1
|
Sato D, Nishiguchi S, Yamamoto D, Ishioka K, Kakutani T, Watai K, Taniguchi M. A Contemporaneous Onset of Eosinophilic Granulomatosis with Polyangiitis and Myasthenia Gravis. Intern Med 2025; 64:786-789. [PMID: 38987194 PMCID: PMC11949664 DOI: 10.2169/internalmedicine.2872-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 05/19/2024] [Indexed: 07/12/2024] Open
Abstract
Autoimmune neuromuscular disorders in patients with eosinophilic granulomatosis with polyangiitis (EGPA) are relatively uncommon. Although two cases of myasthenia gravis (MG) comorbid with EGPA have been reported, both patients developed EGPA several years after starting immunosuppressive treatment for MG. We herein report a 75-year-old man with a rare co-occurrence of EGPA and MG that developed simultaneously and was successfully treated with immunosuppressive therapy. Distinguishing the neurological symptoms of EGPA from complications of other neurological autoimmune diseases, such as MG, is crucial, especially in patients with eosinophilia.
Collapse
Affiliation(s)
- Daisuke Sato
- Division of General Internal Medicine, Shonan Kamakura General Hospital, Japan
- Division of Cardiology, Shonan Kamakura General Hospital, Japan
| | - Sho Nishiguchi
- Division of General Internal Medicine, Shonan Kamakura General Hospital, Japan
| | | | - Kunihiro Ishioka
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Japan
| | - Takuya Kakutani
- Division of Rheumatology, Shonan Kamakura General Hospital, Japan
| | - Kentaro Watai
- Center for Immunology and Allergy, Shonan Kamakura General Hospital, Japan
| | - Masami Taniguchi
- Center for Immunology and Allergy, Shonan Kamakura General Hospital, Japan
| |
Collapse
|
2
|
Mullen KD, Mozzochi KA, Hawkshaw MJ, Sataloff RT. Laryngeal Myasthenia Gravis in Voice Patients: Clinical, Serologic, and Neuromuscular Characterization of Seronegative Patients for Antibodies Against the Acetylcholine Receptor and Muscle-Specific Kinase. J Voice 2024:S0892-1997(24)00331-X. [PMID: 39419706 DOI: 10.1016/j.jvoice.2024.09.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024]
Abstract
OBJECTIVE Laryngeal myasthenia gravis (MG) is a focal manifestation of MG, and most patients are seronegative for antibodies against the acetylcholine receptor (AChR) and muscle-specific kinase (MuSK). The purpose of this study was to determine the incidence of anti-AChR and anti-Musk antibodies in voice patients and to characterize the clinical and neuromuscular profiles of these patients in order to guide the diagnosis of laryngeal MG. STUDY DESIGN This was a retrospective case-control study that included patients over the age of 18 who underwent laryngeal electromyography (LEMG) as part of their evaluation for neuromuscular junction dysfunction. METHODS Cases and controls were evaluated serologically, for the anti-AChR, anti-MuSK, and anti-striational muscle antibodies, and neuromuscularly using the Tensilon test in some patients, repetitive nerve stimulation (RNS) test, and a treatment trial of pyridostigmine bromide. Cases were defined as either (1) positive anti-AChR or anti-MuSK, or (2) a positive Tensilon test or positive pyridostigmine bromide trial. RESULTS Two hundred and eleven patients were screened; 61 (29%) patients were identified with laryngeal MG, and 77 (36%) patients were selected as controls. All case and control patients were seronegative for the anti-AChR and anti-MuSK antibodies with no significant difference between case and control status for seropositivity for anti-striational muscle antibodies. Of the case patients with an electrically positive Tensilon test (80.6%) who completed a treatment trial, 100% had symptom improvement. Of the case patients with a symptomatically positive Tensilon test (16.1%), only 60% of patients had improvement with a treatment trial. The RNS was more likely to be positive in case patients than control patients, and cases had a higher severity of paresis in all laryngeal muscles with LEMG evaluation. CONCLUSIONS Laryngeal MG is an underrecognized condition in the otolaryngology community owing in part to its seronegative presentation. Electrical improvement with a Tensilon test, or electrical or substantial symptomatic improvement with pyridostigmine bromide represents the most robust diagnostic criteria in these patients.
Collapse
Affiliation(s)
- Katherine D Mullen
- Drexel University College of Medicine Department of Otolaryngology Head and Neck Surgery, Philadelphia, Pennsylvania; Geisinger Commonwealth School of Medicine, Scranton, Pennsylvania.
| | - Kathryn A Mozzochi
- Drexel University College of Medicine Department of Otolaryngology Head and Neck Surgery, Philadelphia, Pennsylvania
| | - Mary J Hawkshaw
- Drexel University College of Medicine Department of Otolaryngology Head and Neck Surgery, Philadelphia, Pennsylvania
| | - Robert T Sataloff
- Drexel University College of Medicine Department of Otolaryngology Head and Neck Surgery, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Oury J, Gamallo-Lana B, Santana L, Steyaert C, Vergoossen DLE, Mar AC, Vankerckhoven B, Silence K, Vanhauwaert R, Huijbers MG, Burden SJ. Agonist antibody to MuSK protects mice from MuSK myasthenia gravis. Proc Natl Acad Sci U S A 2024; 121:e2408324121. [PMID: 39288173 PMCID: PMC11441477 DOI: 10.1073/pnas.2408324121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Myasthenia gravis (MG) is a chronic and severe disease of the skeletal neuromuscular junction (NMJ) in which the effects of neurotransmitters are attenuated, leading to muscle weakness. In the most common forms of autoimmune MG, antibodies attack components of the postsynaptic membrane, including the acetylcholine receptor (AChR) or muscle-specific kinase (MuSK). MuSK, a master regulator of NMJ development, associates with the low-density lipoprotein-related receptor 4 (Lrp4) to form the signaling receptor for neuronal Agrin, a nerve-derived synaptic organizer. Pathogenic antibodies to MuSK interfere with binding between MuSK and Lrp4, inhibiting the differentiation and maintenance of the NMJ. MuSK MG can be debilitating and refractory to treatments that are effective for AChR MG. We show here that recombinant antibodies, derived from MuSK MG patients, cause severe neuromuscular disease in mice. The disease can be prevented by a MuSK agonist antibody, presented either prophylactically or after disease onset. These findings suggest a therapeutic alternative to generalized immunosuppression for treating MuSK MG by selectively and directly targeting the disease mechanism.
Collapse
Affiliation(s)
- Julien Oury
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Medical School, New York, NY10016
| | - Begona Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University School of Medicine, New York, NY10016
| | - Leah Santana
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Medical School, New York, NY10016
| | | | - Dana L. E. Vergoossen
- Department of Human Genetics, Leiden University Medical Centre, Leiden2300 RC, The Netherlands
- Department of Neurology, Leiden University Medical Centre, Leiden2300 RC, The Netherlands
| | - Adam C. Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University School of Medicine, New York, NY10016
| | | | | | | | - Maartje G. Huijbers
- Department of Human Genetics, Leiden University Medical Centre, Leiden2300 RC, The Netherlands
- Department of Neurology, Leiden University Medical Centre, Leiden2300 RC, The Netherlands
| | - Steven J. Burden
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Medical School, New York, NY10016
| |
Collapse
|
4
|
Habib AA, Sacconi S, Antonini G, Cortés-Vicente E, Grosskreutz J, Mahuwala ZK, Mantegazza R, Pascuzzi RM, Utsugisawa K, Vissing J, Vu T, Wiendl H, Boehnlein M, Greve B, Woltering F, Bril V. Efficacy and safety of rozanolixizumab in patients with muscle-specific tyrosine kinase autoantibody-positive generalised myasthenia gravis: a subgroup analysis of the randomised, double-blind, placebo-controlled, adaptive phase III MycarinG study. Ther Adv Neurol Disord 2024; 17:17562864241273036. [PMID: 39297052 PMCID: PMC11409299 DOI: 10.1177/17562864241273036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/12/2024] [Indexed: 09/21/2024] Open
Abstract
Background Muscle-specific tyrosine kinase (MuSK) autoantibody-positive (Ab+) generalised myasthenia gravis (gMG) is a rare and frequently severe subtype of gMG. Objectives To assess the efficacy and safety of rozanolixizumab in the subgroup of patients with MuSK Ab+ gMG in the MycarinG study. Design A randomised, double-blind, placebo-controlled phase III study. Methods Patients with acetylcholine receptor (AChR) Ab+ or MuSK Ab+ gMG (aged ⩾18 years, Myasthenia Gravis Foundation of America Disease Class II-IVa, Myasthenia Gravis Activities of Daily Living [MG-ADL] score ⩾3.0 [non-ocular symptoms], Quantitative Myasthenia Gravis score ⩾11.0) were randomly assigned (1:1:1) to receive once-weekly subcutaneous infusions of rozanolixizumab 7 mg/kg, rozanolixizumab 10 mg/kg or placebo for 6 weeks, followed by an 8-week observation period. Randomisation was stratified by AChR and MuSK autoantibody status. The primary study endpoint was change from baseline to Day 43 in MG-ADL score. Treatment-emergent adverse events (TEAEs) were also assessed. Results Overall, 200 patients were randomised, of whom 21 had MuSK Ab+ gMG and received rozanolixizumab 7 mg/kg (n = 5), 10 mg/kg (n = 8) or placebo (n = 8). In patients with MuSK Ab+ gMG, reductions from baseline to Day 43 in MG-ADL scores were observed: rozanolixizumab 7 mg/kg least squares mean (LSM) change (standard error), -7.28 (1.94); 10 mg/kg, -4.16 (1.78); and placebo, 2.28 (1.95). Rozanolixizumab 7 mg/kg LSM difference from placebo was -9.56 (97.5% confidence interval: -15.25, -3.87); 10 mg/kg, -6.45 (-11.03, -1.86). TEAEs were experienced by four (80.0%), five (62.5%) and three (37.5%) patients with MuSK Ab+ gMG receiving rozanolixizumab 7 mg/kg, 10 mg/kg and placebo, respectively. No patients experienced serious TEAEs. No deaths occurred. Conclusion This subgroup analysis of adult patients with MuSK Ab+ gMG enrolled in the MycarinG study supports the use of rozanolixizumab as an effective treatment option for patients with gMG who have MuSK autoantibodies. Trial registration ClinicalTrials.gov: NCT03971422 (https://clinicaltrials.gov/study/NCT03971422); EU Clinical Trials Register: EudraCT 2019-000968-18 (https://www.clinicaltrialsregister.eu/ctr-search/trial/2019-000968-18/GB).
Collapse
Affiliation(s)
- Ali A Habib
- MDA ALS and Neuromuscular Center, University of California, 200 South Manchester Avenue, Suite 110, Irvine, Orange, CA 92868, USA
| | - Sabrina Sacconi
- Peripheral Nervous System & Muscle Department, Pasteur 2 Hospital, Centre Hospitalier Universitaire de Nice, Université Côte d'Azur, Nice, France
| | - Giovanni Antonini
- Department of Neuroscience, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Rome, Italy
| | - Elena Cortés-Vicente
- Neuromuscular Diseases Unit, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Julian Grosskreutz
- Precision Neurology of Neuromuscular Diseases, Department of Neurology, University of Lübeck, Lübeck, Germany
| | - Zabeen K Mahuwala
- Department of Neuromuscular Medicine, Epilepsy and Clinical Neurophysiology, University of Kentucky, Lexington, KY, USA
| | - Renato Mantegazza
- Department of Neuroimmunology and Neuromuscular Diseases, Fondazione IRCCS, Istituto Nazionale Neurologico Carlo Besta, Milan, Italy
| | - Robert M Pascuzzi
- Department of Neurology, Indiana University School of Medicine, Indiana University Health, Indianapolis, IN, USA
| | | | - John Vissing
- Department of Neurology, Copenhagen Neuromuscular Center, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tuan Vu
- Department of Neurology, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Heinz Wiendl
- Department of Neurology, Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | | | | | | | - Vera Bril
- Department of Neurology, University Health Network, Toronto, ON, Canada
| |
Collapse
|
5
|
Kaminski HJ, Sikorski P, Coronel SI, Kusner LL. Myasthenia gravis: the future is here. J Clin Invest 2024; 134:e179742. [PMID: 39105625 DOI: 10.1172/jci179742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024] Open
Abstract
Myasthenia gravis (MG) stands as a prototypical antibody-mediated autoimmune disease: it is dependent on T cells and characterized by the presence of autoantibodies targeting proteins located on the postsynaptic surface of skeletal muscle, known as the neuromuscular junction. Patients with MG exhibit a spectrum of weakness, ranging from limited ocular muscle involvement to life-threatening respiratory failure. Recent decades have witnessed substantial progress in understanding the underlying pathophysiology, leading to the delineation of distinct subcategories within MG, including MG linked to AChR or MuSK antibodies as well as age-based distinction, thymoma-associated, and immune checkpoint inhibitor-induced MG. This heightened understanding has paved the way for the development of more precise and targeted therapeutic interventions. Notably, the FDA has recently approved therapeutic inhibitors of complement and the IgG receptor FcRn, a testament to our improved comprehension of autoantibody effector mechanisms in MG. In this Review, we delve into the various subgroups of MG, stratified by age, autoantibody type, and histology of the thymus with neoplasms. Furthermore, we explore both current and potential emerging therapeutic strategies, shedding light on the evolving landscape of MG treatment.
Collapse
Affiliation(s)
| | | | | | - Linda L Kusner
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| |
Collapse
|
6
|
Ricciardi R, Latini E, Guida M, Koneczny I, Lucchi M, Maestri M, De Rosa A, Vincent A. Acetylcholinesterase inhibitors are ineffective in MuSK-antibody positive myasthenia gravis: Results of a study on 202 patients. J Neurol Sci 2024; 461:123047. [PMID: 38759248 DOI: 10.1016/j.jns.2024.123047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/04/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Myasthenia gravis (MG) with MuSK antibodies (MuSK-MG) represents a distinct subtype with different responses to treatments compared to patients with AChR antibodies, especially in terms of tolerance to acetylcholinesterase inhibitors (AChEI). However, AChEI are often used as first line symptomatic treatment in MuSK-MG, despite reports that they are poorly tolerated, seldom effective or even deleterious. METHODS We analyzed demographic, clinical and therapeutic responses and side-effects in the large cohort of 202 MuSK-MG patients cared for at the MG Clinic of Azienda Ospedaliero-Universitaria Pisana. RESULTS 165 patients had received AChEI at first evaluation. Only 7/165 patients (4.2%) reported an initial clinical benefit. Conversely, 76.9% of patients reported at least one side effect, most commonly neuromuscular hyperexcitability (68.4%), gastrointestinal (53.9%) and neurovegetative (35.8%) disturbances. 56 (33.9%) patients reported a concomitant worsening of muscle weakness and twelve patients (7.3%) suffered a cholinergic crisis. According to these patients, the severity of cholinergic side effects was greater at higher doses of AChEI, but side effects occurred regardless of the dose administered and ceased once the drug was discontinued. CONCLUSIONS This is the largest population of MuSK-MG patients reported for perceived responsiveness and tolerance to AChEI treatment. Our obervations strongly suggest avoiding this treatment in MuSK-MG.
Collapse
Affiliation(s)
- R Ricciardi
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Italy; CardioThoracic and Vascular Surgery Department, University of Pisa, Italy
| | - E Latini
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Italy.
| | - M Guida
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Italy
| | - I Koneczny
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - M Lucchi
- CardioThoracic and Vascular Surgery Department, University of Pisa, Italy
| | - M Maestri
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Italy
| | - A De Rosa
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Italy
| | - A Vincent
- Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
7
|
Rudolf R, Kettelhut IC, Navegantes LCC. Sympathetic innervation in skeletal muscle and its role at the neuromuscular junction. J Muscle Res Cell Motil 2024; 45:79-86. [PMID: 38367152 PMCID: PMC11096211 DOI: 10.1007/s10974-024-09665-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 01/30/2024] [Indexed: 02/19/2024]
Abstract
Neuromuscular junctions are the synapses between motor neurons and skeletal muscle fibers, which mediate voluntary muscle movement. Since neuromuscular junctions are also tightly associated with the capping function of terminal Schwann cells, these synapses have been classically regarded as tripartite chemical synapses. Although evidences from sympathetic innervation of neuromuscular junctions was described approximately a century ago, the essential presence and functional relevance of sympathetic contribution to the maintenance and modulation of neuromuscular junctions was demonstrated only recently. These findings shed light on the pathophysiology of different clinical conditions and can optimize surgical and clinical treatment modalities for skeletal muscle disorders.
Collapse
Affiliation(s)
- Rüdiger Rudolf
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163, Mannheim, Germany.
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69117, Heidelberg, Germany.
- Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim Heidelberg University, 69167, Mannheim, Germany.
| | - Isis C Kettelhut
- Department of Biochemistry & Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, 14049900, Brazil
| | - Luiz Carlos C Navegantes
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, 14049900, Brazil
| |
Collapse
|
8
|
Barahman M, Shamsaei G, Kashipazha D, Bahadoram M, Akade E. Paraneoplastic neurological syndromes of small cell lung cancer. POSTEPY PSYCHIATRII NEUROLOGII 2024; 33:80-92. [PMID: 39119541 PMCID: PMC11304241 DOI: 10.5114/ppn.2024.141157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 03/29/2024] [Indexed: 08/10/2024]
Abstract
Purpose This article reviews the relevant literature on paraneoplastic neurological syndromes of small cell lung cancer and discusses the clinical presentation, pathophysiology, and diagnosis of these syndromes. It also includes a summary of the current treatment options for the management of them. Views Paraneoplastic syndromes are a group of signs and symptoms that develop due to cancer in a remote site, mainly triggered by an autoantibody produced by the tissues involved or lymphocytes during anti-cancer defense. Among the cancers associated with paraneoplastic syndromes, lung cancers are the most common type, with small cell lung cancer being the most common subtype. The most common antibody associated with paraneoplastic syndromes is anti-Hu. Neurological and neuroendocrine syndromes comprise the majority of small cell lung cancer-related paraneoplastic syndromes. Classical paraneoplastic neurological syndromes include inappropriate antidiuretic hormone secretion, Cushing's syndrome, myasthenia gravis, Lambert-Eaton myasthenic syndrome, limbic encephalitis, paraneoplastic cerebellar degeneration, opsoclonus myoclonus ataxia, sensory neuropathy, and chorea. Conclusions Antibodies mediate paraneoplastic syndromes, and antibody detection is a crucial part of diagnosing these entities. Managing the underlying tumor is the best treatment approach for most paraneoplastic syndromes. Therefore, early diagnosis of small cell lung cancer may significantly improve the prognosis of paraneoplastic syndromes associated with it.
Collapse
Affiliation(s)
- Maedeh Barahman
- Department of Radiation Oncology, Firoozgar Hospital, Firoozgar Clinical Research Development Center (FCRDC), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Gholamreza Shamsaei
- Department of Neurology, School of Medicine, Musculoskeletal Rehabilitation Research Center, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Davood Kashipazha
- Department of Neurology, School of Medicine, Musculoskeletal Rehabilitation Research Center, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Bahadoram
- Department of Neurology, School of Medicine, Musculoskeletal Rehabilitation Research Center, Golestan Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Esma’il Akade
- Department of Medical Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
9
|
Ma C, Liu D, Wang B, Yang Y, Zhu R. Advancements and prospects of novel biologicals for myasthenia gravis: toward personalized treatment based on autoantibody specificities. Front Pharmacol 2024; 15:1370411. [PMID: 38881870 PMCID: PMC11177092 DOI: 10.3389/fphar.2024.1370411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/06/2024] [Indexed: 06/18/2024] Open
Abstract
Myasthenia gravis (MG) is an antibody-mediated autoimmune disease with a prevalence of 150-250 cases per million individuals. Autoantibodies include long-lived antibodies against the acetylcholine receptor (AChR), mainly of the IgG1 subclass, and IgG4, produced almost exclusively by short-lived plasmablasts, which are prevalent in muscle-specific tyrosine kinase (MuSK) myasthenia gravis. Numerous investigations have demonstrated that MG patients receiving conventional medication today still do not possess satisfactory symptom control, indicating a substantial disease burden. Subsequently, based on the type of the autoantibody and the pathogenesis, we synthesized the published material to date and reached a conclusion regarding the literature related to personalized targeted therapy for MG. Novel agents for AChR MG have shown their efficacy in clinical research, such as complement inhibitors, FcRn receptor antagonists, and B-cell activating factor (BAFF) inhibitors. Rituximab, a representative drug of anti-CD20 therapy, has demonstrated benefits in treatment of MuSK MG patients. Due to the existence of low-affinity antibodies or unidentified antibodies that are inaccessible by existing methods, the treatment for seronegative MG remains complicated; thus, special testing and therapy considerations are necessary. It may be advantageous to initiate the application of novel biologicals at an early stage of the disease. Currently, therapies can also be combined and individualized according to different types of antibodies. With such a wide range of drugs, how to tailor treatment strategies to patients with various conditions and find the most suitable solution for each MG profile are our necessary and urgent aims.
Collapse
Affiliation(s)
- Chi Ma
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dan Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Benqiao Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yingying Yang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ruixia Zhu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Herbst R, Huijbers MG, Oury J, Burden SJ. Building, Breaking, and Repairing Neuromuscular Synapses. Cold Spring Harb Perspect Biol 2024; 16:a041490. [PMID: 38697654 PMCID: PMC11065174 DOI: 10.1101/cshperspect.a041490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
A coordinated and complex interplay of signals between motor neurons, skeletal muscle cells, and Schwann cells controls the formation and maintenance of neuromuscular synapses. Deficits in the signaling pathway for building synapses, caused by mutations in critical genes or autoantibodies against key proteins, are responsible for several neuromuscular diseases, which cause muscle weakness and fatigue. Here, we describe the role that four key genes, Agrin, Lrp4, MuSK, and Dok7, play in this signaling pathway, how an understanding of their mechanisms of action has led to an understanding of several neuromuscular diseases, and how this knowledge has contributed to emerging therapies for treating neuromuscular diseases.
Collapse
Affiliation(s)
- Ruth Herbst
- Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Maartje G Huijbers
- Department of Human Genetics, Leiden University Medical Centre LUMC, 2300 RC Leiden, the Netherlands
- Department of Neurology, Leiden University Medical Centre LUMC, 2333 ZA Leiden, the Netherlands
| | - Julien Oury
- Helen L. and Martin S. Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, NYU School of Medicine, New York, New York 10016, USA
| | - Steven J Burden
- Neurology Department, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| |
Collapse
|
11
|
Koneczny I, Mané-Damas M, Zong S, De Haas S, Huda S, van Kruining D, Damoiseaux J, De Rosa A, Maestri M, Guida M, Molenaar P, Van Damme P, Fichtenbaum A, Perkmann T, De Baets M, Lazaridis K, Zouvelou V, Tzartos S, Ricciardi R, Losen M, Martinez-Martinez P. A retrospective multicenter study on clinical and serological parameters in patients with MuSK myasthenia gravis with and without general immunosuppression. Front Immunol 2024; 15:1325171. [PMID: 38715598 PMCID: PMC11074957 DOI: 10.3389/fimmu.2024.1325171] [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: 10/23/2023] [Accepted: 03/25/2024] [Indexed: 06/19/2024] Open
Abstract
Introduction Muscle-specific kinase (MuSK)- myasthenia gravis (MG) is caused by pathogenic autoantibodies against MuSK that correlate with disease severity and are predominantly of the IgG4 subclass. The first-line treatment for MuSK-MG is general immunosuppression with corticosteroids, but the effect of treatment on IgG4 and MuSK IgG4 levels has not been studied. Methods We analyzed the clinical data and sera from 52 MuSK-MG patients (45 female, 7 male, median age 49 (range 17-79) years) from Italy, the Netherlands, Greece and Belgium, and 43 AChR-MG patients (22 female, 21 male, median age 63 (range 2-82) years) from Italy, receiving different types of immunosuppression, and sera from 46 age- and sex-matched non-disease controls (with no diagnosed diseases, 38 female, 8 male, median age 51.5 (range 20-68) years) from the Netherlands. We analyzed the disease severity (assessed by MGFA or QMG score), and measured concentrations of MuSK IgG4, MuSK IgG, total IgG4 and total IgG in the sera by ELISA, RIA and nephelometry. Results We observed that MuSK-MG patients showed a robust clinical improvement and reduction of MuSK IgG after therapy, and that MuSK IgG4 concentrations, but not total IgG4 concentrations, correlated with clinical severity. MuSK IgG and MuSK IgG4 concentrations were reduced after immunosuppression in 4/5 individuals with before-after data, but data from non-linked patient samples showed no difference. Total serum IgG4 levels were within the normal range, with IgG4 levels above threshold (1.35g/L) in 1/52 MuSK-MG, 2/43 AChR-MG patients and 1/45 non-disease controls. MuSK-MG patients improved within the first four years after disease onset, but no further clinical improvement or reduction of MuSK IgG4 were observed four years later, and only 14/52 (26.92%) patients in total, of which 13 (93.3%) received general immunosuppression, reached clinical remission. Discussion We conclude that MuSK-MG patients improve clinically with general immunosuppression but may require further treatment to reach remission. Longitudinal testing of individual patients may be clinically more useful than single measurements of MuSK IgG4. No significant differences in the serum IgG4 concentrations and IgG4/IgG ratio between AChR- and MuSK-MG patients were found during follow-up. Further studies with larger patient and control cohorts are necessary to validate the findings.
Collapse
Affiliation(s)
- Inga Koneczny
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Marina Mané-Damas
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Shenghua Zong
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Sander De Haas
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Saif Huda
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
- Department of Neurology, Walton Centre National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Daan van Kruining
- Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, Netherlands
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, Netherlands
| | - Anna De Rosa
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | - Michelangelo Maestri
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | - Melania Guida
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | - Peter Molenaar
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Philip Van Damme
- Neurology Department, University Hospital, Leuven, Belgium
- Department of Neurosciences, Center for Brain & Disease Research, VIB, Leuven, Belgium
| | - Andreas Fichtenbaum
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Marc De Baets
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | | | - Vasiliki Zouvelou
- 1stNeurology Department, National and Kapodistrian University of Athens, Athens, Greece
| | - Socrates Tzartos
- Department of Immunology, Hellenic Pasteur Institute, Athens, Greece
- Department of Neuroimmunology, Tzartos NeuroDiagnostics, Athens, Greece
| | - Roberta Ricciardi
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
- Cardio Thoracic and Vascular Surgery Department, University of Pisa, Pisa, Italy
| | - Mario Losen
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Pilar Martinez-Martinez
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
12
|
Jaime D, Fish LA, Madigan LA, Xi C, Piccoli G, Ewing MD, Blaauw B, Fallon JR. The MuSK-BMP pathway maintains myofiber size in slow muscle through regulation of Akt-mTOR signaling. Skelet Muscle 2024; 14:1. [PMID: 38172960 PMCID: PMC10763067 DOI: 10.1186/s13395-023-00329-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 10/19/2023] [Indexed: 01/05/2024] Open
Abstract
Myofiber size regulation is critical in health, disease, and aging. MuSK (muscle-specific kinase) is a BMP (bone morphogenetic protein) co-receptor that promotes and shapes BMP signaling. MuSK is expressed at all neuromuscular junctions and is also present extrasynaptically in the mouse soleus, whose predominantly oxidative fiber composition is akin to that of human muscle. To investigate the role of the MuSK-BMP pathway in vivo, we generated mice lacking the BMP-binding MuSK Ig3 domain. These ∆Ig3-MuSK mice are viable and fertile with innervation levels comparable to wild type. In 3-month-old mice, myofibers are smaller in the slow soleus, but not in the fast tibialis anterior (TA). Transcriptomic analysis revealed soleus-selective decreases in RNA metabolism and protein synthesis pathways as well as dysregulation of IGF1-Akt-mTOR pathway components. Biochemical analysis showed that Akt-mTOR signaling is reduced in soleus but not TA. We propose that the MuSK-BMP pathway acts extrasynaptically to maintain myofiber size in slow muscle by promoting protein synthetic pathways including IGF1-Akt-mTOR signaling. These results reveal a novel mechanism for regulating myofiber size in slow muscle and introduce the MuSK-BMP pathway as a target for promoting muscle growth and combatting atrophy.
Collapse
Grants
- R41 AG073144 NIA NIH HHS
- T32 MH020068 NIMH NIH HHS
- U01 NS064295, R41 AG073144, R21 NS112743, R21 AG073743, P30 GM103410, P30 RR031153, P20 RR018728, S10 RR02763, R25GM083270, 2T32AG041688, and T32 MH20068 NIH HHS
- P30 GM103410 NIGMS NIH HHS
- T32 AG041688 NIA NIH HHS
- P30 RR031153 NCRR NIH HHS
- U01 NS064295 NINDS NIH HHS
- R21 NS112743 NINDS NIH HHS
- P20 RR018728 NCRR NIH HHS
- R21 AG073743 NIA NIH HHS
- R25 GM083270 NIGMS NIH HHS
- National Institutes of Health
- Carney Institute for Brain Sciences
- ALS Finding a Cure
- AFM-Téléthon
Collapse
Affiliation(s)
- Diego Jaime
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
| | - Lauren A Fish
- Department of Neuroscience, Brown University, Providence, RI, 02912, USA
| | - Laura A Madigan
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, USA
| | - Chengjie Xi
- Department of Neuroscience, Brown University, Providence, RI, 02912, USA
| | - Giorgia Piccoli
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Madison D Ewing
- Department of Neuroscience, Brown University, Providence, RI, 02912, USA
| | - Bert Blaauw
- Veneto Institute of Molecular Medicine (VIMM), Padua, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Justin R Fallon
- Department of Neuroscience, Brown University, Providence, RI, 02912, USA.
- Carney Institute for Neuroscience, Brown University, Providence, RI, USA.
| |
Collapse
|
13
|
Cao M, Liu WW, Maxwell S, Huda S, Webster R, Evoli A, Beeson D, Cossins JA, Vincent A. IgG1-3 MuSK Antibodies Inhibit AChR Cluster Formation, Restored by SHP2 Inhibitor, Despite Normal MuSK, DOK7, or AChR Subunit Phosphorylation. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200147. [PMID: 37582613 PMCID: PMC10427144 DOI: 10.1212/nxi.0000000000200147] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/07/2023] [Indexed: 08/17/2023]
Abstract
BACKGROUND AND OBJECTIVES Up to 50% of patients with myasthenia gravis (MG) without acetylcholine receptor antibodies (AChR-Abs) have antibodies to muscle-specific kinase (MuSK). Most MuSK antibodies (MuSK-Abs) are IgG4 and inhibit agrin-induced MuSK phosphorylation, leading to impaired clustering of AChRs at the developing or mature neuromuscular junction. However, IgG1-3 MuSK-Abs also exist in MuSK-MG patients, and their potential mechanisms have not been explored fully. METHODS C2C12 myotubes were exposed to MuSK-MG plasma IgG1-3 or IgG4, with or without purified agrin. MuSK, Downstream of Kinase 7 (DOK7), and βAChR were immunoprecipitated and their phosphorylation levels identified by immunoblotting. Agrin and agrin-independent AChR clusters were measured by immunofluorescence and AChR numbers by binding of 125I-α-bungarotoxin. Transcriptomic analysis was performed on treated myotubes. RESULTS IgG1-3 MuSK-Abs impaired AChR clustering without inhibiting agrin-induced MuSK phosphorylation. Moreover, the well-established pathway initiated by MuSK through DOK7, resulting in βAChR phosphorylation, was not impaired by MuSK-IgG1-3 and was agrin-independent. Nevertheless, the AChR clusters did not form, and both the number of AChR microclusters that precede full cluster formation and the myotube surface AChRs were reduced. Transcriptomic analysis did not throw light on the pathways involved. However, the SHP2 inhibitor, NSC-87877, increased the number of microclusters and led to fully formed AChR clusters. DISCUSSION MuSK-IgG1-3 is pathogenic but seems to act through a noncanonical pathway. Further studies should throw light on the mechanisms involved at the neuromuscular junction.
Collapse
Affiliation(s)
- Michelangelo Cao
- From the Nuffield Department of Clinical Neurosciences (M.C., W.W.L., S.M., R.W., D.B., J.A.C., A.V.), University of Oxford; Norfolk and Norwich University Hospital (M.C.); The Walton Centre NHS Foundation Trust (S.H.), Liverpool, United Kingdom; and Department of Neuroscience (A.E.), Catholic University, Rome, Italy
| | - Wei-Wei Liu
- From the Nuffield Department of Clinical Neurosciences (M.C., W.W.L., S.M., R.W., D.B., J.A.C., A.V.), University of Oxford; Norfolk and Norwich University Hospital (M.C.); The Walton Centre NHS Foundation Trust (S.H.), Liverpool, United Kingdom; and Department of Neuroscience (A.E.), Catholic University, Rome, Italy
| | - Susan Maxwell
- From the Nuffield Department of Clinical Neurosciences (M.C., W.W.L., S.M., R.W., D.B., J.A.C., A.V.), University of Oxford; Norfolk and Norwich University Hospital (M.C.); The Walton Centre NHS Foundation Trust (S.H.), Liverpool, United Kingdom; and Department of Neuroscience (A.E.), Catholic University, Rome, Italy
| | - Saif Huda
- From the Nuffield Department of Clinical Neurosciences (M.C., W.W.L., S.M., R.W., D.B., J.A.C., A.V.), University of Oxford; Norfolk and Norwich University Hospital (M.C.); The Walton Centre NHS Foundation Trust (S.H.), Liverpool, United Kingdom; and Department of Neuroscience (A.E.), Catholic University, Rome, Italy
| | - Richard Webster
- From the Nuffield Department of Clinical Neurosciences (M.C., W.W.L., S.M., R.W., D.B., J.A.C., A.V.), University of Oxford; Norfolk and Norwich University Hospital (M.C.); The Walton Centre NHS Foundation Trust (S.H.), Liverpool, United Kingdom; and Department of Neuroscience (A.E.), Catholic University, Rome, Italy
| | - Amelia Evoli
- From the Nuffield Department of Clinical Neurosciences (M.C., W.W.L., S.M., R.W., D.B., J.A.C., A.V.), University of Oxford; Norfolk and Norwich University Hospital (M.C.); The Walton Centre NHS Foundation Trust (S.H.), Liverpool, United Kingdom; and Department of Neuroscience (A.E.), Catholic University, Rome, Italy
| | - David Beeson
- From the Nuffield Department of Clinical Neurosciences (M.C., W.W.L., S.M., R.W., D.B., J.A.C., A.V.), University of Oxford; Norfolk and Norwich University Hospital (M.C.); The Walton Centre NHS Foundation Trust (S.H.), Liverpool, United Kingdom; and Department of Neuroscience (A.E.), Catholic University, Rome, Italy
| | - Judith A Cossins
- From the Nuffield Department of Clinical Neurosciences (M.C., W.W.L., S.M., R.W., D.B., J.A.C., A.V.), University of Oxford; Norfolk and Norwich University Hospital (M.C.); The Walton Centre NHS Foundation Trust (S.H.), Liverpool, United Kingdom; and Department of Neuroscience (A.E.), Catholic University, Rome, Italy
| | - Angela Vincent
- From the Nuffield Department of Clinical Neurosciences (M.C., W.W.L., S.M., R.W., D.B., J.A.C., A.V.), University of Oxford; Norfolk and Norwich University Hospital (M.C.); The Walton Centre NHS Foundation Trust (S.H.), Liverpool, United Kingdom; and Department of Neuroscience (A.E.), Catholic University, Rome, Italy.
| |
Collapse
|
14
|
Baisya R, Yerram KV, Baby A, Devarasetti PK, Rajasekhar L. Immunoglobulin G4-Related Lesions in Autoimmune Diseases: Unusual Presentations at Atypical Sites-A Tale of 2 Cases with Literature Review. Eur J Rheumatol 2023; 10:169-175. [PMID: 37873667 PMCID: PMC10765183 DOI: 10.5152/eurjrheum.2023.23052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/13/2023] [Indexed: 10/25/2023] Open
Abstract
Immunoglobulin G4-related disease (IgG4-RD) coexisting with clinically apparent autoimmune diseases, such as rheumatoid arthritis (RA) or antiphospholipid syndrome (APS), is a rarely documented combination in the scientific literature. In this case-based review, we present 2 intriguing cases with preexisting autoimmune diseases, namely, RA and primary APS, who exhibited coexistent IgG4- related lesions at unusual sites. The first case pertains to a patient with known RA who presented with an encasing mass in the esophagus leading to stricture, with histopathological diagnosis of IgG4-RD.The second patient, diagnosed with primary APS, experienced breathlessness, and imaging revealed a right atrial mass. Histopathological examination of the mass confirmed IgG4-RD. Notably, both patients demonstrated significant clinical improvement upon initiation of steroid therapy. Rheumatoid arthritis patients commonly exhibit elevated levels of IgG4 in their sera; however, RA with coexisting IgG4-RD is rarely reported in the literature. Similarly, APS with IgG4-related lesions is exceedingly rare. Although there are few case reports and series on esophageal and cardiac IgG4-RD, the occurrence of such unusual location of IgG4-related lesions in the context of known autoimmunity is presented here for the first time.
Collapse
Affiliation(s)
- Ritasman Baisya
- Clinical Immunology and Rheumatology, Nizam’s Institute of Medical Sciences, Hyderabad, India
| | - Keerthi Vardhan Yerram
- Clinical Immunology and Rheumatology, Nizam’s Institute of Medical Sciences, Hyderabad, India
| | - Arun Baby
- Clinical Immunology and Rheumatology, Nizam’s Institute of Medical Sciences, Hyderabad, India
| | - Phani Kumar Devarasetti
- Clinical Immunology and Rheumatology, Nizam’s Institute of Medical Sciences, Hyderabad, India
| | - Liza Rajasekhar
- Clinical Immunology and Rheumatology, Nizam’s Institute of Medical Sciences, Hyderabad, India
| |
Collapse
|
15
|
Mori S, Suzuki S, Konishi T, Kawaguchi N, Kishi M, Kuwabara S, Ishizuchi K, Zhou H, Shibasaki F, Tsumoto H, Omura T, Miura Y, Mori S, Higashihara M, Murayama S, Shigemoto K. Proteolytic ectodomain shedding of muscle-specific tyrosine kinase in myasthenia gravis. Exp Neurol 2023; 361:114300. [PMID: 36525997 DOI: 10.1016/j.expneurol.2022.114300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/07/2022] [Accepted: 12/10/2022] [Indexed: 12/15/2022]
Abstract
Autoantibodies to muscle-specific tyrosine kinase (MuSK) proteins at the neuromuscular junction (NMJ) cause refractory generalized myasthenia gravis (MG) with dyspnea more frequently than other MG subtypes. However, the mechanisms via which MuSK, a membrane protein locally expressed on the NMJ of skeletal muscle, is supplied to the immune system as an autoantigen remains unknown. Here, we identified MuSK in both mouse and human serum, with the amount of MuSK dramatically increasing in mice with motor nerve denervation and in MG model mice. Peptide analysis by liquid chromatography-tandem-mass spectrometry (LC-MS/MS) confirmed the presence of MuSK in both human and mouse serum. Furthermore, some patients with MG have significantly higher amounts of MuSK in serum than healthy controls. Our results indicated that the secretion of MuSK proteins from muscles into the bloodstream was induced by ectodomain shedding triggered by neuromuscular junction failure. The results may explain why MuSK-MG is refractory to treatments and causes rapid muscle atrophy in some patients due to the denervation associated with Ab-induced disruption of neuromuscular transmission at the NMJ. Such discoveries pave the way for new MG treatments, and MuSK may be used as a biomarker for other neuromuscular diseases in preclinical studies, clinical diagnostics, therapeutics, and drug discovery.
Collapse
Affiliation(s)
- Shuuichi Mori
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Shigeaki Suzuki
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | | | - Naoki Kawaguchi
- Dowa Institute of Clinical Neuroscience, Neurology Clinic Chiba, Chiba, Japan
| | - Masahiko Kishi
- Department of Internal Medicine, Toho University Sakura Medical Center, Chiba, Japan
| | - Satoshi Kuwabara
- Department of Neurology, Chiba University School of Medicine, Chiba, Japan
| | - Kei Ishizuchi
- Department of Neurology, Keio University School of Medicine, Tokyo, Japan
| | - Heying Zhou
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Futoshi Shibasaki
- Department of Genome Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hiroki Tsumoto
- Research Team for Mechanism of Aging, TMIG, Tokyo, Japan
| | - Takuya Omura
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Yuri Miura
- Research Team for Mechanism of Aging, TMIG, Tokyo, Japan
| | - Seijiro Mori
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan
| | - Mana Higashihara
- Department of Neurology, Tokyo Metropolitan Geriatric Hospital, Tokyo (TMGHIG), Japan
| | | | - Kazuhiro Shigemoto
- Research Team for Geriatric Medicine, Tokyo Metropolitan Institute of Gerontology (TMIG), Tokyo, Japan.
| |
Collapse
|
16
|
Jaime D, Fish LA, Madigan LA, Ewing MD, Fallon JR. The MuSK-BMP pathway maintains myofiber size in slow muscle through regulation of Akt- mTOR signaling. RESEARCH SQUARE 2023:rs.3.rs-2613527. [PMID: 36909467 PMCID: PMC10002845 DOI: 10.21203/rs.3.rs-2613527/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
Myofiber size regulation is critical in health, disease, and aging. MuSK (muscle-specific kinase) is a BMP (bone morphogenetic protein) co-receptor that promotes and shapes BMP signaling. MuSK is expressed at all neuromuscular junctions and is also present extrasynaptically in the slow soleus muscle. To investigate the role of the MuSK-BMP pathway in vivo we generated mice lacking the BMP-binding MuSK Ig3 domain. These ΔIg3-MuSKmice are viable and fertile with innervation levels comparable to wild type. In 3-month-old mice myofibers are smaller in the slow soleus, but not in the fast tibialis anterior (TA). Transcriptomic analysis revealed soleus-selective decreases in RNA metabolism and protein synthesis pathways as well as dysregulation of IGF1-Akt-mTOR pathway components. Biochemical analysis showed that Akt-mTOR signaling is reduced in soleus but not TA. We propose that the MuSK-BMP pathway acts extrasynaptically to maintain myofiber size in slow muscle by promoting protein synthetic pathways including IGF1-Akt-mTOR signaling. These results reveal a novel mechanism for regulating myofiber size in slow muscle and introduce the MuSK-BMP pathway as a target for promoting muscle growth and combatting atrophy.
Collapse
|
17
|
Moritz CP, Do LD, Tholance Y, Vallayer PB, Rogemond V, Joubert B, Ferraud K, La Marca C, Camdessanché JP, Honnorat J, Antoine JC. Conformation-stabilizing ELISA and cell-based assays reveal patient subgroups targeting three different epitopes of AGO1 antibodies. Front Immunol 2022; 13:972161. [PMID: 36341350 PMCID: PMC9630334 DOI: 10.3389/fimmu.2022.972161] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/06/2022] [Indexed: 12/20/2024] Open
Abstract
Autoantibodies (Abs) are biomarkers for many disease conditions and are increasingly used to facilitate diagnosis and treatment decisions. To guarantee high sensitivity and specificity, the choice of their detection method is crucial. Via cell-based assays, we recently found 21 patients with neurological diseases positive for antibodies against argonaute (AGO), 10 of which having a neuropathy (NP). Here, we established a simple and conformation-sensitive ELISA with the aim to distinguish between AGO1 Abs against conformational epitopes and non-conformational epitopes and to reveal further characteristics of AGO1 antibodies in NP and autoimmune disease (AID). In a retrospective multicenter case/control and observational study, we tested 434 patients with NP, 274 disease controls with AID, and 116 healthy controls (HC) for AGO1 Abs via conformation-stabilizing ELISA. Seropositive patients were also tested for conformation-specificity via comparative denaturing/stabilizing ELISA (CODES-ELISA), CBA positivity, AGO1 titers and IgG subclasses, and AGO2 reactivity. These parameters were statistically compared among different epitope-specific patient groups. We found Abs in 44 patients, including 28/434 (6.5%) NP, 16/274 (5.8%) AID, and 0/116 (0%) HC. Serum reactivity was consistently higher for AGO1 than AGO2. Globally among the 44 AGO1 Abs-positive patients, 42 were also tested in CBA for AGO1 Abs positivity and 15 (35.7%) were positive. Furthermore, 43 were tested for conformation-specificity and 32 (74.4%) bound a conformational epitope. Among the subgroups of highly positive patients (ELISA z-score >14) with sera binding conformational epitopes (n=23), 14 patient sera were also CBA positive and 9 bound a second conformational but CBA-inaccessible epitope. A third, non-conformational epitope was bound by 11/43 (15.6%). Among the epitope-specific patient subgroups, we found significant differences regarding the Abs titers, IgG subclass, and AGO2 reactivity. When comparing AGO1 Abs-positive NP versus AID patients, we found the conformation-specific and CBA inaccessible epitope significantly more frequently in AID patients. We conclude that 1) conformational ELISA was more sensitive than CBA in detecting AGO1 Abs, 2) serum reactivity is higher for AGO1 than for AGO2 at least for NP patients, 3) AGO1 Abs might be a marker-of-interest in 6.5% of NP patients, 4) distinguishing epitopes might help finding different patient subgroups.
Collapse
Affiliation(s)
- Christian P. Moritz
- Department of Neurology, University hospital of Saint-Etienne, Saint-Etienne, France
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- INMG/Melys team, University Jean Monnet, Saint-Étienne, France
| | - Le-Duy Do
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Bron, France
| | - Yannick Tholance
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- INMG/Melys team, University Jean Monnet, Saint-Étienne, France
- Department of Biochemistry, University Hospital of Saint-Etienne, Saint-Etienne, France
| | | | - Véronique Rogemond
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Bron, France
| | - Bastien Joubert
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Bron, France
| | - Karine Ferraud
- Department of Neurology, University hospital of Saint-Etienne, Saint-Etienne, France
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Coralie La Marca
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Department of Biochemistry, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Jean-Philippe Camdessanché
- Department of Neurology, University hospital of Saint-Etienne, Saint-Etienne, France
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- INMG/Melys team, University Jean Monnet, Saint-Étienne, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Bron, France
- European Reference Network for Rare Neuromuscular Diseases, Paris, France
| | - Jérôme Honnorat
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Bron, France
- European Reference Network for Rare Neuromuscular Diseases, Paris, France
| | - Jean-Christophe Antoine
- Department of Neurology, University hospital of Saint-Etienne, Saint-Etienne, France
- Synaptopathies and autoantibodies (SynatAc) team, Mechanisms In Integrated Life Sciences (MELIS) Laboratory, Institute NeuroMyoGène (INMG), INSERM U1314/CNRS UMR 5284, Universités de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- INMG/Melys team, University Jean Monnet, Saint-Étienne, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Bron, France
- European Reference Network for Rare Neuromuscular Diseases, Paris, France
| |
Collapse
|
18
|
Severe Obstructive Sleep Apnea in an Adolescent with Hypernasality, Dysarthria, and Muscle Weakness. Ann Am Thorac Soc 2022; 19:1764-1768. [PMID: 36178398 DOI: 10.1513/annalsats.202201-030cc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
19
|
Waheed W, Newman E, Aboukhatwa M, Moin M, Tandan R. Practical Management for Use of Eculizumab in the Treatment of Severe, Refractory, Non-Thymomatous, AChR + Generalized Myasthenia Gravis: A Systematic Review. Ther Clin Risk Manag 2022; 18:699-719. [PMID: 35855752 PMCID: PMC9288180 DOI: 10.2147/tcrm.s266031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 07/04/2022] [Indexed: 11/28/2022] Open
Abstract
Myasthenia gravis (MG) is a rare autoimmune disorder caused by specific autoantibodies at the neuromuscular junction. MG is classified by the antigen specificity of these antibodies. Acetylcholine receptor (AChR) antibodies are the most common type (74–88%), followed by anti-muscle specific kinase (MuSK) and other antibodies. While all these antibodies lead to neuromuscular transmission failure, the immuno-pathogenic mechanisms are distinct. Complement activation is a primary driver of AChR antibody-positive MG (AChR+ MG) pathogenesis. This leads to the formation of the membrane attack complex and destruction of AChR receptors and the postsynaptic membrane resulting in impaired neurotransmission and muscle weakness characteristic of MG. Broad-based immune-suppressants like corticosteroids are effective in controlling MG; however, their long-term use can be associated with significant adverse effects. Advances in translational research have led to the development of more directed therapeutic agents that are likely to alter the future of MG treatment. Eculizumab is a humanized monoclonal antibody that inhibits the cleavage of complement protein C5 and is approved for use in generalized MG. In this review, we discuss the pathophysiology of MG; the therapeutic efficacy and tolerability of eculizumab, as well as the practical guidelines for its use in MG; future studies exploring the role of eculizumab in different stages and subtypes of MG subtypes; the optimal duration of therapy and its discontinuation; the characterization of non-responder patients; and the use of biomarkers for monitoring therapy are highlighted. Based on the pathophysiologic mechanisms, emerging therapies and new therapeutic targets are also reviewed.
Collapse
Affiliation(s)
- Waqar Waheed
- Department of Neurological Sciences, The University of Vermont and the University of Vermont Medical Center, Burlington, VT, USA
| | - Eric Newman
- Department of Neurological Sciences, The University of Vermont and the University of Vermont Medical Center, Burlington, VT, USA
| | - Marwa Aboukhatwa
- Pharmacotherapy Department, University of Vermont Medical Center, Burlington, VT, USA
| | - Maryam Moin
- Department of Neurology, University of Illinois College of Medicine, Peoria, IL, USA
| | - Rup Tandan
- Department of Neurological Sciences, The University of Vermont and the University of Vermont Medical Center, Burlington, VT, USA
| |
Collapse
|
20
|
Lenti MV, Rossi CM, Melazzini F, Gastaldi M, Bugatti S, Rotondi M, Bianchi PI, Gentile A, Chiovato L, Montecucco C, Corazza GR, Di Sabatino A. Seronegative autoimmune diseases: A challenging diagnosis. Autoimmun Rev 2022; 21:103143. [PMID: 35840037 DOI: 10.1016/j.autrev.2022.103143] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/10/2022] [Indexed: 12/19/2022]
Abstract
Autoimmune diseases (AID) are increasingly prevalent conditions which comprise more than 100 distinct clinical entities that are responsible for a great disease burden worldwide. The early recognition of these diseases is key for preventing their complications and for tailoring proper management. In most cases, autoantibodies, regardless of their potential pathogenetic role, can be detected in the serum of patients with AID, helping clinicians in making a definitive diagnosis and allowing screening strategies for early -and sometimes pre-clinical- diagnosis. Despite their undoubted crucial role, in a minority of cases, patients with AID may not show any autoantibody, a condition that is referred to as seronegative AID. Suboptimal accuracy of the available laboratory tests, antibody absorption, immunosuppressive therapy, immunodeficiencies, antigen exhaustion, and immunosenescence are the main possible determinants of seronegative AID. Indeed, in seronegative AID, the diagnosis is more challenging and must rely on clinical features and on other available tests, often including histopathological evaluation and radiological diagnostic tests. In this review, we critically dissect, in a narrative fashion, the possible causes of seronegativity, as well as the diagnostic and management implications, in several AID including autoimmune gastritis, celiac disease, autoimmune liver disease, rheumatoid arthritis, autoimmune encephalitis, myasthenia gravis, Sjögren's syndrome, antiphospholipid syndrome, and autoimmune thyroid diseases.
Collapse
Affiliation(s)
- Marco Vincenzo Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Carlo Maria Rossi
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Federica Melazzini
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Matteo Gastaldi
- Neuroimmunology Laboratory, IRCCS Mondino Foundation, Pavia, Italy
| | - Serena Bugatti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Unit of Rheumatology, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Mario Rotondi
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia, Italy
| | - Paola Ilaria Bianchi
- Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Antonella Gentile
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Luca Chiovato
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Istituti Clinici Scientifici Maugeri IRCCS, Unit of Internal Medicine and Endocrinology, Laboratory for Endocrine Disruptors, Pavia, Italy
| | - Carlomaurizio Montecucco
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Unit of Rheumatology, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Gino Roberto Corazza
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia, Italy; Department of Internal Medicine, IRCCS San Matteo Hospital Foundation, Pavia, Italy.
| |
Collapse
|
21
|
Tan Y, Shi J, Huang Y, Li K, Yan J, Zhu L, Guan Y, Cui L. Long-Term Efficacy of Non-steroid Immunosuppressive Agents in Anti-Muscle-Specific Kinase Positive Myasthenia Gravis Patients: A Prospective Study. Front Neurol 2022; 13:877895. [PMID: 35775051 PMCID: PMC9237788 DOI: 10.3389/fneur.2022.877895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/25/2022] [Indexed: 11/27/2022] Open
Abstract
Background and Purpose Anti-muscle-specific kinase (MuSK) positive myasthenia gravis (MG) is characterized by a high relapsing rate, thus, choosing the appropriate oral drug regimen is a challenge. This study aimed to evaluate the efficacy of oral immunosuppressants (IS) in preventing relapse in MuSK-MG. Methods This prospective cohort observational study included patients with MuSK-MG at Peking Union Medical College Hospital between January 1, 2018, and November 15, 2021. The patients were divided into 2 groups: those with (IS+) or without (IS-) non-steroid immunosuppressive agents. The primary outcome was relapsed at follow-up, and the log-rank test was used to compare the proportion of maintenance-free relapse between the groups; hazard ratio (HR) was calculated using the Cox proportional hazards models. Results Fifty-three of 59 patients with MuSK-MG were included in the cohort, 14 were in the IS+ group, and 39 were in the IS- group. Twenty-four cases in the cohort experienced relapse at least once; the relapse rate was 2/14 (14.3%) in the IS+ group and 22/39 (56.4%) in the IS- group. At the end of follow-up, the proportion of maintenance-free relapse was significantly different between the two groups (log-rank χ2 = 4.94, P = 0.02). Of all the potential confounders, only the use of IS was associated with a reduced risk of relapse. The HR for relapse among patients in the IS+ group was 0.21 (95%CI 0.05-0.58) and was 0.23 (95%CI 0.05-0.93) in a model adjusted for age, sex, relapse history, highest Myasthenia Gravis Foundation of America (MGFA), and accumulated time of steroid therapy. Conclusions This study provides evidence that oral non-steroid immunosuppressive agents may be beneficial in reducing relapse in patients with MuSK-MG.
Collapse
Affiliation(s)
- Ying Tan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jiayu Shi
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yangyu Huang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ke Li
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingwen Yan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing, China
| | - Yuzhou Guan
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Liying Cui
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
22
|
Li Y, Yang S, Dong X, Li Z, Peng Y, Jin W, Chen D, Zhou R, Jiang F, Yan C, Yang H. Factors affecting minimal manifestation status induction in myasthenia gravis. Ther Adv Neurol Disord 2022; 15:17562864221080520. [PMID: 35371293 PMCID: PMC8968991 DOI: 10.1177/17562864221080520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 01/25/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Minimal manifestation status (MMS) is an important landmark in the treatment of myasthenia gravis (MG), and predictors of MMS induction have rarely been identified in previous studies. Objective: The objective of this study is to evaluate the clinical factors associated with MMS induction among patients with MG. Design: This two-step retrospective cohort study with a single center investigated the factors that may be associated with MMS induction and retested these predictors in a test cohort. Methods: A total of 388 diagnosed MG patients who visited Xiangya Hospital between 1 July 2015 and 1 July 2019 were involved. We performed detailed chart reviews and recorded all cases achieving MMS. Demographics and clinical characteristics were also collected and their relationships to achieving MMS were investigated. Results: MMS was achieved in 124 patients (50.2%), and the median time to achieve MMS was 26 months. Several factors were found to be associated with MMS induction in exploring cohort, including muscle-specific tyrosine-protein kinase receptor (MuSK) antibody positivity (adjusted hazard ratio, HR = 4.333, 95% confidence interval, CI: 1.862–10.082), isolated ocular involvement (adjusted HR = 1.95, 95% CI: 1.284–2.961), and low baseline quantitative myasthenia gravis score (QMG score; adjusted HR = 2.022, 95% CI: 1.086–3.764). These factors were then retested in the test cohort. Isolated ocular involvement or low baseline QMG scores were factors found to be beneficial for MMS induction were confirmed. Conclusion: Isolated ocular involvement and low baseline QMG score are predictors of MMS induction in MG patients.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Shumei Yang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Xiaohua Dong
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Zhibin Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Yuyao Peng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Wanlin Jin
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Di Chen
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Ran Zhou
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Fei Jiang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Chengkai Yan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, Hunan, P.R. China
| |
Collapse
|
23
|
Koneczny I, Tzartos J, Mané-Damas M, Yilmaz V, Huijbers MG, Lazaridis K, Höftberger R, Tüzün E, Martinez-Martinez P, Tzartos S, Leypoldt F. IgG4 Autoantibodies in Organ-Specific Autoimmunopathies: Reviewing Class Switching, Antibody-Producing Cells, and Specific Immunotherapies. Front Immunol 2022; 13:834342. [PMID: 35401530 PMCID: PMC8986991 DOI: 10.3389/fimmu.2022.834342] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/28/2022] [Indexed: 12/24/2022] Open
Abstract
Organ-specific autoimmunity is often characterized by autoantibodies targeting proteins expressed in the affected tissue. A subgroup of autoimmunopathies has recently emerged that is characterized by predominant autoantibodies of the IgG4 subclass (IgG4-autoimmune diseases; IgG4-AID). This group includes pemphigus vulgaris, thrombotic thrombocytopenic purpura, subtypes of autoimmune encephalitis, inflammatory neuropathies, myasthenia gravis and membranous nephropathy. Although the associated autoantibodies target specific antigens in different organs and thus cause diverse syndromes and diseases, they share surprising similarities in genetic predisposition, disease mechanisms, clinical course and response to therapies. IgG4-AID appear to be distinct from another group of rare immune diseases associated with IgG4, which are the IgG4-related diseases (IgG4-RLD), such as IgG4-related which have distinct clinical and serological properties and are not characterized by antigen-specific IgG4. Importantly, IgG4-AID differ significantly from diseases associated with IgG1 autoantibodies targeting the same organ. This may be due to the unique functional characteristics of IgG4 autoantibodies (e.g. anti-inflammatory and functionally monovalent) that affect how the antibodies cause disease, and the differential response to immunotherapies of the IgG4 producing B cells/plasmablasts. These clinical and pathophysiological clues give important insight in the immunopathogenesis of IgG4-AID. Understanding IgG4 immunobiology is a key step towards the development of novel, IgG4 specific treatments. In this review we therefore summarize current knowledge on IgG4 regulation, the relevance of class switching in the context of health and disease, describe the cellular mechanisms involved in IgG4 production and provide an overview of treatment responses in IgG4-AID.
Collapse
Affiliation(s)
- Inga Koneczny
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - John Tzartos
- Neuroimmunology, Tzartos NeuroDiagnostics, Athens, Greece
- 2nd Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marina Mané-Damas
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Vuslat Yilmaz
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Maartje G. Huijbers
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - Konstantinos Lazaridis
- Department of Immunology, Laboratory of Immunology, Hellenic Pasteur Institute, Athens, Greece
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Erdem Tüzün
- Department of Neuroscience, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Pilar Martinez-Martinez
- Research Group Neuroinflammation and Autoimmunity, Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Socrates Tzartos
- Neuroimmunology, Tzartos NeuroDiagnostics, Athens, Greece
- Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Frank Leypoldt
- Neuroimmunology, Institute of Clinical Chemistry and Department of Neurology, UKSH Kiel/Lübeck, Kiel University, Kiel, Germany
| |
Collapse
|
24
|
Huijbers MG, Marx A, Plomp JJ, Le Panse R, Phillips WD. Advances in the understanding of disease mechanisms of autoimmune neuromuscular junction disorders. Lancet Neurol 2022; 21:163-175. [DOI: 10.1016/s1474-4422(21)00357-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/15/2021] [Accepted: 10/06/2021] [Indexed: 01/19/2023]
|
25
|
Endmayr V, Tunc C, Ergin L, De Rosa A, Weng R, Wagner L, Yu TY, Fichtenbaum A, Perkmann T, Haslacher H, Kozakowski N, Schwaiger C, Ricken G, Hametner S, Klotz S, Dutra LA, Lechner C, de Simoni D, Poppert KN, Müller GJ, Pirker S, Pirker W, Angelovski A, Valach M, Maestri M, Guida M, Ricciardi R, Frommlet F, Sieghart D, Pinter M, Kircher K, Artacker G, Höftberger R, Koneczny I. Anti-Neuronal IgG4 Autoimmune Diseases and IgG4-Related Diseases May Not Be Part of the Same Spectrum: A Comparative Study. Front Immunol 2022; 12:785247. [PMID: 35095860 PMCID: PMC8795769 DOI: 10.3389/fimmu.2021.785247] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/15/2021] [Indexed: 12/18/2022] Open
Abstract
Background IgG4 is associated with two emerging groups of rare diseases: 1) IgG4 autoimmune diseases (IgG4-AID) and 2) IgG4-related diseases (IgG4-RLD). Anti-neuronal IgG4-AID include MuSK myasthenia gravis, LGI1- and Caspr2-encephalitis and autoimmune nodo-/paranodopathies (CNTN1/Caspr1 or NF155 antibodies). IgG4-RLD is a multiorgan disease hallmarked by tissue-destructive fibrotic lesions with lymphocyte and IgG4 plasma cell infiltrates and increased serum IgG4 concentrations. It is unclear whether IgG4-AID and IgG4-RLD share relevant clinical and immunopathological features. Methods We collected and analyzed clinical, serological, and histopathological data in 50 patients with anti-neuronal IgG4-AID and 19 patients with IgG4-RLD. Results A significantly higher proportion of IgG4-RLD patients had serum IgG4 elevation when compared to IgG4-AID patients (52.63% vs. 16%, p = .004). Moreover, those IgG4-AID patients with elevated IgG4 did not meet the diagnostic criteria of IgG4-RLD, and their autoantibody titers did not correlate with their serum IgG4 concentrations. In addition, patients with IgG4-RLD were negative for anti-neuronal/neuromuscular autoantibodies and among these patients, men showed a significantly higher propensity for IgG4 elevation, when compared to women (p = .005). Last, a kidney biopsy from a patient with autoimmune paranodopathy due to CNTN1/Caspr1-complex IgG4 autoantibodies and concomitant nephrotic syndrome did not show fibrosis or IgG4+ plasma cells, which are diagnostic hallmarks of IgG4-RLD. Conclusion Our observations suggest that anti-neuronal IgG4-AID and IgG4-RLD are most likely distinct disease entities.
Collapse
Affiliation(s)
- Verena Endmayr
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Cansu Tunc
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Lara Ergin
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Anna De Rosa
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | - Rosa Weng
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Lukas Wagner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thin-Yau Yu
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Andreas Fichtenbaum
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | | | - Carmen Schwaiger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Gerda Ricken
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Sigrid Klotz
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Lívia Almeida Dutra
- Department of Neurology and Neurosurgery, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Christian Lechner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
- Pediatric Neurology, Department of Pediatric and Adolescent Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Désirée de Simoni
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
- Department of Neurology, University Hospital St. Poelten, St. Poelten, Austria
| | - Kai-Nicolas Poppert
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Georg Johannes Müller
- Department of Neurology and Karl Landsteiner Institute for Neuroimmunological and Neurodegenerative Disorders, Klinik Donaustadt, Vienna, Austria
| | - Susanne Pirker
- Department of Neurology, Klinik Hietzing, Vienna, Austria
| | - Walter Pirker
- Department of Neurology, Klinik Ottakring, Vienna, Austria
| | | | - Matus Valach
- Department of Pathology, Klinik Landstrasse, Vienna, Austria
| | - Michelangelo Maestri
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | - Melania Guida
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | - Roberta Ricciardi
- Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | - Florian Frommlet
- Center for Medical Statistics, Informatics and Intelligent Systems, Section for Medical Statistics, Medical University of Vienna, Vienna, Austria
| | - Daniela Sieghart
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Miklos Pinter
- Wiener Privatklinik – Health Center, Vienna, Austria
| | - Karl Kircher
- Department of Ophthalmology, Medical University of Vienna, Vienna, Austria
| | - Gottfried Artacker
- Department of Pediatrics and Adolescent Medicine, Klinik Donaustadt, Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Inga Koneczny
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
26
|
Spontaneous Remission in Juvenile Myasthenia Gravis: A cohort of 13 cases and review of the literature. Neuromuscul Disord 2022; 32:213-219. [DOI: 10.1016/j.nmd.2021.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/28/2021] [Accepted: 11/29/2021] [Indexed: 11/30/2022]
|
27
|
Amor S, Nutma E, Marzin M, Puentes F. Imaging immunological processes from blood to brain in amyotrophic lateral sclerosis. Clin Exp Immunol 2021; 206:301-313. [PMID: 34510431 PMCID: PMC8561688 DOI: 10.1111/cei.13660] [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: 06/08/2021] [Revised: 08/18/2021] [Accepted: 08/29/2021] [Indexed: 12/12/2022] Open
Abstract
Neuropathology studies of amyotrophic lateral sclerosis (ALS) and animal models of ALS reveal a strong association between aberrant protein accumulation and motor neurone damage, as well as activated microglia and astrocytes. While the role of neuroinflammation in the pathology of ALS is unclear, imaging studies of the central nervous system (CNS) support the idea that innate immune activation occurs early in disease in both humans and rodent models of ALS. In addition, emerging studies also reveal changes in monocytes, macrophages and lymphocytes in peripheral blood as well as at the neuromuscular junction. To more clearly understand the association of neuroinflammation (innate and adaptive) with disease progression, the use of biomarkers and imaging modalities allow monitoring of immune parameters in the disease process. Such approaches are important for patient stratification, selection and inclusion in clinical trials, as well as to provide readouts of response to therapy. Here, we discuss the different imaging modalities, e.g. magnetic resonance imaging, magnetic resonance spectroscopy and positron emission tomography as well as other approaches, including biomarkers of inflammation in ALS, that aid the understanding of the underlying immune mechanisms associated with motor neurone degeneration in ALS.
Collapse
Affiliation(s)
- Sandra Amor
- Department of Pathology, Amsterdam UMC Location VUmc, Amsterdam, the Netherlands.,Department of Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Erik Nutma
- Department of Pathology, Amsterdam UMC Location VUmc, Amsterdam, the Netherlands
| | - Manuel Marzin
- Department of Pathology, Amsterdam UMC Location VUmc, Amsterdam, the Netherlands
| | - Fabiola Puentes
- Department of Neuroscience and Trauma, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
28
|
Gastaldi M, Scaranzin S, Businaro P, Mobilia E, Benedetti L, Pesce G, Franciotta D. Improving laboratory diagnostics in myasthenia gravis. Expert Rev Mol Diagn 2021; 21:579-590. [PMID: 33970749 DOI: 10.1080/14737159.2021.1927715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Myasthenia gravis (MG) is a prototypical autoimmune disease, characterized by pathogenic autoantibodies targeting structures of the neuromuscular junction. Radioimmunoprecipitation assays (RIPAs) represent the gold standard for their detection. However, new methods are emerging to complement, or overcome RIPAs, also with the perspective of eliminating the use of radioactive reagents.Areas covered: We discuss advances in laboratory methods, prompted especially by cell-based assays (CBAs), for the detection of the autoantibodies of MG diagnostics, above all those to the nicotinic acetylcholine receptor (AChR), muscle-specific kinase (MuSK), and low molecular-weight receptor-related low-density lipoprotein-4 (LRP4).Expert opinion: CBA technology makes AChRs aggregate on cell membranes, thus allowing to detect autoantibodies to clustered AChRs, with reduction of seronegative MG cases. The diagnostic relevance of RIPA/CBA-measurable LRP4 antibodies is still unclear, in Caucasian patients at least. Live CBAs for the detection of AChR, MuSK, and LRP4 antibodies might represent an alternative to RIPAs, but first require full validation. CBAs could be used as screening tests, limiting RIPAs for antibody quantification. To this end, ELISAs might be an alternative.Fixation procedures preserving enough degree of antigen conformationality could yield AChR and MuSK CBAs suitable for a wide use in clinical-chemistry laboratories.
Collapse
Affiliation(s)
- Matteo Gastaldi
- Neuroimmunology Laboratory, IRCCS Mondino Foundation, Pavia, Italy
| | - Silvia Scaranzin
- Neuroimmunology Laboratory, IRCCS Mondino Foundation, Pavia, Italy
| | - Pietro Businaro
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Emanuela Mobilia
- Autoimmunity Laboratory, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Luana Benedetti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Giampaola Pesce
- Autoimmunity Laboratory, IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Department of Internal Medicine (Dimi), University of Genova, Genova, Italy
| | - Diego Franciotta
- Autoimmunity Laboratory, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
29
|
Rousseff RT. Diagnosis of Myasthenia Gravis. J Clin Med 2021; 10:jcm10081736. [PMID: 33923771 PMCID: PMC8073361 DOI: 10.3390/jcm10081736] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 11/24/2022] Open
Abstract
The diagnosis of autoimmune Myasthenia Gravis (MG) remains clinical and rests on the history and physical findings of fatigable, fluctuating muscle weakness in a specific distribution. Ancillary bedside tests and laboratory methods help confirm the synaptic disorder, define its type and severity, classify MG according to the causative antibodies, and assess the effect of treatment objectively. We present an update on the tests used in the diagnosis and follow-up of MG and the suggested approach for their application.
Collapse
Affiliation(s)
- Rossen T. Rousseff
- Department of Neurology, Ibn-Sina Hospital, Sabah Health Area,, Kuwait City 13115, Kuwait; ; Tel.: +359-878-417-412
- Science and Research Institute, Medical University of Pleven, 5800 Pleven, Bulgaria
| |
Collapse
|
30
|
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: 117] [Impact Index Per Article: 23.4] [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
|