Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Brain ; 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38696726

RESUMEN

Congenital myasthenic syndromes (CMS) are clinically and genetically heterogeneous diseases caused by mutations affecting neuromuscular transmission. Even if the first symptoms mainly occur during childhood, adult neurologists must confront this challenging diagnosis and manage these patients throughout their adulthood. However, long-term follow-up data from large cohorts of CMS patients are lacking and the long-term prognosis of these patients is largely unknown. We report the clinical features, diagnostic difficulties, and long-term prognosis of a French nationwide cohort of 235 adult patients with genetically confirmed CMS followed in 23 specialized neuromuscular centres. Data were retrospectively analysed. Of the 235 patients, 123 were female (52.3%). The diagnosis was made in adulthood in 139 patients, 110 of whom presented their first symptoms before the age of 18. Mean follow-up time between first symptoms and last visit was 34 years (SD = 15.1). Pathogenic variants were found in 19 disease-related genes. CHRNE-low expressor variants were the most common (23.8%), followed by variants in DOK7 (18.7%) and RAPSN (14%). Genotypes were clustered into four groups according to the initial presentation: ocular group (CHRNE-LE, CHRND, FCCMS), distal group (SCCMS), limb-girdle group (RAPSN, COLQ, DOK7, GMPPB, GFPT1), and a variable-phenotype group (MUSK, AGRN). The phenotypical features of CMS did not change throughout life. Only four genotypes had a proportion of patients requiring intensive care unit (ICU) admission that exceeded 20%: RAPSN (54.8%), MUSK (50%), DOK7 (38.6%) and AGRN (25.0%). In RAPSN and MUSK patients most ICU admissions occurred before age 18 years and in DOK7 and AGRN patients at or after 18 years of age. Different patterns of disease course (stability, improvement and progressive worsening) may succeed one another in the same patient throughout life, particularly in AGRN, DOK7 and COLQ. At the last visit, 55% of SCCMS and 36.3% of DOK7 patients required ventilation; 36.3% of DOK7 patients, 25% of GMPPB patients and 20% of GFPT1 patients were wheelchair-bound; most of the patients who were both wheelchair-bound and ventilated were DOK7 patients. Six patients died in this cohort. The positive impact of therapy was striking, even in severely affected patients. In conclusion, even if motor and/or respiratory deterioration could occur in patients with initially moderate disease, particularly in DOK7, SCCMS and GFPT1 patients, the long-term prognosis for most CMS patients was favourable, with neither ventilation nor wheelchair needed at last visit. CHRNE patients did not worsen during adulthood and RAPSN patients, often severely affected in early childhood, subsequently improved.

2.
J Med Genet ; 56(9): 590-601, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31010831

RESUMEN

BACKGROUND: Subtelomeres are variable regions between telomeres and chromosomal-specific regions. One of the most studied pathologies linked to subtelomeric imbalance is facioscapulohumeral dystrophy (FSHD). In most cases, this disease involves shortening of an array of D4Z4 macrosatellite elements at the 4q35 locus. The disease also segregates with a specific A-type haplotype containing a degenerated polyadenylation signal distal to the last repeat followed by a repetitive array of ß-satellite elements. This classification applies to most patients with FSHD. A subset of patients called FSHD2 escapes this definition and carries a mutation in the SMCHD1 gene. We also recently described patients carrying a complex rearrangement consisting of a cis-duplication of the distal 4q35 locus identified by molecular combing. METHODS: Using this high-resolution technology, we further investigated the organisation of the 4q35 region linked to the disease and the 10q26 locus presenting with 98% of homology in controls and patients. RESULTS: Our analyses reveal a broad variability in size of the different elements composing these loci highlighting the complexity of these subtelomeres and the difficulty for genomic assembly. Out of the 1029 DNA samples analysed in our centre in the last 7 years, we also identified 54 cases clinically diagnosed with FSHD carrying complex genotypes. This includes mosaic patients, patients with deletions of the proximal 4q region and 23 cases with an atypical chromosome 10 pattern, infrequently found in the control population and never reported before. CONCLUSION: Overall, this work underlines the complexity of these loci challenging the diagnosis and genetic counselling for this disease.


Asunto(s)
Cromosomas Humanos Par 10 , Cromosomas Humanos Par 4 , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Distrofia Muscular Facioescapulohumeral/diagnóstico , Distrofia Muscular Facioescapulohumeral/genética , Telómero/genética , Alelos , Deleción Cromosómica , Estudios de Asociación Genética/métodos , Sitios Genéticos , Genotipo , Humanos , Linaje
3.
J Med Genet ; 55(7): 469-478, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29563141

RESUMEN

BACKGROUND: 18p deletion syndrome is a rare disorder caused by partial or full monosomy of the short arm of chromosome 18. Clinical symptoms caused by 18p hemizygosity include cognitive impairment, mild facial dysmorphism, strabismus and ptosis. Among other genes, structural maintenance of chromosomes flexible hinge domain containing 1 (SMCHD1) is hemizygous in most patients with 18p deletions. Digenic inheritance of a SMCHD1 mutation and a moderately sized D4Z4 repeat on a facioscapulohumeral muscular dystrophy (FSHD) permissive genetic background of chromosome 4 can cause FSHD type 2 (FSHD2). OBJECTIVES: Since 12% of Caucasian individuals harbour moderately sized D4Z4 repeats on an FSHD permissive background, we tested if people with 18p deletions are at risk of developing FSHD. METHODS: To test our hypothesis we studied different cellular systems originating from individuals with 18p deletions not presenting FSHD2 phenotype for transcriptional and epigenetic characteristics of FSHD at D4Z4. Furthermore, individuals with an idiopathic muscle phenotype and an 18p deletion were subjected to neurological examination. RESULTS: Primary fibroblasts hemizygous for SMCHD1 have a D4Z4 chromatin structure comparable with FSHD2 concomitant with DUX4 expression after transdifferentiation into myocytes. Neurological examination of 18p deletion individuals from two independent families with a moderately sized D4Z4 repeat identified muscle features compatible with FSHD. CONCLUSIONS: 18p deletions leading to haploinsufficiency of SMCHD1, together with a moderately sized FSHD permissive D4Z4 allele, can associate with symptoms and molecular features of FSHD. We propose that patients with 18p deletion should be characterised for their D4Z4 repeat size and haplotype and monitored for clinical features of FSHD.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Trastornos de los Cromosomas/genética , Epigénesis Genética , Distrofia Muscular Facioescapulohumeral/genética , Adolescente , Adulto , Cromatina/genética , Deleción Cromosómica , Trastornos de los Cromosomas/diagnóstico , Trastornos de los Cromosomas/fisiopatología , Cromosomas Humanos Par 18/genética , Metilación de ADN/genética , Femenino , Haploinsuficiencia/genética , Humanos , Masculino , Persona de Mediana Edad , Monosomía/genética , Monosomía/patología , Distrofia Muscular Facioescapulohumeral/epidemiología , Distrofia Muscular Facioescapulohumeral/fisiopatología , Mutación , Factores de Riesgo , Adulto Joven
4.
Muscle Nerve ; 57(2): 217-221, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28407266

RESUMEN

INTRODUCTION: In young patients with mononeuropathy who lack family history and precipitating factors, hereditary neuropathy with liability to pressure palsy (HNPP) may be a possibility. Our objective is to propose neurophysiological criteria for HNPP in patients <30 years of age. METHODS: We conducted a national multicenter retrospective clinical and neurophysiological study in patients under 30 with genetically confirmed HNPP. RESULTS: All of the 51 patients included in the study had at least 1 demyelinating pattern in 2 asymptomatic nerves, and 3 abnormalities were found in almost 90%, including slowed motor nerve conduction velocity across the elbow in at least 1 ulnar nerve (97.5%), increased distal motor latency (DML) in at least 1 fibular nerve (95.8%), and increased DML in both median nerves (89%). Age influenced DML slightly only in the fibular nerve. DISCUSSION: Dissemination of nerve involvement in HNPP incites to perform a complete nerve conduction study. including bilateral ulnar, fibular, and median nerves. Muscle Nerve 57: 217-221, 2018.


Asunto(s)
Electrodiagnóstico/normas , Neuropatía Hereditaria Motora y Sensorial/fisiopatología , Nervios Periféricos/fisiopatología , Adolescente , Adulto , Edad de Inicio , Envejecimiento , Niño , Enfermedades Desmielinizantes/complicaciones , Enfermedades Desmielinizantes/patología , Femenino , Neuropatía Hereditaria Motora y Sensorial/complicaciones , Neuropatía Hereditaria Motora y Sensorial/diagnóstico , Humanos , Masculino , Nervio Mediano/fisiopatología , Neuronas Motoras , Conducción Nerviosa , Parálisis , Nervio Peroneo/fisiopatología , Presión , Estudios Retrospectivos , Nervio Cubital/fisiopatología , Adulto Joven
5.
Hum Mutat ; 35(7): 779-90, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24692096

RESUMEN

Mutations affecting skeletal muscle isoforms of the tropomyosin genes may cause nemaline myopathy, cap myopathy, core-rod myopathy, congenital fiber-type disproportion, distal arthrogryposes, and Escobar syndrome. We correlate the clinical picture of these diseases with novel (19) and previously reported (31) mutations of the TPM2 and TPM3 genes. Included are altogether 93 families: 53 with TPM2 mutations and 40 with TPM3 mutations. Thirty distinct pathogenic variants of TPM2 and 20 of TPM3 have been published or listed in the Leiden Open Variant Database (http://www.dmd.nl/). Most are heterozygous changes associated with autosomal-dominant disease. Patients with TPM2 mutations tended to present with milder symptoms than those with TPM3 mutations, DA being present only in the TPM2 group. Previous studies have shown that five of the mutations in TPM2 and one in TPM3 cause increased Ca(2+) sensitivity resulting in a hypercontractile molecular phenotype. Patients with hypercontractile phenotype more often had contractures of the limb joints (18/19) and jaw (6/19) than those with nonhypercontractile ones (2/22 and 1/22), whereas patients with the non-hypercontractile molecular phenotype more often (19/22) had axial contractures than the hypercontractile group (7/19). Our in silico predictions show that most mutations affect tropomyosin-actin association or tropomyosin head-to-tail binding.


Asunto(s)
Estudios de Asociación Genética , Enfermedades Musculares/congénito , Enfermedades Musculares/genética , Mutación , Tropomiosina/genética , Actinas/metabolismo , Adolescente , Adulto , Secuencia de Aminoácidos , Niño , Preescolar , Bases de Datos Genéticas , Femenino , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/diagnóstico , Fenotipo , Fosforilación , Unión Proteica , Alineación de Secuencia , Tropomiosina/química , Tropomiosina/metabolismo , Adulto Joven
6.
Eur J Hum Genet ; 32(1): 37-43, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37337091

RESUMEN

Proximal spinal muscular atrophy (SMA) is defined by a degeneration of the anterior horn cells resulting in muscle weakness predominantly in the proximal lower limbs. While most patients carry a biallelic deletion in the SMN1 gene (localized in chromosome 5q), little is known regarding patients without SMN1-mutation, and a genetic diagnosis is not always possible. Here, we report a cohort of 24 French patients with non-5q proximal SMA from five neuromuscular centers who all, except two, had next-generation sequencing (NGS) gene panel, followed by whole exome sequencing (WES) if gene panel showed a negative result. The two remaining patients benefited directly from WES or whole genome sequencing (WGS). A total of ten patients with causative variants were identified, nine of whom were index cases (9/23 families = 39%). Eight variants were identified by gene panel: five variants in DYNC1H1, and three in BICD2. Compound heterozygous causative variants in ASAH1 were identified directly by WES, and one variant in DYNC1H1 was identified directly by WGS. No causative variant was found using WES in patients with a previous panel with negative results (14 cases). We thus recommend using primarily NGS panels in patients with non-5q-SMA and using WES, especially when several members of the same family are affected and/or when trio analyses are possible, or WGS as second-line testing if available.


Asunto(s)
Atrofia Muscular Espinal , Humanos , Secuenciación del Exoma , Mutación , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/diagnóstico , Secuenciación Completa del Genoma
7.
J Cachexia Sarcopenia Muscle ; 13(1): 621-635, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34859613

RESUMEN

BACKGROUND: Facioscapulohumeral dystrophy (FSHD) is a late-onset autosomal dominant form of muscular dystrophy involving specific groups of muscles with variable weakness that precedes inflammatory response, fat infiltration, and muscle atrophy. As there is currently no cure for this disease, understanding and modelling the typical muscle weakness in FSHD remains a major milestone towards deciphering the disease pathogenesis as it will pave the way to therapeutic strategies aimed at correcting the functional muscular defect in patients. METHODS: To gain further insights into the specificity of the muscle alteration in this disease, we derived induced pluripotent stem cells from patients affected with Types 1 and 2 FSHD but also from patients affected with Bosma arhinia and microphthalmia. We differentiated these cells into contractile innervated muscle fibres and analysed their transcriptome by RNA Seq in comparison with cells derived from healthy donors. To uncover biological pathways altered in the disease, we applied MOGAMUN, a multi-objective genetic algorithm that integrates multiplex complex networks of biological interactions (protein-protein interactions, co-expression, and biological pathways) and RNA Seq expression data to identify active modules. RESULTS: We identified 132 differentially expressed genes that are specific to FSHD cells (false discovery rate < 0.05). In FSHD, the vast majority of active modules retrieved with MOGAMUN converges towards a decreased expression of genes encoding proteins involved in sarcomere organization (P value 2.63e-12 ), actin cytoskeleton (P value 9.4e-5 ), myofibril (P value 2.19e-12 ), actin-myosin sliding, and calcium handling (with P values ranging from 7.9e-35 to 7.9e-21 ). Combined with in vivo validations and functional investigations, our data emphasize a reduction in fibre contraction (P value < 0.0001) indicating that the muscle weakness that is typical of FSHD clinical spectrum might be associated with dysfunction of calcium release (P value < 0.0001), actin-myosin interactions, motor activity, mechano-transduction, and dysfunctional sarcomere contractility. CONCLUSIONS: Identification of biomarkers of FSHD muscle remain critical for understanding the process leading to the pathology but also for the definition of readouts to be used for drug design, outcome measures, and monitoring of therapies. The different pathways identified through a system biology approach have been largely overlooked in the disease. Overall, our work opens new perspectives in the definition of biomarkers able to define the muscle alteration but also in the development of novel strategies to improve muscle function as it provides functional parameters for active molecule screening.


Asunto(s)
Células Madre Pluripotentes Inducidas , Distrofia Muscular Facioescapulohumeral , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Contracción Muscular , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Facioescapulohumeral/genética , Sarcómeros/metabolismo
8.
Hum Mutat ; 31(1): E1110-25, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19953646

RESUMEN

Spinal muscular atrophy (SMA) is a common neuromuscular disorder caused by homozygous inactivation of the SMN1 (Survival Motor Neuron 1) gene. The disease severity is mainly influenced by the copy number of SMN2, a nearly identical gene from which only low amounts of full-length mRNA are produced. This correlation is not absolute, suggesting the existence of yet unknown factors modulating disease progression. We identified and characterized the rare variant c.859G>C (p.Gly287Arg) in exon 7 in both SMN2 copies of a male patient affected with type III SMA, a milder form of the disease rarely associated with only two SMN2 copies. We demonstrated in vivo, in this patient and in a second unrelated patient, and ex vivo, using SMN splicing assays, that the variant induces inclusion of exon 7 into SMN2 mRNA. Moreover, we show that the c.859G>C variation is located in a composite splicing regulatory element in the centre of exon 7. The variation does not affect binding of HTra2â nor creates a novel SF2/ASF enhancer, but disrupts an hnRNP A1 binding site. The natural occurrence of enhanced inclusion of SMN2 exon 7 in milder SMA cases supports the current therapeutic strategies based on splicing modulation in SMA patients.


Asunto(s)
Elementos de Facilitación Genéticos , Exones , Variación Genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/fisiopatología , Empalme del ARN , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Adolescente , Adulto , Sitios de Unión , Preescolar , Femenino , Dosificación de Gen , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Humanos , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Índice de Severidad de la Enfermedad
9.
Neuromuscul Disord ; 29(2): 114-126, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30598237

RESUMEN

Spinal muscular atrophy with respiratory distress type 1 (SMARD1) is a rare autosomal recessive neuromuscular disorder characterized by progressive motor and respiratory decline during the first year of life. Early and late-onset cases have recently been reported, although not meeting the established diagnostic criteria, these cases have been genotyped. We thus conducted a national multicenter observational retrospective study to determine the prognosis of children with SMARD1 according to their phenotype. We recorded all known French pediatric cases with mutations identified on the immunoglobulin µ-binding protein 2 gene and the presence of respiratory symptoms. Thirty centers provided 22 observations. A diaphragmatic palsy was diagnosed 1.5 months (p = 0.02) after first respiratory symptoms, and hypotonia preceded areflexia by 4 months (p = 0.02). Early onset of symptoms leading to specialist consultation before the age of 3 months was associated with a significantly worse prognosis (p < 0.01). Among the 6 patients who were still alive, all were tracheostomized. Only one case survived beyond 2 years without artificial ventilation. The remaining patients died at a median age of 7 months. Our results may help pediatricians to provide medical information to parents and improve the decision-making process of setting up life support.


Asunto(s)
Proteínas de Unión al ADN/genética , Atrofia Muscular Espinal/diagnóstico , Síndrome de Dificultad Respiratoria del Recién Nacido/diagnóstico , Factores de Transcripción/genética , Preescolar , Progresión de la Enfermedad , Femenino , Francia , Humanos , Lactante , Recién Nacido , Masculino , Atrofia Muscular Espinal/genética , Mutación , Fenotipo , Pronóstico , Respiración Artificial , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Estudios Retrospectivos
10.
Neurology ; 92(8): e852-e865, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30659139

RESUMEN

OBJECTIVE: To genotypically and phenotypically characterize a large pediatric myotonic dystrophy type 1 (DM1) cohort to provide a solid frame of data for future evidence-based health management. METHODS: Among the 2,697 patients with genetically confirmed DM1 included in the French DM-Scope registry, children were enrolled between January 2010 and February 2016 from 24 centers. Comprehensive cross-sectional analysis of most relevant qualitative and quantitative variables was performed. RESULTS: We studied 314 children (52% females, with 55% congenital, 31% infantile, 14% juvenile form). The age at inclusion was inversely correlated with the CTG repeat length. The paternal transmission rate was higher than expected, especially in the congenital form (13%). A continuum of highly prevalent neurodevelopmental alterations was observed, including cognitive slowing (83%), attention deficit (64%), written language (64%), and spoken language (63%) disorders. Five percent exhibited autism spectrum disorders. Overall, musculoskeletal impairment was mild. Despite low prevalence, cardiorespiratory impairment could be life-threatening, and frequently occurred early in the first decade (25.9%). Gastrointestinal symptoms (27%) and cataracts (7%) were more frequent than expected, while endocrine or metabolic disorders were scarce. CONCLUSIONS: The pedDM-Scope study details the main genotype and phenotype characteristics of the 3 DM1 pediatric subgroups. It highlights striking profiles that could be useful in health care management (including transition into adulthood) and health policy planning.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Debilidad Muscular/fisiopatología , Distrofia Miotónica/fisiopatología , Insuficiencia Respiratoria/fisiopatología , Adolescente , Arritmias Cardíacas/epidemiología , Arritmias Cardíacas/etiología , Niño , Preescolar , Medicina Basada en la Evidencia , Femenino , Deformidades del Pie/epidemiología , Deformidades del Pie/etiología , Francia/epidemiología , Humanos , Lactante , Recién Nacido , Masculino , Debilidad Muscular/epidemiología , Debilidad Muscular/etiología , Distrofia Miotónica/complicaciones , Distrofia Miotónica/epidemiología , Distrofia Miotónica/genética , Sistema de Registros , Insuficiencia Respiratoria/epidemiología , Insuficiencia Respiratoria/etiología , Índice de Severidad de la Enfermedad , Expansión de Repetición de Trinucleótido
11.
Epileptic Disord ; 20(4): 289-294, 2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-30078772

RESUMEN

SCN8A encephalopathy is a newly defined epileptic encephalopathy caused by de novo mutations of the SCN8A gene. We report herein a four-year-old boy presenting with severe non-epileptic abnormal movements, of possibly antenatal onset, progressively associated with pharmacoresistant epilepsy and regression, associated with a de novo heterozygous missense mutation of SCN8A. This case shows that paroxysmal non-epileptic episodes of severe tremor and hyperekplexia-like startles and a striking vegetative component can be the first early symptoms of severe SCN8A developmental and epileptic encephalopathy. Clinicians should be aware of these symptoms in order to avoid misdiagnosis and ensure early appropriate therapeutic management. [Published with video sequences on www.epilepticdisorders.com].


Asunto(s)
Encefalopatías , Epilepsia , Hiperekplexia , Enfermedades del Recién Nacido , Canal de Sodio Activado por Voltaje NAV1.6/genética , Temblor , Encefalopatías/diagnóstico , Encefalopatías/genética , Encefalopatías/fisiopatología , Preescolar , Epilepsia/diagnóstico , Epilepsia/genética , Epilepsia/fisiopatología , Humanos , Hiperekplexia/diagnóstico , Hiperekplexia/genética , Hiperekplexia/fisiopatología , Recién Nacido , Enfermedades del Recién Nacido/diagnóstico , Enfermedades del Recién Nacido/genética , Enfermedades del Recién Nacido/fisiopatología , Masculino , Temblor/diagnóstico , Temblor/genética , Temblor/fisiopatología
12.
J Neurol ; 264(8): 1791-1803, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28712002

RESUMEN

Mutations in GFPT1 (glutamine-fructose-6-phosphate transaminase 1), a gene encoding an enzyme involved in glycosylation of ubiquitous proteins, cause a limb-girdle congenital myasthenic syndrome (LG-CMS) with tubular aggregates (TAs) characterized predominantly by affection of the proximal skeletal muscles and presence of highly organized and remodeled sarcoplasmic tubules in patients' muscle biopsies. We report here the first long-term clinical follow-up of 11 French individuals suffering from LG-CMS with TAs due to GFPT1 mutations, of which nine are new. Our retrospective clinical evaluation stresses an evolution toward a myopathic weakness that occurs concomitantly to ineffectiveness of usual CMS treatments. Analysis of neuromuscular biopsies from three unrelated individuals demonstrates that the maintenance of neuromuscular junctions (NMJs) is dramatically impaired with loss of post-synaptic junctional folds and evidence of denervation-reinnervation processes affecting the three main NMJ components. Moreover, molecular analyses of the human muscle biopsies confirm glycosylation defects of proteins with reduced O-glycosylation and show reduced sialylation of transmembrane proteins in extra-junctional area. Altogether, these results pave the way for understanding the etiology of this rare neuromuscular disorder that may be considered as a "tubular aggregates myopathy with synaptopathy".


Asunto(s)
Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/genética , Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/patología , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología , Unión Neuromuscular/patología , Adolescente , Adulto , Anciano , Femenino , Estudios de Seguimiento , Glicosilación , Humanos , Persona de Mediana Edad , Músculo Esquelético/enzimología , Músculo Esquelético/inervación , Músculo Esquelético/patología , Síndromes Miasténicos Congénitos/tratamiento farmacológico , Síndromes Miasténicos Congénitos/enzimología , Miopatías Estructurales Congénitas/tratamiento farmacológico , Miopatías Estructurales Congénitas/enzimología , Unión Neuromuscular/enzimología , Estudios Prospectivos , Estudios Retrospectivos , Adulto Joven
13.
Child Neurol Open ; 2(4): 2329048X15609053, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-28503595

RESUMEN

Guillain-Barré syndrome is a rare acute polyradiculoneuropathy. Several variants and unusual presentations have been described, particularly in pediatrics. In most cases, making an early diagnosis is challenging due to the treatments that consist in the rapid administration of intravenous immunoglobulin or plasma exchange. The authors present the case of a 7-year-old boy with an atypical and severe axonal Guillain-Barré syndrome, associated with Mycoplasma pneumonia. When he was admitted, febrile respiratory failure was the main focus, and then he presented signs of acute polyneuropathy with cranial nerve palsy and brief hyperreflexia. Mechanical ventilation was required for 48 days as well as 2 cycles of intravenous immunoglobulin. The authors describe all the medical challenges that the authors encountered. This case highlights the fact that respiratory distress can be the main clinical symptom in children. This delays the establishment of a correct diagnosis, even more so when neurological manifestations are abundant and unusual.

14.
Clin Chim Acta ; 448: 146-9, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26148721

RESUMEN

BACKGROUND: Dystrophinopathies, either the severe Duchenne Muscular Dystrophy (DMD) or the milder Becker Muscular Dystrophy (BMD), are X-linked recessive disorders caused by mutations in the DMD gene. DMD is one of the longest human genes. Large deletions or duplications account for 60-80% of the mutations. Remaining anomalies consist in point mutations or small rearrangements. Routinely, the molecular diagnosis is done by a Multiplex Ligation-dependent Probe Amplification (MLPA) or array Comparative Genome Hybridization (aCGH), followed, if negative, by Sanger sequencing of all exons. METHODS: In this study, massive parallel sequencing (MPS) or next generation sequencing (NGS) was used to make a rapid and costless molecular diagnosis in a young boy suspected of DMD. RESULTS: A small deletion: NM_004006.2:c.2803+5_2803+8del was identified. The diagnosis was performed in one single manipulation and within a week. The consequence of this intronic mutation is a skipping of exon 21 confirmed by mRNA and protein analysis. CONCLUSIONS: NGS appears to be an efficient new strategy in DMD molecular diagnosis. It highlights the major evolution of the diagnostic strategy towards high throughput technologies, where bioinformatics analysis becomes the real challenge for variations detection. This is the first study reporting in vivo impact of this intronic mutation.


Asunto(s)
Distrofina/química , Distrofina/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Distrofia Muscular de Duchenne/genética , Mutación/genética , Empalme del ARN/genética , ARN Mensajero/genética , Niño , Humanos , Masculino , Distrofia Muscular de Duchenne/diagnóstico
15.
Spine J ; 14(7): 1214-20, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24200409

RESUMEN

BACKGROUND CONTEXT: Combined monitoring of muscle motor evoked potentials elicited by transcranial electric stimulation (TES-mMEP) and cortical somatosensory evoked potentials (cSSEPs) is safe and effective for spinal cord monitoring during scoliosis surgery. However, TES-mMEP/cSSEP is not always feasible. Predictors of feasibility would help to plan the monitoring strategy. PURPOSE: To identify predictors of the feasibility of TES-mMEP/cSSEP during scoliosis surgery. STUDY DESIGN/SETTING: Prospective cohort study in a clinical neurophysiology unit and pediatric orthopedic department of a French university hospital. PATIENT SAMPLE: A total of 103 children aged 2 to 19 years scheduled for scoliosis surgery. OUTCOME MEASURES: Feasibility rate of intraoperative TES-mMEP/cSSEP monitoring. METHODS: All patients underwent a preoperative neurological evaluation and preoperative mMEP and cSSEP recordings at both legs. For each factor associated with feasibility, we computed sensitivity, specificity, positive predictive value (PPV), and negative predictive value. A decision tree was designed. RESULTS: Presence of any of the following factors was associated with 100% feasibility, 100% specificity, and 100% PPV: idiopathic scoliosis, normal preoperative neurological findings, and normal preoperative mMEP and cSSEP recordings. Feasibility was 0% in the eight patients with no recordable mMEPs or cSSEPs during preoperative testing. A decision tree involving three screening steps can be used to identify patients in whom intraoperative TES-mMEP/cSSEP is feasible. CONCLUSIONS: Preoperative neurological and neurophysiological assessments are helpful for identifying patients who can be successfully monitored by TES-mMEP/cSSEP during scoliosis surgery.


Asunto(s)
Potenciales Evocados Motores/fisiología , Potenciales Evocados Somatosensoriales/fisiología , Monitoreo Intraoperatorio/métodos , Escoliosis/cirugía , Médula Espinal/fisiopatología , Estimulación Transcraneal de Corriente Directa , Adolescente , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Masculino , Escoliosis/fisiopatología , Sensibilidad y Especificidad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA