Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 30(14): 1337-1348, 2021 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-33961044

RESUMEN

Syntaxin-binding protein 1 (STXBP1; also called MUNC18-1), encoded by STXBP1, is an essential component of the molecular machinery that controls synaptic vesicle docking and fusion. De novo pathogenic variants of STXBP1 cause a complex set of neurological disturbances, namely STXBP1 encephalopathy (STXBP1-E) that includes epilepsy, neurodevelopmental disorders and neurodegeneration. Several animal studies have suggested the contribution of GABAergic dysfunction in STXBP1-E pathogenesis. However, the pathophysiological changes in GABAergic neurons of these patients are still poorly understood. Here, we exclusively generated GABAergic neurons from STXBP1-E patient-derived induced pluripotent stem cells (iPSCs) by transient expression of the transcription factors ASCL1 and DLX2. We also generated CRISPR/Cas9-edited isogenic iPSC-derived GABAergic (iPSC GABA) neurons as controls. We demonstrated that the reduction in STXBP1 protein levels in patient-derived iPSC GABA neurons was slight (approximately 20%) compared to the control neurons, despite a 50% reduction in STXBP1 mRNA levels. Using a microelectrode array-based assay, we found that patient-derived iPSC GABA neurons exhibited dysfunctional maturation with reduced numbers of spontaneous spikes and bursts. These findings reinforce the idea that GABAergic dysfunction is a crucial contributor to STXBP1-E pathogenesis. Moreover, gene expression analysis revealed specific dysregulation of genes previously implicated in epilepsy, neurodevelopment and neurodegeneration in patient-derived iPSC GABA neurons, namely KCNH1, KCNH5, CNN3, RASGRF1, SEMA3A, SIAH3 and INPP5F. Thus, our study provides new insights for understanding the biological processes underlying the widespread neuropathological features of STXBP1-E.


Asunto(s)
Encefalopatías , Células Madre Pluripotentes Inducidas , Animales , Encefalopatías/genética , Encefalopatías/metabolismo , Neuronas GABAérgicas/metabolismo , Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Munc18/genética , Proteínas Munc18/metabolismo
2.
Biochem Biophys Res Commun ; 643: 169-174, 2023 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-36610382

RESUMEN

Dravet syndrome (DS) is an infantile-onset epileptic encephalopathy. More than 80% of DS patients have a heterozygous mutation in SCN1A, which encodes a subunit of the voltage-gated sodium channel, Nav1.1, in neurons. The roles played by astrocytes, the most abundant glial cell type in the brain, have been investigated in the pathogenesis of epilepsy; however, the specific involvement of astrocytes in DS has not been clarified. In this study, we evaluated Ca2+ signaling in astrocytes using genetically modified mice that have a loss-of-function mutation in Scn1a. We found that the slope of spontaneous Ca2+ spiking was increased without a change in amplitude in Scn1a+/- astrocytes. In addition, ATP-induced transient Ca2+ influx and the slope of Ca2+ spiking were also increased in Scn1a+/- astrocytes. These data indicate that perturbed Ca2+ dynamics in astrocytes may be involved in the pathogenesis of DS.


Asunto(s)
Epilepsias Mioclónicas , Epilepsia , Animales , Ratones , Astrocitos/metabolismo , Epilepsias Mioclónicas/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Neuronas/metabolismo , Señalización del Calcio
3.
Epilepsy Behav ; 131(Pt B): 108043, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34053869

RESUMEN

Dravet syndrome is a well-established electro-clinical condition first described in 1978. A main genetic cause was identified with the discovery of a loss-of-function SCN1A variant in 2001. Mechanisms underlying the phenotypic variations have subsequently been a main topic of research. Various genetic modifiers of clinical severities have been elucidated through many rigorous studies on genotype-phenotype correlations and the recent advances in next generation sequencing technology. Furthermore, a deeper understanding of the regulation of gene expression and remarkable progress on genome-editing technology using the CRISPR-Cas9 system provide significant opportunities to overcome hurdles of gene therapy, such as enhancing NaV1.1 expression. This article reviews the current understanding of genetic pathology and the status of research toward the development of gene therapy for Dravet syndrome. This article is part of the Special Issue "Severe Infantile Epilepsies".


Asunto(s)
Epilepsias Mioclónicas , Epilepsia , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/terapia , Epilepsia/genética , Síndromes Epilépticos , Terapia Genética , Humanos , Canal de Sodio Activado por Voltaje NAV1.1/genética , Espasmos Infantiles
4.
Pediatr Int ; 64(1): e14949, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34390093

RESUMEN

BACKGROUND: Invasive candidiasis (IC) is a leading cause of morbidity and mortality in preterm infants. The objective of this study was to determine the prevalence of IC infection in newborns in the neonatal intensive care unit (NICU) of a tertiary hospital in Japan, and to identify specific predisposing factors for IC. METHODS: We retrospectively collected data on demographics, clinical characteristics, and outcomes of infants with IC, who were discharged from a tertiary NICU in Japan between January 2009 and December 2020. We compared predisposing factors associated with the occurrence of early-onset IC (EOIC < 72 h) and late-onset IC (LOIC ≥ 72 h) with those of early-onset and late-onset bacterial sepsis. RESULTS: Between January 2009 and December 2020, 3,549 infants were admitted to the NICU, including 344 extremely-low birthweight (ELBW) infants. Eleven infants (including nine ELBW infants) had IC (incidence 0.31%), and the mortality rate of IC was 0%. Four (36%) infants had EOIC and seven (64%) had LOIC. All those with EOIC presented with skin lesions and 86% with LOIC had thrombocytopenia. Maternal vaginal Candida colonization was a more specific predisposing factor for EOIC, while gestational age <26 weeks, broad-spectrum antibiotic use, prior bacterial infection, prior gastrointestinal (GI) surgery, and GI diseases were more specific predisposing factors for LOIC. CONCLUSIONS: The findings suggest that maternal vaginal Candida colonization and skin lesions in ELBW infants may contribute to early recognition of EOIC. LOIC should be suspected if ELBW infants with several predisposing factors of LOIC have thrombocytopenia.


Asunto(s)
Candidiasis Invasiva , Trombocitopenia , Candidiasis , Candidiasis Invasiva/diagnóstico , Candidiasis Invasiva/epidemiología , Candidiasis Invasiva/microbiología , Femenino , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Unidades de Cuidado Intensivo Neonatal , Estudios Retrospectivos
5.
Hum Mutat ; 42(11): 1422-1428, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34405929

RESUMEN

Isolated complex I deficiency is the most common cause of pediatric mitochondrial disease. Exome sequencing (ES) has revealed many complex I causative genes. However, there are limitations associated with identifying causative genes by ES analysis. In this study, we performed multiomics analysis to reveal the causal variants. We here report two cases with mitochondrial complex I deficiency. In both cases, ES identified a novel c.580G>A (p.Glu194Lys) variant in NDUFV2. One case additionally harbored c.427C>T (p.Arg143*), but no other variants were observed in the other case. RNA sequencing showed aberrant exon splicing of NDUFV2 in the unsolved case. Genome sequencing revealed a novel heterozygous deletion in NDUFV2, which included one exon and resulted in exon skipping. Detailed examination of the breakpoint revealed that an Alu insertion-mediated rearrangement caused the deletion. Our report reveals that combined use of transcriptome sequencing and GS was effective for diagnosing cases that were unresolved by ES.


Asunto(s)
Elementos Alu , Complejo I de Transporte de Electrón/deficiencia , Eliminación de Gen , Genoma Humano , Mutación INDEL , Enfermedades Mitocondriales/genética , NADH Deshidrogenasa/genética , Análisis de Secuencia de ARN/métodos , Complejo I de Transporte de Electrón/genética , Femenino , Humanos , Lactante , Masculino , Enfermedades Mitocondriales/diagnóstico , Linaje
6.
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
7.
J Hum Genet ; 66(6): 569-578, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33262389

RESUMEN

Missense and truncating variants in protocadherin 19 (PCDH19) cause PCDH19-related epilepsy. In this study, we aimed to investigate variations in distributional characteristics and the clinical implications of variant type in PCDH19-related epilepsy. We comprehensively collected PCDH19 missense and truncating variants from the literature and by sequencing six exons and intron-exon boundaries of PCDH19 in our cohort. We investigated the distribution of each type of variant using the cumulative distribution function and tested for associations between variant types and phenotypes. The distribution of missense variants in patients was clearly different from that of healthy individuals and was uniform throughout the extracellular cadherin (EC) domain, which consisted of six highly conserved domains. Truncating variants showed two types of distributions: (1) located from EC domain 1 to EC domain 4, and (2) located from EC domain 5 to the cytoplasmic domain. Furthermore, we also found that later onset seizures and milder intellectual disability occurred in patients with truncating variants located from EC domain 5 to the cytoplasmic domain compared with those of patients with other variants. Our findings provide the first evidence of two types of truncating variants in the PCDH19 gene with regard to distribution and the resulting clinical phenotype.


Asunto(s)
Cadherinas/genética , Epilepsia/genética , Estudios de Asociación Genética , Convulsiones/genética , Edad de Inicio , Preescolar , Estudios de Cohortes , Epilepsia/patología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Lactante , Masculino , Mutación Missense/genética , Fenotipo , Protocadherinas , Convulsiones/patología
8.
Mol Cell Neurosci ; 108: 103535, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32758699

RESUMEN

Epilepsy is among the most common neurological disorders, affecting approximately 50 million people worldwide. Importantly, epilepsy is genetically and etiologically heterogenous, but several epilepsy types exhibit similar clinical presentations. Epilepsy-associated genes are being identified. However, the molecular pathomechanisms remain largely unknown. Approximately one-third of epilepsy is refractory to multiple conventional anti-epileptic drugs (AEDs). Induced pluripotent stem cells (iPSCs) provide an excellent tool to study the pathomechanisms underlying epilepsy and to develop novel treatments. Indeed, disease-specific iPSCs have been established for several genetic epilepsies. In particular, the molecular mechanisms underlying certain developmental and epileptic encephalopathies, such as Dravet syndrome, have been revealed. Modeling epilepsy with iPSCs enables new drug development based on the elucidated pathomechanisms. This can also be used to evaluate conventional AEDs and drug repurposing. Furthermore, transplanting neuronal cells derived from iPSCs into the brain has great potential to treat refractory epilepsies. Recent advances in iPSC technology have enabled the generation of neuronal organoids, or "mini brains." These organoids demonstrate electrophysiological activities similar to those of the brain and have the potential for extensive epilepsy research opportunities. Thus, the application of iPSCs in epilepsy provides insight into novel treatments based on the molecular pathomechanisms of epilepsy. In this review, we comprehensively discuss the studies conducted on iPSCs established for genetic epilepsy or epilepsies without major structural dysmorphic features.


Asunto(s)
Anticonvulsivantes/farmacología , Epilepsia/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Evaluación Preclínica de Medicamentos/métodos , Epilepsia/genética , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos
9.
Int J Mol Sci ; 22(8)2021 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-33924373

RESUMEN

A common pathological hallmark of several neurodegenerative diseases, including amyotrophic lateral sclerosis, is cytoplasmic mislocalization and aggregation of nuclear RNA-binding protein TDP-43. Perry disease, which displays inherited atypical parkinsonism, is a type of TDP-43 proteinopathy. The causative gene DCTN1 encodes the largest subunit of the dynactin complex. Dynactin associates with the microtubule-based motor cytoplasmic dynein and is required for dynein-mediated long-distance retrograde transport. Perry disease-linked missense mutations (e.g., p.G71A) reside within the CAP-Gly domain and impair the microtubule-binding abilities of DCTN1. However, molecular mechanisms by which such DCTN1 mutations cause TDP-43 proteinopathy remain unclear. We found that DCTN1 bound to TDP-43. Biochemical analysis using a panel of truncated mutants revealed that the DCTN1 CAP-Gly-basic supradomain, dynactin domain, and C-terminal region interacted with TDP-43, preferentially through its C-terminal region. Remarkably, the p.G71A mutation affected the TDP-43-interacting ability of DCTN1. Overexpression of DCTN1G71A, the dynactin-domain fragment, or C-terminal fragment, but not the CAP-Gly-basic fragment, induced cytoplasmic mislocalization and aggregation of TDP-43, suggesting functional modularity among TDP-43-interacting domains of DCTN1. We thus identified DCTN1 as a new player in TDP-43 cytoplasmic-nuclear transport, and showed that dysregulation of DCTN1-TDP-43 interactions triggers mislocalization and aggregation of TDP-43, thus providing insights into the pathological mechanisms of Perry disease and other TDP-43 proteinopathies.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejo Dinactina/metabolismo , Agregado de Proteínas , Secuencia de Aminoácidos , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Complejo Dinactina/química , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Neuronas/metabolismo , Señales de Localización Nuclear/metabolismo , Mutación Puntual/genética , Unión Proteica , Fracciones Subcelulares/metabolismo
10.
J Hum Genet ; 64(11): 1097-1106, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31471553

RESUMEN

Early-onset developmental and epileptic encephalopathy (DEE) is a group of devastating disorders that appear during the neonatal and infantile periods. Despite great progress in the discovery of genes leading to early-onset DEE, many cases with unexplained etiology remain. Furthermore, to date, the association of copy number variations (CNVs) with early-onset DEE has seldom been addressed. Here, we investigated the contribution of CNVs to epilepsy in a cohort of Japanese children with a variety of early-onset DEEs. Single nucleotide polymorphism (SNP) array analysis was performed for 83 cases that were previously negative for pathogenic single nucleotide variants (SNVs) in 109 genes known or suspected to cause epileptic seizures. Rare CNVs were detected in a total of 12 cases (14.4%), of which three cases (3.6%) involved clearly pathogenic CNVs and nine cases (10.8%) were CNVs of uncertain significance. The three pathogenic CNVs included two de novo heterozygous deletions involving known epileptic encephalopathy genes, such as GABRG2 and PCDH19, and one maternally inherited duplication encompassing MECP2. Our findings indicate rare CNVs are also relevant for the diagnosis of early-onset DEEs, highlighting the importance of not relying only on the investigation of SNVs/small indels at the risk of missing large deletions and duplications.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Discapacidades del Desarrollo/genética , Convulsiones/genética , Espasmos Infantiles/genética , Cadherinas/genética , Niño , Discapacidades del Desarrollo/fisiopatología , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Polimorfismo de Nucleótido Simple/genética , Protocadherinas , Receptores de GABA-A/genética , Convulsiones/fisiopatología , Espasmos Infantiles/fisiopatología
11.
J Hum Genet ; 64(8): 741-755, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31076647

RESUMEN

A newborn screening program for Pompe disease using dried blood spots (DBSs) was initiated in Japan. Here, we summarized this screening program and described the results of the GAA gene analysis. From April 2013 to November 2016, 103,204 newborns were screened; 71 had low acid alpha-glucosidase (AαGlu) activity. GAA sequencing showed that 32 (45.1%) and 37 (52.1%) of these newborns were homozygous and heterozygous for pseudodeficiency alleles c.[1726G>A; 2965G>A], respectively. Moreover, 24 of 32 newborns with homozygous c.[1726G>A; 2965G>A] alleles had no mutations, and the other eight had one mutation each. Thirty-five of 37 newborns with heterozygous c.[1726G>A; 2965G>A] alleles had one mutation, and the other two had two mutations each. Only one newborn who had two mutations did not harbor c.[1726G>A; 2965G>A] alleles. Thus, it was difficult to distinguish newborns with c.[1726G>A; 2965G>A] alleles from newborns with pre-symptomatic Pompe disease using AαGlu assays in DBSs or fibroblasts; GAA gene sequencing was necessary. Seventy-one newborns had 50 variants, including 21 mutations or predictably pathogenic variants, and 29 polymorphisms or predictably non-pathogenic variants. Four of 21 mutations or predictably pathogenic variants and four of 29 polymorphisms or predictably non-pathogenic variants were novel. No infantile-onset Pompe disease was detected, and three newborns were diagnosed with potential late-onset Pompe disease. In the literature, 156 variants have been reported for 296 patients from 277 families in 41 articles from Japan, Korea, Taiwan, and China. Our results provide insights into GAA gene mutation profiles and the relationship between GAA and Pompe disease in Asian populations.


Asunto(s)
Pueblo Asiatico/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/epidemiología , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Mutación , alfa-Glucosidasas/genética , Edad de Inicio , Alelos , Sustitución de Aminoácidos , Análisis Mutacional de ADN , Activación Enzimática , Frecuencia de los Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Genotipo , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Recién Nacido , Japón/epidemiología , Tamizaje Neonatal , Vigilancia de la Población , Flujo de Trabajo , alfa-Glucosidasas/metabolismo
12.
Epilepsia ; 60(9): 1870-1880, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31418850

RESUMEN

OBJECTIVE: Pathogenic variants of KCNQ2, which encode a potassium channel subunit, cause either benign (familial) neonatal epilepsy-B(F)NE)-or KCNQ2 encephalopathy (KCNQ2 DEE). We examined the characteristics of KCNQ2 variants. METHODS: KCNQ2 pathogenic variants were collected from in-house data and two large disease databases with their clinical phenotypes. Nonpathogenic KCNQ2 variants were collected from the Genome Aggregation Database (gnomAD). Pathogenicity of all variants was reevaluated with clinical information to exclude irrelevant variants. The cumulative distribution plots of B(F)NE, KCNQ2 DEE, and gnomAD KCNQ2 variants were compared. Several algorithms predicting genetic variant pathogenicity were evaluated. RESULTS: A total of 259 individuals or pedigrees with 216 different pathogenic KCNQ2 variants and 2967 individuals with 247 different nonpathogenic variants were deemed eligible for the study. Compared to the distribution of nonpathogenic variants, B(F)NE and KCNQ2 DEE missense variants occurred in five and three specific KCNQ2 regions, respectively. Comparison between B(F)NE and KCNQ2 DEE sets showed that B(F)NE missense variants frequently localized to the intracellular domain between S2 and S3, whereas those of KCNQ2 DEE were more frequent in S6, and its adjacent pore domain, as well as in the intracellular domain between S6 and helix A. The scores of Protein Variation Effect Analyzer (PROVEAN) and Percent Accepted Mutation (PAM) 30 prediction algorithms were associated with phenotypes of the variant loci. SIGNIFICANCE: Missense variants in the intracellular domain between S2 and S3 are likely to cause B(F)NE, whereas those in S6 and its adjacent regions are more likely to cause KCNQ2 DEE. With such regional specificities of variants, PAM30 is a helpful tool to examine the possibility that a novel KCNQ2 variant is a B(F)NE or KCNQ2 DEE variant in genetic analysis.


Asunto(s)
Encefalopatías/genética , Epilepsia Benigna Neonatal/genética , Canal de Potasio KCNQ2/genética , Espasmos Infantiles/genética , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Mutación Missense , Linaje , Fenotipo
13.
Am J Med Genet A ; 176(3): 657-662, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29341473

RESUMEN

Somatic mosaicism in single nucleotide variants of SCN1A is known to occur in a subset of parents of children with Dravet syndrome (DS). Here, we report recurrent somatic mosaic microdeletions involving SCN1A in children diagnosed with DS. Through the evaluation of 237 affected individuals with DS who did not show SCN1A or PCHD19 mutations in prior sequencing analyzes, we identified two children with mosaic microdeletions covering the entire SCN1A region. The allele frequency of the mosaic deletions estimated by multiplex ligation-dependent probe amplification and array comparative genomic hybridization was 25-40%, which was comparable to the mosaic ratio in lymphocytes and buccal mucosa cells observed by fluorescence in situ hybridization analysis. The minimal prevalence of SCN1A mosaic deletion is estimated to be 0.9% (95% confidence level: 0.11-3.11%) of DS with negative for SCN1A and PCDH19 mutations. This study reinforces the importance of somatic mosaicism caused by copy number variations in disease-causing genes, and provides an alternative spectrum of SCN1A mutations causative of DS. Somatic deletions in SCN1A should be considered in cases with DS when standard screenings for SCN1A mutations are apparently negative for mutations.


Asunto(s)
Epilepsias Mioclónicas/diagnóstico , Epilepsias Mioclónicas/genética , Mosaicismo , Canal de Sodio Activado por Voltaje NAV1.1/genética , Eliminación de Secuencia , Hibridación Genómica Comparativa , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Hibridación Fluorescente in Situ , Linaje , Fenotipo , Análisis de Secuencia de ADN
14.
J Med Genet ; 54(3): 202-211, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27789573

RESUMEN

BACKGROUND: Early myoclonic encephalopathy (EME), a disease with a devastating prognosis, is characterised by neonatal onset of seizures and massive myoclonus accompanied by a continuous suppression-burst EEG pattern. Three genes are associated with EMEs that have metabolic features. Here, we report a pathogenic mutation of an ion channel as a cause of EME for the first time. METHODS: Sequencing was performed for 214 patients with epileptic seizures using a gene panel with 109 genes that are known or suspected to cause epileptic seizures. Functional assessments were demonstrated by using electrophysiological experiments and immunostaining for mutant γ-aminobutyric acid-A (GABAA) receptor subunits in HEK293T cells. RESULTS: We discovered a de novo heterozygous missense mutation (c.859A>C [p.Thr287Pro]) in the GABRB2-encoded ß2 subunit of the GABAA receptor in an infant with EME. No GABRB2 mutations were found in three other EME cases or in 166 patients with infantile spasms. GABAA receptors bearing the mutant ß2 subunit were poorly trafficked to the cell membrane and prevented γ2 subunits from trafficking to the cell surface. The peak amplitudes of currents from GABAA receptors containing only mutant ß2 subunits were smaller than that of those from receptors containing only wild-type ß2 subunits. The decrease in peak current amplitude (96.4% reduction) associated with the mutant GABAA receptor was greater than expected, based on the degree to which cell surface expression was reduced (66% reduction). CONCLUSION: This mutation has complex functional effects on GABAA receptors, including reduction of cell surface expression and attenuation of channel function, which would significantly perturb GABAergic inhibition in the brain.


Asunto(s)
Síndrome de Opsoclonía-Mioclonía/genética , Receptores de GABA-A/genética , Convulsiones/genética , Espasmos Infantiles/genética , Encéfalo/diagnóstico por imagen , Encéfalo/fisiopatología , Cristalografía por Rayos X , Electroencefalografía , Células HEK293 , Humanos , Lactante , Masculino , Modelos Moleculares , Mutación Missense , Síndrome de Opsoclonía-Mioclonía/fisiopatología , Receptores de GABA-A/química , Convulsiones/fisiopatología , Espasmos Infantiles/fisiopatología
15.
Epilepsia ; 58(8): 1430-1439, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28575529

RESUMEN

OBJECTIVE: Mutations of the KCNQ2 gene, which encodes the Kv 7.2 subunit of voltage-gated M-type potassium channels, have been associated with epilepsy in the neonatal period. This developmental stage is unique in that the neurotransmitter gamma aminobutyric acid (GABA), which is inhibitory in adults, triggers excitatory action due to a reversed chloride gradient. METHODS: To examine whether KCNQ2-related neuronal hyperexcitability involves neonatally excitatory GABA, we examined 1-week-old knockin mice expressing the Kv 7.2 variant p.Tyr284Cys (Y284C). RESULTS: Brain slice electrophysiology revealed elevated CA1 hippocampal GABAergic interneuron activity with respect to presynaptic firing and postsynaptic current frequency. Blockade with the GABAA receptor antagonist bicuculline decreased ictal-like bursting in brain slices with lowered divalent ion concentration, which is consistent with GABA mediating an excitatory function that contributes to the hyperexcitability observed in mutant animals. SIGNIFICANCE: We conclude that excitatory GABA contributes to the phenotype in these animals, which raises the question of whether this special type of neurotransmission has broader importance in neonatal epilepsy than is currently recognized.


Asunto(s)
Epilepsia/genética , Epilepsia/metabolismo , Canal de Potasio KCNQ2/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Transmisión Sináptica/genética , Ácido gamma-Aminobutírico/metabolismo , Animales , Animales Recién Nacidos , Biofisica , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Modelos Animales de Enfermedad , Estimulación Eléctrica , GABAérgicos/farmacología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/genética , Interneuronas/efectos de los fármacos , Interneuronas/fisiología , Magnesio/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Placa-Clamp , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/farmacología
16.
Epilepsia ; 58(2): 282-290, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28012175

RESUMEN

OBJECTIVE: Two major classes of SCN1A variants are associated with Dravet syndrome (DS): those that result in haploinsufficiency (truncating) and those that result in an amino acid substitution (missense). The aim of this retrospective study was to describe the first large cohort of Japanese patients with SCN1A mutation-positive DS (n = 285), and investigate the relationship between variant (type and position) and clinical expression and response to treatment. METHODS: We sequenced all exons and intron-exon boundaries of SCN1A in our cohort, investigated differences in the distribution of truncating and missense variants, tested for associations between variant type and phenotype, and compared these patterns with those of cohorts with milder epilepsy and healthy individuals. RESULTS: Unlike truncation variants, missense variants are found at higher density in the S4 voltage sensor and pore loops and at lower density in the domain I-II and II-III linkers and the first three segments of domain II. Relative to healthy individuals, there is an increased frequency of truncating (but not missense) variants in the noncoding C-terminus. The rate of cognitive decline is more rapid for patients with truncation variants regardless of age at seizure onset, whereas age at onset is a predictor of the rate of cognitive decline for patients with missense variants. SIGNIFICANCE: We found significant differences in the distribution of truncating and missense variants across the SCN1A sequence among healthy individuals, patients with DS, and those with milder forms of SCN1A-variant positive epilepsy. Testing for associations with phenotype revealed that variant type can be predictive of rate of cognitive decline. Analysis of descriptive medication data suggests that in addition to conventional drug therapy in DS, bromide, clonazepam and topiramate may reduce seizure frequency.


Asunto(s)
Epilepsias Mioclónicas/genética , Haploinsuficiencia/genética , Mutación Missense/genética , Canal de Sodio Activado por Voltaje NAV1.1/genética , Adolescente , Adulto , Edad de Inicio , Anticonvulsivantes/uso terapéutico , Pueblo Asiatico/genética , Niño , Preescolar , Estudios de Cohortes , Bases de Datos Factuales/estadística & datos numéricos , Progresión de la Enfermedad , Epilepsias Mioclónicas/complicaciones , Epilepsias Mioclónicas/tratamiento farmacológico , Femenino , Humanos , Lactante , Discapacidad Intelectual/etiología , Discapacidad Intelectual/genética , Masculino , Modelos Moleculares , Adulto Joven
17.
Epilepsia ; 57(7): 1027-35, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27270488

RESUMEN

On April 21, 2015, the first SCN8A Encephalopathy Research Group convened in Washington, DC, to assess current research into clinical and pathogenic features of the disorder and prepare an agenda for future research collaborations. The group comprised clinical and basic scientists and representatives of patient advocacy groups. SCN8A encephalopathy is a rare disorder caused by de novo missense mutations of the sodium channel gene SCN8A, which encodes the neuronal sodium channel Nav 1.6. Since the initial description in 2012, approximately 140 affected individuals have been reported in publications or by SCN8A family groups. As a result, an understanding of the severe impact of SCN8A mutations is beginning to emerge. Defining a genetic epilepsy syndrome goes beyond identification of molecular etiology. Topics discussed at this meeting included (1) comparison between mutations of SCN8A and the SCN1A mutations in Dravet syndrome, (2) biophysical properties of the Nav 1.6 channel, (3) electrophysiologic effects of patient mutations on channel properties, (4) cell and animal models of SCN8A encephalopathy, (5) drug screening strategies, (6) the phenotypic spectrum of SCN8A encephalopathy, and (7) efforts to develop a bioregistry. A panel discussion of gaps in bioregistry, biobanking, and clinical outcomes data was followed by a planning session for improved integration of clinical and basic science research. Although SCN8A encephalopathy was identified only recently, there has been rapid progress in functional analysis and phenotypic classification. The focus is now shifting from identification of the underlying molecular cause to the development of strategies for drug screening and prioritized patient care.


Asunto(s)
Encefalopatías/genética , Epilepsia/etiología , Epilepsia/genética , Canal de Sodio Activado por Voltaje NAV1.6/genética , Simbiosis/genética , Animales , Anticonvulsivantes/uso terapéutico , Encefalopatías/complicaciones , Encefalopatías/tratamiento farmacológico , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Epilepsias Mioclónicas/tratamiento farmacológico , Epilepsias Mioclónicas/genética , Epilepsia/tratamiento farmacológico , Humanos , Modelos Moleculares , Canal de Sodio Activado por Voltaje NAV1.1/genética , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Fenotipo
18.
No To Hattatsu ; 48(2): 133-6, 2016 Mar.
Artículo en Japonés | MEDLINE | ID: mdl-27149744

RESUMEN

A 3-month-old girl exhibited asymmetric abnormal eye movement and unilateral dystonic posture intermittently after the first few days of life. Unilateral ocular deviation or nystagmus were the main signs of abnormal eye movements. She was suspected to have alternating hemiplegia of childhood (AHC) despite the absence of apparent alternating hemiplegic episodes. Gene analysis revealed a de-novo missense mutation (Asp801Asn) of ATP1A3. AHC is a rare neurodevelopmental disorder characterized by recurrent transient attacks of hemiplegia affecting the unilateral or bilateral side of the body; in most cases, these attacks begin in the first 6 months of life. Initial symptoms of AHC are not alternating hemiplegic episodes, but rather asymmetric abnormal eye movement, dystonic posture, or seizures. It is difficult to diagnose AHC early because no specific findings are observed in the diagnostic laboratory or neuroradiological examinations. Early diagnosis is important because flunarizine may have a protective effect on the severe motor deterioration associated with AHC. Asymmetric abnormal eye movement could be an important clinical symptom for the diagnosis of AHC in early infancy.


Asunto(s)
Hemiplejía/diagnóstico , Trastornos de la Motilidad Ocular/fisiopatología , Femenino , Humanos , Lactante , Imagen por Resonancia Magnética , Postura
19.
No To Hattatsu ; 48(5): 351-4, 2016 09.
Artículo en Japonés | MEDLINE | ID: mdl-30010281

RESUMEN

Benign familial infantile epilepsy (BFIE) is characterized by non-febrile focal seizures, which sometimes evolve to secondarily generalized seizures and are usually resolved in the second year. Proline-rich transmembrane protein 2 (PRRT2) is confirmed as the major cause of BFIE, familial paroxysmal kinesigeneic dystonia (PKD) and infantile convulsions and choreoathetosis (ICCA) syndrome. We examined a female patient with a hot spot mutation of PRRT2 gene. She had recurrent tonic seizures when she was three months old. The seizures were controlled by several kinds of anticonvulsants. Then, she had several times of focal seizures daily at nine months old. However, the seizures were stopped by small amounts of carbamazepine. Later, when she was two years old, she experienced frequent motor seizures characterized by truncal flexion and swaying the body with partially disturbed consciousness. Her father also had the same PRRT2 gene mutation and non-febrile seizures in infancy. The patient had mild to moderate mental retardation, whereas her father was mentally normal. Therefore, the patient revealed a quiet different phenotype from that of her father as a carrier of the same PRRT2 gene mutation. We speculate that the PRRT2 mutation had caused the BFIE-like seizures both in the patient and her father, whereas other unknown genetic factors specific for the patient might be associated with the atypical seizures observed only in her.


Asunto(s)
Epilepsia/genética , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Convulsiones/genética , Secuencia de Bases , Preescolar , Electroencefalografía , Epilepsia/fisiopatología , Femenino , Humanos , Convulsiones/fisiopatología
20.
Biochem Biophys Res Commun ; 456(1): 275-81, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25436429

RESUMEN

p230/golgin-245 is a trans-Golgi coiled-coil protein that is known to participate in regulatory transport from the trans-Golgi network (TGN) to the cell surface. We investigated the role of p230 and its interacting protein, microtubule actin crosslinking protein 1 (MACF1), in amino acid starvation-induced membrane transport. p230 or MACF1 knock-down (KD) cells failed to increase the autophagic flow rate and the number of microtubule-associated protein 1 light chain 3 (LC3)-positive puncta under starvation conditions. Loss of p230 or MACF1 impaired mAtg9 recruitment to peripheral phagophores from the TGN, which was observed in the early step of autophagosome formation. Overexpression of the p230-binding domain of MACF1 resulted in the inhibition of mAtg9 trafficking in starvation conditions as in p230-KD or MACF1-KD cells. These results indicate that p230 and MACF1 cooperatively play an important role in the formation of phagophore through starvation-induced transport of mAtg9-containing membranes from the TGN. In addition, p230 itself was detected in autophagosomes/autolysosome with p62 or LC3 during autophagosome biogenesis. Thus, p230 is an important molecule in phagophore formation, although it remains unclear whether p230 has any role in late steps of autophagy.


Asunto(s)
Autoantígenos/metabolismo , Autofagia , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Actinas/metabolismo , Transporte Biológico , Membrana Celular/metabolismo , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Microtúbulos/metabolismo , Fagosomas/metabolismo , Plásmidos/metabolismo , Transporte de Proteínas , ARN Interferente Pequeño/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA