Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
J Clin Neurosci ; 119: 76-84, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37988976

RESUMEN

BACKGROUND: Increased morbidity in many patients with myasthenia gravis (MG) on long-term immunosuppression highlights the need for improved treatments. The aim of this study is to investigate the safety and efficacy of iscalimab (CFZ533), a fully human anti-CD40 monoclonal antibody, in patients with moderate-to-severe MG receiving standard-of-care (SoC) therapies. METHODS: In this double-blind, placebo-controlled phase 2 study, symptomatic patients (n = 44) despite SoC were randomized 1:1 to receive intravenous iscalimab (10 mg/kg; n = 22) or placebo (n = 22) every 4 weeks for 6 doses in total. Patients were followed up for 6 months after the last dose. The total duration of the study was 52 weeks. RESULTS: In total, 34 of 44 patients (77.3 %) completed the study. The primary endpoint, Quantitative MG score, did not change significantly between baseline and week 25 for iscalimab (median [90 % CI], -4.07 [-5.67, -2.47]) versus placebo (-2.93 [-4.53, -1.33]); however, non-thymectomized patients (n = 29) showed more favorable results (iscalimab, -4.35 [-6.07, -2.64] vs placebo, -2.26 [-4.16, -0.36]). A statistically significant difference between iscalimab and placebo groups was observed in MG Composite score (adjusted mean change: -4.19 [-6.67, -1.72]; p = 0.007) at week 13, and MG-Activities of Daily Living score (-1.93 [-3.24, -0.62]; p = 0.018) at week 21. Adverse events were comparable between the iscalimab (91 %) and placebo (96 %) groups. CONCLUSION: Iscalimab showed favorable safety and improvements compared with placebo in non-thymectomized patients with moderate-to-severe MG. It did not show any protective effect in patients with moderate-to-severe MG.


Asunto(s)
Actividades Cotidianas , Miastenia Gravis , Humanos , Resultado del Tratamiento , Anticuerpos Monoclonales/efectos adversos , Miastenia Gravis/tratamiento farmacológico , Miastenia Gravis/inducido químicamente , Método Doble Ciego
2.
JAMA Neurol ; 78(7): 834-841, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33999158

RESUMEN

Importance: This ongoing study assesses long-term safety and durability of response in infants with spinal muscular atrophy (SMA) type 1 after dosing with onasemnogene abeparvovec gene replacement therapy. Objective: The primary objective of this ongoing study is to assess safety. The secondary objective is to determine whether developmental milestones achieved in the START phase 1 clinical trial were maintained and new milestones gained. Design, Setting, and Participants: This study is an ongoing, observational, follow-up study for continuous safety monitoring for 15 years in patients from the START phase I study (conducted May 5, 2014, through December 15, 2017) at Nationwide Children's Hospital in Columbus, Ohio. Participants were symptomatic infants with SMA type 1 and 2 copies of SMN2 previously treated with an intravenous dose of onasemnogene abeparvovec (low dose, 6.7 × 1013 vg/kg; or therapeutic dose, 1.1 × 1014 vg/kg) in START. Thirteen of 15 original START patients are included in this analysis; 2 patients' families declined follow-up participation. Data were analyzed from September 21, 2017, to June 11, 2020. Exposures: Median time since dosing of 5.2 (range, 4.6-6.2) years; 5.9 (range, 5.8-6.2) years in the low-dose cohort and 4.8 (range, 4.6-5.6) years in the therapeutic-dose cohort. Main Outcomes and Measures: The primary outcome measure was the incidence of serious adverse events (SAEs). Results: At data cutoff on June 11, 2020, 13 patients treated in START were enrolled in this study (median age, 38.9 [range, 25.4-48.0] months; 7 females; low-dose cohort, n = 3; and therapeutic-dose cohort, n = 10). Serious adverse events occurred in 8 patients (62%), none of which resulted in study discontinuation or death. The most frequently reported SAEs were acute respiratory failure (n = 4 [31%]), pneumonia (n = 4 [31%]), dehydration (n = 3 [23%]), respiratory distress (n = 2 [15%]), and bronchiolitis (n = 2 [15%]). All 10 patients in the therapeutic-dose cohort remained alive and without the need for permanent ventilation. Prior to baseline, 4 patients (40%) in the therapeutic-dose cohort required noninvasive ventilatory support, and 6 patients (60%) did not require regular ventilatory support, which did not change in long-term follow-up. All 10 patients treated with the therapeutic dose maintained previously acquired motor milestones. Two patients attained the new milestone of "standing with assistance" without the use of nusinersen. Conclusions and Relevance: The findings of this ongoing clinical follow-up of patients with SMA type 1 treated with onasemnogene abeparvovec supports the long-term favorable safety profile up to 6 years of age and provides evidence for sustained clinical durability of the therapeutic dose. Trial Registration: ClinicalTrials.gov Identifier: NCT03421977.


Asunto(s)
Productos Biológicos/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Atrofias Musculares Espinales de la Infancia/tratamiento farmacológico , Niño , Preescolar , Femenino , Estudios de Seguimiento , Terapia Genética/métodos , Humanos , Masculino , Resultado del Tratamiento
3.
Lancet Neurol ; 17(12): 1043-1052, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30337273

RESUMEN

BACKGROUND: Spinal and bulbar muscular atrophy is an X-linked neuromuscular disease caused by CAG repeat expansion in the androgen receptor gene. Patients with this disease have low concentrations of insulin-like growth factor-1 (IGF-1), and studies of overexpression and administration of IGF-1 showed benefit in a transgenic model; thus the IGF-1 pathway presents as a potential treatment target. We assessed safety, tolerability, and preliminary efficacy of BVS857, an IGF-1 mimetic, in patients with spinal and bulbar muscular atrophy. METHODS: In this randomised, double-blind, placebo-controlled trial, we recruited patients from neuromuscular centres in Denmark (Copenhagen), Germany (Ulm), Italy (Padova), and three sites within the USA (Bethesda, MD; Irvine, CA; and Columbus, OH). Eligible patients were 18 years or older with a confirmed genetic diagnosis of spinal and bulbar muscular atrophy, were ambulatory, had symptomatic weakness, and had serum IGF-1 concentrations of 170 ng/mL or lower. Patients were randomly assigned (2:1) to study drug or placebo by a number scheme. Patients, investigators, and study personnel were masked to treatment assignment. After a safety and tolerability assessment with eight patients, BVS857 was administered once a week (0·06 mg/kg intravenously) for 12 weeks. Primary outcome measures were safety, tolerability, and the effects of BVS857 on thigh muscle volume (TMV) measured by MRI. The ratio of TMV at day 85 to baseline was analysed with ANCOVA per protocol. Secondary outcomes of muscle strength and function were measured with the Adult Myopathy Assessment Tool, lean body mass through dual energy x-ray absorptiometry, and BVS857 pharmacokinetics. This trial was registered with ClinicalTrials.gov, NCT02024932. FINDINGS: 31 patients were assessed for eligibility, 27 of whom were randomly assigned to either BVS857 treatment (n=18) or placebo (n=9), and 24 were included in the preliminary efficacy analysis (BVS857 group, n=15; placebo group, n=9). BVS857 was generally safe with no serious adverse events. No significant differences were found in adverse events between the BVS857 and placebo groups. Immunogenicity was detected in 13 (72%) of 18 patients in the BVS857 group, including crossreacting antibodies with neutralising capacity to endogenous IGF-1 in five patients. TMV decreased from baseline to day 85 in the placebo group (-3·4% [-110 cm3]) but not in the BVS857 group (0% [2 cm3]). A significant difference in change in TMV was observed in the BVS857 group versus the placebo group (geometric-mean ratio 1·04 [90% CI 1·01-1·07]; p=0·02). There were no differences between groups in measures of muscle strength and function. INTERPRETATION: TMV remained stable in patients with spinal and bulbar muscular atrophy after being given BVS857 for 12 weeks. The intervention was associated with high incidence of immunogenicity and did not improve muscle strength or function. Additional studies might be needed to assess the efficacy of activating the IGF-1 pathway in this disease. FUNDING: Novartis Pharmaceuticals and the US National Institutes of Health.


Asunto(s)
Atrofia Bulboespinal Ligada al X/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Atrofia Muscular/tratamiento farmacológico , Resultado del Tratamiento , Adulto , Anciano , Biomimética , Atrofia Bulboespinal Ligada al X/complicaciones , Atrofia Bulboespinal Ligada al X/diagnóstico por imagen , Estudios de Cohortes , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Cooperación Internacional , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Atrofia Muscular/complicaciones , Atrofia Muscular/diagnóstico por imagen
4.
J Neuromuscul Dis ; 4(2): 169-173, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28505981

RESUMEN

Monoclonal antibodies that target either PD-1 or PD-L1 have recently been approved for treatment of advanced non-small cell lung cancer. These antibodies are immune checkpoint inhibitors which have been shown to exacerbate Myasthenia Gravis (MG) and other autoimmune diseases. While effective in preventing tumor cells from evading immune attack, immune checkpoint inhibitors such as nivolumab, an antibody directed against the programmed cell death protein-1 (PD-1) receptor located on T-cells, may also cause immune dysregulation and could cause or potentiate pre-existing autoimmune conditions. We present a patient with latent ocular MG treated with nivolumab for her stage IV non-small cell lung cancer who developed generalized MG and severe myasthenic crisis. Providers must be aware of the risks inherent to these novel therapies since they can have life-threatening effects.


Asunto(s)
Antineoplásicos Inmunológicos/efectos adversos , Inmunoterapia/efectos adversos , Miastenia Gravis/inmunología , Nivolumab/efectos adversos , Anciano , Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/complicaciones , Carcinoma de Pulmón de Células no Pequeñas/terapia , Femenino , Humanos , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/terapia , Miastenia Gravis/complicaciones , Nivolumab/uso terapéutico
6.
Neurotherapeutics ; 13(1): 118-31, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26510558

RESUMEN

Myasthenia gravis (MG) is an autoimmune disease associated with the production of autoantibodies against 1) the skeletal muscle acetylcholine receptor; 2) muscle-specific kinase, a receptor tyrosine kinase critical for the maintenance of neuromuscular synapses; 3) low-density lipoprotein receptor-related protein 4, an important molecular binding partner for muscle-specific kinase; and 4) other muscle endplate proteins. In addition to the profile of autoantibodies, MG may be classified according the location of the affected muscles (ocular vs generalized), the age of symptom onset, and the nature of thymic pathology. Immunopathologic events leading to the production of autoantibodies differ in the various disease subtypes. Advances in our knowledge of the immunopathogenesis of the subtypes of MG will allow for directed utilization of the ever-growing repertoire of therapeutic agents that target distinct nodes in the immune pathway relevant to the initiation and maintenance of autoimmune disease. In this review, we examine the pathogenesis of MG subtypes, current treatment options, and emerging new treatments and therapeutic targets.


Asunto(s)
Miastenia Gravis/tratamiento farmacológico , Linfocitos B/efectos de los fármacos , Linfocitos B/fisiología , Inactivadores del Complemento/uso terapéutico , Humanos , Miastenia Gravis/inmunología , Miastenia Gravis/terapia , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/fisiología , Linfocitos T/efectos de los fármacos , Linfocitos T/fisiología , Investigación Biomédica Traslacional
8.
Calcif Tissue Int ; 96(3): 234-42, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25363509

RESUMEN

Skeletal muscle weakness is a leading cause of mobility disability in the elderly (sarcopenia), as a complication of acute or chronic illness (cachexia), and due to inherited or acquired muscle diseases (muscular dystrophies, myositides, etc.). As of now, there are no approved drugs that can reliably increase muscle strength and function. However, with our understanding of the regulation of myocyte signaling and homeostasis evolving rapidly, experimental treatments are now entering the clinic. We review the current status of clinical research in pharmacological therapies for muscle disorders.


Asunto(s)
Debilidad Muscular/tratamiento farmacológico , Debilidad Muscular/fisiopatología , Músculo Esquelético/fisiopatología , Humanos
9.
J Autoimmun ; 52: 64-73, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24388113

RESUMEN

Myasthenia gravis (MG) is an autoimmune disorder caused by target-specific pathogenic antibodies directed toward postsynaptic neuromuscular junction (NMJ) proteins, most commonly the skeletal muscle nicotinic acetylcholine receptor (AChR). In MG, high-affinity anti-AChR Abs binding to the NMJ lead to loss of functional AChRs, culminating in neuromuscular transmission failure and myasthenic symptoms. Intravenous immune globulin (IVIg) has broad therapeutic application in the treatment of a range of autoimmune diseases, including MG, although its mechanism of action is not clear. Recently, the anti-inflammatory and anti-autoimmune activities of IVIg have been attributed to the IgG Fc domains. Soluble immune aggregates bearing intact Fc fragments have been shown to be effective treatment for a number of autoimmune disorders in mice, and fully recombinant multimeric Fc molecules have been shown to be effective in treating collagen-induced arthritis, murine immune thrombocytopenic purpura, and experimental inflammatory neuritis. In this study, a murine model of MG (EAMG) was used to study the effectiveness of this novel recombinant polyvalent IgG2a Fc (M045) in treating established myasthenia, with a direct comparison to treatment with IVIg. M045 treatment had profound effects on the clinical course of EAMG, accompanied by down-modulation of pathogenic antibody responses. These effects were associated with reduced B cell activation and T cell proliferative responses to AChR, an expansion in the population of FoxP3(+) regulatory T cells, and enhanced production of suppressive cytokines, such as IL-10. Treatment was at least as effective as IVIg in suppressing EAMG, even at doses 25-30 fold lower. Multimeric Fc molecules offer the advantages of being recombinant, homogenous, available in unlimited quantity, free of risk from infection and effective at significantly reduced protein loads, and may represent a viable therapeutic alternative to polyclonal IVIg.


Asunto(s)
Linfocitos B/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Inmunoterapia/métodos , Miastenia Gravis Autoinmune Experimental/terapia , Linfocitos T Reguladores/efectos de los fármacos , Animales , Autoanticuerpos/sangre , Linfocitos B/inmunología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Humanos , Inmunidad Humoral/efectos de los fármacos , Fragmentos Fc de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Inmunoglobulinas Intravenosas/administración & dosificación , Interleucina-10/metabolismo , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Miastenia Gravis Autoinmune Experimental/inmunología , Receptores Colinérgicos/inmunología , Proteínas Recombinantes/genética , Linfocitos T Reguladores/inmunología
10.
Ann N Y Acad Sci ; 1274: 68-76, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23252899

RESUMEN

Forkhead box P3 (FOXP3) is a transcription factor necessary for the function of regulatory T cells (T(reg) cells). T(reg) cells maintain immune homeostasis and self-tolerance and play an important role in the prevention of autoimmune disease. Here, we discuss the role of T(reg) cells in the pathogenesis of myasthenia gravis (MG) and review evidence indicating that a significant defect in T(reg) cell in vitro suppressive function exists in MG patients, without an alteration in circulating frequency. This functional defect is associated with a reduced expression of key functional molecules, such as FOXP3 on isolated T(reg) cells, and appears to be more pronounced in immunosuppression-naive MG patients. In vitro administration of granulocyte macrophage-colony-stimulating factor (GM-CSF) enhanced the suppressive function of T(reg) cells and upregulated FOXP3 expression. These findings indicate a clinically relevant T(reg) cell-intrinsic defect in immune regulation in MG that may reveal a novel therapeutic target.


Asunto(s)
Miastenia Gravis/metabolismo , Linfocitos T Reguladores/metabolismo , Antígenos CD4/inmunología , Antígenos CD4/metabolismo , Factores de Transcripción Forkhead/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Tolerancia Inmunológica , Subunidad alfa del Receptor de Interleucina-2/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Miastenia Gravis/inmunología , Autotolerancia , Linfocitos T Reguladores/inmunología
12.
Clin Immunol ; 145(3): 209-23, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23110942

RESUMEN

Previous studies have reported alterations in numbers or function of regulatory T (Treg) cells in myasthenia gravis (MG) patients, but published results have been inconsistent, likely due to the isolation of heterogenous "Treg" populations. In this study, we used surface CD4, CD25(high), and CD127(low/-) expression to isolate a relatively pure population of Tregs, and established that there was no alteration in the relative numbers of Tregs within the peripheral T cell pool in MG patients. In vitro proliferation assays, however, demonstrated that Treg-mediated suppression of responder T (Tresp) cells was impaired in MG patients and was associated with a reduced expression of FOXP3 in isolated Tregs. Suppression of both polyclonal and AChR-activated Tresp cells from MG patients could be restored using Tregs isolated from healthy controls, indicating that the defect in immune regulation in MG is primarily localized to isolated Treg cells, and revealing a potential novel therapeutic target.


Asunto(s)
Miastenia Gravis/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Células Presentadoras de Antígenos/inmunología , Antígenos CD4/metabolismo , Estudios de Casos y Controles , Separación Celular , Citocinas/biosíntesis , Femenino , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Técnicas In Vitro , Interleucina-10/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Miastenia Gravis/genética , Miastenia Gravis/metabolismo , Receptores Colinérgicos/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Adulto Joven
13.
Muscle Nerve ; 46(3): 449-53, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22907239

RESUMEN

INTRODUCTION: In this study we describe a patient with a prolonged myasthenic crisis refractory to conventional immunomodulatory therapy who was treated with GM-CSF (granulocyte macrophage colony-stimulating factor, sargramostim). METHODS: T-regulatory cell (Treg) suppressive function and Foxp3 expression were evaluated before and after treatment with GM-CSF. RESULTS: Treatment with GM-CSF was associated with clinical improvement, expansion in the circulating numbers of Foxp3(+) cells, increase in Foxp3 expression levels in Tregs, early improvement in Treg suppressive capacity for AChR-α-induced T-cell proliferation, and subsequent enhancement in Treg suppression of polyclonal T-cell proliferation. CONCLUSION: Although definitive conclusions cannot be drawn from a single case, the correlation with similar findings in GM-CSF-treated animals with experimental autoimmune myasthenia gravis suggests further exploration of the effects of GM-CSF in myasthenia gravis should be studied in a clinical trial setting.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Miastenia Gravis/tratamiento farmacológico , Linfocitos T Reguladores/efectos de los fármacos , Anciano , Proliferación Celular/efectos de los fármacos , Factores de Transcripción Forkhead/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Masculino , Miastenia Gravis/inmunología , Miastenia Gravis/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Resultado del Tratamiento
14.
Expert Rev Clin Immunol ; 8(5): 427-38, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22882218

RESUMEN

Myasthenia gravis is an autoimmune disorder of the neuromuscular junction. A number of molecules, including ion channels and other proteins at the neuromuscular junction, may be targeted by autoantibodies leading to abnormal neuromuscular transmission. In approximately 85% of patients, autoantibodies, directed against the postsynaptic nicotinic acetylcholine receptor can be detected in the serum and confirm the diagnosis, but in general, do not precisely predict the degree of weakness or response to therapy. Antibodies to the muscle-specific tyrosine kinase are detected in approximately 50% of generalized myasthenia gravis patients who are seronegative for anti-acetylcholine receptor antibodies, and levels of anti-muscle-specific tyrosine kinase antibodies do appear to correlate with disease severity and treatment response. Antibodies to other muscle antigens may be found in the subsets of myasthenia gravis patients, potentially providing clinically useful diagnostic information, but their utility as relevant biomarkers (measures of disease state or response to treatment) is currently unclear.


Asunto(s)
Autoanticuerpos/sangre , Músculo Esquelético/inmunología , Miastenia Gravis/diagnóstico , Miastenia Gravis/inmunología , Receptores Nicotínicos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Humanos , Miastenia Gravis/sangre , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/inmunología
15.
Muscle Nerve ; 45(6): 909-17, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22581550

RESUMEN

The recommendations for clinical research standards published in 2000 by a task force of the Medical Scientific Advisory Board (MSAB) of the Myasthenia Gravis Foundation of America (MGFA) were largely successful in introducing greater uniformity in the recording and reporting of MG clinical trials. Recognizing that changes in clinical trial design and implementation may increase the likelihood that new therapies are developed for MG, the MGFA MSAB Task Force here presents updated recommendations for the design and implementation of clinical trials in MG, including (a) the use of a quantitative measure, such as the MG-Composite, that is weighted for clinical significance and incorporates patient reported outcomes; (b) consideration of nontrial strategies; and (c) development of biomarkers that support mechanistic studies of pharmacotherapies. The hope is that these updated task force recommendations will expedite the development and acceptance of more effective and less noxious therapies for MG.


Asunto(s)
Ensayos Clínicos como Asunto/normas , Miastenia Gravis/terapia , Proyectos de Investigación/normas , Comités Consultivos , Biomarcadores , Humanos , Evaluación de Resultado en la Atención de Salud , Sociedades Médicas , Estados Unidos
17.
J Neuroimmunol ; 240-241: 65-73, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22099723

RESUMEN

We and others have demonstrated the ability of granulocyte-macrophage colony-stimulating factor (GM-CSF) to suppress autoimmunity by increasing the number of CD4(+)CD25(+) regulatory T cells (Tregs). In the current study, we have explored the critical role of induced antigen specific Tregs in the therapeutic effects of GM-CSF in murine experimental autoimmune myasthenia gravis (EAMG). Specifically, we show that Tregs from GM-CSF treated EAMG mice (GM-CSF/AChR-induced-Tregs) adoptively transferred into animals with EAMG suppressed clinical disease more potently than equal numbers of Tregs from either GM-CSF untreated EAMG mice or healthy mice treated with GM-CSF. In addition, GM-CSF/AChR-induced-Tregs selectively suppressed antigen specific T cell proliferation induced by AChR relative to that induced by an irrelevant self antigen, (thyroglobulin) and failed to significantly alter T cell proliferation in response to an exogenous antigen (ovalbumin). These results are consistent with the hypothesized mechanism of action of GM-CSF involving the mobilization of tolerogenic dendritic cell precursors which, upon antigen (AChR) capture, suppress the anti-AChR immune response through the induction/expansion of AChR-specific Tregs.


Asunto(s)
Especificidad de Anticuerpos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Miastenia Gravis Autoinmune Experimental/inmunología , Receptores Colinérgicos/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/sangre , Proliferación Celular , Antagonistas Colinérgicos/uso terapéutico , Células Dendríticas/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Miastenia Gravis Autoinmune Experimental/tratamiento farmacológico , Miastenia Gravis Autoinmune Experimental/patología , Células Madre/inmunología , Linfocitos T Reguladores/trasplante
18.
J Hum Genet ; 56(8): 589-94, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21697856

RESUMEN

Emery-Dreifuss muscular dystrophy (EDMD) is a neuromuscular disorder exhibiting a cardiomyopathy with cardiac conduction defects. X-linked EDMD arises from mutations in the EMD gene, which encodes for a nuclear membrane protein termed emerin. In this study, we describe novel and recurrent EMD mutations identified in 18 probands and three carriers from a cohort of 255 North American patients referred for EDMD genetic mutation analysis. Eight of these mutations are novel including six frameshift mutations (p.D9GfsX24, p.F39SfsX17, p.R45KfsX16, p.F190YfsX19, p.R203PfsX34 and p.R204PfsX7) and two non-sense mutations (p.S143X, p.W200X). Our data augment the number of EMD mutations by 13.8%, equating to an increase of 5.2% in the total known EMD mutations and to an increase of 6.0% in the number of different mutations. Analysis of the exon distribution of mutations within the EMD gene, suggests a nonrandom distribution, with exon 2 as a hot spot. This phenomenon may be due to its high GC content, which at 60% is the most GC-rich exon in the EMD gene.


Asunto(s)
Exones/genética , Proteínas de la Membrana/genética , Distrofia Muscular de Emery-Dreifuss/genética , Mutación , Proteínas Nucleares/genética , Adolescente , Adulto , Niño , Preescolar , Codón sin Sentido , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Mutación del Sistema de Lectura , Humanos , Recién Nacido , Masculino , Persona de Mediana Edad , Mutagénesis Insercional , Mutación Missense , Eliminación de Secuencia , Adulto Joven
19.
Hum Mutat ; 32(2): 152-67, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20848652

RESUMEN

Mutations in LMNA cause a variety of diseases affecting striated muscle including autosomal Emery-Dreifuss muscular dystrophy (EDMD), LMNA-associated congenital muscular dystrophy (L-CMD), and limb-girdle muscular dystrophy type 1B (LGMD1B). Here, we describe novel and recurrent LMNA mutations identified in 50 patients from the United States and Canada, which is the first report of the distribution of LMNA mutations from a large cohort outside Europe. This augments the number of LMNA mutations known to cause EDMD by 16.5%, equating to an increase of 5.9% in the total known LMNA mutations. Eight patients presented with either p.R249W/Q or p.E358K mutations and an early onset EDMD phenotype: two mutations recently associated with L-CMD. Importantly, 15 mutations are novel and include eight missense mutations (p.R189P, p.F206L, p.S268P, p.S295P, p.E361K, p.G449D, p.L454P, and p.W467R), three splice site mutations (c.IVS4 + 1G>A, c.IVS6 - 2A>G, and c.IVS8 + 1G>A), one duplication/in frame insertion (p.R190dup), one deletion (p.Q355del), and two silent mutations (p.R119R and p.K270K). Analysis of 4 of our lamin A mutations showed that some caused nuclear deformations and lamin B redistribution in a mutation specific manner. Together, this study significantly augments the number of EDMD patients on the database and describes 15 novel mutations that underlie EDMD, which will contribute to establishing genotype-phenotype correlations.


Asunto(s)
Análisis Mutacional de ADN , Lamina Tipo A/genética , Distrofia Muscular de Emery-Dreifuss/genética , Secuencia de Aminoácidos , Animales , Canadá , Línea Celular , Humanos , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Alineación de Secuencia , Estados Unidos
20.
Exp Neurol ; 225(2): 320-7, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20637753

RESUMEN

Autoantibodies directed against the skeletal muscle acetylcholine receptor (AChR) play a critical role in the pathogenesis of the autoimmune disease, myasthenia gravis (MG). The pathogenic importance of anti-AChR antibodies is substantiated clinically by the often dramatic clinical improvement that follows removal of circulating antibodies utilizing extracorporeal plasma exchange (PE). Unfortunately, the effects of PE are non-specific as immunoglobulins (IgG) and other plasma proteins are removed in addition to anti-AChR IgG. In this study, we have successfully incorporated the AChR protein purified from Torpedo californicus into a Nanodisc (ND) membrane scaffold protein/phospholipid structure. We go on to demonstrate the effectiveness of this ND-AChR complex, administered intravenously, in the in vivo down-modulation of anti-AChR antibodies and subsequent amelioration of clinical disease in the experimental murine model of MG. These results provide proof-of-principle for the in vivo antigen-specific reduction of pathogenic anti-AChR antibodies utilizing ND-AChR particles. Further development of this strategy may provide an effective, antigen-specific, and readily accessible acute therapy for exacerbating MG or myasthenic crisis.


Asunto(s)
Autoanticuerpos/inmunología , Miastenia Gravis/inmunología , Receptores Colinérgicos/inmunología , Adsorción/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Músculo Esquelético/inmunología , Andamios del Tejido , Torpedo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA