Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Genet Metab ; 112(2): 183-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24745848

RESUMEN

Phospholipase A2 associated neurodegeneration (PLAN) is a major phenotype of autosomal recessive Neurodegeneration with Brain Iron Accumulation (NBIA). We describe the clinical phenotypes, neuroimaging features and PLA2G6 mutations in 5 children, of whom 4 presented with infantile neuroaxonal dystrophy (INAD). One other patient was diagnosed with the onset of PLAN in childhood, and our report highlights the diagnostic challenges associated with this atypical PLAN subtype. In this series, the neuroradiological relevance of classical PLAN features as well as apparent claval hypertrophy' is explored. Novel PLA2G6 mutations were identified in all patients. PLAN should be considered not only in patients presenting with a classic INAD phenotype but also in older patients presenting later in childhood with non-specific progressive neurological features including social communication difficulties, gait disturbance, dyspraxia, neuropsychiatric symptoms and extrapyramidal motor features.


Asunto(s)
Fosfolipasas A2 Grupo VI/genética , Distrofias Neuroaxonales/diagnóstico por imagen , Distrofias Neuroaxonales/patología , Edad de Inicio , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Preescolar , Femenino , Variación Genética , Humanos , Lactante , Irlanda , Masculino , Mutación , Distrofias Neuroaxonales/genética , Fenotipo , Radiografía , Reino Unido
2.
Ann Neurol ; 68(5): 717-26, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20839240

RESUMEN

OBJECTIVE: Centronuclear myopathy (CNM) is a rare congenital myopathy characterized by prominence of central nuclei on muscle biopsy. CNM has been associated with mutations in MTM1, DNM2, and BIN1 but many cases remain genetically unresolved. RYR1 encodes the principal sarcoplasmic reticulum calcium release channel and has been implicated in various congenital myopathies. We investigated whether RYR1 mutations cause CNM. METHODS: We sequenced the entire RYR1 coding sequence in 24 patients with a diagnosis of CNM from South Africa (n = 14) and Europe (n = 10) and identified mutations in 17 patients. The most common genotypes featured compound heterozygosity for RYR1 missense mutations and mutations resulting in reduced protein expression, including intronic splice site and frameshift mutations. RESULTS: The high incidence in South African patients (n = 12/14) in conjunction with recurrent RYR1 mutations associated with common haplotypes suggested the presence of founder effects. In addition to central nuclei, prominent histopathological findings included (often multiple) internalized nuclei and type 1 fiber predominance and hypotrophy with relative type 2 hypertrophy. Although cores were not typically seen on oxidative stains, electron microscopy revealed subtle abnormalities in most cases. External ophthalmoplegia, proximal weakness, and bulbar involvement were prominent clinical findings. INTERPRETATION: Our findings expand the range of RYR1-related phenotypes and suggest RYR1 mutations as a common cause of congenital myopathies with central nuclei. Corresponding to recent observations in X-linked CNM, these findings indicate disturbed assembly and/or malfunction of the excitation-contraction machinery as a key mechanism in CNM and related myopathies.


Asunto(s)
Músculo Esquelético/patología , Miopatías Estructurales Congénitas/etiología , Canal Liberador de Calcio Receptor de Rianodina/genética , Adolescente , Niño , Preescolar , Europa (Continente) , Genotipo , Heterocigoto , Humanos , Masculino , Mutación , Miopatías Estructurales Congénitas/diagnóstico , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología , Fenotipo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Sudáfrica
3.
J Neurol Neurosurg Psychiatry ; 80(7): 706-14, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19531685

RESUMEN

Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD) and limb girdle muscular dystrophies (LGMD) represent a significant proportion of paediatric and adult neuromuscular neurology practice. The proactive symptom-based multidisciplinary team (MDT) management and access to non-invasive ventilation have enabled improved survival into adulthood. Nevertheless the severe disability imposed by conditions such as DMD poses a challenge for successful transition of care and management for paediatric and adult neurology teams. DMD is discussed in detail as a paradigm illustrating diagnosis, management and role for different pharmacological interventions to improve survival, but also challenges in adulthood care, and cutting-edge therapies. LGMDs are much rarer than DMD and BMD, and in addition there is a significant genetic and clinical heterogeneity, which leads to diagnostic difficulties. The clinical and laboratory diagnostic features of seven LGMD subtypes are summarised, and their allelic "non-limb girdle" phenotypes are tabulated to illustrate the theme of one gene causing multiple clinical phenotypes, with the aim of refining the clinician's diagnostic approach. The lessons learnt from DMD MDT management to improve survival are broadly applicable to LGMDs with severe motor disability/multisystem complications.


Asunto(s)
Corticoesteroides/uso terapéutico , Distrofias Musculares/diagnóstico , Distrofias Musculares/terapia , Modalidades de Fisioterapia , Síndromes de la Apnea del Sueño/terapia , Adulto , Niño , Humanos , Distrofias Musculares/complicaciones , Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Distrofia Muscular de Cinturas/diagnóstico , Distrofia Muscular de Cinturas/terapia , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/terapia , Síndromes de la Apnea del Sueño/etiología , Resultado del Tratamiento
4.
Postgrad Med J ; 85(1009): 622-30, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19892898

RESUMEN

Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD) and limb girdle muscular dystrophies (LGMD) represent a significant proportion of paediatric and adult neuromuscular neurology practice. The proactive symptom-based multidisciplinary team (MDT) management and access to non-invasive ventilation have enabled improved survival into adulthood. Nevertheless the severe disability imposed by conditions such as DMD poses a challenge for successful transition of care and management for paediatric and adult neurology teams. DMD is discussed in detail as a paradigm illustrating diagnosis, management and role for different pharmacological interventions to improve survival, but also challenges in adulthood care, and cutting-edge therapies. LGMDs are much rarer than DMD and BMD, and in addition there is a significant genetic and clinical heterogeneity, which leads to diagnostic difficulties. The clinical and laboratory diagnostic features of seven LGMD subtypes are summarised, and their allelic "non-limb girdle" phenotypes are tabulated to illustrate the theme of one gene causing multiple clinical phenotypes, with the aim of refining the clinician's diagnostic approach. The lessons learnt from DMD MDT management to improve survival are broadly applicable to LGMDs with severe motor disability/multisystem complications.

5.
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
6.
Cochrane Database Syst Rev ; (1): CD003725, 2008 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-18254031

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy of childhood. This incurable disease is characterised by muscle wasting and loss of walking ability leading to complete wheelchair dependence by 13 years of age. Prolongation of walking is one of the major aims of treatment. OBJECTIVES: The aim of this review was to assess whether glucocorticoid corticosteroids stabilize or improve muscle strength and walking in boys with DMD. SEARCH STRATEGY: This is an update of the Cochrane systematic review first published in 2004 (Manzur 2004). We searched the Cochrane Neuromuscular Disease Group Trials Register (August 2006) using the term 'Duchenne muscular dystrophy'. We also searched MEDLINE (January 1966 to July 2007), EMBASE (January 1980 to August 2006), CINAHL and LILACS (January 1982 to August 2006). We wrote to authors of published studies and other experts in this disease to help identify other trials, checked the references in the identified trials and hand searched the abstracts of relevant journals. SELECTION CRITERIA: Types of studies: randomised or quasi-randomised trials. TYPES OF PARTICIPANTS: all patients with a definite diagnosis of Duchenne muscular dystrophy. Types of interventions: glucocorticoids such as prednisone, prednisolone, deflazacort or others, with a minimum treatment period of three months. PRIMARY OUTCOME MEASURE: prolongation of walking (independent walking without long leg calipers). SECONDARY OUTCOME MEASURES: strength outcome measures, manual muscle strength testing using Medical Research Council strength scores, functional outcome measures and adverse events. DATA COLLECTION AND ANALYSIS: We identified six randomised controlled trials that met the inclusion criteria for our review, and one of these (Beenakker 2005) is a new addition to this update, as it was published subsequent to our first review (Manzur 2004). Two review authors independently selected the trials for the review and assessed methodological quality. Data extraction and inputting were double-checked. PRIMARY OUTCOME MEASURE: data from one small study used prolongation of walking as an outcome measure and did not show significant benefit. SECONDARY OUTCOME MEASURES: The meta-analysis of the results from four randomised controlled trials with altogether 249 participants showed that glucocorticoid corticosteroids improved muscle strength and function over six months. Improvements were seen in time taken to rise from the floor (Gowers' time), nine metres walking time, four-stair climbing time, ability to lift weights, leg function grade and forced vital capacity. One randomised controlled trial with altogether 28 participants showed that glucocorticoid corticosteroids stabilize muscle strength and function for up to two years. The most effective prednisolone regime appears to be 0.75 mg/kg/day, given in a daily dose regime. Not enough data were available to compare efficacy of prednisone with deflazacort. Adverse effects: Excessive weight gain, behavioural abnormalities, cushingoid appearance and excessive hair growth were all more common with glucocorticoid corticosteroids than placebo. Long-term adverse effects of glucocorticoid therapy could not be evaluated because of the short-term duration of the randomised studies.Non-randomised studies: A number of non-randomised studies with important efficacy and adverse effects data are tabulated and discussed. AUTHORS' CONCLUSIONS: There is evidence from randomised controlled studies that glucocorticoid corticosteroid therapy in Duchenne muscular dystrophy improves muscle strength and function in the short-term (six months to two years). The most effective prednisolone regime appears to be 0.75 mg/kg/day, given daily. In the short term, adverse effects were significantly more common but not clinically severe. Long-term benefits and hazards of glucocorticoid treatment cannot be evaluated from the currently published randomised studies. Non-randomised studies support the conclusions of functional benefits but also identify clinically significant adverse effects of long-term treatment. These benefits and adverse effects have implications for future research studies and clinical practice.


Asunto(s)
Corticoesteroides/uso terapéutico , Glucocorticoides/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Corticoesteroides/efectos adversos , Glucocorticoides/efectos adversos , Humanos , Masculino , Prednisolona/uso terapéutico , Prednisona/uso terapéutico , Pregnenodionas/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Caminata
7.
Neuromuscul Disord ; 28(9): 757-765, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30072201

RESUMEN

Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is a treatable chronic disorder of the peripheral nervous system. We retrospectively studied 30 children with a suspected diagnosis of CIDP. The diagnosis of CIDP was compared against the childhood CIDP revised diagnostic criteria 2000. Of the 30 children, five did not meet the criteria and four others met the criteria but subsequently had alternative diagnosis, leaving a total of 21 children (12 male) with CIDP as the final diagnosis. Thirteen children presented with chronic symptom-onset (>8 weeks). The majority presented with gait difficulties or pain in legs (n = 16). 12 children (57%) met the neurophysiological criteria and 18/19 (94%) met the cerebrospinal fluid criteria. Nerve biopsy was suggestive in 3/9 (33%), with magnetic resonance imaging supportive in 9/20 (45%). Twenty-one children received immuno-modulatory treatment at first presentation, of which majority (n = 19, 90%) received IVIG (immunoglobulin) monotherapy with 13 (68%) showing a good response. 8 children received second line treatment with either IVIG or steroids or plasmapharesis (PE) and 4 needed other immune-modulatory agents. During a median follow-up of 3.6 years, 9 (43%) had a monophasic course and 12 (57%) had a relapsing-remitting course. At last paediatric follow up 7 (33%) were off all treatment, 9 (43%) left with no or minimal residual disability and 6 (28%) children were walking with assistance (n = 3) or were non-ambulant (n = 3). Our review highlights challenges in the diagnosis and management of paediatric CIDP. It also confirms that certain metabolic disorders may mimic CIDP.


Asunto(s)
Marcha/fisiología , Inmunoglobulinas Intravenosas/uso terapéutico , Factores Inmunológicos/uso terapéutico , Plasmaféresis , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/diagnóstico , Adolescente , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Conducción Nerviosa/fisiología , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/fisiopatología , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/terapia , Estudios Retrospectivos , Resultado del Tratamiento
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.
AJNR Am J Neuroradiol ; 28(7): 1223-31, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17698520

RESUMEN

BACKGROUND AND PURPOSE: Brain imaging is an integral part of the diagnostic work-up for metabolic disorders, and the bedside availability of cranial ultrasonography (cUS) allows very early brain imaging in symptomatic neonates. Our aim was to investigate the role and range of abnormalities seen on cUS in neonates presenting with metabolic disorders. A secondary aim, when possible, was to address the question of whether brain MR imaging is more informative by comparing cUS to MR imaging findings. MATERIALS AND METHODS: Neonates with a metabolic disorder who had at least 1 cUS scan were eligible. cUS images were reviewed for anatomic and maturation features, cysts, calcium, and other abnormalities. When an MR imaging scan had been obtained, both sets of images were compared. RESULTS: Fifty-five infants (35 also had MR imaging) were studied. The most frequent findings were in oxidative phosphorylation disorders (21 cUS and 12 MR imaging): ventricular dilation (11 cUS and 6 MR imaging), germinolytic cysts (GLCs; 7 cUS and 5 MR imaging), and abnormal white matter (7 cUS and 6 MR imaging); in peroxisomal biogenesis disorders (13 cUS and 9 MR imaging): GLCs (10 cUS and 6 MR imaging), ventricular dilation (10 cUS and 5 MR imaging), abnormal cortical folding (8 cUS and 7 MR imaging), and lenticulostriate vasculopathy (8 cUS); in amino acid metabolism and urea cycle disorders (14 cUS and 11 MR imaging): abnormal cortical folding (9 cUS and 4 MR imaging), abnormal white matter (8 cUS and 8 MR imaging), and hypoplasia of the corpus callosum (7 cUS and 6 MR imaging); in organic acid disorders (4 cUS and 2 MR imaging): periventricular white matter echogenicity (2 cUS and 1 MR imaging); and in other disorders (3 cUS and 1 MR imaging): ventricular dilation (2 cUS and 1 MR imaging). cUS findings were consistent with MR imaging findings. cUS was better for visualizing GLCs and calcification. MR imaging was more sensitive for subtle tissue signal intensity changes in the white matter and abnormality in areas difficult to visualize with cUS, though abnormalities of cortical folding suggestive of polymicrogyria were seen on cUS. CONCLUSION: A wide range of abnormalities is seen using cUS in neonatal metabolic disorders. cUS is a reliable bedside tool for early detection of cysts, calcium, structural brain abnormalities, and white matter echogenicity, all suggestive of metabolic disorders.


Asunto(s)
Encefalopatías/diagnóstico , Encéfalo/patología , Ecoencefalografía/métodos , Imagen por Resonancia Magnética/métodos , Enfermedades Metabólicas/diagnóstico , Encefalopatías/complicaciones , Femenino , Humanos , Recién Nacido , Masculino , Enfermedades Metabólicas/complicaciones , Tamizaje Neonatal/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
10.
Eur J Paediatr Neurol ; 11(3): 160-6, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17257866

RESUMEN

OBJECTIVE: Scoliosis is a frequent complication (68-90%) of Duchenne muscular dystrophy (DMD). Prevention of limb deformities, rehabilitation in knee-ankle-foot-orthoses (KAFOs) and glucocorticoids prolong walking and standing, and might reduce scoliosis. We evaluated possible predictive factors for scoliosis development in a large DMD population. METHODS: Case notes of 123 DMD boys, > or = 17 years, followed at our centre between 1992 and 2002 were reviewed. Univariate analysis was used to relate two outcome measures (age at onset of scoliosis and severity at 17 years) with (i) glucocorticoids treatment; (ii) ages at (a) loss of independent ambulation, (b) rehabilitation into KAFOs, (c) loss of standing, (iii) forced vital capacity (FVC) (%) between 11 and 12 years and (iv) lower limb contractures. RESULTS: In total, 37/123 boys (30%) received intermittent prednisolone (0.75 mg/kg/day, 10 day/month) for a median 1-year (2 months-9 years), starting between 7.7 and 12.4 years (mean 9.5). About 96/123 (78%) were rehabilitated into KAFOs at 10.2+/-1.6 years. Age at loss of ambulation in KAFOs was 12.3+/-1.9 years and at loss of standing 12.8+/-2.1 years. About 95/123 (77%) boys developed scoliosis (Cobb angle >30 degrees ). Mean age+/-S.D. at scoliosis onset was 12.7+/-1.6 years. Forty-three boys (35%) had scoliosis surgery by 15+/-1.2 years. Later age at loss of ambulation (p<0.0001) and longer duration of prednisolone treatment (p=0.01) related to later scoliosis onset. Ages at loss of ambulation and standing were inversely related to scoliosis severity at 17 years (p<0.005). Hip asymmetry and %FVC at 11-12 years were directly related to scoliosis severity (p=0.02). CONCLUSIONS: Our data indicate a significant association between prolonged ambulation and a reduced risk of scoliosis development. Glucocorticoid administration, in our series, appear to be associated with a later onset of scoliosis, but did not alter the severity at 17 years, probably reflecting the shorter overall glucocorticoid exposure in this population.


Asunto(s)
Distrofia Muscular de Duchenne/complicaciones , Escoliosis/etiología , Adolescente , Factores de Edad , Edad de Inicio , Progresión de la Enfermedad , Humanos , Modelos Logísticos , Masculino , Distrofia Muscular de Duchenne/terapia , Valor Predictivo de las Pruebas , Prednisolona/uso terapéutico , Estudios Retrospectivos , Factores de Riesgo , Escoliosis/prevención & control
11.
J Neuromuscul Dis ; 2(4): 433-438, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-27858751

RESUMEN

BACKGROUND AND OBJECTIVES: Nemaline myopathy may be caused by pathogenic variants in the TPM3 gene and is then called NEM1. All previously identified disease-causing variants are point mutations including missense, nonsense and splice-site variants. The aim of the study was to identify the disease-causing gene in this patient and verify the NM diagnosis. METHODS: Mutation analysis methods include our self-designed nemaline myopathy array, The Nemaline Myopathy Comparative Genomic Hybridisation Array (NM-CGH array), whole-genome array-CGH, dHPLC, Sanger sequencing and whole-exome sequencing. The diagnostic muscle biopsy was investigated further by routine histopathological methods. RESULTS: We present here the first large (17-21 kb) aberration in the α-tropomyosinslow gene (TPM3), identified using the NM-CGH array. This homozygous deletion removes the exons 1a and 2b as well as the promoter of the TPM3 isoform encoding Tpm3.12st. The severe phenotype included paucity of movement, proximal and axial weakness and feeding difficulties requiring nasogastric tube feeding. The infant died at the age of 17.5 months. Muscle biopsy showed variation in fibre size and rods in a population of hypotrophic muscle fibres expressing slow myosin, often with internal nuclei, and abnormal immunolabelling revealing many hybrid fibres. CONCLUSIONS: This is the only copy number variation we have identified in any NM gene other than nebulin (NEB), suggesting that large deletions or duplications in these genes are very rare, yet possible, causes of NM.

12.
Eur J Paediatr Neurol ; 19(4): 395-401, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25843299

RESUMEN

BACKGROUND/PURPOSE: Duchenne Mmuscular Ddystrophy (DMD) related cardiomyopathy is associated with significant perioperative mortality. Cardiac MRI (CMR) has not previously been systematically evaluated as pre-operative assessment tool for heart function in DMD. Our aim was to establish whether CMR versus echocardiography contributes to pre-operative DMD assessment. METHODS: Case records were retrospectively reviewed of 35 consecutive DMD boys who underwent evaluation for surgical procedures between 2010 and 2013. RESULTS: Echocardiography revealed a median left ventricular (LV) shortening fraction (SF) of 29/% (range: 7-44). 37% of boys (13/35) had abnormal SF <25%, 66% (23/35) showed hypokinesia and 26% (9/35) had LV dilatation. CMR revealed a median left ventricular ejection fraction (LVEF) of 52% (range: 27-67%). 57% of boys (20/35) had abnormal LVEF <55%, 71% (25/35) had hypokinesia, and 82% late gadolinium enhancement. Extensive versus minimal late gadolinium enhancement was associated with reduced left ventricular ejection fraction (48% vs 58%; p = 0.003) suggesting more severe cardiomyopathy. Although echocardiography shortening fraction correlated with CMR ejection fraction (rs = 0.67; p < 0.001), three-quarter of echocardiography studies had suboptimal scanning windows and in 26% measurements significantly over- or underestimated left-ventricular function compared to CMR. CONCLUSION: Our findings clearly demonstrate the added value of CMR versus echocardiography in assessing DMD-cardiomyopathy. Particularly when echocardiographic scanning windows are suboptimal, CMR should be considered to allow accurate pre-operative cardiac assessment.


Asunto(s)
Cardiomiopatías/cirugía , Ecocardiografía/métodos , Imagen por Resonancia Cinemagnética/métodos , Distrofia Muscular de Duchenne/complicaciones , Cuidados Preoperatorios/métodos , Cardiomiopatías/etiología , Niño , Humanos , Masculino , Distrofia Muscular de Duchenne/fisiopatología , Estudios Retrospectivos
13.
Neurology ; 59(4): 609-10, 2002 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-12196659

RESUMEN

The aim of this pilot study was to evaluate the effect of albuterol in children with spinal muscular atrophy (SMA). Thirteen patients (five with SMA II and eight with SMA III) were given oral albuterol for 6 months. There was a significant increase in myometry, forced vital capacity, and lean body mass between the baseline and the 6-month assessments (p < 0.05). Albuterol may have a beneficial effect in patients with SMA without causing any significant adverse effects. Larger randomized, placebo-controlled trials are needed to confirm this observation.


Asunto(s)
Agonistas Adrenérgicos beta/uso terapéutico , Albuterol/uso terapéutico , Atrofias Musculares Espinales de la Infancia/tratamiento farmacológico , Absorciometría de Fotón , Agonistas Adrenérgicos beta/efectos adversos , Albuterol/efectos adversos , Presión Sanguínea/efectos de los fármacos , Composición Corporal/efectos de los fármacos , Niño , Preescolar , Estudios de Cohortes , Esquema de Medicación , Electrocardiografía/efectos de los fármacos , Femenino , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Contracción Isométrica/efectos de los fármacos , Masculino , Contracción Muscular/efectos de los fármacos , Proyectos Piloto , Resultado del Tratamiento , Capacidad Vital/efectos de los fármacos
14.
Neurology ; 59(2): 284-7, 2002 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-12136074

RESUMEN

Central core disease (CCD) is a congenital myopathy due to dominant mutations in the skeletal muscle ryanodine receptor gene (RYR1). The authors report three patients from two consanguineous families with symptoms of a congenital myopathy, cores on muscle biopsy, and confirmed linkage to the RYR1 locus. Molecular genetic studies in one family identified a V4849I homozygous missense mutation in the RYR1 gene. This report suggests a congenital myopathy associated with recessive RYR1 mutations.


Asunto(s)
Músculo Esquelético/patología , Enfermedades Musculares/congénito , Enfermedades Musculares/genética , Mutación Missense , Canal Liberador de Calcio Receptor de Rianodina/genética , Biopsia , Preescolar , Consanguinidad , Femenino , Homocigoto , Humanos , Enfermedades Musculares/patología
15.
Neuromuscul Disord ; 14(12): 818-21, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15564039

RESUMEN

Chronic inflammatory demyelinating polyneuropathy (CIPD) is characterised by progressive weakness, hyporeflexia and electrophysiological evidence of demyelination with maximal neurological deficit reached after at least 8 weeks progression. CIPD rarely affects children. We present a neonate with clinical features compatible with congenital CIPD. A term male infant of non-consanguineous parents was referred to us at birth with weakness and contractures affecting his legs, suggesting a prenatal onset of immobility. He also had evidence of bulbar dysfunction with poor suck, recurrent aspiration and requiring nasogastric feeding. He had no antigravity movements in the legs, bilateral wrist drop, distal joint contractures and absent deep tendon reflexes. Electromyography showed neurogenic changes, with nerve conduction velocities markedly reduced, increased distal motor latency and dispersed compound muscle action potentials. Cerebrospinal fluid protein was raised. Sural nerve biopsy demonstrated decreased numbers of myelinated fibres and inflammatory cell infiltrates. Muscle biopsy showed denervation. He only received supportive treatment and by 6 months he had fully recovered, and all electrophysiological parameters had normalised.


Asunto(s)
Sistema Nervioso Periférico/fisiopatología , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/congénito , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/fisiopatología , Progresión de la Enfermedad , Humanos , Recién Nacido , Masculino , Microscopía Electrónica de Transmisión , Debilidad Muscular/congénito , Debilidad Muscular/patología , Debilidad Muscular/fisiopatología , Músculo Esquelético/inervación , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Vaina de Mielina/patología , Vaina de Mielina/ultraestructura , Fibras Nerviosas Mielínicas/patología , Fibras Nerviosas Mielínicas/ultraestructura , Proteínas del Tejido Nervioso/líquido cefalorraquídeo , Conducción Nerviosa/genética , Paresia/congénito , Paresia/patología , Paresia/fisiopatología , Sistema Nervioso Periférico/patología , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/patología , Remisión Espontánea , Nervio Sural/patología , Nervio Sural/ultraestructura
16.
Neuromuscul Disord ; 2(5-6): 379-87, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1300186

RESUMEN

We performed a randomized controlled trial of early surgical treatment of contractures in 20 boys with Duchenne muscular dystrophy, age 4-6 yr. Surgery consisted of release of hip flexors, removal of iliotibial bands, and lengthening of tendo Achilles bilaterally. All patients were monitored for at least 12 months post-randomization, and assessed quantitatively for muscle strength and function. Surgery corrected the deformities, but had no beneficial effect on strength or function. Indeed, data in the second year showed more rapid deterioration of function in some of the operated boys. There appeared to be continued evolution of pathology following surgery, as assessed by sequential muscle ultrasound and muscle biopsy. We cannot recommend this type of surgery as a routine treatment.


Asunto(s)
Articulación de la Cadera/cirugía , Pierna/cirugía , Distrofias Musculares/cirugía , Biopsia , Niño , Preescolar , Estudios de Seguimiento , Marcha , Humanos , Masculino , Contracción Muscular/fisiología , Músculos/diagnóstico por imagen , Músculos/patología , Distrofias Musculares/diagnóstico por imagen , Distrofias Musculares/patología , Distrofias Musculares/fisiopatología , Análisis de Regresión , Ultrasonografía
17.
Neuromuscul Disord ; 8(7): 467-73, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9829276

RESUMEN

Central core disease (CCD) is a well recognized, relatively mild, non- or slowly progressive, dominantly inherited, congenital myopathy due, at least in some families, to mutations in the ryanodine receptor gene on chromosome 19q13.1. We report two unrelated cases with an unusual, early onset congenital myopathy with severe contractures, delayed motor milestones, proximal muscle weakness, normal serum creatine kinase (CK), a non-progressive course, with muscle biopsy findings of central cores and in addition, marked proliferation of connective and adipose tissue, and variation in fibre size. Muscle biopsies from the parents, who were non-consanguineous and healthy, showed minor myopathic changes and uneven staining with oxidative enzymes, but no central cores. The marked histological muscle changes, the distribution of weakness and the non-progressive course of the disease suggest that this is a severe variant of central core disease with secondary dystrophy-like change. The presence of mild changes in the histochemical reactions of biopsies of both parents of these two children supports the hypothesis that they are carriers of a recessive disease gene mutation responsible for this unusually severe form of central core disease.


Asunto(s)
Genes Recesivos/genética , Músculos/patología , Miopatías Nemalínicas/patología , Biopsia , Preescolar , Femenino , Humanos , Lactante , Masculino , Miopatías Nemalínicas/genética
18.
Neuromuscul Disord ; 14(10): 689-93, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15351427

RESUMEN

We report an unusual family in which the same point mutation in the voltage-gated potassium channel gene KCNA1 resulted in markedly different clinical phenotypes. The propositus presented in infancy with marked muscle stiffness, motor developmental delay, short stature, skeletal deformities, muscle hypertrophy and muscle rippling on percussion. He did not experience episodic ataxia. His mother presented some years later with typical features of Episodic Ataxia type 1 (EA1), with episodes of ataxia lasting a few minutes provoked by exercise. On examination she had myokymia, joint contractures and mild skeletal deformities. A heterozygous point mutation in the voltage-gated K(+) channel (KCNA1) gene (ACG-AGG, Thr226Arg) was found in both. We conclude that mutations in the potassium channel gene (KCNA1) can cause severe neuromyotonia resulting in marked skeletal deformities even if episodic ataxia is not prominent.


Asunto(s)
Síndrome de Isaacs/genética , Músculo Esquelético/patología , Fenotipo , Canales de Potasio con Entrada de Voltaje/genética , Adulto , Arginina/genética , Preescolar , Análisis Mutacional de ADN/métodos , Salud de la Familia , Femenino , Técnicas Histológicas/métodos , Humanos , Síndrome de Isaacs/patología , Síndrome de Isaacs/fisiopatología , Canal de Potasio Kv.1.1 , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Mutación , NAD/metabolismo , Treonina/genética
19.
Neuromuscul Disord ; 8(7): 495-501, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9829280

RESUMEN

We report two siblings, an 11-year-old boy and his 7-year-old sister, referred to us with a diagnosis of muscular dystrophy. The boy presented at 22 months with delay in walking. A very high serum creatine kinase (CK) level and a dystrophic muscle biopsy lead to a diagnosis of Duchenne muscular dystrophy prior to the identification of the dystrophin gene. Two years later his sister presented with similar problems. A diagnosis of limb-girdle muscular dystrophy was made when they were shown to have inherited different X-chromosomes and normal expression of dystrophin and all sarcoglycans. Their conditions remained static. Recently a slowing of the peripheral motor nerve conduction velocities and T2-weighted brain magnetic resonance imaging showed increased signal of the white matter, both of which are features of merosin-deficient congenital muscular dystrophy. Immunolabelling using a C-terminal laminin alpha 2 chain antibody showed a reduction in expression, while labelling with another antibody that recognises a 300-kDa fragment showed a very significant reduction. Mutational analysis of the LAMA2 gene showed two mutations: one was a G-->C point mutation at position -1 of intron 28 acceptor splicing site. This mutation induced activation of a cryptic splice at nucleotide 4429 of exon 29 and partial skipping of this exon, with conservation of the open reading frame. The other was a nonsense mutation due to a C_T transition at position 5525 of the cDNA sequence (exon 37), resulting in a stop codon. These data confirm that mutations of the LAMA2 gene that do not completely disrupt the production of the protein can give rise to phenotypes considerably milder than classical merosin-deficient congenital muscular dystrophy. Partial laminin alpha 2 deficiency should be considered in the differential diagnosis of limb-girdle muscular dystrophy.


Asunto(s)
Laminina/genética , Distrofias Musculares/genética , Secuencia de Bases , Biopsia , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Niño , Análisis Mutacional de ADN , Electrofisiología , Salud de la Familia , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Músculos/inervación , Músculos/metabolismo , Músculos/patología , Distrofias Musculares/diagnóstico , Distrofias Musculares/patología , Linaje , Mutación Puntual , Cintigrafía
20.
Neuromuscul Disord ; 13(1): 55-9, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12467733

RESUMEN

X-linked myotubular myopathy is a severe congenital myopathy in males, caused by mutations in the myotubularin (MTM1) gene on chromosome Xq28. In heterozygous carriers of MTM1 mutations, clinical symptoms are usually absent or only mild. We report a 6-year-old girl presenting at birth with marked hypotonia and associated feeding and respiratory difficulties. A muscle biopsy performed at 5 months suggested a diagnosis of myotubular myopathy. On examination at 6 years she had marked facial weakness with bilateral ptosis and external ophthalmoplegia, severe axial and proximal weakness and a mild scoliosis. Muscle magnetic resonance imaging showed a distinctive pattern of muscle involvement. Molecular genetic investigation of the MTM1 gene identified a heterozygous mutation in exon 12. X-inactivation studies in lymphocytes showed an extremely skewed pattern (97:3). This case emphasizes that investigation of the MTM1 gene and X-inactivation studies are indicated in isolated females with histopathological and clinical findings suggestive of myotubular myopathy.


Asunto(s)
Cromosomas Humanos X , Compensación de Dosificación (Genética) , Ligamiento Genético , Miopatías Estructurales Congénitas/genética , Proteínas Tirosina Fosfatasas/genética , Niño , Análisis Mutacional de ADN , Femenino , Mutación del Sistema de Lectura , Heterocigoto , Humanos , Imagen por Resonancia Magnética , Hipotonía Muscular , Miopatías Estructurales Congénitas/patología , Oftalmoplejía , Aberraciones Cromosómicas Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA