Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 206
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Dysphagia ; 37(3): 473-487, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34226958

RESUMO

Autoimmune neurogenic dysphagia refers to manifestation of dysphagia due to autoimmune diseases affecting muscle, neuromuscular junction, nerves, roots, brainstem, or cortex. Dysphagia is either part of the evolving clinical symptomatology of an underlying neurological autoimmunity or occurs as a sole manifestation, acutely or insidiously. This opinion article reviews the autoimmune neurological causes of dysphagia, highlights clinical clues and laboratory testing that facilitate early diagnosis, especially when dysphagia is the presenting symptom, and outlines the most effective immunotherapeutic approaches. Dysphagia is common in inflammatory myopathies, most prominently in inclusion body myositis, and is frequent in myasthenia gravis, occurring early in bulbar-onset disease or during the course of progressive, generalized disease. Acute-onset dysphagia is often seen in Guillain-Barre syndrome variants and slowly progressive dysphagia in paraneoplastic neuropathies highlighted by the presence of specific autoantibodies. The most common causes of CNS autoimmune dysphagia are demyelinating and inflammatory lesions in the brainstem, occurring in patients with multiple sclerosis and neuromyelitis optica spectrum disorders. Less common, but often overlooked, is dysphagia in stiff-person syndrome especially in conjunction with cerebellar ataxia and high anti-GAD autoantibodies, and in gastrointestinal dysmotility syndromes associated with autoantibodies against the ganglionic acetyl-choline receptor. In the setting of many neurological autoimmunities, acute-onset or progressive dysphagia is a potentially treatable condition, requiring increased awareness for prompt diagnosis and early immunotherapy initiation.


Assuntos
Transtornos de Deglutição , Síndrome de Guillain-Barré , Esclerose Múltipla , Autoanticorpos , Transtornos de Deglutição/etiologia , Humanos
2.
Curr Opin Neurol ; 33(5): 545-552, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32833750

RESUMO

PURPOSE OF REVIEW: To provide an update on immunomodulating and immunosuppressive therapies in myasthenia gravis and highlight newly approved, or pending approval, therapies with new biologics. RECENT FINDINGS: Preoperative IVIg is not needed to prevent myasthenic crisis in stable myasthenia gravis patients scheduled for surgery under general anesthesia, based on controlled data. Rituximab, if initiated early in new-onset myasthenia gravis, can lead to faster and more sustained remission even without immunotherapies in 35% of patients at 2 years. Biomarkers determining the timing for follow-up infusions in Rituximab-responding AChR-positive patients are discussed. Most patients with MuSK-positive myasthenia gravis treated with Rituximab have sustained long-term remission with persistent reduction of IgG4 anti-MuSK antibodies. Eculizumb in the extension REGAIN study showed sustained long-term pharmacological remissions and reduced exacerbations. Three new biologic agents showed promising results in phase-II controlled myasthenia gravis trials: Zilucoplan, a subcutaneous macrocyclic peptide inhibiting complement C5; Efgartigimod, an IgG1-derived Fc fragment binding to neonatal FcRn receptor; and Rozanolixizumab, a high-affinity anti-FcRn monoclonal antibody. Finally, the safety of ongoing myasthenia gravis immunotherapies during COVID19 pandemic is discussed. SUMMARY: New biologics against B cells, complement and FcRn receptor, are bringing us closer to successful targeted immunotherapies in the chronic management of myasthenia gravis promising an exciting future for antibody-mediated neurological diseases.


Assuntos
Fatores Imunológicos/uso terapêutico , Imunoterapia/métodos , Miastenia Gravis/tratamento farmacológico , Autoanticorpos/imunologia , COVID-19 , Humanos , Fatores Imunológicos/efeitos adversos , Imunoterapia/efeitos adversos , Miastenia Gravis/imunologia , SARS-CoV-2
3.
J Autoimmun ; 104: 102339, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31611142

RESUMO

Autoimmune encephalitides, with an estimated incidence of 1.5 per million population per year, although described only 15 years ago, have already had a remarkable impact in neurology and paved the field to autoimmune neuropsychiatry. Many patients traditionally presented with aberrant behavior, especially of acute or subacute onset, and treated with anti-psychotic therapies, turn out to have a CNS autoimmune disease with pathogenic autoantibodies against synaptic antigens responding to immunotherapies. The review describes the clinical spectrum of these disorders, and the pathogenetic role of key autoantibodies directed against: a) cell surface synaptic antigens and receptors, including NMDAR, GABAa, GABAb, AMPA and glycine receptors; b) channels such as AQP4 water-permeable channel or voltage-gated potassium channels; c) proteins that stabilize voltage-gated potassium channel complex into the membrane, like the LGI1 and CASPR2; and d) enzymes that catalyze the formation of neurotransmitters such as Glutamic Acid Decarboxylase (GAD). These antibodies, effectively target excitatory or inhibitory synapses in the limbic system, basal ganglia or brainstem altering synaptic function and resulting in uncontrolled neurological excitability disorder clinically manifested with psychosis, agitation, behavioral alterations, depression, sleep disturbances, seizure-like phenomena, movement disorders such as ataxia, chorea and dystonia, memory changes or coma. Some of the identified triggering factors include: viruses, especially herpes simplex, accounting for the majority of relapses occurring after viral encephalitis, which respond to immunotherapy rather than antiviral agents; tumors especially teratoma, SCLC and thymomas; and biological cancer therapies (immune-check-point inhibitors). As anti-synaptic antibodies persist after viral infections or tumor removal, augmentation of autoreactive B cells which release autoantigens to draining lymph nodes, molecular mimicry and infection-induced bystander immune activation products play a role in autoimmunization process or perpetuating autoimmune neuroinflammation. The review stresses the importance of early detection, clinical recognition, proper antibody testing and early therapy initiation as these disorders, regardless of a known or not trigger, are potentially treatable responding to systemic immunotherapy with intravenous steroids, IVIg, rituximab or even bortezomid.


Assuntos
Bortezomib/uso terapêutico , Encefalite/imunologia , Encefalite/terapia , Doença de Hashimoto/imunologia , Doença de Hashimoto/terapia , Imunoglobulinas Intravenosas/uso terapêutico , Imunoterapia , Rituximab/uso terapêutico , Esteroides/uso terapêutico , Autoanticorpos/imunologia , Encefalite/epidemiologia , Encefalite/patologia , Doença de Hashimoto/epidemiologia , Doença de Hashimoto/patologia , Humanos , Incidência , Sinapses/imunologia
4.
BMC Neurol ; 19(1): 1, 2019 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-30606131

RESUMO

BACKGROUND: Stiff Person Syndrome (SPS) is an under-diagnosed disorder that affects mobility and the quality of life of affected patients. The aim of the study is to describe the natural history of SPS, the extent of accumulated disability and the associated clinical and immunological features in patients followed for up to 8 years in a single center. METHODS: Our collective cohort included 57 SPS patients. Additionally, 32 of these patients were examined every 6 months for a two-year period in a longitudinal study protocol, to assess disease progression using quantitative measures of stiffness and heightened sensitivity. RESULTS: The most frequent initial symptom was leg stiffness, followed by paraspinal muscle rigidity and painful spasms in 95% of the patients. Although none of the patients required assistance for ambulation during the first 2 years of disease onset, 46 patients (80%) lost the ability to walk independently during our follow-up, despite symptomatic medications. In the longitudinal cohort, the number of stiff areas increased (p < 0.0001), consistent with worsening functional status and quality of life. High-titer anti-GAD antibodies were present in serum and CSF with elevated intrathecal GAD-specific IgG synthesis, but they did not correlate with clinical severity or progression. CONCLUSIONS: This large study on SPS patients, combining an eight-year follow-up at a single center by the same leading neurologist and his team, is the first to provide longitudinal data in a large patient subgroup using objective clinical measures. One of the main findings is that SPS is a progressive disease leading to physical disability over time.


Assuntos
Rigidez Muscular Espasmódica , Estudos de Coortes , Progressão da Doença , Humanos , Qualidade de Vida
5.
Ann Neurol ; 82(2): 271-277, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28749549

RESUMO

OBJECTIVE: In stiff person syndrome (SPS), an antibody-mediated impaired γ-aminobutyric acidergic (GABAergic) neurotransmission is believed to cause muscle stiffness and spasms. Most patients improve with GABA-enhancing drugs and intravenous immunoglobulin, but some respond poorly and remain disabled. The need for more effective therapy prompted a trial with the anti-CD20 monoclonal antibody rituximab. METHODS: This was a placebo-controlled randomized trial of rituximab (2 biweekly infusions of 1g each). The primary outcome was a change in stiffness scores at 6 months. Secondary outcomes were changes in heightened-sensitivity and quality of life scores. Enrolling 24 patients was calculated to detect 50% change in stiffness scores. RESULTS: Randomization was balanced for age, sex, disease duration, and glutamic acid decarboxylase autoantibody titers. No significant changes were noted at 6 months after treatment in all outcomes. Specifically, no differences were noted in the stiffness index, the primary outcome, or sensitivity scores, the secondary outcome, at 3 or 6 months. Quality of life scores improved significantly (p < 0.01) at 3 months in both groups, but not at 6 months, denoting an early placebo effect. Blinded self-assessment rating of the overall stiffness for individual patients revealed improvement in 4 patients in each group. At 6 months, improvement persisted in 1 patient in the placebo group versus 3 of 4 in the rituximab group, where these meaningful improvements were also captured by video recordings. INTERPRETATION: This is the largest controlled trial conducted in SPS patients and demonstrates no statistically significant difference in the efficacy measures between rituximab and placebo. Rituximab's lack of efficacy could be due to a considerable placebo effect; insensitivity of scales to quantify stiffness, especially in the less severely affected patients; or drug effectiveness in only a small patient subset. Ann Neurol 2017;82:271-277.


Assuntos
Rituximab/uso terapêutico , Rigidez Muscular Espasmódica/tratamento farmacológico , Autoanticorpos/sangue , Método Duplo-Cego , Feminino , Glutamato Descarboxilase/imunologia , Humanos , Fatores Imunológicos/uso terapêutico , Masculino , Pessoa de Meia-Idade , Rigidez Muscular Espasmódica/sangue , Resultado do Tratamento
7.
Biochim Biophys Acta ; 1852(4): 658-66, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24949885

RESUMO

Autoimmune neuropathies occur when immunologic tolerance to myelin or axonal antigens is lost. Even though the triggering factors and the underling immunopathology have not been fully elucidated in all neuropathy subsets, immunological studies on the patients' nerves, transfer experiments with the patients' serum or intraneural injections, and molecular fingerprinting on circulating autoantibodies or autoreactive T cells, indicate that cellular and humoral factors, either independently or in concert with each other, play a fundamental role in their cause. The review is focused on the main subtypes of autoimmune neuropathies, mainly the Guillain-Barré syndrome(s), the Chronic Inflammatory Demyelinating Polyneuropathy (CIDP), the Multifocal Motor Neuropathy (MMN), and the IgM anti-MAG-antibody mediated neuropathy. It addresses the factors associated with breaking tolerance, examines the T cell activation process including co-stimulatory molecules and key cytokines, and discusses the role of antibodies against peripheral nerve glycolipids or glycoproteins. Special attention is given to the newly identified proteins in the nodal, paranodal and juxtaparanodal regions as potential antigenic targets that could best explain conduction failure and rapid recovery. New biological agents against T cells, cytokines, B cells, transmigration and transduction molecules involved in their immunopathologic network, are discussed as future therapeutic options in difficult cases. This article is part of a Special Issue entitled: Neuromuscular Diseases: Pathology and Molecular Pathogenesis.


Assuntos
Síndrome de Guillain-Barré , Doença dos Neurônios Motores , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica , Autoanticorpos/imunologia , Síndrome de Guillain-Barré/imunologia , Síndrome de Guillain-Barré/patologia , Humanos , Doença dos Neurônios Motores/imunologia , Doença dos Neurônios Motores/patologia , Proteínas do Tecido Nervoso/imunologia , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/imunologia , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/patologia , Linfócitos T/imunologia , Linfócitos T/patologia
8.
BMC Neurol ; 16: 48, 2016 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-27083892

RESUMO

BACKGROUND: Mechanisms of inflammation and protein accumulation are crucial in inclusion body myositis (IBM). Recent evidence demonstrated that intravenous immunoglobulin failed to suppress cell-stress mediators in IBM. Here we studied the molecular changes in skeletal muscle biopsies from patients with IBM before and after treatment with alemtuzumab. METHODS: Relevant inflammatory and degeneration-associated markers were assessed by quantitative-PCR and immunohistochemistry in repeated muscle biopsy specimens from patients with IBM, which had been treated in a previously published uncontrolled proof-of-concept trial with alemtuzumab. RESULTS: There were no significant changes of the mRNA expression levels of the pro-inflammatory chemokines CXCL-9, CCL-4, and the cytokines IFN-γ, TGF-ß, TNF-α, and IL-1ß. Similarly, the degeneration-associated molecules ubiquitin, APP and αB-crystallin did not substantially change. Although no overall beneficial treatment effect was noted except for a 6-month stabilization, some patients experienced a transient improvement in muscle strength. In such responders, a trend towards reduced expression of inflammatory markers was noted. In contrast, the expression remained unchanged in the others who did not experience any change. The expression levels of IL-1ß and MHC-I correlated with the positive clinical effect. By immunohistochemistry, some inflammatory mediators like CD8, CXCL-9, and MHC-I were downmodulated. However, no consistent changes were noted for ubiquitin, nitrotyrosin and ß-amyloid. CONCLUSIONS: Alemtuzumab showed a trend towards downregulation of the expression of some inflammatory molecules in skeletal muscle of IBM patients but has no effect on several crucial markers of cell stress and degeneration. The data are helpful to explain the molecular treatment effects of future lymphocyte-targeted immunotherapies in IBM.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Inflamação/patologia , Miosite de Corpos de Inclusão/tratamento farmacológico , Alemtuzumab , Biópsia , Citocinas/metabolismo , Humanos , Imuno-Histoquímica , Imunoterapia/métodos , Força Muscular/efeitos dos fármacos , Músculo Esquelético/metabolismo
9.
Muscle Nerve ; 52(4): 498-502, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25728021

RESUMO

INTRODUCTION: Electrodiagnostic studies (EDX) are not performed routinely before treatment suspension in CIDP, and no data exist regarding their value in predicting clinical relapse. METHODS: Serial EDX (baseline and after IGIV-C therapy) were analyzed from subjects in the ICE clinical trial who responded to IGIV-C treatment and were subsequently re-randomized to placebo in an extension phase. Comparisons were made between subjects who relapsed and those who did not. RESULTS: A total of 55% (6/11) of the Relapse group had an increase in total number of demyelinating findings (DF) versus 8% (1/13) in the No Relapse group (P = 0.023). In the Relapse group, 100% had ≥ 1 new DF and 73% (8/11) had ≥ 4 new DF versus 60% (8/13) and 8% (1/13), respectively, in the No Relapse group. CONCLUSIONS: An increased total number of DF or the occurrence of ≥ 4 new DF may indicate a higher risk of clinical relapse after treatment cessation in IGIV-C-responsive patients.


Assuntos
Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Condução Nervosa/efeitos dos fármacos , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/terapia , Resultado do Tratamento , Potenciais de Ação/efeitos dos fármacos , Adulto , Idoso , Eletrodiagnóstico , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Condução Nervosa/fisiologia , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/fisiopatologia , Tempo de Reação , Recidiva
10.
Cochrane Database Syst Rev ; 7: CD001555, 2015 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-35658164

RESUMO

BACKGROUND: Inclusion body myositis (IBM) is a late-onset inflammatory muscle disease (myopathy) associated with progressive proximal and distal limb muscle atrophy and weakness. Treatment options have attempted to target inflammatory and atrophic features of this condition (for example with immunosuppressive and immunomodulating drugs, anabolic steroids, and antioxidant treatments), although as yet there is no known effective treatment for reversing or minimising the progression of inclusion body myositis. In this review we have considered the benefits, adverse effects, and costs of treatment in targeting cardinal effects of the condition, namely muscle atrophy, weakness, and functional impairment. OBJECTIVES: To assess the effects of treatment for IBM. SEARCH METHODS: On 7 October 2014 we searched the Cochrane Neuromuscular Disease Group Specialized Register, the Cochrane Central Register for Controlled Trials (CENTRAL), MEDLINE, and EMBASE. Additionally in November 2014 we searched clinical trials registries for ongoing or completed but unpublished trials. SELECTION CRITERIA: We considered randomised or quasi-randomised trials, including cross-over trials, of treatment for IBM in adults compared to placebo or any other treatment for inclusion in the review. We specifically excluded people with familial IBM and hereditary inclusion body myopathy, but we included people who had connective tissue and autoimmune diseases associated with IBM, which may or may not be identified in trials. We did not include studies of exercise therapy or dysphagia management, which are topics of other Cochrane systematic reviews. DATA COLLECTION AND ANALYSIS: We used standard Cochrane methodological procedures. MAIN RESULTS: The review included 10 trials (249 participants) using different treatment regimens. Seven of the 10 trials assessed single agents, and 3 assessed combined agents. Many of the studies did not present adequate data for the reporting of the primary outcome of the review, which was the percentage change in muscle strength score at six months. Pooled data from two trials of interferon beta-1a (n = 58) identified no important difference in normalised manual muscle strength sum scores from baseline to six months (mean difference (MD) -0.06, 95% CI -0.15 to 0.03) between IFN beta-1a and placebo (moderate-quality evidence). A single trial of methotrexate (MTX) (n = 44) provided moderate-quality evidence that MTX did not arrest or slow disease progression, based on reported percentage change in manual muscle strength sum scores at 12 months. None of the fully published trials were adequately powered to detect a treatment effect. We assessed six of the nine fully published trials as providing very low-quality evidence in relation to the primary outcome measure. Three trials (n = 78) compared intravenous immunoglobulin (combined in one trial with prednisone) to a placebo, but we were unable to perform meta-analysis because of variations in study analysis and presentation of trial data, with no access to the primary data for re-analysis. Other comparisons were also reported in single trials. An open trial of anti-T lymphocyte immunoglobulin (ATG) combined with MTX versus MTX provided very low-quality evidence in favour of the combined therapy, based on percentage change in quantitative muscle strength sum scores at 12 months (MD 12.50%, 95% CI 2.43 to 22.57). Data from trials of oxandrolone versus placebo, azathioprine (AZA) combined with MTX versus MTX, and arimoclomol versus placebo did not allow us to report either normalised or percentage change in muscle strength sum scores. A complete analysis of the effects of arimoclomol is pending data publication. Studies of simvastatin and bimagrumab (BYM338) are ongoing. All analysed trials reported adverse events. Only 1 of the 10 trials interpreted these for statistical significance. None of the trials included prespecified criteria for significant adverse events. AUTHORS' CONCLUSIONS: Trials of interferon beta-1a and MTX provided moderate-quality evidence of having no effect on the progression of IBM. Overall trial design limitations including risk of bias, low numbers of participants, and short duration make it difficult to say whether or not any of the drug treatments included in this review were effective. An open trial of ATG combined with MTX versus MTX provided very low-quality evidence in favour of the combined therapy based on the percentage change data given. We were unable to draw conclusions from trials of IVIg, oxandrolone, and AZA plus MTX versus MTX. We need more randomised controlled trials that are larger, of longer duration, and that use fully validated, standardised, and responsive outcome measures.


ANTECEDENTES: La miositis por cuerpos de inclusión (MCI) es una enfermedad muscular inflamatoria (miopatía) de aparición tardía asociada con atrofia muscular y debilidad progresivas de los miembros proximales y distales. Las opciones de tratamiento se han intentado dirigir a las características inflamatorias y atróficas de esta afección (por ejemplo, con fármacos inmunosupresores e inmunomoduladores, esteroides anabólicos y tratamientos antioxidantes), aunque hasta ahora no hay un tratamiento eficaz conocido para la reversión o la reducción de la progresión de la miositis por cuerpos de inclusión. En esta revisión se han considerado los efectos beneficiosos, los efectos adversos y los costos del tratamiento dirigido a los efectos fundamentales de la afección, a saber, la atrofia muscular, la debilidad y el deterioro funcional. OBJETIVOS: Evaluar los efectos del tratamiento para la MCI. MÉTODOS DE BÚSQUEDA: El 7 octubre 2014, se hicieron búsquedas en el registro especializado del Grupo Cochrane de Enfermedades Neuromusculares (Cochrane Neuromuscular Disease Group), en el Registro Cochrane Central de Ensayos Controlados (Cochrane Central Register of Controlled Trials) (CENTRAL), MEDLINE y en EMBASE. Además, en noviembre 2014 se realizaron búsquedas de ensayos en curso o terminadas pero no publicados en los registros de ensayos clínicos. CRITERIOS DE SELECCIÓN: Se consideraron para inclusión en la revisión los ensayos aleatorios o cuasialeatorios, incluidos los ensayos cruzados (crossover), del tratamiento para la MCI en adultos en comparación con placebo u otro tratamiento. Se excluyeron específicamente los pacientes con MCI familiar y miopatía por cuerpos de inclusión hereditaria, pero se incluyeron los pacientes con enfermedades del tejido conjuntivo y autoinmunitarias asociadas con MCI, que pueden o no identificarse en los ensayos. No se incluyeron los estudios de terapia con ejercicios o tratamiento de la disfagia, que son los temas de otras revisiones sistemáticas Cochrane. OBTENCIÓN Y ANÁLISIS DE LOS DATOS: Se utilizaron procedimientos metodológicos Cochrane estándar. RESULTADOS PRINCIPALES: La revisión incluyó diez ensayos (249 participantes) que utilizaron diferentes regímenes de tratamiento. Siete de los diez ensayos evaluaron agentes únicos y tres evaluaron agentes combinados. Muchos de los estudios no presentaron datos suficientes para el informe del resultado primario de la revisión, que fue el cambio porcentual en la puntuación de fuerza muscular a los seis meses. Los datos agrupados de dos ensayos de interferón beta­1a (n = 58) no identificaron diferencias importantes en las puntuaciones normalizadas de la suma de la fuerza muscular manual desde el inicio hasta los seis meses (diferencia de medias [DM] ­0,06; IC del 95%: ­0,15 a 0,03) entre IFN beta­1a y placebo (pruebas de calidad moderada). Un único ensayo de metotrexato (MTX) (n = 44) proporcionó pruebas de calidad moderada de que el MTX no detuvo ni enlenteció la progresión de la enfermedad, sobre la base del cambio porcentual informado en las puntuaciones de la suma de la fuerza muscular manual a los 12 meses. Ninguno de los ensayos publicados completamente tuvo poder estadístico suficiente para detectar un efecto del tratamiento. Se consideró que seis de los nueve ensayos publicados completamente aportaron pruebas de calidad muy baja con respecto a la medida de resultado primaria. Tres ensayos (n = 78) compararon la inmunoglobulina intravenosa (combinada en un ensayo con prednisona) con placebo, pero no fue posible realizar el metanálisis debido a las variaciones en el análisis de los estudios y a la presentación de los datos del ensayo, sin acceso a los datos primarios para el reanálisis. Otras comparaciones también se informaron en ensayos individuales. Un ensayo abierto de inmunoglobulina anti­linfocitos T (IgAT) combinada con MTX versus MTX proporcionó pruebas de calidad muy baja a favor del tratamiento combinado, sobre la base del cambio porcentual en las puntuaciones cuantitativas de la suma de la fuerza muscular a los 12 meses (DM 12,50%; IC del 95%: 2,43 a 22,57). Los datos de los ensayos de oxandrolona versus placebo, azatioprina (AZA) combinada con MTX versus MTX y arimoclomol versus placebo no permitieron informar sobre el cambio porcentual o normalizado en las puntuaciones de la suma de la fuerza muscular. Un análisis completo de los efectos del arimoclomol está pendiente de la publicación de los datos. Están en curso estudios de simvastatina y bimagrumab (BYM338). Todos los ensayos analizados informaron eventos adversos. Solamente uno de los diez ensayos interpretó la significación estadística de los eventos adversos. Ninguno de los ensayos incluyó criterios preespecificados para los eventos adversos significativos. CONCLUSIONES DE LOS AUTORES: Los ensayos de interferón beta­1a y MTX proporcionaron pruebas de calidad moderada de que no tienen efectos sobre la progresión de la MCI. Las limitaciones generales del diseño de los ensayos, que incluyen el riesgo de sesgo, los escasos números de participantes y la corta duración, hacen difícil determinar si alguno de los tratamientos farmacológicos incluidos en esta revisión fue eficaz. Un ensayo abierto de ATG combinada con MTX versus MTX aportó pruebas de calidad muy baja a favor del tratamiento combinado sobre la base de los datos de cambio porcentual facilitados. No fue posible establecer conclusiones de los ensayos de IgIV, oxandrolona y AZA más MTX versus MTX. Se necesitan más ensayos controlados aleatorios de mayor tamaño, con una duración más prolongada y que utilicen medidas de resultado completamente validadas, estandarizadas y de interés.

11.
J Clin Immunol ; 34 Suppl 1: S120-6, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24722854

RESUMO

The mechanisms of action of IVIg on immunoregulatory and neuroinflammatory network have been predominantly based on in vitro experiments and animal studies, rather than direct effects on human tissues. Based on clinicopathologic correlations and tissues obtained before and after IVIg therapy, the better documented and clinically-relevant in-vivo actions of IVIg include effects on: a) Antibodies. An extracted antigen-specific anti-immunoglobulin (idiotypic) fraction appears partially responsible for its effect in myasthenia gravis and GBS; b) Complement. Sera from Dermatomyositis (DM) patients responding to IVIg, inhibit complement consumption and intercept MAC formation leading to disappearance of MAC deposits in the repeated muscle biopsies and normalization of muscle tissue; c) Genes. In repeated muscle biopsies from DM patients who improved after IVIg, but not from Inclusion-Body-Myositis (IBM) who did not improve, there is a 2-fold alteration of 2206 tissue genes associated with inflammation, fibrosis, tissue remodeling and regeneration; and d) degenerative-proinflammatory molecules and ß-amyloid, implicated in neurodegenerative CNS diseases and IBM. In repeated muscle biopsies of IBM patients who did not respond to IVIg, the mRNA or protein expression for chemokines, IFN-γ, TGF-ß, IL-10, Ubiquitin and aB-crystallin is reduced, but not for the key molecules ICOS, ICOSL, IL-6, IL1-ß, perforin, APP, nitric oxide synthase and nitrotyrosine, in spite of good IVIg penetration in muscles. Collectively, the selective effectiveness of IVIg in human diseases seems to correlate in vivo with inhibition of causative inflammatory mediators. Study of accessible tissues before and after therapy and clinicopathologic correlations, may help explain the differential effect of IVIg in autoimmune or neuroinflammatory diseases.


Assuntos
Doenças Autoimunes/terapia , Imunoglobulinas Intravenosas/uso terapêutico , Músculos/efeitos dos fármacos , Doenças Neurodegenerativas/terapia , Inflamação Neurogênica/terapia , Animais , Anticorpos Anti-Idiotípicos/farmacologia , Doenças Autoimunes/imunologia , Ativação do Complemento/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica , Humanos , Imunoglobulinas Intravenosas/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , Músculos/patologia , Doenças Neurodegenerativas/imunologia , Inflamação Neurogênica/genética , Inflamação Neurogênica/imunologia
12.
Neurol Neuroimmunol Neuroinflamm ; 11(2): e200192, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38147623

RESUMO

OBJECTIVES: To describe a patient with mild GAD-positive stiff-leg syndrome (SLS) who developed severely disabling stiff-person syndrome (SPS) 1 week after mild COVID-19 and discuss the impact of viral implications. METHODS: Video-documented serial clinical observations at baseline, after acute COVID-19, and after IVIG treatments. RESULTS: A 39-year-old man with left-SLS was stable during a 2-year follow-up with low-dose antispasmodics, working fully and functioning normally, even able to run. One week after mild COVID-19, he started to experience generalized SPS symptomatology that steadily worsened the following 2-3 weeks, becoming unable to walk, requiring a walker, with significant thoracolumbar and bilateral leg stiffness and spasms. GAD ab were very high. After 3 monthly IVIg infusions he showed improvements, but his gait remains significantly stiff. All clinical changes, from baseline to post-Covid, and then post- IVIg have been video-documented. DISCUSSION: This is the first, clearly documented, severe GAD-positive SPS after COVID-19. Although viral or postviral causation can be incidental, the temporal connection with acute COVID-19, the severe disease worsening after symptom-onset, and the subsequent steady improvement after IVIg, suggest viral-triggered autoimmunity. Because COVID-19 reportedly can trigger or worsen GAD-associated diabetes type 1 through proinflammatory mediators, and SPS has been reportedly triggered by West Nile Virus, possibly through molecular mimicry, this case of acutely converting GAD-SLS to GAD-SPS suggest the need to explore viral etiologies in patients with GAD-SPS experiencing acute, long-lasting episodic exacerbations of stiffness and spasms.


Assuntos
COVID-19 , Rigidez Muscular Espasmódica , Masculino , Humanos , Adulto , Rigidez Muscular Espasmódica/complicações , Rigidez Muscular Espasmódica/diagnóstico , Imunoglobulinas Intravenosas , COVID-19/complicações , Espasmo/complicações , Espasmo/terapia
13.
N Engl J Med ; 373(4): 393-4, 2015 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-26200989
15.
BMC Neurol ; 13: 29, 2013 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-23514108

RESUMO

BACKGROUND: Hereditary myopathy with early respiratory failure (HMERF) was described in several North European families and recently linked to a titin gene (TTN) mutation. We independently studied HMERF-like diseases with the purpose to identify the cause, refine diagnostic criteria, and estimate the frequency of this disease among myopathy patients of various ethnic origins. METHODS: Whole exome sequencing analysis was carried out in a large U.S. family that included seven members suffering from skeletal muscle weakness and respiratory failure. Subsequent mutation screening was performed in further 45 unrelated probands with similar phenotypes. Studies included muscle strength evaluation, nerve conduction studies and concentric needle EMG, respiratory function test, cardiologic examination, and muscle biopsy. RESULTS: A novel TTN p.Gly30150Asp mutation was identified in the highly conserved A-band of titin that co-segregated with the disease in the U.S. family. Screening of 45 probands initially diagnosed as myofibrillar myopathy (MFM) but excluded based on molecular screening for the known MFM genes led to the identification of a previously reported TTN p.Cys30071Arg mutation in one patient. This same mutation was also identified in a patient with suspected HMERF. The p.Gly30150Asp and p.Cys30071Arg mutations are localized to a side chain of fibronectin type III element A150 of the 10th C-zone super-repeat of titin. CONCLUSIONS: Missense mutations in TTN are the cause of HMERF in families of diverse origins. A comparison of phenotypic features of HMERF caused by the three known TTN mutations in various populations allowed to emphasize distinct clinical/pathological features that can serve as the basis for diagnosis. The newly identified p.Gly30150Asp and the p.Cys30071Arg mutation are localized to a side chain of fibronectin type III element A150 of the 10th C-zone super-repeat of titin.


Assuntos
Saúde da Família , Doenças Genéticas Inatas/etnologia , Doenças Genéticas Inatas/genética , Proteínas Musculares/genética , Doenças Musculares/etnologia , Doenças Musculares/genética , Mutação/genética , Proteínas Quinases/genética , Insuficiência Respiratória/etnologia , Insuficiência Respiratória/genética , Adolescente , Adulto , Conectina , Análise Mutacional de DNA , Avaliação da Deficiência , Eletromiografia , Exoma/genética , Feminino , Doenças Genéticas Inatas/diagnóstico , Humanos , Masculino , Proteínas Musculares/metabolismo , Força Muscular/genética , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Músculo Esquelético/ultraestrutura , Doenças Musculares/diagnóstico , Polimorfismo de Nucleotídeo Único , Insuficiência Respiratória/diagnóstico , Tomografia Computadorizada por Raios X , Estados Unidos/etnologia , Adulto Jovem
16.
Arthritis Rheum ; 64(12): 4094-103, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22941914

RESUMO

OBJECTIVE: In sporadic inclusion body myositis (IBM), inflammation and accumulation of ß-amyloid-associated molecules cause muscle fiber damage. We undertook this study to determine why intravenous immunoglobulin (IVIG) and prednisone are not effective in sporadic IBM despite their effectiveness in other inflammatory myopathies. METHODS: Relevant inflammatory and degeneration- associated markers were assessed by quantitative polymerase chain reaction and immunohistochemistry in repeated muscle biopsy specimens from patients with sporadic IBM treated in a controlled study with IVIG and prednisone (n = 5) or with prednisone alone (n = 5). Functional effects were assessed in a muscle cell culture model. RESULTS: In muscle biopsy specimens, messenger RNA (mRNA) expression of the proinflammatory chemokines CXCL9, CCL3, and CCL4 and of the cytokines interferon-γ (IFNγ), transforming growth factor ß, interleukin-10 (IL-10), and IL-1ß was significantly reduced after treatment in both groups. No consistent changes were observed for tumor necrosis factor α, IL-6, inducible costimulator (ICOS), its ligand ICOSL, and perforin. Messenger RNA expression of the degeneration-associated molecule ubiquitin and the heat-shock protein αB-crystallin was also reduced, but no changes were noted for amyloid precursor protein (APP) or desmin. By immunohistochemistry, a significant down-modulation of chemokines was observed, but not of inducible nitric oxide (NO) synthase, nitrotyrosine, IL-1ß, APP, and ubiquitin; ß-amyloid was reduced in 6 of 10 patients. Pronounced staining of IgG was observed in the muscle after treatment with IVIG, indicating penetration of infused IgG into the muscle and a possible local effect. In muscle cells exposed to IFNγ plus IL-1ß, IgG and/or prednisone down-regulated mRNA expression of IL-1ß 2.5-fold. Accumulation of ß-amyloid, overexpression of αB-crystallin, and cell death were prevented. In contrast, NO-associated cell stress remained unchanged. CONCLUSION: IVIG and prednisone reduce some inflammatory and degenerative molecules in muscle of patients with sporadic IBM and in vitro, but do not sufficiently suppress myotoxic and cell stress mediators such as NO. The data provide an explanation for the resistance of sporadic IBM to immunotherapy and identify markers that may help to design novel treatment strategies.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Inflamação/metabolismo , Miosite de Corpos de Inclusão/terapia , Prednisona/uso terapêutico , Biópsia , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Quimioterapia Combinada , Humanos , Imunoterapia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miosite de Corpos de Inclusão/metabolismo , Óxido Nítrico/metabolismo , RNA Mensageiro/metabolismo , Resultado do Tratamento
17.
Brain ; 135(Pt 4): 1102-14, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22436237

RESUMO

Sporadic inclusion body myositis is a severely disabling myopathy. The design of effective treatment strategies is hampered by insufficient understanding of the complex disease pathology. Particularly, the nature of interrelationships between inflammatory and degenerative pathomechanisms in sporadic inclusion body myositis has remained elusive. In Alzheimer's dementia, accumulation of ß-amyloid has been shown to be associated with upregulation of nitric oxide. Using quantitative polymerase chain reaction, an overexpression of inducible nitric oxide synthase was observed in five out of ten patients with sporadic inclusion body myositis, two of eleven with dermatomyositis, three of eight with polymyositis, two of nine with muscular dystrophy and two of ten non-myopathic controls. Immunohistochemistry confirmed protein expression of inducible nitric oxide synthase and demonstrated intracellular nitration of tyrosine, an indicator for intra-fibre production of nitric oxide, in sporadic inclusion body myositis muscle samples, but much less in dermatomyositis or polymyositis, hardly in dystrophic muscle and not in non-myopathic controls. Using fluorescent double-labelling immunohistochemistry, a significant co-localization was observed in sporadic inclusion body myositis muscle between ß-amyloid, thioflavine-S and nitrotyrosine. In primary cultures of human myotubes and in myoblasts, exposure to interleukin-1ß in combination with interferon-γ induced a robust upregulation of inducible nitric oxide synthase messenger RNA. Using fluorescent detectors of reactive oxygen species and nitric oxide, dichlorofluorescein and diaminofluorescein, respectively, flow cytometry revealed that interleukin-1ß combined with interferon-γ induced intracellular production of nitric oxide, which was associated with necrotic cell death in muscle cells. Intracellular nitration of tyrosine was noted, which partly co-localized with amyloid precursor protein, but not with desmin. Pharmacological inhibition of inducible nitric oxide synthase by 1400W reduced intracellular production of nitric oxide and prevented accumulation of ß-amyloid, nitration of tyrosine as well as cell death inflicted by interleukin-1ß combined with interferon-γ. Collectively, these data suggest that, in skeletal muscle, inducible nitric oxide synthase is a central component of interactions between interleukin-1ß and ß-amyloid, two of the most relevant molecules in sporadic inclusion body myositis. The data further our understanding of the pathology of sporadic inclusion body myositis and may point to novel treatment strategies.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-1beta/farmacologia , Células Musculares/metabolismo , Miosite de Corpos de Inclusão/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Morte Celular/efeitos dos fármacos , Células Cultivadas , Dactinomicina/análogos & derivados , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Regulação da Expressão Gênica/fisiologia , Humanos , Interferon gama/farmacologia , Células Musculares/efeitos dos fármacos , Miosite de Corpos de Inclusão/metabolismo , Óxido Nítrico Sintase Tipo II/genética , RNA Mensageiro/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
18.
J Infect Dis ; 205(12): 1778-87, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22476717

RESUMO

BACKGROUND: Although human immunodeficiency virus (HIV) infection and antiretroviral therapy (ART) affect mitochondrial DNA (mtDNA) content and function, comprehensive evaluations of their effects on mitochondria in muscle, adipose tissue, and blood cells are limited. METHODS: Mitochondrial DNA quantification, mitochondrial genome sequencing, and gene expression analysis were performed on muscle, adipose tissue, and peripheral blood mononuclear cell (PBMC) samples from untreated HIV-positive patients, HIV-positive patients receiving nucleoside reverse transcriptase inhibitor (NRTI)-based ART, and HIV-negative controls. RESULTS: The adipose tissue mtDNA/nuclear DNA (nDNA) ratio was increased in untreated HIV-infected patients (ratio, 353) and decreased in those receiving ART (ratio, 162) compared with controls (ratio, 255; P < .05 for both comparisons); the difference between the 2 HIV-infected groups was also significant (P = .002). In HIV-infected participants, mtDNA/nDNA in adipose tissue correlated with the level of activation (CD38+ /HLA-DR+) for CD4+ and CD8+ lymphocytes. No significant differences in mtDNA content were noted in muscle or PMBCs among groups. Exploratory DNA microarray analysis identified differential gene expression between patient groups, including a subset of adipose tissue genes. CONCLUSIONS: HIV infection and ART have opposing effects on mtDNA content in adipose tissue; immune activation may mediate the effects of HIV, whereas NRTIs likely mediate the effects of ART.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Fármacos Anti-HIV/administração & dosagem , Terapia Antirretroviral de Alta Atividade , DNA Mitocondrial/metabolismo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/patologia , Mitocôndrias/efeitos dos fármacos , Adolescente , Adulto , Feminino , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Músculos/efeitos dos fármacos , Adulto Jovem
19.
Handb Clin Neurol ; 195: 425-460, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37562881

RESUMO

The autoimmune inflammatory myopathies constitute a heterogeneous group of acquired myopathies that have in common the presence of endomysial inflammation and moderate to severe muscle weakness. Based on currently evolved distinct clinical, histologic, immunopathologic, and autoantibody features, these disorders can be best classified as dermatomyositis, necrotizing autoimmune myositis, antisynthetase syndrome-overlap myositis, and inclusion body myositis. Although polymyositis is no longer considered a distinct subset but rather an extinct entity, it is herein described because its clinicopathologic information has provided over many years fundamental information on T-cell-mediated myocytotoxicity, especially in reference to inclusion body myositis. Each inflammatory myopathy subset has distinct immunopathogenesis, prognosis, and response to immunotherapies, necessitating the need to correctly diagnose each subtype from the outset and avoid disease mimics. The paper describes the main clinical characteristics that aid in the diagnosis of each myositis subtype, highlights the distinct features on muscle morphology and immunopathology, elaborates on the potential role of autoantibodies in pathogenesis or diagnosis , and clarifies common uncertainties in reference to putative triggering factors such as statins and viruses including the 2019-coronavirus-2 pandemic. It extensively describes the main autoimmune markers related to autoinvasive myocytotoxic T-cells, activated B-cells, complement, cytokines, and the possible role of innate immunity. The concomitant myodegenerative features seen in inclusion body myositis along with their interrelationship between inflammation and degeneration are specifically emphasized. Finally, practical guidelines on the best therapeutic approaches are summarized based on up-to-date knowledge and controlled studies, highlighting the prospects of future immunotherapies and ongoing controversies.


Assuntos
Doenças Autoimunes , COVID-19 , Miosite de Corpos de Inclusão , Miosite , Humanos , Miosite de Corpos de Inclusão/diagnóstico , Miosite de Corpos de Inclusão/terapia , Miosite/diagnóstico , Miosite/terapia , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/terapia , Inflamação , Autoanticorpos
20.
Ther Adv Neurol Disord ; 16: 17562864231194821, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37675039

RESUMO

The value of practice guidelines in the three most common autoimmune neuromuscular disorders, namely Chronic Inflammatory Demyelinating Polyneuropathy (CIDP), Myasthenia Gravis (MG) and Autoimmune Inflammatory Myopathies (AIM), has been extensively debated regarding their usefulness in clinical practice, objectivity and universal value considering that guidelines are also established regionally in certain countries. This commentary highlights common concerns on how guidelines are presently generated, pointing out: (a) non-sufficient diversity among Task-Force members to identify and address not only routine clinical and electrophysiology issues but also immunology, imaging, pathology, biomarkers, epidemiology or treatment economics; (b) Task-Force being often comprised by the same or seemingly like-minded members conveying the erroneous impression that experts with opposing views might have been excluded, even if this is clearly not the case; and (c) relying on web-based registries or retrospective data collections from heterogeneous sources. As a result, the existing practice guidelines in CIDP, MG and AIM remain an unfinished business but an excellent base for further enhancement. Guidelines can be extremely helpful not only for clinical trials but also in clinical practice if viewed as a living document with continuously updated versions by experts even with opposing views with precise information on diagnostics, pathomechanisms, therapeutic schemes, evolving biomarkers and economics of new therapies with validation of the post-guidelines criteria. Geographic diversity should be taken into consideration because the availability of biomarker testing, and therapies differ among countries. Patient preferences need to be also considered in therapeutic guidelines because newly marketed drugs offer more options steadily changing the therapeutic algorithms in autoimmune neuromuscular diseases generating also questions as to whether they also influence decisions on insurance coverage. Collectively, these startup considerations are aimed to make practice guidelines more objective, widely acceptable worldwide and more practical or easier to follow in clinical practice.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA