulated pathways. MAPK1 and RAF1 were responsible for most of this crosstalk (80%), likely reflecting their central roles in MG pathogenesis. In addition, most key genes were enriched in immune-related local areas that were strongly disordered in MG. These results provide new insight into the pathogenesis of MG and offer new potential targets for therapeutic intervention.
Collapse
|
28
|
Fan X, Lin C, Han J, Jiang X, Zhu J, Jin T. Follicular Helper CD4 + T Cells in Human Neuroautoimmune Diseases and Their Animal Models. Mediators Inflamm 2015; 2015:638968. [PMID: 26300592 PMCID: PMC4537760 DOI: 10.1155/2015/638968] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 02/16/2015] [Indexed: 01/27/2023] Open
Abstract
Follicular helper CD4+ T (TFH) cells play a fundamental role in humoral immunity deriving from their ability to provide help for germinal center (GC) formation, B cell differentiation into plasma cells and memory cells, and antibody production in secondary lymphoid tissues. TFH cells can be identified by a combination of markers, including the chemokine receptor CXCR5, costimulatory molecules ICOS and PD-1, transcription repressor Bcl-6, and cytokine IL-21. It is difficult and impossible to get access to secondary lymphoid tissues in humans, so studies are usually performed with human peripheral blood samples as circulating counterparts of tissue TFH cells. A balance of TFH cell generation and function is critical for protective antibody response, whereas overactivation of TFH cells or overexpression of TFH-associated molecules may result in autoimmune diseases. Emerging data have shown that TFH cells and TFH-associated molecules may be involved in the pathogenesis of neuroautoimmune diseases including multiple sclerosis (MS), neuromyelitis optica (NMO)/neuromyelitis optica spectrum disorders (NMOSD), and myasthenia gravis (MG). This review summarizes the features of TFH cells, including their development, function, and roles as well as TFH-associated molecules in neuroautoimmune diseases and their animal models.
Collapse
Affiliation(s)
- Xueli Fan
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Chenhong Lin
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Jinming Han
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Xinmei Jiang
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| | - Jie Zhu
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institute, 14186 Stockholm, Sweden
| | - Tao Jin
- Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun 130021, China
| |
Collapse
|
29
|
Cortactin: A new target in autoimmune myositis and Myasthenia Gravis. Autoimmun Rev 2014; 13:1001-2. [DOI: 10.1016/j.autrev.2014.08.037] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 07/05/2014] [Indexed: 01/21/2023]
|
30
|
Sheng JR, Quan S, Soliven B. CD1d(hi)CD5+ B cells expanded by GM-CSF in vivo suppress experimental autoimmune myasthenia gravis. THE JOURNAL OF IMMUNOLOGY 2014; 193:2669-77. [PMID: 25135828 DOI: 10.4049/jimmunol.1303397] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
IL-10-competent subset within CD1d(hi)CD5(+) B cells, also known as B10 cells, has been shown to regulate autoimmune diseases. Whether B10 cells can prevent or suppress the development of experimental autoimmune myasthenia gravis (EAMG) has not been studied. In this study, we investigated whether low-dose GM-CSF, which suppresses EAMG, can expand B10 cells in vivo, and whether adoptive transfer of CD1d(hi)CD5(+) B cells would prevent or suppress EAMG. We found that treatment of EAMG mice with low-dose GM-CSF increased the proportion of CD1d(hi)CD5(+) B cells and B10 cells. In vitro coculture studies revealed that CD1d(hi)CD5(+) B cells altered T cell cytokine profile but did not directly inhibit T cell proliferation. In contrast, CD1d(hi)CD5(+) B cells inhibited B cell proliferation and its autoantibody production in an IL-10-dependent manner. Adoptive transfer of CD1d(hi)CD5(+) B cells to mice could prevent disease, as well as suppress EAMG after disease onset. This was associated with downregulation of mature dendritic cell markers and expansion of regulatory T cells resulting in the suppression of acetylcholine receptor-specific T cell and B cell responses. Thus, our data have provided significant insight into the mechanisms underlying the tolerogenic effects of B10 cells in EAMG. These observations suggest that in vivo or in vitro expansion of CD1d(hi)CD5(+) B cells or B10 cells may represent an effective strategy in the treatment of human myasthenia gravis.
Collapse
Affiliation(s)
- Jian Rong Sheng
- Department of Neurology, University of Chicago, Chicago, IL 60637
| | - Songhua Quan
- Department of Neurology, University of Chicago, Chicago, IL 60637
| | - Betty Soliven
- Department of Neurology, University of Chicago, Chicago, IL 60637
| |
Collapse
|
31
|
Genetic basis of myasthenia gravis – A comprehensive review. J Autoimmun 2014; 52:146-53. [DOI: 10.1016/j.jaut.2013.12.001] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 12/02/2013] [Indexed: 11/24/2022]
|
32
|
Berrih-Aknin S. Myasthenia Gravis: paradox versus paradigm in autoimmunity. J Autoimmun 2014; 52:1-28. [PMID: 24934596 DOI: 10.1016/j.jaut.2014.05.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
Myasthenia Gravis (MG) is a paradigm of organ-specific autoimmune disease (AID). It is mediated by antibodies that target the neuromuscular junction. The purpose of this review is to place MG in the general context of autoimmunity, to summarize the common mechanisms between MG and other AIDs, and to describe the specific mechanisms of MG. We have chosen the most common organ-specific AIDs to compare with MG: type 1 diabetes mellitus (T1DM), autoimmune thyroid diseases (AITD), multiple sclerosis (MS), some systemic AIDs (systemic lupus erythematous (SLE), rheumatoid arthritis (RA), Sjogren's syndrome (SS)), as well as inflammatory diseases of the gut and liver (celiac disease (CeD), Crohn's disease (CD), and primary biliary cirrhosis (PBC)). Several features are similar between all AIDs, suggesting that common pathogenic mechanisms lead to their development. In this review, we address the predisposing factors (genetic, epigenetic, hormones, vitamin D, microbiota), the triggering components (infections, drugs) and their interactions with the immune system [1,2]. The dysregulation of the immune system is detailed and includes the role of B cells, Treg cells, Th17 and cytokines. We particularly focused on the role of TNF-α and interferon type I whose role in MG is very analogous to that in several other AIDS. The implication of AIRE, a key factor in central tolerance is also discussed. Finally, if MG is a prototype of AIDS, it has a clear specificity compared to the other AIDS, by the fact that the target organ, the muscle, is not the site of immune infiltration and B cell expansion, but exclusively that of antibody-mediated pathogenic mechanisms. By contrast, the thymus in the early onset subtype frequently undergoes tissue remodeling, resulting in the development of ectopic germinal centers surrounded by high endothelial venules (HEV), as observed in the target organs of many other AIDs.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- Sorbonne Universités, UPMC Univ Paris 06, Myology Research Center UM76, F-75013 Paris, France; INSERM U974, F-75013 Paris, France; CNRS FRE 3617, F-75013 Paris, France; Institute of Myology, F-75013 Paris, France.
| |
Collapse
|
33
|
Berrih-Aknin S, Frenkian-Cuvelier M, Eymard B. Diagnostic and clinical classification of autoimmune myasthenia gravis. J Autoimmun 2014; 48-49:143-8. [PMID: 24530233 DOI: 10.1016/j.jaut.2014.01.003] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/13/2013] [Indexed: 01/17/2023]
Abstract
Myasthenia gravis is characterized by muscle weakness and abnormal fatigability. It is an autoimmune disease caused by the presence of antibodies against components of the muscle membrane localized at the neuromuscular junction. In most cases, the autoantibodies are against the acetylcholine receptor (AChR). Recently, other targets have been described such as the MuSK protein (muscle-specific kinase) or the LRP4 (lipoprotein related protein 4). Myasthenia gravis can be classified according to the profile of the autoantibodies, the location of the affected muscles (ocular versus generalized), the age of onset of symptoms and thymic abnormalities. The disease generally begins with ocular symptoms (ptosis and/or diplopia) and extends to other muscles in 80% of cases. Other features that characterize MG include the following: variability, effort induced worsening, successive periods of exacerbation during the course of the disease, severity dependent on respiratory and swallowing impairment (if rapid worsening occurs, a myasthenic crisis is suspected), and an association with thymoma in 20% of patients and with other autoimmune diseases such as hyperthyroidism and Hashimoto's disease. The diagnosis is based on the clinical features, the benefit of the cholinesterase inhibitors, the detection of specific autoantibodies (anti-AChR, anti-MuSK or anti-LRP4), and significant decrement evidenced by electrophysiological tests. In this review, we briefly describe the history and epidemiology of the disease and the diagnostic and clinical classification. The neonatal form of myasthenia is explained, and finally we discuss the main difficulties of diagnosis.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- INSERM U974, Paris, France; CNRS FRE3617, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UM76, Paris, France; AIM, Institut de myologie, Paris, France.
| | - Mélinée Frenkian-Cuvelier
- INSERM U974, Paris, France; CNRS FRE3617, Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UM76, Paris, France; AIM, Institut de myologie, Paris, France
| | - Bruno Eymard
- Centre de référence de pathologie neuromusculaire Paris Est, Service de Neurologie 2, Institut de Myologie, Hôpital de la Pitié Salpêtrière, France
| |
Collapse
|
34
|
Autoimmunity: from black water fever to regulatory function. J Autoimmun 2014; 48-49:1-9. [PMID: 24491820 DOI: 10.1016/j.jaut.2013.12.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 12/31/2013] [Indexed: 12/15/2022]
Abstract
Autoimmunity is a field that has only been around for a little over a century. Initially, it was thought that autoimmunity could not happen, that the body would never turn on itself (i.e. "horror autotoxicus"). It was only around the First World War that autoimmunity was recognized as the pathogenesis of various diseases, including rheumatoid arthritis. The discovery of Compound E led to successful treatment of patients with autoimmune diseases, but it was not till later that the adverse effects of this class of drugs were elucidated. The "modern" age of autoimmunity began around 1945 with the description of blackwater fever, and most of the subsequent research on hemolytic anemia and the role of an autoantibody in its pathogenesis led to a description of the anti-globulin reaction. The lupus erythematous (LE) cell was recognized in the mid-1940s by Hargreaves. His research carried on into the 1960s. Rheumatoid factor was also first described in the 1940s as yet another serum factor with activity against globulin-coated sheep red blood cells. The concept of autoimmunity really gained a foothold in the 1950s, when autoimmune thyroid disease and idiopathic thrombocytopenia were first described. Much has happened since then, and our understanding of autoimmunity has evolved now to include mechanisms of apoptosis, signaling pathway derangements, and the discovery of subsets of T cells with regulatory activity. The modern day study of autoimmunity is a fascinating area of research, and full understanding of the pathogenesis of autoimmune diseases is far from being completely elucidated.
Collapse
|
35
|
Cordiglieri C, Marolda R, Franzi S, Cappelletti C, Giardina C, Motta T, Baggi F, Bernasconi P, Mantegazza R, Cavalcante P. Innate immunity in myasthenia gravis thymus: pathogenic effects of Toll-like receptor 4 signaling on autoimmunity. J Autoimmun 2014; 52:74-89. [PMID: 24397961 DOI: 10.1016/j.jaut.2013.12.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 12/15/2013] [Indexed: 12/21/2022]
Abstract
The thymus is the main site of immune sensitization to AChR in myasthenia gravis (MG). In our previous studies we demonstrated that Toll-like receptor (TLR) 4 is over-expressed in MG thymuses, suggesting its involvement in altering the thymic microenvironment and favoring autosensitization and autoimmunity maintenance processes, via an effect on local chemokine/cytokine network. Here, we investigated whether TLR4 signaling may favor abnormal cell recruitment in MG thymus via CCL17 and CCL22, two chemokines known to dictate immune cell trafficking in inflamed organs by binding CCR4. We also investigated whether TLR4 activation may contribute to immunodysregulation, via the production of Th17-related cytokines, known to alter effector T cell (Teff)/regulatory T cell (Treg) balance. We found that CCL17, CCL22 and CCR4 were expressed at higher levels in MG compared to normal thymuses. The two chemokines were mainly detected around medullary Hassall's corpuscles (HCs), co-localizing with TLR4(+) thymic epithelial cells (TECs) and CCR4(+) dendritic cells (DCs), that were present in higher number in MG thymuses compared to controls. TLR4 stimulation in MG TECs increased CCL17 and CCL22 expression and induced the production of Th17-related cytokines. Then, to study the effect of TLR4-stimulated TECs on immune cell interactions and Teff activation, we generated an in-vitro imaging model by co-culturing CD4(+) Th1/Th17 AChR-specific T cells, naïve CD4(+)CD25(+) Tregs, DCs and TECs from Lewis rats. We observed that TLR4 stimulation led to a more pronounced Teff activatory status, suggesting that TLR4 signaling in MG thymic milieu may affect cell-to-cell interactions, favoring autoreactive T-cell activation. Altogether our findings suggest a role for TLR4 signaling in driving DC recruitment in MG thymus via CCL17 and CCL22, and in generating an inflammatory response that might compromise Treg function, favoring autoreactive T-cell pathogenic responses.
Collapse
Affiliation(s)
- Chiara Cordiglieri
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Roberta Marolda
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Sara Franzi
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Cristina Cappelletti
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Carmelo Giardina
- Department of Pathological Anatomy, Azienda Ospedaliera Bolognini Seriate, Via Paterno 21, 24068 Seriate Bergamo, Italy.
| | - Teresio Motta
- Department of Pathological Anatomy, Azienda Ospedaliera Bolognini Seriate, Via Paterno 21, 24068 Seriate Bergamo, Italy.
| | - Fulvio Baggi
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Pia Bernasconi
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Renato Mantegazza
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| | - Paola Cavalcante
- Neurology IV Unit, Neurological Institute 'Carlo Besta', Via Celoria 11, 20133 Milan, Italy.
| |
Collapse
|
36
|
Berrih-Aknin S, Le Panse R. Myasthenia gravis: a comprehensive review of immune dysregulation and etiological mechanisms. J Autoimmun 2014; 52:90-100. [PMID: 24389034 DOI: 10.1016/j.jaut.2013.12.011] [Citation(s) in RCA: 238] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 12/12/2013] [Indexed: 12/31/2022]
Abstract
Autoimmune myasthenia gravis (MG) is characterized by muscle weakness caused by antibodies directed against proteins of the neuromuscular junction. The main antigenic target is the acetylcholine receptor (AChR), but the muscle Specific Kinase (MuSK) and the low-density lipoprotein receptor-related protein (LRP4) are also targets. This review summarizes the clinical and biological data available for different subgroups of patients, who are classified according to antigenic target, age of onset, and observed thymic abnormalities, such as follicular hyperplasia or thymoma. Here, we analyze in detail the role of the thymus in the physiopathology of MG and propose an explanation for the development of the thymic follicular hyperplasia that is commonly observed in young female patients with anti-AChR antibodies. The influence of the pro-inflammatory environment is discussed, particularly the role of TNF-α and Th17-related cytokines, which could explain the escape of thymic T cells from regulation and the chronic inflammation in the MG thymus. Together with this immune dysregulation, active angiogenic processes and the upregulation of chemokines could promote thymic follicular hyperplasia. MG is a multifactorial disease, and we review the etiological mechanisms that could lead to its onset. Recent global genetic analyses have highlighted potential susceptibility genes. In addition, miRNAs, which play a crucial role in immune function, have been implicated in MG by recent studies. We also discuss the role of sex hormones and the influence of environmental factors, such as the viral hypothesis. This hypothesis is supported by reports that type I interferon and molecules mimicking viral infection can induce thymic changes similar to those observed in MG patients with anti-AChR antibodies.
Collapse
Affiliation(s)
- Sonia Berrih-Aknin
- INSERM U974, Paris, France; CNRS UMR 7215, Paris, France; UPMC Univ Paris 6, Paris, France; AIM, Institute of myology, Paris, France.
| | - Rozen Le Panse
- INSERM U974, Paris, France; CNRS UMR 7215, Paris, France; UPMC Univ Paris 6, Paris, France; AIM, Institute of myology, Paris, France.
| |
Collapse
|