Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 146(10): 4233-4246, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37186601

RESUMEN

In utero exposure to maternal antibodies targeting the fetal acetylcholine receptor isoform (fAChR) can impair fetal movement, leading to arthrogryposis multiplex congenita (AMC). Fetal AChR antibodies have also been implicated in apparently rare, milder myopathic presentations termed fetal acetylcholine receptor inactivation syndrome (FARIS). The full spectrum associated with fAChR antibodies is still poorly understood. Moreover, since some mothers have no myasthenic symptoms, the condition is likely underreported, resulting in failure to implement effective preventive strategies. Here we report clinical and immunological data from a multicentre cohort (n = 46 cases) associated with maternal fAChR antibodies, including 29 novel and 17 previously reported with novel follow-up data. Remarkably, in 50% of mothers there was no previously established myasthenia gravis (MG) diagnosis. All mothers (n = 30) had AChR antibodies and, when tested, binding to fAChR was often much greater than that to the adult AChR isoform. Offspring death occurred in 11/46 (23.9%) cases, mainly antenatally due to termination of pregnancy prompted by severe AMC (7/46, 15.2%), or during early infancy, mainly from respiratory failure (4/46, 8.7%). Weakness, contractures, bulbar and respiratory involvement were prominent early in life, but improved gradually over time. Facial (25/34; 73.5%) and variable peripheral weakness (14/32; 43.8%), velopharyngeal insufficiency (18/24; 75%) and feeding difficulties (16/36; 44.4%) were the most common sequelae in long-term survivors. Other unexpected features included hearing loss (12/32; 37.5%), diaphragmatic paresis (5/35; 14.3%), CNS involvement (7/40; 17.5%) and pyloric stenosis (3/37; 8.1%). Oral salbutamol used empirically in 16/37 (43.2%) offspring resulted in symptom improvement in 13/16 (81.3%). Combining our series with all previously published cases, we identified 21/85 mothers treated with variable combinations of immunotherapies (corticosteroids/intravenous immunoglobulin/plasmapheresis) during pregnancy either for maternal MG symptom control (12/21 cases) or for fetal protection (9/21 cases). Compared to untreated pregnancies (64/85), maternal treatment resulted in a significant reduction in offspring deaths (P < 0.05) and other complications, with treatment approaches involving intravenous immunoglobulin/ plasmapheresis administered early in pregnancy most effective. We conclude that presentations due to in utero exposure to maternal (fetal) AChR antibodies are more common than currently recognized and may mimic a wide range of neuromuscular disorders. Considering the wide clinical spectrum and likely diversity of underlying mechanisms, we propose 'fetal acetylcholine receptor antibody-related disorders' (FARAD) as the most accurate term for these presentations. FARAD is vitally important to recognize, to institute appropriate management strategies for affected offspring and to improve outcomes in future pregnancies. Oral salbutamol is a symptomatic treatment option in survivors.


Asunto(s)
Artrogriposis , Miastenia Gravis , Enfermedades Neuromusculares , Embarazo , Femenino , Adulto , Humanos , Inmunoglobulinas Intravenosas , Receptores Colinérgicos , Miastenia Gravis/terapia , Miastenia Gravis/complicaciones , Autoanticuerpos , Artrogriposis/complicaciones
2.
Brain ; 144(2): 584-600, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33559681

RESUMEN

The extracellular matrix comprises a network of macromolecules such as collagens, proteoglycans and glycoproteins. VWA1 (von Willebrand factor A domain containing 1) encodes a component of the extracellular matrix that interacts with perlecan/collagen VI, appears to be involved in stabilizing extracellular matrix structures, and demonstrates high expression levels in tibial nerve. Vwa1-deficient mice manifest with abnormal peripheral nerve structure/function; however, VWA1 variants have not previously been associated with human disease. By interrogating the genome sequences of 74 180 individuals from the 100K Genomes Project in combination with international gene-matching efforts and targeted sequencing, we identified 17 individuals from 15 families with an autosomal-recessive, non-length dependent, hereditary motor neuropathy and rare biallelic variants in VWA1. A single disease-associated allele p.(G25Rfs*74), a 10-bp repeat expansion, was observed in 14/15 families and was homozygous in 10/15. Given an allele frequency in European populations approaching 1/1000, the seven unrelated homozygote individuals ascertained from the 100K Genomes Project represents a substantial enrichment above expected. Haplotype analysis identified a shared 220 kb region suggesting that this founder mutation arose >7000 years ago. A wide age-range of patients (6-83 years) helped delineate the clinical phenotype over time. The commonest disease presentation in the cohort was an early-onset (mean 2.0 ± 1.4 years) non-length-dependent axonal hereditary motor neuropathy, confirmed on electrophysiology, which will have to be differentiated from other predominantly or pure motor neuropathies and neuronopathies. Because of slow disease progression, ambulation was largely preserved. Neurophysiology, muscle histopathology, and muscle MRI findings typically revealed clear neurogenic changes with single isolated cases displaying additional myopathic process. We speculate that a few findings of myopathic changes might be secondary to chronic denervation rather than indicating an additional myopathic disease process. Duplex reverse transcription polymerase chain reaction and immunoblotting using patient fibroblasts revealed that the founder allele results in partial nonsense mediated decay and an absence of detectable protein. CRISPR and morpholino vwa1 modelling in zebrafish demonstrated reductions in motor neuron axonal growth, synaptic formation in the skeletal muscles and locomotive behaviour. In summary, we estimate that biallelic variants in VWA1 may be responsible for up to 1% of unexplained hereditary motor neuropathy cases in Europeans. The detailed clinical characterization provided here will facilitate targeted testing on suitable patient cohorts. This novel disease gene may have previously evaded detection because of high GC content, consequential low coverage and computational difficulties associated with robustly detecting repeat-expansions. Reviewing previously unsolved exomes using lower QC filters may generate further diagnoses.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Neuropatía Hereditaria Motora y Sensorial/genética , Adulto , Anciano , Animales , Conducta Animal/fisiología , Niño , Femenino , Neuropatía Hereditaria Motora y Sensorial/patología , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Mutación , Linaje , Adulto Joven , Pez Cebra
3.
J Neurol Neurosurg Psychiatry ; 89(7): 762-768, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29437916

RESUMEN

BACKGROUND: Defects in glycosylation of alpha-dystroglycan (α-DG) cause autosomal-recessive disorders with wide clinical and genetic heterogeneity, with phenotypes ranging from congenital muscular dystrophies to milder limb girdle muscular dystrophies. Patients show variable reduction of immunoreactivity to antibodies specific for glycoepitopes of α-DG on a muscle biopsy. Recessive mutations in 18 genes, including guanosine diphosphate mannose pyrophosphorylase B (GMPPB), have been reported to date. With no specific clinical and pathological handles, diagnosis requires parallel or sequential analysis of all known genes. METHODS: We describe clinical, genetic and biochemical findings of 21 patients with GMPPB-associated dystroglycanopathy. RESULTS: We report eight novel mutations and further expand current knowledge on clinical and muscle MRI features of this condition. In addition, we report a consistent shift in the mobility of beta-dystroglycan (ß-DG) on Western blot analysis of all patients analysed by this mean. This was only observed in patients with GMPPB in our large dystroglycanopathy cohort. We further demonstrate that this mobility shift in patients with GMPPB was due to abnormal N-linked glycosylation of ß-DG. CONCLUSIONS: Our data demonstrate that a change in ß-DG electrophoretic mobility in patients with dystroglycanopathy is a distinctive marker of the molecular defect in GMPPB.


Asunto(s)
Distroglicanos/metabolismo , Guanosina Difosfato Manosa/genética , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mutación/genética , Nucleotidiltransferasas/genética , Adolescente , Anciano , Biomarcadores/metabolismo , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Masculino , Persona de Mediana Edad , Distrofias Musculares/patología
4.
Muscle Nerve ; 57(3): 380-387, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29053898

RESUMEN

INTRODUCTION: Earlier small case series and clinical observations reported on chronic pain playing an important role in facioscapulohumeral dystrophy (FSHD). The aim of this study was to determine the characteristics and impact of pain on quality of life (QoL) in patients with FSHD. METHODS: We analyzed patient reported outcome measures collected through the U.K. FSHD Patient Registry. RESULTS: Of 398 patients, 88.6% reported pain at the time of study. The most frequent locations were shoulders and lower back. A total of 203 participants reported chronic pain, 30.4% of them as severe. The overall disease impact on QoL was significantly higher in patients with early onset and long disease duration. Chronic pain had a negative impact on all Individualised Neuromuscular Quality of Life Questionnaire domains and overall disease score. DISCUSSION: Our study shows that pain in FSHD type 1 (FSHD1) is frequent and strongly impacts on QoL, similar to other chronic, painful disorders. Management of pain should be considered when treating FSHD1 patients. Muscle Nerve 57: 380-387, 2018.


Asunto(s)
Dolor Crónico/psicología , Distrofia Muscular Facioescapulohumeral/psicología , Calidad de Vida/psicología , Adulto , Anciano , Dolor Crónico/complicaciones , Dolor Crónico/diagnóstico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Distrofia Muscular Facioescapulohumeral/complicaciones , Distrofia Muscular Facioescapulohumeral/diagnóstico , Dimensión del Dolor , Índice de Severidad de la Enfermedad , Adulto Joven
5.
Brain ; 137(Pt 5): 1323-36, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24727571

RESUMEN

Despite being a canonical presenting feature of mitochondrial disease, the genetic basis of progressive external ophthalmoplegia remains unknown in a large proportion of patients. Here we show that mutations in SPG7 are a novel cause of progressive external ophthalmoplegia associated with multiple mitochondrial DNA deletions. After excluding known causes, whole exome sequencing, targeted Sanger sequencing and multiplex ligation-dependent probe amplification analysis were used to study 68 adult patients with progressive external ophthalmoplegia either with or without multiple mitochondrial DNA deletions in skeletal muscle. Nine patients (eight probands) were found to carry compound heterozygous SPG7 mutations, including three novel mutations: two missense mutations c.2221G>A; p.(Glu741Lys), c.2224G>A; p.(Asp742Asn), a truncating mutation c.861dupT; p.Asn288*, and seven previously reported mutations. We identified a further six patients with single heterozygous mutations in SPG7, including two further novel mutations: c.184-3C>T (predicted to remove a splice site before exon 2) and c.1067C>T; p.(Thr356Met). The clinical phenotype typically developed in mid-adult life with either progressive external ophthalmoplegia/ptosis and spastic ataxia, or a progressive ataxic disorder. Dysphagia and proximal myopathy were common, but urinary symptoms were rare, despite the spasticity. Functional studies included transcript analysis, proteomics, mitochondrial network analysis, single fibre mitochondrial DNA analysis and deep re-sequencing of mitochondrial DNA. SPG7 mutations caused increased mitochondrial biogenesis in patient muscle, and mitochondrial fusion in patient fibroblasts associated with the clonal expansion of mitochondrial DNA mutations. In conclusion, the SPG7 gene should be screened in patients in whom a disorder of mitochondrial DNA maintenance is suspected when spastic ataxia is prominent. The complex neurological phenotype is likely a result of the clonal expansion of secondary mitochondrial DNA mutations modulating the phenotype, driven by compensatory mitochondrial biogenesis.


Asunto(s)
ADN Mitocondrial/metabolismo , Metaloendopeptidasas/metabolismo , Enfermedades Mitocondriales/complicaciones , Enfermedades Mitocondriales/genética , Mutación/genética , Oftalmoplejía Externa Progresiva Crónica/complicaciones , Oftalmoplejía Externa Progresiva Crónica/genética , ATPasas Asociadas con Actividades Celulares Diversas , Anciano , Enfermedad Crónica , Análisis Mutacional de ADN , ADN Mitocondrial/genética , Estimulación Eléctrica , Complejo IV de Transporte de Electrones/metabolismo , Potenciales Evocados Motores/genética , Femenino , Estudios de Asociación Genética , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Oftalmoplejía Externa Progresiva Crónica/patología , Fenotipo , Tiempo de Reacción
6.
J Neurol Neurosurg Psychiatry ; 85(5): 538-43, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23757420

RESUMEN

A national U.K. workshop to discuss practical clinical management issues related to pregnancy in women with myasthenia gravis was held in May 2011. The purpose was to develop recommendations to guide general neurologists and obstetricians and facilitate best practice before, during and after pregnancy. The main conclusions were (1) planning should be instituted well in advance of any potential pregnancy to allow time for myasthenic status and drug optimisation; (2) multidisciplinary liaison through the involvement of relevant specialists should occur throughout pregnancy, during delivery and in the neonatal period; (3) provided that their myasthenia is under good control before pregnancy, the majority of women can be reassured that it will remain stable throughout pregnancy and the postpartum months; (4) spontaneous vaginal delivery should be the aim and actively encouraged; (5) those with severe myasthenic weakness need careful, multidisciplinary management with prompt access to specialist advice and facilities; (6) newborn babies born to myasthenic mothers are at risk of transient myasthenic weakness, even if the mother's myasthenia is well-controlled, and should have rapid access to neonatal high-dependency support.


Asunto(s)
Miastenia Gravis/terapia , Complicaciones del Embarazo/terapia , Atención Prenatal/organización & administración , Adolescente , Adulto , Protocolos Clínicos , Parto Obstétrico , Femenino , Humanos , Inmunosupresores/uso terapéutico , Recién Nacido , Miastenia Gravis/complicaciones , Miastenia Gravis/diagnóstico , Guías de Práctica Clínica como Asunto , Embarazo , Complicaciones del Embarazo/diagnóstico , Complicaciones del Embarazo/etiología , Reino Unido , Adulto Joven
8.
J Neurol Neurosurg Psychiatry ; 85(12): 1359-65, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24695763

RESUMEN

OBJECTIVE: GNE myopathy is a rare recessive myopathy associated with inclusion bodies on muscle biopsy. The clinical phenotype is associated with distal muscle weakness with quadriceps sparing. Most of the current information on GNE myopathy has been obtained through studies of Jewish and Japanese patient cohorts carrying founder mutations in the GNE gene. However, little is known about GNE myopathy in Europe where the prevalence is thought to be very low. METHODS: Patients were referred through the National Specialist Commissioning Team service for limb-girdle muscular dystrophies at Newcastle (UK). All patients harbouring mutations in the GNE gene were recruited for our study. Detailed clinical and genetic data as well as muscle MRIs and muscle biopsies were reviewed. RESULTS: We identified 26 patients harbouring mutations in the GNE gene. Two previously reported mutations (c.1985C>T, p.Ala662Val and c.1225G>T, p.Asp409Tyr) were prevalent in the Scottish, Northern Irish and Northern English populations; with 90% of these patients carrying at least one of the two mutations. Clinically, we confirmed the homogenous pattern of selective quadriceps sparing but noted additional features like asymmetry of weakness at disease onset. CONCLUSIONS: GNE myopathy is an important diagnosis to consider in patients presenting with distal leg muscle weakness. We report, for the first time, two common mutations in the north of Britain and highlight the broader spectrum of clinical phenotypes. We also propose that the prevalence of GNE myopathy may be underestimated due to the frequent absence of rimmed vacuoles in the muscle biopsy.


Asunto(s)
Miopatías Distales/genética , Complejos Multienzimáticos/genética , Mutación/genética , Adolescente , Adulto , Niño , Miopatías Distales/epidemiología , Miopatías Distales/patología , Femenino , Mutación del Sistema de Lectura/genética , Humanos , Imagen por Resonancia Magnética , Masculino , Debilidad Muscular/genética , Debilidad Muscular/patología , Músculo Esquelético/patología , Mutación Missense/genética , Reino Unido/epidemiología , Adulto Joven
9.
J Neurol Neurosurg Psychiatry ; 85(3): 345-53, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23606733

RESUMEN

OBJECTIVE: Several families with characteristic features of hereditary myopathy with early respiratory failure (HMERF) have remained without genetic cause. This international study was initiated to clarify epidemiology and the genetic underlying cause in these families, and to characterise the phenotype in our large cohort. METHODS: DNA samples of all currently known families with HMERF without molecular genetic cause were obtained from 12 families in seven different countries. Clinical, histopathological and muscle imaging data were collected and five biopsy samples made available for further immunohistochemical studies. Genotyping, exome sequencing and Sanger sequencing were used to identify and confirm sequence variations. RESULTS: All patients with clinical diagnosis of HMERF were genetically solved by five different titin mutations identified. One mutation has been reported while four are novel, all located exclusively in the FN3 119 domain (A150) of A-band titin. One of the new mutations showed semirecessive inheritance pattern with subclinical myopathy in the heterozygous parents. Typical clinical features were respiratory failure at mid-adulthood in an ambulant patient with very variable degree of muscle weakness. Cytoplasmic bodies were retrospectively observed in all muscle biopsy samples and these were reactive for myofibrillar proteins but not for titin. CONCLUSIONS: We report an extensive collection of families with HMERF with five different mutations in exon 343 of TTN, which establishes this exon as the primary target for molecular diagnosis of HMERF. Our relatively large number of new families and mutations directly implies that HMERF is not extremely rare, not restricted to Northern Europe and should be considered in undetermined myogenic respiratory failure.


Asunto(s)
Enfermedades Genéticas Congénitas/epidemiología , Enfermedades Musculares/epidemiología , Insuficiencia Respiratoria/epidemiología , Adulto , Anciano , Conectina/genética , Exoma/genética , Femenino , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Ligamiento Genético/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Mutación/genética , Linaje , Fenotipo , Insuficiencia Respiratoria/genética , Insuficiencia Respiratoria/patología
10.
Muscle Nerve ; 49(6): 845-57, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24037712

RESUMEN

INTRODUCTION: Myotonia congenita (MC) is caused by congenital defects in the muscle chloride channel CLC-1. This study used muscle velocity recovery cycles (MVRCs) to investigate how membrane function is affected. METHODS: MVRCs and responses to repetitive stimulation were compared between 18 patients with genetically confirmed MC (13 recessive, 7 dominant) and 30 age-matched, normal controls. RESULTS: MC patients exhibited increased early supernormality, but this was prevented by treatment with sodium channel blockers. After multiple conditioning stimuli, late supernormality was enhanced in all MC patients, indicating delayed repolarization. These abnormalities were similar between the MC subtypes, but recessive patients showed a greater drop in amplitude during repetitive stimulation. CONCLUSIONS: MVRCs indicate that chloride conductance only becomes important when muscle fibers are depolarized. The differential responses to repetitive stimulation suggest that, in dominant MC, the affected chloride channels are activated by strong depolarization, consistent with a positive shift of the CLC-1 activation curve.


Asunto(s)
Canales de Cloruro/fisiología , Músculo Esquelético/fisiopatología , Miotonía Congénita/fisiopatología , Recuperación de la Función/fisiología , Adulto , Anciano , Estudios de Casos y Controles , Estimulación Eléctrica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/efectos de los fármacos , Miotonía Congénita/tratamiento farmacológico , Tiempo de Reacción/fisiología , Bloqueadores de los Canales de Sodio/farmacología , Bloqueadores de los Canales de Sodio/uso terapéutico , Factores de Tiempo
11.
Hum Mutat ; 34(8): 1111-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23606453

RESUMEN

Limb girdle muscular dystrophy type 2L or anoctaminopathy is a condition mainly characterized by adult onset proximal lower limb muscular weakness and raised CK values, due to recessive ANO5 gene mutations. An exon 5 founder mutation (c.191dupA) has been identified in most of the British and German LGMD2L patients so far reported. We aimed to further investigate the prevalence and spectrum of ANO5 gene mutations and related clinical phenotypes, by screening 205 undiagnosed patients referred to our molecular service with a clinical suspicion of anoctaminopathy. A total of 42 unrelated patients had two ANO5 mutations (21%), whereas 14 carried a single change. We identified 34 pathogenic changes, 15 of which are novel. The c.191dupA mutation represents 61% of mutated alleles and appears to be less prevalent in non-Northern European populations. Retrospective clinical analysis corroborates the prevalently proximal lower limb phenotype, the male predominance and absence of major cardiac or respiratory involvement. Identification of cases with isolated hyperCKaemia and very late symptomatic male and female subjects confirms the extension of the phenotypic spectrum of the disease. Anoctaminopathy appears to be one of the most common adult muscular dystrophies in Northern Europe, with a prevalence of about 20%-25% in unselected undiagnosed cases.


Asunto(s)
Canales de Cloruro/genética , Distrofia Muscular de Cinturas/genética , Mutación , Adulto , Anciano , Anoctaminas , Canales de Cloruro/metabolismo , Europa (Continente)/epidemiología , Femenino , Variación Genética , Humanos , Masculino , Persona de Mediana Edad , Distrofia Muscular de Cinturas/epidemiología , Distrofia Muscular de Cinturas/metabolismo , Fenotipo , Prevalencia , Estudios Retrospectivos , Factores Sexuales
13.
J Neurol Neurosurg Psychiatry ; 84(10): 1119-25, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23447650

RESUMEN

BACKGROUND: A newly defined congenital myasthenic syndrome (CMS) caused by DPAGT1 mutations has recently been reported. While many other CMS-associated proteins have discrete roles localised to the neuromuscular junction, DPAGT1 is ubiquitously expressed, modifying many proteins, and as such is an unexpected cause of isolated neuromuscular involvement. METHODS: We present detailed clinical characteristics of five patients with CMS caused by DPAGT1 mutations. RESULTS: Patients have prominent limb girdle weakness and minimal craniobulbar symptoms. Tubular aggregates on muscle biopsy are characteristic but may not be apparent on early biopsies. Typical myasthenic features such as pyridostigmine and 3, 4- diaminopyridine responsiveness, and decrement on repetitive nerve stimulation are present. CONCLUSIONS: These patients mimic myopathic disorders and are likely to be under-diagnosed. The descriptions here should facilitate recognition of this disorder. In particular minimal craniobulbar involvement and tubular aggregates on muscle biopsy help to distinguish DPAGT1 CMS from the majority of other forms of CMS. Patients with DPAGT1 CMS share similar clinical features with patients who have CMS caused by mutations in GFPT1, another recently identified CMS subtype.


Asunto(s)
Análisis Mutacional de ADN , Síndromes Miasténicos Congénitos/genética , N-Acetilglucosaminiltransferasas/genética , 4-Aminopiridina/análogos & derivados , 4-Aminopiridina/uso terapéutico , Agonistas de Receptores Adrenérgicos beta 2 , Adulto , Edad de Inicio , Albuterol/uso terapéutico , Amifampridina , Biopsia , Inhibidores de la Colinesterasa/uso terapéutico , Diagnóstico Diferencial , Exoma , Femenino , Pruebas Genéticas , Glicosilación , Humanos , Masculino , Persona de Mediana Edad , Neuronas Motoras/fisiología , Músculo Esquelético/inervación , Músculo Esquelético/patología , Síndromes Miasténicos Congénitos/diagnóstico , Síndromes Miasténicos Congénitos/patología , Síndromes Miasténicos Congénitos/fisiopatología , Examen Neurológico , Unión Neuromuscular/fisiología , Fenotipo , Bloqueadores de los Canales de Potasio/uso terapéutico , Bromuro de Piridostigmina/uso terapéutico
14.
Clin Exp Rheumatol ; 31(5): 767-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23806844

RESUMEN

OBJECTIVES: Various criteria have been proposed to classify the inflammatory myositides (IIMs) polymyositis (PM) and dermatomyositis (DM). However, none have received universal acceptance. Our aim was to assess the performance of the main criteria used to classify IIM. Specialist consultant diagnosis was considered the gold standard. METHODS: Patients attending King's College Hospital (KCH) or Reggio Emilia Hospital (REH) since 1990 with a diagnosis of IIM or non-inflammatory myopathy were identified, and their records and laboratory investigations retrospectively reviewed. Where the complete data required for the classification criteria or a final physician diagnosis was unavailable, patients were excluded. 52 patients with a specialist diagnosis of PM, DM, inclusion body myositis (IBM) or non-inflammatory myopathy were included. Agreement between specialist consultant diagnosis and classification criteria was measured using Cohen's kappa (κ) statistics. Sensitivity and specificity were also calculated. RESULTS: The Dalakas (2003) criteria demonstrated substantial agreement with specialist diagnosis: κ=0.69, sensitivity 77%, specificity 99%. The European Neuromuscular Centre criteria (ENMC) demonstrated fair agreement: κ=0.49, sensitivity 71%, specificity 82%. Other criteria performed less well. In particular, the Bohan and Peter criteria demonstrated a specificity of only 29%. CONCLUSIONS: The criteria of Dalakas (2003) agreed best with specialist consultant diagnosis. The criteria of Bohan and Peter demonstrated very poor specificity. Prospective studies are required to develop improved classification criteria.


Asunto(s)
Dermatomiositis/diagnóstico , Indicadores de Salud , Polimiositis/diagnóstico , Biomarcadores/sangre , Biopsia , Dermatomiositis/sangre , Dermatomiositis/clasificación , Electromiografía , Femenino , Humanos , Italia , Londres , Masculino , Persona de Mediana Edad , Examen Físico , Polimiositis/sangre , Polimiositis/clasificación , Valor Predictivo de las Pruebas , Derivación y Consulta , Estudios Retrospectivos , Índice de Severidad de la Enfermedad
15.
Hum Mutat ; 33(6): 981-8, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22473935

RESUMEN

Ryanodine receptor 1 (RYR1) mutations are a common cause of congenital myopathies associated with both dominant and recessive inheritance. Histopathological findings frequently feature central cores or multi-minicores, more rarely, type 1 predominance/uniformity, fiber-type disproportion, increased internal nucleation, and fatty and connective tissue. We describe 71 families, 35 associated with dominant RYR1 mutations and 36 with recessive inheritance. Five of the dominant mutations and 35 of the 55 recessive mutations have not been previously reported. Dominant mutations, typically missense, were frequently located in recognized mutational hotspot regions, while recessive mutations were distributed throughout the entire coding sequence. Recessive mutations included nonsense and splice mutations expected to result in reduced RyR1 protein. There was wide clinical variability. As a group, dominant mutations were associated with milder phenotypes; patients with recessive inheritance had earlier onset, more weakness, and functional limitations. Extraocular and bulbar muscle involvement was almost exclusively observed in the recessive group. In conclusion, our study reports a large number of novel RYR1 mutations and indicates that recessive variants are at least as frequent as the dominant ones. Assigning pathogenicity to novel mutations is often difficult, and interpretation of genetic results in the context of clinical, histological, and muscle magnetic resonance imaging findings is essential.


Asunto(s)
Mutación , Miopatías Estructurales Congénitas/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Niño , Preescolar , Femenino , Genes Dominantes , Genes Recesivos , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Linaje
16.
J Neurol Neurosurg Psychiatry ; 82(6): 671-3, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20392977

RESUMEN

AIM: To assess the treatment effects of rituximab in a population of patients with myasthenia gravis and Lambert-Eaton myasthenic syndrome. METHODS: Data on all treated patients in the UK were collected from referring physicians, with full case ascertainment and follow-up. RESULTS: Since 2004, 10 patients with generalised myasthenia gravis (three of whom were positive for muscle-specific tyrosine kinase (MuSK) antibodies) and two patients with Lambert-Eaton myasthenic syndrome (LEMS) were treated with rituximab. Using the Myasthenia Gravis Foundation America postintervention status, three patients (25%) achieved remission, and a further five (42%) improved clinically over an 18-month period. Only one patient developed worsening symptoms. The probability of achieving remission was unrelated to the duration of neurological symptoms prior to treatment. All LEMS and MuSK antibody patients improved following rituximab treatment. CONCLUSION: In a relatively large, unselected group of patients with myasthenia gravis and LEMS, rituximab treatment resulted in a significant clinical improvement in two-thirds of cases. As a selective, B cell targeted therapy, rituximab should be considered as a treatment option for patients with either myasthenia gravis or LEMS for whom standard immunosuppressive treatments have been unsuccessful.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Factores Inmunológicos/uso terapéutico , Síndrome Miasténico de Lambert-Eaton/tratamiento farmacológico , Miastenia Gravis/tratamiento farmacológico , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miastenia Gravis/inmunología , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores Colinérgicos/inmunología , Estudios Retrospectivos , Rituximab
18.
Brain ; 132(Pt 11): 3175-86, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19767415

RESUMEN

We have performed a detailed population study of patients with genetic muscle disease in the northern region of England. Our current clinic population comprises over 1100 patients in whom we have molecularly characterized 31 separate muscle disease entities. Diagnostic clarity achieved through careful delineation of clinical features supported by histological, immunological and genetic analysis has allowed us to reach a definitive diagnosis in 75.7% of our patients. We have compared our case profile with that from Walton and Nattrass' seminal study from 1954, also of the northern region, together with data from other more recent studies from around the world. Point prevalence figures for each of the five major disease categories are comparable with those from other recent studies. Myotonic dystrophies are the most common, comprising 28.6% of our clinic population with a point prevalence of 10.6/100,000. Next most frequent are the dystrophinopathies and facioscapulohumeral muscular dystrophy making up 22.9% (8.46/100,000) and 10.7% (3.95/100,000) of the clinic population, respectively. Spinal muscular atrophy patients account for 5.1% or 1.87/100,000 patients. Limb girdle muscular dystrophy, which was described for the first time in the paper by Walton and Nattrass (1954) and comprised 17% of their clinic population, comprises 6.2% of our clinic population at a combined prevalence of 2.27/100,000. The clinic population included patients with 12 other muscle disorders. These disorders ranged from a point prevalence of 0.89/100 000 for the group of congenital muscular dystrophies to conditions with only two affected individuals in a population of three million. For the first time our study provides epidemiological information for X-linked Emery-Dreifuss muscular dystrophy and the collagen VI disorders. Each of the X-linked form of Emery-Dreifuss muscular dystrophy and Ullrich muscular dystrophy has a prevalence of 0.13/100,000, making both very rare. Bethlem myopathy was relatively more common with a prevalence of 0.77/100,000. Overall our study provides comprehensive epidemiological information on individually rare inherited neuromuscular conditions in Northern England. Despite the deliberate exclusion of relatively common groups such as hereditary motor and sensory neuropathy (40/100,000) and mitochondrial disorders (9.2/100,000), the combined prevalence is 37.0/100,000, demonstrating that these disorders, taken as a group, encompass a significant proportion of patients with chronic disease. The study also illustrates the immense diagnostic progress since the first regional survey over 50 years ago by Walton and Nattrass.


Asunto(s)
Instituciones de Atención Ambulatoria , Músculo Esquelético , Distrofias Musculares , Adulto , Niño , Bases de Datos Factuales , Diagnóstico Diferencial , Inglaterra/epidemiología , Humanos , Músculo Esquelético/patología , Músculo Esquelético/fisiología , Músculo Esquelético/fisiopatología , Distrofias Musculares/epidemiología , Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Mutación
19.
Biochim Biophys Acta ; 1772(2): 238-42, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17123791

RESUMEN

The limb-girdle muscular dystrophies are a group of disorders where our understanding of their underlying molecular basis has made huge strides over the past years, revealing great heterogeneity at the clinical and molecular level. The availability of direct protein and/ or gene based approaches to diagnosis means that these disorders can now be precisely defined, and such definition of a precise diagnosis is increasingly allowing directed management for these diseases by the ability to predict specific complications such as those of the cardiac or respiratory systems. An algorithm combining clinical, biochemical and molecular testing is described which will aid precision of diagnosis and direct specific testing towards the cases most likely to benefit. This brings advantages for the patients of today in recognising the specific risks of their disorders, and in the future will be the starting point for specific gene and protein based therapies.


Asunto(s)
Distrofia Muscular de Cinturas/diagnóstico , Animales , Diagnóstico Diferencial , Humanos , Distrofia Muscular de Cinturas/complicaciones , Distrofia Muscular de Cinturas/fisiopatología , Distrofia Muscular de Cinturas/terapia
20.
Acta Neuropathol Commun ; 4(1): 44, 2016 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-27121971

RESUMEN

Mutations of the human plectin gene (PLEC) on chromosome 8q24 cause autosomal recessive epidermolysis bullosa simplex with muscular dystrophy (EBS-MD). In the present study we analyzed the downstream effects of PLEC mutations on plectin protein expression and localization, the structure of the extrasarcomeric desmin cytoskeleton, protein aggregate formation and mitochondrial distribution in skeletal muscle tissue from three EBS-MD patients. PLEC gene analysis in a not previously reported 35-year-old EBS-MD patient with additional disease features of cardiomyopathy and malignant arrhythmias revealed novel compound heterozygous (p.(Phe755del) and p.(Lys1040Argfs*139)) mutations resulting in complete abolition of plectin protein expression. In contrast, the other two patients with different homozygous PLEC mutations showed preserved plectin protein expression with one only expressing rodless plectin variants, and the other markedly reduced protein levels. Analysis of skeletal muscle tissue from all three patients revealed severe disruption of the extrasarcomeric intermediate filament cytoskeleton, protein aggregates positive for desmin, syncoilin, and synemin, degenerative myofibrillar changes, and mitochondrial abnormalities comprising respiratory chain dysfunction and an altered organelle distribution and amount.Our study demonstrates that EBS-MD causing PLEC mutations universally result in a desmin protein aggregate myopathy phenotype despite marked differences in individual plectin protein expression patterns. Since plectin is the key cytolinker protein that regulates the structural and functional organization of desmin filaments, the defective anchorage and spacing of assembled desmin filaments is the key pathogenetic event that triggers the formation of desmin protein aggregates as well as secondary mitochondrial pathology.


Asunto(s)
Epidermólisis Ampollosa Simple/genética , Epidermólisis Ampollosa Simple/metabolismo , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo , Mutación , Plectina/genética , Plectina/metabolismo , Adulto , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Epidermólisis Ampollosa Simple/patología , Femenino , Humanos , Mitocondrias/metabolismo , Mitocondrias/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/patología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA