Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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.

7.
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
8.
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
9.
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
10.
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
11.
J Pathol ; 207(3): 313-23, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16178054

RESUMEN

Tubular aggregates are morphological abnormalities characterized by the accumulation of densely packed tubules in skeletal muscle fibres. To improve knowledge of tubular aggregates, the formation and role of which are still unclear, the present study reports the electron microscopic analysis and protein characterization of tubular aggregates in six patients with 'tubular aggregate myopathy'. Three of the six patients also presented with myasthenic features. A large panel of immunochemical markers located in the sarcoplasmic reticulum, T-tubules, mitochondria, and nucleus was used. Despite differences in clinical phenotype, the composition of tubular aggregates, which contained proteins normally segregated differently along the sarcoplasmic reticulum architecture, was similar in all patients. All of these proteins, calsequestrin, RyR, triadin, SERCAs, and sarcalumenin, are involved in calcium uptake, storage, and release. The dihydropyridine receptor, DHPR, specifically located in the T-tubule, was also present in tubular aggregates in all patients. COX-2 and COX-7 mitochondrial proteins were not found in tubular aggregates, despite being observed close to them in the muscle fibre. The nuclear membrane protein emerin was found in only one case. Electron microscopy revealed vesicular budding from nuclei, and the presence of SAR-1 GTPase protein in tubular aggregates shown by immunochemistry, in all patients, suggests that tubular aggregates could arise from endoplasmic reticulum exit sites. Taken together, these results cast new light on the composition and significance of tubular aggregates.


Asunto(s)
Músculo Esquelético/patología , Miopatías Estructurales Congénitas/patología , Adulto , Calcio/metabolismo , ATPasas Transportadoras de Calcio/análisis , Núcleo Celular/patología , Femenino , Humanos , Inmunohistoquímica/métodos , Masculino , Microscopía Electrónica/métodos , Persona de Mediana Edad , Mitocondrias Musculares/patología , Proteínas Mitocondriales/análisis , Fibras Musculares Esqueléticas/patología , Proteínas Musculares/análisis , Miopatías Estructurales Congénitas/fisiopatología , Retículo Sarcoplasmático/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico
12.
J Soc Biol ; 199(1): 61-77, 2005.
Artículo en Francés | MEDLINE | ID: mdl-16114265

RESUMEN

Congenital myasthenic syndromes (CMS) are rare genetic diseases affecting the neuromuscular junction (NMJ) and are characterized by a dysfunction of the neurotransmission. They are heterogeneous at their pathophysiological level and can be classified in three categories according to their presynaptic, synaptic and postsynaptic origins. We report here the first case of a human neuromuscular transmission dysfunction due to mutations in the gene encoding a postsynaptic molecule, the muscle-specific receptor tyrosine kinase (MuSK). Gene analysis identified two heteroallelic mutations, a frameshift mutation (c.220insC) and a missense mutation (V790M). The muscle biopsy showed dramatic pre- and postsynaptic structural abnormalities of the neuromuscular junction and severe decrease in acetylcholine receptor (AChR) epsilon-subunit and MuSK expression. In vitro and in vivo expression experiments were performed using mutant MuSK reproducing the human mutations. The frameshift mutation led to the absence of MuSK expression. The missense mutation did not affect MuSK catalytic kinase activity but diminished expression and stability of MuSK leading to decreased agrin-dependent AChR aggregation, a critical step in the formation of the neuromuscular junction. In electroporated mouse muscle, overexpression of the missense mutation induced, within a week, a phenotype similar to the patient muscle biopsy: a severe decrease in synaptic AChR and an aberrant axonal outgrowth. These results strongly suggest that the missense mutation, in the presence of a null mutation on the other allele, is responsible for the dramatic synaptic changes observed in the patient.


Asunto(s)
Mutación , Síndromes Miasténicos Congénitos/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Colinérgicos/genética , Animales , Axones/patología , Mutación del Sistema de Lectura , Expresión Génica , Humanos , Masculino , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Mutación Missense , Síndromes Miasténicos Congénitos/enzimología , Síndromes Miasténicos Congénitos/patología , Unión Neuromuscular/química , Unión Neuromuscular/enzimología , Unión Neuromuscular/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/análisis , Receptores Colinérgicos/metabolismo , Sinapsis/química , Sinapsis/patología , Sinapsis/fisiología , Transfección
13.
Curr Opin Neurol ; 17(5): 539-51, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15367858

RESUMEN

PURPOSE OF REVIEW: Congenital myasthenic syndromes are a heterogeneous group of diseases caused by genetic defects affecting neuromuscular transmission. In this article, a strategy that leads to the diagnosis of congenital myasthenic syndromes is presented, and recent advances in the clinical, genetic and molecular aspects of congenital myasthenic syndrome are outlined. RECENT FINDINGS: Besides the identification of new mutations in genes already known to be implicated in congenital myasthenic syndromes (genes for the acetylcholine receptor subunits and the collagen tail of acetylcholinesterase), mutations in other genes have more recently been discovered and characterized (genes for choline acetyltransferase, rapsyn, and the muscle sodium channel SCN4A). Fluoxetine has recently been proposed as an alternative treatment for 'slow channel' congenital myasthenic syndrome. SUMMARY: The characterization of congenital myasthenic syndromes comprises two complementary steps: establishing the diagnosis and identifying the pathophysiological type of congenital myasthenic syndrome. Characterization of the type of congenital myasthenic syndrome has allowed it to be classified as caused by presynaptic, synaptic and postsynaptic defects. A clinically and muscle histopathologically oriented genetic study has identified several genes in which mutations cause the disease. Despite comprehensive characterization, the phenotypic expression of one given gene involved is variable, and the aetiology of many congenital myasthenic syndromes remains to be discovered.


Asunto(s)
Síndromes Miasténicos Congénitos/diagnóstico , Síndromes Miasténicos Congénitos/fisiopatología , Acetilcolinesterasa/deficiencia , Animales , Genotipo , Humanos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutación , Síndromes Miasténicos Congénitos/clasificación , Síndromes Miasténicos Congénitos/genética , Unión Neuromuscular/fisiología , Fenotipo , Pronóstico
14.
J Cell Physiol ; 196(1): 105-12, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12767046

RESUMEN

Regulation of thrombin activity may be required during skeletal muscle differentiation since the thrombin tissue inhibitor protease nexin-1 appears at the myotube stage before being localized at the neuromuscular synapse. Here, we have used a model of rat fetal myotube primary cultures to study the effect of thrombin on acetylcholine receptor (AChR) expression, which is enhanced at the myotube stage. Our results show that thrombin decreases both the number of surface AChRs (AChRn) and AChR alpha-subunit gene expression. Using the agonist peptide SFLLRN, we establish that the AChRn decrease is mediated by the G protein-coupled thrombin receptor "protease-activated receptor-1" (PAR-1). Moreover, the specific thrombin inhibitor hirudin increases AChRn by inhibiting the thrombin intrinsically present in the cultures. We further demonstrate that the activation of PAR-1 by thrombin induces intracellular calcium movements that are blocked by 2-APB, an inhibitor of inositol 1,4,5-triphosphate (IP3)-induced calcium release. These calcium signals are more intense in nuclei than in the cytoplasm and are consistent with the intracellular distribution of IP3 receptor that we find in the cytoplasm in a cross-striated pattern and at a high level in the nuclear envelope zone. Finally, we show that the blockade of these IP3-induced calcium signals by 2-APB prevents the AChRn decrease induced by thrombin. Our results thus demonstrate that thrombin downregulates AChR expression by activating PAR-1 and that this effect is mediated via an IP3 signaling pathway.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas/metabolismo , Inositol 1,4,5-Trifosfato/metabolismo , Receptores Colinérgicos/metabolismo , Receptores de Trombina/metabolismo , Transducción de Señal/efectos de los fármacos , Trombina/farmacología , Acetilcolina/farmacología , Animales , Compuestos de Boro/farmacología , Calcio/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Fluorescencia , Músculo Esquelético/citología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Oligopéptidos/farmacología , Fragmentos de Péptidos/farmacología , Subunidades de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptor PAR-1 , Receptores Colinérgicos/química , Receptores Colinérgicos/genética , Receptores de Trombina/agonistas
15.
Eur J Neurosci ; 19(8): 2099-108, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15090037

RESUMEN

In the course of studies on thrombin and its inhibitor(s) in synaptic plasticity, we addressed the question of their roles in the formation of neuromuscular junctions (NMJ) and used a model of rat neuron-myotube cocultures. We report that the size of acetylcholinesterase (AChE) patches used as a marker of neuromuscular contacts was decreased in the presence of either thrombin or SFLLRN, the agonist peptide of the thrombin receptor PAR-1, whereas it was increased with hirudin, a specific thrombin inhibitor. In an attempt to relate these neuromuscular contact size variations to molecular changes, we studied muscle-specific tyrosine kinase receptor (MuSK), acetylcholine receptor (AChR) and rapsyn expression in the presence of thrombin. We showed that thrombin did not change rapsyn gene and protein expression. However, the expression of MuSK and surface AChR proteins was diminished in both myotube cultures and neuron-myotube cocultures. These reductions in protein expression were associated with a decrease in MuSK and AChR alpha-subunit gene expression in myotube cultures but not in neuron-myotube cocultures. Moreover, the expression of the AChR epsilon-subunit gene, specifically enhanced by neuron-released factors, was not modified by thrombin in neuron-myotube cocultures. This suggests that thrombin did not affect the expression of synaptic AChRs enhanced by neuron-released factors but rather reduced the level of extrasynaptic AChRs. Taken together, these results indicate that thrombin in balance with its inhibitor(s) could modulate the formation of neuromuscular contacts in vitro by affecting the expression of two essential molecules in NMJ postsynaptic differentiation, MuSK and AChR.


Asunto(s)
Unión Neuromuscular/efectos de los fármacos , Unión Neuromuscular/metabolismo , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores Colinérgicos/biosíntesis , Trombina/farmacología , Animales , Células Cultivadas , Femenino , Embarazo , Ratas , Ratas Sprague-Dawley
16.
Hum Mol Genet ; 13(24): 3229-40, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15496425

RESUMEN

We report the first case of a human neuromuscular transmission dysfunction due to mutations in the gene encoding the muscle-specific receptor tyrosine kinase (MuSK). Gene analysis identified two heteroallelic mutations, a frameshift mutation (c.220insC) and a missense mutation (V790M). The muscle biopsy showed dramatic pre- and postsynaptic structural abnormalities of the neuromuscular junction and severe decrease in acetylcholine receptor (AChR) epsilon-subunit and MuSK expression. In vitro and in vivo expression experiments were performed using mutant MuSK reproducing the human mutations. The frameshift mutation led to the absence of MuSK expression. The missense mutation did not affect MuSK catalytic kinase activity but diminished expression and stability of MuSK leading to decreased agrin-dependent AChR aggregation, a critical step in the formation of the neuromuscular junction. In electroporated mouse muscle, overexpression of the missense mutation induced, within a week, a phenotype similar to the patient muscle biopsy: a severe decrease in synaptic AChR and an aberrant axonal outgrowth. These results strongly suggest that the missense mutation, in the presence of a null mutation on the other allele, is responsible for the dramatic synaptic changes observed in the patient.


Asunto(s)
Síndromes Miasténicos Congénitos/genética , Proteínas Tirosina Quinasas Receptoras/genética , Receptores Colinérgicos/genética , Adulto , Análisis Mutacional de ADN , Femenino , Humanos , Inmunohistoquímica , Masculino , Músculos/metabolismo , Mutación , Síndromes Miasténicos Congénitos/metabolismo , Linaje , Polimorfismo Genético , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Colinérgicos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA