Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurol Sci ; 45(4): 1635-1643, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37875597

RESUMEN

Juvenile myoclonic epilepsy (JME) is the most common of the generalized genetic epilepsies, with multiple causal and susceptibility genes; however, its etiopathogenesis is mainly unknown. The toxic effects caused by xenobiotics in cells occur during their metabolic transformation, mainly by enzymes belonging to cytochrome P450. The elimination of these compounds by transporters of the ABC type protects the central nervous system, but their accumulation causes neuronal damage, resulting in neurological diseases. The present study has sought the association between single nucleotide genetic variants of the CYP2C9, CYP2C19, and ABCB1 genes and the development of JME in patients compared to healthy controls. The CC1236 and GG2677 genotypes of ABCB1 in women; allele G 2677, genotypes GG 2677 and CC 3435 in men; the CYP2C19*2A allele, and the CYP2C19*3G/A genotype in both sexes were found to be risk factors for JME. Furthermore, carriers of the TTGGCC genotype combination of the ABCB1 gene (1236/2677/3435) have a 10.5 times higher risk of developing JME than non-carriers. Using the STRING database, we found an interaction between the proteins encoded by these genes and other possible proteins. These findings indicate that the CYP450 system and ABC transporters could interact with other genes in the JME.


Asunto(s)
Epilepsia Generalizada , Epilepsia Mioclónica Juvenil , Masculino , Humanos , Femenino , Epilepsia Mioclónica Juvenil/genética , Citocromo P-450 CYP2C9/genética , Citocromo P-450 CYP2C19/genética , Genotipo , Subfamilia B de Transportador de Casetes de Unión a ATP/genética
2.
N Engl J Med ; 378(11): 1018-1028, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29539279

RESUMEN

BACKGROUND: In juvenile myoclonic epilepsy, data are limited on the genetic basis of networks promoting convulsions with diffuse polyspikes on electroencephalography (EEG) and the subtle microscopic brain dysplasia called microdysgenesis. METHODS: Using Sanger sequencing, we sequenced the exomes of six members of a large family affected with juvenile myoclonic epilepsy and confirmed cosegregation in all 37 family members. We screened an additional 310 patients with this disorder for variants on DNA melting-curve analysis and targeted real-time DNA sequencing of the gene encoding intestinal-cell kinase ( ICK). We calculated Bayesian logarithm of the odds (LOD) scores for cosegregating variants, odds ratios in case-control associations, and allele frequencies in the Genome Aggregation Database. We performed functional tests of the effects of variants on mitosis, apoptosis, and radial neuroblast migration in vitro and conducted video-EEG studies in mice lacking a copy of Ick. RESULTS: A variant, K305T (c.914A→C), cosegregated with epilepsy or polyspikes on EEG in 12 members of the family affected with juvenile myoclonic epilepsy. We identified 21 pathogenic ICK variants in 22 of 310 additional patients (7%). Four strongly linked variants (K220E, K305T, A615T, and R632X) impaired mitosis, cell-cycle exit, and radial neuroblast migration while promoting apoptosis. Tonic-clonic convulsions and polyspikes on EEG resembling seizures in human juvenile myoclonic epilepsy occurred more often in knockout heterozygous mice than in wild-type mice (P=0.02) during light sleep with isoflurane anesthesia. CONCLUSIONS: Our data provide evidence that heterozygous variants in ICK caused juvenile myoclonic epilepsy in 7% of the patients included in our analysis. Variant ICK affects cell processes that help explain microdysgenesis and polyspike networks observed on EEG in juvenile myoclonic epilepsy. (Funded by the National Institutes of Health and others.).


Asunto(s)
Mutación , Epilepsia Mioclónica Juvenil/genética , Proteínas Serina-Treonina Quinasas/genética , Adolescente , Animales , Teorema de Bayes , Estudios de Casos y Controles , Niño , Preescolar , Cromosomas Humanos Par 6 , Modelos Animales de Enfermedad , Electroencefalografía , Femenino , Heterocigoto , Humanos , Lactante , Recién Nacido , Masculino , Malformaciones del Desarrollo Cortical/genética , Ratones , Ratones Noqueados , Epilepsia Mioclónica Juvenil/fisiopatología , Análisis de Secuencia de ADN , Adulto Joven
4.
Genet Med ; 19(2): 144-156, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27467453

RESUMEN

PURPOSE: EFHC1 variants are the most common mutations in inherited myoclonic and grand mal clonic-tonic-clonic (CTC) convulsions of juvenile myoclonic epilepsy (JME). We reanalyzed 54 EFHC1 variants associated with epilepsy from 17 cohorts based on National Human Genome Research Institute (NHGRI) and American College of Medical Genetics and Genomics (ACMG) guidelines for interpretation of sequence variants. METHODS: We calculated Bayesian LOD scores for variants in coinheritance, unconditional exact tests and odds ratios (OR) in case-control associations, allele frequencies in genome databases, and predictions for conservation/pathogenicity. We reviewed whether variants damage EFHC1 functions, whether efhc1-/- KO mice recapitulate CTC convulsions and "microdysgenesis" neuropathology, and whether supernumerary synaptic and dendritic phenotypes can be rescued in the fly model when EFHC1 is overexpressed. We rated strengths of evidence and applied ACMG combinatorial criteria for classifying variants. RESULTS: Nine variants were classified as "pathogenic," 14 as "likely pathogenic," 9 as "benign," and 2 as "likely benign." Twenty variants of unknown significance had an insufficient number of ancestry-matched controls, but ORs exceeded 5 when compared with racial/ethnic-matched Exome Aggregation Consortium (ExAC) controls. CONCLUSIONS: NHGRI gene-level evidence and variant-level evidence establish EFHC1 as the first non-ion channel microtubule-associated protein whose mutations disturb R-type VDCC and TRPM2 calcium currents in overgrown synapses and dendrites within abnormally migrated dislocated neurons, thus explaining CTC convulsions and "microdysgenesis" neuropathology of JME.Genet Med 19 2, 144-156.


Asunto(s)
Proteínas de Unión al Calcio/genética , Epilepsia Mioclónica Juvenil/genética , Convulsiones/genética , Animales , Dendritas/patología , Exoma , Frecuencia de los Genes , Humanos , Ratones , Ratones Noqueados , Mutación , Epilepsia Mioclónica Juvenil/fisiopatología , National Human Genome Research Institute (U.S.) , Neuronas/patología , Linaje , Polimorfismo de Nucleótido Simple , Convulsiones/fisiopatología , Sinapsis/patología , Estados Unidos
5.
Epilepsy Behav ; 61: 34-40, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27300146

RESUMEN

Juvenile myoclonic epilepsy (JME) is a genetic generalized epilepsy accounting for 3-12% of adult cases of epilepsy. Valproate has proven to be the first-choice drug in JME for controlling the most common seizure types: myoclonic, absence, and generalized tonic-clonic (GTC). In this retrospective study, we analyzed seizure outcome in patients with JME using valproate monotherapy for a minimum period of one year. Low valproate dose was considered to be 1000mg/day or lower, while serum levels were considered to be low if they were at or below 50mcg/dl. One hundred three patients met the inclusion criteria. Fifty-six patients (54.4%) were female. The current average age was 28.4±7.4years, while the age of epilepsy onset was 13.6±2.9years. Most patients corresponded to the subsyndrome of classic JME. Forty-six (44.7%) patients were free from all seizure types, and 76 (73.7%) patients were free from GTC seizures. No significant difference was found in seizure freedom among patients using a low dose of valproate versus a high dose (p=0.535) or among patients with low blood levels versus high blood levels (p=0.69). In patients with JME, it seems appropriate to use low doses of valproate (500mg to 1000mg) for initial treatment and then to determine if freedom from seizures was attained.


Asunto(s)
Anticonvulsivantes/administración & dosificación , Anticonvulsivantes/uso terapéutico , Epilepsia Mioclónica Juvenil/tratamiento farmacológico , Ácido Valproico/administración & dosificación , Ácido Valproico/uso terapéutico , Adolescente , Adulto , Edad de Inicio , Anticonvulsivantes/sangre , Niño , Relación Dosis-Respuesta a Droga , Electroencefalografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Cooperación del Paciente , Estudios Retrospectivos , Factores de Riesgo , Resultado del Tratamiento , Ácido Valproico/sangre , Adulto Joven
6.
Hum Mol Genet ; 21(23): 5106-17, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22926142

RESUMEN

Heterozygous mutations in Myoclonin1/EFHC1 cause juvenile myoclonic epilepsy (JME), the most common form of genetic generalized epilepsies, while homozygous F229L mutation is associated with primary intractable epilepsy in infancy. Heterozygous mutations in adolescent JME patients produce subtle malformations of cortical and subcortical architecture, whereas homozygous F229L mutation in infancy induces severe brain pathology and death. However, the underlying pathological mechanisms for these observations remain unknown. We had previously demonstrated that EFHC1 is a microtubule-associated protein (MAP) involved in cell division and radial migration during cerebral corticogenesis. Here, we show that JME mutations, including F229L, do not alter the ability of EFHC1 to colocalize with the centrosome and the mitotic spindle, but act in a dominant-negative manner to impair mitotic spindle organization. We also found that mutants EFHC1 expression disrupted radial and tangential migration by affecting the morphology of radial glia and migrating neurons. These results show how Myoclonin1/EFHC1 mutations disrupt brain development and potentially produce structural brain abnormalities on which epileptogenesis is established.


Asunto(s)
Encéfalo/embriología , Encéfalo/metabolismo , Proteínas de Unión al Calcio/genética , Mutación , Epilepsia Mioclónica Juvenil/embriología , Epilepsia Mioclónica Juvenil/genética , Animales , Proteínas de Unión al Calcio/metabolismo , Ciclo Celular/genética , Línea Celular , Movimiento Celular/genética , Proliferación Celular , Humanos , Espacio Intracelular/metabolismo , Ratones , Neuroglía/metabolismo , Neuronas/metabolismo , Transporte de Proteínas , Ratas , Huso Acromático/genética , Huso Acromático/metabolismo , Células Madre/metabolismo
7.
Hum Mol Genet ; 20(21): 4248-57, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21835885

RESUMEN

Mutations in the EFHC1 gene have been linked to juvenile myoclonic epilepsy. To understand EFHC1 function in vivo, we generated knockout Drosophila for the fly homolog Defhc1.1. We found that the neuromuscular junction synapse of Defhc1.1 mutants displays an increased number of satellite boutons resulting in increased spontaneous neurotransmitter release. Defhc1.1 binds to microtubules in vitro and overlaps in vivo with axonal and synaptic microtubules. Elimination of Defhc1.1 from synaptic terminals reduces the number of microtubule loops, suggesting that Defhc1.1 is a negative regulator of microtubule dynamics. In fact, pharmacological treatment of Defhc1.1 mutants with vinblastine, an inhibitor of microtubule dynamics, suppresses the satellite bouton phenotype. Furthermore, Defhc1.1 mutants display overgrowth of the dendritic arbor and Defhc1.1 overexpression reduces dendrite elaboration. These results suggest that Defhc1.1 functions as an inhibitor of neurite growth by finely tuning the microtubule cytoskeleton dynamics and that EFHC1-dependent juvenile myoclonic epilepsy may result from augmented spontaneous neurotransmitter release due to overgrowth of neuronal processes.


Asunto(s)
Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas de Microtúbulos/metabolismo , Epilepsia Mioclónica Juvenil/genética , Homología de Secuencia de Aminoácido , Animales , Espinas Dendríticas/metabolismo , Proteínas de Drosophila/genética , Potenciales Evocados , Proteínas de Microtúbulos/genética , Microtúbulos/metabolismo , Mutación/genética , Epilepsia Mioclónica Juvenil/patología , Neurotransmisores/metabolismo , Terminales Presinápticos/metabolismo , Unión Proteica
8.
Epilepsy Behav ; 28 Suppl 1: S52-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23756480

RESUMEN

Introduced into a specific population, a juvenile myoclonic epilepsy (JME) mutation generates linkage disequilibrium (LD). Linkage disequilibrium is strongest when the JME mutation is of recent origin, still "hitchhiking" alleles surrounding it, as a haplotype into the next thousands of generations. Recombinations decay LD over tens of thousands of generations causing JME alleles to produce smaller genetic displacements, requiring other genes or environment to produce an epilepsy phenotype. Family-based linkage analysis captures rare epilepsy alleles and their "hitchhiking" haplotypes, transmitted as Mendelian traits, supporting the common disease/multiple rare allele model. Genome-wide association studies identify JME alleles whose linkage disequilibrium has decayed through thousands of generations and are sorting out the common disease/common allele versus rare allele models. Five Mendelian JME genes have been identified, namely, CACNB4, CASR, GABRa1, GABRD, and Myoclonin1/EFHC1. Three SNP alleles in BRD2, Cx-36, and ME2 and microdeletions in 15q13.3, 15q11.2, and 16p13.11 also contribute risk to JME.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Mutación/genética , Epilepsia Mioclónica Juvenil/genética , Alelos , Animales , Estudio de Asociación del Genoma Completo , Haplotipos , Humanos
9.
Epilepsy Behav ; 28 Suppl 1: S87-90, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23756490

RESUMEN

An international workshop on juvenile myoclonic epilepsy (JME) was conducted in Avignon, France in May 2011. During that workshop, a group of 45 experts on JME, together with one of the founding fathers of the syndrome of JME ("Janz syndrome"), Prof. Dr. Dieter Janz from Berlin, reached a consensus on diagnostic criteria and management of JME. The international experts on JME proposed two sets of criteria, which will be helpful for both clinical and scientific purposes. Class I criteria encompass myoclonic jerks without loss of consciousness exclusively occurring on or after awakening and associated with typical generalized epileptiform EEG abnormalities, with an age of onset between 10 and 25. Class II criteria allow the inclusion of myoclonic jerks predominantly occurring after awakening, generalized epileptiform EEG abnormalities with or without concomitant myoclonic jerks, and a greater time window for age at onset (6-25years). For both sets of criteria, patients should have a clear history of myoclonic jerks predominantly occurring after awakening and an EEG with generalized epileptiform discharges supporting a diagnosis of idiopathic generalized epilepsy. Patients with JME require special management because their epilepsy starts in the vulnerable period of adolescence and, accordingly, they have lifestyle issues that typically increase the likelihood of seizures (sleep deprivation, exposure to stroboscopic flashes in discos, alcohol intake, etc.) with poor adherence to antiepileptic drugs (AEDs). Results of an inventory of the different clinical management strategies are given. This article is part of a supplemental special issue entitled Juvenile Myoclonic Epilepsy: What is it Really?


Asunto(s)
Consenso , Manejo de la Enfermedad , Epilepsia Mioclónica Juvenil/diagnóstico , Epilepsia Mioclónica Juvenil/terapia , Humanos , Cooperación Internacional
10.
Nat Genet ; 35(2): 125-7, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12958597

RESUMEN

Lafora progressive myoclonus epilepsy is characterized by pathognomonic endoplasmic reticulum (ER)-associated polyglucosan accumulations. We previously discovered that mutations in EPM2A cause Lafora disease. Here, we identify a second gene associated with this disease, NHLRC1 (also called EPM2B), which encodes malin, a putative E3 ubiquitin ligase with a RING finger domain and six NHL motifs. Laforin and malin colocalize to the ER, suggesting they operate in a related pathway protecting against polyglucosan accumulation and epilepsy.


Asunto(s)
Proteínas Portadoras/genética , Mutación , Epilepsias Mioclónicas Progresivas/genética , Proteínas Tirosina Fosfatasas/genética , Secuencia de Bases , Estudios de Cohortes , Femenino , Homocigoto , Humanos , Enfermedad de Lafora/genética , Masculino , Datos de Secuencia Molecular , Epilepsias Mioclónicas Progresivas/enzimología , Linaje , Proteínas Tirosina Fosfatasas no Receptoras , Eliminación de Secuencia , Ubiquitina-Proteína Ligasas
11.
Nat Genet ; 36(8): 842-9, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15258581

RESUMEN

Juvenile myoclonic epilepsy (JME) is the most frequent cause of hereditary grand mal seizures. We previously mapped and narrowed a region associated with JME on chromosome 6p12-p11 (EJM1). Here, we describe a new gene in this region, EFHC1, which encodes a protein with an EF-hand motif. Mutation analyses identified five missense mutations in EFHC1 that cosegregated with epilepsy or EEG polyspike wave in affected members of six unrelated families with JME and did not occur in 382 control individuals. Overexpression of EFHC1 in mouse hippocampal primary culture neurons induced apoptosis that was significantly lowered by the mutations. Apoptosis was specifically suppressed by SNX-482, an antagonist of R-type voltage-dependent Ca(2+) channel (Ca(v)2.3). EFHC1 and Ca(v)2.3 immunomaterials overlapped in mouse brain, and EFHC1 coimmunoprecipitated with the Ca(v)2.3 C terminus. In patch-clamp analysis, EFHC1 specifically increased R-type Ca(2+) currents that were reversed by the mutations associated with JME.


Asunto(s)
Epilepsia Mioclónica Juvenil/genética , Animales , Apoptosis/genética , Proteínas de Unión al Calcio/genética , Células Cultivadas , Humanos , Ratones , Datos de Secuencia Molecular , Mutación Missense , Linaje
12.
Epilepsia ; 53(8): 1450-6, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22765836

RESUMEN

PURPOSE: The ß3 subunit of the γ-aminobutyric acid type A receptors (GABA(A) -Rs) is an essential component of GABA(A) -Rs in fetal, perinatal, and adult mammalian brain. Various transcripts of the ß3 subunit gene (GABRB3) produce various proteins with different N-termini. Rare variants in this N-terminus (exon 1A and exon 2) of GABRB3 protein segregate in affected family members of two multigeneration-multiplex families with remitting childhood absence epilepsy (rCAE), suggesting GABRB3 is a major Mendelian epilepsy gene for rare families with CAE. Therefore, the N-terminus of GABRB3 could be important for GABRB3 regulation in development, and its alteration could produce rCAE. Herein we determine if single nucleotide polymorphisms (SNPs) within the 1,148-bp region upstream from exon 1A influence the expression of GABRB3. METHODS: We studied luciferase reporter expression for promoter activity, 1,148-bp upstream from exon 1A, using human embryonic kidney 293 cells. We generated constructs of the promoter region and compared different SNP haplotypes in 48 patients with rCAE. Next, we compared frequencies of rs20317, located in the core promoter region, and rs4906902, located in the enhancer region between 48 patients with rCAE and >500 healthy controls matched for ethnicity and ancestral origin. KEY FINDINGS: Highest luciferase expression occurred 230-bp upstream of exon 1A. The construct that excluded this region lost luciferase activity. Therefore, this region contains the core promoter of exon 1A. Allele C but not allele G (rs20317) significantly increased luciferase expression activity. Allele C creates binding motifs for cMYB and EGR-3. Longer constructs overlapping this region have a binding motif for REST (RE1-silencing transcription factor), a critical epigenetic modulator for neuronal genes. REST represses expression of neuronal genes in nonneuronal tissues, resulting in reduced luciferase expression activity. Even in the suppressed condition, the longer construct enhanced luciferase expression activity of the shorter construct, which excluded the distal end containing rs4906902. However, allele frequencies of rs20317 and rs4906902 were not significantly associated with 48 rCAE patients in comparison to >500 controls matched for ethnicity and ancestral origin. SIGNIFICANCE: Common SNPs in the promoter region increase luciferase expression activity. An epigenetic modulator, REST, specifically alters expression of GABRB3 exon 1A transcripts, suggesting epigenetic regulation by REST dominantly controls the expression of GABRB3 variant 2 transcript in early life GABA(A) signaling. Abnormal epigenetic regulation could be involved in absence seizures.


Asunto(s)
Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Receptores de GABA-A/genética , Alelos , Epilepsia Tipo Ausencia/genética , Células HEK293 , Haplotipos , Humanos , Luciferasas/metabolismo , Proteínas Represoras/genética
13.
Child Neurol Open ; 9: 2329048X221126361, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36211619

RESUMEN

Lafora disease is a rare refractory epilepsy that results in death. This report highlights two cases of lafora disease and introduces a novel mutation in the patients. A review of the pathophysiology and future therapies is reviewed.

14.
Hum Mol Genet ; 18(6): 1099-109, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19147686

RESUMEN

Mutations in EFHC1 gene have been previously reported in patients with epilepsies, including those with juvenile myoclonic epilepsy. Myoclonin1, also known as mRib72-1, is encoded by the mouse Efhc1 gene. Myoclonin1 is dominantly expressed in embryonic choroid plexus, post-natal ependymal cilia, tracheal cilia and sperm flagella. In this study, we generated viable Efhc1-deficient mice. Most of the mice were normal in outward appearance, and both sexes were found to be fertile. However, the ventricles of the brains were significantly enlarged in the null mutants, but not in the heterozygotes. Although the ciliary structure was found intact, the ciliary beating frequency was significantly reduced in null mutants. In adult stages, both the heterozygous and null mutants developed frequent spontaneous myoclonus. Furthermore, the threshold of seizures induced by pentylenetetrazol was significantly reduced in both heterozygous and null mutants. These observations seem to further suggest that decrease or loss of function of myoclonin1 may be the molecular basis for epilepsies caused by EFHC1 mutations.


Asunto(s)
Proteínas de Unión al Calcio/deficiencia , Predisposición Genética a la Enfermedad , Mioclonía/complicaciones , Mioclonía/genética , Convulsiones/complicaciones , Convulsiones/genética , Animales , Proteínas de Unión al Calcio/metabolismo , Ventrículos Cerebrales/metabolismo , Ventrículos Cerebrales/patología , Cilios/ultraestructura , Convulsivantes , Giro Dentado/metabolismo , Giro Dentado/patología , Heterocigoto , Ratones , Mutación/genética , Mioclonía/patología , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Convulsiones/patología , Ácidos Siálicos/metabolismo , Ácido gamma-Aminobutírico/metabolismo
15.
Am J Hum Genet ; 82(6): 1249-61, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18514161

RESUMEN

Childhood absence epilepsy (CAE) accounts for 10% to 12% of epilepsy in children under 16 years of age. We screened for mutations in the GABA(A) receptor (GABAR) beta 3 subunit gene (GABRB3) in 48 probands and families with remitting CAE. We found that four out of 48 families (8%) had mutations in GABRB3. One heterozygous missense mutation (P11S) in exon 1a segregated with four CAE-affected persons in one multiplex, two-generation Mexican family. P11S was also found in a singleton from Mexico. Another heterozygous missense mutation (S15F) was present in a singleton from Honduras. An exon 2 heterozygous missense mutation (G32R) was present in two CAE-affected persons and two persons affected with EEG-recorded spike and/or sharp wave in a two-generation Honduran family. All mutations were absent in 630 controls. We studied functions and possible pathogenicity by expressing mutations in HeLa cells with the use of Western blots and an in vitro translation and translocation system. Expression levels did not differ from those of controls, but all mutations showed hyperglycosylation in the in vitro translation and translocation system with canine microsomes. Functional analysis of human GABA(A) receptors (alpha 1 beta 3-v2 gamma 2S, alpha 1 beta 3-v2[P11S]gamma 2S, alpha 1 beta 3-v2[S15F]gamma 2S, and alpha 1 beta 3-v2[G32R]gamma 2S) transiently expressed in HEK293T cells with the use of rapid agonist application showed that each amino acid transversion in the beta 3-v2 subunit (P11S, S15F, and G32R) reduced GABA-evoked current density from whole cells. Mutated beta 3 subunit protein could thus cause absence seizures through a gain in glycosylation of mutated exon 1a and exon 2, affecting maturation and trafficking of GABAR from endoplasmic reticulum to cell surface and resulting in reduced GABA-evoked currents.


Asunto(s)
Epilepsia Tipo Ausencia/genética , Epilepsia Tipo Ausencia/fisiopatología , Mutación Missense , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Secuencia de Aminoácidos , Línea Celular , Niño , Preescolar , Análisis Mutacional de ADN , ADN Complementario/genética , Electroencefalografía , Femenino , Ligamiento Genético , Genotipo , Glicosilación , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Fenotipo , Receptores de GABA-A/química , Homología de Secuencia de Aminoácido , Transfección
16.
Epilepsia ; 52(6): 1177-85, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21275975

RESUMEN

PURPOSE: Epilepsy is highly prevalent in developing countries like Honduras, with few studies evaluating this finding. This population-based study evaluated the impact of an 8-year public health and educational intervention program in reducing symptomatic epilepsies in rural Salamá, Honduras. METHODS: We used the capture and recapture method including review of charts, previous databases, key informants from the community, and a second house-to-house survey for epilepsy. Epilepsy incidence and prevalence day after the interventions was May 5, 2005. Residents with active epilepsy with onset after May 1997 were offered neurologic evaluation, electroencephalography, and brain tomography. New data over 8 years were compared to preintervention data from the initial baseline 1997 study utilizing prevalence ratios and confidence intervals. Other calculations utilized chi square or Fisher's exact tests. KEY FINDINGS: Thirty-three of 36 patients with onset of active epilepsy after 1997 accepted evaluations to determine etiology. Symptomatic etiology was found in 58.3%. Neurocysticercosis (NCC) was again the most frequent cause (13.9%), followed by perinatal insults (11.1%). Epilepsy secondary to NCC was significantly reduced from 36.9% in 1997 (p = 0.02). The incidence (35.7/100,000) and prevalence (11.8/1,000) of active epilepsy were not significantly reduced when compared to the incidence (92.7/100,000) and prevalence (15.4/1,000) of active epilepsy in 1997. SIGNIFICANCE: Our cohort appears to indicate that health and educational community interventions can reduce preventable epilepsy from NCC in a hyperendemic population in a low-resource, developing country. Plans are underway for the Honduran Government to institute this rural model countrywide.


Asunto(s)
Epilepsia/epidemiología , Epilepsia/prevención & control , Neurocisticercosis/epidemiología , Neurocisticercosis/prevención & control , Vigilancia de la Población , Características de la Residencia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Estudios de Cohortes , Recolección de Datos/métodos , Epilepsia/etiología , Femenino , Honduras/epidemiología , Humanos , Lactante , Masculino , Persona de Mediana Edad , Neurocisticercosis/complicaciones , Vigilancia de la Población/métodos , Porcinos , Adulto Joven
17.
Epilepsia ; 51 Suppl 5: 1-5, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21208201

RESUMEN

In 1969, H.H. Jasper, A.A. Ward, and A. Pope and the Public Health Service Advisory Committee on the Epilepsies of the National Institutes of Health (NIH) published the first edition on Basic Mechanisms of the Epilepsies (BME). Since then, basic and clinical researchers in epilepsy have gathered together each decade to assess where epilepsy research has been, what it has accomplished, and where it should go. In 1999, the third edition of BME was named in honor of H.H. Jasper. Projected for publication in 2011, the fourth edition of Jasper's BME will (1) synthesize the role of interactions between neurons, synapses, and glia in the initiation, spread, and arrest of seizures; (2) examine the molecular, cellular, and network plasticity mechanisms that subserve excitability, seizure susceptibility, and ultimately epileptogenesis; (3) provide a framework for expanding the genome of rare mendelian epilepsies and understanding the complex heredity responsible for common epilepsies; (4) explore cellular mechanisms of the two main groups of presently known Mendelian epilepsy genes, namely ion channelopathies and developmental epilepsy genes; and (5) for the first time, describe the current efforts to translate the discoveries in epilepsy disease mechanisms into molecular and cellular therapeutic strategies in order to repair and cure the epilepsies. For an expanded treatment of this topic see Jasper's Basic Mechanisms of the Epilepsies, Fourth Edition (Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV, eds) published by Oxford University Press (available on the National Library of Medicine Bookshelf [NCBI] at http://www.ncbi.nlm.nih.gov/books).


Asunto(s)
Epilepsia , Animales , Epilepsia/etiología , Epilepsia/patología , Epilepsia/terapia , Humanos
18.
Proc Natl Acad Sci U S A ; 104(49): 19262-6, 2007 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-18040046

RESUMEN

Lafora disease is a progressive myoclonus epilepsy with onset typically in the second decade of life and death within 10 years. Lafora bodies, deposits of abnormally branched, insoluble glycogen-like polymers, form in neurons, muscle, liver, and other tissues. Approximately half of the cases of Lafora disease result from mutations in the EPM2A gene, which encodes laforin, a member of the dual-specificity protein phosphatase family that additionally contains a glycogen binding domain. The molecular basis for the formation of Lafora bodies is completely unknown. Glycogen, a branched polymer of glucose, contains a small amount of covalently linked phosphate whose origin and function are obscure. We report here that recombinant laforin is able to release this phosphate in vitro, in a time-dependent reaction with an apparent K(m) for glycogen of 4.5 mg/ml. Mutations of laforin that disable the glycogen binding domain also eliminate its ability to dephosphorylate glycogen. We have also analyzed glycogen from a mouse model of Lafora disease, Epm2a(-/-) mice, which develop Lafora bodies in several tissues. Glycogen isolated from these mice had a 40% increase in the covalent phosphate content in liver and a 4-fold elevation in muscle. We propose that excessive phosphorylation of glycogen leads to aberrant branching and Lafora body formation. This study provides a molecular link between an observed biochemical property of laforin and the phenotype of a mouse model of Lafora disease. The results also have important implications for glycogen metabolism generally.


Asunto(s)
Fosfatasas de Especificidad Dual/deficiencia , Glucógeno/metabolismo , Enfermedad de Lafora/enzimología , Animales , Modelos Animales de Enfermedad , Fosfatasas de Especificidad Dual/genética , Glucógeno Sintasa/análisis , Glucógeno Sintasa/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación , Fosforilación , Proteínas Tirosina Fosfatasas no Receptoras , Conejos , Proteínas Recombinantes/farmacología
19.
Epilepsia ; 50(5): 1184-90, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18823326

RESUMEN

PURPOSE: Juvenile myoclonic epilepsy (JME) accounts for 3 to 12% of all epilepsies. In 2004, we identified a mutation-harboring Mendelian gene that encodes a protein with one EF-hand motif (EFHC1) in chromosome 6p12. We observed one doubly heterozygous and three heterozygous missense mutations in EFHC1 segregating as an autosomal dominant gene with 21 affected members of six Hispanic JME families from California and Mexico. In 2006, similar and three novel missense mutations were reported in sporadic and familial Caucasian JME from Italy and Austria. In this study, we asked if coding single nucleotide polymorphisms (SNPs) of EFHC1 also contribute as susceptibility alleles to JME with complex genetics. METHODS: We screened using denaturing high-performance liquid chromatography (DHPLC) and then directly sequenced the 11 exons of EFHC1 in 130 unrelated JME probands, their 352 family members, and seven exons of EFHC1 in 400-614 ethnically matched controls. We carried out case-control association studies between 124 unrelated Hispanic JME probands and 552-614 ethnically matched controls using four SNPs, rs3804506, rs3804505, rs1266787, and rs17851770. We also performed family-based association on SNPs rs3804506 and rs3804505 in 84 complete JME families using the Family-Based Association Test (FBAT) program. RESULTS: We found no statistically significant differences between JME probands and controls in case-control association and no genetic transmission disequilibria in family-based association for the tested SNPs. In addition, we identified four new DNA variants in the coding region of EFHC1. CONCLUSION: The four coding SNPs, rs3804506, rs3804505, rs1266787, and rs17851770, of EFHC1 may not be susceptibility alleles for JME.


Asunto(s)
Proteínas de Unión al Calcio/genética , Epilepsia Mioclónica Juvenil/genética , Polimorfismo de Nucleótido Simple/genética , Estudios de Casos y Controles , Análisis Mutacional de ADN/métodos , Exones/genética , Salud de la Familia , Femenino , Humanos , Masculino , Datos de Secuencia Molecular
20.
Front Cell Neurosci ; 13: 433, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31611775

RESUMEN

Juvenile myoclonic epilepsy (JME), a lifelong disorder that starts during adolescence, is the most common of genetic generalized epilepsy syndromes. JME is characterized by awakening myoclonic jerks and myoclonic-tonic-clonic (m-t-c) grand mal convulsions. Unfortunately, one third of JME patients have drug refractory m-t-c convulsions and these recur in 70-80% who attempt to stop antiepileptic drugs (AEDs). Behavioral studies documented impulsivity, but also impairment of executive functions relying on organization and feedback, which points to prefrontal lobe dysfunction. Quantitative voxel-based morphometry (VBM) revealed abnormalities of gray matter (GM) volumes in cortical (frontal and parietal) and subcortical structures (thalamus, putamen, and hippocampus). Proton magnetic resonance spectroscopy (MRS) found evidence of dysfunction of thalamic neurons. White matter (WM) integrity was disrupted in corpus callosum and frontal WM tracts. Magnetic resonance imaging (MRI) further unveiled anomalies in both GM and WM structures that were already present at the time of seizure onset. Aberrant growth trajectories of brain development occurred during the first 2 years of JME diagnosis. Because of genetic origin, disease causing variants were sought, first by positional cloning, and most recently, by next generation sequencing. To date, only six genes harboring pathogenic variants (GABRA1, GABRD, EFHC1, BRD2, CASR, and ICK) with Mendelian and complex inheritance and covering a limited proportion of the world population, are considered as major susceptibility alleles for JME. Evidence on the cellular role, developmental and cell-type expression profiles of these six diverse JME genes, point to their pathogenic variants driving the first steps of brain development when cell division, expansion, axial, and tangential migration of progenitor cells (including interneuron cortical progenitors) sculpture subtle alterations in brain networks and microcircuits during development. These alterations may explain "microdysgenesis" neuropathology, impulsivity, executive dysfunctions, EEG polyspike waves, and awakening m-t-c convulsions observed in JME patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA