Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neurol ; 263(3): 517-23, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26754003

RESUMEN

Congenital myopathies are a group of inherited muscle disorders characterized by hypotonia, weakness and a non-dystrophic muscle biopsy with the presence of one or more characteristic histological features. Neuromuscular transmission defects have recently been reported in several patients with congenital myopathies (CM). Mutations in KLHL40 are among the most common causes of severe forms of nemaline myopathy. Clinical features of affected individuals include fetal akinesia or hypokinesia, respiratory failure, and swallowing difficulties at birth. Muscle weakness is usually severe and nearly half of the individuals have no spontaneous antigravity movement. The average age of death has been reported to be 5 months in a recent case series. Herein we present a case of a patient with a nemaline myopathy due to KLHL40 mutations (c.604delG, p.Ala202Argfs*56 and c.1513G>C, p.Ala505Pro) with an impressive and prolonged beneficial response to treatment with high-dose pyridostigmine. Myasthenic features or response to ACEI have not previously been reported as a characteristic of nemaline myopathy or KLHL40-related myopathy.


Asunto(s)
Inhibidores de la Colinesterasa/uso terapéutico , Proteínas Musculares/genética , Miopatías Nemalínicas/tratamiento farmacológico , Miopatías Nemalínicas/genética , Femenino , Humanos , Lactante , Estudios Longitudinales , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miopatías Nemalínicas/patología , Examen Neurológico
2.
Sci Rep ; 6(1): 15, 2016 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-28442759

RESUMEN

Laboratory data interpretation for the assessment of complex biological systems remains a great challenge, as occurs in mitochondrial function research studies. The classical biochemical data interpretation of patients versus reference values may be insufficient, and in fact the current classifications of mitochondrial patients are still done on basis of probability criteria. We have developed and applied a mathematic agglomerative algorithm to search for correlations among the different biochemical variables of the mitochondrial respiratory chain in order to identify populations displaying correlation coefficients >0.95. We demonstrated that coenzyme Q10 may be a better biomarker of mitochondrial respiratory chain enzyme activities than the citrate synthase activity. Furthermore, the application of this algorithm may be useful to re-classify mitochondrial patients or to explore associations among other biochemical variables from different biological systems.


Asunto(s)
Algoritmos , Citrato (si)-Sintasa/análisis , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Mitocondrias Musculares/enzimología , Ubiquinona/análogos & derivados , Adolescente , Biomarcadores/análisis , Niño , Preescolar , Transporte de Electrón , Humanos , Lactante , Enfermedades Mitocondriales/enzimología , Ubiquinona/análisis
3.
Neuromuscul Disord ; 22(2): 139-48, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22075033

RESUMEN

Mutations in COL6A1, COL6A2 and COL6A3 genes result in collagen VI myopathies: Ullrich congenital muscular dystrophy (UCMD), Bethlem myopathy (BM) and intermediate phenotypes. At present, none of the existing diagnostic techniques for evaluating collagen VI expression is quantitative, and the detection of subtle changes in collagen VI expression remains challenging. We investigated flow cytometry analysis as a means of quantitatively measuring collagen VI in primary fibroblasts and compared this method with the standard method of fibroblast collagen VI immunohistochemical analysis. Eight UCMD and five BM molecularly confirmed patients were studied and compared to five controls. Flow cytometry analysis consistently detected a reduction of collagen VI of at least 60% in all UCMD cases. In BM cases the levels of collagen VI were variable but on average 20% less than controls. Flow cytometry analysis provides an alternative method for screening for collagen VI deficiency at the protein level in a quantitative, time and cost-effective manner.


Asunto(s)
Colágeno Tipo VI/deficiencia , Citometría de Flujo , Distrofias Musculares/diagnóstico , Adolescente , Adulto , Niño , Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Fibroblastos/metabolismo , Humanos , Persona de Mediana Edad , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mutación/genética , Adulto Joven
4.
Neurology ; 73(1): 25-31, 2009 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-19564581

RESUMEN

OBJECTIVE: To describe the course, complications, and prognosis of Ullrich congenital muscular dystrophy (UCMD), with special reference to life-changing events, including loss of ambulation, respiratory insufficiency, and death. METHODS: Review of the case notes of 13 patients with UCMD, aged 15 years or older at last visit, followed up at a tertiary neuromuscular centre, London, UK, from 1977 to 2007. Data collected were age at onset of symptoms, presenting symptoms, mobility, contractures, scoliosis, skin abnormalities, respiratory function, and feeding difficulties. RESULTS: The mean age at onset of symptoms was 12 months (SD 14 months). Eight patients (61.5%) acquired independent ambulation at a mean age of 1.7 years (SD 0.8 years). Nine patients (69.2%) became constant wheelchair users at a mean age of 11.1 years (SD 4.8 years). Three patients continued to ambulate indoors with assistance. Forced vital capacity (FVC) values were abnormal in all patients from age 6 years. The mean FVC (% predicted) declined at a mean rate of 2.6% (SD 4.1%) yearly. Nine patients (69.2%) started noninvasive ventilation at a mean age of 14.3 years (SD 5.0 years). Two patients died of respiratory insufficiency. CONCLUSION: In Ullrich congenital muscular dystrophy (UCMD), the decline in motor and respiratory functions is more rapid in the first decade of life. The deterioration is invariable, but not always correlated with age or severity at presentation. This information should be of help to better anticipate the difficulties encountered by patients with UCMD and in planning future therapeutic trials in this condition.


Asunto(s)
Debilidad Muscular/diagnóstico , Distrofias Musculares/diagnóstico , Parálisis Respiratoria/diagnóstico , Adolescente , Adulto , Distribución por Edad , Edad de Inicio , Niño , Estudios de Cohortes , Colágeno Tipo VI/genética , Comorbilidad , Contractura/etiología , Contractura/fisiopatología , Evaluación de la Discapacidad , Progresión de la Enfermedad , Humanos , Estudios Longitudinales , Limitación de la Movilidad , Mortalidad , Debilidad Muscular/mortalidad , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofias Musculares/congénito , Distrofias Musculares/mortalidad , Mutación/genética , Parálisis Respiratoria/mortalidad , Índice de Severidad de la Enfermedad , Capacidad Vital/genética , Adulto Joven
5.
Brain ; 132(Pt 1): 147-55, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19015158

RESUMEN

Mutations in COL6A1, COL6A2 and COL6A3, the genes which encode the extra-cellular matrix component collagen VI, lead to Bethlem myopathy and Ullrich congenital muscular dystrophy (UCMD). Although the Col6a1(-/-) null mouse has an extremely mild neuromuscular phenotype, a mitochondrial defect has been demonstrated, linked to dysregulation of the mitochondrial permeability transition pore (PTP) opening. This finding has been replicated in UCMD muscle cells in culture, providing justification for a clinical trial using cyclosporine A, an inhibitor of PTP opening. We investigated whether PTP dysregulation could be detected in UCMD fibroblasts (the predominant source of muscle collagen VI), in myoblast cells from patients with other diseases and its response to rescue agents other than collagen VI. Although we confirm the presence of PTP dysregulation in muscle-derived cultures from two UCMD patients, fibroblasts from the same patients and the majority of fibroblasts from other well-characterized UCMD patients behave normally. PTP dysregulation is found in limb girdle muscular dystrophy (LGMD) type 2B myoblasts but not in myoblasts from patients with Bethlem myopathy, merosin-deficient congenital muscular dystrophy, LGMD2A, Duchenne muscular dystrophy and Leigh syndrome. In addition to rescue by cyclosporine A and collagen VI, this cellular phenotype was also rescued by other extra-cellular matrix constituents (laminin and collagen I). As the muscle derived cultures demonstrating PTP dysregulation shared poor growth in culture and lack of desmin labelling, we believe that PTP dysregulation may be a particular characteristic of the state of these cells in culture and is not specific to the collagen VI defect, and can in any case be rescued by a range of extra-cellular matrix components. Further work is needed on the relationship of PTP dysregulation with UCMD pathology.


Asunto(s)
Ciclosporina/farmacología , Mitocondrias/fisiología , Distrofias Musculares/patología , Adolescente , Células Cultivadas , Niño , Preescolar , Colágeno Tipo VI/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Lactante , Recién Nacido , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/fisiología , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/metabolismo , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Rodaminas , Piel/metabolismo , Adulto Joven
6.
J Neuroimmunol ; 201-202: 6-12, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18707767

RESUMEN

The Congenital Myasthenic Syndromes (CMS), a group of heterogeneous genetic disorders of neuromuscular transmission, are often misdiagnosed as congenital muscular dystrophy (CMD) or myopathies and present particular management problems. We present our experience of 46 children with CMS, referred to us between 1992-2007 with provisional diagnoses of congenital myopathy (22/46), CMS or limb-girdle myasthenia (9/46), central hypotonia or neurometabolic disease (5/46), myasthenia gravis (4/46), limb-girdle or congenital muscular dystrophy (4/46) and SMA (2/46). Diagnosis was often considerably delayed (up to 18y4 m), despite the early symptoms in most cases. Diagnostic clues in the neonates were feeding difficulties (29/46), hypotonia with or without limb weakness (21/46), ptosis (19/46), respiratory insufficiency (12/46), contractures (4/46) and stridor (6/46). Twenty-five children had delayed motor milestones. Fatigability developed in 43 and a variable degree of ptosis was eventually present in 40. Over the period of the study, the mainstay of EMG diagnosis evolved from repetitive nerve stimulation to stimulation single fibre EMG. The patients were studied by several different operators. 66 EMGs were performed in 40 children, 29 showed a neuromuscular junction abnormality, 7 were myopathic, 2 had possible neurogenic changes and 28 were normal or inconclusive. A repetitive CMAP was detected in only one of seven children with a COLQ mutation and neither of the two children with Slow Channel Syndrome mutations. Mutations have been identified so far in 32/46 children: 10 RAPSN, 7 COLQ, 6 CHRNE, 7 DOK7, 1 CHRNA1 and 1 CHAT. 24 of 25 muscle biopsies showed myopathic changes with fibre size variation; 14 had type-1 fibre predominance. Three cases showed small type-1 fibres resembling fibre type disproportion, and four showed core-like lesions. No specific myopathic features were associated with any of the genes. Twenty children responded to Pyridostigmine treatment alone, 11 to Pyridostigmine with either 3, 4 DAP or Ephedrine and five to Ephedrine alone. Twenty one children required acute or chronic respiratory support, with tracheostomy in 4 and nocturnal or emergency non-invasive ventilation in 9. Eight children had gastrostomy. Another 11 were underweight for height indicative of failure to thrive and required dietetic input. A high index of clinical suspicion, repeat EMG by an experienced electromyographer and, if necessary, a therapeutic trial of Pyridostigmine facilitates the diagnosis of CMS with subsequent molecular genetic confirmation. This guides rational therapy and multidisciplinary management, which may be crucial for survival, particularly in pedigrees where previous deaths have occurred in infancy.


Asunto(s)
Síndromes Miasténicos Congénitos/diagnóstico , Síndromes Miasténicos Congénitos/terapia , Adolescente , Edad de Inicio , Biopsia/métodos , Niño , Preescolar , Análisis Mutacional de ADN , Electromiografía , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Mutación , Síndromes Miasténicos Congénitos/clasificación , Síndromes Miasténicos Congénitos/fisiopatología , Respiración , Estudios Retrospectivos
7.
Hum Mutat ; 29(6): 809-22, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18366090

RESUMEN

Mutations in the genes encoding collagen VI (COL6A1, COL6A2, and COL6A3) cause Bethlem myopathy (BM) and Ullrich congenital muscular dystrophy (UCMD), two related conditions of differing severity. BM is a relatively mild dominantly inherited disorder characterized by proximal weakness and distal joint contractures. UCMD was originally regarded as an exclusively autosomal recessive condition causing severe muscle weakness with proximal joint contractures and distal hyperlaxity. We and others have subsequently modified this model when we described UCMD patients with heterozygous in-frame deletions acting in a dominant-negative way. Here we report 10 unrelated patients with a UCMD clinical phenotype and de novo dominant negative heterozygous splice mutations in COL6A1, COL6A2, and COL6A3 and contrast our findings with four UCMD patients with recessively acting splice mutations and two BM patients with heterozygous splice mutations. We find that the location of the skipped exon relative to the molecular structure of the collagen chain strongly correlates with the clinical phenotype. Analysis by immunohistochemical staining of muscle biopsies and dermal fibroblast cultures, as well as immunoprecipitation to study protein biosynthesis and assembly, suggests different mechanisms each for exon skipping mutations underlying dominant UCMD, dominant BM, and recessive UCMD. We provide further evidence that de novo dominant mutations in severe UCMD occur relatively frequently in all three collagen VI chains and offer biochemical insight into genotype-phenotype correlations within the collagen VI-related disorders by showing that severity of the phenotype depends on the ability of mutant chains to be incorporated in the multimeric structure of collagen VI.


Asunto(s)
Colágeno Tipo VI/genética , Distrofias Musculares/genética , Mutación , Empalme del ARN , Células Cultivadas , Colágeno Tipo VI/metabolismo , Análisis Mutacional de ADN , Exones , Fibroblastos/metabolismo , Eliminación de Gen , Humanos , Músculo Esquelético/metabolismo , Índice de Severidad de la Enfermedad , Piel/citología
8.
Neuromuscul Disord ; 17(2): 174-9, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17236770

RESUMEN

Severe diaphragmatic weakness in infancy is rare. Common causes include structural myopathies, neuromuscular transmission defects, or anterior horn cell dysfunction (spinal muscular atrophy with respiratory distress, SMARD1). We describe a form of infantile diaphragmatic weakness without mutations in the SMARD1 gene, in which pathological and clinical features differ from known conditions, and investigations suggest a myopathy. We identified seven cases in four families. All presented soon after birth with feeding and breathing difficulties, marked head lag, facial weakness, and preserved antigravity movements in the limbs, with arms weaker than legs. All had paradoxical breathing and paralysis of at least one hemi-diaphragm. All required gastrostomy feeding, and all became ventilator-dependent. Investigations included myopathic EMG, muscle biopsy showing myopathic changes, normal electrophysiology and no mutations in SMN1 or IGHMBP2. These seven infants are affected by a myopathic condition clinically resembling SMARD1. However, its pathogenesis appears to be a myopathy affecting predominantly the diaphragm.


Asunto(s)
Proteínas de Unión al ADN/genética , Diafragma/fisiopatología , Debilidad Muscular/congénito , Debilidad Muscular/genética , Enfermedades Musculares/congénito , Enfermedades Musculares/genética , Factores de Transcripción/genética , Creatina Quinasa/metabolismo , Electromiografía , Nutrición Enteral , Extremidades/fisiopatología , Músculos Faciales/fisiopatología , Femenino , Crecimiento/fisiología , Humanos , Lactante , Recién Nacido , Movimiento/fisiología , Debilidad Muscular/fisiopatología , Enfermedades Musculares/fisiopatología , Respiración Artificial , Mecánica Respiratoria/fisiología
9.
Acta Myol ; 26(3): 129-35, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18646561

RESUMEN

Muscular dystrophies are a clinically and genetically heterogeneous group of disorders. Until recently most of the proteins associated with muscular dystrophies were believed to be proteins of the sarcolemma associated with reinforcing the plasma membrane or in facilitating its re-sealing following injury. In the last few years a novel and frequent pathogenic mechanism has been identified that involves the abnormal glycosylation of alpha-dystroglycan (ADG). This peripheral membrane protein undergoes complex and crucial glycosylation steps that enable it to interact with LG domain containing extracellular matrix proteins such as laminins, agrin and perlecan. Mutations in six genes (POMT1, POMT2, POMGnT1, fukutin, FKRP and LARGE) have been identified in patients with reduced glycosylation of ADG. While initially a clear correlation between gene defect and phenotype was observed for each of these 6 genes (for example, Walker Warburg syndrome was associated with mutations in POMT1 and POMT2, Fukuyama congenital muscular dystrophy associated with fukutin mutations, and Muscle Eye Brain disease associated with POMGnT1 mutations), we have recently demonstrated that allelic mutations in each of these 6 genes can result in a much wider spectrum of clinical conditions. Thus, the crucial aspect in determining the phenotypic severity is not which gene is primarily mutated, but how severely the mutation affects the glycosylation of ADG. Systematic mutation analysis of these 6 glycosyltransferases in patients with a dystroglycan glycosylation disorder identifies mutations in approximately 65% suggesting that more genes have yet to be identified.


Asunto(s)
Distroglicanos/metabolismo , Distrofias Musculares/metabolismo , ADN/genética , Distroglicanos/genética , Glicosilación , Humanos , Distrofias Musculares/genética , Mutación
10.
Neuromuscul Disord ; 16(9-10): 571-82, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16935502

RESUMEN

Ullrich congenital muscular dystrophy (UCMD) is caused by recessive and dominant mutations in COL6A genes. We have analysed collagen VI expression in 14 UCMD patients. Sequencing of COL6A genes had identified homozygous and heterozygous mutations in 12 cases. Analysis of collagen VI in fibroblast cultures derived from eight of these patients showed reduced extracellular deposition in all cases and intracellular collagen VI staining in seven cases. This was observed even in cases that showed normal collagen VI labelling in skin biopsies. Collagen VI immunolabelling was reduced in all the available muscle biopsies. When comparisons were possible no correlation was seen between the extent of the reduction in the muscle and fibroblast cultures, the mode of inheritance or the severity of the clinical phenotype. Mutations affecting glycine substitutions in the conserved triple helical domain were common and all resulted in reduced collagen VI. This study expands the spectrum of collagen VI defects and shows that analysis of skin fibroblasts may be a useful technique for the detection of collagen VI abnormalities. In contrast, immunohistochemical analysis of skin biopsies may not always reveal an underlying collagen VI defect.


Asunto(s)
Colágeno Tipo VI/genética , Fibroblastos/metabolismo , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Piel/metabolismo , Adolescente , Adulto , Células Cultivadas , Niño , Colágeno Tipo VI/biosíntesis , Análisis Mutacional de ADN , Progresión de la Enfermedad , Fibroblastos/patología , Genes Dominantes/genética , Genes Recesivos/genética , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Pruebas Genéticas , Humanos , Inmunohistoquímica/normas , Patrón de Herencia/genética , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofias Musculares/fisiopatología , Mutación/genética , Fenotipo , Valor Predictivo de las Pruebas , Piel/patología , Piel/fisiopatología
11.
Cell Mol Life Sci ; 62(7-8): 809-23, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15868406

RESUMEN

The congenital muscular dystrophies are a clinically and genetically heterogeneous group of neuromuscular disorders. Each form has a characteristic phenotype, but there is overlap between some entities and their classification is based on a combination of clinical features and the primary or secondary protein defect. Recent studies have identified the genetic basis of a number of congenital muscular dystrophies (11 genes in total) and have recognised a novel pathological mechanism that highlights the importance of the correct posttranslational processing of proteins, in particular alpha-dystroglycan. Diagnosis of these conditions has been aided by the availability of specific antibodies for each protein and a better understanding of the protein changes that accompany each condition. In this review we present the major molecular, clinical and diagnostic aspects of each group of congenital muscular dystrophy with an emphasis in the more recent developments.


Asunto(s)
Distroglicanos/metabolismo , Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Antígenos CD/metabolismo , Humanos , Cadenas alfa de Integrinas/metabolismo , Laminina/metabolismo , Músculo Esquelético/fisiopatología , Distrofias Musculares/congénito , Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Procesamiento Proteico-Postraduccional
12.
Neuropathol Appl Neurobiol ; 30(5): 540-5, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15488030

RESUMEN

Immunohistochemistry using antibodies to dystrophin is the pathological basis for the diagnosis of Duchenne and Becker muscular dystrophy (DMD and BMD). While the sarcolemma of DMD muscle is negative, BMD muscle generally shows variable labelling because of the translation of a partially functional dystrophin that is localized to the sarcolemma. In rare cases, however, this labelling is equivocal and similar to that observed in controls making diagnosis difficult. We report here that in such instances immunolabelling with antibodies to the neuronal form of nitric oxide synthase (nNOS) can be useful in suspecting a dystrophinopathy with a mutation in the 'hot-spot' rod domain and help to direct molecular analysis. nNOS localizes to the sarcolemma of mature muscle fibres via several components of the dystrophin-associated protein complex (DAPC) including dystrophin but sarcolemmal nNOS is lost when dystrophin levels are very low or absent because of deletions in critical regions of the rod domain. We report three cases who presented with only mild or no muscle weakness but had elevated serum creatine kinase activity and dystrophin immunolabelling indistinguishable from normal, making a pathological diagnosis difficult. All three cases had a complete absence of sarcolemmal nNOS and were subsequently found to have an in-frame deletion in the common rod domain exons (in these cases 48, 45-51, 47-53) compatible with a BMD. In addition, we observed that nNOS appears to be developmentally regulated with the antibody used and was often absent from the sarcolemma of immature fibres. These findings demonstrate the value of including antibodies to nNOS in routine immunohistochemical studies and that absence of nNOS can be a more sensitive marker than up-regulation of utrophin for diagnosis of BMD. Immaturity of fibres, however, needs to be taken into account, especially in neonates.


Asunto(s)
Distrofina/genética , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/metabolismo , Óxido Nítrico Sintasa/metabolismo , Biomarcadores , Biopsia , Niño , Preescolar , Creatina Quinasa/sangre , Diagnóstico Diferencial , Humanos , Inmunohistoquímica , Músculo Esquelético/metabolismo , Mutación , Óxido Nítrico Sintasa de Tipo I , Reacción en Cadena de la Polimerasa , Sarcolema/enzimología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA