Myasthenia Gravis and Related Disorders

[1]  A. Vincent,et al.  IgG-specific cell-based assay detects potentially pathogenic MuSK-Abs in seronegative MG , 2017, Neurology: Neuroimmunology & Neuroinflammation.

[2]  T. Keil,et al.  CD4+ FoxP3+ T regulatory cell subsets in myasthenia gravis patients. , 2017, Clinical immunology.

[3]  Qiang Sun,et al.  Regulatory B Cells in Seropositive Myasthenia Gravis versus Healthy Controls , 2017, Front. Neurol..

[4]  L. Hobson‐Webb,et al.  B10 Cell Frequencies and Suppressive Capacity in Myasthenia Gravis Are Associated with Disease Severity , 2017, Front. Neurol..

[5]  P. van Damme,et al.  IgG4 autoantibodies against muscle-specific kinase undergo Fab-arm exchange in myasthenia gravis patients. , 2017, Journal of autoimmunity.

[6]  F. Shi,et al.  Augmentation of Circulating Follicular Helper T Cells and Their Impact on Autoreactive B Cells in Myasthenia Gravis , 2016, The Journal of Immunology.

[7]  Jiayin Lu,et al.  Imbalance of circulating CD4+CXCR5+FOXP3+ Tfr-like cells and CD4+CXCR5+FOXP3− Tfh-like cells in myasthenia gravis , 2016, Neuroscience Letters.

[8]  R. Rojas-García,et al.  Clinical Characteristics of Patients With Double-Seronegative Myasthenia Gravis and Antibodies to Cortactin. , 2016, JAMA neurology.

[9]  B. Soliven,et al.  Impaired regulatory B cells in myasthenia gravis , 2016, Journal of Neuroimmunology.

[10]  A. Marx,et al.  Randomized Trial of Thymectomy in Myasthenia Gravis. , 2016, The New England journal of medicine.

[11]  M. Ticchioni,et al.  Therapeutic target of memory B cells depletion helps to tailor administration frequency of rituximab in myasthenia gravis , 2016, Journal of Neuroimmunology.

[12]  E. Zwet,et al.  Longitudinal epitope mapping in MuSK myasthenia gravis: implications for disease severity , 2016, Journal of Neuroimmunology.

[13]  J. Lemos,et al.  Clinical Utility of Acetylcholine Receptor Antibody Testing in Ocular Myasthenia Gravis. , 2015, JAMA neurology.

[14]  L. Mei,et al.  Flow Cytofluorimetric Analysis of Anti-LRP4 (LDL Receptor-Related Protein 4) Autoantibodies in Italian Patients with Myasthenia Gravis , 2015, PloS one.

[15]  K. Kleopa,et al.  MuSK autoantibodies in myasthenia gravis detected by cell based assay — A multinational study , 2015, Journal of Neuroimmunology.

[16]  J. Maessen,et al.  Robotic thymectomy in patients with myasthenia gravis: neurological and surgical outcomes. , 2015, European journal of cardio-thoracic surgery : official journal of the European Association for Cardio-thoracic Surgery.

[17]  Jacqueline A Palace,et al.  Clinical Features and Diagnostic Usefulness of Antibodies to Clustered Acetylcholine Receptors in the Diagnosis of Seronegative Myasthenia Gravis. , 2015, JAMA neurology.

[18]  V. Damato,et al.  Efficacy and safety of rituximab for myasthenia gravis: a systematic review and meta-analysis , 2015, Journal of Neurology.

[19]  J. Howard,et al.  Characterization of B cells in muscle-specific kinase antibody myasthenia gravis , 2015, Neurology: Neuroimmunology & Neuroinflammation.

[20]  D. Beeson,et al.  Collagen Q – A potential target for autoantibodies in myasthenia gravis , 2015, Journal of Neurological Sciences.

[21]  N. Gilhus,et al.  Repeated Acetylcholine Receptor Antibody-Concentrations and Association to Clinical Myasthenia Gravis Development , 2014, PloS one.

[22]  R. Balice-Gordon,et al.  Cortactin autoantibodies in myasthenia gravis. , 2014, Autoimmunity reviews.

[23]  K. Kleopa,et al.  A comprehensive analysis of the epidemiology and clinical characteristics of anti-LRP4 in myasthenia gravis. , 2014, Journal of autoimmunity.

[24]  P. Miossec,et al.  Th17 and regulatory T cell balance in autoimmune and inflammatory diseases. , 2014, Autoimmunity reviews.

[25]  Li Yang,et al.  Interleukin‐10 producing‐B cells and their association with responsiveness to rituximab in myasthenia gravis , 2014, Muscle & nerve.

[26]  R. Lewis,et al.  Autoantibodies to Agrin in Myasthenia Gravis Patients , 2014, PloS one.

[27]  A. Engeland,et al.  Myasthenia gravis epidemiology in a national cohort; combining multiple disease registries , 2014, Acta neurologica Scandinavica. Supplementum.

[28]  Amit Bar-Or,et al.  IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases , 2014, Nature.

[29]  B. Eymard,et al.  Antibodies to clustered acetylcholine receptor: expanding the phenotype , 2014, European journal of neurology.

[30]  G. Lauria,et al.  LRP4 antibodies in serum and CSF from amyotrophic lateral sclerosis patients , 2013, Annals of clinical and translational neurology.

[31]  H. Vrolijk,et al.  MuSK IgG4 autoantibodies cause myasthenia gravis by inhibiting binding between MuSK and Lrp4 , 2013, Proceedings of the National Academy of Sciences.

[32]  L. Mei,et al.  Antibodies against low-density lipoprotein receptor-related protein 4 induce myasthenia gravis. , 2013, The Journal of clinical investigation.

[33]  A. Vincent,et al.  MuSK Myasthenia Gravis IgG4 Disrupts the Interaction of LRP4 with MuSK but Both IgG4 and IgG1-3 Can Disperse Preformed Agrin-Independent AChR Clusters , 2013, PloS one.

[34]  M. Benatar,et al.  A randomized, double‐blind, placebo‐controlled phase II study of eculizumab in patients with refractory generalized myasthenia gravis , 2013, Muscle & nerve.

[35]  Gui-xian Zhao,et al.  Impaired regulatory function and enhanced intrathecal activation of B cells in neuromyelitis optica: distinct from multiple sclerosis , 2013, Multiple sclerosis.

[36]  F. Hiepe,et al.  Pathogenic Long-Lived Plasma Cells and Their Survival Niches in Autoimmunity, Malignancy, and Allergy , 2012, The Journal of Immunology.

[37]  J. Bussel,et al.  Defective regulatory B-cell compartment in patients with immune thrombocytopenia. , 2012, Blood.

[38]  R. Howard,et al.  Clinically biphasic myasthenia gravis with both AChR and MuSK antibodies , 2012, Journal of Neurology.

[39]  A. Vincent,et al.  Presence and pathogenic relevance of antibodies to clustered acetylcholine receptor in ocular and generalized myasthenia gravis. , 2012, Archives of neurology.

[40]  D. Lacomis,et al.  Characteristics of late-onset myasthenia gravis , 2012, Journal of Neurology.

[41]  D. Richman,et al.  Acute severe animal model of anti-muscle-specific kinase myasthenia: combined postsynaptic and presynaptic changes. , 2012, Archives of neurology.

[42]  A. Vincent,et al.  Passive and active immunization models of MuSK-Ab positive myasthenia: Electrophysiological evidence for pre and postsynaptic defects , 2012, Experimental Neurology.

[43]  J. Verschuuren,et al.  Muscle-specific kinase myasthenia gravis IgG4 autoantibodies cause severe neuromuscular junction dysfunction in mice. , 2012, Brain : a journal of neurology.

[44]  R. Lewis,et al.  Autoantibodies to lipoprotein-related protein 4 in patients with double-seronegative myasthenia gravis. , 2012, Archives of neurology.

[45]  B. Schoser,et al.  Anti-LRP4 autoantibodies in AChR- and MuSK-antibody-negative myasthenia gravis , 2012, Journal of Neurology.

[46]  A. Miyazawa,et al.  Antibodies against muscle-specific kinase impair both presynaptic and postsynaptic functions in a murine model of myasthenia gravis. , 2012, The American journal of pathology.

[47]  A. Mussi,et al.  PTPN22 and myasthenia gravis: Replication in an Italian population and meta-analysis of literature data , 2012, Neuromuscular Disorders.

[48]  J. Verschuuren,et al.  Long-lasting treatment effect of rituximab in MuSK myasthenia , 2012, Neurology.

[49]  J. Verschuuren,et al.  Lambert–Eaton myasthenic syndrome: from clinical characteristics to therapeutic strategies , 2011, The Lancet Neurology.

[50]  S. Jayawant,et al.  Juvenile Myasthenia Gravis: A Paediatric Perspective , 2011, Autoimmune diseases.

[51]  N. Singhal,et al.  Role of extracellular matrix proteins and their receptors in the development of the vertebrate neuromuscular junction , 2011, Developmental neurobiology.

[52]  J. Buckner,et al.  CD4+FOXP3+ T Regulatory Cells in Human Autoimmunity: More Than a Numbers Game , 2011, The Journal of Immunology.

[53]  J. G. van Dijk,et al.  Clinical Dutch-English Lambert-Eaton Myasthenic syndrome (LEMS) tumor association prediction score accurately predicts small-cell lung cancer in the LEMS. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[54]  Y. Yamanashi,et al.  Autoantibodies to low‐density lipoprotein receptor–related protein 4 in myasthenia gravis , 2011, Annals of neurology.

[55]  D. Campbell,et al.  Phenotypical and functional specialization of FOXP3+ regulatory T cells , 2011, Nature Reviews Immunology.

[56]  A. Marx,et al.  Thymoma and paraneoplastic myasthenia gravis , 2010, Autoimmunity.

[57]  A. Vincent,et al.  Diagnostic use of autoantibodies in myasthenia gravis , 2010, Autoimmunity.

[58]  D. Sanders,et al.  Immunosuppressive therapies in myasthenia gravis , 2010, Autoimmunity.

[59]  D. Hafler,et al.  FOXP3+ regulatory T cells in the human immune system , 2010, Nature Reviews Immunology.

[60]  M. Farrugia,et al.  The use of rituximab in myasthenia gravis and Lambert–Eaton myasthenic syndrome , 2010, Journal of Neurology, Neurosurgery & Psychiatry.

[61]  M. Pasnoor,et al.  Clinical findings in MuSK‐antibody positive myasthenia gravis: A U.S. experience , 2010, Muscle & nerve.

[62]  D. Isenberg,et al.  CD19(+)CD24(hi)CD38(hi) B cells exhibit regulatory capacity in healthy individuals but are functionally impaired in systemic Lupus Erythematosus patients. , 2010, Immunity.

[63]  A. Twijnstra,et al.  SOX antibodies in small-cell lung cancer and Lambert-Eaton myasthenic syndrome: frequency and relation with survival. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[64]  Hulun Li,et al.  Disequilibrium of T helper type 1, 2 and 17 cells and regulatory T cells during the development of experimental autoimmune myasthenia gravis , 2009, Immunology.

[65]  I. Sanz,et al.  Novel Human Transitional B Cell Populations Revealed by B Cell Depletion Therapy1 , 2009, The Journal of Immunology.

[66]  M. Marino,et al.  HLA class II allele analysis in MuSK-positive myasthenia gravis suggests a role for DQ5 , 2009, Neurology.

[67]  H. J. Schelhaas,et al.  The Lambert–Eaton myasthenic syndrome 1988–2008: A clinical picture in 97 patients , 2008, Journal of Neuroimmunology.

[68]  V. Scaioli,et al.  Two cases of thymoma-associated myasthenia gravis without antibodies to the acetylcholine receptor , 2008, Neuromuscular Disorders.

[69]  S. Servidei,et al.  Response to Therapy in Myasthenia Gravis with Anti‐MuSK Antibodies , 2008, Annals of the New York Academy of Sciences.

[70]  O. Gervásio,et al.  Anti‐MuSK patient antibodies disrupt the mouse neuromuscular junction , 2008, Annals of neurology.

[71]  A. Vincent,et al.  Myasthenia Gravis Seronegative for Acetylcholine Receptor Antibodies , 2008, Annals of the New York Academy of Sciences.

[72]  A. Vincent,et al.  Autoimmunizing Mechanisms in Thymoma and Thymus * , 2008, Annals of the New York Academy of Sciences.

[73]  P. Laforêt,et al.  Severe neonatal myasthenia due to maternal anti-MuSK antibodies , 2008, Neuromuscular Disorders.

[74]  A. Vincent,et al.  IgG1 antibodies to acetylcholine receptors in ‘seronegative’ myasthenia gravis† , 2008, Brain : a journal of neurology.

[75]  D. Gray,et al.  Not always the bad guys: B cells as regulators of autoimmune pathology , 2008, Nature Reviews Immunology.

[76]  J. Verschuuren,et al.  A TRANSIENT NEONATAL MYASTHENIC SYNDROME WITH ANTI-MUSK ANTIBODIES , 2008, Neurology.

[77]  A. Gure,et al.  SOX1 antibodies are markers of paraneoplastic Lambert–Eaton myasthenic syndrome , 2008, Neurology.

[78]  S. Fuchs,et al.  Ex Vivo Generated Regulatory T Cells Modulate Experimental Autoimmune Myasthenia Gravis1 , 2008, The Journal of Immunology.

[79]  Chuan-zhen Lu,et al.  Decrease of CD4(+)CD25(high)Foxp3(+) regulatory T cells and elevation of CD19(+)BAFF-R(+) B cells and soluble ICAM-1 in myasthenia gravis. , 2008, Clinical immunology.

[80]  R. Pirskanen,et al.  Two SNPs in the promoter region of the CTLA‐4 gene affect binding of transcription factors and are associated with human myasthenia gravis , 2007, Journal of internal medicine.

[81]  N. Gilhus,et al.  Polygenic disease associations in thymomatous myasthenia gravis. , 2007, Archives of neurology.

[82]  E. Rendina,et al.  Expression of autoimmune regulator gene (AIRE) and T regulatory cells in human thymomas , 2007, Clinical and experimental immunology.

[83]  X. Ke,et al.  An IRF8-binding promoter variant and AIRE control CHRNA1 promiscuous expression in thymus , 2007, Nature.

[84]  W. J. Hamilton,et al.  ANTI-MUSK ANTIBODY AFTER THYMECTOMY IN A PREVIOUSLY SEROPOSITIVE MYASTHENIC CHILD , 2007, Neurology.

[85]  R. Caspi,et al.  C57BL/6 Mice Genetically Deficient in IL-12/IL-23 and IFN-γ Are Susceptible to Experimental Autoimmune Myasthenia Gravis, Suggesting a Pathogenic Role of Non-Th1 Cells1 , 2007, The Journal of Immunology.

[86]  Rob C. G. van de Ven,et al.  Severely impaired neuromuscular synaptic transmission causes muscle weakness in the Cacna1a‐mutant mouse rolling Nagoya , 2007, The European journal of neuroscience.

[87]  S. Ikeda,et al.  Comparison of the histological and immunohistochemical features of the thymus in young- and elderly-onset myasthenia gravis without thymoma , 2007, Journal of Clinical Neuroscience.

[88]  M. Mizuno,et al.  The membrane attack pathway of complement drives pathology in passively induced experimental autoimmune myasthenia gravis in mice , 2006, Clinical and experimental immunology.

[89]  B. Prabhakar,et al.  Suppression of Experimental Autoimmune Myasthenia Gravis by Granulocyte-Macrophage Colony-Stimulating Factor Is Associated with an Expansion of FoxP3+ Regulatory T Cells1 , 2006, The Journal of Immunology.

[90]  Min Zhang,et al.  Clinical and serological study of myasthenia gravis in HuBei Province, China , 2006, Journal of Neurology, Neurosurgery & Psychiatry.

[91]  B. Lecky Transient neonatal Lambert–Eaton syndrome , 2006, Journal of Neurology, Neurosurgery & Psychiatry.

[92]  G. Pearse Normal Structure, Function and Histology of the Thymus , 2006, Toxicologic pathology.

[93]  A. Vincent,et al.  Strong association of MuSK antibody–positive myasthenia gravis and HLA-DR14-DQ5 , 2006, Neurology.

[94]  P. Matthews,et al.  MRI and clinical studies of facial and bulbar muscle involvement in MuSK antibody-associated myasthenia gravis. , 2006, Brain : a journal of neurology.

[95]  Y. Abe,et al.  Induction of myasthenia by immunization against muscle-specific kinase. , 2006, The Journal of clinical investigation.

[96]  A. Vincent,et al.  Thymus changes in anti-MuSK-positive and -negative myasthenia gravis , 2005, Neurology.

[97]  A. Marx,et al.  Pathology of thymic tumors. , 2005, Seminars in thoracic and cardiovascular surgery.

[98]  A. Marx,et al.  Fewer thymic changes in MuSK antibody‐positive than in MuSK antibody‐negative MG , 2005, Annals of neurology.

[99]  A. Vincent,et al.  Acetylcholine receptors loss and postsynaptic damage in MuSK antibody–positive myasthenia gravis , 2005, Annals of neurology.

[100]  A. Saoudi,et al.  Functional defect of regulatory CD4(+)CD25+ T cells in the thymus of patients with autoimmune myasthenia gravis. , 2005, Blood.

[101]  A. Rosenwald,et al.  Selective loss of regulatory T cells in thymomas , 2004, Annals of neurology.

[102]  B. Rees Smith,et al.  Muscle-specific receptor tyrosine kinase autoantibodies--a new immunoprecipitation assay. , 2004, Clinica chimica acta; international journal of clinical chemistry.

[103]  P. Christadoss,et al.  ICOS is essential for the development of experimental autoimmune myasthenia gravis , 2004, Journal of Neuroimmunology.

[104]  A. Vincent,et al.  Low frequency of MuSK antibody in generalized seronegative myasthenia gravis among Chinese , 2004, Neurology.

[105]  A. Vincent,et al.  Detection and characterization of MuSK antibodies in seronegative myasthenia gravis , 2004, Annals of neurology.

[106]  R. Pirskanen,et al.  Circulating CD4+CD25+ and CD4+CD25– T Cells in Myasthenia Gravis and in Relation to Thymectomy , 2004, Scandinavian journal of immunology.

[107]  P. Barzaghi,et al.  Inhibition of synapse assembly in mammalian muscle in vivo by RNA interference , 2004, EMBO reports.

[108]  S. Higgs,et al.  Genetic Evidence for Involvement of Classical Complement Pathway in Induction of Experimental Autoimmune Myasthenia Gravis 1 , 2003, The Journal of Immunology.

[109]  P. Tonali,et al.  Clinical correlates with anti-MuSK antibodies in generalized seronegative myasthenia gravis. , 2003, Brain : a journal of neurology.

[110]  P. Rothwell,et al.  Evidence of underdiagnosis of myasthenia gravis in older people , 2003, Journal of neurology, neurosurgery, and psychiatry.

[111]  A. Vincent,et al.  Clinical aspects of MuSK antibody positive seronegative MG , 2003, Neurology.

[112]  H. Putter,et al.  The epidemiology of myasthenia gravis, Lambert-Eaton myasthenic syndrome and their associated tumours in the northern part of the province of South Holland , 2003, Journal of Neurology.

[113]  M. Wadhwa,et al.  Anti‐cytokine autoantibodies in autoimmunity: preponderance of neutralizing autoantibodies against interferon‐alpha, interferon‐omega and interleukin‐12 in patients with thymoma and/or myasthenia gravis , 2003, Clinical and experimental immunology.

[114]  A. Vincent,et al.  Do titin and cytokine antibodies in MG patients predict thymoma or thymoma recurrence? , 2001, Neurology.

[115]  S. J. Wood,et al.  Safety factor at the neuromuscular junction , 2001, Progress in Neurobiology.

[116]  J. Palace,et al.  Myasthenia gravis , 2001, The Lancet.

[117]  P. Karachunski,et al.  Absence of IFN-γ or IL-12 Has Different Effects on Experimental Myasthenia Gravis in C57BL/6 Mice1 , 2000, The Journal of Immunology.

[118]  J. Lindstrom Acetylcholine receptors and myasthenia , 2000, Muscle & nerve.

[119]  P. Christadoss,et al.  Animal models of myasthenia gravis. , 2000, Clinical immunology.

[120]  M. Bunce,et al.  A susceptibility region for myasthenia gravis extending into the HLA-class I sector telomeric to HLA-C. , 1999, Human immunology.

[121]  A. Vincent,et al.  Monoclonal antibodies raised against human acetylcholine receptor bind to all five subunits of the fetal isoform , 1999, Journal of Neuroimmunology.

[122]  R. Van Ree,et al.  Normal human immunoglobulin G4 is bispecific: it has two different antigen‐combining sites , 1999, Immunology.

[123]  K. Iwasa,et al.  Calcium channel peptide can cause an autoimmune-mediated model of Lambert–Eaton myasthenic syndrome in rats , 1999, Journal of the Neurological Sciences.

[124]  A. Vincent,et al.  Plasma from human mothers of fetuses with severe arthrogryposis multiplex congenita causes deformities in mice. , 1999, Journal of Clinical Investigation.

[125]  P. Karachunski,et al.  Interleukin-4 deficiency facilitates development of experimental myasthenia gravis and precludes its prevention by nasal administration of CD4+ epitope sequences of the acetylcholine receptor , 1999, Journal of Neuroimmunology.

[126]  D. Beeson,et al.  Early‐onset myasthenia gravis: A recurring T‐cell epitope in the adult‐specific acetylcholine receptor ε subunit presented by the susceptibility allele HLA‐DR52a , 1999, Annals of neurology.

[127]  Zeng-Yu Wang,et al.  Myasthenia in SCID mice grafted with myasthenic patient lymphocytes , 1999, Neurology.

[128]  J. Howard,et al.  TCR-Vβ Usage in the Thymus and Blood of Myasthenia Gravis Patients☆ , 1998 .

[129]  P. Christadoss,et al.  The Th2 cytokine IL-4 is not required for the progression of antibody-dependent autoimmune myasthenia gravis. , 1998, Journal of immunology.

[130]  B. He,et al.  Differential requirements for CD28 and CD40 ligand in the induction of experimental autoimmune myasthenia gravis , 1998, Journal of Neuroimmunology.

[131]  N. Gilhus,et al.  The histomorphology of the thymus in late onset, non‐thymoma myasthenia gravis , 1998 .

[132]  J. Changeux,et al.  An acetylcholine receptor alpha subunit promoter confers intrathymic expression in transgenic mice. Implications for tolerance of a transgenic self-antigen and for autoreactivity in myasthenia gravis. , 1998, The Journal of clinical investigation.

[133]  H. Müller-Hermelink,et al.  Abnormal thymocyte development and generation of autoreactive T cells in mixed and cortical thymomas. , 1998, Laboratory investigation; a journal of technical methods and pathology.

[134]  Zeng-Yu Wang,et al.  CD4+ Epitope Spreading and Differential T Cell Recognition of Muscle Acetylcholine Receptor Subunits in Myasthenia Gravis a , 1998, Annals of the New York Academy of Sciences.

[135]  J. Oger,et al.  Rapsyn Antibodies in Myasthenia Gravis a , 1998, Annals of the New York Academy of Sciences.

[136]  Zeng-Yu Wang,et al.  T Cell Recognition of the Acetylcholine Receptor in Myasthenia Gravis a , 1998, Annals of the New York Academy of Sciences.

[137]  N. Gilhus,et al.  Titin transcripts in thymomas. , 1997, Annals of the New York Academy of Sciences.

[138]  P. Christadoss,et al.  Interferon γ (IFN-γ) Is Necessary for the Genesis of Acetylcholine Receptor–induced Clinical Experimental Autoimmune Myasthenia gravis in Mice , 1997, The Journal of experimental medicine.

[139]  K. Iwasa,et al.  Antibodies to synthetic peptides of the alA subunit of the voltage‐gated calcium channel in Lambert‐Eaton myasthenic syndrome , 1997, Neurology.

[140]  Jacqueline Palace,et al.  Incidence of serum anti-P/Q-type and anti-N-type calcium channel autoantibodies in the Lambert-Eaton myasthenic syndrome , 1997, Journal of the Neurological Sciences.

[141]  I. Kockum,et al.  TAP polymorphisms in Swedish myasthenia gravis patients. , 1997, Tissue antigens.

[142]  A. Vincent,et al.  A transfected human muscle cell line expressing the adult subtype of the human muscle acetylcholine receptor for diagnostic assays in myasthenia gravis , 1996, Neurology.

[143]  A. Vincent,et al.  Soluble complement receptor 1 (sCR1) protects against experimental autoimmune myasthenia gravis , 1996, Journal of Neuroimmunology.

[144]  P. Distefano,et al.  The Receptor Tyrosine Kinase MuSK Is Required for Neuromuscular Junction Formation In Vivo , 1996, Cell.

[145]  S. Nagataki,et al.  Antibodies to recombinant synaptotagmin and calcium channel subtypes in Lambert-Eaton myasthenic syndrome , 1995, Journal of the Neurological Sciences.

[146]  K. Mikoshiba,et al.  Thymomas express epitopes shared by the ryanodine receptor , 1995, Journal of Neuroimmunology.

[147]  P. Molenaar,et al.  Acetylcholine release in myasthenia gravis: Regulation at single end‐plate level , 1995, Annals of neurology.

[148]  S. Huson,et al.  Recurrent congenital arthrogryposis leading to a diagnosis of myasthenia gravis in an initially asymptomatic mother , 1995, Neuromuscular Disorders.

[149]  E. Rieber,et al.  Treatment of myasthenia gravis with anti‐CD4 antibody , 1994, Neurology.

[150]  F. Somnier Exacerbation of myasthenia gravis after removal of thymomas , 1994, Acta neurologica Scandinavica.

[151]  H. Garchon,et al.  Involvement of human muscle acetylcholine receptor alpha-subunit gene (CHRNA) in susceptibility to myasthenia gravis. , 1994, Proceedings of the National Academy of Sciences of the United States of America.

[152]  H. Garchon,et al.  Association of Km3 allotype with increased serum levels of autoantibodies against muscle acetylcholine receptor in myasthenia gravis , 1994, Journal of Neuroimmunology.

[153]  M. Shenoy,et al.  Major histocompatibility complex class II gene disruption prevents experimental autoimmune myasthenia gravis. , 1994, Journal of immunology.

[154]  J. Newsom-Davis,et al.  Heterogeneity of calcium channel autoantibodies detected using a small‐cell lung cancer line derived from a Lambert‐Eaton myasthenic syndrome patient , 1994, Neurology.

[155]  C Bona,et al.  Defining criteria for autoimmune diseases (Witebsky's postulates revisited) , 1993, Immunology today.

[156]  A. Drosos,et al.  D-penicillamine induced myasthenia gravis: clinical, serological and genetic findings. , 1993, Clinical and experimental rheumatology.

[157]  C. Aimé,et al.  Antibodies to thymic epithelial cells in myasthenia gravis , 1991, Journal of Neuroimmunology.

[158]  V. Lennon,et al.  Mouse T lymphocyte response to acetylcholine receptor determined by T cell receptor for antigen V beta gene products recognizing Mls-1a. , 1991, Journal of Immunology.

[159]  P Wordsworth,et al.  Critical role for the Val/Gly86 HLA-DR beta dimorphism in autoantigen presentation to human T cells. , 1991, Proceedings of the National Academy of Sciences of the United States of America.

[160]  E. M. Adler,et al.  Strategic location of calcium channels at transmitter release sites of frog neuromuscular synapses , 1990, Neuron.

[161]  J. Wolinsky,et al.  Immune studies in human immunodeficiency virus infection with myasthenia gravis , 1990, Neurology.

[162]  G. Biesecker,et al.  Inhibition of acute passive transfer experimental autoimmune myasthenia gravis with Fab antibody to complement C6. , 1989, Journal of immunology.

[163]  A. Engel,et al.  Lambert‐eaton myasthenic syndrome IgG depletes presynaptic membrane active zone particles by antigenic modulation , 1988, Annals of neurology.

[164]  N. Murray,et al.  The Lambert-Eaton myasthenic syndrome. A review of 50 cases. , 1988, Brain : a journal of neurology.

[165]  P. Whiting,et al.  Antibody Heterogeneity and Specificity in Myasthenia Gravis a , 1987, Annals of the New York Academy of Sciences.

[166]  A. Engel,et al.  Lambert‐Eaton myasthenic syndrome: II. Immunoelectron microscopy localization of IgG at the mouse motor end‐plate , 1987, Annals of neurology.

[167]  R. Aalberse,et al.  Inhibition of complement activation by IgG4 antibodies. , 1986, Clinical and experimental immunology.

[168]  P. Whiting,et al.  Myasthenia gravis , 1986, Neurology.

[169]  A. Engel,et al.  Passive transfer of Lambert-Eaton myasthenic syndrome with IgG from man to mouse depletes the presynaptic membrane active zones. , 1983, Proceedings of the National Academy of Sciences of the United States of America.

[170]  J. Newsom-Davis,et al.  Antibodies to motor nerve terminals: an electrophysiological study of a human myasthenic syndrome transferred to mouse. , 1983, The Journal of physiology.

[171]  A. Vincent,et al.  Acetylcholine receptor antibody and clinical response to thymectomy in myasthenia gravis , 1983, Neurology.

[172]  G. Scadding,et al.  Acetylcholine receptor antibody synthesis by thymic lymphocytes , 1981, Neurology.

[173]  P. Berman,et al.  Linkage between the frequency of muscular weakness and loci that regulate immune responsiveness in murine experimental myasthenia gravis , 1980, The Journal of experimental medicine.

[174]  A. Vincent,et al.  Clinical, pathological, HLA antigen and immunological evidence for disease heterogeneity in myasthenia gravis. , 1980, Brain : a journal of neurology.

[175]  A. Vincent,et al.  Anti-acetylcholine receptor antibodies. , 1980, Journal of neurology, neurosurgery, and psychiatry.

[176]  J. Lindstrom,et al.  Antigenic modulation and receptor loss in experimental autoimmune myasthenia gravis , 1979, Muscle & nerve.

[177]  J. Michelson,et al.  Myasthenic antibodies cross-link acetylcholine receptors to accelerate degradation. , 1978, The New England journal of medicine.

[178]  R. Ulevitch,et al.  Role of complement in the pathogenesis of experimental autoimmune myasthenia gravis , 1978, The Journal of experimental medicine.

[179]  C. Hawkey,et al.  [Plasmapheresis for myasthenia gravis]. , 1978, The New England journal of medicine.

[180]  G. Scadding,et al.  IN-VITRO SYNTHESIS OF ANTI-ACETYLCHOLINE-RECEPTOR ANTIBODY BY THYMIC LYMPHOCYTES IN MYASTHENIA GRAVIS , 1978, The Lancet.

[181]  A. Engel,et al.  ULTRASTRUCTURAL LOCALIZATION OF IMMUNE COMPLEXES (IgG and C3) AT THE END‐PLATE IN EXPERIMENTAL AUTOMMUNE MYASTHENIA GRAVIS , 1977, Journal of neuropathology and experimental neurology.

[182]  A. Engel,et al.  Immune complexes (IgG and C3) at the motor end-plate in myasthenia gravis: ultrastructural and light microscopic localization and electrophysiologic correlations. , 1977, Mayo Clinic proceedings.

[183]  J. Lindstrom,et al.  Antibody to acetylcholine receptor in myasthenia gravis , 1976, Neurology.

[184]  A. Pestronk,et al.  Myasthenia gravis: passive transfer from man to mouse , 1975, Science.

[185]  D. Drachman,et al.  Neuromuscular Junction in Myasthenia Gravis: Decreased Acetylcholine Receptors , 1973, Science.

[186]  J. Patrick,et al.  Autoimmune Response to Acetylcholine Receptor , 1973, Science.

[187]  W. W. Hofmann,et al.  An electrophysiological investigation of neuro‐muscular transmission in myasthenia gravis , 1964, The Journal of physiology.

[188]  J. Simpson Myasthenia Gravis: A New Hypothesis , 1960 .

[189]  M. Walker TREATMENT OF MYASTHENIA GRAVIS WITH PHYSOSTIGMINE , 1934 .

[190]  J. Goldstein,et al.  Durability of the Rituximab Response in Acetylcholine Receptor Autoantibody–Positive Myasthenia Gravis , 2017, JAMA neurology.

[191]  G. Plant,et al.  Ocular Myasthenia Gravis: Controversies and Updates , 2013, Current Neurology and Neuroscience Reports.

[192]  A. Mamalaki,et al.  The main immunogenic region (MIR) of the nicotinic acetylcholine receptor and the anti-MIR antibodies , 2008, Molecular Neurobiology.

[193]  A. Marx,et al.  Immunopathology and Infectious Disease Myasthenia Gravis Thymus Complement Vulnerability of Epithelial and Myoid Cells , Complement Attack on Them , and Correlations with Autoantibody Status , 2007 .

[194]  R. Pirskanen,et al.  Different HLA DR-DQ associations in subgroups of idiopathic myasthenia gravis , 2005, Immunogenetics.

[195]  H. Inoko,et al.  Combinations of HLA-DPB1 and HLA-DQB1 alleles determine susceptibility to early-onset myasthenia gravis in Japan. , 1994, Autoimmunity.

[196]  S. Gammeltoft,et al.  Acetylcholine receptor antibody in myasthenia gravis: predominance of IgG subclasses 1 and 3. , 1987, Clinical and experimental immunology.

[197]  H. Müller-Hermelink,et al.  Immunohistological patterns of non-neoplastic changes in the thymus in Myasthenia gravis , 1986, Virchows Archiv. B, Cell pathology including molecular pathology.

[198]  N. Murray,et al.  Lambert-Eaton myasthenic syndrome: electrophysiological evidence for a humoral factor. , 1982, Muscle & nerve.

[199]  C. Cardwell,et al.  A systematic review of population based epidemiological studies in Myasthenia Gravis , 2010, BMC neurology.