Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Ann Neurol ; 89(3): 573-586, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33325057

RESUMEN

OBJECTIVE: We aimed to characterize the phenotypic spectrum and functional consequences associated with variants in the gene GABRB2, coding for the γ-aminobutyric acid type A (GABAA ) receptor subunit ß2. METHODS: We recruited and systematically evaluated 25 individuals with variants in GABRB2, 17 of whom are newly described and 8 previously reported with additional clinical data. Functional analysis was performed using a Xenopus laevis oocyte model system. RESULTS: Our cohort of 25 individuals from 22 families with variants in GABRB2 demonstrated a range of epilepsy phenotypes from genetic generalized epilepsy to developmental and epileptic encephalopathy. Fifty-eight percent of individuals had pharmacoresistant epilepsy; response to medications targeting the GABAergic pathway was inconsistent. Developmental disability (present in 84%) ranged from mild intellectual disability to severe global disability; movement disorders (present in 44%) included choreoathetosis, dystonia, and ataxia. Disease-associated variants cluster in the extracellular N-terminus and transmembrane domains 1-3, with more severe phenotypes seen in association with variants in transmembrane domains 1 and 2 and the allosteric binding site between transmembrane domains 2 and 3. Functional analysis of 4 variants in transmembrane domains 1 or 2 (p.Ile246Thr, p.Pro252Leu, p.Ile288Ser, p.Val282Ala) revealed strongly reduced amplitudes of GABA-evoked anionic currents. INTERPRETATION: GABRB2-related epilepsy ranges broadly in severity from genetic generalized epilepsy to developmental and epileptic encephalopathies. Developmental disability and movement disorder are key features. The phenotypic spectrum is comparable to other GABAA receptor-encoding genes. Phenotypic severity varies by protein domain. Experimental evidence supports loss of GABAergic inhibition as the mechanism underlying GABRB2-associated neurodevelopmental disorders. ANN NEUROL 2021;89:573-586.


Asunto(s)
Epilepsia/fisiopatología , Trastornos del Movimiento/fisiopatología , Trastornos del Neurodesarrollo/fisiopatología , Receptores de GABA-A/genética , Adolescente , Adulto , Animales , Ataxia/genética , Ataxia/fisiopatología , Atetosis/genética , Atetosis/fisiopatología , Niño , Preescolar , Corea/genética , Corea/fisiopatología , Estudios de Cohortes , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/fisiopatología , Epilepsia Refractaria/genética , Epilepsia Refractaria/fisiopatología , Distonía/genética , Distonía/fisiopatología , Epilepsia/genética , Femenino , Genotipo , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Masculino , Persona de Mediana Edad , Trastornos del Movimiento/genética , Mutación Missense , Trastornos del Neurodesarrollo/genética , Oocitos , Técnicas de Placa-Clamp , Fenotipo , Dominios Proteicos/genética , Xenopus laevis , Adulto Joven
2.
Sci Rep ; 9(1): 5591, 2019 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-30944364

RESUMEN

The intent of this study was to investigate if cerebrospinal fluid (CSF) from autoimmune encephalitis (AE) patients regulates in vitro neuronal network activity differentially to healthy human control CSF (hCSF). To this end, electrophysiological effects of CSF from AE patients or hCSF were measured by in vitro neuronal network activity (ivNNA) recorded with microelectrode arrays (MEA). CSF from patients with either N-methyl-D-aspartate-receptor-antibody (pCSFNMDAR, n = 7) or Leucine-rich-glioma-inactivated-1-Ab (pCSFLGI1, n = 6) associated AE suppressed global spiking activity of neuronal networks by a factor of 2.17 (p < 0.05) or 2.42 (p < 0.05) compared to hCSF. The former also suppressed synchronous network bursting by a factor of 1.93 (p < 0.05) in comparison to hCSF (n = 13). As a functional diagnostic test, this parameter reached a sensitivity of 86% for NMDAR-Ab- and 100% for LGI1-Ab-associated AE vs. hCSF at a specificity of 85%. To explore if modulation at the NMDAR influences effects of hCSF or pathological CSF, we applied the NMDAR-antagonist 2-Amino-5-phosphono-pentanoic acid (AP5). In CSF from NMDAR-Ab-associated AE patients, spike rate reduction by AP5 was more than 2-fold larger than in hCSF (p < 0.05), and network burst rate reduction more than 18-fold (p < 0.01). Recording ivNNA might help discriminating between functional effects of CSF from AE patients and hCSF, and thus could be used as a functional diagnostic test in AE. The pronounced suppression of ivNNA by CSF from NMDAR-Ab-associated AE patients and simultaneous antagonism at the NMDAR by AP5, particularly in burst activity, compared to hCSF plus AP5, confirms that the former contains additional ivNNA-suppressing factors.


Asunto(s)
Líquido Cefalorraquídeo/fisiología , Encefalitis/diagnóstico , Encefalitis/patología , Enfermedad de Hashimoto/diagnóstico , Enfermedad de Hashimoto/patología , Sistema Nervioso/patología , Neuronas/patología , 2-Amino-5-fosfonovalerato/farmacología , Animales , Encefalitis Antirreceptor N-Metil-D-Aspartato/diagnóstico , Encefalitis Antirreceptor N-Metil-D-Aspartato/tratamiento farmacológico , Líquido Cefalorraquídeo/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Encefalitis/metabolismo , Femenino , Enfermedad de Hashimoto/tratamiento farmacológico , Enfermedad de Hashimoto/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Embarazo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores
3.
Lancet Neurol ; 17(8): 699-708, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30033060

RESUMEN

BACKGROUND: Genetic generalised epilepsy is the most common type of inherited epilepsy. Despite a high concordance rate of 80% in monozygotic twins, the genetic background is still poorly understood. We aimed to investigate the burden of rare genetic variants in genetic generalised epilepsy. METHODS: For this exome-based case-control study, we used three different genetic generalised epilepsy case cohorts and three independent control cohorts, all of European descent. Cases included in the study were clinically evaluated for genetic generalised epilepsy. Whole-exome sequencing was done for the discovery case cohort, a validation case cohort, and two independent control cohorts. The replication case cohort underwent targeted next-generation sequencing of the 19 known genes encoding subunits of GABAA receptors and was compared to the respective GABAA receptor variants of a third independent control cohort. Functional investigations were done with automated two-microelectrode voltage clamping in Xenopus laevis oocytes. FINDINGS: Statistical comparison of 152 familial index cases with genetic generalised epilepsy in the discovery cohort to 549 ethnically matched controls suggested an enrichment of rare missense (Nonsyn) variants in the ensemble of 19 genes encoding GABAA receptors in cases (odds ratio [OR] 2·40 [95% CI 1·41-4·10]; pNonsyn=0·0014, adjusted pNonsyn=0·019). Enrichment for these genes was validated in a whole-exome sequencing cohort of 357 sporadic and familial genetic generalised epilepsy cases and 1485 independent controls (OR 1·46 [95% CI 1·05-2·03]; pNonsyn=0·0081, adjusted pNonsyn=0·016). Comparison of genes encoding GABAA receptors in the independent replication cohort of 583 familial and sporadic genetic generalised epilepsy index cases, based on candidate-gene panel sequencing, with a third independent control cohort of 635 controls confirmed the overall enrichment of rare missense variants for 15 GABAA receptor genes in cases compared with controls (OR 1·46 [95% CI 1·02-2·08]; pNonsyn=0·013, adjusted pNonsyn=0·027). Functional studies for two selected genes (GABRB2 and GABRA5) showed significant loss-of-function effects with reduced current amplitudes in four of seven tested variants compared with wild-type receptors. INTERPRETATION: Functionally relevant variants in genes encoding GABAA receptor subunits constitute a significant risk factor for genetic generalised epilepsy. Examination of the role of specific gene groups and pathways can disentangle the complex genetic architecture of genetic generalised epilepsy. FUNDING: EuroEPINOMICS (European Science Foundation through national funding organisations), Epicure and EpiPGX (Sixth Framework Programme and Seventh Framework Programme of the European Commission), Research Unit FOR2715 (German Research Foundation and Luxembourg National Research Fund).


Asunto(s)
Epilepsia Generalizada/genética , Secuenciación del Exoma/métodos , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Receptores de GABA-A/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Niño , Preescolar , Estudios de Cohortes , Epilepsia Generalizada/etnología , Europa (Continente) , Salud de la Familia , Femenino , Humanos , Lactante , Recién Nacido , Cooperación Internacional , Masculino , Persona de Mediana Edad , Modelos Moleculares , Adulto Joven
4.
Brain ; 140(11): 2879-2894, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29053855

RESUMEN

Genetic epilepsies are caused by mutations in a range of different genes, many of them encoding ion channels, receptors or transporters. While the number of detected variants and genes increased dramatically in the recent years, pleiotropic effects have also been recognized, revealing that clinical syndromes with various degrees of severity arise from a single gene, a single mutation, or from different mutations showing similar functional defects. Accordingly, several genes coding for GABAA receptor subunits have been linked to a spectrum of benign to severe epileptic disorders and it was shown that a loss of function presents the major correlated pathomechanism. Here, we identified six variants in GABRA3 encoding the α3-subunit of the GABAA receptor. This gene is located on chromosome Xq28 and has not been previously associated with human disease. Five missense variants and one microduplication were detected in four families and two sporadic cases presenting with a range of epileptic seizure types, a varying degree of intellectual disability and developmental delay, sometimes with dysmorphic features or nystagmus. The variants co-segregated mostly but not completely with the phenotype in the families, indicating in some cases incomplete penetrance, involvement of other genes, or presence of phenocopies. Overall, males were more severely affected and there were three asymptomatic female mutation carriers compared to only one male without a clinical phenotype. X-chromosome inactivation studies could not explain the phenotypic variability in females. Three detected missense variants are localized in the extracellular GABA-binding NH2-terminus, one in the M2-M3 linker and one in the M4 transmembrane segment of the α3-subunit. Functional studies in Xenopus laevis oocytes revealed a variable but significant reduction of GABA-evoked anion currents for all mutants compared to wild-type receptors. The degree of current reduction correlated partially with the phenotype. The microduplication disrupted GABRA3 expression in fibroblasts of the affected patient. In summary, our results reveal that rare loss-of-function variants in GABRA3 increase the risk for a varying combination of epilepsy, intellectual disability/developmental delay and dysmorphic features, presenting in some pedigrees with an X-linked inheritance pattern.


Asunto(s)
Encefalopatías/genética , Fisura del Paladar/genética , Discapacidades del Desarrollo/genética , Epilepsia/genética , Facies , Discapacidad Intelectual/genética , Nistagmo Patológico/genética , Receptores de GABA-A/genética , Adolescente , Adulto , Animales , Niño , Preescolar , Femenino , Variación Genética , Humanos , Masculino , Microcefalia/genética , Mutagénesis Sitio-Dirigida , Oocitos/metabolismo , Técnicas de Placa-Clamp , Linaje , Receptores de GABA-A/metabolismo , Síndrome , Xenopus laevis , Adulto Joven , Ácido gamma-Aminobutírico/metabolismo
5.
Mol Brain ; 10(1): 30, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28716058

RESUMEN

A hallmark of temporal lobe epilepsy (TLE) is hippocampal neuronal demise and aberrant mossy fiber sprouting. In addition, unrestrained neuronal activity in TLE patients induces gene expression including immediate early genes (IEGs) such as Fos and Egr1.We employed the mouse pilocarpine model to analyze the transcription factor (TF) serum response factor (SRF) in epileptogenesis, seizure induced histopathology and IEG induction. SRF is a neuronal activity regulated TF stimulating IEG expression as well as nerve fiber growth and guidance. Adult conditional SRF deficient mice (Srf CaMKCreERT2 ) were more refractory to initial status epilepticus (SE) acquisition. Further, SRF deficient mice developed more spontaneous recurrent seizures (SRS). Genome-wide transcriptomic analysis uncovered a requirement of SRF for SE and SRS induced IEG induction (e.g. Fos, Egr1, Arc, Npas4, Btg2, Atf3). SRF was required for epilepsy associated neurodegeneration, mossy fiber sprouting and inflammation. We uncovered MAP kinase signaling as SRF target during epilepsy. Upon SRF ablation, seizure evoked induction of dual specific phosphatases (Dusp5 and Dusp6) was reduced. Lower expression of these negative ERK kinase regulators correlated with altered P-ERK levels in epileptic Srf mutant animals.Overall, this study uncovered an SRF contribution to several processes of epileptogenesis in the pilocarpine model.


Asunto(s)
Epilepsia/genética , Epilepsia/patología , Hipocampo/patología , Red Nerviosa/patología , Convulsiones/genética , Convulsiones/patología , Factor de Respuesta Sérica/metabolismo , Transcripción Genética , Animales , Modelos Animales de Enfermedad , Fosfatasas de Especificidad Dual/metabolismo , Epilepsia/inducido químicamente , Regulación de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Fibras Musgosas del Hipocampo/metabolismo , Fibras Musgosas del Hipocampo/patología , Red Nerviosa/metabolismo , Neuronas/metabolismo , Neuronas/patología , Pilocarpina , Reproducibilidad de los Resultados
6.
Neurology ; 87(11): 1140-51, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27521439

RESUMEN

OBJECTIVE: To delineate phenotypic heterogeneity, we describe the clinical features of a cohort of patients with GABRA1 gene mutations. METHODS: Patients with GABRA1 mutations were ascertained through an international collaboration. Clinical, EEG, and genetic data were collected. Functional analysis of 4 selected mutations was performed using the Xenopus laevis oocyte expression system. RESULTS: The study included 16 novel probands and 3 additional family members with a disease-causing mutation in the GABRA1 gene. The phenotypic spectrum varied from unspecified epilepsy (1), juvenile myoclonic epilepsy (2), photosensitive idiopathic generalized epilepsy (1), and generalized epilepsy with febrile seizures plus (1) to severe epileptic encephalopathies (11). In the epileptic encephalopathy group, the patients had seizures beginning between the first day of life and 15 months, with a mean of 7 months. Predominant seizure types in all patients were tonic-clonic in 9 participants (56%) and myoclonic seizures in 5 (31%). EEG showed a generalized photoparoxysmal response in 6 patients (37%). Four selected mutations studied functionally revealed a loss of function, without a clear genotype-phenotype correlation. CONCLUSIONS: GABRA1 mutations make a significant contribution to the genetic etiology of both benign and severe epilepsy syndromes. Myoclonic and tonic-clonic seizures with pathologic response to photic stimulation are common and shared features in both mild and severe phenotypes.


Asunto(s)
Epilepsia/genética , Mutación , Receptores de GABA-A/genética , Adolescente , Adulto , Animales , Encéfalo/fisiopatología , Niño , Preescolar , Estudios de Cohortes , Epilepsia/fisiopatología , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Masculino , Potenciales de la Membrana/fisiología , Persona de Mediana Edad , Oocitos , Fenotipo , Receptores de GABA-A/metabolismo , Xenopus laevis , Ácido gamma-Aminobutírico/metabolismo
7.
J Exp Med ; 212(10): 1529-49, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26347470

RESUMEN

Inactivating mutations of the NF-κB essential modulator (NEMO), a key component of NF-κB signaling, cause the genetic disease incontinentia pigmenti (IP). This leads to severe neurological symptoms, but the mechanisms underlying brain involvement were unclear. Here, we show that selectively deleting Nemo or the upstream kinase Tak1 in brain endothelial cells resulted in death of endothelial cells, a rarefaction of brain microvessels, cerebral hypoperfusion, a disrupted blood-brain barrier (BBB), and epileptic seizures. TAK1 and NEMO protected the BBB by activating the transcription factor NF-κB and stabilizing the tight junction protein occludin. They also prevented brain endothelial cell death in a NF-κB-independent manner by reducing oxidative damage. Our data identify crucial functions of inflammatory TAK1-NEMO signaling in protecting the brain endothelium and maintaining normal brain function, thus explaining the neurological symptoms associated with IP.


Asunto(s)
Encéfalo/irrigación sanguínea , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Circulación Cerebrovascular/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Epilepsia/genética , Femenino , Quinasa I-kappa B/metabolismo , Incontinencia Pigmentaria/metabolismo , Incontinencia Pigmentaria/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Quinasa Quinasa PAM/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ocludina/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción ReIA/metabolismo
8.
J Neurochem ; 115(2): 450-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20681951

RESUMEN

Nucleotides act as early signals for microglial recruitment to sites of CNS injury. As microglial motility and activation can be influenced by several local factors at the site of the lesion, we investigated the effects of interferon-gamma, lipopolysaccharide (LPS) or transforming growth factor-ß (TGF-ß) addition to mixed glial cell cultures, on microglial migration in response to ADP, P2Y12 and P2Y1 mRNA expression as well as on the expression of an array of genes associated with the process of microglial activation. First, we demonstrated, by pharmacological inhibition and by using small interfering RNAs, that in addition to P2Y12, P2Y1 is involved in ADP-stimulated microglial migration. The ability of specific agonists to induce Ca(2+) mobilization further confirmed the expression of functional P2Y receptors in microglia. Then, we found that migratory capability and expression of both P2Y receptors were abrogated in microglial cells from LPS-stimulated mixed glial cultures, while TGF-ß increased ADP-induced migration and the expression of P2Y12 and P2Y1 receptors. Interferon-gamma did not influence receptor expression or microglial migration. Finally, the patterns of gene expression induced in microglia by LPS or TGF-ß treatment of mixed glial cultures were clearly distinct. LPS induced a set of classical pro-inflammatory genes, whereas TGF-ß increased the expression of genes associated with atypical microglial phenotype, namely arginase-1 and TGF-ß genes. These results imply that both P2Y1 and P2Y12 may guide microglia toward the lesion. They also suggest that the modulation of microglial purinergic receptors expression by local factors, through direct and/or astrocyte-mediated actions, may represent a novel mechanism affecting neuroinflammatory response.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Animales Recién Nacidos , Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Regulación de la Expresión Génica/efectos de los fármacos , Interferón gamma/farmacología , Microglía/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y1/genética , Receptores Purinérgicos P2Y12 , Tionucleótidos/farmacología , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA