Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Epilepsia ; 52(4): e23-5, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21371021

RESUMEN

Most patients with Dravet syndrome have de novo mutations in the neuronal voltage-gated sodium channel type 1 (SCN1A) gene. We report on two unrelated fathers with severe childhood epilepsy compatible with a possible diagnosis of Dravet syndrome, who both have a child with Dravet syndrome. Analysis of the SCN1A gene revealed a pathogenic mutation in both children. One father exhibited somatic mosaicism for the mutation detected in his son. A relatively favorable cognitive outcome in patients with Dravet syndrome patients may be explained by somatic mosaicism for the SCN1A mutation in brain tissue. A mild form of Dravet syndrome in adult patients is associated with a high recurrence risk and possibly a more severe epilepsy phenotype in their offspring.


Asunto(s)
Epilepsias Mioclónicas/diagnóstico , Epilepsias Mioclónicas/genética , Predisposición Genética a la Enfermedad/genética , Proteínas del Tejido Nervioso/genética , Canales de Sodio/genética , Adolescente , Adulto , Preescolar , Femenino , Humanos , Masculino , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.1 , Síndrome
2.
Epilepsia ; 47(10): 1743-6, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17054699

RESUMEN

PURPOSE: The EFHC1 gene, encoding a protein with a Ca(2+)-sensing EF-hand motif, is localized at 6p12 and was recently reported as mutated in six Mexican juvenile myoclonic epilepsy (JME) families linked to this region. We had previously confirmed linkage between JME and 6p11-12 in 18 Dutch families, and shown exclusionary lod scores at 6p21.3. We therefore evaluated the relevance of EFHC1 in our set of 6p11-12-linked families. METHODS: We screened all coding and regulatory regions of EFHC1 by direct sequencing, and the detected variants were tested in a case-control association study. RESULTS: We found none of the five mutations previously reported in the Mexican families, but identified nine variants, three of which are novel: 5' upstream region (c.-146_147delGC), nonsynonymous (R159W, R182H, M448T, I619L), intronic (IVS3 + 10A>G, IVS8 + 175_176delTT, IVS10 + 59C>T), and 3' UTR (c.+121C>A). These variants did not cosegregate with JME and did not account for the observed linkage at the 6p11-12 locus. Furthermore, no significant association was detected between JME and these variants in 112 unrelated patients and 180 controls. Finally, none of the mutations reported in Mexican families was found in 100 unrelated patients. CONCLUSIONS: We found no evidence that EFHC1 is a major genetic risk factor for JME susceptibility in Dutch patients. The EFHC1 variants reported in Mexican families may be mendelian variants specific for those families, suggesting that for Dutch patients and possibly many other populations, the main disease variant at the 6p11-12 is yet to be identified.


Asunto(s)
Cromosomas Humanos Par 6/genética , Familia , Mutación/genética , Epilepsia Mioclónica Juvenil/genética , Proteínas de Unión al Calcio/genética , Femenino , Heterogeneidad Genética , Ligamiento Genético , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Masculino , México , Epilepsia Mioclónica Juvenil/epidemiología , Países Bajos , Linaje , Factores de Riesgo , Población Blanca
3.
Hum Mol Genet ; 14(1): 171-8, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15548544

RESUMEN

Photoparoxysmal response (PPR) is an abnormal visual sensitivity of the brain in reaction to intermittent photic stimulation. It is an epilepsy-related electroencephalographic trait with high prevalence in idiopathic epilepsies, especially in common idiopathic generalized epilepsies (IGEs), such as childhood absence epilepsy and juvenile myoclonic epilepsy. This degree of co-morbidity suggests that PPR may be involved in the predisposition to IGE. The identification of genes for PPR would, therefore, aid the dissection of the genetic basis of IGE. Sixteen PPR-multiplex families were collected to conduct a genome-wide linkage scan using broad (all PPR types) and narrow (exclusion of PPR types I and II and the occipital epilepsy cases) models of affectedness for PPR. We found an empirical genome-wide significance for parametric (HLOD) and non-parametric (NPL) linkage (Pgw(HLOD)=0.004 and Pgw(NPL)=0.01) for two respective chromosomal regions, 7q32 at D7S1804 (HLOD=3.47 with alpha=1, P(NPL)=3.39x10(-5)) and 16p13 at D16S3395 (HLOD=2.44 with alpha=1, P(NPL)=7.91x10(-5)). These two genomic regions contain genes that are important for the neuromodulation of cortical dynamics and may represent good targets for candidate-gene studies. Our study identified two susceptibility loci for PPR, which may be related to the underlying myoclonic epilepsy phenotype present in the families studied.


Asunto(s)
Cromosomas Humanos Par 16/genética , Cromosomas Humanos Par 7/genética , Epilepsia Tipo Ausencia/genética , Ligamiento Genético , Predisposición Genética a la Enfermedad/genética , Genoma Humano , Epilepsia Mioclónica Juvenil/genética , Mapeo Cromosómico , Humanos
4.
Epilepsia ; 45(3): 211-7, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15009221

RESUMEN

PURPOSE: Previous linkage studies provided evidence for juvenile myoclonic epilepsy (JME) susceptibility loci at 6p11-12, HLA-6p21.3 region, 15q14, and 5q34. These results indicate locus heterogeneity or interpopulation differences, thus underlining the importance of replication studies. METHODS: We describe a replication linkage study of the 6p-q13 region in 18 families ascertained from JME probands of Dutch descent. In the presence of heterogeneity, the definition of the disease status may be crucial, and we therefore used two disease phenotypes: narrow [JME/idiopathic generalized epilepsy (IGE)-"only"] and broad (JME/IGE-plus-fast EEG background activity). RESULTS: We found evidence of linkage at 6p11-12 in multipoint analyses (p < 0.01 in a replication study) for both these disease definitions. Analysis of this region, assuming heterogeneity and autosomal dominant inheritance with a conservative 60% of penetrance, gave a maximum multipoint parametric lod score of 2.07 at D6S1573 for the narrow phenotype and peaked at 2.53 between D6S1623 and D6S1573 for the broad phenotype. The p value for nonparametric linkage reached 0.0013 for the narrow phenotype and 0.0010 for the broad. Significant exclusion (lod score

Asunto(s)
Cromosomas Humanos Par 6/genética , Epilepsia Generalizada/etnología , Epilepsia Generalizada/genética , Ligamiento Genético/genética , Epilepsia Mioclónica Juvenil/etnología , Epilepsia Mioclónica Juvenil/genética , Adolescente , Adulto , Niño , Mapeo Cromosómico/métodos , Electroencefalografía , Epilepsia Generalizada/diagnóstico , Femenino , Heterogeneidad Genética , Marcadores Genéticos , Genoma Humano , Genotipo , Humanos , Masculino , Epilepsia Mioclónica Juvenil/diagnóstico , Países Bajos , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA