Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 92(2): 271-8, 2013 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-23332920

RESUMEN

Tubular aggregates are regular arrays of membrane tubules accumulating in muscle with age. They are found as secondary features in several muscle disorders, including alcohol- and drug-induced myopathies, exercise-induced cramps, and inherited myasthenia, but also exist as a pure genetic form characterized by slowly progressive muscle weakness. We identified dominant STIM1 mutations as a genetic cause of tubular-aggregate myopathy (TAM). Stromal interaction molecule 1 (STIM1) is the main Ca(2+) sensor in the endoplasmic reticulum, and all mutations were found in the highly conserved intraluminal Ca(2+)-binding EF hands. Ca(2+) stores are refilled through a process called store-operated Ca(2+) entry (SOCE). Upon Ca(2+)-store depletion, wild-type STIM1 oligomerizes and thereby triggers extracellular Ca(2+) entry. In contrast, the missense mutations found in our four TAM-affected families induced constitutive STIM1 clustering, indicating that Ca(2+) sensing was impaired. By monitoring the calcium response of TAM myoblasts to SOCE, we found a significantly higher basal Ca(2+) level in TAM cells and a dysregulation of intracellular Ca(2+) homeostasis. Because recessive STIM1 loss-of-function mutations were associated with immunodeficiency, we conclude that the tissue-specific impact of STIM1 loss or constitutive activation is different and that a tight regulation of STIM1-dependent SOCE is fundamental for normal skeletal-muscle structure and function.


Asunto(s)
Calcio/metabolismo , Proteínas de la Membrana/metabolismo , Miopatías Estructurales Congénitas/patología , Proteínas de Neoplasias/metabolismo , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Niño , Femenino , Homeostasis , Humanos , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Músculos/patología , Músculos/ultraestructura , Mutación/genética , Mioblastos/metabolismo , Mioblastos/patología , Miopatías Estructurales Congénitas/genética , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Linaje , Fenotipo , Molécula de Interacción Estromal 1 , Adulto Joven
2.
Brain ; 137(Pt 9): 2429-43, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24951643

RESUMEN

Congenital myasthenic syndromes are a clinically and genetically heterogeneous group of rare diseases resulting from impaired neuromuscular transmission. Their clinical hallmark is fatigable muscle weakness associated with a decremental muscle response to repetitive nerve stimulation and frequently related to postsynaptic defects. Distal myopathies form another clinically and genetically heterogeneous group of primary muscle disorders where weakness and atrophy are restricted to distal muscles, at least initially. In both congenital myasthenic syndromes and distal myopathies, a significant number of patients remain genetically undiagnosed. Here, we report five patients from three unrelated families with a strikingly homogenous clinical entity combining congenital myasthenia with distal muscle weakness and atrophy reminiscent of a distal myopathy. MRI and neurophysiological studies were compatible with mild myopathy restricted to distal limb muscles, but decrement (up to 72%) in response to 3 Hz repetitive nerve stimulation pointed towards a neuromuscular transmission defect. Post-exercise increment (up to 285%) was observed in the distal limb muscles in all cases suggesting presynaptic congenital myasthenic syndrome. Immunofluorescence and ultrastructural analyses of muscle end-plate regions showed synaptic remodelling with denervation-reinnervation events. We performed whole-exome sequencing in two kinships and Sanger sequencing in one isolated case and identified five new recessive mutations in the gene encoding agrin. This synaptic proteoglycan with critical function at the neuromuscular junction was previously found mutated in more typical forms of congenital myasthenic syndrome. In our patients, we found two missense mutations residing in the N-terminal agrin domain, which reduced acetylcholine receptors clustering activity of agrin in vitro. Our findings expand the spectrum of congenital myasthenic syndromes due to agrin mutations and show an unexpected correlation between the mutated gene and the associated phenotype. This provides a good rationale for examining patients with apparent distal myopathy for a neuromuscular transmission disorder and agrin mutations.


Asunto(s)
Agrina/genética , Debilidad Muscular/genética , Atrofia Muscular/genética , Síndromes Miasténicos Congénitos/genética , Adulto , Secuencia de Aminoácidos , Atrofia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Debilidad Muscular/complicaciones , Debilidad Muscular/patología , Atrofia Muscular/complicaciones , Atrofia Muscular/patología , Síndromes Miasténicos Congénitos/complicaciones , Síndromes Miasténicos Congénitos/patología , Linaje
3.
Brain ; 136(Pt 8): 2359-68, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23824486

RESUMEN

Amyotrophic lateral sclerosis is a typically rapidly progressive neurodegenerative disorder affecting motor neurons leading to progressive muscle paralysis and death, usually from respiratory failure, in 3-5 years. Some patients have slow disease progression and prolonged survival, but the underlying mechanisms remain poorly understood. Riluzole, the only approved treatment, only modestly prolongs survival and has no effect on muscle function. In the early phase of the disease, motor neuron loss is initially compensated for by collateral reinnervation, but over time this compensation fails, leading to progressive muscle wasting. The crucial role of muscle histone deacetylase 4 and its regulator microRNA-206 in compensatory reinnervation and disease progression was recently suggested in a mouse model of amyotrophic lateral sclerosis (transgenic mice carrying human mutations in the superoxide dismutase gene). Here, we sought to investigate whether the microRNA-206-histone deacetylase 4 pathway plays a role in muscle compensatory reinnervation in patients with amyotrophic lateral sclerosis and thus contributes to disease outcome differences. We studied muscle reinnervation using high-resolution confocal imaging of neuromuscular junctions in muscle samples obtained from 11 patients with amyotrophic lateral sclerosis, including five long-term survivors. We showed that the proportion of reinnervated neuromuscular junctions was significantly higher in long-term survivors than in patients with rapidly progressive disease. We analysed the expression of muscle candidate genes involved in the reinnervation process and showed that histone deacetylase 4 upregulation was significantly greater in patients with rapidly progressive disease and was negatively correlated with the extent of muscle reinnervation and functional outcome. Conversely, the proposed regulator of histone deacetylase 4, microRNA-206, was upregulated in both patient groups, but did not correlate with disease progression or reinnervation. We conclude that muscle expression of histone deacetylase 4 may be a key factor for muscle reinnervation and disease progression in patients with amyotrophic lateral sclerosis. Specific histone deacetylase 4 inhibitors may then constitute a therapeutic approach to enhancing motor performance and slowing disease progression in amyotrophic lateral sclerosis.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Histona Desacetilasas/genética , MicroARNs/genética , Neuronas Motoras/metabolismo , Músculo Esquelético/inervación , Proteínas Represoras/genética , Adulto , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Progresión de la Enfermedad , Femenino , Histona Desacetilasas/metabolismo , Humanos , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Neuronas Motoras/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Proteínas Represoras/metabolismo , Sobrevivientes , Regulación hacia Arriba
4.
Bull Acad Natl Med ; 198(2): 257-70; discussion 270-1, 2014 Feb.
Artículo en Francés | MEDLINE | ID: mdl-26263703

RESUMEN

Congenital myasthenic syndromes CMS) form a heterogeneous group of genetic diseases characterized by abnormal neuromuscular transmission. The associated muscular weakness is exacerbated by exertion and usually starts during infancy/childhood In 2002 a national Congenital Myasthenic Syndromes Network was created in France, composed of neurologists, neuropediatricians, pathologists, molecular geneticists and neurobiologists. The network has now identified nearly 300 cases of CMS, as well as three new culprit genes. Based on our personal experience and data from the most recent studies, we describe the 18 principal culprit genes so far identified, along with diagnostic pitfalls, the disease course, prognosis and treatment. The underlying genetic defect remains to be identified in nearly half of CMS patients.


Asunto(s)
Síndromes Miasténicos Congénitos , Diagnóstico Diferencial , Progresión de la Enfermedad , Francia/epidemiología , Humanos , Síndromes Miasténicos Congénitos/diagnóstico , Síndromes Miasténicos Congénitos/epidemiología , Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/terapia , Pronóstico , Receptores Nicotínicos/genética
5.
Curr Opin Neurol ; 26(5): 561-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23995276

RESUMEN

PURPOSE OF REVIEW: Congenital myasthenic syndromes (CMSs) form a heterogeneous group of genetic diseases characterized by a dysfunction of neuromuscular transmission because of mutations in numerous genes. This review will focus on the causative genes recently identified and on the therapy of CMSs. RECENT FINDINGS: Advances in exome sequencing allowed the discovery of a new group of genes that did not code for the known molecular components of the neuromuscular junction, and the definition of a new group of glycosylation-defective CMS. Rather than the specific drugs used, some of them having been known for decades, it is the rigorous therapeutic strategy that is now offered to the patient in relation to the identified mutated gene that is novel and promising. SUMMARY: In addition to the above main points, we also present new data on the genes that were already known with an emphasis on the clinic and on animal models that may be of use to understand the pathophysiology of the disease. We also stress not only the diagnosis difficulties between congenital myopathies and CMSs, but also the continuum that may exist between the two.


Asunto(s)
Síndromes Miasténicos Congénitos/diagnóstico , Síndromes Miasténicos Congénitos/genética , Animales , Diagnóstico Diferencial , Modelos Animales de Enfermedad , Humanos , Mutación/genética , Síndromes Miasténicos Congénitos/terapia , Miopatías Estructurales Congénitas/diagnóstico , Miopatías Estructurales Congénitas/genética , Unión Neuromuscular/genética
6.
Am J Hum Genet ; 85(2): 155-67, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19631309

RESUMEN

We report the case of a congenital myasthenic syndrome due to a mutation in AGRN, the gene encoding agrin, an extracellular matrix molecule released by the nerve and critical for formation of the neuromuscular junction. Gene analysis identified a homozygous missense mutation, c.5125G>C, leading to the p.Gly1709Arg variant. The muscle-biopsy specimen showed a major disorganization of the neuromuscular junction, including changes in the nerve-terminal cytoskeleton and fragmentation of the synaptic gutters. Experiments performed in nonmuscle cells or in cultured C2C12 myotubes and using recombinant mini-agrin for the mutated and the wild-type forms showed that the mutated form did not impair the activation of MuSK or change the total number of induced acetylcholine receptor aggregates. A solid-phase assay using the dystrophin glycoprotein complex showed that the mutation did not affect the binding of agrin to alpha-dystroglycan. Injection of wild-type or mutated agrin into rat soleus muscle induced the formation of nonsynaptic acetylcholine receptor clusters, but the mutant protein specifically destabilized the endogenous neuromuscular junctions. Importantly, the changes observed in rat muscle injected with mutant agrin recapitulated the pre- and post-synaptic modifications observed in the patient. These results indicate that the mutation does not interfere with the ability of agrin to induce postsynaptic structures but that it dramatically perturbs the maintenance of the neuromuscular junction.


Asunto(s)
Agrina/genética , Mutación Missense , Síndromes Miasténicos Congénitos/genética , Sinapsis/metabolismo , Adulto , Agrina/química , Agrina/metabolismo , Animales , Biopsia , Línea Celular , Análisis Mutacional de ADN , Distroglicanos/metabolismo , Femenino , Humanos , Masculino , Modelos Químicos , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/cirugía , Músculo Esquelético/ultraestructura , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Unión Neuromuscular/fisiología , Unión Neuromuscular/ultraestructura , Linaje , Estructura Terciaria de Proteína , Ratas , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Receptores Colinérgicos/fisiología , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
7.
J Med Genet ; 47(12): 795-6, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20930056

RESUMEN

Congenital myasthenic syndromes (CMS) are a heterogeneous group of genetic disorders that give rise to a defect in neuromuscular transmission. We described here three patients with a characteristic phenotype of recessive CMS and presenting mutation in the gene encoding rapsyn (RAPSN). Familial analysis showed that one allelic mutation failed to be detected by direct sequencing. An allelic quantification on patient's DNA identified three novel multi-exon deletions of RAPSN. These three genomic rearrangements in RAPSN represent 15% of our CMS patients with RAPSN mutations and we emphasize that single-nucleotide polymorphism markers and a gene dosage method should be performed in addition to DNA direct sequencing analysis particularly when there is a genetic counselling issue.


Asunto(s)
Exones/genética , Proteínas Musculares/genética , Síndromes Miasténicos Congénitos/genética , Análisis de Secuencia de ADN , Eliminación de Secuencia/genética , Adolescente , Adulto , Niño , Femenino , Humanos , Lactante , Recién Nacido , Adulto Joven
8.
Hum Mol Genet ; 17(22): 3577-95, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18718936

RESUMEN

In the muscle-specific tyrosine kinase receptor gene MUSK, a heteroallelic missense and a null mutation were identified in a patient suffering from a congenital myasthenic syndrome (CMS). We generated one mouse line carrying the homozygous missense mutation V789M in musk (musk(V789M/V789M) mice) and a second hemizygous line, resembling the patient genotype, with the V789M mutation on one allele and an allele lacking the kinase domain (musk(V789M/-) mice). We report here that musk(V789M/V789M) mice present no obvious abnormal phenotype regarding weight, muscle function and viability. In contrast, adult musk(V789M/-) mice suffer from severe muscle weakness, exhibit shrinkage of pelvic and scapular regions and hunchback. Musk(V789M/-) diaphragm develops less force upon direct or nerve-induced stimulation. A profound tetanic fade is observed following nerve-evoked muscle contraction, and fatigue resistance is severely impaired upon a train of tetanic nerve stimulations. Electrophysiological measurements indicate that fatigable muscle weakness is due to impaired neurotransmission as observed in a patient suffering from a CMS. The diaphragm of adult musk(V789M/-) mice exhibits pronounced changes in endplate architecture, distribution and innervation pattern. Thus, the missense mutation V789M in MuSK acts as a hypomorphic mutation and leads to insufficiency in MuSK function in musk(V789M/-) mutants. These mutant mice represent valuable models for elucidating the roles of MuSK for synapse formation, maturation and maintenance as well as for studying the pathophysiology of a CMS due to MuSK mutations.


Asunto(s)
Diafragma/inervación , Síndromes Miasténicos Congénitos/genética , Unión Neuromuscular/fisiopatología , Proteínas Tirosina Quinasas Receptoras/genética , Animales , Axones/fisiología , Diafragma/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Cifosis , Locomoción , Masculino , Ratones , Microscopía Electrónica , Placa Motora/fisiopatología , Contracción Muscular , Debilidad Muscular , Mutación Missense , Síndromes Miasténicos Congénitos/metabolismo , Síndromes Miasténicos Congénitos/fisiopatología , Unión Neuromuscular/ultraestructura , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Periodo Refractario Electrofisiológico , Transmisión Sináptica
9.
Neuromuscul Disord ; 17(5): 409-14, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17363247

RESUMEN

Congenital myasthenic syndromes (CMSs) are rare hereditary disorders transmitted in a recessive or dominant pattern, and are caused by mutations in the genes encoding proteins of the neuromuscular junction. They are classified in three groups depending on the origin of the molecular defect. Postsynaptic defects are the most frequent and have been reported to be partly due to abnormalities of the acetylcholine receptor, and particularly to mutations in CHRNE, the gene encoding the acetylcholine receptor epsilon-subunit. In a Portuguese patient with a mild form of recessive CMS, CHRNE sequencing identified an unknown homozygous transition. This variation affects the third nucleotide of the glycine 285 condon, and leads to a synonymous variant. Analysis of transcripts demonstrated that this single change creates a new splice donor site located 4 nucleotides upstream of the normal site, leading to a deletion and generating a frameshift in exon 9 followed by a premature termination codon. This paper relates the identification of a synonymous mutation in CHRNE that creates a new splice donor site leading to an aberrant splicing of pre-mRNAs and so to their instability. This is the first synonymous mutation in CHRNE known to generate a cryptic splice site, and mRNA quantification strongly suggests that it is the disease-causing mutation.


Asunto(s)
Mutación , Síndromes Miasténicos Congénitos/genética , Empalme del ARN , Receptores Nicotínicos/genética , Adolescente , Bungarotoxinas/metabolismo , Análisis Mutacional de ADN/métodos , Femenino , Glicina/genética , Humanos , Síndromes Miasténicos Congénitos/metabolismo , Receptores Nicotínicos/metabolismo
10.
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
11.
Neuromuscul Disord ; 25(10): 773-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26248958

RESUMEN

Brody disease was first described as a benign pseudo-myotonic disorder with muscular stiffness, which increased with exercise. Biochemical and genetic studies have pointed out its close relationship to a functional defect of the fast-twitch sarcoplasmic reticulum Ca(++) ATPase pump (SERCA1) encoded by the ATP2A1 gene located on chromosome 16. The histopathological features in this form of myopathy were generally described as non-specific, i.e. moderate degree of type 2 fibre atrophy and excess of internal nuclei. We here present the clinical and histopathological features of a patient with Brody disease over a 19-year follow-up period. This patient had two heterozygous ATP2A1 mutations and complained about muscle stiffness immediately after effort. He had suffered from this since early childhood and exhibited clinical symptoms mimicking myotonia. Histological, ultrastructural and cytogenetic analyses revealed morphologically abnormal nuclei with polyploidy. In this report, we discuss the possible links between the consequences of the genetic abnormality and the peculiar aspect of the nuclei.


Asunto(s)
Núcleo Celular/patología , Músculo Esquelético/ultraestructura , Miotonía Congénita/patología , Adulto , Estudios de Seguimiento , Humanos , Masculino , Tono Muscular , Mutación , Miotonía Congénita/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética
12.
Ann Clin Transl Neurol ; 2(4): 362-72, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25909082

RESUMEN

OBJECTIVE: Data from mouse models of amyotrophic lateral sclerosis (ALS) suggest early morphological changes in neuromuscular junctions (NMJs), with loss of nerve-muscle contact. Overexpression of the neurite outgrowth inhibitor Nogo-A in muscle may play a role in this loss of endplate innervation. METHODS: We used confocal and electron microscopy to study the structure of the NMJs in muscle samples collected from nine ALS patients (five early-stage patients and four long-term survivors). We correlated the morphological results with clinical and electrophysiological data, and with Nogo-A muscle expression level. RESULTS: Surface electromyography assessment of neuromuscular transmission was abnormal in 3/9 ALS patients. The postsynaptic apparatus was morphologically altered for almost all NMJs (n = 430) analyzed using confocal microscopy. 19.7% of the NMJs were completely denervated (fragmented synaptic gutters and absence of nerve terminal profile). The terminal axonal arborization was usually sparsely branched and 56.8% of innervated NMJs showed a typical reinnervation pattern. Terminal Schwann cell (TSC) morphology was altered with extensive cytoplasmic processes. A marked intrusion of TSCs in the synaptic cleft was seen in some cases, strikingly reducing the synaptic surface available for neuromuscular transmission. Finally, high-level expression of Nogo-A in muscle was significantly associated with higher extent of NMJ denervation and negative functional outcome. INTERPRETATION: Our results support the hypothesis that morphological alterations of NMJs are present from early-stage disease and may significantly contribute to functional motor impairment in ALS patients. Muscle expression of Nogo-A is associated with NMJ denervation and thus constitutes a therapeutic target to slow disease progression.

13.
Neuromuscul Disord ; 13(3): 236-44, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12609505

RESUMEN

Congenital myasthenic syndromes with endplate acetylcholinesterase deficiency are very rare autosomal recessive diseases, characterized by onset of the disease in childhood, general weakness increased by exertion, ophthalmoplegia and refractoriness to anticholinesterase drugs. To date, all reported cases are due to mutations within the gene encoding ColQ, a specific collagen that anchors acetylcholinesterase in the basal lamina at the neuromuscular junction. We identified two new cases of congenital myasthenic syndromes with endplate acetylcholinesterase deficiency. The two patients showed different phenotypes. The first patient had mild symptoms in childhood, which worsened at 46 years with severe respiratory insufficiency. The second patient had severe symptoms from birth but improved during adolescence. In both cases, the absence of acetylcholinesterase was demonstrated by morphological and biochemical analyses, and heteroallelic mutations in the COLQ gene were found. Both patients presented a novel splicing mutation (IVS1-1G-->A) affecting the exon encoding the proline-rich attachment domain (PRAD), which interacts with acetylcholinesterase. This splicing mutation was associated with two different mutations, both of which cause truncation of the collagen domain (a known 788insC mutation belonging to one patient and a novel R236X to the other) and may impair its trimeric organization. The close similarity of the mutations of these two patients with different phenotypes suggests that other factors may modify the severity of this disease.


Asunto(s)
Acetilcolinesterasa/deficiencia , Acetilcolinesterasa/genética , Colágeno/genética , Placa Motora/enzimología , Proteínas Musculares , Mutación , Síndromes Miasténicos Congénitos/genética , Acetilcolinesterasa/metabolismo , Adulto , Alanina/genética , Animales , Bungarotoxinas , Análisis Mutacional de ADN/métodos , Exones , Femenino , Expresión Génica , Glicina/genética , Humanos , Técnicas In Vitro , Microscopía Confocal/métodos , Microscopía Electrónica/métodos , Persona de Mediana Edad , Placa Motora/genética , Placa Motora/ultraestructura , Músculo Esquelético/enzimología , Músculo Esquelético/ultraestructura , Síndromes Miasténicos Congénitos/etiología , Síndromes Miasténicos Congénitos/metabolismo , Síndromes Miasténicos Congénitos/ultraestructura , Oocitos , Reacción en Cadena de la Polimerasa/métodos , Polimorfismo Conformacional Retorcido-Simple , ARN Mensajero/biosíntesis , Ratas , Xenopus
14.
Neuromuscul Disord ; 14(1): 24-32, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14659409

RESUMEN

Congenital myasthenic syndromes are rare heterogeneous hereditary disorders, which lead to defective neuromuscular transmission resulting in fatigable muscle weakness. Post-synaptic congenital myasthenic syndromes are caused by acetylcholine receptor kinetic abnormalities or by acetylcholine receptor deficiency. Most of the congenital myasthenic syndromes with acetylcholine receptor deficiency are due to mutations in acetylcholine receptor subunit genes. Some have recently been attributed to mutations in the rapsyn gene. Here, we report the case of a 28-year-old French congenital myasthenic syndrome patient who had mild diplopia and fatigability from the age of 5 years. His muscle biopsy revealed a marked reduction in rapsyn and acetylcholine receptor at neuromuscular junctions together with a simplification of the subneural apparatus structure. In this patient, we excluded mutations in the acetylcholine receptor subunit genes and identified the homozygous N88K rapsyn mutation, which has already been shown by cell expression to impair rapsyn and acetylcholine receptor aggregation at the neuromuscular junction. The detection of the N88K mutation at the heterozygous state in five of 300 unrelated control subjects shows that this mutation is not infrequent in the healthy population. Electrophysiological measurements on biopsied intercostal muscle from this patient showed that his rapsyn mutation-induced fatigable weakness is expressed not only in a diminution in acetylcholine receptor membrane density but also in a decline of endplate potentials evoked at low frequency.


Asunto(s)
Proteínas Musculares/deficiencia , Músculo Esquelético/fisiopatología , Síndromes Miasténicos Congénitos/genética , Unión Neuromuscular/metabolismo , Receptores Nicotínicos/deficiencia , Adulto , Biopsia , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/fisiopatología , Análisis Mutacional de ADN , Regulación hacia Abajo/genética , Electrofisiología , Femenino , Frecuencia de los Genes , Genes Recesivos/genética , Haplotipos/genética , Homocigoto , Humanos , Técnicas In Vitro , Masculino , Potenciales de la Membrana/genética , Proteínas Musculares/genética , Músculo Esquelético/inervación , Músculo Esquelético/patología , Mutación/genética , Síndromes Miasténicos Congénitos/metabolismo , Síndromes Miasténicos Congénitos/fisiopatología , Unión Neuromuscular/patología , Unión Neuromuscular/ultraestructura , Linaje , Agregación de Receptores/genética , Receptores Nicotínicos/genética , Membranas Sinápticas/metabolismo , Membranas Sinápticas/patología , Membranas Sinápticas/ultraestructura , Transmisión Sináptica/genética
15.
Handb Clin Neurol ; 113: 1469-80, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23622369

RESUMEN

Congenital myasthenic syndromes (CMS) are a heterogeneous group of disorders caused by genetic defects affecting neuromuscular transmission and leading to muscle weakness accentuated by exertion. The characterization of CMS comprises two complementary steps: establishing the diagnosis and identifying the pathophysiological type of CMS. The combination of clinical, electrophysiological, and morphological studies allows the physician to refer a given CMS to mutation(s) in one of the 18 causative genes discovered to date and, in turn, to classify the CMS according to the location of the mutated proteins at the neuromuscular junction into presynaptic compartment, synaptic basal lamina, and postsynaptic compartment CMS. This complete characterization is essential for counseling and therapy of the patient, depending on the molecular background of the respective CMS. Despite comprehensive characterization, the phenotypic expression of one given gene involved is variable, and the etiology of many CMS remains to be discovered.


Asunto(s)
Síndromes Miasténicos Congénitos/diagnóstico , Unión Neuromuscular/fisiopatología , Diagnóstico Diferencial , Electromiografía , Humanos , Mutación , Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/fisiopatología , Unión Neuromuscular/genética , Fenotipo , Transmisión Sináptica/genética
16.
Neuromuscul Disord ; 23(12): 998-1009, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24011702

RESUMEN

Schwartz-Jampel syndrome (SJS) is a recessive disorder with muscle hyperactivity that results from hypomorphic mutations in the perlecan gene, a basement membrane proteoglycan. Analyses done on a mouse model have suggested that SJS is a congenital form of distal peripheral nerve hyperexcitability resulting from synaptic acetylcholinesterase deficiency, nerve terminal instability with preterminal amyelination, and subtle peripheral nerve changes. We investigated one adult patient with SJS to study this statement in humans. Perlecan deficiency due to hypomorphic mutations was observed in the patient biological samples. Electroneuromyography showed normal nerve conduction, neuromuscular transmission, and compound nerve action potentials while multiple measures of peripheral nerve excitability along the nerve trunk did not detect changes. Needle electromyography detected complex repetitive discharges without any evidence for neuromuscular transmission failure. The study of muscle biopsies containing neuromuscular junctions showed well-formed post-synaptic element, synaptic acetylcholinesterase deficiency, denervation of synaptic gutters with reinnervation by terminal sprouting, and long nonmyelinated preterminal nerve segments. These data support the notion of peripheral nerve hyperexcitability in SJS, which would originate distally from synergistic actions of peripheral nerve and neuromuscular junction changes as a result of perlecan deficiency.


Asunto(s)
Unión Neuromuscular/patología , Osteocondrodisplasias/patología , Nervios Periféricos/fisiopatología , Adulto , Proteínas de Unión al Calcio/metabolismo , Electromiografía , Humanos , Masculino , Proteína Básica de Mielina/metabolismo , Conducción Nerviosa/fisiología , Proteínas de Neurofilamentos/metabolismo , Unión Neuromuscular/metabolismo , Unión Neuromuscular/fisiopatología , Unión Neuromuscular/ultraestructura , Nervios Periféricos/metabolismo , Nervios Periféricos/patología , Nervios Periféricos/ultraestructura , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor ErbB-3/metabolismo , Receptores Colinérgicos/metabolismo , Proteínas S100/metabolismo
17.
PLoS One ; 8(1): e53826, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23326516

RESUMEN

Congenital myasthenic syndromes (CMSs) are a heterogeneous group of genetic disorders affecting neuromuscular transmission. The agrin/muscle-specific kinase (MuSK) pathway is critical for proper development and maintenance of the neuromuscular junction (NMJ). We report here an Iranian patient in whom CMS was diagnosed since he presented with congenital and fluctuating bilateral symmetric ptosis, upward gaze palsy and slowly progressive muscle weakness leading to loss of ambulation. Genetic analysis of the patient revealed a homozygous missense mutation c.2503A>G in the coding sequence of MUSK leading to the p.Met835Val substitution. The mutation was inherited from the two parents who were heterozygous according to the notion of consanguinity. Immunocytochemical and electron microscopy studies of biopsied deltoid muscle showed dramatic changes in pre- and post-synaptic elements of the NMJs. These changes induced a process of denervation/reinnervation in native NMJs and the formation, by an adaptive mechanism, of newly formed and ectopic NMJs. Aberrant axonal outgrowth, decreased nerve terminal ramification and nodal axonal sprouting were also noted. In vivo electroporation of the mutated MuSK in a mouse model showed disorganized NMJs and aberrant axonal growth reproducing a phenotype similar to that observed in the patient's biopsy specimen. In vitro experiments showed that the mutation alters agrin-dependent acetylcholine receptor aggregation, causes a constitutive activation of MuSK and a decrease in its agrin- and Dok-7-dependent phosphorylation.


Asunto(s)
Debilidad Muscular , Músculo Esquelético , Síndromes Miasténicos Congénitos , Unión Neuromuscular , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Colinérgicos/genética , Agrina/metabolismo , Animales , Niño , Células HEK293 , Humanos , Masculino , Ratones , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Debilidad Muscular/genética , Debilidad Muscular/metabolismo , Debilidad Muscular/fisiopatología , Músculo Esquelético/inervación , Músculo Esquelético/fisiopatología , Mutación Missense , Síndromes Miasténicos Congénitos/genética , Síndromes Miasténicos Congénitos/metabolismo , Síndromes Miasténicos Congénitos/fisiopatología , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Unión Neuromuscular/fisiopatología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/metabolismo , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal , Transmisión Sináptica/genética
19.
J Cell Biol ; 184(1): 31-44, 2009 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-19124654

RESUMEN

The LMNA gene encodes lamins A and C, two intermediate filament-type proteins that are important determinants of interphase nuclear architecture. Mutations in LMNA lead to a wide spectrum of human diseases including autosomal dominant Emery-Dreifuss muscular dystrophy (AD-EDMD), which affects skeletal and cardiac muscle. The cellular mechanisms by which mutations in LMNA cause disease have been elusive. Here, we demonstrate that defects in neuromuscular junctions (NMJs) are part of the disease mechanism in AD-EDMD. Two AD-EDMD mouse models show innervation defects including misexpression of electrical activity-dependent genes and altered epigenetic chromatin modifications. Synaptic nuclei are not properly recruited to the NMJ because of mislocalization of nuclear envelope components. AD-EDMD patients with LMNA mutations show the same cellular defects as the AD-EDMD mouse models. These results suggest that lamin A/C-mediated NMJ defects contribute to the AD-EDMD disease phenotype and provide insights into the cellular and molecular mechanisms for the muscle-specific phenotype of AD-EDMD.


Asunto(s)
Lamina Tipo A/fisiología , Distrofia Muscular de Emery-Dreifuss/metabolismo , Unión Neuromuscular/patología , Animales , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Modelos Animales de Enfermedad , Humanos , Lamina Tipo A/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Desnervación Muscular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/inervación , Músculo Esquelético/ultraestructura , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/patología , Transporte de Proteínas
20.
J Cell Biol ; 187(6): 859-74, 2009 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-20008564

RESUMEN

Mammalian target of rapamycin (mTOR) is a key regulator of cell growth that associates with raptor and rictor to form the mTOR complex 1 (mTORC1) and mTORC2, respectively. Raptor is required for oxidative muscle integrity, whereas rictor is dispensable. In this study, we show that muscle-specific inactivation of mTOR leads to severe myopathy, resulting in premature death. mTOR-deficient muscles display metabolic changes similar to those observed in muscles lacking raptor, including impaired oxidative metabolism, altered mitochondrial regulation, and glycogen accumulation associated with protein kinase B/Akt hyperactivation. In addition, mTOR-deficient muscles exhibit increased basal glucose uptake, whereas whole body glucose homeostasis is essentially maintained. Importantly, loss of mTOR exacerbates the myopathic features in both slow oxidative and fast glycolytic muscles. Moreover, mTOR but not raptor and rictor deficiency leads to reduced muscle dystrophin content. We provide evidence that mTOR controls dystrophin transcription in a cell-autonomous, rapamycin-resistant, and kinase-independent manner. Collectively, our results demonstrate that mTOR acts mainly via mTORC1, whereas regulation of dystrophin is raptor and rictor independent.


Asunto(s)
Proteínas Portadoras/metabolismo , Distrofina/metabolismo , Músculo Esquelético/enzimología , Distrofia Muscular Animal/enzimología , Fosfotransferasas (Aceptor de Grupo Alcohol)/deficiencia , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Factores de Edad , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Células Cultivadas , Distrofina/genética , Electroporación , Metabolismo Energético , Activación Enzimática , Femenino , Glucosa/metabolismo , Glucógeno/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Musculares/enzimología , Contracción Muscular , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatología , Mutación , Oxidación-Reducción , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Asociada al mTOR Insensible a la Rapamicina , Ratas , Proteína Reguladora Asociada a mTOR , Índice de Severidad de la Enfermedad , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Transducción Genética , Utrofina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA